You are on page 1of 1495

DORFMAN and CZERNIAK’S

BONE TUMORS
ERRNVPHGLFRVRUJ
SECOND EDITION

DORFMAN and CZERNIAK’S

BONE
TUMORS
ERRNVPHGLFRVRUJ
Bogdan Czerniak, MD, PhD
Professor of Pathology
Department of Pathology
The University of Texas MD Anderson Cancer Center
Houston, Texas
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

DORFMAN AND CZERNIAK’S BONE TUMORS  ISBN: 978-0-323-02396-2


Copyright © 2016 by Saunders, an imprint of Elsevier Inc.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic
or mechanical, including photocopying, recording, or any information storage and retrieval system,
without permission in writing from the publisher. Details on how to seek permission, further
information about the Publisher’s permissions policies and our arrangements with organizations such as
the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website:
www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the
Publisher (other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical
treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in
evaluating and using any information, methods, compounds, or experiments described herein. In
using such information or methods they should be mindful of their own safety and the safety of
others, including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the
most current information provided (i) on procedures featured or (ii) by the manufacturer of each
product to be administered, to verify the recommended dose or formula, the method and duration
of administration, and contraindications. It is the responsibility of practitioners, relying on their own
experience and knowledge of their patients, to make diagnoses, to determine dosages and the best
treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products,
instructions, or ideas contained in the material herein.

Library of Congress Cataloging-in-Publication Data

Czerniak, Bogdan, author.


  Dorfman and Czerniak’s bone tumors / Bogdan Czerniak. – 2nd edition.
   p. ; cm.
  Bone tumors
  Preceded by Bone tumors / Howard D. Dorfman, Bogdan Czerniak. c1998.
  Includes bibliographical references and index.
  ISBN 978-0-323-02396-2 (hardcover : alk. paper)
  I.  Dorfman, Howard D. Bone tumors. Preceded by (work):  II.  Title.  III.  Title: Bone tumors.
  [DNLM: 1. Bone Neoplasms. WE 258]
  RC645.7
  616.99′441–dc23
  2014038002

Senior Content Strategist: William Schmitt


Content Development Specialist: Amy Meros
Publishing Services Manager: Patricia Tannian
Project Manager: Ted Rodgers
Designer: Margaret Reid

Printed in China

Last digit is the print number:  9  8  7  6  5  4  3  2  1


To my wife Elzbieta with love

Portrait of Elzbieta Czerniak by Gretchen Van Atta Loro


Preface

More than a decade has passed since the advent of the Henry L. Jaffe is considered the conceptual founder
first edition of Bone Tumors. During that time, we have of modern skeletal pathology. I did not have an opportu-
witnessed the emergence of genomic medicine, which has nity to work with him, but his diagnostic philosophy was
significantly affected clinical practice, including the diag- transmitted to me by Howard Dorfman and is evident in
nosis and treatment of tumors that arise in the skeleton. both editions of the book. My contribution to this phi-
The second edition attempts to address this change but losophy is the extension of the diagnostic concepts to
does not neglect the continuation of nearly 50 years of molecular techniques that, similar to other aspects of
combined experience for the authors of both editions in pathology, require in-depth knowledge and a critical
the conventional practice of skeletal pathology. approach and should be considered in synchrony with
Similar to the first edition, the second edition is based other clinical, radiographic, and pathological features
on large databases of bone tumors from several sources. of the lesion in question in order to be meaningful.
Pathologic and clinical data on 11,500 benign and malig- All molecular and genetic annotations included in the
nant bone tumors from patients treated and followed at book are from the University of California, Santa Cruz
the University of Texas MD Anderson Cancer Center Genome Browser (http://genome.ucsc.edu/). The muta-
in Houston, Texas, represent the core database for this tions in genes are annotated according to the Catalogue
edition. The data on 8,417 malignant bone tumors of Somatic Mutations in Cancer (COSMIC, http://
entered into the National Cancer Institute’s Surveillance, cancer.sanger.ac.uk/cancergenome/projects/cosmic/).
Epidemiology, and End Result (SEER) program (www. The genetic locations and the information concerning
seer.cancer.gov) were analyzed and included in the text as the structure of the genes are according to the Gene-
reflecting malignant bone tumor epidemiology in the Cards Database of Human Genes (http://www.genecards.
U.S. population. Most important, I was given free access org/). The reader is encouraged to use these and other
to Dr. Howard Dorfman’s consultation files, comprising website-based tools in studying the molecular data
more than 9,500 cases, which were used extensively for included in this text for the most updated information,
the new edition. The resulting database totaling nearly which is arriving at an exponential rate.
30,000 primary bone tumors and tumor-like lesions was The second edition is significant for the inclusion
used to provide the reader with the most complete infor- of four chapters written by five additional authors:
mation on the epidemiologic, clinical, pathologic, and Chapter 2, “Clinical Considerations and Imaging of Bone
molecular aspects of bone tumors, even those that are Tumors,” by Colleen M. Costelloe and John E. Madewell;
extremely rare. Chapter 12, “Hematopoietic Tumors,” by April Ewton;
My route to skeletal pathology, to which I was intro- Chapter 14, “Neurogenous Tumors and Neurofibroma-
duced multiple years into my practice, was full of enlight- tosis Affecting Bone,” by Gregory N. Fuller; and Chapter
ening experiences and charismatic influences. I am 19, “Metastatic Tumors of Bone,” by John D. Reith. The
fortunate and proud to include among my initial teachers remaining chapters were extensively reviewed to provide
Stanisław Woyke, Maria Dąbska, and Wenancjusz updated photographic documentation. I am particularly
Domagała, from Poland, and Leopold Koss, Juan Rosai, in debt to Colleen Costelloe, who spent endless hours
and Alberto Ayala, in the United States. However, by far assisting me in updating the radiographic imaging data
the most significant influence, specifically related to for virtually every chapter in this text. Particular atten-
skeletal pathology, was that of Howard Dorfman. It was tion was given to provide the reader with new molecular
a fortunate moment when I walked into his office in and genomic data that are relevant in the current diag-
Montefiore Medical Center in 1988 and expressed my nostic workup.
interest in learning skeletal pathology. He immediately The extensive photographic documentation and
gave me a recent case sent in consultation, and my molecular diagrams necessitated computer processing
lessons began. During the following years, we reviewed and were diligently performed by Kim-Anh T. Vu, a
together most of his extensive consultation files, which computer graphic designer in our department. Multiple
provided the foundation for the first edition of Bone molecular and epidemiologic diagrams for the second
Tumors. His full retirement, several years ago, necessi- edition were prepared by Jolanta Bondaruk and Tadeusz
tated the change of authorship for the second edition; it Majeweski, researchers in my laboratory. Extensive revi-
was decided that I would be the sole author but the book sions of the text were prepared by Stephanie Garza and
would be titled Dorfman and Czerniak’s Bone Tumors to Virginia Hurley. Ms. Hurley also served as an editor to
honor his contributions. the endless versions of the text.
vi
Preface vii

I would also like to acknowledge the assistance of the Practicing pathology in the genomic era necessitates
editorial staff at Elsevier, who tolerated the long process the education of a new generation of pathologists to be
of revision with limited complaints and provided every equally vested in clinical, radiographic, and microscopic
possible assistance to the author to fulfill the vexing details and who are also capable of critical implementa-
mission of the multiple revisions with an attempt to bring tion of molecular data. I hope that Bone Tumors will con-
the reader the most current information available. In tribute to this mission.
particular, I would like to recognize the encouraging col-
laborations with William Schmitt and Amy Meros in the Bogdan Czerniak
revision process as well as with Ted Rodgers during the
final production of the book.
Foreword

Since the publication of the first edition of this book in tumor classification and diagnosis. Dr. Henry L. Jaffe
1998, much new information has become available con- (1896-1979) and Dr. Louis Lichtenstein (1906-1977)
cerning the neoplasms that affect the skeleton. It seems based their groundbreaking contributions to bone tumors
obvious that the greatest advance in our understanding on their understanding of the nature and interrelation-
of these perplexing tumors should have come about ships of bone tumors, a profound knowledge of these
through the explosive expansion of knowledge in the tumors’ clinical features, follow-up data, radiographic
fields of molecular and cellular biology of skeletal tissue correlation using mainly plain radiographs and a primi-
and of the mechanisms of malignant transformation. tive form of tomography, hematoxylin, eosin slide mor-
These dramatic changes have occurred at such a dizzying phology, and a rudimentary array of special stains. During
pace that any textbook treatment of such a complex the following years, electron microscopy was available
subject would be at risk of becoming obsolete in short but rarely used for diagnostic purposes, and cytogenetic
order. or molecular biologic methods that could be readily
The revision of this work has been carried out over applied to pathologic diagnosis, computed tomography,
the last several years largely through the efforts of my magnetic resonance imaging, and PET scanning were
colleague Bogdan Czerniak, whose intellectual vigor still far in the future. It is worth noting that as recently
and generosity of spirit know no bounds. I have offered as the 1970s the only special staining method in wide use
suggestions as to changes in content and have provided for the differential diagnosis of bone neoplasms was the
some of the examples of newly recognized or reinter- PAS stain for demonstrating intracytoplasmic glycogen
preted entities derived from my ongoing consultation in the cells of Ewing’s sarcoma. The introduction of the
practice and the accumulated case material in my files. greatly useful and dramatically informative methods of
The updating and elaboration of new knowledge immunohistochemistry did not occur until the 1980s.
addressed in each of the twenty-four chapters has finally The then-available cryostat for frozen section diagnosis
come to fruition, with the inclusion of some newly written was a rudimentary prototype model.
chapters. These were produced by five authors, specifi- Fortunately, my long career as a surgical pathologist
cally chosen and invited to do so because of their special dealing with bone neoplasms has afforded me the oppor-
expertise in selected areas related to bone neoplasia. It is tunity to witness profound changes for the better in
clear to me that their contributions, as well as the exten- almost every respect. Not only are we better equipped
sive reworking and updating of the remainder of the and informed about the diagnostic features and biologic
existing text through the exhaustive efforts of my col- potential of these rare tumors, but the five-year survival
league Dr. Czerniak, have substantially enhanced and rates after precise diagnosis rise with each new innova-
updated the material covered in this new and more tion. The major turning points in treatment were the
weighty volume. introduction of multidrug neoadjuvant chemotherapy
At this point, I would like to pause to reflect on the and limb-salvage surgery. In modern times, these are
remarkable advances in our knowledge of this vast and complemented with the concepts of targeted therapy
complex discipline, the changes that have taken place in based on genomic profiles.
the field of surgical pathology of bone tumors and related The former edition of this book was based, in part, on
disorders that I personally have encountered during the my consultation files, and recently the entire collection,
more than fifty years of my own practice. I am proud to now totaling more than 9,500 case files, was donated to
include among my teachers and professional collabora- the Departments of Radiology and Orthopaedic Surgery,
tors Paul Klemperer, Sadao Otani, Arthur Purdy Stout, at Montefiore Medical Center, supervised by Dr. Nogah
Raffaele Lattes, and Lauren V. Ackerman. But by far, the Haramati and Dr. David Geller. It is my fervent hope that
most powerful influence upon my work and later profes- this large database will be utilized for many research
sional life has been that of Henry Jaffe. projects and studies supporting new publications in the
I arrived at the Hospital for Joint Diseases where Dr. fields of orthopaedic oncology, muculoskeletal radiology,
Jaffe was retiring after a 40-year career as Chief of Pathol- and pathology.
ogy and, though well trained but modestly experienced In approaching the assigned task of composing a fore-
as a surgical pathologist, I was faced with the rather word to this book, what seemed initially to be a reason-
daunting task of attempting to follow in his footsteps. ably routine one, has proved to be an emotionally
During his career he had succeeded in revolutionizing challenging effort that was fraught with self-revelation.
the understanding of bone tumors and had coined the In looking backward over almost six decades of study
names for many neoplasms. He and Dr. Lichtenstein of the entities described here, it became clear that
became the world’s leading experts on the subject of bone one’s ever-changing, always-evolving view of a specific
viii
Foreword ix

neoplasm grows with the accretion of novel bits of evi- practical background knowledge for precise diagnosis of
dence revealed by each new technological method that these relatively rare and difficult neoplasms, but also as a
we apply to its examination. For me, to have participated basis for appropriate and effective patient care.
in this process, even in a small way as an engaged observer
and over such an extended span of time, has been a Howard D. Dorfman, MD
sometimes daunting but always fascinating experience. Professor Emeritus of Pathology,
It is my fervent hope that the present volume will serve Orthopedic Surgery and Radiology
not only as a comprehensive reference source for that Albert Einstein College of
part of the medical community which is faced with the Medicine of Yeshiva University
need for an in-depth treatment of the theoretical and New York, New York
Preface to the First Edition

This book represents the distillation of more than 30 and End Results program from 1973 to 1986 were ana-
years of experience in the diagnosis of benign and malig- lyzed and used as a representative sample of bone neo-
nant bone lesions. It is an attempt to bring into focus the plasms occurring in the American population. In addition,
lessons learned through long exposure to vexing clinical pathologic and clinical data on 4904 benign and primary
and radiologic challenges. We have attempted to place malignant bone tumors from patients treated and fol-
this body of knowledge into the context of modern surgi- lowed at M.D. Anderson Cancer Center were extensively
cal pathology by the integration of clinicopathologic, reviewed. (Cases seen only in consultation were not
immunohistochemical, ultrastructural, and molecular included.) The resulting compilation of data on 13,403
diagnostic techniques. primary bone tumors and tumorlike lesions from several
The present volume is based on several large databases major sources has been drawn on to provide the reader
composed of personally analyzed cases. The core data- with comprehensive information on various epidemio-
base was the consultation file consisting of 5872 cases logic, clinical, and pathologic aspects of bone neoplasms,
from one of the authors. During the course of this project, even those that are extremely rare.
two additional large case files became available to us. The The acquisition of many of the cases on which the text
data on 2627 malignant bone tumors entered into the and illustrations are based could not have been possible
National Cancer Institute’s Surveillance, Epidemiology, without the cooperation of the following colleagues:

Claus-Peter Adler, MD Timothy A. Jennings, MD Margaret Nuovo, MD


Seena C. Aisner, MD Sonny L. Johansson, MD Nelson G. Ordóñez, MD
Lennart Angervall, MD Craig P. Jones, MD H. Ostertag, MD
Norio Azumi, MD Gernot Jundt, MD Antonio Perez-Atayde, MD
Fidelis A. Barba, MD Leonard B. Kahn, MD A. Kevin Raymond, MD
Thomas Bauer, MD, PhD Bridget C. Kahntroff, MD Wolfgang Remagen, MD
Morgan Berthrong, MD Robert S. Katz, MD Jae Y. Ro, MD, PhD
Belur S. Bhagavan, MD Richard L. Kempson, MD Andrew E. Rosenberg, MD
Peter G. Bullough, MD Demetrios E. Kepas, MD Joel A. Roth, MD
Jane Chatten, MD Peter Klacsmann, MD John M. Rowland, MD, PhD
Janet B. Christman, MD Michael J. Klein, MD C. Peter Schwinn, MD
Robert W. Cihak, MD Carl T. Koenen, MD Marco Semiglia, MD
Karen R. Cleary, MD Harry Kozakewich, MD Hubert A. Sissons, MD
Robert A. Colyer, MD Donald A. Kristt, MD Richard Slavin, MD
Katrina A. Conard, MD Dhruv Kumar, MD Suzane Spanier, MD
Harry C. Cooper, MD Jack P. Lawson, MD German C. Steiner, MD
Jose Costa, MD Alan M. Levine, MD Masayuki Takagi, MD
F. Gonzalez Crussi, MD Mario Luna, MD Jerome B. Taxy, MD
Raffaele David, MD William Martel, MD Felix Tchang, MD
Elina Donskoy, MD Takeo Matsuno, MD Henry Tesluk, MD
Tadashi Hasegawa, MD James A. McAlister, MD Timothy J. Triche, MD
Jack Hasson, MD Edward F. McCarthy, MD Maria Tsokos, MD
K. P. Heidelberger, MD Maria J. Merino, MD Masazumi Tsuneyoshi, MD
William Hicken, MD Markku Miettinen, MD Bruce L. Webber, MD
A. R. Von Hochstetter, MD Sara Milchgrub, MD Sharon W. Weiss, MD
Herbert Ichinose, MD Mark L. Mitchell, MD Gary B. Witkin, MD
Tetsuo Imamura, MD Artemis Nash, MD Benjamin Wittels, MD
Tsuyoshi Ishida, MD Takayuki Nojima, MD Marianne Wolfe, MD
x
Preface to the First Edition xi

The subspecialty of surgical pathology which is in solitude, and we would therefore like to express our
devoted to the study of bone tumors is a relatively recent appreciation to the following for their encouragement
development. It was not until the 1940s, a mere 50 years and support: Alberto G. Ayala, MD; John G. Batsakis,
ago, that meaningful classification systems and precise MD; Edward T. Habermann, MD; and Harold G. Jacob-
terminology were applied to bone and cartilage neo- son, MD.
plasms. The authors of this book were fortunate in having Several of our colleagues reviewed the chapters per-
had the benefit of direct contact, as students, with some taining to their fields of expertise. We would like to thank
of the more celebrated figures in this field. It is often T.J. McDonnell, MD, PhD, and G. Karsenty, MD, PhD,
stated that a teacher’s role should be limited to clearly for the review of Chapter 3, “Molecular Biology of Bone
showing the pupil the goal that science sets for itself and Tumors,” and John T. Manning, MD, for his comments
to pointing out all possible means for reaching it. Our on Chapter 12, “Immunohematopoietic Tumors.” Janet
teachers have done this while leaving us free to move M. Bruner, MD, provided unlimited access to her
about in our own way to reach our goals. They have never collection of neuropathologic specimens and reviewed
acted as the promoters of absolute and immutable truth. Chapter 14, “Neurogenous Tumors and Neurofibroma-
Some of our concepts expressed in the following pages tosis Affecting Bone.” The presentation of ultrastructural
have aroused controversy, but we have been inspired by features of many bone tumors was made possible by
our mentors to push our ideas to their full development, Bruce Mackay, MD, PhD, who provided generous unlim-
provided that we are careful to test them by experiment ited access to his enormous collection of electron micro-
and by experience. We would like to acknowledge the scopic specimens.
grace and magnanimity of our teachers. The senior Extensive use of digitized imaging and computer
author’s debt to his principal mentors, Henry L. Jaffe, graphics was accomplished with the creative techni-
MD; Arthur Purdy Stout, MD; Raffaele Lattes, MD; and cal assistance of Vernon Durke II and Ian Suk. The
Lauren V. Ackerman, MD; cannot be overstated, and it monumental task of printing hundreds of radiologic
was with the persistent encouragement of the late Dr. and photomicrographic images was performed by Elsa
Ackerman that this effort was undertaken. The junior Ramos and Lydia Shanks. Dianne Crawley-Paolillo and
author would like to thank Stanisław Woyke, MD; Donna Sprabary provided skillful secretarial assistance
Leopold G. Koss, MD; and Juan Rosai, MD, who signifi- and patiently typed endless versions of the text. The
cantly contributed to his professional skills and inspired difficult task of editing the manuscript and correcting
his academic interests. Dr. Koss’ role as a principal many errors and inconsistencies was accomplished by
mentor and his inspirational role before and during the Kimberly Herrick. Finally, Mosby’s representatives,
conduct of this project are acknowledged with particular Lynne Gery and Pat Joiner, guided us through tedious
pleasure. stages of book development and production, gracefully
A fortunate concurrence of informal events brought enduring consistent delays in completion of various
the authors of this book together approximately 10 years chapters.
ago. It quickly became evident that we have complemen- The reader will be the ultimate judge of the quality of
tary cultural and professional interests, and our initial this work. The contributors of case material are relieved
interaction rapidly evolved into a professional partner- of all responsibility for any errors of fact, taste, or judg-
ship and friendship. One hardly thanks a partner for ment, which are entirely our own.
partnership and a friend for friendship, but we wish to
place on record our mutual sense of fulfillment in the Howard D. Dorfman
completion of this project. This work was not carried out Bogdan Czerniak
Contributors

Colleen M. Costelloe, MD
Associate Professor of Radiology
Department of Diagnostic Radiology
The University of Texas MD Anderson Cancer Center
Houston, Texas

April Ewton, MD
Associate Professor of Pathology
Associate Medical Director of Hematopathology
Houston Methodist Hospital
Department of Pathology and Genomic Medicine
Weill Cornell Medical College of Cornell University
Houston, Texas

Gregory N. Fuller, MD, PhD


Professor and Chief Neuropathologist
Department of Pathology
The University of Texas MD Anderson Cancer Center
Houston, Texas

John E. Madewell, MD
Professor and Chief of Musculoskeletal Imaging
Department of Diagnostic Radiology
The University of Texas MD Anderson Cancer Center
Houston, Texas

John D. Reith, MD
Professor of Pathology
Departments of Pathology, Immunology, Laboratory Medicine and
Orthopaedics and Rehabilitation
Shands Hospital
University of Florida College of Medicine
Gainesville, Florida

xii
C H A P T E R 1 

General Considerations

CHAPTER OUTLINE

MORPHOLOGY OF NORMAL BONE SPECIAL STAINS


PROCESSING OF BONE SPECIMENS ELECTRON MICROSCOPY
STAGING, GRADING, AND REPORTING OF BONE CYTOMETRY AND HISTOMORPHOMETRY
TUMORS
IMMUNOHISTOCHEMISTRY
GENERAL EPIDEMIOLOGY OF BONE TUMORS
CYTOGENETICS
SPECIAL TECHNIQUES

INTRODUCTION changed the fact that histologic and cytologic character-


istics are the basis for classifying bone tumors. Although
The new edition of this book retains the conventional radiologic features can provide valuable clues about
approach to the classification of bone tumors, dividing predisposing conditions and mineralization or growth
them into the categories of osteoblastic, chondroblastic, patterns, ultimately bone tumors are microscopically cat-
fibrous and fibroosseous, vascular and neurogenic, and egorized on the basis of cell type and matrix production.
others of mesenchymal tissue derivation. In general, the The histogenesis of the tumor usually can be deduced
discussion of the lesions is similar but not identical to from the cell morphology and whether collagen, osteoid,
the World Health Organization’s classification of bone or cartilage matrix production can be identified.
tumors (Table 1-1). The past decade has been marked The need, originally stressed by Jaffe,1 to regard bone
by major advancements in our understanding of the tumors as clinicopathologic entities whose behavior and
molecular biology of sarcomas, and some of these new biologic potential are affected by other clinical factors
developments are of diagnostic as well as of potential such as patient age, location in a particular bone or part
therapeutic significance. The identification of recurrent of a bone, multicentricity, and association with other
chromosomal translocations in many sarcomas, and their (underlying) conditions, is reaffirmed. The familiar diag-
application in the differential diagnosis of these tumors, nostic triangle recommended by Jaffe—the surgeon,
is changing the paradigm of both pathologic and clinical radiologist, and pathologist sharing their points of view
practice. It is surprising and at the same time insightful on a bone lesion to arrive at a rational diagnosis—is still
that such clonal chromosomal translocations and their valid. This approach, which avoids overemphasis on one
respective hybrid genes have been identified in several aspect of a tumor’s presentation (often leading to dispa-
bone and soft tissue lesions that were traditionally con- rate opinions), remains as a standard of practice in
sidered to be reactive in nature. Dramatic advancement in skeletal pathology. However, the advent of molecular
our understanding of the biology of skeletal-forming cell diagnostic techniques has expanded this diagram into a
lineages such as osteoblastic, chondroblastic, and osteo- diagnostic quadrangle, as shown in Figure 1-1.
clastic has further expanded the armamentarium of genes The approach of the so-called diagnostic quadrangle
and their encoded proteins that may be useful as potential postulates a stepwise, analytic approach in which four
biomarkers in the differential diagnosis of bone tumors. distinctive data sets—clinical, radiographic, microscopic,
As a consequence, many tumors of uncertain histogen- and molecular—are considered to establish the diagnosis.
esis have undergone reassessment due to findings from Although an intuitive approach based on fragmentary
these techniques while the well-defined pathologic enti- data can occasionally be very impressive when used to
ties are undergoing molecular sub-classifications. Ewing’s arrive at a diagnosis, a stepwise, analytic approach is more
sarcoma and the family of small round cell malignancies likely to lead to consistency and accuracy. For that spe-
represent a paradigm for the changing practice in skeletal cific reason, we attempt to describe individual neoplastic
pathology, in which the established diagnostic algorithm lesions of bone with an approach that includes clinicora-
based on clinical, radiologic, and microscopic correla- diographic, pathologic, and molecular correlations. The
tions is now coupled with new molecular approaches to description of most lesions is separated into paragraphs
delineate this still mysterious group of tumors. including epidemiologic, radiographic, gross, and micro-
Introduction of newer techniques of immunohisto- scopic data, and pertinent information on any special
chemistry, molecular pathology, and cytogenetics has not techniques required for identification is also provided.
1
ERRNVPHGLFRVRUJ
2 1  General Considerations

TABLE 1-1 WHO Classification of Tumors of Bone*


Tumor Code Tumor Code
Chondrogenic Tumors Osteofibrous dysplasia
Benign Chondromesenchymal hamartoma
Rosai-Dorfman disease
Osteochondroma 9210/0
Chondroma 9220/0 Fibrogenic Tumors
  Enchondroma 9220/0
  Periosteal chondroma 9221/0 Intermediate (locally aggressive)
Osteochondromyxoma 9211/0 Desmoplastic fibroma of bone 8823/1
Subungual exostosis 9213/0
Bizarre parosteal osteochondromatous proliferation 9212/0 Malignant
Synovial chondromatosis 9220/0 Fibrosarcoma of bone 8810/3

Intermediate (locally aggressive) Fibrohistiocytic Tumors


Chondromyxoid fibroma 9241/0 Benign
Atypical cartilaginous tumor Benign fibrous histiocytoma/non-ossifying fibroma 8830/0
  Chondrosarcoma grade 1 9222/1
Hematopoietic Neoplasms
Intermediate (rarely metastasizing) Malignant
Chondroblastoma 9230/1 Plasma cell myeloma 9732/3
Malignant Solitary plasmacytoma of bone 9731/3
Primary non-Hodgkin lymphoma of bone 9591/3
Chondrosarcoma
  Chondrosarcoma grade 2, grade 3 9220/3 Osteoclastic Giant Cell Rich Tumors
Dedifferentiated chondrosarcoma 9243/3
Benign
Mesenchymal chondrosarcoma 9240/3
Clear cell chondrosarcoma 9242/3 Giant cell lesion of the small bones

Osteogenic Tumors Intermediate (locally aggressive, rarely metastasizing)


Benign Giant cell tumor of bone 9250/1
Osteoma 9180/0 Malignant
Osteoid osteoma 9191/0 Malignant giant cell tumor of bone 9250/3
Intermediate (locally aggressive)
Notochordal Tumors
Osteoblastoma 9200/0
Benign
Malignant Benign notochordal tumor 9370/0
Low-grade central osteosarcoma 9187/3
Malignant
Conventional osteosarcoma 9180/3
  Chondroblastic osteosarcoma 9181/3 Chordoma 9370/3
  Fibroblastic osteosarcoma 9182/3
Vascular Tumors
  Osteoblastic osteosarcoma 9180/3
Telangiectatic osteosarcoma 9183/3 Benign
Small cell osteosarcoma 9185/3 Hemangioma 9120/0
Secondary osteosarcoma 9184/3
Parosteal osteosarcoma 9192/3 Intermediate (locally aggressive, rarely metastasizing)
Periosteal osteosarcoma 9193/3 Epithelioid hemangioma 9125/0
High-grade surface osteosarcoma 9194/3
Malignant
Myogenic Tumors Epithelioid hemangioendothelioma 9133/3
Benign Angiosarcoma 9120/3
Leiomyoma of bone 8890/3 Intermediate (locally aggressive)
Malignant Aneurysmal bone cyst 9260/0
Leiomyosarcoma of bone 8890/3 Langerhans cell histiocytosis
  Monostotic 9752/1
Lipogenic Tumors   Polystotic 9753/1
Benign Erdheim-Chester disease 9750/1
Lipoma of bone 8850/0 Miscellaneous Tumors
Malignant Ewing’s sarcoma 9364/3
Adamantinoma 9261/3
Liposarcoma of bone 8850/3 Undifferentiated high-grade pleomorphic sarcoma 8830/3
Tumors of Undefined Neoplastic Nature of bone
Benign
Simple bone cyst 8818/0
Fibrous dysplasia

International Agency for Research on Cancer (IARC): WHO Classification of tumours of soft tissue and bone, ed 4, Lyon Cedex,
France, 2013 (edited by Fletcher CDM, Bridge JA, Hogendoorn PCW, et al).
*The morphology codes are from the International Classification of Diseases for Oncology (ICD-0) {916A}. Behavior is coded /0 for benign
tumors, /1 for unspecified, borderline or uncertain behavior, /2 for carcinoma in situ and grade III intraepithelial neoplasia, and /3 for
malignant tumors.

ERRNVPHGLFRVRUJ
1  General Considerations 3

Clinical Radiologic The protective function of bone is demonstrated by


presentation features the encasement of several vital organs (lungs, heart, and
central nervous system) and of bone marrow, which is
the source of blood cells. Metabolically, bone represents
a reservoir for several ions, predominantly calcium and
Diagnosis phosphorus. Living bone is a highly labile, dynamic tissue
that is able to respond to a number of metabolic, physi-
cal, and endocrine stimuli. At the same time, its relative
simplicity in terms of structural elements allows bone
Molecular Microscopic to restore itself to its normal function and architecture
data features after injury.
FIGURE 1-1  ■  Analytical approach to diagnosis of bone tumor.
Stepwise, analytic approach depicted as a diagnostic quadran-
gle in which four distinct sets of data (clinical, radiographic, Topographic Features
microscopic, and molecular) are taken into consideration in
establishing a diagnosis. Major topographic regions of the skeleton frequently
used in the description of bone tumors are shown in
Figure 1-2. The skeleton forming the central axis (skull,
The frequency distributions in skeletal areas represent vertebral column, and sacrum) is referred to as the axial
approximate compilations based on findings from several skeleton.The bones of the extremities (including the
major published series. Published data from the Mayo scapula and pelvis) are collectively called the appendicular
Clinic, Memorial Sloan-Kettering Cancer Center, and skeleton. The term acral skeleton designates the bones of
The University of Texas M.D. Anderson Cancer Center the hands and feet. In the axial skeleton lesions involving
have been included in the analysis. In addition, national craniofacial bones form a distinct group separate from
epidemiologic data are provided for the most common those of the vertebral column and sacrum. Similarly, in
malignant bone tumors and are based on the most recent the discussion of neoplastic lesions arising in the scapula
analysis of the National Cancer Institute Surveillance, and pelvis, these sites are grouped with other bones of
Epidemiology, and End Results (SEER) Program. The the trunk. On the basis of their gross appearance, bones
description of most lesions is accompanied by a graphic are divided into two main groups: flat and tubular bones.
presentation of the peak age incidence and their typical In general, the bones of the trunk and craniofacial region,
sites of skeletal involvement. This should help readers such as the skull, scapula, clavicle, pelvis, and sternum,
recognize the most typical clinicoradiographic patterns are classified as flat. The bones of the extremities and the
of most bone tumors and tumorlike lesions. The system ribs are tubular. The tubular bones are further subdivided
of graphic depiction of skeletal distribution patterns orig- into the long bones (e.g., femur, tibia, humerus) and the
inally designed by the Mayo Clinic Group is used with short bones (e.g., phalanges, metatarsals, metacarpals)
some modifications in this book. (Figs. 1-3 and 1-4). The carpal and tarsal bones, as well
The intention is to provide a balanced view of current as the patella, are designated as epiphysioid bones, which
pathogenetic and diagnostic concepts on bone tumors are analogous to the epiphyses of long bones with regard
and tumorlike lesions. In reference to several bone lesions to development and tumor predilection.
the author’s concepts, which may differ from those of The tubular bones have several regions or zones:
others, are presented. In such instances, the controversies 1. Epiphysis: The region between the growth plate
are discussed in some detail. Personal opinions in the and the end of bone in skeletally immature indi-
form of recommendations on the basis of experience as viduals or between the growth plate scar and the
to how to address a particular diagnostic problem are end of the bone in skeletally mature individuals
expressed in interspersed paragraphs entitled “Personal 2. Metaphysis: The region adjacent to the growth
Comments.” plate opposite the epiphysis
3. Diaphysis or shaft: The region between metaphyses
4. Physis: The region of bone corresponding to the
MORPHOLOGY OF NORMAL BONE growth plate.
Knowledge of these terms and their definitions is very
Discussion of the morphology of the skeletal system is useful because many tumors have a predilection for par-
restricted to some basic elements important to the ticular regions of bone (Figs. 1-5 and 1-6).
pathologist and helpful in the understanding of basic
gross, radiographic, and microscopic features of bone
tumors and related conditions described in this text. For
Bone
more comprehensive descriptions of the structure of the Bone, cartilage, and fibrous connective tissue differ in
skeletal system, readers should refer to any of the major their visible appearance and mechanical properties
textbooks and monographs strictly dedicated to this because of the various compositions of their matri-
subject. ces.2,7,8,13,14,17,22,30,40 Dense fibrous connective tissue is
Bone and cartilage represent highly specialized tissues formed of well-oriented bundles of collagen, and its prin-
that perform several functions: mechanical, protective, cipal function is to resist tension. Bone and cartilage must
and metabolic. Mechanically, they provide for the integ- also resist compression, torsion, and bending forces.
rity of overall body structure and body movements. Each bone has a peripheral compact layer known as the

ERRNVPHGLFRVRUJ
4 1  General Considerations

Axial types.34,43,45 In woven bone, the collagen fibers are hap-


hazardly organized and form an irregular framework. In
contrast, in lamellar bone the collagen fibrillary network
has an orderly parallel organization. In general, woven
bone is produced during rapid bone growth or repair,
Craniofacial such as a fracture callus. It represents an immature form
of bone in which osteoid is rapidly deposited and is
gradually remodeled into a mature lamellar form. The
mature lamellar bone, within the cortex, is organized into
several distinct architectural patterns referred to as cir-
cumferential, concentric, and interstitial. The circumferen-
tial lamellar bone forms the outer and inner layer of the
cortex. The concentric lamellar bone forms the bulk of
the so-called haversian or osteon systems within the
cortex. It contains the central canal with blood vessels
Trunk

surrounded by a cylindrical concentric lamellae of bone.


The osteocytes within such systems are also somewhat
concentrically arranged within the lacunae and are con-
nected by dendritic processes extending outside of the
main osteocytes’ bodies via the system of canaliculi that
forms an interconnecting mesh within the mineralized
matrix. Volkmann’s canals course through the cortex at
more perpendicular angles with respect to the haversian
Appendicular

systems and contain the connective tissue and feeding


vessels that eventually branch into the vasculature of the
haversian canals.
Axial The microarchitecture of the mineralized deposit and
Acral
fibrular network is still poorly understood. The recently
developed models postulate the tubular nature of basic
structural units in which the mineralized plates of
hydroxyapatite are connected by helical collagen fibers.
(Fig. 1-8) The mineral material provides the structural
stiffness of bone but it is the most brittle component
that is protected by interfibrillary matrix of protein.19,28,42
The intrafibrillary matrix contains both collagen and
non-collagen proteins. The mineralized plates are spa-
tially organized to form fibrils composed of platelets of
minerals and intrafibrillary matrix.

Acral Cartilage
Cartilage consists of specialized cells (chondrocytes) and
FIGURE 1-2  ■  Regional designations of skeleton. Major topo- an extracellular matrix composed of fibers embedded in
graphic regions of the skeleton frequently used in the descrip- an amorphous, eosinophilic, gel-like matrix.15,20,49 On the
tion of bone tumors. basis of the matrix composition, cartilage can be divided
into three major subtypes: hyaline, fibrous, and elastic.
Hyaline cartilage is present at the ends of ribs and covers
cortex (Fig. 1-7). The interior of bone has a network of the joint surfaces. It also is seen in tracheal rings and the
trabeculae called the cancellous (spongy) or trabecular bone larynx. It is composed of collagen fibers and a dense
(see Fig. 1-7). The space inside the bone delineated by amorphous eosinophilic substance. Fibrocartilage is
the cortex is referred to as the medullary cavity. The inter- present at the insertion of tendons. The unique feature
trabecular spaces of the medullary cavity consist of of this type of cartilage is its gradual transition to the
adipose tissue, fibrovascular structures, and hematopoi- dense connective tissue of tendons. Elastic cartilage is
etic tissue. The trabecular bone with its high surface/ present in the external and auditory canal, eustachian
volume ratio is susceptible to rapid turnover, and hence tube, external ear, and cuneiform cartilage of the larynx.
most sensitively reflects alterations in mineral homeosta- It is characterized by the presence of a rich network of
sis.3,4,16,18 The trabecular bone contributes to skeletal sta- elastic fibers.
bility by distributing compressive loads across a joint. On
the other hand, the diaphyseal cortex resists bending and
tension forces.
Extracellular Matrix
Based on the overall organization of type I collagen Elements of the extracellular matrix of the skeletal system,
fibers bone is categorized into woven and lamellar uniquely suited to perform the mechanical function of

ERRNVPHGLFRVRUJ
1  General Considerations 5

Cartilaginous epiphysis

Growth plate
Cartilaginous
epiphysis
Metaphysis

Primary spongiosa

Enchondral ossification
Diaphysis

Developing shaft
Metaphysis
Cartilaginous
epiphysis

FIGURE 1-3  ■  Development of bone. Development of primary ossification center in a long bone (tibia). Cartilaginous epiphyses at
both ends are remnants of cartilage model. Center has been replaced by developing diaphysis with zones of enchondral ossification
at both ends. At this stage primary spongiosa with active enchondral ossification occupies most of the bone length within the
metaphyseal portions while the developing shaft is a relatively minor component of the length. Secondary ossification centers will
develop in cartilage masses at bone ends, and continued ossification will lead to formation of growth plates (physis) (see Fig. 1-5).

ERRNVPHGLFRVRUJ
6 1  General Considerations

Epiphysis

Short
tubilar
bones

Shaft

Base
Epiphysioid
tarsal bones

FIGURE 1-4  ■  Development of bone. Whole-mount section of fetal foot shows basic topographic features of short tubular and epi-
physioid bones.

ERRNVPHGLFRVRUJ
1  General Considerations 7

Epiphyseal ossification
center

Epiphysis

Primary
spongiosa

Metaphysis

Secondary
spongiosa Shaft

FIGURE 1-5  ■  Topographic anatomy of a developing long bone. Growth plate or physis results from formation of a secondary ossi-
fication center in the cartilage mass at the end of the cartilage model. Increase in length results mainly from cartilage-cell prolifera-
tion and interstitial growth in the cartilaginous physis.

the skeleton, consist of several major basic components: replaced by randomly arranged fibers of varying sizes
collagen, proteoglycans, and minerals.5,7,10,13 Metaboli- known as woven bone. Woven bone (fiber bone) is formed
cally they represent a major body reservoir of several at sites of early endochondral and membranous ossifica-
ions, predominantly calcium and phosphorus. tions and in fracture callus, periosteal reactions, endosteal
healing processes, and rapidly formed tumor bone. The
recognition of woven bone and its distinction from
Collagen
mature lamellar bone are greatly facilitated by the use of
Collagen is the most abundant protein in the body and polarization microscopy.
the major organic component of extracellular matrix in
bone. The collagen molecule comprises three chains,
Proteoglycans
each of which contains a repeating tripeptide sequence of
glycine-x-y, in which x and y are frequently proline and Proteoglycans are the major noncollagenous organic
hydroxyproline. These three chains are individually syn- components of skeletal matrices.7,14,20,31,35 They are
thesized on ribosomes and subsequently are assembled present in greatest concentrations in cartilage, resulting
into a triple helix. The cross-linking among these mol- in its intense metachromasia. Proteoglycans consist of a
ecules is responsible for the formation of the fibrillar core of hyaluronic acid with protein side chains lined by
matrix. The architecture of collagen fibers reflects the a variety of sulfated glycosaminoglycans. They are mainly
integrity of bone and its level of maturation. In normal assembled and sulfated in the Golgi apparatus after the
adults, virtually all bone collagen is deposited in parallel protein moieties have been synthesized by the ribosomes.
lamellar bundles as seen by polarizing microscopy, hence In the presence of water, the hydrophilic macromolecule
the term lamellar bone. When metabolism of the skeleton of the protein polysaccharide inflates to form a body with
is accelerated and there is need for rapid formation of a shape analogous to a test tube brush with the consis-
matrix collagen, its lamellar architecture is lost and tency of a stiff gel. The size and consistency of the

ERRNVPHGLFRVRUJ
8 1  General Considerations

On its initial appearance in the skeleton, calcium phos-


phate exists in a relatively poorly crystallized form. In
Epiphysis lamellar bone, it is deposited at the interface of osteoid
and mineralized tissue. The lamellar maturation of bone
Physis
Metaphysis
is associated with conversion of the mineralized deposits
(anatomic)
into a hydroxyapatite with a more distinct crystalline
pattern.
Radiologic
(diagnostic)
metaphysis Cells
The cells of the skeleton system include osteoblasts,
osteocytes, osteoclasts, chondroblasts, and chondrocytes
(Fig. 1-9).

Osteoblasts
Osteoblasts are specialized cells that synthesize the bone
matrix.6,25,41,44 They are cuboidal or columnar and are
invariably found lining osteoid seams. They most likely
arise from precursor cells present in the peritrabecular
Diaphysis
marrow. Osteoblasts have a prominent Golgi apparatus
and are rich in rough endoplasmic reticulum, resulting in
prominent basophilia. These features reflect their active
participation in mineralization and in the process of
organic matrix production. Osteoblasts contain large
amounts of alkaline phosphatase. When engaged in the
synthesis of lamellar bone, osteoblasts are polarized in
relationship to the underlying osteoid seam. In woven
bone, this orderly anatomic arrangement of osteoblasts is
absent.
Radiologic Although there is no question that osteoblasts as just
(diagnostic) described actively synthesize bone, most bone surfaces
metaphysis are covered by flat, fusiform cells, variously called inactive
Metaphysis
(anatomic)
osteoblasts or bone-lining cells. These cells are capable of
Physis skeletal synthesis at a slower rate than activated cuboidal
or columnar osteoblasts. They also may act as a barrier
Epiphysis that separates the bone fluid compartment both anatomi-
cally and functionally from the general extracellular fluid.

FIGURE 1-6  ■  Topographic regions of long tubular bones. Ana-


tomically and developmentally, the metaphysis is defined as a
Osteocytes
narrow zone just adjacent to the cartilaginous growth plate Osteocytes represent specialized cells that have been
(physis) in which primary spongiosa is first formed in the
process of enchondral ossification. In radiologic terms, the incorporated into the bone matrix. They are involved in
metaphysis is more loosely defined as a broad region enclosed the maintenance and turnover of bone at a slow rate (i.e.,
by the flare of cortex on the shaft side of the growth plate. This slower than activated osteoblasts).9,24,27,29 They are the
less precisely designated area is diagnostically useful because most numerous of bone cells and, with their cytoplasmic
of the predilection of some bone tumors to develop there.
processes, which extend through canaliculi, constitute
the major portion of the bone cell syncytium. Young
osteocytes often resemble osteoblasts ultrastructurally.
hydrated molecule are responsible for many of the They are capable of perilacunar matrix synthesis and
mechanical and physicochemical properties of cartilage. mineralization, which result in progressive diminution
of lacunar size. Osteocytes that are older and hence
deeper in the matrix may assume osteoclastic features and
Bone Mineral
resorb bone.
The bony skeleton is made rigid by the addition
of mineral to the deposited extracellular organic Osteoclasts
matrix.2,8,10,13,31 The inorganic phase of mature bone
mineral is a carbonate-containing analog of hydroxyapa- Osteoclasts are large, multinucleated cells that are
tite [Ca10 (PO4)6 (OH)2] that forms submicroscopic irreg- responsible for the resorption of bone and calcified car-
ular crystals. The precise mechanism of mineral deposition tilage.11,12,26,32,33,36,44 An abundance of enzymes that play a
in the skeleton is not clear, but there is evidence that the role in bone lytic activities, including acid phosphatase
organic components of bone play a role in the process. and various proteolytic enzymes such as collagenases, is

ERRNVPHGLFRVRUJ
1  General Considerations 9

B C
FIGURE 1-7  ■  Compact bone versus cancellous or spongy bone: Microscopic features. A, Dense cortical compact bone shows
haversian canals surrounded by concentric lamellae-forming units (osteons). Inset: Osteons with concentric lamellae under
polarization microscopy. B, Cancellous bone consists of connecting plates of lamellar bone separated by mature adipose tissue.
C, Higher magnification of A shows connecting and branching plates of lamellar bone. (A, B, and Inset ×50; C, ×160.) (A to
C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10 1  General Considerations

collagen matrix
mineral platelet
A

B C
FIGURE 1-8  ■  Architectural organization of collagen tissue fibers and mineralized deposits. A, The fibrils are made of organized
mineral platelets bound by noncollagenous proteins. The helical structures of proteins absorb and dissipate energy during tensile
strain. B, Scanning electron microscope image of a fractured surface of human bone shows filaments (arrows) connecting the
neighboring fibrils. C, Atomic force microscope image of a fractured surface of human bone showing filaments (arrows) connecting
the neighboring fibrils. (A, Reprinted with permission from Gupta HS, et al: PNAS 108:17741–17746, 2006. B and C, Reprinted with per-
mission from Fantner GE, et al: Nature Materials 4:612–616, 2005.)

present in these cells. Osteoclasts are derived from the typically not seen in the adult normal skeleton. During
monocyte macrophage precursors and share some of fetal development, areas of cartilaginous differentiation
their antigenic features. Osteoclasts have a convoluted occur within mesenchymal tissue. The earliest forms of
cell membrane, or “ruffled border,” that is juxtaposed to chondroblastic differentiation are difficult to recognize
the bone surface. The formation of the ruffled border and microscopically because they do not differ significantly
its adherence to the bone surface are stimulated by para- from fetal myxoid mesenchymal tissue. However, they
thormone and inhibited by calcitonin. In addition, the are at least weakly positive for S-100 protein, an immu-
activity of osteoclasts is mediated by several ubiquitous nohistochemical hallmark of cartilaginous differentiation
cytokines. that is expressed on all cells of cartilage lineage. Young
cartilage cells are relatively small compared with chon-
drocytes. They may have a flattened or irregular contour,
Chondroblasts
and the surface may show multiple projections or filopo-
Chondroblasts represent immature cells of cartilage dia. The nucleus usually contains a prominent nucleolus,
and are precursors of chondrocytes.15,20,25 They are and it may show a prominent paranuclear Golgi zone.

ERRNVPHGLFRVRUJ
1  General Considerations 11

C
FIGURE 1-9  ■  Histology of cartilage and bone. A, Hyaline cartilage from articular end of a long bone. Individual chondrocytes are
seen within lacunae surrounded by a zone of basophilic chondroid matrix that is rich in glycosaminoglycan. Inset, Higher magnifica-
tion of A. B, Osteoblasts that actively synthesize bone matrix are seen bordering trabeculae of newly formed (woven) bone. These
mononuclear cells are cuboidal and have basophilic cytoplasm with a paranuclear clear zone (Golgi center). Osteoid matrix produced
by these cells is deposited in a seam just inside the rim of osteoblasts. Inset, Higher magnification of B. C, Osteoclasts (bone-
resorbing cells) are the multinucleated giant cells shown in concavities (Howship’s lacunae). (A, B, and C, ×100; Insets, ×200.) (A to
C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12 1  General Considerations

A pericellular lacuna is usually absent or indistinct, and surface apposition. Intramembranous bone formation
the amount of intercellular matrix is less abundant than first appears as many separate centers of ossification that
that associated with chondrocytes. In general, immature enlarge and fuse to form a single plate (Fig. 1-11). Mem-
cartilage is highly cellular. The morphology of immature branous bones are directly formed from the mesenchy-
cartilage cells is best studied in lesions that recapitulate mal tissue without a preexisting cartilage model. Growth
embryonal stages of cartilaginous differentiation, such in the diameter of a bone continues principally by the
as chondromyxoid fibroma, chondroblastoma, clear cell deposition of osteoid on the outer convex surface of the
chondrosarcoma, and myxoid chondrosarcoma. A pro- shaft through membranous ossification in the cambium
totype chondroblast is a cell typically seen in a benign layer of the periosteum. Tubulation and remodeling are
cartilage tumor designated as chondroblastoma. It has a achieved by osteoclastic activity resorption on the inner
dense eosinophilic cytoplasm with an oval nucleus that concave surface.
has a prominent longitudinal groove, often seen under
light microscopic examination.
PROCESSING OF BONE SPECIMENS
Chondrocytes
The nature of bone specimens sent for pathologic evalu-
Chondrocytes represent mature cartilage cells that are ation requires some special procedures that are usually
derived from mesenchymal precursor cells.15,20,46 They not required for other specimens.48-50,53,75,91,92 For the
are located in lacunae surrounded by the cartilaginous purpose of this description, the bone specimens are
matrix. Chondrocytes tend to be clustered in small, loose divided into several major categories:
groups that are isogenous or monoclonal because they 1. Intraoperative diagnostic procedures
represent progeny of a single chondrocyte. In the epiphy- 2. Diagnostic or therapeutic biopsies and curettings
seal plates of long bones, the cartilage cells are arranged 3. Resection specimens.
in long columns. During the skeletal growth phase, car- We will discuss the processing of orthopedic speci-
tilage cells in the epiphyseal plates undergo transient mens relevant for tumor-containing specimens, focusing
proliferative activity followed by deposition of a cartilagi- on some practical handling aspects of general interest.
nous matrix and programmed cell death (apoptosis). Pro- The basic steps in handling diagnostic bone specimens
liferation of cartilage cells followed by apoptosis is the are shown in Fig. 1-12.
most important mechanism governing skeletal growth.
Mature chondrocytes have small, dense nuclei. Open
nuclear chromatin with small nucleoli is present in pro-
Intraoperative Diagnostic Procedures
liferating cartilage cells. The ultrastructure of chondro- The pathologist is requested to comment on the nature
cytes is characterized by numerous branched cytoplasmic of biopsy specimens intraoperatively for two reasons: to
processes, a well-developed endoplasmic reticulum, and establish the preliminary diagnosis and to evaluate the
a Golgi center. adequacy of the specimen for future diagnosis to be
established on permanent sections.
The ideal recommended approach begins with preop-
Development of Bone erative consultation between the surgeon and pathologist
Fetal bone formation and postnatal growth occur in one to understand the clinical setting and to establish the
of two ways. In intramembranous ossification, clusters of optimal diagnostic and therapeutic plan. Moreover, it
fetal mesenchymal cells differentiate directly into osteo- reduces miscommunication at the time of surgery. Most
blasts. In enchondral bone formation, a predeposited important, it enables the pathologist to answer more spe-
cartilaginous matrix (cartilage model) serves as a scaffold cifically any questions regarding the therapeutic conse-
for the deposition of osteoid (Fig. 1-10). In the develop- quences of the diagnosis.
ing epiphyseal centers, this cartilage model undergoes Gross specimens submitted for frozen sections must
focal calcification, followed by vascular invasion and the be carefully evaluated for the presence of heavily mineral-
appearance of bone-synthesizing osteoblasts.21,23,38,39 ized tissue, such as fragments of cortical bone. These
Thus the cartilaginous matrix is replaced by bone, except fragments must be removed from the specimen because
at the growth plate and articular surfaces.37-39 At the they cannot be submitted for sectioning without prior
mesenchymal-vascular junction of the epiphyseal growth decalcification. Heavily mineralized specimens are
plate and metaphyseal bone, invasion of continuously unsuitable for frozen sections. Conversely, most bone
growing cartilage is followed by osteoblastic differentia- tumors can be sectioned without prior decalcification
tion and deposition of osteoid. The devitalized, calcified despite matrix mineralization. When the intraoperative
cartilage serves as a scaffolding for the deposition of bone frozen sections are planned, use of a less mineralized
matrix and is resorbed by osteoblasts at the same rate at (softer) portion of the lesion for the biopsy specimen is
which the growth plate is internally expanded. Conse- recommended. It is also important to remember that
quently, long bone growth occurs while the thickness of some heavily mineralized lesions may be unsuitable for
the epiphyseal plate remains constant. The cessation of frozen sections.
interstitial expansion of the epiphyseal plate results in Intraoperative diagnosis is usually based on frozen sec-
its gradual obliteration and the termination of growth. tions stained with hematoxylin-eosin. Occasionally, it
Cartilage serves as the mediator of long bone growth may be superseded by the evaluation of cytologic prepa-
because it is capable of interstitial matrix deposition and rations. Cytologic preparations (touch smears, scrape,

ERRNVPHGLFRVRUJ
1  General Considerations 13

Apoptotic
cartilage cells

Columns
of cartilage
cells

Enchondral
ossification
(primary
spongiosa)

Calcified cartilage matrix

Osteoid

C
FIGURE 1-10  ■  Enchondral ossification: Microscopic features. A, Overall view of anatomy of growth plate and zone of primary spon-
giosa formation below. B, Zone of cartilage-cell hypertrophy at base of cartilage-cell columns where programmed cell death (apop-
tosis) supervenes. C, High-power view of metaphyseal side of growth plate shows osteoid deposition on surface of calcified
chondroid by rimming osteoblasts. (A, ×25; B and C, ×100.) (A to C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14 1  General Considerations

FIGURE 1-11  ■  Intramembranous ossification: Microscopic features. A, Low-power photomicrograph of fetal calvarial bone formation
by direct osteoblastic differentiation from primitive mesenchymal cells. B, Medium-power photomicrograph shows formation of
woven bone trabeculae with osteoblastic rimming without cartilage stage. (A, ×13; B, ×25.) (A and B, hematoxylin-eosin.)

aspirations) can be made from the surface of material that These techniques are recommended as the initial diag-
is too heavily mineralized for frozen section. More often, nostic approaches, but in many cases they yield adequate
cytology is used as a supplement for frozen-section diag- material to establish the final diagnosis. However, some
nosis, which provides an opportunity to evaluate the mor- lesions are extremely difficult or even impossible to
phology of individual cells without freezing artifacts. diagnose on the basis of a small amount of material
obtained by closed biopsy techniques. In such instances,
open biopsy must be performed. Planning the open
Biopsy and Curettage biopsy approach is a complex process that must take into
The specimen for diagnosis can be obtained by various account the clinical presentation, the imaging context,
transcutaneous closed biopsy instruments. The use of and the technical aspects of subsequent definitive
“closed” biopsy techniques has increased, and the proce- surgery.51,65,69,71,80 For example, an inappropriately placed
dure is often assisted by various radiographic imaging open biopsy incision may put an optimal limb-sparing
techniques.47,52,54,59,60,62-64,70,73,74,76-79,90 In many institutions, procedure at risk because of problems such as tissue con-
aspiration cytology often is used as a preliminary diag- tamination and inflammatory complication. Therefore it
nostic approach and, similar to closed biopsy techniques, is generally recommended that an open biopsy be per-
is frequently assisted by radiographic imaging techniques. formed at a medical institution that can provide definitive

ERRNVPHGLFRVRUJ
1  General Considerations 15

Preoperative Analysis of
Clinical and Radiographic Data
(Diagnostic/Therapeutic Plan)

Biopsy/Curettings/Resection

Gross Examination

Processing with Processing without


demineralization demineralization

Microscopic Analysis

Special microscopic
techniques
(immunohistochemistry, etc)
Sterile material for
microbiological,
Diagnostic
tissue cultures, and
cytogenetics

Electron microscopic
Fresh tissue for
Immunohistochemistry
special techniques
on frozen tissue

Frozen tissue for


nonmicroscopic
special techniques
(DNA, RNA, protein, etc.)

FIGURE 1-12  ■  Algorithm for diagnostic plan after clinical and radiographic identification of a suspected bone tumor. Systematic
approach with procurement of adequate samples will lead to a complete workup and more precise diagnosis.

treatment. In general, the incision site and the biopsy situ hybridization or polymerase chain reaction–based
track should be carefully selected so that if the lesion genetic testing. In general, most routine immunohisto-
proves to be malignant, it can be excised en bloc with the chemical studies can be performed on decalcified tissue
segment of affected bone. This prevents tissue contami- with satisfactory results. For that reason, it is impor-
nation and is of particular importance for limited local tant to divide the specimen into several sections and to
excision in limb-salvage procedures (Fig. 1-13). Not process the softer sections without demineralization.
every lesion requires microscopic verification before Demineralization also can destroy some diagnostically
treatment and in some instances imaging techniques are important structures (e.g., chicken wire–type calcification
used to guide the placement of therapeutic devices such in chondroblastoma). Occasionally, a one-step diagnos-
as a radiofrequency electrode for ablation of osteoid tic and therapeutic procedure is chosen on the basis of
osteoma.86 the clinicoradiologic data. In such instances, the material
Open biopsy specimens are submitted in their entirety consists of curettings of the entire lesion. A portion of the
for histologic examination. At the time of gross exami- specimen is typically submitted for microscopic examina-
nation, a decision must be made as to whether the tion. All tissue fragments that appear different should
specimen requires decalcification or can be processed be submitted for microscopic examination because they
without demineralization. Routine demineralization may contain diagnostically important and histologically
of all bone biopsy specimens is inappropriate because distinct components.
this procedure destroys some cellular and extracellu- Small biopsy fragments containing only cancellous
lar components. Demineralization of the entire mate- bone spicules or tumor tissue containing small amounts
rial may render the tissue unsuitable for some special of bone often can be decalcified and fixed in one over-
techniques. Nucleic acids, in particular, are degraded night step through the use of Bouin’s solution. This fixa-
by acid-based demineralization procedures. As a conse- tive contains glacial acetic acid in addition to picric acid,
quence, the demineralized tissue cannot subsequently be and sufficient demineralization is often achieved simulta-
used for certain molecular genetic procedures such as in neously with fixation.

ERRNVPHGLFRVRUJ
16 1  General Considerations

tissue. They also help in the selection of the optimal


plane of bone dissection and may identify special areas of
interest, such as skip metastases, involvement of other
structures such as a neurovascular bundle, and closest
Proposed incision for margin of resection. The best approach is to have the
definitive surgery radiographic documentation available for review during
dissection.
To accomplish the goals of gross examination, it is
important to perform the dissection in an organized
manner:
1. Review the clinical and specimen radiographs.
Biopsy incision 2. Examine the external surface of the specimen.
3. Check the margins of excision (e.g., bone, soft
tissue, synovium, biopsy tract).
Proposed incision for
4. Expose the involved bone.
definitive surgery 5. Expose and examine the tumor.
The specimen’s external surface should be examined
before dissection. Any attached soft tissue or skin should
be inspected for induration, soft tissue masses, and other
changes. Areas of previous incisions and biopsies should
be identified. Margins should be identified and sampled
on the basis of gross examination and radiographic data.
Areas of the closest soft tissue margin of excision, bone
resection margin, and synovial articular resection margin
should be sampled. In limb-sparing procedures, the soft
tissue resection margins around the tumor are of particu-
lar importance. The potential closest margins are best
identified by reviewing the clinicopathologic data in con-
sultation with the surgeon. The bone resection margins
are routinely sampled. In long bone resection specimens,
an en face proximal bone margin of excision is sufficient.
Occasionally multiple sections from large and complex
margins are necessary (e.g., pelvic resection specimens).
Prior biopsy tracks should be left attached to bone and
FIGURE 1-13  ■  Example of incision site for biopsy of distal appropriately sampled to verify their margins and involve-
femoral tumor. Planning of incision site so that tumor can be ment by the tumor. Figure 1-14 shows a sampling plan
excised en bloc with the segment of affected bone and biopsy for osteosarcoma in a distal femoral resection specimen.
tract without contaminating remaining tissue. After examination and sampling, the attached soft
tissue is dissected down to the periosteum and is removed.
Areas of extension into soft tissue identified on radio-
Resection and Amputation Specimens graphs or during dissection should be left attached to the
bone in continuity with the main tumor mass. Bisected
Resection (limb-sparing procedure) or amputation is per- specimens expose the tumor cut surface, and their gross
formed after the malignant or locally aggressive nature examination can reveal features such as epiphyseal
of the lesion has been confirmed by biopsy. involvement, intramedullary invasion and extension into
Dissection of a bone resection or amputation specimen soft tissue, areas of disrupted cortex, and periosteal reac-
should facilitate the following objectives:51,57,58,61,68,72,85,93 tion. The basic features of the two most frequent primary
1. Assessment of the margins of excision bone tumors—osteosarcoma and chondrosarcoma—seen
2. Confirmation of the preoperative diagnosis or its in typical major resection or amputation specimens are
modification on the basis of new information shown in Figures 1-15 to 1-18. Pathologic assessment of
3. Extent of involvement of bone and adjacent the chemotherapy effect in postoperative specimens is an
structures integral element of the multi-model approach to the
4. Assessment of therapeutic effect. management of bone tumors, and it was originally
The preoperative radiographs or specimen radio- designed and clinically validated for osteosarcoma resec-
graphs are extremely helpful in planning the dissection tion specimens.55,56,66,67,81-84,87-89 Subsequent data indicates
of resected or amputated specimens.48,50,51,65,68,75,93 There- that such an assessment may provide important prognos-
fore the preoperative radiographs (plain x-rays, comput- tic and management information for other malignant
erized tomograms, or magnetic resonance images) should bone tumors, including Ewing’s sarcoma.94 The current
always be reviewed before dissection. These images may standards of practice indicate that so-called histologic
be supplemented by specimen radiographs. The radio- mapping of bone resection specimens provides important
graphic data assist in localizing the tumor within the insights for prognosis and management for virtually
specimen and identifying any areas of extension into soft all malignant tumors of bone. Therefore, virtually all

ERRNVPHGLFRVRUJ
1  General Considerations 17

Distance from
bone resection

2
margin
4
5
10 11

6
12 13
14 15 16
17 18 19 20 21

7
22
23
24
25 26
27
28 8
29
30

9
FIGURE 1-14  ■  Gross evaluation of bone tumors. Sampling plan for mapping procedure of central slab to provide precise histologic
information on margins of resection and extent of tumor necrosis after chemotherapy (also see Chapter 5).

resection specimens for primary bone tumors are pro- by Enneking et al.101,102 in 1986 and adopted by the
cessed for mapping. Theoretically the assessment of American Joint Committee Task Force on Bone Tumors.
chemotherapy-related necrosis requires submission of The TNM system can be used to assess any intramed-
the entire tumor for microscopic evaluation, but such an ullary tumors except immunohematopoietic malignan-
approach is impractical and dramatically increases both cies.100,125 The surface bone and soft tissue lesions do not
the workload and the technical cost of pathologic assess- fit well into this system. Stages I and II represent local
ment. For practical purposes, it is performed from the disease. Stage IV designates disseminated disease with
central slice of the tumor which is subjected to specimen local or distant metastases. Stage III identifies lesions that
radiography and is subsequently divided into smaller sec- cannot be defined by the system. Assessment of the tumor
tions submitted in individual cassettes typically averaging is based on three parameters: T (tumor), N (node), and
approximately 2.0 cm2. The general sampling plan for M (metastasis). The tumor is designated as Tx if it cannot
such mapping can be found in Figure 1-14. A detailed be assessed. T1 represents a tumor confined within the
description of pathologic assessment of preoperative che- cortex, and T2 extends beyond the cortex. Nx designates
motherapy effect is provided in Chapter 5. the lymph nodes that cannot be assessed, N0 indicates no
detectable lymph node metastases, and N1 designates the
presence of metastases. Mx indicates an unknown status
of metastases, M0 indicates the absence of detectable
STAGING, GRADING, AND REPORTING distant metastases, and M1 designates the presence of
OF BONE TUMORS distant metastases. In addition, the tumors are histologi-
cally classified into two major categories according to
Staging Systems
their histologic grade: low (grades 1 and 2) and high
Several staging and grading systems have been developed (grades 3 and 4). A high-grade tumor places the lesion at
with the overall idea to stratify tumors according to least in stage II. A summary of the TNM staging system
prognostic factors such as tumor size, compartmentaliza- is provided in Table 1-2.
tion, histologic grade, and the presence or absence of The Enneking system may be applied to both bone
metastasis. and soft tissue tumors. It is based on the assessment
Two staging systems are currently used to assess the of two basic parameters: biologic aggressiveness and
clinicopathologic parameters of bone tumors. The first is local extent of the disease based on the compartmental-
the common tumor, node, metastasis (TNM) staging ization concept of bone and soft tissue(Table 1-3). The
system designed by the American Joint Committee on biologic and clinical aggressiveness is a combination of
Cancer.100,105 It was adopted by the International Union histologic grading and specific histologic types.100-102,122
Against Cancer in 1987. A second system was proposed Text continued on p. 22

ERRNVPHGLFRVRUJ
18 1  General Considerations

Epiphysis

Growth plate

Intramedullary
ivory-like
tumor

Disrupted cortex

Elevated periosteum
(Codman's triangle)

B
FIGURE 1-15  ■  Description of gross findings in resected specimens of osteosarcoma. A, Coronally cut specimen of proximal end of
tibia shows intramedullary lesion delimited by the cartilaginous growth plate in a skeletally immature patient. There is total sparing
of the epiphysis. B, Cortical breakthrough by an intramedullary osteosarcoma with elevation of the periosteum and formation of
Codman’s triangle. This specimen shows crossing of the epiphyseal scar by the tumor and invasion of the epiphysis. Measurements
of the tumor size, the depth of extraosseous penetration, and its distance from the margin of the resection should be obtained.

ERRNVPHGLFRVRUJ
1  General Considerations 19

Bone resection
margin

Soft tissue
extension

Soft tissue
extension

Epiphyseal
extension

B
FIGURE 1-16  ■  Gross examination of resected specimens of bone tumor. A, Femoral osteosarcoma confined distally by cartilaginous
growth plate but with extensive invasion into soft tissue. Measurements of tumor size, length of resection margin, and extent of
penetration into soft tissue can be determined. Foci of cartilage differentiation can be discerned grossly. B, Specimen of osteosar-
coma of distal femur shows extensive epiphyseal penetration distally and a large soft tissue mass that exceeds the size of intra-
medullary tumor. Blood-filled cystic spaces within the intramedullary tumor and in the extraosseous extension suggest telangiectatic
features.

ERRNVPHGLFRVRUJ
20 1  General Considerations

Intramedullary
extension

A B

Soft tissue extension

Epiphyseal extension
FIGURE 1-17  ■  Gross examination of resected specimens of bone tumor. A, Overall view of coronally cut femur containing a very
large osteosarcoma arising in the distal shaft and extending proximally the full length of the medullary cavity of the diaphysis.
Distally the cortex is breached, as is the cartilaginous growth plate. There is circumferential extension into soft tissue. B, Close-up
view of intramedullary growth in the diaphysis. The tumor appears to be discontinuous grossly, but microscopic evidence of con-
tinuous medullary involvement was found.

ERRNVPHGLFRVRUJ
1  General Considerations 21

Myxoid area

Intramedullary tumor

Cystic change

Soft tissue extension


FIGURE 1-18  ■  Gross examination of resected specimens of bone tumors. A, Chondrosarcoma involving the pelvis (hemipelvectomy
specimen) that was cut coronally through the iliac wing and acetabulum. Note extensive intramedullary involvement with hyaline
cartilage tumor nodules and extracortical extension. Tumor breakdown with cyst formation, a common finding in large chondro-
sarcoma, is seen in the supraacetabular region. B, Proximal femoral resected specimen shows extensive intramedullary growth of
a chondrosarcoma that extruded from the bone on the medial aspect of the shaft to form a large extraosseous tumor mass. There
is gross evidence of bone formation (enchondral ossification) in tumor cartilage, as well as cystic degeneration.

ERRNVPHGLFRVRUJ
22 1  General Considerations

TABLE 1-2  TNM Classification of Bone Sarcomas


T—Primary Tumor G—Histopathologic Grading
Translation table for three- and four-grade systems to a two-grade
(low grade vs high grade) system
Tx Primary tumor cannot be assessed
T0 No evidence of primary tumor TNM TWO-GRADE THREE-GRADE FOUR-GRADE
SYSTEM SYSTEMS SYSTEM
T1 Tumor 8 cm or less in greatest dimension Low grade Grade 1 Grade 1
T2 Tumor more than 8 cm in greatest dimension Grade 2
T3 Discontinuous tumors in the primary bone site High grade Grade 2 Grade 3
Grade 3 Grade 4
N—Regional Lymph Nodes Note: If grade cannot be assessed, Ewing’s sarcoma is classified
as high grade. If grade cannot be assessed, classify as low
grade.
Nx Regional lymph nodes cannot be assessed
N0 No regional lymph-node metastasis Stage Grouping
N1 Regional lymph-node metastasis
Stage IA T1 N0 M0 Low grade
Stage IB T2-3 N0 M0 Low grade
M—Distant Metastasis Stage IIA T1 N0 M0 High grade
Stage IIB T2 N0 M0 High grade
Mx Distant metastasis cannot be assessed Stage III T3 N0 M0 High grade
M0 No distant metastasis Stage IVA Any T N0 M0 Any grade
M1 Distant metastasis Stage IVB Any T N1 Any M Any grade
M1a Lung Any T Any N M1b Any grade
M1b other distant sites Note: use N0 for Nx. For T1 and T2, use low grade if no grade is
stated.

Adapted from American Joint Committee on Cancer (AJCC) cancer staging manual, ed 7, New York, 2010, Springer (edited by Edge
SB, Byrd DR, Compton CC, et al),and International Union against Cancer (UICC): TNM classification of malignant tumors, ed 7,
Oxford, 2010, Wiley-Blackwell (edited by Sobin LH, Gospodarowicz MK, Wittekind CH, et al).

TABLE 1-3 Definitions of Anatomic Extent in TABLE 1-4 Musculoskeletal Tumor Society
the Musculoskeletal Tumor Society Staging System
Staging System
Stage Grade Site Metastasis
Intracompartmental (T1) Extracompartmental (T2) IA G1 T1 M0
Intraarticular Soft tissue extension IB G1 T2 M0
Superficial to deep fascia Deep fascial extension IIA G2 T1 M0
Paraosseous Intraosseous or extrafascial IIB G2 T2 M0
extension III G1 or G2 T1 or T2 M1
Intrafascial compartment Extrafascial compartment
Modified from Enneking WF, et al: A system for the surgical
Modified from Enneking WF, et al: A system for the surgical staging of musculoskeletal sarcoma. Clin Orthop 153:106–
staging of musculoskeletal sarcoma. Clin Orthop 153:106– 120, 1980 and Peabody TD, et al: Evaluation and staging of
120, 1980 and Peabody TD, et al: Evaluation and staging of musculoskeletal neoplasms. J Bone Joint Surg 80:1204–
musculoskeletal neoplasms. J Bone Joint Surg 80:1204– 1218, 1998.
1218, 1998.

The designations of this parameter are G0, G1, and G2. lesions are of high histologic grade. Stage III disease is
G0 is a benign neoplasm, and G1 is a locally aggres- any kind of tumor with metastases. The suffixes A and B
sive tumor with a low probability of metastases. The in this system indicate intracompartmental or extracom-
examples of tumors in the G1 category include giant cell partmental lesions, respectively. A summary of Ennek-
tumor, low-grade intraosseous osteosarcoma, parosteal ing’s staging system is provided in Table 1-4.
osteosarcoma, or grade 1 or 2 chondrosarcoma. G2 des- These two systems provide valuable guides to therapy
ignates aggressive tumors with high metastatic potential, and help to stratify tumors of the same categories accord-
such as high-grade conventional osteosarcoma, Ewing’s ing to clinically validated prognostic information. The
sarcoma, grade 3 chondrosarcoma, or malignant fibrous American Joint Commission on Cancer system can be
histiocytoma. T1 tumors involve only one compartment. used for staging of tumors at any anatomic site, but it is
An individual bone with its medullary cavity is consid- difficult to compare the lesions, especially those involving
ered a single compartment. Hence, extension beyond the the extremities as compared with those that affect the
cortex into adjacent soft tissue, joint, or another bone is axial skeleton given the differences in the ability to eradi-
considered extracompartmental and this lesion is desig- cate tumors by surgical excisions in these distinct ana-
nated T2. With these parameters, lesions are separated tomic locations. This system uses a cutoff point of 5 cm
into three stages. Low-grade lesions are stage I. Stage II as an important factor to predict the prognosis. It has to

ERRNVPHGLFRVRUJ
1  General Considerations 23

be understood that this discrimination is somewhat arbi-


TABLE 1-5 Definitions of Grading Parameters
trary, and the size of the tumor is rather a continuous,
for the FNCLCC System
incremental variable. The Enneking system adopted by
the American Joint Committee Task Force on Bone Parameter Criterion
Tumors (also referred to as the Musculoskeletal Tumor
Tumor Differentiation
Society Staging System) places the emphasis on compart-
Score 1 Sarcoma closely resembling normal adult
mentalization and is preferable, especially for lesions mesenchymal tissue (e.g., well-
affecting the extremities. Regardless which staging system differentiated liposarcoma)
is used, a multidisciplinary approach is needed to accu- Score 2 Sarcomas for which histologic typing is
rately stratify the tumors using the information generated certain (e.g., myxoid liposarcoma)
by different modalities (i.e., clinical presentation, radio- Score 3 Embryonal and undifferentiated sarcomas;
sarcoma of uncertain type
graphic imaging, and pathology).
Mitosis Count
Score 1 0-9/10 HPF
Grading Systems Score 2 10-19/10 HPF
Score 3 ≥20/10 HPF
Grading of bone sarcomas represents the evolution of
historical concepts of grading proposed almost a century Tumor Necrosis (Microscopic)
ago by Broders and originally designed for a fibrosar- Score 0 No necrosis
coma.95 Following this publication, numerous studies Score 1 ≤50% tumor necrosis
reaffirmed the importance of grading in a variety of Score 2 >50% tumor necrosis
tumors. In general, grading systems include the state Histologic Grade
of differentiation, the degree of atypia, and the mitotic Grade 1 Total score 2, 3
activity assessed based on the mitotic count in conven- Grade 2 Total score 4, 5
tional pathology preparations or with the assistance of Grade 3 Total score 6, 7, 8
proliferation markers.98,99,107,108,110-114,116-118,121,128 A proto- Modified from Coindre JM, et al: Reproducibility of a
typic large-scale study based on the analysis of stage and histolopathologic grading system for adult soft tissue
grade of 1000 sarcomas was published by Russell et al.124 sarcomas, Cancer 58:306–309, 1986.
The stratification of sarcomas in various systems is based FNCLCC, Federation Nationale de Centres de Lutte Contre le
on two-, three-, or four-tier concepts. None of these Cancer; HPF, high-power field.
systems are precise. The four-tier system usually shows
minimal differences between the two lowest grades. The
three-tier system has a problem with the intermediate TABLE 1-6 Grading of Bone Sarcoma
grade, which may behave as a fully malignant tumor
capable of metastasis or as a locally aggressive tumor only. Grade Sarcoma Type
The overall tendency is to classify the tumors into two Grade 1 Parosteal osteosarcoma
grades, designated as either a low-grade tumor, signifying Grade 1 chondrosarcoma
a locally aggressive tumor, or a high-grade tumor, signify- Clear cell chondrosarcoma
ing a lesion capable of metastasis. The problem with the Low-grade intramedullary osteosarcoma
Grade 2 Periosteal osteosarcoma
two-tier stratification is that it is not always possible to Grade 2 chondrosarcoma
classify the tumor as definitively low or high grade. Classic adamantinoma
The system that is gaining increased popularity was Chordoma
originally published by Trojani et al. in 1984126 and was Grade 3 Osteosarcoma (conventional, telangiectatic,
subsequently adopted by the French Federation of Cancer small cell, secondary, high-grade surface)
Undifferentiated high-grade pleomorphic
Centers Sarcoma Group.96,97,103,109 The system is based on sarcoma
multivariate analysis of microscopic features and utilizes Ewing’s sarcoma
a combination of tumor differentiation, mitotic rate, and Grade 3 chondrosarcoma
necrosis as parameters for grading.(Table 1-5) It is being Dedifferentiated chondrosarcoma
Mesenchymal chondrosarcoma
utilized with increasing popularity both in Europe and Dedifferentiated chordoma
the United States.120 It assigns a score to each parameter Malignancy in giant cell tumor of bone
and then the scores are added to define a combined grade.
The principal weakness of the system is based in the
assignment of the differentiation score, in which the
tumor is compared with its benign tissue origin. In those grade. For example, Ewing’s sarcoma, mesenchymal
tumors in which the benign tissue or cell counterpart is chondrosarcoma, and conventional osteosarcomas are
not known or hypothetical, the differentiation score is highly aggressive tumors considered as high grade.115,127
not very reliable.120 Disregarding this, the system has On the other hand, specific subtypes of osteosarcoma
reasonably good interobserver reproducibility. such as parosteal osteosarcoma are defined as low-grade
The use of grading systems in general, including the tumors. In addition, chondrosarcomas are graded accord-
above listed French system, has never been validated for ing to their own system originally proposed by Evans
bone sarcomas.106 In bone sarcomas, similar to many soft et al., which is described in detail in Chapter 7.104 The
tissue sarcomas, the identification of a specific type deter- grade assignments proposed by the WHO Classification
mines the biologic behavior and by itself defines the System for tumors of bone are listed in Table 1-6.

ERRNVPHGLFRVRUJ
24 1  General Considerations

TABLE 1-7 Checklist for Standard Reporting of Bone Biopsy


Specimen Tumor Size Lymph-Vascular Invasion
Specify bone involved: ___________ Greatest dimension: ___ cm ___ Not identified
___ Not specified Additional dimensions: ___ × ___ cm ___ Present
___ Cannot be determined (see “Comment”) ___ Indeterminate
Procedure
___ Core needle biopsy Histologic Type (World Health Organization Additional Pathologic Findings
___ Curettage Classification) Specify: ___________
___ Excisional biopsy Specify: _____________
___ Other (specify): ____________ ___ Cannot be determined Ancillary Studies
___ Not specified Immunohistochemistry
Mitotic Rate Specify: __________
Tumor Site Specify: ___ /10 high-power fields (HPF) ___ Not performed
___ Epiphysis or apophysis (1 HPF × 400 = 0.1734 mm2; X40 objective; Cytogenetics
___ Metaphysis most proliferative area) Specify: __________
___ Diaphysis ___ Not performed
___ Cortex Necrosis Molecular Pathology
___ Medullary cavity ___ Not identified Specify: __________
___ Surface ___ Present ___ Not performed
___ Tumor involves joint Extent: ___%
___ Tumor extension into soft tissue ___ Cannot be determined Radiographic Findings
___ Cannot be determined Specify: __________
Histologic Grade ___ Not available
Specify: ___
___ Cannot be determined

Rubin BP, et al: Protocol for the examination of specimens from patients with tumors of bone, College of American Pathology,
Based on AJCC/UICC TNM, ed 7, 2013.

Reporting of Bone Tumors Formulation of a pathology report is a complex task


that requires the integration of data from various
A pathologist reporting on a bone tumor is confronted resources, including clinical, imaging, molecular, gross,
with the task of categorizing them into three major sub- and microscopic. The current recommendations for the
types: benign, locally aggressive, and malignant. formulation of pathology reports of bone biopsies and
Benign tumors are defined as having a limited growth resection specimens by the College of American Pathol-
potential. Although they may recur, the recurrence is ogy are summarized in Table 1-7 and Table 1-8.123
typically nondestructive and can be cured by complete
reexcision or curettage. Examples of tumors in this cat-
egory include osteoid osteoma and chondroblastoma. GENERAL EPIDEMIOLOGY
Intermediate, locally aggressive tumors are subdivided OF BONE TUMORS
into two groups. The first are those tumors that often
recur in an infiltrative, locally destructive manner but do Primary bone tumors are comparatively uncommon
not have any evident potential to metastasize. Such among the wide array of human neoplasms. This has
lesions require wide, complete excision to accomplish contributed to a paucity of meaningful data concern-
local control. The exemplary lesion in this category is ing the relative frequency and incidence rates of the
grade 1 chondrosarcoma. The second group of interme- various subtypes of bone tumors, as well as only a
diate tumors, in addition to having locally aggressive rudimentary understanding of risk factors. In previous
recurrence, has the capacity for distant metastasis studies, some regional cancer centers and cancer regis-
although the risk for such metastasis is minimal, typically tries have provided information on the epidemiology of
less than 5%. The prototypic tumor of this category is a bone tumors and some predisposing conditions, which
conventional giant cell tumor of bone, which may occa- include relative frequency of occurrences by age and sex
sionally metastasize, typically to the lung. in addition to skeletal localization.129-134,136,142,146,150,156
Malignant tumors are defined by their ability to grow Such reports have also provided potentially important
locally in a destructive pattern, as well as to have a high pathogenetic information concerning age and gender,
propensity for both local recurrence and distant metasta- as well as ethnic and racial predispositions for bone
sis. Many of the tumors in this category have a risk for sarcomas.135,137,139,140-142,144,150,155
distant metastasis exceeding 20%; in some of them the The data presented here are from 37 years (1973-
metastasis develops in virtually every instance without 2010) of the National Cancer Institute’s SEER Program.152
therapeutic intervention. The SEER Program reports long term incidence, preva-
The recommended grouping of bone tumors into lence, and survival data provided by medically oriented,
these three categories does not correspond to histologic nonprofit organizations, such as universities and state
grading. Specifically, it is important to mention that the health departments, to obtain epidemiologic data on all
intermediate category in this grouping does not corre- cancers diagnosed in residents of selected geographic
spond to the histologic intermediate grade. areas of the United States referred to as the Centers for

ERRNVPHGLFRVRUJ
1  General Considerations 25

TABLE 1-8 Checklist for Standard Reporting of Bone Sarcoma Resection Specimens
Specimen Histologic Grade Number of Lymph Nodes Involved
Specify bone involved (if known): Specify: ___ Specify: ___
_______ ___ Not applicable ___ Number cannot be determined
___ Not specified ___ Cannot be determined (explain): ________
Margins Distant Metastasis (pM)
Procedure ___ Cannot be assessed ___ Not applicable
___ Intralesional resection ___ Margins uninvolved by sarcoma ___ pM1a: Lung
___ Marginal resection   Distance of sarcoma from closest ___ pM1b: Metastasis involving distant
___ Segmental/wide resection margin: ___ cm sites other than lung
___ Radical resection   Specify margin (if known): ___________   +Specify site(s), if known:
___ Other (specify): ________ ___ Margin(s) involved by sarcoma _______________
___ Not specified   Specify margin(s) (if known): _________
Additional Pathologic Findings
Tumor Site Lymph-Vascular Invasion Specify: ___________
___ Epiphysis or apophysis ___ Not identified
Ancillary Studies (required only if
___ Metaphysis ___ Present
applicable)
___ Diaphysis ___ Indeterminate
___ Cortex Immunohistochemistry
___ Medullary cavity Pathologic Staging (pTNM) Specify: ___________
___ Surface TNM Descriptors (required only if ___ Not performed
___ Tumor involves joint applicable) (select all that apply) Cytogenetics
___ Tumor extension into soft tissue ___ m (multiple) Specify: ___________
___ Cannot be determined ___ r (recurrent) ___ Not performed
___ y (posttreatment) Molecular Pathology
Tumor Size Primary Tumor (pT) Specify: ___________
Greatest dimension: ___ cm ___ pTx: Primary tumor cannot be ___ Not performed
Additional dimensions: ___ × ___ cm assessed
Radiographic Findings (if available)
___ Cannot be determined ___ pT0: No evidence of primary tumor
___ Multifocal tumor/discontinuous ___ pT1: Tumor ≤8 cm in greatest Specify: ___________
tumor at primary site (skip dimension ___ Not performed
metastasis) ___ pT2: Tumor >8 cm in greatest
Preresection Treatment
dimension
Histologic Type (World Health ___ pT3: Discontinuous tumors in the ___ No therapy
Organization Classification) primary bone site ___ Chemotherapy performed
Specify: ____________ Regional Lymph Nodes (pN) (Note K) ___ Radiation therapy performed
___ Cannot be determined ___ pNx: Regional lymph nodes cannot ___ Therapy performed, type not specified
be assessed ___ Unknown
Mitotic Rate ___ pN0: No regional lymph node
Treatment Effect
Specify: ___ /10 high-power fields metastasis
(1 HPF × 400 = 0.1734 mm2; X40 ___ pN1: Regional lymph node metastasis ___ Not identified
objective; most proliferative area) ___ No nodes submitted or found ___ Present
Necrosis (macroscopic or microscopic) Number of Lymph Nodes Examined   +Specify percentage of necrotic tumor
___ Not identified Specify: ___ (compared with pretreatment biopsy, if
___ Present ___ Number cannot be determined available): ___%
  Extent: ____% (explain): ________ ___ Cannot be determined

Rubin BP, et al: Protocol for the examination of specimens from patients with tumors of bone, College of American Pathology,
Based on AJCC/UICC TNM, ed 7, 2013.

Disease Control and Prevention’s National Program of SEER program added five additional areas beginning in
Cancer Registries. The long term incidence and survival 2000; these include greater California, greater Georgia,
trends are available for 1975 to 2010 and are based on Kentucky, Louisiana, and New Jersey, bringing the cov-
the data collected from the nine original geographic erage to 28% of the population in the United States.
SEER areas that comprised the entire states of Connecti- Unfortunately this program does not register the data on
cut, Iowa, New Mexico, Utah, and Hawaii, and the benign tumors, including those that arise in the skeleton,
metropolitan areas of Detroit (Michigan), San Francisco– which are considered to be more frequent than bone
Oakland (California), Seattle–Puget Sound (Washing- sarcomas and are as mysterious as their malignant coun-
ton), and Atlanta (Georgia). The Seattle and Atlanta areas terparts.138 For the purpose of SEER data analysis, bone
joined the program in 1974 and 1975, respectively. These tumors were classified according to the International
areas represent approximately 10% of the population in Classification of Diseases (ICD) as shown in Table 1-9.
the United States. As of 1992, SEER data has been col- The total number of bone sarcomas diagnosed among
lected from four additional populations (Alaska Natives, residents of SEER areas from 1973 to 2010 was 8417.
Los Angeles County, San Jose–Monterey, and rural Bone sarcomas are rare when compared with other
Georgia) that increase the representation of minority cancers, constituting only 0.2% of all tumors for which
groups and allow better stratification by race and ethnic- data were obtained by the SEER Program during the
ity. The data from all 13 SEER registries are the source 1973 to 2010 period. Comparison of the incidence of
of incidence and survival rates from 1992 to 2010. The bone sarcomas with that of the closely related group of

ERRNVPHGLFRVRUJ
26 1  General Considerations

TABLE 1-9 Bones and Joints (T-170.0-170.9): Frequency and Percent Distribution by Histology and
Race, Both Sexes, Microscopically Confirmed Cases, SEER Data 1973-2010
All Races White Black
Histology Frequency % Frequency % Frequency %
Bones and Joints 8417 100.00 6930 100.00 825 100.00
  Sarcoma 8322 98.87 6854 98.90 815 98.79
   Malignant fibrous histiocytoma/high-grade 138 1.64 115 1.66 13 1.58
pleomorphic sarcoma (8830-8831)
   Fibrosarcoma (8810-8812) 67 0.80 54 0.78 8 0.97
   Osteosarcoma 3005 35.70 2281 32.91 468 56.73
   Osteosarcoma, NOS (9180) 2011 23.89 1529 22.06 304 36.85
   Specified osteosarcomas (9181-9185, 9190) 752 8.93 583 8.41 115 13.94
   Osteosarcoma in Paget’s disease of bone (9184) 32 0.38 25 0.36 5 0.61
   Other 210 2.49 144 2.08 44 5.33
   Chondrosarcoma, NOS (9220-9240) 2305 27.39 1987 28.67 165 20.00
   Ewing’s sarcoma (9260) 1142 13.57 1023 14.76 42 5.09
   Adamantinoma of long bones (9261) 46 0.55 34 0.49 5 0.61
   Hemangioasarcoma and malignant 116 1.38 96 1.39 2 0.24
hemangioendothelioma (9120-9134)
   Chordoma (9370) 650 7.72 570 8.23 18 2.18
   Other 853 10.13 694 10.01 94 11.39
Unspecified 95 1.13 76 1.10 10 1.21
Bones and Joints—in Situ 0 0.00 0 0.00 0 0.00

Bone Sarcomas
1.8 14

1.6
12
1.4
(cases/100,000 persons)

10

Age distribution (%)


1.2
Indcidence rate

1 8

0.8 6

0.6
4
0.4
2
0.2

0 0
0

9
4

+
9

4
5-
1-

-1

85
-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80

Age at diagnosis
FIGURE 1-19  ■  Bone and soft tissue sarcomas: Epidemiology. Comparison of age-specific incidence rates of bone and soft tissue
sarcomas, all races, both sexes, SEER data, 1973-2010.

soft tissue sarcomas indicates that bone sarcomas occur malignant neoplasms of bone constituted 1.1% of all
at a rate that is approximately one tenth that of soft cases. The number of cases in five major groups of bone
tissue sarcomas.154 The incidence rate of bone sarcomas sarcomas—osteosarcoma (3005), chondrosarcoma (2305),
diagnosed between 1973 and 2010 fluctuated around Ewing’s sarcoma (1142), chordoma (650), and malignant
0.8/100,000 and showed no tendency to increase or fibrous histiocytoma combined with fibrosarcoma (205),
decrease. Osteosarcoma was the most frequently diag- was sufficient to analyze incidence rates and patient sur-
nosed primary sarcoma of bone (35.7%), followed by vival. The age-specific frequencies and incidence rates of
chondrosarcoma (27.3%), Ewing’s sarcoma (13.5%), and bone sarcomas as a group are clearly bimodal (Fig. 1-19).
chordoma (7.7%) (Table 1-9). Malignant fibrous histio- The first well-defined peak occurs during the second
cytoma and fibrosarcoma formed another distinct group decade of life. The second peak occurs in patients older
of lesions and when combined constituted 4.0% of all than 60 years. The risk of development of bone sarcomas
bone sarcomas. Angiosarcomas of bone were extremely during the second decade of life is close to that seen in
rare and constituted 1.3% of cases. Unspecified primary the population older than 60 years, but there are more

ERRNVPHGLFRVRUJ
1  General Considerations 27

TABLE 1-10 Relative Frequencies of Bone Sarcomas by Histologic Type, Sex, and Race: SEER Data
1973-2010
Total Male Female White Black
No. % No. % No. % No. % No. %
Osteosarcoma 3005 35.70 1651 35.13 1354 36.43 2281 32.91 468 56.73
Chondrosarcoma 2305 27.38 1228 26.13 1077 28.98 1987 28.67 165 20.00
Ewing’s Sarcoma 1142 13.57 710 15.11 432 11.62 1023 14.76 42 5.09
Chordoma 650 7.72 386 8.21 264 7.10 570 8.23 18 2.18
Fibrosarcoma/Malignant Fibrous Histiocytoma 205 2.44 105 2.23 100 2.69 169 2.44 21 2.55
Angiosarcoma 116 1.38 72 1.53 44 1.18 96 1.39 2 0.24
Adamantinoma of Long Bones 46 0.55 24 0.51 22 0.59 34 0.49 5 0.61
Unspecified 95 1.13 53 1.13 42 1.13 76 1.10 10 1.21
Others (Sarcomas) 853 10.13 471 10.02 382 10.28 694 10.01 94 11.39
Total 8417 100 4700 100.00 3717 100.00 6930 100.00 825 100.00

TABLE 1-11 Incidence Rates of Bone Sarcomas by Histologic Type, Race, and Sex: SEER Data,
1973-2010
All Races White Black
Total Male Female Total Male Female Total Male Female
Osteosarcoma 0.31 0.35 0.28 0.3 0.34 0.26 0.39 0.42 0.37
Chondrosarcoma 0.25 0.27 0.23 0.26 0.28 0.25 0.14 0.17 0.11
Ewing’s Sarcoma 0.12 0.15 0.08 0.13 0.17 0.1 0.03 0.05 0.02
Chordoma 0.07 0.08 0.05 0.08 0.09 0.06 0.01 0.02 0.01
Malignant Fibrous Histiocytoma 0.01 0.02 0.01 0.02 0.01 0.02 0.01 0.01 0.01
Total 0.76 0.87 0.65 0.79 0.89 0.69 0.58 0.67 0.52

TABLE 1-12 Median Age at Diagnosis for Bone Sarcomas by Histologic Type, Race, and Sex:
SEER Data 1973-2010
All Races White Black
Total Male Female Total Male Female Total Male Female
Osteosarcoma, NOS 21 20 23 23 23 23 18.5 17 23
Osteosarcoma, Paget’s Disease 73 71.5 74 73.5 73 74 59.5 60 _
Chondrosarcoma 52 52 53 54 53 54 45 44 45
Ewing’s Sarcoma 17 17 16 17 17 16 19 20 16
Chordoma 57.5 57 57.5 58 57.5 59 42 45 36
Malignant Fibrous Histiocytoma 68.5 68.5 68 69 69 69 59 59 58

cases in the second decade. The bimodal age-specific However, there are several populations that appear
incidence rate pattern of bone sarcomas is clearly to have increased incidence rates of bone sarcomas par-
different from that of soft tissue sarcomas, which ticularly evident for osteosarcoma, including parts of
shows a gradual increase of incidence with age. General Brazil and Italy, as well as Argentina, Finland, and
epidemiologic data on relative frequencies, incidence, Israel.131,148,149 It is uncertain whether these differences
and survival rates as well as the mean age at diagnosis by reflect increased ethnically based predisposition, environ-
histologic type, sex, and race are summarized in Table 1-9 mental factors, or industrial factors or are simply the
to Table 1-13. Age-related incidence rates and frequency results of inaccuracies related to disease and population
distribution patterns for four major groups of primary monitoring. The most striking difference is in the inci-
bone sarcomas—osteosarcoma, chondrosarcoma, Ewing’s dence of Ewing’s sarcoma, which shows a tenfold higher
sarcoma, and chordoma—are shown in Figures 1-20 and incidence in the white population compared with the
1-21. The overall incidence rates of osteosarcoma, chon- black population. This difference has been repeatedly
drosarcoma, chordoma, and malignant fibrous histiocy- shown in SEER data and regional analyses in the United
toma for the white and black populations are similar. The States.134,143-145,150 The paucity of reports for Ewing’s
incidence rates for the black population, especially for sarcoma from Africa, although the exact population data
males, are somewhat higher for osteosarcoma but the is not available for this continent, confirms the extreme
difference is relatively small.145-147,151,153 In general, the rarity of Ewing’s sarcoma in the black population. The
incidence rates for major groups of bone sarcomas comparison of survival rates for major groups of bone
reported in North America and Europe are similar.131,148,149 sarcoma at the beginning of the SEER program and

ERRNVPHGLFRVRUJ
28 1  General Considerations

TABLE 1-13 Five-year Relative Survival Rates (%) for Bone Sarcomas by Histologic Type, Sex,
and Race: SEER Data 1973-2010
Calendar Period Sex Race
Total 1973-1977 1978-1982 1983-1987 1988-1992 1993-1997 1998-2005 Male Female White Black
Osteosarcoma, 50.6 38.7 40.14 40.42 55.74 53.96 53.2 48.7 53.4 49.8 51.7
NOS
Osteosarcoma, 18.8 * * * * * * 15.6 21.5 20.3 *
Paget’s
Disease
Chondrosarcoma 79.3 73.3 70.42 67.02 79.24 82.92 80.65 76.4 82.2 79.4 77.1
Ewing’s 46.8 34.6 41.46 49.58 46.36 48.2 49.46 44.6 50.9 47.1 37.7
Sarcoma
Chordoma 73.1 63 52.04 73.26 67.16 75.02 77.55 72.5 73.9 73.5 71.7
Malignant 65.4 51.8 54.58 62.12 65.04 65.52 67.88 65.6 65.2 66.1 53.7
Fibrous
Histiocytoma

*<25 cases.

1.0

Osteosarcoma
Chondrosarcoma
0.8 Ewing's sarcoma
(cases/100,000 persons)

Chordoma
Malignant fibrous histiocytoma
Incidence rate

0.6

0.4

0.2

0.0
0

4
+
-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8
1-

5-

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80

Age at diagnosis
FIGURE 1-20  ■  Bone sarcomas: Epidemiology. Age-specific incidence rates and frequency distribution, all races, both sexes, SEER
data, 1973-2010.

currently indicates improvement in 5-year survival rates with hematoxylin-eosin, interpreted in conjunction with
for osteosarcoma, Ewing’s sarcoma, chordoma, and clinical and radiographic data. Hematoxylin-eosin has
malignant fibrous histiocytoma. There has not been proved to be one of the most enduring and reliable stains
improvement in survival rates for chondrosarcoma (Fig. in the entire arsenal of techniques used in diagnostic
1-22). A detailed analysis of the epidemiologic features of pathology, including the diagnosis of skeletal conditions.
bone tumors is provided in addition to a description of The majority of bone tumors and tumorlike conditions
any specific tumors. can be accurately diagnosed with the use of this simple
staining technique. Hematoxylin stains nuclei blue, and
subsequent counterstaining with eosin provides red stain-
SPECIAL TECHNIQUES ing of the cytoplasm and of various extracellular compo-
nents. In brief, hematoxylin must be oxidized with
Fundamental to the diagnosis of bone tumors is the formalin to a purple compound (hematin) and positively
microscopic examination of formalin-fixed (calcified or charged with metallic salt (mordant). The second com-
decalcified) tissue embedded in paraffin and stained ponent used in the technique (eosin) is an anionic

ERRNVPHGLFRVRUJ
1  General Considerations 29

30

Osteosarcoma
Chondrosarcoma
25
Ewing's sarcoma
Chordoma
Age distribution (%)
20 Malignant fibrous histiocytoma

15

10

0
0

4
+
-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8
1-

5-

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80
Age at diagnosis
FIGURE 1-21  ■  Bone sarcomas: Epidemiology. Age-specific frequency distribution by histologic subtype, all races, both sexes, SEER
data, 1973-2010.

90

80
Five-years relative survival rate (%)

70

60

50

40

30 Osteosarcoma
Chondrosarcoma
20
Ewing's sarcoma
10
Chordoma
Malignant fibrous histiocytoma
0
1973-1977 1978-1982 1983-1987 1988-1992 1993-1997 1998-2005
FIGURE 1-22  ■  Bone sarcomas: Epidemiology. Disease specific, 5-year survival rates. Comparison of survival rates by histologic
subtype in all races and both sexes. SEER data, 1973-2010.

ERRNVPHGLFRVRUJ
30 1  General Considerations

dye that electrostatically binds various proteins. The immunohistochemistry has dramatically reduced the
hematoxylin-eosin stain enables accurate microscopic applicability of these techniques in current diagnostic
evaluation of a tissue’s architecture and cells. This tech- pathologic routines. A family of small cell malignancies
nique is certainly not perfect and occasionally must be is the prototype for which the use of special stains was
supplemented with so-called special techniques. From replaced with modern immunohistochemical biomark-
the very beginning of microscopic inspections of tissue ers and molecular tests in their differential diagnosis.169
specimens for both investigative and diagnostic purposes The special stains that are still of value and are at least
there was the necessity to identify specific components of occasionally used in diagnostic skeletal pathology are
extracellular matrix as well as subcellular elements. The described here.
original techniques were based on the combination of
organic dyes and chemicals intended to identify specific Periodic Acid-Schiff Stain
tissue components. Collectively, this approach is referred
to as histochemistry or special stains, which identify various The principle of periodic acid-Schiff (PAS) stain is based
components of tissue based on color reactions. Although on the oxidizing reaction in the presence of periodic
many of these reactions are nonspecific, the special stains acid and Schiff’s reagent.177 Molecules containing vicinal
represent a valuable auxiliary diagnostic tool. The tech- glycol groups form in the presence of periodic acid dial-
niques of histochemistry, especially the color detection dehydes that combine with Schiff’s reagent to produce a
systems, were subsequently combined with immunologic reddish (magenta) substance. PAS stain is principally used
detection of individual molecules with the use of antibod- to demonstrate intracytoplasmic glycogen. The stain by
ies; they now represent the mainstay of the differential itself is nonspecific but with diastase digestion (reduction
diagnostic workup widely used in pathology, including or elimination of a substance positive for PAS after enzy-
skeletal pathology, referred to as immunohistochemistry. matic treatment) it demonstrates the presence of glyco-
Because microscopic inspections of both cellular features gen. In bone tumors, it is most often used to demonstrate
and tissues architecture is subjective, the intention to glycogen in Ewing’s sarcoma and clear cell chondrosar-
quantify the microscopic images has evolved into the coma. In general, PAS stain can be used to demonstrate
technology of cytometry and histomorphology, in which the presence of neutral mucin to outline the basement
individual cellular and extracellular components are membrane and to visualize fungal and parasitic organisms.
quantitatively measured by machines referred to as cyto-
photometers or image analyzers. The necessity to identify Mucins
and elaborate upon details of tissue architecture on a
higher resolution level has evolved into the technique Stains for mucins are frequently used in the diagnosis
collectively referred to as electron microscopy. Various of adenocarcinoma. In bone, they are occasionally used
machines using different electron-based beams were in the diagnosis of metastatic adenocarcinoma when the
developed and the so-called transmission electron microscopy tumor cells do not form conspicuous glandular struc-
has significantly contributed to our understanding of tures. Alcian blue combined with PAS stain is used to
pathogenetic concepts of tumors and has some limited demonstrate neutral and slightly acidic types of muco-
applications in modern diagnostic pathology. Finally, the substances.167,171,172 Mayer’s mucicarmine is probably
investigations of the genetic origin of human cancer have the stain used most frequently to demonstrate acidic
identified disease-specific aberrant chromosomes as well mucin.176,183,198 These stains are useful in demonstrating
as the hybrid genes, providing the foundations of the both intracellular and extracellular mucins in chordoma.
discipline of cytogenetics. The concepts of special tech-
niques listed have evolved in parallel with the concepts
of both investigative and diagnostic pathology. In general,
Trichrome Stain
these special techniques are used when specific diagnos- The trichrome stain is frequently used to demonstrate the
tic, pathogenetic, or etiologic questions cannot be cate- presence of extracellular substances such as collagen.175,179
gorically or satisfactorily answered with the use of the It is sometimes used in research investigation of soft and
standard approach. In this section, we describe the fun- skeletal tissue, but it has minimal diagnostic applicabili-
damental principles of these special techniques, focusing ties. As the name implies, the technique uses three dyes
on their relevance for skeletal pathology. that stain nuclei, cytoplasm, and extracellular matrix, pri-
marily the collagen. This technique is rarely used in the
diagnosis of tumors, but it is frequently used for histomor-
SPECIAL STAINS phometric evaluation of mineralized versus nonmineral-
ized osteoid in metabolic bone diseases (Fig. 1-23).175
Numerous special stains are listed in histotechnology
textbooks, but relatively few provide specific diagnostic
information that cannot be obtained from hematoxylin-
Stains for Microorganisms
eosin–stained sections. Many of these stains are nonspe- Of a long list of stains used to visualize infectious organ-
cific, but historically they represent the first auxiliary isms, only a few are routinely used in diagnostic pathol-
tools available to the pathologist to identify extracel- ogy. Gram stain is used to classify bacteria into two major
lular and subcellular components as well as microor- categories: those that retain crystal violet-iodine dye
ganisms aiding in the diagnosis.165,168,176,186 The advent after alcohol and acetone treatment (gram positive)
of modern biomarkers detected with antibodies and and those that are decolorized by this technique (gram

ERRNVPHGLFRVRUJ
1  General Considerations 31

A B

C D
FIGURE 1-23  ■  Special stains. A, Modified trichrome (Goldner’s stain) of undecalcified bone documents mineralized (blue) and unmin-
eralized (red) bone. Note increased amount of mineralized osteoid (hyperosteoidosis). B, Trichrome stain shows thick rim of unmin-
eralized osteoid in area with increased osteoblastic activity. C, von Kossa’s stain on undecalcified bone demonstrates mineralized
osteoid as black substance. Increased resorptive (osteoclastic) activity is present and is due to secondary hyperparathyroidism.
D, von Kossa’s stain of undecalcified bone with hyperosteoidosis (increased amount of unmineralized osteoid) in tumorigenic
osteomalacia. (A, ×50; B, ×200.)

ERRNVPHGLFRVRUJ
32 1  General Considerations

negative).160,185 The acid-fast stain (Ziehl-Neelsen) is used frequently used techniques. The dye binds the β-pleated
to document the presence of mycobacteria.157 The degree arrangement of amyloid and has no chemical specificity.
of resistance to acid hydrolysis depends on the lipid This is seen as a reddish deposit under nonpolarized
content in the wall of the bacteria. Bacteria with a high light. Green birefringence is present when the sections
content of complex lipids retain carbolfuchsin after are examined under polarized light. Excessive or pro-
decolorization with acid alcohol. Other stains occasion- longed exposure to Congo red can cause binding to the
ally used to document the presence of infectious organ- tissue that is unrelated to the presence of amyloid.
isms are Grocott’s silver (fungi),180,191,194 PAS (fungi),
Dieterle’s (spirochetes),161,196 and Warthin-Starry (spiro-
chetes and rickettsiae).162,192
Hemosiderin
The most frequently used stain for hemosiderin is an iron
stain. In this technique the acid hydrolysis separates the
Argentaffin and Argyrophilic Stains iron from the protein.163 Potassium ferrocyanide is sub-
Before the advent of immunohistochemistry, argentaffin sequently used and forms an insoluble ferric ferrocyanide
and argyrophilic reactions were frequently used to dis- also known as Prussian blue.
close the neuroendocrine and neural differentiation of
cells.158,184,189,197 In skeletal pathology, these reactions
were used mainly in the diagnosis of metastatic tumors.
Melanin Stain
In modern diagnostic pathology, they have been replaced The most frequently used Fontana-Masson technique is
by more specific immunohistochemical stains such as for a silver stain without an external reducing substance
synaptophysin and chromogranin. The Fontana-Masson (argentaffin reaction). In this technique, metallic silver is
modification of the argentaffin reaction is used as a stain directly reduced by melanin.159,164,200
for melanin (see later section). In the argentaffin reaction,
the presence of the phenolic groups, mainly in catechol-
amines and indolamines, reduces the silver salt concen-
Calcium Stain
tration and generates an insoluble black precipitate. The Goldner and von Kossa’s technique for calcium is another
argyrophilic reaction requires an external reducing agent, variant of a silver stain.193 Calcified tissue such as mineral-
usually hydroquinone or formalin. For this reason, the ized osteoid is identified by the substitution of calcium
argyrophilic reaction produces the best results when with potassium and is reduced to an insoluble black silver
tissue is fixed in formalin-free fixative such as Bouin’s precipitate with the photographic developer (Fig. 1-23).
solution. von Kossa’s technique is frequently used on nondecalci-
fied bone sections for histomorphometric assessment of
metabolic bone disorders.
Reticulin Stain
Reticulin fibers represent fine fibrillary material deposited
in the intercellular matrix throughout the body. The
Enzyme Histochemistry
major component of these fibers is type III collagen. The Histochemical identification of enzymatic activity in
histochemical stains used to identify reticulin fibers are diagnostic pathology has been largely replaced by the
silver-based, such as Gomori’s, Wilder’s, or Gordon and immunohistochemical identification of specific proteins.
Sweet’s stains.174,178,190 These stains are nonspecific, and a Histochemical identification requires that the enzyme
positive reaction is related to the presence of interstitial retain its activity, and complex, largely nonspecific color
proteoglycans. Therefore all elements rich in proteogly- reactions are used to visualize the enzyme in ques-
cans stain positive with these stains. Before the discovery tion.181,187 In skeletal and general pathology, a few histo-
of cell markers and immunohistochemistry, reticulin chemical reactions are still being used. A good example
stains were used to disclose the patterns of reticulin fibers, of the use of this technique is the identification of chlo-
to distinguish epithelial and nonepithelial neoplasia, and roacetate esterase (Leder’s esterase) to document myeloid
to subclassify some of the mesenchymal tumors. Epithelial or mast cell differentiation of tumor cells.166,182,188,199 In
neoplasms are characterized by the presence of reticulin bone tumors, acid phosphatase can be used as a marker
fibers surrounding nests of tumor cells. In mesenchymal of osteoclastic cells. Both Leder’s esterase and acid phos-
tumors, reticulin fibers are sparsely distributed among phatase retain their activity after routine fixation and
individual tumor cells. In addition, some of the mesenchy- paraffin embedding. The identification of tyrosinase by
mal tumors have a distinct pattern of reticulin fibers (e.g., the dihydroxyphenylalanine (DOPA) reaction was used
longitudinal parallel arrangement in tumors of Schwann in the past to identify melanocytic differentiation.195
cells). In fibrosarcoma, reticulin fibers encircle individual With the advent of immunohistochemistry, this reaction
tumor cells. In the era of immunohistochemistry, reticulin is rarely used for diagnostic purposes.
stains are seldom used for diagnostic purposes.

Amyloid Stain ELECTRON MICROSCOPY


Staining for amyloid is one of the most useful special Examination of the ultrastructural features of cellular
techniques.170,173 The Congo red stain and examination and extracellular structures was and continues to be a
with nonpolarized and polarized light are the most powerful investigative tool. The introduction of the

ERRNVPHGLFRVRUJ
1  General Considerations 33

transmission electron microscope in the early decades of Ultrastructural studies conducted without a definite
the twentieth century dramatically expanded the investi- list of differential diagnoses and specific questions are
gative capabilities to study the submicroscopic details of likely to provide disappointing results. Such studies have
diseased tissue, including bone tumors and tumorlike the best chance of being useful if conducted along with
conditions.201,202 Use of the transmission electron micro- light microscopy and other applicable special techniques,
scope, in which the electron beam passes through a thin with the formulation of specific questions that need to be
section of tissue impregnated with electron-dense mate- answered.
rial and embedded in plastic medium, provided new The major limitations of ultrastructural studies in the
understanding of the subcellular organization of cells and diagnosis of bone tumors are similar to those listed in
extracellular matrixes. The scanning electron microscope general pathology textbooks:
provides an opportunity to examine the cell surface and 1. Only a small proportion of tissue can be sampled
intracytoplasmic membranous structures, as well as the and examined. Relatively few ultrastructural fea-
three-dimensional composition of extracellular compo- tures are diagnostically specific; as with light
nents. Transmission electron microscopy represented a microscopy, the entire picture is more informative.
substantial diagnostic tool aiding the differential diagno- 2. It is occasionally difficult to distinguish neoplastic
sis of tumors including those in the skeleton. The from nonneoplastic cells in ultrastructural studies.
so-called scanning and analytical instruments had little Therefore the entrapment of normal elements with
use in routine diagnosis. With the advent of modern specific ultrastructural features of cellular differen-
immunohistochemistry, coupled with the dramatic expan- tiation can be misleading. This pitfall can be largely
sion of biomarkers as well as the introduction of molecu- eliminated by careful examination of the so-called
lar diagnostic tests, even the most enthusiastic electron semithin section.
microscopist must admit that as far as diagnostic pathol- 3. The consensus is that in the modern era of immu-
ogy is concerned, the era of electron microscopy is mainly nohistochemistry and molecular pathology, elec-
of historical interest.203,207 The emergence of immunohis- tron microscopy has minimal application in the
tochemistry and molecular pathology has significantly routine diagnosis of bone tumors.
changed the approach to the diagnosis of bone tumors,
and many diagnostic questions that in the past required
ultrastructural studies are now addressed with the use of CYTOMETRY AND
antibodies and molecular tests. It is premature, however, HISTOMORPHOMETRY
to close the diagnostic chapter of electron microscopy in
pathology; it is still being used and the good examples The concept that quantitative analysis of cellular and
represent its application to glomerular disorders of the extracellular tissue components significantly expands the
kidneys and myopathies.204,206,210 In this book, the descrip- investigative capabilities of cell biology and pathogenesis
tion of certain ultrastructural features of bone tumors by of diseases is a very old one. The prototype of modern
electron microscopy is retained because it provides the quantitative instruments was the simple ocular microm-
ultrastructural basis for cellular and extracellular phe- eter. The idea that quantitative cytometry could be used
nomena observed on the light microscope level. to diagnose tumors failed because of the lack of known
In bone tumors, as in general tumor pathology, ultra- parameters that could be used to formulate measurable
structural studies have provided valuable information criteria to distinguish malignant from benign tumors and
about tumor histogenesis and differentiation pathways, to delineate their different categories. Still, the tech-
complementing the immunohistochemical biomarker- niques of quantitative cytometry and histomorphometry
based observations. Such studies were essential to formu- provide reliable information that can be used to study
late novel pathogenetic concepts concerning the tissue tumor cells, and some of this information has prognostic
and cellular origin of many bone tumors. Unfortunately, significance. Moreover, histomorphometry of bone is a
like other special techniques, ultrastructural studies are very useful tool for the diagnosis of metabolic diseases of
not useful in distinguishing benign versus malignant bone. In the past, flow cytometry and image analysis were
tumors and reactive conditions. In the past, the most primarily used to assess the deoxyribonucleic acid (DNA)
frequent applications of this technique in bone tumor ploidy and proliferation rates of the tumors, which were
pathology were in the differential diagnosis of (1) spindle- shown to have some prognostic value. These applications
cell neoplasms, (2) small blue cell neoplasms, (3) vascular continue to be of some interest for practical value, but in
versus nonvascular neoplasms, and (4) a wide range of modern diagnostic pathology these techniques are more
metastatic tumors of bone. These four main areas of often used as investigative rather than diagnostic tools.
electron microscopy have numerous specific applications. The assessment of DNA ploidy and proliferation rates
For educational purposes, we retained the sections have been largely replaced by more accurate techniques
describing ultrastructural features that are of potential such as SNP-based microarray and DNA sequencing.
diagnostic significance for individual bone tumors and For diagnostic purposes, the assessment of tumor prolif-
tumorlike conditions. Electron microscopy continues to eration with the use of immunohistochemistry and pro-
be used as a valuable investigative tool.205,208,209 liferation specific biomarkers is much simpler and cost
Generally, tumors that cannot be diagnosed by light effective. Today, flow cytometry is mainly used for immu-
microscopy and that also have an inconclusive biomarker- nophenotyping of cell populations in lymphohematopoi-
based immunohistochemical profile very likely are not etic malignancies, and the description of such application
diagnosable by electron microscopy. is beyond the scope of this book.

ERRNVPHGLFRVRUJ
34 1  General Considerations

Cell suspension

Sheath fluid

Signal
Light scatter detector processors
Fluorescence
detectors

Integral
computer
Dichroic mirror

Obscuration bar

Remote computers

Laser

FIGURE 1-24  ■  Flow cytometry. Diagrammatic representation of flow cytometric measurements of cell fluorescence stained with fluo-
rochrome and excited by laser beam. Cell size is assessed by measurements of light scatter at low angles. Fluorescence signal is
separated by dichroic mineralization, and several (typically two) fluorescence signals at different wavelengths can be measured by
fluorescence detectors. Finally, data are stored and analyzed by computer.

Flow Cytometry To circumvent these problems, methods have been devel-


oped for the isolation of intact nuclei that permit flow
Flow cytometers are machines constructed to measure cytometric analysis of nuclear components such as DNA.
and record fluorescence on particles (cells) stained with The techniques of nuclear isolation from formalin-fixed,
fluorochrome and flowing in suspension past an excita- paraffin-embedded tissue are widely used for retrospec-
tion source (typically a laser beam) (Fig. 1-24). The fluo- tive studies.
rescence levels of the individual cells are captured by a Analyses of the cell cycle and of DNA ploidy distribu-
photomultiplier tube, converted into an electric pulse, tion patterns are the most common applications of this
and stored and analyzed by a computer. The cells can also technique in diagnostic pathology. For investigative pur-
be stained with multiple fluorochromes and can be excited poses, any cellular component that can be specifically
by two different laser beams. This technique, known as bound with the use of free fluorochromes or in conjunc-
multiparameter flow cytometry, helps analyze several cel- tion with an antibody can be measured and analyzed with
lular components and their relationships. In addition to this approach.219,224,236,237
specific cellular components, some other cellular fea- Flow cytometric analysis of DNA content can be
tures, such as light scattering at small angles, pulse width, accomplished with several fluorochromes. Propidium
or electrical conductivity, related to some extent to cell iodide, ethidium bromide, and diamidine phenylindole
size, can also be measured. (DAPI) are normally used. Acridine orange, which binds
A major limitation of flow cytometry is that it does not differentially to DNA and ribonucleic acid (RNA), can
permit morphologic corroboration of the identity of the be used for the simultaneous analysis of both compo-
measured objects. A second limiting factor is the need to nents. The results of single-parameter measurements are
prepare cell suspensions from solid tumors for analysis. displayed as frequency histograms (Fig. 1-25). With
Cells that normally lack intercellular junctions (e.g., simultaneous measurement of several parameters (usually
peripheral blood cells, bone marrow cells) are ideal for two), the results are presented as scattergraphs or contour
this technique. Single-cell suspensions can also be pre- maps that show the relationship between the measured
pared from cultured cells. On the other hand, the prepa- parameters. In immunofluorescence studies, the proper-
ration of single-cell suspensions from most solid tumors ties of cells binding a given fluorescent antibody are dis-
cannot be done without significant damage to cell mem- played as a percentage of the cell population screened. In
branes. Therefore, the measurement of cytoplasmic com- reference to DNA, its content is usually calculated as an
ponents of solid tumors is more difficult and less accurate. expression of ploidy by comparison with the known

ERRNVPHGLFRVRUJ
1  General Considerations 35

ploidy strands (e.g., 2C = diploid, 4C = tetraploid). The


DNA index is a formula used to express the position of
2C 4C histogram peaks (DNA index 1 = diploid content of
G1 = 60.73%
G1 DNA). The principle of total DNA content analysis is
G2 M S = 19.42%

Time (h)
S G2 + M = 19.84% based on the fact that normal nondividing cells have a
Relative cell number

diploid DNA content. The growing or proliferating cells


G2/G1 = 2.01
G1 can be divided into the three basic cell-cycle compart-
ments: G1, S, and G2 + M. The DNA content of normal
DNA content and nondividing cells (G1) or quiescent (G0) cells is
G2 + M
diploid. During the S phase (synthesis), a duplication of
DNA occurs. During the G2 + M phase, DNA duplica-
S
tion is completed and corresponds to 4C or a tetraploid
value (Fig. 1-25).
DNA content In contrast to normal tissues, neoplastic lesions often
A
undergo chromosomal aberrations that result in the
appearance of aneuploid clones. From a practical point
of view, human tumors can be divided into two major
Aneuploid
groups: diploid and aneuploid. If the main peak of the
(DNA index = 1.32)
DNA histogram centers on the 2C region, the DNA
80 Diploid distribution pattern is similar to that of normal somatic
70 (DNA index = 1.0) cells. Tumors with such a normal or near-normal DNA
60 histogram are classified as diploid or, better, near-diploid.
50 Many tumors exhibit an abnormal position of one or
Number

40 several cell populations that corresponds to the abnormal


30
chromosome complement of their cell populations. Such
tumors are classified as aneuploid. The presence of a
20 diploid or nondiploid DNA histogram, however, is not
10 synonymous with a normal diploid chromosomal com-
0 plement by karyotyping standards. Many of the so-called
10 30 50 70 90 110 140 170 200 230 diploid tumors may in fact exhibit structural chromo-
B DNA somal alterations or can even have an increased number
of chromosomal copies that are too small to be identified
by measurements of total DNA content.
40 p = 0.00002 In addition to DNA ploidy, cell proliferation can also
Diploid
be analyzed. Cell-cycle analysis based on DNA histo-
Aneuploid
grams is not always easy or possible, especially in aneu-
30 ploid tumors, in which several overlapping abnormal cell
populations may be present. The DNA ploidy and cell-
Cases (%)

20 cycle analyses have been repeatedly proven to provide


reliable prognostic information in many tumors of differ-
ent organs, including some bone tumors. The most reli-
10 able data are available in reference to several common
epithelial malignancies such as carcinomas of the breast,
urinary bladder, prostate, and colon. The data in refer-
0 ence to some soft tissue and bone tumors are less
1 2 3
Histologic grade
convincing, but it seems that in at least two of the
C most frequently occurring primary bone sarcomas—
FIGURE 1-25  ■  Flow cytometry. A, Changes of DNA content in
osteosarcoma and chondrosarcoma—DNA ploidy pro-
individual cells traversing the cell cycle and calculation of pro- vides prognostically valuable information.214,223,229,231,233
portion of cells in different components of the cell cycle is Preliminary data indicate that the assessment of DNA
shown. B, Aneuploid DNA histogram in high-grade osteosar- ploidy is also a prognostic factor for small cell malignan-
coma. Note presence of prominent cell population with diploid cies, including Ewing’s sarcoma.232,238
DNA content (DNA index = 1) that accompanies population of
cells with abnormal DNA content (DNA index = 1.32). C, DNA Generally, tumors with modal patterns close to diploid
distribution patterns in chondrosarcoma of bone in relation have a more favorable prognosis compared with tumors
to histologic grade. Virtually all grade 1 chondrosarcomas that have an aneuploid DNA distribution.226,227 Normal
are diploid, and majority of grade 3 tumors are aneuploid. or near-normal total DNA content correlates with a high
(A, B, Courtesy Dr. A.K. El-Naggar, Houston.)
degree of histologic differentiation. In bone tumors,
most high-grade conventional osteosarcomas are aneu-
ploid. On the other hand, well-differentiated low-grade
osteosarcomas, such as low-grade intramedullary or par-
osteal osteosarcomas, are diploid or near-diploid. Dedif-
ferentiation of low-grade precursor conditions with the

ERRNVPHGLFRVRUJ
36 1  General Considerations

30 30

24 24

18 18

12 12

6 6

0 0

Cell count
Cell count

2c 4c 8c 2c 4c 8c

20 20

16 16

12 12

8 8

4 4

0 0
2c 4c 8c 2c 4c 8c
DNA ploidy DNA ploidy
A B
FIGURE 1-26  ■  Image analysis. A, DNA histograms in two components of dedifferentiated chondrosarcoma. Diploid DNA histogram
of low-grade cartilaginous component of tumor(top panel). Aneuploid DNA histogram of high-grade sarcomatous component of
tumor (bottom panel). B, Image analysis of DNA content in cartilage lesions. Diploid histogram in enchondroma (top panel). Aneu-
ploid DNA histogram in grade 3 chondrosarcoma (bottom panel).

development of high-grade sarcomatous phenotypes is of cells in histologic sections or, even better, of whole
typically associated with the development of pronounced cells spread on the glass. Compared with flow cytometry,
aneuploidy (typically, several abnormal cell populations the technique is much slower and fewer objects can be
are present).217,235 Virtually all conventional high-grade measured. The main advantage of this approach is the
osteosarcomas are aneuploid.239 Conventional osteosar- ability to identify a measured object microscopically and
comas that have a prominent near-diploid cell popula- to perform the measurements separately on different cell
tion, in addition to abnormal (aneuploid) cell clones, populations and tissue components. The technique is
are less aggressive than those without prominent near- uniquely suited for the measurements of distinct (micro-
diploid cell populations (Fig. 1-25 and Fig. 1-26).217,229,230 scopically recognizable) tumor cell populations (e.g., dif-
In chondrosarcoma, most grade 1 tumors are diploid, ferent components of dedifferentiated chondrosarcoma)
and nearly all grade 3 chondrosarcomas are aneuploid (Fig. 1-26). Originally, only the smears and cytologic
(Fig. 1-26).212,213,240 Approximately 30% to 40% of grade preparations of whole cells could be measured, but recent
2 chondrosarcomas are aneuploid. In chondrosarcoma of developments in computerized tissue-reconstruction pro-
bone, the presence of a diploid DNA distribution pattern grams have made it possible to perform the quantitations
correlates with lower recurrence and metastasis rates as on histologic sections. The information provided is some-
well as longer disease-free and overall survival rates com- what similar to data gained by flow cytometry. In pathol-
pared with aneuploid tumors.218,222,223,228 The correlation ogy, including that of bone tumors, this technique is most
of DNA ploidy with clinical behavior in other primary frequently used to generate DNA ploidy distribution pat-
bone tumors is less clear and has not been proven to terns and to perform cell-cycle analysis (Fig. 1-26). For
represent a strong prognostic factor. this type of analysis, the cells are typically stained with
the Feulgen reaction. As with flow cytometry, any cellular
Image Analysis component that can be visualized directly by an appropri-
ate color reaction or with the use of antibodies can be
Cytophotometry, or image analysis, is conceptually quantitated for investigative purposes. In addition, several
similar to flow cytometry but requires a different prepara- other parameters, such as nuclear size, shape, volume, and
tion of cells for the measurements.217,227,232 The principle chromatin texture, can be measured. In summary, the
of this technique is the measurement of the optical density main advantage of this technique is its ability to verify

ERRNVPHGLFRVRUJ
1  General Considerations 37

the measured objects microscopically in standard cyto- The parameters indicative of bone resorptive activity
logic and histologic preparations. are as follows:
1. Trabecular resorptive surface; the percent of bone
surface that shows current or prior osteoclastic
Histomorphometry activity
Histomorphometry is similar to image analysis and rep- 2. Cortical resorptive surface; the percent of the corti-
resents a microscopic planimetry or stereology. In skel- cal surface that shows current or prior osteoclastic
etal pathology, it is used to study homeostasis of the activity
skeleton, mainly in metabolic disorders of bone.211,215,216,221 3. Periosteal resorptive surface; the percent of perios-
The application of histomorphometry to the diagnosis of teal surface with osteoclastic activity
bone tumors is minimal. In bone tumors, the technique 4. Trabecular osteoclast count; the number of osteo-
is occasionally used to evaluate the skeletal status and clasts per area (millimeters squared) of medullary
treatment effect in rickets and osteomalacia associated cavity or the number of osteoclasts per length (in
with tumors (see Chapter 22). centimeters) of the trabecular bone
For histomorphometry, undecalcified bone sections 5. Osteoclastic index; the number of osteoclasts per
stained with techniques that enable the visualization of length of the trabecular surface with evidence of
calcified and uncalcified osteoid, such as von Kossa’s or present or prior resorptive activity
Goldner’s (modified trichrome) stains, are used.220,225 6. Cortical porosity; the percentage of the cortex that
Another frequently used technique is tetracycline-pulse contains pores without osteocytic cells.
labeling for epifluorescence.234 Two short cycles (3 days) Accurate measurements of bone resorption usually
of tetracycline approximately 2 weeks apart allow two require specimens taken from two consecutive biopsies.
separate mineralization fronts to be visualized as epifluo- A description of specific features of metabolic disorders
rescence lines outlining the bone trabeculae of cancellous that can be assessed by this technique is beyond the scope
medullary and endosteal cortical bone. This is due to the of this textbook.
ability of tetracyclines to incorporate at the border of
osteoid, which is actively mineralized. The width between
the two lines of fluorescence (pulse labels) reflects the IMMUNOHISTOCHEMISTRY
rate of mineralization.
A number of parameters can be assessed by histomor- The immunohistochemical detection of various extra-
phometry of bone. They can be separated into three cellular and intracellular markers with antibodies has
major categories: (1) measurements that assess the struc- become a generally accepted and widely used auxiliary
tural integrity of the skeleton, (2) parameters that measure method of diagnostic pathology, including the pathol-
bone formation and mineralization, and (3) measure- ogy of bone tumors and tumorlike lesions. This method
ments that reflect the bone’s resorptive activity. has emerged as a diagnostically useful technique because
The most important and frequently measured of the development of highly specific antibodies and
parameters that reflect the structural integrity of the the invention of sensitive immune and enzymatic detec-
skeleton (i.e., the amount of mineralized bone) are as tion systems. The fluorescence detection methods are
follows: more often used in investigative studies and are rarely
1. Total bone volume; the content of mineralized and used in diagnostic pathology. During the past decade,
nonmineralized bone calculated as the percentage the discipline of immunohistochemistry has developed
of the total area of tissue examined novel, more efficient detection systems complemented
2. Cancellous bone volume; mineralized and nonmin- by an exponentially increasing roster of biomarkers
eralized cancellous bone calculated as the percent- that can be used to assess various aspects of differen-
age of the total area of tissue examined or as the tial diagnosis and biologic assessment of the lesion in
percentage of the area of the medullary cavity question.
3. Trabecular osteoid volume; nonmineralized cancel- The immunohistochemical method is based on the
lous bone calculated as a percentage of total cancel- binding of a specific cellular or extracellular substance by
lous bone. the antibody, with subsequent visualization of the bound
The parameters indicative of osteoblastic activity antibody by a color-based detection system. Subsequent
consist of measurements of bone formation and use of a counterstain such as hematoxylin or toluidine
mineralization: blue enables the precise microscopic localization of a
1. Trabecular osteoid surface; the percent of cancel- positive reaction in various components of the tissue.
lous bone surface covered by osteoid Historically, the first identification of the antigen in tissue
2. Mineralizing surface; the proportion of trabecular visualized with a fluorescent antibody was reported in
osteoid surface that exhibits tetracycline labeling 1942; the horseradish peroxidase detection system was
3. Mineral apposition value; the distance (in microns) developed in the mid-1960s.336 It subsequently evolved
between two tetracycline-pulse labels per day into the immunoperoxidase bridge method and the per-
4. Bone formation value; the mineralized bone pro- oxidase antiperoxidase method. The next generation
duced in 1 year detection methods emerged in the early 1980s with the
5. Mineralization lag time; the time (delay) of miner- development of the avidin-biotin complex (ABC) detec-
alization of the matrix after its deposition by tion system, which is still in use today. In this system,
osteoblasts. secondary antibodies are conjugated to biotin and act as

ERRNVPHGLFRVRUJ
38 1  General Considerations

Avidin-biotin
enzyme complex
Enzyme

Secondary Biotinylated
Label antibody secondary
antibody

Primary Primary
antibody antibody
Tissue
antigen
Tissue
antigen

Direct staining Indirect staining B


A

Dextran backbone
Streptavidin
enzyme complex
HRP enzyme

Biotinylated
secondary antibody, Secondary antibody,
mouse/rabbit mouse/rabbit

Primary Primary
antibody antibody
Tissue Tissue
antigen antigen

C D
FIGURE 1-27  ■  Immunohistochemistry: Visualization of bound antibodies with different reactions. A, Direct and indirect staining
methods using primary and secondary antibodies. B, Avidin-biotin complex (ABC) method, in which preformed complex reacts with
the secondary antibody. C, Labeled streptavicin-biotin (LSAB) method, in which biotinylated secondary antibody links a primary
antibody with a streptavidin-peroxidase conjugate. D, Two-step polymer method. The visualization system contains a dextran
polymer backbone to which enzyme molecules are attached. It also contains secondary antibodies with anti-mouse immunoglobulin
and anti-rabbit immunoglobulin specificity facilitating universal application as a detection reagent. (Modified and reprinted with per-
mission from Taylor CR and Rudbeck L, eds, Education Guide: Immunohistochemical Staining Methods, 6th ed, Carpinteria, California, 2013,
Dako.)

a link between tissue-bound primary antibodies and an treatment, or both. At the level of current advancement,
avidin-biotin-peroxidase compound. A closely related immunohistochemistry provides consistent results with
system utilizes the labeled streptavidin-biotin (LSAB) most fixatives. Satisfactory results can be obtained in
complex and utilizes a biotin-related secondary antibody decalcified tissue or even on decolorized, previously
as a link between the primary antibody and a streptavidin- stained microscopic sections. It can also be used in most
peroxidase complex. The modern detection systems are cytologic preparations.
polymer based and contain a dextran backbone attached There are many pitfalls in immunohistochemistry that
with multiple enzyme molecules. It also contains second- to some extent can be avoided by continuous use of
ary antibodies with anti-mouse and anti-rabbit immuno- quality-control methods in the laboratory. The patholo-
globulin specificities. Such a universal polymer-based gist should be familiar with typical patterns of staining
reagent can be used to detect any primary antibody that that are diagnostically helpful and should also be able to
is of mouse or rabbit origin. This system is currently  recognize the most frequent artifactual (nonspecific)
used in the immunohistochemical laboratory in the staining patterns. Constant daily comparison of the
Department of Pathology at the University of Texas MD immunohistochemical results with external and internal
Anderson Cancer Center. The principles of immunohis- positive and negative controls helps prevent misinterpre-
tochemical visualization of the tissue-bound primary tation of ambiguous results. The pitfalls of immunohis-
antibodies using various detection systems are graphically tochemistry are in general caused by false-negative or
depicted in Fig. 1-27. false-positive results.270,336 The major causes of false-
In formalin-fixed, paraffin-embedded tissue, the negative results are as follows:
so-called antigen retrieval techniques are important steps 1. The epitopes of antigens in the tissue are lost
in increasing the sensitivity of the test.336 The goal of this because of inadequate fixation. The amount of
step is to expose the epitopes of the studied antigen and antigen is reduced by degradation and cannot be
to facilitate antigen-antibody binding. The antigen detected by immunohistochemistry.
retrieval techniques are simple and typically include 2. The antigens are misplaced from the specific tissue
limited digestion with proteolytic enzymes, microwave or cellular location because of diffusion. This is

ERRNVPHGLFRVRUJ
1  General Considerations 39

most frequently observed in reference to endothe- designated as orphan comprises filensin and phakinin. All
lial antigens such as factor VIII–related antigen. intermediate filaments have a propensity for self-assembly
3. The antibody is inappropriately used (too low a into ~10-nm wide filaments as heteropolymers with con-
concentration) or destroyed, or its affinity for the served tripartite domain consisting of a central α-helical
antigen is inadequate. rod forming heptad repeats that is flanked by highly vari-
4. The components of the detection system are inad- able end domains comprising their N- and C-termini.
equate or are inappropriately used. This structural uniformity is coupled with pronounced
The major causes of false-positive results are as heterogeneity of their N- and C-termini, resulting in
follows: highly variable mass ranging from 40 kDa (type I keratin
1. Cross-reactivity (lack of specificity) of the antibody 19) to 240 kDa (type IV nestin). Another signature
with other antigens or its nonspecific binding to the feature of intermediate filaments is their tissue type and
tissue the differentiation program-dependence that is in part
2. Nonspecific color reaction caused by the presence retained in the neoplastic process. Although there is a
of unblocked endogenous peroxidase considerable degree of homology among the various
3. Nonspecific binding of detection system compo- groups of intermediate filaments, the differences are suf-
nents, such as the avidin-biotin complex, to the ficient for distinct antigenicity. Therefore the major
tissue (typically caused by excessive use of detection groups of intermediate filaments and even their various
system components) subcategories can be identified by their respective anti-
4. Positive reaction on normal tissue entrapped among bodies. These two features (i.e., tissue specific expression-
the tumor cells and interpreted as an integral com- dependence and specific antigenicity) make them model
ponent of the tumor markers widely used in the differential diagnosis of
5. Misplacement of the antigen from normal tissue tumors including bone cancers. Out of the diverse group
that is subsequently absorbed or phagocytized by of intermediate filaments the keratins, vimentin, desmin,
tumor cells. and GFaP are widely used in the differential diagnosis of
False-positive results are in fact more misleading tumors.
than false-negative results and probably occur more Keratins. The keratins are prototypic intermediate
frequently. filaments of epithelial cells that show a high degree of
Immunohistochemistry is a powerful tool used to molecular diversity.306 They frequently form heteropoly-
provide diagnostically valuable information on the histo- mers by pairing type I and II keratins. In humans, over
genesis and differentiation of cells. Similar to other auxil- 50 keratin genes exist, and they are expressed in a highly
liary techniques, the immunophenotypic profiles of cells specific manner in various types of epithelial cells and in
are not used to distinguish among benign, malignant, and relation to their stage of differentiation. The recent con-
reactive conditions. sensus nomenclature for mammalian keratin genes and
The number of antibodies with potential diagnostic proteins has been established by the Keratin Nomencla-
applications is huge, and new antibodies are constantly ture Committee and is summarized in Table 1-14.326 In
being developed. The immunophenotypic markers of the human genome, the keratin genes cluster in two chro-
hematopoietic lesions of bone and their diagnostic appli- mosomal sites: chromosome 17q21.2 for type I keratins
cations are discussed and tabulated in Chapter 12. The and chromosome 12q13.13 for type II keratins. In some
specific applications of immunohistochemical stains and epithelial cells, they form bundles of structures referred
the so-called immunophenotypic features of bone tumors to as tonofilaments. In a typical epithelial cell, they form a
are provided in the sections on special techniques that network inside the cytoplasm that braids the nucleus and
accompany the discussion of each specific bone tumor. spans through the cytoplasm (Fig. 1-29). These filaments
The markers most frequently used in the diagnosis of are attached to the cytoplasmic plaques at the areas of
bone tumors are described in the sections that follow. cell-to-cell junctions such as desmosomes and hemides-
mosomes. In general, they play a major functional role in
preserving cell structural integrity and mechanical stabil-
Intermediate Filaments ity. They are also important components of cell-to-cell
Intermediate filaments are ubiquitous cytoplasmic struc- and cell-to-stroma interactions. Different epithelia (i.e.,
tures that are 10 nm thick (i.e., they are between 6-nm stratified and simple) have different keratin profiles.
microfilaments and 25-nm thick microtubular struc- Moreover, epithelia in different organs have different
tures). Ultrastructurally, they have a uniform appearance compositions of their keratin, and their expression is
and represent a nonbranching, fine filamentous, cytoplas- retained to some extent in neoplasms derived from these
mic material.259,332 They were originally described nearly organs.306,319 Therefore knowledge of the so-called organ-
half a century ago in a muscle cell. On the basis of their specific keratin profile can be helpful in the differential
chemical composition and presumptive biologic function, diagnosis of neoplasms. In general, keratins are markers
they can be separated into five major groups (Fig. 1-28). of epithelial differentiation.274 However, the presence of
The five groups of intermediate filaments comprise kera- keratin is not absolute proof of epithelial phenotype and
tins, (types I and II); vimentin, desmin, glial fibrillary is occasionally observed in tumors of mesenchymal
acidic protein (GFaP), peripherin, and syncoilin (type origin.319 In reference to bone tumors, it is important to
III); and neurofilaments L, M, and H (NF-L, NF-M, and bear in mind that tumors such as chordoma and, to some
NF-H), α-internexin, synemin, and nestin (type IV); and extent, chondrosarcoma (predominantly those located in
lamins A, B, and C (type V). An additional subgroup VI the central axis) express keratins. The use of highly

ERRNVPHGLFRVRUJ
40 1  General Considerations

Subgrouping Proteins Cell type specificity


Type I Keratins Soft complex epithelia (skin, oral mucosa, etc.)
Type II Soft simple epithelia (liver, gut, kidney, etc.)
Hard epithelia (hair, nail, oral papillae)
Type III Vimentin, Desmin Various (fibroblasts, leukocytes, edothelium
GFAP, Peripherin muscle, astrocytes, glia, peripheral nerves)
syncoilin
Type IV NF-L, NF-M, NF-H CNS & neurons
a-internexin CNS & neurons
synemin, nestin Muscle, neural stem cells
Type V Lamins A, B & C Nucleus
Orphan Filensin, Phakinin Lens
A

Head Rod Tail


1A 1B 2A 2B

N C
L1 L12 L2
B
C
FIGURE 1-28  ■  Intermediate filaments. A, Classification of intermediate filaments according to sequence homology and cell-type
specificity of their expression patterns. B, Schematic representation of the common tripartite domain structure for all intermediate
filaments. A central rod-domain is comprised of heptad repeat-containing α-helical coils 1A, 1B and 2A, 2B. They are separated by
nonheptad repeat-containing linkers L1, L12, and L2. The central rod domain is flanked by head and tail domains of variable length
and structure at their N- and C-termini. C, Assembled 10-nm wide intermediate filament structures reconstituted from recombinant
protein visualized by negative staining and transmission electron microscopy. Bar = 100 nm. GFaP, Glial fibrillary acidic protein; NF,
neurofilament. (Reprinted with permission from Chung G-M, et al: Curr Opin Cell Biol 25:600–612, 2013.)

sensitive reverse transcriptase polymerase chain reaction antigenicity of cells in question when other markers are
(RT-PCR) techniques has shown that low levels of mes- negative. More recent evidence indicates that it is not
senger RNA that code for keratins (predominantly kera- only performing a function of the mesenchymal cell
tins 8 and 18) are expressed in a wide range of mesenchymal cytoskeleton, but it is also involved in the progression
cells and tumors of mesenchymal origin. Fortunately, of several common human malignancies and plays an
these low levels of keratins cannot be detected by tech- important role in phenotypic plasticity of tumor cells,
niques routinely used in diagnostic immunohistochemis- being a signature marker of epithelial to mesenchymal
try. Examples of positivity for keratin have been described transition.290
for virtually every nonepithelial tumor, including many Desmin is a 55-kDa filamentous molecule expressed
bone tumors. Still, for practical purposes, a strong, in smooth, skeletal, and cardiac muscle fibers.248 In skel-
uniform positivity of tumor cells for keratin typically is etal muscle fibers, it is located in a Z band and acts as an
seen in epithelial tumors. adhesion molecule for contractile filaments.248,292,320 In
For diagnostic purposes, several antibodies reacting smooth muscle fibers, it is located in dense bodies and
with keratin are often mixed (AE1/AE3). In skeletal submembranous dense plaques. In general, desmin is a
pathology, keratins are used in the diagnosis of adaman- marker of muscle differentiation, but it is also present in
tinoma, chordoma, chondrosarcoma, epithelioid heman- other cells that have contractile properties, such as myo-
gioendothelioma, and sarcomatoid carcinoma and in the fibroblasts (Fig. 1-31). Desmin is also expressed in some
differential diagnosis of metastatic neoplasms (Fig. 1-30). fetal cells, such as embryonal mesothelium, stromal cells
Vimentin is a 57-kDa filamentous protein universally of fetal kidney, and chorionic villi. It belongs to the cat-
expressed in mesenchymal cells and in some epithelial egory of type III intermediate filaments. In mammalian
cells and their neoplasms.325 In epithelial cells, it is typi- skeletal muscle, including in humans, desmin is one
cally coexpressed with other epithelial markers, such as of the earliest proteins expressed in muscle lineage dif-
keratins. For these two reasons, the specific diagnostic ferentiation and can be detected in somites and early
applicability of vimentin in the differential diagnosis of myoblasts. During skeletal muscle differentiation, the
tumors is minimal. It is most often used to verify the expression of desmin precedes the expression of other

ERRNVPHGLFRVRUJ
1  General Considerations 41

differ in their molecular weight: NF-H (200 kDa), NF-M


TABLE 1-14 The Human Keratin Nomenclature
(160 kDa), and NF-L (68 kDa). Typically, all three poly-
Type I Type II peptides are expressed, but some neuronal cells may lack
all or some of the neurofilament proteins. In general
New Former New Former
Keratin Types Name Name Name Name
pathology, neurofilaments are used as markers of neural
differentiation.305
Epithelial Keratins K9 K9 K1 K1 With reference to bone tumors, neurofilament pro-
K10 K10 K2 K2
K12 K12 K3 K3 teins are used in the differential diagnosis of primary and
K13 K13 K4 K4 metastatic small cell tumors (e.g., neuroblastoma). Poorly
K14 K14 K5 K5 differentiated tumor cells may express undetectable levels
K15 K15 K6a K6a of neurofilament protein. Moreover, fixation and paraffin
K16 K16 K6b K6b
K17 K17 K6c K6e/h
embedding significantly reduce the stainability of cells
K18 K18 K7 K7 for this marker.
K19 K19 K8 K8
K20 K20 K76 K2p
K23 K23 K77 K1b Epithelial Markers
K24 K24 K78 K5b
K79 K6l Keratins are the markers most frequently used in the
K80 Kb20 identification of epithelial phenotypes (see the section on
Hair Follicle-Specific K25 K25irs1 K71 K6irs1 intermediate filaments).
Epithelial Keratins K26 K25irs2 K72 K6irs2 Epithelial membrane antigen (EMA) represents a
(Root Sheath) K27 K25irs3 K73 K6irs3
membrane glycoprotein that was originally identified in
K28 K25irs4 K74 K6irs4
K75 K6hf milk fat globule membranes and is most likely similar or
Hair Keratins K31 Ha1 K81 Hb1 identical to the casein fraction of human milk.278,331 It is
K32 Ha2 K82 Hb2 expressed by virtually all epithelial cells and neoplasms of
K33a Ha3-I K83 Hb3 epithelial origin. However, it is also expressed on a wide
K33b Ha3-II K84 Hb4
K34 Ha4 K85 Hb5
range of tumors of mesenchymal and neural origin and
K35 Ha5 K86 Hb6 even on some lymphomas.276,302
K36 Ha6
K37 Ha7
K38 Ha8 Muscle Markers
K39 Ka35
K40 Ka36 In addition to desmin (described in the section on inter-
mediate filaments), actin and myoglobin are the most
From Moll R et al. Histochem Cell Biology 129:705-733, 2008 frequently used markers of muscle differentiation.
based on new consensus nomenclature from Schweizer J Actin is a major component of the cytoskeleton that
et al. J Cell Biol 174:169-174, 2006.
The respective gene names (“KRT”) by the human genome is ubiquitously present within the cells involved in practi-
consortium utilize the same numbers, e.g. “KRT20”. cally all biologic activities, especially those related to cell
motility. It is an indispensable component of cell integ-
rity, shape, and movement in all eukaryotic cells.251,324
The basic subunit of actin is a monomeric globular
protein (G-actin) that contains an adenosine triphosphate
muscle-specific markers, including the transcriptional (ATP) or adenosine diphosphate (ADP) molecule. Its
regulators of the MyoD family. This expression pattern function depends on the ability to polymerize and form
makes desmin one of the most useful markers of muscle filamentous F-actin. F-actin is composed of two strands
differentiation. In pathology, desmin is used as a marker forming a left-handed double helix. The F-actin is a
for the diagnosis of tumors that exhibit muscle, predomi- unique polar structure that contains a fast-expanding
nantly skeletal, differentiation.243,323 In bone tumors, it is polymerizing plus-end and a minus-end in which depo-
typically used in the differential diagnosis of primary lymerization (referred to as treadmilling) occurs. Actin
and metastatic spindle-cell neoplasms and in the docu- binds numerous partners that participate in various bio-
mentation of rhabdomyoblastic differentiation in dedif- logic functions involved in cell motility and signal trans-
ferentiated chondrosarcoma. It is also frequently used in ductions. Actins are divided into three major subgroups
the differential diagnosis of small cell tumors (Fig. 1-31). of similar molecular weight, 42-kDa: α, β, and γ. The
Glial fibrillary acidic protein is an intermediate fila- three forms of α actin are organ or tissue specific and are
ment expressed in the cytoplasm of glial cells.289 It is designated as α-skeletal, α-cardiac, and α−smooth
useful in the differential diagnosis of cranial and spinal muscle. The two forms of γ actin are γ–smooth muscle
lesions (i.e., glial versus nonglial tumors). It is also used and γ-cytoplasmic. There is only one form of β actin, and
in the diagnosis of malignant and benign peripheral nerve it is designated as β-cytoplasmic. β- and γ-cytoplasmic
sheath tumors.289 The applicability of this marker in the actins are ubiquitous and are expressed in virtually all
diagnosis of bone tumors is minimal. It is rarely used in cells. On the other hand, the expressions of α actins
lesions of the axial skeleton in the differential diagnosis (skeletal, cardiac, and smooth muscle) as well as γ–smooth
of meningeal versus ependymal or glial lesions. muscle actin are tissue specific. The antibody HHF-35 is
Neurofilament protein is expressed in most neuronal most frequently used and reacts with α− and γ–smooth
cells. It is subdivided into three distinct polypeptides that muscle actins. Therefore it is frequently used as a marker

ERRNVPHGLFRVRUJ
42 1  General Considerations

A B

C D

FIGURE 1-29  ■  Cytoskeleton of epithelial cells. A, Immunofluorescence staining of keratin K18 (red, nuclei stained in blue by DAPI)
in PLC (liver carcinoma) cells in vitro. B, Keratin filaments (in red) and the desmosomal component desmoplakin (in green) are
labeled in cultured keratinocytes of line HaCaT. C, Electron microscopic image of tonofilament (keratin) bundles (arrowhead) of
HaCaT keratinocytes. D, Keratin intermediate filaments (black arrowhead) insert at desmosomes (white arrowhead) at cell–cell
contact sites of keratinocytes of the epidermal stratum spinosum (electron microscopy). (Reprinted with permission from Moll R, et al:
Histochem Cell Biology 129:705–733, 2008.)

ERRNVPHGLFRVRUJ
1  General Considerations 43

A B

C D
FIGURE 1-30  ■  Immunohistochemistry. A, Expression of S-100 protein in normal metaphyseal chondrocytes. B, Expression of S-100
protein in chordoma. C, Positivity of endothelial cells for factor VIII–related antigen in hemangioma of bone. D, Strong positivity for
cytokeratins in adamantinoma of bone. (A, ×400; B and D, ×200; C, ×50.)

ERRNVPHGLFRVRUJ
44 1  General Considerations

A B

C D
FIGURE 1-31  ■  Immunohistochemistry. A, Expression of desmin in normal skeletal muscle. B, Expression of smooth muscle actin in
normal smooth muscle of small intestine. C, Expression of desmin in dedifferentiated component of chondrosarcoma. D, Expression
of desmin in metastatic round-cell rhabdomyosarcoma in bone. (A-D, ×200.)

ERRNVPHGLFRVRUJ
1  General Considerations 45

of muscle differentiation (smooth and skeletal). As with Human progenitor cell antigen (CD34) is a member
desmin, actin is expressed on various cells that perform of a large, superfamily of lymphoid markers. It is expressed
contractile functions such as myofibroblasts, myoepithe- early in lymphoid differentiation and also by normal and
lial cells, and pericytes. In bone tumors, antibodies against neoplastic (benign and malignant) endothelial cells.312,322
actin are frequently used in the differential diagnosis of The expression of CD34 is not restricted to endothelial
primary and metastatic spindle- and round-cell tumors. cells. Unfortunately, it is also present in a wide range of
Myoglobin is the heme metalloprotein that binds spindle cells and peripheral nerve sheath tumors.317
oxygen and is expressed in skeletal muscle fibers. In con- Therefore the applicability of CD34 in the differential
trast to other muscle markers, it is not expressed in diagnosis of vascular lesions is limited.
smooth muscle cells. Therefore it serves as a marker of ERG, an ETS-family transcription factor, is a less
skeletal muscle differentiation and is used in the diagnosis frequent partner of EWSR1 translocations in Ewing’s
of rhabdomyosarcoma.252,272,287 However, its expression is sarcoma and is a common translocation partner for
restricted to tumor cells with more differentiation, and TMPRSS2-ERG fusions in prostatic carcinoma.335 Immu-
most poorly differentiated tumors do not express suffi- nohistochemical evidence of ERG overexpression in
cient amounts of this protein to be identified in paraffin- Ewing’s sarcoma and prostatic adenocarcinoma is indica-
embedded, formalin-fixed tissue.307 tive of translocations. In contrast it is constitutively
MyoD is a member of a family of four myogenic expressed in endothelial cells, and its nuclear expression
transcriptional regulator factors (MyoD, Myf5, Mrf4, can be used as a marker of endothelial differentia-
and Myogenin) that control the formation of skeletal tion.263,304,313 It is also commonly overexpressed in epithe-
muscle. They belong to a family of basic-helix-loop- lioid sarcoma.303,334
helix factors that, when overexpressed in undifferentiated
mesenchymal cells, will activate the myogenic differen-
tiation program. MyoD and Myogenin are expressed
Neural Markers
relatively early in skeletal muscle differentiation and Several proteins expressed in neural tissues are used in
can be used as markers in the differential diagnosis of the differential diagnosis of tumors with putative neural
rhabdomyosarcoma.252,253,329 origin. The most frequently used neural markers are
described below.
S-100 protein is an acidic nuclear protein that binds
Vascular Markers calcium and is composed of two (α and β) subunits. It was
A group of heterogeneous proteins with diverse functions originally identified in glial and Schwann cells but is also
commonly expressed in endothelial cells are used as vas- expressed by melanocytes, fat cells, myoepithelial and
cular markers to facilitate the differential diagnosis of Langerhans cells, and cartilage cells.309 In general pathol-
tumors of putative endothelial origin. The most fre- ogy, S-100 protein is often used in the differential diag-
quently used endothelial markers are described below. nosis of tumors of neural origin and melanoma.310
Factor VIII–associated antigen, or von Willebrand In bone tumors, S-100 protein is more frequently used
factor, is a protein expressed by endothelial cells, most as a marker of cartilaginous differentiation. It is expressed
likely in areas corresponding to Weibel-Palade bodies.280,322 early during cartilage lineage differentiation and can be
The factor can be demonstrated in all normal endothelial identified in tumors exhibiting relatively primitive
cells and is expressed by virtually all benign vascular cartilaginous differentiation such as in chondromyxoid
lesions. However, there is great variability of its expres- fibroma, chondroblastoma, and mesenchymal chondro-
sion in malignant vascular lesions.273 High-grade angio- sarcoma. S-100 protein is expressed by chordal tissue,
sarcoma may show only focal positive staining. In skeletal and its expression is retained in chordomas. Therefore
pathology, it is most often used to disclose the endothelial together with epithelial markers (keratins and epithelial
nature of tumor cells in vascular lesions such as heman- membrane antigen), it is frequently used in the differen-
gioendothelioma. It is also used in the differential diag- tial diagnosis of chordoma, chondrosarcoma, and the
nosis of spindle-cell proliferations with a presumed so-called chondroid chordoma. It is also frequently used
vascular origin. in the differential diagnosis of Langerhans cell histiocy-
False-positive staining of nonendothelial cells as a tosis because it is expressed by antigen-presenting cells.
result of diffusion of the antigen from nontumor endo- Myelin basic protein is a major polypeptide compo-
thelial cells is a frequent pitfall in the diagnostic applica- nent of the myelin. Hence, it is expressed in tumors
tion of factor VIII–associated antigen. Nonspecific derived from the Schwann cells, both benign and
staining as a result of leakage of the antigen can be sus- malignant.
pected if diffuse extracellular and cellular staining is Leu-7 was identified as a marker of natural killer cells,
observed. but it is also present in the nerve sheath and neuroendo-
Platelet-endothelial adhesion molecule (CD31) is crine cells.283 It is typically used in the differential diag-
a member of the immunoglobulin family and represents nosis of spindle-cell neoplasm with a presumed nerve
a transmembrane protein of endothelial cells.262,286,322 It is sheath origin.284 Leu-7 is also expressed in tumors such
also expressed by megakaryocytes and platelets and can as leiomyosarcoma, synovial sarcoma, and occasionally in
be detected in some plasma cells. Like CD34, it is rhabdomyosarcoma. It is also used in the differential
expressed by virtually all benign vascular tumors and by diagnosis of small blue-cell neoplasms.
a high percentage of malignant vascular lesions (approxi- Synaptophysin is expressed in neural cells and is
mately 80%).317 found in the presynaptic vesicles. It is also expressed in

ERRNVPHGLFRVRUJ
46 1  General Considerations

neuroendocrine cells. Synaptophysin is generally used as ectopic bone formation.242,246,255 They play an important
a sensitive marker to document neural or neuroendocrine role as molecular mediators of bone and cartilage forma-
differentiation.264,271,305 In bone tumors, it is most often tion in skeletal development as well as in the morpho-
used in the differential diagnosis of metastatic lesions genesis of other organs.315 Preliminary studies have
such as neuroblastoma, paraganglioma, and other neuro- shown that, in addition to osteosarcomas, they are
endocrine tumors. expressed in several other soft tissue and bone neoplasms.
Chromogranin A is an acidic protein belonging to a Their applicability in the differential diagnosis of bone
granin family that is expressed by several normal and tumors is unclear at this time.
neoplastic cells of neuroendocrine origin. It is encoded Proliferating cell nuclear antigen (PCNA) belongs
by a gene located on 14q32. In physiologic conditions, to the cyclin family of nuclear proteins and plays an aux-
chromogranin A is stored and released with catechol- iliary role for DNA polymerase-δ.258,261,265,321 It appears in
amines from storage granules in the adrenal medulla the late G1 phase of the cell cycle.300 In pathology, it is
or with the parathyroid hormone in response to hypocal- frequently used for the immunohistochemical identifica-
cemia from the parathyroid glands.260 In diagnostic tion of proliferating cells.
pathology it is used as a marker of neuroendocrine Ki-67 is similar to PCNA and is expressed in all phases
differentiation.264,283 of the cell cycle. Ki-67, together with PCNA, is fre-
Oligodendrocyte lineage transcription factor 2 quently used to assess the proliferative fraction of tumor
(Olig2) is a 32 kD transcription factor that contains a cells.257,269,339 Because mitotic count is included in several
basic helix-loop-helix DNA binding domain and serves a grading systems for sarcomas assessment of proliferation
critical role in the developing nervous system by deter- rate Ki-67 is frequently used as a prognostic marker in
mining lineage development, particularly of motor sarcomas including bone tumors.245,258,277,281,285,311,327,333
neurons and oligodendrocytes.296 It is highly expressed in Lectins are plant proteins that have an affinity for
diffuse gliomas and is involved in a translocation found carbohydrate substances on the cell surface.328,337 The two
in T-cell acute lymphoblastic leukemia.296 most frequently used lectins are concanavalin A and the
Feminizing locus, Fox-3, or hexaribonucleotide lectin of Ulex europaeus. Concanavalin A has an affinity
binding protein-3 on X3 was originally discovered as a for lymphoid cells.337 The Ulex europaeus lectin binds
marker for neurons that strongly labeled their nuclei the H antigen of the blood O complex. It binds to the
(NeuN, “Neuronal Nuclei”) and was later shown to endothelial cells of all individuals regardless of blood
correspond to Fox-3. NeuN/Fox-3 is involved in RNA group.279
splicing.291 Within the nervous system, NeuN expression
is confined to neurons and labels the vast majority of Lineage-Specific Transcription Factors
neuron subpopulations, with a few notable exceptions
such as cerebellar Purkinje neurons.308 NeuN is com- A large number of transcription factors have been identi-
monly used in oncologic neuropathology to identify fied during the past decade that activate a cascade of
neuronal differentiation in neuronal and glioneuronal genes that control various important cell functions
tumors. ranging from proliferation to differentiation. Some of
these factors are involved in lineage-specific differentia-
tion and organogenesis. Their expression is retained in
Miscellaneous Antigens the neoplasms derived from their specific tissues and
Multiple additional markers related to different aspects organs. Hence, they represent useful markers in the dif-
of cell biology, including proliferation and differentia- ferential diagnosis of various tumors, including those that
tion programs, are frequently used in the differential affect the skeleton.314 Here we describe two groups of
diagnosis of various tumors. The most frequently used such factors that belong to the SOX and PAX families as
markers assessing cell proliferation and differentiation, well as RUNX2 and Osterix, focusing on their relevance
as well as markers overexpressed in a transformed cel- in differential diagnosis of skeletal neoplasms.247
lular state, referred to as tumor-associated antigens, are The SOX genes arise from the founding member Sry,
described below. the mammalian testis-determining factor. More than 20
MIC2 gene product is located in the Xp22.32-pter SOX proteins are known; they are defined by the pres-
region, which corresponds to the p11.2- pter of chromo- ence of the similar HMG (high mobility group) box
some X.244,267,268 The product of this gene is a glycopro- domain.256,282 These genes play important roles in embry-
tein of approximately 30 kDa molecular weight (p30/32) onic development of many tissues and organs and are
that serves as an adhesion molecule of the T cell. The frequently used as lineage differentiation markers.288,301
MIC2 gene is expressed by a majority of Ewing’s sarcomas Of major relevance for skeletal pathology is SOX9, a
and primitive neuroectodermal tumors.267,268,298 It is also marker of osteoprogenitor cells expressed in mesenchy-
expressed by some pediatric lymphomas, leukemias, and mal chondrosarcoma with retention of expression in all
rarely rhabdomyosarcomas. The MIC2 gene product is benign and malignant skeletal neoplasms exhibiting car-
the most significant immunohistochemical marker in the tilage differentiation.241,249,266,316,338,342 Its expression in
differential diagnosis of Ewing’s sarcoma and related osteosarcoma, primarily in the chondroblastic compo-
tumors, as well as other small round-cell malignancies of nent, adversely affects the prognosis.344 It is also involved
bone and soft tissue. in the development of testis, and its expression is retained
Bone morphogenetic proteins represent a class of in some of the germ cell tumors.282 The expression of
heterogeneous polypeptides that are potent inducers of SOX9 is also present in a wide range of carcinomas as a

ERRNVPHGLFRVRUJ
1  General Considerations 47

part of the phenomenon referred to as epithelial to mesen- as clinical cytogenetics. The first specific abnormal chro-
chymal transition.288 SOX9 is also expressed in some soft mosome was identified a half century later by Nowell; it
tissue sarcomas; most importantly it is consistently was originally called a minute chromosome in chronic
expressed in synovial sarcoma.254 In contrast, SOX10 granulocytic leukemia but was later renamed as the Phila-
plays a role in neural differentiation and is also expressed delphia chromosome.365-367 The discovery by Rowley a
in melanomas.275 decade later that this abnormality represented in fact
The PAX genes play an important role in body pat- a unique translocation (9;22)(q34;q11.2) has provided a
terning and organogenesis and, similar to SOX genes, new paradigm for the function of chromosomal abnor-
their expression is retained in neoplasms derived from the malities in human cancer.366,371 The subsequent cloning
specific tissues and organs where they originate.250,295 of the breakpoint and the identification of the BRD/ABL
Consequently, similar to SOX genes they are frequently hybrid gene opened a new era in cancer genetics.355 Simi-
used in differential diagnosis of various tumors. There larly, the identification of t(11;22)(q24;q12) in Ewing’s
are nine PAX genes referred to as PAX1/9.318 PAX2 and sarcoma in 1983 represented a prototypic specific chro-
PAX8 are expressed in the genitourinary tract, and PAX5 mosomal abnormality for sarcomas.346,372 The parallel
is expressed in lymphoid tissue of B cell lineage. PAX cloning of the Ewing’s sarcoma breakpoint and the iden-
genes are also partners in chromosomal rearrangements. tification of the EWSR1/FLI1 hybrid gene facilitated the
PAX3 is a partner of FOXO1 chromosomal rearrange- application of this finding to differential diagnosis of
ment in alveolar rhabdomyosarcoma; PAX8 undergoes small cell malignancies.353 The advent of genome sequenc-
rearrangement with PPARγ in thyroid carcinomas. In ing technologies and high throughput mapping platforms
skeletal pathology, PAX genes and their encoded proteins in the 21st century has dramatically increased the effi-
are used as biomarkers in the differential diagnosis of ciency of identification of chromosomal translocations
metastatic lesions. and specific DNA abnormalities in human cancer includ-
Multiple transcription factors involved in regulation ing skeletal neoplasms.345,350,352,363,374 The number of such
of cell lineages participating in skeletal development have abnormalities identified during the last decade has entered
been identified. Some of these factors are being explored an exponential phase, and their application to differential
as potential biomarkers in differential diagnosis of bone diagnosis of sarcomas, including those that affect the
tumors. In addition to SOX9 described above, such tran- skeleton, have changed the standards of pathology prac-
scriptional factors as RUNX2 and Osterix, involved in tice.345,349,354,356,361,370 In this section, we briefly outline the
osteoblastic lineage differentiation, are of potential inter- techniques of cytogenetics; their details are described in
est as biomarkers in skeletal pathology. Chapter 3. In addition, the specific molecular tests are
RUNX2, also referred to as Cbfa1 (anti-core-binding discussed together with the description of individual
factor α1), regulates the recruitment of preosteoblastic tumors throughout the book.
cells from multipotent mesenchymal cells.293,294,297 Its The conventional cytogenetic studies based on cultur-
expression is retained in all benign and malignant bone- ing of tumor cells are still being used and are comple-
forming neoplasms. (Fig. 1-32) In general, in skeletal mented with several newly developed techniques.351,356,358
pathology it can be used as a marker of osteoblastic Technical advances such as more efficient short-term cul-
lineage differentiation. It has been shown that in osteo- tures and techniques of tumor cell dissociation have made
sarcoma RUNX2 modulates the proliferation rate.299,340,341 solid tumor genetics more applicable to diagnosis.
Similar to other transcription factors, it can be upregu- However, the overgrowth of tumor cells by normal tissue
lated in a wide range of nonskeletal neoplasms as a part and the frequent inability to grow tumor cells are still
of epithelial to mesenchymal transition. major limitations of this technique for everyday diagno-
Osterix is a transcription factor essential for osteo- sis. For example, only approximately 50% of Ewing’s
blastic differentiation.343 Its expression is controlled by sarcoma samples can be successfully cultured for diagnos-
RUNX2, which binds to the promoter of the Osterix tic purposes. The conventional cytogenetic analysis based
gene and upregulates its expression.330 The expression on karyotyping of metaphase chromosomes after their
pattern of Osterix is similar to that for RUNX2, and staining with Giemsa (G-banding) is the mainstay of this
in skeletal pathology it can be used as a marker of osteo- still valid approach.364 It is efficient for the identification
blastic lineage differentiation (Fig. 1-32). Similar to of gross chromosomal translocations and other chromo-
RUNX2 it can also be aberrantly expressed in nonskeletal somal abnormalities. The analysis of karyograms for
neoplasms. translocations is efficient in a background of clean
karyotype. However, when specific chromosomal trans-
locations are associated with complex chromosomal aber-
CYTOGENETICS rations, their identification by this approach may be
difficult. In such instances, fluorescent techniques with
The fundamental observations made at the turn of the chromosome painting probes or the so-called spectral
20th century by von Hansemann and Boveri, that chro- karyotyping (SKY) can be used.359,362
mosomes are asymmetrically distributed during the divi- Because of technical difficulties and the time-
sion of cancer cells and that cancer cells behave abnormally consuming nature of the cytogenetic studies based on
because they have an improper chromosomal comple- culturing of tumor cells, other techniques that are capable
ment, have initiated a quest to unravel the mechanisms of interrogating chromosomes and their genes in nondi-
of the genetic origin of cancer.347,357,373 They also pro- viding cells have been developed. These techniques are
vided the foundation for the discipline referred to today collectively designated as interphase genetics. The use of

ERRNVPHGLFRVRUJ
48 1  General Considerations

SOX9 SOX9high
RUNX2low

SOX9 Chondrocyte

SOX9
Mesenchymal Skeletal RUNX2high
Cells Progenitor
RUNX2

A Preosteoblast
Osx
B

C D SOX9
E RUNX2
F Osterix
Chondromyxoid fibroma Chondroblastoma Osteoid osteoma Osteoblastoma
SOX9
RUNX2
Osterix

G
FIGURE 1-32  ■  Immunohistochemistry: expression of master regulatory genes as chondroblast and osteoblast lineage markers.
A, Schematic diagram depicting the role of SOX9, RUNX2, and Osterix in osteoblastic and chondroblastic differentiated pathways.
B, Representative cores of tissue microarray corresponding to (1 through 4) chondromyxoid fibroma, chondroblastoma, osteoid
osteoma, and osteoblastoma, respectively. C, Whole-mount section of mouse embryo (embryonic day 15 stained with hematoxylin-
eosin). D, Expression of SOX9 in proliferating chondrocytes of the skeletal developmental center. E and F, Expression of RUNX2
and Osterix, respectively, primarily in the perichondrium of the skeletal developmental center. G, Expression of SOX9, Osterix, and
RUNX2 in benign cartilage and bone forming tumors. Strong nuclear expression of SOX9 is evident in chondromyxoid fibroma and
chondroblastoma. Osteoid osteoma and osteoblastoma show strong nuclear expression of RUNX2 and Osterix (DAB and hematoxy-
lin, ×300). (Modified and reprinted with permission from Dancer JY, et al: Hum Pathol 41:1788–1793, 2010.)

ERRNVPHGLFRVRUJ
1  General Considerations 49

fluorescence in situ hybridization (FISH) with specific 19. Fantner G, Hassenkam T, Kindt JH, et al: Sacrificial bonds and
DNA probes to identify chromosomal abnormalities and hidden length dissipate energy as mineralized fibrils separate
during bone fracture. Nat Mater 4:612–616, 2005.
specific breakpoints of genes involved in translocation is 20. Fosang AJ, Beier F: Emerging frontiers in cartilage and chondro-
among the most frequent molecular tests used in differ- cyte biology. Best Pract Res Clin Rheumatol 25:751–766, 2011.
ential diagnosis of sarcomas.348,349,354,362 An alternative 21. Gardner E, Gray DJ: The prenatal development of the human
approach is based on extracted RNA from tumor cells, femur. Am J Anat 129:121–140, 1970.
22. Glimcher MJ: A basic architectural principle in the organization
which is amplified with specific probes using RT-PCR to of mineralized tissues. Clin Orthop 61:16–36, 1968.
detect specific fusion transcripts of translocated genes. 23. Gray DJ, Gardner E: The prenatal development of the human
This approach can be combined with PCR-based humerus. Am J Anat 124:431–445, 1969.
sequencing to identify the specific sequences of fusion 24. Kalajzic I, Matthews BG, Torreggiani E, et al: In vitro and
partners. In addition, the PCR-based amplifications of in vivo approaches to study osteocyte biology. Bone 54:296–306,
2013.
both coding exons and expressed RNA can be performed 25. Karaplis AC: Embryonic development of bone and regulation of
on a genomic scale and after combining with the next intramembranous and endochondral bone formation. In Bilezikian
generation sequencing can provide more complete infor- JP, Raisz LG, Martin TJ, editors: Principles of bone biology, vol 1,
mation on both translocations and mutations.360,368,369 ed 3, Amsterdam, 2008, Elsevier, pp 53–84.
26. Kikuta J, Ishii M: Osteoclast migration, differentiation and func-
tion: novel therapeutic targets for rheumatic diseases. Rheumatol-
REFERENCES ogy 52:226–234, 2013.
27. Klein-Nulend J, Bonewald LF: The osteocyte. In Bilezikian JP,
Introduction Raisz LG, Martin TJ, editors: Principles of bone biology, vol 1, ed 3,
1. Jaffe HL: Tumors and tumorous conditions of the bones and joints, Amsterdam, 2008, Elsevier, pp 153–174.
Philadelphia, 1972, Lea & Febiger. 28. Klein-Nulend J, Bacabac RG, Bakker AD: Mechanical loading
and how it affects bone cells: the role of the osteocyte cytoskeleton
Morphology of Normal Bone in maintaining our skeleton. Eur Cell Mater 24:278–291, 2012.
2. Aaron JE, Shore PA, Shore RC, et al: Trabecular architecture 29. Komori T: Functions of the osteocyte network in the regulation
in women and men of similar bone mass with and without verte- of bone mass. Cell & Tissue Research 352:191–198, 2013.
bral fracture: II Three-dimensional histology. Bone 27:277–282, 30. Lamps L: Diagnostic pathology: normal histologyl. Salt Lake City,
2000. 2013, Amirsys.
3. Ahlborg HG, Johnell O, Turner CH, et al: Bone loss and bone 31. Laros G: Normal structure of bone: light and electron micros-
size after the menopause. N Engl J Med 349:327–334, 2003. copy. In Ackerman LV, Spjut HJ, Abell MR, editors: Bones and
4. Akkus O, Polyakova-Akkus A, Adar F, et al: Aging of microstruc- joints, Baltimore, 1976, Williams & Wilkins.
tural compartments in human compact bone. J Bone Miner Res 32. Malkani K, Luxembourger M-M, Rebel A: Cytoplasmic modifica-
18:1012–1019, 2003. tions at the contact zone of osteoclasts and calcified tissue in
5. Atkins GJ, Findlay DM: Osteocyte regulation of bone mineral: a diaphyseal growing plate of fetal guinea-pig tibia. Calcif Tissue Res
little give and take. Osteoporos Int 23:2067–2079, 2012. 11:258–264, 1973.
6. Aubin JE: Mesenchymal stem cells and osteoblast differentiation. 33. Martin TJ, Suda T: Osteoclast generation. In Bilezikian JP, Raisz
In Bilezikian JP, Raisz LG, Martin TJ, editors: Principles of bone LG, Martin TJ, editors: Principles of bone biology, vol 1, ed 3,
biology, vol 1, ed 3, Amsterdam, 2008, Elsevier, pp 85–108. Amsterdam, 2008, Elsevier, pp 175–192.
7. Baker-LePain JC, Lane NE: Role of bone architecture and 34. Mills SE: Histology for pathologists, ed 4, Philadelphia, 2012, Lip-
anatomy in osteoarthritis. Bone 51:197–203, 2012. pincott Williams & Wilkins.
8. Bauer JS, Kohlmann S, Eckstein F, et al: Structural analysis of 35. Morgan EF, Bouxsein ML: Biomechanics of bone and age-related
trabecular bone of the proximal femur using multislice computed fractures. In Bilezikian JP, Raisz LG, Martin TJ, editors: Principles
tomography: a comparison with dual x-ray absorptiometry for of bone biology, vol 1, ed 3, Amsterdam, 2008, Elsevier, pp 29–51.
predicting biomechanical strength in vitro. Calcif Tissue Int 78:78– 36. Nakamura I, Takahashi N, Jimi E, et al: Regulation of osteoclast
89, 2006. function. Mod Rheumatol 22:167–177, 2012.
9. Bonewald LF: The amazing osteocyte. J Bone Mine Res 26:229– 37. O’Rahilly R: The ossification of the skeleton of the limbs. Ir J
238, 2011. Med Sci 434:78–81, 1962.
10. Boutroy S, Bouxsein ML, Munoz F, et al: In vivo assessment of 38. O’Rahilly R, Gardner E: The embryology of bone and bones. In
trabecular bone microarchitecture by high-resolution peripheral Ackerman LV, Spjut HJ, Abell MR, editors: Bones and joints, Bal-
quantitative computed tomography. J Clin Endocrinol Metab 90: timore, 1976, Williams & Wilkins.
6508–6515, 2005. 39. O’Rahilly R, Gray DJ, Gardner E: Chondrification in the hands
11. Boyce BF: Advances in the regulation of osteoclasts and osteoclast and feet of staged human embryos. Contrib Embryol Carnegie Inst
functions. J Dent Res 92:860–867, 2013. 36:183–192, 1957.
12. Boyce BE, Rosenberg E, de Papp AE, et al: The osteoclast, bone 40. Ross MH, Wojciech P: Histology: a text and atlas, with correlated
remodeling and treatment of metabolic bone disease. Eur J Clin cell and molecular biology, ed 6, Philadelphia, 2010, Lippincott
Invest 42:1332–1341, 2012. Williams & Wilkins.
13. Brandi ML: Microarchitecture, the key to bone quality. Rheuma- 41. Schinke T, Karsenty G: Transcriptional control of osteoblast dif-
tology 48:3–8, 2009. ferentiation and function. In Bilezikian JP, Raisz LG, Martin TJ,
14. Chen C, Tambe DT, Deng L, et al: Biomechanical properties and editors: Principles of bone biology, vol 1, ed 3, Amsterdam, 2008,
mechanobiology of the articular chondrocyte. Am J Physiol Cell Elsevier, pp 109–119.
Physiol 305:1202–1228, 2013. 42. Seeman E: Modeling and remodeling: the cellular machinery
15. Cole AG: A review of diversity in the evolution and development responsible for the gain and loss of bone’s material and structural
of cartilage: the search for the origin of the chondrocyte. Eur Cell strength. In Bilezikian JP, Raisz LG, Martin TJ, editors: Principles
Mater 21:122–129, 2011. of bone biology, vol 1, ed 3, Amsterdam, 2008, Elsevier, pp 3–28.
16. Delmas PD: Treatment of postmenopausal osteoporosis. Lancet 43. Sternberg SS: Histology for pathologists, ed 2, Philadelphia, 1997,
359:2018–2026, 2002. Lippincott-Raven.
17. Engelke K, Libanati C, Fuerst T, et al: Advanced CT-based in 44. Tamma R, Zallone A: Osteoblast and osteoclast crosswalks:
vivo methods for the assessment of bone density, structure, and from OAF to Ephrin. Inflamm Allergy Drug Targets 11:196–200,
strength. Curr Osteoporos Rep 11:246–255, 2013. 2012.
18. Ericksen EF: Normal and pathological remodeling of human tra- 45. Teitelbaum SL, Bullough PG: The pathophysiology of bone and
becular bone: three dimensional reconstruction of the remodeling joint disease. Am J Pathol 96:283–354, 1979.
sequence in normals and in metabolic bone disease. Endocr Rev 46. Wuelling M, Vortkamp A: Chondrocyte proliferation and differ-
7:379–408, 1986. entiation. Endocr Dev 21:1–11, 2011.

ERRNVPHGLFRVRUJ
50 1  General Considerations

75. Mirra JM: Bone tumors: clinical, radiologic, and pathologic correlations,
Processing of Bone Specimens Philadelphia, 1989, Lea & Febiger.
47. Adapon BD, Legada BD, Lim EVA, et al: CT guided closed 76. Monfardini L, Preda L, Aurilio G, et al: CT-guided bone biopsy
biopsy of the spine. J Comput Assist Tomogr 5:73–78, 1981. in cancer patients with suspected bone metastases: retrospective
48. Adler CP, Kozlowski K, Sissons H, et al: Primary bone tumors and review of 308 procedures. Radiol Med 119:852–860, 2014.
tumorous conditions in children: pathologic and radiologic diagnosis, 77. Murphy WA: Radiologically guided percutaneous musculoskeletal
London, 2011, Springer-Verlag. biopsy. Orthop Clin North Am 14:233–241, 1983.
49. An YH, Martin KL: Handbook of histology methods for bone and 78. Murphy WA, Destouet JM, Gilula LA: Percutaneous skeletal
cartilage, Totowa, NJ, 2003, Humana. biopsy 1981: a procedure for radiologists: results, review, and
50. Anderson C: Manual for the examination of bone, Boca Raton, Fla, recommendations. Radiology 139:545–549, 1981.
1982, CRC Press, Inc. 79. Nouh MR, Abu Shady HM: Initial CT-guided needle biopsy of
51. Ayala AG, Raymond AK, Jaffe N: The pathologist’s role in the extremity skeletal lesions: diagnostic performance and experience
diagnosis and treatment of osteosarcoma in children. Hum Pathol of a tertiary musculoskeletal center. Eur J Radiol 83:360–365,
15:258–266, 1984. 2014.
52. Ayala AG, Zornosa J: Primary bone tumors: percutaneous needle 80. Pettersson H, Springfield DS, Enneking WF: Radiologic manage-
biopsy. Radiology 149:675–679, 1983. ment of musculoskeletal tumors, Berlin, 2013, Springer-Verlag.
53. Bancroft JD, Stevens A: Theory and practice of histological techniques, 81. Picci P, Bacci G, Campanacci M, et al: Histologic evaluation of
ed 3, New York, 1990, Churchill Livingstone. necrosis in osteosarcoma induced by chemotherapy: regional
54. Bender CE, Berquist TH, Wold LE: Imaging assisted percutane- mapping of viable and nonviable tumor. Cancer 56:1515–1521,
ous biopsy of the thoracic spine. Mayo Clin Proc 61:942–950, 1986. 1985.
55. Benjamin RS, Chawla SP, Murray JA, et al: Preoperative chemo- 82. Raymond AK, Ayala AG: Specimen management after osteosar-
therapy for osteosarcoma: a treatment approach facilitating limb coma chemotherapy. In Unni KK, editor: Bone tumors, New York,
salvage with major prognostic implications. In Jones SE, Salmon 1988, Churchill Livingstone.
SE, editors: Adjuvant therapy of cancer IV, Orlando, Fla, 1984, 83. Raymond AK, Ayala AG, Carrasco H, et al: Osteosarcoma preop-
Grune & Stratton. erative chemotherapy: specimen management. Cancer Bull 42:
56. Bilbao JI, Martin Algarra S, Martinez de Negri J, et al: Osteosar- 317–331, 1990.
coma: correlation between radiological and histological changes 84. Raymond AK, Chawla SP, Carrasco CH, et al: Osteosarcoma che-
after intra-arterial chemotherapy. Eur J Radiol 11:98, 1990. motherapy effect: a prognostic factor. Semin Diagn Pathol 4:212–
57. Bullough PG: Orthopaedic pathology: expert consult, ed 5, Maryland 236, 1987.
Heights, Mo, 2009, Mosby Elsevier. 85. Raymond AK, Simms W, Ayala AG: Osteosarcoma: management
58. Bullough PG: Atlas of orthopedic pathology: with clinical and radiologic following primary chemotherapy. Hematol Oncol Clin North Am
correlations, Philadelphia, 1992, Lippincott Williams & Wilkins. 9:841–866, 1995.
59. DeSantos LA, Murray JA, Ayala AG: The value of percutaneous 86. Rehnitz C, Sprengel SD, Lehner B, et al: CT-guided radiofre-
needle biopsy in the management of primary bone tumors. Cancer quency ablation of osteoid osteoma: correlation of clinical
43:735–744, 1979. outcome and imaging features. Diagn Interv Radiol 19:330–339,
60. Dollahite HA, Tatum L, Moinuddin SM, et al: Aspiration biopsy 2013.
of primary neoplasms of bone. J Bone Joint Surg 71A:1166–1169, 87. Rosen G, Caparros B, Groshen S, et al: Primary osteogenic
1989. sarcoma of the femur: a model for the use of preoperative chemo-
61. Folpe AL, Inwards CY: Bone and soft tissue pathology: a volume in therapy in high risk malignant tumors. Cancer Invest 2:181–192,
the foundations in diagnostic pathology series, expert consult, Philadel- 1984.
phia, 2009, Saunders Elsevier. 88. Rosen G, Caparros B, Huvos AG, et al: Preoperative chemo-
62. Frager DH, Goldman MJ, Seimon LP, et al: Computed tomog- therapy for osteogenic sarcoma: a selection of postoperative adju-
raphy guidance for skeletal biopsy. Skeletal Radiol 16:644–646, vant chemotherapy based on the response of the primary tumor
1987. to preoperative chemotherapy. Cancer 49:1221–1230, 1982.
63. Gatenby RA, Mulhern CB, Jr, Moldofsky PJ: Computed tomog- 89. Rosen G, Marcove RC, Caparros B, et al: Primary osteogenic
raphy guided thin needle biopsy of small lytic bone lesions. Skel- sarcoma: the rationale for preoperative chemotherapy and delayed
etal Radiol 11:289–291, 1984. surgery. Cancer 43:2163–2177, 1979.
64. Hardy DC, Murphy WA, Gilula LA: Computed tomography in 90. Thapa M, Vo JN, Shiels WE, 2nd: Ultrasound-guided musculo-
planning percutaneous bone biopsy. Radiology 134:447–450, 1980. skeletal procedures in children. Pediatr Radiol 43:55–60, 2013.
65. Hudson TM, Schiebler M, Springfield DS, et al: Radiologic 91. Unni KK, Inwards CY: Dahlin’s bone tumors: general aspects and
imaging of osteosarcoma: role in planning surgical treatment. data on 10,165 cases, Philadelphia, 2009, Lippincott Williams &
Skeletal Radiol 10:137, 1983. Wilkins.
66. Huvos AG: Bone tumors: diagnosis, treatment, prognosis, ed 2, Phila- 92. Vigorita VJ: Orthopaedic pathology, ed 2, Philadelphia, 2007,
delphia, 1990, WB Saunders. Lippincott Williams & Wilkins.
67. Huvos AG, Rosen G, Marcove RC: Primary osteogenic sarcoma: 93. Weatherby RP, Unni KK: Practical aspects of handling orthopedic
pathologic aspect in 20 patients treated with chemotherapy, en specimens in the surgical pathology laboratory. Pathol Annu 17:1–
bloc resection, and prosthetic bone replacement. Arch Pathol Lab 31, 1982.
Med 101:14–18, 1977. 94. Wunder JS, Paulian G, Huvos AG, et al: The histological response
68. Jundt G, Baumhoer D: Pathological assessment of bone sarcomas. to chemotherapy as a predictor of the oncological outcome of
Unfallchirurg 117:510–516, 2014. operative treatment of Ewing sarcoma. J Bone Joint Surg Am
69. Kiatisevi P, Thanakit V, Sukunthanak B, et al: Computed 80:1020–1033, 1998.
tomography-guided core needle biopsy versus incisional biopsy in
diagnosing musculoskeletal lesions. J Orthop Surg 21:204–208, Staging, Grading, and Reporting of Bone Tumors
2013. 95. Broders A, Hargrave R, Meyerding H: Pathological features of
70. Li Y, Du Y, Luo TY, et al: Factors influencing diagnostic yield of soft tissue fibrosarcoma: with special reference to the grading of
CT-guided percutaneous core needle biopsy for bone lesions. Clin its malignancy. Surg Gynecol Obstet 69:267, 1939.
Radiol 69:43–47, 2014. 96. Coindre JM, Terrier P, Bui NB, et al: Prognostic factors in adult
71. Mankin HJ, Lange TA, Spanier SS: The hazards of biopsy in patients with locally controlled soft tissue sarcoma. A study of 546
patients with malignant primary bone and soft tissue tumors. patients from the French Federation of Cancer Centers Sarcoma
J Bone Joint Surg 64A:1121–1127, 1982. Group. J Clin Oncol 14:869–877, 1996.
72. Marcove RC, Rosen G: En bloc resections for osteogenic sarcoma. 97. Coindre JM, Trojani M, Contesso G, et al: Reproducibility of a
Cancer 45:3040–3044, 1980. histopathologic grading system for adult soft tissue sarcoma.
73. Mavrogenis AF, Rimondi E, Rossi G, et al: CT-guided biopsy for Cancer 58:306–309, 1986.
musculoskeletal lesions. Orthopedics 36:416–418, 2013. 98. Costa J, Wesley RA, Glatstein E, et al: The grading of soft tissue
74. Mink J: Percutaneous bone biopsy in the patient with known or sarcomas. Results of a clinicohistologic correlation in a series of
suspected osseous metastases. Radiology 161:191–194, 1986. 163 cases. Cancer 53:530–541, 1984.

ERRNVPHGLFRVRUJ
1  General Considerations 51

99. Deyrup AT, Haydon RC, Huo D, et al: Myoid differentiation and 125. Sobin LH, Gospodarowicz MK, Wittekind CH: TNM classification
prognosis in adult pleomorphic sarcomas of the extremity: an of malignant tumors, ed 7, Oxford, 2010, Wiley-Blackwell.
analysis of 92 cases. Cancer 98:805–813, 2003. 126. Trojani M, Contesso G, Coindre JM, et al: Soft-tissue sarcomas
100. Edge S, Byrd D, Compton C, et al: American Joint Committee on of adults: study of pathological prognostic variables and definition
Cancer: cancer staging manual, ed 7, New York, 2010, Springer. of a histopathological grading system. Int J Cancer 33:37–42, 1984.
101. Enneking WF, Spanier SS, Goodman MA: A system for the surgi- 127. Unni KK, Dahlin DC: Grading of bone tumors. Semin Diagn
cal staging of musculoskeletal sarcoma. Clin Orthop Relat Res Pathol 1:165–172, 1984.
415:4018, 2003. 128. van Unnik JA, Coindre JM, Contesso C, et al: Grading of soft
102. Enneking WF, Spanier SS, Goodman MA: A system for the surgi- tissue sarcomas: experience of the EORTC soft tissue and bone
cal staging of musculoskeletal sarcoma. Clin Orthop 153:106–120, sarcoma group. Eur J Cancer 29A:2089–2093, 1993.
1980.
103. European Sarcoma Network Working Group: Soft tissue and General Epidemiology of Bone Tumors
visceral sarcomas: ESMO clinical practice guidelines for diagno- 129. Campanacci M, Bacci G, Boriani S, et al: Ewing’s sarcoma: a
sis, treatment and follow-up. Ann Oncol 23:92–99, 2012. review of 195 cases. Ital J Orthop Traumatol 5:293–302, 1979.
104. Evans HL, Ayala AG, Romsdahl MM: Prognostic factors in chon- 130. Coventry MB, Dahlin DC: Osteogenic sarcoma: a critical analysis
drosarcoma of bone: a clinicopathologic analysis with emphasis on of 430 cases. J Bone Joint Surg 39A:741–757, 1957.
histologic grading. Cancer 40:818–831, 1977. 131. Curado MP, Edwards B, Shin HR, et al: Cancer incidence in five
105. Fleming ID, Cooper J, Henson D: AJCC cancer staging manual, continents, vol IX, ed 1, 2009, IARC Scientific Publications, World
ed 5, Philadelphia, 1997, Lippincott-Raven. Health Organization.
106. Fletcher CDM, Bridge JA, Hogendoorn PCW, et al: WHO clas- 132. Dahlin DC, Coventry MB: Osteogenic sarcoma: a study of 600
sification of tumours of soft tissue and bone, Lyons, 2013, International cases. J Bone Joint Surg 49A:101–110, 1967.
Agency for Research on Cancer. 133. Dahlin DC, Cupps RE, Johnson EW, Jr: Giant-cell tumor: a study
107. Goldblum JR, Folpe AL, Weiss SW: Enzinger & Weiss’s soft tissue of 195 cases. Cancer 25:1061–1070, 1970.
tumors, ed 6, Philadelphia, 2014, Elsevier Saunders. 134. Damron TA, Ward WG, Stewart A: Osteosarcoma, chondrosar-
108. Golouh R, Bracko M: What is the current practice in soft tissue coma, and Ewing’s sarcoma: National Cancer Data Base report.
sarcoma grading? Radiol Oncol 35:47, 2001. Clin Orthop Relat Res 459:40–47, 2007.
109. Guillou L, Coindre JM, Bonichon F, et al: Comparative study of 135. Eyre R, Feltbower RG, James PW, et al: The epidemiology of
the National Cancer Institute and French Federation of Cancer bone cancer in 0-39 year olds in northern England, 1981-2002.
Centers Sarcoma Group grading systems in a population of 410 BMC Cancer 10:357, 2010.
adult patients with soft tissue sarcomas. J Clin Oncol 15:350–362, 136. Eyre R, Feltbower RG, Mubwandarikwa E, et al: Epidemiology
1997. of bone tumours in children and young adults. Pediatr Blood Cancer
110. Gustafson P: Soft tissue sarcoma. Epidemiology and prognosis in 53:941–952, 2009.
508 patients. Acta Orthop Scand Suppl 259:1–31, 1994. 137. Eyre R, Feltbower RG, Mubwandarikwa E, et al: Incidence and
111. Hasegawa T: Histological grading and MIB-1 labeling index of survival of childhood bone cancer in northern England and the
soft tissue sarcomas. Pathol Int 57:121–125, 2007. West Midlands, 1981-2002. Br J Cancer 100:188–193, 2009.
112. Hasegawa T, Yamamoto S, Yokoyama R, et al: Prognostic signifi- 138. Garcia RA, Inwards CY, Unni KK: Benign bone tumors—recent
cance of grading and staging systems using MIB-1 score in adult developments. Semin Diagn Pathol 28:73–85, 2011.
patients with soft tissue sarcoma of the extremities and trunk. 139. Grimer RJ, Cannon SR, Taminiau AM, et al: Osteosarcoma over
Cancer 95:843–851, 2002. the age of forty. Eur J Cancer 39:157–163, 2003.
113. Hasegawa T, Hirose T, Seki K, et al: Histological and immuno- 140. Homa DM, Sowers MFR, Schwartz AG: Incidence and survival
histochemical diversities, and proliferative activity and grading in rates of children and young adults with osteogenic sarcoma.
osteosarcomas. Cancer Detect Prev 21:280–287, 1997. Cancer 67:2219–2223, 1991.
114. Hashimoto H, Daimaru Y, Takeshita S, et al: Prognostic signifi- 141. Huvos AG: Osteogenic sarcoma of bones and soft tissues in older
cance of histologic parameters of soft tissue sarcomas. Cancer persons. A clinicopathologic analysis of 117 patients older than 60
70:2816–2822, 1992. years. Cancer 57:1442–1449, 1986.
115. Inwards CY, Unni KK: Classification and grading of bone sarco- 142. Huvos AG, Butler A, Bretsky SS: Osteogenic sarcoma in the
mas. Hematol Oncol Clin North Am 9:545–569, 1995. American black. Cancer 52:1959–1965, 1983.
116. Jensen V, Sorensen FB, Bentzen SM, et al: Proliferative activity 143. Jawad MU, Cheung MC, Min ES: Ewing sarcoma demonstrates
(MIB-1 index) is an independent prognostic parameter in patients racial disparities in incidence-related and sex-related differences
with high-grade soft tissue sarcomas of subtypes other than malig- in outcome: An analysis of 1631 cases from the SEER database,
nant fibrous hstiocytomas: a retrospective immunohistological 1973-2005. Cancer 115:3526–3536, 2009.
study. Histopathology 32:536–546, 1998. 144. Lee J, Hoang BH, Ziogas A, et al: Analysis of prognostic factors
117. Jensen OM, Hogh J, Ostgaard SE, et al: Histopathological in Ewing sarcoma using population-based cancer registry. Cancer
grading of soft tissue tumors. Prognostic significance in a 116:1964–1973, 2010.
prospective study of 278 consecutive cases. J Pathol 163:19–24, 145. Mirabellow L, Troisi RJ, Savage SA: Osteosarcoma incidence and
1991. survival rates from 1973 to 2004: data from the Surveillance,
118. Koea JB, Leung D, Lewis JJ, et al: Histopathologic type: an inde- Epidemiology, and End Results Program. Cancer 115:1531–1543,
pendent prognostic factor in primary soft tissue sarcoma of the 2009.
extremity. Ann Surg Oncol 10:432–440, 2003. 146. Mirabello L, Troisi RJ, Savage SA: International osteosarcoma
119. Markhede G, Angervall L, Stener B: A multivariate analysis of the incidence patterns in children and adolescents, middle aged and
prognosis after surgical treatment of malignant soft tumor tumors. elderly persons. Int J Cancer 125:229–234, 2009.
Cancer 49:1721–1733, 1982. 147. Ottaviani G, Jaffe N: The epidemiology of osteosarcoma. Cancer
120. Oliveira AM, Nascimento AG: Grading in soft tissue tumors: Treat Res 152:3–13, 2009.
principles and problems. Skeletal Radiol 30:543–559, 2001. 148. Parkin DM, Whelan SL, Ferlay J, et al: Cancer incidence in five
121. Parham DM, Webber BL, Jenkins JJ, 3rd, et al: Nonrhabdomyo- continents: volume VII IARC Scientific Publications, ed 1, 1997,
sarcomatous soft tissue sarcomas of childhood: formulation of a World Health Organization.
simplified system for grading. Mod Pathol 8:705–710, 1995. 149. Parkin DM, Stiller CA, Nectoux J: International variations in the
122. Peabody TD, Gibbs CP, Jr, Simon MA: Evaluation and staging of incidence of childhood bone tumours. Int J Cancer 53:371–376,
musculoskeletal neoplasms. J Bone Joint Surg Am 80:1204–1218, 1993.
1998. 150. Polednak AP: Primary bone cancer incidence in black and white
123. Rubin BP, Antonescu CR, Cooper K, et al: Protocol for the examina- residents of New York State. Cancer 55:2883–2888, 1985.
tion of specimens from patients with tumors of bone, based on AJCC/ 151. Rainusso N, Wang LL, Yustein JT: The adolescent and young
UICC TNM, ed 7, 2013, College of American Pathologists. adult with cancer: state of the art—bone tumors. Curr Oncol Rep
124. Russell WO, Cohen J, Enzinger F, et al: A clinical and pathologi- 15:296–307, 2013.
cal staging system for soft tissue sarcomas. Cancer 40:1562–1570, 152. Siegel R, Ma J, Zou Z, et al: Cancer statistics, 2014. CA Cancer J
1977. Clin 64:9–29, 2014.

ERRNVPHGLFRVRUJ
52 1  General Considerations

153. Smoll NR, Gautschi OP, Radovanovic I, et al: Incidence and rela- 182. Katayama I, Mochino T: Naphthol AS-D chloroacetate esterase
tive survival of chordomas: the standardized mortality ratio and reaction. Systemic modifications for optimization to smears and
the impact of chordomas on a population. Cancer 119:2029–2037, sections. Acta Pathol Jpn 33:381–393, 1983.
2013. 183. Kiernan J: Histological and histochemical methods: theory and practice,
154. Toro JR, Travis LB, Wu HJ, et al: Incidence patterns of soft tissue ed 4, Oxfordshire, 2008, Scion Publishing.
sarcomas, regardless of primary site, in the Surveillance, Epide- 184. Lack EE, Mercer L: A modified Grimelius argyrophil technique
miology and End Results program, 1978-2001: an analysis of for neurosecretory granules. Am J Surg Pathol 1:275–277,
26,758 cases. Int J Cancer 119:2922–2930, 2006. 1977.
155. Whelan J, McTiernan A, Cooper N, et al: Incidence and survival 185. Lauer BA, Reller LB, Mirrett S: Comparison of acridine orange
of malignant bone sarcomas in England 1979-2007. Int J Cancer and Gram stains for detection of microorganisms in cerebrospinal
131:508–517, 2011. fluid and other clinical specimens. J Clin Microbiol 14:201–205,
156. Wilkins RM, Pritchard DJ, Burgert EO, Jr, et al: Ewing’s sarcoma 1981.
of bone: experience with 140 patients. Cancer 58:2551–2555, 1986. 186. Lyon H, Andersen AP, Hasselager E, et al: Theory and strategy in
histochemistry: a guide to the selection and understanding of techniques,
Special Stains 2011, Springer.
157. Angra P, Ridderhof J, Smithwick R: Comparison of two different 187. Meier-Ruge WA, Bruder E: Current concepts of enzyme histo-
strengths of carbol fuchsin in Ziehl-Neelsen staining for detecting chemistry in modern pathology. Pathobiology 75:233–243, 2008.
acid-fast bacilli. J Clin Microbiol 41:3459, 2003. 188. Mourad W, Kfoury H, Al Husseini H: The value of CD34,
158. Ascoli V, Newman GA, Kline TS: Grimelius stain for cytodiag- myeloperoxidase and chloroacetate esterase (Leder) stain in the
nosis of carcinoid tumor. Diagn Cytopathol 2:157–159, 1986. diagnosis of granulocytic sarcoma. Ann Saudi Med 21:287–291,
159. Barbosa AJ, Castro LP, Margarida A, et al: A simple and economi- 2001.
cal modification of the Masson-Fontana method for staining 189. Najjar D, Naryshkin S, Ascol V, et al: Commentary on the rapid
melanin granules and enterochromaffin cells. Stain Technol microwave Grimelius stain for aspiration biopsy cytology. Diagn
59:193–196, 1984. Cytopathol 4:86, 1988.
160. Bottone EJ: The Gram stain: the century-old quintessential rapid 190. Naoumenko J, Feigin I: A simple silver solution for staining retic-
diagnostic test. Lab Med 19:288–291, 1988. ulin. Stain Technol 49:153–155, 1974.
161. Brady JG, Schultze GE, Seibert R, et al: Detection of mycobacte- 191. Naryshkin S, Daniels J, Freno E, et al: Cytology of treated and
rial infections using the Dieterle stain. Pediatr Dev Pathol 1:309– minimal Pneumocystis carinii pneumonia and a pitfall of the Grocott
313, 1998. methenamine silver stain. Diagn Cytopathol 7:41–47, 1991.
162. Bridges CH, Luna L: Kerr’s improved Warthin-Starry technique; 192. Pounder DJ: Warthin-Starry for Legionella. Am J Clin Pathol
study of the permissible variations. Lab Invest 6:357–367, 1957. 80:276, 1983.
163. Byard RW, Bellis M: The effect of decalcifying solutions on hemo- 193. Rungbyi J, Kassem M, Erisksen EF, et al: The von Kossa reaction
siderin staining. J Forensic Sci 55:1356–1358, 2010. for calcium deposits: silver lactate staining increases sensitivity and
164. Carriel VS, Aneiros-Fernandez J, Arias-Santiago S, et al: A novel reduces background. Histochem 25:446–451, 1993.
histochemical method for a simultaneous staining of melanin and 194. Swisher BL, Chandler FW: Grocott-Gomori methenamine silver
collagen fibers. J Histochem Cytochem 59:270–277, 2011. method for detecting fungi: practical considerations. Lab Med
165. Carson FL, Hladik C: Histotechnology: a self-instructional text, ed 3, 13:568–570, 1982.
Hong Kong, 2009, American Society for Clinical Pathology Press. 195. Tomita Y, Hariu A, Kato C, et al: Radical production during
166. Chinprasertsuk S, Piankijagum A, Issaragrisil S: Cytochemical tyrosinase reaction, dopa-melanin formation, and photoirradia-
stainings in acute promyelocytic leukemia: chloroacetate esterase tion of dopa-melanin. J Invest Dermatol 82:573–576, 1984.
reaction as a prognostic index. J Med Assoc Thai 74:87–91, 1991. 196. Van Orden AE, Greer PW: Modification of the Dieterle spiro-
167. Cohen RE, Aquirre A, Neiders ME, et al: Immunochemistry of chete stain. J Histotechnol 1:51–56, 1977.
high molecular-weight human salivary mucin. Arch Oral Biol 197. Vialli DM: Argentaffinity and argentophilia. I. A critical review
35:127–136, 1990. about the technique and the possibilities for histochemical
168. Culling CFA, Allison RT, Barr WT: Cellular pathology technique, research. Acta Histochem 60:103–120, 1977.
ed 4, Oxford, 1985, Butterworth-Heinemann. 198. Wick MR: Diagnostic histochemistry, New York, 2008, Cambridge
169. D’Cruze L, Dutta R, Rao S, et al: The role of immunohistochem- University Press.
istry in the analysis of the spectrum of small round cell tumours 199. Wong E, Morgan EW, MacDonald DM: The chloroacetate ester-
at a tertiary care centre. J Clin Diagn Res 7:1377–1382, 2013. ase reaction for mast cells in dermatopathology: a comparison
170. Elghetany MT, Saleem A: Methods for staining amyloid in tissues: with metachromatic staining methods. Acta Derm Venereol 62:431–
a review. Stain Technol 63:201–212, 1988. 434, 1982.
171. Eskelund V: Mucin staining with alcian blue. Acta Pathol Microbiol 200. Zappi E, Lombardo W: Combined Fontana-Masson/Perls’ stain-
Scand 40:107–109, 1957. ing. Am J Dermatopathol 6:143–145, 1984.
172. Filipe MI: Mucins in the human gastrointestinal epithelium: a
review. Invest Cell Pathol 2:195–216, 1979. Electron Microscopy
173. Fernandez-Flores A: A review of amyloid staining: methods and 201. Erlandson RA: Application of transmission electron microscopy
artifacts. Biotech Histochem 86:293–301, 2011. to human tumor diagnosis: an historical perspective. Cancer Invest
174. Gallyas F: Silver staining of collagen and reticulin fibres and 5:487–505, 1987.
cerebral capillaries by means of physical development. J Microsc 202. Erlandson RA: Diagnostic transmission electron microscopy of tumors
91:119–124, 1970. with clinicopathological, immunohistochemical and cytogenetic correla-
175. Garvey W: Modified elastic tissue—Masson trichrome stain. Stain tions, New York, 1994, Raven.
Technol 59:213–216, 1984. 203. Fisher C: The value of electron microscopy and immunohisto-
176. Gomori G: Microscopic histochemistry; principles and practice, 2012, chemistry in the diagnosis of soft tissue sarcomas: a study of 200
Ulan Press. cases. Histopathology 16:441–454, 1990.
177. Gomori G: The periodic-acid Schiff stain. Am J Clin Pathol 204. Goebel HH, Stenzel W: Ultrastructural myopathology in the
22:277–281, 1952. molecular era. Ultrastruct Pathol 37:328–331, 2013.
178. Gomori G: Aldehyde fuchsin: a new stain for elastic tissue. Am J 205. Latimer E, Anderson G, Sebire NJ: Ultrastructural features of
Clin Pathol 20:665–666, 1950. neuroblastic tumours in relation to morphological, and molecular
179. Gomori G: A rapid one-step trichrome stain. Am J Clin Pathol findings: a retrospective review study. BMC Clin Pathol 14:13,
20:661–664, 1950. 2014.
180. Grocott RG: A stain for fungi in tissue sections and smears using 206. Liapis H: Electron microscopy in kidney research: seeing is
Gomori’s methenamine-silver nitrate technique. Am J Clin Pathol believing. Ultrastruct Pathol 37:340–345, 2013.
25:975–979, 1955. 207. Mackay B, Ordóñez NG: Electron microscopy in the immunocy-
181. Hardonk MJ, Koudstaal J: Enzyme histochemistry as a link tochemical era. Adv Pathol 9:277–310, 1996.
between biochemistry and morphology. Prog Histochem Cytochem 208. Ordonez NG: Mesothelioma with signet-ring cell features: report
8:1–68, 1976. of 23 cases. Mod Pathol 26:370–384, 2013.

ERRNVPHGLFRVRUJ
1  General Considerations 53

209. Redemann S, Muller-Reichert T: Correlative light and electron Ewing’s sarcoma/primitive neuroectodermal tumor. Anal Quant
microscopy for the analysis of cell division. J Microsc 251:109–112, Cytol Histol 33:101–110, 2011.
2013. 233. Marshall T, Rutledge JC: Flow cytometry DNA applications
210. Stirling J, Curry A, Eyden B: Diagnostic electron microscopy: a practi- in pediatric tumor pathology. Pediatr Dev Pathol 3:314–334,
cal guide to tissue preparation and interpretation, ed 1, London, 2000.
United Kingdon, 2013, Wiley. 234. Melsen F, Mosekilde L: Tetracycline double-labelling of the iliac
trabecular bone in 41 normal adults. Calcif Tissue Res 26:99–102,
Cytometry and Histomorphometry 1978.
211. Arlot ME, Delmas PD, Chappard D, et al: Trabecular and endo- 235. Murata H, Kusuzaki Kuzuhara A, Takeshita H, et al: DNA ploidy
cortical bone remodeling in postmenopausal osteoporosis: com- alterations detected during dedifferentiation of periosteal chon-
parison with normal postmenopausal women. Osteoporos 1:41–49, drosarcoma. Anticancer Res 19:2285–2288, 1999.
1990. 236. Nunez R: DNA measurement and cell cycle analysis by flow
212. Bovee JV, van Royen M, Bardoel AF, et al: Near-haploidy and cytometry. Curr Issues Mol Biol 3:67–70, 2001.
subsequent polyploidization characterize the progression of 237. Pozarowski P, Darzynkiewicz Z: Analysis of cell cycle by flow
peripheral chondrosarcoma. Am J Pathol 157:1587–1595, 2000. cytometry. Methods Mol Biol 281:301–311, 2004.
213. Bovee JV, Cleton-Jansen AM, Kupers-Dijkshoorn NJ, et al: Loss 238. Roberts P, Burchill SA, Brownhill S, et al: Ploidy and karyotype
of heterozygosity and DNA ploidy point to a diverging genetic complexity are powerful prognostic indicators in the Ewing’s
mechanism in the origin of peripheral and central chondrosar- sarcoma family of tumors: a study by the United Kingdom Cancer
coma. Genes Chromosomes Cancer 26:237–246, 1999. Cytogenetics and the Children’s Cancer and Leukaemia Group.
214. Brahmi U, Rajwanshi A, Joshi K, et al: Role of immunocytochem- Genes Chromosomes Cancer 47:207–220, 2008.
istry and DNA flow cytometry in the fine-needle aspiration diag- 239. Serra M, Tarkkanen M, Baldini N, et al: Simultaneous paired
nosis of malignant small round-cell tumors. Diagn Cytopathol analysis of numerical chromosomal aberrations and DNA content
24:233–239, 2001. in osteosarcoma. Mod Pathol 14:710–716, 2001.
215. Bressot C, Meunier PJ, Chapuy MC, et al: Histomorphometric 240. Shih CS, Wang LS, Yang SS, et al: DNA flow cytometric analysis
profile, pathophysiology and reversibility of corticosteroid- of chest-wall chondroma and chondrosarcoma. Scand J Thorac
induced osteoporosis. Metab Bone Dis Relat Res 1:303–311, 1979. Cardiovasc Surg 30:157–161, 1996.
216. Chavassieux PM, Arlot ME, Reda C, et al: Histomorphometric
assessment of the long-term effects of alendronate on bone quality Immunohistochemistry
and remodeling in patients with osteoporosis. J Clin Invest 241. Akiyama H, Lefebvre V: Unraveling the transcriptional regulatory
100:1475–1480, 1997. machinery in chondrogenesis. J Bone Miner Metab 29:390–395,
217. Coughlan B, Feliz A, Ishida T, et al: p53 expression and DNA 2011.
ploidy of cartilage lesions. Hum Pathol 26:620–624, 1995. 242. Ali IH, Brazil DP: Bone morphogenetic proteins and their antago-
218. Darzynkiewicz Z, Juan G, Bedner E: Determining cell cycle stages nists: current and emerging clinical uses. Br J Pharmacol 171:3620–
by flow cytometry. Curr Protoc Cell Biol chapter 8: unit 8.4 2001. 3632, 2014.
219. Darzynkiewicz Z, Bedner E, Smolewski P: Flow cytometry in 243. Altmannsberger M, Weber K, Droste R, et al: Desmin is a specific
analysis of cell cycle and apoptosis. Semin Hematol 38:179–193, marker for rhabdomyosarcomas of human and rat origin. Am J
2001. Pathol 118:85–95, 1985.
220. Dempster DW: Histomorphometric analysis of bone remodeling. 244. Ambros IM, Ambros PF, Strehl S, et al: MIC2 is a specific marker
In Bilezikian JP, Raisz LG, Martin TJ, editors: Principles of bone for Ewing’s sarcoma and peripheral primitive neuroectodermal
biology, vol 1, ed 3, Amsterdam, 2008, Elsevier, pp 85–108. tumors: evidence for a common histogenesis of Ewing’s sarcoma
221. Eriksen EF, Langdahl B, Vesterby A, et al: Hormone replacement and peripheral primitive neuroectodermal tumors from MIC2
therapy prevents osteoclastic hyperactivity: a histomorphometric expression and specific chromosome aberration. Cancer 67:1886–
study in early postmenopausal women. J Bone Miner Res 14:1217– 1893, 1991.
1221, 1999. 245. Amir G, Issakov J, Meller I, et al: Expression of 053 gene product
222. Helio H, Karaharju E, Bohling T, et al: Chondrosarcoma of bone. and cell proliferation marker Ki-67 in Ewing’s carcoma: correla-
A clinical and DNA flow cytometric study. Eur J Surg Oncol tion with clinical outcome. Hum Pathol 33:170–4174, 2002.
21:408–413, 1995. 246. Arrabal PM, Visser R, Santos-Ruiz L, et al: Osteogenic molecules
223. Herget GW, Neuburger M, Adler CP: Prognostic significance of for clinical applications: improving the BMP-collagen system. Biol
nuclear DNA content in chondrosarcoma. Ann Diagn Pathol 4:11– Res 46:421–429, 2013.
16, 2000. 247. Baek WY, Kim JE: Transcriptional regulation of bone formation.
224. Jayat C, Ratinaud MH: Cell cycle analysis by flow cytometry: Front Biosci 3:126–135, 2011.
principles and applications. Biol Cell 78:15–25, 1993. 248. Bar H, Strelkov SV, Gjoberg G, et al: The biology of desmin fila-
225. Karantzoulis V, Liapi C, Papagelopoulos P: Large-scale bone ments: how do mutations affect their structure, assembly and
mineral histomorphometry: report of a simplified technique. His- organization? J Struct Biol 148:137–152, 2004.
topathology 61:1174–1182, 2012. 249. Barrionuevo F, Scherer G: SOX E genes: SOX9 and SOX8 in
226. Koss LG, Czerniak B, Herz F, et al: Flow cytometric measure- mammalian testis development. Int J Biochem Cell Biol 42:433–436,
ments of DNA and other cellular components in human tumors: 2010.
a critical appraisal. Hum Pathol 20:528–548, 1989. 250. Blake JA, Ziman MR: Pax genes: regulators of lineage specifica-
227. Kreicbergs A, Silvferswärd C, Tribukait B: Flow DNA analysis of tion and progenitor cell maintenance. Development 141:737–751,
primary bone tumors: relationship between cellular DNA content 2014.
and histopathologic classification. Cancer 53:129–136, 1984. 251. Blanchoin L, Boujemaa-Paterski R, Sykes C, et al: Actin dynam-
228. Kusuzaki K, Murata H, Takeshita H, et al: Usefulness of cytofluo- ics, architecture, and mechanics in cell motility. Physiol Rev
rometric DNA ploidy analysis in distinguishing benign cartilagi- 94:235–263, 2014.
nous tumors from chondrosarcomas. Mod Pathol 12:863–872, 252. Braun T, Gautel M: Transcriptional mechanisms regulating skel-
1999. etal muscle differentiation, growth and homeostasis. Nat Rev Mol
229. Kusuzaki K, Takeshita H, Murata H, et al: Prognostic value of Cell Biol 12:349–361, 2011.
DNA ploidy response to chemotherapy in human osteosarcomas. 253. Buckingham M, Rigby PW: Gene regulatory networks and tran-
Cancer Lett 141:131–138, 1999. scriptional mechanisms that control myogenesis. Dev Cell 28:225–
230. Kusuzaki K, Takeshita H, Murata H, et al: Prognostic significance 238, 2014.
of DNA ploidy pattern in osteosarcomas in association with che- 254. Cajaiba MM, Luo J, Goodman MA, et al: Sox9 expression is not
motherapy. Cancer Lett 137:27–33, 1999. limited to chondroid neoplasms: variable occurrence in other soft
231. Look AT, Douglass EC, Meyer WH: Clinical importance of near- tissue and bone tumors with frequent expression by synovial sar-
diploid tumor stem lines in patients with osteosarcoma of an comas. Int J Surg Pathol 18:319–323, 2010.
extemity. N Engl J Med 318:1567–1572, 1988. 255. Carreira AC, Lojudice FH, Halcsik E, et al: Bone morphogenetic
232. Machado I, Ruiz-Sauri A, Lopez-Guerrero JA, et al: Morphomet- proteins: facts, challenges, and future perspectives. J Dent Res
ric analysis of DNA ploidy and nuclear cell cycle regulators in 93:335–345, 2014.

ERRNVPHGLFRVRUJ
54 1  General Considerations

256. Castillo SD, Sanchez-Cespedes M: The SOX family of genes in 279. Holthofer H, Virtanen I, Kariniemi AL, et al: Ulex europaeus I
cancer development: biological relevance and opportunities for lectin as a marker for vascular endothelium in human tissues. Lab
therapy. Expert Opin Ther Targets 16:903–919, 2012. Invest 47:60–66, 1982.
257. Celis JE, Celis A: Cell cycle-dependent variations in the distribu- 280. Hoyer LW: The factor VIII complex: structure and function. Blood
tion of the nuclear protein cyclin proliferating cell nuclear antigen 58:1–13, 1981.
in cultured cells: subdivision of S phase. Proc Natl Acad Sci USA 281. Huuhtanen RL, Blomqvist CP, Wiklund TA, et al: Comparison
82:3262–3266, 1985. of the Ki-67 score and S-phase fraction as prognostic variables in
258. Choong PF, Akerman M, Willen H, et al: Expression of prolifer- soft-tissue sarcoma. Br J Cancer 79:945–951, 1999.
ating cell nuclear antigen (PCNA) and Ki-67 in soft tissue 282. Jiang T, Hou CC, She ZY, et al: The SOX gene family: function
sarcoma. Is prognostic significance histiotype-specific? APMIS and regulation in testis determination and male fertility mainte-
103:797–805, 1995. nance. Mol Biol Rep 40:2187–2194, 2013.
259. Chung BM, Rotty JD, Coulombe PA: Networking galore: inter- 283. Jirasek T, Hozak P, Mandys V: Different patterns of chromogranin
mediate filaments and cell migration. Curr Opin Cell Biol 25:600– A and Leu-7 (CD57) expression in gastrointestinal carcinoids:
612, 2013. immunohistochemical and confocal laser scanning microscopy
260. D’amico MA, Ghinassi B, Izzicupo P, et al: Biological function study. Neoplasm 50:107, 2003.
and clinical relevance of chromogranin A and derived peptides. 284. Johnson MD, Glick AD, Davis BW: Immunohistochemical evalu-
Endocr Connect 3:45–54, 2014. ation of Leu-7, myelin basic-protein, S100-protein, glial-fibrillary
261. De Biasio A, Blanco FJ: Proliferating cell nuclear antigen struc- acidic-protein, and LN3 immunoreactivity in nerve sheath tumors
ture and interactions: too many partners for one dancer? Adv and sarcomas. Arch Pathol Lab Med 112:155–160, 1988.
Protein Chem Struct Biol 91:1–36, 2013. 285. Jong R, Davis AM, Mendes MG, et al: Proliferative activity
262. De Young BR, Wick MR, Fitzgibbon JF, et al: CD31: an immu- (KI-67 expression) and outcome in high grade osteosarcomas: a
nospecific marker for endothelial differentiation in human neo- study of 27 cases. Sarcoma 4:4755, 2000.
plasms. Appl Immunohistochem 1:97, 1993. 286. Jones CI, Moraes LA, Gibbins JM: Regulation of platelet biology
263. Dryden NH, Sperone A, Martin-Almedina S, et al: The transcrip- by platelet endothelial cell adhesion molecule-1. Platelets 23:331–
tion factor ERG controls endothelial cell quiescence by repressing 335, 2012.
activity of nuclear factor. J Biol Chem 287:12331–12342, 2012. 287. Kanatous SB, Mammen PP: Regulation of myoglobin expression.
264. Duque M, Modlin IM, Gupta A, et al: Biomarkers in neuroendo- J Exp Biol 213:2741–2747, 2010.
crine tumors. JOP 14:372–376, 2013. 288. Kawaguchi Y: Sox9 and programming of liver and pancreatic
265. Emanuele MJ, Ciccia A, Elia AE, et al: Proliferating cell nuclear progenitors. J Clin Invest 123:1881–1886, 2013.
antigen (PCNA)-associated KIAA0101/PAF15 protein is a cell 289. Kawahara E, Oda Y, Ooi A, et al: Expression of glial fibrillary
cycle-regulated anaphase-promoting complex/cyclosome sub- acidic protein (GFAP) in peripheral nerve sheath tumors: a com-
strate. PNAS 108:9845–9850, 2011. parative study of immunoreactivity of GFAP, vimentin, S-100
266. Fanburg-Smith JC, Auerbach A, Marwaha JS, et al: Reappraisal protein, and neurofilament in 38 schwannomas and 18 neurofi-
of mesenchymal chondrosarcoma: novel morphologic observa- bromas. Am J Surg Pathol 12:115–120, 1988.
tions of the hyaline cartilage and endochondral ossification and 290. Kidd ME, Shumaker DK, Ridge KM: The role of vimentin inter-
beta-catenin, Sox9, and osteocalcin immunostaining of 22 cases. mediate filaments in the progression of lung cancer. Am J Respir
Hum Pathol 41:653–662, 2010. Cell Mol Biol 50:1–6, 2014.
267. Fellinger EJ, Garin-Chesa P, Triche TJ, et al: Immunohistochem- 291. Kim KK, Adelstein RS, Kawamoto S: Identification of neuronal
ical analysis of Ewing’s sarcoma cell surface and antigen p30/32MIC2. nuclei (NeuN) as Fox-3, a member of the Fox-1 gene family of
Am J Pathol 139:317–325, 1991. splicing factors. J Biol Chem 284:31052–31061, 2009.
268. Folpe AL, Hill CE, Parham DM, et al: Immunohistochemical 292. Kiss B, Karsai A, Kellermayer MSZ: Nanomechanical properties
detection of FLI-1 protein expression: a study of 132 round of desmin intermediate filaments. J Struct Biol 155:327–339, 2006.
cell tumors with emphasis on CD99-positive mimics of Ewing’s 293. Komori T: Signaling networks in RUNX2-dependent bone devel-
sarcoma/primitive neuroectodermal tumor. Am J Surg Pathol opment. J Cell Biochem 112:750–755, 2011.
24:1657–1662, 2000. 294. Komori T: Regulation of bone development and extracellular
269. Gerdes J, Lemke H, Baisch H, et al: Cell cycle analysis of a matrix protein genes by RUNX2. Cell Tissue Res 339:189–195,
cell proliferation-associated human nuclear antigen defined by 2010.
the monoclonal antibody Ki-67. J Immunol 133:1710–1715, 295. Li CG, Eccles MR: PAX genes in cancer: friends or foes. Frontiers
1984. Genet 3:1–7, 2012.
270. Goldstein NS, Hewitt SM, Taytlor CR, et al: Recommendation 296. Ligon KL, Alberta JA, Kho AT, et al: The oligodendroglial
for improved standardization of immunohistochemistry. Appl lineage marker OLIG2 is universally expressed in diffuse gliomas,.
Immunohistochemistry Mol Morphol 15:124–133, 2007. J Neuropathol Exp Neurol 63:499–509, 2004.
271. Gould VE, Wiedenmann B, Lee I, et al: Synaptophysin expression 297. Liu TM, Lee EH: Transcriptional regulatory cascades in RUNX2-
in neuroendocrine neoplasms as determined by immunocyto- dependent bone development. Tissue Eng Part B Rev 19:254–263,
chemistry. Am J Pathol 126:243–257, 1987. 2013.
272. Gros G, Wittenberg BA, Jue T: Myoglobin’s old and new clothes: 298. Llombart-Bosch A, Navarro S: Immunohistochemical detection
from molecular structure to function in living cells. J Exp Biol of EWS and FLI-1 proteins in Ewing sarcoma and primitive
213:2713–2725, 2010. neuroectodermal tumors: comparative analysis with CD99
273. Guarda LA, Ordóñez NG, Smith JL, Jr, et al: Immunoperoxidase (MIC-2) expression. Appl Immunohistochem Mol Morphol 9:255–
localization of factor VIII in angiosarcoma. Arch Pathol Lab Med 260, 2001.
106:515–516, 1982. 299. Lucero CM, Vega OA, Osorio MM, et al: The cancer-related
274. Haines RL, Lane EB: Keratins and disease at a glance. J Cell Sci transcription factor RUNX2 modulates cell proliferation in
125:3923–3928, 2012. human osteosarcoma cell lines. J Cell Physiol 228:714–723, 2013.
275. Harris ML, Baxter LL, Loftus SK, et al: Sox proteins in melano- 300. Maga G, Hubscher U: Proliferating cell nuclear antigen (PCNA):
cyte development and melanoma. Pigment Cell Melanoma Res a dancer with many partners. J Cell Science 116:3051–3060, 2003.
23:496–513, 2010. 301. Malki S, Boizet-Bonhoure B, Poulat F: Shuttling of SOX proteins.
276. Hasselblatt M, Paulus W: Sensitivity and specificity of epithelial Int J Biochem Cell Biol 42:411–416, 2010.
membrane antigen staining patterns in ependymomas. Acta Neu- 302. Manivel JC, Wick MR, Swanson PE, et al: Epithelial membrane
ropathol 106:385–388, 2003. antigen in sarcomas. Lab Invest 56:46A, 1987.
277. Hernandez-Rodriguez NA, Correa E, Sotelo R, et al: Ki-67: a 303. Miettinen M, Wang Z, Sarlommo-Rikala M, et al: ERG expres-
proliferative marker that may predict pulmonary metastases and sion in epitheloid sarcoma: a diagnostic pitfall. Am J Surg Pathol
mortality of primary osteosarcoma. Cancer Detect Prev 25:210– 37:1580–1585, 2013.
215, 2001. 304. Miettinen M, Wang ZF, Paetau A, et al: ERG transcription factor
278. Heyderman E, Steele K, Ormerod MG: A new antigen on the as an immunohistochemical marker for vascular endothelial
epithelial membrane: its immunoperoxidase localization in normal tumors and prostatic carcinoma. Am J Surg Pathol 35:432–441,
and neoplastic tissue. J Clin Pathol 32:35–39, 1979. 2011.

ERRNVPHGLFRVRUJ
1  General Considerations 55

305. Miettinen M: Synaptophysin and neurofilament proteins as 331. Sloane JP, Ormerod MG: Distribution of epithelial membrane
markers for neuroendocrine tumors. Arch Pathol Lab Med 111: antigen in normal and neoplastic tissue and its value in diagnostic
813–818, 1987. tumor pathology. Cancer 47:1786–1795, 1981.
306. Moll R, Divo M, Langbein L: The human keratins: biology and 332. Snider NT, Omary MB: Post-translational modifications of inter-
pathology. Histochem Cell Biol 129:705–733, 2008. mediate filament proteins: mechanisms and functions. Nat Rev
307. Mukai K, Rosai J, Hallaway BE: Localization of myoglobin in Mol Cell Biol 15:163–177, 2014.
normal and neoplastic human skeletal muscle cells using an immu- 333. Soderstrom M, Palokangas T, Vahlberg T, et al: Expression of
noperoxidase method. Am J Surg Pathol 3:373–376, 1979. ezrin, Bcl-2, and Ki-67 in chondrosarcomas. APMIS 118:769–776,
308. Mullen RJ, Buck CR, Smith AM: NeuN, a neuronal specific 2010.
nuclear protein in vertebrates. Development 116:201–211, 1992. 334. Sperone A, Dryden NH, Birdsey GM, et al: The transcription
309. Nakajima T, Watanabe S, Sato Y, et al: An immunoperoxidase factor ERG inhibits vascular inflammation by repressing
study of S-100 protein distribution in normal and neoplastic NF-kappaB activation and proinflammatory gene expression in
tissues. Am J Surg Pathol 6:715–727, 1982. endothelial cells. Arterioscler Thromb Vasc Biol 31:142–150, 2011.
310. Nakajima T, Watanabe S, Sato Y, et al: Immunohistochemical 335. Sreenath TL, Dobi A, Petrovics G, et al: Oncogenic activation of
demonstration of S-100 protein in malignant melanoma and pig- ERG: a predominant mechanism in prostate cancer. J Carcinog
mented nevus and its diagnostic application. Cancer 50:912–918, 10:37, 2011.
1982. 336. Taylor CR, Rudbeck L: Immunohistochemical staining methods, ed
311. Nawa G, Ueda T, Mori S, et al: Prognostic significance of Ki67 6, Dako Denmark A/S, 2013.
(MIB1) proliferation index and p53 over-expression in chondro- 337. Walker RA: The use of lectins in histopathology. Pathol Res Pract
sarcomas. Int J Cancer 69:86–91, 1996. 185:826–835, 1989.
312. Nickoloff BJ: The human progenitor cell antigen (CD-34) is 338. Wehrli BM, Huang W, de Crombrugghe B, et al: Sox9, a master
localized on endothelial cells, dermal dendritic cells, and perifol- regulator of chondrogenesis, distinguishes mesenchymal chon-
licular cells in formalin-fixed normal skin, and on proliferating drosarcoma from other small blue round cell tumors. Hum Pathol
endothelial cells and stromal spindle-shaped cells in Kaposi’s 34:263–269, 2003.
sarcoma. Arch Dermatol 127:523–529, 1991. 339. Weinstein LJ, McCarthy EF: Ki-67 immunostaining as a tool in
313. Nikolova-Krstevski V, Yuan L, Le Bras A, et al: ERG is required the diagnosis of central cartilage lesions. Iowa Orthop 16:39–45,
for the differentiation of embryonic stem cells along the endothe- 1996.
lial lineage. BMC Dev Biol 9:72, 2009. 340. Won KY, Park HR, Park YK: Prognostic implication of immuno-
314. Nishimura R, Hata K, Takashima R, et al: Modulation of tran- histochemical Runx2 expression in osteosarcoma. Tumori 95:311–
scriptional regulation during bone and cartilage development and 316, 2009.
their disease. Clin Calcium 23:1585–1593, 2013. 341. Yang J, Zhao L, Tian W, et al: Correlation of WWOX, RUNX2,
315. Nishimura R, Hata K, Matsubara T, et al: Regulation of bone and and VEGFA protein expression in human osteosarcoma. BMC
cartilage development by network between BMP signaling and Med Genomics 6:56, 2013.
transcription factors. J Biochem 151:247–254, 2012. 342. Zehentner BK, Dony C, Burtscher H: The transcription factor
316. Oakley GJ, Fuhrer K, Seethala RR: Brachyury, Sox9, and podo- Sox9 is involved in BMP-2 signaling. J Bone Miner Res 14:1734–
planin, new markers in the skull base chordoma vs chondrosar- 1741, 1999.
coma differential: a tissue microarray-based comparative analysis. 343. Zhang C: Molecular mechanisms of osteoblast-specific transcrip-
Mod Pathol 21:1461–1469, 2008. tion factor Osterix effect on bone formation. Beijing Da Xue Xue
317. Ordonez NG: Immunohistochemical endothelial markers: a Bao 44:659–665, 2012.
review. Adv Anat Pathol 19:281–295, 2012. 344. Zhu H, Tang J, Tang M, et al: Upregulation of Sox9 in osteosar-
318. Paixao-Cortes VR, Salzano FM, Bortolini MC: Evolutionary coma and its association with tumor progression and patients’
history of chordate PAX genes: dynamics of change in a complex prognosis. Diagn Pathol 8:183, 2013.
gene family. PLoS ONE 8:e73560, 2013.
319. Pan X, Hobbs RP, Coulombe PA: The expanding significance of Cytogenetics
keratin intermediate filaments in normal and diseased epithelia. 345. Antonescu C: Round cell sarcomas beyond Ewing: emerging enti-
Curr Opin Cell Biol 25:47–56, 2013. ties. Histopathology 64:26–37, 2014.
320. Paulin D, Li Z: Desmin: a major intermediate filament protein 346. Aurias A, Rimbaut C, Buffe C, et al: Chromosomal translocations
essential for the structural integrity and function of muscle. Exp in Ewing’s sarcoma. N Engl J Med 309:496–498, 1983.
Cell Res 301:1–7, 2004. 347. Boveri T: Zur Frage der Entstehung maligner tumoren, Germany,
321. Paunesku T, Mittal S, Protic M, et al: Proliferating cell nuclear 1914, Gustav Fischer Jena, p 64.
antigen (PCNA): ringmaster of the genome. Int J Radiat Biol 348. Bridge JA: The role of cytogenetics and molecular diagnostics
77:1007–1021, 2001. in the diagnosis of soft-tissue tumors. Mod Pathol 27:80–97,
322. Pusztaszeri MP, Seelentag W, Bosman FT: Immunohistochemical 2014.
expression of endothelial markers CD31, C34, von Willebrand 349. Bridge JA, Cushman-Vokoun AM: Molecular diagnostics of soft
factor, and Fli-1 in normal human tissues. J Histochem Cytochem tissue tumors. Arch Pathol Lab Med 135:588–601, 2011.
54:385–395, 2006. 350. Byrne M, Wray J, Reinert B, et al: Mechanisms of oncogenic
323. Rangdaeng S, Truong LD: Comparative immunohistochemical chromosomal translocations. Ann NY Acad Sci 1310:89–97, 2014.
staining for desmin and muscle specific actin: a study of 576 cases. 351. Campbell LJ: Cancer cytogenetics: methods and protocols (methods in
Am J Clin Pathol 96:32–45, 1991. molecular biology), ed 2, New York, 2011, Humana Press.
324. Sandbo N, Dulin N: Actin cytoskeleton in myofibroblast differ- 352. Chen X, Bahrami A, Pappo A, et al: St. Jude’s Children’s Research
entiation: ultrastructure defining form and driving function. Hospital-Washington University Pediatric Cancer Genome
Transl Res 158:181–196, 2011. Project. Cell Rep 7:104–112, 2014.
325. Satelli A, Li S: Vimentin in cancer and its potential as a molecular 353. Delattre O, Zucman J, Plougastel B, et al: Gene fusion with an
target for cancer therapy. Cell Mol Life Sci 68:3033–3046, 2011. ETS DNA-binding domain caused by chromosome translocation
326. Schweizer J, Bowden PE, Coulombe PA, et al: New consensus in human tumours. Nature 359:162–165, 1992.
nomenclature for mammalian keratins. J Cell Biol 174:169–174, 354. Das K, Tan P: Molecular cytogenetics: recent developments and
2006. applications in cancer. Clin Genet 87:315–325, 2013.
327. Scotlandi K, Serra M, Manara MC, et al: Clinical relevance of 355. Groffen J, Stephenson JR, Heisterkamp N, et al: Philadelphia
Ki-67 expression in bone tumors. Cancer 75:806–814, 1995. chromosomal breakpoints are clustered within a limited region,
328. Sharon N, Lis H: Lectins as cell recognition molecules. Science bcr, on chromosome 22. Cell 36:93–99, 1984.
246:227–234, 1989. 356. Heim S, Mitelman F: Cancer cytogenetics: chromosomal and molec-
329. Singh K, Dilworth FJ: Differential modulation of cell cycle pro- ular genetic aberrations of tumor cells, ed 3, New York, 2009,
gression distinguishes members of the myogenic regulatory factor Wiley-Blackwell.
family of transcription factors. FEBS J 280:3991–4003, 2013. 357. Holland AJ, Cleveland DW: Boveri revisited: chromosomal insta-
330. Sinha KM, Zhou X: Genetic and molecular control of Osterix in bility, aneuploidy and tumorigenesis. Nat Rev Mol Cell Biol 10:478–
skeletal formation. J Cell Biochem 114:975–984, 2013. 487, 2009.

ERRNVPHGLFRVRUJ
56 1  General Considerations

358. Howe B, Umrigar A, Tsien F: Chromosome preparation from 367. Nowell P, Hungerford D: A minute chromosome in human
cultured cells. J Vis Exp 83:50203, 2014. chronic granulocytic leukemia. Science 132:1497, 1960.
359. Imataka G, Arisaka O: Chromosome analysis using spectral karyo- 368. Panagopoulos I, Thorsen J, Gorunova L, et al: Sequential com-
typing (SKY). Cell Biochem Biophys 62:13–17, 2012. bination of karyotyping and RNA-sequencing in the search for
360. Jour G, Scarborough JD, Jones RL, et al: Molecular profiling of cancer-specific fusion genes. Int J Biochem Cell Biol 53:462–465,
soft tissue sarcomas using next-generation sequencing: a pilot 2014.
study toward precision therapeutics. Hum Pathol 46:1563–1571, 369. Qadir MA, Zhan SH, Kwok B, et al: ChildSeq-RNA: a next-
2014. generation sequencing-based diagnostic assay to identify known
361. Kuijjer ML, Hogendoorn PC, Cleton-Jansen AM: Genome-wide fusion transcripts in childhood sarcomas. J Mol Diagn 16:361–370,
analysis on high-grade osteosarcoma: making sense of a genomi- 2014.
cally most unstable tumor. Int J Cancer 133:2512–2521, 2013. 370. Roukos V, Misteli T: The biogenesis of chromosome transloca-
362. Liehr T, Weise A, Hamid AB, et al: Kosyakova N: multicolor tions. Nat Cell Biol 16:293–300, 2014.
FISH methods in current clinical diagnostics. Expert Rev Mol 371. Rowley JD: Letter: a new consistent chromosomal abnormality in
Diagn 13:251–255, 2013. chronic myelogenous leukaemia identified by quinacrine fluores-
363. Lynnn M, Wang Y, Slater J, et al: High-resolution genome-wide cence and Giemsa staining. Nature 243:290–293, 1973.
copy-number analyses identify localized copy-number alterations 372. Turc-Carel C, Philip I, Berge RMP, et al: Chromosomal translo-
in Ewing sarcoma. Diagn Mol Pathol 22:76–84, 2013. cations in Ewing’s sarcoma. N Engl J Med 309:497–498, 1983.
364. MacLeod RA, Drexler HG: Classical and molecular cytogenetic 373. Von Hansemann D: Ueber asymmetrische Zelltheilung in epithel-
analysis. Methods Mol Biol 946:39–60, 2013. hresbsen und deren biologische bedeutung. Virchows Arch A Pathol
365. Nowell PC: Discovery of the Philadelphia chromosome: a per- Anat 119:299–326, 1890.
sonal perspective. J Clin Invest 117:2033–2035, 2007. 374. Zheng J: Oncogenic chromosomal translocations and human
366. Nowell PC, Rowley JD, Knudson AG, Jr: Cancer genetics, cancer. Oncol Rep 30:2011–2019, 2013.
cytogenetics—defining the enemy within. Nat Med 4:1107–1111,
1998.

ERRNVPHGLFRVRUJ
C H A P T E R 2 

Clinical Considerations and


Imaging of Bone Tumors
Colleen M. Costelloe, MD  •  John E. Madewell, MD

CHAPTER OUTLINE

CLINICAL CONSIDERATIONS RESPONSE TO THERAPY


Age Primary Bone Tumors
Location Bone Metastases
INITIAL STAGING RESTAGING AND ONCOLOGIC SURVEILLANCE
Diagnostic Principles Local Recurrence
Imaging Modalities Distant Metastases
Imaging Characterization of Primary
Bone Tumors

INTRODUCTION pathologist work in concert and share their information.


The clinical history, age of the patient, location of the
Bone lesions lend themselves to radiographic assessment lesion, and radiographic appearance provide a foundation
because of the fact that bone has a built-in contrast agent, for the analytic approach to the diagnosis of bone tumors.
the calcium hydroxyapatite crystal. Much information It is important to state that the identification of a bone
about bone destruction, bone production, matrix calcifi- abnormality per se does not definitively indicate that a
cation and ossification, and the reactive response of the biopsy or indeed any surgical intervention is required.
surrounding bone and periosteum is available from plain Easily recognizable fibrous cortical defects, small osteo-
radiographs. Although other imaging techniques play a chondromas, phalangeal enchondromas, and common
role in the diagnostic process, the plain film is still the traumatic and avulsion lesions can be treated by observa-
method of choice and should never be bypassed. Imaging tion alone. Often these are incidental findings observed
is an important tool for diagnosis, staging, therapeutic on radiographs taken for other reasons. Lesions that
response evaluation, and oncologic surveillance of bone cannot be confidently assigned to a benign radiologic
tumors and can be used to augment histopathologic find- category or that have aggressive or malignant appear-
ings. Imaging modalities such as radiography, computed ances require biopsy.
tomography (CT), and magnetic resonance imaging
(MRI) have major roles in the initial characterization and
identification of bone lesions through the assessment of CLINICAL CONSIDERATIONS
characteristics such as margin, bone expansion, periosteal
reaction, soft tissue extension, and matrix mineralization. The analytic approach begins with an appreciation that
Additional imaging modalities such as bone scan, single- tumors of bone have different predilections for certain age
photon emission computed tomography with fused CT groups, anatomic locations, and even specific sites within
(SPECT/CT), 18F-fluorodeoxyglucose positron emission a bone.42,72-81,83,84 Some lesions are painless, and some
tomography with fused CT (FDG PET/CT), and whole- present with specific pain patterns. Thus a small, painful
body MRI can be used to stage patients for distant metas- lesion in the neck of the femur in an adolescent is far more
tases. These modalities, in addition to ultrasonography, likely to be an osteoid osteoma than a chondroblastoma.
can also be used for restaging and oncologic surveillance. A large lytic lesion in the end of a long bone of an adult
In this chapter, we discuss the general imaging and diag- is likely to be a giant cell tumor and not a nonossifying
nostic features of bone tumors, assessment of therapeutic fibroma. Computerized radiographic techniques, such as
responses, and staging of both primary and metastatic CT and MRI, facilitate analysis of bone lesions in greater
bone tumors. Specific imaging features of individual bone detail and are indispensable techniques in planning
tumors and radiographic-pathologic correlations are dis- therapy. They also provide reliable clues for the patholo-
cussed in their respective chapters. gist.71,78,82 The clinical, radiographic, and pathologic rec-
In general, at least 95% of bone tumors can be diag- ognition patterns of the most common bone tumors and
nosed with precision when the clinician, radiologist, and tumorlike lesions are summarized in Table 2-1.
57
ERRNVPHGLFRVRUJ
58 2  Clinical Considerations and Imaging of Bone Tumors

TABLE 2-1 Summary of Clinicopathologic Recognition Patterns of Most Common Bone Tumor and
Tumorlike Lesions
Peak Age
Incidence
Type of Tumor (years) Most Frequent Sites Radiographic Features Microscopic Features
Malignant osteosarcoma 10-20 Metaphyseal parts of Destructive lytic or blastic Highly atypical sarcomatoid
long bones (knee, lesion with cloudlike cells exhibiting tumor
proximal humerus) opacities bone formation
Chondrosarcoma >50 Pelvis, proximal femur Destructive lesion with Exclusively cartilaginous
punctate calcifications, tumor with atypical
cortical scalloping, or cartilage cells
disruption
Ewing’s sarcoma 10-20 Shafts of long bones Permeative, lytic pattern of Small cell tumor
bone destruction
Malignant fibrous >50 Pelvis, long bones, Lytic moth-eaten or Highly atypical spindle or
histiocytoma predominantly lower permeative bone pleomorphic cell sarcoma
extremity (femur, destruction with cortical (no tumor bone
tibia) disruption production)
Adamantinoma of long >20 Predominantly tibia Lytic lesion with Nests of epithelioid cells in
bone with synchronous predominant fibrous stroma
involvement of involvement of
fibula anterolateral cortex
Borderline giant cell 20-40 Ends of long tubular Eccentric epiphyseal lytic Multiple osteoclast-like giant
tumor bones (knee regions, lesion cells with mononuclear
distal radius) stromal cells
Epithelioid >30 Lower extremity, Multifocal, sharply Vascular neoplasm with
hemangioendothelioma multifocal demarcated lytic lesions epithelioid endothelial
involving bones of one cells
extremity (predominantly
lower extremity)
Benign osteoid osteoma 10-30 Long bones of lower Well-demarcated Interlacing network of bone
extremity (femoral intracortical lytic lesion trabeculae in vascular
neck) with sclerosis of adjacent stroma
cortex
Osteoblastoma 5-45 Posterior neural arch Similar to osteoid osteoma Same as osteoid osteoma
of vertebral column (radius >1.5 cm), less
and craniofacial sclerosis
bones
Enchondroma Wide- Small bones of hands Radiolucent defect with Mature cartilage (low
range and feet, long punctate or stippled cellularity)
tubular bones calcifications (<2 cm)
Chondroblastoma 10-20 Epiphyses of long Sharply demarcated Tumor composed of
tubular bones in epiphyseal defect chondroblasts with focal
skeletally immature immature cartilage
patients formation (chicken wire
calcifications)
Chondromyxoid fibroma 10-30 Metaphyses of long Eccentric cortical-based Myxoid tissue with septa
tubular bones lytic defect with containing chondroblasts
scalloped margin and giant cells
Hemangioma >40 Craniofacial bones and Lytic defect with prominent Vascular lesion with
vertebral bodies striations; honeycomb capillary or cavernous
appearance vessels
Nonneoplastic fibrous 0-20 Craniofacial bones, Central, lytic defect with Woven bone trabeculae in
dysplasia ribs, femur ground-glass appearance fibrous stoma
Nonossifying fibroma 0-20 Metaphyses of long Eccentric cortex-based lytic Fibrohistiocytic lesion with
tubular bones defect with sharp giant cells
(femur, tibia) scalloped margins
Aneurysmal bone cyst 10-25 Metaphyses of long Expansile lytic defect Cystic lesion with
tubular bones hemorrhage; septa
containing histiocytic and
giant cells
Synovial chondromatosis >40 Major joints of lower Punctate or stippled Island of atypical cartilage in
extremity calcifications in synovial membrane
periarticular soft tissue
Pigmented villonodular >40 Periarticular synovial Ill-defined, multinodular Histiocytic cells, giant cells,
tenosynovitis tissue and tendons periarticular mass hemosiderin deposits
Osteofibrous dysplasia 0-15 Tibia, fibula Multilocular lytic defects of Fibroosseous lesion with
anterolateral tibial cortex zonal architecture
associated with sclerosis
and tibial bowing;
synchronous fibular
lesions

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 59

TABLE 2-1 Summary of Clinicopathologic Recognition Patterns of Most Common Bone Tumor and
Tumorlike Lesions  (Continued)
Peak Age
Incidence
Type of Tumor (years) Most Frequent Sites Radiographic Features Microscopic Features
Giant cell reparative Craniofacial bones, Central lytic defect Giant cell lesion with
granuloma acral skeleton fibrohistiocytic cells and
reactive bone
Myositis ossificans >20 Parosteal or periosteal Zonal maturation, Histiocytic cells; reactive
soft tissue peripheral shell of bone bone formation maturing
toward periphery
Florid reactive periostitis 20-40 Acral skeleton Ill-defined bone surface Metaplastic cartilage;
lesion reactive bone and giant
cells
Avulsion/stress fracture 20-40 Attachment of major Ill-defined surface bone Reactive process with
tendons, long lesion; presence of metaplastic cartilage and
bones, and pelvis fracture line reactive bone
Pubic osteolysis Females Pubic bones Lytic lesion with florid Reparative process with
>50 periosteal reaction prominent metaplastic
cartilage and reactive
bone

Age (epiphysioid) bones. Periosteal chondroma has a strong


predilection for the surface of the proximal humeral
Most benign tumors and tumorlike lesions, including metaphysis. Solitary bone cysts in childhood are almost
osteoid osteoma, chondroblastoma, chondromyxoid always found in the proximal humeral shaft or upper end
fibroma, solitary bone cysts, aneurysmal bone cysts, and of the femur. Nonossifying fibromas in childhood are
nonossifying fibromas, occur in adolescents and young lesions that favor the metaphyses of long bones of the
adults (second and third decades of life). Giant cell tumor lower extremities.
is a lesion that almost exclusively occurs in skeletally Similar generalizations can be made for bone sarco-
mature patients (i.e., those with growth plates fused) mas. Chondrosarcomas have a predilection for the femur,
between ages 20 and 50 years. Of the malignant bone pelvis, ribs, and sternum and are almost never found in
tumors, osteosarcoma and Ewing’s sarcoma are tumors of the spine or short tubular bones. Osteosarcomas arise
children and young adults. On the other hand, chondro- most often in the distal femoral shaft or proximal tibial
sarcoma, malignant fibrous histiocytoma, plasma cell shaft before closure of the growth plates, and they seldom
myeloma, metastatic tumors to the skeleton, and second- penetrate the cartilaginous physis to involve the epiphy-
ary malignancies in Paget’s disease of bone, after radia- sis. Ewing’s sarcoma tends to have a diaphyseal point of
tion injury, and adjacent to bone infarcts usually develop origin, although it may also involve the metaphysis.
during the sixth through the eighth decades of life. Ewing’s sarcoma seldom affects the epiphyseal ends of
In this text the age distribution of various tumors and bones. Figure 2-1 shows the growth patterns of the most
tumorlike lesions is discussed under the separate diagnos- frequent primary malignant bone tumors, osteosarcoma,
tic headings, and the peak incidence for most is presented chondrosarcoma, and Ewing’s sarcoma.
graphically. Although exceptional cases of individual
bone tumors occur at unusual ages (e.g., chondrosarco-
mas in children or Ewing’s sarcoma in middle-aged INITIAL STAGING
patients), it is best to be circumspect with regard to these
statistical guidelines. Diagnostic Principles
Bone lesions come to clinical attention due to symptoms
Location such as swelling and local pain with or without pathologic
The fact that tumors of bone have predilections for fracture, or they can be incidentally discovered on
certain bones and even for characteristic locations in an imaging studies performed for other reasons. Bone
individual bone provides an additional clue about the lesions can be divided into benign and malignant catego-
nature of the lesion. Figure 2-1 shows the sites of predi- ries. Benign lesions occur more frequently than malig-
lection of the more common benign bone tumors. In nant tumors, and the initial diagnostic maneuvers should
some instances, the location alone strongly suggests the include obtaining a complete medical history and per-
diagnosis, as in a heavily ossified tumor on the posterior forming both physical examination and radiography.1
surface of the distal femur. That will most likely prove to The radiographic appearance of a tumor often suggests
be a parosteal osteosarcoma. Adamantinoma and osteofi- benignity or malignancy. When the radiographic analysis
brous dysplasia almost exclusively involve the tibia or is coupled with clinical information, decisions can be
fibula. Chondroblastoma is most often seen in the epiph- made concerning the need for additional imaging studies
yses of long bones in children or in tarsal or carpal and procedures. Radiologic imaging reflects the gross

ERRNVPHGLFRVRUJ
60 2  Clinical Considerations and Imaging of Bone Tumors

Fibrous dysplasia

Enchondroma

Giant cell
tumor
Nonossifying fibroma
Chondromyxoid fibroma

Chondroblastoma
OSTEOSARCOMA

A B

CHONDROSARCOMA EWING’S SARCOMA


C D
FIGURE 2-1  ■  Sites of predilection and growth pattern of the most common bone tumors. A, Diagrammatic representation of sites
of predilection in a long bone for more common benign bone tumors. B, Osteosarcoma tends to provoke little or no intraosseous
or cortical bone sclerosis and shows early breakthrough into soft tissue. Periosteal reactions tend to be linear and interrupted either
parallel or perpendicular. C, Chondrosarcoma tends to grow within the medullary cavity until very late in its course and provokes
irregular cortical thickening, sometimes with concave scalloping on the inner surface of thickened cortex. Periosteal reactions tend
to be solid and incorporated into cortex, indicating slow growth. D, Ewing’s sarcoma grows permeatively and shows breakthrough
in cortex that provokes multilayered periosteal reactions. Tumor mass shows no evidence of matrix mineralization.

anatomy and pathology of the tumor. Although the final enchondromas, osteochondromas, fibroxanthomas (non-
diagnosis of the tumor ultimately resides with the pathol- ossifying fibromas), and fibrous dysplasia. The growth of
ogist, the overall process is best performed as a multidis- most of these tumors is indolent, allowing bony remodel-
ciplinary correlation of clinical, imaging, and pathologic ing that limits the loss of structural integrity of the bone.
factors. Although some benign bone lesions can grow rapidly
Benign primary bone tumors are common but not (e.g., aneurysmal bone cyst), they rarely if ever metasta-
typically troublesome. Because these lesions are often size. One of the most common benign bone tumors that
incidentally found on imaging studies performed for can (rarely) produce distant metastases is giant cell tumor
other purposes, their true incidence is unknown. Common of bone.2,3 These lesions rarely cause death.4 Conse-
benign bone tumors include bone islands (enostoses), quently, it is hypothesized that the metastases occur due

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 61

A B

C
FIGURE 2-2  ■  Meniscal tear and incidental enchondroma. A, Lateral radiograph of a 61-year-old female with knee pain demonstrates
an intramedullary lesion of the distal femoral metaphysis with stippled and arc-and-ring matrix mineralization, characteristic of
cartilaginous tumors (arrow). No aggressive imaging features are detected, but pain may indicate chondrosarcoma. B, Sagittal fat-
saturated (FS) T2-weighted magnetic resonance image shows a round lesion that is heterogeneously high in signal intensity. C, A
complex tear of the posterior horn of the medial meniscus was responsible for the knee pain, which resolved after meniscal surgery
(arrow, postoperative image). The lesion was an incidental enchondroma that was stable after 2 years of follow-up imaging.

to the passive venous embolism of tumor cells.5,6 This is of the tumor are two of the most important demographic
in contradistinction to secondary sarcomas that develop features in diagnosing bone tumors. The typical age
at the site of treated giant cell tumors, actively metasta- range for Ewing’s sarcoma and osteosarcoma is in child-
size, and carry a poor prognosis.7-9 Benign bone tumors hood or adolescence10,11 with a second peak of osteosar-
are a rare cause of patient demise, but they can cause coma in adults.12 Chondrosarcoma, fibrosarcoma, and
morbidity through mass effect (e.g., osteochondromas malignant fibrous histiocytoma most commonly occur in
abrading adjacent tendons, muscles, or neurovascular middle age or older age groups.13-15
bundles), by predisposing to pathologic fracture due to Bone sarcomas occur in predictable locations within
cortical thinning, or by mimicking malignancy when they the bone. Primary chondrosarcomas and osteosarcomas
arise in a location that is in close proximity to an undi- typically occur in the metaphysis, whereas Ewing’s
agnosed source of pain (e.g., distal femoral enchondroma sarcoma most commonly arises in the diaphysis of long
in a patient with a meniscal tear of the knee in Figure bones. Fibrosarcoma and malignant fibrous histiocytoma
2-2). In the case of an enchondroma, the lack of aggres- can occur in many sites but are preferentially found in
sive imaging features such as osteolysis, periosteal reac- the metaphysis or metaphyseal/diaphyseal junction. The
tion, or cortical breakthrough may aid the pathologist in dominant anatomic sites of many of these tumors have
rendering a benign interpretation of the sample, particu- been described by Johnson et al.16,17 The locations in
larly when advanced imaging demonstrates an alternative which these bone tumors arise tend to be areas of current
source of pain. Pathologists and radiologists must be or previous rapid skeletal growth (Fig. 2-3), and tumor
familiar with the strengths and weaknesses of both spe- development may be related to the high rate of cell divi-
cialties for both disciplines to augment each other and sion. The distal femur and proximal tibia are most com-
increase the likelihood of correct patient diagnosis and monly involved. However, bone sarcomas may arise in
treatment. other areas and any bone may be involved. Most benign
Malignant primary bone tumors are not common but bone tumors also arise in or near the metaphysis of
are of critical importance due to their high likelihood of long bones. Some lesions, such as osteochondromas or
metastasizing and causing death in the absence of proper nonossifying fibromas/fibroxanthomas, may arise in the
treatment. The five most common primary bone sarco- metaphysis and “migrate” away with skeletal growth.
mas account for the majority of these lesions and include Due to lack of symptoms they may be incidentally dis-
osteosarcoma, chondrosarcoma, Ewing’s sarcoma, fibro- covered in the diaphysis of older patients.
sarcoma, and malignant fibrous histiocytoma. Neverthe- Biopsy is an integral part of the diagnostic process and
less, bone metastasis and multiple myeloma are much is indicated in tumors that are suspicious for malignancy.
more common. The age of the patient and the location Percutaneous core biopsy is less invasive, less costly, and

ERRNVPHGLFRVRUJ
62 2  Clinical Considerations and Imaging of Bone Tumors

A C
FIGURE 2-3  ■  Cellular activity of the epiphyseal growth plate. A, Frontal radiograph of the distal femur in 9-year-old male with
osteosarcoma that involves the epiphysis, growth plate, and metadiaphysis with osteoid mineralization in all three sites. Tumor
breakout is shown laterally with moth-eaten osteolysis, layered Codman’s triangle, and spiculated periosteal reaction. B, The high
degree of normal cellular activity in the epiphyseal growth plate of skeletally immature individuals may predispose to later tumor
development at the ends of the long bones, such as in the metaphyses. C, Higher power photomicrograph demonstrating that active
enchondral bone formation is most prominent near the metaphysis and that tumor extends into the growth plate. (B, ×10; C, ×25)
(B and C, hematoxylin-eosin.)

typically more convenient for patients than open surgical responsible for forming the image by exposing/darkening
biopsy. Contrast-enhanced MRI is commonly used to the film or other receiving device.24 Dense anatomic
guide selection of the biopsy site. Enhancing portions of structures such as bone impede the course of the x-rays,
the tumor are viable and preferable for sampling, as leaving the corresponding areas unexposed. Bone is
opposed to nonenhancing areas, which are likely cystic therefore white in appearance on conventional radio-
or necrotic.18,19 Because the biopsy track must be excised graphs. Unlike CT, which provides anatomic cross-
for complete resection of malignancies, the biopsy must sections (see the discussion of CT below), radiographs
be performed in such a way as to minimize contamination are a “summation” of the density of all points in the plane
of the anatomic compartments, neurovascular structures, (frontal, lateral, or oblique). This quality portrays the
or tissues that may be necessary for surgical reconstruc- tumors in a unique fashion that is often essential for
tion.20 This advanced surgical planning can be greatly properly assessing the appearance of key diagnostic attri-
facilitated by the excellent soft tissue contrast resolution butes of the tumors such as margins, periosteal reactions,
provided by MRI.21,22 All intervention must be planned and matrix mineralization. Radiography is recommended
in light of the surgical options. Therefore, the diagnosis for all bone tumors because it provides well-established,
of bone tumors cannot be separated from treatment. A reliable information on which to build further analyses,
multidisciplinary approach is critical in evaluation and such as the cross-sectional and multiplanar imaging
treatment planning for bone sarcomas. studies discussed below.
CT is a useful adjunct to radiography for staging
Imaging Modalities primary bone tumors. Unlike radiography, in which a
single x-ray source strikes a film or detector, most CT
Radiography is essential for the initial staging of primary scanners utilize multiple x-ray sources that strike multiple
bone tumors and is the least expensive and most readily detectors positioned opposite their respective sources on
available of the imaging modalities.23 Radiographic a circular frame. This hardware is located in a circular
images are produced by bombarding anatomic structures gantry through which the patient table moves during the
with x-rays. The x-rays that pass through the body are scan. The majority of CT scanners utilize this design to

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 63

either rotate source/detector pairs around the patient or of soft tissue masses.21,22 MRI can allow unparalleled pre-
to rotate an x-ray source within a ring of stationary detec- surgical evaluation of the relationship of the tumor to
tors embedded in the gantry. These source/detector com- vital structures such as nerves and blood vessels. Most
binations allow the imaging of “slices” of anatomy per untreated bone tumors demonstrate T1 signal that is
revolution. Most modern scanners are operated in helical similar (isointense) to muscle, as well as heterogeneously
mode, allowing rapid scanning while the patient table high T2 signal, and enhance on administration of a
moves continuously through the gantry.25 CT scanning gadolinium-based intravenous contrast medium. The
eliminates the effect of overlapping structures and can entire length of the bone can be imaged in detail; MRI
provide sharp delineation of the anatomy. CT is the most is also considered the imaging method of choice for eval-
efficacious imaging modality for evaluating mineralized uating for skip metastases.37 Heavily mineralized struc-
structures, such as tumor matrix.21,26 For example, CT tures such as cortex and matrix mineralization do not
can be used to detect small areas of mineralized matrix typically contain a sufficient number of free protons to
that are not evident on radiography. Nevertheless, the produce a magnetic resonance signal. Therefore, bone
indistinct quality of aggressive bone tumor margins can cortex appears as a black signal void on MRI (Fig. 2-4),
be underestimated with CT; it is not recommended that and fine cortical detail is not well discerned. MRI is
CT be substituted for radiography during the initial therefore best used in combination with radiography or
evaluation of bone tumors. CT with which cortical detail and other mineralized fea-
Skeletal scintigraphy, commonly known as bone scan, is tures can be more fully evaluated. The combined strengths
a whole-body method of surveying the entire skeleton in of each imaging modality permit excellent evaluation of
a single imaging session and is often used to detect metas- the many and varied features of bone tumors.
tases to bone. Bone scan typically plays a limited role in
the initial staging of bone tumors unless the disease is Imaging Characterization of Primary
metastatic to bone or multifocal at presentation. Uptake
of technetium-based tracers such as technium-99m meth-
Bone Tumors
ylene diphosphonate (MDP) is dependent on a combina- The initial diagnosis of bone tumors on imaging studies
tion of factors such as blood flow and the binding of the begins by dividing lesions into “aggressive” or “nonag-
diphosphonate molecules to hydroxyapatite found in gressive” categories on the basis of characteristics such as
areas of new bone formation.27,28 Diphosphonate uptake lesion margin, expansion of the bone, periosteal reaction,
is nonspecific and can be seen with benign and malignant and soft tissue extension. Aggressive lesions often cor-
primary bone tumors, as well as with metastases. For respond to malignancy with a small number of notable
example, uptake in benign cartilaginous lesions, such as exceptions, such as giant cell tumors of bone, that are
enchondromas, can be problematic because similar uptake often aggressively destructive but rarely result in patient
can also found in chondrosarcomas.29 Enchondromas demise. Conversely, nonaggressive lesions are typically
may also be difficult to differentiate from low-grade benign with the exception of indolent malignancies such
chondrosarcomas on radiographs, other imaging modali- as low-grade chondrosarcomas. Further refinements to
ties,30 and even on histopathologic analysis.31 Although the differential diagnosis are made through features such
radiography, CT, and MRI can each be limited regarding as the presence or absence of mineralized matrix and the
the diagnosis of enchondromas versus low-grade cartilage appearance of the periosteal reaction.
tumors, the combined information provided by multiple The most important factor in determining the aggres-
imaging studies can be valuable in management decisions sive or nonaggressive nature of a lesion is the appearance
and can often aid the diagnostic process. of its margin, which is a product of the osteoclastic activ-
Bone scan can be used for initial staging for skip ity of the host bone in response to the tumor and of
metastases. Skip metastases occur in the same bone as the reparative, osteoblastic activity. The radiographic appear-
primary malignancy but in a noncontiguous location or ance of the margin is an indicator of the growth rate of
across the adjacent joint32 and are associated most com- the lesion.38,39 Three main types of lesion margins have
monly with osteosarcoma33 and Ewing’s sarcoma.34 The been identified on radiographs. Greater tumor aggres-
ability to survey large anatomic regions makes bone scan sion is found with progression up the scale (Fig. 2-5).17
a useful imaging modality for this purpose. Type I margins are geographic (round or ovoid) and have
MRI is capable of producing greater soft tissue resolu- been divided into three subcategories. Type IA margins
tion than other imaging modalities and is commonly used are well defined and have a narrow zone of transition,
to assess the extent of disease prior to the surgical treat- which is defined by the presence of normal cortical bone
ment of bone tumors. MRI scanners manipulate free in close proximity to the lesion. A sclerotic rim is present,
protons. Protons precess, or spin, randomly. When indicative of a successful reparative response by the host
placed in a strong magnetic field, the majority of protons bone. Type IA margins are commonly seen with tumors
align with the field. Radiofrequency pulses or magnetic such as fibrous dysplasia, chondroblastoma, enchon-
gradients are used to reorient proton spins.35 As the spins droma, and fibroxanthoma/nonossifying fibroma (Fig.
return to normal, radio waves are released and interact 2-6). Type IB margins are well defined but have no scle-
with a receiver coil. An electric current is generated, from rotic rim and are therefore of indeterminate biologic
which computer-generated images are derived.36 potential (Fig. 2-7). The tumor may be aggressive and
High soft tissue resolution makes MRI the optimal growing only slightly faster than the bone’s ability to
imaging modality for evaluating the extent of tumors produce a reparative response. Alternatively, the tumor
inside the medullary cavity of bone as well as the extent may produce factors that inhibit osteoblastic activity,

ERRNVPHGLFRVRUJ
64 2  Clinical Considerations and Imaging of Bone Tumors

A B
FIGURE 2-4  ■  Magnetic resonance image of a skip metastasis. A, Lateral radiograph of the ankle in a 10-year-old male with primary
osteosarcoma of the calcaneus. Dense osteoid formation gives the calcaneus the appearance of an “ivory” bone. B, Coronal fat-
saturated T1-weighted magnetic resonance image of the ankle reveals an enhancing soft tissue mass extending from the primary
tumor. The tumor has not broken through the inferior calcaneal cortex, and the intact cortex is represented as a black signal void
(arrow). A skip metastasis to the distal fibular diaphysis is also demonstrated. The skip metastasis (arrowheads on A and B) is dif-
ficult to see on radiography but is well visualized on MRI, which is a highly sensitive method for evaluating local extent of disease.

IA Sclerotic IB Non-Sclerotic IC Ill-Defined

ll-Moth-Eaten lIl Permeative


FIGURE 2-5  ■  Classification of bone tumor margins.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 65

A B
FIGURE 2-6  ■  Type IA margin (geographic, sclerotic rim). A, Frontal radiograph of the proximal lower leg in a 16-year-old female
with an incidental nonossifying fibroma (fibroxanthoma). B, A sclerotic rim encircles the nonaggressive lesion on coronal computed
tomography reformation.

such as in myelomatous lesions.40 Type IB margins are Mild expansion is commonly seen with benign fibrous
commonly seen with tumors such as aneurysmal bone lesions such as fibrous dysplasia, fibrous cortical defects,
cyst, giant cell tumor, chondromyxoid fibroma, low- or enchondromas (Fig. 2-13). Greater degrees of cortical
grade chondrosarcoma, myeloma, and early metastasis. expansion can be seen with other benign lesions such as
Type IC margins are typically seen with aggressive bone nonossifying fibromas/fibroxanthomas and desmoplastic
tumors. While retaining a geographic round or ovoid fibromas. Gross expansion of the cortex with thinning
shape, the margin is ill-defined and indistinct, demon- beyond radiographic visibility can be seen with benign
strating a wide zone of transition with no sharp margin- tumors such as aneurysmal bone cyst, giant cell tumor of
ation (Fig. 2-8). Type IC margins are commonly seen bone, and chondromxyoid fibroma (Fig. 2-14). Malignant
with lesions such as chondrosarcoma, osteosarcoma, lesions will often precipitously destroy cortex and are
fibrosarcoma/MFH, metastasis/myeloma, and giant cell more likely to extend directly into the soft tissues than to
tumors (Fig. 2-9). expand the bone. The cortex surrounding a large, malig-
Type II and III margins are frankly aggressive, repre- nant soft tissue mass may appear expanded but is typically
sent disorganized areas of osteolysis, and are typical of noncontinuous and fragmented, with numerous areas of
high-grade sarcomas such as osteosarcoma and Ewing’s frank cortical destruction. A notable exception to the
sarcoma. Type II is described as moth-eaten; either the benign nature of continuous cortical expansion can be
periphery or the entire lesion is composed of lucent areas seen in low-grade chondrosarcomas. These lesions are
of varying size and shape, representing nonuniform oste- indolent and slowly expand the circumference of the
olysis. MRI will typically demonstrate complete or near- bone, producing a generally thickened cortex with vari-
complete invasion of the marrow cavity (see Figs. 2-10 able areas of thinning (Fig. 2-15). Nevertheless, the
and 2-11). The type III margin corresponds to the most majority of tumors that continuously expand cortex are
highly aggressive bone tumors and is described as perme- benign.
ative. This radiographic appearance is primarily caused Extension of tumor into the soft tissues is typically an
by tumor-stimulated osteoclastic cutting cones that aggressive feature that is most commonly seen with
tunnel rapidly through the bone, producing lesions with malignancy. Primary bone malignancies are often distin-
wide zones of transition and a more uniform pattern of guished from metastatic lesions by the presence of a large
cortical lysis than does the moth-eaten margin (Fig. soft tissue mass extending from a solitary bone tumor,
2-12). The margin classification system is most appropri- and they most commonly demonstrate a type IC, II, or
ately applied to radiographs because CT can produce a III margin. On tumor extension into the soft tissues, MRI
“smoothing” effect that can underestimate the aggres- can be used to evaluate for abutment or encasement of
siveness of the margin of geographic lesions, and MRI adjacent structures such as other bones, nerves, blood
does not directly image calcified structures. vessels, and visceral organs, as well as extension across
Continuous cortical expansion is a feature that is more fascial planes (Fig. 2-16).21,22,26
commonly seen with benign than with malignant tumors. Text continued on p. 75

ERRNVPHGLFRVRUJ
66 2  Clinical Considerations and Imaging of Bone Tumors

A B

C
FIGURE 2-7  ■  Type IA/IB margin (geographic, well-defined). Enchondroma of the fourth metacarpal head in a 22-year-old female.
A, Mild expansion of the circumference of the bone is demonstrated with minimal sclerotic rim. B, Photomicrograph of the lesion
with close-up (C) shows a thin rim of continuous sclerotic bone around the distal margin with thicker (arrows) and thinner (arrow-
head) areas, some of which may not be radiographically apparent. The cortex is thinned but expanded and intact, reflecting endosteal
erosion that is balanced with periosteal reaction. Incidentally noted are small foci of enchondral bone formation within the enchon-
droma. (B, C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 67

FIGURE 2-8  ■  Type IB and IC margins. Frontal radiograph of the left proximal femur in a 53-year-old male with multiple myeloma
demonstrates a lytic lesion involving the proximal femoral neck, the greater trochanter, and intertrochanteric region. The distal
aspect of the lesion is sharply marginated and well-defined but no sclerotic rim is present (IB margin, arrow). The proximal margin
is ill-defined, though the lesion retains an oval, geographic shape (IC margin, arrowheads). The malignant lesion was internally fixed
due to the high risk of pathologic fracture.

ERRNVPHGLFRVRUJ
68 2  Clinical Considerations and Imaging of Bone Tumors

B
FIGURE 2-9  ■  Type IC margin (geographic, ill-defined). A, Photomicrograph of the proximal margin of a tibial malignant fibrous
histiocytoma in a 50-year-old female. The aggressive lesion demonstrates an infiltrating rim with tumor encasing the partially
destroyed trabeculae with a central area of necrosis and cyst formation. B, Specimen radiograph shows infiltrating bony destruction
with transition from complete trabecular osteolysis on the inner edges through a progressive partial trabecular osteolytic zone to
normal trabecular bone at the lesion margin. These zones blend radiographically to form the perception of an ill-defined border.
(A, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 69

A B
FIGURE 2-10  ■  Type II margin. A, Frontal radiograph of the lower leg in a 53-year-old male with osteosarcoma of the proximal tibial
diaphysis. Areas of osteolysis vary in size and shape, indicating a moth-eaten, or type II margin. The aggressive tumor has under-
mined the structural integrity of the bone and an incomplete pathologic fracture has occurred in the medial tibial cortex (arrow).
B, Cortical erosions that correspond to the moth-eaten radiographic pattern are seen in greater detail on coronal computed tomog-
raphy reformation. Inset, Axial fat-saturated T1-weighted magnetic resonance imaging after the administration of intravenous
contrast shows a centrally necrotic medullary tumor, with varying depth of cortical invasion (arrowheads).

ERRNVPHGLFRVRUJ
70 2  Clinical Considerations and Imaging of Bone Tumors

FIGURE 2-11  ■  Type II margin (moth-eaten). A, Specimen radiograph of a calcaneal fibrosarcoma. Patchy foci of moth-eaten osteolysis
are present with sparing of areas of normal bone. B, Macroscopic photomicrograph demonstrates bone invasion with areas of
marrow infiltrated by small islands of tumor corresponding to the moth-eaten osteolytic foci in the radiograph. A central area of
coalescent osteolysis corresponds to a more cohesive focus of tumor and uniform radiographic lucency. (B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 71

A B
FIGURE 2-12  ■  Type III margin (permeative). A and B, Frontal and lateral radiographs of the knee in a 63-year-old female with a
chondrosarcoma of the distal femoral metaphysis. The proximal margin is permeative (arrows), the most aggressive margin type.
Marked osteolysis with soft tissue extension is seen at the level of the metaphysis, resulting in abnormal angulation of the distal
femur.

ERRNVPHGLFRVRUJ
72 2  Clinical Considerations and Imaging of Bone Tumors

A B

FIGURE 2-13  ■  Cortical expansion in a nonaggressive benign lesion. A, Well-corticated expansion of the proximal fibular metadiaphy-
sis with areas of focal calcification are demonstrated on a frontal radiograph of the lower leg in a 44-year-old male with an intraos-
seous lipoma. B, The expanded cortex is contiguous and intact on the macroscopic photomicrograph. Inset, Background fatty tissue
(lipoma) on higher power photomicrograph with central ossification and ischemic bone correspond to the mineralization seen on
the radiograph. (B and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 73

FIGURE 2-14  ■  Cortical expansion in an aggressive benign lesion. Frontal radiograph of the pelvis in a 19-year-old female with a giant
cell tumor of bone. A lytic, expansile lesion is centered at the level of the acetabulum. The expanded cortex is well demonstrated
laterally. The cortex is thinned nearly beyond radiographic detection superiorly and is less severely thinned inferiorly (arrows). A
type IB margin is seen proximally (arrowhead). Giant cell tumors can demonstrate an unusual combination of locally aggressive
yet overall benign behavior.

ERRNVPHGLFRVRUJ
74 2  Clinical Considerations and Imaging of Bone Tumors

FIGURE 2-15  ■  Low-grade chondrosarcoma. Frontal radiograph of the proximal humerus in a 68-year-old male with a lytic lesion that
has mildly expanded the bone and produced variable cortical thickening and thinning. The thickened cortex is the bone’s attempt
to contain the slowly and relentlessly growing tumor. The lesion is a secondary chondrosarcoma that has arisen from an enchon-
droma. The cartilaginous matrix mineralization of the enchondroma can be seen in the nonexpansile, proximal portion of the tumor
(arrow). No sclerotic margin is seen.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 75

FIGURE 2-16  ■  MRI of local disease extent: neural involvement. Axial fat-saturated T1-weighted magnetic resonance image of the
pelvis in a 21-year-old male with mesenchymal chondrosarcoma of the sacrum demonstrates a large tumor arising from the sacrum.
The greatest bulk of the lesion extends anteriorly into the pelvis. A smaller component invades the sacral spinal canal and encases
the left S2 nerve root (arrow). Also observe that the enhancement of the tumor is more heterogeneous anteriorly, corresponding
to a greater degree of central necrosis in the larger portion of the tumor (arrowheads).

Periosteal reaction is another indicator of the aggres- seen with osteoid, cartilaginous, or fibrous tumors. The
sive or nonaggressive nature of tumors.41 During devel- mineralization that occasionally occurs in myxoid matrix
opment, the circumference of the bone enlarges through is indistinguishable from dystrophic calcification, is non-
the mechanism of periosteal bone formation; this mecha- specific, and is not helpful for the diagnosis of bone
nism plays a major role in the healing of fractures that tumors on imaging studies.
experience small degrees of motion at the fracture site.42,43 The presence or absence of mineralized matrix is not
The periosteum is more active in children than adults, an indicator of benign or malignant behavior. Osteoid
and the appearance of adult periosteal reactions typically matrix can be seen in bone islands/endosteomas, osteoid
parallels that of those reactions in children but are lesser osteomas, osteoblastomas, or osteosarcomas and is most
in magnitude. Rapidly aggressive tumors often result in commonly described as “fluffy” or “cloudlike” on radio-
multilaminar or interrupted periosteal reactions. The graphs and CT although dense, ivorylike osteoid can also
mass effect of the tumor can lift the periosteum from the be produced (Fig. 2-21). Cartilaginous matrix is the result
cortex, producing a triangular interface that is termed a of enchondral bone formation and is often “stippled” or
Codman’s triangle (Fig. 2-17). The periosteal reaction of demonstrates a curvilinear “arc-and-ring” quality that
Ewing’s sarcoma is commonly parallel to the long axis of conforms to the rounded contour of cartilage lobules
the bone and resembles an onion skin (Fig. 2-18), though (Fig. 2-22). The lobular growth of cartilaginous tumors
osteosarcoma can produce a similar appearance (Fig. is often reflected in a lobulated pattern of endosteal scal-
2-19). The periosteal reaction of osteosarcomas often loping on radiographs and a septal enhancement pattern
extends perpendicularly from the long axis of the bone, on MRI (Fig. 2-23) that is common but not exclusive to
giving a spiculated, “hair-on-end” or “sunburst” appear- cartilaginous tumors. Distinguishing between benign and
ance (Fig. 2-20). Indolent processes often produce a malignant cartilaginous tumors by means of imaging or
thicker, more solid, or unilaminar appearance. Knowl- histopathology can be difficult. Not all chondrosarcomas
edge of the medical history is important because the demonstrate frankly aggressive imaging findings such as
periosteal reactions of treated malignant primary bone a type II or III margin. A comparative imaging study of
tumors or metastases can thicken or solidify with success- 92 enchondromas and 95 chondrosarcomas45 found that
ful therapy and can mimic a benign feature. chondrosarcomas were typically larger than the enchon-
Matrix is an acellular substance located in the extracel- dromas (>5-6 cm in length), produced deeper endosteal
lular space between tumor cells; the presence of mineral- scalloping (scalloping of at least two-thirds the thickness
ized matrix can aid in the identification of bone tumors.44 of the cortex over at least two-thirds the length of the
Characteristic patterns of matrix mineralization can be Text continued on p. 82

ERRNVPHGLFRVRUJ
76 2  Clinical Considerations and Imaging of Bone Tumors

A B

FIGURE 2-17  ■  Codman’s triangle. A, Radiograph with aggressive, multilaminar periosteal reaction caused by a fibrosarcoma lifting
the periosteum from the anterior tibial diaphysis in a 56-year-old female. B and Inset, Photomicrograph and close-up of the tumor
show the elevated periosteum, its osteoid product beneath it, and the extracortical but still subperiosteal tumor arising from deep
within the femur (arrow). The periosteum is intact, but detectable mineralization is only seen at the peripherial edge where it has
formed and mineralized bone. (B, Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 77

FIGURE 2-18  ■  “Onionskin” periosteal reaction: Ewing’s sarcoma. Frontal radiograph of a 12-year-old male with Ewing’s sarcoma of
the humerus. An aggressive, multilaminar periosteal reaction (arrow) is associated with the aggressive tumor, which produces
moth-eaten and permeative osteolysis of the distal metaphysis of the elbow.

ERRNVPHGLFRVRUJ
78 2  Clinical Considerations and Imaging of Bone Tumors

A B
FIGURE 2-19  ■  “Onionskin” periosteal reaction: osteosarcoma. A, Multilaminar, “onionskin” periosteal reaction is seen on a specimen
radiograph of the proximal humeral diaphysis (arrow) of a 20-year-old male with osteosarcoma. A combination of moth-eaten
osteolysis and mineralized osteoid is seen within the humerus, while foci of mineralized tumor osteoid are seen within the layered
periosteal reaction. B, Macroscopic photomicrograph closely parallels the radiographic image, showing both mineralized tumor
osteoid and nonmineralized tumor and osteoid in the osteolytic areas. These same features are demonstrated in the periosteum
and the layered periosteal reaction. (B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 79

A B
FIGURE 2-20  ■  “Hair-on-end” periosteal reaction: osteosarcoma. A, The periosteal reaction extends perpendicularly from the cortex
on a specimen radiograph of a 14-year-old male with osteosarcoma of the distal femoral metaphysis. The distal growth plate is
involved, with extension of mineralized tumor into the epiphysis. The proximal periosteal reaction is more contiguous and mineral-
ized, corresponding to less biologic growth at this site. B, Macroscopic photomicrograph demonstrates inhomogeneous but diffuse
mineralized tumor osteoid throughout the metadiaphysis and extending through the growth plate into the epiphysis. Foci of unmin-
eralized osteoid in the intraosseous and extraosseous tumor contribute to the inhomogeneous appearance. The hair-on-end min-
eralization shows varying lengths and widths of spicule formation. (B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
80 2  Clinical Considerations and Imaging of Bone Tumors

FIGURE 2-21  ■  Osteoid matrix. Frontal radiograph of the shoulder in a 15-year-old male with osteosarcoma. The tumor produces
aggressive bone destruction and a large soft tissue mass with osteoid matrix that is “fluffy” or “cloudlike” in appearance (arrow).

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 81

A B
FIGURE 2-22  ■  Chondroid matrix. A, Radiograph of the scapula in a 22-year-old male with chondrosarcoma of the scapular body.
Cartilaginous matrix mineralization is characteristically curvilinear with an “arc-and-ring,” or stippled appearance. Also note the
stalk at the base of the tumor, indicating that it is a secondary chondrosarcoma that has arisen from an osteochondroma. B, These
characteristics are seen in better detail on the axial computed tomography image of the tumor.

ERRNVPHGLFRVRUJ
82 2  Clinical Considerations and Imaging of Bone Tumors

A B

C
FIGURE 2-23  ■  Septal enhancement of chondrosarcoma. A, Axial T1-weighted, B, fat-saturated T2-weighted, and C, fat-saturated
T1-weighted postcontrast MRI of the same chondrosarcoma as in FIGURE 2-22. A, T1 signal is lower than muscle. B, T2 signal is
predominately high and the lesion is lobulated. C, Enhancement is seen predominately at the periphery of the lesion in the “septae”
extending about the cartilaginous lobules. The matrix does not enhance. This unusual pattern of tumor enhancement is commonly
seen in cartilage tumors and can be contrasted with the more solidly enhancing tumor in FIGURE 2-16.

lesion), and were more likely to be painful. Other studies posttherapeutic appearance can mimic chondroid matrix
have found malignant cartilaginous tumors to be more (Fig. 2-26). These potentially confusing changes in the
intensely FDG avid than benign cartilage lesions on PET imaging appearance of treated tumors serve to emphasize
imaging,46,47 implying that malignant tumors are more the importance of the medical history in the proper inter-
metabolically active than benign ones. FDG PET is pretation of diagnostic images. Multiple lesion character-
further described below in the section on response to istics identified on imaging studies can combine to aid
therapy. the appropriate diagnosis of bone tumors.
Matrix mineralization can exhibit a spectrum of
appearances. Fibrous dysplasia may be lucent or may
demonstrate ground-glass or dense mineralization
depending on the amount of woven bone formed by the
RESPONSE TO THERAPY
lesion (Fig. 2-24). An increase in lesion mineralization Primary Bone Tumors
can occur over time in nonossifying fibromas, fibrous
dysplasia, osteochondromas, Langerhans cell histiocyto- The treatment for bone tumors is closely related to their
sis, bone islands, and osteoid osteomas (Fig. 2-25).48 The likelihood of recurrence. Benign, symptomatic bone
cartilaginous matrix of benign tumors can also increase tumors with a high likelihood of local recurrence (giant
in density over time, in a process termed maturation. Suc- cell tumor, aneurysmal bone cyst) are treated with curet-
cessful treatment of malignant tumors will often result in tage, which involves tumor removal while leaving the
increased tumor mineralization. The “fluffy” or “cloud- surrounding bone intact. A high-speed burr is used on all
like” matrix that is found in untreated osteosarcoma surfaces of the curetted cavity. Numerous adjunctive
coarsens and becomes more solid after treatment, and the techniques can be used to destroy remaining tumor cells,

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 83

A B
FIGURE 2-24  ■  Variation in matrix mineralization. Frontal radiographs of the proximal femora in two patients with fibrous dysplasia.
A, The mildly expansile lesion demonstrates ground-glass matrix that is similar in density to the normal trabeculae. Two cannulated
screws span a healed, pathologic fracture of the femoral neck. B, The lesion in the second patient demonstrates denser matrix with
more maturation (ossification) and sclerosis about the periphery. This lesion also weakened the structure of the bone, resulting in
an abnormal curvature termed the shepherd’s crook deformity.

such as the application of liquid nitrogen, hydrogen per- become more densely mineralized on radiographs53 and
oxide, phenol,49 or an argon laser.50 The cavity is then CT54 with successful systemic or radiation therapy. The
packed with autologous bone graft, cadaveric bone graft, MRI appearance of a positive therapeutic response
or polymethylmethacrylate cement. typically results in decreased enhancement following
Benign bone tumors that do not typically recur can be gadolinium-based intravenous contrast administration
treated with lesion destruction rather than removal. and a decrease in T2 signal intensity. T1 signal may also
Osteoid osteoma can be treated through radiofrequency diminish. These changes correspond to increased miner-
ablation, which is a percutaneous technique that utilizes alization, diminished vascular perfusion, and decreased
heat necrosis to destroy the painful tumor nidus in a lesional water content. Central necrosis is identified as
minimally invasive fashion.51,52 Because systemic therapy peripheral enhancement in the absence of central
is typically not available or necessary for benign bone enhancement. Central necrosis in a pretherapeutic or
tumors, imaging plays no role in evaluating response to recurrent lesion is an aggressive feature, implying that
therapy. Posttherapeutic imaging of benign lesions is per- the tumor has outgrown its blood supply (Fig. 2-27). The
formed for the purpose of detecting local recurrence or posttherapeutic development of central necrosis, or
complications such as fractures or infections. This increase in central necrosis, typically indicates a positive
imaging strategy also applies to bone malignancies for response, reflecting therapy-induced cell death. In the
which no standard extrasurgical therapy has been devel- past, bone scan played an important role in evaluating
oped, such as chondrosarcoma. therapeutic response in bone tumors. Primary bone
Systemic therapy is available for some malignant tumors have been shown to increase or decrease in tracer
primary bone tumors, such as osteosarcoma and Ewing’s uptake on bone scan with negative or positive response,
sarcoma, and the resultant changes that are observed on respectively. Nevertheless, bone scan has largely been
imaging studies have been correlated with either a posi- supplanted by MRI and CT for primary bone tumor
tive response or progression of disease. Many tumors, staging. Skeletal scintigraphy continues to play a large
such as osteosarcomas and plasmacytomas, typically role in staging for skeletal metastases.

ERRNVPHGLFRVRUJ
84 2  Clinical Considerations and Imaging of Bone Tumors

A B
FIGURE 2-25  ■  Healing nonossifying fibroma (fibroxanthoma). Frontal radiographs in an 8-year-old female with a nonossifying
fibroma of the distal femur. A, The lesion demonstrates a combination of internal lucency and mature mineralization with a sclerotic
type IA, nonaggressive margin. B, One year later, the lucent areas have partially mineralized, indicating interval maturation and
ossification within the lesion. The lesion appears to have “migrated” proximally with growth of the bone distally.

A B
FIGURE 2-26  ■  Posttherapeutic osteoid mimicking chondroid matrix. Axial computed tomography images of the humerus in a
15-year-old male with osteosarcoma. A, “Fluffy” osteoid matrix is evident in the large soft tissue mass extending from the humeral
diaphysis (arrow). B, After chemotherapy, the osteoid has coarsened, simulating the stipples and arcs and rings of chondroid matrix
in some portions of the tumor (arrowheads). The lesion has also decreased in size, corresponding to a positive response to therapy.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 85

A B
FIGURE 2-27  ■  Central necrosis in progressing, recurrent disease. A, Axial fat-saturated T1-weighted magnetic resonance image of
a 19-year-old female with recurrent spinal Ewing’s sarcoma following intravenous contrast demonstrates a solidly enhancing recur-
rent tumor mass. B, Two months later, the lesion enhances less uniformly but has enlarged (arrowheads), indicating central necrosis
in a progressing lesion that now also involves the adjacent right L4-5 foramen (arrow).

Tumor response criteria have been developed to scanning can provide an alternative method of measuring
provide a standardized, uniform methodology for deter- tumor response. Currently, lack of standardization in
mining response in patients undergoing therapy in dif- PET scan acquisition in different institutions has limited
ferent clinical trials and at different institutions. Response its inclusion into major response criteria.
criteria define the minimal allowable tumor change that The soft tissue component of Ewing’s sarcoma and
constitutes a response and delineate the methods by plasmacytomas predictably undergoes a marked decrease
which change can be measured. The most commonly in size with successful therapy. Nevertheless, some soft
used response criteria, exemplified by the Response Eval- tissue sarcomas, such as gastrointestinal stromal tumors,
uation Criteria in Solid Tumors (RECIST), rely primar- have a tendency to increase in size with successful therapy
ily on anatomic change in tumor size.55 Osteosarcoma due to loss of structural integrity.67 This is a rare phe-
typically demonstrates an initial decrease in size that cor- nomenon in bone tumors, in which an increase in size on
relates with a positive outcome, whereas an increase in any imaging modality after therapy is usually a sign of
size during therapy correlates with a poor prognosis.56 disease progression. The determination between positive
Nevertheless, the tumor may not continue to diminish in or negative response can be difficult when the lesion
size or may diminish only slightly (not meeting minimal increases in central necrosis because this could either be
change in size for a “measurable” response as per the caused by outgrowth of blood supply in a progressing
criteria), despite continued positive histologic response. tumor or desired cell death after effective therapy. Typi-
Lack of size correlation with successful chemotherapy is cally, the progressing, centrally necrotic lesion will dem-
a potential pitfall in determining treatment response in onstrate stable or increasing soft tissue nodularity along
primary bone tumors on conventional, anatomic imaging. the enhancing rim. Rim nodularity in a responding lesion
FDG PET/CT is a form of functional imaging that will often diminish.
demonstrates cellular uptake of the glucose analog FDG. Imaging has become an essential tool for evaluating
The FDG molecule is transported into cells using GLUT therapeutic response in primary bone tumors. Therapy
transporters located in cell membranes.57,58 FDG is a can be modified based on imaging results, potentially
phosphorylated molecule that cannot be metabolized by allowing the patient to be changed from ineffective to
the cell and is broken down slowly. The majority of effective therapeutic agents before surgical resection and
malignancies demonstrate a high rate of glucose metabo- histopathologic analysis of the specimen.
lism. Therefore, the resultant intracellular accumulation
of FDG in tumor cells permits functional, metabolic
imaging. As a reflection of tumor metabolism that dimin-
Bone Metastases
ishes with cell death, FDG has been shown to effectively Bone sarcomas metastasize most commonly to the lungs
anticipate therapeutic response in primary bone sarcomas but demonstrate a predilection for metastasizing to other
such as osteosarcoma,59-64 Ewing’s sarcoma,65 and chon- bones.68,69 Muliple foci of osteosarcoma may be found
drosarcoma.66 The change in FDG uptake, when mea- only in the skeleton, raising the possibility of multicentric
sured before and after chemotherapy, may also have primary disease in the absence of a dominant lesion.70,71
prognostic value, as has been shown in studies of osteo- Bone metastases can be osteolytic and lucent, osteoblastic
sarcoma (Fig. 2-28).59,61,63,64 Change in FDG uptake can and sclerotic, or they can be mixed lytic and sclerotic
occur independently of change in tumor size; FDG PET (Fig. 2-29). Many parallels can be seen in the imaging

ERRNVPHGLFRVRUJ
86 2  Clinical Considerations and Imaging of Bone Tumors

A C

B D
FIGURE 2-28  ■  Response to therapy on FDG PET/CT. A, Axial computed tomography (CT) portion of a positron emission tomography
(PET)/CT scan in a 17-year-old male demonstrates an osteoid-containing mass extending from the left iliac bone that was biopsy-
proven osteosarcoma. B, The maximum standardized uptake value (SUVmax) of the lesion on the fused PET/CT image was 8.5 before
therapy. C, After chemotherapy, the lesion increased in mineralization but only slightly decreased in size. D, The posttherapeutic
PET/CT image shows a marked decrease in fluorodeoxyglucose uptake, with an SUVmax of 2.5. The functional imaging reflected the
histologic response, which included 99% necrosis in the resected specimen.

A B
FIGURE 2-29  ■  Types of metastases. A, Axial computed tomography (CT) scan of the L2 vertebra in a 53-year-old female with breast
cancer. The blastic/sclerotic metastasis is dense on CT (arrow) and radiography (not shown). B, The lytic metastasis to the T9 ver-
tebra is lucent (arrowheads). Additional metastases on both images are mixed.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 87

A C E

B D F
FIGURE 2-30  ■  Flare phenomenon. A and B, Computed tomography (CT) of the pelvis before and after therapy demonstrates the
interval appearance of a sclerotic lesion in the right sacral ala (arrow) in a 38-year-old female with metastatic breast cancer. C and
D, Technetium-99m methylene diphosphonate (MDP) bone scans contemporaneous with A and B show an interval development of
tracer uptake in the area of interest (arrowhead). The CT and bone scans appear to indicate disease progression. E and F, Neverthe-
less, contemporaneous fused positron emission tomography/CT images show a marked reduction in fluorodeoxyglucose uptake on
the latter examination, indicating that the sclerotic lesion seen on CT and the associated MDP uptake were a deceptive flare phe-
nomenon caused by healing sclerosis within the lesion that was initially occult on CT and bone scan.

appearance of therapeutic response in bone metastases as and it can be diagnosed by observing a healing response
compared to primary bone tumors. on other imaging modalities (Fig. 2-30).
Different imaging modalities demonstrate different The majority of bone metastases arise in the medullary
physical characteristics of the neoplastic process. Radio- cavity, with a small minority arising in the cortex.78 As
graphs, CT, and nuclear medicine bone scans primarily with primary bone tumors, MRI can directly image the
image the secondary response of the host bone to the metastatic tumor mass inside the medullary cavity. After
tumor rather than the lesion itself. On radiographs and successful therapy, T2 signal, T1 signal, and enhance-
CT, lytic lesions frequently heal with peripheral (rim) or ment often diminish while the size of the lesion may or
internal sclerosis irrespective of the histology of the may not diminish. MRI can detect metastases earlier than
primary tumor, with the exception of nonexpansile radiography or CT79 because MRI does not depend on a
myelomatous lesions, which typically remain lucent. secondary host bone response such as the development
Nevertheless, expansile plasmacytomas will develop of the reparative new bone that is dense on x-ray based
peripheral mineralization after successful therapy.72 imaging modalities and “hot” on bone scan. PET scan-
Enlarging lucencies caused by tumor osteolysis indicate ning reflects metabolic uptake of FDG in the tumors,
progression.73,74 Stability of size typically comprises a which is another direct method of evaluating response in
positive response in sclerotic metastases, whereas an FDG-avid lesions. Studies have shown FDG PET to be
increase in size is seen with progression. Healing sclerotic efficacious in determining the response of the bone
lesions may increase in density but rarely fade. Tracer metastases of breast cancer.80-82 Although not primarily
uptake on bone scan will typically decrease or increase investigated at the time of this writing, FDG uptake was
with healing or progression, respectively.75 A pitfall asso- seen to decrease with therapy in seven bone metastases
ciated with positive therapeutic response can occur on from three Ewing’s sarcoma patients in a study that evalu-
bone scan. Increasing sclerosis within effectively treated ated the response of the primary tumors.61
lesions can cause a spurious increase in tracer uptake The most commonly used response criteria, such as
within the first 3 months of therapy.76,77 This potential RECIST, are often applied to soft tissue metastases such
diagnostic conundrum is designated the flare phenomenon, as lymph nodes or lung nodules and, as mentioned

ERRNVPHGLFRVRUJ
88 2  Clinical Considerations and Imaging of Bone Tumors

TABLE 2-2 MD Anderson (MDA) Criteria*


Response Category Criteria
Complete response Complete sclerotic fill-in of lytic lesions on XR or CT
Normalization of bone density on XR or CT
Normalization of signal intensity on MRI
Normalization of tracer uptake on SS
Partial response Development of a sclerotic rim or partial sclerotic fill-in of lytic lesions on XR or CT
Sclerotic flare: Interval visualization of lesions with sclerotic rims or new sclerotic lesions in the setting
of other signs of partial response and absence of progressive bony disease
≥50% decrease in measurable lesions on XR, CT, or MRI
≥50% subjective decrease in the size of ill-defined lesions on XR, CT, or MRI
≥50% subjective decrease in tracer uptake on SS
Progressive disease ≥25% increase in size of measurable lesions on XR, CT, or MRI
≥25% subjective increase in the size of ill-defined lesions on XR, CT, or MRI
≥25% subjective increase in tracer uptake on SS
New bone metastases
Stable disease No change
<25% increase or <50% decrease in size of measurable lesions
<25% subjective increase or <50% subjective decrease in size of ill-defined lesions
No new bone metastases

*Measurements are based on the sum of a perpendicular, bidimensional measurement of the greatest diameters of each individual
lesion. Abbreviations: CT, computed tomography; MRI, magnetic resonance imaging; SS, skeletal scintigraphy; XR, radiography.
Table modified from Hamaoka T, et al. and published with permission from J Cancer 1:80–92, 2010.

previously, are primarily based on change in size. The Response was not identified with the WHO criteria until
successful therapy of bone metastases rarely results in a 23.5 months (P = 0.025). Early response identification
decrease in size on radiography or CT. A measurable can allow timely modification of therapeutic regimens,
decrease in the size of positively responding bone metas- implying that the behavior of bone metastases can be
tases is more likely to be demonstrated on MRI but does used to guide therapy in these patients.
not occur in a consistent fashion. Additionally, the irregu-
lar shape of many bones and the inability to ensure that
the lesions will be imaged at consistent obliquity renders RESTAGING AND
most bone lesions “unmeasurable” according to RECIST.
Patients who have undergone resection of their primary
ONCOLOGIC SURVEILLANCE
tumor and have metastases only to bone could be denied Local Recurrence
enrollment into clinical trials due to the absence of mea-
surable disease by which to gauge the efficacy of the The restaging and surveillance of patients with bone
therapeutic agents. To address this need, bone-specific tumors can be divided into two main categories, includ-
response criteria have been developed at the University ing local recurrence and distant metastases. The most
of Texas MD Anderson Cancer Center (MDA Criteria; common imaging modality used to evaluate for local
Table 2-2, Fig. 2-31).23 The MDA criteria incorporate recurrence is radiography. Serial radiographs are used to
the changes in imaging appearance of bone metastases on restage malignancies and benign tumors with a tendency
radiographs, CT, bone scan, and MRI into a system that to recur, such as aneurysmal bone cyst or giant cell tumor
has been used to successfully predict progression-free of bone (10%-20% recurrence rates with modern
survival in breast cancer patients with bone-only metas- therapeutic techniques).84,85,86,87 Radiographs of recurrent
tases.83 This retrospective study included 41 breast cancer tumor may show interval development of osteolysis (Fig.
patients who received standardized, systemic therapeutic 2-32), matrix mineralization in the soft tissues, periosteal
regimens. Medical records and patient images were reaction, or expansion of cortex.88 Suspicious radio-
analyzed at 2 to 6 months or 11 to 13 months after the graphic findings are often further investigated with MRI
initiation of therapy. Two musculoskeletal radiologists to assess disease extent and corroborate the diagnosis of
analyzed the response of bone metastases on each imaging recurrence. Most recurrent primary bone tumors have
modality for each patient at each time point using the T1 signal that is isointense to muscle, T2 signal that
MDA, World Health Organization (WHO), and Union is high or heterogeneous, and demonstrate internal
for International Cancer Control (UICC) response cri- enhancement after the administration of intravenous
teria. The UICC criteria provide response guidelines contrast.88 Most small recurrent nodules show solid or
only for radiography and were excluded due to low predominantly solid enhancement. Even tumors that are
numbers of radiographs. The WHO criteria include radi- characteristically cystic or hemorrhagic, such as giant
ography and bone scan but not CT or MRI. The study cell tumors, will typically demonstrate foci of nodular
found that using the MDA criteria, patients who enhancement. An exception is aneurysmal bone cyst,
responded to therapy could be differentiated from those which is entirely cystic and enhancement is seen only in
who did not (progression-free survival) at 5.5 months. the rim. The cystic appearance of recurrent aneurysmal

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 89

A: 12.7mm

B: 11.7mm

A B

B: 26.4mm

A: 26.4mm

C D
FIGURE 2-31  ■  MDA bone response criteria for metastatic disease. Axial computed tomography images of a 24-year-old female with
a primary osteosarcoma of the left humerus. A, A blastic metastasis is observed in the L3 vertebral body. B, The lesion initially
measured 12 mm × 13 mm (25 mm sum of diameters). C and D, Four months later, the lesion enlarged to 26 mm × 26 mm (52 mm
sum of diameters), having undergone a 108% change. This meets the minimum MD Anderson (MDA) size criteria of ≥25% increase
in the sum of the diameters needed for disease progression. The most commonly used response criteria, such as RECIST, would
not have categorized this bone metastasis as measurable disease. The MDA response criteria can be used to quantify response to
therapy in bone metastasis.

bone cysts is corroborated by the clinical context. Con- fifth year. Sarcoma patients are then followed at least once
trast enhanced MRI scans can be helpful in distinguishing per year for a total of 10 years.90 This surveillance regimen
between simple, postoperative cysts (thin, uniform rim is designed to facilitate early detection of recurrent tumor
enhancement) and other types of recurrent tumors (solid with the cost-effective distribution of imaging studies and
or nodular internal enhancement), both of which can clinic visits in the years immediately after the surgery.
demonstrate similar low T1 and homogeneous, high T2 The risk of recurrence diminishes with time; the clinical
signal intensity.89 Ultrasonography is useful for oncologic emphasis of later visits shifts to the integrity of the ortho-
surveillance of superficial or soft tissue resection sites and pedic reconstruction.
can be used to guide biopsy of superficial or palpable
masses. Ultrasound waves cannot penetrate cortex, and
MRI is the most sensitive modality for evaluating recur-
Distant Metastases
rent tumor in the marrow cavity. Malignant bone tumors typically metastasize in a predict-
The local restaging of high-risk sarcoma patients is able fashion, and various imaging studies are used to
often performed with a combination of radiography and evaluate the anatomic locations in which metastatic
MRI or ultrasonography. Clinic visits, which may or may lesions are most likely to occur. Most primary malignant
not include imaging, are typically performed every 6 bone tumors are sarcomas and metastasize preferentially
months for 3 to 5 years for lower grade lesions. For high- to the lungs. Chest CT is more sensitive for detecting
grade sarcomas, the patients are followed every 3 months pulmonary nodules than is radiography and can be used
for 2 years, every 4 months for the third year, every 4 to to monitor high-risk patients, whereas lower risk patients
6 months for the fourth year, and every 6 months for the can be followed with posteroanterior (PA) and lateral

ERRNVPHGLFRVRUJ
90 2  Clinical Considerations and Imaging of Bone Tumors

A B

C
FIGURE 2-32  ■  Local recurrence. A, Frontal radiograph of the proximal humerus in a 7-year-old male with pain in the proximal upper
arm demonstrates a unicameral bone cyst (UBC) with an undisplaced pathologic fracture (arrowheads). B, The lesion was curetted
and ablated with hydrogen peroxide, and the cavity was packed with bone graft. C, Four months later, a rounded lucency indicates
recurrent UBC (arrows). The interventional procedure was repeated and no further recurrence occurred.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 91

A B
FIGURE 2-33  ■  Radiography for indeterminate bone scan. A, Frontal planar technetium-99m MDP bone scan of a 15-year-old male
who underwent resection of an osteosarcoma of the distal right femur. A globular area of tracer uptake at the medial aspect of the
distal right femur is indeterminate and could be potentially related to the resection arthroplasty. B, Knee radiography was then
obtained, and a small focus of osteoid at the area of the area of interest (arrowhead) was indicative of recurrent osteosarcoma.
(Images courtesy of Hubert H. Chuang, MD, PhD, University of Texas MD Anderson Cancer Center.)

chest radiography. Visceral and lymph node metastases followed by planar bone scan to stage the remainder of
may occur later in the course of the disease and are most the skeleton.96 Direct imaging of the marrow cavity with
commonly evaluated with CT of the abdomen and pelvis. MRI allows early detection of bony metastases,79 but
These CT examinations are optimized by use of oral, MRI is more expensive than the combination of radio-
rectal, and intravenous contrast agents. graphs and bone scan.23 If the radiographs are inconclu-
The majority of bone metastases occur in the axial sive and bone scan is not definitive for bone metastases,
skeleton, although metastases to the extremities are not MRI is recommended in sarcoma patients as in patients
uncommon.91,92 Therefore, whole-body imaging modali- with breast cancer.96
ties are necessary to properly evaluate the entire skeleton Another option for evaluating indeterminate areas of
of patients at risk for bone metastases. Imaging of the tracer uptake on planar bone scan is SPECT/CT. A
entire skeleton can also aid in the diagnosis of metastases SPECT scan is essentially a cross-sectional bone scan.
because multiplicity of lesions is common. Bone scan is Tracer uptake from degenerative change is more easily
the most commonly used imaging modality for this indi- identified on SPECT than on a planar bone scan.97 For
cation and is sensitive but not specific for the detection example, SPECT can more readily differentiate an
of bony metastases.73 Nonmalignant processes such as abnormality in the pedicle of a vertebra, which is often
arthritis, degenerative joint disease, and posttraumatic metastatic, from one in a facet joint, which is usually
findings, such as fractures, can induce tracer uptake.93-95 degenerative.98 Modern SPECT scanners are hybrid
This lack of tracer uptake specificity is exacerbated by a devices that also perform a CT to create fused SPECT/
relative lack of anatomic resolution that further limits CT images. The CT further increases the specificity
differentiation between benign and malignant processes. of the scan by providing anatomic information that
Radiographs are specific but not sensitive and can be used can allow even more precise identification of affected
to augment planar bone scans for further evaluation of structures (Fig. 2-34).99
indeterminate areas of tracer uptake73 (Fig. 2-33). Con- PET/CT is an imaging modality that can also provide
versely, radiography is the initial imaging modality of whole-body coverage. Advantages over bone scan include
choice to evaluate a specific area of bone pain in cancer increased anatomic resolution and multiorgan system
patients at risk for osseous metastases. This can be evaluation. PET/CT has become standard of care for the

ERRNVPHGLFRVRUJ
92 2  Clinical Considerations and Imaging of Bone Tumors

A B
FIGURE 2-34  ■  SPECT/CT for indeterminate bone scan. A, Bone scan in a 58-year-old breast cancer patient with tracer uptake in the
left distal femoral diaphysis that is suspicious for a bone metastasis (arrow). B, Coronal fused single-photon emission computed
tomography/computed tomography (SPECT/CT) of the area of interest, obtained in the same imaging session as the planar scan,
demonstrates an intramedullary lesion with characteristic arc-and-ring cartilaginous matrix mineralization. No aggressive features
were seen on the CT, and the diagnosis was an incidental enchondroma. Fused CT adds specificity to functional examinations such
as bone scan and positron emission tomography. Osteoarthritic changes are incidentally seen at the medial compartment of both
knees (arrowheads).

staging of many types of lymphoma.100 As with SPECT/ Conventional MRI is most commonly obtained as a
CT, modern PET scanners are hybrid devices that acquire regional rather than whole-body examination due to the
PET and CT scans in the same imaging session, allowing effect of image-distorting artifacts that increase as the
the fusion of functional and anatomic data sets. PET by size of the area to be scanned enlarges and due to exces-
itself has limited anatomic resolution, and the addition of sive scan times. Technologic developments have permit-
the fused CT data set has been shown to facilitate the ted diagnostic MR scanning of the entire body in
identification of bony metastases with high sensitivity and approximately 1 hour.103 Whole-body MRI has been
specificity101 and has also been found useful in detecting shown to be more sensitive than bone scan for detecting
extraskeletal metastases, such as to the mediastinum and osseous metastases in numerous malignancies.91,104-109 In
liver in patients with Ewing’s sarcoma.102 FDG PET/CT addition to the skeleton, whole-body MRI has been
has been approved for the initial staging of sarcoma shown capable of evaluating organs, such as liver, brain,
patients by the US Centers for Medicare and Medicaid and lymph nodes.91
Services (CMS) at the time of this writing, and its use in Numerous imaging modalities are available to evaluate
the care of patients with bone sarcoma is expected to patients for local recurrence and distant metastases. The
increase in the United States. unique strengths of specific imaging modalities are used

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 93

to perform oncologic staging in the most therapeutically 18. Erlemann R, Reiser MF, Peters PE, et al: Musculoskeletal neo-
and cost effectively optimal manner. plasms: static and dynamic Gd-DTPA–enhanced MR imaging.
Radiology 171(3):767–773, 1989.
19. May DA, Good RB, Smith DK, et al: MR imaging of muscu-
loskeletal tumors and tumor mimickers with intravenous gado-
CONCLUSION linium: experience with 242 patients. Skeletal Radiol 26(1):2–15,
1997.
20. Liu PT, Valadez SD, Chivers FS, et al: Anatomically based guide-
Imaging studies provide important information regard- lines for core needle biopsy of bone tumors: implications for
ing the biologic activity and unique characteristics of limb-sparing surgery. Radiographics 27(1):189–205, 2007.
bone tumors that often aids in the initial diagnosis of the 21. Zimmer WD, Berquist TH, McLeod RA, et al: Bone tumors:
lesions. Initial staging and local restaging is performed magnetic resonance imaging versus computed tomography. Radi-
with techniques such as radiography, CT, and conven- ology 155(3):709–718, 1985.
22. Aisen AM, Martel W, Braunstein EM, et al: MRI and CT evalu-
tional MRI. The entire skeleton can be evaluated for ation of primary bone and soft-tissue tumors. AJR Am J Roentgenol
metastases with bone scan. Modalities such as PET/CT 146(4):749–756, 1986.
and whole-body MRI permit evaluation of multiple organ 23. Hamaoka T, Madewell JE, Podoloff DA, et al: Bone imaging
systems for distant metastases in a single imaging session. in metastatic breast cancer. J Clin Oncol 22(14):2942–2953,
2004.
Oncologic imaging can greatly aid the pathologist and 24. Bushberg BT, Seibert AJ, Leidholdt EM, Jr, et al: Screen-
other specialists in the care of cancer patients. film radiography. The essential physics of medical imaging, ed 2,
Philadelphia, 2002, Lippincott Williams & Wilkins, pp 145–174.
25. Bushberg BT, Seibert AJ, Leidholdt EM, Jr, et al: Computed tomog-
REFERENCES raphy. The essential physics of medical imaging, Philadelphia, 2002,
1. Morrison WBDM, Daffner RH, et al: Bone tumors. In ACR Lippincott Williams & Wilkins, pp 327–372.
appropriateness criteria, Reston, VA, 2005, American College of 26. Tehranzadeh J, Mnaymneh W, Ghavam C, et al: Comparison of
Radiology, pp 1–5. CT and MR imaging in musculoskeletal neoplasms. J Comput
2. Cheng JC, Johnston JO: Giant cell tumor of bone. Prognosis and Assist Tomogr 13(3):466–472, 1989.
treatment of pulmonary metastases. Clin Orthop Relat Res 338:205– 27. Arano Y: Recent advances in 99mTc radiopharmaceuticals. Ann Nucl
214, 1997. Med 16(2):79–93, 2002.
3. Tubbs WS, Brown LR, Beabout JW, et al: Benign giant-cell 28. Koenigsberg M, Freeman LM: Radionuclide bone imaging. Curr
tumor of bone with pulmonary metastases: clinical findings and Probl Diagn Radiol 6(5):1–54, 1976.
radiologic appearance of metastases in 13 cases. AJR Am J Roent- 29. Hudson TM, Chew FS, Manaster BJ: Scintigraphy of benign
genol 158(2):331–334, 1992. exostoses and exostotic chondrosarcomas. AJR Am J Roentgenol
4. Maloney WJ, Vaughan LM, Jones HH, et al: Benign metastasiz- 140(3):581–586, 1983.
ing giant-cell tumor of bone. Report of three cases and review of 30. Lee FY, Yu J, Chang SS, et al: Diagnostic value and limitations of
the literature. Clin Orthop Relat Res 243:208–215, 1989. fluorine-18 fluorodeoxyglucose positron emission tomography for
5. Goldenberg RR, Campbell CJ, Bonfiglio M: Giant-cell tumor of cartilaginous tumors of bone. J Bone Joint Surg Am 86-A(12):2677–
bone. An analysis of two hundred and eighteen cases. J Bone Joint 2685, 2004.
Surg Am 52(4):619–664, 1970. 31. Wang XL, De Beuckeleer LH, De Schepper AM, et al: Low-
6. Joly MA, Vazquez JJ, Martinez A, et al: Blood-borne spread of a grade chondrosarcoma vs enchondroma: challenges in diagnosis
benign giant cell tumor from the radius to the soft tissue of the and management. Eur Radiol 11(6):1054–1057, 2001.
hand. Cancer 54(11):2564–2567, 1984. 32. Enneking WF, Kagan A: “Skip” metastases in osteosarcoma.
7. Kapoor SK, Jain V, Agrawal M, et al: Primary malignant giant cell Cancer 36(6):2192–2205, 1975.
tumor of bone: a series of three rare cases. J Surg Orthop Adv 33. Kager L, Zoubek A, Kastner U, et al: Skip metastases in osteosar-
16(2):89–92, 2007. coma: experience of the Cooperative Osteosarcoma Study Group.
8. Bertoni F, Bacchini P, Staals EL: Malignancy in giant cell tumor J Clin Oncol 24(10):1535–1541, 2006.
of bone. Cancer 97(10):2520–2529, 2003. 34. Jiya TU, Wuisman PI: Long-term follow-up of 15 patients with
9. Anract P, De Pinieux G, Cottias P, et al: Malignant giant-cell non-metastatic Ewing’s sarcoma and a skip lesion. Acta Orthop
tumours of bone. Clinico-pathological types and prognosis: a 76(6):899–903, 2005.
review of 29 cases. Int Orthop 22(1):19–26, 1998. 35. Bushberg BT, Seibert AJ, Leidholdt EM, Jr, et al: Nuclear
10. Lee J, Hoang BH, Ziogas A, et al: Analysis of prognostic factors magnetic resonance. The essential physics of medical imaging, ed 2,
in Ewing’s sarcoma using a population-based cancer registry. Philadelphia, 2002, Lippincott Williams & Wilkins, pp 373–414.
Cancer 116(8):1964–1973, 2010. 36. Bushberg BT, Seibert AJ, Leidholdt EM, Jr, et al: Magnetic
11. Harting MT, Lally KP, Andrassy RJ, et al: Age as a prognostic resonance imaging (MRI). The essential physics of medical imaging,
factor for patients with osteosarcoma: an analysis of 438 patients. Philadelphia, 2002, Lippincott Williams & Wilkins, pp 415–468.
J Cancer Res Clin Oncol 136(4):561–570, 2010. 37. Sajadi KR, Heck RK, Neel MD, et al: The incidence and prog-
12. deSantos LA, Rosengren JE, Wooten WB, et al: Osteogenic nosis of osteosarcoma skip metastases. Clin Orthop Relat Res
sarcoma after the age of 50: a radiographic evaluation. AJR Am J 426:92–96, 2004.
Roentgenol 131(3):481–484, 1978. 38. Lodwick GS, Wilson AJ, Farrell C, et al: Determining growth
13. Giuffrida AY, Burgueno JE, Koniaris LG, et al: Chondrosarcoma rates of focal lesions of bone from radiographs. Radiology 134(3):
in the United States (1973 to 2003): an analysis of 2890 cases 577–583, 1980.
from the SEER database. J Bone Joint Surg Am 91(5):1063–1072, 39. Oudenhoven LF, Dhondt E, Kahn S, et al: Accuracy of radiogra-
2009. phy in grading and tissue-specific diagnosis—a study of 200 con-
14. Papagelopoulos PJ, Galanis E, Frassica FJ, et al: Primary fibro- secutive bone tumors of the hand. Skeletal Radiol 35(2):78–87,
sarcoma of bone. Outcome after primary surgical treatment. Clin 2006.
Orthop Relat Res 373:88–103, 2000. 40. Taube T, Beneton MN, McCloskey EV, et al: Abnormal bone
15. Bielack SS, Schroeders A, Fuchs N, et al: Malignant fibrous his- remodelling in patients with myelomatosis and normal biochemi-
tiocytoma of bone: a retrospective EMSOS study of 125 cases. cal indices of bone resorption. Eur J Haematol 49(4):192–198,
European Musculo-Skeletal Oncology Society. Acta Orthop Scand 1992.
70(4):353–360, 1999. 41. Ragsdale BD, Madewell JE, Sweet DE: Radiologic and pathologic
16. Johnson LC: A general theory of bone tumors. Bull NY Acad Med analysis of solitary bone lesions. Part II: periosteal reactions.
29(2):164–171, 1953. Radiol Clin North Am 19(4):749–783, 1981.
17. Madewell JE, Ragsdale BD, Sweet DE: Radiologic and pathologic 42. Carter DR, Beaupre GS, Giori NJ, et al: Mechanobiology of
analysis of solitary bone lesions. Part I: internal margins. Radiol skeletal regeneration. Clin Orthop Relat Res 355(Suppl):S41–S55,
Clin North Am 19(4):715–748, 1981. 1998.

ERRNVPHGLFRVRUJ
94 2  Clinical Considerations and Imaging of Bone Tumors

43. Goodship AE, Cunningham JL, Kenwright J: Strain rate and 65. Hawkins DS, Schuetze SM, Butrynski JE, et al: [18F]Fluorodeoxy-
timing of stimulation in mechanical modulation of fracture glucose positron emission tomography predicts outcome for
healing. Clin Orthop Relat Res 355(Suppl):S105–S115, 1998. Ewing sarcoma family of tumors. J Clin Oncol 23(34):8828–8834,
44. Sweet DE, Madewell JE, Ragsdale BD: Radiologic and pathologic 2005.
analysis of solitary bone lesions. Part III: matrix patterns. Radiol 66. Brenner W, Conrad EU, Eary JF: FDG PET imaging for grading
Clin North Am 19(4):785–814, 1981. and prediction of outcome in chondrosarcoma patients. Eur J Nucl
45. Murphey MD, Flemming DJ, Boyea SR, et al: Enchondroma Med Mol Imaging 31(2):189–195, 2004.
versus chondrosarcoma in the appendicular skeleton: differentiat- 67. Benjamin RS, Choi H, Macapinlac HA, et al: We should desist
ing features. Radiographics 18(5):1213–1237, 1998. using RECIST, at least in GIST. J Clin Oncol 25(13):1760–1764,
46. Feldman F, Vanheertum R, Saxena C: 18Fluoro-deoxyglucose 2007.
positron emission tomography evaluation of benign versus malig- 68. Aparicio J, Munarriz B, Pastor M, et al: Long-term follow-up
nant osteochondromas: preliminary observations. J Comput Assist and prognostic factors in Ewing’s sarcoma. A multivariate analysis
Tomogr 30(5):858–864, 2006. of 116 patients from a single institution. Oncology 55(1):20–26,
47. Aoki J, Watanabe H, Shinozaki T, et al: FDG-PET in differential 1998.
diagnosis and grading of chondrosarcomas. J Comput Assist Tomogr 69. Bacci G, Longhi A, Bertoni F, et al: Bone metastases in osteosar-
23(4):603–608, 1999. coma patients treated with neoadjuvant or adjuvant chemother-
48. Yanagawa T, Watanabe H, Shinozaki T, et al: The natural history apy: the Rizzoli experience in 52 patients. Acta Orthop 77(6):
of disappearing bone tumours and tumour-like conditions. Clin 938–943, 2006.
Radiol 56(11):877–886, 2001. 70. Hopper KD, Moser RP, Jr, Haseman DB, et al: Osteosarcomato-
49. Balke M, Schremper L, Gebert C, et al: Giant cell tumor of bone: sis. Radiology 175(1):233–239, 1990.
treatment and outcome of 214 cases. J Cancer Res Clin Oncol 71. Shinozaki T, Chigira M, Watanabe H, et al: Osteosarcoma with
134(9):969–978, 2008. multiple skeletal metastases. A case of “nonstochastic” metastasis.
50. Lewis VO, Wei A, Mendoza T, et al: Argon beam coagulation as Arch Orthop Trauma Surg 112(6):292–295, 1993.
an adjuvant for local control of giant cell tumor. Clin Orthop Relat 72. Roodman GD: Mechanisms of bone metastasis. N Engl J Med
Res 454:192–197, 2007. 350(16):1655–1664, 2004.
51. Cioni R, Armillotta N, Bargellini I, et al: CT-guided radiofre- 73. Hortobagyi GN, Libshitz HI, Seabold JE: Osseous metastases
quency ablation of osteoid osteoma: long-term results. Eur Radiol of breast cancer. Clinical, biochemical, radiographic, and scinti-
14(7):1203–1208, 2004. graphic evaluation of response to therapy. Cancer 53(3):577–582,
52. Hoffmann RT, Jakobs TF, Kubisch CH, et al: Radiofrequency 1984.
ablation in the treatment of osteoid osteoma: 5-year experience. 74. Libshitz HI, Hortobagyi GN: Radiographic evaluation of thera-
Eur J Radiol 73(2):374–379, 2010. peutic response in bony metastases of breast cancer. Skeletal Radiol
53. Smith J, Heelan RT, Huvos AG, et al: Radiographic changes in 7(3):159–165, 1981.
primary osteogenic sarcoma following intensive chemotherapy. 75. Janicek MJ, Shaffer K: Scintigraphic and radiographic patterns
Radiological-pathological correlation in 63 patients. Radiology of skeletal metastases in breast cancer: value of sequential
143(2):355–360, 1982. imaging in predicting outcome. Skeletal Radiol 24(8):597–600,
54. Ueda T, Naka N, Araki N, et al: Validation of radiographic 1995.
response evaluation criteria of preoperative chemotherapy for 76. Chao HS, Chang CP, Chiu CH, et al: Bone scan flare phenom-
bone and soft tissue sarcomas: Japanese Orthopaedic Association enon in non-small-cell lung cancer patients treated with gefitinib.
Committee on Musculoskeletal Tumors Cooperative Study. Clin Nucl Med 34(6):346–349, 2009.
J Orthop Sci 13(4):304–312, 2008. 77. Schneider JA, Divgi CR, Scott AM, et al: Flare on bone scintig-
55. Eisenhauer EA, Therasse P, Bogaerts J, et al: New response evalu- raphy following Taxol chemotherapy for metastatic breast cancer.
ation criteria in solid tumours: revised RECIST guideline (version J Nucl Med 35(11):1748–1752, 1994.
1.1). Eur J Cancer 45(2):228–247, 2009. 78. Coerkamp EG, Kroon HM: Cortical bone metastases. Radiology
56. Kim MS, Lee SY, Cho WH, et al: Effect of increases in tumor 169(2):525–528, 1988.
volume after neoadjuvant chemotherapy on the outcome of stage 79. Avrahami E, Tadmor R, Dally O, et al: Early MR demonstration
II osteosarcoma regardless of histological response. J Orthop Sci of spinal metastases in patients with normal radiographs and CT
14(3):292–297, 2009. and radionuclide bone scans. J Comput Assist Tomogr 13(4):598–
57. Pauwels EK, Sturm EJ, Bombardieri E, et al: Positron-emission 602, 1989.
tomography with [18F]fluorodeoxyglucose. Part I. Biochemical 80. Tateishi U, Gamez C, Dawood S, et al: Bone metastases in
uptake mechanism and its implication for clinical studies. J Cancer patients with metastatic breast cancer: morphologic and metabolic
Res Clin Oncol 126(10):549–559, 2000. monitoring of response to systemic therapy with integrated PET/
58. Tateishi U, Yamaguchi U, Seki K, et al: Glut-1 expression and CT. Radiology 247(1):189–196, 2008.
enhanced glucose metabolism are associated with tumour grade 81. Du Y, Cullum I, Illidge TM, et al: Fusion of metabolic function
in bone and soft tissue sarcomas: a prospective evaluation by and morphology: sequential [18F]fluorodeoxyglucose positron-
[18F]fluorodeoxyglucose positron emission tomography. Eur J emission tomography/computed tomography studies yield new
Nucl Med Mol Imaging 33(6):683–691, 2006. insights into the natural history of bone metastases in breast
59. Costelloe CM, Macapinlac HA, Madewell JE, et al: 18F-FDG cancer. J Clin Oncol 25(23):3440–3447, 2007.
PET/CT as an indicator of progression-free and overall survival 82. Stafford SE, Gralow JR, Schubert EK, et al: Use of serial FDG
in osteosarcoma. J Nucl Med 50(3):340–347, 2009. PET to measure the response of bone-dominant breast cancer to
60. Hawkins DS, Conrad EU, 3rd, Butrynski JE, et al: [F-18]- therapy. Acad Radiol 9(8):913–921, 2002.
fluorodeoxy-d-glucose–positron emission tomography response is 83. Hamaoka T, Costelloe CM, Madewell JE, et al: Tumour response
associated with outcome for extremity osteosarcoma in children interpretation with new tumour response criteria vs the World
and young adults. Cancer 115(15):3519–3525, 2009. Health Organisation criteria in patients with bone-only metastatic
61. Franzius C, Sciuk J, Brinkschmidt C, et al: Evaluation of chemo- breast cancer. Br J Cancer 102(4):651–657, 2010.
therapy response in primary bone tumors with F-18 FDG posi- 84. Gibbs CP, Jr, Hefele MC, Peabody TD, et al: Aneurysmal bone
tron emission tomography compared with histologically assessed cyst of the extremities. Factors related to local recurrence after
tumor necrosis. Clin Nucl Med 25(11):874–881, 2000. curettage with a high-speed burr. J Bone Joint Surg Am 81(12):
62. Franzius C, Bielack S, Flege S, et al: Prognostic significance of 1671–1678, 1999.
18
F-FDG and 99mTc-methylene diphosphonate uptake in primary 85. Varshney MK, Rastogi S, Khan SA, et al: Is sclerotherapy better
osteosarcoma. J Nucl Med 43(8):1012–1017, 2002. than intralesional excision for treating aneurysmal bone cysts?
63. Nair N, Ali A, Green AA, et al: Response of Osteosarcoma to Clin Orthop Relat Res 468(6):1649–1659, 2009.
Chemotherapy. Evaluation with F-18 FDG-PET scans. Clin Posi- 86. Errani C, Ruggieri P, Asenzio MA, et al: Giant cell tumor of the
tron Imaging 3(2):79–83, 2000. extremity: a review of 349 cases from a single institution. Cancer
64. Schulte M, Brecht-Krauss D, Werner M, et al: Evaluation of Treat Rev 36(1):1–7, 2010.
neoadjuvant therapy response of osteogenic sarcoma using FDG 87. Mendenhall WM, Zlotecki RA, Scarborough MT, et al: Giant cell
PET. J Nucl Med 40(10):1637–1643, 1999. tumor of bone. Am J Clin Oncol 29(1):96–99, 2006.

ERRNVPHGLFRVRUJ
2  Clinical Considerations and Imaging of Bone Tumors 95

88. Costelloe CM, Kumar R, Yasko AW, et al: Imaging characteristics 100. Wahl RL, Jacene H, Kasamon Y, et al: From RECIST to
of locally recurrent tumors of bone. AJR Am J Roentgenol 188(3): PERCIST: evolving considerations for PET response criteria in
855–863, 2007. solid tumors. J Nucl Med 50(Suppl 1):122S–150S, 2009.
89. Costelloe CM, Murphy WA, Jr, Haygood TM, et al: Comparison 101. Costelloe CM, Chuang HH, Madewell JE: FDG PET for the
of half-dose and full-dose gadolinium MR contrast on the detection of bone metastases: sensitivity, specificity and compari-
enhancement of bone and soft tissue tumors. Skeletal Radiol son with other imaging modalities. PET Clinics 5(3):281–295,
40(3):327–333, 2010. 2010.
90. Costelloe C, Lin PP: Follow-up evaluation and surveillance. Bone 102. Franzius C, Sciuk J, Daldrup-Link HE, et al: FDG-PET for
sarcoma (MD Anderson Cancer Care Series), New York, in press, detection of osseous metastases from malignant primary bone
Springer. tumours: comparison with bone scintigraphy. Eur J Nucl Med
91. Costelloe C, Kundra V, Ma J, et al: Fast Dixon whole-body 27(9):1305–1311, 2000.
MRI for detecting distant cancer metastasis: a preliminary clinical 103. Ma J, Costelloe CM, Madewell JE, et al: Fast Dixon-based mul-
study. JMRI 35(2):399–408, 2012. tisequence and multiplanar MRI for whole-body detection of
92. Clain A: Secondary Malignant Disease of Bone. Br J Cancer 19: cancer metastases. J Magn Reson Imaging 29(5):1154–1162, 2009.
15–29, 1965. 104. Eustace S, Tello R, DeCarvalho V, et al: A comparison of whole-
93. Corcoran RJ, Thrall JH, Kyle RW, et al: Solitary abnormalities in body turboSTIR MR imaging and planar 99mTc-methylene
bone scans of patients with extraosseous malignancies. Radiology diphosphonate scintigraphy in the examination of patients with
121(3 Pt 1):663–667, 1976. suspected skeletal metastases. AJR Am J Roentgenol 169(6):1655–
94. Citrin DL, Bessent RG, Greig WR: A comparison of the sensitiv- 1661, 1997.
ity and accuracy of the 99TCm-phosphate bone scan and skeletal 105. Horvath LJ, Burtness BA, McCarthy S, et al: Total-body echo-
radiograph in the diagnosis of bone metastases. Clin Radiol 28(1): planar MR imaging in the staging of breast cancer: comparison
107–117, 1977. with conventional methods—early experience. Radiology 211(1):
95. Lee YT: Bone scanning in patients with early breast carcinoma: 119–128, 1999.
should it be a routine staging procedure? Cancer 47(3):486–495, 106. Steinborn MM, Heuck AF, Tiling R, et al: Whole-body bone
1981. marrow MRI in patients with metastatic disease to the skeletal
96. Costelloe CM, Rohren EM, Madewell JE, et al: Imaging bone system. J Comput Assist Tomogr 23(1):123–129, 1999.
metastases in breast cancer: techniques and recommendations for 107. Walker R, Kessar P, Blanchard R, et al: Turbo STIR magnetic
diagnosis. Lancet Oncol 10(6):606–614, 2009. resonance imaging as a whole-body screening tool for metastases
97. Sedonja I, Budihna NV: The benefit of SPECT when added to in patients with breast carcinoma: preliminary clinical experience.
planar scintigraphy in patients with bone metastases in the spine. J Magn Reson Imaging 11(4):343–350, 2000.
Clin Nucl Med 24(6):407–413, 1999. 108. Lauenstein TC, Goehde SC, Herborn CU, et al: Whole-body
98. Han LJ, Au-Yong TK, Tong WC, et al: Comparison of bone MR imaging: evaluation of patients for metastases. Radiology
single-photon emission tomography and planar imaging in the 233(1):139–148, 2004.
detection of vertebral metastases in patients with back pain. Eur 109. Thomson V, Pialat JB, Gay F, et al: Whole-body MRI for metas-
J Nucl Med 25(6):635–638, 1998. tases screening: a preliminary study using 3D VIBE sequences
99. Romer W, Nomayr A, Uder M, et al: SPECT-guided CT for with automatic subtraction between noncontrast and contrast
evaluating foci of increased bone metabolism classified as indeter- enhanced images. Am J Clin Oncol 31(3):285–292, 2008.
minate on SPECT in cancer patients. J Nucl Med 47(7):1102–
1106, 2006.

ERRNVPHGLFRVRUJ
C H A P T E R 3 

Molecular Biology of Bone Tumors


CHAPTER OUTLINE

MOLECULAR TECHNIQUES CELL-CYCLE AND MITOTIC REGULATION


MOLECULAR BIOLOGY OF BONE FORMATION MOLECULAR CONCEPTS OF BONE NEOPLASIA
MECHANISMS OF MALIGNANT
TRANSFORMATION

During the past two decades, our knowledge of the MOLECULAR TECHNIQUES
molecular events that are responsible for the develop-
ment and progression of many human neoplasms, includ- The most frequently used techniques to elucidate the
ing those that affect the skeleton, has dramatically genetic abnormalities discussed here are based on the
improved. This has been made possible by numerous extraction and analysis of deoxyribonucleic acid (DNA),
technologic developments that have contributed to the ribonucleic acid (RNA), or proteins. To facilitate the
understanding of cellular biology and that shed light on separation and identification of the genetic material, long
some molecular aspects of the malignant transformation strands of DNA are usually cut with specific restriction
of cells. The past decade is defined as the postgenomic enzymes (endonucleases). Restriction endonucleases are
era, in which many diseases, including bone tumors, are a unique class of enzymes that recognize specific nucleo-
beginning to be understood in the context of genome tide sequences and cut double-stranded DNA at specific
changes rather than only in relation to individual genes points. These enzymes are used in nearly all gene recon-
and their respective pathways. This is due to the comple- struction and gene transfection (gene transfer) experi-
tion of the sequencing of the human genome released to ments, collectively known as DNA recombinant technology
the public as the so-called reference human genome sequence or genetic engineering. The many techniques used to iden-
in 2004.9,10,23 This accomplishment has altered the land- tify normal or altered components of a genome are
scape of biomedical science including medicine and has assembled into strategies capable of identifying both the
changed the environment in which health care is pro- structural and functional aspects of the genetic element
vided.7,3,20,23,27 It also has accelerated improvements in our under the study.
understanding of skeletal physiology as a complex body- The techniques of modern molecular genetics can
supporting organ and has provided new insights into the be divided into three main groups: DNA hybridization
biology of bone tumors. techniques, technologies that involve polymerase chain
In this chapter, we focus on some of the most promis- reaction (PCR), and genomic sequencing with high
ing areas of molecular research related to the understand- throughput genomic profiling. Commonly used terms
ing of the molecular pathogenesis of bone tumors and and techniques are described in the following paragraphs.
that are relevant to their diagnosis, as well as representing
novel, still to be fully explored, therapeutic targets. Hybridization Techniques
The pathogenesis of neoplasia cannot be understood
without basic knowledge of the biology of the cells Hybridization techniques were originally designed to
involved in the process of transformation. Therefore the identify individual components of our genetic material
description of the molecular biology of bone tumors is such as genes and expressed species of RNA such as
preceded by a section describing some fundamental tech- mRNA or proteins.11,25 In general, the material is first
niques used to investigate the function of cells and on the separated by gel electrophoresis and transferred to a
molecular biology of physiologic bone formation. The membrane; then the specific DNA, RNA, or proteins are
description of both skeletal physiology and the biology identified with a detection probe. These techniques
of bone tumors is restricted to a few fundamental aspects involve Southern blotting, in which specific components
of cell function in physiology and disease. An interested of DNA are identified, Northern blotting, in which spe-
reader is referred to comprehensive textbooks concern- cific species of RNA are analyzed, and finally Western
ing the biology of the skeleton as well as the biology of blotting for the identification of specific proteins. These
cancer. The two recommended highly respected and techniques are described here for historical purposes.
periodically reviewed texts are by JP Bilezikian, et al, However, today Southern blotting is rarely used, most
Principles of Bone Biology, and by RA Weinberg, The Biology often in the identification of the ablated regions of the
of Cancer. genome in the development of genetically engineered
96
ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 97

animals. Northern blotting has almost been completely label and is visualized by autoradiography to identify posi-
eliminated from the modern armamentarium of molecu- tions of bands that are complementary to the probe.
lar techniques and has been replaced by more effective
techniques such as PCR and quantitative PCR. On the Northern Blotting
other hand, Western blotting remains the most popular
technique used in molecular biology and pathology for Northern blotting is used to determine the size and
diagnostic clinical workups. amount of specific messenger RNA (mRNA). As in South-
ern blotting, the RNA is separated by electrophoresis on
the basis of size using denaturing agarose gels and is
Southern Blotting
transferred to a solid support (nylon membranes, nitrocel-
Southern blotting is used to identify particular sequences lulose filters). The RNA of interest is located by hybrid-
within genomic DNA. Traditionally, DNA is digested ization with an appropriately labeled DNA or RNA probe.
with restriction endonucleases, and the resulting DNA
fragments are separated by size using electrophoresis Western Blotting
through agarose gels. After in situ denaturation of the
fragments by an alkaline solution or heat, the DNA is Western blotting is used for the immunologic identification
transferred from the gel onto a solid support (nylon mem- of proteins extracted from tissues or cells. For this proce-
branes, nitrocellulose filters). The relative positions of the dure, protein extracts are separated by electrophoresis and
fragments are preserved during this transfer. Subsequently, then transferred to a solid support and identified with
the DNA blotted to the solid support is hybridized with labeled or unlabeled antibodies that recognize the antigenic
a known, specific DNA or RNA probe with a radioactive epitopes displayed by the target protein (Fig. 3-1). When

Sample Preparation

Well
Samples
Electrode −

Larger fragments Buffer

Smaller fragments
Gel

Electrode +

Plastic frame

Electrophoresis
A B Transfer

Detection signal
(colorimetric or chemiluminescent)

Enzyme substrate cathode (−)


Enzyme covalenty
attached to 2nd antibody filter papers
Secondary antibody
against primary polyacrylamide gel
Anti-target protein antibody
antibody (IgG) membrane
(primary antiobody)
Target protein
filter papers
anode (+)
Membrane blo
t Membrane Staining
C
Fluorescent Detection
D
FIGURE 3-1  ■  Identification of individual proteins by Western blotting. A, Separation of protein lysates by electrophoresis. B, Western
blotting machine facilitating electrophoresis and membrane transfer. C, Transfer of proteins from polyacrylamide gel to nitrocellulose
membrane. D, Identification of proteins with primary antibodies and colorimetric or chemiluminescent secondary detection antibodies.

ERRNVPHGLFRVRUJ
98 3  Molecular Biology of Bone Tumors

unlabeled antibodies are used, the identification of the of the target template DNA and flank the DNA segment
bands is done with a labeled secondary antibody to the being amplified. The reaction mixture contains the four
immunoglobulin class of the animal species in which the nucleotides required for DNA synthesis in the presence
first antibody was raised. In general, the Western blotting of a heat-stable DNA polymerase. The procedure com-
procedure depends on three key steps: (1) the separation prises three basic steps: heat denaturation of the DNA,
of proteins by size using gel electrophoresis, (2) the trans- cooling and annealing of the primers to their target
fer of separated proteins to a membrane, and (3) the sequences, and extension by a heat-stable DNA poly-
detection of a target protein by specific antibodies. Typi- merase (Fig. 3-2). Because the product generated in the
cally, the antibodies used in Western blotting recognize first PCR round serves as the template for subsequent
a short specific linear sequence of amino acids within the rounds, an exponential amplification of the DNA segment
target protein. The target protein is visualized as a band of interest may be obtained by repeating the cycles. The
on a blotting membrane using radiography or an imaging usual amplification level is on the order of 106 after 25 to
system with secondary antibodies labeled with chemilu- 30 cycles. Greater amplification (109 to 1011) of DNA
minescence or fluoroluminescence. The thickness of the copies may be achieved by repeating the cycles with the
band or, in general, the staining intensity corresponds to product generated in the first run used as a template
the amount of the target protein. Western blotting is one and fresh DNA polymerase. PCR can also be used to
of the most efficient and popular techniques, utilizing generate amplified DNA from mRNA after initial reverse
simple equipment and inexpensive reagents. transcription.
Real-time reverse transcription polymerase chain
reaction (RT PCR), or quantitative polymerase chain
Polymerase Chain Reaction reaction (qPCR), is based on the concept of standard
PCR is an in vitro technique used to amplify a DNA PCR and facilitates the quantitative assessment of a tem-
segment lying between two regions of known sequences plate DNA (Fig. 3-3). It utilizes fluorescent dyes tagged
by using two oligonucleotide primers.5,8 The primers are to a probe, which anneal to the amplified product; the
complementary to the sequences on the opposite strands signal intensity is proportional to the amount of DNA

A 5’ 3’ B
3’ 5’

Denaturation

p53 exon 9
5’ 3’ 3’ 5’

Size marker
Annealing
5’ 3’ 3’ 5’
Extending
5’ 3’
3’ 5’
Elongation
5’ 3’
x30 3’ 5’

etc

C
T

mutation: CAG TAG (Gln330STOP)


C
FIGURE 3-2  ■  Polymerase chain reaction. A, Schematic drawing showing the design of polymerase chain reaction and exponential
amplification of DNA in each thermal cycle comprising denaturation, annealing, and elongation repeated 30 times with the use of
forward and reverse primers, depicted as forward and reverse arrows. B, Amplified DNA fragments shown on fluorescent electro-
phoresis gel. C, DNA sequence of the PCR fragment shown in B with C-T mutation results in STOP codon.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 99

TaqMan being amplified.1,14,29 RT PCR utilizes the standard PCR


Fluorophore Probe Quencher sequence, specific forward and reverse primers, and a
5’ fluorescently labeled probe that provides quantitative
3’
assessment of the template DNA. In general, the inten-
reverse PCR primer sity of the fluorescence emitted during the amplification
cycle correlates with the amount of the DNA that is
amplification assay
Polymerization amplified. Fluorescence intensity increases proportion-
ally to the amplification cycles; after a few initial cycles
5’
3’ it surpasses a threshold level set above the background
and starts to increase exponentially. The Cq value (quan-
Fluorescence Probe displacement tification cycle), which represents the number of PCR
and cleavage
cycles that pass before the threshold is reached, is a
measure of the original template DNA. By comparing the
5’
3’ results of samples of unknown concentration with a series
of standards, the amount of template DNA in the mea-
Result sured sample with unknown concentration can be deter-
Fluorescence mined. RT PCR has wide applications in gene expression
analysis, pathogen detection, and genetic testing. It can
also be performed in a multiplex form, assessing the
PCR Cleavage expression level of many genes.
A products products

Genomic Sequencing
0.3
Exponential phase With the completion of the Human Genome Project, the
Non-
exponential release of the first draft of the human genome sequence
Fluorescence

plateau in 2001, and the final publication of a reference human


0.2 phase genome sequence in 2004, genomics became a main-
stream discipline of biomedical research.9,10 The rapid
Cq value advances in genomic sequencing technologies facilitated
0.1 the ability to sequence the genome of an individual
Threshold line human being in 2008, offering genomic profiling of
tumor samples and their paired germ line DNA to cancer
0
patients.12,16,19,21,24,28 This approach has provided new
0 10 20 30 40
insights into the biology of many diseases, including
B Cycle
cancer, potentially improving human health. Technologic
4,000
development is the driving force of genomics and is
Target responsible for the invention of new methods, reagents,
Relavive Fluorescence

input and instruments.23,24,30 It also inspires the improvement


3,000 108 of efficiency and output, with consistently decreasing
107 costs making genomic profiling possible for individual
106
2,000
105 patients (Fig. 3-4). The novel genome sequencing tech-
104 nologies collectively referred to as next-generation sequenc-
1,000 Baseline Fluorescence 103 ing enable assessment of mutations in several hundreds
102 of potential therapeutically targetable genes and are also
101
0 NTC capable of applying total genome sequencing to indi-
vidual patients. It is predicted that during the next decade
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 we will witness an unprecedented genomic characteriza-
C Cycle tion of many human cancers. This will be accomplished
FIGURE 3-3  ■  Quantitative polymerase chain reaction. A, Ampli- in parallel with a more in-depth understanding of the
fication cycles of the quantitative polymerase chain reaction biology of our genome and how it functions in a disor-
(PCR), which utilize the standard forward and reverse primers dered state in cancer development and progression.
and the third fluorescently labeled primers that are used to Genomic approaches are also anticipated to open novel
quantitate the amplified template DNA. B, The fluorescence
intensity curve during amplification of template DNA. The
avenues for the improvement of cancer therapy and the
number of PCR cycles that pass before the threshold fluores- overall effectiveness of cancer patient outcomes. The
cence level is reached (Cq value) reflects the amount of the most advanced genome sequencing technologies are
original template DNA. C, Fluorescence intensity curves after capable of analyzing the entire human genome sequence
titration of the template DNA. The Cq values for each titration within a few days at reasonable cost (Fig. 3-5). Such
are indicated by the arrows.
technologies offer the detection of a full spectrum of
genomic abnormalities that include point mutations,
copy number alterations including homozygous and
hemizygous deletions, copy gain, translocation break-
points, and pathogen identification.

ERRNVPHGLFRVRUJ
100 3  Molecular Biology of Bone Tumors

ps
gr of
ou
e e
tiv ag
io se

g
ec av
er

llin
ns a
n
te e b
rim

o t le
g

ca
pr C
in
ex ngl
.p

ag

se
q

Si

Im

Ba
Se
T
G
C
T G
G A
C A
G T
A T
A
T
T

A B
Reference sequence Non-human
Chr 1 Chr 5 sequence
A
c
c
c
c
c
c
c
c
c
c
c
c
c
c
c

Homozygous Hemizygous Translocation


Point mutation Indel deletion deletion Gain breakpoint Pathogen

Copy number alterations


C
FIGURE 3-4  ■  Principles of DNA sequencing using next-generation technologies. A, Illumina 2000 Hi-Seq DNA Sequencer. B, DNA
sequencing with fluorescent chain elongation used in modern next-generation sequencing strategies. The base calling is accom-
plished on the addition of the fluorescently labeled nucleotide pairing the template DNA attached to solid support. C, Types of
genomic alterations that can be analyzed on a total genome scale using genome profiling platforms. (Reprinted with permission from
Meyerson M, et al: Nat Rev Genet 11:685–696, 2010.)

High Throughput Genomic Profiling using the cDNA microarray.4,15 Similarly, the expression
levels of various regulatory RNA species such as
The completion of the Human Genome Project and the microRNA can also be assessed on a total genome scale.22
knowledge of the complete sequence of our genome Finally, the epigenetic modifications of the genome,
enabled the development of a full roster of high through- including methylation and histone acetylation, can be
put genomic platforms that facilitate the analysis of analyzed with specific strategies and microarrays.18 The
various structural and functional aspects of our genome genomic high throughput platforms are complemented
both in physiology and disease. They include the single with proteomic and metabolomic technologies that are
nucleotide polymorphism-based, high density microar- capable of analyzing the expression profiles and status of
rays with which we can assess precisely the copy number metabolic activities in the entire cellular proteome.2,13,17,26
change of specific genomic regions and analyze it on a All of these platforms are capable of generating tsunami
global genomic scale.6,15,27 The expression patterns of waves of data that represent a major analytical challenge
genes can be investigated by testing the levels of mRNA to the health care system in our time.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 101

NRAS
FAM46C
FAF1
CDKN2C ANP32E
CKS1B

40

40
120

80

120
0
0

160
DTNB

80
y

200
40
IGL@ 1 DNMT3A

240
x

0
40

0
IGK@

40
0
22

40
MAFB

80
21

0
12
40
2 16
0
40
0 20
0
20
0 0
19 24 ULK4
0
40
TRAK1
Translocation Partner 40
18
800 80
Deleted
3
40 Mutated 120
TP53 17
WWOX 0 Deleted and Mutated 160
80
FGFR3
MAF MMSET
Gained 0
CYLD 40 16
Inherited 40
0
80
80 4
15 120
40
160
IGH@ 0
TRAF3 80 0
14
40
40 DNAH5
0
5 80

120
DIS3 80 13
16
0
40
RBQ1 0
0
0 40
12 6
12 80 CCND3
80
12
40 0
KRAS
0

16
11

0
0
0

CCND2 7
12

40
80

BIRC2 80 DNAH11
40

BIRC3 10 8 CDCA7L
120
0
120

9
0
80

40

BRAF
40

80

CCND1
0

120
120

0
80

40

MYC
FIGURE 3-5  ■  Example of genome sequencing data in multiple myeloma. Circos diagram summarizing the sequencing and copy
number variation data in multiple myeloma. The chromosomes are aligned in a clockwise circle and the key mutations are depicted.
The inner circle shows the copy number change. Solid lines designate the positions and partners for chromosomal translocation.
(Reprinted with permission from Morgan GJ, et al: Nat Rev Cancer 12:335–348, 2012.)

template undergo stepwise differentiation into hyper-


MOLECULAR BIOLOGY tropic chondrocytes.70,73 This maturation process requires
OF BONE FORMATION cessation of proliferation. Hypertropic chondrocytes,
after exiting the cell cycle, die by apoptosis and their
The human skeleton is a complex body-supporting organ cartilage matrix is used as a template for bone formation.
with more than 200 bones adapted to bipedal locomo- This process requires precise zonal orchestration of two
tion. The majority of the human skeleton is formed main cell lineages involved in skeletal development—
by endochondral ossification, which is responsible for chondroblasts and osteoblasts—which are mediated in
growth and patterning of the long bones of the limbs and this process through Indian hedgehog (IHH) signal-
elements of the axial skeleton (Fig. 3-6).60,84 Fewer bones, ing.52,53 In intramembranous ossification, flat bones
predominantly the flat bones and the clavicle, are formed develop directly from mesenchymal condensation and no
by intramembranous ossification.60,84,112 Endochondral interaction with a preformed cartilage template is needed
ossification starts as condensations of mesenchymal cells for bone development. During the past decade, there has
that form cartilaginous templates subsequently remod- been a dramatic development of our understanding of the
eled into bone. The condensation centers have peculiar molecular mechanisms that orchestrate these processes.
zonal patterns of differentiation in which the central The genes and their respective proteins that play a role
component differentiates into cartilage while the periph- in skeletal development can be, in part, used as biomark-
eral cells form perichondrium, which differentiates into ers for differential diagnosis of bone tumors and can
osteoblastic precursors and in the mature phase forms provide new insights, not only into the physiology of
the periosteum. The central components of the cartilage bone, but also on the pathogenesis of bone tumors.

ERRNVPHGLFRVRUJ
102 3  Molecular Biology of Bone Tumors

da

B li
B

li

da

C da

li
D

cm

oc oc

cm cm
ob

A F
FIGURE 3-6  ■  Morphology of tissue undergoing endochondral ossification. A, Light micrograph of the growth plate of an equine
fetus. Labels B-D indicate the regions corresponding to electron micrographs of chondrocytes shown in parts B-D of this figure. The
invading ossification front is seen in the lower part of the image and includes blood vessels (arrowheads) and multinucleated
osteoclasts (oc); arrows indicate the bone matrix being deposited on remnants of cartilage matrix (cm) by a row of osteoblasts.
B-F, Electron micrographs of different cell types seen in part A. Light (li) and dark (da) chondrocytes are seen in the late zone of
proliferation (B), the middle zone of hypertrophy (C), and the last few lacunae before the ossification front, where the cells are dying
(D). E, An osteoclast adherent to a remnant of cartilage matrix, adjacent to an erythrocyte (arrowhead) in a blood capillary. F, Two
osteoblasts (ob) depositing bone matrix (arrows) on a remnant of cartilage matrix. (Bar in A = 50  µm. The magnification for parts
B-D is the same, and the magnification for parts E and F is the same; bar = 10  µm.) (Reprinted with permission from Mackie EJ, et al:
IJBCB 40:46–62, 2008.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 103

Three distinct cell lineages of mesenchymal origin chondrocytes. The expression level of Osterix is con-
participate in the formation of the skeleton: osteoblastic, trolled by RUNX2 and is stimulated by interaction with
chondroblastic, and osteoclastic.74,85 These cells cooper- the nuclear factor of activated T-cells (NFAT). RUNX2
ate and influence one another in the formation and is required in early phases of osteoblastic progenitor cells
remodeling of the skeleton. For example, osteoblasts and is downregulated in mature osteoblasts. The activity
produce factors that affect the function of osteoclasts. In of RUNX2 is enhanced by multiple factors such as
the enchondral ossification model, the formation of car- SATB2, ATF/CREB family members, and is reduced by
tilage matrix is a prerequisite event for osteoid deposition DLX3 and MSX2. The transcription factors SP3 and
in the primary spongiosa. Subsequent resorption of the ATF4 play a role in the late phases of osteoblastic dif-
primary spongiosa by osteoclastic cells is an important ferentiation. ATF4 is expressed at high levels in osteo-
step in bone remodeling and formation of the secondary blasts because of its inhibited proteasomal degradation.
spongiosa. Other factors that regulate bone-forming and The activation of osteoblasts is also modified by phos-
bone-remodeling functions of cells are produced in phorylation of ATF4 by the kinase RSK2 and interaction
response to extracellular growth factors and hormonal with SATB2. Finally, the matrix production of osteoblasts
stimulation. It is speculated that the osteoblastic cells is likely controlled by FRA1. Osteocalcin gene upregula-
participating in intramembranous and enchondral bone tion is a feature of late osteoblastic and osteocytic dif-
formation may be phenotypically distinct or may even ferentiation. Similar patterns of expression in association
originate from another sublineage differentiation with later stages of osteoblastic differentiation are seen
pathway.42,64,65,103,107,108 In general, bone development and in reference to osteopontin, galectin-3, and CD44. The
its maintenance are regulated by conserved pathways production of collagen type I starts with the onset of dif-
linked to the interactive network of growth factors, regu- ferentiation and increases with the cell’s progression to a
lated by hormones, their respective receptors, and vita- mature osteocytic state.
mins.36,65 The major hormones and their receptors In summary, there is a reciprocal relationship between
involved in regulating bone development include para- two distinct groups of genes that predominantly act on
thyroid hormone, parathyroid hormone-like peptide, one another in a proliferative and postmitotic differenti-
thyroid hormones, and steroid hormones as well as ated stage. The high levels of histone proteins and other
vitamin D.109 In addition to contributing to the develop- genes related to the proliferative stage gradually approach
ment of the skeleton, these factors also control bone mass residual constituent levels or are completely suppressed
and bone maintenance during adult life.90,91,100,108,112 with the onset of osteoblastic differentiation (see the
The unique capacity of bone for complete structural section in this chapter on cell-cycle regulation). One of
and functional renewal depends on the presence of stem the earliest events that signifies the onset of osteoblastic
cells that can proliferate rapidly and differentiate into maturation is the upregulation of collagen type I genes,
different bone-forming cell lineages. It is postulated that followed by the upregulation of alkaline phosphatase and
the unique ability for renewal is in the bone itself.104,105 A matrix GLA protein. High levels of osteopontin, osteo-
unique class of matrix polypeptides, collectively desig- calcin, and collagenase are characteristic of the mineral-
nated as bone morphogenetic proteins (BMPs), is implicated ized phase of osteoblastic differentiation.
as potent early inducers of bone formation and cartilage
development, as well as of bone reconstitution after
injury.41,44,46-49,66,77,82,87 BMPs also seem to play a ubiqui-
Chondroblastic Lineage
tous role in skeletal patterning, limb development, and A number of ubiquitous growth factors have been
organogenesis of some nonskeletal structures.55,58,71,72 shown to mediate early phases of skeletal development.68,84
They show a significant sequence homology to the beta Their role in the regulation of skeletal growth and
family of transforming growth factors (TGFs).45-49,51,67 cartilage formation is graphically depicted in Figure
3-8.61,69,73,76,78,80,82,99 One of the transcription factors that
acts very early and is responsible for rescuing of pluripo-
Osteoblastic Lineage tential mesenchymal cells into skeletal progenitor cells is
Osteoblastic lineage cells, as they participate in bone SOX9.83,107 SOX9 is a member of the SOX gene family
production, are separated into several distinctive sub- of transcription factors characterized by high-mobility-
groups on the basis of their gene expression patterns, group box DNA binding motif showing homology with
ability to produce extracellular matrix, and their relation- sex determining factor SRY. Skeletal progenitor cells
ship to bone (Fig. 3-7). The development of osteoblastic with upregulated SOX9 and RUNX2, a Runt domain
lineage cells and bone production are regulated by transcription factor, are capable of bimodal differentia-
a complex cascade of growth factors, which are graphi- tion into chondrocytes and osteoblasts. The chondro-
cally depicted in Figure 3-8.37,38,53,60,61,63,68,69,100,112 Skeletal blastic fate of the skeletal progenitor cells is ensured by
progenitor cells that co-express SOX9 and RUNX2 direct interaction of SOX9 transcriptionally inhibiting
are not fully committed to osteoblastic lineage.56,83 It RUNX2 and indirectly by the repression of NKX3.2/
appears that the osteoblastic lineage fate in the early BAPX1. Collective in vitro and in vivo evidence indi-
phases of lineage commitment is controlled by the tran- cate that SOX9 activity is required for chondrocyte dif-
scription factor Osterix/SP7.54,81,107,110 It belongs to a ferentiation, but it requires cooperation with additional
super family of Kruppel-like zinc-finger transcription transcription factors such as PAX1 and PAX9, paired-box
factors and is expressed at high levels in osteoblasts. Low family members, and NKX3-2 and NKX3-1, homebox
levels of Osterix/SP7 are also found in prehypertrophic family members. In addition, such transcription factors as

ERRNVPHGLFRVRUJ
104 3  Molecular Biology of Bone Tumors

Multipotential
stem cell
Progenitors
Unlimited of other
renewal mesenchymal
cells

Limited
Osteoprogenitor
renewal
cells

Limited
proliferation Preosteoblasts

Mature osteoblasts
Bone-lining cells

Osteoblasts

Osteoid

Osteocyte
Limited
proliferation

FIGURE 3-7  ■  Differentiation of osteoblastic cells. Stages of differentiation and their location imply phenotypically recognizable
phases of osteoblastic lineage cells that originate from multipotential mesenchymal stem cells and, through osteoprogenitor and
osteoblastic cells, result in formation of mature osteocytic or bone-lining cells.

RUNX1, DLX5, and DLX6 as well as BARX2 have been process is again mediated by SOX genes, which further
implicated in chondrocyte differentiation. SOX9 has been signifies their central role in cartilage development. Mat-
shown to control expression of important cartilage matrix uration into hypertrophic chondrocytes is accomplished
proteins, including COL2A1, COL9A1, COL27A1, through NKX3-2 mediated downregulation of RUNX2
and matrilins. Cooperation with additional SOX family and the β-catenin/TCF complex. Downregulation of
members such as SOX5 and SOX6 are required to dif- RUNX2, IHH, and FGFR3 plays a role in the timing of
ferentiate chondroblasts into early chondrocytes. The the chondrocyte hypertrophic stage. Multiple regulatory
development of fully matured cartilage requires matu- loops control the sequential suppression of these factors
ration of chondroblastic/early chondrocytic cells into via the helix-loop-helix transcription factor, TWIST1,
prehypertrophic and hypertrophic chondrocytes. This and other cofactors, such as GRG5, HDAC4, DLX5,

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 105

Resting Proliferating Hypertrophic


chondrocyte chondrocyte chondrocyte Chondroclast Woven bone Blood vessel Periosteum
Lamellar
Cartilaginous Calcified bone
matrix cartilage Osteoblast Osteoclast

MMP9

VEGF

ANG1

PTC SMO PTHR PTHR

FGFR3 FGFR2
IHH LRP5

PTHrP FGF18 PTH PTHrP FGF18


WNT

RANK
OPG
RANKL

Primary spongiosa Secondary spongiosa


A
ATF4 FIAT

Sox8 HDAC3,7
SATB2

Osterix Runx2 Fra1 ATF4 Runx2 Sp3


Sox8 TWIST HDAC3,7

Dlx5
Runx2
Msx2 osteoprogenitor committed immature mature
osteoprogenitor osteoblast Msx2 osteoblast
osteocyte
MSX2nc Sox9 Sox9+
Runx2+

skeletal
precursor Sox9 GRG5 HDAC4
mesenchymal SOX9 Sox5/6
Pax1 Sox9
cell Runx3
Pax9 L-Sox5
Nkx3.1 Sox6 Atf2 -CATENIN Runx2 Runx2c Mef2 cMaf
Runx2 Fra2
Nkx3.2 Dlx5/6
Dlx
D 5/6
Nkx3.2

Sox9 chondroblast chondrocyte prehypertrophic Fgf18


F hypertrophic mineralized
Msx2 chondrocyte chondrocyte hypertrophic
p chondrocyte
Sox5, Sox6 Runx2
B
TWIST
FIGURE 3-8  ■  Molecular regulation of endochondral bone formation. A, Selected molecular regulations and their zonal effects stimu-
lating bone growth through endochondral bone formation. (A, Reprinted with permission from Goltzman D: Nat Rev Drug Discov
1:784–796, 2002.) B, Schematic summary of the activities of the most important transcriptional factors involved in differentiation
and maturation of the osteoblastic and chondroblastic cell lineages. Positive influences are indicated by green arrows and positive
factors are labeled in green. Negative influences are indicated by blunted red lines and negative acting factors are labeled in red.
(B, Reprinted with permission from Hartmann C: Curr Opin Genet Dev 19:437–443, 2009.)

ERRNVPHGLFRVRUJ
106 3  Molecular Biology of Bone Tumors

and DLX6. Additional factors such as MSX2, MEF2C, several activation-resorption cycles. It is speculated that
MEF2D, and FRA2 are likely involved in the regulation cessation of bone resorptive activities must be accom-
of the chondrocyte hypertrophic phase. Terminal matu- plished by programmed cell death (apoptosis).
ration and progression to apoptosis is not fully under- Surprisingly, receptors that mediate osteoclastic for-
stood at this time but appears to be regulated by the mation are present on osteoblastic cells.39,75,86,90-96,101
leucine zipper protein c-Maf. Chondrocytic cells express It appears that interaction with mesenchymal cells is
the genes such as collagen type II (COL2A1) that are important for early deposition for osteoclast progenitor
responsible for the formation of the cartilage matrix and cells.97 Three major signal transduction pathways,
the genes that contribute to the formation of the primary involving cyclic adenosine monophosphate, gp130, and
proteoglycans in the matrix, which are aggrecan, decorin, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], are present
biglycan, and fibromodulin.50,52,57,98 These glycoproteins in the plasma membrane of osteoblasts and mediate
are responsible for the osmotic properties of the matrix the formation of osteoclastic cells.75,86,106 These major
and its interaction with collagen II fibers, and provide transduction pathways mediate the formation of osteo-
the matrix with structural integrity.43,62,76,79,88 Extracellular clasts in response to various extracellular factors (Fig.
matrix synthesis is in part regulated by C-type natriuretic 3-9).31-35,59,89,90,94-96 Therefore the destruction of bone can
peptide (CNP) and the MAPK pathway of fibroblast proceed only in the presence of bone-forming cells (i.e.,
growth factor (FGF) signaling.64,111 matrix lysis occurs only when restoration or remodeling
of the structures is securely under way).
Osteoclasts are derived from mononuclear precursors
Osteoclastic Lineage that are of monocyte or macrophage hematogenous
Orchestration of skeletal development requires not only origin.40,60,91-93,106 These cells can proliferate and fuse to
bone production, but also resorption of mineralized form multinucleated osteoclasts. The transcription factor
matrix to achieve functionally optimal patterning of indi- PU.1 is essential for macrophage and osteoclast precur-
vidual bones.40,86,87 The cells in bone that perform this sor commitment to osteoclastic lineage. In addition, such
function and are capable of resorbing mineralized matrix transcription factors as c-Fos, NFκB, and microphthalmia-
are multinucleated osteoclasts (Fig. 3-9). It is generally associated transcription factor are essential for the matu-
accepted that osteoclasts develop by means of a differen- ration of osteoclasts. The cell-bound cytokine RANKL
tiation pathway distinct from that of osteoprogenitor and plays an essential role in activating the osteoclastic
cartilage progenitor cells. Numerous experimental data lineage. RANKL is a member of the tumor necrosis
indicate that osteoclasts are derived from hematopoietic factor family and interacts with its receptor RANK. Che-
precursor cells present in the bone marrow, spleen, and motactic stimulation of bone resorption is mediated via
peripheral blood.91-93,102 They have a common differentia- osteoblastic stromal cells, which can enhance RANKL
tion pathway with macrophages and develop distinct phe- expression. On the other hand, interferon-γ primarily
notypic features in the final steps of their maturation. produced by T-lymphocytes inhibits RANKL by causing
These cells, in response to chemotactic stimulation, degradation of the RANK-adapter protein TRAF6
migrate to areas of exposed matrix where the bone lining resulting in activation of NFκB and the kinase JNK.
cells have retracted and form sealing zones on the matrix
by interactions of αvβ3-integrins with matrix proteins.
Three of these receptors—, αvβ3 (fibronectin receptor), MECHANISMS OF MALIGNANT
α2β1 (collagen receptor), and αvβ1—are essential for TRANSFORMATION
adherence and subsequent bone resorption. Osteoclasts
secrete acid to the sealed pockets on the surface of bone Among the more than 100 transforming and tumor sup-
to dissolve the mineralized matrix and protein degrading pressor genes that have been identified, only a few have
enzymes to disintegrate exposed matrix proteins. These been shown to play a significant role in the biology of
functions are performed in a specialized apical cell mem- human neoplasia, including that of bone cancers.165,197,226
brane surface referred to as a ruffled border. Only a few The process of tumorigenesis is initiated by mutations
biochemical reactions can solubilize mineralized deposits that affect the so-called gatekeeping pathways, which
of the bone matrix (i.e., hydroxyapatite), and for these provide growth advantage.238 This process may represent
reactions to proceed, the environment must be acidic a single event or an acquisition of multiple alterations in
(low pH).60,106 In fact, sealed bone lacunae are acidic and a sequence. Many of these initiating events were identi-
reach this state by a V-type proton pump that uses ade- fied by studies that examined families with unique pre-
nosine triphosphatases (ATPases) that operate in the dispositions for certain tumor types. In many solid human
ruffled border of osteoclasts. The levels of these enzymes tumors such initiating events are still unknown. In this
are extremely high in activated osteoclasts. It can be envi- section, we discuss the biologic significance of a few
sioned that without the sealing of the resorptive lacunae, tumor suppressors and oncogenes that play a potential
the proton pump is very unlikely to be effective in role in the development of bone cancers. Thus the RAS
lowering the pH; thus solubilization of the mineral family of genes, TP53 genes, genes of the so-called
cannot be accomplished. After solubilization of the inor- p53regulatory pathway, and the retinoblastoma (RB) gene
ganic component, the remaining protein material is seem to play roles in the biology of many human neo-
degraded with the use of multiple proteolytic enzymes plasms. In addition, in this section we describe the role
such as collagenases, lysosomal proteinases, and cathep- of helicases in several bone cancer predisposing syn-
sins. There is evidence that osteoclasts can go through dromes, as well as the molecular biology of chromosomal

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 107

PTH IL-6 + sIL-6R


PGE2 IL-11
1α,25(OH)2D3 IL-1 (PGE2) OSM
LIF

Cell membrane
of osteoblasts

Adenylate
cyclase

gp130
1α,25(OH)2D3 receptor cAMP ATP
(VDR)

Osteoclastic
differentiation
factors

Osteoclasticc
precursor

Osteo lasts
Osteoblasts
Integrins’
Osteoclast
A receptor
N
N

N CO2 + H2O
Golgi
ATP
CAll*
ADP
Secretory Primary
vesicle lysosome H+ + HCO3-
Cl- Ruffled
*αγβ3-integrins* border

Cl-
Metallo-
proteinases Cysteine
proteinases H+
(cathepsin K)*,
phosphatases

Matrix proteins and hydroxyapatite

Bone

B
FIGURE 3-9  ■  Osteoclast differentiation and action. A, Three major signal transduction pathways that mediate formation of osteo-
blastic cells are present in cell membrane of osteoblastic cells. These pathways involve vitamin D receptor (1α,25[OH]2D3 receptor)
adenylate cyclase and cyclic adenosine monophosphate pathway and the gp130 protein. Pathways respond to variety of extracel-
lular signals that include 1α,25(OH)2D3; parathormone (PTH); prostaglandins (PGE2); IL-1, IL-6, and IL-11; and oncostatin M (OSM)
and leukemia inhibitory factor (LIF) factors. Bone lysis is mediated by integrin type receptors that seal bone resorptive lacunae. Bone
mineral is solubilized by V-type proton pump operating in ruffled border of osteoclastic cells. (A, Adapted from Suda T, et al: Cells
of bone: osteoblast generations. In Bilezikian JP, Raisz LG, Rodan GA, editors: Principles of bone biology, San Diego, 1996, Academic.)
B, Multinucleated osteoclasts (N) adhere to bone through αvβ3-integrins. Carbonic anhydrase II (CAII) can generate H+ and HCO−3.
HCO−3 is extruded in exchange for Cl− at the basolateral membrane. H− can be secreted through an H+-ATPase on the apical mem-
brane (ruffled border) into the resorption space and Cl− is secreted through a Cl− channel. HCl will dissolve matrix mineral. Phos-
phatases and cysteine proteinases—notably cathepsin K—will be released from lysosomes and degrade matrix proteins.
Metalloproteinases that are released from secretory vesicles will also degrade matrix proteins. Asterisks (*) indicate potential sites
for the inhibition of osteoclast action. (B, Adapted and reprinted with permission from Goltzman D: Nat Rev Drug Discov 1:784–796, 2002.)

ERRNVPHGLFRVRUJ
108 3  Molecular Biology of Bone Tumors

translocations that play an important role in the develop- regulatory role, causing overexpression of p21 and sig-
ment of many sarcomas. Finally, we address the possible nificantly increasing transforming efficiency. Thus a
role of a novel mechanism of cancer development referred mutation at position 2719 of the Ha-ras gene intron is
to as chromothripsis that may play a role in the develop- analogous in function to that of a strong promoter-
ment of a significant proportion of such important bone enhancer in that it causes overexpression of the gene
tumors as osteosarcoma and chordoma. Several lines of product. Although point mutations at coding sequences
evidence indicate that fully developed malignant pheno- alone are not essential for transformative activity, their
types result from the disruption of distinct regulatory presence enhances the transforming efficiency of overex-
circuits. These acquired capabilities of cancerous cells are pressed RAS gene products.
signified by the following key functional characteristics: Ras proteins bind GDP molecules in their inactive
(1) self-sufficiency in growth signals, (2) insensitivity to state and release the bound GDP molecules after being
antigrowth signals, (3) tissue invasion and metastases, (4) stimulated by the upstream regulatory partners and
limitless replicative potential, (5) sustained angiogenesis, absorb GTP molecules.243 The GTP bound state is
and (6) evasion of apoptosis.156,157 an activated configuration, and the molecule emits a
signal activating its downstream regulatory partners. The
active GTP binding state is transient and the protein
RAS Genes cleaves the bound GTP using its intrinsic GTPase func-
The human RAS gene family (Ha-ras, Ki-ras, and N-ras) tion and returns to an inactive nonsignal emitting state
has served as a prototype of cellular genes whose muta- (Fig. 3-11).
tions, overexpression, or both, can lead to malignant The mutant Ras proteins were found to have lost the
transformation (Fig. 3-10). These genes encode a group GTPase activity, abolishing its ability to return to the
of closely related 21 kDa proteins (p21) that belong to a inactive state. In such conditions, the proteins can be
super family of proteins that bind guanine nucleotides pushed to their signal emitting active states by the
with high affinity and have guanosine triphosphatase upstream regulatory partners, hyperactivating the down-
(GTPase) activity.116 The genes encoding these proteins stream pathways.
map to chromosomes 11, 12, and 1, respectively. The It is evident that Ras-regulated signaling pathways are
proteins encoded by these genes have a high degree of cascades of kinases that represent three important down-
homology and differ primarily in their hypervariable stream signaling pathways (Fig. 3-11).243 The main and
anchoring region. These proteins are bound to the cyto- originally discovered pathway is the Ras-Raf-MAP kinase
plasmic surface of the cell membrane and serve as trans- pathway. It represents a signature kinase signaling cascade
ducer molecules for signals that affect cell proliferation referred to as the mitogen-activated protein kinase (MAPK)
and differentiation. Two mechanisms have been proposed pathway. The activation of this pathway upregulates the
for RAS genes to transform cells. One involves a single so-called immediate early genes encoding such transcrip-
nucleotide mutation in the coding sequence that occurs tion factors as Fos and Jun. Together, these proteins form
most frequently at codon 12, 13, 59, or 61.120,141,149,213,233 the AP1 transcription factor, which is activated in many
This alteration results in an amino acid substitution of human tumors.
the gene product p21 that affects the GTP binding The second Ras-regulated pathway governs inositol
domain and reduces its GTPase activity. The second pos- lipids and Akt/PKB kinase. In this instance, the Ras pro-
sible mechanism involves overexpression of the normal teins activate phosphatidylinositol 3-kinase (PI3K) result-
RAS gene product. There is evidence that overexpression ing in downstream activation of Akt and Rho guanine
of an altered RAS gene product may play a role in the nucleotide exchange factor (RhoGEF), which results in
biology of some human tumors. the stimulation of several important downstream path-
It has been shown that introns play a regulatory role ways that affect cell proliferation and apoptosis, including
in the function of RAS genes. The DNA sequence around mTOR.
nucleotide 2719 within intron D functions as an alterna- The third Ras-regulated downstream pathway involves
tive splicing site.126,127,130 It suppresses gene expression by RalGEF, which includes the distant cousins of Ras pro-
destabilizing the transcript and causing it to enter a non- teins referred to as Ral-A and Ral-B. Similar to the pro-
productive pathway. From the functional point of view, totypic Ras proteins, activation of Ral proteins involves
the intron D splicing-recognition sequence acts as a neg- the GDP-GTP switch. The activated Ral-A and Ral-B
ative regulatory element by suppressing gene expression upregulate multiple downstream partners, including
and presumably protecting the cell from the eventual Sec5 and Exo84, which promote anchorage-independent
transforming activity of the mutated (altered) gene growth.
product. As a result of the alternative splicing pathway, a Activation of RAS genes by point mutation occurs in
minimal amount of a different gene product, 19 kDa approximately 15% to 20% of some common epithelial
(p19), may be produced. It is possible that this protein malignancies.116,118,149 Rearrangement of RAS genes has
acts as an antioncogene and protects the cell from the been sporadically identified in some soft tissue sarcomas.
effects of the oncogene. Hence Ha-ras appears to repre- In general, activation of the RAS gene by point mutation
sent a gene capable of encoding two antagonistic prod- occurs relatively infrequently in human cancers and espe-
ucts: one with transforming capability and the other cially in bone sarcomas, but it plays a major role in the
with antioncogenic (tumor suppressor gene) activity. A development of gastrointestinal malignancies, especially
mutation at position 2719 of intron D (A–T point muta- colon cancer.141,211 Experimental transfection of the RAS
tion) abolishes the alternative splicing pathway and its gene suggests that activated RAS genes can alter the

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 109

Chr 12
13.3 13.2
KRAS
13.1

Exon1

Exon2

Exon3
Exon4

Exon5

Exon6
12.3

cds
12.2 12.1
11.2 KRAS
11 11.1
12
13.1
13.3 13.2

11
14
15 KRAS

18
21.1

6
21.2
21.3

6
24.902

132
22

6
4.365
23

497

133
25

24

28

15
12
24.1
6

4
5

5
5

5
24.2
24.31 24.32
24.33
188/9 aa
A 1

Chr 11
15.5
HRAS
15.4
HRAS
15.3
Exon1

Exon2

Exon3

Exon4

Exon5

Exon6
15.1 15.2

cds
14
13
12
11.2
11.11 11.12
11
12
13.1
13.2
13.4 13.3
13.5
HRAS
237
423

402

14.1
26

4
14.2
14.3
5

21
22.1 22.2
22.3
23.1
23.2
23.3 1 188/9 aa
24
25

NRAS
Chr 1
Exon1

Exon2

Exon3

Exon4

Exon5
Exon6

Exon7
cds

36.3
36.2
36.1
35
34.3
34.2
34.1
33
32.3
4

32.2
NRAS
4
4

32.1
31.3
2287

31.2
877
424

13

31.1
5
9

5
5

22.3
22.2
22.1
21 1 188/9 aa
13.3
13.2
13.1
11
NRAS Small GTP binding protein domain 1-159
12
11 P-loop containing nucleoside triphosphate hydrolase domain 3-164 Linker
12 Ancor
Hypervariable region 165-188/9
21.1
21.2
21.3 Missense substitution
22
23
Nonsense substitution - Stop K-ras KKKKKKSKTK
KKKKKKS CVIM
24 Insertion inframe H-ras GCMSCK
C C CVLS
25 Deletion inframe
Insertion frameshift N-ras GCMGLP
C CVVM
31 Deletion frameshift
Complex - deletion inframe CAAX
32.1
32.2
Unknown motif
32.3 Substitution - coding silent
41
42.1
Complex - frameshift
42.2
43
42.3 Complex - compound substitution
44 Complex
C
FIGURE 3-10  ■  Chromosomal location, exonal structure and functional domains of the RAS genes. A, Chromosomal location and
exonal structure of the K-ras gene. The functional domains of the encoded protein and the distribution of mutations are shown.
B, Chromosomal location and exonal structure of the H-ras gene. The functional domains of the encoded protein and the distribu-
tion of mutations are shown. C, Chromosomal location and exonal structure of the N-ras gene. The functional domains of the
encoded protein and the distribution of mutations are shown.

ERRNVPHGLFRVRUJ
110 3  Molecular Biology of Bone Tumors

upstream stimulatory
INACTIVE signal and Ras activation
GTP triggered by GEF
hydrolysis Ras
Switch I and Ras Pi
inactivation
induced GDP GEF
by GAP

GAP GTP

GDP

Switch II
blockage
caused by Ras
oncogenic
mutation GTP downstream
A B C signaling
ACTIVE

Ras

GTP

PI3K Raf (MAPKKK) Ral-GEFs

PIP3
MEK (MAPKK) RalA RalB

Akt/PKB Rho-GEFs
Erk1 or 2 (MAPK)
Sec5 Exo84 RalB

Cdc42 Rac
Bad mTOR GSK-3β FOXO Msk1 RSK Mnk1 Ets Elk-1 SAP-1
inhibition
of apoptosis stimulation stimulation cell-cycle chromatin
of protein of cell progression; remodeling (transcription) filopodia lamellipodia
synthesis proliferation inhibition of
(cell growth) apoptosis elF4E
(protein synthesis)
D
FIGURE 3-11  ■  The structure and biologic effects of the Ras protein. A, The ribbon diagram of the structure of a Ras protein as
determined by x-ray crystallography as it interacts with the guanosine 5′-triphosphate (GTP) molecule. The α-helical part is in red
and β-pleated polypeptide is in green. The GTP molecule is shown as a stick figure and the two amino acid residues that are often
mutated (glycine 12 and glutamine 61) are highlighted. B, The substitution of GDP by GTP resulting in the activation of RAS signal-
ing causes a conformational shift in the two regions of the Ras protein that interact with the GDP-GTP molecules. The conformational
shift activating the protein is indicated by the magenta arrows. The shift facilitates the interaction with the downstream effectors
activating the pathways. (A and B, Courtesy of A. Wittinghofer.) C, The Ras signaling cycle shows that the Ras proteins operate as
switches binding GDP (inactive state) and GTP (active state) in which they emit signals activating the downstream effectors. The
inactive GTP binding protein is stimulated by GEF (guanine nucleotide exchange factor) and they release the GDP in exchange for
a GTP. The active state is very transient and the signaling is halted by the GTPase activity intrinsic to the Ras protein, which hydro-
lyzes GTP to GDP. The GTPase activity is stimulated by GTPase activating proteins (GAP). D, The signaling pathways activated by
the Ras protein. The Ras-Raf-MAP kinase pathway, depicted in pink, represents the main signaling cascade activated by the Ras
proteins. In addition, the two alternative kinase pathways, referred to as the PI3 and Ral pathways, depicted in lavender and blue,
respectively, are shown. (C and D, Modified and reprinted with permission from Weinberg RA: The biology of cancer, 2d ed, New York,
2014, Garland Science.)

tumorigenicity of osteosarcoma cells.119,121 The increased TP53 Gene


tumorigenicity of Ras-transformed osteosarcoma cells
may be related to the activation of metalloproteinases. The TP53 gene, located on the short arm of chromosome
Analysis of a larger series of human osteosarcoma, chon- 17, has been a constant source of fascination since its
drosarcoma, and Ewing’s sarcoma indicates that the acti- discovery more than two decades ago (Fig. 3-12).186,193
vation of RAS genes by point mutation does not play a The gene product was originally identified as a 53 kDa
major role in the pathogenesis of these tumors.114 On the nuclear phosphoprotein in cells transformed by simian
other hand, upregulation of Ras regulatory pathways is a virus 40 (SV40).186 The protein formed a complex with
common event in many human tumors, including bone the large T antigen of SV40, suggesting that this interac-
cancers. tion was important for transformation. The insights

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 111

TP53
Chr 17

Exon 10

Exon 11
Exon 1

Exon 2
Exon 3
Exon 4

Exon 5
Exon 6

Exon 7
Exon 8
Exon 9

cds
13 TP53
12
11.2
11.1 11.1
11.2
12
21.2 21.1
21.3 P53
22
1 393 aa
23
24
6 29 94 297 318 359
25 p53 transactivation domain
p53/RUNT-type transcription factor, DNA-binding domain
A p53, tetramerisation domain

C termini
Checkpoint kinases

ATP ADP
Mdm2 P
P
p53 p53
Tetramerization domain
Degradation
P P
P P

Flexible linker region p53 p53


Core domains DNA

Cdk p21 Puma


inhibitor
Inhibits

Cdk
Bcl-2
Cyclin
N termini (TAD) Cannot Cannot
with bound Taz2 phosphorylate inhibit
Rb protein apoptosis

B
CELL CYCLE APOPTOSIS
(arrest) (cell death)

C
FIGURE 3-12  ■  The structure and biologic effects of the TP53 gene and its encoded protein. A, Chromosomal location, exonal struc-
ture, and functional domains of the encoded protein of the TP53 gene. B, Model of a full length p53 protein forming the ternary
complex with cognate DNA. The p53 DNA-binding domains are shown in blue and green, and the tetramerization domain is shown
in red. (B, Modified and printed with permission from Joerger AC, et al: Cold Spring Harb Perspect Biol 1–20, 2010.) C, p53 DNA-binding
motif. (C, Modified and printed with permission from Smeenk L, et al: Nucleic Acids Res 36:3639–3654, 2008.) D, p53 regulatory pathway.

concerning the function of p53 in cellular physiology and activity.143,188 Because of the gain of a new function, the
malignant transformations were provided by two main gene can act in the heterozygous state and does not
lines of investigation. The first group of studies involved require allelic deletion for its transforming activity. p53
the biologic effects of altered (mutant) TP53 gene in is a DNA-binding protein and a transcription activator
model organisms and tumor cells grown in culture. The factor capable of inducing so-called p53-dependent genes.186
second group of studies concerned biochemical and Biochemical and structural analyses of the p53 protein
structural analyses of the p53 protein. It was shown that showed that it functions as a transcription factor that
the transforming activity of TP53 is related to gene point exists in normal cells as a homotetramer (Fig. 3-12).170
mutations and that the mutated protein has a much Chromatin precipitation studies have revealed that it
longer half-life than the wild-type gene product. More- binds to a specialized DNA sequence. It appears that
over, it was established that the wild-type protein there are more than 1500 p53 binding sites in the human
acts more like a tumor suppressor gene, negatively regu- genome, confirming that p53 in both cell physiologic
lating the cell cycle by loss of function, gain of a new and malignant stages can control the expression of a
function, or both capacities to perform its transforming vast of genes capable of significantly modifying cell

ERRNVPHGLFRVRUJ
112 3  Molecular Biology of Bone Tumors

behavior.191,224 Because a mutant p53 protein frequently cancer-prone families, especially those with Li-Fraumeni
acts in a heterozygotic state, it is very likely that tetra- syndrome. The functional domains of TP53 and the dis-
meric complexes contain a mixture of mutant and wild- tribution of mutation sites in human tumors are shown
type polypeptide subunits. in Figure 3-12.
It is evident that, under normal conditions, the p53 The TP53 gene is one of the most frequently altered
protein is unstable and cells have to continuously synthe- genes in various human cancers.201 In various common
size new molecules at a high rate and degrade them with epithelial malignancies, its overexpression being associ-
the same efficiency. The system permits a rapid increase ated with an altered gene can be documented in up to
of p53 by simply blocking its degradation. In normal 50% of cases. It is typically associated with a more aggres-
cells, the p53 protein level is controlled by a complex sive variant of the tumor. The altered gene, overexpressed
system in which MDM2 plays an essential role. The p53- p53 protein, or both are frequent findings in osteosar-
MDM2 complex triggers the degradation of p53 by ubiq- coma and chondrosarcoma.122,135,136,211,237,245 Mutations of
uitination. This identifies an alternative mechanism of the TP53 gene are infrequently present in Ewing’s
p53 loss of function by hyperactivated MDM2, which is sarcoma, and its role in the pathogenesis of this tumor
frequently amplified and overexpressed in sarcomas. and its clinical significance are not well established.176,178
Another mechanism that affects MDM2 has been identi- Overexpression or alterations of p53 can be documented
fied by the discovery of a gene that is antagonistic to in approximately 30% of high-grade conventional osteo-
MDM2 termed p16INK4A.139,222 The upstream promotor sarcomas. As in epithelial malignancies, altered TP53 is
and alternative splicing program of the p16INK4A produces associated with highly aggressive variants of osteosar-
a different protein referred to as p19ARF and in humans coma.212 In a small percentage of cases, alterations of
p14ARF, which acts as a strong inhibitor of MDM2. Over- TP53 are associated with the amplification of the MDM2
expression of ARF by inhibiting MDM2 increases p53 gene.189,198 The amplification of the MDM2 gene is more
levels. Conversely, the mutations involving the p16INK4A frequent in metastatic osteosarcoma.180,198
locus, observed in many human tumors, represent another Alteration of the TP53 gene and overexpression of
mechanism contributing to the p53 loss of function. p53 protein can be documented in approximately 30%
Phosphorylation of the MDM2 binding domain is yet of conventional chondrosarcomas of bone. It is pre­
another mechanism controlling the p53 protein levels dominantly present in high-grade, clinically aggressive
which is mediated by a number of kinases including the tumors.136 It can be identified in a majority of grade 3
family of Aurora kinases. These mechanisms position the tumors and in approximately half of grade 2 tumors. In
p53 protein and its function centrally in controlling cell chondrosarcomas, the presence of altered TP53 or over-
survival, proliferation, genetic stability, and differentia- expression of its protein is accompanied by allelic dele-
tion signifying its key role in malignant transformation. tions in the TP53 locus. In chondrosarcoma, the presence
The mutations that change the properties of TP53 are of altered TP53 correlates with an aggressive clinical
usually missense substitutions.163,239 They are clustered in variant (high propensity for metastases as well as shorter
one particular region of the gene product located between disease-free and overall survival times). The presence of
amino acids 130 and 290 and mainly in the residue of p53 overexpression in virtually all cases of dedifferenti-
amino acids 117 through 142, 171 through 181, 239 ated chondrosarcoma studied, and in which it is restricted
through 258, and 270 through 286.186 These regions are to the high-grade sarcomatous component of the tumor,
highly conserved among species and are most likely suggests a critical role of this gene in the development of
important to the normal function of TP53. The human high-grade secondary malignancies of bone.
TP53 gene is located on chromosome 17p13.1, and muta-
tions have been documented in a wide range of tumors.
Loss of heterozygosity of the TP53 locus has been found
RB Gene
in many human cancers. Therefore TP53 most likely The RB gene was originally identified as a gene of reti-
contributes to human carcinogenesis when its normal noblastoma, a rare intraocular malignant neoplasm of
allele is deleted or inactivated. The transforming activity early childhood. It was the first human tumor suppres-
in the heterozygous state can be the result of the forma- sor gene (i.e., a gene whose loss of function leads to the
tion of an oligomeric complex between mutant and wild- development of malignant neoplasms) discovered.153,248 In
type TP53. It has been shown that in cells transformed humans, germ line alterations of the RB1 gene represent
with the mutant TP53 gene, the altered protein com- the prototypic genetic predisposition to cancer. The main
plexes with endogenous TP53, and the complex remains tumor associated with this predisposition is retinoblas-
in the cytoplasm.192 In general, mutations affecting dif- toma. The tumor is composed of poorly differentiated
ferent portions of the gene may result in the formation cells that form rosettes and are referred to as retino-
of the gene product with distinct biologic activity.155 blasts. The survivors of childhood retinoblastoma carry
Mutated TP53 genes can cooperate with RAS genes to an increased lifetime risk of secondary cancers, which
transform primary cultured fibroblasts in the presence of include osteosarcoma, fibrosarcoma, chondrosarcoma,
endogenous wild-type p53 protein. Because deletions of Ewing’s sarcoma, melanoma, leukemia and lymphoma,
chromosome 17 loci may occur simultaneously with brain tumors, and a long list of common epithelial
other chromosomal abnormalities in certain human solid malignancies such as bladder cancer. The RB gene
tumors, it has been suggested that the TP53 mutation is codes for a 105 kDa protein that belongs to a family of
a late event during carcinogenesis. Germline transmis- three proteins, including RBL2/p130 and RBL1/p107
sion of TP53 mutations has been described in certain (Fig. 3-13).166,218 These proteins are structurally and

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 113

RB1

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17

Exon 18
Exon 19
Exon 20
Exon 21
Exon 22
Exon 23

Exon 24
Exon 25
Exon 26
Exon 27
Chr 13

Exon 1
Exon 2

Exon 3
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8
Exon 9

cds
13
12
11.2
11 11.1
12.1
12.2
12.3
13 14.1
14.2 RB1
21.1 14.3 RB1
21.3
21.2 638 786
1 928 aa
22
31
111 228 373 576 768 927
32
33 Retinoblastoma-associated protein, N-terminal domain
34
Cyclin-like – Retinoblastoma-associated protein, A-box domain
Cyclin-like – Retinoblastoma-associated protein, B-box domain
Retinoblastoma-associated protein, C-terminal
A

Growth factor

Ras pathway
P
Rb
P

Rb Cdk
Cyclin
E2F E2F

Genes needed ATP ADP


for S phase are
not transcribed Gene transcription

mRNA translation

Enzymes and other


proteins required
for S phase
D

B
FIGURE 3-13  ■  The structure and biologic effects of the RB1 gene and its encoded protein. A, Chromosomal location, exonal structure,
and the functional domains of the encoded protein of the RB1 gene. B, The model of a full length RB1 protein forming the complex
with cognate DNA. C, The RB1 DNA-binding motif. D, RB1 regulatory pathway. (B, Modified and printed with permission from Rubin
SM, et al: Cell 123:1093–1106, 2005 and Hassler M, et al: Molecular Cell 28:371–385, 2007. C, Based on Marinescu VD, et al: Nucleic Acids
Research 33:91–97, 2005, and Marinescu VD, et al: BMC Bioinformatics 6:1–20, 2005.)

functionally very similar to RB1/p105 and control the transported to the nucleus by interaction with RBL1/
same pathways but may show distinctive functions in p107 and RBL2/p130. Similar to p53, the function of all
specific tissue environments.133,160 The most studied and, Rb proteins are modulated by phosphorylation mediated
likely, the most significant biologic effect of Rb proteins by cyclin-dependent kinase-cyclin complexes. Hyper-
is their regulation of cell cycle and inhibition of G1-S phosphorylation of Rb proteins inhibits binding to E2F
phase transition.132,137,230 They interact with transcription proteins and releases cell cycle progression. A number
factors such as E2F1, E2F2, and F2F3a, which are the acti- of stimuli can reactivate Rb proteins, including the
vators of transcriptions inhibited by RB1/p105.191,218,219 In induction of cyclin-dependent kinase inhibitors such as
contrast, E2F4 and E2F5 are repressors of transcriptions CIP/KIP family members, including p21, p27, and p57

ERRNVPHGLFRVRUJ
114 3  Molecular Biology of Bone Tumors

and INK4 family members such as p15, p16, p18, and their unique role in cancer development, including
p19. In addition to the well-known function of Rb pro- sarcoma predisposing syndromes such as Rothmund-
teins as regulators of cell cycle progression, emerging Thomson, Baller-Gerold, RAPADILINO, and Werner
evidence postulates that they are involved in chromo- syndromes (Fig. 3-14).117,125,244 Helicases are defined as
somal stability, senescence, apoptosis, differentiation, and molecular engines that are coupling nucleoside triphos-
angiogenesis, all of which may contribute to malignant phate hydrolysis, typically of ATP, to unwind the double
transformation.174 helix of nucleic acids.124,200,210,221 These enzymes move
In retinoblastoma, the RB gene initiates tumorigenesis along the double strand directionally from 5′ to 3′ or in
when both alleles are altered in retinal cells.184 In other reverse direction. They catalytically disrupt base pairs
types of tumors, loss of a functional Rb protein may have between complementary strands of DNA in a manner
a similar mechanism, or the RB gene product may be that is dependent on ATP. They also participate in anneal-
inhibited by other mechanisms, such as the overexpres- ing of the double-stranded DNA by promoting pairing
sion of cyclin D1.150 Human patients with one altered RB of the base pairs. In general, these enzymes help cells to
allele are normal except that they have an increased risk recover from both endogenous and exogenous stress-
of developing cancer. Their risk for the development of induced DNA damage. Therefore, it is understandable
retinoblastoma is 36,000 times that of people without this that their impaired function by mutations causes unique
mutation. Human patients with germline RB mutations clinical syndromes characterized by genomic instability
also have 2000 times the normal risk for osteosarcoma and sensitivity to environmental DNA damage such as
and an undefined but increased risk for other tumors, sun exposure, resulting in premature aging and an
predominantly melanoma and soft tissue sarcomas. increased risk for cancer.128,206 There are nearly 100
Alterations of the RB gene in human cancers are of known helicases and an additional 30 putative helicases
two main types: deletions and mutations (see Fig. 3-8). encoded by the human genome.117,181 The list of helicases
The first consists of major deletions of large segments of whose defects have been linked to unique clinical syn-
the gene, which cause the absence of a properly function- dromes and increased risk for cancer are provided in
ing gene product. The second consists of substitutions of Table 3-1.
nucleotides, which alter the function of the gene by creat- The two helicases on which we focus in this section
ing improperly located initiation signals, splicing sites, are referred to as RECQL4 and WRN helicases, whose
stop codons and amino acid substitutions.135,147,150,240 They mutations are associated with the predisposition to a
destabilize the transcript, produce a truncated gene number of solid and hematologic malignancies, including
product, or otherwise modify mRNA, causing the absence osteosarcomas. RECQL4 plays a role in several DNA
of a functional Rb protein. repair pathways, which are mediated by its specific
The RB gene represents a model tumor suppressor helicase domain.134 Mutations of RECQL4 have been
gene with frequent alterations in a variety of common identified in three distinct cancer predisposing syn-
human malignant neoplasms.129,164,207 It serves as a para- dromes: as Rothmund-Thomson, RAPADILINO, and
digm of molecular alterations that predispose the human Baller-Gerold.168
body to the development of osteosarcoma as the second The WRN helicase gene on chromosome 8p12 encodes
malignancy in families affected by retinoblastoma. Similar RecQ helicase, which possesses a helicase and DNA exo-
to TP53, the RB gene is also frequently altered in sporadic nuclease domains.148 The gene is mutated in the Werner
osteosarcomas not associated with retinoblastoma.234,235,240 syndrome, which is a mendelian autosomal recessive dis-
The alterations of RB in sporadic osteosarcoma are order manifested by features of accelerated aging and
similar to those seen in retinoblastoma when the retino- increased risk for multiple malignancies, including osteo-
blastoma is associated with osteosarcoma (allelic loss and sarcoma.155,227 The WRN helicase functions as a key
functional absence of the protein as a result of altered regulator of recovery from replication arrest at replica-
remaining alleles).115,235 The absence of the RB gene tran- tion forks and acts at the replication checkpoint. The
script or the presence of its truncated fragments is a basic biologic effect of malfunctioning WRN helicase is
frequent finding in sporadic osteosarcomas. The pres- a lack of recovery from arrested replication forks, result-
ence of an altered RB gene and the presence of allelic ing in chromosomal rearrangements.208,217,228
deletions in the RB locus are associated with more aggres-
sive variants of osteosarcoma.115,142 Loss of heterozygosity
in the RB gene locus has also been documented in chon-
Chromosomal Translocations
drosarcomas and chordomas.138 The significance of recurrent chromosomal transloca-
tions to understanding the biology of sarcomas and espe-
cially their use in the differential diagnosis of these
DNA Helicases malignancies warrants a more detailed discussion.337 The
Genome maintenance is a complex physiologic mecha- transfer of one fragment of a chromosome to another
nism that involves multiple proteins that are essential for chromosome may put two genes together in one place.
the preservation of the structural and functional integrity This in turn can fuse the two coding sequences near the
of cellular genetic material. This system involves multiple breakpoint, creating a hybrid messenger RNA (mRNA)
genes and their respective encoded proteins organized in coding for a new protein, which can have a profound
several closely interacting regulatory pathways. In this effect on cell biology.179,195,196 The biologic effect of the
paragraph, we describe the role of one particular class of hybrid gene results from overexpression of a fused protein
enzymatic proteins, referred to as helicases because of driven by a new strong promoter or may be related to a

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 115

RECQL4

Exon 10
Exon 11
Exon 12

Exon 13
Exon 14
Exon 15

Exon 16

Exon 17
Exon 18

Exon 19
Exon 20

Exon 21

Exon 22
Exon 1
Exon 2
Exon 3

Exon 4

Exon 5

Exon 6

Exon 7
Exon 8
Exon 9
cds
Chr 8
23.3 23.2
23.1 tel cen
c.118+27del25
22
21.3 21.2 c.84+6del16 c.1048_1049AG
21.1
12 c.1391-1g>A c.1390+2delT c.1878+5G>A
c.2492-2493delAT
11.2 11.1 c.1483+25del11 c.1483+27del11 c.1878+32del24
11.1 c.2506_2518del13bp
11.22 11.21 c.1885del4
11.23 c.1568delG c.1573delT c.2547-2548delGT
12 c.1887del4
13 c1650del7 c.2059-1G>A c.2552delC
21.1 c.2059-1G>T c.2886-2A>T
c.1704G>A c.1705-1G>A
21.2 c.3056-2A>C
21.3 c.1718delA c.2207insC
c.3072delA
22.1 c.2335del22
22.2 c.3072-3073delAG
22.3 Rothmund-Thomson Syndrome c.2419ins5
23 c.3270delG
RAPADILINO Syndrome c.2464-1G>C
c.3276delG
24.1
Baller-Gerold Syndrome c.3599_3600delCG
24.2 24.3
RECQL4
RECQL4
492 820
1 1208 aa
402418 477 677 683 850
p.Gln166X p.Gln253His p.Trp383X p.Pro466Leu
p.Phe637Ser p.Leu926Arg
p.Leu638Pro p.Leu927Arg
A p.Phe697Leu
p.Ala741Thr
p.Arg1021Trp
p.Ile1051Val
p.Gln757X#
p.Arg1072X
p.Gln800X
p.Gln810X p.Gln1091X
p.Gln821X p.Pro1170Leu
p.Arg826X# p.Gln1175X

DNA
RecA-like domain 2
replication 3’
P466L 5’
F697L 5’
5’ 3’
3’ 5’
flap Homologous
recombinatiom D-loop
F637S

5’ Telomere 3’
3’ 5’
maintenance
5’
RecA-like domain 1
3’
fork Holliday junction
B DNA repair
C
FIGURE 3-14  ■  The structure and biologic effects of the RECQL4 helicase gene and its encoded protein. A, Chromosomal location,
exonal structure, and the functional domains of the encoded protein of the RECQL4 helicase gene. The distribution of mutations in
the Rothmund-Thomson, RAPADILINO, and Baller-Gerold syndromes are superimposed over the gene and protein diagrams.
(A, Adapted and reprinted with permission from Larizza L, et al: Orphanet J Rare Dis 5:1750–1172, 2010.) B, The crystal structure of
RECQL4. The important motifs are highlighted in yellow and the RecA-like domains 1 and 2 are indicated. Labels indicate the
examples of mutations in the protein. (B, Reprinted with permission from Jensen MB, et al: Aging (Albany NY) 4:790–802, 2012.) C, The
inferred RECQL helicase substrates and their roles in DNA metabolism. RECQ helicases are able to unwind and release DNA flaps
(upper left); promote replication fork progression, regression, or remodeling (lower left); release an invading 3′ DNA tail in a D-loop
(upper right); and branch migrate and, in the case of BLM in conjunction with topoisomerase IIIα, resolve separate DNA duplexes
joined in a Holliday junction (lower right). (C, Reprinted with permission from Monnat RJ Jr: Semin Cancer Biol 20:328–339, 2010.)

new function of a protein that does not exist in normal in bone and soft tissue, including the prototypic translo-
cells. The molecular structures of the vast majority of cations in Ewing’s sarcoma and related lesions, result in
cytogenetically detectable translocations have been iden- the production of a new fusion transcript that consists of
tified. New translocations continue to be discovered for a portion of a gene that has an RNA binding domain
rare variants or for cytogenetically silent translocations (EWSR1) replaced by another gene exhibiting overall
or those occurring in the background of complex aber- biologic activity of the transcription factor (FLI1, ERG,
rant karyotypes by genome profiling and sequencing WT1).113,131,151,185,194,216,223,241 Moreover, it seems that the
techniques.167,173,209,232,242 The list of currently known EWSR1 gene is fused with other genes that code for
translocations in both soft tissue and bone sarcomas is DNA binding molecules in several distinct tumors appar-
provided in Table 3-2. Elucidation of the molecular ently not related to Ewing’s sarcoma. Thus the translo-
structures of breakpoints and identification of the genes cational patterns of this gene represent a unique model
affected by the translocations in many bone or soft tissue of molecular mechanism associated with several distinct
malignancies have revealed some striking functional simi- types of bone and soft tissue sarcomas (Fig. 3-15). It is
larities. It appears that a majority of translocations found evident that the same fusion partner can be involved in

ERRNVPHGLFRVRUJ
116 3  Molecular Biology of Bone Tumors

TABLE 3-1  DNA Helicase Gene Defects Linked to Cancer


Genome Metabolic
Gene Disease Cancer Type Pathway Biochemical Function
WRN Werner syndrome Thyroid neoplasms, DSB repair and 3′-5′ helicase, 3′-5′
melanomas, replication distress exonuclease, HJ
meningiomas, sarcomas, response branch migration, G4
hematologic and resolution, replication
lymphoid neoplasms, fork regression, and
and osteosarcoma strand annealing
BLM Bloom syndrome Adult epithelial tumors, DSB repair and repair of 3′-5′ helicase, HJ branch
leukemias, lymphomas, replication-associated migration, G4
and rare pediatric DNA damage resolution, replication
tumors fork regression, and
strand annealing
RECQL4 RTS, BGS, and Osteosarcomas and Replication, mitochondrial 3′-5′ helicase and strand
RAPADILINO syndromes lymphomas genome stability, and annealing
repair of endogenous
base damage
RECQL1 Pancreatic cancer Replication and oxidative 3′-5′ helicase, HJ branch
(polymorphisms) DNA damage response migration, and strand
annealing
FANCJ FA Acute myeloid leukemia DSB repair and ICL repair 5′-3′ helicase and G4
and breast cancer resolution
(heterozygotes)
XPD Xeroderma pigmentosum, Skin cancer NER and transcription 5′-3′ helicase
xeroderma pigmentosum
with Cockayne syndrome,
TTD, and COFS
XPB Xeroderma pigmentosum, Skin cancer NER and transcription 3′-5′ helicase
xeroderma pigmentosum
with Cockayne syndrome,
and TTD
RTEL1 Dyskeratosis congenita Adult glioma Telomere maintenance 5′-3′ helicase and
(polymorphisms) and homologous disassembly of
recombination D-loops and T-loops
PIF1 Breast cancer Replication fork 5′-3′ helicase and G4
predisposition progression, telomere resolution
maintenance, and
mitochondrial DNA
metabolism

BGS, Baller-Gerold syndrome; COFS, cerebro-oculo-facial-skeletal syndrome; D-loop, displacement loop; DSB, double-strand break;
FA, Fanconi anemia; G4, G-quadruplex; HJ, Holliday junction; ICL, interstrand crosslink; NER, nucleotide-excision repair; RAPADILINO
syndrome involving radial hypoplasia/aplasia, patellar hypoplasia/aplasia and cleft or highly arched palate, diarrhea and dislocated joints,
little size (height at least 2 standard deviations smaller than average) and limb malformation, nose slender and normal intelligence;
RTS, Rothmund-Thomson syndrome; T-loop, telomeric displacement loop; TTD, trichothiodystrophy.
From Brosh RM Jr: Nature Reviews Cancer 13:542–558, 2013.

TABLE 3-2  Specific Genetic Aberrations in Bone and Soft Tissue Tumors
Neoplasm Translocation Involved gene(s)
Ewing’s sarcoma/PNET t(11;22)(q24;q12) EWSR1-FLI1
t(21;22)(q22;q12) EWSR1-ERG
t(7;22)(p22;q12) EWSR1-ETV1
t(17;22)(q12;q12) EWSR1-ETV4
t(2;22)(q33;q12) EWSR1-FEV
t(16;21)(p11;q22) FUS-ERG
t(2;16)(q35;p11) FUS-FEV
Ewing’s sarcoma-like tumors t(6;22)(p21;q12) EWSR1-POU5F1
without ETS involvement Inv(22)(q12) EWSR1-ZSG (ZNF278)
t(4;22)(q31;q12) EWSR1-SMARCA
t(2;22)(q31;q12) EWSR1-SP3
t(1;22)(p36.1;q12) EWSR1-ZNF278
Complex ring chromosome with EWSR1-NFATc2
amplification of the translocated segments
t(4;19)(q35;q13) CIC-DUX4
Inversion involving Xp11.4; Xp11.22 BCOR-CCNB3

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 117

TABLE 3-2  Specific Genetic Aberrations in Bone and Soft Tissue Tumors—cont’d
Neoplasm Translocation Involved gene(s)
Angiomatoid fibrous histiocytoma t(2;22)(q34;q12) EWSR1-CREB1
t(12;22)(q13;q12) EWSR1-ATF1
t(12;16)(q13;p11) FUS-ATF1
Clear cell sarcoma t(12;22)(q13;q12) EWSR1-ATF1
t(2;22)(q34;q12) EWSR1-CREB1
Desmoplastic small round cell tumor t(11;22)(p13;q12) EWSR1-WT1
Extraskeletal myxoid t(9;22)(q22;q12) EWSR1-NR4A3
chondrosarcoma t(9;17)(q22:q11) TAF15-NR4A3
t(9;15)(q22;q21) TCF12-NR4A3
t(3;9)(q12;q22) TFG-NR4A3
Myoepithelioma, soft tissue t(19;22)(q13;q12) EWSR1-ZNF444
t(1;22)(q23;q12) EWSR1-PBX1
t(6;22)(p21;q12) EWSR1-POU5F1
t with 16p11 FUS
Myxoid/round cell liposarcoma t(12;16)(q13;p11) FUS-DDIT3
t(12;22)(q13;q12) EWSR1-DDIT3
Aneurysmal bone cyst t(16;17)(q22;p13) CDH11-USP6 (Tre2)
t(1;17)(p34.1-34.3;p13) TRAP150-USP6
t(3;17)(q21;p13) ZNF9-USP6
t(9;17)(q22;p13) OMD-USP6
t(17;17)(q12;p13) COL1A1-USP6
Mesenchymal chondrosarcoma NA HEY1-NCOA2
t(1;5)(q42;q32) IRF2BP2-CDX1
Myoepithelial tumor of bone t(6;22)(p21;q12) EWSR1-POU5F1
t(19;22)(q13;q12) EWSR1-ZNF444
t(1;22)(q23;q12) EWSR1-PBX1
Bizarre parosteal t(1;17)(q32;q21) RDC1
osteochondromatous proliferation
(Nora’s lesion)
Subungual exostosis t(X;6)(q24-q26;q15-21) COL12A1-COL4A5
Alveolar rhabdomyosarcoma t(2;13)(q35;q14) PAX3-FOXO1
t(1;13)(p36;q14) PAX7-FOXO1
Other t with 2q35 PAX3
Alveolar soft part sarcoma t(X;17)(p11.2;q25) ASPSCR1-TFE3
Angiofibroma t(5;8)(p15;q13) AHRR-NCOA2
Dermatofibrosarcoma protuberans/ +ring/marker chromosome from t(17;22) COL1A1-PDGFB
giant cell fibroblastoma (q22;q13)
Desmoplastic fibroblastoma t(2;11)(q31;q12) Unknown
Epithelioid sarcoma-like t(7;9)(q22;q13) Unknown
hemangioendothelioma
Epithelioid hemangioendothelioma t(1;3)(p36.3;q25) WWTR1-CAMTA1
t(X;11)(p11.2;q22.1) TFE3-YAP1
Infantile fibrosarcoma t(12;15)(p13;q25) ETV6-NTRK3
Inflammatory myofibroblastic tumor t with 2p23 ALK fusion with:
TPM4 (19p13.1), TPM3 (1q21), CLTC
(17q23), RANBP2 (2q13), ATIC (2q35),
SEC31A (4q21), CARS (11p15)
Lipoblastoma t with 8q12 PLAG1 fusions
Lipoma, ordinary t with 12q14.3 HMGA2 fusions
t with 6p21 HMGA1 fusions
Lipoma chondroid t(11;16)(q13;p13) C11orf95-MKL2
Low-grade fibromyxoid sarcoma t(7;16)(q33;p11) FUS-CREB3L2
t(11;16)(p11;p11) FUS-CREB3L1
Mesenchymal chondrosarcoma del(8)(q13.3q21.1) HEY1-NCOA2
Myxoinflammatory fibroblastic der(10)t(1;10)(p22;q24) TGFBR3-MGEA5
sarcoma/hemosiderotic
fibrolipomatous tumor
Nodular fasciitis t(17;22)(p13;q13.1) MYH9-USP6
Ossifying fibromyxoid tumor t with 6p21 PHF1 fusions
Pericytoma t(7;12)(7p22;q13) ACTB-GLI1
Sclerosing epithelioid fibrosarcoma t(7;16)(q33;p11.2) FUS-CREB3L2
t(11;16)(p13;p11.2) FUS-CREB3L1
Solitary fibrous tumor inv(12)(q13q13) NAB2-STAT6
Spindle cell rhabdomyosarcoma t with 8q13 NCOA2 fusion with: SRF(6p21), TEAD1
(11p15)
Synovial sarcoma t(X;18)(p11.2;q11.2) SS18-SSX1
SS18-SSX2
SS18-SSX4
Tenosynovial giant cell tumor t(1;2)(p13;q37) CSF1-COL6A3

Other t with 1p13 CSF1

ERRNVPHGLFRVRUJ
118 3  Molecular Biology of Bone Tumors

Myoepithelial *
POU5F1
tumors

Ewing-like sarcoma
NFATc2

20
6

Myoepithelial tumors ZNF444


19

Myoepithelial tumors PBX1


1
Desmoplastic small round cell tumor

Angiomatoid fibrous histiocytoma

Angiomatoid fibrous histiocytoma


Acute myeloid leukemia
Clear cell sarcoma

Clear cell sarcoma


EWSR1

Extraskeletal myxoid chondrosarcoma

Myxoid liposarcoma
WT1 22

Ewing sarcoma Ewing-like sarcoma

FLI1
11

ERG
21 ETV1
SP3
TAF15
Acute myeloid leukemia
Ewing sarcoma

E1AF CREB1
CREB3L2 17 FEV
2
7
myxoid sarcoma

NR4A3
Low-grade fibro-

9
Acute myeloid
NMP4
leukemia
ETS TF family
DDIT3 ATF1
FUS
CREB/ATF TF family Ewing sarcoma
12
TET family of proteins
16
Angiomatoid fibrous histiocytoma
FIGURE 3-15  ■  An overview of translocation partnerships of the TET family of genes in various tumors. Translocation to various
genes in the same gene family is believed to be pathognomonic to a given entity (e.g., EWSR1 translocation to members of the ETS
family of transcription factors in Ewing’s sarcoma). Complementation by another TET member, FUS, has taken away this specificity
and the presence of FUS/ERG translocation both in Ewing’s sarcoma and acute myeloid leukemia brings additional complications
to molecular diagnostics. Similar shared translocation variants have been observed in clear cell sarcoma and angiomatoid fibrous
histiocytoma. Arrows point from the TET family translocation partners to translocation partners; members of a gene family are
indicated with identical colors. Annotation on arrows indicates associated tumor entities. The asterisk (*) indicates myoepithelial
tumors, which include soft tissue myoepithelioma and myoepithlelial carcinoma. (Modified and printed with permission from Szuhai
K, et al: Cancer Genet 205:193–204, 2012.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 119

many, sometimes pathogenetically unrelated, entities; technique certainly can produce valid and reproducible
therefore the identification of the second fusion partner results, it requires the often-difficult culture of fresh
by additional tests provides more precise information and tumor tissue and highly skilled personnel to produce and
can be helpful in classifying the lesion. In general, the interpret the karyotypes. In the case of simple balanced
results of genetic and molecular testing should always be translocations or the involvement of large chromosomal
interpreted within the histologic, clinical, radiographic, segments, this traditional method is reliable and repre-
as well as other biomarker contexts. In the chimeric sents the historic gold standard. However, in cases with
genes, the EWSR1 component provides the promoter complex karyotypes, demonstration of characteristic
activity that upregulates the production of the chimeric translocations can be more challenging. A new technique
transcript. The second partner of the chimeric gene sup- known as spectral karyotyping (SKY) can more defini-
plies the DNA binding domain and is responsible for the tively demonstrate chromosomal aberrations, especially
transcriptional activity of the chimeric protein. It is in complex karyotypes, and can identify complicated
very interesting that unique chromosomal translocations translocations that involve several chromosomes (Figs.
and fusion partner genes have also been identified in 3-16 and 3-17). This technique uses chromosome-specific
several soft tissue and bone lesions that were considered probes that create a unique color spectrum for each of
to be, by conventional pathogenetic concepts, of rather the 22 autosomal pairs and the two sex chromosomes that
reactive than neoplastic origin. Such translocations were is then optimized by computer-generated pseudocolor-
identified in myositis ossificans, aneurysmal bone cyst, ation. In complex karyotypes, the color-coding of SKY
nodular fasciitis, bizarre parosteal osteochondromatous improves recognition of individual chromosomes and
proliferation (Nora’s lesion), and subungual exosto- translocation events by the trained eye.169,214 Like
sis.140,182,202-205,229,231,246,247 Because of the presence of clonal G-banding, SKY requires a chromosomal metaphase
chromosomal translocations, these conditions should be preparation for analysis. The strength of both of these
considered to represent unique neoplastic proliferations techniques is that they will reveal all karyotypic aberra-
with self-limiting growth potential. The translocations tions within their limits of detection since all the chro-
have been identified in a wide range of neoplastic lesions, mosomes are examined. On the other hand, both
including soft tissue and bone tumors as well as common techniques can miss cryptic translocations, in which the
solid malignancies, due to the wide use of genomic translocated chromosomal segments are minute. Unfor-
sequencing, which is capable of detecting the hybrid tunately, these techniques are not very effective for diag-
genes even in the absence of cytogenetically detectable nostic purposes because in clinical practice only about
abnormalities or in the background of complex genomic 50% of sarcoma samples can be successfully cultured.
rearrangements. Therefore the absence of cytogenetically detectable spe-
For practical purposes to help detect chromosomal cific chromosomal translocations may provide a false
translocations and their respective chimeric genes in negative result. Results that yield a normal chromosomal
diagnostic workups of individual cases, several ancillary complement are particularly suspicious and suggest that
genetic techniques are now widely used. In addition to the cells analyzed may not represent tumor cells.
chromosomal karyotyping by conventional cytogenetics
and spectral karyotyping, fluorescence in situ hybridiza- Fluorescence In Situ Hybridization
tion with the fusion and break-apart probes as well as
sequencing of fusion transcripts are now routinely used This technique, fluorescence in situ hybridization (FISH),
in differential diagnosis of both soft tissue and bone sar- uses fluorescently labeled nucleic acid probes that hybrid-
comas. In describing these techniques we use the proto- ize to regions flanking the loci of interest and can detect
typic translocation in Ewing’s sarcoma involving the aberrant localization of these probes. FISH can be used
EWSR1 and FLI1 genes as a model translocation used in to identify abnormalities in cases where conventional
differential diagnosis of small cell sarcomas. karyotypic analysis is inconclusive or where metaphase
spreads are not available; for example, in cases with only
formalin-fixed, paraffin-embedded tissue. The advantage
Karyotyping
of FISH is that it can be applied easily to touch prepara-
Chromosomal karyotyping is the classic methodology for tions, fresh tissue, karyotype preparations, frozen speci-
demonstrating translocations and traditionally represents mens and formalin-fixed, paraffin-embedded samples.
the initial approach for identifying genetic markers in The signal is visualized by fluorescent microscopy and
these tumors.220 Fresh tumor cells are cultured, and cells computerized image analysis. It is particularly useful in
in metaphase are obtained for staining and visualization mapping studies in which the hypervariable probes for
under microscopy. The preparations are treated with chromosomes are used to identify chromosomal dele-
trypsin and Giemsa stains to produce G-banding, allow- tions, numerical aberrations, and translocations that
ing an experienced cytogeneticist to identify individual cannot be identified by conventional banding techniques.
chromosomes and often visualize translocations between The two basic strategies used to design the FISH probes
two chromosomes (Fig. 3-16). Computer-aided imaging are referred to as fusion and break-apart strategies.183
and initial analysis represent a clear improvement over With the fusion strategy, one uses two probes (labeled
traditional techniques that involve photographing the with two distinct chromophores) targeted to the two loci
cells in metaphase, cutting out individual chromosomes, that are known to recombine (giving two red signals and
organizing the chromosomes in a standard format, and two green signals in cells that are negative for the fusion
examining differences between them. Although this event) (Fig. 3-18). Translocation involving two probed

ERRNVPHGLFRVRUJ
120 3  Molecular Biology of Bone Tumors

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

der(21)

19 20 21 22 X X

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

19 20 21 22 X Y

B
FIGURE 3-16  ■  Chromosomal abnormalities in Ewing’s sarcoma and osteosarcoma. A, Karyotype of a Ewing’s sarcoma with the
t(11;22)(q24;q12). Extra copies of chromosomes 2, 7, and 9 are also present. Trisomy 8 is a frequent secondary abnormality found
in Ewing’s sarcoma and PNET. (A, Reprinted with permission from Sandberg AA, Bridge JA: Cancer Genet Cytogenet 123:1–26, 2000.)
B, Representative spectral karyotype from an osteosarcoma showing multiple chromosomal rearrangements. (B, Courtesy of Lu X:
MD Anderson Cancer Center, Houston, TX.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 121

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

der(21)

19 20 21 22 X Y
A

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18
der(21)

19 20 21 22 X Y
B
FIGURE 3-17  ■  Complex chromosomal rearrangements in Ewing’s sarcoma. A, A G-banded karyotype showing the three-way trans-
location 46, XX, t(1;21;7)(q25;q22.3;q22) (black arrows). B, Computerized spectral karyotype confirmed the three-way translocation
with rearrangement of chromosomes 1, 21, and 7 (white arrows). (A and B, Reprinted with permission from Jinwath N, et al: Cancer
Genet Cytogenet 201:42–47, 2010.)

loci generates nuclei with three signals, one yellow (indi- With the break-apart strategy, one can use probes
cating the recombination product),and one orange and labeled with two different fluorescent chromophores,
one green signal (corresponding to the two other loci/ often orange/red and green that hybridize to sequences
chromosomes not involved in the translocation). The that flank the fusion partner gene, EWSR1 (Fig. 3-19).
fusion probes are highly specific and can precisely iden- When these two probes hybridize at an intact gene/locus,
tify both partners of the chimeric gene, but a different they are in close proximity and appear as a combined or
probe pair would be needed to query for each of the pos- single signal due to the mixture of the dye spectra of
sible partners in the translocation product, usually requir- the two probes (when the probes are orange and green,
ing multiple tests. the combined signal approximates yellow). Two such

ERRNVPHGLFRVRUJ
122 3  Molecular Biology of Bone Tumors

11t
22

FLI-1 Probe (green)

Probe

R EWSR1

Probe
G

FLI1

A EWSR1 Probe (red)


11t B
22t

EWSR1/FLI-1 Fusion
Probes
G

R EWSR1 FLI1
breakpoint

FLI1 EWSR1
silent breakpoint

C 11

t(11:22)
22
22
Normal 22
Normal 11

11
E
t(11:22)

FIGURE 3-18  ■  Schematic diagram of fluorescence in situ hybridization (FISH) using fusion probes for the EWSR1 and FLI1 loci in
diagnosis of Ewing’s sarcoma. A, Specific DNA components of the probe hybridize to the telomeric (fluorescently labeled green)
region flanking the FLI1 gene and centromeric (fluorescently labeled red) region flanking the EWSR1 gene. B, In the absence of
the chimeric gene (the fusion of EWSR1 and FLI1 genes) these probes generate two green and two red hybridization signals in
cells with normal diploid (two copies) complement in the EWSR1 and FLI1 loci. C, The t(11;22)(q24;q12) translocation results in
transfer of the telomeric portion of the q-arm of chromosome 22 to the q-arm of chromosome 11, generating no hybridization signal.
In contrast, the transfer of the telomeric portion of chromosome 11 to chromosome 22 fuses the hybridization signals from the
centromeric side of the ESWR1 gene with the telomeric side of the FLI1 gene. D, The translocation depicted in C generates an
overlapped green and red (sometimes yellow) hybridization signal. In the diploid complement the remaining nonfused copies of
the ESWR1 and FLI1 genes generate additional split green and red hybridization signals. E, FISH performed on a normal control
metaphase cell with the fusion probe described in A-D showing two duplicates of hybridization signals flanking the FLI1 gene on
chromosome 11 (green) and the EWSR1 gene on chromosome 22 (red) (left panel). FISH performed on a Ewing’s sarcoma metaphase
cell showing the fusion of green and red signals on chromosome 22 (right panel). Inset, Enlarged image of chromosome 22 showing
the fusion of hybridization signals. (E and Inset; Modified and reprinted with permission from Sandberg AA, et al: Cancer Genet Cytogenet
123:1–26, 2000.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 123

22
22 22

Probes
G

R EWSR1
B

C
11t

22t

11t

22 22t
Probe Probe
G

FLI1 EWSR1 R EWSR1 FLI1


silent breakpoint breakpoint

E
D

F
FIGURE 3-19  ■  Schematic diagram of fluorescence in-situ hybridization (FISH) using break-apart probes for the EWSR1 locus in
diagnosis of Ewing’s sarcoma. A, Specific DNA components of the probe hybridize to the centromeric (fluorescently labeled red)
and telomeric (fluorescently labeled green) regions flanking the EWSR1 gene. B, The two components of the probe are in close
proximity when there is no rearrangement within the EWSR1 locus. The overlapping emission spectra yield a yellow signal. C, The
EWSR1 locus at both chromosomes is intact in these three depicted nuclei, and two overlapping pairs of red and green signals are
present (yielding two yellow signals) as seen using FISH in formalin-fixed, paraffin-embedded tumor sections. The nucleus is identi-
fied by DAPI stain and appears blue. D, The t(11;22)(q24;q12) translocation results in transfer of the telomeric portion of the q-arm
of chromosome 22 (with green hybridization signal) to the q-arm of chromosome 11. In addition, the telomeric portion of the q-arm
of chromosome 11 (with red hybridization signal) is transferred to the q-arm of chromosome 22. E, This translocation involves only
one of the two chromosomes, and the nonrearranged chromosome 22 has adjacent hybrization signals (green and red overlap to
produce a yellow signal). The rearranged chromosomes 11 and 22 have only green and red signals, respectively. F, Chromosomes
11 and 22 segregate independently and thus separate green and yellow signals are visualized in the nucleus when examined by
FISH. (Modified and reprinted with permission from Lazar A, et al: Arch Pathol Lab Med 130:1199–1207, 2006.)

ERRNVPHGLFRVRUJ
124 3  Molecular Biology of Bone Tumors

Ewing’s Sarcoma

SK-ES1
ARhSa
EWSR1 mRNA

A4573
TC71
UC2

UC6

PBL
RNA binding domain

SM

SM
NC
forward primer
FLI1 mRNA
ETS
reverse primer

EWSR1-FLI1
Fusion transcript
ETS

EWSR1 portion FLI1 portion B


A

EWSR1 (exon 7) FLI1 (exon 6)

Q Q S S S Y G Q Q N P S Y D S V R R V

CAACAGAGCAGCAGCTACGGGCAGCAGAACCCTTCTTATGACTCAGTCAGAAGAGG
A

EWSR1 (exon 10) FLI1 (exon 6)

M D E G P D L D L D P S Y D S V R R G

ATGGATGAAGGACCAGATCTTGATCTAGACCCTTCTTATGACTCAGTCAGAAGAGGA

EWSR1 FLI1 EWSR1 FLI1


1 23 4 5 6 7 6 7 8 9 1 23 4 5 6 7 89 6 7 8 9

C E 10

EWSR1 (exon 7) FLI1 (exon 5)

Q Q S S S Y G Q Q S S L L A Y N T T S

CAACAGAGCAGCAGCTACGGGCAGCAGAGTTCACTGCTGGCCTATAATACAACCTCC

EWSR1 FLI1
123 4 5 6 7 5 6 7 8 9

D
FIGURE 3-20  ■  Reverse transcriptase polymerase chain reaction (RT-PCR) analysis to demonstrate chimeric fusion transcripts in
Ewing’s sarcoma cell lines. A, Using primers specific to 5′ sequences of EWSR1 mRNA and 3′ sequences of FLI1 mRNA, the fusion
transcripts produced by chimeric EWSR1/FLI1 genes were amplified by RT-PCR. B, The results of RT-PCR reaction performed on
RNA extracted from Ewing’s sarcoma cell lines. Note the presence of fusion transcripts in RNA corresponding to Ewing’s sarcoma
cell lines. C through E, Verification of the identity of the amplified fragments by DNA sequencing, disclosing fusion transcripts cor-
responding to alternative breakpoints within EWSR1 and FLI1 chimeric genes.
ARhSa, Alveolar rhabdomyosarcoma; NC, negative control without template DNA; PBL, normal peripheral blood lymphocytes;
SM, size marker; TC71, SK-ES1, A4573, Ewing’s sarcoma cell lines; UC2, UC6, human urothelial carcinoma cell lines. (Reprinted with
permission from Lazar A, et al: Arch Pathol Lab Med 130:1199–1207, 2006.)

combined signals (one for each chromosome) will appear Sequencing of Fusion Transcripts
in cells in which the locus of interest is intact. When a
translocation occurs in the gene between the two probes, The DNA sequences of the genes that recombine to
the signals are split and distinct and separate orange and produce the chimeric fusion genes are known, and enough
green signals are detected along with the remaining intact examples have been characterized and sequenced to
yellow signal representing the nonrearranged locus/ determine where in the gene breaks tend to occur. The
chromosome. In practice, the break-apart methodology identification of long exonal/intronal DNA sequences of
is more common and is used in most commercially avail- the chimeric genes by polymerase chain reaction (PCR)
able products for sarcoma. The break-apart strategy is technically challenging and is practically never used
offers the advantage of demonstrating any rearrangement for diagnostic purposes. Instead, the amplification of
at the locus probed, but it cannot identify the transloca- fused mRNA (fusion transcripts) encoded by the specific
tion partner. Thus, the same set of probes can be used chimeric gene can be easily accomplished after its conver-
to demonstrate a rearrangement at the EWSR1 gene sion into cDNA using reverse transcriptase (Fig. 3-20).
locus in any tumor associated with this event, but cannot This methodology, the reverse transcriptase polymerase
identify the specific fusion partners involved in a wide chain reaction (RT-PCR) assay, can be used with fresh,
range of tumors. frozen, or paraffin-embedded tissue. The identity of the

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 125

amplified fragment can be confirmed using multiple tech- of bone cancers, especially osteosarcomas and chordo-
niques, including DNA gel electrophoresis, restriction mas, demonstrate chromothripsis, making this newly dis-
fragment digestion or, most rigorously, direct DNA covered mechanism contributory to the development of
sequencing (Figure 3-20). RT-PCR can be used to deter- a significant proportion of sarcomas arising in bone.225
mine which binding partner is participating in the forma- Chromothripsis occurs in a single catastrophic shattering
tion of the fusion gene when multiple potential partners event and it is followed by the disorderly stitching
are possible. The technology is limited in that it probes together of genomic fragments.187,190 The features of
for very specific forms of chimeric gene formations, chromothripsis include multiple and complex rearrange-
requiring multiple probe sets to examine all of the most ments that alter selected chromosomes, which can be
common sites of recombination within the gene; rare or restricted to one chromosomal arm or its region (Fig.
aberrant chimeric gene types thus can be missed or 3-21). In some instances, there is an exchange of multiple
require multiple to numerous amplifications to detect. fragments between the involved chromosomes. Copy
Moreover, the RT-PCR technique is prone to produce number changes associated with chromothripsis are
false positive results due to annealing of the primers to minimal and show either heterozygous deletions or no
alternative homologous genomic sites, resulting in the copy change. The rearrangements within the involved
amplification of nonspecific fragments. Therefore, it is regions are typically very complex and there is a pro-
recommended that the identity of amplified fragments nounced clustering of breakpoints. The arrangement of
should be verified by sequencing. broken chromosomal fragments is not related to their
In practice, it is wise to use a combination of various proximity in the normal genome.177
techniques in the diagnostic setting. We recommend The mechanisms of double-stranded DNA breaks that
using karyotype analysis complemented with FISH or trigger chromothripsis are still unclear. Experimental evi-
SKY for complicated cases when fresh tumor is available. dence suggest that exposure to ionizing radiation while
FISH is used as the first-line diagnostic modality for the chromosomes are condensed during mitosis is a pos-
paraffin-embedded tissue, with appeal to PCR-based sibility. Another possibility is a stress which causes DNA
testing when FISH results are uninterpretable or confus- replication forks to collapse in the S phase, triggering
ing. Many laboratories successfully use PCR-based multiple breakpoints. Breakpoints in chromothripsis typ-
testing of fresh, frozen, or paraffin-embedded tumors as ically involve the telomeric regions and therefore short-
first-line modalities. None of these tests can be inter- ened telomeres can contribute to the initiation of a
preted in isolation and cannot be reliably applied outside breakage-fusion cycle. The shattering of chromosomes
the context of a sensible morphologic and clinical dif- found in chromothripsis is similar to the previously iden-
ferential diagnosis. tified phenomenon of premature chromosomal compac-
tion.162 Chromothripsis may also represent an incomplete
apoptotic event, in which cells survive programmed cell
Chromothripsis death by initiating DNA repair with a random assembly
The generally accepted model of cancer development of chromosomal fragments.
postulates a progression of malignant transformation The mechanisms that put the shattered fragments
through a series of progressively increasing waves together are as interesting as those that trigger the entire
of mutational and epigenetic changes that gradually process. A template-independent DNA double-strand
change cell behavior from normal to malignant states. break repair is the main mechanism involved. In addition,
Such models of progression were developed for many such replicative processes that have been previously iden-
common solid human cancers; they involve progression tified in complex genomic rearrangements, including
through microscopically recognizable precursor condi- fork stalling, template switching and microhomology-
tions referred to as dysplasia and carcinoma in situ. These mediated break-inducing repair, are contributing factors.
microscopically recognizable phases of cancer develop- Interestingly, there is solid evidence that chromothrip-
ment are biologically underpinned by waves of clonal sis plays a role in the development of germline transmis-
expansion as cells acquire and accumulate multiple sion of genetic disorders and may have a profound effect
genetic and epigenetic changes. Such modifications of not only on tumor progression, but also on human
genetic material are believed to be essentially random, development.175
but they generate phenotypic variations in cell popula-
tions that are subject to selection through Darwinian
evolution. Recently, the phenomenon referred to as chro- CELL-CYCLE AND MITOTIC REGULATION
mothripsis was identified in a subset of human cancers; this
postulates that myriads of mutations involving selected It is generally agreed that during cell growth and espe-
chromosomes can occur in one catastrophic mitotic cially in the exponential growth phase, cells double their
cycle that can trigger the initiation of the carcinogenic constituents before mitosis. Most housekeeping proteins
process.123,146,172,225 This phenomenon explains the double their content before the onset of DNA replica-
well-known puzzle of many human cancers, especially tion. It is also of interest that minimum threshold levels
sarcomas developing in relatively young patients or of total RNA and proteins are essential for entry into the
often in childhood without any recognizable precursor S phase. On the basis of these parameters, it is possible
conditions.215,236 In general, this phenomenon is operat- to separate cells in the G phase into subpopulations of
ing in a small fraction of all human cancers, not exceeding those that are in the early G phase (G1A) and those that
2% to 3% of investigated cases. In contrast, up to 25% are in the late G phase (G1B).254-256 Similar minimum

ERRNVPHGLFRVRUJ
A
PD3808a: Chr 9p Chr 8p Chr 4q

To chr 7q To chr 8p To chr 7q


* * *

2
1
0
1
0

CDKN2A WRN FBXW7

17 20 23 26 29 32 35 38 152 154 156 158 160


Genomic location (Mb) Genomic location (Mb) Genomic location (Mb)
B
8505C: Chr 9p PD3175a (CLL)
CDKN2A
CDKN2A

2 Chr 9
0
18Mb 24Mb
4
2 To chr 10
0
1
0

2
0
21.5 21.75 22 22.25 22.5 45Mb 52Mb 98Mb 101Mb168Mb 171Mb

Genomic location (Mb) Chr 13 Chr 4


miR-15a / 16-1
C D
FIGURE 3-21  ■  The concept of chromothripsis and its biologic effects. A, Stimulating stress initiates the shattering of chromosomes
in localized regions, which are subsequently recombined together at random (left). The double-strand breaks caused by the stressed
stimuli are reconnected and may result in the deletion of some of the regions. The breakpoint junctions reveal homology without
additional insertions, supporting the concept that they are being generated by nonhomologous end-joining (right). (A, Reprinted with
permission from Maher CA, Wilson RK: Cell 148:29–32, 2012.) B, Microhomology-mediated break-induced repair results in small dupli-
cation or triplication of the reassembled segment, represented by additional open rectangles above the reassembled segments of
the altered chromosome. C, Examples of chromothripsis involving selected chromosomes in osteosarcoma (left) and chordoma
(right). The diagrams shows rearrangements of chromosomal segments and their position on the chromosomal map depicted by
continuous arcs. The inner ring shows SNP-based allelic ratios and the outer ring shows the copy number profile of the selected
chromosomes. D, Loss of tumor suppressor genes, CDKN2A and the microRNA (miR-15a/16-1) cluster by rearrangements involving
chromosomes 4, 9, and 13. (B through D, Reprinted with permission from Stephens PJ, et al: Cell 144:27–40, 2011.)
ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 127

thresholds exist for individual protein levels.254-258 Further, level of G1. The overall sequence homology of CDKs
many cell proteins follow the same overall pattern of cell across the species is minimal, but the cyclin binding
cycle-related expression. The most common qualitative domain is highly conserved. In view of this fact, it is
difference between benign and transformed cells is that surprising that the regulatory network, especially of G1-S
transformed cells have increased variability of postmitotic control, has a strikingly similar network architecture
G1 cells and an increased ratio in gene expression levels across species. The transcriptional regulation of E2F/RB
among G1A and G1B cell populations. According to a pos- in animals and SBF/Whi5 in yeast are controlled by one
tulated concept of asymmetric distribution of gene prod- CDK-cyclin complex (Cdk4/6CycD in animals and
ucts after mitosis, this can be the result of increased Cdc28-Cin3 in yeast) and then by a second CDK-cyclin
asymmetry of malignant post­mitotic daughter cells com- complex (CDK2-CycE in animals and Cdc28-Cin2 in
pared with benign cells.257 This phenomenon was docu- yeast). A third CDK-cyclin complex is activated by
mented in reference to some tumor-transforming and Cdk2-CycA in animals and Cdc-Cib5 in yeast. The
tumor suppressor genes expressed in human cancer cell third complex also requires the inactivation of CDK
lines growing in vitro.267 The increased postmitotic asym- inhibitors such as p27 in animals and Sic1 in yeast to
metry of cancer cells and the uneven distribution of their complete the S phase. In addition, Cdk4/6-CycD plays a
regulatory proteins account for the asynchrony of the role in titrating p27, preventing the inhibition of
cell-cycle transverse by placing the cells with higher Cdk2-CycE complexes. New layers of complexity are
levels of regulatory protein closer to the G1-S phase tran- added to this regulatory network by the orchestration of
sition.257,267 This explains why the cells with a lower level mRNA stability, including modulation by miRNA.280,293
of these constituents have a longer time of residency in The main tumor suppressor genes, TP53 and RB1, play
the G1 phase. On the other hand, the increased asym- a central role in the regulation of cell-cycle progression
metry of the postmitotic distribution of cell-cycle regula- by interactions with the transcription factors, cyclins,
tory elements increases the pool of postmitotic cells with and DNA responsive motifs connecting the direct
levels of multiple proteins sufficient for immediate entry cell-cycle regulatory machinery to diverse cellular path-
into the S phase. ways controlled by growth factors and extracellular
Mitotic cycle and cell cycle have several variants that signaling.171,259,261,266,276
are characteristic of specific developmental stages (Fig. The regulatory mechanisms of cell-cycle progression
3-22).158 A prototypic cell cycle of all eukarytotic cells has and especially of the G1 and S phase transits have been
three major checkpoints referred to as G1-S, G2+M, and extensively studied, but the molecular machinery of the
spindle checkpoints. The cycles of embryonic cells are mitotic spindle has only recently been elucidated.281
rapid, and some phases of the full cell cycle can be These studies disclose a formidable complex molecular
skipped. In mature differentiated cells, they often repeat machine built by the cells to segregate chromosomes into
the endoreplication in which the S phase is not concluded two daughter cells after cell division.199,252,263,284,291 The
by M phase but is followed by another S phase, resulting biologic function of mitosis is to accomplish the segrega-
in polyploidy. tion of one copy of each chromosome into daughter cells.
The traverse of a cell across the cell cycle can be envi- The apparatus responsible for this process, the so-called
sioned as a sequence of checkpoints that act in concert. mitotic spindle, is composed of hundreds of proteins with
On the individual cell level, the phenomenon of cancer the overall architecture of microtubules attached to the
results from a profound deregulation of this system, and specific part of the chromosome referred to as the centro-
almost certainly, each cell population has some distinctive mere and to the pole of the mitotic spindle referred to as
alterations and biologic features. This explains why it is the centrosome.268,291,294 Similar to chromosomes, the cen-
so difficult, and most likely even impossible, to link the trosomes are delivered into the daughter cell after cell
development and progression of most human cancers, division and are duplicated in the following S phase. The
including those that affect the skeleton, to a single genetic centrosome duplication cycle shares common regulatory
or molecular alteration. pathways with the cell-cycle regulators and involve
A central dogma of cell-cycle regulation is that its CDKs.277,292 The microtubules forming the spindle can
progression is controlled by the activity of a specific class be considered as microengines acting as springs and dash-
of serine/threonine kinases referred to as CDKs, which pots. They are arranged longitudinally and laterally and
contain a highly conserved PSTAIRE sequence in their propagate the segregation of chromosomes due to their
domain that binds cyclins (Fig. 3-22).158,266,271,279,287,290 viscoelastic nature and tiled-array architecture. The
CDK1 controls mitosis and CDK2 controls entry into S overall orientation of the mitotic spindle is regulated by
phase in concert with CDK4 and CDK6. Specific CDK- a highly conserved set of molecules that govern cell
cyclin complexes have specific functions, but it appears polarity. In both mesenchymal and epithelial cells, the
that the overall level of CDK activity is more important apical to basal polarity regulates the position of the
for controlling the cell-cycle progression than their spe- mitotic spindle and secures equal segregation of chromo-
cific function.159,249,260,261,264,265,266,282 In this model devel- somes into daughter cells. Overall, more than 1000 pro-
oped primarily for yeast but also likely functional for all teins show a mitotic-specific phosphorylation and many
eukaryotic cells, the CDK activity is at its lowest in G1, of them belong to the superfamily of kinases. Among
is moderate at the start of S phase, and rises steadily them, the Aurora kinases play a prominent role as regula-
through G2, preventing reinitiation of replication and tors of the mitotic spindle and progression of mitosis
reaches its peak level at the start of mitosis. After mitosis, (Fig. 3-23).161,269,273 Aurora kinase A, a major component
the CDK activity is shut down and it returns to the lowest of centrosomes, controls mitotic entry, centrosome

ERRNVPHGLFRVRUJ
128 3  Molecular Biology of Bone Tumors

(a) (b) (c)

S G2 S

G1-S G2-M
check check G S
point P point P
M M
M M
A
G1 T T A
spindle
check
point
A

Threshold level for mitosis Mitosis

M Cyclin Cdc2
chromosomes
S Cyclin Cdc2

G1 Cyclin Cdc2
DNA licensing

G 1 S G2 M

B
CYCDs CycD Cln3

CDKA;1 Cdk4/6 Cdc28

E2F RBR1 E2f Rb SBF Whi5

FBL17 Skp2 Cdc4

Kip1
KRPs p27 Sic1

CDKA;1 Cdk2 Cdc28

CYCDs CycE Cln2

CDKB1s

CYCAs CycA Clb5

Arabidopsis Mammals Budding yeast


C
FIGURE 3-22  ■  Cell-cycle control. A, Mitotic cell cycle and its variants typically associated with different cell stages. (a) A typical
eukaryotic cell cycle with three major checkpoints. (b) Embryonic cell cycle with skip phases, resulting in rapid multiplication. (c)
Cell cycle of differentiating tissue with frequent and replicative cycles in which S phase is followed by M phase, resulting in poly-
ploidy. P, M, A, and T designate prophase, metaphase, anaphase, and telophase, respectively. B, Quantitative changes of major
regulatory components of the cell cycle. Total cyclin-dependent kinase (CDK) activity is shown in relation to cell-cycle phases. The
model was developed for Schizosaccharomyces pombe but is anticipated to be relevant to all eukaryotic cells. C, Cell-cycle control
networks at the G1-S transition. Note considerable homology among the regulatory networks across the species. (A through
C, Reprinted with permission from Harashima H, et al: Trends Cell Biol 23:345–356, 2013.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 129

_ +

+ _

+ _
+ _

B
Key
Mechanical symbols Molecular symbols
Spring Microtubule Kinesin-14
Dashpot Centrosome
Kinesin-5
Force Chromosome
arm Kinetochore
A
kinetochore
sister chromatid
interpolar MT

centrosome

K-fibre

Aurora A Aurora B Aurora A + B

Mitotic entry Chromosome biorientation


Microtubule stability
Centrosome separation Spindle assembly checkpoint
Anaphase spindle disassembly
Centrosome maturation Sister chromatid cohesion
Chromosome alignment Cytokinesis

C
FIGURE 3-23  ■  Mitotic spindle micromechanics. A, The spindle can effectively be considered to be many springs and dashpots in
parallel arranged both longitudinally and laterally. Both elastic and viscous elements originate from the viscoelastic nature of
molecular motors (upper left), microtubule elasticity and turnover, and the tiled-array architecture of the spindle. Multiple motors
cooperate and slide antiparallel microtubules apart, but oppose each other and lock and parallel microtubules together (upper right).
A large external force can lead to rapid detachment of half of the motors, breaking microtubule-microtubule crosslinking and leading
to a plastic spindle response. B, Robust spindle architecture depends on properly homogeneous “filing” of the spindle interior by
microtubules. This cannot be achieved by random microtubule nucleation with the RanGTP cloud (shaded region, top); the micro-
tubules are too spread out and disjointed. Nor can this be achieved by the autocatalytic nucleation (bottom) of short microtubules,
which leads to dense, disjointed clumps of microtubules. The optimal regimen (middle) is a combination of autocatalytic microtu-
bules branching with a RanGTP-mediated enhancing effect and fast microtubule growth and turnover dynamics. (A and B, Mogilner
A, Craig E: J Cell Sci 123:3435–3445, 2013.) C, Summary of Aurora kinases in the mitotic spindle mechanics and function. (C, Hocheg-
ger H, et al: Open Biol 3:120185, 2013.)

separation, maturation, and chromosome alignment.275,277 that is observed in many human cancers, including bone
On the other hand, Aurora kinase B is implicated in cancers. The overactive Aurora kinase A produces super-
chromosomal orientation, spindle assembly checkpoint, numerary centrosomes, resulting in multipolar mitosis
sister chromatin adhesion, and cytokinesis.275 Both of the and chromosomal misseggregation with aneuploidy.277
Aurora kinases collaborate to accomplish microtubule These observations provide new insights into molecular
stability and disassembly of the mitotic spindle in ana- mechanisms of aneuploidy and explain the microscopic
phase. Lack of function of Aurora kinases in experimental hallmark of malignant cells, the so-called atypical mitosis.
systems contributes to various mitotic dysfunctions and In summary, complex regulatory mechanisms that
may be lethal. Interestingly, the overall activity of Aurora operate on the promoter level of cell cycle-mediating
kinases, and especially of Aurora kinase A, is a frequent genes influence the proliferation activities of cells.
phenomenon due to amplification and overexpression The regulation of histone gene expression is used as a

ERRNVPHGLFRVRUJ
130 3  Molecular Biology of Bone Tumors

Histone Gene

S1-PHASE: MAXIMAL TRANSCRIPTION


IRF-1
HinF-D (CDC2/CLN-A/RB)
HS IRF-2
HS
Nuclear
AFF1 matrix SP-1
NMP-1 ATF1 RNA pol II
H4UA-1
H4UA-3 YY-1 HinFA mRNA

Site IV Site III Site I Site II HinFA

Distal promoter Proximal promoter

DIFFERENTIATED: INACTIVE
mRNA

Site IV Site III Site I Site II

Distal promoter Proximal promoter


A

Osteocalcin Gene

DHS-1 Positioned DHS-1


nucleosome AP-1 TFIIB TAFS
AP-1 YY-1
OCBP2 OCBP1 RNA pol II

AML VDR/ AML


AP-1 GR AP-1 AML HLH MSX TFIID mRNA
RXR
GRE A VDRE B TGRE C E-box TATA GRE

-0.6 kB AP-1 YY-1 -0.4 kB -0.2 kB OC-BOX II OC-BOX I SIL

Far distal Distal promoter Proximal promoter


promoter
B
FIGURE 3-24  ■  Organization of regulatory elements (promoters) of histone and osteocalcin genes. A, Maximal transcription in S
phase is associated with binding of multiple transcription factors in sites I and II of proximal promoter and sites III and IV of distal
promoter. Differentiated (inactive) state is associated with complete inhibition of transcription and dissociation of multiple growth
factors from sites I, II, and IV. Occupation at site I is retained in differentiated (inactive) state. B, In contrast, promoter of osteocalcin
gene has several overlapping motives that respond to multiple transcription factors in differentiated nonproliferative state. They
contain elements responsible for tissue-specific expression (silencer domains, TATAboxes, and HOX homeodomain). Several AP-1
binding sites regulate expression in relation to cell differentiation. In addition, hormone-related expression is mediated by vitamin
D–responsive elements (VDRE) and multiple glucocorticoid-responsive elements (GRE). (Modified and reprinted with permission from
Stein GS, et al: Mechanisms regulating osteoblast proliferation and differentiation. In Bilezikian JP, Raisz LG, Rodan GA, eds. Principles of
bone biology, San Diego, 1996, Academic Press.)

paradigm of ubiquitous regulatory mechanisms in prolif- of the histone gene from the nuclear matrix and from
erating cells (Fig. 3-24).286 In general, the transcription transcription factors at several binding sites within the
of histone genes is upregulated at the beginning of the promoter.
S phase and is suppressed in differentiated or quiescent In bone, the regulation of the osteocalcin gene in
cells.251,286 Accordingly, the promoter of the histone gene osteoblastic cells signifies a model regulatory mechanism
responds to various regulatory proliferation signals such in the transition from proliferation to differentiation
as those from CDC2, cyclin A, the Rb-related protein, phases (Fig. 3-24). In contrast to the histone gene, the
and 1RF-2, among many others.278,283,285,289 Suppression organization of the osteocalcin gene promoter contains
of histone gene expression at transition to the differen- several elements that upregulate the gene in a differenti-
tiation or quiescent state is associated with dissociation ated postproliferative state and enhance its expression

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 131

in parallel to matrix mineralization and differentiation In subsequent paragraphs, we briefly discuss some
of osteoblastic cells. The osteocalcin gene contains general molecular concepts of osteosarcoma, chondrosar-
three major groups of DNA motives that bind factors coma, chordoma and Ewing’s sarcoma and examine the
responsible for (1) tissue-specific expression (silencer significance of so-called dedifferentiation as a unique
domain, TATA box, HOX homeodomain), (2) regula- phenomenon observed in several low-grade skeletal
tion of its expression in parallel to cell differentiation neoplasms.
(AP-1 sites), and (3) regulation of hormone-dependent
expression (vitamin D–responsive element and multiple Osteosarcoma
glucocorticoid-responsive elements).250,253,262,270,272,274,288
The individual elements of the osteocalcin gene pro- The vast majority of osteosarcomas are sporadic de
moter have an overlapping organization. It has been sug- novo tumors that develop without any known predis-
gested that the unique organization of the regulatory posing conditions. Small fractions of osteosarcomas
motives of the osteocalcin gene maintains responses to develop, however, in the background of well defined,
multiple factors. typically familial, predisposing syndromes. These syn-
dromes provide important clues for molecular mecha-
nisms involved in the development of these aggressive
mysterious malignancies. They include the retinoblas-
MOLECULAR CONCEPTS toma and Li-Fraumeni syndromes, which are asso-
OF BONE NEOPLASIA ciated with the mutation of the RB and TP53 genes,
respectively, as well as a group of cancer predisposing
From the molecular point of view, human malignant neo- syndromes associated with germline mutations of heli-
plasms, including those that affect the skeleton, can be cases.295,305,307,308,312,318,325,329-331,333,339,342 The mutations of
divided into two broad groups: (1) tumors that exhibit helicases, primarily the RECQL4, are the hallmarks of
distinct molecular alterations that have been implicated several cancer predisposing syndromes, the most impor-
as playing a significant role in tumor pathogenesis and tant of which include Rothmund-Thomson, RAPADI-
(2) tumors with widespread genomic alterations that LINO, Baller-Gerold, and Werner. The unifying features
seem to develop by means of a multistep cumulative of these syndromes represent genomic instability, prema-
process. In tumors with distinct molecular alterations, ture aging, and increased risk for multiple cancers includ-
the changes are typically not single alterations, and ing osteosarcoma. Although the mutations of helicases
additional secondary hits are required for the develop- are unusual in sporadic osteosarcomas, the mutations of
ment of the complete malignant phenotype. They also RB and TP53 genes, as well as their respective regula-
occur less frequently than those that seem to develop by tory pathways, are among the most frequent molecu-
a complex multistep process. lar alterations seen in these tumors.309 Osteosarcomas
The molecular mechanisms that play a role in the are extremely unstable tumors and exhibit pronounced
pathogenesis of human malignant tumors are of several aneuploidy with alterations of multiple chromosomes
major types: and their extra copies.296,310,311,313,315,316,321-323,334,341,361 A
1. A gene with a negative growth-regulatory effect novel mechanism involved in the development of several,
(tumor suppressor gene) is altered or lost. malignancies, both solid and hematologic, including
2. A gene with positive regulation of growth is acti- osteosarcoma, has been proposed and is referred to as
vated by upregulation or alteration (mutation) that chromothripsis (described in more detail above).123,172,225
enhances its transforming activity. In general, the key regulatory pathways involved in
3. The fusion of two genes by translocation results in osteosarcoma represent a component of cell cycle check-
a new hybrid gene that exhibits a strong activity as points.324,360,363 Some of these alterations were shown to
a transforming factor. be of prognostic value (e.g., the deletions in the CDKN2A
4. A gene involved in the stabilization of DNA tran- locus are associated with poor survival in patients
scriptional fidelity (DNA repair gene) is altered or with osteosarcoma).350,358 Similarly, the molecular mecha-
lost. This results in failure to repair DNA tran- nisms involved in the maintenance of telomeres, such
scriptional mistakes and in turn causes a cascade of as overexpression of mRNA for telomerase reverse
alterations that affects many genes. transcriptase, also predict poor outcome. Several gene
This simplified view of the role of individual genetic expression and miRNA studies have identified specific
alterations should be placed in the context of the complex signatures associated with an outcome in osteosarcoma.
landscape of genomic alterations that affect multiple inter- The miRNA network studies have shown that miR-1
acting pathways contributing to the phenomenon referred and miR-133b are involved in proliferation and cell cycle
to as malignant transformation. Because tumors are begin- control.348 The involvement of long noncoding RNAs
ning to be investigated with increasing frequency by as a gene regulatory network has also been identified in
genome profiling rather than by individual gene or pathway osteosarcoma.328
approaches, it is appropriate to view tumors, including The frequent presence of cartilaginous differentiation
skeletal neoplasms, as complex genomic disorders. This and a large proportion of undifferentiated mesenchymal
view does not eliminate the concept and importance of key cells in most high-grade osteosarcomas suggest that early
driver type alterations and potential targetable genes as osteoprogenitor cells with the ability for chondroblastic
well as their respective pathways that offer new, hopefully differentiation are affected in the development of osteo-
more effective, therapeutic interventions. sarcoma. On the other hand, a phenotypic switch to

ERRNVPHGLFRVRUJ
132 3  Molecular Biology of Bone Tumors

another mesenchymal cell lineage is extremely rare in percentage of primary and secondary central as well as
osteosarcoma. This suggests that cells with a commit- periosteal chondrosarcomas. The progression mecha-
ment to perform skeleton-forming functions are affected nisms of chondrosarcoma are largely unknown, but
in the development of osteosarcoma. This observation high-grade lesions are characterized by aneuploidy and
also strongly suggests the existence of such a mechanism multiple chromosomal abnormalities. As in osteosar-
that seems to be highly protected and remains unaltered coma, TP53 and RB genes are relatively frequently altered
in osteosarcoma, but its molecular mediators remain to in chondrosarcoma, and these alterations are associated
be elucidated. with the clinical aggressiveness of the tumor.345 Allelic
Most conventional osteosarcomas produce easily rec- losses of 17p are ubiquitous in chondrosarcomas and may
ognizable amounts of osteoid but lack its organization play an important role in their pathogenesis. The activa-
and the lamellar maturation seen in normal bone. This tion of several oncogenic pathways that may represent
is in keeping with the phenotypic heterogeneity of various targets for novel therapeutic interventions have been
osteosarcoma cells and their functions, such as their identified; they involve IHH/PTHLH/Bcl-2, Src, Akt,
ability to produce individual elements of extracellular and PDGRF, IGF, and estrogen signaling as well
matrix. Osteosarcomas can produce or even overexpress as hypoxic and glycolytic pathways.299,300,301,303,317 The
some elements of the matrix. On the other hand, they are Hedgehog signaling pathway plays a major role in carti-
not able to produce the whole gamut of matrix compo- lage development and continues to be activated in the
nents in the appropriate amount and sequence required progression of chondrosarcomas, possibly contributing
for the complete maturation of bone. The discovery of to their proliferative states.307 Overall, the alterations of
differentiation activities of matrix polypeptides related to TP53 and RB pathways can be identified in a significant
growth factors (BMPs) explains a microscopically recog- proportion of high-grade chondrosarcomas.304,345 This is
nizable phenomenon referred to as normalization (i.e., concurrent with the frequent amplification of 12q13 and
osteocyte-like appearance of osteosarcoma cells embed- loss of 9p21, which contain many genes involved in the
ded in solid areas of osteoid). regulation of these pathways. The PI3K-AKT pathway
and MAPK-AP-1-Runx2 axis are the most frequently
activated kinase pathways in chondrosarcoma.350,351 Alter-
Chondrosarcoma ations of programmed cell death play a significant role in
In general, chondrosarcomas develop from cells com- the pathogenesis of low- to intermediate-grade chondro-
mited to cartilaginous differentiation. They represent a sarcoma. On the other hand, high-grade lesions most
heterogeneous group of neoplasms with diverse biologic likely develop by means of a multistep mechanism that
behaviors. They range from low-grade, locally aggressive involves multiple transforming and tumor suppressor
malignancies (grade 1) to highly aggressive tumors with genes. In general, aneuploidy and overexpression of
a high propensity for metastases (grade 3). Grade 2 Aurora kinases are adverse prognostic factors.332,338The
tumors represent an intermediate group. Some grade 2 complex network of regulatory miRNA contributed to
tumors metastasize, and some behave as locally aggressive this process.362
lesions. A subset of chondrosarcomas exhibit myxoid The identification of EXT1 and EXT2 genes, which
changes. Most skeletal myxoid chondrosarcomas are are implicated in the development of hereditary multiple
grade 2 lesions and are pathogenetically distinct from soft exostoses, suggests that their alterations may play a role
tissue myxoid chondrosarcomas. A small subset of bone in secondary malignancies (i.e., chondrosarcomas that
myxoid chondrosarcomas are high-grade lesions and may develop in this syndrome).314 Secondary peripheral chon-
be pathogenetically similar to extraskeletal myxoid chon- drosarcomas, especially those associated with osteochon-
drosarcomas. Mesenchymal chondrosarcoma is an outlier droma, carry the same mutations of EXT1 and EXT2
and has more similarities with small cell malignancies, genes as an underlying osteochondroma.
including the presence of the two fusion genes HEY1- In chondrosarcoma, a number of genetic alterations
NCOA2 and IRF2BP2-CDX1 than with the rest of the (altered genes) correlate with the clinical aggressiveness
cartilaginous neoplasm. Central and especially secondary of the lesion, and most high-grade tumors are aneuploid.
peripheral chondrosarcomas evolve via distinct molecular Karyotyping studies indicate that alterations involving
pathways. Both central chondromas, commonly referred multiple chromosones are frequent in chondrosarcoma
to as enchondromas, and central chondrosarocmas share of bone.302,352 Moreover, recent differential genomic
some molecular similarities and are characterized by het- hybridization studies show losses and gains of genetic
erozygous mutations of isocitrate dehydrogenase 1 material on multiple chromosomes that may play a role
(IDH1) and isocitrate dehydrogenase 2 (IDH2).300,349 in the pathogenesis of these neoplasms.326
There are two hot spots where most of the mutations The progression of a low-grade, locally aggressive
occur, and they involve R132C and R132H in IDH1 and neoplasm to a high-grade malignancy is referred to as
R172S in IDH2. These genes are also frequently involved dedifferentiation; dedifferentiated chondrosarcoma is a
in Ollier’s disease and Maffucci’s syndrome. They cata- prototype of this phenomenon. Dedifferentiation may
lyze the oxidative decarboxylation of isocitrate into also occur in low-grade intramedullary osteosarcoma,
α-ketoglutarate of the Krebs tricarboxylic acid cycle. parosteal osteosarcoma, giant cell tumor, and chordoma.
IDH1 is a predominantly cytoplasmic and peroxisomal The development of a similar, highly lethal sarcoma in
enzyme; IDH2 catalyzes the mitochondrial Krebs cycle. several distinct precursor conditions may in fact repre-
The mutations of these genes are not only present in sent a pathogenetically similar process that has common
benign cartilage lesions, but are also identified in a high molecular pathways of development.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 133

Cytogenetic studies and our own observations with involved in approximately 10% of Ewing’s sarcomas.152
the use of hypervariable chromosomal probes indicate In very rare cases, the FUS gene fuses with ERG or FEV
that the two components of dedifferentiated chondrosar- (ETS family members). In rare instances EWSR1 forms
coma are closely related and that the high-grade sarco- a chimeric gene with non-ETS partners.144,306 Although
matous component represents a subclone of a low-grade the molecular structures of common translocations
precursor condition.340,357,359 Virtually all reported cases are elucidated, new subgroups of small cell malignancies
of dedifferentiated chondrosarcoma exhibit strong over- with unique translocations continue to be identified.306
expression of the p53 protein in the dedifferentiated An example is a novel class of small cell malignancy
components.304,355 Lack of an immunohistochemically with Ewing’s sarcoma-like microscopic features and
detectable Rb protein is seen in more than 50% of dedif- characterized by the translocation of CIC-DUX4 and
ferentiated sarcomatous components. The bcl-2 protein BCOR-CCNB3 genes.154,167,173,209 Typically, cytogeneti-
is typically present in the low-grade precursor conditions cally detectable translocations occur in the background
but is absent in the sarcomatous component. Our data of an almost normal karyotype with a limited number of
indicate that extensive losses of genetic material on secondary alterations usually representing chromosomal
chromosome 17p play a role in the development of duplications. There is, however, a subset of Ewing’s sar-
dedifferentiation. comas that exhibit complex copy number alterations and
chromosomal rearrangements.319,320,335 At the time of this
writing, there is not much information on the biologic
Chordoma effects of alternative fusion partners; the bulk of the
The signature molecular mechanism involved in the information on target partners and involved pathways is
development of chordoma is chromothripsis. In fact, available for the signature translocation involving EWSR1
chordoma is the prototypic tumor in which this novel and FLI1.353,356 Multiple upregulated and downregulated
mechanism of cancer development has been identi- genes responsible for transcriptional activation and a
fied.123,172,225 Hence, the signature of chordoma is the repressor function of EWSR1-FLI1 have been identified.
presence of complex rearrangements involving selected Several direct targets of EWSR1-FLI1 have been identi-
chromosomes. On the other hand, brachyury is a major fied via a demonstration of binding to their upstream
regulator of notochord development and displays major promotor regions.336 It has been documented that
biologic effects on the behavior of chordoma cells (Fig. EWSR1-FLI1 acts as an aberrant transcription factor
3-25).346 Elucidation of the molecular structure of brachy- and binds to GGAA microsatellites within the regulatory
ury provided novel insights into its function as a tran- elements of the target genes. The direct target genes
scription factor binding to a unique DNA motif and of EWSR1-FLI1 include hsRPB7, UPP1, tenascin-C,
controlling the expression of multiple genes.344 In experi- ID2, p21, PTPL1, phospholipase D2, TGFBR1, IGFBP3,
mental systems, silencing of brachyury induces growth MK-STYX, TERT, GLI1, and Aurora A and B. There is a
arrest of chordoma cells in vitro. Brachyury binds to the long list of so-called secondary interacting genes that are
T-box motif that is highly conserved among species. It affected downstream of the primary target genes; their
directly binds to nearly 100 target genes and influences direct interactions with EWSR1-FLI1 have not been
the expression patterns of a myriad of downstream targets. identified (Table 3-3).
It has been shown that brachyury, in fact, acts as a master The interest in these genes and their respective path-
regulator of an elaborate oncogenic network involving ways has two aspects. The first is the elucidation of
diverse pathways affecting the cell cycle and cellular Ewing’s sarcoma biology. The second, and most impor-
interactions with matrix components. In addition, tant, is the identification of possible therapeutic targets
PDGRFB and RTCs, belonging to Flt3 and MCSFR1,
as well as the EGFR family (HER1, HER2/neu, and
HER4), are activated in a majority of chordomas. It has TABLE 3-3 A Summary of EWSR1/FLI1 Genetic
also been shown that PI3K/AKT and RAS/ERK 1-2 Targets
pathways are also frequently activated in chordomas, but
Regulate Gene Name
such major players in these pathways as PI3Kp110, PTEN,
RAS, and BRAF are not mutated. Moreover, the mTOR Direct Targets
complex 1 (raptor) is also frequently overexpressed and   Upregulated RPB7/POLR2G, TNC, UPP1, ID2,
hyperactivated in chordomas.354 These findings provide TERT, PTPL1/PTPN13, PLD2,
MK-STYX, FLI1, Aurora A and B
novel insights into the biology of chordomas and may   Downregulated p21/CDKN1A, TGFBR2, IGFBP3
represent novel therapeutic targets.
Indirect Targets
  Upregulated EAT-2/SH21B, MFNG, UBE2C,
Ewing’s Sarcoma CCND1, MAPT, PP1R1A, NEK2,
MYC, PIM3, NKX2-2, CCK, CAV1,
The biology of Ewing’s sarcoma focuses on the effects of CD99, VEGF-A, EZH2, TOPK/PBK,
the EWSR1 gene fused to one of five ETS (erythroblast IGF1/IGF1R, DAX1/NR0B1
transformation-specific) factors (Fig. 3-26).145,297,298,343,347,353   Downregulated p21/CDKN1B, p57/CDKN1C, ZYX,
FLI1 (Friend leukemia virus integration 1) is the domi- NOTCH-p53, thrombospondin 1
and 2
nant fusion partner involved in more than 80% of Ewing’s
sarcomas. ERG (v-ets erythroblastosis virus E26 oncogene From Mackintosh C, et al: Cancer Biol Therapy 9:655–667,
homolog) is the second most important fusion partner, 2010.

ERRNVPHGLFRVRUJ
134 3  Molecular Biology of Bone Tumors

Chr 6
25 Brachyury (T)
24
23
22.3

Exon 1

Exon 2

Exon 3

Exon 4
Exon 5

Exon 6

Exon 7

Exon 8
22.2 22.1

cds
21.3
21.2
21.1
12
11.2
11.1 tel
11.1 cen
12
13
14
15 Brachyury (T)
16.1
16.2 16.3
21
1 435 aa
42 222
22.1
22.2 Transcription factor, T-box domain
22.3
23.1 23.2
23.3
24
25.1 25.2
25.3 26
27 Brachyury (T)

A B

Xenopus
in vitro

Zebrafish
in vitro

Mouse
in vitro

U-CH1
in vitro

C D
80 IL8 FZD4
60 30
40
20 10

20 10

5kb 5kb

60
MTMR7 FGF1
30 40

10 20

10 20

5kb 5kb

E
FIGURE 3-25  ■  Structure and biologic effects of brachyury. A, Chromosomal location and exonal structure of the brachyury (T) gene
as well as the functional domains of the encoded protein. B, Molecular structure of the brachyury protein bound to DNA. A ribbon
diagram of the dimer protein bound to DNA is shown in a view perpendicular to the DNA axis. C, The binding motif for brachyury
identified in the chordoma cell line, U-CH1, showing sequence homology across the species. (C, Reprinted with permission from Muller
CW, Herrmann BG: Nature 389:884–888, 1997.) D, Representative chromatin precipitation peaks (ChiP-seq peaks) surrounding the
brachyury target genes. E, Representative ChIP-seq peaks surrounding brachyury target genes. (D and E, Reprinted with permission
from Nelson AC, et al: J Pathol 228:274–785, 2012.)

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 135

Break Point Break Point

IGFBP3
NH2 COOH NH2 COOH c-KIT
DHR RRM AD ETS-DBD IGF1
AMG 479
EWSR1 (chromosome 22) FLI1 (chromosome 11)
t(11;22) IMC-A12
SCH 717454 PDGFR VEGFR
R1507 IGF1R
NH2 COOH CP-751,871
DHR variable ETS-DBD
EWSR1-FLI1 t(11;22)(q24;q12)
A PIP2
PIP3 PI3K
Ras
EWSR1 Akt
U1C Pazopanib
NH2 hsRBP7 Imatinib
YK-4-279 Raf
TSC2
activation Rheb Dasatinib
domain TSC1
Pol II MEK1/2
DBD
GBL Rictor GBL Raptor Temsirolimus
GGAA mTOR mTOR Everolimus
ERK1/2
mTORC2 mTORC1 G1
RHA COOH
M
FLI1 MLN8237
Aurora
kinase
Transcriptional activation and repression G2 Cyclin
C D1
Alteration of splice site selection S
Modulation of RNA half-life
B
FIGURE 3-26  ■  Molecular genetics and its implication for targeted therapies in Ewing’s sarcoma. A, Schematic drawing summarizing
the EWSR1-FLI1 t(11;22)(q24;q12) translocation. The EWSR1-FLI1 fusion includes the N-terminal activation domain of EWSR1, which
contains multiple degenerate hexapeptide repeats (consensus SYGQQS), and the C-terminal ETS DNA-binding domain (ETS-DBD)
of FLI1. Variation in the sites of chromosomal breakpoints leads to multiple fusion types (bracketed region). B, Putative molecular
function of the EWSR1-FLI1 protein and selected protein-protein interactions. As an aberrant transcription factor, EWS-FLI1 regulates
genes in part by binding to GGAA microsatellites upstream of target genes. EWSR1-FLI1 has been shown to interact with the splic-
ing factor U1C (also known as SNRPC, small nuclear ribonucleoprotein polypeptide c), RNA helicase A (RHA), and the hRBP7 subunit
of RNA polymerase II (PolII), which links the protein to splicing and transcription. The small molecule that blocks the EWSR1-FLI1-
RHA interaction is indicated in red. C, Signaling pathways and targeted therapies in Ewing’s sarcoma. EWSR1-FLI1 modulation of
IGFBP3 and IGF1 and overexpression of IGF1R promote increased IGF1 signaling. ES also expresses PDGFR, c-KIT, and VEGFR.
Activation of IFG1R, PDGFR, c-KIT, and VEGFR leads to downstream signaling through the PI3K and MAPK pathways (indicated by
gray dashed line and arrows). EWSR1-FLI1 upregulates Aurora kinase A and cyclin D1, promoting progression through the cell cycle.
Targeted therapeutic agents used in recent clinical trials for Ewing’s sarcoma are indicated in red. Genes modulated by EWSR1-FLI1
are indicated in purple. Receptors overexpressed in Ewing’s sarcoma are indicated in red. ES, Ewing’s sarcoma; ETS, erythroblast
transformation-specific; FLI1, Friend leukemia virus integration 1; IGFBP, insulin-like growth factor binding protein; IGF1, insulin-like
growth factor 1; IFG1R, IGF1 receptor; MAPK, mitogen-activated protein kinase; PDFG, platelet-derived growth factor; PDGFR, PDGF
receptor; PI3K, phophoinositol3-kinase; VEGFR, vascular endothelial growth factor receptor. (Reprinted with permission from Anderson
JL, et al: Pediatric Res 72:112–121, 2012.)

and the design of more effective management for these biologically driven therapeutic interventions open some
highly aggressive tumors (Fig. 3-26).113,327 Downregula- new avenues for targeted therapies, but their clinical
tion of the IGF binding protein 3 connects the tumor application remains to be proven.
pathogenesis to the IGF pathway. Because Ewing’s Systems biology approaches to biologic effects of the
sarcoma cell lines ubiquitously express IGF1 receptor, EWSR1-FLI1 fusion protein have identified a complex
and EWSR1-FLI1 hybrid protein has been shown to crosstalk network of pathways affecting cell proliferation
upregulate IGF1 in mesenchymal progenitor cells, the and apoptosis. The network containing the signaling
anti-IGF1R therapies are of major interest. Because pathways involved in apoptosis signaling includes CASP3
EWSR1-FLI1 upregulates Aurora A and B kinases, which and cytochrome C as well as such cell cycle regulators as
act as major regulators of mitosis, the inhibition of Aurora TNF, TFGβ, MAPK, IGF, NFκB, c-Myc, and RB/E2F
kinases represents another experimental therapeutic among others.
approach. The preclinical studies with Aurora kinase A
inhibitor in a Ewing’s sarcoma animal xenograft model REFERENCES
show promising responses and open an avenue for clinical Molecular Techniques
applications. The direct binding of EWSR1-FLI1 with 1. Bernard PS, Wittwer CT: Real-time PCR technology for cancer
RNA helicase A, which increases the transcriptional diagnostics. Clin Chem 48:1178–1185, 2002.
activity of the complex, offers another therapeutic 2. Bernardini G, Laschi M, Geminiani M, et al: Proteomics of osteo-
sarcoma. Expert Rev Proteomics 2014 (epub ahead of print).
approach, and it has been shown that the block of RNA 3. Bignell GR, Greenman CD, Davies H, et al: Signatures of muta-
helicase A-EWSR1-FLI1 interaction induces apoptosis tion and selection in the cancer genome. Nature 463:893–898,
and inhibits tumor growth in xenograft models. These 2010.

ERRNVPHGLFRVRUJ
136 3  Molecular Biology of Bone Tumors

4. Brehme M, Hantschel O, Colinge J, et al: Charting the molecular 32. Abe E, Shiina Y, Miyaura C, et al: Activation and fusion by 1α,25-
network of the drug target Bcr–Abl. Proc Natl Acad Sci USA dihydroxyvitamin D3 and their relation in alveolar macrophages.
106:7414–7419, 2009. Proc Natl Acad Sci USA 81:7112–7116, 1984.
5. Bridge JA, Sandberg AA: Cytogenetic and molecular genetic tech- 33. Akatsu T, Takahashi N, Debari K, et al: Prostaglandins promote
niques as adjunctive approaches in the diagnosis of bone and soft osteoclast-like cell formation by a mechanism involving cyclic
tissue tumors. Skeletal Radiol 29:249–258, 2000. adenosine 38,58-monophosphate in mouse bone marrow cell cul-
6. Fan JB, Gunderson KL, Bibikova M, et al: Illumina universal bead tures. J Bone Miner Res 4:29–35, 1989.
arrays. Methods Enzymol 410:57–73, 2006. 34. Akatsu T, Takahashi N, Udagawa N, et al: Role of prostaglandins
7. Green ED, Guyer MS: National Human Genome Research Insti- in interleukin-1–induced bone resorption in mice in vitro. J Bone
tute: Charting a course for genomic medicine from base pairs to Miner Res 6:183–190, 1991.
bedside. Nature 470:204–213, 2011. 35. Akatsu T, Takahashi N, Udagawa N, et al: Parathyroid hormone
8. Griffiths AJF, Wessler SR, Carroll SB, et al: Introduction to genetic (PTH)–related protein is a potent stimulator of osteoclast-like
analysis, Winslow S, editor, ed 10, New York, 2011, WH Freeman. multi-nucleated cell formation to the same extent as PTH in
9. International Human Genome Sequencing Consortium: Finish- mouse marrow cultures. Endocrinology 125:20–27, 1989.
ing the euchromatic sequence of the human genome. Nature 36. Amizuka N, Davidson D, Liu H, et al: Signalling by fibroblast
431:931–945, 2004. growth factor receptor 3 and parathyroid hormone-related
10. International Human Genome Sequencing Consortium: Initial peptide coordinate cartilage and bone development. Bone 34:13–
sequencing and analysis of the human genome. Nature 409:860– 25, 2004.
921, 2001. 37. Aubin JE, Liu F: The osteoblast lineage. In Bilezikian JP,
11. Kennedy S, Oswald N: PCR troubleshooting and optimization: the Raisz LG, Rodan GA, editors: Principles of bone biology, San Diego,
essential guide, Norfolk UK, 2011, Caister Academic Press. 1996, Academic.
12. Koboldt DC, Steinberg KM, Larson DE, et al: The next- 38. Aubin JE, Liu F, Malaval L, et al: Osteoblast and chondroblast
generation sequencing revolution and its impact on genomics. Cell differentiation. Bone 17(2 Suppl):77S–83S, 1995.
155:27–38, 2013. 39. Berger U, Wilson P, McClelland RA, et al: Immunocytochemical
13. Kolch W, Pitt A: Functional proteomics to dissect tyrosine kinase detection of 1α,25-dihydroxyvitamin D receptors in normal
signalling pathways in cancer. Nat Rev Cancer 10:618–629, 2010. human tissues. J Clin Endocrinol Metab 67:607–613, 1988.
14. Kubista M, Andrade JM, Bengtsson M, et al: The real-time poly- 40. Bilezikian JP, Raisz LG, Martin TJ: Principles of bone biology, ed 3,
merase chain reaction. Mol Aspects Med 27(2–3):95–125, 2006. vol 2, San Diego, 2008, Elsevier.
15. Kuijjer ML, Rydbeck H, Kresse SH, et al: Identification of osteo- 41. Bostrom MPG, Lane JM, Berberian WS, et al: Immunolocaliza-
sarcoma driver genes by integrative analysis of copy number and tion and expression of bone morphogenetic proteins 2 and 4 in
gene expression data. Genes Chromosomes Cancer 51(7):696–706, fracture healing. J Orthop Res 13:357–367, 1995.
2012. 42. Bovee JV, van den Broek LJ, Cleton-Jansen AM, et al:
16. Ley TJ, Mardis ER, Ding L, et al: DNA sequencing of a cytoge- Up-regulation of PTHrP and Bcl-2 expression characterizes the
netically normal acute myeloid leukaemia genome. Nature progression of osteochondroma towards peripheral chondrosar-
456:66–72, 2008. coma and is a late event in central chondrosarcoma. Lab Invest
17. Liesenfeld DB, Habermann N, Owen RW, et al: Review of mass 80:1925–1934, 2000.
spectrometry-based metabolomics in cancer research. Cancer Epi- 43. Buckwalter JA, Rosenberg LC: Electron microscopic studies
demiol Biomarkers Prev 22(12):2182–2201, 2013. of cartilage proteoglycans. Electron Microscopy Rev 1:87–112,
18. Ma X, Wang YW, Zhang MQ, et al: DNA methylation data analy- 1988.
sis and its application to cancer research. Epigenomics 5:301–316, 44. Carey DE, Liu X: Expression of bone morphogenetic protein-6
2013. messenger RNA in bovine growth plate chondrocytes of different
19. Mardis ER: Next-generation DNA sequencing methods. Annu size. J Bone Miner Res 10:401–405, 1995.
Rev Genomics Hum Genet 9:387–402, 2008. 45. Celeste AJ, Iannazzi JA, Taylor RC, et al: Identification of trans-
20. Meyerson M, Gabriel S, Getz G: Advances in understanding forming growth factor beta family members present in bone-
cancer genomes through second-generation sequencing. Nat Rev inductive protein purified from bovine bone. Proc Natl Acad Sci
Genet 11:685–696, 2010. USA 87:9843–9847, 1990.
21. Morgan GJ, Walker BA, Davies FE: The genetic architecture of 46. Celeste AJ, Song JJ, Cox K, et al: Bone morphogenetic protein-9,
multiple myeloma. Nat Rev Cancer 12:335–348, 2012. a new member of the TGF-β superfamily. J Bone Miner Res
22. Morin RD, Zhao Y, Prabhu AL, et al: Preparation and analysis of 9:S137, 1994.
microRNA libraries using the Illumina massively parallel sequenc- 47. Centrella M, Rosen V, Horowitz MC, et al: Transforming growth
ing technology. Methods Mol Biol 50:173–199, 2010. factor-β gene family members, their receptors, and bone cell func-
23. National Research Council: Mapping and sequencing the human tion. Endocrine Rev 4:211–226, 1995.
genome, Washington, D.C., 1988, National Academy Press. 48. Chang SC, Hoang B, Thomas JT, et al: Cartilage derived mor-
24. Pareek CS, Smoczynski R, Tretyn A: Sequencing technologies and phogenetic proteins: new members of the transforming growth
genome sequencing. J Appl Genet 52:413–435, 2011. factor-β superfamily predominantly expressed in long bone during
25. Sandberg AA: Cytogenetics and molecular genetics of bone and human embryonic development. J Biol Chem 269:28227–28234,
soft-tissue tumors. Am J Med Genet 115:189–193, 2002. 1994.
26. Spratlin JL, Serkova NJ, Eckhardt SG: Clinical applications of 49. Chen P, Carrington JL, Hammonds RG, et al: Stimulation of
metabolomics in oncology: a review. Clin Cancer Res 15:431–440, chondrogenesis in limb bud mesoderm cells by recombinant
2009. human bone morphogenetic protein 2b (BMP-2b) and modula-
27. Stankiewicz P, Lupski JR: Structural variation in the human tion by transforming growth factor-β1 and β2. Exp Cell Res
genome and its role in disease. Annu Rev Med 61:437–455, 2010. 195:509–515, 1991.
28. The Cancer Genome Atlas Research Network: Comprehensive 50. Chen Q, Johnson DM, Haudenschild DR, et al: Cartilage matrix
genomic characterization defines human glioblastoma genes and protein: expression patterns in chicken, mouse, and human. Ann
core pathways. Nature 455:1061–1068, 2008. NY Acad Sci 785:238–240, 1996.
29. VanGuilder HD, Vrana KE, Freeman WM: Twenty-five years of 51. Chimal-Monroy J, Bravo-Ruiz MT, Diaz de Leon L: Regulation
quantitative PCR for gene expression analysis. Bio Techniques of chondrocyte differentiation by transforming growth factors β1,
44:619–626, 2008. β2, β3, and β5. Ann NY Acad Sci 785:241–244, 1996.
30. Wu W, Choudhry H: Next generation sequencing in cancer research, 52. Cortes M, Baria AT, Schwartz NB: Sulfation of chondroitin
vol 2, Decoding the cancer genome, Springer, 2013, New York. sulfate proteoglycans is necessary for proper Indian hedgehog
signaling in the developing growth plate. Development 136:1697–
Molecular Biology of Bone Formation 1706, 2009.
31. Abe E, Miyaura C, Tanaka H, et al: 1α,25-Dihydroxyvitamin D3 53. Day TF, Yang Y: Wnt and Hedgehog signaling pathways in bone
promotes fusion of alveolar macrophages both by a direct mecha- development. J Bone Joint Surg 90:19–24, 2008.
nism and by a spleen cell–mediated indirect mechanism. Proc Natl 54. Ducy P, Zhang R, Geoffroy V, et al: Osf2/Cbfa1: a transcriptional
Acad Sci USA 80:5583–5587, 1983. activator of osteoblast differentiation. Cell 89:747–754, 1997.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 137

55. Dudley AT, Lyons KM, Robertson EJ: A requirement for bone 81. Otto F, Thornell AP, Crompton T, et al: Cbfa1, a candidate gene
morphogenetic protein-7 during development of the mammalian for cleidocranial dysplasia syndrome, is essential for osteoblast
eye. Genes Dev 9:2795–2807, 1995. differentiation and bone development. Cell 89:765–772, 1997.
56. Dy P, Wang W, Bhattaram P, et al: Sox9 directs hypertrophic 82. Pogue R, Lyons K: BMP signaling in the cartilage growth plate.
maturation and blocks osteoblast differentiation of growth plate Curr Topics Dev Biol 76:1–48, 2006.
chondrocytes. Dev Cell 22:597–609, 2012. 83. Prior HM, Walter MA: SOX genes: architects of development.
57. Eyre DR: The collagens of articular cartilage. Semin Arth Rheum Mol Med 2:405–412, 1996.
21:2–11, 1991. 84. Provot S, Schipani E: Molecular mechanisms of endochondral
58. Francis PH, Richardson MK, Brickell PM, et al: Bone morpho- bone development. Biochem Biophys Res Commun 328:658–665,
genetic proteins and a signalling pathway that controls patterning 2004.
in the developing chick limb. Development 120:209–218, 1994. 85. Rodan GA, Harada SI: The missing bone. Cell 89:677–680,
59. Girasole G, Passeri G, Jilka RL, et al: Interleukin-11: a new cyto- 1997.
kine critical for osteoclast development. J Clin Invest 93:1516- 86. Rodan GA, Martin TJ: Role of osteoblasts in hormonal control
1524, 1994. of bone resorption: a hypothesis. Calcif Tissue Int 33:349–351,
60. Goltzman D: Discoveries, drugs and skeletal disorders. Nat Rev 1981.
Drug Discov 1:784–796, 2002. 87. Rosen V, Cox K, Hattersley G: Bone morphogenetic proteins. In
61. Hartmann C: Transcriptional networks controlling skeletal devel- Bilezikian JP, Raisz LG, Rodan GA, editors: Principles of bone
opment. Curr Opin Genet Dev 19:437–443, 2009. biology, San Diego, 1996, Academic.
62. Hardingham TE, Fosang AJ, Dudhia J: The structure, function 88. Roughley PJ, Lee ER: Cartilage proteoglycans: structure and
and turnover of aggrecan, the large aggregating proteoglycan potential functions. Microsc Res Tech 28:385–397, 1994.
from cartilage. Eur J Clin Chem Clin Biochem 32:249–257, 1994. 89. Rouleau MF, Mitchell J, Goltzman D: Characterization of the
63. Hillarby MC, King KE, Brady G, et al: Localization of gene major parathyroid hormone target cell in the endosteal metaphy-
expression during endochondral ossification. In De Crombrugghe sis of rat long bone. J Bone Miner Res 5:1043–1053, 1990.
B, Horton WA, Olsen BR, et al, editors: Molecular and developmen- 90. Suda T, Takahashi N, Abe E: Role of vitamin D in bone resorp-
tal biology of cartilage, New York, 1996, New York Academy of tion. J Cell Biochem 49:53–58, 1992.
Sciences. 91. Suda T, Takahashi N, Martin TJ: Modulation of osteoclast dif-
64. Horton WA, Degnin CR: FGFs in endochondral skeletal develop- ferentiation. Endocr Rev 13:66–80, 1992.
ment. Trends Endocrinol Metab 20:341–348, 2009. 92. Suda T, Takahashi N, Martin TJ: Modulation of osteoclast
65. Isaksson OG, Nilsson A, Isgaard J, et al: Cartilage as a target differentiation: update 1995. Endocr Rev Monogr 4:266–270,
tissue for growth hormone and insulin-like growth factor I. Acta 1995.
Paediatr Scand 367:137–141, 1990. 93. Suda T, Udagawa N, Takahashi N: Cells of bone: osteoclast gen-
66. Katagiri T, Yamaguchi A, Ikeda T, et al: The non-osteogenic eration. In Bilezikian JP, Raisz LG, Rodan GA, editors: Principles
mouse pluripotent cell line C3H10T1/2 is induced to differen- of bone biology, San Diego, 1996, Academic.
tiate into osteoblastic cells by recombinant human bone mor- 94. Taga T, Hibi M, Hirata Y, et al: Interleukin-6 triggers the associa-
phogenetic protein-2. Biochem Biophys Res Commun 172:295–299, tion of its receptor with a possible signal transducer, gp130. Cell
1990. 58:573–581, 1989.
67. Kishigami S, Mishina Y: BMP signaling and early embryonic pat- 95. Taga T, Kishimoto T: Role of a two-chain IL-6 receptor system
terning. Cytokine Growth Factor Rev 16:265–278, 2005. in immune and hematopoietic cell regulation. Crit Rev Immunol
68. Komori T, Yagi H, Nomura S, et al: Targeted disruption of Cbfa1 11:265–280, 1992.
results in a complete lack of bone formation owing to maturational 96. Tamura T, Udagawa N, Takahashi N, et al: Soluble interleukin-6
arrest of osteoblasts. Cell 89:755–764, 1997. receptor triggers osteoclast formation by interleukin-6. Proc Natl
69. Kronenberg HM: Developmental regulation of the growth plate. Acad Sci USA 90:11924–11928, 1993.
Nature 423:332–336, 2003. 97. Thesingh CW, Burger EH: The role of mesenchyme in embry-
70. Loveridge N, Farquharson C: Studies on growth plate chondro- onic long bones as early deposition site for osteoclast progenitor
cytes in situ: cell proliferation and differentiation. Acta Paediatr cells. Dev Biol 95:429–438, 1983.
82(Suppl 391):42–48, 1993. 98. Thomas JT, Ayad S, Grant ME: Cartilage collagens: strategies for
71. Luo G, Hofmann C, Bronckers ALJJ, et al: BMP-7 is an inducer the study of their organization and expression in the extracellular
of nephrogenesis and is also required for eye development and matrix. Ann Rheum Dis 53:488–496, 1994.
skeletal patterning. Genes Dev 9:2808–2820, 1995. 99. Thornemo M, Jansson ES, Lindahl A: Expression of the ID1 and
72. Lyons KM, Hogan BLM, Robertson EJ: Colocalization of BMP-7 ID3 genes during chondrocyte differentiation. Ann NY Acad Sci
and BMP-2 RNAs suggests that these factors cooperatively 785:337–339, 1996.
mediate tissue interactions during murine development. Mech Dev 100. Triffitt JT: The stem cell of the osteoblast. In Bilezikian JP, Raisz
50:71–83, 1995. LG, Rodan GA, editors: Principles of bone biology, San Diego, 1996,
73. Mackie EJ, Ahmed YA, Tatarczuch L, et al: Endochondral ossifi- Academic.
cation: how cartilage is converted into bone in the developing 101. Udagawa N, Takahashi N, Akatsu T, et al: Origin of osteoclasts:
skeleton. IJBCB 40:46–62, 2008. mature monocytes and macrophages are capable of differentiating
74. Marks SC, Jr, Popoff SN: Bone cell biology: the regulation of into osteoclasts under a suitable microenvironment prepared by
development, structure, and function in the skeleton. Am J Anat bone marrow–derived stromal cells. Proc Natl Acad Sci USA
183:1–44, 1988. 87:7260–7264, 1990.
75. Martin TJ, Ng KW: Mechanisms by which cells of the osteoblast 102. Udagawa N, Takahashi N, Katagiri T, et al: Interleukin (IL)-6
lineage control osteoclast formation and activity. J Cell Biochem induction of osteoclast differentiation depends on IL-6 receptors
56:357–366, 1994. expressed on osteoblastic cells but not on osteoclast progenitors.
76. Muir H: The chondrocyte, architect of cartilage: biomechanics, J Exp Med 182:1461–1468, 1995.
structure, function and molecular biology of cartilage matrix mac- 103. Ueda K, Yamanaka Y, Harada D, et al: PTH has the potential to
romolecules. Bioessays 17:1039–1048, 1995. rescue disturbed bone growth in achondroplasia. Bone 41:13–18,
77. Mundy GR: Peptides and growth regulatory factors in bone. 2007.
Rheum Dis Clin North Am 20:577–588, 1994. 104. Urist MR: Bone: formation by autoinduction. Science 150:893–
78. Nurminskaya M, Linsenmayer TF: Analysis of up-regulated genes 899, 1965.
during chondrocyte hypertrophy. In De Crombrugghe B, Horton 105. Urist MR, Iwata H, Ceccotti PL, et al: Bone morphogenesis in
WA, Olsen BR, et al, editors: Molecular and developmental biology implants of insoluble bone gelatin. Proc Natl Acad Sci USA
of cartilage, New York, 1996, New York Academy of Sciences. 70:3511–3515, 1973.
79. Olson MC, Scott EW, Hack AA, et al: PU 1 is not essential for 106. Väänänen K: Osteoclast function: biology and mechanisms. In
early myeloid gene expression but is required for terminal myeloid Bilezikian JP, Raisz LG, Rodan GA, editors: Principles of bone
differentiation. Immunity 3:703–714, 1995. biology, San Diego, 1996, Academic.
80. Ornitz DM: FGF signaling in the developing endochondral skel- 107. Wagner EF, Karsenty G: Genetic control of skeletal development.
eton. Cytokine Growth Factor Rev 16:205–213, 2005. Curr Opin Genetics Dev 11:527–532, 2001.

ERRNVPHGLFRVRUJ
138 3  Molecular Biology of Bone Tumors

108. Waung JA, Bassett JH, Williams GR: Thyroid hormone metabo- 134. Dietschy T, Shevelev I, Stagljar I: The molecular role of the
lism in skeletal development and adult bone maintenance. Trends Rothmund-Thomson-, RAPADILINO- and Baller-Gerold-gene
Endocrinol Metab 23:155–162, 2012. product, RECQL4: recent progress. Cell Mol Life Sci 64:796–802,
109. Wojcicka A, Bassett JH, Williams GR: Mechanisms of action of 2007.
thyroid hormones in the skeleton. Biochim Biophys Acta 1830:3979– 135. Diller L, Kassel J, Nelson CE, et al: p53 functions as a cell cycle
3986, 2013. control protein in osteosarcomas,. Mol Cell Biol 10:5772–5781,
110. Yamada Y, Horton W, Miyashita T, et al: Expression and structure 1990.
of cartilage proteins. J Craniofacial Genet Dev Biol 11:350–356, 136. Dobashi Y, Sugimura H, Sato A, et al: Possible association of p53
1991. overexpression and mutation with high-grade chondrosarcoma.
111. Yasoda A, Komatsu Y, Chusho H, et al: Overexpression of CNP Diagn Mol Pathol 2:257–263, 1993.
in chondrocytes rescues achondroplasia through a MAPK- 137. Dowdy SF, Hinds PW, Louie K, et al: Physical interaction of the
dependent pathway. Nat Med 10:80–86, 2004. retinoblastoma protein with human D cyclins. Cell 73:449–511,
112. Zelzer E, Olsen BR: The genetic basis for skeletal diseases. Nature 1993.
423:343–348, 2003. 138. Eisenberg MB, Woloschak M, Sen C, et al: Loss of heterozygosity
in the retinoblastoma tumor suppressor gene in skull base chor-
Mechanisms of Malignant Transformation domas and chondrosarcomas. Surg Neurol 47:156–160, 1997.
113. Anderson JL, Denny CT, Tap WD, et al: Pediatric sarcomas: 139. El-Deiry WS, Tokino T, Velculescu VE, et al: WAF-1, a potential
translating molecular pathogenesis of disease to novel therapeutic mediator of p53 tumor suppression. Cell 75:817–825, 1993.
possibilities. Pediatr Res 72:112–121, 2012. 140. Erickson-Johnson MR, Chou MM, Evers BR, et al: Nodular fas-
114. Antillón-Klussmann F, García-Delgado M, Villa-Elizaga I, et al: ciitis: a novel model of transient neoplasia induced by MYH9-
Mutational activation of ras genes is absent in pediatric osteosar- USP6 gene fusion. Lab Invest 91:1427–1433, 2011.
coma. Cancer Genet Cytogenet 79:49–53, 1995. 141. Feinberg AP, Vogelstein B, Droller MJ, et al: Mutation affecting
115. Belchis DA, Meece CA, Benko FA, et al: Loss of heterozygosity the 12th amino acid of the c-Ha-ras oncogene product occurs
and microsatellite instability at the retinoblastoma locus in osteo- infrequently in human cancer. Science 220:1175–1177, 1983.
sarcomas. Diagn Mol Pathol 5:214–219, 1996. 142. Feugeas O, Guriec N, Babin-Biolletot A, et al: Loss of heterozy-
116. Bos JL: Ras oncogene in human cancer: a review. Cancer Res gosity of the RB gene is a poor prognostic factor in patients with
49:4682–4689, 1989. osteosarcoma. J Clin Oncol 14:467–472, 1996.
117. Brosh RM, Jr: DNA helicases involved in DNA repair and their 143. Finlay C, Hinds PW, Levine AJ: The p53 proto-oncogene can act
roles in cancer. Nat Rev Cancer 13:542–558, 2013. as a suppressor of transformation. Cell 57:1083–1093, 1989.
118. Burchill SA, Neal DE, Lunec J: Frequency of H-ras mutations in 144. Fisher C: Soft tissue sarcomas with non-EWS translocations:
human bladder cancer detected by direct sequencing. Br J Urol molecular genetic features and pathologic and clinical correla-
73:516–521, 1994. tions. Virchows Arch 456:153–166, 2010.
119. Carloni G, Venuat AM, Daya-Grosjean L, et al: Integration and 145. Folpe Al, Goldblum JR, Rubin BP, et al: Morphologic and immu-
loss of a single v-Ki-ras gene affects tumorigenic potential of nophenotypic diversity in Ewing family tumors: a study of 66
human osteosarcoma cells. FEBS Lett 229:333–339, 1988. genetically confirmed cases. Am J Surg Pathol 29:1025–1033,
120. Chakraborty AK, Cichutek K, Duesberg PH: Transforming func- 2005.
tion of proto-ras genes depends on heterologous promoters and 146. Forment JV, Kaidi A, Jackson SP: Chromothripsis and cancer:
is enhanced by specific point mutations. Proc Natl Acad Sci USA causes and consequences of chromosome shattering. Nat Rev
88:2217–2221, 1991. Cancer 12:663–670, 2012.
121. Chan JC, Song WT: Activated production of metalloproteinase 147. Friend SH, Horowitz JM, Gerber MR, et al: Deletions of a DNA
in Ki-ras–transformed human osteosarcoma cells. Leukemia sequence in retinoblastomas and mesenchymal tumors: organiza-
8(Suppl 1):S30–S35, 1994. tion of the sequence and its encoded protein. Proc Natl Acad Sci
122. Chandar N, Billig B, McMaster J, et al: Inactivation of p53 gene USA 84:9059–9063, 1987.
in human and murine osteosarcoma cells. Br J Cancer 65:208–214, 148. Friedrich K, Lee L, Leistritz DF, et al: WRN mutations in Werner
1992. syndrome patients: genomic rearrangements, unusual intronic
123. Chen JM, Ferec C, Cooper DN: Transient hypermutability, chro- mutations and ethnic-specific alterations. Hum Genet 128:103–
mothripsis and replication-based mechanisms in the generation of 111, 2010.
concurrent clustered mutations. Mutat Res 750:52–59, 2012. 149. Fujita J, Yoshida O, Yuasa Y, et al: Ha-ras oncogenes are activated
124. Chu WK, Hickson ID: RecQ helicases: multifunctional genome by somatic alterations in human urinary tract tumors. Nature
caretakers. Nat Rev Cancer 9:644–654, 2009. 309:464–466, 1984.
125. Chun SG, Shaeffer DS, Bryant-Greenwood PK: The Werner’s 150. Gallie BL: Retinoblastoma gene mutations in human cancer.
syndrome RecQ helicase/exonuclease at the nexus of cancer and N Engl J Med 330:786–787, 1994.
aging. Hawaii Med J 70:52–55, 2011. 151. Gerald WL, Haber DA: The EWS-WT1 gene fusion in desmo-
126. Cohen JB, Broz SD, Levinson AD: Expression of the Ha-ras plastic small round cell tumor. Semin Cancer Biol 15:197–205,
proto-oncogene is controlled by alternative splicing. Cell 58:461– 2005.
472, 1989. 152. Ginsberg JP, de Alava E, Ladanyi M, et al: EWS-FLI1 and EWS-
127. Cohen JB, Levinson AD: A point mutation in the last intron ERG gene fusions are associated with similar clinical phenotypes
responsible for increased expression and transforming activity for in Ewing’s sarcoma. J Clin Oncol 17:1809–1814, 1999.
the c-Ha-ras oncogene. Nature 334:119–124, 1988. 153. Goodrich DW: The retinoblastoma tumor-suppressor gene, the
128. Coppede F: Prematuring aging syndrome. Adv Exp Med Biol exception that proves the rule. Oncogene 25:5233–5243, 2006.
724:317–331, 2012. 154. Graham C, Chilton-MacNeill S, Zielenska M, et al: The CIC-
129. Cordon-Cardo C, Wartinger D, Petrylak D, et al: Altered expres- DUX4 fusion transcript is present in a subgroup of pediatric
sions of the retinoblastoma gene product: prognostic indicator in primitve round cell sarcomas. Hum Pathol 43:180–189, 2012.
bladder cancer. J Natl Cancer Inst 84:1251–1256, 1992. 155. Halevy O, Michalovitz D, Oren M: Different tumor-derived p53
130. Czerniak B, Cohen GL, Etkind P, et al: Concurrent mutations of mutants exhibit distinct biological activities. Science 250:113–116,
coding and regulatory sequences of the Ha-ras gene in urinary 1990.
bladder carcinomas. Hum Pathol 23:1199–1204, 1992. 156. Hanahan D, Weinberg RA: Hallmarks of cancer: the next genera-
131. De Camargo VP, van de Rijn M, de Alava E, et al: Other targe- tion. Cell 144:646–674, 2011.
table sarcomas. Semin Oncol 36:358–371, 2009. 157. Hanahan D, Weinberg RA: The hallmarks of cancer. Cell 100:57–
132. De Caprio JA, Ludlow JW, Lynch D, et al: The product of the 70, 2000.
retinoblastoma susceptibility gene has properties of a cycle regula- 158. Harashima H, Dissmeyer N, Schnittger A: Cell cycle control
tory element. Cell 58:1085–1095, 1989. across the eukaryotic kingdom. Trends Cell Biol 23:345–356,
133. Di Fiore R, D’Anneo A, Tesoriere G, et al: RB1 in cancer: 2013.
different mechanisms of RB1 inactivation and alterations of 159. Harper JW, Adami GR, Wei N, et al: The p21 cdk–interacting
pRb pathway in tumorigenesis. J Cell Physiol 228:1676–1687, protein CIP-1 is a potent inhibitor of G1 cyclin-dependent kinases.
2013. Cell 75:805–816, 1993.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 139

160. Hassler M, Singh S, Yue WW, et al: Crystal structure of the reti- 187. Liu P, Erez A, Nagamani SC, et al: Chromosome catastrophes
noblastoma protein N domain provides insight into tumor sup- involve replication mechanisms generating complex genomic
pression, ligand interaction, and holoprotein architecture. Mol rearrangements. Cell 146:889–903, 2011.
Cell 28:371–385, 2007. 188. Livingstone LR, White A, Sprouse J, et al: Altered cell cycle arrest
161. Hochegger H, Hegarat N, Pereira-Leal JB: Aurora at the pole and gene amplification potential accompanying loss of wild-type
and equator: overlapping functions of Aurora kinases in the p53. Cell 70:923–935, 1992.
mitotic spindle. Open Biology 3:120185, 2014. 189. Lonardo F, Ueda T, Huvos AG, et al: p53 and MDM2 alterations
162. Holland AJ, Cleveland DW: Chromoanagenesis and cancer: in osteosarcomas: correlation with clinicopathologic features and
mechanisms and consequences of localized, complex chromo- proliferative rate. Cancer 79:1541–1547, 1997.
somal rearrangements. Nat Med 18:1630–1638, 2012. 190. Maher CA, Wilson RK: Chromothripsis and human disease:
163. Hollstein M, Sidransky D, Vogelstein G, et al: p53 mutations in piecing together the shattering process. Cell 148:29–32, 2012.
human cancers. Science 253:49–53, 1991. 191. Matys V, Kel-Margoulis OV, Fricke E, et al: TRANSFAC and its
164. Horowitz JM, Park SH, Bogenmann E, et al: Frequent inactiva- module TRANSCompel: transcriptional gene regulation in
tion of the retinoblastoma anti-oncogene is restricted to a subset eukaryotes. Nucleic Acids Res 34:108–110, 2006.
of human tumor cells. Proc Natl Acad Sci USA 87:2775–2779, 192. Martinez J, Georgoff I, Martinez J, et al: Cellular localization and
1990. cell cycle regulation by a temperature sensitive p53 protein. Genes
165. Hunter T: Oncoprotein networks. Cell 88:333–346, 1997. Dev 5:151–159, 1991.
166. Indovina P, Marcelli E, Casini N, et al: Emerging roles of RB 193. McBride OW, Merry D, Givol D: The gene for human p53 cel-
family: new defense mechanisms against tumor progression. J Cell lular tumor antigen is located on chromosome 17 short arm. Proc
Physiol 228:525–535, 2013. Natl Acad Sci USA 83:130–134, 1986.
167. Italiano A, Sung YS, Zhang L, et al: High prevalence of CIC 194. McKeon C, Thiele CJ, Ross RA, et al: Indistinguishable patterns
fusion with doube-homeobox (DUX4) transcription factors in of protooncogene expression in two distinct but closely related
EWSR1-negative undifferentiated small blue round cell sarcomas. tumors: Ewing’s sarcoma and neuroepithelioma. Cancer Res 48:
Genes Chromosomes Cancer 51:207–218, 2012. 4307–4311, 1988.
168. Jensen MB, Dunn CA, Keijzers G, et al: The helicase and ATPase 195. Mertens F, Panagopoulos I, Mandahl N: Genomic chracteristics
activities of RECQL4 are compromised by mutations reported in of soft tissue sarcomas. Virchows Archiv 456:129–139, 2010.
three human patients. Aging (Albany NY) 4:790–802, 2012. 196. Mertens F, Antonescu CR, Hohenberger P, et al: Translocation-
169. Jinawath N, Morsberger L, Norris-Kirby A, et al: Complex rear- related sarcomas. Semin Oncol 36:312–323, 2009.
rangement of chromsomes 1, 7, 21, 22 in Ewing sarcoma. Cancer 197. Michiels L, Merregaert J: Retroviruses and oncogenes associated
Genet Cytogenet 201:42–47, 2010. with osteosarcomas. Cancer Treat Res 62:7–18, 1993.
170. Joerger AC, Fersht AR: The tumor suppressor p53: from struc- 198. Miller CW, Aslo A, Won A, et al: Alterations of the p53, Rb and
tures to drug discovery. Cold Spring Harbor Perspectives in Biology MDM2 genes in osteosarcoma. J Cancer Res Clin Oncol 122:559–
1–20, 2010. 565, 1996.
171. Jiang Z, Jones R, Liu JC, et al: RB1 and p53 at the crossroad of 199. Mogilner A, Craig E: Towards a quantitative understanding of
EMT and triple-negative breast cancer. Cell Cycle 10:1563–1570, mitotic spindle assembly and mechanics. J Cell Science 123:3435–
2011. 3445, 2010.
172. Jones MJ, Jallepalli PV: Chromothripsis: chromosomes in crisis. 200. Monnat RJ, Jr: Human RECQ helicases: roles in DNA metabo-
Dev Cell 23:908–917, 2012. lism, mutagenesis and cancer biology. Semin Cancer Biol 20:329–
173. Kawamura-Saito M, Yamazaki Y, Kaneko K, et al: Fusion between 339, 2010.
CIC and DUX4 up-regulates PEA3 family genes in Ewing-like 201. Nigro JM, Baker SJ, Preisinger AC, et al: Mutations in the
sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet p53 gene occur in diverse tumor types. Nature 342:705–708,
15:2125–2137, 2006. 1989.
174. Khidr L, Chen PL: RB, the conductor that orchestrates life, death 202. Nilsson M, Domanski HA, Mertens F, et al: Molecular cytoge-
and differentiation. Oncogene 25:5210–5219, 2006. netic characterization of recurrent translocation breakpoints in
175. Kloosterman WP, Cuppen E: Chromothripsis in congenital dis- bizarre parosteal osteochondromatous proliferation (Nora’s
orders and cancer: similarities and differences. Curr Opin Cell Biol lesion). Hum Pathol 35:1063–1069, 2004.
25:341–348, 2013. 203. Oliveira AM, Chou MM, Perez-Atyade AR, et al: Aneurysmal
176. Komuro H, Hayashi Y, Kawamura M, et al: Mutations of the p53 bone cyst: a neoplasm driven by upregulation of the USP6 onco-
gene are involved in Ewing’s sarcomas but not in neuroblastomas,. gene. J Clin Oncol 24:e1, 2006.
Cancer Res 53:5284–5288, 1993. 204. Oliveira AM, Perez-Atayde AR, Dal Cin P, et al: Aneurysmal bone
177. Korbel JO, Campbell PJ: Criteria for inference of chromothripsis cyst variant translocations upregulate USP6 transcription by pro-
in cancer genomes. Cell 152:1226–1236, 2013. moter swapping with the ZNF9, COL1A1, TRAP150, and OMD
178. Kovar H, Auinger A, Jug G, et al: Narrow spectrum of infrequent genes. Oncogene 24:3419–3426, 2005.
p53 mutations and absence of MDM2 amplificaton in Ewing 205. Oliveira AM, Perez-Atayde AR, Inwards CY, et al: USP6 and
tumours. Oncogene 8:2683–2690, 1993. CDH11 oncogenes identify the neoplastic cell in primary aneu-
179. Ladanyi M, Fletcher JA, Dal Cin P: Cytogenetic and molecular rysmal bone cysts and are absent in so-called secondary aneurys-
genetic pathology of soft tissue tumors. In Goldblum JR, Folpe mal bone cysts. Am J Pathol 165:1773–1780, 2004.
AL, Weiss SW, editors: Enzinger & Weiss’s soft tissue tumors, ed 6, 206. Ouyang KJ, Woo LL, Ellis NA: Homologous recombination and
Philadelphia, 2014, Elsevier Saunders, pp 76–109. maintenance of genome integrity: cancer and aging through the
180. Ladanyi M, Cha C, Lewis R, et al: MDM2 gene amplification in prism of human RecQ helicases. Mech Ageing Dev 129:425–440,
metastatic osteosarcoma. Cancer Res 53:16–18, 1993. 2008.
181. Larizza L, Roversi G, Volpi L: Rothmund-Thomson syndrome. 207. Ozaki T, Ikeda S, Kawai A, et al: Alterations of retinoblastoma
Orphanet J Rare Dis 5:1–16, 2010. susceptible gene accompanied by c-myc amplification in human
182. Lau AW, Pringle LM, Quick L, et al: TRE17/ubiquitin-specific bone and soft tissue tumors. Cellular Mol Biol 39:235–242, 1993.
protease 6 (USP6) oncogene translocated in aneurysmal bone cyst 208. Pichierri P, Ammazzalorso F, Bignami M, et al: The Werner syn-
blocks osteoblastic maturation via an autocrine mechanism involv- drome protein: linking the replication checkpoint response to
ing bone morphogenetic protein dysregulation. J Biol Chem 285: genome stability. Aging 3:311–318, 2011.
37111–37120, 2010. 209. Pierron G, Tirode F, Lucchesi C, et al: A new subtype of bone
183. Lazar A, Abruzzo LV, Pollock RE, et al: Molecular diagnosis of sarcoma defined by BCOR-CCNB3 gene fusion. Nat Genet 44:461–
sarcomas. Arch Pathol Lab Med 130:1199–1207, 2006. 466, 2012.
184. Lee WH: The molecular basis of cancer suppression by the reti- 210. Pike AC, Shrestha B, Popuri V, et al: Structure of the human
noblastoma gene. Princess Takamatsu Symp 20:159–170, 1989. RECQ1 helicase reveals a putative strand-separation pin. Proc
185. Lessnick SL, Tos APD, Sorensen PHB, et al: Small round cell Natl Acad Sci USA 106:1039–1044, 2009.
sarcomas. Semin Oncol 36:338–346, 2009. 211. Pompetti F, Rizzo P, Simon RM, et al: Oncogene alterations in
186. Levine AJ, Momand J, Finlay CA: The p53 tumor suppressor primary, recurrent, and metastatic human bone tumors. J Cell
gene. Nature 351:435–456, 1991. Biochem 63:37–50, 1996.

ERRNVPHGLFRVRUJ
140 3  Molecular Biology of Bone Tumors

212. Radig K, Schneider-Stock R, Oda Y, et al: Mutation spectrum of 236. Tubio JM, Estivill X: Cancer: when catastrophe strikes a cell.
p53 gene in highly malignant human osteosarcomas. Gen Diagn Nature 470:476–477, 2011.
Pathol 142:25–32, 1996. 237. Ueda Y, Dockhorn-Dworniczak B, Blasius S, et al: Analysis
213. Reddy EP, Reynolds RK, Santos E, et al: A point mutation is of mutant p53 protein in osteosarcomas and other malignant
responsible for the acquisition of transforming properties by the and benign lesions of bone. J Cancer Res Clin Oncol 119:172–178,
T24 human bladder carcinoma oncogene. Nature 300:149–152, 1993.
1982. 238. Vogelstein B, Kinzler KW: Cancer genes and the pathways they
214. Ried T: Cytogenetics—in color and digitized. N Engl J Med control. Nat Med 10:789–799, 2004.
350:1597–1600, 2004. 239. Vogelstein B, Kinzler KW: p53 function and dysfunction. Cell
215. Righolt C, Mai S: Shattered and stitched chromosomes— 70:523–526, 1992.
chromothripsis and chromoanasynthesis—manifestations of a new 240. Wadayama B, Toguchida J, Shimizu T, et al: Mutation spectrum
chromosome crisis? Genes Chromosomes Cancer 51:975–981, 2012. of the retinoblastoma gene in osteosarcomas. Cancer Res 54:3042–
216. Romeo S, Dei Tos AP: Soft tissue tumors associated with EWSR1 3048, 1994.
translocation. Virchows Arch 456:219–234, 2010. 241. Wang WL, Patel NR, Caragea M, et al: Expression of ERG, an
217. Rossi ML, Ghosh AK, Bohr VA: Roles of Werner syndrome Ets family transcription factor, identifies ERG-rearranged Ewing
protein in protection of genome integrity. DNA Repair (Amst) sarcoma. Mod Pathol 25:1378–1383, 2012.
9:331–344, 2010. 242. Wang L, Bhargava R, Zheng T, et al: Undifferentiated small
218. Rubin SM, Gall AL, Zheng N, et al: Structure of the Rb round cell sarcomas with rare EWS gene fusions: identification
C-terminal domain bound to E2F1-DP1: A mechanism for of a novel EWS-SP3 fusion and of additional cases with the
phosphorylation-induced E2F release. Cell 123:1093–1106, 2005. EWS-ETV1 and EWS-FEV fusions. J Mol Diagn 9:489–509,
219. Sachdeva UM, O’Brien JM: Understanding pRb: toward the nec- 2007.
essary development of targeted treatments for retinoblastoma. 243. Weinberg RA: The biology of cancer, ed 2, New York, 2014, Garland
J Clin Invest 122:425–434, 2012. Science.
220. Sandberg AA, Bridge JA: Updates on cytogenetics and molecular 244. Wu Y: Unwinding and rewinding: double faces of helicase?
genetics of bone and soft tissue tumors: Ewing sarcoma and J Nucleic Acids 140601:2012, 2012.
peripheral primitive neuroectodermal tumors. Cancer Genet Cyto- 245. Yamaguchi T, Toguchida J, Wadayama B, et al: Loss of heterozy-
genet 123:1–26, 2000. gosity and tumor suppressor gene mutations in chondrosarcomas.
221. Seki M, Otsuki M, Ishii Y, et al: RecQ family helicases in genome Anticancer Res 16:2009–2015, 1996.
stability: lessons from gene disruption studies in DT40 cells. Cell 246. Ye Y, Pringle LM, Lau AW, et al: TRE17/USP6 oncogene trans-
Cycle 7:2472–2478, 2008. located in aneurysmal bone cyst induces matrix metalloproteinase
222. Sheikh MS, Li XS, Chen JC, et al: Mechanisms of regulation of production via activation of NF-kappaB. Oncogene 29:3619–3629,
WAF-1/Cip-1 gene expression in human breast carcioma: role of 2010.
p53 dependent and independent signal transduction pathways. 247. Zambrano E, Nose V, Perez-Atayde AR, et al: Distinct chromo-
Oncogene 9:3407–3415, 1994. somal rearrangements in subungual (Dupuytren) exostosis and
223. Shing DC, McMullan DJ, Roberts P, et al: FUS/ERG gene fusions bizarre parosteal osteochondromatous proliferation (Nora lesion).
in Ewing’s tumors. Cancer Res 63:4568–4576, 2003. Am J Surg Pathol 28:1033–1039, 2004.
224. Smneek L, van Heeringen SJ, Koeppel M, et al: Characterization 248. Zhou Y, Li J, Xu K, et al: Further characterization of retinoblas-
of genome-wide p53-binding sites upon stress reponse. Nucleic toma gene–mediated cell growth and tumor suppression in human
Acids Res 36:3639–3654, 2008. cancer cells. Proc Natl Acad Sci USA 91:4165–4169, 1994.
225. Stephens PJ, Greenman CD, Fu B, et al: Massive genomic rear-
rangement acquired in a single catastrophic event during cancer Cell-Cycle and Mitotic Regulation
development. Cell 144:27–40, 2011. 249. Agathocleous M, Harris WA: Metabolism in physiological cell
226. Stratton MR, Campbell PJ, Futreal PA: The cancer genome. proflieration and differentiation. Trends Cell Biol 23:484–492,
Nature 458:719–724, 2009. 2013.
227. Sugimoto M: A cascade leading to premture aging phenotypes 250. Aslam F, Shalhoub V, van Wijnen AJ, et al: Contributions of
including abnormal tumor profiles in Werner syndrome. Int J Mol distal and proximal promoter elements to glucocorticoid regula-
Med 33:247–253, 2014. tion of osteocalcin gene transcription. Mol Endocrinol 9:679–690,
228. Sugimoto M, Furuichi Y, Ide T, et al: Involvement of WRN heli- 1995.
case in immortalization and tumorigenesis by the telomeric crisis 251. Baumbach LL, Marashi F, Plumb M, et al: Inhibition of DNA
pathway. Oncol Lett 2:609–611, 2011. replication coordinately reduces cellular levels of core and H1
229. Sukov WR, Franco MF, Erickson-Johnson M, et al: Frequency of histone mRNAs: requirement for protein synthesis. Biochemistry
USp6 rearrangements in myositis ossificans, brown tumor, and 23:1618–1625, 1984.
cherubism: molecular cytogenetic evidence that a subset of “myo- 252. Bolanos-Garcia VM: Assessment of the mitotic spindle assembly
sitis ossificans-like lesions” are the early phases in the formation checkpoint (SAC) as the target of anticancer therapies. Curr
of soft-tissue aneurysmal bone cyst. Skeletal Radiol 37:321–327, Cancer Drug Targets 9:131–141, 2009.
2008. 253. Bortell R, Owen TA, Bidwell JP, et al: Vitamin D–responsive
230. Sun A, Bagella L, Tutton S, et al: From G0 to S phase: a view of protein-DNA interactions at multiple promoter regulatory ele-
the roles played by the retinoblastoma (Rb) family members in the ments that contribute to the level of rat osteocalcin gene expres-
Rb-E2F pathway. J Cell Biochem 102:1400–1404, 2007. sion. Proc Natl Acad Sci USA 89:6119–6123, 1992.
231. Szuhai K, Cleton-Jansen AM, Hogendoorn PCW, et al: Molecu- 254. Czerniak B, Darzynkiewicz Z, Staiano-Coico L, et al: Expression
lar pathology and its diagnostic use in bone tumors. Cancer Genet- of Ca antigen in relation to cell cycle in cultured human tumor
ics 205:193–204, 2012. cells. Cancer Res 44:4342–4346, 1984.
232. Szuhai K, Ijszenga M, de Jong D, et al: The NFATc2 gene is 255. Czerniak B, Herz F, Wersto R, et al: Expression of H-ras onco-
involved in a novel cloned translocation in a Ewing sarcoma gene p21 protein in relation to the cell cycle of cultured human
variant that couples its function in immunology to oncology. Clin tumor cells. J Am Pathol 126:411–416, 1987.
Cancer Res 15:2259–2268, 2009. 256. Czerniak B, Herz F, Wersto R, et al: Modification of Ha-ras onco-
233. Taparowsky E, Suard Y, Fasano O, et al: Activation of the T24 gene expression and cell cycle progression in the human colonic
bladder carcinoma transforming gene is linked to a single amino cancer cell line HT-29. Cancer Res 47:2826–2830, 1987.
acid change. Nature 300:762–765, 1982. 257. Czerniak B, Herz F, Wersto RP, et al: Asymmetric distribution of
234. Toguchida J, Ishizaki K, Sasaki MS, et al: Chromosomal reorga- oncogene products at mitosis. Proc Natl Acad Sci USA 89:4860–
nization for the expression of recessive mutation of retinoblastoma 4863, 1992.
susceptibility gene in the development of osteosarcoma. Cancer 258. Darzynkiewicz Z, Gong J, Juan G, et al: Cytometry of cyclin
Res 48:3939–3942, 1988. proteins. Cytometry 25:1–13, 1996.
235. Toguchida J, Ishizaki K, Sasaki MS, et al: Preferential mutation 259. Dowdy SF, Hinds PW, Louie K, et al: Physical interaction of the
of paternally derived Rb gene as the initial event in sporadic osteo- retinoblastoma protein with human D cyclins. Cell 73:499–511,
sarcoma. Nature 338:156–158, 1989. 1993.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 141

260. Duncan JS, Turowec JP, Vilk G, et al: Regulation of cell prolifera- A–activated G1 extracts is catalyzed by cdk–cyclin A complex and
tion and survival: convergence of protein kinases and caspases. DNA-dependent protein kinase. Proc Natl Acad Sci USA 91:8343–
Biochim Biophys Acta 1804:505–510, 2010. 8347, 1994.
261. Duronio RJ, Xiong Y: Signaling pathways that control cell prolif- 286. Pauli U, Chrysogelos S, Stein G, et al: Protein-DNA interactions
eration. Cold Spring Harbor Perspec Biol 5:a008904, 2013. in vivo upstream of cell cycle regulated human H4 histone gene.
262. Frenkel B, Montecino M, Green J, et al: Basal and vitamin D– Science 236:1308–1311, 1987.
responsive activity of the rat osteocalcin promoter in stably trans- 287. Satyanarayana A, Kaldis P: Mammalian cell-cycle regulation:
fected osteosarcoma cells: requirement of upstream sequences several Cdks, numerous cyclins and diverse compensatory mecha-
for control by the proximal regulatory domain. Endocrinology nisms. Oncogene 28:2925–2939, 2009.
137:1080–1088, 1996. 288. Schule R, Umesono K, Mangelsdorf DJ, et al: Jun-fos and recep-
263. Gabrielli B, Brown M: Histone deacetylase inhibitors disrupt the tors for vitamins A and D recognize a common response element
mitotic spindle assembly checkpoint by targeting histone and non- in the human osteocalcin gene. Cell 61:497–504, 1990.
histone proteins. Adv Cancer Re 116:1–37, 2012. 289. Serrano M, Hannon GJ, Beach D: A new regulatory motif in cell-
264. Girard F, Strausfeld U, Fernandez A, et al: Cyclin A is required cycle control causing specific inhibition of cyclin D/CDK4.
for the onset of DNA replication in mammalian fibroblasts. Cell Nature 366:704–707, 1993.
67:1169–1179, 1991. 290. Sherr CJ: G1 phase progression: cycling on cue. Cell 79:551–555,
265. Gong J, Traganos F, Darzynkiewicz Z: Threshold expression of 1994.
cyclin E but not D type cyclins characterizes normal and tumour 291. Stevermann L, Liakopoulos D: Molecular mechanisms in spindle
cells entering S phase. Cell Prolif 28:337–346, 1995. positioning: structures and new concepts. Curr Opin Cell Biol
266. Hallstrom TC, Nevins JR: Balancing the decision of cell profliera- 24:816–824, 2012.
tion and cell fate. Cell Cycle 8:532–535, 2009. 292. Vitre BD, Cleveland DW: Centrosomes, chromosome instability
267. Herz F, Czerniak B, Deitch D, et al: Protein expression in relation (CIN) and aneuploidy. Curr Opin Cell Biol 24:809–815, 2012.
to the cell cycle of exponentially growing human prostatic epithe- 293. Wang Y, Blelloch R: Cell cycle regulation by microRNAs in stem
lial cells. Cell Prolif 24:321–330, 1991. cells. Results Probl Cell Differ 53:459–472, 2011.
268. Hornick JE, Karanjeet K, Collins ES, et al: Kinesins to the 294. Wordeman L: How kinesin motor proteins drive mitotic spindle
core: the role of microtubule-based motor proteins in building function: lessons from molecular assays. Semin Cell Devel Biol
the mitotic spindle midzone. Semin Cell Devel Biol 21:290–299, 21:260–268, 2010.
2010.
269. Kelly KR, Ecsedy J, Mahalingam D, et al: Targeting aurora Molecular Concepts of Bone Neoplasia
kinases in cancer treatment. Curr Drug Targets 12:2067–2078, 295. Abramson DH, Ellsworth RM, Kitchin FD, et al: Second nonocu-
2011. lar tumors in retinoblastoma survivors: are they radiation induced?
270. Kerner SA, Scott RA, Pike JW: Sequence elements in the human Ophthalmology 91:1351–1355, 1984.
osteocalcin gene confer basal activation and inducible response to 296. Bauer HC, Kreicbergs A, Silfversward C, et al: DNA analysis in
hormonal vitamin D3. Proc Natl Acad Sci USA 86:4455–4459, the differential diagnosis of osteosarcoma. Cancer 61:2532–2540,
1989. 1988.
271. Koff A, Ohtsuki M, Polyack K, et al: Negative regulation of G1 297. Berger M, Dirksen U, Braeuninger A, et al: Genomic EWS-FLI1
in mammalian cells: inhibition of cyclin E–dependent kinase by fusion sequences in Ewing sarcoma resemble breakpoint charac-
TGF-β. Science 260:536–539, 1993. teristics of immature lymphoid malignancies. PLoS ONE 8:e56408,
272. Kuno H, Kurian SM, Hendy GN, et al: Inhibition of 1,25- 2013.
dihydroxyvitamin D3 stimulated osteocalcin gene transcription by 298. Blau O: Molecular investigation of Ewing sarcoma: about detect-
tumor necrosis factor-alpha: structural determinants within the ing translocations. EMBO Molec Med 4:449–450, 2012.
vitamin D response element. Endocrinology 134:2524–2531, 1994. 299. Boeuf S, Bovee JVMG, Lehner B, et al: BMP and TGFβ pathways
273. Lens SM, Voest EE, Medema RH: Shared and separate functions in human central chondrosarcoma: enhanced endoglin and Smad
of polo-like kinases and Aurora kinases in cancer. Nature Rev 1 signaling in high grade tumors. BMC Cancer 12:488, 2012.
Cancer 10:825–841, 2010. 300. Bovee JVMG, Hogendoorn PCW, Wunder JS, et al: Cartilage
274. Lian JB, Stewart C, Puchacz E, et al: Structure of the rat osteo- tumours and bone development: molecular pathology and possible
calcin gene and regulation of vitamin D–dependent expression. therapeutic targets. Nature Rev Cancer 10:481–488, 2010.
Proc Natl Acad Sci USA 86:1143–1147, 1989. 301. Bovee JVMG, Cleton-Jansen AM, Taminiau AHM, et al: Emerg-
275. Libertini S, Abagnale A, Passaro C, et al: Aurora A and B kinases— ing pathways in the development of chondrosarcoma of bone and
targets of novel anticancer drugs. Recent Patents on Anti-Cancer implications for targeted treatment. Lancet Oncol 6:599–607, 2005.
Drug Discovery 5:219–241, 2010. 302. Bridge JA: Cytogenetic and molecular cytogenetic techniques in
276. Liu H, Jiang D, Chi F, et al: The Hippo pathway regulates stem orthopaedic surgery. J Bone Joint Surg 75A:606–614, 1993.
cell proliferation, self-renewal, and differentiation. Protein Cell 303. Campbell V, Nadesan P, Ali SA, et al: Hedghog pathway inhibi-
3:291–304, 2012. tion in chondrosarcoma using the smoothened inhibitor IPI-926
277. Lukasiewicz KB, Lingle WL: Aurora A, centrosome structure, directly inhibits sarcoma cell growth. Mol Cancer Ther 2014. (epub
and the centrosome cycle. Environm Molec Mutagen 50:602–619, ahead of print).
2009. 304. Coughlan B, Feliz A, Ishida T, et al: p53 expression and DNA
278. MacLachlan TK, Sang N, Giordano A: Cyclins, cyclin-dependent ploidy of cartilage lesions. Hum Pathol 26:620–624, 1995.
kinases and Cdk inhibitors: implications in cell cycle control and 305. Cumin I, Cohen JY, David A, et al: Rothmund-Thomson syn-
cancer. Crit Rev Eukaryot Gene Expr 5:127–156, 1995. drome and osteosarcoma. Med Pediatr Oncol 26:414–416, 1996.
279. Malumbres M: Physiological relevance of cell cycle kinases. Physiol 306. Deng FM, Galvan K, de la Roza G, et al: Molecular characteriza-
Rev 91:973–1007, 2011. tion of an EWSR1-POU5F1 fusion associated with a t(6;22) in an
280. Mazzoni C, Falcone C: mRNA stability and control of cell pro- undifferentiated soft tissue sarcoma. Cancer Genet 204:423–429,
liferation. Biochem Soc Trans 39:1461–1465, 2011. 2011.
281. Mogilner A, Craig E: Towards a quantitative understanding of 307. Der Kinderen DJ, Koten JW, Wolterbeck R, et al: Nonocular
mitotic spindle assembly and mechanics. J Cell Sci 123:3435–3445, cancer in hereditary retinoblastoma survivors and relatives.
2010. Ophthal Paediatr Genet 8:23–25, 1987.
282. Moncada S, Higgs EA, Colombo SL: Fulfilling the metabolic 308. Dryja TP, Friend S, Weinberg RA: Genetic sequences that pre-
requirements for cell proliferation. Biochem J 446:1–7, 2012. dispose to retinoblastoma and osteosarcoma. Symp Fund Cancer
283. Nigg EA: Targets of cyclin-dependent protein kinases. Curr Opin Res 39:115–119, 1986.
Cell Biol 5:187–193, 1993. 309. Feugeas O, Guriec N, Babin-Boilletot A, et al: Loss of heterozy-
284. Noatynska A, Gotta M, Meraldi P: Mitotic spindle (DIS)orienta- gosity of the Rb gene is a poor prognostic factor in patients with
tion and DISease: cause or consequence? J Cell Biol 199:1025– osteosarcoma. J Clin Oncol 14:467–472, 1996.
1035, 2012. 310. Fletcher JA, Gebhardt MC, Kosakewich HP: Cytogenetic aber-
285. Pan ZQ, Amin AA, Gibbs E, et al: Phosphorylation of the p34 rations in osteosarcomas: nonrandom deletions, rings, and double-
subunit of human single-stranded, DNA-binding protein in cyclin minute chromosomes. Cancer Genet Cytogenet 77:81–88, 1994.

ERRNVPHGLFRVRUJ
142 3  Molecular Biology of Bone Tumors

311. Forus A, Weghuis DO, Smeets D, et al: Comparative genomic 335. Lynn M, Wang Y, Slater J, et al: High-resolution genome-wide
hybridization analysis of human sarcomas. II. Identification of copy-number analyses identify localized copy-number alterations
novel amplicons at 6p and 17p in osteosarcomas. Genes Chromo- in Ewing sarcoma. Diagn Mol Pathol 22:76–84, 2013.
somes Cancer 14:15–21, 1995. 336. Mackintosh C, Madoz-Gurpide J, Ordonez JL, et al: The molecu-
312. Frebourg T, Barbier N, Yan YX, et al: Germ-line p53 mutations lar pathogenesis of Ewing sarcoma. Cancer Biol Ther 9:655–667,
in 15 families with Li-Fraumeni syndrome. Am J Hum Genet 2010.
56:608–615, 1995. 337. Mandahl N, Baldetorp B, Femö M, et al: Comparative cytoge-
313. Hansen MF: Molecular genetic considerations in osteosarcoma. netic and DNA flow cytometric analysis of 150 bone and soft
Clin Orthop 270:237–246, 1991. tissue tumors. Int J Cancer 53:258–364, 1993.
314. Hecht JT, Hogue D, Wang Y, et al: Hereditary multiple 338. Mankin HJ, Connor JF, Schiller AL, et al: Grading of bone
exostoses (EXT): mutational studies of familial EXT1 cases tumors by analysis of nuclear DNA content using flow cytometry.
and EXT-associated malignancies. Am J Hum Genet 60:80–86, J Bone Joint Surg 67A:404–413, 1985.
1997. 339. McIntyre JF, Smith-Sorensen B, Friend SH, et al: Germline
315. Helio H, Karaharju E, Nordling S: Flow cytometric determina- mutations of the p53 suppressor gene in children with osteosar-
tion of DNA content in malignant and benign bone tumors. coma. J Clin Oncol 12:925–930, 1994.
Cytometry 6:165–171, 1985. 340. Meijer D, de Jong D, Pansuriya TC, et al: Genetic characteriza-
316. Hiddemann W, Roessner A, Wormann B, et al: Tumor heteroge- tion of mesenchymal, clear cell, and dedifferentiated chondrosar-
neity in osteosarcoma as identified by flow cytometry. Cancer coma. Genes Chromosomes Cancer 51:899–909, 2012.
59:324–328, 1987. 341. Mertens F, Mandahl N, Örndal C, et al: Cytogenetic findings in
317. Ho L, Stojanovski A, Whetstone H, et al: Gli2 and p53 cooperate 33 osteosarcomas. Int J Cancer 55:44–50, 1993.
to regulate IGFBP-3-mediated chondrocyte apoptosis in the pro- 342. Mirzayans R, Enns L, Dietrich K, et al: Faulty DNA polymerase
gression from benign to malignant cartilage tumors. Cancer Cell delta/epsilon-mediated excision repair in response to gamma
16:126–136, 2009. radiation of ultraviolet light in p53-deficient fibroblast strains
318. Issing WJ, Wustrow TP, Oeckler R, et al: An association of the from affected members of a cancer-prone family with Li-Fraumeni
Rb gene with osteosarcoma: molecular genetic evaluation of a case syndrome. Carcinogenesis 17:691–698, 1996.
of hereditary retinoblastoma. Eur Arch Otorhinolaryngol 250:277– 343. Moller E, Hornick JL, Magnusson L, et al: FUS-CREB3L2/
280, 1993. L1-positive sarcomas show a specific gene expression profile with
319. Jahromi MS, Putnam AR, Druzgal C, et al: Molecular inversion upregulation of CD24 and FOXL1. Clin Cancer Res 17:2646–2656,
probe analysis detects novel copy number alterations in Ewing 2011.
sarcoma. Cancer Genet 205:391–404, 2012. 344. Muller CW, Herrmann BG: Crystallographic structure of the T
320. Jinawath N, Morsberger L, Norris-Kirby A, et al: Complex rear- domain-DNA complex of the Brachyury transcription factor.
rangement of chromosomes 1, 7, 21, 22 in Ewing sarcoma. Cancer Nature 389:884–888, 1997.
Genet Cytogenet 201:42–47, 2010. 345. Nawa G, Ueda T, Mori S, et al: Prognostic significance of Ki67
321. Judge MR, Kilby A, Harper JI: Rothmund-Thomson syndrome (MIB1) proliferation index and p53 over-expression in chondro-
and osteosarcoma. Br J Dermatol 129:723–725, 1993. sarcoma. Int J Cancer 69:86–91, 1996.
322. Kreicbergs A, Silfversward C, Tribukait B: Flow DNA analysis 346. Nelson AC, Pillay N, Henderson S, et al: An integrated functional
of primary bone tumors: relationship between cellular DNA genomics approach identifies the regulatory network directed by
content and histopathologic classification. Cancer 53:129–136, brachyury (T) in chordoma. J Pathol 228:274–285, 2012.
1984. 347. Newby R, Rowe D, Paterson L, et al: Cryptic EWSR1-FLI1
323. Kuijjer ML, Hogendoorn PCW, Cleton-Jansen AM: Genome- fusions in Ewing sarcoma: potential pitfalls in the diagnostic use
wide analyses on high-grade osteosarcoma: making sense of of fluorescence in situ hybridization probes. Cancer Genet Cyto-
genomically most unstable tumor. Int J Cancer 133:2512–2521, genet 200:60–64, 2010.
2013. 348. Novello C, Pazzaglia L, Cingolani C, et al: miRNA expression
324. Kuijjer ML, Rydbeck H, Kresse SH, et al: Identification of osteo- profile in human osteosarcoma: role of miR-1 and miR-133b in
sarcoma driver genes by integrative analysis of copy number and proliferation and cell cycle control. Int J Oncol 42:667–675, 2013.
gene expression data. Genes Chromosomes Cancer 51:696–706, 349. Pansuriya TC, van Eijk R, d’Adamo P, et al: Somatic mosaic IDH1
2012. and IDH2 mutations are associated with enchondroma and spindle
325. Larralde M, Morales MS, Gonzalez JA, et al: What syndrome is cell hemangioma in Ollier disease and Maffucci syndrome. Nature
this? Rothmund-Thomson syndrome. Pediatr Dermatol 12:277– Genet 43:1256–1261, 2011.
280, 1995. 350. Papachristou DJ, Papavassiliou AG: Osteosarcoma and chondro-
326. Larramendy ML, Tarkkanen M, Valle J, et al: Gains, losses, and sarcoma: new signaling pathways as targets for novel therapeutic
amplifications of DNA sequences evaluated by comparative interventions. Int J Biochem Cell Biol 39:857–862, 2007.
genomic hybridization in chondrosarcomas. Am J Pathol 150:685– 351. Papachristou DJ, Papachristou GJ, Papefthimiou OA, et al: The
691, 1997. MAPK-AP-1/-Runx2 signalling axes are implicated in chondro-
327. Lessnick SL, Ladanyi M: Molecular pathogenesis of Ewing sarcoma pathobiology either independently or via up-regulation
sarcoma: new therapeutic and transcriptional targets. Annu Rev of VEGF. Histopathology 47:565–574, 2005.
Pathol Mech Dis 7:145–159, 2012. 352. Sandberg AA: Genetics of chondrosarcoma and related tumors.
328. Li JP, Liu LH, Li J, et al: Microarray expression profile of long Curr Opin Oncol 16:342–354, 2004.
noncoding RNAs in human osteosarcoma. Biochem Biophys Res 353. Sankar S, Lessnick SL: Promiscuous partnerships in Ewing’s
Commun 433:200–206, 2013. sarcoma. Cancer Genet 204:351–365, 2011.
329. Li FP, Fraumeni JF, Jr: Soft tissue sarcomas, breast cancer, and 354. Scheil-Bertram S, Kappler R, Von Baer A, et al: Molecular profil-
other neoplasms: a familial syndrome? Ann Intern Med 71:747– ing of chordoma. Int J Oncol 44:1041–1055, 2014.
752, 1969. 355. Simms WW, Ordóñez NG, Johnston D, et al: p53 expression in
330. Li FP, Fraumeni JF, Jr: Prospective study of a family cancer syn- dedifferentiated chondrosarcoma. Cancer 76:223–227, 1995.
drome. JAMA 247:2692–2694, 1982. 356. Stoll G, Surdez D, Tirode F, et al: Systems biology of Ewing
331. Li FP, Fraumeni JF, Jr, Mulvihill JJ, et al: A cancer family syn- sarcoma: a network model of EWS-FLI1 effect on proliferation
drome in 24 kindreds. Cancer Res 48:5358–5362, 1988. and apoptosis. Nucleic Acids Res 41:8853–8871, 2013.
332. Liang X, Wang D, Wang Y, et al: Expression of Aurora Kinase A 357. Swarts SJ, Neff JR, Johansson SL, et al: Cytogenetic analysis of
and B in chondrosarcoma and its relationship with the prognosis. dedifferentiated chondrosarcoma. Cancer Genet Cytogenet 89:49–
Diagn Pathol 7:84, 2012. 51, 1996.
333. Liu PK, Kraus E, Wu TA, et al: Analysis of genomic instability in 358. Tarkkanen M, Karhu R, Kallioniemi A, et al: Gains and losses of
Li-Fraumeni fibroblasts with germline p53 mutations. Oncogene DNA sequences in osteosarcomas by comparative genomic
12:2267–2278, 1996. hybridization. Cancer Res 55:1334–1338, 1995.
334. Lopez-Gines C, Carda-Batalla C, Lopez-Terrada L, et al: Pres- 359. Tarkkanen M, Wiklund T, Virolainen M, et al: Dedifferentiated
ence of double minutes and monosomy 17p in xenografted human chondrosarcoma with t(9;22)(q34;q11-12). Genes Chromosomes
osteosarcomas. Cancer Genet Cytogenet 90:57–62, 1996. Cancer 9:136–140, 1994.

ERRNVPHGLFRVRUJ
3  Molecular Biology of Bone Tumors 143

360. Trougakos IP, Chondrogianni N, Amarantos I, et al: Genome- 362. Yoshitaka T, Kawai A, Miyaki S, et al: Analysis of microRNAs
wide transcriptome profile of the human osteosarcoma Sa OS, and expressions in chondrosarcoma. J Ortho Res 31:1992–1998,
U-2 OS cell lines. Cancer Genet Cytogenet 196:109–118, 2010. 2013.
361. Yamaguchi T, Tolguchida J, Yamamuro T, et al: Allelotype analysis 363. Zou C, Shen J, Tang Q, et al: Cancer-testis antigens expressed in
in osteosarcomas: frequent allele loss on 3q, 13q, 17p, and 18q. osteosarcoma identified by gene microarray correlate with a poor
Cancer Res 52:2419–2423, 1992. patient prognosis. Cancer 118:1845–1855, 2012.

ERRNVPHGLFRVRUJ
C H A P T E R 4 

Benign Osteoblastic Tumors


CHAPTER OUTLINE

OSTEOID OSTEOMA AGGRESSIVE OSTEOBLASTOMA


BENIGN OSTEOBLASTOMA

Benign osteoblastic tumors were first recognized as a lesions have been referred to by a variety of terms, includ-
distinct group by Jaffe and Mayer in 1932.54 The identi- ing aggressive osteoblastoma,42 malignant osteoblastoma,110 or
fication of osteoid osteoma as a separate entity came even low-grade osteosarcoma.15,78,79,82,84,98 Therefore there
later in a 1935 report by Jaffe.52 Once osteoid osteoma are three main categories of benign osteoblastic tumors:
had been established as a diagnostic category, other osteoid osteoma, benign osteoblastoma, and aggressive
benign bone-forming lesions were recognized. The term osteoblastoma. However, benign osteoblastic tumors
benign osteoblastoma was introduced independently by represent a continuum of lesions that have different
Jaffe and Lichtenstein in 1956 to delineate a benign growth potentials and different levels of extrinsic humoral
osteoblastic tumor that has greater growth potential than activity. This results in a mosaic of lesions that vary con-
osteoid osteoma.53,70,71 Most authors now regard osteoid siderably in size and in their ability to induce secondary
osteoma and osteoblastoma as separate, though related, changes in the adjacent tissue (Fig. 4-1). In such lesions,
entities.44,71 In the majority of cases, fairly consistent hemorrhage, secondary reparative changes, and bizarre
differences in size, location, degree of reactive sclerosis, degenerating nuclei can raise the suspicion of malignancy
and pain pattern are useful features for selecting the (“pseudoanaplasia”).9,83
proper diagnostic category.46 However, there are cases Almost a century after the first conceptual identifica-
with composite features that defy classification. More- tion of this group of bone forming tumors, relatively little
over, a few cases of transition from osteoid osteoma is known about their biology. Cytogenetic studies on
to osteoblastoma have been reported, supporting the a few benign osteoblastic tumors have demonstrated
concept of a close relationship between these two clonal chromosomal alterations involving 22q in osteoid
lesions.16,17,25,122 It is important to realize that osteoid osteoma, benign osteoblastoma, and aggressive osteo-
osteomas and osteoblastomas have nearly identical micro- blastoma.11,34 A single cytogenetic report on an aggressive
scopic features and must be distinguished from one osteoblastoma showed a distinct pseudodiploid clone
another by some means other than microscopy. Several with balanced translocations involving chromosomes 4,
arbitrary but still useful diagnostic criteria have been 7, and 14.10 The study of alterations in cell cycle regulator
proposed to resolve this diagnostic dilemma. Maximum genes such as mutations in the TP53 and RAS genes, loss
diameters for the osteoid osteoma nidus of 1 and 2 cm of heterozygosity at the p53, p16, and Rb-locus, and
have been proposed as criteria.110 McLeod et al.81 arbi- amplification of the MDM2 gene and the c-myc gene, as
trarily designated all lesions measuring less than 1 cm in indicators of graded genetic instability in borderline
diameter as osteoid osteomas and those larger than 2 cm osteoblastic tumors, may prove useful in their differential
in diameter as osteoblastomas. These authors also pro- diagnosis.100 A recent study of genomic aberrations in
posed an arbitrary dividing line of 1.5 cm for lesions with several osteoblastomas and two examples of aggressive
composite features. osteoblastoma disclosed multiple chromosomal rear-
More recent experience with benign osteoblastic rangements in aggressive osteoblastomas.93 Moreover,
tumors has indicated that some lesions may reach a con- deletions of 22q were seen in several conventional osteo-
siderable size, usually exceeding 4 cm in diameter. These blastomas and in aggressive osteoblastomas, confirming
lesions have a locally destructive growth pattern and a the relationship between these two entities. The chromo-
high recurrence rate after curettage. Although the basic somal alterations, especially those involving 22q, involve
histologic pattern of these tumors is similar to that of the genes controlling osteogenesis and are implicated to
conventional osteoblastoma, the presence of so-called have a role in the development of several solid and hema-
epithelioid osteoblasts is their distinct diagnostic topoietic malignancies. The genomic alterations point
feature.1,42,59,60 We have designated these tumors as aggres- toward the involvement of MN1 and NF2 as well as
sive osteoblastomas, a term tentatively proposed in 1973.39 ZNRF3 and KREMEN1, known inhibitors of the Wnt/
The more definitive recognition of aggressive osteoblas- beta-catenin signaling pathway. In line with these obser-
toma as an entity was made in a 1984 report by Dorfman vations increased levels of beta-catenin were detected in
and Weiss.42 Such intermediate or borderline osteoblastic osteoblastomas.93
144
ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 145

Local Continuous
Growth

1.5 cm
Benign with
Benign with Increased Size, but
Limited Growth Limited Growth

Bone Expansion
Adjacent bone
Sclerosis of

Size >3 cm
Osteoid Osteoblastoma
Osteoma
Aggressive
Osteoblastoma
FIGURE 4-1  ■  Benign osteoblastic tumors. Comparative features of biologic behavior and growth potential of osteoid osteoma,
benign osteoblastoma, and aggressive osteoblastoma.

OSTEOID OSTEOMA skeletal involvement are shown in Figure 4-2. The skel-
etal and age distribution patterns of individual osteoid
Definition osteomas are shown in Figure 4-3.

Osteoid osteoma is a benign tumor that consists of a


well-demarcated osteoblastic mass called a nidus that is
Clinical Symptoms
surrounded by a distinct zone of reactive bone sclerosis. Osteoid osteoma is associated with characteristic and
The zone of sclerosis is not an integral part of the tumor quite often virtually diagnostic symptoms. The most fre-
and represents a secondary reversible change that gradu- quently reported symptom is pain of increasing severity
ally disappears after removal of the nidus. This zone that is relieved by aspirin and other nonsteroidal anti-
should be specifically excluded in taking measurements inflammatory agents. The typical clinical symptoms
of these lesions. The nidus tissue has a limited local occur in approximately 80% of patients.5,13,48 The pain
growth potential and usually is less than 1 cm in is frequently worse at night and is sometimes referred
diameter. to the nearest joint. If the tumor is located in the
proximity of a joint, the patient may have symptoms of
arthritis. If the involved bone is superficial, painful swell-
Incidence and Location ing of the adjacent soft tissue may be present. This often
Osteoid osteomas are common lesions that account for occurs in the small bones of the hands and feet, where
about 10% to 12% of all benign bone tumors. They osteoid osteoma can be clinically mistaken for an inflam-
usually occur in teenagers and young adults. The male/ matory process. With vertebral involvement, there is
female sex ratio is approximately 2 : 1. Familial occur- often an associated painful scoliosis that results from a
rence has been reported rarely.55 More than 80% of unilateral spasticity of spinal muscles. Some patients
patients are between age 5 and 25 years, and the peak with vertebral lesions may have clinical symptoms sug-
incidence is in the second decade of life. About 50% of gestive of a neurologic disorder, lumbar disk disease, or
all osteoid osteomas occur in the long bones of the lower both.8,62 Lesions of the extremities in young patients with
extremities, and the femoral neck is the single most fre- open growth plates may produce significant length dis-
quent anatomic site. In the long bones, osteoid osteoma crepancy, caused by the increased growth rate of the
is usually located near the end of the shaft. Osteoid affected bone.95 In neglected cases, the patient may have
osteoma occurs less frequently in the long bones of the severe dysfunction of the extremity and muscle atrophy.
upper extremities, and the bones of the elbow are the Some degree of muscle atrophy is present in nearly 20%
most frequent anatomic site in the upper extremity. of cases.
Osteoid osteomas are often present in the small bones of In the past, the presence of nerves within the nidus,
the hands and feet. They occur rarely in the axial skeleton documented by the axonal silver stain and electron
but if present are usually found in the lumbar portion of microscopy, was thought to be the cause of the patient’s
the spine. In vertebrae, they are nearly exclusively located pain.113 More recently, high levels of prostaglandin E2 and
in the posterior arch. The primary location in the verte- prostacyclin have been found directly in the nidus tissue
bral body is very rare. Osteoid osteomas occur very rarely and its explants incubated in vitro.47,76,130 Large amounts
in flat bones and almost never in craniofacial bones.13,36,110 of prostaglandin E2 and prostacyclin released from the
The peak age incidence and the most frequent sites of nidus have since been implicated as being responsible for

ERRNVPHGLFRVRUJ
Peak
incidence
5 25
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 4-2  ■  Osteoid osteoma. Peak age incidence and frequent sites of skeletal involvement. The most frequent site is indicated
by the solid black arrow.

Osteoid Osteoma
30 40
Mean Age: 17.3
35 Male/Female Ratio: 1.56
25
30
Number of Cases

Number of Cases

20
25

15 20

15
10
10
5
5

0 0
Rib

1 2 3 4 5 6 7 8 9
Vertebra

Ilium

Tibia
Humerus
Ulna
Fibula
Radius
Phalanx
Carpal
Tarsal
r al
Femur

Metatars

Age in Decades
B

Flat Major Small bones of


bones long bones hands and feet

Axial Appendicular
skeleton skeleton
A
FIGURE 4-3  ■  Skeletal sites and age frequency distribution patterns of osteoid osteoma. A, Skeletal distribution of osteoid osteoma.
B, Age frequency distribution of osteoid osteoma.
ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 147

reactive sclerosis, nonspecific inflammatory changes of minimal or even completely absent. The nidus is rarely
soft tissue, and pain in osteoid osteomas.47 Moreover, it surrounded by normal cancellous bone with no evidence
has been documented that the urinary excretion rate of of sclerosis. Sometimes, it may be difficult to identify the
the major metabolite of prostacyclin (i.e., 2,3-dinor-6- nidus grossly, particularly in sclerotic intracortical lesions.
keto-prostaglandin F1α) is significantly higher in patients Preoperative tetracycline labeling is therefore of great
with osteoid osteoma and returns to normal after the assistance because the nidus tissue stands out from the
nidus is removed.30 Recent studies on the expression of reactive bone when it is examined under ultraviolet
the cyclooxygenases (COX-1 and COX-2), which were light.6,68 Intraoperative scintillation probes after the
thought to be the source of the excess prostaglandins in administration of a radioisotope may be used to confirm
osteoid osteomas, have shown that COX-2 is one of the identification.64,68,96,124
main mediators of pain and inflammatory effects on sur-
rounding tissues, including adjacent synovium.57,87 Microscopic Findings
At low magnification the nidus consists of an interlacing
Radiographic Imaging network of bone trabeculae with different levels of min-
The radiographic features of osteoid osteoma are eralization (Figs. 4-9 to 4-11). The trabeculae are usually
characteristic and diagnostic. Conventional radiographs thin and uniformly distributed in loose stromal vascular
reveal a well-demarcated lytic lesion (nidus) surrounded connective tissue. The size, thickness, and mineralization
by a distinct zone of sclerosis. A zone of central opacity of the trabeculae can vary considerably in different lesions
that represents a more sclerotic portion of the nidus and (Figs. 4-9 to 4-13). The trabeculae also may vary in dif-
is surrounded by a lucent halo may be present within the ferent areas of the same nidus (Fig. 4-9). The entire nidus
nidus. The intracortical lesions of long bones produce may be composed of densely sclerotic bone tissue with a
extensive fusiform thickening of the cortex with dense disordered (pagetoid) pattern of cement lines. Prominent
radiopacity that sometimes obscures the nidus (Figs. 4-4 osteoblasts rim the osteoid trabeculae and are accompa-
and 4-5). In such instances the nidus may not be visible nied by numerous osteoclast-like, multinucleated giant
on conventional radiographs, so additional imaging tech- cells. The nidus may have a clearly recognizable zonal
niques, such as computed tomography, radioisotope scan- architecture with a more completely ossified central
ning, and magnetic resonance imaging, may be necessary portion (Fig. 4-9). The central portion, which is heavily
to document the lesion.18,49,50,75,123,129 Occasionally, several ossified, is usually less cellular compared with the less
distinct foci of osteoid osteoma occur at the same ana- mineralized and highly cellular peripheral zone. As a rule,
tomic site, giving the appearance of a multifocal nidus.7 cartilage is not an integral component of the nidus and
The tumors may exhibit different levels of matrix miner- should not be present in typical osteoid osteoma. In some
alization and may have different degrees of radiographic exceptional cases, however, small foci of clearly recogniz-
density and scintigraphic activity.48 The adjacent perios- able cartilage with occasional myxoid features may be
teal reaction sometimes mimics a stress fracture. The present within the nidus (Fig. 4-14).
intramedullary lesion and the lesion located in cancellous The bone surrounding the nidus may be cancellous
bone produce less distinctive sclerosis. However, such with thickened trabeculae and stroma consisting of rich
lesions may provoke a multilaminar, exuberant periosteal vascular connective tissue and rarely small foci of mono-
reaction that may involve long segments of the diaphysis nuclear inflammatory cell infiltrate. Prominent large
(Fig. 4-5). Minimal or absent perilesional sclerosis is also blood vessels may be present in the connective tissue
a common feature of osteoid osteomas that are located surrounding the nidus and within enlarged haversian
near the end of bone (juxtaarticular) and in subperiosteal canals in the surrounding densely sclerotic cortical bone
lesions. In vertebral locations, conventional radiographs (Fig. 4-15). The adjacent synovium may be thickened,
show increased density of the pedicle, loss of a distinct with prominent chronic inflammatory cell infiltrates that
contour, or both features. In vertebrae the nidus is often have lymphofollicular features. The changes in the syno-
not seen on conventional radiographs. Exact anatomic vial membrane occasionally simulate rheumatoid arthritis
localization of the nidus usually requires computed (Fig. 4-20). Special techniques such as immunohisto-
tomography (Figs. 4-6 and 4-7). Most frequently, the chemistry and ultrastructural investigations are practi-
nidus is present in the area of the posterior arch or at the cally never used in the diagnosis of osteoid osteoma.3,120,121
base of a pedicle. In very unusual instances, it is present Osteoid osteomas express master regulatory proteins
within the transverse or spinous process. involved in skeletal development, such as Runx2 and
Osterix, confirming the osteoblastic nature of the lesion.36
Gross Findings
The nidus tissue, when examined intact in its anatomic
Differential Diagnosis
setting, appears as a distinct oval or round, reddish area Osteoid osteoma should be differentiated from chronic
that can be easily separated from its bed (Fig. 4-8). This and acute osteomyelitis, bone abscess, intracortical hemangi-
tissue varies in consistency from friable, soft, and granu- oma, bone island, stress fracture, Ewing’s sarcoma, and intra-
lar to densely sclerotic. The central portion of the nidus cortical osteosarcoma. This long list of diverse entities
is sometimes more sclerotic than its periphery. The nidus indicates that in some instances the differential diagnosis
is usually surrounded by a large zone of dense sclerotic of this well-known lesion can be very complex and
bone (Fig. 4-8). The surrounding sclerotic bone may be difficult.

ERRNVPHGLFRVRUJ
148 4  Benign Osteoblastic Tumors

A B

C D

FIGURE 4-4  ■  Intracortical osteoid osteomas of long bones. A, A 15-year-old boy whose pain was worse at night and relieved by
aspirin was found to have a well-defined lytic intracortical lesion in the proximal femoral shaft. Note fusiform thickening around,
and extending for several centimeters above and below, nidus. B, Computed tomogram shows marked perilesional sclerosis of
bone (same case as A). C, Lateral radiograph of distal femur of 41-year-old man who had symptoms typical of osteoid osteoma for
3 months. Cortical thickening and sclerosis of bone failed to show lucent nidus; inset, computed tomogram of patient shown in
C reveals lucent nidus within markedly thickened posterior cortex. D, Photomicrograph of nidus containing irregular, randomly
oriented trabeculae of osteoid and woven bone. Abundant osteoblasts and osteoclasts border trabeculae (same case as A and B)
(D, ×100) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 149

A B

C D

FIGURE 4-5  ■  Osteoid osteoma: radiographic features. A, Radiograph with diffuse fusiform cortical thickening and sclerosis of tibial
shaft. No nidus tissue can be seen on this plain radiograph. B, Specimen radiograph of same case shows resected fragment of
anterolateral tibial cortex with dense sclerotic area representing nidus (arrows). C, Anteroposterior radiograph of intramedullary
nidus of osteoid osteoma in femoral shaft. D, Lateral radiograph of the same case shown in C. Note mild sclerosis of adjacent cortex
and prominent multilayered periosteal new bone formation.
ERRNVPHGLFRVRUJ
150 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-6  ■  Osteoid osteoma: radiographic and microscopic features. A, Computed tomogram of osteoid osteoma shows well-
circumscribed nidus involving the lateral elements of the neural arch. B, Low power photomicrograph of the nidus tissue shows
haphazardly arranged bony trabeculae of woven bone in a background of hypercellular stroma with abundant osteoblast and mul-
tinucleated osteoclastic giant cell (B, ×70). C and D, Intermediate power photmicrographs of the nidus tissue showing trabeculae of
woven bone rimmed by plump osteoblasts and occasional osteoclastic cells (C and D, ×100). (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 151

A C
FIGURE 4-7  ■  Osteoid osteoma: radiographic features. A, Anteroposterior plain radiograph with ill-defined sclerosis of the right
pedicle of the L1 lumbar vertebra. B, Computed tomogram shows well-circumscribed nidus with mineralized center in lamina.
C, Whole-mount specimen of the resected posterior neural arch showing a well-circumscribed nidus (arrows).

Radiographically and clinically, osteoid osteoma can osteomas by the absence of clinical symptoms, lack of
be confused with inflammatory conditions, such as bone perilesional sclerosis, and usually negative bone isotope
abscess and osteomyelitis of acute or chronic sclerosing scans.
types. This problem is most evident in osteoid osteomas Periosteal new bone formation associated with osteoid
affecting the small bones of the hands and feet. In juxta- osteomas can simulate stress fractures and even avulsion
articular locations, monoarticular rheumatoid arthritis injuries on radiographs when the nidus is not clearly
can be simulated microscopically and radiographically discernible on plain films.
because of the associated prominent lymphoproliferative Exuberant periosteal reactions also can mimic Ewing’s
synovitis. In each of these cases, histologic identification sarcoma or acute osteomyelitis on plain radiographs. This
of the nidus tissue disposes of the diagnostic difficulty. problem may be accentuated by the associated prominent
Lesions that present as intracortical lucencies resem- swelling of surrounding soft tissue.
bling osteoid osteoma include abscesses, hemangiomas and, Rarely, accessory growth centers can be confused
in very unusual circumstances, intracortical osteosarcomas. radiologically with osteoid osteoma. Such growth centers
In all these entities, positive findings on bone scans can are most likely to occur in tarsal or carpal bones.
be anticipated; therefore bone scans are of limited diag- The histologic appearance of osteoid osteoma nidus
nostic usefulness in discriminating between osteoid tissue is usually so distinctive that differential diagnostic
osteoma and the other lesions. In most cases, enostoses, problems on this level rarely occur. However, when taken
or small bone islands, can be separated from osteoid Text continued on p. 160

ERRNVPHGLFRVRUJ
152 4  Benign Osteoblastic Tumors

A B

C D

E G
FIGURE 4-8  ■  Gross, radiographic, and microscopic features of resected osteoid osteoma. A, Resected specimen with central round,
well-demarcated nidus tissue. Note ivory appearance of surrounding sclerotic bone. B, Low power photomicrograph shows topog-
raphy of intracortical nidus and surrounding bone. Note sharply demarcated nidus surrounded by sclerotic cortical bone with
thickened medullary and periosteal bone trabeculae. C, Resected specimen with a well-demarcated nidus tissue and sclerotic ivory
appearance of surrounding bone. D, Radiograph of the specimen shown in C shows lucent well-demarcated nidus surrounded by
sclerotic bone with thickened trabeculae. E, Radiograph of resected fragment of bone with sharply demarcated oval nidus surrounded
by densely sclerotic bone. F, Radiograph of perpendicular section of long bone with nidus. There is prominent periosteal reaction
with numerous parallel spiculae of newly formed bone. Nidus shows prominent dense center surrounded by less ossified peripheral
zone. Note dense sclerotic bone with coarse trabecular pattern around nidus. G, Low power photomicrograph shows topography
of nidus and surrounding bone. Note sharply demarcated nidus with peripheral lucency and sclerotic trabeculae of surrounding
bone. (B and G, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 153

A B

C D

FIGURE 4-9  ■  Osteoid osteoma: microscopic features. A, Low power view showing topographic features of nidus in osteoid osteoma.
B, Higher power of A showing a well-delineated somewhat sclerotic center of the nidus and less sclerotic peripheral zone. C, Higher
magnification of B showing the periphery of the nidus composed of uniformly distributed bony trabeculae in cellular and vascular
stroma. Note merging of trabeculae structures of osteoid osteoma with adjacent sclerotic bone. D, Sclerotic bone at the periphery
of osteoid osteoma (A, ×100; B, ×25; C and D, ×100) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
154 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-10  ■  Microscopic features of osteoid osteoma. A, Intermediate power view of nidus with numerous bony trabeculae uni-
formly distributed in cellular and vascular stroma. B, Intermediate power view of nidus with numerous irregular bony trabeculae.
Supporting tissue consists of numerous osteoblasts and prominent vessels (×100). C, Intermediate power view of nidus with promi-
nent irregular bony trabeculae. D, Intermediate power view shows irregular interconnecting bony trabeculae. Note variable pattern
of matrix deposition in osteoid osteomas. (A-D, ×100) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 155

A B

C D
FIGURE 4-11  ■  Patterns of bony trabeculae with different levels of mineralization in nidus of osteoid osteoma. A, Poorly mineralized
ill defined areas of early osteoid deposition (×100). B, Poorly mineralized irregular areas of woven type haphazardly distributed in
highly cellular stroma with numerous osteoblasts, prominent vessels, and occasional osteoclast-like giant cells. C, Intermediate
power photomicrograph shows trabeculae of woven bone rimmed by osteoblasts and occasional osteoclasts. D, Low power pho-
tomicrograph showing irregular trabeculae of woven bone in highly cellular hemorrhagic stroma with numerous osteoblasts,
prominent vessels, and occasional osteoclast-like giant cells. (A, B, and D, ×100; C, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
156 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-12  ■  Patterns of bony trabeculae and osteoid deposition in nidus of osteoid osteoma. A, Bony trabeculae forming intercon-
necting network with prominent dialated vascular channels in stroma. B, Higher magnification of specimen A shows interconnecting
well mineralized bony trabeculae rimmed by plump osteoblasts (×200). C, Thick, irregular bony trabeculae in a highly cellular stroma
with plump osteoblastic cells and occasional osteoclast-like giant cells. D, Thin, poorly developed bony trabeculae and irregular
osteoid depositions in a background of highly cellular stroma composed of osteoblastic cells, dilated vessels, and occasional
osteoclast-like giant cells. (A, ×100; B-D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 157

A B

C D
FIGURE 4-13  ■  Patterns of osteoid depositions in osteoid osteoma. A, Thick, irregular bony trabeculae of well-mineralized
bone obliterating highly cellular stromal tissue. B, Higher magnification of A shows irregular areas of osteoid deposition (×100).
C, Well-mineralized trabeculae form interconnecting network and solid areas of osteoid. D, Irregular, poorly developed osteoid
with collagen-like features produces interconnecting network in a highly vascular spindle cell stromal tissue. (A, ×100; B-D, ×200)
(A and B, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
158 4  Benign Osteoblastic Tumors

B
FIGURE 4-14  ■  Cartilage in osteoid osteoma, rare finding. A, Unusual case of osteoid osteoma with area of cartilage in center of
otherwise typical nidus. B, Higher magnification of area marked in A shows well-developed cartilage with myxoid changes. Caution:
This finding is extremely rare in nidus tissue of osteoid osteomas. Diagnosis in such cases should be made only after careful cor-
relation with clinical and radiographic data. (A, ×50; B, ×100) (A and B, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 159

A B

C D
FIGURE 4-15  ■  Reactive changes adjacent to osteoid osteoma. A and B, Sclerotic cortex at the periphery of osteoid osteoma.
C, Extensive remodeling and sclerosis of cortex adjacent to osteoid osteoma. D, Sclerosis and remodeling of medullary bone in the
vicinity of osteoid osteoma. (A-D, ×100) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
160 4  Benign Osteoblastic Tumors

out of clinical and radiologic context, it can be confused tomography should be performed before introducing the
with osteosarcoma. Reactive new bone formation usually electrode with percutaneous thermocoagulation therapy
can be distinguished by its parallel or radially oriented of osteoid osteomas, because other intracortical or sub-
bone trabeculae, which often show gradual peripheral periosteal lesions can mimic their radiologic appearance.
maturation to lamellar bone. These can be easily distin- Among these are intracortical abscess and intracortical
guished from the random disorderly pattern of osteoid chondroma.
and woven bone trabeculae in nidus tissue. In curetted
material, sclerotic nidus tissue may present special prob- Osteoid Osteoma in Different
lems in microscopic recognition.
Anatomic Sites
The microscopic appearance of nidus tissue in osteoid
Treatment and Behavior osteoma is identical regardless of the anatomic location.
Osteoid osteomas have limited growth potential and There is no correlation between the pain pattern and the
rarely exceed 1 cm. The majority of lesions are about degree of associated bone sclerosis. In different anatomic
0.5 cm in diameter. Some tumors may even sponta- sites and age groups, osteoid osteomas may induce quite
neously regress.51,116 The primary treatment is surgi- distinct secondary changes in the adjacent bone and soft
cal removal of the nidus and some of the surrounding tissue that lead to various diagnostic and therapeutic
bone by en bloc excision after precise localization of the dilemmas. Therefore it is important from both the patho-
nidus.99,128 Curettage with bone grafting is an acceptable logic and the clinical points of view to discuss osteoid
modality of treatment in anatomic sites where en bloc osteoma separately in three major anatomic locations:
excision cannot be performed. However, this approach is long tubular bones, small bones of the hands and feet,
generally discouraged because of the risk of incomplete and the vertebral column. In addition, there are separate
removal of the nidus and recurrence of symptoms. Recur- descriptions of subperiosteal and juxtaarticular lesions
rence may occasionally happen many years after removal because of their distinct clinicoradiologic and pathologic
of the primary lesion.103 In some cases, no nidus tissue can features.
be found in the resected specimen. According to Mayo
Clinic data, this occurs in nearly 20% of patients with Osteoid Osteoma of Long Tubular Bones
osteoid osteomas that have a radiologically documented
nidus.117 It is important to consider whether the lesion Osteoid osteomas that occur in long tubular bones are
was missed at surgery or the pathologist failed to identify most often found within the proximal or distal portions
it. The persistence of symptoms suggests that the nidus of the bone shafts (Fig. 4-16). Approximately 50% of
was not removed. Such patients need reevaluation and cases are diagnosed in the lower extremities; the femoral
further surgery if there is evidence of a nidus. The nidus neck is the single most frequent anatomic site. The tibia
tissue can be found in a relatively high proportion of is the second most frequently involved long tubular bone;
patients who undergo further surgery.117 Still, in some fibular lesions are very rare. In the upper extremities, the
patients whose symptoms are relieved, no nidus tissue humerus is the most frequently involved bone, and a
or other abnormality can be documented in the resected majority of cases occur around the elbow joint.108 Involve-
specimen other than bone sclerosis. ment of the distal ulna and radius is very rare. Radio-
Although en bloc excision is the preferred treat- graphically, the intracortical lesions of long bones are
ment, in some cases a lesion may be inaccessible to usually associated with distinct fusiform cortical thicken-
the surgeon. In such instances, its complete removal ing that is sometimes accompanied by periosteal reaction.
may cause more disability than that associated with Occasionally, the periosteal reaction can be very exuber-
the original disease. Some of these patients can be ant, with multiple layers of new bone formation. The
managed with prolonged treatment with nonsteroidal exuberant periosteal reaction can mimic that seen in
anti-inflammatory drugs. As reported by Kneisl and stress fracture or that associated with Ewing’s sarcoma
Simon,65 prolonged anti-inflammatory treatment (30-40 (Figs. 4-5 and 4-16). The intramedullary lesions usually
months) can induce permanent relief of symptoms, and produce less surrounding sclerosis. The absence of dis-
regression of the nidus may be seen on radiographs. Abla- tinct sclerosis around the intramedullary nidus is proba-
tion of the nidus with a percutaneously placed radiofre- bly related to its distant location from labile periosteum.
quency electrode that produces thermocoagulation has A rare complication of osteoid osteoma of long tubular
become widely accepted. This technique may be of value bones is localized overgrowth of bone, angular deformity,
in anatomic sites where it is difficult to surgically remove or both95 (Fig. 4-17). These complications typically
the tumor.4,5,20,22,31,80,81,106,107 Osteoid osteomas of the verte- happen in very young patients whose symptoms begin
brae present special problems with this type of treatment before age 5 years. Long-standing hyperemia induced by
because of the risk of nerve root damage, but to date, a prostaglandins is probably a factor in accelerated growth
number of tumors at this site have been treated success- of bone in osteoid osteoma. The resulting bone defor-
fully without nerve root or spinal cord injury. Reported mity and discrepancy in limb length may disappear after
success rates for treatment by thermocoagulation with excision of the nidus. In some patients, however, the
radiofrequency electrodes have ranged between 80% deformity and length discrepancy never completely
and 90% (no need for additional procedures or medica- resolve. This is particularly true in cases of many years’
tions for 2 years). A needle biopsy guided by computed duration.95

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 161

A B C

D E
FIGURE 4-16  ■  Osteoid osteoma in long bones. A, Radiograph of hip of an 8-year-old girl shows nidus with moderate bone sclerosis
surrounding lesion. B, Computed tomogram at this level shows nidus more clearly with cortical thickening and medullary sclerosis.
C, Bone scan shows large zone of increased uptake corresponding to nidus and perilesional sclerosis. D, Radiograph of a 17-year-
old athletic boy with proximal leg pain and localized cortical thickening that at first was thought to represent a stress fracture.
E, Computed tomogram of tibia shown in D reveals punctate lucency diagnostic for intracortical osteoid osteoma.

ERRNVPHGLFRVRUJ
162 4  Benign Osteoblastic Tumors

A B C

D E F
FIGURE 4-17  ■  Osteoid osteoma with prominent overgrowth and deformity of bone. A, Osteoid osteoma of the proximal humerus
with prominent diffuse sclerosis and periosteal reaction deforming bone contour. B, Anteroposterior radiograph showing diffuse
sclerosis and length discrepancy of the left femur. C, Radiograph of a 1-year-old boy with painful swelling of left leg. Note sclerosing
lesion of left tibial diaphysis and pronounced length discrepancy. Biopsy showed reactive bone formation. Lesion was diagnosed
as osteomyelitis and was treated with antibiotics. D and E, Five years later, same patient had recurrent pain. Note pronounced
bowing deformity of left tibia and diffuse cortical sclerosis. A biopsy documented nidus of osteoid osteoma. F, Clinical photograph
of the same patient 1 year later, showing discrepancy in limb length and persistent bowing deformity of involved tibia.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 163

Osteoid Osteoma of Small Bones to other common joint disorders. Pain is usually less
of Hands and Feet intense, and its response to nonsteroidal antiinflamma-
tory drugs is less dramatic. On plain radiographs the
In our mainly consultation series of osteoid osteomas, intraarticular nidus is difficult to identify (Fig. 4-19). In
slightly more than 20% of osteoid osteomas occur in the addition, the adjacent sclerosis is typically absent, and
small bones of the hands and feet, with tumors occurring there is no periosteal reaction. Consequently, there is
twice as frequently in the hands as in the feet. In the usually a considerable delay between the onset of
hands, the phalanges are the most frequently involved symptoms and identification of the nidus. Bone scintig-
sites.23,88 In small bones, the nidus is usually surrounded raphy, computed tomography, and magnetic resonance
by a distinct zone of sclerosis. The bone contour can be imaging are virtually essential for early detection of
expanded, and the entire bone can be markedly enlarged. the nidus. In addition, these techniques usually reveal
Osteoid osteomas of small peripheral bones are almost minimal periosteal reaction and sclerosis at a distance
always associated with a distinct swelling of soft tissue and from the lesion on either side of the joint. The absence
synovitis of the adjacent joints. Therefore in this location, of distinct sclerosis around these lesions is probably
long delays in arriving at the correct diagnosis may occur related to a lack of functional periosteum in the immedi-
because the tumor may frequently mimic an inflamma- ate vicinity of the intracapsular nidus. To the contrary,
tory process.26 Thus it is not unusual for patients with intraarticular osteoid osteoma can provoke periarticular
osteoid osteoma of the hands and feet to be treated ini- osteoporosis similar to that associated with inflammatory
tially for chronic osteomyelitis or other inflammatory arthropathy.
conditions (Fig. 4-18). In rare instances, the intraarticular lesions may even
induce degenerative changes within the articular carti-
Osteoid Osteoma of Vertebral Column lage, promote proliferation of chondrocytes with angio-
genesis, and promote migration of mesenchymal cells in
Approximately 10% to 15% of osteoid osteomas occur in the subchondral bone. These changes are responsible for
the vertebral column. Most frequently, they are located the development of secondary osteoarthritis with osteo-
in the lumbar and lower thoracic portions of the spine. phyte formation and occasionally extensive joint destruc-
Spinal lesions are difficult to detect on plain radiographs tion.94 In unusual instances, osteoid osteoma may induce
and are known to be responsible for unexplained back- premature fusion of the epiphysis.19
ache and painful scoliosis.8,21,58,62,97,102,115 In some patients, The intraarticular lesions typically are associated with
clinical symptoms may suggest a neurologic disorder, an inflammatory response of the synovium, which is
lumbar disk disease, or both conditions. It is recom- edematous and thickened and shows villous overgrowth.
mended that an underlying osteoid osteoma be ruled out Histologically, it is characterized by a lymphoprolifera-
in virtually all cases of unexplained back pain and painful tive synovitis with multiple lymphoid follicles indistin-
scoliosis that occur in children and young adults.8,20,62,109,115 guishable from rheumatoid arthritis (Fig. 4-20). However,
Although removal of the lesion usually results in gradual destruction of the joint with formation of pannus and
regression of the curve, in neglected cases a prolonged subsequent ankylosis does not occur. Synovial changes
delay in treatment may result in persistent structural sco- spontaneously subside after removal of the nidus. It is
liosis.102 As mentioned, a vertebral nidus of osteoid postulated that increased levels of prostaglandins can
osteoma is difficult to detect on plain radiographs. At play a major role in secondary synovitis and soft tissue
best, the radiographs may show an area of sclerosis, a edema commonly associated with intraarticular osteoid
thickened and distorted sclerotic transverse process, or a osteoma.119 It has also been found that the occurrence of
deformed sclerotic pedicle with indistinct borders. Locat- synovitis in patients with osteoid osteoma coincides with
ing the exact anatomic site of the nidus requires com- the presence of DR4, DR7, and MT3 types of histocom-
puted tomography (Figs. 4-6 and 4-7). In vertebrae the patibility antigens (HLA), the types frequently found in
nidus is most frequently located in the posterior arch. patients with rheumatoid arthritis.119
Magnetic resonance imaging also may facilitate the iden-
tification of an inconspicuous nidus. High signal in T2- Subperiosteal Osteoid Osteoma
weighted images of nidus tissue correlates well with its
increased vascularity. Other frequent locations include Subperiosteal osteoid osteoma is a relatively rare lesion
articular facets and pedicles. Less frequently the nidus is that has unique radiographic and microscopic features.
located in the transverse and spinous processes. A primary Conventional radiographs demonstrate a lucent lesion on
tumor in the vertebral body is extremely rare. Within the the surface of bone with adjacent periosteal reaction. The
body the nidus is usually located eccentrically near the nidus is typically radiolucent with no evidence of central
base of a transverse process. opacity. It has been suggested that the absence of restrict-
ing compressive force exerted on the lesion growing on
the surface of bone is at least in part responsible for the
Juxtaarticular Osteoid Osteoma
generally larger size of subperiosteal osteoid osteoma.63
Osteoid osteomas located within the articular capsule are The lack of confinement may also explain other distinct
rare and difficult to detect.12,16,24,28,37,45,56,61,94,112,114,126,132 histologic features of surface lesions. The subperiosteal
Any joint may be involved, but the most vexing diagnostic nidi are on average less sclerotic and have thinner tra-
problems involve the elbow, hip, and ankle joints. They beculae and a greater proportion of stromal tissue com-
are typically associated with nonspecific symptoms similar pared with intracortical lesions.

ERRNVPHGLFRVRUJ
164 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-18  ■  Osteoid osteoma in short tubular bones masquerading as chronic osteomyelitis. A, Radiograph of sclerotic nidus in
proximal phalanx of long finger (arrows) of a 26-year-old woman who noted pain and swelling for 2 years before diagnosis was
established. She had been followed and treated for suspected osteomyelitis throughout this period. B, Clinical photograph of case
A showing soft tissue edema and deformity of long finger. C, Radiograph showing lucent nidus with sclerosis in the adjacent bone
in proximal phalanx of fifth finger. D, Radiograph of a 22-year-old man with pain, swelling of soft tissue, and expanded radiolucent
nidus in proximal phalanx of index finger (arrows). Diagnosis of chronic osteomyelitis resulted in prolonged treatment with antibiot-
ics. Cultures were always negative, and even after synovial biopsy, chronic inflammation was diagnosed. Finally, after 5 years
radiologic diagnosis of osteoid osteoma was suggested, and curettage resulted in permanent relief of symptoms.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 165

A B

C D
FIGURE 4-19  ■  Juxtaarticular osteoid osteoma of elbow joint. A, Radiograph of elbow joint with diffuse, poorly demarcated area of
sclerosis of proximal ulnar metaphysis (arrows). No nidus tissue can be identified. B, Computed tomogram of case shown in
A documents juxtaarticular sclerotic nidus in ulna that is surrounded by lucent halo (arrow). Nidus is not centrally located in sclerotic
area. C, Radiograph of elbow showing ill defined sclerosis and prominent periosteal reaction involving the distal end of humerus.
D, Magnetic resonance imaging shows inhomogeneous subarticular nidus of intermediate signal intensity (arrows). Note prominent
periosteal reaction of the distal humerus and fluid accumulation in the elbow joint.

ERRNVPHGLFRVRUJ
166 4  Benign Osteoblastic Tumors

B
FIGURE 4-20  ■  Synovial changes associated with osteoid osteoma. A, Lymphoproliferative synovitis with lymphocytic infiltrate of
synovium and multiple lymphoid follicles with germinal centers. B, Higher magnification of follicle shown in A. Note scattered
predominantly perivascular inflammatory infiltrate (A, ×50; B, ×100) (A and B, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 167

Personal Comments decades of life. The male/female ratio is the same as that
for osteoid osteoma, approximately 2 : 1.
Problems with the identification of nidus tissue and the Benign osteoblastoma has a unique predilection for
simulation of reactive, posttraumatic, and inflammatory the axial skeleton, with more than 40% of cases involving
conditions have predominated in diagnostic case material the vertebral column, sacrum, and the proximal parts
submitted for consultation. The recurring theme of of the appendicular skeleton such as pelvis and femur
confusion between reactive bone and suspected nidus (Figs. 4-24 and 4-25).21,81,85,111,125 The second most fre-
tissue, when the nidus has not actually been sampled, is quent site is the jaw bone, followed by other craniofacial
exemplified by Figure 4-21. The humoral effects of bones (Figs. 4-26 and 4-27). Benign osteoblastoma also
osteoid osteoma on surrounding bone and soft tissue occurs frequently in the extremities, where its overall
have often masked the presence of a nidus and have distribution is similar to that of osteoid osteoma. Conse-
led to long delays in diagnosis and treatment. As a con- quently the proximal femur in the area of the femoral
sequence, the patient may undergo inappropriate therapy neck is its most frequent site in the appendicular skeleton.
for many months or even years under the mistaken diag- In the upper extremities, the humerus is most frequently
nostic impression of an infection or inflammatory condi- involved. Benign osteoblastoma is rarely present in ribs
tion. Frequent use of computerized imaging techniques and flat bones. Rare instances of osteoblastomas associ-
in diagnostic workup in bone lesion in modern practice ated with severe systemic symptoms of fever, weight loss,
has dramatically reduced the incidence of such lesions. cachexia, clubbing of fingers and toes, and diffuse peri-
Our own experience with the exceptional cases in ostitis (“toxic osteoblastoma”) have been observed.35 A
which nidus tissue is never documented microscopically case of a conventional osteoblastoma arising extraskele-
in the face of typical clinical and radiologic features coin- tally in the laryngeal cricoid cartilage has been reported.67
cides with that described by Sim et al.117 This remains an Osteoblastomas are uncommonly associated with onco-
unresolved issue, but recent evidence of spontaneous or genic phosphaturic syndromes.29
therapy-induced regression of osteoid osteoma may
provide an explanation for this discrepancy.65 Radiographic Imaging
The radiographic features of some benign osteoblasto-
mas are similar to those of osteoid osteoma. Benign
BENIGN OSTEOBLASTOMA osteoblastoma produces a round or oval, well-demarcated
Definition metaphyseal lytic defect surrounded by a zone of reactive
sclerosis.66,77,81,85,125 Less commonly in long bones, it may
Benign osteoblastoma is a rare osteoblastic tumor that arise entirely within the epiphysis.132 The main difference
is closely related to osteoid osteoma. In fact, the micro- from osteoid osteoma is that the nidus is larger, exceeding
scopic features of the nidus in osteoid osteoma are similar 1.5 cm in diameter.81,110 Moreover, the sclerotic zone is
and quite often identical to those of osteoblastoma. frequently less extensive compared with that of an ordi-
Benign osteoblastoma differs from osteoid osteoma in nary osteoid osteoma. In fact, sclerosis may not be present
that it has a higher growth potential because it exceeds at all. It is believed that the predilection of benign osteo-
2 cm in diameter. Moreover, it frequently lacks distinct blastoma for cancellous bone may at least in part account
nocturnal pain that can be relieved by aspirin. Secondary for the absence of distinct sclerosis. In the vertebral
peripheral sclerosis may be minimal or absent. Still, there column and the sacrum, the tumor is located posteriorly
are cases with composite features that fall between osteoid within the dorsal elements of the arch (Figs. 4-24 and
osteoma and osteoblastoma. These have been designated 4-25).89,91 A primary tumor within the vertebral body is
in the past as giant osteoid osteomas. This problem has been unusual.92 The osteoblastic nature of the lesion can be
resolved by arbitrarily designating all lesions as osteoblas- documented on radiographs in nearly 50% of all cases by
tomas when the nidus is more than 1.5 cm in diameter. the presence of patchy or cloudlike radiopacities. Exten-
The lesion designated as cementoblastoma in the oral sive reactive changes in the surrounding tissue and appar-
pathology literature is an osteoblastoma located in jaw ent soft tissue masses on magnetic resonance imaging
bones in the vicinity of the roots of teeth. Benign osteo- may overestimate the extent of the lesion, so computed
blastomas of the mandible do not differ in their morpho- tomography should continue to be the imaging modality
logic features and behavior from those located in other of choice for the demonstration and local staging of sus-
anatomic sites. They are included among the osteoblas- pected vertebral osteoblastomas.26 Osteoblastomas of the
tomas of the craniofacial bones in our series. spine frequently develop a secondary aneurysmal bone
cyst and may present as expansile destructive lesions
involving the adjacent vertebra and ribs.
Incidence and Location Mandibular lesions are located in the vicinity of a
The peak age incidence and frequent sites of skeletal tooth root and almost invariably show central opacity
involvement are shown in Figure 4-22. The skeletal and surrounded by a lucent halo (Fig. 4-26). Their pathoge-
age distribution patterns of individual osteoblastomas are netic relationship to the root of the tooth is most likely
shown in Figure 4-23. Benign osteoblastoma is a very incidental. In other words, they do not arise from the
rare tumor that accounts for less than 1% of all primary odontogenic apparatus itself but rather from adjacent
bone tumors. Most patients are between age 5 and 45 bone.
years, and the peak incidence is in the second and third Text continued on p. 174

ERRNVPHGLFRVRUJ
168 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-21  ■  Eighteen-year-old man who noted pain in right hip region on weight bearing and developed progressively worsening
limp. A, Plain radiograph shows bone sclerosis of ischium with prominent periosteal new bone formation. Inset shows bone scan
with high uptake of isotope. B, Computed tomogram reveals central nidus, which was obscured by profuse reactive changes. Inset,
Magnetic resonance image shows central nidus surrounded by sclerotic bone and extensive reactive changes. C and D, This lesion
was originally considered to be benign reactive bone-forming process related to trauma even after two biopsies. These showed
only reactive bone formation. Later, on third exploration for continuing symptoms and persistent sclerosis, easily distinguished
nidus tissue was excised from interior of ischium. (C and D ×100; hematoxylin eosin)

ERRNVPHGLFRVRUJ
Peak
incidence
10 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 4-22  ■  Osteoblastoma. Peak age incidence and frequent sites of skeletal involvement.

Osteoblastoma
30 35
Mean Age: 25
30 Male/Female Ratio: 1.43
25

25
Number of Cases

Number of Cases

20

20
15
15
10
10

5 5

0 0
1 2 3 4 5 6 7 8 9
Vertebra
Mandible
Skull
Sacrum
Maxilla
Rib
Ilium

Tibia
Humerus
Fibula
Radius
Ulna
Tarsal
Carpal
Phalanx
Femur

Age in Decades
B
Flat Major Small bones of
bones long bones hands and feet

Axial Appendicular
skeleton skeleton
A
FIGURE 4-23  ■  Skeletal sites and age frequency distribution patterns of osteoblastoma. A, Skeletal distribution of osteoblastoma.
B, Age frequency distribution of osteoblastoma.

ERRNVPHGLFRVRUJ
170 4  Benign Osteoblastic Tumors

A C
FIGURE 4-24  ■  Osteoblastoma radiographic and microscopic features. A, Anteroposterial plain radiograph of lumbar spine showing
ill defined sclerotic lesion involving the left pedicle of L5 lumbar vertebra (arrows). B, Computed tomogram shows well-circumscribed
lytic tumor with foci of mineralized bone matrix in lateral body and neural arch. C, Trabeculae of woven bone with irregular scal-
loped borders in hypercellular stroma with abundant osteoblasts and osteoclast-like giant cells. (C, ×100). (C, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 171

A B

C D
FIGURE 4-25  ■  Osteoblastoma of lumbar and cervical vertebrae. A, Osteoblastoma of lumbar vertebra in a 27-year-old man with
radicular pain related to L5 computed tomogram shows lobulated posterior extradural mass displacing dural sac to left and anteri-
orly. Origin of tumor from within lamina is shown by central lucency (arrow). B, Magnetic resonance sagittal image shows destruc-
tion of lamina with soft tissue mass in spinal canal. Mass demonstrates high signal with foci of signal void corresponding to tumor
matrix mineralization. Same case as shown in A. C, Osteoblastoma of third cervical vertebra of a 36-year-old man. Magnetic reso-
nance image shows high signal at site of eccentric tumor involving posterolateral portion of third cervical vertebral body and pedicle
on left. D, Computed tomogram shows well-circumscribed lytic tumor containing foci of mineralized bone matrix in base of pedicle,
lamina, and body. Dural sac is displaced to right. Same case as shown in C.

ERRNVPHGLFRVRUJ
172 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-26  ■  Osteoblastoma of mandible. A, Nineteen-year-old man who reported pain at site of previously extracted lower tooth.
Well-circumscribed radiolucent area with central opacity represents benign bone-forming tumor. B, Bone-forming mass with ill
defined punctuated mineralization pattern at the root of bicuspid. C, Twenty-two-year-old man with tumor arising adjacent to root
of first molar tooth. D, Gross photograph of tumor shown in C; tumor consists of firm, gritty mass attached to tooth root.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 173

A B

E
FIGURE 4-27  ■  Osteoblastoma of skull: radiographic features. A and B, Plain lateral and anteroposterior radiographs of osteoblastoma
arising in left frontoparietal region of skull of 18-year-old woman. Notice beveled edge surrounding totally radiolucent tumor. Double
contour of margin is caused by unequal destruction of inner and outer tables of calvarium. Mass was present for 4 years without
significant increase in size. Throbbing headaches and blurring of vision were present. There was incomplete relief with aspirin.
C, Computed tomogram shows shell of reactive bone peripherally and smaller intracranial bulge. D, Gross photograph of resected
specimen showing well-circumscribed, tan external surface of osteoblastoma. E, Gross photograph of bisected resection specimen
of calvarial osteoblastoma. Note sharp circumscription and greater destruction of outer table than of inner cortex.

ERRNVPHGLFRVRUJ
174 4  Benign Osteoblastic Tumors

Osteoblastoma may expand the bone contour with a to as atypical or pseudosarcomatous osteoblastomas (Fig.
markedly thinned cortex (Fig. 4-27). The cortex can be 4-33). The atypia is of a degenerative type similar to
at least focally destroyed with or without periosteal new that more often seen in ancient schwannomas. The
bone formation. These features are responsible for the absence of mitotic activity and specifically of atypical
radiologic confusion with malignant tumors that may mitoses is helpful in distinguishing such lesions from an
occur in approximately 20% of osteoblastomas.81 In a osteosarcoma.9
large study of osteoblastomas comprising over 300 cases,
radiographic evidence of bone expansion and destruction
was present in nearly 40% of the cases.74 In 12% of
Differential Diagnosis
the lesions, radiographic presentation was suggestive of Osteoblastoma should be differentiated from osteosar-
malignancy.74 Occasionally, osteoblastomas are located coma, osteoid osteoma, and aneurysmal bone cyst. The distin-
on the surface of bone (subperiosteal osteoblastomas) and guishing features of osteoid osteoma and osteoblastoma
produce a lucent lesion that may erode the cortex.90 were discussed in the opening paragraph of this chapter.
Surface lesions may be surrounded by a thin shell of The main differences of size, pain pattern, and perile-
periosteal new bone (Fig. 4-28). sional sclerosis are usually sufficient to separate these two
entities, which are histologically indistinguishable. An
arbitrary size limit of 1.5 cm has been proposed for
Gross Findings lesions with composite features; those exceeding this
The gross appearance of the nidus in benign osteoblas- dimension are classified as osteoblastomas.
toma is similar to that of osteoid osteoma. Because of its Osteoblastomas that attain sizes exceeding 4 cm and
rich stromal vasculature, the tissue is granular, friable, and that show prominent or exuberant periosteal new bone
reddish and may bleed profusely when curetted. The formation present problems in their radiographic dif-
nidus is well demarcated at its periphery and can be easily ferentiation from osteosarcoma. This group of osteoblas-
separated from the surrounding bone. The bone contours tomas may also show histologic features that can be
can be markedly expanded, especially in small bones. The misinterpreted as signs of malignancy by the pathologist.
cortex is thinned and may be focally destroyed. There These features include foci of lacelike osteoid deposition,
may be gross evidence of hemorrhage and cystic degen- high cellularity, and more than a few scattered mitotic
eration with blood-filled spaces characteristic of second- figures. This may be further complicated by the rare
ary aneurysmal bone cyst. The hemorrhage and secondary finding of small foci of cartilage in otherwise benign-
reparative changes are particularly frequent in larger appearing osteoblastomas. Fortunately, these exceptional
lesions and may significantly alter the gross appearance. disturbing features usually occur separately and focally in
a tumor that otherwise represents a conventional benign
osteoblastoma. A tumor that exhibits all the mentioned
Microscopic Findings atypical features is best regarded as an osteosarcoma.
The microscopic features of osteoblastoma are similar to Although aneurysmal bone cysts have not been
those of ordinary osteoid osteoma (Figs. 4-29 to 4-31). At observed in association with osteoid osteomas, they
low magnification the nidus tissue consists of an interlac- present a distinct problem because they frequently occur
ing network of bone trabeculae evenly distributed in a as secondary phenomena superimposed on underlying
loose fibroblastic stroma with a prominent vasculature. osteoblastomas. Moreover, primary aneurysmal bone
The osteoid formed may be deposited in coarse trabecu- cysts not associated with underlying lesions have a pre-
lae or sparsely cellular sheets. Areas focally approaching dilection for the same anatomic sites as osteoblastomas.
a lacelike appearance can be present but are uncom- This is particularly true with regard to their occurrence
mon. Prominent rimming osteoblasts and multinucle- in the posterior neural arch of vertebrae.
ated osteoclast-like giant cells are present. In general, the Reactive bone formation in solid areas or septa of an
osteoblasts do not form solid sheets that fill the intertra- aneurysmal bone cyst should not be interpreted as evi-
becular spaces. The mitotic rate can be focally quite high, dence of an underlying osteoblastoma. Aneurysmal bone
but atypical mitoses are not seen. Although the lesion is cysts may contain microscopic zones of reactive osteoid
grossly well demarcated, microscopically the osteoid tra- and new bone that differ from the small, irregular, anas-
beculae merge gradually with the adjacent normal bone. tomosing trabeculae of nidus tissue.
Varying degrees of mineralization of osteoid can be seen. Similar to osteoid osteomas, osteoblastomas express
When the tumor extends beyond the cortex, the soft transcription factors controlling osteoblastic lineage dif-
tissue margin usually has a clearly recognizable shell of ferentiation, such as Runx2 and Osterix.36
reactive bone. Large, expanded lesions usually have gross
and microscopic evidence of secondary aneurysmal bone Treatment and Behavior
cyst with multiple cystic spaces filled with blood.
As a rule, cartilage should not be present in benign Osteoblastomas have a higher growth potential than
osteoblastoma. However, in our consultation material, osteoid osteomas and may occasionally attain consider-
foci of benign cartilage were found in several otherwise able size. Typical lesions are approximately 2 cm in
typical osteoblastomas with proven benign clinical behav- diameter. Similar to osteoid osteoma, osteoblastomas fre-
ior (Fig. 4-32). quently present as painful lesions.91 Lesions located in the
In rare instances stromal cells in osteoblastoma axial skeleton frequently present as back pain, neurologic
may show pronounced atypia, such lesions are referred Text continued on p. 181

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 175

A B C

D E

FIGURE 4-28  ■  Two examples of subperiosteal osteoblastoma. A, Radiograph of 30-year-old woman who reported posterior thigh
pain above knee. Lateral radiograph of femur with surface lesion (arrow) attached to posterior cortex. B and C, Sagittal and axial
magnetic resonance T1-weighted images show subperiosteal location with cortical erosion. Note low signal in adjacent marrow and
signal void of normal cortical bone. D, Radiograph of 15-year-old boy with knee pain, who was found to have eccentric lytic lesion
of distal femoral metaphysis. Anteroposterior view shows periosteal elevation with cortical erosion (arrow). E, Computed tomogram
shows encapsulation by peripheral shell of subperiosteal bone and cortical erosion. Central opacity is present. Note unusual degree
of adjacent sclerosis.

ERRNVPHGLFRVRUJ
176 4  Benign Osteoblastic Tumors

B C
FIGURE 4-29  ■  Osteoblastoma of pelvis: radiographic and microscopic features. A, Axial computed tomogram of pelvis showing
well-circumscribed osteoblastoma involving the iliac bone in the vicinity of the sacroiliac joint. B, Low power photomicrograph of
osteoblastoma showing irregular ill defined trabeculae of woven bone in hypercellular stromal tissue. C, Intermediate power pho-
tomicrograph of osteoblastoma with irregular bony trabeculae surrounded by prominent rims of osteoblasts. Note hypercellular
spindle cell stromal tissue. (B and C, ×100) (B and C, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 177

A B

C D
FIGURE 4-30  ■  Osteoblastoma: patterns of osteoid deposition. A, Irregular bony trabeculae in highly cellular vascular stroma.
B, Irregular, ill defined sheets of osteoid with entrapped osteoblastic cells (×100). C, Ill defined irregular areas of poorly mineralized
osteoid. D, Higher power view shows prominent osteoblastic cells bordering irregular poorly mineralized bony trabeculae. (A-C,
×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
178 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-31  ■  Osteoblastoma: patterns of osteoid deposition. A, Irregular haphazardly arranged bony trabeculae of woven
bone surrounded by plump osteoblastic cells in a highly cellular spindle cell stromal tissue with dilated vessels. B, Irregular,
thick trabeculae of woven bone in osteoblastoma. C, Small focus of lacelike poorly mineralized osteoid with entrapped
osteoblastic cells. D, Small focus of ill defined osteoid deposition with entrapped osteoblastic cells. (A and B, ×100; C and D, ×200).
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 179

B
FIGURE 4-32  ■  Cartilage in osteoblastoma. A, Unusual case of osteoblastoma with area of well-developed cartilage in otherwise
typical osteoblastoma. B, Higher power view of A. Well-developed areas of endochondral ossification. (A, ×50; B, ×100) (A and
B, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
180 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-33  ■  Atypical osteoblastoma with pseudosarcomatous changes: microscopic features. A, Low power photomicrograph
shows well-developed haphazardly oriented trabeculae of bone in hypercellular stroma with atypia. B, High power photomicrograph
of stromal tissue shows prominent nuclear atypia. C and D, High power photomicrographs of bony trabeculae in stromal tissue
showing prominent atypia. Note the degenerative nature of atypia and the absence of mitotic activity in this osteoblastoma with
pseudosarcomatous change. (A, ×100; B-D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 181

disorders, or painful scoliosis.43,89,91 In unusual cases, the associated with vitamin D-resistant rickets.69 The peak
tumor may induce a pronounced inflammatory reaction age incidence and the most frequent sites of skeletal
in the surrounding soft tissue similar to the reaction that involvement of aggressive osteoblastoma are shown in
occurs in osteoid osteoma.32 The preferred primary treat- Figure 4-34. The skeletal and age distribution patterns
ment is surgical en bloc excision. Complete removal of of individual aggressive osteoblastomas are shown in
the tumor is not always possible, especially in the spine. Figure 4-35.
In such instances, radiotherapy is sometimes used to
control the disease. The recurrence rate after surgical
removal may be as high as 25%. In some cases, there may
Radiographic Imaging
be multiple recurrences. Malignant transformation has Aggressive osteoblastomas share many of the radio-
been reported rarely for osteoblastoma.39,40,78,79,82,114,131 graphic features of conventional osteoblastoma. The
lesion consists of a circumscribed lytic defect sometimes
surrounded by a rim of sclerosis (Fig. 4-36). The bone
contour can be markedly expanded and have a rim of
AGGRESSIVE OSTEOBLASTOMA reactive bone. The main difference from conventional
Definition osteoblastoma is that aggressive osteoblastomas are
larger, usually exceeding 4 cm in diameter.42 In our series,
Aggressive osteoblastoma is a rare tumor that represents the range of maximum dimensions was 2 to 8.5 cm. The
a borderline lesion between benign osteoblastoma and vertebral lesions do not differ in appearance from con-
osteosarcoma. These tumors are likely to recur, do not ventional osteoblastoma and represent lucent masses
metastasize, and are characterized microscopically by the located in the posterior elements of the neural arch
presence of so-called epithelioid osteoblasts. Transition to (Fig. 4-37). Similarly, the lesions of the sacrum are located
osteosarcoma has not been observed to date, and these within its dorsal elements and expand posteriorly
rare tumors therefore are not considered to be precursors (Fig. 4-38). The lesions of long bones and the skull may
of conventional osteosarcoma. have a distinct rim of moderate sclerosis. In small bones,
the soft tissue is frequently involved (Fig. 4-39).
In small anatomic structures (vertebral column, bones
Incidence and Location of the hands and feet), the lesion may cross a joint space
This is a very rare tumor, and its true incidence and its to involve the adjacent bone, providing clear evidence of
age distribution are not exactly known. The original its local aggressive nature. In some cases, prominent peri-
series, reported in 1984, consisted of 15 cases,39 and an osteal new bone formation can be present, raising the
update published in 199641 dealt with 21 additional cases radiologic suspicion of malignancy. The osteoblastic
of aggressive osteoblastoma. A further 11 cases observed nature of the lesion can sometimes be documented by the
as consultations since 1996 are included in the present presence of patchy, cloudlike opacities (Fig. 4-39). Similar
chapter. The limited experience with these tumors based to conventional osteoblastomas, aggressive osteoblas-
on the analyses of 47 cases indicates that they occur in a toma can lead to the development of secondary aneurys-
slightly older group of patients than conventional osteo- mal bone cysts that present as expansile destructive
blastomas. The peak age incidence is in the second and lesions (Fig. 4-40).
third decade of life. The ages of 47 patients with aggres-
sive osteoblastoma ranged between 7 and 80 years. Only
3 patients were younger than age 10 years. The male-to-
Gross Findings
female ratio in this group was 1.5 : 1 Fifteen of the tumors The gross appearance of aggressive osteoblastoma is
were in the vertebral column, three in the sacrum, one in similar to that of osteoblastoma. Intact in its anatomic
the scapula, five in the proximal femur, one in the setting, the lesion is oval or round with well-defined
humerus, four in the ilium, one in the pubic ramus, three margins (Figs. 4-41 and 4-48). The tissue is granular,
in the skull, two in the mandible, and one in the maxilla. friable, and reddish in color. Similar to conventional
Six tumors were located in small bones of the hands and osteoblastoma, it may bleed profusely during curettage
feet. This indicates that the overall distribution pattern because of its rich stromal vasculature. The bone contour
of aggressive osteoblastoma is similar to that of conven- can be markedly expanded with a thinned and focally
tional osteoblastoma, with clear predilection for the disrupted cortex. Extension into the adjacent soft tissue
axial skeleton. The second most frequent location of may be present, often with an encasing shell of reactive
occurrence is in the small bones of the hands and feet. bone peripherally.42 The main difference from conven-
This distribution pattern is clearly different from that tional osteoblastoma is the size of the lesion, which typi-
of conventional osteosarcoma, further supporting the cally exceeds 4 cm in diameter.
close pathogenetic relationship between aggressive
osteoblastoma and conventional osteoblastoma. Aside
from the personally collected series reported here and
Microscopic Findings
based largely on consultation material, the recent litera- Aggressive and conventional osteoblastomas share many
ture contains individual case reports of these very rare of the same microscopic features. The main difference
tumors.1,2,10,73,101,118,127 Multifocal, primary soft tissue, and and the most prominent feature of aggressive osteoblas-
cutaneous aggressive osteoblastomas have also been toma is the presence of so-called epithelioid osteoblasts that
described.27,33,38,72 Aggressive osteoblastomas can also be Text continued on p. 189

ERRNVPHGLFRVRUJ
Peak
Incidence
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 4-34  ■  Aggressive osteoblastoma. Peak age incidence and frequent sites of skeletal involvement.

Aggressive Osteoblastoma
16 20
Mean Age: 25.4
14 18 Male/Female Ratio: 1.61
16
12
14
Number of Cases

Number of Cases

10
12

8 10

8
6
6
4
4
2
2

0 0
1 2 3 4 5 6 7 8 9
Vertebra

Maxilla

Sacrum

Skull

Mandible

Pelvis

Scapula

Humerus

Tarsal

Phalanx

Metatarsal
Femur

Age in Decades
B

Flat Major Small bones of


bones long bones hands and feet

Axial Appendicular
skeleton skeleton
A
FIGURE 4-35  ■  Skeletal sites and age frequency distribution patterns of aggressive osteoblastoma. A, Skeletal distribution of aggres-
sive osteoblastoma. B, Age frequency distribution of aggressive osteoblastoma.
ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 183

A B

C D
FIGURE 4-36  ■  Aggressive osteoblastoma: radiographic features. A, Ill defined tumor with irregular mineralization pattern and diffuse
sclerosis of the adjacent bone involving proximal tibia. B, Mixed lytic and sclerotic tumor with ill defined margins and sclerosis in
the adjacent bone of proximal tibia. C, Ill defined lytic tumor of distal femur with sclerosis in the adjacent bone. D, Mixed lytic and
sclerotic tumor with ill defined margins involving the medial condyle of femur.

ERRNVPHGLFRVRUJ
184 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-37  ■  Aggressive osteoblastoma of vertebra. A, Radiograph of cervical spine in a 19-year-old man who had pain and par-
esthesias for 112 years. Expanded lytic lesion involves mainly transverse process of C5. B, Computed tomogram of case in A shows
expanded but well-delimited lesion with focal radiodensities in lateral aspect of vertebra. Although body is involved, lesion appears
to originate in lateral and posterior vertebral elements. C, Axial computed tomogram (bottom) and sagittal cut scout film (top).
Patient was a 53-year-old man with 3-week history of back pain radiating to lower extremities. D, Axial section of vertebra shown
in A documents eccentric lytic lesion primarily involving vertebral body and extending posteriorly into spinal canal.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 185

B C
FIGURE 4-38  ■  Aggressive osteoblastoma of sacrum with secondary aneurysmal bone cyst. Patient, a 16-year-old girl, incidentally
was found to have osteopoikilosis, which is shown in radiographs of pelvis. A, Anteroposterior view reveals lytic lesion in left side
of sacrum. Punctate opacities are not related and merely represent osteopoikilosis. B, Lateral view of sacrum shows posteriorly
expanded contour of tumor with superimposed aneurysmal bone cyst. C, Computed tomogram reveals expanded lytic lesion with
radiopaque delimiting capsule.

ERRNVPHGLFRVRUJ
186 4  Benign Osteoblastic Tumors

A B C

D E
FIGURE 4-39  ■  Aggressive osteoblastoma of short tubular bones. A, Radiograph of right long finger of a 19-year-old woman shows
lytic area of recurrent tumor in middle phalanx. Benign osteoblastic tumor was removed from this site 1 year previously. Review
of this material and histologic study of recurrence showed aggressive osteoblastoma. B, Second recurrence, 2 years later, shows
extension to adjacent soft tissue. This led to ray amputation because of likelihood of further recurrences. C, Sagittally cut amputa-
tion specimen shows tumor in middle phalanx that extends into soft tissue on volar aspect. D, Radiograph of thumb metacarpal
tumor in a 21-year-old man who had pain and swelling for 5 months. The patient received radiation therapy, and amputation was
done 1 year later. Note expanded cortical contour and dense radiopacities within lesion. E, Forty-six-year-old woman with painless
swelling of her right second toe of several years’ duration. Proximal phalanx shows that bone-forming tumor has caused extensive
destruction and has extended into soft tissue.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 187

A B

C D
FIGURE 4-40  ■  Aggressive osteoblastoma involving right superior pubic ramus. A, Radiograph of a 18-year-old man 3 months before
admission shows 5-cm lytic lesion at junction of acetabulum and ramus. B, Large, expanded mass at same site 3 months later is
shown in this anteroposterior film. C, Computed tomogram of pelvis. The “blown-out” area of juxtaacetabular bone represents
secondary aneurysmal bone cyst superimposed on aggressive osteoblastoma. D, Radioisotope scan shows marked uptake at site
of combined bone-forming tumor and secondary aneurysmal bone cyst.

ERRNVPHGLFRVRUJ
188 4  Benign Osteoblastic Tumors

A C

D E
FIGURE 4-41  ■  Aggressive osteoblastoma involving left acetabular region of ilium. A, Anteroposterior radiograph of a 61-year-old
woman with 6-month history of increasing left hip pain shows well-circumscribed lytic lesion with central opacity (arrows). B and
C, Computed tomograms show lesion adjacent to acetabulum and reactive sclerosis in surrounding bone. D, Specimen radiograph
shows multiple areas of patchy density not visible on computed tomogram or plain film. E, Specimen photograph shows granular
hemorrhagic tumor with bony central foci.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 189

rim osteoid trabeculae and occasionally form loosely sheets that fill intertrabecular spaces as they do in aggres-
cohesive solid sheets that fill the intertrabecular spaces sive osteoblastoma.
(Fig. 4-42). Large solid areas of epithelioid osteoblasts The distinction from osteosarcoma is based on the
with mild to moderate atypia can be present (Figs. 4-43 absence of cellular atypia, high mitotic rate, atypical
to 4-45).1,28,29,31,86,98,104,105 mitotic figures, prominent and abundant lacelike osteoid
At low magnification, the lesion consists of an irreg- deposition, permeative growth into adjacent bone and
ular network of bone trabeculae haphazardly distrib- soft tissue, and presence of neoplastic cartilage. The pres-
uted in a loose fibroblastic stroma with a prominent ence of a peripheral shell of reactive bone over the soft
vasculature. The trabeculae occasionally appear wider tissue extensions of aggressive osteoblastomas also helps
and more irregular than those of conventional osteo- distinguish this lesion from osteosarcoma. Sampling of
blastoma. The deposition of osteoid may focally be in the borders of the lesion is necessary to establish its pres-
a nontrabecular, lacelike pattern surrounding individual ence. Furthermore, all the comments in the discussion of
cells. There may also be broad zones of solid osteoid differential diagnosis of benign osteoblastoma also apply
enveloping individual epithelioid osteoblasts (Figs. 4-46 to aggressive osteoblastoma.
and 4-47).
Chondroid differentiation has not been observed in
aggressive osteoblastomas. Numerous benign osteoclast-
Personal Comments
type multinucleated giant cells are often present. In some The principal diagnostic problem with regard to the enti-
cases, they may be present only focally. ties classified as osteoblastoma and aggressive osteoblas-
As mentioned, the presence of epithelioid osteoblasts toma centers around their distinction from osteosarcoma.
is a hallmark of aggressive osteoblastoma. These cells are This problem is further complicated by a recently pro-
round; are at least twice the size of ordinary osteoblasts; posed category of low-grade osteosarcoma that resembles
and show abundant eosinophilic cytoplasm and an eccen- osteoblastoma (osteoblastoma-like osteosarcoma).14
tric large, oval or round nucleus with a prominent nucle- We believe that aggressive osteoblastoma is a separate
olus (Fig. 4-48). Within the cytoplasm, there is usually a entity and should not be confused with osteosarcoma.
large, clear area that sometimes compresses and displaces Furthermore, there is as yet no evidence that it undergoes
the nucleus. Ultrastructurally, this structure represents an spontaneous transformation to osteosarcoma. In our col-
enlarged Golgi center. The remaining cytoplasm has a lected series, 19 of the 47 cases have recurred one or
few mitochondria and numerous prominent channels of more times (40%), which is significantly higher than the
rough endoplasmic reticulum. The cells form loosely 25% risk for benign osteoblastomas. We have not seen a
cohesive sheets, with the formation of small, irregular, well-documented case of aggressive osteoblastoma that
glandlike spaces seen ultrastructurally. The mitotic activ- has shown metastatic behavior.
ity within the epithelioid osteoblasts ranges from 1 to 4 All the diagnostic criteria cited are important in evalu-
mitoses per 20 high-power fields. Atypical mitoses are ating the biologic potential of an osteoblastic tumor. The
not present. The mitotic activity of the associated stromal mere finding of epithelioid osteoblasts does not qualify
cells is very low or absent. No mitotic activity is present the lesion as an aggressive osteoblastoma. Indeed, it is
within the giant osteoclast-like cells. possible to find plump epithelioid osteoblastic cells in
Gross and microscopic evidence of secondary aneurys- conventional osteosarcomas otherwise containing frankly
mal bone cyst is frequently present. Microscopic evidence malignant histologic features. This approach was the
of a peripheral shell of reactive bone bordering soft basis for an assertion in one published series that aggres-
tissue extensions is an interesting feature of this tumor sive osteoblastomas are merely low-grade osteosarcomas,
and one that it shares with giant cell tumor of bone superficially resembling osteoblastomas.15 This concept
(Fig. 4-40). has since been modified because the series on which it
was based included clearly identifiable high-grade osteo-
sarcomas that focally simulated osteoblastomas. The same
Differential Diagnosis authors have recently embraced the concept of aggressive
osteoblastoma as a distinct entity.14 The established histo-
Aggressive osteoblastoma should be differentiated from logic criteria for the recognition of aggressive osteoblas-
osteoid osteoma, benign osteoblastoma, and osteosarcoma. The toma were found to be predictive of a significantly higher
distinction from osteoid osteoma and osteoblastoma is recurrence rate than that of conventional osteoblastoma
based on size of the lesion and the presence of epithelioid and of an absence of metastasizing potential.9
osteoblasts. We believe that all aggressive osteoblastomas should
Although occasional isolated large osteoblasts with be treated by wide local excision when technically
abundant cytoplasm can be seen in osteoid osteoma or feasible.
benign osteoblastoma, they do not occur in cohesive Text continued on p. 197

ERRNVPHGLFRVRUJ
190 4  Benign Osteoblastic Tumors

A C
FIGURE 4-42  ■  Aggressive osteoblastoma: radiographic and microscopic features. A, Anteroposterior radiograph showing well-
circumscribed lytic tumor involving lateral malleolus. Note periosteal reaction proximally and expansion of bone contour. B, Mag-
netic resonance imaging shows tumor mass of low signal intensity expanding the bone contour. Note sharp demarcation of the
medullary margin. C, Photomicrograph of the curetted specimen showing irregular trabeculae or bone with intertrabecular spaces
filled by plump epithelioid osteoblasts. (C, ×100). (C, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 191

A B

C D
FIGURE 4-43  ■  Aggressive osteoblastoma: microscopic features. A, Solid areas of so-called epithelioid osteoblasts (×100). B, Higher
magnification of A. Note numerous epithelioid osteoblasts that are at least 2 times the size of those of ordinary osteoid osteoma
and osteoblastoma. C, Solid areas of epithelioid osteoblasts. D, Higher magnification of epithelioid osteoblasts with small foci of
osteoid-like deposits. Note large eccentrically located nuclei with prominent irregular nucleoli. (A and C, ×100; B and D, ×200) (A-D,
hematoxylin-eosin)

ERRNVPHGLFRVRUJ
192 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-44  ■  Aggressive osteoblastoma: microscopic features. A, Low power magnification of osteoblastoma with solid areas
of epithelioid osteoblasts. B, Higher magnification of A showing solid proliferation of large epithelioid osteoblasts.
Note some mild degree of atypia. C, Low power magnification of osteoblastoma with solid areas of epithelioid osteoblasts. D, Higher
magnification of C shows solid proliferation of osteoblasts with immature osteoid deposition. (A and C, ×100; B and D, ×200) (A-D,
hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 193

A B

C D
FIGURE 4-45  ■  Aggressive osteoblastoma: microscopic features. A, Low power magnification of osteoblastoma with solid areas of
epithelioid osteoblasts. B, Higher magnification of A showing solid proliferation of large epithelioid osteoblasts with prominent
nucleoli. C, Low power magnification of osteoblastoma with solid areas of epithelioid osteoblasts and irregular deposition of imma-
ture osteoid. D, Higher magnification of C shows solid proliferation of epithelioid osteoblasts and deposition of unmineralized
immature osteoid. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
194 4  Benign Osteoblastic Tumors

A B

C D
FIGURE 4-46  ■  Aggressive osteoblastoma: patterns of osteoid deposition. A, Low power magnification shows confluent sclerotic
areas of osteoid and solid proliferations of epithelioid osteoblasts. B, Higher magnification of A shows confluent areas of osteoid
and solid foci of epithelioid osteoblasts. C and D, Patterns of osteoid deposition in form of irregular trabeculae of woven bone with
intertrabecular spaces filled by solid proliferations of epithelioid osteoblasts. Such patterns of osteoid deposition associated with
solid proliferation of atypical epithelioid osteoblastic cells may be misinterpreted as malignancy, leading to an incorrect diagnosis
of osteosarcoma. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 195

A B

C D
FIGURE 4-47  ■  Aggressive osteoblastoma: patterns of osteoid deposition. A and B, Low and intermediate power photomicrographs
showing osteoid deposition associated with atypical epithelioid osteoblastic cells. C and D, Low and intermediate power photomi-
crographs showing osteoid deposition associated with atypical epithelioid osteoblastic cells. Such patterns of tumor osteoid associ-
ated with atypical epithelioid osteoblasts may be misinterpreted as malignancy, leading to an incorrect diagnosis of osteosarcoma.
(A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
196 4  Benign Osteoblastic Tumors

A B

D E
FIGURE 4-48  ■  Aggressive osteoblastoma of talus. A, Lateral radiograph of foot of a 27-year-old man with painful lesion of antero-
superior portion of talus. Note soft tissue calcifications. B, Tomogram shows sharply demarcated lytic lesion. C, Gross photograph
of bisected specimen of excised talus. Note granular reddish tumor tissue within medullary cavity and bulging into adjacent soft
tissue (arrows). D, Whole-mount photomicrograph shows intraosseous and extraosseous components of aggressive osteoblastoma.
Shell of reactive bone is present at periphery of tumor extension into soft tissue (arrow). E, High power photomicrograph showing
solid proliferation of epithelioid osteoblasts. Note prominent nucleoli and dense eozynophilic cytoplasm with prominent Golgi
centers seen as cytoplasmic halo displacing nucleus. (E, ×400) (E, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 197

29. Chun KA, Cho IH, Won KJ, et al: Osteoblastoma as a cause of
REFERENCES osteomalacia assessed by bone scan. Ann Nucl Med 17(5):411–414,
1. Adler M, Hnatuk L, Mock D, et al: Aggressive osteoblastoma 2003.
of the temporal bone: a case report. J Otolaryngol 19:307–310, 30. Ciabattoni G, Tamburrelli F, Greco F: Increased prostacyclin bio-
1990. synthesis in patients with osteoid osteoma. Eicosanoids 4:165–167,
2. Angervall L, Persson S, Stenman G, et al: Large cell, epithelioid, 1991.
telangiectatic osteoblastoma: a unique pseudosarcomatous variant 31. Cribb GL, Goude WH, Cool P, et al: Percutaneous radiofre-
of osteoblastoma. Hum Pathol 30(10):1254–1259, 1999. quency thermocoagulation of osteoid osteomas: factors affecting
3. Aparisi T, Arborgh B, Ericsson JL: Studies on the fine structure therapeutic outcome. Skeletal Radiol 34(11):702–706, 2005.
of osteoblastoma with notes on the localization of nonspecific acid 32. Crim JR, Mirra JM, Eckardt JJ, et al: Widespread inflammatory
and alkaline phosphatase. Cancer 41:1811–1822, 1978. response to osteoblastoma: the flare phenomenon. Radiology 177:
4. Atesok KI, Alman BA, Schemitsch EH, et al: Osteoid osteoma and 835–836, 1990.
osteoblastoma. J Am Acad Orthop Surg 19:678–689, 2011. 33. Vinuth DP, Agarwal P, Gadewar D, et al: A large multifocal
5. Athwal P, Stock H: Osteoid osteoma: a pictorial review. Conn Med aggressive osteoblastoma of mandible: an immunohistochemistry
48:233–235, 2014. case study report. J Dent Res Dent Clin Dent Prospects 8:51–55,
6. Ayala AG, Murray JA, Erling MA, et al: Osteoid-osteoma: intra- 2014.
operative tetracycline-fluorescence demonstration of the nidus. 34. Dal Cin P, Sciot R, Samson I, et al: Osteoid osteoma and osteo-
J Bone Joint Surg 68A:747–751, 1986. blastoma with clonal chromosome changes. Br J Cancer 78(3):344–
7. Aynaci O, Turgutoglu O, Kerimoglu S, et al: Osteoid osteoma 348, 1998.
with a multicentric nidus: a case report and review of the litera- 35. Dale S, Breidahl WH, Baker D, et al: Severe toxic osteoblastoma
ture. Arch Orthop Trauma Surg 127:863–866, 2007. of the humerus associated with diffuse periostitis of multiple
8. Azouz EM, Kozlowski K, Marton D, et al: Osteoid osteoma and bones. Skeletal Radiol 30(8):464–468, 2001.
osteoblastoma of the spine in children: report of 22 cases with 36. Dancer JY, Henry SP, Bondaruk J, et al: Expression of master
brief literature review. Pediatr Radiol 16:25–31, 1986. regulatory genes controlling skeletal development in benign car-
9. Bahk WJ, Mirra JM: Pseudoanaplastic tumors of bone. Skeletal tilage and bone forming tumors. Hum Pathol 41:1788–1793, 2010.
Radiol 33(11):641–648, 2004. 37. Davidson RS, Mahboubi S, Heyman S, et al: Nondiaphyseal
10. Baker AC, Rezeanu L, Kelin MJ, et al: Aggressive osteoblastoma: osteoid osteomas in the pediatric patient. Clin Orthop 243:230–
a case report involving a unique chromosomal aberration. Int J 234, 1989.
Surg Pathol 18:219–224, 2010. 38. Deyrup AT, Monson DK, Dorfman HD: Aggressive (epithelioid)
11. Baruffi MR, Volpon JB, Neto JB, et al: Osteoid osteoma with osteoblastoma arising in soft tissue. Int J Surg Pathol 16:308–310,
chromosome alterations involving 22q. Cancer Genet Cytogenet 2008.
124(2):127–131, 2001. 39. Dorfman HD: Malignant transformation of benign bone lesions.
12. Bauer TW, Zehr JR, Belhobek GH, et al: Juxta-articular osteoid Proc Natl Cancer Conference 7:901–913, 1972.
osteoma. Am J Surg Pathol 15:381–387, 1991. 40. Dorfman HD: Case records of the Massachusetts General Hos-
13. Berry M, Mankin H, Gebhardt M, et al: Osteoblastoma: a 30-year pital, case 40. N Engl J Med 303:866–873, 1980.
study of 99 cases. J Surg Oncol 98:179–183, 2008. 41. Dorfman HD: Aggressive osteoblastoma revisited: twenty years
14. Bertoni F, Donati D, Bacchini P, et al: The morphologic spectrum experience with a still controversial entity. J. Jpn Orthop Assoc
of osteoblastoma: is its “aggressive” nature predictable? Lab Invest 70(6):S1013, 1996.
66:3A, 1992 (abstract). 42. Dorfman HD, Weiss SW: Borderline osteoblastic tumors: prob-
15. Bertoni F, Unni KK, McLeod RA, et al: Osteosarcoma resembling lems in the differential diagnosis of aggressive osteoblastoma and
osteoblastoma. Cancer 55:416–426, 1985. low-grade osteosarcoma. Semin Diagn Pathol 1:215–234, 1984.
16. Bettelli G, Capanna R, van Horn JR, et al: Osteoid osteoma 43. Fakharani-Hein M, Griss P, Ludke A, et al: Rapidly developing
and osteoblastoma of the pelvis. Clin Orthop 247:261–271, scoliosis in an adolescent due to spinal osteoblastoma: a case
1989. report. Arch Orthop Trauma Surg 107:259–262, 1988.
17. Bettelli G, Tigani D, Picci P: Recurring osteoblastoma initially 44. Garcia RA, Inwards CY, Unni KK: Benign bone tumors—recent
presenting as a typical osteoid osteoma: report of two cases. Skel- developments. Semin Diagn Pathol 28:73–85, 2011.
etal Radiol 20:1–4, 1991. 45. Gille P, Gross P, Brax P, et al: Osteoid osteoma of the acetabulum:
18. Bilchik T, Heyman S, Siegel A, et al: Osteoid osteoma: the role two cases. J Pediatr Orthop 10:416–418, 1990.
of radionuclide bone imaging, conventional radiography and com- 46. Gitelis S, Schajowicz F: Osteoid osteoma and osteoblastoma.
puted tomography in its management. J Nucl Med 33:269–271, Orthop Clin North Am 20:313–325, 1989.
1992. 47. Greco F, Tamburrelli F, Ciabattoni G: Prostaglandins in osteoid
19. Bordelon RL, Cracco A, Book MK: Osteoid osteoma producing osteoma. Int Orthop 15:35–37, 1991.
premature fusion of the epiphysis of the distal phalanx of the big 48. Healy JH, Ghelman B: Osteoid osteoma and osteoblastoma:
toe. J Bone Joint Surg 57A:120–122, 1975. current concepts and recent advances. Clin Orthop 204:76–85,
20. Boriani S, Amendola L, Bandiera S, et al: Staging and treatment 1986.
of osteoblastoma in the mobile spine: a review of 51 cases. Eur 49. Helms CA: Osteoid osteoma: the double density sign. Clin Orthop
Spine J 21:2003–2010, 2012. 222:167–173, 1987.
21. Boriani S, Capanna R, Donati D, et al: Osteoblastoma of the 50. Helms CA, Hattner RS, Vogler JB, III: Osteoid osteoma: radio-
spine. Clin Orthop 278:37–45, 1992. nuclide diagnosis. Radiology 151:779–784, 1984.
22. Boscainos PJ, Cousins GR, Kulshreshtha R, et al: Osteoid 51. Jackson RP, Reckling FW, Mants FA: Osteoid osteoma and osteo-
osteoma. Orthopedics 36:792–800, 2013. blastoma: similar histologic lesions with different natural histories.
23. Bowen CV, Dzus AK, Hardy DA: Osteoid osteomata of the distal Clin Orthop 128:303–313, 1977.
phalanx. J Hand Surg 12:387–390, 1987. 52. Jaffe H: Osteoid osteoma”: a benign osteoblastic tumor composed
24. Brabants K, Geens S, van Damme B: Subperiosteal juxtaarticular of osteoid and atypical bone. Arch Surg 31:709–728, 1935.
osteoid osteoma. J Bone Joint Surg 68B:320–324, 1986. 53. Jaffe HL: Benign osteoblastoma. Bull Hosp J Dis 17:141–151,
25. Bruneau M, Polivka M, Cornelius JF, et al: Progression of an 1956.
osteoid osteoma to an osteoblastoma. Case report. J Neurosurg 54. Jaffe HL, Mayer L: An osteoblastic-osteoid tissue forming tumor
Spine 3:238–241, 2005. of a metacarpal bone. Arch Surg 24:550–564, 1932.
26. Burger IM, McCarthy EF: Phalangeal osteoid osteomas in the 55. Kalil RK, Antunes JS: Familial occurrence of osteoid osteoma.
hand: a diagnostic problem. Clin Orthop Relat Res 427:198–203, Skeletal Radiol 7:416–419, 2003.
2004. 56. Kattapuram SV, Kushner DC, Phillips WC, et al: Osteoid
27. Cannavo SP, Marafioti T: Cutaneous aggressive osteoblastoma. osteoma: an unusual cause of articular pain. Radiology 147:383–
Int J Dermatol 35:504–505, 1996. 387, 1983.
28. Cassar-Pullicino VN, McCall IW, Wan S: Intra-articular osteoid 57. Kawaguchi Y, Sato C, Hasegawa T, et al: Intraarticular osteoid
osteoma. Clin Radiol 45:153–160, 1992. osteoma associated with synovitis: a possible role of

ERRNVPHGLFRVRUJ
198 4  Benign Osteoblastic Tumors

cyclooxygenase-2 expression by osteoblasts in the nidus. Mod 86. Morton KS, Quenville NF, Beauchamp CP: Aggressive osteoblas-
Pathol 10:1086–1091, 2000. toma: a case previously reported as a recurrent osteoid osteoma.
58. Keim HA, Reina EG: Osteoid osteoma as a cause of scoliosis. J Bone Joint Surg 71B:428–431, 1989.
J Bone Joint Surg 57A:159–163, 1975. 87. Mungo DV, Zhang X, O’Keefe RJ, et al: COX-1 and COX-2
59. Kenan S, Floman Y, Robin GC, et al: Aggressive osteoblastoma: expression in osteoid osteomas. J Orthop Res 20(1):159–162,
a case report and review of the literature. Clin Orthop 195:294– 2002.
298, 1985. 88. Muren C, Hoglund M, Engkvist O, et al: Osteoid osteomas of the
60. Keret D, Harcke HT, MacEwen D, et al: Multiple osteoid osteo- hand: report of three cases and review of the literature. Acta Radiol
mas of the fifth lumbar vertebra: a case report. Clin Orthop 32:62–66, 1991.
248:163–168, 1989. 89. Myles ST, MacRae ME: Benign osteoblastoma of the spine in
61. Khurana JS, Mayo-Smith W, Kattapuram SV: Subtalar arthralgia childhood. J Neurosurg 68:884–888, 1988.
caused by juxtaarticular osteoid osteoma. Clin Orthop 252:205– 90. Nakatani T, Yamamoto T, Akisue T, et al: Periosteal osteoblas-
208, 1990. toma of the distal femur. Skeletal Radiol 33(2):107–111, 2004.
62. Kiers L, Shield LK, Cole WG: Neurological manifestations of 91. Nemoto O, Moser RP, Jr, Van Dam BE, et al: Osteoblastoma of
osteoid osteoma. Arch Dis Child 65:851–855, 1990. the spine: a review of 75 cases. Spine 15:1272–1280, 1990.
63. Klein MH, Shankman S: Osteoid osteoma: radiologic and patho- 92. Nguyen VD, Hersh M: A rare bone tumor in an unusual location:
logic correlation. Skeletal Radiol 21:23–31, 1992. osteoblastoma of the vertebral body. Comput Med Imaging Graph
64. Klonecke AS, Licho R, McDougall IR: A technique for intraop- 16:11–16, 1992.
erative bone scintigraphy: a report of 17 cases. Clin Nucl Med 93. Nord KH, Nilsson J, Arbajian E, et al: Recurrent chromosome 22
16:482–486, 1991. deletions in osteoblastoma affect inhibitors of the Wnt/beta-
65. Kneisl JS, Simon MA: Medical management compared with oper- catenin signaling pathway. PLoS ONE 8:e80725, 2013.
ative treatment for osteoid osteoma. J Bone Joint Surg 74A:179– 94. Norman A, Abdelwahab IF, Buyon J, et al: Osteoid osteoma of the
185, 1992. hip stimulating an early onset of osteoarthritis. Radiology 158:417–
66. Kroon HM, Schurmans J: Osteoblastoma: clinical and radiologic 420, 1986.
findings in 98 new cases. Radiology 175:783–790, 1990. 95. Norman A, Dorfman HD: Osteoid osteoma inducing pronounced
67. Ledeboer QC, Kerrebijn JD, Govaere F, et al: Osteoblastoma of overgrowth and deformity of bone. Clin Orthop 10:233–238,
the larynx: a case report. Int J Surg Pathol 13(4):365–367, 2005. 1975.
68. Lee DH, Malawer MM: Staging and treatment of primary and 96. Papanicolaou N: Osteoid osteoma: operative confirmation of
persistent (recurrent) osteoid osteoma: evaluation of intraopera- complete removal by bone scintigraphy. Radiology 154:821–822,
tive nuclear scanning, tetracycline fluorescence, and tomography. 1985.
Clin Orthop 281:229–238, 1992. 97. Pettine KA, Klassen RA: Osteoid osteoma and osteoblastoma of
69. Lee DY, Choi IH, Lee CK, et al: Acquired vitamin D-resistant the spine. J Bone Joint Surg 68A:354–361, 1986.
rickets caused by aggressive osteoblastoma in the pelvis: a case 98. Pieterse AS, Vernon-Roberts B, Paterson DC, et al: Osteoid
report with 10 years’ follow-up and review of the literature. osteoma transforming to aggressive (low grade malignant) osteo-
J Pediatr Orthop 14:793–798, 1994. blastoma: a case report and literature review. Histopathology 7:789–
70. Lichtenstein L: Benign osteoblastoma: a category of osteoid- and 800, 1983.
bone-forming tumors other than classical osteoid osteoma, which 99. Poey C, Clement JL, Baunin C, et al: Joomye H, Chicoisne MP,
may be mistaken for giant-cell-tumor or osteogenic sarcoma. Bonnevialle P, Railhac JJ: Percutaneous extraction of an osteoid
Cancer 9:1044–1052, 1956. osteoma of the lumbar spine under CT guidance. J Comput Assist
71. Lichtenstein L, Sawyer WF: Benign osteoblastoma: further obser- Tomogr 15:1056–1058, 1991.
vations and report of twenty additional cases. J Bone Joint Surg 100. Radig K, Schneider-Stock R, Mittler U, et al: Genetic instability
45A:755–765, 1964. in osteoblastic tumors of the skeletal system. Pathol Res Pract
72. Lin JH, Lin EY, Liu MY: Double aggressive osteoblastomas of 194(10):669–677, 1998.
calvarium. Br J Neurosurg 21:626–628, 2007. 101. Ragois P, Leclerc P, Hallonet D: Aggressive osteoblastoma of
73. Lu ZH, Cao WH, Qian WX: Aggressive osteoblastoma of the the carpal scaphoid bone. Rev Chir Orthop Reparatrice Appar Mot
temporal bone: a case report and review of the literature. Clin 86(8):851, 2000.
Imaging 37:386–389, 2013. 102. Ransford AO, Pozo JL, Hutton PA, et al: The behaviour pattern
74. Lucas DR, Unni KK, McLeod RA, et al: Osteoblastoma: clinico- of the scoliosis associated with osteoid osteoma or osteoblastoma
pathologic study of 306 cases. Hum Pathol 25:117–134, 1994. of the spine. J Bone Joint Surg 66B:16–20, 1984.
75. Magnan B, Caudana R, Morelli N, et al: The contribution of 103. Regan MW, Galey JP, Oakeshott RD: Recurrent osteoid osteoma:
magnetic resonance imaging in a rare ischiatic localization of case report with a ten-year asymptomatic interval. Clin Orthop
osteoid osteoma. Ital J Orthop Traumatol 17:407–411, 1991. 253:221–224, 1990.
76. Makley JT, Dunn M: Prostaglandins: a mechanism for pain medi- 104. Revell PA, Scholtz CL: Aggressive osteoblastoma. J Pathol 127:
ation in osteoid osteoma. Orthop Trans 6:72, 1982. 195–198, 1979.
77. Marsh BW, Bonfiglio M, Brady LP, et al: Benign osteoblastoma: 105. Roessner A, Metze K, Heymer B: Aggressive osteoblastoma.
range of manifestations. J Bone Joint Surg 57A:1–9, 1975. Pathol Res Pract 179:433–438, 1985.
78. Mayer L: Malignant degeneration of so-called benign osteoblas- 106. Rosenthal DI, Alexander A, Rosenberg AE, et al: Ablation of
toma. Bull Hosp J Dis 28:4–13, 1967. osteoid osteomas with a percutaneously placed electrode: a new
79. Mayer L: Benign osteoblastoma. Bull Hosp J Dis 29:236–240, procedure. Radiology 183:29–33, 1992.
1968–1969 (letter). 107. Rosenthal DI, Hornicek FJ, Toriani M, et al: Osteoid osteoma:
80. Mazoyer JF, Kohler R, Bossard D: Osteoid osteoma: CT-guided percutaneous treatment with radiofrequency energy. Radiology
percutaneous treatment. Radiology 181:269–271, 1991. 232(1):304, 2003.
81. McLeod RA, Dahlin DC, Beabout JW: The spectrum of osteo- 108. Ruggieri P, Biagini R, Ferraro A, et al: Osteoid osteoma of the
blastoma. Am J Roentgen 126:321–335, 1976. elbow: a study of twelve cases. Ital J Orthop Traumatol 15:154–163,
82. Merryweather R, Middlemiss JH, Sanerkin NG: Malignant trans- 1989.
formation of osteoblastoma. J Bone Joint Surg 62B:381–384, 1980. 109. Saccomanni B: Osteoid osteoma and osteoblastoma of the spine:
83. Mirra JM, Kendrick RA, Kendrick RE: Pseudomalignant osteo- a review of the literature. Curr Rev Musculoskelet Med 2:65–67,
blastoma versus arrested osteosarcoma. Cancer 37:2005–2014, 2009.
1976. 110. Schajowicz F, Lemos C: Osteoid osteoma and osteoblastoma. Acta
84. Mitchell ML, Ackerman LV: Metastatic and pseudomalignant Orthop Scand 41:272–291, 1970.
osteoblastoma: a report of two unusual cases. Skeletal Radiol 15: 111. Schajowicz F, Lemos C: Malignant osteoblastoma. J Bone Joint
213–218, 1986. Surg 58B:202–211, 1976.
85. Mohan V, Sabri T, Marklund T, et al: Clinicoradiological diagno- 112. Schlesinger AE, Hernandez RJ: Intracapsular osteoid osteoma of
sis of benign osteoblastoma of the spine in children. Arch Orthop the proximal femur: findings on plain film and CT. AJR Am J
Trauma Surg 110:260–264, 1991. Roentgenol 154:1241–1244, 1990.

ERRNVPHGLFRVRUJ
4  Benign Osteoblastic Tumors 199

113. Schulman L, Dorfman HD: Nerve fibers in osteoid osteoma. 124. Szypryt EP, Hardy JG, Colton CL: An improved technique of
J Bone Joint Surg 52A:1351–1356, 1970. intraoperative bone scanning. J Bone Joint Surg 68B:643–646,
114. Seki T, Fukuda H, Ishii Y, et al: Malignant transformation of 1986.
benign osteoblastoma: a case report. J Bone Joint Surg 57A:424– 125. Tonai M, Campbell CJ, Ahn GH, et al: Osteoblastoma: classifica-
426, 1975. tion and report of 16 patients. Clin Orthop 167:222–235, 1982.
115. Shaikh MI, Saifuddin A, Pringle J, et al: Spinal osteoblastoma: CT 126. van Horn JR, Karthaus RP: Epiphyseal osteoid osteoma: two case
and MR imaging with pathological correlation. Skeletal Radiol reports. Acta Orthop Scand 60:625–627, 1989.
28(1):33–40, 1999. 127. Vigneswaran N, Fernandes R, Rodu B, et al: Aggressive osteoblas-
116. Sherman MS, McFarland G: Mechanism of pain in osteoid osteo- toma of the mandible closely simulating calcifying epithelial
mas. South Med J 58:163–166, 1965. odontogenic tumor. Report of two cases with unusual histopatho-
117. Sim FH, Dahlin CD, Beabout JW: Osteoid osteoma: diagnostic logic findings. Pathol Res Pract 197(8):569–576, 2001.
problems. J Bone Joint Surg Am 57:154–159, 1975. 128. Voto SJ, Cook AJ, Weiner DS, et al: Treatment of osteoid osteoma
118. Singh DK, Das KK, Mehrotra A, et al: Aggressive osteoblas- by computed tomography guided excision in the pediatric patient.
toma involving the craniovertebral junction: a case report and J Pediatr Orthop 10:510–513, 1990.
review of the literature. J Craniovertebr Junction Spine 4:69–72, 129. Wells RG, Miller JH, Sty JR: Scintigraphic patterns in osteoid
2013. osteoma and spondylolysis. Clin Nucl Med 12:39–44, 1987.
119. Snarr JW, Abell MR, Martel W: Lymphofollicular synovitis with 130. Wold LE, Pritchard DJ, Bergert J, et al: Prostaglandin synthesis
osteoid osteoma. Radiology 106:557–560, 1973. by osteoid osteoma and osteoblastoma. Mod Pathol 1:129–131,
120. Steiner GC: Ultrastructure of osteoid osteoma. Hum Pathol 7: 1988.
309–325, 1976. 131. Wozniak AW, Nowaczyk MT, Osmola K, et al: Malignant trans-
121. Steiner GC: Ultrastructure of osteoblastoma. Cancer 39:2127– formation of an osteoblastoma of the mandible: case report and
2136, 1977. review of the literature. Eur Arch Otorhinolaryngol 267:845–849,
122. Sung HW, Liu C: Can osteoid osteoma become osteoblastoma? 2010.
A case report. Arch Orthop Trauma Surg 95:217–219, 1979. 132. Zhang X, Bukata SV, Healey JH, et al: Epiphyseal osteoblastoma
123. Swee RG, McLeod RA, Beabout JW: Osteoid osteoma: detection, of tibia with xanthomatous stromal reaction. Ann Diagn Pathol
diagnosis, and localization. Radiology 130:117–123, 1979. 7(6):360–364, 2003.

ERRNVPHGLFRVRUJ
C H A P T E R 5 

Osteosarcoma
CHAPTER OUTLINE

INTRAMEDULLARY OSTEOSARCOMA SURFACE OSTEOSARCOMA


Conventional Osteosarcoma Parosteal Osteosarcoma
Telangiectatic Osteosarcoma Periosteal Osteosarcoma
Well-differentiated Intramedullary High-grade Surface Osteosarcoma
Osteosarcoma
INTRACORTICAL OSTEOSARCOMA
OSTEOSARCOMA ASSOCIATED WITH SPECIFIC
EXTRASKELETAL OSTEOSARCOMA
CLINICAL SYNDROMES
SECONDARY OSTEOSARCOMA

Osteosarcoma is the most common primary malignant be profuse osteoid matrix production and extensive min-
tumor of bone. When all aspects of its presentation are eralization throughout the tumor, or both may be minimal
taken into consideration, it is evident that this term is in extent and very focally distributed. The osteoblastic
used to describe a heterogeneous group of lesions with nature of the tumor can be easy to identify both radio-
diverse morphology and clinical behavior. From the graphically and microscopically, or extensive sampling
morphologic point of view, osteosarcoma can be divided and a great deal of expertise may be required for its rec-
into several subgroups (e.g., osteoblastic, chondroblastic, ognition. In the past, the term osteogenic sarcoma (i.e.,
fibroblastic). These terms reflect the great microscopic sarcoma arising in bone) was used in a more general sense
variability of the tumor, and almost invariably a mixture to designate all sarcomatous neoplasms arising in bone.134
of several different components is present in one lesion. More recently, it was used interchangeably with osteosar-
It is understood that these terms are used in a descriptive coma, but this use of the term is no longer acceptable
sense only and do not necessarily imply difference in from the pathogenetic point of view.
prognosis or clinical behavior. With regard to the growth
pattern of these lesions, osteosarcomas can be subdivided
Incidence and Location
into lesions that originate and grow primarily inside the
bone (intramedullary osteosarcoma) and those that grow Osteosarcoma accounts for approximately 20% of all
on the surface of bone (surface osteosarcoma) within primary sarcomas of bone, excluding multiple myeloma
periosteal or parosteal tissue. Radiographically, lesions and other hematopoietic neoplasms. The most frequent
can be predominantly lytic or sclerotic, but usually there sites of skeletal involvement and the peaks of age inci-
is a combination of these features. There is no uniformity dences are shown in Figure 5-1. The age distribution is
among the many current classification systems of osteo- bimodal, with the first major peak occurring during the
sarcoma. However, all systems, including the one adopted second decade of life, and the second, much smaller, peak
in this chapter, are based on a combination of micro- being observed in patients older than age 50 years. The
scopic, radiographic, and clinical features that help sepa- presence of the second peak was originally documented
rate osteosarcoma into several categories with a distinct in the Memorial Sloan-Kettering Cancer Center series96
clinical behavior and prognosis. The system of classifica- and has been confirmed by data from the National Cancer
tion used here recognizes the diversity of osteosarcomas Institute’s Surveillance, Epidemiology, and End Results
and may reflect, at least to some extent, the different (SEER) Program56 (Fig. 5-2). The SEER Program, con-
pathways of their development (Table 5-1). taining population-based data on approximately 5000
cases of osteosarcoma, provides a detailed view on epide-
miologic trends and survival rates for osteosarcoma.56,157
The overall incidence rates are clearly age specific and
INTRAMEDULLARY OSTEOSARCOMA indicate that individuals, during the first two decades of
life and over age 60, have similar risk for the development
Conventional Osteosarcoma of osteosarcoma with incidence rates of 4.4 and 4.2
per million respectively. The population between the
Definition
third and sixth decades of life have a lower incidence
Osteosarcoma can be defined as a malignant tumor of bone rate of osteosarcoma, in a range of 1.7 per million. Osteo-
in which malignant mesenchymal tumor cells have the sarcoma is rare in the first decade of life (<5% of cases).
ability to produce osteoid or immature bone. There may Most patients (~60%) are between 10 and 20 years old.
200
ERRNVPHGLFRVRUJ
5  Osteosarcoma 201

involves the appendicular skeleton. Approximately 50%


TABLE 5-1 Types of Osteosarcoma and
of cases are located in the knee region. The distal femoral
Descriptive Terms Used in
metaphysis is affected 2.5 times more frequently than
Their Diagnosis
the proximal tibial metaphysis. The humerus is the third
Intramedullary Osteosarcoma most frequently involved bone in young patients and is
Conventional the site of 15% of all cases, with the majority of osteo-
Osteoblastic sarcomas developing in the proximal humeral metaphy-
Chondroblastic sis and diaphysis. The distal portion of the humerus is
Fibroblastic seldom involved. Osteosarcoma is unusual in the bones of
Malignant fibrous histiocytoma-like
Osteoblastoma-like the forearm and in the small bones of the hands and feet.
Giant cell-rich In older patients, the predilection for the appendicular
Small cell skeleton and the knee region is less clear59,94 (Figs. 5-4
Epithelioid and 5-5). In patients older than age 50 years, only 15% of
Telangiectatic
Well-differentiated (low-grade intraosseous)
the tumors occur in the knee area. In this age group, the
axial skeleton and flat bones are more frequently affected
Secondary Osteosarcoma (~40% of cases). Approximately 10% of osteosarcomas
Paget’s disease occur in the pelvis, and the ilium is the most frequently
Postradiation involved flat bone. Less than 10% of osteosarcomas occur
Bone infarct
Fibrous dysplasia in the mandible and other craniofacial bones.
Metallic implant
Chronic osteomyelitis
Clinical Symptoms
Osteosarcoma Associated with Specific Clinical Syndromes
Familial
The most common symptom is pain that has usually con-
Rothmund-Thomson syndrome tinued for several weeks to months. The pain gradually
Retinoblastoma becomes more severe and eventually is accompanied by
Multifocal swelling. The overlying skin feels warm and has promi-
Surface Osteosarcoma nent superficial vasculature. Swelling is often accompa-
Parosteal (juxtacortical) nied by some limitation of motion. Pain in the adjacent
Periosteal joint and accumulation of fluid may also be present. At
High-grade surface (de novo and dedifferentiated) the time of presentation, some patients may have weight
loss; this is usually associated with disseminated disease,
Intracortical Osteosarcoma
which is most frequently in the form of lung metastases.
Extraskeletal Osteosarcoma The serum alkaline phosphatase level is frequently
elevated in patients with osteosarcoma. It can be particu-
larly high in patients with a heavy tumor burden or in
those who have tumors that exhibit prominent osteoblas-
The sex ratio varies among the series but a male predomi- tic differentiation. Elevation of the alkaline phosphatase
nance is clear (1.3 : 1 to 1.6 : 1). The higher rate in males level after surgery indicates persistent, recurrent, or met-
is attributed to their longer period of skeletal growth. It astatic disease.
appears that osteosarcoma has a tendency to develop
earlier in females than in males, with a median age of 17
Radiographic Imaging
years compared with 18 years for males,96 but the distri-
bution of the age-related incidence and frequency from The radiographic presentation of osteosarcoma varies
the SEER Program data do not confirm this trend56 (Fig. greatly. The tumor may be completely lytic or sclerotic,
5-3). There are no major differences among races (white but usually a combination of these features enables a
versus black), but it appears that the peak incidence preoperative radiographic diagnosis of osteosarcoma in
during the second decade of life is somewhat higher in the majority of cases99 (Fig. 5-6). The mineralization of
black males than in white males or in black or white tumor matrix produces cloudy opacities that vary in size,
females (Fig. 5-3). shape, and density. These opacities can be relatively uni-
The peak incidence of osteosarcoma in adoles- formly distributed throughout the lesion or can cluster
cents corresponds to the peak period of skeletal in one area to form large, irregular sclerotic masses (Figs.
growth.64,65,71,76,90,163,174,175 The portions of the skeleton 5-7 to 5-10). Occasionally, the tumor may appear to have
with the highest growth rate are the most frequently a uniform ivory-like density with minimal or no lytic
affected. The frequency of tumor occurrence within a component (Figs. 5-11 and 5-12). The tumor exhibits
specific bone corresponds to the site of greatest growth a destructive growth pattern with a gradual transition
rate. Accordingly, the distal femoral and proximal tibial from lytic or sclerotic areas to normal bone, which makes
metaphyses, where most growth occurs during adoles- the borders of the lesion ill-defined. The process can
cence, are the most common sites for osteosarcoma. be limited to the medullary space, but in most instances
Some studies have documented that, on average, patients the cortex is also involved and often is destroyed, at
with osteosarcoma are taller than their peers in the corre- least focally. In the area of penetration, the outer limits
sponding age group and have increased levels of somato- of the cortex are indistinct, but its original outline can
medin.71,136 In adolescents, osteosarcoma predominantly usually still be traced. Usually the tumor extends into the

ERRNVPHGLFRVRUJ
202 5  Osteosarcoma

Peak
incidence
10 20
Incidence

Second
peak
50

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-1  ■  Conventional osteosarcoma. Peak age incidence and frequent sites of skeletal involvement. The most frequent sites
are indicated by solid black arrows.

soft tissue, and its mineralized shadow can be seen overly- without radiographic evidence of extension into soft
ing the area of cortical penetration (Figs. 5-13 and 5-14). tissue but is usually observed within the soft tissue com-
Tumors that extend beyond the cortex can vary in size ponent of the tumor that overlies an area of cortical
and degree of mineralization. In general, the size of the disruption. The tumor growing on the surface of bone
extracortical soft tissue component corresponds with the can elevate the periosteum and induce a periosteal reac-
size of intramedullary tumor and to the extent of cortical tion in the form of an open triangle overlying the diaphy-
destruction. The greater the extent of tumor within the seal side of the lesion. This type of reaction is known as
bone and the more extensive the cortical disruption, the Codman’s triangle (see Figs. 5-6 and 5-15). Sometimes the
larger the extracortical soft tissue component is likely to periosteal reaction can be in the form of multiple layers
be. In exceptional cases, long plugs of tumor can extend (“onion skin”), which are more typically seen in small-cell
within the medullary cavity well beyond the level of corti- tumors involving bone and in osteosarcomas that are in
cal discontinuity. The pattern of destructive growth and a diaphyseal rather than metaphyseal location.
extension into soft tissue is usually asymmetric with one Rapidly growing tumors can produce predominantly
side of the bone being more involved (see Figs. 5-6 and lytic masses with a permeative pattern of bone destruc-
5-8). Initially there is a single area of cortical penetration tion and minimal or no periosteal reaction53 (see Figs. 5-8
and expansion into soft tissue, but eventually with tumor and 5-14). This type of osteosarcoma is more difficult to
progression, it may form an extensive soft tissue mass that diagnose from radiographs and requires special care to
circumferentially involves the affected long bone (see distinguish it from other destructive lesions of bone, such
Figs. 5-7, 5-9, 5-10, 5-12, 5-14, and 5-16). as osteomyelitis. Osteosarcoma is usually suspected
The outer surface of the cortex overlying the tumor because of the presence of a destructive lesion in the
may demonstrate a prominent periosteal reaction that metaphyseal area of a long bone in an adolescent, even
can be in the form of parallel periosteal new bone, when the characteristic pattern of tumor matrix mineral-
hazy cortical irregularity and fuzziness, or both features ization is lacking. Occasionally osteosarcoma of quite
(Fig. 5-15). Occasionally the tumor may form perpen- high histologic grade can produce deceptively innocent
dicular or radiating striations (“sunburst”). This type of radiographic defects with sharp or even sclerotic margins.
periosteal reaction can be seen on the bone surface Text continued on p. 215

ERRNVPHGLFRVRUJ
5  Osteosarcoma 203

1 25

0.8 20
(cases/100,000 persons)

Age distribution (%)


Incidence rate

0.6 15

0.4 10

0.2 5

0 0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

85+
Age at diagnosis
FIGURE 5-2  ■  Epidemiology of conventional osteosarcoma. National Cancer Institute Surveillance, Epidemiology, and End Result
Program 1973-2009. Age-adjusted incidence rate and age-specific frequency, all races, both sexes, 4962 cases.

Black, male
Black, female
Incidence rate (cases/100,000 persons)

1.5 White, male


White, female

0.5

0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

85+

Age at diagnosis
FIGURE 5-3  ■  Epidemiology of conventional osteosarcoma. National Cancer Institute Surveillance, Epidemiology, and End Result
Program 1973-2009. Age-adjusted incidence rates by race and sex, 4962 cases.

ERRNVPHGLFRVRUJ
204 5  Osteosarcoma

0.5

Bones of skull and face and associated joints


Vertebral column
0.4 Rib, sternum, clavicle and associated joints
Extremities
Pelvic bones, sacrum, coccyx and associated joints
(cases/100,000 persons)

0.3
Incidence rate

0.2

0.1

0
00-19 20-49 50+
Age at diagnosis
FIGURE 5-4  ■  Epidemiology of conventional osteosarcoma. National Cancer Institute Surveillance, Epidemiology, and End Result
Program 1973-2009. Age-adjusted incidence rates by sites of skeletal involvement, all races, both sexes, 4962 cases.

120

Bones of skull and face and associated joints


Vertebral column
100 Rib, sternum, clavicle and associated joints
Extremities
Skeletal distribution by age (%)

Pelvic bones, sacrum, coccyx and associated joints


80

60

40

20

0
00-19 20-49 50+
Age at diagnosis
FIGURE 5-5  ■  Epidemiology of conventional osteosarcoma. National Cancer Institute Surveillance, Epidemiology, and End Result
Program 1973-2009. Age-adjusted frequency distribution by sites of skeletal involvement, all races, both sexes, 4962 cases.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 205

A B

C D
FIGURE 5-6  ■  Conventional intramedullary osteosarcoma of distal femoral metaphysis. A, Anteroposterior plain distal femoral ra-
diograph showing a destructive sclerotic lesion with soft tissue extension. B, Fat-saturated T2-weighted magnetic resonance image
(MRI) showing distal femoral lesion with inhomogeneous signal, cortical destruction, and extension of tumor into soft tissue. The
tumor penetrates the growth plate and extends to the epiphysis. C, Fat-saturated T1-weighted axial MRI with contrast documents the
eccentric intramedullary lesion with partial circumferential extension into soft tissue medially. D, Frontal section, after limb-salvage
procedure, shows sclerotic metaphyseal lesion with soft tissue extension medially and penetration of the growth plate.

ERRNVPHGLFRVRUJ
206 5  Osteosarcoma

A B

C D
FIGURE 5-7  ■  Conventional intramedullary osteosarcoma of proximal tibia. A, Coronal computed tomography reformation of tibia
showing intramedullary mixed-sclerotic and lytic lesion with soft tissue extension. B, Fat-saturated T2-weighted coronal magnetic
resonance image showing extensive intramedullary lesion with cortical penetration and circumferential soft tissue extension.
C, Gross photograph of sclerotic intramedullary tumor extensively involving metaphyseal proximal tibia and showing massive
circumferential extension into the soft tissue. D, Microscopic features of the same tumor, showing extensive tumor osteoid disposi-
tion and anaplastic mesenchymal cells. (D, ×100) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 207

A B

C D
FIGURE 5-8  ■  Conventional intramedullary osteosarcoma of distal femur. A, Anteroposterior plain radiograph shows an eccentric
predominantly lytic destructive process in the lateral aspect of the distal femoral metaphysis. A discreet shadow of the tumor
extending to the soft tissue is seen medially (arrows). B, Fat-saturated T2-weighted coronal magnetic resonance image showing
destructive tumor with high signal intensity involving the lateral aspects of distal femoral metaphysis with extension into epiphysis
and involvement of soft tissue medially. C, Gross photograph showing intramedullary tumor involving posterior aspects of femoral
metaphysis with extension to the epiphysis and massive involvement of retrofemoral soft tissue. D, Microscopic features of the
tumor, showing highly pleomorphic mesenchymal tumor cells and tumor osteoid deposition. (D, ×100) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
208 5  Osteosarcoma

A B

C D
FIGURE 5-9  ■  Intramedullary osteosarcoma of distal femur. A, Lateral plain radiograph showing extensive mixed lytic and sclerotic
tumor of the distal femur with circumferential soft tissue extension. B, T1-weighted sagittal magnetic resonance image (MRI) with
contrast, showing extensive intramedullary lesion with irregular patches of signal void corresponding to extensive mineralization
of tumor matrix. Note extensive circumferential involvement of paraosseous soft tissue. C, Coronal MRI showing similar features
as in B, documenting massive circumferential involvement of the paraosseous soft tissue. D, Sagittal section of the resection speci-
men, showing extensive intramedullary tumor with circumferential soft tissue extension.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 209

A C
FIGURE 5-10  ■  Conventional intramedullary osteosarcoma of the proximal tibia. A, Plain radiograph showing mixed sclerotic and
lytic lesion of the proximal tibia. B, Sagittal fat-saturated T2-weighted magnetic resonance image of the proximal tibia, showing
intramedullary tumor with high signal intensity and cortical penetration anteriorly and posteriorly. C, Gross photograph showing
sagittal image of a fleshy tumor involving the proximal end of the tibia.

ERRNVPHGLFRVRUJ
210 5  Osteosarcoma

A B

C D
FIGURE 5-11  ■  Conventional intramedullary osteosarcoma of the distal femur. A, Fat-saturated T2-weighted coronal magnetic reso-
nance image (MRI) showing intramedullary tumor with signal enhancement. B, Fat-saturated T2-weighted axial MRI showing low
signal intensity in the intramedullary tumor. C, Gross photograph showing sagittal section of a highly sclerotic intramedullary tumor
involving the distal femur. D, Closer view of the image shown in C, documenting the penetration of the growth plate.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 211

A B

C D
FIGURE 5-12  ■  Osteosarcoma of femoral diaphysis. A, Lateral plain radiograph shows highly sclerotic destructive tumor involving
femoral shaft and extending into adjacent soft tissue. B, Fat-saturated T2-weighted coronal magnetic resonance image (MRI) showing
extensive intramedullary lesion with inhomogeneous signal and circumferential extension into adjacent soft tissue. C, T1-weighted
axial MRI with contrast, documenting extensive circumferential involvement of adjacent soft tissue. D, Gross photograph showing
intramedullary tumor with massive involvement of adjacent soft tissue.

ERRNVPHGLFRVRUJ
212 5  Osteosarcoma

A B

C D

FIGURE 5-13  ■  Conventional intramedullary osteosarcoma of the distal femur. A, Anteroposterior plain radiograph showing the
destructive mixed sclerotic and lytic lesion with cortical breakthrough medially and soft tissue extension. B, Fat-saturated T2-weighted
coronal magnetic resonance image (MRI) showing intramedullary tumor with areas of signal void and patches of signal enhance-
ment penetrating the cortex circumferentially. C, T1-weighted axial MRI with contrast, showing extensive circumferential involve-
ment of soft tissue. D, Gross photograph disclosing extensive tumor of the distal femur with variegated mineralization pattern
extending to the epiphysis and soft tissue.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 213

A B

C D

FIGURE 5-14  ■  Conventional intramedullary osteosarcoma of the proximal femur. A, Anteroposterior plain radiograph showing a
destructive lytic lesion of the proximal femur. B, Fat-saturated T2-weighted axial magnetic resonance image (MRI) showing extensive
tumor with signal enhancement of the proximal femur extending to the adjacent soft tissue. C, T1-weighted axial MRI with contrast,
documenting foci of signal void in the extensive tumor involving soft tissue adjacent to proximal femoral area. D, Gross photograph
documenting extensive fleshy and mucinous tumor involving the femoral neck and intertrochanteric area with extensive involve-
ment of paraosseous tissue.

ERRNVPHGLFRVRUJ
214 5  Osteosarcoma

A B C

D E
FIGURE 5-15  ■  Conventional intramedullary osteosarcoma of the distal femur. A, Anteroposterior plain radiograph showing sclerotic
destructive tissue involving the distal femur. B, Bilateral T1-weighted coronal magnetic resonance image (MRI) showing extensive
destructive tumor mass of the distal femur with involvement of the epiphysis and adjacent soft tissue. C, Fat-saturated T2-weighted
coronal MRI showing patches of signal enhancement within the tumor mass. D, Gross photograph showing extensive, partially
necrotic, tumor mass involving distal femur and extension to epiphysis and adjacent soft tissue. Note transverse pathologic
fracture. E, Microscopic image showing extensive tumor osteoid deposition and pleomorphic mesenchymal tumor cells (×100,
hematoxylin-eosin).

ERRNVPHGLFRVRUJ
5  Osteosarcoma 215

In very rare instances, osteosarcoma can originate within More advanced lesions fill the medullary cavity and prog-
the epiphysis (epiphyseal osteosarcoma) and may be mis- ress toward the shaft and growth plate (Figs. 5-21 and
taken on radiographs for chondroblastoma or clear-cell 5-22). At this stage, lesions usually exhibit clear areas of
chondrosarcoma205 (see Figs. 5-19 and 5-20). cortical destruction, elevation of periosteum, expansion
Computed tomography and especially magnetic reso- into the soft tissue, or a combination of the features.
nance imaging (MRI) are used to evaluate the extent of These features correspond to the deposition of periosteal
disease.52,74,83,193 These types of imaging are especially new bone, which can be detected on radiographs. Further
useful in evaluating the extent of intramedullary involve- growth produces an eccentric soft tissue mass that over-
ment and extension into soft tissue. Of particular impor- lies the large cortical defect. Signs of periosteal impinge-
tance for the therapy plan (limb-sparing procedure) is the ment with associated periosteal bony reaction often can
relationship between the soft tissue extension and the be seen at the periphery of the lesion, particularly in the
neurovascular bundle. MRI is particularly useful to evalu- diaphysis. This corresponds to the Codman’s triangle
ate the extent of the tumor and plan the resection planes seen in radiographs (see Fig. 5-18). As a rule, the intra-
for surgical excisions.102 MRI reveals the heterogeneous medullary progression of the tumor is more extensive
nature of the tumor by exhibiting variable levels of signal than can be appreciated on plain radiographs. The intra-
intensity in different areas of the tumor tissue. Heavily medullary border may be irregular or may form a sharply
mineralized areas typically show no signal on the demarcated dome-shaped structure. The growth plate
T2-weighted images. On the contrary, well-vascularized serves as a substantial barrier to tumor growth and is
sarcomatous areas may exhibit various degrees of signal seldom penetrated except in more advanced lesions (see
enhancement. In general, in MRI techniques, contrast Figs. 5-13 and 5-15). Serial block sections of the epiphy-
enhancement, combined with fat saturation, discloses the seal area may document foci of perforation of the physis
heterogeneous nature of the tumor and is helpful to eval- and microscopic extension into the epiphysis, even in less
uate tumor extent and its relationship to anatomic struc- advanced cases.62,187 Rarely, the articular cartilage can be
tures in the surrounding normal tissue (Fig. 5-16; see also destroyed with extension of tumor into the joint cavity.
Figs. 5-6 to 5-15). The synovium is more frequently involved in advanced
cases by tumor growth along the bone surface rather than
Gross Findings by transarticular penetration.

The gross appearance of osteosarcoma is best described


Microscopic Findings
in its typical location (i.e., in the metaphyseal portion of
a long bone). Osteosarcoma can be composed of pre- Osteosarcoma represents one of the most heterogeneous
dominantly ossified or nonossified tissue, but usually a tumors known in human pathology. The microscopic
combination of bony and soft tissue areas is responsible features may vary considerably among different lesions
for the characteristic gross appearance of this tumor. In and in different areas of the same tumor. However, some
most instances, the tumor’s cut surface is very heteroge- common microscopic patterns can be used to subdivide
neous, with areas that vary in color, consistency, and osteosarcomas into several morphologically distinct
degree of ossification (Fig. 5-17). Highly ossified ivory- groups.
like areas are yellow-white and may be as hard as normal The two basic microscopic components of osteosar-
cortical bone (see Fig. 5-11). The less ossified areas are coma are the sarcomatous tumor cells and the extracel-
soft and less yellow in appearance. Areas with minimal or lular matrix (see Fig. 5-23). The relationship between the
no ossification are tan, fleshy, or of chondroid consis- tumor cells and the matrix is very important for diagno-
tency. They can also exhibit features of hemorrhage and sis. Evidence of direct production of osteoid matrix by
necrosis (see Figs. 5-17 and 5-18). sarcomatous tumor cells is required to classify the lesion
The large, densely ossified areas are usually the result as an osteosarcoma (Figs. 5-23 to 5-25). Osteosarcomas
of interaction between tumor osteoid and preexisting can be subdivided into three main categories on the basis
nontumor bone. In the central intramedullary portions of their predominant matrix product: osteoblastic, chondro-
of the tumor, large areas of bony condensation are pro- blastic, and fibroblastic.47-50,94,96,100,121,209 Osteosarcoma often
duced by superimposition of tumor osteoid on the pre- presents as an undifferentiated sarcoma with predomi-
existing cancellous bone of the medullary cavity. Outside nance of giant pleomorphic, round or spindle cells
the medullary cavity, within the soft tissue extension, with little intervening matrix (Figs. 5-26 to 5-28). Usually
solid bony areas are formed by the deposition of tumor a mixture of several cellular components leads to a varie-
bone between spicules of reactive nontumor bone of peri- gated pattern of matrix production. The tumor cells may
osteal origin. Overall, the tumor exhibits an invasive and have densely eosinophilic cytoplasm with eccentrically
bone-destroying pattern of growth (Figs. 5-18 to 5-20). placed nuclei and resemble osteoblasts, but they are
The borders between the tumor tissue and adjacent usually much larger than normal or even activated osteo-
structures are indistinct and irregular. This is particularly blasts. They vary considerably in size and often exhibit
evident in the areas where heavily ossified tumor tissue pronounced nuclear atypia. These cells grow in large,
merges with the adjacent cortex. cohesive sheets and may form areas with epithelioid fea-
The smaller lesions are usually eccentrically located tures.85,86,108 Focal epithelioid features are quite common
within the medullary cavity. Even relatively small lesions in conventional osteosarcoma. Tumors with predominant
that do not fill the medullary cavity have a high propen- organoid growth of cells with epithelioid features are
sity to invade the cortex and to induce periosteal reaction. Text continued on p. 229

ERRNVPHGLFRVRUJ
216 5  Osteosarcoma

B C

D
FIGURE 5-16  ■  Conventional intramedullary osteosarcoma of the proximal humerus. A, Anteroposterior radiograph showing a
destructive predominantly lytic tumor of the proximal humerus with pathologic fracture. B, Fat-saturated T2-weighted coronal mag-
netic resonance image of an extensive destructive process involving the medullary cavity of the proximal humerus with massive
soft tissue extension in the shoulder area. C, Gross photograph showing a fleshy tumor involving proximal humerus. D, Closer view
of specimen shown in C, disclosing fleshy destructive tumor with areas of necrosis and displacement of the bone axis due to patho-
logic fracture and fragmentation of proximal humerus.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 217

A B

C D
FIGURE 5-17  ■  Gross morphology of conventional osteosarcoma. A, Distal femur containing conventional osteosarcoma in metaphy-
sis. There is extensive cortical disruption and subperiosteal expansion. Tumor is confined to metaphyseal side of cartilaginous
growth plate. Hemorrhagic area represents biopsy site. B, Osteosarcoma of distal femoral metaphysis with epiphyseal extension
through incomplete, partially fused growth plate. C, Densely sclerotic osteosarcoma of tibial metaphysis extending into diaphysis.
Note that tumor is limited proximally by growth plate. D, Sclerotic osteosarcoma of distal femur with extensive involvement of the
epiphyseal end of bone. (B-D, courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
218 5  Osteosarcoma

A B

C D
FIGURE 5-18  ■  Periosteal reactions in osteosarcoma with extension into soft tissue. A, Gross photograph of tibial metaphyseal
osteosarcoma with lateral cortical breakthrough and subperiosteal mass. Note perpendicular spicules of reactive bone and angular
elevation of periosteum at lower aspect of extracortical mass. B, Specimen radiograph of slab of specimen shown in A. Cortex is
breached laterally with periosteal elevation. Resulting periosteal new bone is in form of Codman’s triangle below and horizontal
striations of reactive bone above. C, Whole-mount low power photomicrograph of extracortical extension of osteosarcoma delimited
by elevated periosteum. Note acute angle formed by stripped and elevated periosteum, under which reactive bone is being depos-
ited. D, Specimen radiograph of tumor shown in C demonstrates perpendicular striation of reactive bone traversing extracortical
tumor and oblique lines of subperiosteal bone forming Codman’s triangle below. (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 219

A B

C D
FIGURE 5-19  ■  Osteosarcoma of tibia and epiphyseal osteosarcoma. A and B, Thirteen-year-old girl fell 2 months before these antero-
posterior (AP) and lateral radiographs. Pain and swelling were interpreted as osteomyelitis, and the patient was treated with anti-
biotics without beneficial effect. Note ill defined radiodense lesion with associated periosteal reactive bone and large lucent
extraosseous tumor mass posterior to tibia. Prominent Codman’s triangles are seen in both views. C, AP radiograph of distal femur
shows totally epiphyseal, circumscribed lesion with cloudy opacities in a 15-year-old boy. D, Oblique radiograph of the epiphyseal
high-grade osteoblastic osteosarcoma in medial femoral condyle shown in C.

ERRNVPHGLFRVRUJ
220 5  Osteosarcoma

E F
FIGURE 5-20  ■  Epiphyseal osteosarcoma. A, T1-weighted coronal magnetic resonance image (MRI) showing low signal intensity
region in the epiphysis of the distal femur. B, T1-weighted sagittal MRI showing the epiphyseal lesion involving the lateral condyle.
C, T2-weighted sagittal MRI showing signal enhancement of the lesion. D, Axial computed tomogram discloses patchy signal
enhancement corresponding to tumor matrix mineralization. E, Gross photograph showing gelatinous well demarcated tumor
involving the femoral epiphysis with gritty and granular mineralization pattern. F, Microscopic image showing anaplastic tumor cells
and irregular tumor osteoid deposition. (D, ×100) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 221

A B

C D E
FIGURE 5-21  ■  Osteosarcoma: gross morphology. A, Coronal section of conventional medullary osteosarcoma of distal femoral shaft.
There is extensive tumor necrosis in both intramedullary and soft tissue components. Note extension beyond periosteum. B, Speci-
men radiograph of tumor shown in A. Extraosseous component contains cloudlike densities representing tumor bone matrix min-
eralization. C, Extensively necrotic metaphyseal osteosarcoma of distal femur delineated by zone of hemorrhagic tumor. Note growth
plate barrier blocking distal growth of tumor. D, Osteosarcoma of proximal femur with involvement of intertrochanteric region and
femoral neck. E, Osteosarcoma of the proximal femur with extensive intramedullary involvement spanning nearly the entire femur.
(A, courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
222 5  Osteosarcoma

A B

C D
FIGURE 5-22  ■  Osteosarcoma: gross morphology. A, Osteosarcoma of proximal humerus with extensive cortical disruption and
extension into soft tissue. B, Osteosarcoma of distal tibial metaphysis with circumferential extension into soft tissue and elongated
proximal intramedullary component occupying tibial shaft. C, Metaphyseal osteosarcoma of distal tibia demarcated distally by intact
cartilaginous growth plate. D, Whole-mount photomicrograph of tumor shown in C. Note that tumor has destroyed cortex and
extended into soft tissue but does not violate growth plate barrier. (D, hematoxylin-eosin.) (A and B, courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 223

A B

C D
FIGURE 5-23  ■  Osteoblastic osteosarcoma, high grade. A, High-grade osteoblastic osteosarcoma with an abundance of tumor bone
(×100). B, Higher power of A showing irregular tumor osteoid deposits and anaplastic tumor cells (×200). C, High-grade osteoblastic
osteosarcoma with abundant irregular tumor bone (×100). D, High-grade osteoblastic osteosarcoma with abundant tumor osteoid.
Note fragments of residual normal lamellar bone surrounded by the tumor. (A, C, and D, ×100; B, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
224 5  Osteosarcoma

A B

C D
FIGURE 5-24  ■  Osteoblastic osteosarcoma, high grade. A, Low power photomicrograph of sclerosing osteosarcoma fused with the
cortex on the right and obliterating marrow spaces. B, Higher magnification of A showing sheetlike osteoid fused with normal
lamellar bone on the lower left. Note the presence of highly pleomorphic atypical tumor cells. C, Low power photomicrograph
of sclerosing osteosarcoma producing abundant osteoid fused with lamellar medullary bone, obliterating marrow spaces.
D, Higher magnification of C showing abundant tumor osteoid fused with the lamellar medullary bone. Note hyperchromatic pleo-
morphic tumor cells. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 225

A B

C D
FIGURE 5-25  ■  Osteoblastic osteosarcoma, high grade: variations in osteoids and bone formation patterns. A, Irregular osteoid
bands interspaced with islands of tumor cells. B, Irregular deposits of unmineralized tumor osteoid. C, Prominent solid areas
of osteoid. D, Interconnected seams of osteoid and islands of anaplastic tumor cells. (A and C, ×100; B and D, ×200)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
226 5  Osteosarcoma

A B

C D
FIGURE 5-26  ■  Fibroblastic osteosarcoma, high grade. A and B, Low power microphotograph showing proliferations of atypical
spindle cells characteristic of fibroblastic osteosarcoma. C and D, Intermediate power views of A and B, respectively, showing con-
fluent proliferations of atypical spindle cells and irregular deposits of unmineralized matrix. Inset shows nuclear details of fibroblastic
cells with mitotic activity. (A and B, ×50; C and D, ×100; inset, ×200) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 227

A B

C D
FIGURE 5-27  ■  Conventional osteosarcoma, high grade. A, Pleomorphic sarcomatoid cells. B, Higher magnification of highly pleo-
morphic sarcomatoid cells. C, Densely packed anaplastic sarcomatoid tumor cells. D, Higher magnification of C showing nuclear
pleomorphism of tumor cells. (A and C, ×100; B, ×400; D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
228 5  Osteosarcoma

A B

C D
FIGURE 5-28  ■  Conventional osteosarcoma, high grade: morphologic variants of tumor cells. A, Densely packed epithelioid anaplastic
tumor cells. B, Sarcomatoid spindle cells with scattered multinucleated giant cells. C, Mixture of atypical spindle and pleomorphic
cells. D, Loosely arranged epithelioid tumor cells in a background of hemorrhage. (A-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 229

refered to as epithelioid osteosarcoma, described in more osteoid and they more closely resemble normal osteo-
detail below. The osteoblastic nature of the tumor cells blasts. Jaffe100 described this phenomenon as normal-
is best recognized by their close apposition to trabeculae ization in referring to the less malignant appearance of
of tumor bone or by their entrapment in lacelike osteoid tumor cells deeply embedded in osteoid. Normalization
deposits (Figs. 5-29 and 5-30). A full gamut of sarcoma- can also be used in a general sense to describe a more
tous features ranging from undifferentiated round, oval, mature appearance of osteosarcoma in highly ossified
or polygonal cells through various patterns of pleomor- solid areas (see Fig. 5-32). The trabecular pattern of
phic sarcoma and spindle-cell sarcoma may be seen. This tumor bone sometimes focally mimics the appearance of
mixture of patterns is best observed in predominantly benign reactive woven bone or that of benign osteoblastic
lytic lesions with minimal osteoid production. Those tumors (i.e., osteoid osteoma and osteoblastoma). The
lesions typically exhibit pronounced nuclear atypia and major difference is in the cells that fill the intertrabecular
brisk mitotic activity with numerous atypical mitoses (see spaces, which in osteosarcoma exhibit atypical, sarcoma-
Figs. 5-27 and 5-38) and are easily diagnosed as malig- tous features. The infiltrating pattern within such areas,
nant, but their bone-forming features can be very focal with destruction or engulfment of preexisting nontumor
and inconspicuous. Such cases may simulate pleomorphic bone, facilitates the diagnosis of a malignant process.
malignant fibrous histiocytoma of bone. As in other high- These tumors may have a deceptively benign look and
grade sarcomas, prominent stromal vascular patterns that may be mistaken for osteoblastomas of the conventional
mimic hemangiopericytoma can be found in osteosar- or aggressive type (for a more detailed description see
coma (see Fig. 5-38). At the opposite end of the spectrum Chapter 4 and the section on osteosarcoma with unique
are highly ossified, sclerosing lesions in which a full range microscopic features in this chapter).
of tumor bone production can be observed. Osteosarcomas can also exhibit cartilaginous differen-
The production of tumor bone matrix begins with the tiation. In some instances, the cartilaginous component
elaboration of extracellular fibrillar collaginous matrix can be quite extensive and dominant throughout the
(osteoid). In its early stages, it can be recognized by lesion (Figs. 5-39 and 5-40). The cartilaginous differen-
the presence of a dense, fibrillar eosinophilic substance tiation in osteosarcoma can be very heterogeneous and
deposited between small groups of cells. At this stage, represents all stages of cartilaginous matrix formation.
seams of dense eosinophilic matrix material separate and The earliest stages represent the formation of a loose
enmesh individual tumor cells. Larger areas of osteoid basophilic intercellular substance with cells lying in
produce a lacelike pattern, in which rows of tumor cells lacunar spaces. Mineralization of the matrix produces
are surrounded by a fibrillar ground substance that may bluish granular or linear deposits of calcium. Other pat-
begin to show early mineralization (Figs. 5-29 to 5-31). terns of cartilage differentiation parallel those seen in
Further advance of ossification produces wide sheets of chondrosarcoma and vary from areas similar to those seen
osteoid that separate tumor cells and show deposits of in high-grade chondrosarcoma through myxoid change
calcification (Figs. 5-32 and 5-33). More advanced min- to well-differentiated areas with a deceptively benign
eralization produces clearly recognizable trabeculae of appearance. Often a full spectrum of cartilage differentia-
woven tumor bone. The tumor bone trabeculae differ tion representing different degrees of matrix maturation
considerably in size, shape, and degree of mineralization. and cellular atypia is seen in one tumor. Even in a pre-
They are haphazardly arranged, have highly irregular dominantly chondroid tumor, at least focal direct osteoid
borders, and merge gradually with areas of less mature production by sarcomatous tumor cells is required for the
osteoid (Figs. 5-34 to 5-37). The tumor bone frequently recognition of its osteosarcomatous nature. The cartilage
is in direct contact with preexisting nontumor bone that and bone-forming areas are frequently intermingled and
represents cancellous bone trabeculae of the medul- there can be a gradual transition between them with
lary cavity, the cortex, or the reactive bone of perios- zones in which it is difficult to classify the matrix.
teal origin. The tumor bone characteristically fuses and Fibroblastic differentiation in osteosarcoma is charac-
merges with the preexisting nontumor lamellar bone and terized by the presence of a predominant spindle-cell
often forms large, ossified, solid areas (see Fig. 5-24). In component similar to that seen in fibrosarcoma. In
such instances, the nontumor bone serves as a scaffold for conventional osteosarcomas, the spindle-cell component
the tumor bone and both occasionally form large solid usually shows high-grade cytologic features (see Fig.
areas of ossified tissue as dense as the normal cortex. 5-26). The production of tumor bone by fibroblast-like
Usually, however, there is a clearcut separation of the cells discloses the bone-forming nature of these predomi-
immature, woven tumor bone where it attaches to the nantly spindle-cell lesions and helps differentiate them
lamellar, nontumor bone. In contradistinction, a gradual from other spindle-cell neoplasms of bone. Occasionally,
conversion of woven bone into mature lamellar bone these tumors resemble pure fibrosarcomas that contain
can be seen in reactive lesions, such as fracture callus. inconspicuous foci of homogenous eosinophilic material
The advancing tumor can partially or completely destroy (see Fig. 5-26). These foci cannot be identified with cer-
the original bone, replacing it with tumor bone or with tainty as early osteoid when mineralization is not present.
nonossified tumor tissue. The tumor osteoid can form Such tumors, if found in an appropriate clinical setting,
well-defined islands that mimic trabecular bone or can are still best classified as osteosarcoma. Furthermore,
be deposited in irregular sheets that contain entrapped these almost purely fibroblastic lesions occasionally can
tumor cells (see Fig. 5-32). Interestingly, the tumor produce very heavily ossified metastases. Low-grade
cells entrapped by the osteoid are usually smaller and fibroblastic osteosarcomas (intramedullary and surface)
less pleomorphic than the tumor cells lying outside the Text continued on p. 242

ERRNVPHGLFRVRUJ
230 5  Osteosarcoma

A B

C D
FIGURE 5-29  ■  Conventional osteosarcoma, high grade: variants of osteoids and primitive bone formation by tumor cells. A, Discrete
lacelike pattern of osteoid deposition. B, Higher magnification of A showing lacelike pattern of osteoid deposition and courts of
anaplastic tumor cells. C, Lacelike pattern of osteoid formation by tumor cells. D, Higher magnification of C showing interconnected
seams of osteoid encircling tumor cells. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 231

A B

C D
FIGURE 5-30  ■  Conventional osteosarcoma, high grade: variants of osteoid and primitive bone formation by tumor cells. A, Nearly
solid sheetlike osteoid encircling small clusters of tumor cells with lacuna spaces. B, Delicate seams of osteoid and clusters of
anaplastic tumor cells. C and D, Variants of seamlike osteoid deposition encircling small clusters of tumor cells and occasionally
individual tumor cells. (A, ×100; B-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
232 5  Osteosarcoma

A B

C D
FIGURE 5-31  ■  Conventional osteosarcoma, high grade. A, Loosely arranged anaplastic tumor cells with epithelioid features. B, Higher
magnification of A showing loosely arranged epithelioid tumor cells with oval densely eosinophilic cytoplasm. C, Early osteoid
deposition by tumor cells. D, Higher magnification of C showing early seams of unmineralized osteoid among tumor cells. (A and
C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 233

A B

C D
FIGURE 5-32  ■  Conventional osteosarcoma, high grade: patterns of osteoid deposition. A, Coarse deposits of osteoid separating
groups of malignant cells. B, Interconnected coarse deposits of osteoid encircling and separating clusters of tumor cells. C, Clearly
defined interconnected osteoid separating clusters of tumor cells. D, Well developed interconnected irregular trabecular pattern
produced by tumor osteoid. (A and B, ×100; C and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
234 5  Osteosarcoma

A B

C D
FIGURE 5-33  ■  Conventional osteosarcoma, high grade: patterns of osteoid deposition and mineralization. A, Irregular separated
trabecular pattern of osteoid deposition. B, Prominent trabecular pattern of well mineralized osteoid. C, Profuse matrix
deposition in osteosarcoma. D, Coarse trabecular pattern of well mineralized osteoid. (A and B, ×100; C and D, ×200)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 235

A B

C D
FIGURE 5-34  ■  Conventional osteosarcoma, high grade: patterns of osteoid deposition and mineralization. A, Coarse interconnected
trabecular pattern of well mineralized osteoid. B, Higher magnification of A showing well mineralized coarse osteoid. Note entrap-
ment of individual tumor cells residing within lacunar spaces of osteoid. C, Conspicuous, well mineralized, osteoid deposition pattern
separating and encircling larger clusters of loosely arranged tumor cells. D, Higher magnification of C showing well mineralized
osteoid and loosely arranged anaplastic tumor cells. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
236 5  Osteosarcoma

A B

C D
FIGURE 5-35  ■  Conventional osteosarcoma, high grade: patterns of osteoid deposition and mineralization. A, Sheetlike osteoid with
early mineralization. B, Higher magnification of A showing a less developed area of osteoid deposition with sparse seams of matrix
deposition. C, Coarser deposition of osteoid with early mineralization. D, Coarse sheetlike deposition of osteoid. (A and D, ×100;
B and C, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 237

A B

C D
FIGURE 5-36  ■  Conventional osteosarcoma, high grade: patterns of osteoid deposition and mineralization and epithelioid features
of tumor cells. A, Large irregular areas of osteoid with entrapped tumor cells. B, Higher power of A showing irregular solid areas
of osteoid with entrapped tumor cells. Note well demarcated dense eosinophilic cytoplasm of tumor cells. C, Irregular solid
areas of osteoid separating large clusters of epithelioid tumor cells. Note dense eosinophilic cytoplasm responsible for epithelioid
appearance of tumor cells. D, Higher power of C showing irregular areas of osteoid with entrapped tumor cells. (A and C, ×100;
B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
238 5  Osteosarcoma

A B

C D
FIGURE 5-37  ■  Conventional osteosarcoma, high grade. A and B, Infiltrative growth pattern of high-grade osteosarcoma permeating
intratrabecular marrow spaces. C, Higher power of B showing permeation of intratrabecular marrow spaces by an osteosarcoma.
D, Variegated osteoid deposition and mineralization patterns in osteosarcoma permeating marrow spaces. (A and B, ×50; C and
D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 239

A B

C D
FIGURE 5-38  ■  Conventional osteosarcoma, high grade with hemangiopericytoma-like pattern. A, Hemangiopericytoma-like pattern
of tumor growth bordering endothelial-lined spaces. B, Higher magnification of A showing prominent vascular spaces
and sheetlike osteoid deposition. C, Hemangiopericytoma-like pattern with prominent vasculature and sheetlike osteoid deposition.
D, Higher magnification of C showing prominent engorged vessels and osteoid deposition by tumor cells. (A and C, ×100; B and
D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
240 5  Osteosarcoma

A B

C D
FIGURE 5-39  ■  Chondroblastic osteosarcoma, high grade. A, Mixed chondroid and osteoid matrix in high-grade osteosarcoma.
B, Higher magnification of A showing gradual transition between hypercellular sarcomatoid component and areas of more solid
matrix deposition. C, Islands of cartilaginous differentiation. D, Higher power of C showing a gradual transition between solid areas
of anaplastic tumor cells and the cartilage matrix. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 241

A B

C D
FIGURE 5-40  ■  Chondroblastic osteosarcoma, high grade. A and B, Low and intermediate power views of osteoid deposition by
anaplastic tumor cells. C and D, Low and intermediate power views of cartilaginous differentiation in the same tumor as shown in
A and B. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
242 5  Osteosarcoma

represent distinct types of osteosarcoma, and these are and immunohistochemical markers of greater specificity,
described separately. Occasionally a highly anaplastic the diagnosis of small cell osteosarcoma is made with
tumor, typically located in the metaphysis of a long decreasing frequency. In general, our diagnostic philoso-
bone, shows no osteoid production but is otherwise phy is to diagnose tumors of bone that are composed
cytologically similar to an osteosarcoma. These lesions, of small cells that mimic those of Ewing’s sarcoma and
previously classified as undifferentiated (osteolytic) osteo- show focal tumor osteoid production as well as being
sarcoma, are now designated as high-grade, pleomorphic, characterized, molecularly, by the presence of the gene
malignant fibrous histiocytoma of bone. The distinction fusions involving EWSR1 reported in Ewing’s sarcoma
is an academic one because both tumors are treated with as variants of this tumor. This is not to deny the pres-
similar chemotherapeutic regimens. ence of small cell osteosarcoma composed of small cells
By convention, a four-tier system has been used with non-Ewing’s sarcoma morphology that are negative
for histologic grading of conventional osteosar- for the gene fusions involving EWSR1. Such tumors, in
coma.137,141,207,208 However, the majority of grade 1 osteo- our opinion, represent true small cell variants of osteo-
sarcomas seem to belong in the categories of distinct sarcoma. The diagnostic controversy related to the clas-
types of osteosarcoma described as separate entities. sification of small cell osteosarcoma is, however, further
Moreover, separation of conventional osteosarcoma into complicated by the recent report of a novel EWSR1-
four grades is based on subjective evaluation of nuclear CREB3L1 fusion transcript in small cell osteosarcoma.51
atypia and seems to have minimal clinical value. The This unique chimeric gene has not been reported in
current trend is to separate conventional osteosarcoma Ewing’s sarcoma or in any other tumors at the time of
into two major categories: low and high grade. This sim- this writing. The fusion of CREB3L1 with FUS, creat-
plified grading system seems to be more reproducible and ing the FUS-CREB3L1 fusion transcript, however has
clinically valid. been reported in low-grade fibromyxoid sarcomas of soft
tissue. Therefore, it may well be that there is a subset
Osteosarcoma with Unique Microscopic Features.  of small cell osteosarcomas with molecular aberrations
The vast majority of osteosarcomas have obvious features distinct from Ewing’s sarcoma. More detailed discussion
of atypia and osteoblastic differentiation, which make of this problem can be found in Chapters 3 and 11.
them readily recognizable as malignant bone-forming Epithelioid osteosarcoma is a rare variant of osteosar-
tumors. In rare instances, these tumors may present with coma that is composed of epithelioid cells frequently
microscopic features that overlap with other malignant growing in cords and forming well defined nests (Fig.
or even benign conditions. In this section, we describe 5-42).38,45,105,167,222 The cells may have polarized nuclei and
microscopic variants of conventional intramedullary dense eosinophilic cytoplasm. The growth pattern can
osteosarcoma that mimic small cell malignancies, includ- be trabecular, nested, or organoid, and rosette-like struc-
ing Ewing’s sarcoma or lymphoma, metastatic carcinoma, tures can be present focally.38,45,105,151 Solid areas exhibiting
myxoid sarcoma, and giant cell tumor of bone, as well as such a growth pattern can dominate the tumor; evidence
benign bone-forming tumors such as osteoblastoma and of tumor osteoid and bone deposition may be focal and
chondroblastoma. may require extensive sampling to be documented. Some
Small cell osteosarcoma is a microscopic variant char­ of these tumors were reported to be focally or even dif-
acterized by a proliferation of undifferentiated small fusely strongly positive for cytokeratins.86,108,152 Overall,
round cells that contain minimal amounts of cyto- the microscopic features and positivity for epithelial
plasm.6,26,29,54,132,146,168,186,190 These tumors, in which the markers mimic epithelial malignancy and may cause the
bone and osteoid matrix production is usually incon- misdiagnosis of metastatic carcinoma in bone. Meticu-
spicuous, can be confused with other small cell malig- lous correlation with radiographic presentation that, in
nancies such as lymphoma and Ewing’s sarcoma (Fig. the vast majority of cases, shows typical features con-
5-41). Typically, unequivocal tumor osteoid formation sistent with a malignant bone-forming tumor and the
can be identified focally, which clearly defines the entity fact that these tumors usually affect patients of younger
as a malignant bone-forming tumor. Whether small cell age, in whom metastatic carcinoma is exceedingly rare,
osteosarcoma represents a distinct entity, with its own are helpful in making the correct diagnosis. Microscopic
characteristic clinical and radiologic features, or is merely identification of direct tumor bone production is the most
a histologic variant of conventional medullary osteosar- reliable discriminatory feature that aids in the diagnosis
coma continues to be the subject of debate. On the other in this rare variant of osteosarcoma.
hand, the presence of translocation between chromo- Myxoid osteosarcoma is a rare variant of osteosarcoma
somes 11 and 22, which characterize Ewing’s sarcoma and that mimics myxoid sarcoma or the myxoid variant of
related tumors, has been documented in some of these malignant fibrous histiocytoma (Fig. 5-43). We recently
cases.149 More modern data indicates that a significant encountered several examples of this form of osteosar-
proportion of small cell osteosarcomas have gene fusions coma; all of them originated in the maxillary bone. The
involving EWSR1 similar to those of Ewing’s sarcoma.127 mesenchymal component of the tumor is composed of
In addition, these tumors may have microscopic features loosely arranged, undifferentiated short plump spindle
overlapping with those of Ewing’s sarcoma. Therefore cells growing in abundant myxoid stroma. Large areas
some of the small cell osteosarcomas diagnosed in the of the tumor are composed of paucicellular mucinous
past on the basis of purely morphologic criteria may actu- lakes. Nuclear atypia and pleomorphism are minimal to
ally represent variants of Ewing’s sarcoma. In fact, with moderate, and the tumor may have a deceptively benign
the availability of cytogenetic studies, molecular probes, appearance. There are also ill-defined areas of increased

ERRNVPHGLFRVRUJ
5  Osteosarcoma 243

A B

C D
FIGURE 5-41  ■  Small cell variant of osteosarcoma. A, Monotonous proliferation of small tumor cells. B, Higher magnification of
A showing undifferentiated tumor cells with oval cytoplasm and somewhat eccentrically located nuclei. C, Interconnected trabeculae
of tumor osteoid engulfing small clusters of tumor cells in the same tumor as shown in A and B. D, Coarse deposits of osteoid in
the same tumor. (A, C, and D, ×200; B, ×400) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
244 5  Osteosarcoma

A B

C D
FIGURE 5-42  ■  Epithelioid osteosarcoma. A, Trabecular and organoid growth pattern of epithelioid tumor cells interspersed by well
developed interconnected trabecular pattern of tumor bone. B-D, Higher magnification showing epithelioid tumor cells growing in
a nested and trabecular pattern that mimic carcinoma. (A, x100; B-D, x200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 245

A B

C D

FIGURE 5-43  ■  Myxoid osteosarcoma. A, Low power photomicrograph shows tumor cells dispersed in myxoid stroma and focal
deposition of tumor osteoid. B, Higher power photomicrograph of A showing interface between myxoid tumor and osteoid
deposits. C, More cellular area of the tumor composed of undifferentiated tumor cells dispersed in myxoid stroma (×100).
D, Low power photomicrograph showing lakes of mucin and overall blunt appearance of the tumor. Inset shows higher magnifica-
tion of D, depicting mucin lakes and paucity of tumor cells. (A and D, ×50; B, C, and inset, ×100) (A-D and inset,
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
246 5  Osteosarcoma

cellularity. Evidence of tumor bone production is difficult invariably challenging, and a generous sampling of the
to appreciate and requires extensive sampling. Correla- tumor using open biopsy is often required to disclose the
tion with radiographic imaging discloses the destructive malignant nature of the process.
aggressive growth pattern of the lesion and may alert the Chondroblastoma-like osteosarcoma is most likely the
pathologist to radiographic evidence of the bone-forming rarest of the variants of osteosarcoma described in this
nature of the tumor. section and only a handful of single case reports are avail-
Giant cell-rich osteosarcoma accounts for approximately able in the literature.7,177 The overall microscopic features
3% of all cases of osteosarcoma and most of them appear of this lesion mimic that of chondroblastoma. The irreg-
to arise in the appendicular skeleton.24,73,145,176 The char- ular matrix deposition areas resemble those frequently
acteristic feature is the presence of large numbers of seen in chondroblastoma and the cellular composition
osteoclast-like giant cells distributed among sarcomatous with mononuclear cells having eccentric nuclei and well
tumor cells (Fig. 5-44).24,73,145,148,211 The stromal cells of defined eosinophilic cytoplasm admixed with scattered
such tumors may have a deceptively benign appearance osteoclast-like giant cells are responsible for frequent
and the lesion may be mistaken for a giant cell tumor. confusion of this variant of osteosarcoma with a benign
Similar to other variants of osteosarcoma described in chondroblastoma on initial biopsies (Figs. 5-46 and 5-47).
this section the bone-forming nature of giant cell-rich The presence of obvious atypia and infiltrating growth
osteosarcoma may require extensive sampling, correla- pattern of the tumor differentiate this lesion from a chon-
tion with radiographic features, or both. Highly atypical droblastoma. In addition, virtually all of the reported
pleomorphic giant cells are a constant feature in a large chondroblastoma-like osteosarcomas show an ill defined
proportion of conventional osteosarcoma. Tumors con- aggressive growth pattern radiographically. Therefore, as
taining such cells are not designated as giant cell-rich in virtually every instance of diagnostic difficulties in
osteosarcoma. The term giant cell-rich osteosarcoma is skeletal pathology, strict correlation with radiographic
reserved only for those tumors that contain a prominent imaging data is the best aid for correct diagnosis.135
population of benign appearing multinucleated osteo-
clastic giant cells and in which there is overall mimicry
Pathologic Assessment of Preoperative
of the giant cell tumor of bone. The features helpful in
Chemotherapy Effect
differentiating this variant of osteosarcoma from giant
cell tumor consist of the presence of malignant stromal The clinical and radiographic data, as well as postopera-
cells as well as tumor bone production. tive microscopic examination, indicate that current mul-
Osteoblastoma-like osteosarcoma accounts for less than tidrug chemotherapeutic regimens result in substantial
1% of all osteosarcomas; two small series of this entity reduction of tumor mass in at least 50% of patients with
were described from the Rizzoli Institute and the Mayo osteosarcoma. In addition, in some of these patients,
Clinic.25,27 Most of the reports represent single case complete or nearly complete (90%) necrosis of the
communications.1,30,89,113,194 The difficulty in interpreting tumor can be accomplished.5,8,19,48,77,96,123,162 The latter is
published results is related, in part, to the fact that associated with substantially better prognosis (i.e., longer
the diagnostic philosophy to classify the lesion as relapse-free time and survival rates) than in tumors that
osteoblastoma-like osteosarcoma differs among various respond less favorably to chemotherapy. These observa-
schools of pathology and some of the cases in these tions provided the foundation for the pathologic assess-
reports may overlap with our concept of aggressive osteo- ment of the effect of therapy in postoperative specimens.
blastoma. We reserve the term osteoblastoma-like osteosar- Pathologic assessment of chemotherapy effect is now
coma for a tumor with overall trabecular osteoid pattern universally accepted as an integral element of the multi-
that mimics that of osteoblastoma but is distinct from an modal approach (Table 5-2).5,8,9,21,77,162,165,166 Moreover,
aggressive osteoblastoma, characterized by solid prolif- the degree of response (necrosis) is used as a factor in
eration of epithelioid osteoblasts (Fig. 5-45). A more modifying the subsequent (postoperative) treatment pro-
detailed description of this problem is included in tocol. Spontaneous necrosis, however, is a common event
Chapter 4. In addition, the stromal composition with in a variety of malignant neoplasms, including osteosar-
scattered giant cells and prominent vasculature further coma.29,212 Therefore the presence of necrosis may not
mimics that of osteoblastoma. The tumor, however, has
an infiltrating pattern of the host bone and there are areas
of more solid and irregular deposits of osteoid with obvi-
ously atypical osteoblastic cells. The microscopic evi- TABLE 5-2 Histologic Grading of Chemotherapy
dence of an aggressive growth pattern is further supported Effect in Osteosarcoma
by poorly defined infiltrating borders and frequent corti-
cal destruction documented radiographically. The domi- Grade Tumor Response
nance of an osteoblastoma-like pattern creates diagnostic
1 None or minimal
difficulties; a significant proportion of the reported cases 2 Extensive necrosis with more than 10% of viable
are initially diagnosed as benign lesions. This further tumor
amplifies the need for strict correlations between micro- 3 Extensive necrosis with scattered foci of viable
scopic and radiographic presentations of the lesion that tumor (<10%)
4 Complete necrosis
can alert the pathologist to the aggressive growth pattern
inconsistent with the diagnosis of a benign osteoblastic Modified from Huvos A: Bone tumors: diagnosis, treatment
lesion. Small core needle biopsies of these lesions are and prognosis, ed 2, Philadelphia, 1991, WB Saunders.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 247

A B

C D
FIGURE 5-44  ■  Giant cell-rich osteosarcoma. A and B, Prominent population of osteoclast-like multinucleated giant cells. Note back-
ground anaplastic tumor cells. C, Higher magnification of B documenting the microscopic features of numerous osteoclastic multi-
nucleated giant cells. Note highly atypical anaplastic tumor cells in the background. D, Evidence of tumor osteoid production in
another area of the same tumor. (A and B, ×50; C and D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
248 5  Osteosarcoma

A B

C D
FIGURE 5-45  ■  Osteoblastoma-like osteosarcoma. A, Low power photomicrograph showing interconnected coarse trabeculae of
tumor osteoid and prominent vascular stroma resembling osteoblastoma. B, Higher magnification of A showing interconnected
trabeculae of tumor osteoid. Note prominent dilated vessels in the stroma. C, Tumor osteoid deposits and vascularized stromal
tissue with osteoblastic cells growing in the intertrabecular spaces. Note scattered multinucleated giant cells. D, Low power photo-
micrograph showing extensive tumor osteoid deposits comprised of interconnected trabecular pattern and solid areas of osteoid
deposition. Inset shows higher magnification of solid areas of osteoid with numerous atypical osteoblastic cells. Note the overall
resemblance to osteoblastoma; the distinguishing microscopic feature is the presence of solid areas of osteoid with highly atypical
osteoblastic cells. (A and D, ×50; B and C, ×100; inset, x200) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 249

A B

C D

FIGURE 5-46  ■  Chondroblastoma-like osteosarcoma. A, Anteroposterior radiograph showing destructive lesion in the periacetabular
area. B, T1-weighted magnetic resonance image of tumor shown in A reveals heterogeneous signal enhancement in the
tumor, infiltrating medullary cavity of the periacetabular area. C, Low power magnification with matrix deposition resembling
chondroblastoma. D, Irregular areas of matrix deposition and more cellular areas of tumor with solid growth pattern. (C, ×100;
D, ×200) (C-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
250 5  Osteosarcoma

A B

C D
FIGURE 5-47  ■  Chondroblastoma-like osteosarcoma. A and B, Low power photomicrographs showing matrix deposition and the
overall cytoarchitectural features resembling chondroblastoma. Insets show higher magnification of matrix deposits. C and D, Higher
power photomicrographs of the tumor shown in A and B depicting solid growth pattern with the cellular compositions comprising
mononuclear and scattered multinucleated giant cells with distinct eosinophilic cytoplasm. Note that the overall pattern of the matrix
deposition and cellular components resemble osteoblastoma. The distinguishing features represent an aggressive growth pattern
documented radiographically and the pronounced atypia throughout the lesion. (A and B, ×100; B, C, and inset, ×200) (A-D and
insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 251

necessarily reflect the effect of treatment. It has been mens. In summary, MRI, possibly combined with PET,
estimated that up to 60% of necrosis can be spontaneous may have the potential to provide estimates of residual
and may not necessarily reflect a treatment effect, but viable tumor before surgery, although its true clinical
subtotal (90%) or complete necrosis almost never occurs value continues to be investigated. The most accurate
spontaneously in osteosarcoma and can be attributed to assessment of therapeutic effect is still based on patho-
treatment. logic mapping of resection specimens.
In this section, we discuss some of the technical aspects
of the pathologic evaluation of a therapeutic effect
Special Techniques
according to the approach used at the University of
Texas MD Anderson Cancer Center. Theoretically, the Ultrastructurally, osteosarcoma is composed of pleomor-
accurate assessment of the extent of necrosis requires phic, undifferentiated mesenchymal cells (Fig. 5-52).
complete sampling of the entire tumor. However, this Features of osteoblastic differentiation consist of eccen-
approach is impractical and in many cases could require tric nuclei, abundant and frequently dilated rough
examination of several hundred histologic sections. For endoplasmic reticulum, and prominent Golgi centers.
practical purposes, complete sampling (the so-called his- However, these features are nonspecific and of no diag-
tologic mapping) of the central slice of the tumor provides nostic value. Large amounts of intracytoplasmic lipid
a reasonably good and clinically valid assessment of material may be present. Foci of cartilaginous differentia-
the effect of therapy. The postoperative specimen is tion with prominent glycogen and deposition of loose
bivalved along the central plane of tumor growth. In extracellular fibrillar matrix may be seen. Two types of
smaller specimens, such as those shown in Figure 5-48, multinucleated giant cells may be present. One repre-
a whole-mount embedding technique can be used. In the sents a large undifferentiated sarcoma cell, and the other
majority of cases, the central slice is further subdivided type has features of a benign osteoclast with prominent
into smaller samples suitable for embedding in conven- mitochondria and cytoplasmic projections.67,104,159,185 The
tional cassettes (Fig. 5-49). The position of each sample extracellular matrix in osteosarcoma has features of dis-
can be recorded on the gross photograph, specimen organized, incomplete ossification. It is composed of col-
radiograph, or sketch of the tumor. lagen fibrils with deposition of calcium hydroxyapatite
Tumor necrosis is characterized by the death of sarco- crystals. The process of tumor matrix ossification takes
matous cells with pyknosis and fragmentation of their place along or among collagen fibrils and in close associa-
nuclei or by complete disappearance of tumor tissue. tion with the so-called matrix vesicles. The crystals grow
The prenecrotic changes attributed to therapy consist of in size, coalesce, and form irregular solid aggregates216
the development of bizarre nuclei, homogenization of the (Fig. 5-53). Scanning electron microscopic studies reveal
chromatin structure, and vacuolization of the cytoplasm. that the calcium deposits form globules of calcification
The complete elimination of sarcomatous cells within that grow in size by the deposition on their surfaces of
tumor osteoid or residual host bone is often seen. In such smaller globular deposits that are 0.1 to 0.3 µm.184
areas, the sarcomatous tissue is replaced by a hyalinized Immunohistochemistry continues to be of marginal
vascular stroma (Fig. 5-50). Cystic cavities filled with help in the diagnosis of osteosarcoma and is predomi-
mucoid material are remnants of more solid sarcomatous nantly used to rule out other neoplasms. Osteosarcomas
areas with minimal or no tumor bone formation. express a full spectrum of osteoblastic lineage biomark-
For research purposes, planimetric techniques are used ers, including bone morphogenetic proteins, osteocalcin,
to calculate the percentage of necrosis and the amount of and osteopontin, as well as master regulatory proteins
residual viable tumor. Figure 5-48 shows an example of involved in skeletal development such as Osterix and
the assessment of treatment effect from a planimetric Runx2. 2,3,23,32,36,37,81,85,106,117,126,147,183,188,192,198,202,213,217
reconstruction of a whole-mount preparation. This labo- Although these proteins are involved in the development
rious approach, which requires special equipment that is of osteoblastic lineage cells, their role in differential diag-
not available in standard pathologic laboratories, is not nosis of osteosarcoma is hampered by the fact that they are
absolutely necessary to evaluate the therapeutic effect on also expressed in other human tumors, playing a role in
routine cases. Visual assessment of individual sections and organogenesis of their respective tissue lineages. In addi-
rough estimates of the extent of necrosis are sufficient tion, they can be upregulated in many solid tumors as a
and clinically valid. The refinement of imaging tech- part of the so-called epithelial to mesenchymal transition
niques, especially of sequential postcontrast MRI, may phenomenon. Therefore, they can be used to evaluate
permit preoperative imaging assessment of a therapeutic the osteoblastic nature of the cells but can not absolutely
effect. This technique is based on the identification identify malignant cells in question as osteosarcoma.
of signal enhancement by viable tumor after contrast Osteosarcoma cells are consistently and strongly positive
injection versus signal void in areas of necrosis. Various for vimentin. Strong positivity for S-100 protein is typi-
combinations of contrast enhanced MRI techniques cally seen in the areas that exhibit cartilaginous differen-
have been applied to evaluate tumor necrosis after che- tiation, but it can also be present focally in osteoblasts
motherapy.60,80,82,170,206 More recently, a combination of of undifferentiated sarcomatous areas. Focal positivity
18
F-fluorodeoxyglucose positron emission tomography for epithelial markers (keratins and epithelial membrane
(FDG PET) and MRI was used to formulate a prediction antigen) and muscle markers (smooth muscle actin and
model of chemotherapy response in osteosarcoma.41 desmin) have been reported.42,43,54 However, intense and
Figure 5-51 shows examples of MRI compared with con- diffuse positivity for these markers is practically never
ventional pathologic evaluation of postoperative speci- Text continued on p. 258

ERRNVPHGLFRVRUJ
252 5  Osteosarcoma

A B

C D
FIGURE 5-48  ■  Preparation of postoperative resection specimen and assessment of therapeutic effect in conventional osteosarcoma
on basis of whole-mount embedding and planimetry. A, Specimen radiograph. B, Gross photograph. C, Whole-mount histologic
section of proximal humerus. D, Extent of tumor necrosis and amount of residual viable tumor (blue) based on planimetric evalu-
ation of specimen shown in B. Amount of residual viable tumor (>10%) is considered as an unfavorable (grade 2) response. (Courtesy
Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 253

A B

C D
FIGURE 5-49  ■  Assessment of therapeutic effect in conventional osteosarcoma on basis of conventional embedding. A, Specimen
radiograph of osteosarcoma of distal femur. B, Gross photograph of central tumor slice after sectioning to create smaller fragments
suitable for embedding in conventional cassettes. C, Specimen radiograph after central tumor slice was sectioned. D, Graphic
reconstruction of specimen shows residual viable tumor (blue areas). (Courtesy Dr. K. Raymond, Houston.)

ERRNVPHGLFRVRUJ
254 5  Osteosarcoma

A B

C D
FIGURE 5-50  ■  Chemotherapeutic effect in osteosarcoma. A-C, Replacement of sarcomatous with amorphous stromal material among
tumor osteoid. D, Necrotic tumor infiltrating marrow spaces. Note occasional residual tumor cells with pyknotic nuclei. (A-D, ×100)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 255

A B

C D
FIGURE 5-51  ■  Magnetic resonance imaging of osteosarcoma of proximal tibia for evaluation of therapeutic effect in comparison
with histologic mapping. A, T1-weighted sagittal magnetic resonance image (MRI) of proximal tibia reveals marrow replacing tumor
with epiphyseal extension. B, Sagittal spin-echo T2-weighted MRI at slightly different plane reveals abnormal high signal intensity
within tumor replacing medullary cavity. C, T2-weighted sagittal MRI with contrast reveals focal areas of high signal intensity that
were presumed to contain residual viable neoplasm. D, Histologic mapping of extent of tumor necrosis. Blue areas represent residual
viable tumor. Note that overall there is good correlation between pathologic and MRI assessment of therapeutic effect. (A-C, courtesy
Dr. D.G. Varna, Houston.)

ERRNVPHGLFRVRUJ
256 5  Osteosarcoma

B
FIGURE 5-52  ■  Conventional osteosarcoma: ultrastructural features. A and B, Undifferentiated pleomorphic sarcomatous cells are
the most frequent cellular component seen in majority of high-grade conventional osteosarcoma. (A, ×4500; B, ×6000.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 257

FIGURE 5-53  ■  Conventional osteosarcoma: ultrastructural features. A and B, Ultrastructural features of matrix mineralization with
deposition of calcium hydroxyapatite crystals along collagen fibers. (A, ×3500; B, ×9000; inset, ×14,000.)

ERRNVPHGLFRVRUJ
258 5  Osteosarcoma

seen in osteosarcoma. If present, other tumors should be binding function was frequently found in metastatic
seriously considered. lesions.115 This finding indicates that in metastatic
DNA ploidy studies have documented that a majority osteosarcoma, p53 can be functionally inactive because of
of conventional osteosarcomas have prominent, often high levels of MDM2 products. Alterations of other
multiple, aneuploid clones.13,16,31,88,91,109,110,130,131,138 The genes such as MYC seem to be infrequent in osteosar-
degree of aneuploidy has some prognostic implica- coma.116 A recent study from the Rizzoli Orthopedic
tions.109,110,130 The relapse-free time and survival rates are Institute suggests that an upregulation of the multidrug
better in a tumor with a prominent, near-diploid cell resistance gene (MDRI) coding for P-glycoprotein
population (as a sole clone or dominant but coexistent (a transmembrane adenosine triphosphate–dependent
with more aneuploid cell clones).125 The conversion from efflux pump) in osteosarcoma is associated with a
aneuploidy to near diploidy after chemotherapy corre- significantly increased risk of adverse events.11 Increased
lates with a good histologic response (complete or almost levels of P-glycoprotein may confer chemotherapy resis-
complete necrosis).11,22 tance and are associated with significantly lower survival
On the chromosomal level, most conventional osteo- rates of patients with osteosarcoma. These original
sarcomas show multiple rearrangements, multiplications, reports concerning the molecular prognostic factors in
and the presence of marker chromosomes.34 Chromo- osteosarcoma were followed by numerous publications
somal complements in osteosarcoma can be divided into reporting the relationship between the expression
two major groups: grossly aneuploid and near diploid. pattern of numerous genes and their encoded proteins,
Grossly aneuploid complements show multiple extra including VEGF, CXCL12, c-kit, NDRG1, URG4, EBF2,
chromosomes, chromosomal rearrangements, and marker RUNX2, ERBB2, WWOX, KAI1, Ezrin (EZR), and
chromosomes. This complement corresponds to highly cancer-testis antigen as potential prognostic factors
aneuploid cell populations identified by flow cytometry in osteosarcoma.10-12,14,42,63,92,93,106,114,118,158,161,173,218,220,221,224
or image analysis. The other complement is character- These reports provide interesting clues on the biology of
ized by multiple chromosomal deletions, rearrangements, osteosarcoma, but their role in the diagnosis and manage-
and a lesser degree of chromosomal multiplication. This ment of this disease is uncertain.
pattern corresponds to the so-called near-diploid or even The past decade of research on osteosarcoma
occasionally hypodiploid cell populations documented by is highlighted by the applications of various high
flow cytometry. Several chromosomal alterations seem to throughput genomic mapping platforms, includ-
be distributed in a nonrandom fashion. Structural rear- ing genome sequencing, to investigate the myriad of
rangements cluster to 1p, 1q, 3p, 3q, 11p, 17p, and 22q. genetic and epigenetic alterations observed in these
The following bands were documented to be most fre- malignancies.40,68,69,103,111,120,124,156
quently altered: 1q11, 1q21, 1q42, and 7q11. Loss of Comparative genomic hybridization (CGH) microar-
chromosomes 3, 10, 12, and 15 are the most frequent ray analysis confirmed earlier-known large amplicons at
numerical abnormalities; the most frequent complex 12q11-115, 8q24, 6p12-p13, and 17p11-p13 and identi-
recurrent rearrangements involve chromosomes 8, 17, fied novel amplicons at 14q11, 17q25, and 22q11-q13.4
and 20. These data implicate the TOM1L2 and CYP27B1 genes
On the molecular level, the retinoblastoma (RB) gene as novel targets for 17p and 12q amplicons. The inte-
seems to play the most important role in the pathogenesis grated use of high resolution DNA methylation, genomic
of osteosarcoma (see indices for osteosarcoma associated imbalance, and gene expression profiling platforms
with retinoblastoma).18,44,57,178-180 Several studies have reveals a complex genetic and epigenetic deregulation of
shown that the patterns of alterations (mutations) of the the MYC network in the cell lines of osteosarcoma.171,204
RB gene in osteosarcoma are similar to those seen in Similar approaches applied to human samples disclose
retinoblastoma. These alterations result in the absence of multiple osteosarcaroma driver genes, many of which
a functional RB gene product. The development of these belong to the superfamily of genes controlling genomic
changes follows the double-hit kinetics postulated for the stability.46,68,111,112,203 Interactive analysis of gene networks
development of retinoblastoma. The first mutation or discloses upregulation of histone cluster 2 genes at chro-
deletion alters one allele, and the subsequent “hit” results mosome 1q21.1q21.3, loss of chromosome 8p21.2-p21.3,
in the loss of function of the remaining allele. These data and inhibition of the DOCK5 and TNFRSF10A/D genes,
are consistent with clinical observations that document complemented by the amplification-related overexpres-
an increased incidence of osteosarcoma in patients with sion of RUNX2 at chromosome 6p12.3-p21.1.172 Activa-
retinoblastoma. In addition to the loss of heterozygosity tion of RUNX2 disrupts G2/M cell-cycle checkpoints;
at the RB locus on chromosome 13, osteosarcoma fre- inactivation of DOCK5 signaling in synchrony with TP53
quently shows loss of heterozygosity of loci on 3q, 17p, and TNFRSF10A/D-related changes plays a role in inac-
and 18q.219 tivating cell death pathways. MicroRNA analyses identi-
Alteration of TP53 is frequently present in osteosar- fied several novel miRNA species, which include the
comas.133,144,154,179,180,201 Germ cell line mutations of TP53 MIR21, MIR93, MIR143 and MIR145, miRNA199, and
were identified in some cancer-predisposing syndromes, the miRNA-17-92 cluster, as potential regulatory targets
such as the Li-Fraumeni syndrome, with high incidence of in osteosarcoma.15,58,66,143,155,223 Genome-wide analyses
osteosarcoma.200 Tumors with altered TP53 are also more disclose complex miRNA deregulation involving signal-
aggressive and have a high propensity for metastases. ing pathways important for the development of osteosar-
Amplification of the human homolog of the Mouse coma, including Notch, RAS/p21, MAPK, Wnt, and Jun/
double minute 2 gene (MDM2) with a p53 protein– FOS.128 Preliminary genome sequencing data shows that

ERRNVPHGLFRVRUJ
5  Osteosarcoma 259

a subset of osteosarcomas can develop through a novel The differentiation of osteosarcoma from osteoblas-
genetic mechanism referred to as chromothripsis.139,191 It toma and aggressive osteoblastoma is discussed in Chapter
postulates that if, in one catastrophic event, apoptosis is 4. It should be emphasized that some osteosarcomas may
not completed, the shattered chromosomes are reassem- contain focal areas that have a superficial microscopic
bled at random by nonhomologous end-joining. Such resemblance to osteoblastoma. In very rare instances, the
reassembly creates hundreds of translocations that involve entire tumor may exhibit a histologic pattern similar to
multiple target genes with oncogenic potentials. An that seen in osteoblastoma, with relatively uniform tumor
example of such a mechanism evidenced by genomic bone trabeculae and less conspicuous nuclear atypia. Such
sequencing of an osteosarcoma is shown in Fig. 5-54. variants of osteosarcoma are referred to as osteoblastoma-
The conceptual implication of this finding is that, in the like osteosarcoma. These variants usually contain atypi-
predisposed individual, a single event can trigger the cal mitoses, grow to a larger size than osteoblastomas
development of cancer without the long pathway of pre- (generally >5 cm), and exhibit a permeative and destruc-
neoplastic conditions. This concept is in keeping with the tive growth pattern with engulfment of preexistent bone
clinical development of the majority of de novo osteosar- trabeculae. These tumors in particular can be confused
comas, which affect young individuals who are without histologically with aggressive osteoblastoma.
any evidence of longstanding predisposing lesions. Chondrosarcoma is usually easily distinguished from
In summary, molecular techniques, including recently osteosarcoma on clinical, radiologic, and pathologic
applied genomic platforms, provide some interesting grounds. Problems in distinguishing between these
clues about the pathogenesis of osteosarcoma. The recent tumors arise when an osteosarcoma is predominantly car-
data contribute to our understanding of the complex tilaginous or when a low-grade chondrosarcoma exhibits
biology of this tumor; there is hope that such approaches enchondral ossification. The cartilaginous component of
in the future may identify novel diagnostic, prognostic, a chondroblastic osteosarcoma is usually of high grade
and therapeutic targets. histologically and is accompanied by foci of direct tumor
bone production from sarcomatous stromal cells. Enchon-
dral ossification is usually seen within cartilage lobules
Differential Diagnosis
of an otherwise typical low-grade chondrosarcoma. Fur-
Conventional intramedullary osteosarcoma can be con- thermore, clear cell chondrosarcomas characteristically
fused with both benign and malignant bone lesions. show microscopic features of bone formation that may
Careful correlation with radiographic findings and mimic osteosarcoma. The typical epiphyseal location
knowledge of the clinical setting will serve to avoid these and unique clear cell population in the intertrabecular
pitfalls in most cases. spaces help to distinguish this lesion from osteosarcoma.
Fracture callus can simulate a malignant bone-forming The anaplastic sarcomatous component of a dedifferenti-
neoplasm when the bone-forming features include ated chondrosarcoma may take the form of a high-grade
enlarged, highly active, immature but uniform osteo- osteoblastic osteosarcoma. This form of chondrosarcoma
blasts that have prominent mitotic activity. The osteoid can be distinguished from de novo osteosarcoma only
and woven bone trabeculae in callus usually show a when the low-grade cartilaginous component is identi-
pattern of parallel arrangement with prominent osteo- fied. This distinction is aided by careful radiographic cor-
blastic rimming. This purposeful orientation is not seen relation, which usually reveals the presence of a heavily
in osteosarcoma, where the bone matrix is deposited in a calcified low-grade chondrosarcoma component.
disorganized and haphazard fashion. Atypical (multipo- Malignant fibrous histiocytoma of bone has some over-
lar) mitotic figures are often present in osteosarcoma lapping features with osteosarcoma.97 Indeed, this tumor
together with pronounced nuclear atypia. Neither of was classified as a highly anaplastic osteolytic form of
these cytologic abnormalities should be present in frac- osteosarcoma before it was recognized as a distinct tumor
ture callus. Fracture callus frequently contains foci of entity. However, some forms of osteosarcoma closely
cartilage matrix production that may blend imperceptibly resemble malignant fibrous histiocytoma because their
with woven bone–producing osteochondroid material. principal cell population is highly pleomorphic and
The latter finding sometimes contributes to its confusion osteoid and tumor bone formation is focal and incon-
with osteosarcoma. spicuous (malignant fibrous histiocytoma-like osteosar-
In some cases of osteogenesis imperfecta tarda, incon- coma). The prevailing criteria for the diagnosis of
spicuous fractures can be associated with exuberant callus malignant fibrous histiocytoma of bone include the
that can mimic osteosarcoma both radiologically and absence of bone or cartilage matrix production by the
histologically. tumor cells, typical occurrence in an older population,
Acute suppurative osteomyelitis characteristically involves paucity of periosteal reactive bone, and frequent associa-
the metaphyses of long bones of children and is associ- tion with underlying bone conditions. When this tumor
ated with prominent periosteal new bone formation that occurs in the metaphyses of long bones in children or
simulates Codman’s triangles. For this reason, patients adolescents, the distinction from conventional osteosar-
with this inflammatory lesion may have a misdiagnosis of coma may be only academic because their biologic poten-
osteosarcoma; the misdiagnosis is based on radiographs. tials and treatments do not differ.
Similarly, osteosarcomas in the same clinical setting can Giant cell tumor of bone may be simulated histologically
be radiographically mistaken for osteomyelitis, and unless in cases of osteosarcoma that are rich in osteoclast-like
a biopsy is performed, patients may receive inappropriate giant cells and show minimal bone matrix produc-
antibiotic therapy. tion. Radiographic studies showing the characteristic

ERRNVPHGLFRVRUJ
260 5  Osteosarcoma

AA BB CC D D EE FF GG HH I I J J

Telomere Centromere
Shattering

F
B
A

E
C

F
J
H

H
A
E D

I
C

G
B D

Stitching

D C E F H G
AB CH I A B A B I J D E C D E F H I J

Deletion-type
4
2 Tandem dup-type
0
Tail-to-tail inverted
1
Head-to-head inverted
0
Interchromosomal

0 5 10 15 20 25 30 35 40 45 50 55 60 65
Genomic location (Mb)
B
Chr 8
Chr 14

C Chr 12

FIGURE 5-54  ■  Genome sequencing provides evidence for chromothripsis involved in the development of osteosarcoma. A, Diagram-
matic representation of chromosomal shattering and random reassembly resulting in a myriad of translocations. B, An example of
chromosomal maps documenting complex rearrangements that involve deletions, tandem duplications, tail-to-tail inversion, head-
to-head inversion, and interchromosomal insertions. C, Circos diagram of representative chromosomes, documenting multiple
chromosomal translocations depicted as colored arch lines.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 261

localization of giant cell tumor in the epiphyseal end disease-free survival rate. The overall 5-year survival rate
of a long bone in a skeletally mature patient serve to in this group was 64%, whereas the continuous disease-
distinguish it from giant cell–containing osteosarcoma. free survival rate was 58%.
Furthermore, osteosarcomas usually affect metaphyseal The introduction of chemotherapy in the 1970s
sites in skeletally immature individuals in whom the car- resulted in a gradual increase in the national survival rates
tilaginous growth plate acts as a barrier to epiphyseal for osteosarcoma, which improved significantly between
extension. Sampling also plays a role in this distinction 1973 and 1983 and again between 1984 and 1993.140
because the majority of giant cell–rich osteosarcomas More modern chemotherapy protocols, including stem
also contain more typical conventional osteosarcoma-like cell support, improved the rates of pathologic response
areas and the dominant giant cells are usually focally dis- but had minimal impact on survival.87,119,160 In summary,
tributed. The distinction is further aided by the presence there has been no dramatic improvement in survival rates
of nuclear anaplasia and atypical mitoses in the stromal since 1993.140
cells of osteosarcoma. Osteosarcomas frequently metastasize, and this
The rare small cell variant of osteosarcoma can be outcome is observed in approximately 80% of patients
confused microscopically with other round-cell tumors, treated by surgical excision alone. Such high metastatic
such as Ewing’s sarcoma, peripheral neuroectodermal rate is related to high propensity of osteosarcoma for early
tumor, and lymphoma. The small cell components of lymphatic spread.215 The most frequent sites of metastases
osteosarcoma are usually more pleomorphic than the are the lungs and liver. Apparently, there is a somewhat
cells that comprise Ewing’s sarcoma, lymphoma, and increased rate of skeletal metastases in patients treated
other round-cell tumors, the differential diagnosis of with chemotherapy. The presence of pulmonary metas-
which is aided by appropriate cell markers and electron tases can be associated with hypertrophic pulmonary
microscopic study. For details of the differential diagnosis osteoarthropathy.55,78 Surgical excision of metastases with
of small cell bone tumors, the reader is referred to adjuvant chemotherapy prolongs life.17,33,84 In the group
Chapters 11 and 12. of surgically resectable, especially solitary, metastases, this
Metastatic carcinoma rarely presents problems in dif- approach sporadically results in cure. The prognosis in
ferential diagnosis when the epithelial tumor provokes osteosarcoma is also related to the site of involvement.35,42
marked osteoblastic reaction, as is most frequently seen Surgically resectable lesions of the appendicular skeleton
in prostate or breast carcinomas. On the other hand, are associated with a nearly 50% 5-year survival rate. On
some osteosarcomas may contain large osteoblasts with the other hand, osteosarcoma that involves the trunk
densely eosinophilic or vacuolated cytoplasm that grows bones is associated with a less favorable survival rate (e.g.,
in cohesive sheets or nests and may simulate metastatic only 20% at 5 years for patients with pelvic osteosar-
carcinoma. Appropriate immunohistochemical studies coma).56 Osteosarcomas of the craniofacial bones, espe-
for epithelial markers and tumor-associated antigens may cially those of the mandible, were originally reported to
be useful in facilitating this diagnosis. have a more indolent course than other conventional
osteosarcomas.207,208 Satellite intramedullary foci (“skip”
metastases) were considered to be a major source of local
Treatment and Behavior
recurrence.61,129 The extensive use of MRI has shown that
Conventional osteosarcoma belongs to the category of the so-called skip foci are in fact extremely rare.
the most aggressive and highly lethal osseous neoplasms.19 Originally, it was postulated that classification of con-
Before the advent of chemotherapy, treatment by surgery ventional osteosarcoma into histologic subtypes had
alone resulted in low 5-year survival rates, within the some prognostic value. With the advent of modern che-
range of 10% to 20%.214 With the introduction of che- motherapeutic protocols, the consensus is that histologic
motherapy and especially with the development of subtypes of conventional osteosarcoma should be used
modern multiple-modality treatment protocols, this only in a descriptive sense and have no prognostic impli-
dismal prognosis has improved.8,9,19,20,21,28,39,101,123,169,182 cations. In summary, the final outcome in osteosarcoma
The initial evaluations of the treatment effect and sur- is related to multiple factors, such as site of involvement,
vival rates in osteosarcoma were complicated by the fact tumor size, resectability, presence of metastases, and
of spontaneously changing survival rates disclosed by perhaps to some extent the histologic grade or even the
analysis of data from the Mayo Clinic.75,195,196 Higher age of the patient and the dose of chemotherapy that can
5-year survival rates (60% to 70%) can be achieved only be tolerated.
in a subset of patients with osteosarcoma—those who Based on the recent analysis of SEER data, it appears
present with surgically resectable lesions and without evi- that survival rates are age specific.140 The relative 5-year
dence of metastases.181,189,197 The general treatment plan survival rate of young-onset osteosarcoma (aged 0-24
consists of preoperative (neoadjuvant) chemotherapy and years) is 61%. A similar survival rate of 58% was observed
sometimes irradiation followed by limb-sparing surgical for patients aged 29 to 59 years. A dramatic drop in the
excision (if possible) and a postoperative chemotherapeu- survival rate, to 24%, was recorded for patients older
tic regimen modified according to the extent of tumor than age 60 years.
necrosis.72,164 Data from the University of Texas MD The most important factor is the biologic potential of
Anderson Cancer Center indicate that 90% and more of the tumor, which is beginning to be investigated on a
tumor necrosis is associated with nearly 90% 5-year genome-wide scale by novel high throughput techniques.
disease-free survival.165,166 On the contrary, less than 90% There is hope that this approach may identify novel
of tumor necrosis is associated with only a 14% 5-year therapeutic targets and will guide the design of more

ERRNVPHGLFRVRUJ
262 5  Osteosarcoma

effective chemotherapeutic protocols that may further in the literature. Radiographic data strongly indicate that
improve the outcomes of patients affected by these highly most osteosarcomas originate as intramedullary metaph-
aggressive and still poorly understood malignancies. yseal lesions with secondary involvement of the cortex.
The extremely rare intracortical osteosarcoma has been
Conventional Osteosarcoma in Different reported only in the shafts of the tibia and femur. This
Anatomic Sites curious lesion is not a precursor or an in situ lesion of
the majority of osteosarcomas and is discussed in further
Osteosarcoma has a clear predilection for the metaphyses detail at the end of this chapter.
of long bones in adolescent patients. It is important
to remember, however, that osteosarcoma sporadically Osteosarcoma of Craniofacial Bones.  Osteosarcoma
occurs in virtually every part of the skeleton regardless of of craniofacial bones accounts for 6% to 10% of all
the patient’s age. It appears that there is a significant dif- cases.43,70,79,92,107,122,150 The mandible and the maxilla are
ference in anatomic distribution between young and the most frequently involved bones, representing 50%
elderly patients. In the older age group, the tumor is less and 25%, respectively, of cases involving craniofacial
likely to occur in long bones and more frequently involves bones (see Figs. 5-58, 5-59 and 5-62).210 Among the man-
flat bones and the axial skeleton.56,95 There is disagree- dibular cases, the body is more frequently involved than
ment as to the age and anatomic distribution of major the ramus (see Figs. 5-59 and 5-62), and the alveolar
histologic subtypes of osteosarcoma (i.e., osteoblastic, ridge is the most common location in the maxilla (see
chondroblastic, and fibroblastic). Some data indicate that Figs. 5-59 and 5-62). Osteosarcoma of the skull is very
chondroblastic osteosarcoma has a predilection for the rare and accounts for less than 2% of all cases located in
trunk bones and more frequently occurs in older patients. this site.98 It typically involves the cranial vault and practi-
The discrepancies in overall distribution of histologic cally never occurs in the base (see Figs. 5-58 and 5-60).
types of osteosarcoma are at least partially related to Osteosarcoma of the skull is often related to Paget’s
interobserver differences in classifying lesions. They are disease. This association is reported in nearly half of the
also related to the fact that some believe almost totally cases in this location. When osteosarcoma occurs in the
chondroblastic osteosarcomas are better classified as cranium, it almost always arises in bone formed by mem-
chondrosarcomas. Some authors postulate that osteosar- branous ossification and practically never occurs in the
coma of the jaws, especially the mandible, has a distinctly bones of the base of the skull that are preformed in car-
different natural history and perhaps should be regarded tilage. Interestingly, in this connection, the mandibular
as a separate clinicopathologic entity.50,207,208 body, formed by membranous ossification, gives rise to
most of the osteosarcomas at this site. The involvement
Osteosarcoma of Appendicular Skeleton.  Nearly 80% of other craniofacial bones such as the palate, shown in
of osteosarcomas occur in the long tubular bones of ado- Fig. 5-61, is extremely rare.
lescents. Almost 50% of cases are located in the knee These osteosarcomas typically affect skeletally mature
area. The distal femoral metaphysis is 2.5 times more patients; nearly 80% of the cases are reported in patients
frequently involved than the proximal tibial metaphysis. older than age 20 years. The age range is wide, but rare
The relative frequency of tumor occurrence can be cor- cases are seen in children younger than age 10 years. The
related with the growth potential of individual long peak incidence of craniofacial osteosarcoma is during the
bones. The femur is the most frequently affected bone third and fourth decades of life, and there is no sex
(nearly 50% of cases) and is followed by the tibia (~20%) predominance.
and the humerus (~10%). More specifically, the part of The radiographic appearance of craniofacial osteosar-
the bone with the highest growth rate is most frequently comas does not differ significantly from that of tumors
involved. Accordingly, the distal femoral metaphysis, located in the long tubular bones. However, the complex
where most growth occurs during adolescence, is approx- anatomic structure of craniofacial bones requires special
imately 8 times more frequently involved than the proxi- expertise in evaluating lesions involving this site. The
mal femur. The ratio between tumor occurrence in the constellation of bone destruction, mineralized matrix
proximal and the distal tibia is approximately 10 : 1. A production, and periosteal new bone formation is typical
similar 10 : 1 ratio of the frequency of involvement by for the radiographic presentation of osteosarcoma (see
osteosarcoma is observed between the proximal and distal Figs. 5-57 to 5-62). A thickened vault and blurred cortical
ends of the humerus. The fibula, which is even more outlines, especially when associated with a focus of bone
rarely involved, accounts for approximately 3% of cases, sclerosis, can be early radiographic signs of osteosarcoma.
with tumor clustered in the proximal portion. Osteosar- Small lytic or sclerotic foci, especially in a background of
coma is extremely rare in the bones of the forearm. The Paget’s disease, are difficult to interpret and can be easily
diaphyses of long bones are unusual sites for osteosar- overlooked. Periosteal changes in the form of laminated
coma, and no more than 6% of osteosarcomas are exclu- or perpendicular new bone formation and cortical irregu-
sively diaphyseal lesions. The femoral diaphysis is most larities are quite often present but may require special
frequently involved, followed by the diaphyses of the tibia views for demonstration on plain radiographs.
and humerus. Fewer than 1% of osteosarcomas occur in Cortical destruction in the mandible is almost invari-
short tubular bones of the hands and feet (Figs. 5-55 and ably associated with the disappearance of the lamina dura
5-56).142,153 Osteosarcoma is exceptionally rare distal to of the adjacent teeth and with secondary invasion of
the ankle and wrist joints. Very few well-documented the mandibular nerve canal. Computed tomography and
cases of primary epiphyseal osteosarcoma are described Text continued on p. 267

ERRNVPHGLFRVRUJ
5  Osteosarcoma 263

A B

C D
FIGURE 5-55  ■  Osteosarcoma of the acral skeleton. A, Plain radiograph showing a mineralized intramedullary lesion involving the
head of the fifth left metacarpal bone. B, Gross photograph of the bisected resection specimen showing gritty intramedullary lesion
involving the head of the fifth left metacarpal bone. C, Whole-mount specimen showing heavily mineralized intramedullary growth
pattern of an osteosarcoma extensively infiltrating the medullary cavity. D, Low power photomicrograph showing coarse depositions
of osteoid and anaplastic tumor cells characteristic of a high-grade osteosarcoma. (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
264 5  Osteosarcoma

A B

C
FIGURE 5-56  ■  Osteosarcoma of the calcaneus. A, Lateral plain radiograph showing heavily mineralized tumor involving the entire
calcaneus. B, Fat-saturated T2-weighted sagittal magnetic resonance image showing inhomogeneous signal enhancement involving
the entire calcaneus and extending to the soft tissue. C, Gross photograph of sagittally bisected resection specimen depicting heavily
mineralized mass involving almost the entire medullary cavity. Note soft tissue extension plantar and around calcaneal
tuberosity.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 265

B
FIGURE 5-57  ■  Osteosarcoma of navicular bone. A, Lateral radiograph of right foot shows expansile destructive lesion in navicular
bone. Inset, Computed tomogram of foot shows lytic destructive process with cortical disruption in navicular bone. B, Sagittal cut
section (amputation specimen) of tarsal (navicular) bone shows high-grade osteoblastic osteosarcoma that has broken out into the
soft tissue.

ERRNVPHGLFRVRUJ
266 5  Osteosarcoma

A B

C
FIGURE 5-58  ■  Osteosarcoma of craniofacial bones. A, Radiograph of large destructive mass in right maxilla of a 22-year-old woman
who had a recurrence of a lesion previously thought to represent fibrous dysplasia. Note cloudlike opacity in upper and central area
of lytic tumor mass. B, Coronal computed tomogram of the lesion in A shows maxillary replacement with bone-forming tumor that
extends to nasal cavity, orbit, and buccal soft tissue. C, Lateral radiograph showing de novo osteosarcoma of a skull in a child. A
destructive lesion involves the cranial vault of an 11-year-old boy who had noted a painless lump in frontoparietal region for 1 year.
Note homogeneous density anteriorly and cloudlike density within lytic area.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 267

B C
FIGURE 5-59  ■  Osteosarcoma of mandible. A, Panorex radiograph of jaw of a 26-year-old man who reported right-sided numbness
of chin for several months. Note patchy radiodensity (arrows) adjacent to roots of molar tooth with impingement on mandibular
canal. B, Computed tomogram shows radiodense tumor in right side of mandible (arrow). Periosteal new bone formation is present
on inner aspect. C, T1-weighted magnetic resonance image of mandible shows replacement of marrow by osteosarcoma in right
side of mandible. Tumor mass produces a low signal consistent with tumor bone formation.

MRI greatly enhance the preoperative assessment of behavior of these tumors compared with that of conven-
these lesions. These techniques make it possible to evalu- tional high-grade osteosarcomas. In addition, chondro-
ate the degree of involvement of adjacent anatomic struc- blastic osteosarcomas of the craniofacial bones affect the
tures and of the extent of soft tissue involvement. In same age population as the more typical osteoblastic
reference to calvarial lesions, the techniques are particu- osteosarcomas in the same anatomic site, and there is no
larly useful in demonstrating intracranial extension of the significant difference in survival and response to therapy
tumor. These imaging techniques permit better preop- between patients with these two lesions.
erative planning of the definitive surgical procedure in If secondary osteosarcomas related to Paget’s disease
the case of craniofacial osteosarcomas. and radiation therapy are excluded, cellular atypia is less
Osteosarcomas of craniofacial bones do not differ evident in craniofacial lesions, especially in the mandible.
microscopically from those located in the long bones. Nearly half of the cases involving the jaw bone are his-
The only significant difference is the preponderance of tologic grades 1 and 2. Whether the less aggressive
the chondroblastic type, which, in the mandible and the behavior of osteosarcoma of the jaws is related to their
maxilla, accounts for approximately 30% of the cases. chondroblastic features or to their better differentiation
These lesions produce tumor osteoid, but this can be very is still unclear. Regardless of minimal atypia, chondroid
focal and minimal in extent. In some cases, the cartilagi- differentiation in craniofacial bone tumors, especially in
nous differentiation of the tumor is so dominant that the mandible and maxilla, should be considered a warning
some observers postulate that these tumors should be signal of malignancy. Exclusive of callus, cartilaginous
classified as chondrosarcomas. This disagreement may be differentiation is almost never seen in benign conditions
academic but highlights the less aggressive biologic of these bones.

ERRNVPHGLFRVRUJ
268 5  Osteosarcoma

A B

C D
FIGURE 5-60  ■  Osteosarcoma of the skull. A, T2-weighted sagittal magnetic resonance image (MRI) with contrast showing a destruc-
tive intramedullary lesion of the cranial vault extending to the paraosseous soft tissue externally and internally. B, T1-weighted
coronal MRI with contrast of the tumor shown in A. C, Gross photograph of the bisected resection specimen showing mineralized
intramedullary lesion with destructive growth pattern and extension to the paraosseous soft tissue. Note cap-shaped extension to
the subcutaneous tissue externally. D, Sagittal computed tomography reformation of the same lesion documenting the extent of
intramedullary bone involvement.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 269

A B

C D
FIGURE 5-61  ■  Osteosarcoma of the palate. A, Axial computed tomogram with contrast, soft tissue window, depicting a mass pro-
truding into the oral cavity. B, Coronal CT, bone window, showing the tumor involving the hard palate and protruding into the oral
cavity. C, Gross photograph of the resection specimen depicting lobulated mass extending from roof of oral cavity. D, Bisected
specimen showing a fleshy mass originating in the hard palate and growing downward from the roof of the oral cavity.

ERRNVPHGLFRVRUJ
270 5  Osteosarcoma

B
FIGURE 5-62  ■  Osteosarcoma of the jaw. A, Specimen radiograph of mandibular resection specimen. Note destructive
intramedullary lesion and prominent radiating (sunburst) of periosteal new bone formation. B, Gross photograph of bisected man-
dibular specimen shown in A with high-grade chondroblastic osteosarcoma. Note lobules of tumor cartilage and ill-defined borders
of intraosseous component. Extension into soft tissue has radiating sunburst appearance of reactive bone spicules.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 271

The most important reason for therapeutic failure in is typically located in the vertebral body and grows into
craniofacial osteosarcoma is local recurrence. Nearly 50% the spinal canal and the paraspinal soft tissue. Densely
of mandibular osteosarcomas recur locally after resection. sclerotic osteosarcomas, especially those involving the
The recurrence rate is even higher with maxillary and posterior arch, can occasionally mimic a benign osteo-
skull lesions (80% and 75%, respectively). Metastases are blastic lesion (i.e., osteoid osteoma and osteoblastoma)
less frequent than local recurrence and occur in about on radiographs (see Fig. 5-65). Because chondroblastic
one third of patients with craniofacial osteosarcomas. osteosarcoma has some predilection for the trunk bones
They typically occur within 2 years of initial treatment, (i.e., the vertebral column and pelvis), the punctate cal-
predominantly in the lungs. Occasionally cervical lymph cifications seen in some osteosarcomas of this location
nodes can be involved, necessitating a radical neck dissec- may simulate chondrosarcoma on radiographs.
tion. Once metastases are identified, the average survival
rate is less than 1 year. The overall 5-year survival rate for
Personal Comments
mandibular tumors after combined modality treatment is
approximately 75%. The survival rates for maxillary and The majority of cases of conventional osteosarcoma
skull de novo lesions are similar. However, analysis of the present with clinical and radiologic features that strongly
SEER data indicates a much lower 5-year survival rate suggest the diagnosis on the basis of the patient’s age,
(in the range of 40%). Osteosarcomas of the craniofacial tumor location, and radiographic patterns of bone
bones that develop in association with Paget’s disease are destruction, matrix mineralization, and periosteal new
clearly much more aggressive than de novo lesions. bone formation. Diagnostic difficulties arise when occa-
sional osteosarcomas present in an older age group, in
Osteosarcoma of Flat Bones and Ribs.  Approximately nonmetaphyseal sites, or with a histologic pattern that
10% of osteosarcomas are located in flat bones. Almost deviates significantly from the classic one. Often, the
90% of osteosarcomas involving flat bones are located in diagnostic impression can be confirmed by the use of
the pelvis. The scapula is rarely involved, and less than closed biopsy techniques using computed tomographic
2% of all osteosarcomas occur in that site. Osteosarcoma control and a large-bore needle or trephine. Because
of flat bones has clinicopathologic characteristics similar most, if not all, high-grade osteosarcomas are now treated
to those of osteosarcoma of craniofacial bones and osteo- with one or more courses of multidrug chemotherapy
sarcomas arising in the vertebral column. They typically before definitive surgery, the adequacy of the initial
occur in skeletally mature patients and are rare during biopsy material is more important than ever. Effective
the second decade of life. In older patients, flat bones in chemotherapy may render it impossible to do special
general and the ilium in particular are relatively frequent studies, such as cytogenetics, DNA analysis, immunos-
sites of osteosarcoma. As in the osteosarcomas that arise taining, or electron microscopy of the tumor when the
in craniofacial bones and the vertebral column, approxi- resected specimen becomes available. It may even be
mately 30% of tumors have a predominant chondroblas- impossible to identify any viable tumor cells in a post-
tic component. A further similarity is the frequent chemotherapy specimen. Even more than in other bone
association with underlying conditions, such as Paget’s tumors, sampling problems are of particular importance
disease and radiation damage. In contrast to the fre- with regard to osteosarcoma because of its variability. For
quency of low histologic grade of osteosarcoma in cra- example, when the radiographic findings point toward a
niofacial sites, the pelvic and scapular lesions are usually highly aggressive bone-forming tumor, the biopsy sample
of high grade. Approximately 2% of osteosarcomas occur may show only an anaplastic sarcoma composed of pleo-
in the ribs, sternum, and clavicle (Figs. 5-63 and 5-64). morphic spindle cells and tumor giant cells without either
They are far less common than chondrosarcomas of these osteoid or tumor bone. In these cases, it may be necessary
sites. to resort to a presumptive pathologic diagnosis of osteo-
sarcoma on the basis of the radiographic findings in con-
Osteosarcoma of Vertebral Column.  Osteosarcoma is junction with these microscopic features. Perhaps even
extremely rare in the vertebral column: less than 4% of more important, a dilemma arises when the biopsy mate-
cases occur in this location.199 A significant proportion of rial contains a sample of the tumor that exhibits a decep-
these lesions represent secondary osteosarcoma related to tively benign appearance or nonneoplastic tissue related
previous radiation therapy or Paget’s disease. The sacrum to secondary phenomena (e.g., fracture callus and reac-
is the most frequently involved bone of the vertebral tive fibrosis). Careful attention to the radiographic find-
column. It appears that osteosarcoma is more frequent in ings and knowledge of the biopsy site help to resolve this
the lower thoracic and lumbar portion of the vertebral difficulty.
column. In the spine, osteosarcoma occurs in a much The widespread use of MRI in the diagnosis of bone
older population than osteosarcoma of the appendicular tumors has contributed greatly to the planning of ade-
skeleton. In patients older than age 60 years, osteosar- quate surgical resections because it demonstrates exqui-
coma of the flat bones and the vertebral column accounts sitely the intramedullary and extramedullary extent of
for approximately 40% of cases in this age group. On tumor involvement. Even though the method does not
radiographs, osteosarcoma of the vertebral column typi- image bone and other mineralized tissues, it is useful in
cally presents as a mixed lytic and sclerotic destructive picking up separate sites of marrow involvement. Skip
mass (Fig. 5-65). The exact location and extent of involve- areas are now known to be much rarer in osteosarcoma
ment of adjacent structures are best evaluated by com- as a result of widespread use of MRI. Refinements of
puted tomography, MRI, or both modalities. The mass this technique continue to allow for better assessment

ERRNVPHGLFRVRUJ
272 5  Osteosarcoma

A B

C D
FIGURE 5-63  ■  Osteosarcoma of rib. A and B, Axial computed tomogram with contrast showing a destructive lesion involving the
anterior portion of the rib. C, Gross photograph of the bisected specimen shows heavily mineralized tumor involving the rib and
circumferentially involving the paraosseous soft tissue. D, Low power microphotograph showing malignant bone-forming tumor
with irregular patches of osteoid deposits consistent with high-grade osteosarcoma. (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 273

A B

D E
FIGURE 5-64  ■  Osteosarcoma of sternum and clavicle. A, Axial computed tomogram (CT) showing mineralized mass of the sternal
manubrium. Note circumferential extension to the paraosseous soft tissue. B, Gross photograph of the resection specimen in
A shows heavily mineralized tumor with destructive growth pattern and extension to the parasternal soft tissue involving the sternal
manubrium consistent with osteosarcoma. C and D, Axial CTs showing destructive tumor involving the sternal end of the clavicle.
E, Bisected resection specimen showing destructive tumor involving the sternal end of the clavicle with variegated mineralization
pattern, cystic changes, and mucinous changes consistent with osteosarcoma.

ERRNVPHGLFRVRUJ
274 5  Osteosarcoma

A D

B F

FIGURE 5-65  ■  Osteosarcoma of vertebra. A and B, Anteroposterior (AP) and lateral radiographs of lumbar spine of a 15-year-old boy
with back pain and extradural block after collapse of body of second lumbar vertebra. C, Computed tomogram (CT) of L2 in A and
B shows tumor destruction with radiodense tissue extending into spinal canal and paraspinal soft tissue. D and E, AP and lateral
radiographs of lumbar spine of a 59-year-old man with 3-month history of back pain. Level of serum alkaline phosphatase was
elevated to twice normal level. Irregular, dense lesion involves right pedicle of third lumbar vertebra, suggesting osteoblastoma.
F, CT of L3 in D and E shows radiodense tumor mass impinging on spinal canal and extending into paraspinal soft tissue with
pattern more consistent with osteosarcoma.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 275

of tumor response to chemotherapy before definitive motion. In general, the symptoms are similar to those of
surgery. Pathologic assessment of postchemotherapy conventional osteosarcoma. The high level of alkaline
resection specimens by mapping continues to be the phosphatase frequently seen in osteosarcoma is not a
mainstay in evaluating tumor response to treatment and feature of telangiectatic osteosarcoma.
in planning alternative or further treatment in cases
where there has been an unfavorable response to the
Radiographic Imaging
initial treatment plan.
The explosion of data generated by high throughput Telangiectatic osteosarcoma is typically a purely lytic
genomic approaches provides interesting insights into lesion with a permeative destructive growth pat­
the complexity of molecular alterations involved in the tern.230,231,237,241,251 This rapidly growing tumor often
development of osteosarcoma. There is hope that these expands the contour of bone and shows features of corti-
approaches may identify novel diagnostic, prognostic, cal disruption with minimal or no periosteal new bone
and therapeutic targets for these highly aggressive formation (Fig. 5-67). In this type of osteosarcoma, the
malignancies. periosteal new bone formation is often a multilayered
structure referred to as onion skin. Overall, the radio-
graphic appearance is not that of typical osteosarcoma,
Telangiectatic Osteosarcoma which has a mixture of blastic and lytic areas. Indeed, a
purely lytic radiographic appearance is one of the diag-
Definition
nostic requirements for this designation (Figs. 5-67 and
Telangiectatic osteosarcoma is a malignant bone-forming 5-68). The radiographic features of telangiectatic osteo-
tumor microscopically composed of blood-filled spaces sarcoma are less diagnostically specific than those of
divided by septa containing neoplastic sarcomatous cells. conventional osteosarcoma and are similar to other lytic,
There may be only minimal and focal osteoid production. rapidly growing malignant tumors of bone, such as
Radiologically as well as microscopically, it superficially Ewing’s sarcoma. The massive expansile growth pattern
mimics aneurysmal bone cyst. of telangiectatic osteosarcoma can superficially simulate
aneurysmal bone cyst (Figs. 5-69 and 5-70). This type of
osteosarcoma, in particular, can have a deceptively inno-
Incidence and Location
cent radiographic appearance with sharply demarcated
Telangiectatic osteosarcoma is rare, accounting for less margins, simulating a benign bone cyst.234 The multi-
than 4% of all cases of osteosarcoma. Originally, it was locular cystic nature of the lesion and the so-called fluid
thought to be a variant of conventional osteosarcoma, level sign are best documented by MRI (see Figs. 5-68
but recently it has been categorized as a distinct form and 5-69).
of osteosarcoma. The peak incidence and the anatomic
distribution are generally similar to those of conven-
Gross Findings
tional osteosarcoma.233,239,252 Telangiectatic osteosarcoma
most frequently occurs during the second decade of life Telangiectatic osteosarcoma is very hemorrhagic, and its
and is dominated by males (1.9 : 1 male-to-female sex gross appearance is frequently described as a bag of blood.
ratio). It usually involves the appendicular skeleton, with A fleshy sarcomatous component is typically not present.
more than 50% of cases located in the knee area. The The tumor grossly mimics aneurysmal bone cyst and pres-
distal femoral metaphysis is the single most common ents as a lytic lesion filled with hemorrhagic cystic or
anatomic site followed by the proximal tibia and the spongy tissue or both (Figs. 5-69 to 5-71). The cystic
proximal humerus. Approximately 10% of telangiectatic spaces can vary considerably in size and may occasionally
osteosarcomas are exclusively diaphyseal lesions that measure several centimeters in diameter. More often, there
predominantly involve the femur, tibia, and humerus. is a mixture of large cystic and spongy areas. The spongy
Individual cases have been reported in flat bones, the areas represent tissue honeycombed with smaller cysts that
axial skeleton, and craniofacial bones, but for practi- measure up to several millimeters in size. The borders of
cal purposes, telangiectatic osteosarcoma is a disease of the lesion are usually well demarcated, but often there are
long tubular bones.225,227,249 Rare examples of multicentric features of invasive growth with extensive irregular cortical
and extraskeletal telangiectatic osteosarcoma have also erosion, complete disruption of cortical continuity, and
been described.232,243 The peak age incidence and the invasion of soft tissue. The soft tissue component is ini-
most frequent skeletal sites of involvement are shown in tially outlined by a rim of elevated periosteum. With tumor
Figure 5-66. In rare instances, telangiectatic osteosar- progression, there is usually massive destruction of the
coma may involve extraskeletal sites or may develop as involved bone and formation of a large palpable mass.
dedifferentiation of preexisting low-grade lesions such
as parosteal osteosarcoma and chondrosarcoma or other
Microscopic Findings
solid extraskeletal tumors.233,235,238,240,242,244,250,253
Typically this tumor consists of cystlike spaces divided
Clinical Symptoms by septa similar to those seen in aneurysmal bone cyst
(Figs. 5-72 and 5-73). However, the septa are composed
Most patients initially have pain in the affected extremity of highly atypical sarcomatous tissue (Figs. 5-74 to 5-78).
that has continued for several weeks to months. The pain Less frequently, instead of cystlike spaces divided by
is often accompanied by swelling and limitation of septa, there are large areas of hemorrhage with “floating”

ERRNVPHGLFRVRUJ
276 5  Osteosarcoma

Peak
incidence
10 20
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-66  ■  Telangiectatic osteosarcoma. Peak age incidence and frequent sites of skeletal involvement.

individual pleomorphic sarcomatous cells (Figs. 5-79 and observed in small peripheral foci of this tumor. The
5-80). The cystic spaces have no endothelial lining, and advancing edge of the tumor, as it permeates marrow
the tumor cells are in direct contact with areas of hemor- spaces, sometimes shows an affinity for small, preexisting
rhage. Usually there is a high degree of nuclear atypia, blood vessels.
cellular pleomorphism, and brisk mitotic activity with Occasionally, the tissue within the septa can be similar
numerous atypical mitoses. In addition to multinucleated to that seen in aneurysmal bone cyst with very few
sarcomatous giant cells with bizarre nuclei, there may atypical cells (see Fig. 5-78). In such cases, a careful
be clearly benign osteoclast-like giant cells, which can search for atypical mitoses can disclose the true nature of
contribute to the mimicry of aneurysmal bone cyst by the lesion. Telangiectatic osteosarcoma with a deceptively
telangiectatic osteosarcoma. The osteoid production is benign histologic appearance is sometimes referred to as
usually minimal and focal (see Figs. 5-73 and 5-74). In low-grade telangiectatic osteosarcoma. This term is not
fact, often the osteoid formation can only be found after widely accepted in the literature, but it is sometimes
prolonged search. If the tumor has overall telangiectatic applied to cases in which repeated biopsies over time are
architecture (i.e., produces septa) or if highly pleomor- misdiagnosed as recurrent aneurysmal bone cyst until the
phic sarcoma cells are found floating in the large areas of sarcomatous nature of the tumor becomes obvious.229,234,248
hemorrhage, the lesion should be classified presump- Controversial cases of telangiectatic osteosarcomas that
tively as a telangiectatic osteosarcoma, even without evi- develop within a preexisting aneurysmal bone cyst have
dence of osteoid production. also been described in the literature.236
Typically, telangiectatic osteosarcoma is a high-grade
lesion with easily recognizable sarcomatous septa (Fig. Special Techniques
5-80). The concept that telangiectatic osteosarcoma is an
entity sui generis and not simply a variant of conventional Special techniques do not aid in the diagnosis of telangi-
osteosarcoma is borne out by the fact that the telangiec- ectatic osteosarcoma. Ultrastructurally, the tumor shows
tatic architecture is frequently reproduced in metastatic undifferentiated pleomorphic sarcomatous cells with
lesions from this tumor. The formation of telangiectatic minimal focal matrix mineralization.245 DNA ploidy mea-
blood-filled spaces is therefore not considered to be a surements typically show a high degree of aneuploidy,
secondary degenerative phenomenon. It can even be Text continued on p. 291

ERRNVPHGLFRVRUJ
5  Osteosarcoma 277

A B

C D
FIGURE 5-67  ■  Telangiectatic osteosarcoma. A, Slightly expanded, apparently well-circumscribed lytic lesion is shown in this plain
radiograph of femoral diaphysis of young adult. It was initially thought to represent a bone cyst or fibrous dysplasia. B, Purely lytic
lesion of proximal end of humerus shows cortical destruction of medial aspect. C, Anteroposterior plain radiograph shows ill-defined
radiolucent tumor in distal femoral shaft. D, Lateral view of specimen radiograph of same case in C documenting pathologic fracture
(arrow).

ERRNVPHGLFRVRUJ
278 5  Osteosarcoma

A B

C D
FIGURE 5-68  ■  Telangiectatic osteosarcoma: radiographic features. A and B, Anteroposterior and lateral plain radiographs show ill-
defined lytic and permeative lesion of distal femoral metaphysis with pathologic fracture. C, Lytic lesion with sclerotic, well-defined
margin. Note deceptively benign radiographic appearance of this high-grade telangiectatic osteosarcoma. D, T1-weighted coronal
magnetic resonance image shows multiloculated architecture of lesion with different levels of signal intensity.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 279

C D
FIGURE 5-69  ■  Telangiectatic osteosarcoma. A, Anteroposterior plain radiograph shows ill-defined lytic and permeative lesion of
distal metaphysis. Note extension to the soft tissue laterally with elevation of the paraosteum (Codman’s triangle) proximal to lesion.
Inset, Fat-saturated T2-weighted axial magnetic resonance image showing multiloculated cystic architecture of the lesion with fluid
levels. B, Gross photograph of the lesion in A shows expansile hemorrhagic tumor of the distal femoral metaphysis extending to
the soft tissue laterally. Note elevation of the periosteum proximal to lesion corresponding to Codman’s triangle shown in A.
C, Low power microphotography showing multilocular cystic architecture of the lesion. Inset, Higher power of septum
with histiocyte-like neoplastic cells showing nuclear atypia. D, Microscopic features of telangiectatic osteosarcoma septations
containing pleomorphic neoplastic cells and scattered multinucleated giant cells. (C, ×50; D, ×100; inset, ×100) (C, D, and
inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
280 5  Osteosarcoma

A B

C D
FIGURE 5-70  ■  Telangiectatic osteosarcoma. A, Lateral plain radiograph showing expansile blowout lesion with permeative growth
pattern of the proximal tibia. B, Gross photograph of A showing hemorrhagic and partially necrotic tumor of the proximal tibial
metaphysis with massive soft tissue expansion posteriorly. C, Low power photomicrograph showing multilocular cystic architecture
of the lesion. D, Higher magnification of C showing a small cystic space lined by a mantle of highly atypical neoplastic cells. Some
of the tumor cells are free floating in the cystic space. (C, ×50; D, ×200) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 281

A B

C D
FIGURE 5-71  ■  Telangiectatic osteosarcoma. A, Expansile hemorrhagic tumor replacing proximal end of fibula. Note cortical blowout
destruction. B, Massive extension into soft tissue from intramedullary osteosarcoma of humerus forms large encircling mass with
hemorrhagic loculi in fleshy background. C, Septa bordering blood field spaces. D, Higher magnification of C shows nuclear atypia
of tumor cells within the septum. (C, ×50; D, ×200) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
282 5  Osteosarcoma

A B

C D
FIGURE 5-72  ■  Telangiectatic osteosarcoma: microscopic features. A, Low power photomicrograph shows multilocular cystic archi-
tecture of the lesion. B, Higher magnification of A showing a meandering septum containing neoplastic cells and scattered multi-
nucleated giant cells. Inset shows higher magnification of B. Note irregular mantles of anaplastic tumor cells lining the septum and
a multinucleated giant cell in the center. C, Collapsed cystic spaces with clustered septations, a common feature in telangiectatic
osteosarcoma. D, Higher magnification of C shows nuclear atypia of neoplastic cells within the septa. Note scattered multinucleated
giant cells. (A, ×20; B and C, ×50; D, x200; inset, ×400) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 283

A B

C D
FIGURE 5-73  ■  Telangiectatic osteosarcoma: microscopic features. A and B, Low power photomicrograph showing blood filled spaces
separated by irregular septa. C, Higher magnification of B showing tumor osteoid deposits within the septum. D, Microscopic cyst-
like space surrounded by an irregular mantle of histiocyte-like neoplastic cells and scattered multinucleated giant cells. (A and
B, ×20; C, ×50; D, ×200) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
284 5  Osteosarcoma

A B

C D
FIGURE 5-74  ■  Telangiectatic osteosarcoma: microscopic features. A, A low power photomicrograph showing cystlike spaces of
various sizes and a more solid component of the tumor. Note delicate osteoid deposits. B, Higher magnification of A showing osteoid
matrix within the septum. C, Higher magnification of A showing an irregular cystlike space lined by a mantle of partially free-floating
tumor cells. D, Higher magnification showing the interface between the septum and cystic space lined by a mantle of partially free-
floating, highly atypical tumor cells. (A, ×50; B and C, ×100; D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 285

A B

C D
FIGURE 5-75  ■  Telangiectatic osteosarcoma: microscopic features. A, Low power photomicrograph showing irregular blood-filled
spaces of different sizes within the cellular solid component of the tumor. B, Medium powered magnification of septum containing
sheets of malignant cells. C, Solid area of the tumor composed of highly pleomorphic tumor cells. D, Higher magnification of
C documenting atypia and pleomorphism of tumor cells. (A, ×50; B and C, ×100; D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
286 5  Osteosarcoma

B C
FIGURE 5-76  ■  Telangiectatic osteosarcoma: microscopic features. A, Low power photomicrograph showing partially collapsed
irregular blood-filled spaces separated by meandering septations. B, Higher magnification of A showing meandering septations
bordering blood-filled spaces. C, Higher magnification of B showing atypia of histiocyte-like tumor cells within the septa. (A, ×20;
B, ×100; C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 287

A B

C D
FIGURE 5-77  ■  Telangiectatic osteosarcoma: microscopic features. A, Low power photomicrograph showing cystlike spaces and solid
components of the tumor. B, Intermediate power magnification of A showing interface between solid and cystic components of the
tumor. C, Another intermediate power magnification of A showing smaller cystic space within the solid component of the tumor.
D, Higher magnification of C showing the solid area of the tumor composed of histiocyte-like malignant cells with scattered multi-
nucleated giant cells. (A, ×20; B and C, ×100; D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
288 5  Osteosarcoma

B C
FIGURE 5-78  ■  Telangiectatic osteosarcoma: microscopic features. A, Large partially collapsed cystic spaces separated by irregular
septations. Inset, Cellular composition of the septum. Note overall bland appearance of cellular components within the septum that
may cause misdiagnosis and confusion with aneurysmal bone cyst. B, Low power photomicrograph showing interface between
cystic and solid components of the tumor. C, Higher magnification of A showing high cellularity and atypia of tumor cells within
the septa. (A and B, ×20; C, ×100; inset, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 289

A B

C D
FIGURE 5-79  ■  Telangiectatic osteosarcoma: microscopic features. A, Solid component of the tumor with extensive interstitial fresh
hemorrhage (upper right) and smaller cystic spaces (lower left). B, Higher magnification of A showing mineralized osteoid bands
within the solid component of the tumor bordering the area of fresh hemorrhage. C, Partially collapsed hemorrhagic spaces sepa-
rated by irregular septations. D, Higher magnification of C showing bands of osteoid matrix within the septum. (A and C, ×20;
B and D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
290 5  Osteosarcoma

B C
FIGURE 5-80  ■  Telangiectatic osteosarcoma: microscopic features. A, Low power photomicrograph showing irregular cystlike spaces
with fresh hemorrhage and septations composed of highly cellular tumor tissue. B, Higher magnification of A showing cystlike
spaces filled with hemorrhage and septations composed of anaplastic tumor cells. Inset, Higher magnification of septum composed
of histiocyte-like atypical tumor cells. C, Areas of interstitial fresh hemorrhage with floating, noncohesive tumor cells. (A, ×20;
B and C, ×100; inset, ×200) (A-C and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 291

which is consistent with morphometric analysis that dis- sarcoma is characterized microscopically by deceptively
closes great variability in the nuclear area and volume.247 benign cytologic features in the spindle-cell component
Immunohistochemically, the tumor cells of telangiectatic and relative maturity of the tumor bone. It also follows
osteosarcoma are positive for vimentin and negative for an indolent course.
other markers.
Incidence and Location
Differential Diagnosis
This very rare variant of osteosarcoma accounts for
This uncommon form of osteosarcoma must be distin- approximately 1% of all osteosarcomas.255,258,264 The peak
guished principally from aneurysmal bone cyst and con­ incidence is during the third decade of life, in which
ventional osteosarcoma with focal telangiectatic features.248 nearly 50% of the cases occur. Individual cases are diag-
At times, it may suggest the possibility of malignant forms nosed during the second decade of life and in people
of giant cell tumor, but this distinction can be made on the older than age 50 years. There is no sexual predilection.
basis of age (skeletal maturity) and localization in epiphy- Most cases occur in the major long bones of the lower
seal sites characteristic of giant cell tumors. It is important extremities, the femur being the most frequently involved
to apply strict radiologic and microscopic criteria. Radio- (approximately 50%). The tibia is the second most fre-
logic absence of sclerotic features and histologic paucity quently involved bone. As in other forms of osteosar-
of tumor bone formation are required to distinguish tel- coma, the knee area is involved in the majority of cases.
angiectatic osteosarcoma from otherwise conventional Intraosseous well-differentiated osteosarcoma has also
osteosarcoma that may exhibit focal and minor histologic been reported in the bones of the trunk and the cranio-
features of high vascularity or blood-filled channels. Simi- facial bones. This tumor typically involves the metaphy-
larly, the microscopic feature of hemangiopericytoma- seal parts of the long bones of skeletally mature patients.
like areas with branching endothelial-lined vascular Rare examples of intracortical or multicentric well-
channels described in the section on conventional osteo- differentiated osteosarcoma have been described.254,260
sarcoma should not be misinterpreted as telangiectatic The peak age incidence and the most frequently involved
osteosarcoma. The most important aspect of differential skeletal sites are shown in Figure 5-81.
diagnosis is the mimicry of the benign aneurysmal bone
cyst. Errors in diagnosis can occur in both directions
Clinical Symptoms
when close attention is not paid to the cytologic features
of the septal cells or if there is undue alarm over the Most patients initially have pain in the affected extremity
presence of reactive osteoid or reactive bone formation with a duration of many months to several years, in
in aneurysmal bone cyst. Bands of osteoid or delicate contrast to conventional osteosarcoma, in which the
woven bone trabeculae oriented parallel to the long axes duration of symptoms is measured in weeks to months.
of aneurysmal bone cyst septa should not be overinter- Occasionally, the patient notices a palpable mass as the
preted as features of malignancy. Careful scrutiny of the initial symptom. Some patients have a history of a recur-
surrounding cells helps avoid this pitfall. rent lesion, previously diagnosed pathologically as fibrous
dysplasia or other benign spindle-cell lesions.
Clinical Behavior
Radiographic Imaging
Originally, it was thought that telangiectatic osteosar-
coma had a particularly doleful prognosis, much worse These tumors exhibit a wide range of radiographic fea-
than conventional osteosarcoma. Its pattern of metastatic tures, but most show a variable pattern of lytic foci
spread is similar to that seen in conventional osteosar- and dense cloudlike areas that are poorly demarcated
coma. The original Mayo Clinic data indicated that more (Figs. 5-82 to 5-86).256,259,263,265 In contrast to conventional
than 80% of patients with this form of osteosarcoma die osteosarcoma, periosteal new bone formation is minimal
of the disease within 2 years of diagnosis. On the other or absent, even in advanced lesions that have significant
hand, the experience with modern combined modality cortical involvement. The ill-defined medullary tumor
treatment indicates that the particularly bad prognosis can frequently extend to the end of the bone to involve
formerly associated with telangiectatic osteosarcoma the subarticular bony end plate. The shaft may be exten-
probably no longer applies.225,226,228,246 In fact, telangiec- sively involved for a considerable distance from the
tatic osteosarcoma usually shows a better response to metaphyseal area. In addition to alternating areas of
chemotherapy than conventional osteosarcoma. Survival amorphous density and lytic foci, a coarse trabeculated
rates with current chemotherapy protocols are within the pattern of ossification has been observed (see Figs. 5-83
range of 65% after 5 years. to 5-86). Expansion of the bone contour indicating slow
growth may be seen. Complete cortical destruction may
be followed by significant extraosseous extension with an
Well-differentiated ill-defined soft tissue mass (Fig. 5-86). The fact that the
Intramedullary Osteosarcoma radiologic interpretation of these lesions is overwhelm-
ingly that of a malignant process highlights the necessity
Definition
for the pathologist to obtain radiologic correlative data
A distinct form of intramedullary low-grade fibroblastic in the interpretation of any microscopically atypical
osteosarcoma, well-differentiated intramedullary osteo- fibroosseous bone lesion.

ERRNVPHGLFRVRUJ
292 5  Osteosarcoma

Peak
incidence
20 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-81  ■  Low-grade intramedullary osteosarcoma. Peak age incidence and frequent sites of skeletal involvement.

Gross Findings (Fig. 5-89). Even the branching and curved “Chinese
character” appearance of woven bone trabeculae seen in
The tumors are predominantly centrally located in fibrous dysplasia can be found. The fibroblast-like spindle
the medullary cavity of the long bone metaphysis and cells are plump with elongated nuclei that contain promi-
diaphysis. Some are eccentrically placed, and the cortical nent nucleoli, but they are generally uniform in appear-
destruction may be striking. The tumor tissue is grayish ance (Figs. 5-89 and 5-90). Atypia is usually minimal.
and gritty with fleshy areas, reflecting the fibrous and Mitoses are infrequent but easily found, and atypical
osseous makeup (Fig. 5-84). Poor demarcation from mitoses are rare. The bone formed in this tumor varies
uninvolved bone is most frequently present, although in its degree of maturation from slender woven bone
some tumors may have sharply demarcated margins in trabeculae to thick, irregular bone spicules that can
heavily ossified areas. The tumor tends to fuse with the coalesce to form solid bony areas, which can approach
inner surface of the cortex. Extension into soft tissue is lamellar maturation. True lamellar architecture, if it is
usually associated with minimal periosteal bone forma- present, implies residual host bone. An interesting affin-
tion, and Codman’s triangles are rarely present. ity for peritrabecular growth by this tumor may be
observed at its periphery. In some cases, small foci of
atypical cartilage may be present in the tumor.
Microscopic Findings
Areas of high-grade pleomorphic osteosarcoma can
This tumor differs markedly in its histologic appearance be found within a low-grade intramedullary osteosar-
from conventional osteosarcoma because its fibroblastic coma.258,264 This can be observed in the primary tumor
and osseous components are well differentiated and but more frequently occurs in recurrences after incom-
reproduce within the medullary cavity, the pattern seen plete removal of the primary tumor. The low- and high-
in parosteal osteosarcoma (Fig. 5-87).255,258,261-263 The grade components are sharply demarcated (Fig. 5-91), as
mature fibroblast-like appearance of tumor cells embed- is generally the case when low-grade tumors dedifferenti-
ded in a dense collaginous stroma is best seen under the ate. These tumors are referred to as dedifferentiated
electron microscope (Fig. 5-88). In some cases the bland low-grade intramedullary osteosarcomas.257 The current
hypercellular spindle-cell components and the delicate pathogenetic concept postulates that dedifferentiation
osseous trabeculae can closely simulate fibrous dysplasia Text continued on p. 297

ERRNVPHGLFRVRUJ
5  Osteosarcoma 293

A B

C D

FIGURE 5-82  ■  Low-grade intramedullary osteosarcoma: radiographic features. A and B, Anteroposterior (AP) and lateral plain radio-
graphs of distal femur in a 12-year-old girl. Tumor is expansile and shows trabeculated pattern of bone destruction. No periosteal
reactive bone is seen, and proximal border is indistinct. C, AP view of low-grade intramedullary osteosarcoma of distal femoral
shaft. Note expanded contour produced by this slow-growing tumor, which had been previously biopsied and diagnosed as fibrous
dysplasia. D, Lateral radiograph of C shows extramedullary extension posteriorly and trabeculated radiolucent appearance of intra-
medullary component.

ERRNVPHGLFRVRUJ
294 5  Osteosarcoma

A B

C D
FIGURE 5-83  ■  Low-grade intramedullary osteosarcoma. A, Lateral plain radiograph shows ill-defined sclerotic intramedullary lesion
involving the proximal tibial metaphysis. B, Reformatted computed tomogram in coronal plane of A shows ill-defined intramedullary
lesion of the proximal tibial metaphysis. C, Anteroposterior plain radiograph of sclerotic ill-defined intramedullary lesion of the
proximal tibia extending to the soft tissue laterally and medially. D, Low power photomicrograph of the tumor shown in C, with
well developed bone trabeculae that simulates those seen in fibrous dysplasia. (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 295

A B

C D
FIGURE 5-84  ■  Low-grade intramedullary osteosarcoma. A, Reformatted sagittal computed tomogram (CT) in sagittal plane showing
intramedullary lesion of the mandible. B, Reformatted CT in coronal plane showing intramedullary lesion of the mandible with ill-
defined borders and increased signals, centrally. C, Axial CT showing intramedullary mental protuberance region of the mandible.
Note ill-defined borders and cortical penetration anteriorly. D, Gross photographs of the bisected resection specimens showing
overall fibrous appearance of the lesion with focal granularity. Note ill-defined borders and cortical penetration anteriorly.

ERRNVPHGLFRVRUJ
296 5  Osteosarcoma

B
FIGURE 5-85  ■  Low-grade intramedullary osteosarcoma: radiographic features. A, radiograph showing extensive predominantly lytic
destructive lesion of the left ilium. B, Axial computed tomogram showing extensive destructive process involving the entire iliac
bone and extending to the sacroiliac region.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 297

A B
FIGURE 5-86  ■  Low-grade intramedullary osteosarcoma: radiographic features. A, Anteroposterior plain radiograph showing sclerotic
intramedullary lesion expanding the distal end of the ulna. B, Lateral plain radiograph showing a mixed lytic and sclerotic lesion
with ill-defined margins involving the distal femur. Note soft tissue extension by sclerotic tumor posteriorly.

represents a progression of a preexisting low-grade fibro- particularly common (e.g., the craniofacial bones), the
blastic osteosarcoma to a high-grade sarcomatoid malig- recognition of well-differentiated osteosarcoma may be
nancy. The dedifferentiated component typically has especially challenging. Microscopically, the coarseness
features of high-grade osteoblastic osteosarcoma (Fig. of the tumor bone trabeculae, compared with the deli-
5-92). Less frequently, it may have features of a high- cate branching and curved woven trabeculae of fibrous
grade unclassified sarcoma with overall architecture dysplasia, provides a useful guide. Careful search for
similar to malignant fibrous histiocytoma. A similar phe- atypical nuclei and mitotic activity in the spindle-cell ele-
nomenon occurs in parosteal osteosarcoma (dedifferenti- ments is essential for the diagnosis of well-differentiated
ated parosteal osteosarcoma), which is described later. osteosarcoma.
Other malignant spindle-cell tumors of bone such as
fibrosarcoma, malignant fibrous histiocytoma, and leiomyosar-
Differential Diagnosis
coma can sometimes be confused with this tumor, but they
The striking difference in biologic behavior and survival do not show evidence of tumor bone formation, and
when this tumor is compared with conventional intramed- specific markers can be useful in their recognition. The
ullary osteosarcoma mandates its distinct separation from benign but locally aggressive desmoplastic fibroma of bone
the more common high-grade tumor. Often, it overlaps is also devoid of tumor bone components, although it
with the fibroblastic type of medullary osteosarcoma, may contain reactive bone at its periphery.
but the degree of nuclear pleomorphism and the rate of
mitosis are far less in the well-differentiated variant. In
Treatment and Behavior
addition, the tumor bone trabeculae tend to be larger,
more abundant, and more mature. These distinguishing Inadequate treatment of this indolent, low-grade malig-
features of well-differentiated medullary osteosarcoma nancy in the form of curettage or partial resection inevi-
can produce a superficially benign microscopic appear- tably leads to recurrence.255,258,264 Difficulty in making the
ance that can lead to its misdiagnosis as fibrous dysplasia of histologic diagnosis and confusion with benign entities
bone. This is one of the most common misinterpretations contribute to the delay in diagnosis and inappropriate
and is sometimes carried through several serial biopsies treatment. Furthermore, the likelihood of increase in
of recurrences until the malignant nature of the tumor grade (dedifferentiation) rises with each recurrence.
becomes apparent. Metastasis occurs in patients whose tumors show areas of
Fibrous dysplasia can usually be recognized by its dis- high-grade transformation and hence in those whose
tinctly nonaggressive appearance on radiographs, but initial mode of therapy was inadequate. The overall
in certain anatomic sites where fibrous dysplasia is Text continued on p. 304

ERRNVPHGLFRVRUJ
298 5  Osteosarcoma

A B

C D
FIGURE 5-87  ■  Low-grade intramedullary osteosarcoma: microscopic features. A, Low power photomicrograph shows branching
well-developed bone trabeculae that simulate the pattern of fibrous dysplasia. B, Intermediate power magnification of A shows
interconnected bone trabeculae in fibrous stroma. Lack of rimming by osteoblastic cells contributes to the similarity of fibrous
dysplasia. C, Low power photomicrograph shows similar features in another area of the same tumor with well-developed intercon-
nected bone trabeculae resembling fibrous dysplasia. D, Intermediate power magnification of C showing branching bone trabeculae
without osteoblastic rimming in a fibrous stroma. (A and C, ×50; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 299

B
FIGURE 5-88  ■  Low-grade intramedullary osteosarcoma: ultrastructural features. A, Fibroblast-like spindle tumor cells surrounded
by dense collagen stroma. B, Higher magnification of A. (A, ×3500; B, ×9000) (A-B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
300 5  Osteosarcoma

A B

C D
FIGURE 5-89  ■  Low-grade intramedullary osteosarcoma: microscopic features. A, Low power photomicrograph showing well-
developed bone trabeculae resembling fibrous dysplasia. B, Higher magnification of A showing branching bone trabeculae
in somewhat hypercellular stromal tissue with spindle cells. C, Low power photomicrograph showing the bone trabecular
pattern mimicking fibrous dysplasia. D, Trabecular osteoid pattern of low-grade intramedullary osteosarcoma resembling fibrous
dysplasia. Note somewhat hypercellular stromal proliferation of spindle cells with loose texture. (A and C, ×50; B and D, ×100)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 301

A B

C D

FIGURE 5-90  ■  Low-grade intramedullary osteosarcoma: microscopic features. A, Osteoid deposition pattern with well-developed
branching bone trabeculae. B, Intermediate power magnification of A showing branching and somewhat parallel tumor bone tra-
beculae with well mineralized core and unmineralized surface osteoid deposits. Note somewhat hypercellular stromal tissue com-
posed of spindle cells. C, Low power photomicrograph showing branching pattern of tumor bone trabeculae. D, Slightly higher
magnification of C showing osteoid deposition in the form of interconnected and branching bony trabeculae. Inset shows higher
magnification of D and depicts focal parallel arrangement of bone trabeculae. (A and D, x50; B and inset, x100; C, x20) (A-D and
inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
302 5  Osteosarcoma

B
FIGURE 5-91  ■  Low-grade intramedullary osteosarcoma with dedifferentiation: microscopic features. A and B, Low and high power
views of junction of well-differentiated tumor (right) and high-grade dedifferentiated osteosarcoma (lower left). Inset, Marked nuclear
pleomorphism, hyperchromatism, and tumor giant cells in high-grade dedifferentiated component. (A, ×100; B, ×200; inset ×200)
(A, B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 303

A B

C D
FIGURE 5-92  ■  Low-grade intramedullary osteosarcoma with dedifferentiation: microscopic features. A and B, Intermediate power
magnification showing low-grade fibroblastic osteosarcoma infiltrating medullary cavity. The tumor is composed of spindle cell
stromal tissue and well developed branching bone trabeculae. Note residual fat cells of marrow infiltrated by the tumor.
C and D, High-grade osteosarcoma corresponding to a dedifferentiated component of the same tumor shown in A and B. Note
pronounced atypia and pleomorphism of tumor cells and coarse irregular tumor osteoid deposits consistent with high-grade osteo-
sarcoma. (A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
304 5  Osteosarcoma

incidence of metastasis is 15%. The treatment of choice Rothmund-Thomson syndrome, or congenital poikilo-
is wide excision or amputation. The use of adjuvant derma, is a rare autosomal recessive familial syndrome
chemotherapy is reserved for those cases showing dedif- characterized by erythematous and maculopapular skin
ferentiation in the primary tumor or in a recurrence. lesions with areas of hyperpigmentation, cataracts, pho-
tosensitivity, hypogonadism, psychomotor retardation,
and various skeletal abnormalities. Extensive data indi-
OSTEOSARCOMA ASSOCIATED WITH cate that Rothmund-Thomson syndrome belongs to a
SPECIFIC CLINICAL SYNDROMES group of unique diseases caused by missense mutations
in DNA helicases. A subset of patients with Rothmund-
High-grade conventional osteosarcomas can be Thomson syndrome have inactivating mutations in the
associated with certain clinical syndromes, such as RECQL4 helicase linked to chromosomal instability, a
familial osteosarcoma, retinoblastoma-associated osteo- hallmark of this disorder.291,303,325 RECQL4 mutations
sarcoma, Rothmund-Thomson syndrome, and multifocal are also seen in related disorders such as Baller-Gerold
osteosarcoma. syndrome or RAPADILINO.318,319 Rothmund-Thomson
Osteosarcoma in siblings is extremely rare and occurs syndrome predisposes an individual to the development
in 1 : 1000 to 1 : 3000 cases of conventional (sporadic) of squamous cell carcinoma of the skin and also to osteo-
osteosarcoma.307,308,314,363 An autosomal dominant predis- sarcoma, which may be multicentric.278,289 The osteosar-
position can be anticipated when it affects several genera- comas associated with this syndrome are of conventional
tions of one family.281 Familial osteosarcoma has two basic intramedullary type and show a tendency to develop in
forms. It may occur sporadically in siblings without an unusual skeletal sites. Autosomal recessive inheritance
inherited pattern of transmission.275,276,283,284,296,304,307 The has been suggested in this condition (see Chapter 24).
more frequent form is inherited familial osteosarcoma, Multifocal osteosarcoma can be subdivided into two clin-
which occurs in several generations of families, who are ical forms: the synchronous type that more often occurs
most frequently affected by retinoblastoma.282,298,315 This in childhood and adolescence (Figs. 5-93 and 5-94) and
peculiar association between two apparently unrelated the metachronous type that typically occurs in adults (see
malignant neoplasms led to the conclusion that there Fig. 5-96).268,270,280,287,290,292,297,300,310,313,322 But there is such
may be a common molecular genetic mechanism.326 For a significant overlap between these two forms of clinical
that reason, osteosarcoma associated with retinoblastoma presentation of multifocal osteosarcoma that metachro-
seems to be separate from other conventional osteosar- nous lesions can occur in childhood and adolescence and
comas and perhaps should be considered as a distinct synchronous lesions may affect adults. The synchronous
entity. type is practically always a high-grade sclerosing intra-
Families affected by retinoblastoma carry the germ medullary osteosarcoma that principally involves long
line alteration in the predisposing locus on chromosome bones in a symmetric distribution. The metachronous
13, which contains the RB gene. This alteration also type is asymmetrically distributed in adults and does not
increases the risk of developing osteosarcoma many hun- necessarily have sclerosing features on radiographs. It
dreds of times more than the chance of its occurring more often affects trunk bones and is associated with
in an individual who does not carry the genetic longer survival compared with the highly lethal synchro-
alteration.279,295,298,299,301 This genetic mutation not only nous juvenile type (Fig. 5-95). Average survival for the
increases the risk for development of de novo osteosar- latter is only 6 to 8 months. The less aggressive behavior
coma, but also potentiates the osteosarcoma-inducing of the adult metachronous type may be related to the
modality of external beam irradiation.266,273,274,277,302,311,321 lower histologic grade of these tumors. The synchronous
A potential ancillary role of TP53 in the development type of multifocal osteosarcoma with symmetric involve-
of osteosarcoma in association with retinoblastoma has ment of long bones and without a dominant mass and
also been postulated.285 Familial forms of osteosarcomas initial absence of pulmonary metastases almost certainly
without retinoblastoma have also been described.306,316 represents separately developing primary tumors. On the
Li-Fraumeni syndrome is an autosomal dominant contrary, the adult metachronous form of this condition
familial disorder of a complex cancer predisposing syn- most likely represents sequentially developing skeletal
drome and the missense germline mutations of the TP53 metastases.267,270 This concept is further supported by the
gene predominantly involving the DNA binding domain fact that some of the metachronous skeletal foci may
represent its underlying molecular defect causing genetic have features of skip metastases or may develop as soft
instability.271,286,293-295,305,309,312,317,320,324 Cancer risk in muta- tissue lesions (Fig. 5-96). Multifocal osteosarcoma result-
tion carriers is very high and has been estimated to be ing from heavy tumor burden can be associated with
50% by age 40 and nearly 90% by age 60. It is estimated particularly high levels of serum alkaline phosphatase.
that only 70% of families with Li-Fraumeni syndrome
carry germline mutations of TP53. A subset of patients
with this syndrome, who are negative for T53 mutations, SECONDARY OSTEOSARCOMA
carry mutations of CHEK2 in chromosome 22q11 or may
show alteration in PTEN.272 In addition, some Li-Fraumeni Osteosarcoma can develop in association with several
families show positive linkage to 1q23, possibly repre- neoplastic and nonneoplastic precursors.360 This subject
senting the third predisposing locus in this disorder.269 is covered in detail in Chapter 24. Paget’s disease is
Li-Fraumeni syndrome predisposes to a wide range of the most common precursor and is the prototype
sarcomas, including osteosarcoma. Text continued on p. 309

ERRNVPHGLFRVRUJ
5  Osteosarcoma 305

B
FIGURE 5-93  ■  Multifocal osteosarcoma (juvenile/adolescent type). A and B, Sixteen-year-old boy was first seen with multifocal pelvic
lesions and subsequently (7 months later) developed sclerotic lesions in vertebrae, ribs, clavicles, and skull. Death from widespread
metastases of this high-grade osteosarcoma occurred 2 years and 4 months after onset of symptoms.

ERRNVPHGLFRVRUJ
306 5  Osteosarcoma

A C
FIGURE 5-94  ■  Multifocal osteosarcoma (juvenile/adolescent type). A, Radioisotopic bone scan showing increased uptake in the
primary osteosarcoma of the distal right femur and multifocal skeletal lesions involving the right distal tibia, pelvis, left proximal
humerus, and the sternal end of the left clavicle. In addition, there are bilateral axillary metastases. B, Anteroposterior plain radio-
graph showing matrix-producing lesion in the proximal humerus, the sternal end of the clavicle, and in the soft tissue of axilla.
C, Sclerotic destructive tumor with extensive involvement of the paraosseous soft tissue laterally, most likely representing the
primary tumor site.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 307

B
FIGURE 5-95  ■  Multifocal osteosarcoma (adult type). A, Plain radiograph of pelvis shows two separate foci of sclerosing osteosarcoma
involving ilium and fifth lumbar vertebra in young woman. B, Computed tomogram of pelvis shows intraosseous and exophytic
radiodense tumor seen in A.

ERRNVPHGLFRVRUJ
308 5  Osteosarcoma

FIGURE 5-96  ■  Multifocal osteosarcoma (adult type). A, Radioisotopic bone scan showing increased uptake in the proximal humerus
and the soft tissue in the right proximal tibial/fibular region. B, Anteroposterior (AP) radiograph showing calcified tumor mass in
the intertibial/fibular region. Note external scalloping of the underlying tibial and fibular cortex consistent with soft tissue metastasis.
Inset, Positron emission tomography/computed tomography (PET/CT) fused image showing fluorodeoxyglucose (FDG) avid lesion
in the intertibial/fibular soft tissue. C, AP radiograph showing a mixed lytic sclerotic ill-defined lesion of the proximal humerus with
extension into the soft tissue. Note skipped metastasis in the humeral shaft (arrows). Inset, PET/CT shows FDG-avid tumor in the
proximal humerus. Collectively the imaging data implicates the primary osteosarcoma of the left humerus with skipped metastases
in the same humeral shaft and a distant soft tissue metastasis in the right lower leg.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 309

nonneoplastic condition that precedes the development of radiodensity. Radiolucent lesions were of higher his-
of osteosarcoma.328,335,336,339,343,347,350,353,359,361,362 The tumors tologic grade and exhibited a higher recurrence rate than
are usually of high grade, and the skeletal sites most low-grade radiodense lesions. Subsequent publications
frequently involved are the pelvis, humerus, and femur, from the Mayo Clinic and others resulted in the current
in that order. The radiologic pattern is usually that of a classification of malignant osteoblastic tumors of bone
lytic destructive focus within an area of involvement by surface. The present concept recognizes the heterogene-
Paget’s disease. The prognosis for Paget-related sarcomas ity of this group in terms of their microscopic and radio-
is significantly worse than that for de novo conventional graphic features, as well as their clinical behavior. Those
high-grade osteosarcoma. features are related to the particular growth pattern of
Radiation-induced osteosarcomas may develop after both each tumor and its putative site of origin on the surface
external and internal radiation exposure. Dosage in excess of bone. The clinical behavior of these lesions correlates
of 3000 Gy and with a latency period of more than 3 years with their histologic grade of malignancy.363
is recorded in most cases, but shortening of the latency Surface osteosarcomas are divided into two main cat-
period has been noted in patients treated with chemother- egories: parosteal and periosteal. The term paraosseous
apy. The tumors are of high grade and follow an aggres- osteosarcoma is sometimes used in reference to tumors
sive course with short survival.288,302,323,338,342,344,355,357,358 arising adjacent to the bone within the soft tissue. His-
Bone infarcts rarely give rise to sarcomas, and torically the term juxtacortical osteosarcoma was used as a
most are either malignant fibrous histiocytomas or synonym for parosteal osteosarcoma. Recently, it has
fibrosarcomas.331-333 The latter form is disappearing been used in a more general sense to designate all osteo-
as a diagnostic designation as the current trend is to sarcomas growing on the surface of bone. The terms
classify virtually all high-grade spindle-cell sarcomas periosteal and parosteal osteosarcoma imply that these tumors
of bone as malignant fibrous histiocytoma rather than originate in the distinct structures of a bone surface (Fig.
fibrosarcoma. A small number of high-grade osteo- 5-97). It is postulated that periosteal osteosarcoma origi-
sarcomas have been reported in patients with bone nates in the deep layer of the periosteum or in the outer
infarcts and in patients who have had metallic prosthetic layer of the cortex. Hence, its growth is manifested by a
implants.327,334,337,340,346,348,349,351 separation and elevation of the periosteum from the
Fibrous dysplasia is associated with a very low incidence cortex. Parosteal osteosarcoma is derived from the outer
(0.4%) of secondary malignancy and most of these layer of the periosteum and grows in an exophytic
patients had prior irradiation.329,330,341,345,352,354,356 Most mushroom-like pattern without elevation of the perios-
patients are in the third and fourth decade of life, and teum or evidence of periosteal new bone formation.
osteosarcomas of high grade are the most frequent his- These tumors are typically of low to intermediate histo-
tologic type. logic grade, but sometimes they may exhibit marked ana-
Chronic osteomyelitis with sinus track formation associ- plasia and are then referred to as high-grade surface
ated with reactive squamous hyperplasia is a predispos- osteosarcomas. Occasionally, parosteal osteosarcoma may
ing factor for the development of squamous carcinoma dedifferentiate into a high-grade lesion in which low- and
associated with this condition. Rarely do sarcomas, high-grade parosteal osteosarcomas are present within
including osteosarcoma, develop in association with this the same tumor. Such tumors are referred to as dediffer-
condition. entiated parosteal osteosarcomas.
Osteosarcoma rarely complicates treatment of hemato-
logic malignancies. Typically these tumors develop after
the treatment of childhood hematologic malignancies Parosteal Osteosarcoma
such as non-Hodgkin’s lymphoma and lymphoblastic leu-
Definition
kemia. It is uncertain whether there is a real pathogenetic
relationship between chemotherapy during the first Parosteal osteosarcoma is a low-grade bone-forming
decade of life and later occurrence of osteosarcoma. It tumor that grows predominantly on the surface of
cannot be excluded that such cases represent mere coin- long bones in an exophytic pattern without elevating
cidences rather than a pathogenic relationship. the periosteum. A spindle-cell fibroblast-like compo-
nent and well-developed trabeculae of tumor bone are
characteristic.
SURFACE OSTEOSARCOMA
Incidence and Location
Osteosarcomas that originate and grow predominantly
on the surface of a bone were recognized more than 40 This rare neoplasm accounts for approximately 3% of all
years ago. Jaffe and Selin,367 Geschickter and Copeland,365 osteosarcomas. It typically occurs in skeletally mature
as well as Dwinnell, Dahlin, and Ghormley,364 recognized patients. The peak incidence is during the third and
this variety of osteosarcoma in the early 1950s and coined fourth decades of life. Approximately 20% of cases occur
the term parosteal or juxtacortical osteogenic sarcoma for during the second decade of life, and there is a clear
these tumors. Later, this group of lesions was recognized female sex predominance.
to have diverse radiographic and microscopic features Parosteal osteosarcoma has a peculiar anatomic distri-
and clinical behavior. In 1958, Jaffe366 separated them bution, with more than 80% of cases located in the distal
into low- and high-grade surface osteosarcomas and cor- portion of the femoral shaft on its posterior aspect, within
related the increasing histologic grades with the degree the superior popliteal area. The second most common

ERRNVPHGLFRVRUJ
310 5  Osteosarcoma

Cartilaginous
cap

- No elevation of
periosteum

- No new periosteal
bone
Elevated periosteum with
new bone formation

PAROSTEAL PERIOSTEAL

FIGURE 5-97  ■  Surface osteosarcomas. Diagrammatic presentation of differences in growth patterns between parosteal and periosteal
osteosarcomas.

site of involvement is in the tibia, where nearly all cases mineralized than its periphery. These features, along with
involve the proximal shaft and the proximal metaphyseal its unique ability to encircle the shaft of the affected
region. Individual cases have been reported in the bone, are best demonstrated by computed tomography
proximal humerus and other long tubular bones. Rare and MRI. In rare instances, parosteal osteosarcoma can
examples of parosteal osteosarcoma involving the acral present as a relatively radiolucent lesion with only small
skeleton and the craniofacial bones have also been irregular opacities (Fig. 5-100).
described.368,371,380,387,388 The development of parosteal As mentioned, the tumor grows on the surface of the
osteosarcoma has also been linked to previous radiation periosteum. Hence, there is no associated elevation of
exposure.386 The age distribution and most common skel- the periosteum and no radiographically recognizable
etal sites are shown in Figure 5-98. periosteal new bone formation. The unique ability of
parosteal osteosarcoma to encircle the shaft may result in
the formation of a large, heavily ossified lobulated mass
Clinical Symptoms
that forms a cuff around the bone and involves a large
These slow-growing tumors usually present as painless, segment of the shaft. The lack of penetration into the
firm, fixed masses of long duration. Pain may be present, medullary cavity may only be demonstrable through the
and the location adjacent to a joint can limit motion. use of computed tomography and MRI. The latter
There may be a history of previous biopsy and attempts methods are used predominantly to document the extent
at excision of what was thought to be a benign, reactive of bone and soft tissue involvement. They usually show
process. an intact cortex beneath the lesion and a broad area
of attachment in the central portion of the tumor
(Figs. 5-100 to 5-103).
Radiographic Imaging
The presence of a cartilaginous cap partially covering
The tumor grows on the surface of bone attached to the the tumor is usually not demonstrated on conventional
cortex along a broad base (Fig. 5-99). Peripherally, it radiographs, but MRI may render it visible. Computed
grows in a mushroom-like fashion with a space separating tomography and MRI also may reveal satellite nodules
the tumor from the underlying cortex.375,382,383,400 The within the soft tissue that are usually present in recurrent
tumor mass is typically densely mineralized and has lobu- lesions.383 The presence of lucent areas at the periphery
lated outlines. Often the mineralization is uneven and of lesions can be associated with the development
has irregularly coalesced opacities. If this occurs, the base of large, predominantly cartilaginous areas. The main
and the central portions of the tumor are usually more significance of this finding is that it may represent a sign

ERRNVPHGLFRVRUJ
5  Osteosarcoma 311

Peak
incidence
20 30
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-98  ■  Parosteal osteosarcoma. Peak age incidence and frequent sites of skeletal involvement. Most frequent site is indicated
by solid black arrow.

of dedifferentiation (i.e., transformation to a high-grade Figs. 5-100 to 5-104). The latter may form a peripheral
sarcoma).370 irregular cap. Heavily ossified areas have a tendency to
Medullary involvement is difficult to evaluate on plain coalesce at the base of the lesion centrally and to fuse with
radiographs. It is usually present in advanced stages and the outer layer of the cortex. The periphery of the mass is
is best documented by MRI and computed tomography softer and fleshier than the base. The soft tissue border of
(Fig. 5-104). Cases with medullary involvement may pres- the lesion is usually lobulated and sharply demarcated.
ent a difficult diagnostic problem centering on whether
the lesion represents a medullary well-differentiated Microscopic Findings
osteosarcoma with extension into soft tissue (Fig. 5-101).
In surface lesions, the main tumor mass is usually within This tumor is characterized by the presence of a
the soft tissue and has a relatively minor intramedullary spindle-cell fibroblastic component that is deceptively
component (Fig. 5-104).390 benign appearing and admixed with relatively well
developed tumor bone trabeculae (Figs. 5-105 and
5-106).372,384,389,393,397 The latter may coalesce and form
Gross Findings
solid bony areas. The spindle cells show minimal atypia,
Parosteal osteosarcoma typically presents as a firm, exo- and although mitoses can be found with difficulty, atypi-
phytic, polypoid, bony mass attached to the cortex of a cal forms are not present. Similar to low-grade intra-
long bone by a broad base (Fig. 5-99). The surface is medullary osteosarcoma, the tumor cells have features
lobulated or bosselated with overhanging edges. It can of fibroblasts embedded in a dense collagenous stroma.
grow along the long axis of the bone or form an encircl- Foci of cartilage are frequently found in parosteal osteo-
ing cuff. One or more satellite nodules may be found sarcoma (Figs. 5-107 and 5-108). They range in size from
completely separate from the main tumor mass or partly small ill defined areas of immature cartilaginous matrix to
attached to it at the periphery. Multiple satellite nodules large irregular areas of well developed hyaline cartilage.
are more often found in recurrent lesions. The cut surface In some instances, large irregular areas of gradual tran-
presents a tan to ivory bony mass that may contain fleshy sition between cartilage and tumor bone can be present
areas or foci of grossly recognizable hyaline cartilage (see Text continued on p. 322

ERRNVPHGLFRVRUJ
312 5  Osteosarcoma

A B

C D
FIGURE 5-99  ■  Parosteal osteosarcoma. A, Parosteal osteosarcoma of distal femoral shaft with large focus of radiolucency peripher-
ally. Such areas should be preferentially sampled to exclude possibility of dedifferentiation. B, Large bulky tumor shows peripheral
lobulation. C, Specimen radiograph of distal femoral tumor shows central attachment to cortex, overhanging edges, and cancellous
bonelike trabeculation distally. Early penetration of medullary cavity at base was associated with reactive bone formation. D, Lobu-
lated parosteal osteosarcoma of distal femur extending into intercondylar notch.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 313

A B

C D
FIGURE 5-100  ■  Parosteal osteosarcoma. A, Anteroposterior radiograph shows focally mineralized bone surface lesion in the medial
supracondylar aspect. B, Fat-saturated T2-weighted coronal magnetic resonance image showing relatively homogeneous high signal
intensity of the bone surface lesion. Note focal penetration of the underlying cortex and involvement of medullary cavity (arrow).
C, Coronally bisected resection specimen showing dense fibrous bone surface mass involving the distal medial aspect of the femur.
Note focal invasion into medullary cavity (arrow). D, Low power photomicrograph of the same tumor showing parallel arrangement
of well developed tumor bone trabeculae and low cellular bland-appearing fibrous stromal tissue. (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
314 5  Osteosarcoma

A B

C D
FIGURE 5-101  ■  Parosteal osteosarcoma. A, Anteroposterior radiograph showing sclerotic lesion encircling both the tibia and the
fibula. B, Sagittally bisected resection specimen showing dense fibrous bone surface mass encircling the tibia and invading the
underlying medullary cavity. C and D, Low power photomicrographs show various patterns of tumor osteoid forming interconnected
well developed bone trabeculae in fibroblastic stromal tissue. (C and D, ×50) (C and D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
5  Osteosarcoma 315

A B

C D
FIGURE 5-102  ■  Parosteal osteosarcoma. A, Lateral radiograph showing mineralized bone surface lesion encircling the distal femoral
metastasis. B, Fat-saturated T2-weighted sagittal magnetic resonance image with contrast of A showing a bulky tumor encircling
the distal femoral metastasis with signal enhancement and large patches of signal void. C, Gross photograph of sagittally bisected
resection specimen showing bulky tumor mass encircling the distal femoral metastasis. Note, the overall fibrous appearance of the
lesion and small cystic changes in the central portion of the posterior tumor mass. D, Low power photomicrograph shows well
mineralized coarse tumor bone trabeculae in fibrous stroma. (D, ×20) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
316 5  Osteosarcoma

A B

C D
FIGURE 5-103  ■  Parosteal osteosarcoma. A, Anteroposterior radiograph showing a sclerotic tumor mass involving the proximal
humeral metaphysis. B, Fat-saturated T2-weighted coronal magnetic resonance image showing inhomogeneous enhancement in
the tumor encircling the surface of the proximal humeral metaphysis. C, Gross photograph of coronally bisected resection specimen
showing a fleshy and fibrous tumor mass encircling the proximal humerus. D, Low power photomicrograph showing well developed
coarse bone trabeculae in fibrous stroma. (D, ×20) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
5  Osteosarcoma 317

A B

C D
FIGURE 5-104  ■  Parosteal osteosarcoma. A, Lateral plain radiograph showing heavily mineralized tumor attached to the posterior
distal aspect of the femoral bone. B, Fat-saturated T2-weighted sagittal magnetic resonance image of A showing inhomogeneous
signal enhancement in the tumor involving the posterior aspect of the distal femoral bone. Note high signal intensity in the tumor
penetrating the underlying cortex and invading the medullary cavity (arrow). C, Gross photograph of the same tumor showing sagit-
tally bisected resection specimen. Note fibrous tumor growing on the surface of posterior distal femur. The tumor penetrates
underlying cortex and invades the medullary cavity (arrow). D, Low power photomicrograph showing well developed interconnected
tumor bone trabeculae and inconspicuous fibrous stromal tissue. (D, ×20) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
318 5  Osteosarcoma

A B

C D
FIGURE 5-105  ■  Parosteal osteosarcoma: microscopic features. A, Low power photomicrography showing tumor bone trabeculae
pattern and fibroblastic stromal tissue. B-D, Higher power magnifications showing well developed bony trabeculae of various shapes
and spindle-cell fibroblastic stromal tissue. (A, ×20; B-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 319

A B

C D
FIGURE 5-106  ■  Parosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing interconnected tumor bone
trabeculae and inconspicuous, well vascularized stromal tissue. B, Higher magnification of A showing somewhat parallel arrange-
ment of tumor bone trabeculae and low cellular fibroblastic stromal tissue. C, Low power photomicrograph corresponding to a
heavily mineralized sclerotic portion of the tumor with large solid areas of well developed tumor bone. D, Advancing tumor edge
growing into the skeletal muscle. Note a continuous shell-like bone outlining the peripheral aspect of the tumor. (A, C, and D, ×20;
B, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
320 5  Osteosarcoma

A B

C D
FIGURE 5-107  ■  Parosteal osteosarcoma: microscopic features. A and B, Intermediate power views showing somewhat hypercellular
spindle-cell stromal tissue and various patterns of tumor bone formation in a low-grade parosteal osteosarcoma. C, Low
power photomicrograph showing ill-defined areas of cartilaginous differentiation in parosteal osteosarcoma. D, Higher magnification
of C showing a focus of cartilaginous differentiation. Note nuclear atypia of cartilage cells. (A, B, and D, ×100; C, ×20)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 321

A B

C D
FIGURE 5-108  ■  Parosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing coarse irregular bone tra-
beculae in fibrous stroma. Note engorged prominent vessels in the lower aspect of the photomicrograph. B, Higher power of A
showing irregular well-developed bone trabeculae and somewhat hypercellular spindle cell stromal tissue. C, Low power photomi-
crograph showing large areas of cartilaginous differentiation in parosteal osteosarcoma. D, Higher magnification of C showing
mineralized cartilage matrix and tumor cartilage cells occupying lacunar spaces. Note variability in size and nuclear atypia of cartilage
cells. (A and C, ×20; B and D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
322 5  Osteosarcoma

(Fig. 5-109). Some of those areas may form large solid phenomenon is similar to that observed in dedifferenti-
masses of gradually merging osteochondroid matrix. ated chondrosarcoma.
The cartilage contains atypical chondrocytes and such
areas do not show the orderly enchondral ossification
Differential Diagnosis
sequence found in osteochondromas. A unique feature
found in some parosteal osteosarcomas is the presence The most important factor underlying the misdiagnosis
of large cartilage caps that can be seen on radiographs of parosteal osteosarcoma is the deceptively innocent
as radiolucent areas. They represent solid areas of well appearance of the well-differentiated fibroblastic compo-
developed hyaline cartilage that have an overall archi- nent and the relative maturity of the tumor bone in this
tectural arrangement similar to cartilaginous caps seen low-grade neoplasm. Consequently, it is commonly mis-
in osteochondromas. The cap-shaped outer surface is interpreted as a benign reactive lesion. Among the condi-
oriented toward the periphery of the lesion; the more tions with which it can be confused are posttraumatic
central or baseline aspects of the cartilaginous areas may periostitis, florid reactive periostitis, juxtacortical myositis ossi-
display disorganized areas of enchondral ossification ficans, and postavulsive lesions such as cortical irregularity
similar to the base of an osteochondroma (Fig. 5-110). syndrome in children. Clinical and radiologic correlations
The cartilage cells of the cartilaginous cap reside in are essential in distinguishing these conditions, even
well developed lacunar spaces and have some columnar when the microscopic features are equivocal. Generally,
arrangement. The overall mimicry of osteochondroma the reactive surface lesions show greater radiodensity
is quite striking and biopsies containing such areas can peripherally and central lucency. This pattern is the
be confused with a benign cartilage lesion if evaluated reverse of that seen in parosteal osteosarcomas. For
without correlation to a radiographic presentation of the details of the differential diagnoses of these conditions,
lesion. In most cases, the diagnosis of parosteal osteosar- see Chapter 23.
coma is evident on radiographic imaging and radiolucent Periosteal and high-grade surface osteosarcomas must be
areas are specifically targeted for preoperative biopsies to distinguished from parosteal osteosarcoma. The radio-
rule out high-grade dedifferentiation.370 Therefore, well graphic pattern of growth of periosteal osteosarcoma is
developed cartilage areas may be present in such biop- significantly different because the tumor elevates the
sies as they typically present radiographically as radio- periosteum to produce Codman’s triangles and a fusiform
lucent lesions. At the base of the lesion centrally, an surface enlargement. In addition, the mineralization
intact cortex, to which the tumor bone trabeculae fuse, is pattern of periosteal osteosarcoma is more delicate, is
usually present. However, in some cases, erosion through feathery, and often shows perpendicular striations. His-
the cortex with tumor invasion of the medullary cavity tologically, the tumor usually contains abundant hyaline
may be present. Because parosteal osteosarcoma grows in cartilage matrix, and the areas of osteogenesis are
an exophytic fashion without elevating the periosteum, of intermediate to high grade. These tumors also tend
histologic evidence of periosteal new bone deposition is to occur in young patients. Parosteal osteosarcoma
lacking, as is the Codman’s triangle found in other forms can usually be distinguished from high-grade surface
of osteosarcoma. This tumor differs completely from the osteosarcoma on purely histologic grounds in biopsy
histologic appearance of conventional osteosarcoma, but samples, but these tumors may contain residual areas of
cellular foci of high-grade tumor may be found. Such low-grade appearance if they developed through dedif-
foci represent a progression of a low-grade fibroblastic ferentiation of parosteal osteosarcoma. Such areas can be
osteosarcoma to a high-grade sarcoma and are referred anticipated radiologically if attention is paid to localized
to as dedifferentiation.369,374,377,378,391,398,399 Radiographically, areas of low density or complete lucency in otherwise
foci of dedifferentiation are seen as radiolucent areas typical parosteal osteosarcomas.
with a mineralization pattern that differs from that of Occasionally an intramedullary low-grade osteosarcoma
low-grade parosteal osteosarcoma, which typically pres- has a significant soft tissue component that is histologi-
ents as a heavily mineralized bone surface lesion (Figs. cally indistinguishable from parosteal osteosarcoma of
5-111 to 5-113).376 On gross inspection, such areas may low grade. The distinction is made easier through com-
have fleshy gelatinous or myxoid appearance (see Figs. puted tomography and MRI, which demonstrate that the
5-111 to 5-113). In general, dedifferentiated parosteal tumor’s major component is intramedullary.
osteosarcoma represents an evolution of a preexisting Sessile osteocartilaginous exostoses are among the benign
low-grade surface tumor to a high-grade sarcomatoid lesions most frequently mistaken for parosteal osteosar-
malignancy and is seen in approximately 10% to 15% coma on radiographs, but the intact cortex beneath the
of all parosteal osteosarcomas. Foci of dedifferentiation parosteal sarcoma can be easily demonstrated in com-
most frequently have features of high-grade conven- puted tomographic and MRI studies. Similarly, when the
tional osteosarcoma. Dedifferentiation may also have lesion is an osteochondroma, continuity between the fatty
features of high-grade sarcoma with malignant fibrous marrow of the stalk and the underlying medullary bone
histiocytoma–like features. Rare examples of dedifferen- is evident. The cartilage cap, which can be found in either
tiation resembling giant cell tumors or with features of lesion, may contribute to the confusion, but the distinc-
telangiectatic osteosarcoma have been described.373,395 A tive microscopic morphology of parosteal osteosarcoma
phenotypic switch to rhabdomyosarcoma can also occur is lacking in an osteochondroma stalk.
in a dedifferentiated component.392 It can also be in the Fibromatosis and fasciitis can secondarily involve the
form of high-grade spindle-cell or pleomorphic sarcoma adjacent bone and occasionally originate from the
with malignant fibrous histiocytoma–like features. The Text continued on p. 328

ERRNVPHGLFRVRUJ
5  Osteosarcoma 323

A B

C D
FIGURE 5-109  ■  Parosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing irregular large deposits of
tumor osteoid. B, Higher magnification of A showing irregular ill-defined areas of osteoid. C, Highly variable and irregular pattern
of tumor bone trabeculae in parosteal osteosarcoma. D, Highly sclerotic area of parosteal osteosarcoma showing confluent solid
areas of osteoid gradually transitioning into chondroid matrix. (A, C, and D, ×50; B, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
324 5  Osteosarcoma

A B

C D
FIGURE 5-110  ■  Parosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing a large confluent area of
cartilaginous differentiation in parosteal osteosarcoma referred to as a cartilaginous cap. Solid area of well developed hyaline car-
tilage facing the outer peripheral surface of the tumor is in the upper portion of the photomicrograph. The lower portion shows a
disorganized pattern of enchondral ossification mimicking the growth plate. B, Outer surface of the cartilaginous cap covered by
fibrous tissue. C, Intermediate power photomicrograph showing the baselike aspect of the cartilaginous cap with a growth platelike
enchondral ossification pattern. D, Architectural features of the outer peripheral areas of the cartilaginous cap showing cartilage
cells of variegated size located in well defined lacunar spaces. (A and B, ×20; C, ×50; D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 325

A B

C D
FIGURE 5-111  ■  Dedifferentiated parosteal osteosarcoma. A, Lateral radiograph of distal femur showing heavily mineralized tumor
encircling the distal femoral surface. Note the bulky tumor with an increasing mineralization pattern toward the base of the lesion
in the posterior aspect of the femoral bone. Arrows indicate a focus with changed fluffy mineralization pattern corresponding to
dedifferentiation. B, Fat-saturated T2-weighted sagittal magnetic resonance image showing inhomogeneous signal enhancement.
Arrows indicate a focus of signal enhancement in the lower posterior aspect of the lesion corresponding to dedifferentiation.
C, Gross photograph of sagittally bisected resection specimen showing a heavily mineralized tumor involving the distal femoral
surface with the bulky tumor mass in the posterior aspect (arrows). D, Magnification of the lower posterior aspect of the tumor
showing a discrete fleshy, somewhat myxoid, area of the tumor corresponding to dedifferentiation (arrows). Note heavily mineral-
ized upper portion of the tumor with grossly visible trabeculae pattern.

ERRNVPHGLFRVRUJ
326 5  Osteosarcoma

A B

C D
FIGURE 5-112  ■  Dedifferentiated parosteal osteosarcoma. A, Lateral radiograph showing heavily mineralized tumor involving the
posterior distal aspect of the femoral surface. Note change in the mineralization pattern with a loosened fluffy area in the upper
aspect of the tumor corresponding to dedifferentiation (arrows). B, Gross photograph of sagittally bisected resection specimen
showing bulky tumor of the posterior aspect of the distal femoral surface. Note the fleshy appearance of the tumor corresponding
to dedifferentiation of the upper aspects of the lesion (arrows). C, Low power photomicrograph showing well developed intercon-
nected tumor bone trabeculae pattern and fibrous stroma corresponding to preexisting low-grade parosteal osteosarcoma.
D, Dedifferentiated area of the tumor showing high-grade osteosarcoma with lacelike osteoid and anaplastic tumor cells. (C, ×20;
D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 327

A B

C D
FIGURE 5-113  ■  Dedifferentiated parosteal osteosarcoma. A, Gross photograph of coronally bisected distal femoral resection speci-
men showing parosteal osteosarcoma growing on the medial aspect of the distal femoral surface (arrows). B, Magnified view of
A showing interface between the low-grade well mineralized upper portion of the tumor and an area of dedifferentiation with fleshy
myxoid appearance (arrows). A cystic area in the medullary cavity represents a regressed tumor after preoperative chemotherapy.
C, Low-grade component of the tumor with well developed tumor trabeculae. D, Focus of viable postchemotherapy tumor
with features of a high-grade osteosarcoma corresponding to the dedifferentiated portion of the tumor. (C, ×50; D, ×100) (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
328 5  Osteosarcoma

periosteum itself. Such lesions can present formidable Clinical Symptoms


problems in differential diagnosis from parosteal osteo-
sarcoma, particularly when reactive bone is produced as Swelling of an extremity with or without pain of relatively
in ossifying parosteal fasciitis, a rare but clearly benign short duration (several weeks to months) is characteristic
condition. The spindle-cell component usually differs for this tumor.
sufficiently from parosteal osteosarcoma to permit its
recognition. Radiographic Imaging
Periosteal osteosarcoma is a predominantly radiolucent
Treatment and Behavior
fusiform lesion that presents on the surface of a long
Parosteal osteosarcoma is a slowly enlarging tumor that bone401-404,411 (Figs. 5-115 to 5-118). The periosteum
recurs if it is not completely excised and in which areas appears to have been elevated by the tumor’s expansion
of dedifferentiation are associated with the capacity to external to the cortex, which provokes prominent perios-
metastasize to distant sites.379,381,385,394,396,398 Overall the teal new bone formation in the form of both perpendicu-
5-year survival rate ranges from 85% to 90%, and tumors lar striae and peripheral Codman’s triangles seen on
in patients in whom metastases develop almost invariably plain radiographs (Fig. 5-119). In addition to the linear
show dedifferentiation to a higher grade. On the other densities external to the cortex, patchy focal cartilage
hand, patients who had radical excision of a completely calcification may be visible. The subperiosteal location of
low-grade tumor had small risks of local recurrence and the tumor is clearly demonstrated in cross-sectional com-
metastasis. Medullary involvement occurs more fre- puted tomographic scans and MRI (see Figs. 5-115 and
quently in patients with higher-grade tumors, including 5-118). The outer layer of the cortex beneath the tumor
dedifferentiated ones; it is related to multiple recurrences may show irregularity and erosion, but the medullary
and does not, in itself, correlate with a poor prognosis. cavity is usually not invaded. Some authors use the
Its demonstration in cross-sectional radiographs can indi- absence of medullary involvement as a sine qua non for
cate the necessity for more extensive excision. Higher- the diagnosis of periosteal osteosarcoma. They argue that
grade and dedifferentiated lesions are treated with if the cortex is completely disrupted, small primary med-
chemotherapy in addition to radical surgery. ullary osteosarcomas with subperiosteal extension cannot
be ruled out.

Periosteal Osteosarcoma Gross Findings


Definition
The fusiform tumor is well demarcated and attached to
Periosteal osteosarcoma is a low- to intermediate-grade the surface of the cortex. Periosteal elevation with reac-
bone-forming sarcoma with predominantly chondroblas- tive bone spicules may be seen traversing the tumor cen-
tic differentiation that develops on the surfaces of long trally and parallel to the surface peripherally where it
bones in children. It arises beneath the periosteum, ele- attaches to the cortex (Fig. 5-118). Grossly visible carti-
vating it and provoking prominent periosteal new bone lage is frequently present, and a lobular architecture may
formation. Some authors designated these extensively be seen.
cartilaginous osteosarcomas as juxtacortical chondrosarco-
mas.417 This term is no longer acceptable for these lesions Microscopic Findings
because it confuses the entity of periosteal osteosarcoma
with a true periosteal chondrosarcoma, which is described This tumor exhibits predominant features of cartilage
in Chapter 7. differentiation that may be in the form of poorly delin-
eated lobules separated by bands of primitive sarcoma-
tous cells. In these areas, direct tumor osteoid and
Incidence and Location
immature bone production can be found (Figs. 5-120 to
Periosteal osteosarcoma is a rare tumor that represents 5-122).401-403,413,418,419 The perpendicular striations seen
less than 2% of osteosarcomas. The peak incidence is grossly are represented by spicules of reactive bone that
during the second decade of life, and the tumor occurs have prominent osteoblastic rimming. The cartilaginous
more commonly in female patients, with a 1 : 1.7 male- component usually shows obvious nuclear atypia equiva-
to-female ratio. It occurs almost exclusively on the long lent to a grade 2 or 3 chondrosarcoma (Figs. 5-123
tubular bones of a lower extremity.401-403,406,407 The tibia is to 5-125). It frequently contains areas of heavy calcifica-
most frequently involved, followed by the femur, with tion. Areas of primitive tumor bone in the undifferenti-
diaphyseal location predominating over metaphyseal. ated spindle-cell component identify this surface tumor
More rarely, the long bones of an upper extremity are as an osteosarcoma. Special techniques do not aid in
involved, and individual cases have been reported in the the diagnosis of this form of osteosarcoma as areas of
acral skeleton and craniofacial bones (the mandible).412,420 solid prolipherations of anaplastic mesenchymal cells can
Multifocal synchronous and metachronous involvement be present (Fig. 5-126). Ultrastructural study reveals
of the long bones of the lower extremities has also been undifferentiated sarcomatous cells, areas of osteoblastic
reported.408,409 The most common skeletal sites and peak differentiation with matrix mineralization, and promi-
age incidence are shown in Figure 5-114. nent cartilaginous areas.410

ERRNVPHGLFRVRUJ
5  Osteosarcoma 329

Peak
incidence
10 35
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-114  ■  Periosteal osteosarcoma. Peak age incidence and frequent sites of skeletal involvement.

Differential Diagnosis surface component is sampled without full visualization


of the intramedullary extent of the tumor. This problem
Periosteal osteosarcoma is distinguished from parosteal is easily solved through the use of computed tomography
osteosarcoma on the basis of distinct differences in loca- and MRI, which should confirm the absence of medullary
tion, age of the patient, and radiographic growth pattern. involvement in periosteal osteosarcoma.
Recognition is usually made easier by the fact that the In the past, periosteal osteosarcomas were not fully
periosteal osteosarcoma is predominantly cartilaginous accepted as bone-forming neoplasms, and some authors
and of intermediate- to high-grade differentiation. In suggested that they were better classified as juxtacortical
contrast, parosteal osteosarcomas are predominantly chondrosarcomas. In some cases the inconspicuous nature
spindle-cell (fibroblastic) tumors of low grade and contain of the tumor bone formation contributed to this contro-
abundant tumor bone. versy, but it is now generally agreed that a distinction
High-grade surface osteosarcoma presents greater diffi- can be made between these two lesions on the bases of
culties in differential diagnosis from this tumor because clinical, radiologic, and histologic findings. Juxtacortical
some periosteal osteosarcomas have high-grade differen- chondrosarcoma tends to occur in older patients, is
tiation. Although the distinction may be purely academic, usually more metaphyseal in location, and shows a differ-
periosteal osteosarcoma generally contains much more ent (coarser) pattern of matrix calcification and little per-
prominent cartilaginous differentiation and usually shows turbation of the periosteum. Histologically, it differs
a more pronounced lobular pattern than high-grade from periosteal osteosarcoma in that the cartilage is
surface osteosarcoma. The latter shows a predominantly usually very well differentiated and there is no direct
osteoblastic histologic pattern with greater anaplasia. tumor bone formation.
The frequency of metastasis is significantly greater in Whereas periosteal chondroma is usually a small, well-
high-grade surface osteosarcoma, and the prognosis defined surface lesion that is easily distinguished from the
is correspondingly poorer than that for periosteal more irregular periosteal osteosarcoma, some larger
osteosarcoma. examples of this benign cartilage tumor may suggest the
Conventional intramedullary osteosarcoma can occasion- diagnosis of periosteal osteosarcoma. The distinction is
ally present a problem in differential diagnosis when the made without difficulty on histologic grounds.

ERRNVPHGLFRVRUJ
330 5  Osteosarcoma

C
FIGURE 5-115  ■  Periosteal osteosarcoma: radiographic features. A, Anteroposterior plain radiograph of periosteal osteosarcoma
shows no matrix mineralization but fusiform soft tissue density that erodes external cortical surface. B, T1-weighted coronal magnetic
resonance image of lesion shows low signal in subperiosteal mass. Medullary cavity is not involved. C, Axial computed tomogram
of tumor shown in A and B.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 331

A B
FIGURE 5-116  ■  Periosteal osteosarcoma: radiographic features. A, Anteroposterior radiograph shows fusiform surface tumor of tibial
shaft in a 13-year-old girl. B, Close-up view of periosteal osteosarcoma of proximal tibial shaft shows radiating spicules at the base
of the fusiform mass.

Treatment and Behavior similar to conventional medullary osteosarcoma, with


most cases arising on the surface of the metaphyses. The
Radical surgical excision with wide margins and amputa- distal femur and proximal humerus are involved most
tion are the treatment methods that provide the best frequently. The peak age incidence and the most frequent
results for patients with this tumor.414,415 Chemotherapy sites of skeletal involvement are shown in Figure 5-127.
appears to have questionable effects on the development
of metastatic deposits in periosteal osteosarcoma. The Clinical Symptoms
limited clinical experience with this variant indicates that
approximately 25% of the patients die with metastatic Pain is the most common presenting symptom, in addi-
tumor, usually to the lungs, within 2 to 3 years after tion to swelling. The duration varies from months to
primary treatment.401,403,416,419 These original observations several years.
from the Mayo Clinic and Rizzoli Institute were recently
confirmed by the European Musculo Skeletal Oncology Radiographic Imaging
Society study.405
High-grade surface osteosarcoma typically presents
radiographically as a mineralized bone surface mass. The
High-grade Surface Osteosarcoma pattern of matrix mineralization tends to mimic that of a
Definition conventional medullary osteosarcoma with irregularly
distributed cloudlike opacities (Fig. 5-128). The degree
This de novo tumor is an exclusively high-grade osteo- of mineralization varies considerably. Cortical erosion
sarcoma that develops on the surface of a long bone may be present, but the medullary cavity is usually not
without medullary involvement. involved. As in periosteal osteosarcoma, Codman’s tri-
angles may be present.
Incidence and Location
Gross Findings
This is the least common variety of surface osteosarcoma,
constituting less than 1% of all osteosarcomas.421-424 As The gross features generally mimic those of a conventional
with conventional osteosarcoma, there seems to be a male medullary osteosarcoma as described previously. This
predominance. The limited experience with this tumor tumor presents as a multilobulated fungating mass on the
indicates that it principally occurs in adolescents and bone surface with a broad-based attachment. The cortex is
young adults. The skeletal distribution is somewhat usually roughened and focally deeply eroded, but any

ERRNVPHGLFRVRUJ
332 5  Osteosarcoma

C
FIGURE 5-117  ■  Periosteal osteosarcoma: radiographic features. A, Periosteal osteosarcoma of tibial shaft in skeletally immature
patient. Note Codman’s triangles at upper and lower limits of tumor. B and C, Axial and coronal T1-weighted magnetic resonance
images of tumor shown in A. Note absence of medullary involvement and irregular cortical surface. Subperiosteal location of tumor
is well demonstrated.

significant medullary penetration should exclude the tumor Differential Diagnosis


from the diagnostic category of surface osteosarcoma.
The main lesions to be distinguished from this tumor are
parosteal osteosarcoma, dedifferentiated parosteal osteosarcoma,
Microscopic Findings
periosteal osteosarcoma, and extramedullary extension from
High-grade surface osteosarcoma is indistinguishable a conventional central osteosarcoma. The prognosis for
from conventional medullary osteosarcoma because it dedifferentiated parosteal osteosarcoma and for high-
shows anaplastic cellular features and osteoid and imma- grade surface osteosarcoma is the same as that for high-
ture bone and cartilage formation without any of the grade medullary osteosarcoma. Therefore the distinction
low-grade fibroblastic features seen in parosteal osteosar- among these entities has no therapeutic implications.
coma (Fig. 5-128). If microscopic evidence of medullary The distinction from periosteal osteosarcoma is of more
extension is present, it is minimal. Text continued on p. 342

ERRNVPHGLFRVRUJ
5  Osteosarcoma 333

A B

C D
FIGURE 5-118  ■  Periosteal osteosarcoma: radiographic and gross features. A, Lateral radiograph shows radiolucent lesion of the tibial
shaft with elevation of periosteum on both sides of the lesion. B, Fat-saturated T2-weighted axial magnetic resonance image showing
periosteal signal enhancing lesion in the anterior aspect of the tibia. Note elevation of the periosteum in the peripheral aspects of
the lesion. C, Gross photographs of sagittally bisected tibial shaft resection specimen showing an oval lesion originating in the outer
layers of the cortex and elevating the overlying periosteum. D, Magnified view of C showing gritty ill-defined lesion originating in
the periosteal outer aspects of the cortex beneath the periosteum. Note the elevation of the periosteum forming a buttress-like eleva-
tion in the peripheral aspects of the lesion with a thin layer of fibrous periosteal tissues covering the entire outer surface of the
lesion.

ERRNVPHGLFRVRUJ
334 5  Osteosarcoma

A B

C D
FIGURE 5-119  ■  Periosteal osteosarcoma: gross and microscopic features. A, Bisected segmentally resected tibial shaft tumor shows
surface location and subperiosteal growth. Peripheral hemorrhagic defect represents biopsy site. B, Whole-mount photomicrograph
of periosteal osteosarcoma shows peripheral reactive bone formation at site of the Codman’s triangle (arrows). Cartilage is discern-
ible even in low power views. C, Photomicrograph of tumor in B shows atypical cartilage with calcifications in periosteal osteosar-
coma. D, Whole-mount photomicrograph of periosteal osteosarcoma with reactive periosteal new bone formation at the periphery
(arrows). (C, ×100) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 335

B
FIGURE 5-120  ■  Periosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing the overall architecture of
the lesion. The peripheral zone shows hypercellular mantle while the portion at the bottom of the photograph contains areas of
cartilaginous and osseous differentiation. Inset, Higher magnification of A showing more of the hypercellular area of the tumor.
B, Deeper area of the tumor showing areas of cartilaginous matrix gradually blending with well developed irregular areas of osteoid.
(A, ×20; B, ×50; inset, ×100) (A, B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
336 5  Osteosarcoma

A B

C D
FIGURE 5-121  ■  Periosteal osteosarcoma: microscopic features. A-D, Areas of cartilaginous and osseous differentiation with gradual
transition from cartilaginous to osteoid matrix deposition in periosteal osteosarcoma. (A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 337

A B

C D
FIGURE 5-122  ■  Periosteal osteosarcoma: microscopic features. A-D, Extensive cartilaginous differentiation gradually blending
with well developed areas of osteoid matrix and irregular trabeculae of tumor bone in periosteal osteosarcoma. (A-D, ×100)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
338 5  Osteosarcoma

B
FIGURE 5-123  ■  Periosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing peripheral aspects of
periosteal osteosarcoma with large areas of cartilaginous differentiation forming bulging lobules. B, Intermediate power
photomicrograph of cartilage matrix area gradually blending with more cellular areas of the tumor. (A, ×20; B, ×100) (A and
B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 339

B
FIGURE 5-124  ■  Periosteal osteosarcoma: microscopic features. A, Low power photomicrograph showing large areas of cartilaginous
differentiation in peripheral parts of the tumor. Note hypercellular zone at the tumor periphery in upper portion of the photomicro-
graph. B, Higher magnification of A showing the gradual transition of well developed cartilage areas with the spindle-cell component
of the tumor. (A, ×50; B, ×100) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
340 5  Osteosarcoma

FIGURE 5-125  ■  Periosteal osteosarcoma: microscopic features. A, Low power microphotograph showing myxoid areas of the tumor
with irregular focal calcifications. Note the overall lobular arrangement of the tumor. B, Tumor periphery with extensive areas of
cartilaginous differentiation and hypercellular areas. Inset, Higher magnification of B showing a hypercellular area. (A, ×20; B, ×50;
inset, ×100) (A, B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 341

A B

C D
FIGURE 5-126  ■  Periosteal osteosarcoma: microscopic features. A-D, Low and high power views of cellular solid areas of a tumor
composed of undifferentiated mesenchymal cells with nuclear atypia and brisk mitotic activity (A, ×100; B, ×200; C, ×100; D, ×200).
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
342 5  Osteosarcoma

Probable
peak incidence
20 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 5-127  ■  High-grade surface osteosarcoma. Peak age incidence and most frequent sites of skeletal involvement.

prognostic importance and is based on the fact that high- osteosarcoma is a distinctly rare, high-grade osteosar-
grade surface osteosarcoma shows more anaplasia and coma that is confined to the cortex of a major long bone.
less prominent chondroid differentiation than periosteal
osteosarcoma. The microscopic picture of periosteal Incidence and Location
osteosarcoma is typically dominated by prominent chon-
droid differentiation. Fewer than 10 cases of intracortical osteosarcoma have
been described.425-431 The age range is 10 to 30 years, with
Treatment and Behavior all reported cases diagnosed in patients who were in
the second and third decades of life. Four of six cases
High-grade surface osteosarcoma has the same prognosis occurred in the tibial diaphysis and the other two in the
as its conventional (high-grade) intramedullary counter- femoral shaft. This lesion appears to affect male more
part. This tumor has a great propensity for distant metas- often than female patients. The majority of conventional
tasis, principally to the lungs. The same combined osteosarcomas originate within the medullary cavity.
modality treatment with chemotherapy and radical surgi- Intracortical osteosarcoma represents a medical curiosity
cal procedures used for conventional intramedullary rather than a precursor or incipient form of conventional
tumors is indicated. osteosarcoma.

INTRACORTICAL OSTEOSARCOMA Clinical Symptoms


Pain, tenderness, and swelling over the affected bone of
Definition
less than 1 year’s duration are characteristic.
In 1960, Jaffe426 described a peculiar form of osteosar-
coma that originated within the cortex and at the time Radiographic Imaging
of clinical presentation was still confined to the cortical
bone. He separated his original two cases from ordinary The radiographic findings are distinct and consist of
medullary osteosarcoma and proposed the term intracor- intracortical lucency with sclerosis of the surrounding
tical osteosarcoma to designate these lesions. Intracortical bone (Fig. 5-129). The outline of the defect is somewhat

ERRNVPHGLFRVRUJ
5  Osteosarcoma 343

A B

FIGURE 5-128  ■  High-grade surface osteosarcoma: radiographic and microscopic features. A, Anteroposterior plain radiograph shows
ill-defined lesion with multiple opacities attached to proximal femoral shaft. B and C, Photomicrographs of the same case show
high-grade osteoblastic osteosarcoma (B, ×200; C, ×400). (B and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
344 5  Osteosarcoma

A B

C D
FIGURE 5-129  ■  Intracortical osteosarcoma: radiographic and microscopic features. A, Small radiolucent defect in cortex of tibia 7
months after onset of pain in a 10-year-old girl. Because of the benign appearance of this tumor, it was at first considered to be
osteoid osteoma and the lesion was treated conservatively for several months before an en bloc resection revealed high-grade
osteosarcoma. B, Low power photomicrograph shows high-grade osteoblastic osteosarcoma. C, Low power photomicrograph shows
border between high-grade osteoblastic tumor and cortical bone. D, Permeative growth of high-grade osteoblastic osteosarcoma.
(B-D, ×100) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 345

irregular. The radiologic appearance typically simulates tinguishable from a conventional high-grade osteosar-
a benign process that is typically 1.0 to 2.0 cm in diam- coma arising in bone.
eter. There is no evidence of intramedullary or soft tissue
involvement. Periosteal reactive changes are minimal or
Incidence and Location
absent.
In contrast to osteosarcomas arising in bone, extraskeletal
Gross Findings osteosarcoma is typically seen in patients older than
age 40 years and male or female predominance is
An intracortical, well-demarcated tumor with irregular uncertain.438-441 Deep muscles of the thigh and the large
borders is seen within a thickened and expanded cortex. muscles of the pelvic and shoulder girdles as well as ret-
The mass is gray to tan with a gritty consistency. roperitoneum are typical sites.432-434 High-grade osteosar-
comas were also described in breast, bladder, prostate,
and virtually every other organ. It is, in general, believed
Microscopic Findings
that many of these lesions represent a progression of
All tumors described to date have been high-grade preexisting epithelial malignancies.443 Osteosarcoma can
osteoblastic osteosarcomas with abundant tumor bone also present as a progression of preexisting soft tissue
formation and surrounding sclerosis425-431 (Fig. 5-129). malignancies such as liposarcoma.
Microscopic invasiveness is demonstrated at the borders,
with engulfed remnants of cortical bone separated by Radiographic Presentation
layers of anaplastic cells. Cartilage matrix production by
tumor cells is minimal. These tumors present as soft tissue masses with at least
focal cloudlike mineralization patterns. Typically, the
center of the lesion is more heavily mineralized than
Differential Diagnosis
the periphery. MRI discloses the heterogeneous nature
This tumor has been confused on radiographs with a of the lesion with irregular signal enhancement pattern
variety of benign conditions, such as stress fracture, osteoid and permits accurate assessment of its relationship to the
osteoma, osteoblastoma, intracortical abscess, fibrous dysplasia, surrounding normal anatomic structures such as neuro-
and nonossifying fibroma. Because of its frequent origin vascular bundles, important for planning surgical exci-
in the diaphyseal cortex of the tibia, adamantinoma of sions (Fig. 5-130).
long bones is also included among the lesions considered
radiographically. Gross Findings
Microscopically, the obvious osteoblastic nature of the
lesion excludes all possible lesions except osteoid osteoma The gross appearance of extraskeletal osteosarcoma is
and osteoblastoma. The presence of prominent nuclear similar to that of high-grade conventional osteosarcoma.
atypia and an invasive growth pattern indicates the malig- The tumors may have fleshy sarcomatoid appearance and
nant nature of this lesion. areas of necrosis and hemorrhage can be present. The
mineralization pattern can vary from discrete areas dis-
playing a gritty pattern to solid irregular masses of heavily
Treatment and Behavior
mineralized tumor bone.
The clinical and follow-up data of single case reports
on intracortical osteosarcoma were summarized by Microscopic Findings
Kyriakos.427 Half of the reported patients who were
treated initially by excisional biopsy were disease-free 3 Similar to a conventional high-grade osteosarcoma of
to 10 years after treatment. Patients treated by curettage, bone, the tumor is composed of two basic microscopic
irradiation, or both experienced recurrences locally, and components representing sarcomatous tumor cells and
two had lethal metastases. extracellular matrix, which may represent osteoid depos-
its, immature tumor bone, and cartilaginous matrix.436,437
All features described in the section on conventional
EXTRASKELETAL OSTEOSARCOMA high-grade osteosarcoma arising in bone can be present
in extraskeletal osteosarcoma (Fig. 5-130). Rarely,
Extraskeletal osteosarcoma arising in soft tissue is extraskeletal osteosarcoma may have features of well dif-
extremely rare and accounts for not more than 1% to 2% ferentiated fibroblastic osteosarcoma similar to parosteal
of all soft tissue sarcomas. The detailed description of this osteosarcoma or telangiectatic osteosarcoma.435,442
entity is beyond the scope of this textbook; the interested
reader is referred to textbooks on soft tissue sarcomas for Differential Diagnosis
a more comprehensive description of this entity.
Differential diagnosis of extraskeletal osteosarcoma
includes myositis ossificans and reactive reparative pro-
Definition
cesses that may show metaplastic bone. Differential
Extraskeletal osteosarcoma is defined as a malignant mes- diagnosis of malignant tumors include synovial sarcoma,
enchymal neoplasm that produces osteoid, immature epithelioid sarcoma, malignant fibrous histiocytoma, liposar-
bone, and chondroid matrix. It is microscopically indis- coma, and other soft tissue tumors that may contain

ERRNVPHGLFRVRUJ
346 5  Osteosarcoma

A B

C D
FIGURE 5-130  ■  Soft tissue osteosarcoma. A, Fat-saturated T2-weighted coronal magnetic resonance image (MRI) showing hetero-
geneous high signal in soft tissue mass of the upper arm. Inset, T1-weighted axial MRI showing low signal intensity in a well-
circumscribed soft tissue mass of the upper arm. B, Gross photograph of coronally bisected resection specimen showing extensively
necrotic gritty well-circumscribed tumor mass in the deep muscle of the upper arm. C, Low power photomicrograph showing irregu-
lar interconnected tumor osteoid deposits and anaplastic tumor cells consistent with high-grade osteosarcoma. D, Well developed
tumor bone trabeculae and pleomorphic tumor cells of high-grade osteosarcoma. (C, ×50; D, ×100) (C-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
5  Osteosarcoma 347

metaplastic bone or may show osteosarcoma as a sign of 19. Benjamin RS: Chemotherapy for osteosarcoma. In Unni KK,
their progression. editor: Bone tumors, New York, 1988, Churchill Livingstone.
20. Benjamin RS, Chawla SP, Carrasco CH, et al: Preoperative che-
motherapy for osteosarcoma with intravenous adriamycin and
Treatment and Behavior intra-arterial cis-platinum. Ann Oncol 2:S3–S6, 1992.
21. Benjamin RS, Patel SR: Pediatric and adult osteosarcoma: com-
Extraskeletal osteosarcomas are highly aggressive and parisons and contrasts in presentation and therapy. Cancer Treat
Res 152:355–363, 2009.
most patients develop distant metastases, typically to the 22. Bentzen SM, Poulsen HS, Kaae S, et al: Prognostic factors in
lungs within 2 to 3 years after original diagnosis. The osteosarcomas: a regression analysis. Cancer 62:194–202, 1988.
survival rates are dismal; 5-year survival rates are within 23. Berge G, Pettersen S, Grotterod I, et al: Osteopontin—an impor-
the 20% range. tant downstream effector of S100A4-mediated invasion and
metastasis. Int J Cancer 129:780–790, 2011.
24. Bertoni F, Bacchini P, Staals EL: Giant cell-rich osteosarcoma.
REFERENCES Orthopedics 26:179–181, 2003.
Intramedullary Osteosarcoma 25. Bertoni F, Bacchini P, Donati D, et al: Osteoblastoma-like osteo-
sarcoma: the Rizzoli Institute experience. Mod Pathol 6:707–716,
Conventional Osteosarcoma 1993.
1. Abramovici L, Kenan S, Hytiroglou P, et al: Osteoblastoma-like 26. Bertoni F, Present D, Bacchini P, et al: The Istituto Rizzoli experi-
osteosarcoma of the distal tibia. Skeletal Radiol 31:179–182, 2002. ence with small cell osteosarcoma. Cancer 64:2591–2599, 1989.
2. Ahmed M, Behera R, Chakraborty G, et al: Osteopontin: a poten- 27. Bertoni F, Unni KK, McLeod RA, et al: Osteosarcoma resembling
tially important therapeutic target in cancer. Expert Opin Ther osteoblastoma. Cancer 55:416–426, 1985.
Target 15:1113–1126, 2011. 28. Bielack SS, Kempf-Bielack B, Delling G, et al: Prognostic factors
3. Anderson PH, Atkins GJ, Findlay DM, et al: RNAi-mediated in high-grade osteosarcoma of the extremities or trunk: an analysis
silencing of CYP27B1 abolishes 1,25(OH)2D3 synthesis and of 1,702 patients treated on neoadjuvant cooperative osteosar-
reduces osteocalcin and CYP24 mRNA expression in human coma study group protocols. J Clin Oncol 20:776–790, 2002.
osteosarcoma (HOS) cells. J Steroid Biochem Mol Biol 103:601–605, 29. Bjornsson J, Inwards CY, Wold LE, et al: Prognostic significance
2007. of spontaneous tumor necrosis in osteosarcoma. Virchows Arch
4. Atiye J, Wolf M, Kaur S, et al: Gene amplifications in 423:195–199, 1993.
osteosarcoma-CGH microarray analysis. Gene Chromosome Cancer 30. Bonar SF, McCarthy S, Stalley P, et al: Epiphyseal osteoblastoma-
42:158–163, 2005. like osteosarcoma. Skeletal Radiol 33:46–50, 2004.
5. Ayala AG, Raymond AK, Jaffe N: The pathologist’s role in the 31. Bosing T, Roessner A, Hiddemann W, et al: Cytostatic effects in
diagnosis and treatment of osteosarcoma in children. Hum Pathol osteosarcoma as detected by flow cytometric DNA analysis after
15:258–266, 1984. preoperative chemotherapy according to the COSS 80/82 proto-
6. Ayala AG, Ro JY, Raymond AK, et al: Small cell osteosarcoma: a col. J Cancer Res Clin Oncol 113:369–375, 1987.
clinicopathologic study of 27 cases. Cancer 64:2162–2173, 1989. 32. Bragdon B, Moseychuk O, Saldanha S, et al: Bone morphogenetic
7. Bacchini P, Inwards C, Biscaglia R, et al: Chondroblastoma-like proteins: a critical review. Cellul Signal 23:609–620, 2011.
osteosarcoma. Orthopedics 22:337–339, 1999. 33. Briccoli A, Rocca M, Salone M, et al: High grade osteosarcoma
8. Bacci G, Ferrari S, Tienghi A, et al: A comparison of methods of of the extremities metastatic to the lung: long-term results in 323
loco-regional chemotherapy combined with systemic chemother- patients treated combining surgery and chemotherapy, 1985-
apy as neo-adjuvant treatment of osteosarcoma of the extremity. 2005. Surg Oncol 19:193–199, 2010.
Eur J Surg Oncol 27:98–104, 2001. 34. Bridge JA: Cytogenetic and molecular cytogenetic techniques in
9. Bacci G, Ferrari S, Delepine N, et al: Predictive factors of histo- orthopaedic surgery. J Bone Joint Surg 75A:606–614, 1993.
logic response to primary chemotherapy in osteosarcoma of the 35. Brostrom LA, Strander H, Nilsonne U: Survival in osteosarcoma
extremity: study of 272 patients preoperatively treated with high- in relation to tumor size and location. Clin Orthop 167:250–254,
dose methotrexate, doxorubicin, and cisplatin. J Clin Oncol 16: 1982.
658–663, 1998. 36. Buijs JT, Petersen M, van der Horst G, et al: Bone morphogenetic
10. Bajpai J, Sharma M, Sreenivas V, et al: VEGF expression as a proteins and its receptors; therapeutic targets in cancer progres-
prognostic marker in osteosarcoma. Pediatr Blood Cancer 53:1035– sion and bone metastasis? Curr Pharmaceut Design 16:1291–1300,
1039, 2009. 2010.
11. Baldini N, Gebhardt MC, Springfield DS, et al: Effect of preop- 37. Cao Y, Zhou Z, de Crombrugghe B, et al: Osterix, a transcription
erative chemotherapy on nuclear DNA content in osteosarcoma. factor for osteoblast differentiation, mediates antitumor activity
Chir Organi Mov 75:22–24, 1990. in murine osteosarcoma. Cancer Res 65:1124–1128, 2005.
12. Baldini N, Scotlandi K, Barbanti-Brodano G, et al: Expression of 38. Carlos-Bregni R, Contreras E, Hiraki KR, et al: Epithelioid
P-glycoprotein in high-grade osteosarcoma in relation to clinical osteosarcoma of the mandible: a rare case with unusual immuno-
outcome. N Engl J Med 333:1380–1385, 1995. profile. Oral Surg Oral Med Pathol Oral Radiol Endod 105:e47–e52,
13. Bauer HC, Kreicbergs A, Silfversward C, et al: DNA analysis in 2008.
the differential diagnosis of osteosarcoma. Cancer 61:2532–2540, 39. Carrle D, Bielack SS: Current strategies of chemotherapy in
1988. osteosarcoma. Int Orth 30:445–451, 2006.
14. Baumhoer D, Smidda J, Zillmer S, et al: Strong expression of 40. Chen X, Yang TT, Qiu XC, et al: Gene expression profiles of
CXCL12 is associated with a favorable outcome in osteosarcoma. human osteosarcoma cell sublines with different pulmonary meta-
Modern Pathol 25:522–528, 2012. static potentials. Cancer Biol Ther 11:287–292, 2011.
15. Baumhoer D, Zillmer S, Unger K, et al: MicroRNA profiling with 41. Cheon GJ, Kim MS, Lee JA, et al: Prediction model of chemo-
correlation to gene expression revealed the oncogenic miR-17-92 therapy response in osteosarcoma by 18F-FDG PET and MRI.
cluster to be up-regulated in osteosarcoma. Cancer Genet 205:212– J Nucl Med 50:1435–1440, 2009.
219, 2012. 42. Clark JC, Dass CR, Choong PF: A review of clinical and molecu-
16. Bayani J, Zielenska M, Pandita A, et al: Spectral karyotyping lar prognostic factors in osteosarcoma. J Cancer Res Clin Oncol
identifies recurrent complex rearrangements of chromosomes 8, 134:281–297, 2008.
17, and 20 in osteosarcomas. Gene Chromosome Cancer 36:7–16, 43. Clark JL, Unni KK, Dahlin DC, et al: Osteosarcoma of the jaw.
2003. Cancer 51:2311–2316, 1983.
17. Belli L, Scholl S, Livartowski A, et al: Resection of pulmonary 44. Cohen R: Metachronous sarcomas in a patient with bilateral reti-
metastases in osteosarcoma: a retrospective analysis of 44 patients. noblastomas: a case report. S Afr Med J 76:117–118, 1989.
Cancer 63:2546–2550, 1989. 45. Cozza R, Devito R, De loris MA, et al: Epithelioid osteosarcoma
18. Benedict WF, Fung YK, Murphree AL: The gene responsible for of the jaw. Pediatr Blood Cancer 52:877–879, 2009.
the development of retinoblastoma and osteosarcoma. Cancer 46. Cui J, Wang W, Li Z, et al: Epigenetic changes in osteosarcoma.
62:1691–1694, 1988. Bull Cancer 98:E62–E68, 2011.

ERRNVPHGLFRVRUJ
348 5  Osteosarcoma

47. Dahlin DC: Pathology of osteosarcoma. Clin Orthop 111:23–32, MR imaging with pathologic macroslides. Radiology 167:765–767,
1975. 1988.
48. Dahlin DC: Osteosarcoma of bone and a consideration of prog- 75. Giuliano AE, Feig S, Eilber FR: Changing metastatic patterns of
nostic variables. Cancer Treat Rep 62:189–192, 1978. osteosarcoma. Cancer 54:2160–2164, 1984.
49. Dahlin DC: Problems in the interpretation of results of treatment 76. Glass AG, Fraumeni JF, Jr: Epidemiology of bone cancer in chil-
for osteosarcoma. Mayo Clin Proc 54:621–622, 1979. (editorial). dren. J Natl Cancer Inst 44:187–199, 1970.
50. Dahlin DC, Unni KK: Osteosarcoma of bone and its important 77. Glasser DB, Lane JM, Huvos AG, et al: Survival, prognosis, and
recognizable varieties. Am J Surg Pathol 1:61–72, 1977. therapeutic response in osteogenic sarcoma. Cancer 69:698–708,
51. Debelenko LV, McGregor LM, Shivakumar BR, et al: A novel 1992.
EWSRI-CREB3LI fusion transcript in a case of small cell osteo- 78. Goidanich IF, Battaglia L, Lenzi L, et al: Osteogenic sarcoma:
sarcoma. Gene Chromosome Cancer 50:1054–1062, 2011. analysis of factors influencing prognosis in 100 cases. Clin Orthop
52. deSantos LA, Bernardino ME, Murray JA: Computed tomogra- 48:209–222, 1966.
phy in the evaluation of osteosarcoma: experience with 25 cases. 79. Guadagnolo BA, Zagars GK, Raymond AK, et al: Osteosarcoma
Am J Roentgenol 132:535–540, 1979. of the jaw-craniofacial region: outcomes after multimodality treat-
53. deSantos LA, Edeiken B: Purely lytic osteosarcoma. Skeletal Radiol ment. Cancer 115:3262–3270, 2009.
9:1–7, 1982. 80. Guo J, Reddick WE, Glass JO, et al: Dynamic contrast-enhanced
54. Dickersin GR, Rosenberg AE: The ultrastructure of small-cell magnetic resonance imaging as a prognostic factor in predicting
osteosarcoma, with a review of the light microscopy and differen- event-free and overall survival in pediatric patients with osteosar-
tial diagnosis. Hum Pathol 22:267–275, 1991. coma. Cancer 118:3776–3785, 2012.
55. Diwan RV: Hypertrophic pulmonary osteoarthropathy and its 81. Guo J, Wu G: The signaling and functions of heterodimeric bone
occurrence with pulmonary metastasis from osteogenic sarcoma: morphogenetic proteins. Cytokine Growth Factor Rev 23:61–67,
case report and review of literature. J Assoc Physicians India 26:651– 2012.
655, 1978. 82. Han G, Wang Y, Bi WZ, et al: Magnetic resonance imaging is
56. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210, appropriate for determining the osteotomy plane for appendicular
1995. osteosarcoma after neoadjuvant chemotherapy. Med Oncol 29:
57. Dryja TP, Rapaport JM, Epstein J, et al: Chromosome 13 homo- 1347–1353, 2012.
zygosity in osteosarcoma without retinoblastoma. Am J Hum 83. Hao YK, Zhang YK, Yang ZP, et al: The accuracy of magnetic
Genet 38:59–66, 1986. resonance imaging in determining the osteotomy plane in osteo-
58. Duan Z, Choy E, Harmon D, et al: MicroRNA-199a-3p is down- sarcoma. Orthopedics 31:544, 2008.
regulated in human osteosarcoma and regulates cell proliferation 84. Harting MT, Blakely ML, Jaffe N, et al: Long-term survival after
and migration. Molec Cancer Ther 10:1337–1345, 2011. aggressive resection of pulmonary metastases among children and
59. Edeiken J, Raymond AK, Ayala AG, et al: Small-cell osteosar- adolescents with osteosarcoma. J Pediatr Surg 41:194–199, 2006.
coma. Skeletal Radiol 16:621–628, 1987. 85. Hasegawa T, Hirose T, Kudo E, et al: Immunophenotypic het-
60. Eftekhari F: Imaging assessment of osteosarcoma in childhood erogeneity in osteosarcomas. Hum Pathol 22:583–590, 1991.
and adolescence: diagnosis, staging, and evaluating response to 86. Hasegawa T, Shibata T, Hirose T, et al: Osteosarcoma with epi-
chemotherapy. Cancer Treat Res 152:33–62, 2009. thelioid features: an immunohistochemical study. Arch Pathol Lab
61. Enneking WF, Kagan A: “Skip” metastases in osteosarcoma. Med 117:295–298, 1993.
Cancer 36:2192–2205, 1975. 87. Hawkins DS, Felgenhauer J, Park J, et al: Peripheral blood stem
62. Enneking WF, Kagan A, II: Transepiphyseal extension of osteo- cell support reduces the toxicity of intensive chemotherapy for
sarcoma: incidence, mechanism, and implications. Cancer 41: children and adolescents with metastatic sarcomas. Cancer 95:
1526–1537, 1978. 1354–1365, 2002.
63. Entz-Werle N, Marcellin L, Gaub MP, et al: Prognostic signifi- 88. Helio H, Karaharju E, Nordling S: Flow cytometric determina-
cance of allelic imbalance at the c-kit gene locus and c-kit over- tion of DNA content in malignant and benign bone tumors.
expression by immunohistochemistry in pediatric osteosarcoma. Cytometry 6:165–171, 1985.
J Clin Oncol 23:2248–2255, 2005. 89. Hermann G, Klein MJ, Springfield D, et al: Osteoblastoma like
64. Eyre R, Feltbower RG, James PW, et al: The epidemiology of osteosarcoma. Clin Radiol 59:105–108, 2004.
bone cancer in 0-39 year olds in northern England, 1981-2002. 90. Herzog CE: Overview of sarcomas in the adolescent and young
BMC Cancer 10:357, 2010. adult population. J Pediatr Hematol Oncol 27:215–218, 2005.
65. Eyre R, Feltbower RG, Mubwandarikwa E, et al: Epidemiology 91. Hiddemann W, Roessner A, Wormann B, et al: Tumor heteroge-
of bone tumours in children and young adults. Pediatr Blood Cancer neity in osteosarcoma as identified by flow cytometry. Cancer
53:941–952, 2009. 59:324–328, 1987.
66. Fan L, Wu Q, Xing X, et al: MicroRNA-145 targets vascular 92. Hua Y, Jia X, Sun M, et al: Plasma membrane proteomic analysis
endothelial growth factor and inhibits invasion and metastasis of of human osteosarcoma and osteoblastic cells: revealing NDRG1
osteosarcoma cells. Acta Biochim Biophys Sin 44:401–414, 2012. as a marker for osteosarcoma. Tumour Biol 32:1013–1021, 2011.
67. Ferguson RJ, Yunis EJ: The ultrastructure of human osteosar- 93. Huang J, Zhu B, Lu L, et al: The expression of novel gene URG4
coma: a study of 9 cases. Clin Orthop 131:234–246, 1978. in osteosarcoma: correlation with patients’ prognosis. Pathology
68. Flores RJ, Li Y, Yu A, et al: A systems biology approach reveals 41:149–154, 2009.
common metastatic pathways in osteosarcoma. BMC Systems Biol 94. Huvos AG: Clinicopathologic spectrum of osteogenic sarcoma:
6:50, 2012. recent observations. Pathol Annu 14:123–144, 1979.
69. Folio C, Mora MI, Zalacain M, et al: Proteomic analysis of che- 95. Huvos AG: Osteogenic sarcoma of bones and soft tissues in older
monaive pediatric osteosarcomas and corresponding normal bone persons: a clinicopathologic analysis of 117 patients older than 60
reveals multiple altered molecular targets. J Proteome Res 8:3882– years. Cancer 57:1442–1449, 1986.
3888, 2009. 96. Huvos AG: Bone tumors: diagnosis, treatment and prognosis, ed 2,
70. Foltz J, Jackson D: Osteogenic sarcoma of the mandible: a 24-year Philadelphia, 1991, WB Saunders.
follow-up study. J Oral Maxillofac Surg 40:48–51, 1982. 97. Huvos AG, Heilweil M, Bretsky SS: The pathology of malignant
71. Fraumeni JF, Jr: Stature and malignant tumor of bone in child- fibrous histiocytoma of bone: a study of 130 patients. Am J Surg
hood and adolescence. Cancer 20:967–973, 1967. Pathol 9:853–871, 1985.
72. French Bone Tumor Study Group: Age and dose of chemotherapy 98. Huvos AG, Sundaresan N, Bretsky SS, et al: Osteogenic sarcoma
as major prognostic factors in a trial of adjuvant therapy of osteo- of the skull: a clinicopathologic study of 19 patients. Cancer 56:
sarcoma combining two alternating drug combinations and early 1214–1221, 1985.
prophylactic lung irradiation. Cancer 61:1304–1311, 1988. 99. Jacobs P: Radiological diagnosis of osteosarcoma. Proc R Soc Med
73. Fu HH, Zhuang QW, He J, et al: Giant cell-rich osteosarcoma or 69:543–546, 1976.
giant cell reparative granuloma of the mandible? J Craniofac Surg 100. Jaffe HL: Tumors and tumerous conditions of the bones and joints,
22:1136–1139, 2011. Philadelphia, 1958, Lea & Febiger.
74. Gillespy T, III, Manfrini M, Ruggieri P, et al: Staging of intraos- 101. Jaffe N, Patel SR, Benjamin RS: Chemotherapy in osteosarcoma.
seous extent of osteosarcoma: correlation of preoperative CT and Basis for application and antagonism to implementation: early

ERRNVPHGLFRVRUJ
5  Osteosarcoma 349

controversies surrounding its implementation. Hematol Oncol Clin 126. Luo X, Chen J, Song WX, et al: Osteogenic BMPs promote
North Am 9:825–840, 1995. tumor growth of human osteosarcomas that harbor differentiation
102. Jones KB, Ferguson PC, Lam B, et al: Effects of neoadjuvant defects. Lab Invest 88:1264–1277, 2008.
chemotherapy on image-directed planning of surgical resection 127. Machado I, Alberghini M, Giner F, et al: Histopathological
for distal femoral osteosarcoma. J Bone Joint Surg 94A:1399–1405, characterization of small cell osteosarcoma with immunohisto-
2012. chemistry and molecular genetic support. A study of 10 cases.
103. Kansara M, Thomas DM: Molecular pathogenesis of osteosar- Histopathology 57:162–167, 2010.
coma. DNA Cell Biol 26:1–18, 2007. 128. Maire G, Martin JW, Yoshimoto M, et al: Analysis of miRNA-
104. Katenkamp D, Stiller D, Waldmann G: Ultrastructural cytology gene expression-genomic profiles reveals complex mechanisms of
of human osteosarcoma cells. Virchows Arch [A] 381:49–61, 1978. microRNA deregulation in osteosarcoma. Cancer Genet 138–146,
105. Kaveri H, Rekha K, Punnya VA: Epithelioid osteosarcoma of the 2011.
maxilla: report of an unusual case. Br J Oral Maxillofac Surg 129. Malawer MM, Dunham WK: Skip metastases in osteosarcoma:
47:143–145, 2009. recent experience. J Surg Oncol 22:236–245, 1983.
106. Korpi JT, Jagstrom J, Lehtonen N, et al: Expression of matrix 130. Mandahl N, Baldetorp B, Ferno M, et al: Comparative cytoge-
metalloproteinases-2, -8, -13, -26, and tissue inhibitors of netic and DNA flow cytometric analysis of 150 bone and soft-
metalloproteinase-1 in human osteosarcoma. Surg Oncol 20:18– tissue tumors. Int J Cancer 53:358–364, 1993.
22, 2011. 131. Mankin HJ, Connor JF, Schiller AL, et al: Grading of bone
107. Koteshwer Rao K, Pillai KG, Sripathi Rao BH, et al: Osteosar- tumors by analysis of nuclear DNA content using flow cytometry.
coma of facial bones. Int J Oral Surg 12:106–109, 1983. J Bone Joint Surg 67A:404–413, 1985.
108. Kramer K, Hicks DG, Palis J, et al: Epithelioid osteosarcoma of 132. Martin SE, Dwyer A, Kissane JM, et al: Small-cell osteosarcoma.
bone: immunocytochemical evidence suggesting divergent epithe- Cancer 50:990–996, 1982.
lial and mesenchymal differentiation in a primary osseous neo- 133. Masuda H, Miller C, Koeffler HP, et al: Rearrangement of the
plasm. Cancer 71:2977–2982, 1993. p53 gene in human osteogenic sarcomas. Proc Natl Acad Sci USA
109. Kreicbergs A, Brostrom LA, Cewrien G, et al: Cellular DNA 84:7716–7719, 1987.
content in human osteosarcoma: aspects on diagnosis and prog- 134. McKenna RJ, Schwinn CP, Soong KY, et al: Sarcomata of the
nosis. Cancer 50:2476–2481, 1982. osteogenic series (osteosarcoma, fibrosarcoma, chondrosarcoma,
110. Kreicbergs A, Silfversward C, Tribukait B: Flow DNA analysis of parosteal osteogenic sarcoma, and sarcomata arising in abnormal
primary bone tumors: relationship between cellular DNA content bone): an analysis of 552 cases. J Bone Joint Surg 48A:1–26,
and histopathologic classification. Cancer 53:129–136, 1984. 1966.
111. Kubista B, Klinglmueller F, Bilban M, et al: Microarray analysis 135. McLeod RA, Dahlin DC, Beabout JW: The spectrum of osteo-
identifies distinct gene expression profiles associated with histo- blastoma. AJR Am J Roentgenol 126:321–325, 1976.
logical subtype in human osteosarcoma. Int Ortho 35:401–411, 136. McMaster JH, Scranton PE, Jr, Drash AL: Growth and hormone
2011. control mechanisms in osteosarcoma: evidence for a new thera-
112. Kuijjer ML, Rydbeck H, Kresse SH, et al: Identification of osteo- peutic approach. Clin Orthop 106:366–376, 1975.
sarcoma driver genes by integrative analysis of copy number and 137. Meister P, Konrad E, Lob G, et al: Osteosarcoma: histological
gene expression data. Gene Chromosome Cancer 51:696–706, 2012. evaluation and grading. Arch Orthop Trauma Surg 94:91–98, 1979.
113. Kumar NL, Rosenberg AE, Raskin KA: Osteoblastoma-like 138. Mertens F, Mandahl N, Örndal C, et al: Cytogenetic findings in
osteosarcoma of the cuboid: a case report. J Orthop Surg Res 5:52, 33 osteosarcomas. Int J Cancer 55:44–50, 1993.
2010. 139. Meyerson M, Pellman D: Cancer genomes evolve by pulverizing
114. Kurek KC, Del Mare S, Salah Z, et al: Frequent attenuation of single chromosomes. Cell 144:9–10, 2011.
the WWOX tumor suppressor in osteosarcoma is associated with 140. Mirabello L, Troisi RJ, Savage SA: Osteosarcoma incidence and
increased tumorigenicity and aberrant RUNX2 expression. Cancer survival rates from 1973 to 2004: data from the Surveillance,
Res 70:5577, 5586, 2010. Epidemiology, and End Results Program. Cancer 115:1531–1543,
115. Ladanyi M, Cha C, Lewis R, et al: MDM2 gene amplification in 2009.
metastatic osteosarcoma. Cancer Res 53:16–18, 1993. 141. Mirra JM: Bone tumors: diagnosis and treatment, Philadelphia, 1980,
116. Ladanyi M, Park CK, Lewis R, et al: Sporadic amplification of the JB Lippincott.
MYC gene in human osteosarcomas. Diagn Mol Pathol 2:163–167, 142. Mirra JM, Kameda N, Rosen G, et al: Primary osteosarcoma of
1993. toe phalanx: first documented case: review of osteosarcoma of
117. Laflamme C, Curt S, Rouabhia M: Epidermal growth factor and short tubular bones. Am J Surg Pathol 12:300–307, 1988.
bone morphogenetic proteins upregulate osteoblast proliferation 143. Montanini L, Lasagna L, Barili V, et al: MicroRNA cloning and
and osteoblastic markers and inhibit bone nodule formation. Arch sequencing in osteosarcoma cell lines: differential role of miR-93.
Oral Biol 55:689–701, 2010. Cell Oncol 35:29–41, 2012.
118. Leavey PJ, Timmons C, Frawley W, et al: KAI-1 expression in 144. Mulligan LM, Matlashewski GJ, Scrable HJ, et al: Mechanisms of
pediatric high-grade osteosarcoma. Pediatr Dev Pathol 9:219–224, p53 loss in human sarcomas. Proc Natl Acad Sci USA 87:5863–
2006. 5867, 1990.
119. Lewis IJ, Nooij MA, Whelan J, et al: Improvement in histologic 145. Nagata S, Nishimura H, Uchida M, et al: Giant cell-rich osteo-
response but not survival in osteosarcoma patients treated with sarcoma of the distal femur: radiographic and magnetic resonance
intensified chemotherapy: a randomized phase III trial of the imaging findings. Radiat Med 24:228–232, 2006.
European Osteosarcoma Intergroup. JNCI 99:112–128, 2007. 146. Nakajima H, Sim FH, Bond JR, et al: Small cell osteosarcoma of
120. Li Y, Meng G, Guo QN: Changes in genomic imprinting and bone. Review of 72 cases. Cancer 79:2095–2106, 1997.
gene expression associated with transformation in a model of 147. Nathan SS, Pereira BP, Zhou YF, et al: Elevated expression of
human osteosarcoma. Exp Molec Pathol 84:234–239, 2008. Runx2 as a key parameter in the etiology of osteosarcoma. Molec
121. Lindbom Å, Söderberg G, Spjut HJ: Osteosarcoma: a review of Biol Rep 36:153–158, 2009.
96 cases. Acta Radiol 56:1–19, 1961. 148. Negrin A, Bernardini M, Diana A, et al: Giant cell osteosarcoma
122. Lindqvist C, Teppo L, Sane J, et al: Osteosarcoma of the man- in the calvarium of a cat. Vet Pathol 43:179–182, 2006.
dible: analysis of nine cases. J Oral Maxillofac Surg 44:759–764, 149. Noguera R, Navarro S, Triche TJ: Translocation (11;22) in small
1986. cell osteosarcoma. Cancer Genet Cytogenet 45:121–124, 1990.
123. Link MP, Goorin AM, Miser AW, et al: The effect of adjuvant 150. Nora FE, Unni KK, Pritchard DJ, et al: Osteosarcoma of extrag-
chemotherapy on relapse-free survival in patients with osteosar- nathic craniofacial bones. Mayo Clin Proc 58:268–272, 1983.
coma of the extremity. N Engl J Med 314:1600–1606, 1986. 151. Okada K, Hasegawa T, Yokoyama R: Rosette-forming epithelioid
124. Lisle JW, Choi JY, Horton JA, et al: Metastatic osteosarcoma gene osteosarcoma: a histologic subtype with highly aggressive clinical
expression differs in vitro and in vivo. Clin Orth Rel Res 466:2071– behavior. Hum Pathol 32:726–733, 2001.
2080, 2008. 152. Okada K, Hasegawa T, Yokoyama R, et al: Osteoarcoma with
125. Look AT, Douglass EC, Meyer WH: Clinical importance of near- cytokeratin expression: a clinicopathological study of six cases
diploid tumor stem lines in patients with osteosarcoma of an with an emphasis on differential diagnosis from metastatic cancer.
extremity. N Engl J Med 318:1567–1572, 1988. J Clin Pathol 56:742–746, 2003.

ERRNVPHGLFRVRUJ
350 5  Osteosarcoma

153. Okada K, Wold LE, Beabout JW, et al: Osteosarcoma of the 177. Schajowicz F, de Prospero JD, Cosentino E: Case report 641:
hand: a clinicopathologic study of 12 cases. Cancer 72:719–725, Chondroblastoma-like osteosarcoma. Skeletal Radiol 19:603–606,
1993. 1990.
154. Oliner JD, Kinzler KW, Meltzer PS, et al: Amplification of a gene 178. Scheffer H, Haandrikman A, Oosterhuis JW, et al: Loss of het-
encoding a p53-associated protein in human sarcomas. Nature erozygosity of chromosome 13 in osteosarcoma. Pediatr Res 20:
358:80–83, 1992. 1045, 1986. (abstract).
155. Osaki M, Takeshita F, Sugimoto Y, et al: MicroRNA-143 regu- 179. Scheffer H, Kruize YC, Osinga J, et al: Complete association of
lates human osteosarcoma metastasis by regulating matrix loss of heterozygosity of chromosomes 13 and 17 in osteosarcoma.
metalloprotease-13 expression. Molec Ther 19:1123–1130, 2011. Cancer Genet Cytogenet 53:45–55, 1991.
156. Ottaviani G, Jaffe N: The etiology of osteosarcoma. Cancer Treat 180. Scholz RB, Kabisch H, Weber B, et al: Studies of the RB1 gene
Res 152:15–32, 2009. and the p53 gene in human osteosarcomas. Pediatr Hematol Oncol
157. Ottaviani G, Jaffe N: The epidemiology of osteosarcoma. Cancer 9:125–137, 1992.
Treatment & Research 152:3–13, 2009. 181. Schrager J, Patzer RE, Mink PJ, et al: Survival outcomes of pedi-
158. Park HR, Jung WW, Bacchini P, et al: Ezrin in osteosarcoma: atric osteosarcoma and Ewing’s sarcoma: a comparison of surgery
comparison between conventional high-grade and central low- type within the SEER database, 1988-2007. J Registr Manage
grade osteosarcoma. Pathol Res Pract 202:509–515, 2006. 38:153–161, 2011.
159. Paschall HA, Paschall MM: Electron microscopic observations of 182. Schuetze SM: Chemotherapy in the management of osteosarcoma
20 human osteosarcomas. Clin Orthop 111:42–56, 1975. and Ewing’s sarcoma. J Natl Comprehens Cancer Network 5:449–
160. Patel SR, Papadopolous N, Raymond AK, et al: A phase II study 455, 2007.
of cisplatin, doxorubicin, and ifosfamide with peripheral blood 183. Schwinn CP, McKenna RJ: The biologic behavior of osteosar-
stem cell support in patients with skeletal osteosarcoma and coma. In Proceedings of the Seventh National Cancer Conference of the
variant bone tumors with a poor prognosis. Cancer 101:156–163, American Cancer Society, Philadelphia, 1973, JB Lippincott.
2004. 184. Sela J: Bone remodeling in pathologic conditions: a scanning
161. Patino-Garcia A, Zalacain M, Folio C, et al: Profiling of che- electron microscopy study. Calcif Tissue Res 23:229–234, 1977.
monaive osteosarcoma and paired-normal cells identifies EBF2 as 185. Sela J, Boyde A: Further observations on the relationship between
a mediator of osteoprotegerin inhibition to tumor necrosis factor- the matrix and the calcifying fronts in osteosarcoma. Virchows Arch
related apoptosis-inducing ligand-induced apoptosis. Clin Cancer [A] 376:175–180, 1977.
Res 15:5082–5091, 2009. 186. Sim FH, Unni KK, Beabout JW, et al: Osteosarcoma with small
162. Picci P, Bacci G, Campanacci M, et al: Histologic evaluation of cells simulating Ewing’s tumor. J Bone Joint Surg 61A:207–215,
necrosis in osteosarcoma induced by chemotherapy: regional 1979.
mapping of viable and nonviable tumor. Cancer 56:1515–1521, 187. Simon MA, Bos GD: Epiphyseal extension of metaphyseal osteo-
1985. sarcoma in skeletally immature individuals. J Bone Joint Surg
163. Price CH: Primary bone-forming tumours and their relationship 62A:195–204, 1980.
to skeletal growth. J Bone Joint Surg 40B:574–593, 1958. 188. Singh A, Morris RJ: The Yin and Yang of bone morphogenetic
164. Pritchard DJ: Surgical management of osteosarcoma. In proteins in cancer. Cytokine Growth Factor Rev 21:299–313, 2010.
Unni KK, editor: Bone tumors, New York, 1988, Churchill 189. Spanier SS, Shuster JJ, Vander Griend RA: The effect of local
Livingstone. extent of the tumor on prognosis in osteosarcoma. J Bone Joint
165. Raymond AK, Ayala AG: Specimen management after osteosar- Surg 72A:643–653, 1990.
coma chemotherapy. In Unni DD, editor: Bone tumors, New York, 190. Stea B, Cavazzana A, Kinsella TJ: Small-cell osteosarcoma: cor-
1988, Churchill Livingstone. relation of in vitro and clinical radiation response. Int J Radiat
166. Raymond AK, Chawla SP, Carrasco H, et al: Osteosarcoma che- Oncol Biol Phys 15:1233–1238, 1988.
motherapy effect: a prognostic factor. Semin Diagn Pathol 4:212– 191. Stephens PJ, Greenman CD, Fu B, et al: Massive genomic rear-
236, 1987. rangement acquired in a single catastrophic event during cancer
167. Rinaggio J, Kewitt GF, McGuff HS: Epithelioid osteosarcoma development. Cell 144:27–40, 2011.
presenting as a rapidly expanding maxillary mass. Head Neck 192. Sugawara M, Kato N, Tsuchiya T, et al: RUNX2 expression in
29:705–709, 2007. developing human bones and various bone tumors. Pathol Int
168. Roessner A, Immenkamp M, Hiddemann W, et al: Case report 61:565–571, 2011.
331: small cell osteosarcoma of the tibia with diffuse metastatic 193. Sundaram M, McGuire MH, Herbold DR: Magnetic resonance
disease. Skeletal Radiol 14:216–225, 1985. imaging of osteosarcoma. Skeletal Radiol 16:23–29, 1987.
169. Ryan J, Baker L, Benjamin R, et al: Long-term follow-up in the 194. Tani T, Okada K, Shoji K, et al: Osteoblastoma-like osteosar-
cure of osteogenic sarcoma. The Southwest Oncology Group. coma. Skeletal Radiol 29:656–659, 2000.
Chir Organi Mov 75:48–49, 1990. 195. Taylor WF, Ivins JC, Dahlin DC, et al: Trends and variability in
170. Sabourin SM, Jayashankar A, Mullins ME: Imaging of osteosar- survival from osteosarcoma. Mayo Clin Proc 53:695–700, 1978.
coma after irradiation: self-assessment module. AJR 191:S28–S30, 196. Taylor WF, Ivins JC, Pritchard DJ, et al: Trends and variability in
2008. survival among patients with osteosarcoma: a 7-year update. Mayo
171. Sadikovic B, Yoshimoto M, Al-Romaih K, et al: In vitro analysis Clin Proc 60:91–104, 1985.
of integrated global high-resolution DNA methylation profiling 197. Taylor WF, Ivins JC, Unni KK, et al: Prognostic variables in
with genomic imbalance and gene expression in osteosarcoma. osteosarcoma: a multi-institutional study. J Natl Cancer Inst 81:21–
PLoS ONE 3:e2834, 2008. 30, 1989.
172. Sadikovic B, Yoshimoto M, Chilton-MacNeill S, et al: Identifica- 198. Thawani JP, Wang AC, Than KD, et al: Bone morphogenetic
tion of interactive networks of gene expression associated with proteins and cancer: review of the literature. Neurosurg 66:233–
osteosarcoma oncogenesis by integrated molecular profiling. Hum 246, 2010.
Mol Genet 18:1962–1975, 2009. 199. Tigani D, Pignatti G, Picci P, et al: Vertebral osteosarcoma. Ital
173. Sadikovic B, Thorner P, Chilton-Macneill S, et al: Expression J Orthop Traumatol 14:5–13, 1988.
analysis of genes associated with human osteosarcoma tumors 200. Toguchida J, Yamaguchi T, Dayton SH, et al: Prevalence and
shows correlation of RUNX2 overexpression with poor response spectrum of germline mutations of the p53 gene among patients
to chemotherapy. BMC Cancer 10:202, 2010. with sarcoma. N Engl J Med 326:1301–1308, 1992.
174. Sampo MM, Tarkkanen M, Kivioja AH, et al: Osteosarcoma in 201. Toguchida J, Yamaguchi T, Ritchie B, et al: Mutation spectrum of
Finland from 1971 through 1990: a nationwide study of epidemi- the p53 gene in bone and soft tissue sarcomas. Cancer Res 52:6194–
ology and outcome. Acta Orthop 79:861–866, 2008. 6199, 1992.
175. Sampo M, Koivikko M, Taskinen M, et al: Incidence, epidemiol- 202. Tohmonda T, Miyauchi Y, Ghosh R, et al: The IRE1-XBP1
ogy and treatment results of osteosarcoma in Finland—a nation- pathway is essential for osteoblast differentiation through promot-
wide population-based study. Acta Oncol (Madr) 50:1206–1214, ing transcription of Osterix. EMBO Rep 12:451–457, 2011.
2011. 203. Toledo SR, Zago MA, Oliveira ID, et al: Insights on PRAME and
176. Sato K, Yamamura S, Iwata H, et al: Giant cell-rich osteosarcoma: osteosarcoma by means of gene expression profiling. J Ortho Sci
a case report. Nagoya J Med Sci 59:151–157, 1996. 16:458–466, 2011.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 351

204. Trougakos IP, Chondrogianni N, Amarantos I, et al: Genome- 231. Gomes H, Menanteau B, Gaillard D, et al: Telangiectatic osteo-
wide transcriptome profile of the human osteosarcoma Sa OS and sarcoma. Pediatr Radiol 16:140–143, 1986.
U-2 OS cell lines. Cancer Genet Cytogenet 196:109–118, 2010. 232. Hussain MI, Al-Akeely MH, Alam MK, et al: Extraskeletal osteo-
205. Tsuneyoshi M, Dorfman HD: Epiphyseal osteosarcoma: distin- sarcoma, telangiectatic variant arising from the small bowel mes-
guishing features from clear cell chondrosarcoma, chondroblas- entery. Saudi Med J 32:958–961, 2011.
toma, and epiphyseal enchondroma. Hum Pathol 18:644–651, 233. Huvos AG, Rosen G, Bretsky SS, et al: Telangiectatic osteogenic
1987. sarcoma: a clinicopathologic study of 124 patients. Cancer 49:
206. Uhl M, Saueressig U, van Buiren M, et al: Osteosarcoma: prelimi- 1679–1689, 1982.
nary results of in vivo assessment of tumor necrosis after 234. Kaufman RA, Towbin RB: Telangiectatic osteosarcoma simulating
chemotherapy with diffusion- and perfusion-weighted magnetic the appearance of an aneurysmal bone cyst. Pediatr Radiol 11:102–
resonance imaging. Investig Radiol 41:618–623, 2006. 104, 1981.
207. Unni KK: Dahlin’s bone tumors: general aspects and data on 11,087 235. Klein MJ: Parosteal osteosarcoma dedifferentiating into telangi-
cases, ed 5, Philadelphia, 1996, Lippincott-Raven. ectatic osteosarcoma: importance of lytic changes and fluid cavi-
208. Unni KK, Dahlin DC: Osteosarcoma: pathology and classifica- ties at imaging. Skeletal Radiol 39:69, 2010.
tion. Semin Roentgenol 24:143–152, 1989. 236. Kyriakos M, Hardy D: Malignant transformation of aneurysmal
209. Uribe-Botero G, Russell WO, Sutow WW, et al: Primary osteo- bone cyst, with an analysis of the literature. Cancer 68:1770–1780,
sarcoma of bone: clinicopathologic investigation of 243 cases, with 1991.
necropsy studies in 54. Am J Clin Pathol 67:427–435, 1977. 237. Larsson SE, Lorentzon R, Boquist L: Telangiectatic osteosar-
210. Vaquero J, Carrillo R, Leunda G, et al: Primary osteogenic coma. Acta Orthop Scand 49:589–594, 1978.
sarcoma of the skull. Acta Neurochir (Wien) 51:97–104, 1979. 238. Lee KH, Joo JK, Kim DY, et al: Mesenteric extraskeletal osteo-
211. Verma RK, Gupta G, Bal A, et al: Primary giant cell rich osteo- sarcoma with telangiectatic features: a case report. BMC Cancer
sarcoma of maxilla: an unusual case report. J Maxillofac Oral Surg 7:82, 2007.
10:159–162, 2011. 239. Matsuno T, Unni KK, McLeod RA, et al: Telangiectatic osteo-
212. von Hochstetter AR: Spontaneous necrosis in osteosarcomas. Vir- genic sarcoma. Cancer 38:2538–2547, 1976.
chows Arch [A] 417:5–8, 1990. 240. Mukherjee P, Kalish LH, Delprado W, et al: Telangiectatic osteo-
213. Weber GF: The cancer biomarker osteopontin: combination with sarcomatous differentiation in a phyllodes tumour. ANZ J Surg
other markers. Cancer Genom Proteom 8:263–288, 2011. 74:707–709, 2004.
214. Weinfeld MS, Dudley HR, Jr: Osteogenic sarcoma: a follow-up 241. Murphey MD, wan Jaovisidha S, Temple HT, et al: Telangiectatic
study of the 94 cases observed at the Massachusetts General Hos- osteosarcoma: radiologic-pathologic comparison. Radiology 229:
pital from 1920 to 1960. J Bone Joint Surg 44A:269–276, 1962. 545–553, 2003.
215. Weingrad DN, Rosenberg SA: Early lymphatic spread of osteo- 242. Okada K, Hasegawa T, Tateishi U, et al: Dedifferentiated chon-
genic and soft-tissue sarcomas. Surgery 84:231–240, 1978. drosarcoma with telangiectatic osteosarcoma-like features. J Clin
216. Williams AH, Schwinn CP, Parker JW: The ultrastructure of Pathol 59:1200–1202, 2006.
osteosarcoma: a review of 20 cases. Cancer 37:1293–1301, 1976. 243. Park YK, Joo M: Multicentric telangiectatic osteosarcoma. Pathol
217. Wittrant Y, Lamoureux F, Mori K, et al: RANKL directly induces Int 51:200–203, 2001.
bone morphogenetic protein-2 expression in RANK-expressing 244. Radhi JM, Loewy J: Dedifferentiated chondrosarcoma with
POS-1 osteosarcoma cells. Int J Oncol 28:261–269, 2006. features of telangiectatic osteosarcoma. Pathology 31:428–430,
218. Won KY, Park HR, Park YK: Prognostic implication of immuno- 1999.
histochemical Runx2 expression in osteosarcoma. Tumori 95:311– 245. Roessner A, Hobik HP, Immenkamp M, et al: Ultrastructure of
316, 2009. telangiectatic osteosarcoma. J Cancer Res Clin Oncol 95:197–207,
219. Yamaguchi T, Toguchida J, Yamamuro T, et al: Allelotype analysis 1979.
in osteosarcomas: frequent allele loss on 3q, 13q, 17p, and 18q. 246. Rosen G, Huvos AG, Marcove R, et al: Telangiectatic osteogenic
Cancer Res 52:2419–2423, 1992. sarcoma: improved survival with combination chemotherapy. Clin
220. Yalcin B, Gedikoglu G, Kutluk T, et al: C-erbB-2 expression and Orthop 207:164–173, 1986.
prognostic significance in osteosarcoma. Pediatr Blood Cancer 51: 247. Ruiter DJ, Cornelisse CJ, van Rijssel TG, et al: Aneurysmal bone
222–227, 2008. cyst and telangiectatic osteosarcoma: a histopathological and mor-
221. Yang J, Yang D, Sun Y, et al: Genetic amplification of the vascular phometric study. Virchows Arch [A] 373:311–325, 1977.
endothelial growth factor (VEGF) pathway genes, including 248. Saito T, Oda Y, Kawaguchi K, et al: Five-year evolution of a tel-
VEGFA, in human osteosarcoma. Cancer 117:4925–4938, 2011. angiectatic osteosarcoma initially managed as an aneurysmal bone
222. Yoshida H, Yumoto T, Adachi H, et al: Osteosarcoma with cyst. Skeletal Radiol 34:290–294, 2005.
prominent epithelioid features. Acta Pathol Jpn 39:439–445, 249. Sangle NA, Layfield LJ: Telangiectatic osteosarcoma. Arch Pathol
1989. Lab Med 136:572–576, 2012.
223. Ziyan W, Shuhua Y, Xiufang W, et al: MicroRNA-21 is involved 250. Sirikulchayanonta V, Jaovisidha S: Soft tissue telangiectatic osteo-
in osteosarcoma cell invasion and migration. Med Oncol 28:1469– sarcoma in a young patient: imaging and immunostains. Skeletal
1474, 2011. Radiol 34:295–298, 2005.
224. Zou C, Shen J, Tang Q, et al: Cancer-testis antigens expressed in 251. Vanel D, Tcheng S, Contesso G, et al: The radiological appear-
osteosarcoma identified by gene microarray correlate with a poor ances of telangiectatic osteosarcoma: a study of 14 cases. Skeletal
patient prognosis. Cancer 118:1845–1855, 2012. Radiol 16:196–200, 1987.
252. Weiss A, Khoury JD, Hoffer FA, et al: Telangiectatic osteosar-
Telangiectatic Osteosarcoma coma: the St. Jude Children’s Research Hospital’s experience.
225. Bacci G, Ferrari S, Ruggieri P, et al: Telangiectatic osteosarcoma Cancer 109:1627–1637, 2007.
of the extremity: neoadjuvant chemotherapy in 24 cases. Acta 253. Wines A, Bonar F, Lam P, et al: Telangiectatic dedifferen-
Orthop Scand 72:167–172, 2001. tiation of a parosteal osteosarcoma. Skeletal Radiol 29:597–600,
226. Bertoni F, Bacchini P, Pignatti G, et al: Telangiectatic osteosar- 2000.
coma: survival compared to that of “conventional” osteosarcoma.
Mod Pathol 1:10A, 1988. (abstract). Well-differentiated Intramedullary Osteosarcoma
227. Chan CW, Kung TM, Ma L: Telangiectatic osteosarcoma of the 254. Arkader A, Huvos AG, Healey JH: Multifocal synchronous low-
mandible. Cancer 58:2110–2115, 1986. grade intramedullary and juxtacortical osteogenic sarcoma. Clin
228. Chawla SP, Benjamin RS: Effectiveness of chemotherapy in the Orthop Relat Res 465:265–270, 2007.
management of metastatic telangiectatic osteosarcoma. Am J Clin 255. Bertoni F, Bacchini P, Fabbri N, et al: Osteosarcoma: low-grade
Oncol 11:177–180, 1988. intraosseous-type osteosarcoma, histologically resembling paros-
229. Chowdhury K, Bachynski B, Alport EC: Telangiectatic osteosar- teal osteosarcoma, fibrous dysplasia, and desmoplastic fibroma.
coma: unusual behavior. Can J Surg 29:29–31, 1986. Cancer 71:338–345, 1993.
230. Discepola F, Powell TI, Nahal A: Telangiectatic osteosarcoma: 256. Ellis JH, Siegel CL, Martel W, et al: Radiologic features of well-
radiologic and pathologic findings. Radiographics 29(2):380–383, differentiated osteosarcoma. AJR AmJ Roentgenol 151:739–742,
2009. 1988.

ERRNVPHGLFRVRUJ
352 5  Osteosarcoma

257. Iemoto Y, Ushigome S, Fukunaga M, et al: Case report 679: 284. Hillman A, Ozaki T, Winkelmann W: Familial occurrence of
central low-grade osteosarcoma with foci of dedifferentiation. osteosarcoma. A case report and review of the literature. J Cancer
Skeletal Radiol 20:379–382, 1991. Res Clin Oncol 126:497–502, 2000.
258. Kurt AM, Unni KK, McLeod RA, et al: Low-grade intraosseous 285. Hovig E, Andreassen A, Fangan BM, et al: A TP53 mutation
osteosarcoma. Cancer 65:1418–1428, 1990. detected in cells established from an osteosarcoma, but not in the
259. Lodwick GS: Case report 169: low-grade osteosarcoma of the retinoblastoma of a patient with bilateral retinoblastoma and mul-
tibia. Skeletal Radiol 7:139–141, 1981. tiple primary osteosarcomas. Cancer Genet Cytogenet 64:178–182,
260. Sailor J, McCarthy EF, Weber KL, et al: Low-grade intramedul- 1992.
lary osteosarcoma of the tibia presenting as an intracortical lesion. 286. Iwakuma T, Lozano G, Flores ER: Li-Fraumeni syndrome: a p53
Orthopedics 31(7):714, 2008. family affair. Cell Cycle 4:865–867, 2005.
261. Schwab JH, Antonescu CR, Athanasian EA, et al: A comparison 287. Jaffe N, Pearson P, Yasko AW, et al: Single and multiple meta-
of intramedullary and juxtacortical low-grade osteogenic sarcoma. chronous osteosarcoma tumors after therapy. Cancer 98:2457–
Clin Orthop Relat Res 466:1318–1322, 2008. 2466, 2003.
262. Sundaram M, Herbold DR, McGuire MH: Case report 370: low- 288. Koshy M, Paulino AC, Mai WY, et al: Radiation-induced osteo-
grade (well-differentiated) intramedullary osteosarcoma. Skeletal sarcomas in the pediatric population. Int J Radiation Oncol Biol
Radiol 15:338–342, 1986. Physics 63:1169–1174, 2005.
263. Unni KK: Case report 136: central low-grade osteosarcoma of 289. Kozlowski K, Scougall JS, Oates RK: Osteosarcoma in a boy with
tibia. Skeletal Radiol 6:65–67, 1981. Rothmund-Thomson syndrome. Pediatr Radiol 10:42–44, 1980.
264. Unni KK, Dahlin DC, McLeod RA, et al: Intraosseous well- 290. Kunze B, Burkle S, Kluba T: Multifocal osteosarcoma in child-
differentiated osteosarcoma. Cancer 40:1337–1347, 1977. hood. Chi Organi Mov 93:27–31, 2009.
265. Xipell JM, Rush J: Case report 340: well-differentiated intraosse- 291. Larizza L, Magnani I, Roversi G: Rothmund-Thomson syndrome
ous osteosarcoma of the left femur. Skeletal Radiol 14:312–316, and RECQL4 defect: splitting and lumping. Cancer Lett 232:107–
1985. 120, 2006.
292. Lee HJ, Kim IO, Kim WS, et al: Metachronous multifocal osteo-
Osteosarcoma Associated with Specific Clinical Syndromes sarcoma: a case report and literature review. Clin Imaging 26:63–
266. Abramson DH, Ellsworth RM, Kitchin FD, et al: Second nonocu- 68, 2002.
lar tumors in retinoblastoma survivors: are they radiation induced? 293. Li FP, Fraumeni JF, Jr: Soft tissue sarcomas, breast cancer, and
Ophthalmology 91:1351–1355, 1984. other neoplasms: a familial syndrome? Ann Intern Med 71:747–
267. Amstutz HC: Multiple osteogenic sarcomata: metastatic or mul- 752, 1969.
ticentric? Report of two cases and review of the literature. Cancer 294. Li FP, Fraumeni JF, Jr: Prospective study of a family cancer syn-
24:923–931, 1969. drome. JAMA 247:2692–2694, 1982.
268. Bacci G, Fabbri N, Balladelli A, et al: Treatment and prognosis 295. Li FP, Fraumeni JF, Jr, Mulvihill JJ, et al: A cancer family syn-
for synchronous multifocal osteosarcoma in 42 patients. J Bone- drome in 24 kindreds. Cancer Res 48:5358–5362, 1988.
Joint Surg Br 88:1071–1075, 2006. 296. Longhi A, Benassi MS, Molendini L, et al: Osteosarcoma in blood
269. Bachinski LL, Olufemia SE, Zhou X, et al: Genetic mapping of a relatives. Oncol Rep 8:131–136, 2001.
third Li-Fraumeni syndrome predisposition locus to human chro- 297. Longhi A, Fabbri N, Donati D, et al: Neoadjuvant chemotherapy
mosome 1q23. Cancer Res 65:427–431, 2005. for patients with synchronous multifocal osteosarcoma: results in
270. Bhutani M, Pathak AK, Sengar M, et al: Multifocal osteosarcoma eleven cases. J Chemother 13:324–330, 2001.
involving unusual sites. Cancer Investig 24:278–282, 2006. 298. MacCarthy A, Bayne AM, Draper GJ, et al: Non-ocular tumours
271. Boyle JM, Spreadborough A, Greaves MJ, et al: Chromosome following retinoblastoma in Great Britain 1951 to 2004. Br J
instability in fibroblasts derived from Li-Fraumeni syndrome Ophthal 93:1159–1162, 2009.
families without TP53 mutations. Br J Cancer 83:1136–1138, 299. Mahajan S, Juneja M, George T: Osteosarcoma as a second neo-
2000. plasm after chemotherapeutic treatment of hereditary retinoblas-
272. Brown LT, Sexsmith E, Malkin D: Identification of a novel PTEN toma: a case report. Quintessence Int 39:439–445, 2008.
intronic deletion in Li-Fraumeni syndrome and its effect on RNA 300. Mahoney JP, Spanier SS, Morris JL: Multifocal osteosarcoma: a
processing. Cancer Genet Cytogenet 123:65–68, 2000. case report with review of the literature. Cancer 44:1897–1907,
273. Chan LL, Czerniak BA, Ginsberg LE: Radiation-induced osteo- 1979.
sarcoma after bilateral childhood retinoblastoma. AJR 174:1288, 301. Matsumoto H, Kobayashi O, Tamura K, et al: Novel germline
2000. mutation in the RB1 gene with multifocal bone tumors following
274. Chauveinc L, Mosseri V, Quintana E, et al: Osteosarcoma follow- retinoblastoma. Pediatr Int 45:728–730, 2003.
ing retinoblastoma: age at onset and latency period. Ophthal Genet 302. Meadows AT, Strong LC, Li F, et al: Bone sarcoma as a second
22:77–88, 2001. malignant neoplasm in children: influence of radiation and genetic
275. Chin KR, Mankin HJ, Gebhardt MC: Primary osteosarcoma of predisposition for the Late Effects Study Group. Cancer 46:2603–
the distal femur in two consecutive brothers. Clin Orthop Rel Res 2606, 1980.
382:191–196, 2001. 303. Mehollin-Ray AR, Kozinetz CA, Schlesinger AE, et al: Radio-
276. Colyer RA: Osteogenic sarcoma in siblings. Johns Hopkins Med J graphic abnormalities in Rothmund-Thomson syndrome and
145:131–135, 1979. genotype-phenotype correlation with RECQL4 mutation status.
277. Der kinderen DJ, Koten JW, Wolterbeek R, et al: Nonocular AJR Am J Roentgenol 191:W62–W66, 2008.
cancer in hereditary retinoblastoma survivors and relatives. 304. Miller CW, McLaughlin RE: Osteosarcoma in siblings: report of
Ophthal Paediatr Genet 8:23–25, 1987. two cases. J Bone Joint Surg 59A:261–262, 1977.
278. Dick DC, Morley WN, Watson JT: Rothmund-Thomson syn- 305. Moule RN, Jhavar SG, Eeles RA: Genotype phenotype correla-
drome and osteogenic sarcoma. Clin Exp Dermatol 7:119–123, tion in Li-Fraumeni syndrome kindreds and its implications for
1983. management. Fam Cancer 5:129–133, 2006.
279. Draper GJ, Sanders BM, Kingston JE: Second primary neoplasms 306. Mulvihill JJ, Gralnick HR, Whang-Peng J, et al: Multiple child-
in patients with retinoblastoma. Br J Cancer 53:661–671, 1986. hood osteosarcomas in an American Indian family with erythroid
280. Fitzgerald RH, Jr, Dahlin DC, Sim FH: Multiple metachronous macrocytosis and skeletal anomalies. Cancer 40:3115–3122, 1977.
osteogenic sarcoma: report of 12 cases with 2 long term survivors. 307. Olsen JH: Osteogenic sarcoma in siblings. Ugeskr Laeger 145:
J Bone Joint Surg 55A:595–605, 1973. 1065–1066, 1983.
281. Gilman PA, Wang N, Fan SF, et al: Familial osteosarcoma associ- 308. Ottaviani G, Jaffe N: Clinical and pathologic study of two siblings
ated with 13;14 chromosomal rearrangement. Cancer Genet Cyto- with osteosarcoma. Med Pediatr Oncol 38:62–64, 2002.
genet 17:123–132, 1985. 309. Palmero EI, Achatz MI, Ashton-Prolla P, et al: Tumor protein 53
282. Hansen MF, Koufos A, Gallie BL, et al: Osteosarcoma and reti- mutations and inherited cancer: beyond Li-Fraumeni syndrome.
noblastoma: a shared chromosomal mechanism revealing recessive Curr Opin Oncol 22:64–69, 2010.
predisposition. Proc Natl Acad Sci USA 82:6216–6220, 1985. 310. Parham DM, Pratt CB, Parvey LS, et al: Childhood multifocal
283. Harmon TP, Morton KS: Osteogenic sarcoma in four siblings. osteosarcoma: clinicopathologic and radiologic correlates. Cancer
J Bone Joint Surg Br 48:493–498, 1966. 55:2653–2658, 1985.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 353

311. Reissmann PT, Simon MA, Lee WH, et al: Studies of the retino- 336. Haibach H, Farrell C, Dittrich FJ: Neoplasms arising in Paget’s
blastoma gene in human sarcomas. Oncogene 4:839–843, 1989. disease of bone: a study of 82 cases. J Clin Pathol 83:594–600,
312. Ruijs MW, Verhoef S, Rookus MA, et al: TP53 germline mutation 1985.
testing in 180 families suspected of Li-Fraumeni syndrome: muta- 337. Hamblen DL, Carter RL: Sarcoma and joint replacement. J Bone
tion detection rate and relative frequency of cancers in different Joint Surg Br 66:625–627, 1984.
familial phenotpyes. J Med Genet 47:421–428, 2010. 338. Hamre MR, Severson RK, Chuba P, et al: Osteosarcoma as a
313. Sato H, Hayashi N, Yamamoto H, et al: Synchronous multifocal second malignant neoplasm. Radiother Oncol 65:153–157, 2002.
osteosarcoma involving the skull presenting with intracranial 339. Hansen MF, Seton M, Merchant A: Osteosarcoma in Paget’s
hemorrhage: case report. Neurol Med-Chir 50:407–409, 2010. disease of bone. J Bone Mineral Res 2:P58–P63, 2006.
314. Schey WL, Pinsky SM, Post M: Bone tumors in identical twins. 340. Heater K, Collins PA: Osteosarcoma in association with infarction
Clin Nucl Med 2:82–86, 1977. of bone. J Bone Joint Surg 69A:300–302, 1987.
315. Schimke RN, Lowman JT, Cowan AB: Retinoblastoma and osteo- 341. Hoshi M, Matsumoto S, Manabe J, et al: Malignant change
genic sarcoma in siblings. Cancer 34:2077–2079, 1974. secondary to fibrous dysplasia. Int J Clin Oncol 11:229–235,
316. Smith JW, Ali K, Caces JN, et al: Familial cancer: the occurrence 2006.
of bone cancer in male members of a family in multiple genera- 342. Hutchison GB: Late neoplastic changes following medical irradia-
tions. Clin Res 28:869A, 1980. [abstract]. tion. Radiol 105:645–652, 1972.
317. Srivastava S, Zou ZQ, Pirollo K, et al: Germline transmission of 343. Huvos AG, Butler A, Bretsky SS: Osteogenic sarcoma associated
a mutated p53 gene in a cancer-prone family with Li-Fraumeni with Paget’s disease of bone: a clinicopathologic study of 65
syndrome. Nature 348:747–749, 1990. patients. Cancer 52:1489–1495, 1983.
318. Suhasini AN, Brosh RM, Jr: Disease-causing missense mutations 344. Kalra S, Grimer RJ, Spooner D, et al: Radiation-induced sarco-
in human DNA helicase disorders. Mutat Res 2012 [Epub ahead mas of bone: factors that affect outcome. J Bone Joint Surg Br
of print]. 89:808–813, 2007.
319. Suhasini AN, Brosh RM, Jr: DNA helicases associated with 345. Kaushik S, Smoker WR, Frable WJ: Malignant transformation of
genetic instability, cancer, and aging. Adv Exp Med Biol 767:123– fibrous dysplasia into chondroblastic osteosarcoma. Skel Radiol
144, 2013. 31:103–106, 2002.
320. Tainsky MA, Bischoff FZ, Strong LC: Genomic instability due to 346. Keel SB, Jaffe KA, Petur NG, et al: Orthopaedic implant-related
germline p53 mutations drives preneoplastic progression toward sarcoma: a study of twelve cases. Mod Pathol 14:969–977, 2001.
cancer in human cells. Cancer Metastasis Rev 14:43–48, 1995. 347. Langston AL, Ralston SH: Management of Paget’s disease of
321. Toguchida J, Ishizaki K, Sasaki MS, et al: Chromosomal reorga- bone. Rheumatology 43:955–959, 2004.
nization for the expression of recessive mutation of retinoblastoma 348. Martin A, Bauer TW, Manley MT, et al: Osteosarcoma at the site
susceptibility gene in the development of osteosarcoma. Cancer of total hip replacement: a case report. J Bone Joint Surg 70A:1561–
Res 48:3939–3943, 1988. 1567, 1988.
322. Tsai MH, Yang CP, Jaing TH, et al: Synchronous multifocal 349. Mirra JM, Bullough PG, Marcove RC, et al: Malignant fibrous
osteosarcoma: report of one case. Acta Paediatr Taiwanica 47:146– histiocytoma and osteosarcoma in association with bone infarcts:
149, 2006. report of four cases, two in caisson workers. J Bone Joint Surg Am
323. Tucker MA, D’Angio GJ, Boice JR, et al: Bone sarcomas linked 56:932–940, 1974.
to radiotherapy and chemotherapy in children. N Engl J Med 350. Naot D: Paget’s disease of bone: an update. Curr Opin Endorcinol
317:588–593, 1987. Diabetes Obesity 18:352–358, 2011.
324. Upton B, Chu Q, Li BD: Li-Fraumeni syndrome: the genetics 351. Penman HG, Ring PA: Osteosarcoma in association with total hip
and treatment considerations for the sarcoma and associated neo- replacement. J Bone Joint Surg Br 66:632–634, 1984.
plasms. Surg Oncol Clin North Am 18:145–156, 2009. 352. Present D, Bertoni F, Enneking WF: Osteosarcoma of the man-
325. Wang LL, Gannavarapu A, Kozinetz CA, et al: Association dible arising in fibrous dysplasia. Clin Orthop 204:238–244, 1986.
between osteosarcoma and deleterious mutations in the RECQL4 353. Price CH, Goldie W: Paget’s sarcoma of bone: a study of 80 cases
gene in Rothmund-Thomson syndrome. JNCI 95:669–674, from the Bristol and the Leeds bone tumor registries. J Bone Joint
2003. Surg 51B:203–224, 1969.
326. Weichselbaum RR, Beckett M, Diamond A: Some retinoblasto- 354. Reis C, Genden EM, Bederson JB, et al: A rare spontaneous
mas, osteosarcomas, and soft tissue sarcomas may share a common osteosarcoma of the calvarium in a patient with long-standing
etiology. Proc Natl Acad Sci USA 85:2106–2109, 1988. fibrous dysplasia: CT and MR findings. Br J Radiol 81:e31–e34,
2008.
Secondary Osteosarcoma 355. Rosemann M, Kuosaite V, Nathrath M, et al: The genetics of
327. Brien WW, Salvati EA, Healey JH, et al: Osteogenic sarcoma radiation-induced and sporadic osteosarcoma: a unifying theory?
arising in the area of a total hip replacement: a case report. J Bone J Radiol Protection 22:A113–A116, 2002.
Joint Surg 72A:1097–1099, 1990. 356. Sadeghi SM, Hosseini SN: Spontaneous conversion of fibrous
328. Colina M, La Corte R, De Leonardis F, et al: Paget’s disease of dysplasia into osteosarcoma. J Craniofacial Surg 22:959–961, 2011.
bone: a review. Rheum Int 28:1069–1075, 2008. 357. Shaheen M, Deheshi BM, Riad S, et al: Prognosis of radiation-
329. Doganavsargil B, Argin M, Kececi B, et al: Secondary osteosar- induced bone sarcoma is similar to primary osteosarcoma. Clin
coma arising in fibrous dysplasia, case report. Arch Orthop Trauma Orthop Rel Res 450:76–81, 2006.
Surg 129:439–444, 2009. 358. Sim FH, Cupps RE, Dahlin DC, et al: Postradiation sarcoma of
330. Dorfman HD: Malignant transformation of benign bone lesions. bone. J Bone Joint Surg 54A:1479–1489, 1972.
In Proceedings of the Seventh National Cancer Conference of the Ameri- 359. Smith J, Botet JF, Yeh SD: Bone sarcomas in Paget disease: a study
can Cancer Society, Philadelphia, 1973, JB Lippincott. of 85 patients. Radiology 152:583–590, 1984.
331. Dorfman HD, Habermann ET: The risk of bone sarcoma in 360. Unni KK, Dahlin DC: Premalignant tumors and conditions of
patients with bone infarction (abstract). In Proceedings of the 55th bone. Am J Surg Pathol 3:47–60, 1979.
Annual Meeting of the American Academy of Orthopaedic Surgery, 361. Vuillemin-Bodaghi V, Parlier-Cuau C, Cywiner-Golenzer C,
Atlanta, February 1988. et al: Multifocal osteogenic sarcoma in Paget’s disease. Skeletal
332. Frierson HG, Jr, Fechner RE, Stallings RG, et al: Malignant Radiol 29:349–353, 2000.
fibrous histiocytoma in bone infarct. Association with sickle cell 362. Wick MR, Siegal GP, Unni KK, et al: Sarcomas of bone compli-
trait and alcohol abuse. Cancer 59:496–500, 1987. cating osteitis deformans (Paget’s disease). Am J Surg Pathol
333. Galli SJ, Weintraub HP, Proppe KH: Malignant fibrous histiocy- 5:47–59, 1981.
toma and pleomorphic sarcoma in association with medullary
bone infarcts. Cancer 41:607–619, 1978. Surface Osteosarcoma
334. Gillespie WJ, Frampton CM, Henderson RJ, et al: The incidence 363. Ahuja SC, Villacin AB, Smith J, et al: Juxtacortical (parosteal)
of cancer following total hip replacement. J Bone Joint Surg Br osteogenic sarcoma: histological grading and prognosis. J Bone
70:539–542, 1988. Joint Surg 59A:632–647, 1977.
335. Grimer RJ, Cannon SR, Taminiau AM, et al: Osteosarcoma over 364. Dwinnell LA, Dahlin DC, Ghormley RK: Parosteal (juxtacortical)
the age of forty. Eur J Cancer 39:157–163, 2003. osteogenic sarcoma. J Bone Joint Surg 36A:732–744, 1954.

ERRNVPHGLFRVRUJ
354 5  Osteosarcoma

365. Geschickter CF, Copeland MM: Parosteal osteoma of bone: a new 394. Sheth DS, Yasko AW, Raymond AW, et al: Conventional and
entity. Ann Surg 133:790–807, 1951. dedifferentiated parosteal osteosarcoma. Diagnosis, treatment,
366. Jaffe HL: Tumors and tumorous conditions of the bones and joints, and outcome. Cancer 78:2136–2145, 1996.
Philadelphia, 1958, Lea & Febiger. 395. Shuhaibar H, Friedman L: Dedifferentiated parosteal osteosar-
367. Jaffe HL, Selin G: Tumors of bones and joints. Bull NY Acad Med coma with high-grade osteoclast-rich osteogenic sarcoma at pre-
27:165, 1951. sentation. Skeletal Radiol 27:574–577, 1998.
396. Temple HT, Scully SP, O’Keefe RJ, et al: Clinical outcome of 38
Parosteal Osteosarcoma patients with juxtacortical osteosarcoma. Clin Orthop Relat Res
368. Banerjee SC: Juxtacortical osteosarcoma of mandible: review of 373:208–217, 2000.
literature and report of case. J Oral Surg 39:535–538, 1981. 397. Unni KK, Dahlin DC, Beabout JW, et al: Parosteal osteogenic
369. Bertoni F, Bacchini P, Staals EL, et al: Dedifferentiated parosteal sarcoma. Cancer 37:2644–2675, 1976.
osteosarcoma: the experience of the Rizzoli Institute. Cancer 398. van Oven MW, Molenaar WM, Freling NJ, et al: Dedifferenti-
103:2373–2382, 2005. ated parosteal osteosarcoma of the femur with aneuploidy and
370. Bertoni F, Present D, Hudson T, et al: The meaning of radiolu- lung metastases. Cancer 63:807–811, 1989.
cencies in parosteal osteosarcoma. J Bone Joint Surg 67A:901–910, 399. Wold LE, Unni KK, Beabout JW, et al: Dedifferentiated parosteal
1985. osteosarcoma. J Bone Joint Surg 66A:53–59, 1984.
371. Bras JM, Donner R, van der Kwast WA, et al: Juxtacortical osteo- 400. Wolfel DA, Carter PR: Parosteal osteosarcoma. AJR Am J Roent-
genic sarcoma of the jaws: review of the literature and report of a genol 105:142–146, 1969.
case. Oral Surg Oral Med Oral Pathol 50:535–544, 1980.
372. Campanacci M, Picci P, Gherlinzoni F, et al: Parosteal osteosar- Periosteal Osteosarcoma
coma. J Bone Joint Surg Br 66:313–321, 1984. 401. Bertoni F, Boriani S, Laus M, et al: Periosteal chondrosarcoma
373. Cardona DM, Knapik JA, Reith JD: Dedifferentiated parosteal and periosteal osteosarcoma: two distinct entities. J Bone Joint
osteosarcoma with giant cell tumor component. Skeletal Radiol Surg Br 64:370–376, 1982.
37:367–371, 2008. 402. Campanacci M, Giunti A: Periosteal osteosarcoma: review of 41
374. Dunham WK, Wilborn WN, Zarzour RJ: A large parosteal osteo- cases, 22 with long-term follow-up. Ital J Orthop Traumatol 2:23–
sarcoma with transformation to high-grade osteosarcoma: a case 35, 1976.
report. Cancer 44:1495–1500, 1979. 403. Cesari M, Alberghini M, Vanel D, et al: Periosteal osteosarcoma:
375. Edeiken J, Farrell C, Ackerman LV, et al: Parosteal sarcoma. AJR a single-institution experience. Cancer 117:1731–1735, 2011.
Am J Roentgenol 111:579–583, 1971. 404. deSantos LA, Murray JA, Finklestein JB, et al: The radiographic
376. Futani H, Okayama A, Maruo S, et al: The role of imaging modal- spectrum of periosteal osteosarcoma. Radiology 127:123–129,
ities in the diagnosis of primary dedifferentiated parosteal osteo- 1978.
sarcoma. J Orthop Sci 6:290–294, 2001. 405. Grimer RJ, Bielack S, Flege S, et al: Periosteal osteosarcoma—
377. Gamberi G, Ragazzini P, Benassi MS, et al: Analysis of 12q13-15 a European review of outcome. Eur J Cancer 41:2806–2811, 2005.
genes in parosteal osteosarcoma. Clin Orthop Relat Res 377:195– 406. Hall RB, Robinson LH, Malawar MM, et al: Periosteal osteosar-
204, 2000. coma. Cancer 55:165–171, 1985.
378. Haeckel C, Ayala AG, Radig K, et al: Protease expression in dedif- 407. Hoshi M, Matsumoto S, Manabe J, et al: Three cases with peri-
ferentiated parosteal osteosarcoma. Arch Pathol Lab Med 123:213– osteal osteosarcoma arising from the femur. J Orthop Sci 11:224–
221, 1999. 228, 2006.
379. Han I, Oh JH, Na YG, et al: Clinical outcome of parosteal osteo- 408. Howat AJ, Dickens DR, Boldt DW, et al: Bilateral metachronous
sarcoma. J Surg Oncol 97:146–149, 2008. periosteal osteosarcoma. Cancer 58:1139–1143, 1986.
380. Hinton CE, Turnbull AE, O’Donnell HD, et al: Parosteal osteo- 409. Maheshwari AV, Jelinek JS, Seibel NL, et al: Bilateral synchro-
sarcoma of the skull. Histopathology 14:322–323, 1989. nous tibial periosteal osteosarcoma with familial incidence. Skel-
381. Hoshi M, Matsumoto S, Manabe J, et al: Oncologic outcome of etal Radiol 41:1005–1009, 2012.
parosteal osteosarcoma. Int J Clin Oncol 11:120–126, 2006. 410. Martínez-Tello FJ, Navas-Palacios JJ: The ultrastructure of con-
382. Levine E, De Smet AA, Huntrakoon M: Juxtacortical osteosar- ventional, parosteal, and periosteal osteosarcomas. Cancer 50:949–
coma: a radiologic and histologic spectrum. Skeletal Radiol 14:38– 961, 1982.
46, 1985. 411. Murphey MD, Jelinek JS, Temple HT, et al: Imaging of periosteal
383. Lindell MM, Jr, Shirkhoda A, Raymond AK, et al: Parosteal osteosarcoma: radiologic-pathologic comparison. Radiology 233:
osteosarcoma: radiologic-pathologic correlation with emphasis on 129–138, 2004.
CT. AJR Am J Roentgenol 148:323–328, 1987. 412. Patterson A, Greer RO, Jr, Howard D: Periosteal osteosarcoma
384. Lorentzon R, Larsson SE, Boquist L: Parosteal (juxtacortical) of the maxilla: a case report and review of the literature. J Oral
osteosarcoma: a clinical and histopathological study of 11 cases Maxillofac Surg 48:522–526, 1990.
and a review of the literature. J Bone Joint Surg Br 62:86–92, 413. Raymond AK: Surface osteosarcoma. Clin Orthop 270:140–148,
1980. 1991.
385. Luck JV, Jr, Luck JV, Schwinn CP: Parosteal osteosarcoma: a 414. Revell MP, Deshmukh N, Grimer RJ, et al: Periosteal osteosar-
treatment-oriented study. Clin Orthop 153:92–105, 1980. coma: a review of 17 cases with mean follow-up of 52 months.
386. Masuda S, Murakawa Y: Postirradiation parosteal osteosarcoma: Sarcoma 6:123–130, 2002.
a case report. Clin Orthop 184:204–207, 1984. 415. Ritts GD, Pritchard DJ, Unni KK, et al: Periosteal osteosarcoma.
387. Millar BG, Browne RM, Flood TR: Juxtacortical osteosarcoma of Clin Orthop 219:299–307, 1987.
the jaws. Br J Oral Maxillofac Surg 28:73–79, 1990. 416. Rose PS, Dickey ID, Wenger DE, et al: Periosteal osteosarcoma:
388. Newland JR, Ayala AG: Parosteal osteosarcoma of the maxilla. long-term outcome and risk of late recurrence. Clin Orthop Relat
Oral Surg Oral Med Oral Pathol 43:727–734, 1977. Res 453:314–317, 2006.
389. Okada K, Frassica FJ, Sim FH, et al: Parosteal osteosarcoma: a 417. Schajowicz F: Juxtacortical chondrosarcoma. J Bone Joint Surg Br
clinicopathological study. J Bone Joint Surg 76A:366–378, 1994. 59:473–480, 1977.
390. Picci P, Campanacci M, Bacci G, et al: Medullary involvement in 418. Spjut HJ, Ayala AG, deSantos LA, et al: Periosteal osteosarcoma.
parosteal osteosarcoma: a case report. J Bone Joint Surg 69A:131– In Management of primary bone and soft tissue tumors, St Louis, 1977,
136, 1987. Mosby.
391. Pintado SO, Lane J, Huvos AG: Parosteal osteogenic sarcoma of 419. Unni KK, Dahlin DC, Beabout JW: Periosteal osteogenic
bone with coexistent low- and high-grade sarcomatous compo- sarcoma. Cancer 37:2476–2485, 1976.
nents. Hum Pathol 20:488–491, 1989. 420. Zarbo RJ, Regezi JA, Baker SR: Periosteal osteogenic sarcoma of
392. Reith JD, Donahue FI, Hornicek FJ: Dedifferentiated parosteal the mandible. Oral Surg Oral Med Oral Pathol 57:643–647, 1984.
osteosarcoma with rhabdomyosarcomatous differentiation. Skele-
tal Radiol 28:527–531, 1999. High-grade Surface Osteosarcoma
393. Schajowicz F, McGuire MH, Santini Araujo E, et al: Osteosarco- 421. Hoshi M, Matsumoto S, Manabe J, et al: Report of four cases with
mas arising on the surfaces of long bones. J Bone Joint Surg high-grade surface osteosarcoma. Jpn J Clin Oncol 36:180–184,
70A:555–564, 1988. 2006.

ERRNVPHGLFRVRUJ
5  Osteosarcoma 355

422. Sonneland PR, Unni KK: Case report 258: high-grade “surface” 433. Bane BL, Evans HL, Ro JY, et al: Extraskeletal osteosarcoma:
osteosarcoma arising from the femoral shaft. Skeletal Radiol 11:77– a clinicopathologic review of 26 cases. Cancer 65:2762–2770,
80, 1984. 1990.
423. Wold LE, Unni KK, Beabout JW, et al: High-grade surface osteo- 434. Chung EB, Enzinger FM: Extraskeletal osteosarcoma. Cancer
sarcomas. Am J Surg Pathol 8:181–186, 1984. 60:1132–1142, 1987.
424. Wold LE, Unni KK, Raymond AK, et al: High-grade surface 435. Dubec JJ, Munk PL, O’Connell JX, et al: Soft tissue osteosarcoma
osteosarcoma. Lab Invest 48:94A, 1983. (abstract). with telangiectatic features: MR imaging findings in two cases.
Skeletal Radiol 26:732–736, 1997.
Intracortical Osteosarcoma 436. Fang Z, Yokoyama R, Mukai K, et al: Extraskeletal osteosarcoma:
425. Anderson RB, McAlister JA, Jr, Wrenn RN: Case report 585: a clinicopathologic study of four cases. Jpn J Clin Oncol 25:55–60,
intracortical osteosarcoma of tibia. Skeletal Radiol 18:627–630, 1995.
1989. 437. Fine G, Stout AP: Osteogenic sarcoma of the extraskeletal soft
426. Jaffe HL: Intracortical osteogenic sarcoma. Bull Hosp J Dis 21: tissues. Cancer 9:1027–1043, 1956.
189–197, 1960. 438. Huvos AG: Osteogenic sarcoma of bones and soft tissues in older
427. Kyriakos M: Intracortical osteosarcoma. Cancer 46:2525–2533, persons: a clinicopathologic analysis of 117 patients older than 60
1980. years. Cancer 57:1442–1449, 1986.
428. Lichtenstein L: Bone tumors, ed 5, St Louis, 1977, Mosby. 439. Jensen ML, Schumacher B, Myhre Jensen O, et al: Extraskeletal
429. López-Barea F, Rodriguez-Peralto JL, González-López J, et al: osteosarcomas: a clinicopathologic study of 25 cases. Am J Surg
Intracortical osteosarcoma: a case report. Clin Orthop 268:218– Pathol 22:588–594, 1998.
222, 1991. 440. Lee JSY, Fetsch JF, Wasdhal DA, et al: A review of 40
430. Picci P, Gherlinzoni F, Guerra A: Intracortical osteosarcoma: rare patients with extraskeletal osteosarcoma. Cancer 76:2253–2259,
entity or early manifestation of classical osteosarcoma? Skeletal 1995.
Radiol 9:255–258, 1983. 441. Sordillo PP, Hajdu SI, Magill GB, et al: Extraosseous osteo-
431. Vigorita VJ, Jones JK, Ghelman B, et al: Intracortical osteosar- genic sarcoma. A review of 48 patients. Cancer 51:727–734,
coma. Am J Surg Pathol 8:65–71, 1984. 1983.
442. Yi ES, Shmookler BM, Malawer MM, et al: Well-differentiated
Extraskeletal Osteosarcoma extraskeletal osteosarcoma. A soft-tissue homologue of parosteal
432. Allan CJ, Soule EH: Osteogenic sarcoma of the somatic soft osteosarcoma. Arch Pathol Lab Med 115:906–909, 1991.
tissues: clinicopathologic study of 26 cases and review of the lit- 443. Weiss SW, Goldblum JR: Enzinger and Weiss’s soft tissue tumors,
erature. Cancer 27:1121–1133, 1971. ed 4, St. Louis, 2001, Mosby.

ERRNVPHGLFRVRUJ
C H A P T E R 6 

Benign Cartilage Lesions

CHAPTER OUTLINE

ENCHONDROMA CHONDROMYXOID FIBROMA


PERIOSTEAL CHONDROMA OSTEOCHONDROMA
ENCHONDROMATOSIS Solitary Osteochondroma
(Osteocartilaginous Exostosis)
Ollier’s Disease
Multiple Hereditary Exostoses
Maffucci’s Syndrome
Malignant Transformation in
Malignant Transformation in Osteochondroma
Enchondromatosis
CHONDROBLASTOMA

Traditionally, cartilage lesions are considered to be for malignant neoplasms. Although this chapter deals
of neoplastic, dysplastic, hamartomatous, and reactive with benign cartilage lesions, secondary bone sarcomas
origin. Reactive cartilage containing lesions are described developing in association with some preexisting benign
in Chapters 20 and 23. Enchondroma is an example of a cartilage lesions are also discussed.
benign cartilage neoplasm that most frequently occurs
within the medullary cavity. It rarely presents as a bone
surface subperiosteal (juxtacortical) lesion such as a peri-
osteal chondroma. Enchondromatosis is considered to ENCHONDROMA
represent a dysplastic cartilage condition rather than a Definition
true neoplasm. It occurs in two main clinical settings:
Ollier’s disease and Maffucci’s syndrome. Although soli- Enchondroma is a common intramedullary benign neo-
tary enchondromas and enchondromatosis in Ollier’s plasm composed of mature cartilage. It has a limited
disease and Maffucci’s syndrome are clinically distinct, growth potential, and a majority of enchondromas are
they share similar genetic abnormalities with frequent small, asymptomatic lesions less than 3 cm in diameter.
mutations of IDH1 and IDH2 genes. More recent experi-
ence discloses the heterogeneous nature of enchondro-
Incidence and Location
matosis, which, in addition to Ollier’s disease and
Maffucci’s syndrome, can be separated into at least five Enchondroma is a frequently occurring benign tumor
unique clinical syndromes with different patterns of skel- that, in different series, accounts for 12% to 24% of
etal involvement, genetic background, and means of benign bone tumors and 3% to 10% of all bone tumors.
familial transmission. Chondroblastomas and chondro- The age of patients varies widely, and in some series,
myxoid fibromas are two examples of benign cartilage enchondromas are fairly evenly distributed throughout
neoplasms that are characterized by immature cartilage all decades of life.5 In other series, the peak age incidence
cells and an extracellular matrix component. Osteochon- seems to be during the third and fourth decades of life.
droma, or osteocartilaginous exostosis, is traditionally More than 60% of patients are age 10 to 40 years.7,10,29,30
presented as a developmental anomaly of the hamarto- There is no clear sex predilection, and the male-to-female
matous type. The most frequent expression is a solitary sex ratio is about 1 : 1. The small bones of the hands and
exostosis. The less frequent form is a clinical syndrome feet are the most frequent anatomic sites for enchon-
of multiple hereditary exostosis in which osteochondro- droma, and approximately 60% of cases are located in
mas are associated with other anomalies of skeletal mod- these sites. The small bones of the hands are more fre-
eling. In view of recent identification of unique genetic quently involved than the bones of the feet (ratio approxi-
abnormalities characterized by the silencing of EXT1 and mately 7 : 1). Involvement of the long tubular bones is
EXT2 genes in both forms of the disease, the traditional next in frequency. The femur is the most frequently
hamartomatous concept has to be revisited and an osteo- involved long tubular bone and makes up approximately
chondroma is better described as a unique benign neo- 17% of all cases. Enchondromas of the femur are typi-
plasm recapitulating epiphyseal skeletal development. cally located near the proximal or distal ends. Midshaft
Some of the benign cartilage lesions are precursor lesions involvement is very rare. The proximal humerus is
356
ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 357

Wide age distribution

10 40
Incidence

10 70

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-1  ■  Enchondroma. Age distribution and common sites of skeletal involvement. Most common sites are indicated by large
black arrows.

involved in approximately 7% of cases. Enchondromas Therefore, if such growth cannot be explained by recent
occur less commonly in the bones of the forearm, tibia, injury, pathologic fracture, or the presence of other unre-
and fibula. They are very rare in the pelvis, ribs, scapulae, lated conditions, pain should raise the suspicion of
and vertebrae. Moreover, in several major series of bone malignancy.
tumors, there is not a single report of an enchondroma
in the craniofacial bones. In general, enchondromas are Radiographic Imaging
extremely rare in the sites most commonly affected by
chondrosarcoma: the trunk bones. To the contrary, they Enchondroma produces a localized, radiolucent defect
occur frequently in the acral skeleton, where chondrosar- with punctate or less frequently linear calcifications (see
comas almost never occur. The site where significant Figs. 6-2 to 6-5). Whether the lesion is sharply defined
overlap between the skeletal distribution of enchondroma or has indefinite outlines within bone depends on the
and chondrosarcoma occurs is in the long tubular bones, skeletal site. The degree of radiographic calcification
where both lesions occur with similar frequency. The can vary considerably. Occasionally, there may not be a
peak age incidence and most common sites of enchon- sufficient amount of mineralization to be detected on
droma are depicted in Figure 6-1. plain radiographs, and the lesion can be purely lytic in
appearance (see Figs. 6-6 and 6-7). In some radiolucent
lesions, computed tomographic (CT) imaging may reveal
Clinical Symptoms
the presence of calcifications. On magnetic resonance
Typically, enchondroma is an asymptomatic lesion inci- imaging (MRI), calcifications appear as small signal voids.
dentally discovered on radiographs or radioisotope scans MRI is better suited to demonstrate the noncalcified
performed for other reasons. In the small bones of the chondroid lesions within the marrow cavity as a low-
hands and feet, an enchondroma can expand the bone signal–intensive area on T1-weighted images and a high-
contour and present as a palpable mass. Pathologic signal–intensive area on T2-weighted images (see Figs.
fracture can be the presenting sign of enchondroma in 6-2 to 6-5). In the major long bones, the cortex and bone
phalanges, metacarpals, and metatarsal bones. Pain in contour are usually not altered, and there is no periosteal
cartilage lesions often correlates with growth activity. Text continued on p. 364

ERRNVPHGLFRVRUJ
358 6  Benign Cartilage Lesions

A B C D

E G

FIGURE 6-2  ■  Enchondroma: radiographic features. A, Anteroposterior (AP) radiograph of distal femur with heavily calcified intra-
medullary tumor occupying the diaphysis. B, AP radiograph of proximal femur with calcified intramedullary tumor occupying
diaphysis and metaphyseal area. C, Lateral radiograph of distal tibia showing calcified intramedullary lesion involving the metaphy-
sis. D, AP radiograph of proximal humerus with calcified lesion of humeral head and neck. E, AP radiograph of distal femur showing
intramedullary lesion with patchy calcifications. F, Fat-saturated T2-weighted axial magnetic resonance image (MRI) showing irregu-
lar signal enhancement within a well delineated intramedullary lesion. G, Fat-saturated T2-weighted axial MRI with contrast showing
mild irregular signal enhancement within a well delineated intramedullary lesion.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 359

A B

C D

E F

FIGURE 6-3  ■  Enchondroma: radiographic features. A and B, Anteroposterior and lateral radiographs of proximal humerus showing
a calcified intramedullary lesion extending to the intercondylar eminence. C, Reformatted coronal computed tomogram (CT) shows
patchy signal intensity of intramedullary lesion. D, Fat-saturated T2-weighted sagittal magnetic resonance image (MRI) showing
patchy signal enhancement of the intramedullary lesion. E, Fat-saturated T1-weighted axial MRI with contrast showing patchy
enhancements of signal intensity. F, Axial CT showing intramedullary lesion with linear and patchy signal.

ERRNVPHGLFRVRUJ
360 6  Benign Cartilage Lesions

A B

C E

FIGURE 6-4  ■  Enchondroma: radiographic features. A and B, Anteroposterior and lateral radiographs of distal femur showing calcified
intramedullary lesion involving distal femoral metaphysis. C, T1-weighted coronal magnetic resonance image (MRI) showing well
demarcated intramedullary lesion of low signal intensity involving the distal femoral diaphysis. Note lobulated well demarcated
borders of the lesion. D, T1-weighted axial MRI showing low signal intensity in the intramedullary lesion. The outline of the lesion
shows lobulated architecture with well demarcated satellite nodules at the periphery. E, Fat-saturated T1-weighted axial MRI with
contrast shows mild signal enhancement and discloses lobular/nodular architecture of the lesion.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 361

A B C D

E F G H
FIGURE 6-5  ■  Enchondroma: radiographic features. A and B, Anteroposterior (AP) and lateral radiographs of proximal tibia show
intramedullary calcifications involving the diaphysis. C, Fat-saturated T2-weighted coronal magnetic resonance image (MRI) of lesion
shown in A and B showing patchy signal enhancement within extensive medullary lesion. D, T1-weighted coronal MRI showing
intramedullary lesion of low signal intensity. Note well delineated lesion with scalloped outline. E and F, AP and lateral view of
distal femur showing extensive intramedullary lesions with punctate calcification. G and H, AP and oblique views of proximal
humerus showing extensive intramedullary lesion involving humeral neck and metaphyseal/diaphyseal area.

ERRNVPHGLFRVRUJ
362 6  Benign Cartilage Lesions

A B

C D E

FIGURE 6-6  ■  Enchondroma of short tubular bones: radiographic features. A, Expanded lucent area at distal metaphyseal area of
index metacarpal bone in adult (arrow). B, Base of third proximal phalanx shows well-circumscribed expanded lesion with punctate
matrix calcification (arrow). C and D, Plain radiographs of expanded lucencies in middle phalanges of fingers in two patients who
sustained pathologic fractures through thin cortical shells of bone around enchondromas. E, Pathologic fracture through enchon-
droma in fifth metatarsal shaft.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 363

A B C

F G
FIGURE 6-7  ■  Enchondroma of short tubular bones: radiographic features. A and B, Anteroposterior and lateral radiographs showing
a mildly calcified and expansile lesion of the index proximal phalanx. C, T2-weighted sagittal magnetic resonance image (MRI)
showing a lesion of the index proximal phalanx involving almost the entire medullary cavity. Note the expanded contour of bone
with thin but intact overlying cortex. D, E, and F, Axial fat-saturated T2-weighted MRI, T1-weighted MRI, and fat-saturated T1-weighted
MRI with contrast showing various degrees of signal intensity in the intramedullary lesion involving the proximal index phalanx.
Note a characteristic lacunar pattern of signal enhancement and the void in MRI shown in F. G, Expanded radiolucent lesion involv-
ing the base of the metacarpal bone of thumb.

ERRNVPHGLFRVRUJ
364 6  Benign Cartilage Lesions

new bone formation (see Figs. 6-2 to 6-6). Enchondro- loosely arranged clusters, at least focally (Fig. 6-16). The
mas that fill the medullary cavity of short tubular bones individual chondrocytes are small, lie in lacunar spaces,
may show some cortical thinning (see Figs. 6-6 and 6-7). and have small, dark nuclei (see Figs. 6-14 and 6-16). In
The contour of the small bones is typically expanded general, the chondrocytes of enchondromas are similar
along with cortical thinning. Radiographic evidence of to those seen in normal hyaline cartilage. Occasional
true extension into soft tissue with a completely disrupted binucleated cells and cells with so-called open nuclear
cortex, as well as the presence of reactive periosteal new chromatin can be present. The presence of more than
bone formation, should be regarded as indicators of malig- one chondrocyte in a lacuna should not be considered an
nancy. In rare cases, eccentric expansion of one cortex ominous finding and is not equated with binucleation or
can produce the picture of enchondroma protuberans multinucleation of cartilage cells. The chondroid matrix
(Fig. 6-8).4,5,8 In the small tubular bones, the lesion is is typically of hyaline type. Foci of fine or coarse calcifica-
usually diaphyseal, but epiphyseal extension is also fre- tions and even of enchondral ossification can be present.
quently present. In the long tubular bones, enchondro- Prominent myxoid change should not be present in
mas are most frequently located in the metaphysis. A enchondroma. If present, even when the cellularity is low,
midshaft or epiphyseal localization is extremely uncom- it usually suggests malignancy. One exception to this is
mon (see Figs. 6-6 and 6-7).22 In the long bones, enchon- in the small bones of the hands and feet, which may show
dromas typically do not show distinct outlines on plain prominent foci of myxoid change. In addition, enchon-
radiographs as they tend to do in short tubular bones. On dromas of the acral skeleton may show focal areas of
the other hand, MRIs demonstrate their sharply defined, hypercellularity with open chromatin architecture (Fig.
often lobulated borders (see Figs. 6-2 to 6-5). 6-17). Such features would be considered a sign of malig-
nancy in cartilage lesions of the major tubular bones but
Gross Findings are within the spectrum of changes observed in enchon-
dromas of the acral skeleton when combined with nonag-
Although curettage specimen material is the rule, in the gressive radiographic features. However, even at these
occasionally received intact resection specimen the car- skeletal sites, the change should be focal and the overall
tilaginous nature of the lesion is easy to recognize on matrix should be hyaline. The lesion is usually clearly
gross examination (Figs. 6-9 and 6-10). Enchondroma is demarcated from the surrounding bone, and the periph-
composed of confluent lobules of cartilage. The lobules ery of cartilage lobules often shows encasement by a rim
vary in size from less than 1 mm through several milli- of lamellar bone (Fig. 6-13). In addition, especially in the
meters to more than 1 cm. The periphery of the lesion is short tubular bones, mild focal scalloping of the inner
often somewhat irregular because the individual lobules cortical surface can be present.
can bulge into the adjacent marrow spaces or separate
satellite foci can be present. Occasionally enchondroma
Special Techniques
can grow in the form of sparsely separated small cartilage
nodules. Calcifications, ossifications, or both are respon- Special techniques are of little or no help in the diagnosis
sible for foci of ivory-white, and they may accentuate the of enchondroma. Ultrastructurally, the cartilage cells
overall lobular architecture of the lesion when they are have irregular cell surfaces and centrally located nuclei
concentrated on the periphery of cartilage islands. with condensed chromatin and a convoluted nuclear
envelope (Fig. 6-18).
Microscopic Findings Similar to other cartilage lesions, in enchondroma, a
thickening of the inner nuclear membrane is present.
Enchondromas are composed of mature cartilage that Immunohistochemically, the cells of enchondroma are
has lobular architecture (Figs. 6-11 and 6-12).7,10,26 The positive for S-100 protein and vimentin. Enchondromas
individual lobules can be separated by normal bone express cartilage lineage differentiation markers and
marrow spaces with hematopoietic elements (Fig. 6-12). invariably show nuclear positivity for SOX9 protein.
More frequently, the individual lobules are incompletely SOX9 protein is also positive on all other tumors, benign
separated by thin fibrovascular septa.7,10,29,30 Individual or malignant, that express cartilage lineage differentia-
lobules of cartilage are often partially encased by tion. The cartilaginous nature of enchondroma is clearly
mature lamellar bone. This feature is well preserved in evident on conventional hematoxylin-eosin sections, and
incidentally resected enchondromas (see Figs. 6-11 and special techniques are practically never required to
6-12), but can also be observed in curettage specimens support the diagnosis. Special techniques are also of no
(Fig. 6-13). Lobules of cartilage, especially at the periph- help in differentiating an enchondroma from a low-grade
ery of the lesion, can be sparsely separated and may chondrosarcoma.
appear as satellite nodules. In rare instances, lobules of Image analysis and DNA flow cytometry show a
cartilage can grow around the lamellar medullary bone diploid DNA histogram pattern with low proliferation
(Fig. 6-12). Such a growth pattern should not be confused rate.1,3,13,15 Cytogenetic studies detect diploid or near-
with the aggressive infiltration of the medullary cavity diploid chromosomal complement with structural aber-
frequently seen in chondrosarcoma. The overall cellular- rations and translocations most often involving
ity is very low, and in general, enchondromas appear very chromosomes 6, 12, and 24.12,28 Occasionally enchondro-
bland and have relatively few chondrocytes more or less mas may exhibit complex translocations involving several
evenly distributed within the hyaline cartilage matrix (see chromosomes such as 12, 15, and 21.12,24,28 Interestingly,
Figs. 6-14 and 6-15). More frequently, the cells form Text continued on p. 376

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 365

A C

D E

FIGURE 6-8  ■  Enchondroma of proximal humerus: radiographic features. A, Anteroposterior (AP) radiograph of proximal humerus
showing patchy and punctate calcifications within the lesion involving femoral head and neck and extending to humeral metaphysis.
B, T1-weighted axial magnetic resonance image (MRI) showing intramedullary lesion of low signal intensity with overall lobular/
nodular architecture. C, Fat-saturated T2-weighted axial MRI showing irregular signal enhancement within the intramedullary lesion.
D and E, AP radiograph and fat-saturated T2-weighted coronal MRI of an eccentric intramedullary cartilage tumor that causes medial
cortex of shaft of fibula to bulge consistent with enchondroma protuberans.

ERRNVPHGLFRVRUJ
366 6  Benign Cartilage Lesions

D E
FIGURE 6-9  ■  Enchondroma: radiographic and gross features. A, Anteroposterior radiograph shows calcified intramedullary tumor
with ill-defined borders in metaphyseal area. Note absence of any cortical abnormality. B, Axial computed tomogram of enchon-
droma of proximal humerus shows discrete, punctate intramedullary calcifications. C, Gross photograph of enchondroma in
metaphysis of proximal humerus discovered incidentally in chest radiograph. Note sharply circumscribed lobulated hyaline cartilage.
D and E, Low power and whole-mount photomicrographs showing nodular architecture of enchondroma. (D and E, ×10 and ×4,
respectively) (D and E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 367

A B

C D
FIGURE 6-10  ■  Enchondroma: gross features. A, Enchondroma of fibular head. Note lobular hyaline cartilage tumor occupying medul-
lary cavity. B, Proximal fibular enchondroma producing expansile contour of diaphysis designated as enchondroma protuberans
(same case as shown in Fig. 6-11). C, Coronal section of humeral head shows intramedullary enchondroma with confluent lobules
of hyaline cartilage. D, Specimen radiograph of humerus shown in C with prominent matrix calcification in cartilage lobules.

ERRNVPHGLFRVRUJ
368 6  Benign Cartilage Lesions

B C
FIGURE 6-11  ■  Enchondroma: microscopic features. A-C, Low power photomicrographs showing lobular/nodular architecture of
enchondroma. Note well delineated cartilage lobules. Some of the cartilage lobules are outlined by medullary bony trabeculae
forming narrow shells of bone partially encasing tumor lobules (A and B, ×10; C, ×20). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 369

A B

C D
FIGURE 6-12  ■  Enchondroma: microscopic features. A-D, Low power photomicrographs showing lobular/nodular architecture of
enchondroma, which grows in the form of well demarcated sparse lobular/nodular structures. Some nodules (shown in A and B)
grow around intramedullary bony trabeculae. The presence of bony trabeculae within the well demarcated nodules of enchondroma
should not be confused with an aggressive infiltrating growth pattern diagnostic of chondrosarcoma (A, C, and D, ×10; B, ×20).
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
370 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-13  ■  Enchondroma: microscopic features. A-D, Medium power photomicrographs of lobules of hyaline cartilage in a speci-
men obtained by curettage. Note that the lobular/nodular architecture of the lesion with narrow shells of bone partially encasing
tumor lobules can be reconstructed in curettage specimens (A-D, ×40). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 371

C D
FIGURE 6-14  ■  Enchondroma: microscopic features. A-D, Low power photomicrographs show low cellularity and small uniform nuclei
of chondrocytes residing in lacunar spaces. Inset, Microscopic details of chondrocytes showing uniform nuclei with condensed
chromatin (A-D, ×100; inset, ×400). (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
372 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-15  ■  Enchondroma: microscopic features. A-D, Low power photomicrographs of enchondroma show low to intermediate
cellularity and small chondrocytes residing in lacunar spaces with uniform nuclei (A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 373

A B

FIGURE 6-16  ■  Enchondroma: microscopic features. A and B, Low and intermediate power photomicrographs of enchondroma
showing clustering pattern of chondrocytes residing in lacunar spaces without nuclear pleomorphism or size variation. C and
D, Low and intermediate power photomicrographs show low cellularity and uniform nuclei of chondrocytes. Insets, Higher magni-
fication documenting nuclear and cytoplasmic details of chondrocytes residing in lacunar spaces. Note and example of a double
nucleated chondrocyte in D. (A, C, and D, ×100; B, ×200; insets, ×400.) (A-D and insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
374 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-17  ■  Enchondroma: microscopic features. A-D, Low and medium photomicrographs show increased cellularity and cluster-
ing of chondrocytes. Note the enlarged nuclei with open chromatin architecture without nuclear pleomorphism and cell size variation.
The levels of cellularity with the enlargement of nuclei are worrisome features and should be correlated carefully with radiographic
presentation. In this particular instance, the lesion was accepted as an enchondroma because it involved a finger and there was no
radiographic feature of an aggressive growth pattern. Such features in a cartilage lesion involving long tubular bones that was clini-
cally symptomatic (pain) and showed an aggressive growth pattern documented radiographically would be diagnostic of a low-grade
chondrosarcoma. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 375

B
FIGURE 6-18  ■  Enchondroma: ultrastructural features. A, Chondrocyte with multiple cytoplasmic processes and markedly indented
nuclear contour corresponding to dense homogeneous nuclear appearance in light microscopy (×3500). B, Higher magnification of
enchondroma in A. Note rough endoplasmic reticulum adjacent to highly indented nuclear membrane (×12,000).

ERRNVPHGLFRVRUJ
376 6  Benign Cartilage Lesions

chromosomal aberrations involving 6q13-21 have been Enchondromas of the short tubular bones are typically
associated with locally aggressive behavior of several diaphyseal lesions that sometimes involve the bone
benign cartilage lesions, including enchondroma.25 ends.14,21 They regularly produce endosteal scalloping
Recent identification of isocitrate dehydrogenases 1 and and expand the bone contour (see Figs. 6-6 and 6-7). In
2 (IDH1 and IDH2) gene mutations mapping to the long the bones of the acral skeleton, these radiographic find-
arms of chromosomes 2 and 15, respectively, in solitary ings are not considered to be indicative of malignancy.
enchondromas, chondrosarcomas, and multiple enchon- Moreover, enchondromas in these sites are typically more
dromas of Ollier’s disease and Maffucci’s syndrome cellular than enchondromas of other parts of the skele-
provide interesting clues concerning the biology of car- ton, and they may exhibit some nuclear atypia. The car-
tilage neoplasia. Approximately 40% of solitary enchon- tilage matrix is typically hyaline, but foci of myxoid
dromas show mutations of IDH1 or IDH2, whereas change can be present in acral enchondromas. Regardless
nearly 80% of multiple enchondromas and hemangiomas of the site, lesions that are exclusively myxoid, even if
associated with Ollier’s disease and Maffucci’s syndrome they have low cellularity, are suspicious for malignancy.
show mutations of those genes, suggesting an important In general, in the small bones of the hands and feet, a
and possible driver role of these mutations in the devel- cartilage lesion can show features of endosteal scalloping,
opment of cartilage neoplasia. Surprisingly, the same bone expansion, and increased cellularity and still behave
genes are frequently mutated in gliomas of the central as a benign enchondroma.2,7,10,15 If the same features are
nervous system and acute myeloid leukemia and less fre- present in other anatomic sites, such as long tubular and
quently in some other solid tumors. More detailed flat bones, they should be considered suggestive of a low-
descriptions of the role of the IDH1 and IDH2 genes in grade malignant cartilage lesion. Typically small bone
the biology of cartilage neoplasia is provided in the enchondromas are made clinically manifest by pain
section on enchondromatosis below. related to small infractions of the thinned cortex or com-
plete pathologic fracture with displacement (see Fig. 6-6).
Differential Diagnosis
Enchondroma of Long Tubular Bones.  Enchondro-
Enchondromas must be distinguished from low-grade mas of the long tubular bones account for approximately
chondrosarcomas, particularly when they involve the 45% of cases.7,26,30 The femur and humerus are the most
metaphyses of long bones in middle-aged to elderly frequently involved long bones. The proximal metaphysis
patients. The distinction can usually be made on the basis and shaft of the humerus and distal metaphysis of the
of absence of pain, no disturbance of the architecture of femur are the most frequently involved sites (see Figs.
the surrounding cancellous bone or adjacent cortex, and 6-2 to 6-5). Enchondromas located in the midshaft are
a lack of cytologic atypia.7,10,11,29,30 The nuclei of cartilage rare. Enchondromas are also considerably less common
cells in an enchondroma are small and uniform, and a in the fibula and bones of the forearm.
homogeneous chromatin pattern is present.9,19 Multinu- Enchondromas of the long tubular bones present dif-
cleated chondrocytes are infrequent, and the chondroid ferential diagnostic problems with low-grade chondro-
matrix is well formed, without prominent myxoid sarcomas, which also occur in this part of the skeleton
features.9,19 with comparable frequency. They may be microscopically
Chondroid differentiation in fibrous dysplasia can be indistinguishable from low-grade chondrosarcomas. The
distinguished from large solitary enchondromas on following are benign features of a solitary intramedullary
radiographs by the more diaphyseal localization and cartilage lesion of the long bones. Such lesions are typi-
ground-glass appearance of the fibroosseous areas. The cally asymptomatic and are incidentally discovered on
presence of fibroosseous elements in the sections adjacent radiographic images or isotope scans performed for other
to cartilaginous nodules is diagnostic for fibrocartilagi- reasons. They are small and usually measure less than
nous dysplasia. 3 cm. The adjacent bone and the overlying cortex are
normal. Microscopically, the cellularity is low, the chon-
drocytes have small dark nuclei, the matrix is hyaline, and
Enchondroma in Different Anatomic Sites
the lesion is well demarcated (i.e., the islands of cartilage
Enchondromas have a very characteristic anatomic dis- are bordered by bone trabeculae and there is no evidence
tribution that differs significantly from that of chondro- of invasion of the haversian canals of the cortex). Any
sarcoma. For that reason, the specific anatomic location deviation from this pattern should be considered suspi-
of the lesion and its radiographic features are important cious for malignancy.
and often decisive elements of the differential diagno-
sis.7,10,29,30 Therefore we provide separate descriptions of Enchondroma of Trunk Bones.  Enchondromas of the
enchondroma with some principles of differential diag- pelvis, spine, ribs, and sternum are very rare. Any carti-
nosis in various anatomic sites. lage lesion in these sites that is not reactive or metaplastic
in nature and belongs in the neoplastic cartilage category
Enchondroma of the Small Bones of the Hands and should be considered potentially clinically aggressive.
Feet.  As previously stated, the small bones of the hands This is especially true for lesions that measure more than
and the feet are the most frequent anatomic sites for 3 cm in diameter. Rare cases of enchondromas reported
enchondroma, with approximately 60% of all cases at these sites should be diagnosed after complete excision
located in these sites.2,7,10,11,29,27,30 The hands are more and thorough evaluation of their clinical, radiographic,
frequently involved than are the feet. and pathologic features. They represent well-demarcated

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 377

lesions (less than 3 cm in diameter) that uniformly have borders, the presence or absence of endosteal scalloping
low cellularity, exhibit no nuclear atypia, and produce or thickening of the adjacent cortex, the involvement of
mature hyaline matrix. Any—even minimal—deviation any associated periosteal bone, and possible extension
from this pattern should suggest a clinically aggressive of the tumor to the surrounding soft tissue. Evidence of
lesion (low-grade chondrosarcoma). Cartilaginous tumors multiple lesions or characteristic features of a cartilage
of the jaws, facial bones, and base of the skull should be dysplasia, such as Ollier’s disease, should be sought. Only
approached with particular circumspection. No major after these factors have been considered should the his-
series has reported enchondromas arising in the cranio- tologic features be studied and a diagnostic conclusion
facial bones. reached in the context of all available clinical and radio-
graphic data. Omission of any of these steps or taking
Treatment and Behavior “shortcuts” to the diagnosis of a cartilage tumor is fraught
with danger. Serious errors have been made in overdiag-
Enchondromas of long bones that are small and asymp- nosis of incidentally discovered enchondromas, as well as
tomatic require no treatment. The patient is advised to underestimation of medullary cartilage tumors, which, in
report the onset of symptoms, particularly any pain in the retrospect, showed indisputable radiologic features of
affected area, and is followed by serial radiographs and aggressiveness.
clinical evaluation. Lesions that are borderline in size, Central cartilage tumors of acral parts of the skeleton
symptomatic, or predominantly lytic or that appear are almost invariably benign but may become malignant
otherwise suspicious in nature should be curetted and if they are part of the picture of dyschondroplasia (Ollier’s
evaluated under the microscope. It is important to evalu- disease). Central cartilage tumors of the ribs, sternum,
ate the entire lesion. Limited sampling may not reveal and pelvis are unlikely to be benign, particularly if they
the focal features indicative of malignancy. Enchondro- exceed 4 cm in size.
mas of the small tubular bones are frequently treated with
curettage and bone grafting, especially if they expand the
bone contour or disturb the function of the affected site PERIOSTEAL CHONDROMA
in any way.16,18,20 The approach to biopsy of suspected Definition
cartilaginous lesions of the trunk bones should take into
account the fact that most of them are clinically aggres- Periosteal chondroma is a benign cartilage neoplasm that
sive. In fact, lesions that involve the ribs, sternum, scapula, develops on the surface of bone under the periosteum.42
or pelvis are best treated by wide local excision on the It is also referred to as juxtacortical chondroma.38,46
basis of clinical and radiographic evidence and should be
subjected to careful pathologic evaluation. This reduces Incidence and Location
the likelihood of local tissue contamination and thereby
decreases the risk of local recurrence if the lesion proves Periosteal chondromas are rare lesions and account for
to be low-grade chondrosarcoma. Enchondromas typi- less than 1% of all chondromas.32,41 The majority of cases
cally heal with consolidation after curettage and bone are diagnosed during the second and third decades of
grafting. Recurrence of enchondroma suggests malig- life. The limited experience with these lesions indicates
nancy, especially in lesions that affect the long bones.23 that male patients are probably more frequently affected
Nevertheless, an incompletely curetted enchondroma, than are female patients. Periosteal chondromas typically
especially in the short tubular bones, can occasionally affect the long tubular bones of the extremities.32,40,43,44,50
recur and can be successfully treated by a second curet- The proximal humerus is the single most frequently
tage.9,17,19,20,22,26 Rare and somewhat questionable exam- affected site; nearly 50% of cases are diagnosed at this
ples of enchondroma progressing to a chondrosarcoma site. The acral skeleton with the involvement of the short
have been described.6,17 tubular bones of the hands is the second most frequently
affected site. Rare examples of periosteal chondroma
Personal Comments have been described in the spine, clavicle, ribs, and
toes.31,35-37,39,49,52,53 The peak age incidence and most com-
In the assessment of cartilaginous lesions of bone in monly affected skeletal sites are shown in Figure 6-19.
general and of enchondromas in particular, it is essential
that an orderly and stepwise procedure be followed. The Clinical Symptoms
age of the patient and the anatomic site of the lesion
should first be considered in addition to the presence or This surface lesion most often presents as a palpable
absence of clinical symptoms. The presence of pain mass. Because periosteal chondromas are frequently
is extremely important and may be the only indicator found near tendon insertion sites and disturb their func-
of malignancy. Other causes of pain should be ruled tion, pain and local discomfort on activity can be initial
out before factoring this finding into the equation. Patho- symptoms. Periosteal chondromas are sometimes discov-
logic fractures in short tubular bones containing enchon- ered incidentally on radiographs.
dromas are very common and do not suggest unusual
aggressiveness. Radiographic Imaging
Next, radiographs should be examined to determine
the precise anatomic site, the size of the lesion, the pres- Periosteal chondroma presents as a well-circumscribed
ence of matrix calcification, the character of the lesion’s surface lesion of bone with punctate calcifications on

ERRNVPHGLFRVRUJ
378 6  Benign Cartilage Lesions

Peak
incidence
10 30
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-19  ■  Periosteal chondroma. Peak age incidence and most common anatomic sites of involvement. Most common site is
indicated by a large black arrow.

radiographs33,34 (see Figs. 6-20 to 6-22). The degree Gross Findings


of matrix mineralization can vary from purely lytic to
heavily calcified (see Figs. 6-20 to 6-22). In a typical On gross examination, periosteal chondroma is a lobu-
case, the cartilaginous nature of juxtacortical chon- lated cartilaginous mass covered by fibrous tissue that
droma is easy to recognize on plain radiographs. The represents an elevated periosteum (see Figs. 6-21 and
cortex beneath the lesion is usually eroded, with elevated 6-23). Calcifications can often be seen within the lobules.
edges forming a craterlike excavation (see Figs. 6-20 to The cortex beneath is eroded and usually smoothly exca-
6-22). The presence of an elevated periosteum overlying vated, but sometimes it has a scalloped border. The lesion
the lesion is best documented on MRI (see Figs. 6-20 is clearly separated from the medullary cavity by a rim of
and 6-22). It can sometimes be clearly seen on plain sclerotic cortical bone. The size of periosteal chondromas
radiographs or CT (Fig. 6-21). The affected area may varies from less than 1 cm to 5 cm.
also show minimal evidence of a zone of subcortical scle-
rosis beneath the lesion. Although the cortex underly- Microscopic Findings
ing the lesion is excavated, there is no complete cortical
disruption, and the lesion is clearly demarcated from Microscopic features are those typical of a chondroma
the medullary cavity by a continuous rim of cortical with a hyaline cartilage matrix (see Figs. 6-22 to 6-24).
bone (see Figs. 6-20 to 6-22). CT or MRI may be On average, juxtacortical chondromas are more cellular
needed to evaluate the extent of involvement and the than are enchondromas of long bones and can show mild
relationship between the lesion and adjacent structures.51 nuclear atypia or binucleated chondrocytes. Scalloping
The elevated periosteum surrounding the lesion is and erosion of the outer cortex are seen microscopically.
represented on plain radiographs by solid buttresses of A sectioning artifact may give the impression of small
mature subperiosteal bone that can sometimes slightly satellite cartilaginous foci of cellular cartilage in the scle-
overhang the edges of the central excavation. This rotic border (see Figs. 6-25 and 6-26). This does not
appearance is quite characteristic for smaller periosteal indicate invasiveness. Some periosteal chondromas may
chondromas. Text continued on p. 386

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 379

A B

C D

FIGURE 6-20  ■  Periosteal chondroma: radiographic features. A and B, Lateral and oblique plain radiographs of unusually large peri-
osteal chondroma of proximal humerus in young man. Note sharply defined borders and intralesional matrix calcification. C and
D, Fat-saturated T1-weighted and T2-weighted sagittal magnetic resonance images (MRI) of a large femoral periosteal chondroma
in a 13-year-old boy. Inset, T1-weighted axial MRI with contrast of lesion shown in C and D. Cortex beneath lesion is thinned but
intact.

ERRNVPHGLFRVRUJ
380 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-21  ■  Periosteal chondroma: radiographic and gross features. A, Plain radiograph shows concave cortical erosion near
insertion of biceps muscle on lateral aspect of humeral shaft. B, Computed tomogram of lesion shown in A. External aspect of cortex
is eroded with solid periosteal bone buttressing. C, Specimen radiograph of lesion shown in A and B. Note sharply defined borders
of cortical erosion and intact cortex beneath lesion. D, Gross photograph of periosteal chondroma of humerus shows chondroid
tumor beneath periosteum, eroding underlying cortex. Medullary cavity is not involved.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 381

A B

C D

FIGURE 6-22  ■  Periosteal chondroma: radiographic and microscopic features. A, T1-weighted sagittal magnetic resonance image
shows periosteal lesion growing on the popliteal surface of the femur. The lesion is covered by elevated periosteum and the under-
lying cortex is eroded but the medullary cavity is not involved. B, Whole-mount photomicrograph showing a fibrous capsule cor-
responding to elevated periosteum covering the surface of the lesion and the partially eroded cortical bone beneath the lesion.
C and D, Low power photomicrographs showing increased cellularity and clustered growth pattern of chondrocytes (C and D, ×50).
(B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
382 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-23  ■  Periosteal chondroma: gross and microscopic features. A, Gross photograph of periosteal chondroma shows chon-
droid tumor beneath periosteum eroding underlying cortex. B, Composite whole-mount photomicrograph of periosteal chondroma
showing a cap-shaped lesion growing beneath the periosteum and eroding the underlying cortex. C and D, Low power photomi-
crographs showing base of periosteal chondroma with interface between lesion and underlying cortex. Note scalloping of underlying
cortical bone, but there is in general a sharp demarcation between periosteal chondroma and cortex with no feature of infiltrative
aggressive growth pattern (C and D, ×40). (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 383

A B

C D

FIGURE 6-24  ■  Periosteal chondroma: microscopic features. A and B, Low power photomicrographs of the lateral aspect of periosteal
chondroma showing elevations of periosteum and erosion of the underlying cortex. C and D, Low power photomicrographs of the
base of periosteal chondroma showing erosion of underlying cortex. Note that the underlying cortex is thin but there is a sharp
demarcation between the lesion and the underlying cortical bone. (A and C, ×20; B and D, ×40.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
384 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-25  ■  Periosteal chondroma: microscopic features. A-D, Low power photomicrographs showing satellite nodules in perios-
teal chondromas. The satellite nodules are typically present in the superficial and lateral aspects of the lesion. Occasionally they
can be present at the base of the periosteal chondroma. The presence of satellite nodules typically represent tangentially cut
marginal irregularities and should not be considered as signs of aggressive invasive growth. (A, B, and D, ×20; C, ×60.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 385

B
FIGURE 6-26  ■  Periosteal chondroma: microscopic features. A, Lobulated cartilaginous tumor bordered by extensive subperiosteal
lamellar bone. Apparent satellite nodules at periphery (arrows) represent tangentially cut marginal irregularities. B, Nodular marginal
extension of periosteal chondroma with central calcification and endochondral ossification. Inset, High cellularity and mild atypia
of chondrocytes (A, ×20; B, ×60; inset, ×200). (A, B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
386 6  Benign Cartilage Lesions

bulge for a short distance into the underlying medullary to as Ollier’s disease and Maffucci’s syndrome, and should
cavity, but there is always a border of intervening lamellar not be regarded simply as multifocal enchondromas of
bone. bone (i.e., as multifocal benign cartilage neoplasms).
Enchondromatosis represents a pathogenetically distinct
developmental disorder of enchondral ossification that is
Special Techniques
probably more accurately described as cartilage dysplasia.
Similar to enchondromas, periosteal chondromas ex- The term dyschondroplasia was introduced by Ollier in his
presses a full roster of markers characteristic of cartilage description of the entity in 1900.66 Maffucci’s syndrome,
lineage differentiation. In such a sense, these lesions are in which multifocal skeletal cartilage lesions coexist with
immunohistochemically indistinguishable from ordinary extraskeletal hemangiomas of skin, soft tissue, and viscera,
enchondromas. Like enchondroma, periosteal chondro- was described 19 years earlier in 1881.63
mas harbor mutations of IDH1 and IDH2 genes. Several More recent experience indicates that enchondro-
distinct clonal chromosomal abnormalities have been matosis is more heterogeneous than originally thought.
described in periosteal chondromas; they include rear- In addition to the two main forms of enchondroma-
rangements of 2q37, 4q21-25, 11q13-15, and 12q13. tosis referred to as Ollier’s disease and Maffucci’s
syndrome, which are somatic noninheritable disorders,
there are at least five additional forms of enchondroma-
Differential Diagnosis
tosis, referred to as metachondromatosis, genochondromato-
Juxtacortical chondrosarcoma can usually be distinguished sis, spondyloenchondrodysplasia, dysspondyloenchondromatosis,
with ease by the large size of the lesion (>5 cm) and the and cheirospondyloenchondromatosis.52-62,64,65,67-70,95 They are
absence of radiologic evidence of solid periosteal new characterized by distinct patterns of transmission (auto-
bone buttressing at the margins. The latter is a charac- somal dominant vs. recessive), clinical presentations, and
teristic feature of periosteal chondroma. Microscopically, radiographic presentations, as well as unique genetic
the degree of cellularity, variation in size and shape of backgrounds known for some of the disorders. Moreover,
nuclei, and frequent multinucleate chondrocytes are the they are associated with dissimilar risks for secondary
basis for differentiating these two tumors. Juxtacortical chondrosarcoma. Detailed description of these syndromes
chondrosarcomas are also more likely to show irregular is beyond the scope of this textbook, but their main
invasion of surrounding soft tissue and lack the limiting differentiating features are summarized in Table 6-1
periosteal shell that is present in periosteal chondromas. (Figs. 6-27 and 6-28).
Periosteal osteosarcoma, a predominantly cartilaginous form
of surface osteosarcoma, can be recognized by the feath-
ery perpendicular calcific striae seen on radiographs. Ollier’s Disease
Although Codman’s triangles may be seen at the limits
Definition
of this tumor, periosteal osteosarcoma usually lacks the
solid buttresses seen in periosteal chondroma. Periosteal Ollier’s disease is a rare, nonhereditary skeletal disorder
osteosarcoma is diagnosed from the presence of sheets of characterized by a multifocal intramedullary proliferation
primitive mesenchymal cells between the cartilage lobules, of hypercellular dysplastic cartilage. The lesions have a
with tumor osteoid and bone deposition between the cells. tendency to be metaphyseal and are sometimes eccentri-
cally placed, with predominant unilateral involvement of
the appendicular skeleton. The clinical manifestations
Treatment and Behavior
typically appear during childhood, and the extent of skel-
Because periosteal chondromas may demonstrate radio- etal involvement is variable.
logic overlap with juxtacortical chondrosarcoma, the
preferable mode of treatment is wide local excision that Incidence and Location
includes the underlying cortex.47 Earlier reports on fre-
quent local recurrences after curettage of juxtacortical As stated, Ollier’s disease is rare, and its true incidence is
chondroma are controversial because some of these unknown. The largest series reported from the Rizzoli
lesions might have been surface chondrosarcomas.48 In Institute consisted of 51 cases of multiple en­chondromas
selected skeletal sites such as the ribs and fibula, segmen- presumably representing enchondromatosis compared
tal resection is the preferable mode of treatment. In with 334 solitary enchondromas encountered in the same
other sites, en bloc excision is preferred over curettage. period.77 There is no clear sex difference in incidence.
The latter may be followed by recurrence and require The extent of involvement of the skeleton varies
reoperation. Complete local excision of juxtacortical widely from case to case.77,82,86 At one end of the spectrum
chondroma is a curative procedure. In some instances are the cases in which there is limited involvement of one
periosteal chondroma may cause growth disturbance of bone or only a few short tubular bones. On the other end
the affected bone. In such cases the presence of the lesion of the spectrum are the cases with massive involvement
is associated with shortening of the bone.50 of multiple bones and severe deformities. The most fre-
quent presentation is a tumor affecting one extremity,
but bilateral involvement is often present. Cases with
ENCHONDROMATOSIS symmetric involvement of both extremities are seen
infrequently. Even in cases with diffuse involvement of
Enchondromatosis, also called dyschondroplasia and mul- multiple bones, the disease has a tendency to predomi-
tiple enchondromas, occurs in two clinical settings, referred nate on one side of the body.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 387

TABLE 6-1  The Classification of Different Types of Enchondromatosis


Involved
Enchondromatoses Clinical Subtypes Genes
Nonhereditary
Ollier’s Disease IDH1, IDH2,
• Manifests in early childhood PTH1R
• Enchondromas primarily affect the short and long tubular bones
of the extremities
• Craniofacial bones and vertebrae usually not involved
Maffuci’s Syndrome IDH1
• Manifests in early childhood
• Skeletal involvement by enchondromas identical to Ollier’s disease
• Cavernous and spindle-cell hemangiomas in the dermis, subcutis,
and internal organs
Autosomal Dominant
Metachondromatosis PTPN11
• Manifests in early childhood
• Skeletal involvement by enchondromas involving iliac crest and
metaphyses of long bones of lower extremities
• Osteochondroma-like lesions involving hands and feet
Genochondromatosis TYPE 1 PTHLH
• Manifests in early childhood • Additional enchondromas in clavicle
• Symmetric enchondromas in the metaphyses of the proximal TYPE 2
humerus and distal femur
• Additional enchondromas in short
• Enchondromas do not cause bone deformities and tend to
tubular bones of hand, wrist, and feet
regress in adulthood

Autosomal Recessive
Spondyloenchondrodysplasia TYPE 1 ACP5
• Manifests at birth to later infancy • Classic
• Vertebral dysplasia combine with enchondromas involving pelvis TYPE 2
and long tubular bones
• Central nervous system involvement
• Short stature associated with short limbs, lumbar lordosis, and
with spasticity and developmental
facial abnormalities
delay, late-onset cerebral classifications

Unknown Pattern of Transmission


Cheirospondyloenchondromatosis Unknown
• Manifests in early age and associated with mental retardation
• Symmetrically distributed enchondromas with preponderance for
involvement of metacarpals and phalanges, resulting in short
hands and feet with platyspondyly
• Moderate dwarfism and enlarged joints
Dysspondyloenchondromatosis Unknown
• Manifests at birth and during infancy
• Enchondromas involving the long tubular and flat bones
• Bones of hands and feet are not or only mildly affected
• Malformations of spine with severe segmentation of vertebral
column
• Unequal limb length and facial deformations

Ollier’s disease has a predilection for the appendicular skeletal sites involved in Ollier’s disease are shown in
skeleton, but in more severe forms the trunk bones can Figure 6-29.
also be involved. The short tubular bones of the hands
are most frequently affected. The bones most often Clinical Symptoms
affected after the hand are the short tubular bones of
the feet, femur, and humerus and the bones of the In general the more diffuse and severe the skeletal
forearm. The femur is the most frequently involved long involvement, the earlier in life symptoms appear.77,82
tubular bone, followed by the tibia and humerus. The Typically, individuals with presentation in early child-
pelvis is the most frequently involved trunk bone. The hood develop extensive and severe forms of the disease.
lesions in Ollier’s disease can sporadically occur in In general, symptoms appear early, and most cases are
other trunk bones, such as the scapula and ribs. Typically, typically diagnosed in childhood. The symptoms that
they do not involve the vertebral column or craniofacial lead to discovery of the disease vary with the anatomic
bones. The peak age incidence and most common distribution and the extent to which they alter the

ERRNVPHGLFRVRUJ
388 6  Benign Cartilage Lesions

Onset of
symptoms
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-27  ■  Ollier’s disease. Peak age incidence at onset of symptoms and most common anatomic sites of involvement. Most
frequent sides are indicated by arrows.

anatomy and growth of the affected bone. The most tic.77,82,86,91,96,99,111 The lesions have a tendency to be
common symptom is slowly increasing swelling of the metaphyseal and diaphyseal, eccentric, and multifocal
fingers. With the involvement of the long tubular bones, (see Figs. 6-30 to 6-33). Metaphyseal involvement is less
angular deformity of the affected extremity can be a pre- evident in the short tubular bones, where the eccentricity
senting symptom. The involvement of long tubular bones of the lesions with multiple lytic defects oriented perpen-
is typically associated with length discrepancy.72,94,110 The dicularly to the long axis and extending toward the soft
affected bones grow more slowly. Retarded growth and tissue is most pronounced. The lesions often show punc-
length discrepancy are particularly evident in the lower tate calcifications that are typical for the radiographic
extremities. In severe cases, the discrepancy can be in a appearance of the cartilaginous matrix. The combined
range of several centimeters in early childhood (age 2 to metaphyseal and diaphyseal involvement is best seen in
3 years). Pathologic fracture may be a presenting the long tubular bones (see Figs. 6-31 and 6-32). In these
symptom, but more often occurs later in the course of locations, the lesion forms elongated grooves or longitu-
the disease with the progression of bone involvement. If dinal lucent columns along the long axis of bone. The
one of the patient’s bones, such as in the forearm, is radiographic appearance is best understood if the changes
altered, the retarded growth of the affected bone can are envisioned as a parallel arrangement of rows of dys-
produce bony deformity of the unaffected or less affected plastic cartilage that extend from the growth plate toward
paired bone. Patients with severe enchondromatosis may the diaphysis. The radiologic appearance is sometimes
be seen in adult life with shortening and bowing deformi- referred to as the fluted-column sign (Fig. 6-31). With
ties of the extremities that severely affect motor function. progression of the lesion as a result of the continuous
Other secondary changes, such as compensatory scolio- growth of cartilage, larger expanding masses that extend
sis, can be present. to involve the diaphysis are formed. At this stage, the
parallel arrangement of the cartilaginous lesion may
Radiographic Imaging become so distorted that it presents as a large multilobu-
lar mass that involves the bone end. Severe involvement
Enchondromatosis presents with radiographic features of both proximal and distal metaphyses can produce a
that are distinctive and in some cases diagnos- Text continued on p. 395

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 389

A B

C D

FIGURE 6-28  ■  Ollier’s disease: radiographic features. A-D, Radiographic manifestations of Ollier’s disease involving the hands of
four different patients with multiple disfiguring lucent lesions corresponding to dysplastic cartilage involving multiple bones.

ERRNVPHGLFRVRUJ
390 6  Benign Cartilage Lesions

B
FIGURE 6-29  ■  Ollier’s disease: radiographic features. A, Lateral radiograph of distal femur of a 13-year-old girl with Ollier’s disease
shows typical pattern of intramedullary linear lucencies that extend from growth plate upward into shaft (fluted-column sign). In
addition, small eccentric subperiosteal lucency is seen in posterior cortex. B, Radiograph of proximal tibia and fibula of the same
patient shown in A. Healed pathologic fracture of tibial shaft with angular deformity and bowing of fibula. Fluted-column sign is
present, but epiphysis is uninvolved.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 391

A B

C
FIGURE 6-30  ■  Ollier’s disease: radiographic features. A, Fan-shaped radiolucencies are seen in anteroposterior radiograph of the
pelvis of an 8-year-old boy with Ollier’s disease. Elongated columns of dysplastic cartilage extend from iliac crest growth plate into
body of ilium. Proximal femoral shaft is also involved. B, Radiograph of right hip shows characteristic medial intertrochanteric
lucency seen in about one third of cases of Ollier’s disease. Pelvis is involved as well. C, Asymmetric involvement of right pelvis
and proximal femur in 5-year-old girl with Ollier’s disease.

ERRNVPHGLFRVRUJ
392 6  Benign Cartilage Lesions

A B C D

E F G

FIGURE 6-31  ■  Ollier’s disease: radiographic features. A-G, Extensive disfiguring Ollier’s disease involving multiple bones in one
patient.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 393

A B C

FIGURE 6-32  ■  Ollier’s disease: radiographic features. A-D, Anteroposterior radiographs showing extensive disfiguring Ollier’s disease
predominantly involving right pelvis and bones of right lower extremity.

ERRNVPHGLFRVRUJ
394 6  Benign Cartilage Lesions

Chr 2
25.3
IDH1 IDH2
25.1 25.2
24 Exon 10

Exon 11
Exon 10
Chr 15
Exon 9
Exon 8

Exon 7

Exon 6

Exon 5

Exon 4

Exon 3

Exon 2
Exon 1

Exon 9
Exon 8

Exon 7
Exon 6
Exon 5
Exon 4

Exon 3
Exon 2

Exon 1
23
22
209100952

209119806

90627211

90645708
13
21 12
16 11.2
11.1
11.1 11.2
15 12
14 13
13 14

12 IDH1 21.1
15
IDH2
21.2
11.2 Isopropylmalate dehydrogenase-like domain 21.3 Isopropylmalate dehydrogenase-like domain
11.1 22.2 22.1
11.1
11.2 22.3
12 1 414 aa 23 1 452 aa
13 11 399 24 45 440
14.1 25
14.3
14.2 IDH2 208
21.1 26.1
21.2 26.2
21.3 5148 26.3
766
22
23
102531392 bp
24.1
24.2
24.3
1 414 aa 1 452 aa
31
32.1 Missense substitution Synonymous substitution
32.2
32.3 Deletion Missense substitution
33
Nonsense substitution - Stop codon Nonsense substitution - Stop codon
34 IDH1
35
36 B
37.1
37.2
37.3

243199373 bp

A Tumor cell

Citrate Histone
Mitochondria
demethylases
Isocitrate
Citrate
IDH1
Isocitrate
TCA cycle

IDH3 IDH2
-KG TET1 KDM2a
-KG TET2
IDH2 IDH1
mut
Nucleus
mut
2-HG
OH
2-HG
Nucleosome

HIF-1 H3K4me1, H3K4me3,


stabilization H3K9me2, H3K27me2,
Endostatin H3K79me2
5-Hydroxymethylcytosine
HIF1-
Cancer biomarker?

VEGF Dysregulation of
epigenetic and
Cytoplasm gene expression profiles

C
Glucose Acetyl-CoA Fatty acids

Lactate Pyruvate Citrate Isocitrate


NADP
IDH1 or
Acetyl-CoA IDH2
TCA NADPH NADP
KG 2-HG
IDH1 mutant or
Glutamine or glutamate IDH2 mutant
D
FIGURE 6-33  ■  Molecular mechanisms of enchondromatosis. A, Chromosomal location, exonal structure, functional domain, and the
distribution of mutations of the IDH1 gene. B, Chromosomal location, exonal structure, functional domain, and the distribution of
mutations of the IDH2 gene. C, Molecular mechanisms controlled by IDH1 and IDH2 genes and functional implications of their muta-
tions. (C, Reprinted with permission from Prensner JR, Chinnaiyan AM: Nature Med 17:291-293, 2011.) D, Significance of IDH1 and IDH2
mutations for NADP/NADPH energy cycle within the mitochondria. (D, Reprinted with permission from Cairns RA, et al: Nature Rev
Cancer 11:85-95, 2011.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 395

dumbbell appearance of the affected long bone. Some- disease are similar to solitary enchondromas.89,104 The
times the dysplastic changes may affect only a portion of hallmark genetic abnormality of Ollier’s disease is the
the growth plate, leading to asymmetric involvement presence of somatic mutations involving the isocitrate
with uneven growth and resulting bowing deformities. A dehydrogenase genes, IDH1 and IDH2, mapping to the
very characteristic radiologic finding is an exophytic long arms of chromosomes 2 and 15 repectively71,97,98
eccentric lucency in the intertrochanteric region of the (Fig. 6-33). These mutations are seen in nearly 80% of
femur (Fig. 6-32). It shows associated radiolucent stria- patients with Ollier’s disease but are also present in soli-
tions that are oriented oblique to the long axis of the tary enchondromas, periosteal enchondromas, and some
femur. The broad base is usually centered over the lesser central chondrosarcomas, further confirming the unified
trochanter. In the flat bones, the parallel arrangement of pathologic mechanisms contributing to the development
an elongated lytic lesion that originates at the junction of of these neoplasms. The mutations affecting the IDH
the secondary ossification center, such as the iliac crest, genes are also frequently found in gliomas of the central
can be seen (see Figs. 6-30 and 6-32). This produces nervous system and acute myeloid leukemia.83,93,114 This
characteristic fan-shaped grooves that extend into the explains some known clinical associations showing that
metaphysis. patients with Ollier’s disease are prone to developing
central nervous system gliomas.73,92 The mutations of
the IDH genes are less frequently present in some other
Gross Findings
solid tumors. These genes encode the cytoplasmic and
The affected area is typically expanded and the entire mitochondrial NADP+-dependent isocitrate dehydroge-
bone is shortened. On a cut section, the affected metaph- nases, which convert isocitrate to α-ketoglutarate (Fig.
yseal regions show extensive involvement and contain 6-33). Mutations in enchondromatosis most frequently
longitudinal extensions composed of numerous pea-sized affect the Arg132 position in exon 4 of IDH1 and also
cartilage masses. Parallel arrangement of rows of carti- include R132C, R132H, R132G, or rare variants affect-
lage masses can be focally present, but in many cases of ing R132. The mutations in enchondromatosis of Olli-
severe involvement the lesion can be grossly distorted. It er’s type typically involve the IDH1 gene. The IDH2
is composed of irregular masses of cartilage that range in gene (R172S) appears to be rarely involved. Maffucci’s
length from 1 cm to several centimeters, located in the syndrome is characterized by exclusive involvement of
metaphyseal parts of the long bone and extending into IDH1. Mutations of IDH genes result in the new ability
the diaphysis. Eccentric subperiosteal hyaline cartilage of the enzyme to catalyze the NADPH-dependent
nodules can be seen. reduction of α-ketoglutarate to (R)-2-hydroxyglutarate
(2HG).81,100,102,108 Excess accumulation of 2HG acts as an
oncometabolite and contributes to the development of
Microscopic Findings
enchondromas76,113,115 (Fig. 6-33). In addition, the IDH
The morphology of lesions in Ollier’s disease is some- mutations impair histone demethylation and are associ-
what similar to that of solitary enchondroma. Enchon- ated with a DNA hypermethylator phenotype, resulting
dromas in Ollier’s disease have overall lobular architecture in defective cellular differentiation.83,88
with encasement of individual cartilage lobules by mature A small fraction of patients (less than 5%) with Ollier’s
medullary bone similar to solitary enchondromas (Fig. disease carry mutations of the PTH1R gene encoding a
6-35). In general, the lesions in Ollier’s disease are more receptor for parathyroid hormone, resulting in upregula-
cellular and less calcified than most typical solitary tion of the Indian hedgehog (IHH) pathway.80,85,105,106
enchondromas. Moreover, the cartilage cells in enchon- Mutations of PTH1R include R150C, G121E, A122T, and
dromatosis are larger than the cells of solitary enchon- R255H. Mutant PTH/PTHrP1 receptor complex consti-
droma. They have an open chromatin structure, and tutively activates IHH signaling and is sufficient to trigger
double nucleated cells are also frequently present (see the formation of enchondroma-like lesions in animal
Figs. 6-36 and 6-37). Features of nuclear atypia and models (Fig. 6-34).
immaturity of the extracellular matrix with frequent
myxoid change further complicate the microscopic Differential Diagnosis
pattern, making the microscopic differential diagnosis
of enchondromatosis and low-grade chondrosarcoma The dysplastic chondroid tissue in this condition charac-
extremely difficult. teristically extends in columns from the physis through
In summary, the cartilage in Ollier’s disease has fea- the metaphysis into the diaphysis. Although such lesions
tures consistent with grade 1 chondrosarcoma if cytology can simulate low-grade chondrosarcomas microscopically,
of the lesion is evaluated out of context without careful close correlation with the radiologic pattern of involve-
consideration of its radiographic and clinical setting. ment usually provides a solid basis for distinguishing them
from chondrosarcoma. The richly cellular dysplastic car-
tilage may show mild nuclear atypia and multinucleation
Special Techniques
of chondrocytes, which may raise questions of secondary
Similar to ordinary enchondromas, chondromas of Olli- malignant change. Although the lesions of Ollier’s disease
er’s disease express markers of cartilage lineage differen- may be eccentrically placed in bone, the soft tissue itself is
tiation and are immunohistochemically indistinguishable not involved. Conversion to low-grade chondrosarcoma
from solitary enchondromas. In fact, genome-wide is usually signaled by a change in symptoms and exten-
expression studies show that enchondromas of Ollier’s sion beyond the bony cortex into the adjacent soft tissue.

ERRNVPHGLFRVRUJ
396 6  Benign Cartilage Lesions

Chr 3
26
PTH1R
25

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
24.3
24.2

Exon 1

Exon 2

Exon 3

Exon 4

Exon 5
Exon 6
Exon 7
Exon 8
Exon 9
24.1
23
22

46919235

46945289
21.3
21.2 PTH1R
21.1
14.3 14.2
14.1
13
12
PTH1R
11.2
11.1
11.2 11.1
12
13.1 1 594 aa
13.2
13.3 47 179 187 463
21
22
23 GPCR, family 2, extracellular hormone receptor domain
24
25.1
GPCR, family 2-like domain
25.2
25.3
26.1
26.2
Synonymous substitution
26.3 Missense substitution
27
28 Non stop extension
29
Deletion frameshift
198022430 bp
FIGURE 6-34  ■  Molecular mechanisms of enchondromatosis. Chromosomal location, exonal structure, functional domains, and the
distribution of mutations of the PTH1R gene.

Radiologically, there is some overlap between enchon- Vascular lesions in Maffucci’s syndrome most typically
dromatosis and polyostotic fibrous dysplasia because both represent cavernous hemangiomas with frequent throm-
involve multiple skeletal sites. Enchondromatosis more bosis and calcified thrombi that are often seen on plain
frequently involves the small bones of the hands and also radiographs. Hemangiomas in Maffucci’s syndrome
shows a more pronounced unilateral predominance than have a tendency to be more pronounced in the areas
fibrous dysplasia. The frequency of craniofacial involve- of skeletal involvement by enchondromatosis but are
ment in fibrous dysplasia and the fact that enchondro- not restricted to these regions. Usually the cartilaginous
matosis is limited to bones preformed in cartilage aids in skeletal dysplastic changes manifest earlier, and the dis-
the differential diagnosis of these two lesions. Enchon- order is initially classified as Ollier’s disease. Vascular
dromatosis almost never involves the skull, and the skin lesions develop later in life in some patients. Minimal soft
pigmentation seen in polyostotic fibrous dysplasia is not tissue hemangiomas may not be recognized initially.
seen in Ollier’s disease. Other soft tissue anomalies include arteriovenous aneu-
rysms or fistulas, lymphedema, and lymphangiomas.
Vascular anomalies also can involve the gastrointestinal
Treatment and Behavior
tract. Maffucci’s syndrome most likely represents more
The cartilage of Ollier’s disease has the propensity for severe mesodermal dysplasia in which both the skeletal
continuous slow growth. In most cases, the progression and extraskeletal-forming tissues are altered. In keeping
of the lesions has stabilized at puberty, but occasionally with this concept, patients with Maffucci’s syndrome are
they continue to grow even during adulthood. The prone to develop cartilage neoplasm in extraskeletal sites
potential for progressive growth of dysplastic cartilage in such as the trachea.125,129 Coexistence of nodular goiter
enchondromatosis constitutes a basic biologic difference and adrenal insufficiency has also been reported.128 As
between Ollier’s disease and solitary enchondroma, which stated, the risk of secondary malignancy in Maffucci’s
as a benign neoplasm has limited growth potential. There syndrome is even higher than that in Ollier’s disease.128
is no specific treatment available and affected patients Similarly to Ollier’s disease, enchondromas and spindle-
require surveillance for their entire lifetime because of cell hemangiomas in Maffucci’s syndrome carry the
the possibility of secondary sarcomatous change. Correc- mutations of the IDH genes.97 It appears that in Maf-
tive surgery is sometimes performed for deformities and fucci’s syndrome the IDH1 gene is almost exclusively
length discrepancy, and occasionally severely stunted involved.
(nonfunctioning) extremities have to be amputated. More Similar to Ollier’s disease, patients with Maffucci’s
commonly, osteotomies and lengthening procedures are syndrome may develop low-grade conventional chondro-
required to correct growth disturbances. sarcomas.128,130,131,133 The manifestation and diagnostic
criteria of chondrosarcoma in Maffucci’s syndrome are
identical to those of chondrosarcoma complicating Olli-
Maffucci’s Syndrome er’s disease. Rare instances of dedifferentiated chondro-
Maffucci’s syndrome is a rare condition in which enchon- sarcoma and soft tissue angiosarcoma in Maffucci’s
dromatosis identical to that of Ollier’s disease coexists syndrome have been reported.119,132 Earlier studies
with extraskeletal, predominantly soft tissue, angioma- have postulated that the overexpression of neurotrans-
tosis. The angiomas in Maffucci’s syndrome typically mitters with mitogenic activities may play a role in the
involve the soft tissues or skin but occasionally can biology of the soft tissue and bone lesions of Maffucci’s
involve viscera, such as the lungs and liver.118,120,122,123,127,133 syndrome.126

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 397

A B

C D

FIGURE 6-35  ■  Ollier’s disease: microscopic features. A and B, Low power photomicrographs showing lobular growth pattern of
cartilage in enchondroma. Note encasement of cartilage lobules by well developed medullary bone trabeculae. C and D, Intermedi-
ate power photomicrographs of hyaline cartilage nodules in Ollier’s disease. Inset, Whole-mount photograph showing a growth
pattern of cartilage within the intramedullary cavity in Ollier’s disease. (A and B, ×20; C and D, ×50; inset, ×100.) (A-D, inset,
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
398 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-36  ■  Ollier’s disease: microscopic features. A-D, Low and medium power photomicrographs show increased cel-
lularity of dyschondroplastic tissue involving metaphyseal and diaphyseal areas in Ollier’s disease (A-C, ×100; D, ×200).
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 399

A B

C D

FIGURE 6-37  ■  Ollier’s disease: microscopic features. A-D, Low and intermediate power photomicrographs showing dysplastic
cartilage with high cellularity and minimal variation in size and shape of nuclei. Note the presence of binucleated chondrocytes
(A-C, ×100; D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
400 6  Benign Cartilage Lesions

Malignant Transformation nuclear atypia, and myxoid change that are more pro-
in Enchondromatosis nounced than in the benign cartilage of Ollier’s disease
(Fig. 6-41). Therefore the diagnosis of chondrosarcoma
In 1958, Jaffe86 suggested that 50% or more of patients developing in Ollier’s disease requires familiarity with the
with Ollier’s disease would have secondary malignancy, morphologic features of benign lesions in this disorder.
usually in the form of one or more chondrosarcomas, Strict correlation with radiographic and clinical data is
but the actual risk of malignant change is difficult to of great importance. The prognosis in secondary chon-
assess. More recent studies have led to a revised view drosarcoma of Ollier’s disease is exactly the same as in
of this risk. In 1987, Liu et al.88 showed that malignant ordinary conventional chondrosarcoma and is related to
neoplasms develop in approximately 30% of patients its histologic grade and affected site.
with Ollier’s disease. Usually these lesions are low-grade
conventional chondrosarcomas, but the series also con-
tained two dedifferentiated chondrosarcomas, one osteo-
sarcoma, and one chordoma. In the series reported by CHONDROBLASTOMA
Schwartz et al.,107 there was a 25% incidence of second- Definition
ary malignancy in Ollier’s disease, but the incidence in
Maffucci’s syndrome was considerably higher. A total Chondroblastoma is a distinct benign tumor character-
of 7 patients with soft tissue hemangiomas associated ized by a proliferation of immature cartilage cells with
with skeletal enchondromatosis had a total of 10 second- foci of cartilage matrix formation that typically involves
ary malignancies, 3 of which were nonskeletal. Ollier’s the epiphyses of long bones and predominantly occurs in
disease is associated with an increased risk for the devel- skeletally immature patients.
opment of extraskeletal neoplasms, particularly of central
nervous gliomas and rare ovarian tumors (granulosa cell Incidence and Location
and Sertoli-Leydig cell tumors).67,79,101,103,109,112,116 Indi-
viduals affected by Maffucci’s syndrome have a much Chondroblastoma is much less common than giant cell
higher risk for the development of extraskeletal neo- tumor, with which it has been confused in the past. It
plasms and, in addition to angiosarcoma have, similar to makes up less than 1% of bone tumors. The peak inci-
Ollier’s disease, increased risk for central nervous system dence is during the second decade of life, and approxi-
gliomas and juvenile granulosa cell tumors. In addition, mately 50% of cases are diagnosed in skeletally immature
a long list of extraskeletal neoplasms, including pituitary patients. Reported cases have been diagnosed during the
adenoma, pancreatic adenocarcinoma, and fibroadenoma first and sixth decades of life, but fewer than 10% are
of the breast, were reported in patients with Maffucci’s diagnosed during the fifth decade of life or older. There
syndrome.67,117,119,121,124 Patients with widespread skeletal is a definite male sex predominance, and the male-to-
involvement are more prone to have malignant transfor- female ratio is approximately 1.4 : 1. Chondroblastomas
mation than those who have less severe lesions or those have a predilection for the epiphyses of the major long
with localized disease limited to a few bones.84,88,99 Malig- tubular bones.141,150,204,210
nant transformation typically becomes manifest during Similar to giant cell tumor, chondroblastoma most
adult life. Therefore it can be stated that it usually occurs frequently occurs in the distal femoral and proximal tibial
after many years of continuous apparently benign growth. epiphyses. Approximately 30% of cases are diagnosed in
Cases in which secondary chondrosarcoma developed in the knee area. The third most frequently involved bone
two separate sites have also been reported.74,75,78,84,87,88,90 is the proximal humerus, followed by the proximal femur.
In summary the development of secondary malignan- Chondroblastomas also occur in the epiphyseal-equivalent
cies in Ollier’s disease and Maffucci’s syndrome is well sites of flat bones. The acetabular area of the pelvis is a
documented in the literature. In contrast, primarily due frequent site, followed by the iliac crest. Chondroblasto-
to the rarity of other variants of enchondromatosis listed mas also occur in the scapula, spine, and ribs.137,146,165,218
in Table 6-1, it is not well established. In fact, malig- Less frequent but typical sites are in the patella and
nant transformation of enchondromas has not yet been calcaneus or other tarsal bones.152,176,195 Occasionally
reported for metachondromatosis, genochondromatosis, chondroblastomas involve the craniofacial bones, most
or dysspondyloenchondromatosis. typically the base of the skull and the temporal
Clinically and radiographically, chondrosarcoma in fossa.140,157,209 Very rarely the tumor is found entirely
Ollier’s disease manifests with rapid enlargement of one within the metaphysis or diaphysis of a long bone.138,147,208
of the affected sites and the development of a mass that Rare cases of multifocal chondroblastomas with synchro-
breaks out of the bone and invades the adjacent soft tissue nous involvement of several typical sites have been
(see Figs. 6-38 and 6-39). Microscopically, it has fea- reported.198 Extremely rare, exclusively soft tissue chon-
tures of a conventional chondrosarcoma. In fact, some of droblastomas have also been reported.134,159,167 The peak
those lesions retain a lobular architecture and the overall age incidence and most frequent skeletal sites are shown
microscopic features of Ollier’s enchondromas. They are in Figure 6-42.
classified as low-grade chondrosarcomas on the basis
of their continuous locally destructive growth pattern Clinical Symptoms
and invasion of adjacent soft tissue (see Figs. 6-38 and
6-40). In other instances, chondrosarcomas developing Pain in the affected area is a constant initial symptom of
in association with Ollier’s disease show obvious micro- chondroblastoma. Pain can last for several months or
scopic features of malignancy with increased cellularity, Text continued on p. 405

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 401

A B C

F G
FIGURE 6-38  ■  Ollier’s disease with secondary chondrosarcoma: radiographic and gross features. A, Extensive involvement of lower
extremity bones by Ollier’s disease. Note the destructive lesion involving the distal femur corresponding to a secondary chondro-
sarcoma. B and C, Fat-saturated T2-weighted coronal magnetic resonance image (MRI) with contrast showing extensive involvement
of the femur and tibia by Ollier’s disease. Note a large destructive tumor mass of the distal femur (arrow) corresponding to second-
ary chondrosarcoma. D, T1-weighted axial MRI showing a low signal intensity tumor involving the distal femur. E, Fat-saturated
T2-weighted axial MRI showing lobular signal enhancement within a tumor mass of the distal femur. F, T1-weighted axial MRI with
contrast showing irregular signal enhancement corresponding to the lobular architecture of the tumor. G, Coronally bisected resec-
tion specimen of the distal femur showing a destructive cartilage mass with lobular architecture. Inset, Whole-mount photograph
showing a lobular growth pattern of a cartilage mass.

ERRNVPHGLFRVRUJ
402 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-39  ■  Ollier’s disease with secondary chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph of
proximal humerus shows focally mineralized bone surface lesion. Note the presence of intramedullary calcifications corresponding
to Ollier’sdisease. Inset, Fat-saturated T1-weighted axial magnetic resonance image (MRI) with contrast showing low signal intensity
bone surface lesion corresponding to secondary chondrosarcoma. B, Fat-saturated T2-weighted coronal MRI showing high signal
intensity within the bone surface lesion corresponding to a chondrosarcoma and the presence of multifocal high signal nodularities
within the medullary cavity representing underlying Ollier’s disease. C, Bisected resection specimen showing the proximal humerus
with a partially cystic mucinous bone surface lesion corresponding to a chondrosarcoma and the presence of a more extensive
intramedullary cartilage lesion representing underlying Ollier’s disease. D, Enlargement of the specimen in C showing intramedul-
lary Ollier’s disease growing in the form of multiple focally coalescent cartilage nodules.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 403

A B

C D
FIGURE 6-40  ■  Ollier’s disease with secondary chondrosarcoma shown in Figure 6-38: microscopic features. A-D, Low and intermedi-
ate power photomicrographs showing a hypercellular cartilage lesion corresponding to a low-grade chondrosarcoma developing
in a background of Ollier’s disease (A and C, ×100; B and D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
404 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-41  ■  Ollier’s disease with secondary chondrosarcoma shown in Figure 6-39: microscopic features. A and B, Atypical carti-
lage cells with open chromatin structure in a myxoid stroma characteristic of secondary chondrosarcoma developing in Ollier’s
disease. C and D, Hypercellular cartilage lesion with proliferation of atypical cartilage cells in a secondary chondrosarcoma develop-
ing in Ollier’s disease (A, C, and D, ×200; B, 400). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 405

Peak
incidence
10 20
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-42  ■  Chondroblastoma. Peak age incidence and most common skeletal sites of involvement. Most frequent site is indicated
by a large solid black arrow.

many years (>10 years). Chondroblastomas are epiphy- an abundance of chondroid matrix, hypercellularity, cal-
seal lesions and can be associated with joint symptoms cifications, and hemosiderin deposits.169
from the involvement of the articular cartilage or the In a typical case, the bony end plate, the cortex, and
synovium with joint effusion. the bone contour are not altered, but larger lesions may
expand the bone end and provoke some periosteal new
bone formation. These changes are most frequently seen
Radiographic Imaging
in the flat bones, such as the pelvis, scapula, and cranio-
Chondroblastoma usually presents as a sharply demar- facial bones (Figs. 6-48 and 6-49). They are also usually
cated oval or round lytic epiphyseal defect surrounded by present if chondroblastoma involves smaller bones, such
a rim of sclerotic bone.141,150,179,203,204,210,216 The lesion is as the ribs and the fibula. Chondroblastoma is one of the
usually relatively small, most frequently within the range rare lesions that can occur in the patella and in the acral
of 3 to 6 cm in diameter. In contrast to giant cell tumor, skeleton, predominantly in the calcaneus or talus (see
which is frequently an eccentric lesion, chondroblastoma Figs. 6-50 and 6-51). Marked expansion with blowout
is typically centrally located within the epiphysis (Figs. features may be present with secondary aneurysmal bone
6-43 to 6-46). In a typical case, the lesion is radiolucent, cyst formation. This complication is seen in about 15%
but occasionally it can have fine trabeculations. Occa- to 20% of chondroblastomas and is most common in
sionally some irregular calcifications can be seen on tarsal bones. Associated synovitis with collection of fluids
radiographs (Fig. 6-45). In rare cases, the lesion can in the joint adjacent to chondroblastoma is a frequent
present as a heavily calcified epiphyseal mass. On finding, but occasionally florid synovitis can be a domi-
T1-weighted MRIs, chondroblastoma is typically of low nant presenting sign.172
signal intensity.169,171 T2-weighted images typically show
signal enhancement. Multilocular signal enhancement Gross Findings
with fluid levels are typically seen when secondary aneu-
rysmal bone cyst develops in association with chondro- Tumor tissue typically represents curetted material that
blastoma (Fig. 6-47). On gadolinium-enhanced images, is reddish, gritty, and friable with calcific foci (Fig. 6-52).
lesions show marginal or septal enhancements.169 Low In rare instances when an involved bone is resected,
signal intensity on T2-weighted images correlates with Text continued on p. 416

ERRNVPHGLFRVRUJ
406 6  Benign Cartilage Lesions

A B

C E

FIGURE 6-43  ■  Chondroblastoma: radiographic features. A and B, Anteroposterior and lateral radiographs showing a well demarcated
lytic lesion of the proximal tibial epiphysis. C, Fat-saturated T1-weighted sagittal magnetic resonance image (MRI) with contrast
showing well demarcated lesion of low signal intensity involving the proximal tibial epiphysis. D and E, Fat-saturated T1-weighted
axial MRI with contrast and axial computed tomogram showing well demarcated intramedullary tibial lesion. Note mild patchy
signal enhancement in D and signal void in E.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 407

FIGURE 6-44  ■  Chondroblastoma: radiographic features. A, Anteroposterior radiograph of distal end of femur of a 29-year-old man
with heavily calcified, mummified chondroblastoma in femoral epiphysis. B, Lateral radiograph shows epiphyseal and metaphyseal
extent of tumor and its anterior location. C, Axial computed tomogram of lesion shown in A and B.

ERRNVPHGLFRVRUJ
408 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-45  ■  Chondroblastoma: radiographic features. A and B, Anteroposterior and lateral radiographs of the right knee of a
12-year-old boy with chondroblastoma of proximal tibial epiphysis. C and D, Coronal and sagittal T2-weighted magnetic resonance
image showing high signal intensity in a well demarcated intramedullary lesion involving the proximal tibial epiphysis. The sagittal
image documents the posterior epiphyseal location of chondroblastoma.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 409

A B

D E

FIGURE 6-46  ■  Chondroblastoma: radiographic features and microscopic features. A and B, Anteroposterior radiographs of proximal
humerus with a lytic well circumscribed lesion involving the humeral head. C, Axial computed tomogram showing a well demarcated
intramedullary lesion involving the humeral head. D and E, Proliferation of chondroblastic cells with well delineated polygonal or
oval cytoplasm characteristic of a chondroblastoma (D, ×400; E, ×200). (D-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
410 6  Benign Cartilage Lesions

A B C D

F G

FIGURE 6-47  ■  Chondroblastoma: radiographic features and microscopic features. A and B, Anteroposterior and oblique radiographs
of proximal femur showing a lytic lesion involving the greater trochanter. Note well demarcated intramedullary border and a thin
shell of bone outlining the expanded trochanter. C and D, Fat-saturated T2-weighted and T1-weighted coronal magnetic resonance
images (MRIs) showing low signal intensity lesion of the greater trochanter with focal signal enhancement. E and F, T1-weighted
and fat-saturated T2-weighted axial MRIs showing low signal intensity lesion with focal signal enhancement involving greater
trochanter. G, Proliferation of chondroblastic cells and scattered multinucleated osteoclastic giant cells characteristic of a chondro-
blastoma. Inset, Higher magnification showing microscopic details of chondroblastic cells (G, ×200; inset, ×400). (G and inset,
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 411

B
FIGURE 6-48  ■  Chondroblastoma: radiographic features. A, Anteroposterior radiograph of pelvis shows large chondroblastoma in-
volving ischium and acetabular portion of ilium in a 7-year-old boy. B, Axial computed tomogram shows radiolucent lesion expand-
ing into left ischium.

ERRNVPHGLFRVRUJ
412 6  Benign Cartilage Lesions

B
FIGURE 6-49  ■  Chondroblastoma: radiographic features, unusual sites. A and B, Two cases of temporal bone chondroblastomas in
men. Tumors are clearly demonstrated in magnetic resonance imaging (MRI), showing low-signal intensity in axial T1-weighted
MRI and high signal intensity in T2-weighted MRI with contrast.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 413

A C

D E
FIGURE 6-50  ■  Chondroblastoma: radiographic features, unusual sites. A and B, Lateral and sunrise views of left knee with circum-
scribed lucent lesion of patella that proves to be chondroblastoma with extensive secondary cystic changes. C, High power photo-
micrograph showing chondroblastic cells with discrete cytoplasm. D and E, Proliferation of mononuclear chondroblastic cells and
scattered multinucleated giant cells characteristic of chondroblastoma. (C, ×400; D and E, ×200.) (C-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
414 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-51  ■  Chondroblastoma: radiographic features. A, Lateral radiograph showing a lytic lesion of a 23-year-old man with chon-
droblastoma of the anterior two thirds of calcaneus. B, Conventional tomogram showing a well demarcated lytic lesion of the cal-
caneus. C, Axial computed tomogram showing a well delineated lesion within the calcaneus. D, Fat-saturated T2-weighted sagittal
magnetic resonance image showing high signal intensity in a well demarcated chondroblastoma of calcaneus.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 415

A B

C D
FIGURE 6-52  ■  Chondroblastoma: gross and microscopic features. A, Curetted tissue from chondroblastoma of humeral head shows
soft, gelatinous, red-tan tumor with flecks of chalky calcification. B, Prominent osteoclast-like giant cell component in chondroblas-
toma can suggest giant cell tumor of bone. However, intervening tumor cells are plump and polygonal. C, Typical chondroblastoma
tissue with fine, linear streaks of calcification (chicken wire calcification) outlining cells. D, Higher power photomicrograph shows
delicate chicken wire calcifications. (B and C, ×100; D, ×400). (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
416 6  Benign Cartilage Lesions

chondroblastoma examined in situ represents a well cells dominate the lesion. In such predominantly spindle-
demarcated, gray-tan mass with foci of calcification and cell lesions, typical features of chondroblastoma usually
hemorrhage; cystic change can be present. In fact, the are focally present. Extensive myxoid change and accu-
lesion can be almost entirely cystic with only a small focus mulation of pools of intercellular proteinaceous fluid can
of solid tumor tissue at the periphery. occur in some examples of chondroblastoma.
Secondary aneurysmal bone cyst in chondroblastoma
Microscopic Findings is found very frequently (in 15% to 20% of cases). In
fact, chondroblastoma is considered the prototype pre-
The current universally accepted concept of chondro- cursor condition for the development of secondary aneu-
blastoma as a separate entity distinct from giant-cell rysmal bone cyst (see Fig. 6-56 to 6-58). Small foci of
tumor was postulated by Jaffe and Lichenstein in 1942.168 blood-filled cystic spaces are frequently identified micro-
The basic cells are considered to represent chondro- scopically in these tumors. A significant component
blasts.141,179,182,185,210,220 The cytoplasm of these cells is of aneurysmal bone cyst, which can be the dominant
round to oval, and the cells have a centrally or eccentri- radiographic feature, is often present. In such cases of
cally placed oval or round nucleus that frequently exhibits secondary aneurysmal bone cyst, the presence of a chon-
a longitudinal cleft (see Figs. 6-47, 6-50 and 6-52). The droblastoma precursor may not be suspected before his-
cells are closely packed in solid areas in which scattered tologic study (also see Chapter 15).
osteoclast-like giant cells are usually seen (see Figs. 6-52 The cystic change in chondroblastoma also may be in
to 6-56). The presence of principally mononuclear cells the form of unilocular or multilocular cystic spaces filled
with scattered, multinucleated giant cells is responsible with serous fluid.166 Occasionally this type of change can
for the mimicry of giant cell tumor of bone. In fact, dominate the tumor, and a lesion can radiographically
before chondroblastoma was separated from that group, and clinically present as a cystic lesion filled with fluid.
it was designated as a calcifying epiphyseal variant of Small foci of residual chondroblastoma can be identified
giant cell tumor (Codman’s tumor).148 Within these areas by careful microscopic examination of these lesions. This
of immature cartilage cells are foci that exhibit more phenomenon should be distinguished from the secondary
distinct cartilage differentiation and formation of an aneurysmal bone cyst. Tumors with these changes are
immature cartilaginous matrix (see Figs. 6-53 to 6-55). referred to as cystic chondroblastomas.
The proportion of differentiated and immature areas can
vary considerably. In typical cases, cartilaginous matrix Special Techniques
differentiation is easily discerned. The level of cartilagi-
nous differentiation can vary, and mature hyaline matrix Ultrastructurally, chondroblastomas are composed of
is often not present. Calcification is an integral part of round or ovoid cells with deep indentations of the nuclear
chondroblastoma and serves as an important diagnostic membrane corresponding to nuclear grooves seen under
feature. The most typical pattern of calcification is in a light microscopy156,164,187,188,193,211 (Fig. 6-59). These cells
form of fine linear deposits between and around imma- are focally surrounded by a mineralized stroma that
ture mononuclear cells (chicken wire pattern) (Fig. 6-52). represents the so-called chicken wire calcifications
More abundant and coarse calcifications typically involve (Fig. 6-60). The presence of a thick nuclear lamina along
the areas with more mature hyaline cartilage matrix for- the inner membrane of the nuclear envelope is an ultra-
mation. In fact, some of the tumor can be heavily calci- structural hallmark of chondroblastoma. However, it is
fied. The patterns of calcifications in chondroblastoma also present in chondromyxoid fibroma and in a variety
are best examined with tissue that is processed without of other cells. The functional significance of this peculiar
decalcification. It is particularly important to docu- structure is not known. Immunohistochemically, chon-
ment small foci of so-called chicken wire type calcifica- droblastomas are positive for S-100 protein and vimen-
tion within the undifferentiated mononuclear areas of tin. In a typical case the S-100 protein is strongly positive
the tumor. Extensive calcification of the cartilaginous in the majority of mononuclear cells. The multinucleated
matrix affects the viability of the immature cartilage cells. giant cells are negative for S-100 protein.142,154,173,189,219
The chondrocytes within such heavily calcified areas are In cases with secondary changes (aneurysmal bone cyst,
usually dead. In well-preserved areas, the chondroblasts cystic change, and fibrohistiocytic reaction), the S-100
may show mitotic activity in excess of the usual 1 or 2 protein is positive focally only. Reports on positivity
per 10 high-power fields. Atypical mitoses are not seen. of chondroblastomas for keratins are questionable and
Chondroblastoma can have overlapping features with are subject to further verification.205 As chondroblasto-
chondromyxoid fibroma, and hybrid lesions with chon- mas develop from immature cells of cartilage lineage
droma and chondroblastoma-like appearance were also they express SOX9 protein but are in general negative
described.145 for RUNX2 and Osterix.151,175 They also express carti-
Several deviations from this classic pattern may alter lage growth plate signaling molecules such as FGFR1,
the microscopic appearance of chondroblastoma and FGFR3, bcl2, p21, parathyroid hormone-related
make its diagnosis more difficult. Spindle-cell change in hormone (PTHrP), and parathyroid hormone-related
chondroblastoma is frequently present, at least focally. peptide receptor (PTHR1).201 Some chondroblastomas
The spindle cells can represent transformed mononu- may showfocal positivity for osteoblastic lineage markers
clear cells that are an integral part of the tumor. Some- such as RUNX2 and Osterix, however. Type II colla-
times they can be of fibroblastic origin and part of a gen, the main component of cartilage matrix, is diffusely
secondary reparative process. Occasionally the spindle Text continued on p. 425

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 417

A B

C D
FIGURE 6-53  ■  Chondroblastoma: microscopic features. A, Typical chondroblastoma tissue composed of plump polygonal mono-
nuclear chondroblastic cells and prominent osteoclast-like giant cell component. B, Higher magnification of A showing loosely
arranged plump and polygonal chondroblastic cells and scattered giant cells. C, Irregular sheets of eosinophilic matrix in chondro-
blastoma. D, Higher magnification of C showing irregular deposits of eosinophilic matrix separating sheets of chondroblastic cells.
(A and C, ×100; B and D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
418 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-54  ■  Chondroblastoma: microscopic features. A, Sheets of chondroblasts and scattered osteoclast-like giant cells (×200).
B, Higher magnification of A showing nuclear details of chondroblastic cells (×400). Some of them have characteristic grooves.
C, Chondroblastoma showing ill circumscribed areas of chondroid matrix depositions (×100). D, Higher magnification showing
ill defined areas of eosinophilic matrix deposits with chondroblastic cells residing in the lacunar spaces. (A and D, ×200; B, ×400;
D, ×100). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 419

A B

C D
FIGURE 6-55  ■  Chondroblastoma: microscopic features. A, Sheets of chondroblasts in chondroid matrix (upper portion of photo-
graph). B, Higher magnification shows tumor cells within well-defined lacunae of chondroid matrix. C, Higher magnification of
chondroblastic cells with discrete cell borders. D, Cytologic details of chondroblastic cells. Note indented nuclei and occasional
nuclear grooves. (A and B, ×200; C and D, ×400). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
420 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-56  ■  Chondroblastoma: microscopic features. A and B, Cystic change chondroblastoma associated with reactive bone
formation consistent with secondary aneurysmal bone cyst in chondroblastoma. C and D, Higher magnifications of B showing
cystic changes and adjacent sheets of chondroblasts with scattered multinucleated giant cells. (A and B, ×50; C and D, ×100.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 421

A B

C D
FIGURE 6-57  ■  Chondroblastoma: microscopic features. A and B, Low and intermediate power photomicrographs showing typical
features of chondroblastoma, composed of sheets of chondroblastic cells with scattered multinucleated giant cells. C and D, Sec-
ondary aneurysmal bone cyst area in the same case of chondroblastoma. Note cystic change and the composition of thick
septa showing features of chondroblastoma with irregular chondroid matrix deposits. (A, ×100; B, ×200; C, 50; D, 100.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
422 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-58  ■  Chondroblastoma: microscopic features. A and B, Cystic change consistent with secondary aneurysmal bone cyst in
chondroblastoma. Note reactive bone formation and proliferation of sheets of chondroblastic cells within the septum. C and D, Low
and intermediate photomicrographs showing large eosinophilic cartilage matrix deposits in chondroblastoma. Note clustering of
multinucleated giant cells at the border of the matrix deposits. (A, ×50; B, ×100; C, ×100; D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 423

B
FIGURE 6-59  ■  Chondroblastoma: ultrastructural features. A, Low power magnification of chondroblastic cells in loose stromal matrix
(×5000). B, Higher magnification of A shows chondroblasts with deep indentations of nuclear membranes corresponding to nuclear
grooves seen under light microscopy (arrows). Note prominent rough endoplasmic reticulum and mitochondria. Inset, Glycogen
granules in cytoplasm. (B, ×9500; inset, ×22,000).

ERRNVPHGLFRVRUJ
424 6  Benign Cartilage Lesions

B
FIGURE 6-60  ■  Chondroblastoma: ultrastructural features. A, Low magnification of calcified area of chondroblastoma corresponding
to chicken wire mineralization shows chondroblastic cells in collagenous matrix impregnated with hydroxyapatite crystals. B, Higher
magnification of A shows a portion of chondroblast with numerous mitochondria and surface microvilli in a mineralized collagenous
stroma. (A, ×5000; B, ×19,000).

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 425

expressed in chondroblastoma.136,192 The more myxoid epiphyseal localization of chondroblastoma, helps distin-
bluish, pseudochondroid areas are predominantly com- guish such cases.
posed of aggrecan proteoglycan.135,177 Mononuclear chon- In older patients an epiphyseal cartilaginous lesion
droblastic cells express receptor activator of NFκB ligand seen radiologically should raise the question of clear-cell
(RANKL), which is a key molecule essential for regulat- chondrosarcoma. These are often well-circumscribed lytic
ing osteoclast formation and activity.160 This suggests that lesions with sclerotic borders in the end of a long bone,
RANKL may be involved in the tumor cell recruitment and they are usually more heavily calcified than chondro-
of osteoclast-like cells to chondroblastoma.160 blastomas. Furthermore, the microscopic features of
Flow cytometric studies show that typical chondro- chondroid tissue that contains clearly malignant chon-
blastomas are diploid with a low proliferation index.149,155 drocytes and the characteristic large cells with clear cyto-
Cytogenetic studies document heterogeneous arrange- plasm are not seen in chondroblastomas.
ments of individual chromosomes, with chromosomes 5
and 8 being most frequently involved.143,144,184,213,215 Recur-
Treatment and Behavior
rent breakpoints involving 2q34, 3q21-23, and 18q21 as
well as ring chromosome 4 have also been identified in Chondroblastomas are benign lesions, so curettage and
chondroblastomas.207,217 Additional rare rearrangements bone grafting are usually sufficient treatment. About 10%
such as t(5;17) (p15;q22-23) have been also identified of cases recur after curettage. They can recur within the
in chondroblastomas.200 Surprisingly, chondroblastomas bone or in the adjacent soft tissue. The recurrence of
share loss of heterozygosity on 5q, 9p, 11p, 13q, and 19q benign chondroblastoma may require a second curet-
with chondrosarcomas.191 tage.153,163,181,183,194,212 Satisfactory results of treatment of
chondroblastoma with radiofrequency ablation have been
reported; specifically, out of 17 patients treated, only 2
Differential Diagnosis
required additional surgical intervention.202 Similar to
Chondromyxoid fibroma generally affects the same age giant cell tumor, benign chondroblastoma can metasta-
group as chondroblastoma, but it usually involves the size to the lungs.158,178,190,197,214,221 These nodules are
metaphysis of long bones rather than the epiphysis. Nev- appropriately referred to as benign lung implants rather
ertheless, areas of chondroblastic differentiation can be than as true metastatic deposits. They are clinically non-
found microscopically in chondromyxoid fibroma, which progressive; that is, they do not result in the development
can lead to morphologic overlap. Calcification, which of disseminated, clinically malignant pulmonary disease.
is very common in chondroblastomas, is usually absent The benign lung implants of chondroblastoma have been
from chondromyxoid fibroma. The myxoid pseudolobu- successfully treated by surgery.
lation of the latter lesion is not seen in chondroblastomas. The term aggressive chondroblastoma is sometimes used
Although giant cells of the osteoclastic type can be in reference to lesions that are microscopically typically
very numerous in chondroblastomas, true giant cell tumors chondroblastoma but that radiographically or clinically
usually occur in skeletally mature individuals, in contrast exhibit features of local destructive growth. They usually
to chondroblastomas. The presence of chondroid matrix are larger than the typical chondroblastoma, erode the
with or without calcification in chondroblastoma excludes cortex, and invade the soft tissue at the time of diagno-
giant cell tumor. sis.139,161,162,174,188,196 Extremely rare cases of malignant
Eosinophilic granuloma of bone and intraosseous ganglion transformation in chondroblastoma have been reported
can present radiologically as epiphyseal lucencies in in the literature.170,176,180,186,199,206
children and young adults and may simulate the radio-
logic appearance of chondroblastoma. However, their Personal Comments
microscopic features render them easily separable from
this tumor. Even though chondroblastoma usually presents no diffi-
Both aneurysmal bone cyst and solitary bone cyst enter into culties in histologic diagnosis, extensive cystic change in
the differential diagnosis of chondroblastoma. Aneurys- these tumors can present diagnostic problems. Particular
mal bone cysts can become engrafted on chondroblasto- care should be exercised in evaluating aneurysmal blowout
mas and can enlarge rapidly, partially or almost completely lesions of tarsal bones, which frequently contain incon-
obliterating the chondroblastic precursor. When this spicuous chondroblastic components.
happens, the radiologic features of the lesion may be The fact that chondroblastic osteosarcomas can rarely
completely converted to those of an aneurysmal blowout present as epiphyseal lesions can occasionally lead to mis-
lesion. Only the localization to an epiphysis or a tarsal diagnoses of chondroblastoma and inadequate treatment.
bone and the young age of the patient may provide clues
that a chondroblastoma may be present. In such instances,
careful histologic study of all the curettings, including CHONDROMYXOID FIBROMA
immunohistochemical markers (S-100 protein), may be Definition
necessary to identify the chondroblasts. Solitary bone
cyst can be simulated by a chondroblastoma that has Chondromyxoid fibroma is a benign tumor composed of
undergone extensive secondary cystic change with a immature myxoid mesenchymal tissue with features of
serous-filled cavity largely replacing the tumor. The early primitive cartilaginous differentiation. It has a defi-
location of a solitary bone cyst in the metaphysis and nite predilection for the metaphyseal regions of long
diaphysis of long bones, in contrast to the typically tubular bones and is often eccentrically placed. The

ERRNVPHGLFRVRUJ
426 6  Benign Cartilage Lesions

Peak
incidence
10 30
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-61  ■  Chondromyxoid fibroma. Peak age incidence and common anatomic sites. Most frequent sites are indicated by black
arrows.

current concept and the term chondromyxoid fibroma, as fibromas also have been reported to involve the cranio-
well as its distinction from chondrosarcoma, were postu- facial bones, the ribs, the clavicle and sternum, the spine,
lated by Jaffe and Lichtenstein in 1948.240 and the short tubular bones of the hands and
feet.222,226,232,252,257,264 The peak age incidence and most
common sites of skeletal involvement are shown in
Incidence and Location
Figure 6-61.
Chondromyxoid fibroma is a rare tumor, accounting for
less than 1% of all bone tumors.228,234,247,254,266,269 It is Clinical Symptoms
definitely less common than chondroblastoma. Chondro-
myxoid fibroma has a predilection for males, with a male- Pain is the most common presenting symptom in chon-
to-female sex ratio of about 1.5 : 1, and it preferentially dromyxoid fibroma and is sometimes of several years’
affects young patients during the second or third decades duration. In some cases, local swelling of the soft tissue
of life. Individual cases, however, are widely scattered overlying the lesion can be noted clinically.
throughout the later decades, and some cases are diag-
nosed in patients who are age 50 years and older. Chon-
Radiographic Imaging
dromyxoid fibroma occurs in the metaphyseal parts of the
major tubular bones, predominantly of the lower extrem- Chondromyxoid fibromas are typically benign-appearing,
ity. Approximately one third of the cases are diagnosed oval, eccentric, metaphyseal lesions with long axes paral-
around the knee, where the proximal tibial metaphysis is lel to that of the bone.231,234,247,265,278 They may extend into
the most frequently involved site, followed by the distal the epiphyseal and diaphyseal regions, but it is unusual
femoral metaphysis. The small bones of the feet are also for the tumor to cross an open growth plate (Fig. 6-62).
involved. The bones of the upper extremities are rarely The intramedullary margins of the tumor are character-
affected. The pelvis, especially the ilium, is the most istically sharply defined with sclerotic and scalloped
frequently affected flat bone. In general, the knee area, borders (see Fig. 6-62 to 6-64). When the narrow zone
the small bones of the feet, and the ilium are the three of marginal sclerosis is lacking, the radiologic diagnosis
most frequent sites of involvement. Chondromyxoid is more difficult.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 427

C D
FIGURE 6-62  ■  Chondromyxoid fibroma: radiographic and microscopic features. A, Plain radiograph of large chondromyxoid fibroma
of distal femoral metaphysis in a 10-year-old girl. Lesion is completely radiolucent, sharply marginated, and eccentric in location.
B, Axial computed tomogram of lesion in A shows eccentric location on medial aspect of metaphysis. C and D, Intermediate and
low power photomicrographs showing loose fibromyxoid tissue gradually merging with hypercellular areas characteristic of chon-
dromyxoid fibroma (C, ×100; D, ×50) (C-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
428 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-63  ■  Chondromyxoid fibroma: radiographic features. A-D, Chondromyxoid fibromas of ilium in four adults: A, A 55-year-old
man; B, a 19-year-old man; C, a 36-year-old woman; and D, a 60-year-old man. All four lesions are contiguous with iliac crest or
sacroiliac joint.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 429

A B

C D

FIGURE 6-64  ■  Chondromyxoid fibroma: radiographic features. A and B, Lateral and oblique radiographs of eccentric metaphyseal
distal tibial chondromyxoid fibroma. Note sclerotic scalloped medullary margin. C, Lytic lesion involving the third proximal phalan-
geal base. Note the absence of eccentricity, a frequent feature of chondromyxoid fibromas when it involves short tubular bones.
D, Lytic lesion in distal phalanx of great toe of a 30-year-old man. Note central location of lesion, typical in small bones.

ERRNVPHGLFRVRUJ
430 6  Benign Cartilage Lesions

The lesions are usually totally lytic, but matrix cal- are present. Because of these cellular characteristics, a
cification can rarely be seen on radiographs. Histologic diagnosis of chondrosarcoma may be considered. Mitoses
evidence of calcification is more frequently seen than are extremely rare in this tumor. Areas of more mature
radiographic evidence. Eccentric expansion of the cortex cartilaginous differentiation with fine or coarse calcifica-
may be present; when marked, it may represent the for- tions may be present. In general, some of the microscopic
mation of a secondary aneurysmal bone cyst. Computed features of chondromyxoid fibroma overlap with those of
tomography and MRI are useful to evaluate precisely chondroblastoma. In fact, in some cases it can be difficult
the intramedullary and soft tissue components of chon- to distinguish the two entities on purely microscopic
dromyxoid fibroma. On T1-weighted MRIs, chondro- grounds. In such instances, the radiographic features help
myxoid fibromas have hypointense to intermediate signal resolve the ambiguity.
intensity. On T2-weighted images, lesions typically show Deviation from this classic pattern may cause diagnos-
some degree of signal enhancement. Contrast-enhanced tic difficulties. Extensive coarse calcification occasionally
images show either diffuse or nodular signal enhance- obscures the lobular structure of the lesion. In general,
ment.242,247,267 On fluorodeoxyglucose positron emission heavily calcified lesions are more often found in older
tomography (FDG PET) chondromyxoid fibromas are patients and in unusual sites such as the pelvis. Multi-
metabolically active and show an increased uptake of nucleated giant cells may not be present, and the exten-
FDG.237 sive myxoid change can alter the pseudolobulated
In small bones of the hands and feet, chondromyxoid structure. Often, but not universally, lesions that lack
fibromas are usually central, uniformly expanding the giant cells do not exhibit chondroblastic differentiation.
bone, but eccentric lesions can also occur104 (Fig. 6-64). An aneurysmal bone cyst may be present in association
Lesions in the flat bones (e.g., pelvis) may be large and with chondromyxoid fibroma and typically correlates
irregular in outline with prominent intralesional locula- with the radiographic features of an expansile multilocu-
tions (Fig. 6-63). Rib lesions can show fusiform expansion lated lesion.
or marked eccentricity when associated with secondary
aneurysmal bone cyst. Involvement of vertebrae may be
Special Techniques
associated with radiographic findings that deviate from
the benign appearance usually seen with chondromyxoid Ultrastructurally, chondromyxoid fibromas are composed
fibroma. The vertebral and sacral lesions may appear to of immature stellate mesenchymal cells with long, irregu-
extend beyond the cortical confines of the bone into the lar processes embedded in a loose myxoid matrix (Fig.
spinal canal.255 Examples of intracortical, juxtacortical 6-70). The cells of the pseudosepta have ultrastructural
subperiosteal as well as epiphyseal chondromyxoid features similar to those seen in chondroblastoma; that
fibroma have been described.225,233,241,246,258 is, they show inner nuclear membrane thickening and
aggregates of glycogen in their cytoplasm260,262,263 (Fig.
6-71). Immunohistochemically, the cells of the myxoid
Gross Findings
zones and the cells of hypercellular pseudosepta are
Chondromyxoid fibroma presents as a firm, gray-white weakly positive for S-100 protein.223,263,269 The cells within
lobulated mass that is sharply demarcated. The lesion is the areas that exhibit more mature chondroblastic dif-
eccentrically located and is always confined by intact ferentiation are typically strongly positive for S-100,
periosteum, even when the cortical shell is not docu- whereas the cells of the loose myxoid areas show only
mented on radiographs. Areas of semisolid, myxoid focal positivity for S-100 protein. All of these features
appearance may be identified, but chondroid foci are not indicate that chondromyxoid fibroma is cartilaginous in
usually seen grossly. nature, but it represents a less mature, earlier level of
cartilaginous differentiation than that seen in chondro-
blastoma. The chondroblastic nature of cells in chondro-
Microscopic Findings
myxoid fibroma is further confirmed by the expression of
Chondromyxoid fibroma typically has an overall pseudol- SOX9 protein.151,239,243 Similar to chondroblastomas,
obulated architecture with myxomatous and chondroid chondromyxoid fibromas are in general negative for
areas separated by zones of hypercellular mononuclear osteoblastic lineage markers such as RUNX2 and
tissue containing sparse multinucleated giant cells (Fig. Osterix.151 Expression and morphologic comparative
6-65).229,234,244,247,254,266,269 The basic component of this studies with fetal bone indicate that there is substantial
lesion seems to be immature myxoid mesenchymal tissue similarity between chondromyxoid fibroma and vascular-
that has undergone cartilaginous differentiation (see Figs. ized fetal cartilage canals.270 Chondromyxoid fibromas
6-66 to 6-68). The myxoid areas form a geographic or may show focal positivity for SMA and MSA, a feature
pseudolobulated pattern. The cellularity of myxoid areas of their partial myofibroblastic differentiation that may
composed of stellate or plump spindle cells increases at contribute to confusing this tumor with other myxoid
their peripheries (see Figs. 6-65 and 6-66). Areas of solid mesenchymal neoplasms.249 Genome-wide expression
cellularity are composed of plump, oval mononuclear studies indicate that chondromyxoid fibroma has a profile
cells representing chondroblasts; sparse multinucleated distinct from that of other cartilage neoplasms such as
giant cells may intervene between pseudolobules (Fig. chondroblastoma, enchondroma, and chondrosarcoma.250
6-69). The tumor cells within pseudolobules may be Chondromyxoid fibromas show focal deposition of type
pleomorphic and show hyperchromatism and multinu- II collagen. In addition, they may also show expression
cleation. Sometimes, large, bizarre hyperchromatic nuclei Text continued on p. 438

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 431

A B

C D
FIGURE 6-65  ■  Chondromyxoid fibroma: microscopic features. A and B, Periphery of pseudolobule shows both stellate and spindle
cells in loose myxoid stroma and sheets of more rounded cells resembling chondroblasts mixed with multinucleated giant cells.
C and D, Higher magnification of peripheral and central areas of pseudolobule shows details of chondromyxoid fibroma cells.
(A, ×100; B-D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
432 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-66  ■  Chondromyxoid fibroma: microscopic features. A and B, Intermediate and low power photomicrographs showing
loose fibromyxoid stromal tissue with scattered chondromyxoid fibroma cells. Note gradual transition from hypocellular to
hypercellular areas. C and D, Paucicellular mucinous areas of the tumor shown in A and B. (A, C, and D, ×50; B, ×100.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 433

B
FIGURE 6-67  ■  Chondromyxoid fibroma: microscopic features. A and B, Medium power photomicrographs showing stellate and
spindle cells in myxoid stroma. (A and B, ×200) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
434 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-68  ■  Chondromyxoid fibroma: microscopic features. A and B, Ill defined hypercellular areas showing chondromyxoid
fibroma cells dispersed in somewhat eosinophilic stromal tissue. C and D, Variants of stromal background in chondromyxoid fibroma
showing more eosinophilic matrix in C and a mixture of hyalinized and myxoid tissue in D resembling early chondroid matrix with
lacunar spaces. (A and C, ×50; B and D, ×100.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 435

B C
FIGURE 6-69  ■  Chondromyxoid fibroma: microscopic features. A, Hypercellular areas of chondromyxoid fibroma gradually transition-
ing to an immature chondroid matrix. B, Higher magnification of A showing early cartilage matrix formation. C, Higher magnification
of A showing hypercellular area. Note the similarity of hypercellular area of chondromyxoid fibroma to chondroblastoma. (A, x100;
B and C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
436 6  Benign Cartilage Lesions

B
FIGURE 6-70  ■  Chondromyxoid fibroma: ultrastructural features. A, Tumor cells at periphery of pseudolobule with primitive spindle
and stellate cells (left) and rounded chondroblast-like cells in septal zone (right). B, Higher magnification of stellate cell from myxoid
central zone (A, ×6000; B, ×14,000).

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 437

FIGURE 6-71  ■  Chondromyxoid fibroma: ultrastructural features. A, Septal cells of chondromyxoid fibroma show spindled
chondroblast-like cells. Inset, Abundant cytoplasmic glycogen. B, Higher magnification of A shows spindle chondroblastic cells with
granular endoplasmic reticulum and mitochondria. Inset, Indented nucleus and thick nuclear fibrous lamina (arrowheads). (A, ×6000;
inset, ×16,000; B, ×12,000; inset, ×30,000.)

ERRNVPHGLFRVRUJ
438 6  Benign Cartilage Lesions

of collagen types I, III, and VI. The less cellular myxoid site of curettage, in the adjacent soft tissue, or rarely
portions of the tumor are characterized by the presence in both.238,245,261
of hydrated proteoglycans and only minor amounts of Radiation therapy has been used to treat surgically
collagens.256 Cytogenetically, chondromyxoid fibromas inaccessible tumors. However, in rare instances the devel-
are characterized by chromosome 6 aberrations that are opment of high-grade sarcomas have been reported in
heterogeneous in nature. The commonly involved regions association with this approach.230 One case in our consul-
include 6p23-25, 6q12-15, and 6q23-27.227,235,236,248,251,259,268 tation series illustrated a potential complication of this
The rearrangements involving inv(6)(p25q13) and t(6,9) approach.269 A 20-year-old woman with chondromyxoid
(q25; q22) are the most frequent and may represent fibroma of a cervical vertebra was treated with intrale-
sole cytogenetic abnormalities or may be present in the sional excision and radiation therapy. Seven years later
background of other karyotypic anomalies. In general, she had an acute cervical spinal compression caused by a
the cytogenetic findings imply the involvement of malignant fibrous histiocytoma.
chimeric genes in the development of chondromyxoid Spontaneous malignant transformation in chondro-
fibroma, but the genetic alterations are heterogeneous myxoid fibroma is an extremely rare and somewhat con-
and result in cryptic rearrangements.224,248 Abberations troversial phenomenon. An example of such complications
involving 6q13-21 were linked to the locally aggressive is shown in Figure 6-72, which documents a chondro-
behavior of cartilage tumors, including chondromyxoid myxoid fibroma of the scapula with a history of multiple
fibroma.253 local recurrences spanning almost a decade with the
involvement of adjacent soft tissue in the form of satellite
nodules. Microscopically, the tumor showed obvious
Differential Diagnosis
atypia but retained readily recognizable features of chon-
Conventional chondrosarcoma may be confused with chon- dromyxoid fibroma.
dromyxoid fibroma when myxoid stromal changes pre-
dominate and there is prominent peripheral cellularity in Personal Comments
the lobules. The presence of large amounts of well-
formed hyaline cartilage matrix helps distinguish chon- Chondromyxoid fibromas are among the rarest of bone
drosarcoma from chondromyxoid fibroma. The latter tumors, and yet errors in diagnosis are relatively common.
usually contains little or none of that matrix. Further- Most of these errors involve myxoid chondrosarcomas
more, the central multinucleated atypical cells in chon- that are misinterpreted as chondromyxoid fibromas. If a
drosarcoma lobules most often lie within lacunae and lack significant period of time intervenes between the diagno-
the long cytoplasmic processes characteristic of chondro- sis and the first evidence of malignant behavior, these
myxoid fibroma. The resemblance to dedifferentiated chon- instances may be classified incorrectly as malignant trans-
drosarcoma can be resolved by the absence of an abrupt formation of chondromyxoid fibromas.
transition between the spindle cells and the gradual Conversely, chondromyxoid fibromas may be overdi-
merging with the central myxoid areas. In both forms of agnosed as chondrosarcomas because of the nuclear pleo-
chondrosarcoma, the radiologic findings differ markedly morphism of the stellate cells in myxoid areas and the
from the sharply defined chondromyxoid fibroma with high cellularity of the pseudosepta. Careful attention to
scalloped sclerotic borders. the radiographic features of sharp circumscription of the
The presence of zones of chondroblastic differentia- scalloped sclerotic borders in a young patient are helpful
tion surrounding the myxoid pseudolobules in some in avoiding this diagnostic pitfall. An eccentrically placed,
chondromyxoid fibromas may raise the possibility of crisply demarcated metaphyseal lesion in the long bone
chondroblastoma as the diagnosis. This morphologic of an extremity of a child or adolescent is likely to be a
overlap may extend to the radiologic features when the chondromyxoid fibroma in spite of any abundance of
normally metaphyseal chondromyxoid fibroma occupies myxoid change.
a position that bridges the growth plate into the epiphysis
of a long bone. The presence of abundant calcification
both radiologically and histologically favors the diagnosis OSTEOCHONDROMA
of chondroblastoma.
Myxomas of bone, also referred to as extragnathic fibro- By convention, osteochondroma or osteocartilaginous
myxomas, may be distinguished from chondromyxoid exostosis is considered an anomaly of the hamartomatous
fibroma by their lack of pseudolobulation and the type that occurs in two distinct clinical settings: as a
absence of pleomorphism or atypia in the stellate or solitary lesion (solitary osteochondroma) or as multiple
spindle cells. hereditary exostoses associated with other anomalies of
skeletal modeling. Because both conditions are linked to
defects in EXT1 and EXT2 genes, which are involved in
Treatment and Behavior
the development of solitary, sporadic, and hereditary
Primary and preferable treatment of chondromyxoid multifocal forms of the disease, an osteochondroma
fibroma should be en bloc resection. Simple curettage is should be considered a unique form of a benign bone
likely to leave tumor behind and contribute to recur- tumor rather than a hamartoma (Fig. 6-73). The cartilage
rence. The overall recurrence rate is approximately 20% cap in the majority of sporadic and hereditary multifocal
and does not correlate with any histologic features of osteochondromas is composed of a mixture of wild-type
the lesion. The recurrence can be within the bone at the and mutated cells.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 439

A B

C D
FIGURE 6-72  ■  Chondromyxoid fibroma with aggressive clinical behavior: microscopic features. A, Irregular paucicellular myxoid
areas gradually transitioning to areas of hypercellularity. B, Higher magnification of A showing a transition between paucicellular
and hypercellular areas of the tumor. C, Large hypercellular areas of the tumor with atypia (×100). D, Higher magnification of
C showing cellular pleomorphism and nuclear atypia of tumor cells. Inset, Higher magnification of two highly atypical tumor
cells with dense oval cytoplasm. After original treatment, the tumor showed multiple local recurrences with invasion and
satellite nodules in the adjacent soft tissue over a period of nearly a decade. (A and C, ×100; B, ×200; D, ×400; inset, ×600)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
440 6  Benign Cartilage Lesions

EXT1 EXT2
Chr 8 Chr 11
Exon 11

Exon 10

Exon 10
Exon 11

Exon 12
Exon 13
Exon 14
Exon 15
Exon 9

Exon 8

Exon 7
Exon 6
Exon 5

Exon 4

Exon 3
Exon 2

Exon 1

Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6
Exon 7

Exon 8

Exon 9
23.3 23.2 15.5
118810000

119120000
15.4

44117098

44270000
23.1 15.3
15.2
22 15.1
21.3 21.2 14
21.1
12 13
12
11.2
11.1 11.1 11.2 EXT2
11.22 11.21
11.23
EXT1 11.11 11.12
11
EXT2
12 12
13 Transmembrane helicos 13.1
13.2
21.1 Low complexity 13.4 13.3
13.5 alpha-1,4-N-
21.2 Exostosin Glycosyl transferase 64 14.1
14.2 Exostosin acetylhexosaminyltransferase
21.3 14.3
21
22.1 1 746 aa 22.1 1 718 aa
22.2 22.2
22.3 22.3
111 396 480 729 23.1 101 380 456 701
23.2
23 EXT1 7 26 29 41 23.3
24.1 24
24.2 25
24.3
1 746 aa 135006516 bp 1 718 aa
146364022 bp
Synonymous substitution Synonymous substitution
Missense substitution Missense substitution
A B
HS6ST1
HS6ST2
EXTL3
HS6ST3
FGFR HSGLCE Antithrombin GLCAT1 GALT1
6S 6S 6S 6S 6S
1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-4 1-3 1-4 1-4 

NS NS 2S FGF 2S NS NS 2S NS n
3S
EXT1 GALT2 XYLT1
NDST1 HS2ST EXT2 XYLT2
NDST2 HS3ST1–6
NDST3
NDST4 Gal Xyl GlcA IdoA GlcNAc GalNAc

C
Disaccharide repeating region Linkage region
i) in the presence of EXT1
EXT1/EXT2
Polymerization Initiation
GlcNAcT-II/GlcAT-II GlcNAcT-I

GLcUA GlcNAC GLcUA GlcNAC GLcUA GlcNAC GlcUA Gal Gal Xyl Ser

EXTL1 ? EXTL3
EXTL3 ? (EXTL2 ?)

ii) in the absence of EXT1 EXT2 EXTL2


Polymerization Initiation
GlcNAcT-II/GlcAT-II GlcNAcT-I

GLcUA GlcNAC GLcUA GlcNAC GLcUA GlcNAC GlcUA Gal Gal Xyl Ser
D

FIGURE 6-73  ■  Molecular mechanisms of multiple hereditary exostosis. A, Chromosomal location, exonal structure, and functional
domains of the EXT1 gene. B, Chromosomal location, exonal structure, and functional domains of the EXT2 gene. C, Assembly of
heparin sulfate and formation of binding sites for ligands. (C, Reprinted with permission from Bishop JR, et al: Nature 446:1030-1037,
2007.) D, Deficiency of heparin sulfate synthesis when one EXT gene is deficient in its function caused by a mutation. An example
shows the assembly of heparin sulfate when EXT1 is inactivated by a mutation. (D, Reprinted with permission from Okada M, et al:
Biochem J 428, 463-471, 2010.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 441

Initial cytogenetic studies performed on patients solitary osteochondromas are sporadic de novo lesions, a
and families affected by multiple hereditary exostosis small number have been reported to develop after exter-
identified putative loci of the disease mapping to chro- nal irradiation for malignant nonosseous tumors in child-
mosomes 8, 11, and 19.299,315,325 These data guided the hood such as Wilms’ tumor and neuroblastoma.283,300 The
cloning of EXT1 and EXT2 genes mapping to 8q24 and secondary osteochondromas have developed in metaphy-
11p11-13, respectively.271,320 Subsequently, it was shown seal areas of the vertebral column and pelvis, up to 16
that EXT1 and EXT2 genes were also involved in the years after irradiation.
development of solitary osteochondromas.
The EXT genes encode exostosin glycosylase 1 and 2, Definition
which are type II transmembrane glycoproteins that form
a Golgi-localized heterooligomeric complex involved in Solitary osteochondroma is a developmental anomaly of
heparin sulfate (HS) proteoglycan biosynthesis.274,276,286,296 bone that results in the formation of an exophytic out-
In the presence of functional EXT genes, an EXT1-EXT2 growth on the surface of bone. It is composed of a mature
complex has dual functions to initiate HS biosynthesis bony stalk that is covered by a cartilaginous cap. Other
and to polymerize HS chains. HS proteoglycans are terms applied include osteocartilaginous exostosis and simple
essential regulators of endochondral ossification, forming exostosis.
an osmotic gradient around epiphyseal chondrocytes and
controlling critical signal transduction pathways involved Incidence and Location
in the longitudinal growth of bone.274,323 In the absence
of one of the EXT genes, most often the EXT1 gene, the Solitary osteochondroma is the most common benign
remaining gene alone cannot effectively execute the HS tumorlike lesion, accounting for approximately 35% of
polymerization and a shorter HS form is synthesized in benign bone tumors and 10% of all bone tumors in any
EXT-deficient cells.287,308 The EXT genes are inactivated major series. Osteochondromas have a definite male sex
by mutations and large deletions, most likely in cartilage predominance, and the male-to-female sex ratio is 1.8 : 1.
precursor cells involved in the development of an epiphy- Osteochondromas have a predilection for the appendicu-
seal cap.289,293,294,301,311,312 Loss of HS related signal trans- lar skeleton and most frequently occur on the surface of
duction is responsible for the proliferative advantage metaphyseal portions of major long tubular bones. The
of epiphyseal chondrocytes via activation of Hedgehog knee area is most frequently affected, with approximately
and Wnt signaling. The perturbation of Hedgehog and 35% of cases occurring in this site. The proximal femoral
Wnt signaling pathways is considered to disrupt the and humeral metaphyses are the next most common sites.
pattern of enchondral ossification in the growth plate, Osteochondromas also frequently occur in the flat bones
resulting in defective formation of the bony collar.306 and often involve the ilium and scapula. They are rare
As a result, EXT mutated and HS deficient cells lose in small tubular bones of the hands and feet, in the ribs,
polarity, continue to grow, and by recruiting other normal and in the vertebral column.273,285,328 Osteochondromas
cells form an osteochondroma.279,286 In solitary osteo- practically never occur in the craniofacial bones. Figure
chondromas the inactivation of EXT genes is somatic 6-74 depicts the peak age incidence and most common
and restricted to the cartilage cap of the lesion.312 Fami- skeletal sites.
lies affected by multifocal hereditary exostosis carry
germline alterations of EXT genes in the form of point Clinical Symptoms
mutations or large deletions.312,318,322,326 In such instances,
the germline alterations are heterozygotic while the A hard swelling, usually of many years’ duration, is the
lesional tissue frequently, but not always, shows homozy- most common symptom. In rare cases a fracture, usually
gous loss of EXT function.294,302,316,329 Interestingly not at the base of the stalk, can be a presenting symptom. A
all patients with multiple hereditary osteochondromas bursa may be present over the cap, which may become
carry the alterations of EXT1/EXT2 genes, and alterna- inflamed or accumulate synovial fluid or loose bodies,
tive mechanisms with involvement of microRNA that thereby producing symptoms.275,280,284 Nerve or blood
disturbs the cartilage development signaling have been vessel impingement also may call attention to the pres-
postulated.298,327,330 ence of an osteochondroma.288,319

Solitary Osteochondroma Radiographic Imaging


(Osteocartilaginous Exostosis) The typical radiographic presentation is that of a pedun-
Osteochondromas are classified as benign bone tumors, culated surface lesion of bone arising from the metaphy-
and it was originally postulated that they result from sis (see Figs. 6-75 and 6-76). The lesion is composed of
aberrant epiphyseal development with displacement of a mature bony stalk that is continuous with the adjacent
physeal cartilage through the perichondrial fibrous ring cortex and underlying spongiosa. The long axis of the
and subsequent growth at right angles to the long axis of stalk points away from the nearest joint. The stalk is
the bone.304 Indeed, lesions indistinguishable from osteo- frequently slender, and its extremity is covered by a
chondromas have been produced experimentally by sur- lobulated cartilaginous cap that may contain calcifica-
gical implantation of growth plate cartilage beneath tions.291,305,313,317 Especially in the flat bones, the lesion can
the periosteum in animals.277 Although the majority of have a broad base that presents as a sessile surface mass

ERRNVPHGLFRVRUJ
442 6  Benign Cartilage Lesions

Peak
incidence
10 30
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 6-74  ■  Osteochondroma. Peak age incidence and most frequent sites of skeletal involvement. Most common side is indicated
by a large solid black arrow.

(see Figs. 6-76 and 6-77). The stalk can measure several This phenomenon may raise the radiographic suspicion
centimeters along its long axis, but the cartilage cap is of secondary chondrosarcoma. The body and stalk of an
typically thin (2 to 3 mm). The sessile lesions are larger, osteochondroma also may show irregular calcific densi-
and occasionally they can have a base that measures ties that represent unresorbed calcified cartilage. These
more than 10 cm in greatest dimension. A long bone may also contribute to the impression of secondary neo-
harboring a solitary osteochondroma can show slight plastic change.
localized distortion of the contour at its base, but the
overall modeling deformity, clubbing of the bone end,
Gross Findings
and growth disturbance typical of multiple hereditary
exostoses are absent. The thickness of the cartilaginous The external surface of an osteochondroma shows a lobu-
cap can occasionally not be evaluated on plain radio- lated cartilage cap covered by a fibrous membrane (peri-
graphs, but MRI can provide a more precise assessment chondrium) that is continuous with the periosteum
of thickness. Large sessile osteochondromas may be dif- covering the stalk (Fig. 6-78). The cartilage cap covers
ficult to distinguish from other cartilage-containing bone the distal part of a pedunculated osteochondroma and the
surface lesions (see Figs. 6-76 to 6-79). In such instances, elevated surface of a sessile one (see Figs. 6-78 to 6-81).
CT and MRI may be needed to evaluate the architecture The cut surface shows the hyaline composition of the
of the lesion in question. These scans are also helpful in cap, which usually measures from 2 mm to 1 cm in thick-
establishing the continuity of the lesion with the adjacent ness. At the junction of the cartilage cap with underlying
cortex and underlying spongiosa. spongiosa, a narrow, wavy, chalklike zone that corre-
An overlying bursa (exostosis bursata) may be present sponds to the zone of provisional calcification of an
external to the cartilaginous cap. The bursa can develop epiphyseal plate is present (see Figs. 6-80 and 6-81). This
synovial chondromatosis and produce a mass adjacent to reflects the endochondral ossification sequences present
the lesion that contains punctate calcifications (Fig. 6-75). Text continued on p. 450

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 443

A B

C D
FIGURE 6-75  ■  Osteochondroma: radiographic features. A, Pedunculated osteochondroma of the proximal tibial metaphysis in
a skeletally immature individual. B, Sessile osteochondroma of the proximal fibula and in a skeletally immature individual. Inset.
Axial computed tomogram (CT) showing a sessile osteochondroma originating in the fibula. Note the continuity between the
internal space of osteochondroma and the underlying medullary cavity of the involved bone. Inset, Axial CT showing an osteochon-
droma arising from the fibula. C, Pedunculated osteochondroma of the distal femoral metaphysis in a skeletally immature individual.
D, Pedunculated osteochondroma of the distal femoral metaphysis with synovial chondromatosis in bursa overlying the
cartilage cap.

ERRNVPHGLFRVRUJ
444 6  Benign Cartilage Lesions

B C

D E
FIGURE 6-76  ■  Osteochondroma: radiographic features. A, Osteochondroma involving the clavicle. B, Axial computed tomogram (CT)
showing an osteochondroma originating in the clavicle. C, Osteochondroma of the costal aspect of the scapula. D, Anteroposterior
radiograph showing a large broad-based osteochondroma arising from the neck of the left femur. E, Lateral radiograph showing a
large broad-based osteochondroma arising from the posterior aspect of the proximal tibia. Inset, T1-weighted axial magnetic reso-
nance image showing a large osteochodromatous mass arising from the posterior aspect of the tibial surface.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 445

B
FIGURE 6-77  ■  Osteochondroma: radiographic features. A, A large osteochondroma originating from the superior pubic ramus.
B, Large osteochondroma originating from the plantar surface of the calcaneus.

ERRNVPHGLFRVRUJ
446 6  Benign Cartilage Lesions

A B

C D

E F
FIGURE 6-78  ■  Osteochondroma: gross features. A and B, Surface and bisected broad-based osteochondroma arising from the proxi-
mal fibular metaphysis. Note lobulated cartilage cap. C and D, Surface and bisected peduculated osteochondroma arising from the
proximal femoral metaphysis. E and F, Bisected broad-based osteochondroma with a corresponding specimen radiograph arising
from the proximal fibular metaphysis.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 447

A B

C E
FIGURE 6-79  ■  Osteochondroma: radiographic and gross features. A and B, Oblique radiograph and axial computed tomogram (CT)
of a large osteochondroma arising from calcaneus. Inset, Bisected osteochondroma. Note the thick cartilaginous cap. C and
D, Anteroposterior radiograph and axial CT showing a large sessile osteochondroma of the clavicle. E, Bisected resection specimen
of the case in C and D showing an irregular thick cartilage cap covering the surface of this osteochondroma.

ERRNVPHGLFRVRUJ
448 6  Benign Cartilage Lesions

A B

C D

FIGURE 6-80  ■  Osteochondroma: gross features. A-D, External surface and serial sections of a large broad-based osteochondroma
arising from the iliac surface.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 449

A B

C D

FIGURE 6-81  ■  Osteochondroma: gross features. A and B, Cut surface and macrophotograph of histologic sections show relationship
of cartilage cap to underlying cancellous bone in sessile osteochondroma. C and D, Cut surface and whole-mount macrophotograph
of narrow pedicled osteochondroma show cartilage cap at surface.

ERRNVPHGLFRVRUJ
450 6  Benign Cartilage Lesions

in all osteochondromas in the growing skeleton. In adults differs from that of an osteochondroma because it con-
the cartilage cap may be extremely thin or absent because tains cytologically malignant cartilage cells without evi-
endochondral ossification and growth of the osteochon- dence of enchondral ossification at the interface
droma cease at skeletal maturation or shortly thereafter. with the underlying bony and spindle-cell components.
In young children, actively growing osteochondromas Unless a fracture has occurred in the stalk of an osteo-
typically , have a cartilaginous cap that exceeds 1 cm in cartilaginous exostosis, the cancellous bone is composed
thickness (Fig. 6-79). In such instances, the thickness of of mature lamellar trabeculae with fatty marrow. Conti-
the cap exceeding 1cm should not be used as a feature nuity with the marrow cavity of the underlying bone
of malignant transformation and the development of can be found consistently in osteochondromas, and
chondrosarcoma. Deep to the cap, the central portion of an intact cortex is usually present beneath a parosteal
the spongy bone may contain irregular islands of calcified osteosarcoma.
cartilage that can be seen grossly as gritty, opaque white Juxtacortical myositis ossificans and other posttraumatic
deposits. The spongiosum of the stalk is continuous with surface lesions (florid reactive periostitis, subungual exostosis,
the cancellous bone of the underlying medullary cavity bizarre parosteal osteocartilaginous proliferation) can also be
and is bordered by compact cortical bone that flares from confused with an osteocartilaginous exostosis. All these
the adjacent cortex. reactive processes have the distinguishing feature of an
intact cortex beneath their attachments to bone. There
Microscopic Findings is no flaring of the cortical bone into the lesion and no
continuity with the undisturbed cancellous bone of the
The cartilage cap is composed of moderately cellular adjacent medullary cavity that characterizes osteochon-
hyaline cartilage that in a young individual recapitulates dromas. The reactive bony excrescences are most com-
all the zones of an epiphyseal growth plate (see Figs. 6-82 monly found on the surfaces of short tubular bones of the
and 6-83). The periphery is covered by a fibrous peri- hands and feet.
chondrium that blends imperceptibly with the outer layer Periosteal chondroma can occasionally simulate a sessile
of hyaline cartilage. At the junction with the underlying osteochondroma on radiographs, but this lesion rests on
cancellous bone, the cartilage of the cap shows all of the an intact cortex and lacks the cancellous bone composi-
microscopic features of endochondral ossification, and it tion of an osteochondroma’s stalk.
is through this mechanism that an osteochondroma
grows during childhood and adolescence (see Figs. 6-82
Treatment and Behavior
and 6-83). This process decelerates and ceases at skeletal
maturation (Fig. 6-84). As cell replication diminishes in Osteochondromas are benign lesions with self-limited
the cartilage cap, its thickness diminishes, and it may growth that ceases after skeletal maturity. In fact, some
entirely disappear in older adults. The spongy bone osteochondromas may spontaneously regress.272,310,324
beneath the cap may contain one or more irregular They are usually treated by simple excision for cosmetic
masses of necrotic calcified chondroid, which represent reasons or when symptoms of pain, limitation of motion,
unresorbed portions of the calcified zone of the cap. or impingement on adjacent structures such as nerves and
These masses often can be seen on plain radiographs as blood vessels become clinically manifest.288,295,309,319,325
jagged opacities. The cartilage cap may show foci of Recurrence after surgical excision is rare but may
increased cellularity, and individual atypical and even develop when a portion of the cartilage cap or attached
multinucleated chondrocytes may be present. These foci perichondrium is left in situ. In solitary osteochondro-
do not form discrete nodules but blend imperceptibly mas, the risk of secondary malignant change is low
with the rest of the cap (Fig. 6-85). and is discussed later in connection with multiple
The bursa that may form at the periphery of some osteochondromas.
osteochondromas is usually intimately attached to the
perichondrium of the cap.277,284 The bursal wall is lined Multiple Hereditary Exostoses
by synovium that may show inflammatory changes with
the formation of fibrinous rice bodies. In some cases, Synonyms for multiple hereditary exostoses include
chondroid metaplasia in this bursal synovial lining can diaphyseal aclasia and multiple osteochondromas. Mul-
be associated with numerous cartilaginous loose bodies tiple hereditary exostoses are an autosomal dominant
in the bursa. If calcified, these metaplastic chondroid hereditary disorder transmitted by both sexes with
synovial nodules can simulate secondary chondrosarcoma incomplete penetrance in female patients, which results
on radiographs. in a slight male predominance. It is characterized by the
presence of multiple osteochondromas associated with
bone deformities of the affected sites (see Figs. 6-86 to
Differential Diagnosis
6-88). Growth disturbances with shortening of the
The most important lesion to distinguish from osteo- affected bones and bowing deformities of paired bones
chondroma is parosteal osteosarcoma. This surface malig- are frequently present.291,292,313,317 Osteochondromas in
nancy of bone is characteristically more radiodense this syndrome tend to be larger and more sessile
than an osteochondroma, especially at the base and with abundant lobulated cartilage caps than typical soli-
center of the bony excrescence. The cartilage cap some- tary exostoses. Similar to solitary osteochondroma,
times found at the periphery of a parosteal osteosarcoma Text continued on p. 458

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 451

B
FIGURE 6-82  ■  Osteochondroma: microscopic features. A, Low power microphotograph showing cartilage cap that is undergoing
enchondral ossification at the base with overall architectural features recapitulating the growth plate. B, Higher magnification of
A showing a zone of enchondral ossification at the interface of the cartilage cap and cancellous bone. Inset, Columns of hypertrophic
chondrocytes at the base of the cartilage cap (×200). (A,B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
452 6  Benign Cartilage Lesions

B
FIGURE 6-83  ■  Osteochondroma: microscopic features. A and B, Zones of ossification at the base of cartilage caps in enchondroma.
Inset, Whole-mount photomicrograph showing cap-shaped cartilage areas on the surface of osteochondroma transitioning via
the enchondral ossification pattern to cancellous bone within stalk. Note nodular growth pattern on some of the cartilage on surface
of osteochondroma. (A and B, ×100.) (A,B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 453

B
FIGURE 6-84  ■  Osteochondroma: microscopic features. A and B, Low power photomicrographs of two osteochondromas with thin
cartilage layer showing no active enchondral ossification pattern. Such lesions are sometimes referred to as senescent osteochon-
dromas. Insets document low cellularity within the cartilage caps. Note the absence of columnar arrangements of hypertrophic
chondrocytes and the absence of enchondral ossification, both characteristic of actively growing osteochondromas. (A and B, ×50;
insets, ×100.) (A,B, and insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
454 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-85  ■  Osteochondroma: microscopic features. A-D, Microscopic features of cartilage caps composed of mineralized hyaline
cartilage matrix with disorganized growth pattern of chondrocytes residing in lacunar spaces. Enchondral ossification is present in
A. Note the absence of columnar arrangement recapitulating epiphysis. Such features are typically seen in independently growing
nodular cartilage areas on the surface of osteochondroma (A and B, ×100; C and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 455

A B
FIGURE 6-86  ■  Multiple hereditary exostoses: radiographic features. A and B, Anteroposterior and lateral radiographs of knee and
leg of teenage patient showing involvement of proximal and distal tibial metaphyses and fibular metaphysis. Note modeling defor-
mities of metaphysis resulting in broadening and loss of metaphyseal flare.

ERRNVPHGLFRVRUJ
456 6  Benign Cartilage Lesions

A B
FIGURE 6-87  ■  Multiple hereditary exostoses: radiographic features. A, Radiograph of tibia and fibula shows proximal and distal
metaphyseal osteochondromas and modeling defects. B, Lateral radiograph of left tibia and fibula shows symmetric proximal and
distal metaphyseal involvement (same patient as in A).

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 457

A B

C D

FIGURE 6-88  ■  Osteochondroma: radiographic and gross features. A, Anteroposterior radiograph of large proximal fibular osteochon-
droma projecting medially and posteriorly to tibia. B, Arteriogram of patient in A demonstrates deviation of major vessels and
compression. C, Specimen radiograph of osteochondroma of proximal fibula in A. Cortex of stalk of this broad-based exostosis is
continuous with that of fibula. D, Gross photograph of specimen shows unremodeled calcified cartilage remnants in osteochondroma
stalk.

ERRNVPHGLFRVRUJ
458 6  Benign Cartilage Lesions

multiple hereditary exostoses have a predilection for suggest that the risk may be as low as 1% to 2%. The
the metaphyseal parts of the long tubular bones, espe- average age at the onset of secondary malignancy in
cially of the lower extremity. The knee region is invari- osteochondromas is 30 years, about 15 years earlier than
ably affected, so plain radiographs of the knees can be that of de novo conventional chondrosarcoma.
used in the initial screening for this entity. The proximal The radiographic and pathologic criteria for the diag-
femur and the proximal humerus are the next most nosis of secondary malignancy in osteochondromas are
common sites. The flat bones, such as the scapula and the same for solitary and multiple types. Clinically, malig-
pelvis, also are frequently involved. The membranous nancy can be suspected when a sudden increase in size or
bones of the skull are not affected. The disorder usually the onset of pain is noticed at the site of an osteochon-
involves bones of the extremities symmetrically. In severe droma.307 Radiographic signs of secondary malignancy
forms, widespread involvement of multiple skeletal sites include the development of areas of lucency or the pres-
with severe bony deformities and functional impairment ence of an overlying soft tissue mass containing new and
is present. separate areas of calcification (see Figs. 6-89 to 6-92).
The disorder usually is diagnosed during childhood Destruction of the base or adjacent bone is a clear-cut
at an earlier age than solitary osteochondroma, often sign of malignancy of high histologic grade. The devel-
because of the associated deformities and growth distur- opment of chondrosarcoma in large sessile osteochon-
bances. Secondary deformities of the long tubular bones dromas may be difficult to document on plain radiographic
include the so-called pseudo-Madelung’s deformity of the images (see Figs. 6-90 and 6-91). CT and MRI are typi-
forearm with ulnar shortening and bowing of the radius. cally used for precise assessment of the thickness of the
A similar deformity of the lower extremity results from cartilage cap and the presence of an independently
fibular shortening. Large osteochondromas of paired growing cartilage mass signifying the progression to
bones can produce impingement excavations on the adja- chondrosarcoma (Figs. 6-90 to 6-94). The malignancies
cent bone, a finding that can also occasionally be observed associated with solitary osteochondromas are typically
with solitary osteochondromas. In addition to the gross low-grade conventional chondrosarcomas.278,282,290,321 High-
deformity produced by large osteochondromas, shorten- grade sarcomas (malignant fibrous histiocytoma or
ing of long bones, and bowing of long bones, there is osteosarcoma) develop rarely in multiple hereditary exos-
usually clubbing of long bone ends caused by altered toses.281,303 The series reported by Garrison et al.282 indi-
bone remodeling. This results in loss of the normal cates that secondary malignancies in solitary lesions are
metaphyseal-diaphyseal flare and produces a shortened, evenly distributed regardless of anatomic site of the
broadened appearance of the femoral necks and other osteochondroma, whereas those that complicate multiple
long bone metaphyses. osteochondromas typically occur in the flat bones of the
The gross and microscopic features of osteochondro- trunk.
mas in multiple hereditary exostoses do not differ signifi- The pathologic criteria for diagnosis of low-grade
cantly from those described for solitary osteochondroma. chondrosarcoma arising in osteochondroma are based
Corrective surgery is often necessary in the manage- on gross (thickness of the cap) and microscopic features.
ment of these patients to deal with the multiple secondary Increased thickness of the cartilage cap with the forma-
deformities, growth disturbances, and functional impair- tion of discrete, grossly detectable peripheral nodules
ment. Individual symptomatic osteochondromas are that may extend outside the fibrous perichondrium is
treated by surgical removal as in the management of soli- present in the majority of typical cases. The thickened
tary osteochondroma.295,297 Lifetime surveillance of these cap usually exceeds 2 cm in the area of nodular prolif-
patients is mandated by the relatively high risk for the eration (see Figs. 6-95 and 6-96). In a typical case, the
development of secondary malignancy. development of secondary chondrosarcoma in an osteo-
chondroma is associated with the presence of a bulky
Malignant Transformation cartilaginous mass and the diagnosis is obvious on radio-
graphic imaging and gross inspection of the resected
in Osteochondroma specimen (see Figs. 6-93 and 6-94). Microscopically, the
Because a significant number of solitary osteochondro- proliferating nodular areas show increased cellularity and
mas are clinically silent and are undiagnosed, the real risk clustering of plump chondrocytes that contain enlarged
of secondary malignancy is difficult to determine. More- nuclei with an open chromatin structure. Mild to mod-
over, the data on the incidence of malignant change in erate nuclear atypia with frequent binucleate and mul-
both solitary and multiple osteochondromas are derived tinucleate cells are usually present (see Figs. 6-95 and
from surgically treated subpopulations and thereby add a 6-96). Most secondary tumors show the features of grade
selection factor. This bias contributes to the artificially 1 to 2 chondrosarcoma. Rare examples of dedifferenti-
high estimates of risk for the development of chondro- ated chondrosarcomas associated with osteochondroma
sarcoma in published reports. The reported risk for soli- have been described. In such instances, the most aggres-
tary osteochondromas is 1% to 2%, and it is 5% to 25% sive component of the lesion with features of malignant
for multiple osteochondromas. The figures for solitary fibrous histiocytoma or osteosarcoma can dominate the
osteochondroma are more likely to be inflated for the radiographic and clinical presentation.281,303,307,314 Careful
reason stated previously than those quoted for multiple analysis of radiographic data and meticulous inspection of
hereditary exostoses, a condition that is almost always the resected specimen are required to disclose the pres-
clinically symptomatic. Even in this group, analyses of ence of a preexisting condition.
pedigrees in families with multiple hereditary exostoses Text continued on p. 467

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 459

A
FIGURE 6-89  ■  Secondary chondrosarcoma arising in osteochondroma: radiographic features. A, Anteroposterior (AP) radiograph
showing a large mass arising in the intertrochanteric region of the femur representing a chondrosarcoma complicating a preexisting
osteochondroma. B, AP radiograph of a large buttock mass in a 47-year-old man with extensive chondroid matrix calcification in
grade 1 chondrosarcoma arising in an osteochondroma of the ilium.

ERRNVPHGLFRVRUJ
460 6  Benign Cartilage Lesions

B C
FIGURE 6-90  ■  Secondary chondrosarcoma arising in osteochondroma: radiographic features. A, Anteroposterior radiograph of pelvis
shows large peripheral chondrosarcoma arising in a cap of an osteochondroma. B, Enlarged view of secondary chondrosarcoma
shown in A. C, Axial computed tomogram shows large, calcified, soft tissue mass on external surface of ilium.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 461

B
FIGURE 6-91  ■  Radiation-induced osteochondroma: radiographic features. A, Anteroposterior radiograph of pelvis shows large,
broad-based osteochondroma that developed 16 years after irradiation for Wilms’ tumor when patient was 2 years old. Previous
pelvis radiographs had shown no lesion. B, Axial computed tomogram of iliac osteochondroma that developed within the portal of
radiation for Wilms’ tumor. Microscopic islands of secondary low-grade chondrosarcoma were present focally in the cartilage cap
of this radiation-induced osteochondroma. (Courtesy Jane Chatten, MD, Philadelphia.)

ERRNVPHGLFRVRUJ
462 6  Benign Cartilage Lesions

B C
FIGURE 6-92  ■  Secondary chondrosarcoma arising in osteochondroma: radiographic features. A, Anteroposterior (AP) radiograph of
pelvis showing a large calcified mass representing a chondrosarcoma arising in a broad-based osteochondroma of the pubic bone.
Inset, Axial computed tomogram of pelvis showing a calcified mass arising in osteochondroma. B, AP radiograph showing a large
calcified mass representing a secondary chondrosarcoma arising in an osteochondroma of the ischium. C, T1-weighted coronal
magnetic resonance image of the case shown in B documenting a lobulated mass representing a chondrosarcoma arising in an
osteochondroma of the ischium.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 463

B C
FIGURE 6-93  ■  Secondary chondrosarcoma arising in osteochondroma: radiographic and gross features. A, Anteroposterior radio-
graph showing a large calcified mass of the right ilium representing a chondrosarcoma arising in an osteochondroma. B, Axial
computed tomogram showing a large sessile osteochondroma of the ilium. Note a calcified mass attached to the base of osteo-
chondroma representing a secondary chondrosarcoma. C, Bisected resection specimen showing a large cartilaginous mass associ-
ated with a sessile osteochondroma of the ilium.

ERRNVPHGLFRVRUJ
464 6  Benign Cartilage Lesions

B C
FIGURE 6-94  ■  Secondary chondrosarcoma arising in osteochondroma: radiographic and gross features. A, Oblique radiograph of
pelvis showing a calcified mass arising in the osteochondroma of the right iliac crest. B, T2-weighted coronal magnetic resonance
image showing signal enhancement in a cartilage mass associated with an osteochondroma of the right iliac crest. C, Bisected
resected specimen showing a cartilage mass associated with an osteochondroma.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 465

A B

C D
FIGURE 6-95  ■  Secondary chondrosarcoma arising in osteochondroma: microscopic features. A-D, Hypercellular hyaline cartilage
mass on the surface of osteochondroma consistent with a secondary chondrosarcoma. Note cellular pleomorphism and nuclear
atypia of cartilage cells. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
466 6  Benign Cartilage Lesions

A B

C D
FIGURE 6-96  ■  Secondary chondrosarcoma arising in osteochondroma: microscopic features. A-D, Hypercellular cartilage with atypi-
cal cartilage cells consistent with a secondary chondrosarcoma associated with osteochondroma. (A and C, ×100; B and D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 467

REFERENCES 27. Takigawa K: Chondroma of the bones of the hand: a review of


110 cases. J Bone Joint Surg 53A:1591–1600, 1971.
Enchondroma 28. Teyssier JR, Ferre D: Frequent clonal chromosomal changes in
1. Alho A, Skjeldal S, Pettersen EO, et al: Aneuploidy in benign human non-malignant tumors. Int J Cancer 44:828–832, 1989.
tumors and nonneoplastic lesions of musculoskeletal tissues. 29. Unni KK: Dahlin’s bone tumors: general aspects and data on 11,087
Cancer 73:1200–1205, 1994. cases, ed 5, Philadelphia, 1996, Lippincott-Raven.
2. Bonnevialle P, Mansat M, Durroux R, et al: Chondromas of the 30. Unni KK, Inwards CY, Bridge JA, et al: Tumors of the bones and
hand: a report of 35 cases. Ann Chir Main 7:32–44, 1988. joints, Washington, DC, 2005, Armed Forces Institute of Pathol-
3. Bridge JA, Persons DL, Neff JR, et al: Clonal karyotypic aberra- ogy. series 4.
tions in enchondromas. Cancer Detect Prev 16:215–219, 1992.
4. Caballes RL: Enchondroma protuberans masquerading as osteo- Periosteal Chondroma
chondroma. Hum Pathol 13:734–739, 1982. 31. Al-Qudah AS, Abu-Ali HM, Al-Hussaini MA, et al: Periosteal
5. Crim JR, Mirra JM: Enchondroma protuberans: report of a chondroma of the clavicle: case report and review of the literature.
case and its distinction from chondrosarcoma and osteochon- Int J Surg 7:140–141, 2009.
droma adjacent to an enchondroma. Skeletal Radiol 19:431–434, 32. Bauer TW, Dorfman HD, Latham JT, Jr: Periosteal chondroma:
1990. a clinicopathologic study of 23 cases. Am J Surg Pathol 6:631–637,
6. Culver JE, Jr, Sweet DE, McCue FC: Chondrosarcoma of the 1982.
hand arising from a preexistent benign solitary enchondroma. Clin 33. Cooke GM, Pearce JG: Periosteal chondroma: report of two cases
Orthop 113:128–131, 1975. with atypical radiologic features. J Can Assoc Radiol 27:301–303,
7. Dahlin DC, Unni KK: Bone tumors, ed 4, Springfield, Ill, 1986, 1976.
Charles C Thomas. 34. DeSantos LA, Spjut HJ: Periosteal chondroma: a radiologic spec-
8. Degreef I, De Smet L: Enchondroma protuberans of the trum. Skeletal Radiol 6:15–20, 1981.
phalanx. Scand J Plastic Reconstruct Surg Hand Surg 39:315–317, 35. Fahim DK, Johnson KK, Whitehead WE, et al: Periosteal chon-
2005. droma of the pediatric cervical spine. J Neurosurg Pediatr 3:151–
9. F’eaux de Lacroix W, Dietlein M, Schmidt J, et al: Histological 156, 2009.
investigation for comparison of cartilaginous tumors of unknown 36. Hagiwara Y, Hatori M, Abe A, et al: Periosteal chondroma
biological course with unequivocal chondrosarcomas. Zentralbl of the fifth toe—a case report. Upsala J Med Sci 109:65–70,
Pathol 138:339–343, 1992. 2004.
10. Fechner RE, Mills SE: Atlas of tumor pathology: tumors of the bones 37. Inoue S, Fujino S, Kontani K, et al: Periosteal chondroma of the
and joints, Washington, DC, 1993, Armed Forces Institute of rib: report of two cases. Surg Today 31:1074–1078, 2001.
Pathology. 3rd series, fascicle 8. 38. Jaffe HL: Juxtacortical chondroma. Bull Hosp J Dis 17:20–29,
11. Fletcher CDM, Unni KK, Mertens F: Pathology & genetics: tumours 1956.
of soft tissue and bone, Lyon, 2002, World Health Organization 39. Karabakhtsian R, Heller D, Hameed M, et al: Periosteal chon-
Classification of Tumours, IARC Press. droma of the rib—report of a case and literature review. J Pediatr
12. Gunawan B, Weber M, Bergmann F, et al: Clonal chromosome Surg 40:1505–1507, 2005.
abnormalities in enchondromas and chondrosarcomas. Cancer 40. Kirchner SG, Pavlov H, Heller RM, et al: Periosteal chondromas
Genet Cytogenet 120:127–130, 2000. of the anterior tibial tubercle: two cases. AJR Am J Roentgenol
13. Ishida T, Kikuchi F, Machinami R: Histological grading and mor- 131:1088–1089, 1978.
phometric analysis of cartilaginous tumours. Virchows Arch [A] 41. Lewis MM, Kenan S, Yabut SM, et al: Periosteal chondroma:
418:149–155, 1991. a report of ten cases and review of the literature. Clin Orthop
14. Jacobson ME, Ruff ME: Solitary enchondroma of the phalanx. 256:185–192, 1990.
J Hand Surg [Am] 36:1845–1847, 2011. 42. Lichtenstein L, Hall JE: Periosteal chondroma: a distinctive
15. Kusuzaki K, Murata H, Takeshita H, et al: Usefulness of cytofluo- benign cartilage tumor. J Bone Joint Surg 34A:691–697, 1952.
rometric DNA ploidy analysis in distinguishing benign cartilagi- 43. Lisanti M, Buongiorno L, Bonnicoli E, et al: Periosteal chon-
nous tumors from chondrosarcomas. Mod Pathol 12:863–872, droma of the proximal radius: a case report. Chir Degli Organi de
1999. Movimento 90:403–407, 2005.
16. Kuur E, Hansen SL, Lindequist S: Treatment of solitary enchon- 44. Luevitoonvechkij S, Arphornchayanon O, Leerapun T, et al:
dromas in fingers. J Hand Surg 14B:109–112, 1989. Periosteal chondroma of the proximal humerus: a case report
17. Mahajan RH, Dalal RB, Sahu A, et al: Secondary chondrosarcoma and review of the literature. J Med Assoc Thailand 89:1970–1975,
in an enchondroma in a proximal phalanx of toe. A diagnostic 2006.
problem. A case report. Foot 19:62–64, 2009. 45. Mandahl N, Mertens F, Willen H, et al: Rearrangement of band
18. Matysiakiewicz J, Tomasik P, Miszczyk L, et al: Manifestations, q13 on both chromosomes 12 in a periosteal chondroma. Gene
diagnosis and surgical treatment of enchondroma—own experi- Chromosome Cancer 6:121–123, 1993.
ence. Ortop Trauma Rehabil 12:155–159, 2010. 46. Marmor L: Periosteal chondroma (juxtacortical chondroma). Clin
19. Mirra JM, Gold R, Downs J, et al: A new histologic approach Orthop 37:150–153, 1964.
to the differentiation of enchondroma and chondrosarcoma of 47. Nojima T, Unni KK, McLeod RA, et al: Periosteal chondroma
the bones: a clinicopathologic analysis of 51 cases. Clin Orthop and periosteal chondrosarcoma. Am J Surg Pathol 9:666–677,
201:214–237, 1985. 1985.
20. Morii T, Mochizuki K, Tajima T, et al: Treatment outcome of 48. Nosanchuk JS, Kaufer H: Recurrent periosteal chondroma: report
enchondroma by simple curettage without augmentation. J Orthop of two cases and a review of the literature. J Bone Joint Surg
Sci 15:112–117, 2010. 51A:375–380, 1969.
21. Noble J, Lamb DW: Enchondromata of bones of the hand: a 49. Parodi KK, Farrett W, Paden MH, et al: A report of a rare pha-
review of 40 cases. Hand 6:275–284, 1974. langeal periosteal chondroma of the foot. J Foot Ankle Surg
22. Potter BK, Freedman BA, Lehman RA, Jr, et al: Solitary epiphy- 50:122–125, 2011.
seal enchondromas. J Bone Joint Surg Am 87:1551–1560, 2005. 50. Pazzaglia UE, Ceciliani L: Periosteal chondroma of the humerus
23. Ryzewicz M, Manaster BJ, Naar E, et al: Low-grade cartilage leading to shortening: a case report. J Bone Joint Surg Br 67:290–
tumors: diagnosis and treatment. Orthopedics 30:35–46, 2007. 292, 1985.
24. Sakai Junior N, Abe KT, Formigli LM, et al: Cytogenetic findings 51. Woertler K, Blasius S, Brinkschmidt C, et al: Periosteal chon-
in 14 benign cartilage neoplasms. Cancer Genet 204:180–186, droma: MR characteristics. J Computer Assisted Tomogr 25:425–
2011. 430, 2001.
25. Sawyer JR, Swanson CM, Lukacs JL, et al: Evidence of an associa- 52. Yildirim C, Ynay K, Rodop O, et al: Periosteal chondroma that
tion between 6q13-21 chromosome aberrations and locally presented as a subcutaneous mass in the ring finger. J Plastic Surg
aggressive behavior in patients with cartilage tumors. Cancer Hand Surg 45:117–120, 2011.
82:474–483, 1998. 53. Zhao XG, Chen G, Chen XF: Giant periosteal chondroma of the
26. Schajowicz F: Tumors and tumorlike lesions of bone, ed 2, Berlin, rib associated with mediastinum deviation. Ann Thoracic Surg
1994, Springer-Verlag. 94:1014, 2012.

ERRNVPHGLFRVRUJ
468 6  Benign Cartilage Lesions

Enchondromatosis 81. Dang L, White DW, Gross S, et al: Cancer-associated IDH1


54. Bhargava R, Leonard NJ, Chan AK, et al: Autosomal dominant mutations produce 2-hydroxyglutarate. Nature 462:739–744,
inheritance of spondyloenchondrodysplasia. Am J Med Genet A 2009.
135:282–288, 2005. 82. Fairbank HAT: Dyschondroplasia. Synonyms: Ollier’s disease,
55. Chagnon S, Lacert P, Blery M: Spondyloenchondrodysplasia. multiple enchondromata. J Bone Joint Surg Br 30:689–708, 1948.
J Radiol 66:75–77, 1985. 83. Figueroa ME, Abdel-Wahab O, Lu C, et al: Leukemic IDH1 and
56. Freisinger P, Finidori G, Maroteaux P: Dysspondylochondroma- IDH2 mutations result in a hypermethylation phenotype, disrupt
tosis. Am J Med Genet 45:460–464, 1993. TET2 function, and impair hematopoietic differentiation. Cancer
57. Isidor B, Guillard S, Hamel A, et al: Genochondromatosis type Cell 18:553–567, 2010.
II: report of a new patient and further delineation of the pheno- 84. Goodman SB, Bell RS, Fornasier VL, et al: Ollier’s disease with
type. Am J Med Genet A 143A:1919–1921, 2007. multiple sarcomatous transformations. Hum Pathol 15:91–93,
58. Kennedy LA: Metachondromatosis. Radiology 148:117–118, 1983. 1984.
59. Kozlowski K, Brostrom K, Kennedy J, et al: Dysspondyloenchon- 85. Hopyan S, Gokgoz N, Poon R, et al: A mutant PTH/PTHrP type
dromatosis in the newborn. Report of four cases. Pediatr Radiol I receptor in enchondromatosis. Nat Genet 30:306–310, 2002.
24:311–315, 1994. 86. Jaffe HL: Tumors and tumorous conditions of the bones and joints,
60. Kozlowski K, Jarrett J: Genochondromatosis II. Pediatr Radiol Philadelphia, 1958, Lea & Febiger.
22:593–595, 1992. 87. Kosaki N, Yabe H, Anazawa U, et al: Bilateral multiple malignant
61. Kozlowski KS, Masel J: Distinctive enchondromatosis with spine transformation of Ollier’s disease. Skelet Radiol 34:477–484, 2005.
abnormality, regressive lesions, short stature, and coxa vara: 88. Liu J, Hudkins PG, Swee RG, et al: Bone sarcomas associated
importance of long-term follow-up. Am J Med Genet 107:227– with Ollier’s disease. Cancer 59:1376–1385, 1987.
232, 2002. 89. Lu C, Ward PS, Kapoor GS, et al: IDH mutation impairs histone
62. Le Merrer M, Fressinger P, Maroteaux P: Genochondromatosis. demethylation and results in a block to cell differentiation. Nature
J Med Genet 28:485–489, 1991. 483:474–478, 2012.
63. Maffucci A: Di un caso di enchondroma et angioma multiplo: 90. Lucas D, Tupler R, Enneking WF: Multicentric chondrosarcomas
contribuzione alla genesi embrionale dei tumori. Mov Med Chir associated with Ollier’s disease: review and case report. J Fla Med
Nap 13:399–412, 565-575, 1881. Assoc 77:24–28, 1990.
64. Maroteaux P: Metachondromatosis. Z Kinderheilkd 109:246–261, 91. Ly JQ, Beall DP: A rare case of infantile Ollier’s disease demon-
1971. strating bilaterally symmetric extremity involvement. Skel Radiol
65. Menger H, Kruse K, Spranger J: Spondyloenchondrodysplasia. 32:227–230, 2003.
J Med Genet 26:93–99, 1989. 92. Mahafza WS: Multiple enchondromatosis Ollier’s disease with
66. Ollier L: De la dyschondroplasie. Bull Soc De Chir Lyon 3:22–24, two primary brain tumors. Saudi Med J 25:1261–1263, 2004.
1900. 93. Mardis ER, Ding L, Dooling DJ, et al: Recurring mutations
67. Pansuriya TC, Kroon HM, Bovee JV: Enchondromatosis: insights found by sequencing an acute myeloid leukemia genome. N Engl
on the different subtypes. Int J Clin Exper Pathol 3:557–569, J Med 361:1058–1066, 2009.
2010. 94. Martson A, Haviko T, Kirjanen K: Extensive limb lengthening in
68. Schorr S, Legum C, Ochshorn M: Spondyloenchondrodysplasia. Ollier’s disease: 25-year follow-up. Medicina 41:861–866, 2005.
Enchondromatosis with severe platyspondyly in two brothers. 95. McKusick VA: Mendelian inheritance in man: Catalogs of human
Radiology 118:133–139, 1976. genes and genetic disorders. ed 12, Baltimore, 1998, Johns Hopkins
69. Spranger J, Kemperdieck H, Bakowski H, et al: Two peculiar University Press.
types of enchondromatosis. Pediatr Radiol 7:215–219, 1978. 96. Mitchell ML, Ackerman LV: Case report 405: Ollier disease
70. Zack P, Beighton P: Spondyloenchondromatosis: syndromic iden- (enchondromatosis). Skeletal Radiol 16:61–66, 1987.
tity and evolution of the phenotype. Am J Med Genet 55:478–482, 97. Pansuriya TC, van Eijk R, d’Adamo P, et al: Somatic mosaic IDH1
1995. and IDH2 mutations are associated with enchondroma and spindle
cell hemangioma in Ollier disease and Maffucci syndrome. Nature
Genet 43:1256–1261, 2011.
Ollier’s Disease 98. Pansuriya TC, Oosting J, Krenacs T, et al: Genome-wide analysis
71. Amary MF, Bacsi K, Maggiani F, et al: IDH1 and IDH2 mutations of Ollier disease: is it all in the genes? Orphanet J Rare Dis 6:2,
are frequent events in central chondrosarcoma and central and 2011.
periosteal chondromas but not in other mesenchymal tumours. 99. Pravda J, Dorfman HD: Radiologic manifestations of Ollier’s
J Pathol 224:334–343, 2011. disease. Einstein Quart J Biol Med 7:80–93, 1989.
72. Baumgart R, Burklein D, Hinterwimmer S, et al: The manage- 100. Prensner JR, Chinnaiyan AM: Metabolism unhinged: IDH muta-
ment of leg-length discrepancy in Ollier’s disease with a fully tions in cancer. Nat Med 17:291–293, 2011.
implantable lengthening nail. J Bone Joint Surg Br 87:1000–1004, 101. Ranger A, Szymczak A: The association between intracranial
2005. tumours and multiple dyschondroplasia (Ollier’s disease or Maf-
73. Bendel CJ, Gelmers HJ: Multiple enchondromatosis (Ollier’s fucci’s syndrome): do children and adults differ? J Neuro-Oncol
disease) complicated by malignant astrocytoma. Eur J Radiol 95:165–173, 2009.
12:135–137, 1991. 102. Reitman ZJ, Jin G, Karoly ED, et al: Profiling the effects of iso-
74. Bovee JV, van Roggen JF, Cleton-Jansen AM, et al: Malignant citrate dehydrogenase 1 and 2 mutations on the cellular metabo-
progression in multiple enchondromatosis (Ollier’s disease): an lome. PNAS 108:3270–3275, 2011.
autopsy-based molecular genetic study. Hum Pathol 31:1299–1303, 103. Rietveld L, Nieboer TE, Kluivers KB, et al: First case of juvenile
2000. granulose cell tumor in an adult with Ollier disease. Int J Gynecol
75. Bukte Y, Necmioglu S, Nazaroglu H, et al: A case of multiple Pathol 28:464–467, 2009.
chondrosarcomas secondary to severe multiple symmetrical 104. Rozeman LB, Hameetman L, van Wezel T, et al: cDNA expres-
enchondromatosis (Ollier’s disease) at an early age. Clin Radiol sion profiling of chondrosarcomas: Olllier disease resembles soli-
60:1306–1310, 2005. tary tumours and alteration in genes coding for components of
76. Cairns RA, Harris IS, Mak TW: Regulation of cancer cell metabo- energy metabolism occurs with increasing grade. J Pathol 207:61–
lism. Nat Rev 11:85–95, 2011. 71, 2005.
77. Campanacci M: Bone and soft tissue tumors, New York, 1986, 105. Rozeman LB, Sangiorgi L, Briaire-de Bruijn IH, et al: Enchon-
Springer-Verlag. dromatosis (Ollier disease, Maffucci syndrome) is not caused by
78. Cannon SR, Sweetnam DR: Multiple chondrosarcomas in dys- the PTHR1 mutation p.R150C. Hum Mutat 24:466–473, 2004.
chondroplasia (Ollier’s disease). Cancer 55:836–840, 1985. 106. Schipani E, Kruse K, Juppner H: A constitutively active mutant
79. Chang S, Prados MD: Identical twins with Ollier’s disease and PTH-PHTrP receptor in Jansen-type metaphyseal chondrodys-
intracranial gliomas: case report. Neurosurgery 34:903–906, 1994. plasia. Science 268:98–100, 1995.
80. Couvineau A, Wouters V, Bertrand G, et al: PTHR1 mutations 107. Schwartz HS, Zimmerman NB, Simion MA, et al: The malignant
associated with Ollier disease result in receptor loss of function. potential of enchondromatosis. J Bone Joint Surg 69A:269–274,
Hum Molec Genet 17:2766–2775, 2008. 1987.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 469

108. Smolkova K, Jezek P: The role of mitochondrial NADPH- Chondroblastoma


dependent isocitrate dehydrogenase in cancer cells. Int J Cell Biol 134. Abdul-Karim FW, Ayala AG, Spjut HJ: Case report 321: extraos-
273947:2012, 2012. seous chondroblastoma in the subcutaneous tissues of the right
109. Tamimi HK, Bolen JW: Enchondromatosis (Ollier’s disease) and shoulder. Skeletal Radiol 14:73–75, 1985.
ovarian juvenile granulosa cell tumor. Cancer 53:1605–1608, 1984. 135. Aigner T, Loos S, Inwards C, et al: Chondroblastoma is an
110. Tellisi N, Llizarov S, Fragomen AT, et al: Humeral lengthening osteoid-forming, but not cartilage-forming neoplasm. J Pathol
and deformity correction in Ollier’s disease: distraction osteogen- 189:463–469, 1999.
esis with a multiaxial correction frame. J Pediatr Orthop 17:152– 136. Aigner T, Dertinger S, Vornehm SI, et al: Phenotypic diversity of
157, 2008. neoplastic chondrocytes and extracellular matrix gene expression
111. Tomlinson PJ, Turner J, Monsell FP: The distribution of enchon- in cartilaginous neoplasms. Am J Pathol 150:2133–2141, 1997.
dromata in the hands of patients with Ollier’s disease. J Hand Surg 137. Akai M, Tateishi A, Machinami R, et al: Chondroblastoma of the
Eur 35:154–155, 2010. sacrum: a case report. Acta Orthop Scand 57:378–381, 1986.
112. Velasco-Oses A, Alonso-Alvaro A, Blanco-Pozo A, et al: Ollier’s 138. Azorin D, Gonzalez-Mediero I, Colmenero I, et al: Diaphyseal
disease associated with ovarian juvenile granulosa cell tumor. chondroblastoma in a long bone: first report. Skelet Radiol 35:49–
Cancer 62:222–225, 1988. 52, 2006.
113. Wallace DC: Mitochondria and cancer. Nat Rev 12:685–697, 139. Bandyopadhyay A, Chakraborty J, Choudhuri MK, et al: Recur-
2012. rent and aggressive chondroblastoma: a case report. Ind J Pathol
114. Wang P, Dong Q, Zhang C, et al: Mutations in isocitrate dehy- Microbiol 50:780–782, 2007.
drogenase 1 and 2 occur frequently in intrahepatic cholangiocar- 140. Bertoni F, Unni KK, Beabout JW, et al: Chondroblastoma of the
cinomas and share hypermethylation targets with gliobastomas. skull and facial bones. Am J Clin Pathol 88:1–9, 1987.
Oncogene 1-10:2012. 141. Bloem JL, Mulder JD: Chondroblastoma: a clinical and radiologi-
115. Ward PS, Cross JR, Lu C, et al: Identification of additional IDH cal study of 104 cases. Skeletal Radiol 14:1–9, 1985.
mutations associated with oncometabolite (R)-2-hydroxyglutarate 142. Brecher ME, Simon MA: Chondroblastoma: an immunohisto-
production. Oncogene 31:2491–2498, 2012. chemical study. Hum Pathol 19:1043–1047, 1988.
116. Weyl-Ben Arush M, Oslander L: Ollier’s disease associated with 143. Bridge JA, Bhatia PS, Anderson JR, et al: Biologic and clinical
ovarian Sertoli-Leydig cell tumor and breast adenoma. Am J significance of cytogenetic and molecular cytogenetic abnormali-
Pediatr Hematol Oncol 13:49–51, 1991. ties in benign and malignant cartilaginous lesions. Cancer Genet
Cytogenet 69:79–90, 1993.
Maffucci’s Syndrome 144. Buddingh EP, Naumann S, Nelson M, et al: Cytogenetic findings
117. Bach AD, Walgenbach KJ, Horch RE: Hemangiosarcoma of the in benign cartilaginous neoplasms. Cancer Genet Cytogenet 141:
left hand in a patient with the rare combination of Maffucci’s and 164–168, 2003.
Stewart Treves syndrome. Vasa 29:71–73, 2000. 145. Cates JM, Rosenberg AE, O’Connell JX, et al: Chondroblastoma-
118. Bean WB: Dyschondroplasia and hemangiomata (Maffucci’s syn- like chondroma of soft tissue: an underrecognized variant and its
drome). Arch Intern Med 102:544–550, 1958. differential diagnosis. Am J Surg Pathol 25:661–666, 2001.
119. Davidson TI, Kissin MW, Bradish CF, et al: Angiosarcoma arising 146. Chung OM, Yip SF, Ngan KC, et al: Chondroblastoma of the
in a patient with Maffucci syndrome. Eur J Surg Oncol 11:381– lumbar spine with cauda equine syndrome. Spinal Cord 41:359–
384, 1985. 364, 2003.
120. Jermann M, Eid K, Pfammatter T, et al: Maffucci’s syndrome. 147. Clapper AT, DeYoung BR: Chondroblastoma of the femoral
Circulation 104:2001, 1693. diaphysis: report of a rare phenomenon and review of literature.
121. Lewis RJ, Ketcham AS: Maffucci’s syndrome: functional and neo- Hum Pathol 38:803–806, 2007.
plastic significance. J Bone Joint Surg 55A:1465–1479, 1973. 148. Codman EA: Epiphyseal chondromatous giant cell tumors of
122. Lobo-Mueller E, Amaral JG, Babyn PS, et al: Complex combined the upper end of the humerus. Surg Gynecol Obstet 52:543–548,
vascular malformations and vascular malformation syndromes 1931.
affecting the extremities in children. Semin Musculoskel Radiol 149. Cuvelier CA, Roels HJ: Cytophotometric studies of the nuclear
13:255–276, 2009. DNA content in cartilaginous tumors. Cancer 44:1363–1374,
123. McDermott AL, Dutt SN, Chavda SV, et al: Maffucci’s syndrome: 1979.
clinical and radiological features of a rare condition. J Laryngol 150. Dahlin DC, Ivins JC: Benign chondroblastoma: a study of 125
Otol 115:845–847, 2001. cases. Cancer 30:401–413, 1972.
124. Miyake M, Tateishi U, Maeda T, et al: MR features of angiosar- 151. Dancer JY, Henry SP, Bondaruk J, et al: Expression of master
coma in a patient with Maffucci’s syndrome. Radiat Med 23:508– regulatory genes controlling skeletal development in benign car-
512, 2005. tilage and bone forming tumors. Hum Pathol 41:1788–1793, 2010.
125. Moore BA, Rutter MJ, Cotton R, et al: Maffucci’s syndrome and 152. Davila JA, Amrami KK, Sundaram M, et al: Chondroblastoma of
cartilaginous neoplasms of the trachea. Otolaryngol Head Neck Surg the hands and feet. Skelet Radiol 33:582–587, 2004.
128:583–586, 2003. 153. De Silva MV, Reid R: Chondroblastoma: varied histologic appear-
126. Robinson D, Tieder M, Halperin N, et al: Maffucci’s syndrome— ance, potential diagnostic pitfalls, and clinicopathologic features
the result of neural abnormalities? Evidence of mitogenic neu- associated with local recurrence. Ann Diagn Pathol 7:205–213,
rotransmitters present in enchondromas and soft tissue 2003.
hemangiomas. Cancer 74:949–957, 1994. 154. Edel G, Ueda Y, Nakanishi J, et al: Chondroblastoma of bone:
127. Shepherd V, Godbolt A, Casey T: Maffucci’s syndrome with a clinical, radiological, light and immunohistochemical study.
extensive gastrointestinal involvement. Australas J Dermatol Virchows Arch [A] 421:355–366, 1992.
46:33–37, 2005. 155. El Naggar AK, Hurr K, Tu ZN, et al: DNA and RNA content
128. Sugiura I: Maffucci’s syndrome, Nagoya, Japan, 1992, Maruzen analysis by flow cytometry in the pathobiologic assessment of
Nagoya. bone tumors. Cytometry 19:256–262, 1995.
129. Sun GH, Myer CM, 3rd: Otolaryngologic manifestations of Maf- 156. Fadda M, Manunta A, Rinonapoli G, et al: Ultrastructural appear-
fucci’s syndrome. Int J Pediatr Otorhinolaryngol 73:1015–10158, ance of chondroblastoma. Int Orthop 18:389–392, 1994.
2009. 157. Feely M, Keohane C: Chondroblastoma of the skull. J Neurol
130. Sun TC, Swee RG, Shives TC, et al: Chondrosarcoma in Maf- Neurosurg Psychiatry 47:1348–1350, 1984.
fucci’s syndrome. J Bone Joint Surg 67A:1214–1219, 1985. 158. Green P, Whittaker RP: Benign chondroblastoma: case report
131. Tachibana E, Saito K, Takahashi M, et al: Surgical treatment of a with pulmonary metastasis. J Bone Joint Surg 57A:418–420, 1975.
massive chondrosarcoma in the skull base associated with Maf- 159. Hameed MR, Blacksin M, Das K, et al: Cortical chondroblas-
fucci’s syndrome: a case report. Surg Neurol 54:165–169, 2000. toma: report of a case and literature review of this lesion reported
132. Tsuchiya H, Tomita K, Yasutake H, et al: Maffucci’s syndrome in unusual locations. Skel Radiol 35:295–297, 2006.
combined with dedifferentiated chondrosarcoma. Arch Orthop 160. Huang L, Cheng YY, Chow LT, et al: Receptor activator of
Trauma Surg 110:269–272, 1991. NF-kappaB ligand (RANKL) is expressed in chondroblastoma:
133. Williams DS: Maffucci’s syndrome. J Insurance Med 40:146–147, possible involvement in osteoclastic giant cell recruitment. Mol
2008. Pathol 56:116–120, 2003.

ERRNVPHGLFRVRUJ
470 6  Benign Cartilage Lesions

161. Hull MT, Gonzalez-Crussi F, DeRosa GP, et al: Aggressive chon- 186. McLaughlin RE, Sweet DE, Webster T, et al: Chondroblastoma
droblastoma: report of a case with multiple bone and soft tissue of the pelvis suggestive of malignancy. J Bone Joint Surg 57A:549–
involvement. Clin Orthop 126:261–265, 1977. 551, 1975.
162. Huvos AG, Higinbotham NL, Marcove RC, et al: Aggressive 187. Mii Y, Miyauchi Y, Morishita T, et al: Ultrastructural cytochemi-
chondroblastoma: review of the literature on aggressive behavior cal demonstration of proteoglycans and calcium in the extracel-
and metastases with a report of one new case. Clin Orthop lular matrix of chondroblastomas. Hum Pathol 25:1290–1294,
126:266–272, 1977. 1994.
163. Huvos AG, Marcove RC: Chondroblastoma of bone: a critical 188. Mirra JM, Ulich TR, Eckardt JJ, et al: “Aggressive” chondroblas-
review. Clin Orthop 95:300–312, 1973. toma: light and ultramicroscropic findings after en bloc resection.
164. Huvos AG, Marcove RC, Erlandson RA, et al: Chondroblastoma Clin Orthop 178:276–284, 1983.
of bone: a clinicopathologic and electron microscopic study. 189. Monda L, Wick MR: S-100 protein immunostaining in the dif-
Cancer 29:760–771, 1972. ferential diagnosis of chondroblastoma. Hum Pathol 16:287–293,
165. Ilaslan H, Sundaram M, Unni KK: Vertebral chondrobastoma. 1985.
Skel Radiol 32:66–71, 2003. 190. Ozkoc G, Gonlusen G, Ozalay M, et al: Giant chondroblastoma
166. Ippolito E, Tudisco C, Mariani PP: Rare forms of chondroblas- of the scapula with pulmonary metastases. Skeletal Radiol 35:42–
toma: cystic and diaphyseal types. Ital J Orthop Traumatol 12:455– 48, 2006.
460, 1986. 191. Papachristou DJ, Goodman MA, Cieply K, et al: Comparison of
167. Ishida T, Goto T, Motoi N, et al: Intracortical chondroblastoma allelic losses in chondroblastoma and primary chondrosarcoma of
mimicking intra-articular osteoid osteoma. Skelet Radiol 31:603– bone and correlation with fluorescence in situ hybridization analy-
607, 2002. sis. Hum Pathol 37:890–898, 2006.
168. Jaffe HL, Lichtenstein L: Benign chondroblastoma of bone: a 192. Park HR, Park YK, Jang KT, et al: Expression of collagen type II,
re-interpretation of the so-called calcifying or chondromatous S100B, S100A2 and osteocalcin in chondroblastoma and chondro-
giant cell tumor. Am J Pathol 18:969–991, 1942. myxoid fibroma. Oncol Rep 9:1087–1091, 2002.
169. Jee WH, Park YK, McCauley TR, et al: Chondroblastoma: MR 193. Povysil C, Matejovsky Z: Ultrastructure of benign chondroblas-
characteristics with pathologic correlation. J Computer Assisted toma. Pathol Res Pract 166:80–89, 1979.
Tomography 23:721–726, 1999. 194. Ramappa AJ, Lee FY, Tang P, et al: Chondroblastoma of bone.
170. Kahn LB, Wood FM, Ackerman LV: Malignant chondroblastoma: J Bone Joint Surg Am 82-A:1140–1145, 2000.
report of two cases and review of the literature. Arch Pathol 195. Remagen W, Schafer R, Roggatz J: Chondroblastoma of the
88:371–376, 1969. patella. Arch Orthop Trauma Surg 96:157–158, 1980.
171. Kaim AH, Hugli R, Bonel HM, et al: Chondroblastoma and clear 196. Reyes CV, Kathuria S: Recurrent and aggressive chondroblastoma
cell chondrosarcoma: radiological and MRI characteristics with of the pelvis with late malignant neoplastic changes. Am J Surg
histopathological correlation. Skel Radiol 31:88–95, 2002. Pathol 3:449–455, 1979.
172. Kaneko H, Kitoh H, Wasa J, et al: Chondroblastoma of the 197. Riddell RJ, Louis CJ, Bromberger NA: Pulmonary metastases
femoral neck as a cause of hip synovitis. J Pediatr Orthop B 21:179– from chondroblastoma of the tibia: report of a case. J Bone Joint
182, 2012. Surg Br 55:848–853, 1973.
173. Karabela-Bouropoulou V, Markaki S, Prevedorou D, et al: A com- 198. Roberts PF, Taylor JG: Multifocal benign chondroblastomas:
bined immunohistochemical and histochemical approach on the report of a case. Hum Pathol 11:296–298, 1980.
differential diagnosis of giant cell epiphyseal neoplasms. Pathol Res 199. Rodgers WB, Mankin JH: Metastatic malignant chondroblas-
Pract 184:184–187, 1989. toma. Am J Orthop 25:846–849, 1996.
174. Kirchhoff C, Buhmann S, Mussack T, et al: Aggressive scapular 200. Romeo S, Szyhai K, Nishimori I, et al: A balanced t(5;17)
chondroblastoma with secondary metastasis—a case report and (p15;q22-23) in chondroblastoma: frequency of the rearrange-
review of literature. Eur J Med Res 11:128–134, 2006. ment and analysis of the candidate genes. BMC Cancer 9:393,
175. Konishi E, Nakashima Y, Iwasa Y, et al: Immunohistochemical 2009.
analysis for Sox9 reveals the cartilaginous character of chondro- 201. Romeo S, Bovee JV, Jadnanansing NA, et al: Expression of carti-
blastoma and chondromyxoid fibroma of the bone. Hum Pathol lage growth plate signaling molecules in chondroblastoma.
41:208–213, 2010. J Pathol 202:113–120, 2004.
176. Kricun ME, Kricun R, Haskin ME: Chondroblastoma of the 202. Rybak LD, Rosenthal DI, Wittig JC: Chondroblastoma: radiofre-
calcaneus: radiographic features with emphasis on location. Am J quency ablation—alternative to surgical resection in selected
Roentgenol 128:613–616, 1977. cases. Radiology 251:599–604, 2009.
177. Kuhlman RE, McNamee MJ: Quantitative microchemical studies 203. Sailhan F, Chotel F, Parot R, et al: Chondroblastoma of bone
of chondroblastoma, giant-cell tumor, chondromyxoid fibroma in a pediatric population. J Bone Joint Surg Am 91:2159–2168,
and desmoplastic fibroma. Clin Orthop 69:264–270, 1970. 2009.
178. Kunze E, Graewe T, Peitsch E: Histology and biology of meta- 204. Schajowicz F, Gallardo H: Epiphyseal chondroblastoma of bone:
static chondroblastoma: report of a case with a review of the lit- a clinicopathological study of 69 cases. J Bone Joint Surg Br
erature. Pathol Res Pract 182:113–123, 1987. 52:205–226, 1970.
179. Kurt AM, Unni KK, Sim FH, et al: Chondroblastoma of bone. 205. Semmelink HJ, Pruszczynski M, Wiersma-van Tilburg A, et al:
Hum Pathol 20:965–976, 1989. Cytokeratin expression in chondroblastomas. Histopathology 16:
180. Kyriakos M, Land VJ, Penning HL, et al: Metastatic chondroblas- 257–263, 1990.
toma: report of a fatal case with a review of the literature on 206. Sirsat MV, Doctor VM: Benign chondroblastoma of bone: report
atypical, aggressive, and malignant chondroblastoma. Cancer of a case of malignant transformation. J Bone Joint Surg Br 52:741–
55:1770–1789, 1985. 745, 1970.
181. Lehner B, Witte D, Weiss S: Clinical and radiological long-term 207. Sjogren H, Orndal C, Tingby O, et al: Cytogenetic and spectral
results after operative treatment of chondroblastoma. Arch Orthop karyotype analyses of benign and malignant cartilage tumours. Int
Trauma Surg 131:45–52, 2011. J Oncol 24:1385–1391, 2004.
182. Levine GD, Bensch KG: Chondroblastoma: the nature of the 208. Sotelo-Avila C, Sundaram M, Kyriakos M, et al: Case report 373:
basic cell: a study by means of histochemistry, tissue culture, diametaphyseal chondroblastoma of the upper portion of the left
electron microscopy, and autoradiography. Cancer 29:1546–1562, femur. Skel Radiol 15:387–390, 1986.
1972. 209. Spahr J, Elzay RP, Kay S, et al: Chondroblastoma of the temporo-
183. Lin PP, Thenappan A, Deavers MT, et al: Treatment and prog- mandibular joint arising from articular cartilage: a previously
nosis of chondroblastoma. Clin Orthop Rel Res 438:103–109, 2005. unreported presentation of an uncommon neoplasm. Oral Surg
184. Mark J, Wedell B, Dahlenfors R, et al: Human benign chondro- Oral Med Oral Pathol 54:430–435, 1982.
blastoma with a pseudodiploid stemline characterized by complex 210. Springfield DS, Capanna R, Gherlinzoni F, et al: Chondroblas-
and balanced translocation. Cancer Genet Cytogenet 58:14–17, toma: a review of 70 cases. J Bone Joint Surg 67A:748–755,
1992. 1985.
185. Masui F, Ushigome S, Kamitani K, et al: Chondroblastoma: a 211. Steiner GC: Ultrastructure of benign cartilaginous tumors of
study of 11 cases. Eur J Surg Oncol 28:869–874, 2002. intraosseous origin. Hum Pathol 10:71–86, 1979.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 471

212. Suneja R, Grimer RJ, Belthur M, et al: Chondroblastoma of bone: 239. Horvai AE, Roy R, Borys D, et al: Regulators of skeletal develop-
long-term results and functional outcome after intralesional ment: a cluster analysis of 206 bone tumors reveals diagnostically
curettage. J Bone Joint Surg Br 87:974–978, 2005. useful markers. Mod Pathol 25:1452–1461, 2012.
213. Swarts SJ, Neff JR, Johansson SL, et al: Significance of abnormali- 240. Jaffe HL, Lichtenstein L: Chondromyxoid fibroma of bone: a
ties of chromosomes 5 and 8 in chondroblastoma. Clin Orthop distinctive benign tumor likely to be mistaken especially for chon-
189-193:1998. drosarcoma. Arch Pathol 45:541–551, 1948.
214. Tamura M, Oda M, Matsumoto I, et al: Chondroblastoma with 241. Jhala D, Coventry S, Rao P, et al: Juvenile juxtacortical chondro-
pulmonary metastasis in a patient presenting with spontaneous myxoid fibroma of bone: a case report. Hum Pathol 39:960–965,
bilateral pneumothorax: report of a case. Surg Today 41:1439– 2008.
1441, 2011. 242. Kim HS, Jee WH, Ryu KN, et al: MRI of chondromyxoid
215. Tarkkanen M, Nordling S, Bohling T, et al: Comparison of cyto- fibroma. Acta Radiol 52:875–880, 2011.
genetics, interphase cytogenetics, and DNA flow cytometry in 243. Konishi E, Nakashima Y, Iwasa Y, et al: Immunohistochemical
bone tumors. Cytometry 26:185–191, 1996. analysis of Sox9 reveals the cartilaginous character of chondro-
216. Turcotte RE, Kurt AM, Sim FH, et al: Chondroblastoma. Hum blastoma and chondromyxoid fibroma of the bone. Hum Pathol
Pathol 24:944–949, 1993. 41:208–213, 2010.
217. Van Zelderen-Bhola SL, Bovee JV, Wessels HW, et al: Ring chro- 244. Kreicbergs A, Lonnquist PA, Willems J: Chondromyxoid fibroma:
mosome 4 as the sole cytogenetic anomaly in a chondroblastoma: a review of the literature and a report on our own experience. Acta
a case report and review of the literature. Cancer Genet Cytogenet Pathol Microbiol Immunol Scand [A] 93:189–197, 1985.
105:109–112, 1998. 245. Kyriakos M: Soft tissue implantation of chondromyxoid fibroma.
218. Vialle R, Feydy A, Rillardon L, et al: Chondroblastoma of the Am J Surg Pathol 3:363–372, 1979.
lumbar spine. Report of two cases and review of the literature. 246. Park SH, Kong KY, Chung HW, et al: Juxtacortical chondromyx-
J Neurosurg Spine 2:596–600, 2005. oid fibroma arising in an apophysis. Skel Radiol 29:466–469,
219. Weiss AP, Dorfman HD: S-100 protein in human cartilage lesions. 2000.
J Bone Joint Surg 68A:521–526, 1986. 247. Rahimi A, Beabout JW, Ivins JC, et al: Chondromyxoid fibroma:
220. Welsh R, Meyer AT: A histogenetic study of chondroblastoma. a clinicopathologic study of 76 cases. Cancer 30:726–736, 1972.
Cancer 17:578–589, 1964. 248. Romeo S, Duim RA, Bridge JA, et al: Heterogeneous and complex
221. Wirman JA, Crissman JD, Aron BF: Metastatic chondroblastoma: rearrangements of chromosome arm 6q in chondromyxoid
report of an unusual case treated with radiotherapy. Cancer 44:87– fibroma: delineation of breakpoints and analysis of candidate
93, 1979. target genes. Am J Pathol 177:1365–1376, 2010.
249. Romeo S, Eyden B, Prins FA, et al: TGF-beta1 drives partial
Chondromyxoid Fibroma myofibroblastic differentiation in chondromyxoid fibroma of
222. Batsakis JG, Raymond AK: Chondromyxoid fibroma. Ann Otol bone. J Pathol 208:26–34, 2006.
Rhinol Laryngol 98:571–572, 1989. 250. Romeo S, Oosting J, Rozeman LB, et al: The role of noncartilage-
223. Bleiweiss IJ, Klein MJ: Chondromyxoid fibroma: report of six specific molecules in differentiation of cartilaginous tumors:
cases with immunohistochemical studies. Mod Pathol 3:664–666, lessons from chondroblastoma and chondromyxoid fibroma.
1990. Cancer 110:385–394, 2007.
224. Bridge JA, Sanger WG, Neff JR: Translocations involving chro- 251. Safar A, Nelson M, Neff JR, et al: Recurrent anomalies of 6q25
mosomes 2 and 13 in benign and malignant cartilaginous neo- in chondromyxoid fibroma. Hum Pathol 31:306–311, 2000.
plasms. Cancer Genet Cytogenet 38:83–88, 1989. 252. Sakamoto A, Tanaka K, Matsuda S, et al: Chondromyxoid fibroma
225. Choi YS, Kim BS, Joo JE, et al: A rare case of epiphyseal chon- of the clavicle. J Orthop Sci 11:533–536, 2006.
dromyxoid fibroma of the proximal tibia. Kor J Radiol 12:761–764, 253. Sawyer JR, Swanson CM, Lukacs JL, et al: Evidence of an associa-
2011. tion between 6q13-21 chromosome aberrations and locally
226. Cilli F, Ozyurek S, Erler K, et al: Chondromyxoid fibroma of the aggressive behavior in patients with cartilage tumors. Cancer
clavicle. J Orthop Sc 12:193, 2007. 82:474–483, 1998.
227. Dadfarnia T, Velagaleti GV, Carmichael KD, et al: A t(1;9) 254. Schajowicz F, Gallardo H: Chondromyxoid fibroma (fibromyxoid
(q10;q10) translocation with additional 6q23 and 9q22 rearrange- chondroma) of bone: a clinico-pathological study of 32 cases.
ments in a case of chondromyxoid fibroma. Cancer Genet 204:666– J Bone Joint Surg Br 53:198–216, 1971.
670, 2011. 255. Shulman L, Bale P, de Silva M: Sacral chondromyxoid fibroma.
228. Dahlin DC: Chondromyxoid fibroma of bone, with emphasis on Pediatr Radiol 15:138–140, 1985.
its morphological relationship to benign chondroblastoma. Cancer 256. Soder S, Inwards C, Muller S, et al: Cell biology and matrix
9:195–203, 1956. biochemistry of chondromyxoid fibroma. Am J Clin Pathol
229. Desai SS, Jambhekar NA, Samanthray S, et al: Chondromyxoid 116:271–277, 2001.
fibromas: a study of 10 cases. J Surg Oncol 89:28–31, 2005. 257. Takao E, Morioka H, Yabe H, et al: Chondromyxoid fibroma of
230. Dorfman HD: Malignant transformation of benign bone lesions. the sternum. J Thorac Cardiovasc Surg 132:430–431, 2006.
In Proceedings of the Seventh National Cancer Conference of the Ameri- 258. Takenaga RK, Frassica FJ, McCarthy EF: Subperiosteal chondro-
can Cancer Society, Philadelphia, 1973, Lippincott. myxoid fibroma: a report of two cases. Iowa Orthop J 27:104–107,
231. Feldman F, Hecht HL, Johnston AD: Chondromyxoid fibroma of 2007.
bone. Radiology 94:249–260, 1970. 259. Tallini G, Dorfman H, Brys P, et al: Correlation between clinico-
232. Fujii N, Eliseo ML: Chondromyxoid fibroma of the maxilla. pathological features and karyotype in 100 cartilaginous and chor-
J Oral Maxillofac Surg 46:235–238, 1988. doid tumours. A report from the Chromosomes and Morphology
233. Fujiwara S, Nakamura I, Goto T, et al: Intracortical chondromyx- (CHAMP) Collaborative Study Group. J Pathol 196:194–203,
oid fibroma of humerus. Skel Radiol 32:156–160, 2003. 2002.
234. Gherlinzoni F, Rock M, Picci P: Chondromyxoid fibroma: the 260. Tornberg DN, Rice RW, Johnston AD: The ultrastructure of
experience at the Istituto Ortopedico Rizzoli. J Bone Joint Surg chondromyxoid fibroma: its biologic and diagnostic implications.
65A:198–204, 1983. Clin Orthop 95:295–299, 1973.
235. Granter SR, Renshaw AA, Kozakewich HP, et al: The pericentro- 261. Troncoso A, Ro JY, Edeiken J, et al: Case report 798: recurrent
meric inversion, inv (6)(p25q13), is a novel diagnostic marker in chondromyxoid fibroma in connective tissue of leg. Skel Radiol
chondromyxoid fibroma. Mod Pathol 11:1071–1074, 1998. 22:445–448, 1993.
236. Halbert AR, Harrison WR, Hicks MJ, et al: Cytogenetic analysis 262. Ushigome S, Takakuwa T, Shinagawa T, et al: Chondromyxoid
of a scapular chondromyxoid fibroma. Cancer Genet Cytogenet fibroma of bone: an electron microscopic observation. Acta Pathol
104:52–56, 1998. Jpn 32:113–122, 1982.
237. Hamada K, Tomita Y, Konishi E, et al: FDG-PET evaluation of 263. Ushigome S, Takakuwa T, Shinagawa T, et al: Ultrastructure of
chondromyxoid fibroma of left ilium. Clin Nucl Med 34:15–17, cartilaginous tumors and S-100 protein in the tumors: with refer-
2009. ence to the histogenesis of chondroblastoma, chondromyxoid
238. Heydemann J, Gillespie R, Mancer K: Soft tissue recurrence of fibroma and mesenchymal chondrosarcoma. Acta Pathol Jpn
chondromyxoid fibroma. J Pediatr Orthop 5:725–727, 1985. 34:1285–1300, 1984.

ERRNVPHGLFRVRUJ
472 6  Benign Cartilage Lesions

264. Wang J, Zhang Y, Li W, et al: Chondromyxoid fibroma of the 289. Heinritz W, Huffmeier U, Strenge S, et al: New mutations of
petrous apex. J Neuroradiol 38:255–256, 2011. EXT1 and EXT2 genes in German patients with multiple osteo-
265. Wilson AJ, Kyriakos M, Ackerman LV: Chondromyxoid fibroma: chondromas. Ann Hum Genet 73:283–291, 2009.
radiographic appearance in 38 cases and a review of the literature. 290. Hudson TM, Chew FS, Manaster BJ: Scintigraphy of benign
Radiology 179:513–518, 1991. exostoses and exostotic chondrosarcomas. Am J Roentgenol
266. Wu CT, Inwards CY, O’Laughlin S, et al: Chondromyxoid 140:581–586, 1983.
fibroma of bone: a clinicopathologic review of 278 cases. Hum 291. Jaffe HL: Hereditary multiple exostosis. Arch Pathol 36:335–357,
Pathol 29:438–446, 1998. 1943.
267. Yalniz E, Alicioglu B, Yalcin O, et al: Non-specific magnetic reso- 292. Jager M, Westhoff B, Portier S, et al: Clinical outcome and geno-
nance featuring of chondromyxoid fibroma of the iliac bone. type in patients with hereditary multiple exostoses. J Orthop Res
J Balkan Union Oncol 12:407–409, 2007. 25:1541–1551, 2007.
268. Yasuda T, Nishio J, Sumegi J, et al: Aberrations of 6q13 mapped 293. Jennes I, de Jong D, Mees K, et al: Breakpoint characterization
to the COL12A1 locus in chondromyxoid fibroma. Mod Pathol of large deletions in EXT1 or EXT2 in 10 multiple osteochondro-
22:1499–1506, 2009. mas families. BMC Med Genet 12:85, 2011.
269. Zillmer DA, Dorfman HD: Chondromyxoid fibroma of bone: 36 294. Jennes I, Pedrini E, Zuntini M, et al: Multiple osteochondromas:
cases with clinicopathologic correlation. Hum Pathol 20:952–964, mutation update and description of the multiple osteochondromas
1989. mutation database (MOdb). Hum Mutation 30:1620–1627, 2009.
270. Zustin J, Akpalo H, Gambarotti M, et al: Phenotypic diversity in 295. Khare GN: An analysis of indications for surgical excision and
chondromyxoid fibroma reveals differentiation pattern of tumor complications in 116 consecutive cases of osteochondroma. Mus-
mimicking fetal cartilage canals development: an immunohisto- culoskel Surg 95:121–125, 2011.
chemical study. Am J Pathol 177:1072–1078, 2010. 296. Kitagawa H, Izumikawa T, Mizuguchi S, et al: Expression of
rib-1, a Caenorhabditis elegans homolog of the human tumor
Osteochondroma suppressor EXT genes, is indispensable for heparin sulfate synthe-
271. Ahn J, Lüdecke H-J, Lindow S, et al: Cloning of the putative sis and embryonic morphogenesis. J Biol Chem 282:8533–8544,
tumour suppressor gene for hereditary multiple exostoses (EXT1). 2007.
Nature Genet 11:137–143, 1995. 297. Kitsoulis P, Galani V, Stefanaki K, et al: Osteochondromas: review
272. Arkader A, Dormans JP, Gaugler R, et al: Spontaneous regression of the clinical, radiological and pathological features. In Vivo
of solitary osteochondroma: reconsidering our approach. Clin 22:633–646, 2008.
Orthop Rel Res 460:253–257, 2007. 298. Kojima H, Wada T, Seki H, et al: One third of Japanese patients
273. Atik OS, Sarikaya B, Kunat C, et al: Osteochondroma of the talus. with multiple osteochondromas may have mutations in genes
Joint Dis Rel Surg 21:116–117, 2010. other than EXT1 and EXT2. Genet Test 12:557–561, 2008.
274. Bishop JR, Schuksz M, Esko JD: Heparan sulphate proteogly- 299. Le Merrer M, Legeai-Mallet L, Jeannin PM, et al: A gene for
cans fine-tune mammalian physiology. Nature 446:1030–1037, hereditary multiple exostoses maps to chromosome 19p. Hum Mol
2007. Genet 3:717–722, 1994.
275. Borges AM, Huvos AG, Smith J: Bursa formation and synovial 300. Libshitz HI, Cohen MA: Radiation-induced osteochondromas.
chondrometaplasia associated with osteochondromas. Am J Clin Radiology 142:643–647, 1982.
Pathol 75:648–653, 1981. 301. Lonie L, Porter DE, Fraser M, et al: Determination of the muta-
276. Busse M, Feta A, Presto J, et al: Contribution of EXT1, EXT2, tion spectrum of the EXT1/EXT2 genes in British Caucasian
and EXTL3 to heparin sulfate chain elongation. J Biol Chem patients with multiple osteochondromas, and exclusion of six can-
282:32802–32810, 2007. didate genes in EXT negative cases. Hum Mutation 27:1160, 2006.
277. D’Ambrosia R, Ferguson AB: The formation of osteochondroma 302. Matsumoto K, Irie F, Mackem S, et al: A mouse model of
by epiphyseal cartilage transplantation. Clin Orthop 61:103–115, chondrocyte-specific somatic mutation reveals a role for Ext1
1968. loss of heterozygosity in multiple hereditary exostoses. PNAS
278. De Andrea CE, Reijnders CM, Kroon HM, et al: Secondary 107:10932–10937, 2010.
peripheral chondrosarcoma evolving from osteochondroma as a 303. Matsuno T, Ichioka Y, Yagi T, et al: Spindle-cell sarcoma in
result of outgrowth of cells with functional EXT. Oncogene 31: patients who have osteochondromatosis: a report of two cases.
1095–1104, 2012. J Bone Joint Surg 70A:137–141, 1988.
279. De Andrea CE, Wiweger M, Prins F, et al: Primary cilia organiza- 304. Milgram JW: The origins of osteochondromas and enchondro-
tion reflects polarity in the growth plate and implies loss of polar- mas. Clin Orthop 174:264–284, 1983.
ity and mosaicism in osteochondroma. Lab Investig 90:1091–1101, 305. Murphy MD, Choi JJ, Kransdorf MJ, et al: Imaging of osteochon-
2010. droma: variants and complications with radiologic-pathologic cor-
280. El-Khoury GY, Bassett GS: Symptomatic bursa formation with relation. Radiographics 20:1407–1434, 2000.
osteochondromas. Am J Roentgenol 133:895–898, 1979. 306. Nadanaka S, Ishida M, Ikegami M, et al: Chondroitin 4-O-
281. Engel EE, Nogueira-Barbosa MH, Brassesco MS, et al: Osteosar- sulfotransferase-1 modulates Wnt-3a signaling through control of
coma arising from osteochondroma of the tibia: case report and E disaccharide expression of chondroitin sulfate. J Biol Chem
cytogenetic findings. Genet Mol Res 11:448–454, 2012. 283:27333–27343, 2008.
282. Garrison RC, Unni KK, McLeod RA, et al: Chondrosarcoma 307. Nojima T, Yamashiro K, Fujita M, et al: A case of osteosarcoma
arising in osteochondroma. Cancer 49:1890–1897, 1982. arising in a solitary osteochondroma. Acta Orthop Scand 62:290–
283. Gorospe L, Madrid-Muniz C, Royo A, et al: Radiation-induced 292, 1991.
osteochondroma of the T4 vertebra causing spinal cord compres- 308. Okada M, Nadanaka S, Shoji N, et al: Biosynthesis of heparin
sion. Eur Radiol 12:844–848, 2002. sulfate in EXT1-deficient cells. Biochem J 428:463–471, 2010.
284. Griffiths HJ, Thompson RC, Jr, Galloway HR, et al: Bursitis in 309. Paprottka FJ, Machens HG, Lohmeyer JA: Partially irreversible
association with solitary osteochondromas presenting as mass paresis of the deep peroneal nerve caused by osteocartilaginous
lesions. Skel Radiol 20:513–516, 1991. exostosis of the fibula without affecting the tibialis anterior
285. Gunay C, Atalar H, Yildiz Y, et al: Spinal osteochondroma: a muscle. J Plast Reconstr Aesthet Surg 65:e223–e225, 2012.
report on six patients and a review of the literature. Arch Orthop 310. Passanise AM, Mehlman CT, Wall EJ, et al: Radiographic evi-
Trauma Surg 130:1459–1465, 2010. dence of regression of a solitary osteochondroma: a report of 4
286. Hacker U, Nybakken K, Perrimon N: Heparan sulphate proteo- cases and a literature review. J Pediatr Orthop 31:312–316, 2011.
glycans: the sweet side of development. Nat Rev 6:530–541, 2005. 311. Pedrini E, De Luca A, Valente EM, et al: Novel EXT1 and EXT2
287. Hameetman L, David G, Yavas A, et al: Decreased EXT expres- mutations identified by DHPLC in Italian patients with multiple
sion and intracellular accumulation of heparin sulphate proteogly- osteochondromas. Human Mutations 26:280, 2005.
can in osteochondromas and peripheral chondrosarcomas. 312. Pei Y, Wang Y, Huang W, et al: Novel mutations of EXT1 and
J Pathol 211:399–409, 2007. EXT2 genes among families and sporadic cases with multiple
288. Heilman RS, Topuzlu C, Molloy M: Femoral arterial injury sec- exostoses. Genet Testing Mol Biomark 14:865–872, 2010.
ondary to osteochondroma of the distal femur. Am J Roentgenol 313. Peterson HA: Multiple hereditary osteochondromata. Clin Orthop
100:533–536, 1967. 239:222–230, 1989.

ERRNVPHGLFRVRUJ
6  Benign Cartilage Lesions 473

314. Picci P, Stilli S: Malignant fibrous histiocytoma arising from an in EXT gene mutation negative multiple osteochondromas
exostosis in a patient affected by multiple exostoses. Ital J Orthop patients. Hum Mutation 32:E2036–E2049, 2011.
Traumatol 8:205–206, 1982. 323. Trebicz Geffen M, Robinson D, Evron Z, et al: The molecular
315. Raskind WH, Conrad EU, Chansky H, et al: Loss of heterozygos- and cellular basis of exostoses formation in hereditary multiple
ity in chondrosarcomas for markers linked to hereditary multiple exostoses. Int J Exper Pathol 89:321–331, 2008.
exostoses loci on chromosomes 8 and 11. Am J Hum Genet 324. Valdivielso-Ortiz A, Barber I, Soldado F, et al: Solitary osteochon-
56:1132–1139, 1995. droma: spontaneous regression. Pediatr Radiol 40:1699–1701,
316. Reijnders CMA, Waaijer CJF, Hamilton A, et al: No haploinsuf- 2010.
ficiency but loss of heterozygosity for EXT in multiple osteochon- 325. Vanhegan IS, Shehzad KN, Bhatti TS, et al: Acute popliteal pseu-
dromas. Am J Pathol 177:1946–1956, 2010. doaneurysm rupture secondary to distal femoral osteochondroma
317. Shapiro F, Simon S, Glimcher MJ: Hereditary multiple exostoses: in a patient with hereditary multiple exostoses. Ann R Coll Surg
anthropometric, roentgenographic, and clinical aspects. J Bone Engl 94:134–136, 2012.
Joint Surg 61A:815–824, 1979. 326. Waaijer CJF, Winter MGT, Reijnders CMA, et al: Intronic dele-
318. Signori E, Massi E, Matera MG, et al: A combined analytical tion and duplication proximal of the EXT1 gene: a novel causative
approach reveals novel EXT1/2 gene mutations in a large cohort mechanism for multiple osteochondromas. Gene Chromosome
of Italian multiple osteochondromas patients. Gene Chromosome Cancer 52:431–436, 2013.
Cancer 46:470–477, 2007. 327. Wilson WG, Wyandt HE, Shah H: Interstitial deletion of 8q:
319. Stevenson CA, Zuska JJ: Aneurysm of the popliteal artery from occurrence in a patient with multiple exostoses and unusual facies.
perforation by a solitary exostosis of the femur: a case report. Am J Dis Child 137:444–448, 1983.
J Bone Joint Surg 39A:431–434, 1957. 328. Yildirim C, Rodop O, Kuskucu M, et al: Giant solitary osteochon-
320. Stickens D, Clines G, Burbee D, et al: The EXT2 multiple exos- droma arising from the fifth metatarsal bone: a case report. J Foot
toses gene defines a family of putative tumour suppressor genes. Ankle Surg 49:298, 2010.
Nature Genet 14:25–32, 1996. 329. Zuntini M, Pedrini E, Parra A, et al: Genetic models of osteo-
321. Strovski E, Ali R, Graeb DA, et al: Malignant degeneration of chondroma onset and neoplastic progression: evidence for
a lumbar osteochondroma into a chondrosarcoma which mim- mechanisms alternative to EXT genes inactivation. Oncogene 29:
icked a large retroperitoneal mass. Skel Radiol 41:1319–1322, 3827–3834, 2010.
2012. 330. Zuntini M, Salvatore M, Pedrini E, et al: MicroRNA profiling of
322. Szuhai K, Jennes I, de Jong D, et al: Tiling resolution array-CGH multiple osteochondromas: identification of disease-specific and
shows that somatic mosaic deletion of the EXT gene is causative normal cartilage signatures. Clin Genet 78:507–516, 2010.

ERRNVPHGLFRVRUJ
C H A P T E R 7 

Malignant Cartilage Tumors


CHAPTER OUTLINE

INTRAMEDULLARY CHONDROSARCOMA JUXTACORTICAL CHONDROSARCOMA


Conventional Chondrosarcoma SECONDARY CHONDROSARCOMAS
Dedifferentiated Chondrosarcoma
Clear Cell Chondrosarcoma
Mesenchymal Chondrosarcoma

Chondrosarcoma is the second most frequent primary behavior. Virtually all de novo chondrosarcomas of bone
malignant tumor of bone. It represents one fourth of all are intramedullary lesions. Primary juxtacortical (surface)
primary bone sarcomas. The term chondrosarcoma is used chondrosarcomas are extremely rare, but malignant
to describe a heterogeneous group of lesions with diverse change is a known secondary complication of osteochon-
morphologic features and clinical behavior. The behavior dromas. Secondary chondrosarcomas that complicate
of these lesions ranges from slowly growing, nonmetas- other conditions (e.g., in Ollier’s disease) are morpho-
tasizing tumors to very aggressive metastasizing sarco- logically similar to conventional chondrosarcoma, but
mas. More than 90% of these tumors are designated as they form a distinct group of lesions and are defined by
conventional chondrosarcomas. On the basis of histo- the clinical setting in which they occur. They provide
logic features (nuclear atypia and cellularity), conven- useful models in which to study the factors (clinical,
tional chondrosarcoma is further subdivided into three pathologic, and molecular) that predispose to the devel-
grades, 1 through 3, which correlate well with their clini- opment of malignant lesions of cartilage. The special
cal behavior. The majority of conventional chondrosar- types of chondrosarcomas include dedifferentiated, mes-
comas are low- to intermediate-grade tumors that have enchymal, and clear-cell chondrosarcomas. These tumors
indolent clinical behavior and low metastatic potential. have distinct morphologic features and clinical behaviors
High-grade (grade 3) tumors are less frequent and have and should be considered as entities separate from con-
high metastatic potential.27,38 The current analysis of ventional chondrosarcoma. The differences in the bio-
data from the National Cancer Institute Surveillance, logic potential of cartilage lesions as related to their
Epidemiology and End Results (SEER) Program com- degree of differentiation are provided in Figure 7-1.
prising 2048 conventional chondrosarcomas indicates
that 80% of them represent low-grade to intermediate-
grade (grade 1 or 2) lesions and the remaining 20% are
classified as high-grade (grade 3), potentially aggressive INTRAMEDULLARY CHONDROSARCOMA
tumors.
The fundamental biologic difference between a grade Conventional Chondrosarcoma
1 chondrosarcoma and a benign enchondroma is signified Definition
by the limited growth potential of the enchondroma and
the slow but continuous locally invasive growth of a low- Chondrosarcoma can be defined as a malignant tumor
grade chondrosarcoma. A distinction between these two of cartilage in which the matrix formed is uniformly
conditions solely on the basis of microscopic cytologic and entirely chondroid in nature. This definition is per-
details is often impossible. Therefore the pathologist tinent to all conventional chondrosarcomas irrespective
should consider other data, such as clinical presentation of the site of the tumor. Tumors that exhibit bone-
and radiographic features, in arriving at a diagnosis. For forming capability and the presence of primitive mesen-
example, it is known that cartilage lesions involving some chymal sarcomatous elements in addition to cartilaginous
parts of the skeleton (small bones of the hands and feet) differentiation should not be classified as chondrosarco-
are almost always benign, whereas cartilaginous lesions mas. Such tumors most frequently represent chondro-
of the ribs, sternum, and flat bones such as the pelvis and blastic variants of osteosarcoma. The validity of this
scapula frequently behave in a clinically aggressive distinction is confirmed by the observed differences in
manner. On the microscopic level, the pattern of growth clinical behavior and therapeutic response between con-
and the relationship of the lesion to adjacent structures ventional chondrosarcoma and predominantly chondro-
should also be taken into consideration. All these data are blastic osteosarcoma. The latter is a tumor that is
used to provide clues about the biologic potential of the predominantly cartilaginous in nature but focally shows
lesion in question and about the lesion’s ultimate clinical tumor osteoid being directly produced by sarcomatous
474
ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 475

Type of Differentiation
Mesenchymal Chondroblastic Chondrocytic

Matrix production

Myxoid Hyaline

Limited Growth
Chondromyxoid Fibroma

Chondroblastoma

Enchondroma
Cartilage Lesions

Enchondromatosis

Chondrosarcoma grade 1-2

Continuous Growth
Chondrosarcoma grade 3

Clear Cell Chondrosarcoma

Mesenchymal Chondrosarcoma

Dedifferentiated Chondrosarcoma

Metastatic
Potential
Degree of Differentiation
FIGURE 7-1  ■  Differences in biologic potential of cartilage lesions. Graphic representation related to their degree of differentiation
and clinical behavior.

cells. It is important to realize that large portions of chondrosarcoma shows a gradual age-related increase,
conventional chondrosarcoma can be composed of calci- with the peak incidence occurring during the sixth and
fied tumor matrix, can exhibit myxomatous change, or seventh decades of life (Fig. 7-3). The majority of patients
can show enchondral ossification. The development of are older than age 50 years. Chondrosarcomas in people
ossification in the preexisting cartilage (enchondral ossi- younger than age 45 years are rare. Individual cases are
fication) in a conventional chondrosarcoma should not be reported in very young patients (age 20 years and
considered diagnostic of osteosarcoma. younger), but the occurrence of chondrosarcoma in chil-
dren is very rare.46,98 It is therefore recommended that
other, more frequent chondroid lesions of bone be ruled
Incidence and Location
out before the diagnosis of chondrosarcoma is rendered
Chondrosarcomas represent the second most common in young patients with cartilage tumors. The male-to-
group of primary bone sarcomas, and their frequency of female ratio is almost equal. There is no major difference
occurrence varies in different series from approximately in incidence between black and white subjects (Fig. 7-4).
20% to 27% of all primary bone sarcomas.6,27,35,38 The Chondrosarcoma has a predilection for the trunk bones,
most common sites of skeletal involvement and the peak which are involved in nearly 40% of cases (pelvis ~20%
age incidence are shown in Figure 7-2. The analysis of and ribs ~20%) (Fig. 7-5). The ilium is the most fre-
SEER data indicates that chondrosarcomas represent quently involved bone (~20% of all cases), followed by
25% of all primary sarcomas of bone. The currently the femur (15%) and humerus (10%). Chondrosarcoma
available SEER data, which comprise 2757 chondrosar- is extremely rare in the spine and craniofacial bones. The
comas indicates that 74% of them represent conventional majority of cartilage lesions of the small bones of the
chondrosarcomas. The age distribution of patients with hands and feet are benign. However, chondrosarcomas

ERRNVPHGLFRVRUJ
476 7  Malignant Cartilage Tumors

Peak
incidence
50
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 7-2  ■  Conventional chondrosarcoma. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites are
indicated by solid black arrows.

1 25

0.8 20
(cases/100,000 persons)

Age distribution (%)


Incidence rate

0.6 15

0.4 10

0.2 5

0 0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

85+

Age at diagnosis
FIGURE 7-3  ■  Epidemiology of conventional chondrosarcoma. National Cancer Institute Surveillance, Epidemiology and End Results
Program, 1973-2010. Age-adjusted incidence rate and age-specific frequency for all races and both sexes in 2048 cases.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 477

Incidence rate (cases per 100,000 persons)


Black, male
Black, female
0.8 White, male
White, female

0.6

0.4

0.2

0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

85+
Age at diagnosis
FIGURE 7-4  ■  Epidemiology of conventional chondrosarcoma. National Cancer Institute Surveillance, Epidemiology and End Results
Program, 1973-2010. Age-adjusted incidence rate by race and sex for 2048 cases.

60

50

40
Frequency (%)

30

20

10

0
Craniofacial Spine Ribs Extremities Pelvis
FIGURE 7-5  ■  Skeletal distribution of chondrosarcoma. National Cancer Institute Surveillance, Epidemiology and End Results
Program, 1973-2010. Skeletal distribution of 2048 cases of conventional chondrosarcoma.

ERRNVPHGLFRVRUJ
478 7  Malignant Cartilage Tumors

do occasionally occur in these acral sites, and it is impor- many chondrosarcomas provoke cortical thickening that
tant to pay close attention to the correlation of clinical may be very striking in degree (Figs. 7-6 and 7-7). The
and radiologic details in the differential diagnosis between outer surface of the cortex overlying the tumor may show
benign and malignant cartilage lesions in these locations. a prominent periosteal reaction. This reaction may be in
Chondrosarcomas also occur in extraskeletal sites. Chon- the form of hazy cortical irregularity and fuzziness or of
drosarcomas that occur in soft tissue are typically myxoid parallel periosteal new bone formation. The multiple
and represent a pathogenetically distinct group.18,35,39,80 A perpendicular striations (“sunburst”) often seen in osteo-
detailed description of extraskeletal chondrosarcoma sarcoma are usually not present in cartilage lesions. The
is beyond the scope of this book. The involvement of lesion may have a more or less lobulated contour. Because
laryngeal cartilaginous structures by a chondrosarcoma most chondrosarcomas grow slowly, they do not readily
is a rare but well-recognized feature of these neo- erupt from the bone. Initially, mild expansion of the bone
plasms.24,44,75,78,79 Rare examples of chondrosarcomas in contour is observed with focal thinning of the cortex (Fig.
the parenchymal organs most frequently represent diver- 7-7). An area of complete cortical disruption with an
gent cartilaginous differentiation in the sarcomatoid extension into soft tissue is usually present in more
components of epithelial neoplasms.16,66 New and unex- advanced lesions. Nearly all lesions of flat bones show
pected association between breast cancer and chondro- features of cortical disruption and significant extension
sarcoma has been identified by epidemiologic studies of into soft tissue at the time of clinical presentation (Figs.
the European cohort. It appears that a subset of estrogen 7-8 and 7-9). The slow growth of chondrosarcoma is
positive breast cancer patients is prone to develop central associated with a clear radiographic demarcation of the
conventional chondrosarcomas suggesting a putative lesion and its low tendency for cortical disruption (Figs.
novel genetic trait distinct from BRCA1 and BRCA2 7-6 and 7-7). Indeed, in long bones, marked thickening
predisposition.20,68 of the cortex adjacent to a low-grade chondrosarcoma
(Fig. 7-6), which reflects the microscopic permeation of
Clinical Symptoms haversian and Volkmann’s canals with subsequent bone
apposition over a prolonged course, is a definite radio-
Pain is usually the presenting complaint in chondrosar- graphic indication of malignancy. Early cortical disrup-
coma. It is typically described as a dull aching that is tion, a destructive (permeative or moth-eaten) pattern of
sometimes intermittent. The pain is noted at rest and growth, or both features are radiographic signs that indi-
may become severe at night. The symptoms are usually cate a high-grade lesion (Figs. 7-7 and 7-8).
of several months’ duration, but it is not uncommon to Magentic resonance imaging (MRI) and computed
obtain a history of pain that has continued for several tomography (CT) are indispensable techniques used to
years. If the lesion is located near the end of a bone in evaluate the extent of bone and soft tissue involve-
proximity to a joint, some restriction of motion can be ment.32,36 In addition, MRI typically shows a low signal
present. A local swelling may be palpable as a conse- (signal void) on T1 images and a high signal on T2
quence of the expansion of a bone contour or extension images. Although not entirely specific, this feature helps
into soft tissue. Pain is an important element in the dif- identify the presumptive cartilaginous nature of the lesion
ferential diagnosis between malignant and benign carti- in question when the typical pattern of punctate calcifica-
lage lesions. Benign intramedullary cartilage lesions, such tion is present on plain radiographs. In addition, CT
as enchondroma, are typically asymptomatic, and many scans can identify discrete calcifications of the lesion that
appear as incidental findings on radiographs. cannot be resolved on plain radiographs or that are invis-
ible on MRI. This is usually present as punctate signal
Radiographic Imaging voids on T1-weighted images.

The presence of discrete calcified opacities is a radio-


Gross Findings
graphic hallmark of cartilage lesions.8,17,21,45,76 Typically,
the cartilage lesion presents as an area of radiolucency In a typical case, the cartilaginous nature of the lesion is
with more or less evenly distributed punctate or ringlike readily recognized on gross examination of the bisected
opacities (Figs. 7-6 and 7-7). The level of mineralization tumor. The lesion has a grossly lobulated architecture
may vary from lesion to lesion. At one end of the that is composed of translucent hyaline nodules that, to
spectrum are predominantly lytic lesions, and their car- some extent, resemble normal cartilage (Figs. 7-10 and
tilaginous nature can be difficult to identify radiographi- 7-11). The lobulated nature of the lesion is accentuated
cally (Figs. 7-7 to 7-9). On the opposite end are heavily by more intense mineralization of the peripheral parts of
calcified lesions with consolidated areas of opacity that the lobules. The mineralized areas are opaque, chalklike,
can be difficult to distinguish from bone-forming lesions. or granular and yellow. The presence of extensive enchon-
Typically the lesion appears as a radiolucent area with dral ossification can be grossly identified as focal, ivory-
moderate numbers of punctate opacities, and its cartilagi- like bony areas in a chondrosarcoma (Fig. 7-29). On the
nous nature can be easily recognized on plain radio- other hand, the tumor may be soft and myxomatous, and
graphs. In the long bones, a large segment of the hemorrhage or necrosis can be present (Fig. 7-11). The
medullary cavity may be involved (Fig. 7-7). The bone low- to intermediate-grade lesions typically have a grossly
contour in the affected region is at least somewhat recognizable cartilaginous nature, but the central por-
expanded. Typically, the cortex is thinned or shows mul- tions of large tumors may become cystic (Fig. 7-11). The
tiple inner surface erosions (endosteal scalloping), but Text continued on p. 485

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 479

A B C
FIGURE 7-6  ■  Chondrosarcoma: radiographic features. A, Anteroposterior (AP) radiograph of a low-grade chondrosarcoma of tibia
has punctate intramedullary calcifications. B, AP radiograph of a heavily calcified grade 1 chondrosarcoma of proximal femoral
shaft. Discrete ringlike and coarse punctate opacities predominate. Note irregularities of inner cortical margins. C, AP radiograph
of discrete punctate calcifications in intramedullary low-grade chondrosarcoma occupying upper humeral shaft.

ERRNVPHGLFRVRUJ
480 7  Malignant Cartilage Tumors

A B C D

E F

FIGURE 7-7  ■  Chondrosarcoma: radiographic features. A, Anteroposterior (AP) radiograph of a low-grade chondrosarcoma of the
femoral shaft with punctate intramedullary calcifications and thickening of the overlying cortex. B, Lateral radiograph of a low-grade
chondrosarcoma of the distal femoral shaft with discrete punctate and ringlike opacities and more lytic component seen proximally.
C and D, AP and lateral radiographs show large intramedullary heavily calcified lesion that has ringlike, punctate, and curvilinear
opacities in low-grade chondrosarcoma. E and F, AP radiographs of high-grade chondrosarcoma presenting as lytic destructive
lesion of the proximal humeral end. Such radiographic presentations of chondrosarcoma have overlapping features with giant cell
tumor of bone.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 481

B C
FIGURE 7-8  ■  Chondrosarcoma: radiographic features. A, Anteroposterior radiograph showing a destructive lytic lesion of the left
periacetabular area representing a high-grade chondrosarcoma. B, Axial computed tomogram showing a large periacetabular mass
with involvement of soft tissue of the lower pelvis. C, Fat-saturated T1-weighted coronal magnetic resonance image with contrast
showing a mass of low signal intensity with extensive involvement of the adjacent soft tissue and the displacement of the pelvic
organs.

ERRNVPHGLFRVRUJ
482 7  Malignant Cartilage Tumors

A C

D E

FIGURE 7-9  ■  Chondrosarcoma: radiographic features. A, Anteroposterior radiograph of a well delineated lesion with discrete min-
eralization pattern involving the glenoid region of the right scapula. B, Axial computed tomogram showing a destructive process
involving the glenoid region of the scapula with expansion and focal penetration of the overlying cortex. Note punctate mineraliza-
tion pattern in the central portion of the lesion. C, T1-weighted axial magnetic resonance image (MRI) showing signal enhancement
of the lesion in the glenoid region of the scapula. D, T1-weighted coronal MRI showing a destructive mass of low signal intensity
originating in the scapula and extending to the adjacent soft tissue. E, Fat-saturated T2-weighted coronal MRI showing lobular signal
enhancement in chondrosarcoma of the scapula.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 483

A B

C D
FIGURE 7-10  ■  Conventional chondrosarcoma: gross features. A, Bisected sternum contains cartilaginous tumor with focal chalklike
gritty areas. B, Specimen radiograph of tumor shown in A demonstrates irregular discrete calcifications in lobules of chondrosar-
coma and extension of tumor into soft tissue. C, Bisected proximal femur contains low-grade chondrosarcoma involving neck and
intertrochanteric area. D, Pronounced lobulation in grade 1 chondrosarcoma of rib. (C, Courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
484 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-11  ■  Conventional chondrosarcoma: gross features. A, Large low-grade chondrosarcoma involving inner surface of ilium
shows central cystic changes. B, Chondrosarcoma involving outer surface of ilium shows prominent lobulated growth pattern.
C, Chondrosarcoma of scapula with prominent cavitation. D, Chondrosarcoma of tibia with extensive cortical disruption and exten-
sion into soft tissue. (B and C, Courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 485

presence of gray, friable, and hemorrhagic tissue with a cases, chondrosarcoma exhibits some unusual micro-
fleshy sarcomatous appearance is indicative of more scopic features such as the signet-ring appearance or the
aggressive high-grade lesions (Fig. 7-12). presence of prominent intranuclear inclusions.25,29,47
In the long tubular bones, large portions of the medul- From the diagnostic point of view, chondrosar-
lary cavity may be filled with a lobulated cartilaginous comas can be divided into two main groups: low- to
tissue. The cortex overlying the affected area is thick- intermediate-grade differentiated tumors and high-grade
ened, roughened, and pitted. These features are the result tumors. Those that require supportive evidence other
of the slow infiltrative advance of the tumor, with scal- than microscopic features to be definitely classified as
loping of the inner cortex and deposition of reactive bone chondrosarcomas comprise the group of borderline, low-
on the outer cortical surface. Initially the lesion grows grade tumors. Those that can be independently identified
within the medullary canal, but eventually complete cor- by microscopic features as malignant cartilage lesions
tical disruption ensues, and the tumor advances through include grade 2 tumors, and those that are frankly ana-
this soft tissue area (Fig. 7-12). Eventually a large lobu- plastic are grade 3 tumors. Low-grade chondrosarcoma
lated extraosseous mass that is attached to the bone manifests cytologic features similar to those of benign
is formed. The extraosseous component often grows on cartilage lesions such as enchondroma. It is important
the bone surface, encircling the affected area. Cortical to mention that some benign lesions of cartilage, such
disruption develops earlier in the flat bones, such as as synovial chondromatosis, multiple enchondromatosis
the pelvis, scapula, and cranium, which have relatively (Ollier’s disease), and metaplastic cartilage of reactive
narrow medullary cavities. Virtually all chondrosarcomas conditions, may exhibit levels of nuclear atypia and cel-
that occur at these sites develop a significant extraosse- lularity approximating or even exceeding those of a low-
ous component by the time they are clinically symptom- grade chondrosarcoma. Therefore it is mandatory that
atic (Figs. 7-11 and 7-12). In the pelvis, they typically nuclear atypia and cellularity of the lesion be considered
present as sessile lobulated masses with large extraosseous in light of the overall clinical, radiographic, and patho-
components. logic patterns of the lesion in question.
In general, a solitary cartilage lesion should be sus-
pected of being a low-grade chondrosarcoma if it
Microscopic Findings
shows, even focally, hypercellularity, plump cells with the
To be classified as a chondrosarcoma, the tumor should so-called open nuclear chromatin pattern and prominent
be uniformly cartilaginous. The cartilaginous nature of nucleoli, the presence of nuclear pleomorphism, and
the lesion is typically easy to recognize (Fig. 7-13). The more than occasional double nuclei. As stated, this type
tumor cells resemble normal chondrocytes and lie in of lesion requires clinical and radiologic data to support
lacunar spaces embedded within hyaline cartilage matrix the diagnosis of chondrosarcoma.17,21,29,33,34,72,76,83
that may be partially calcified or myxoid or that may A cartilage lesion should be regarded as chondrosar-
exhibit foci of enchondral ossification. The level of min- coma microscopically if it shows prominent nuclear
eralization can vary in different lesions and in different atypia, mitotic activity with atypical mitoses, and multiple
areas of the same tumor, but typically chondrosarcoma pleomorphic or multinuclear cartilage cells.
shows mild to moderate levels of calcification. Foci of Contrary to other bone and soft tissue sarcomas, his-
myxoid change can be present, and some lesions are pre- tologic grading of chondrosarcomas correlates well with
dominantly myxoid. Chondrosarcoma has an overall their clinical behavior.6,8,10,17,31,35,37,42,58,64,81,82 Chondrosar-
lobulated architecture. The individual lobular structures comas of different grades most likely represent pathoge-
may vary in size, ranging from less than 1 mm to several netically distinct conditions with different biologic
millimeters in diameter. The individual lobules can potential and clinical behavior.58-60,96 The most widely
be separated by narrow fibrovascular bands. At the accepted method of grading is based on a three-tier
periphery, lobules of tumor can be seen permeating the system (Fig. 7-23). The diagnostic criteria for the three
marrow spaces and engulfing cancellous trabeculae of grades of chondrosarcomas are described next.
bone (Figs. 7-14 to 7-16). There is usually little or no
evidence of reactive bone at the periphery of tumor Grade 1 Chondrosarcoma.  Grade 1 chondrosarcoma
lobules in the marrow spaces. The lesion can be com- is cytologically very similar to enchondroma. The micro-
posed of homogeneous areas of varying size centrally or scopic differences are minimal, and the distinction
can have a mixed homogeneous and lobular architecture. may be a subjective one.21,29,33,37,49 Overall the cellularity
Deposition of periosteal new bone can be seen micro- is higher than in enchondroma, and there are more
scopically in areas of complete cortical disruption. than occasional plump nuclei with an open chromatin
The cells can be more or less uniformly distributed structure and double nuclei (Fig. 7-13). The lesion typi-
in the cartilaginous matrix or more typically form cally has an infiltrative growth pattern with features of
small clusters (Figs. 7-17 to 7-19). The chondrocyte cyto- endosteal erosion and engulfment of the adjacent cancel-
plasm may show marked multivesicular vacuolization and lous bone. Clinical and radiologic features of extraosse-
even ballooning of cells (Figs. 7-19, 7-21, and 7-23). ous extension can be present. The pattern of growth and
These swollen cells may take on features that suggest the relationship of the lesion to the adjacent bone and
clear cell differentiation in chondrosarcoma. However, soft tissue often cannot be evaluated in limited biopsy
this finding in an otherwise conventional chondrosar- material. The diagnosis of grade 1 chondrosarcoma
coma should not be construed as evidence for the diag- nearly always requires supportive evidence from the
nosis of the clear cell chondrosarcoma variant. In rare Text continued on p. 494

ERRNVPHGLFRVRUJ
486 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-12  ■  High-grade conventional chondrosarcoma. A, Myxoid appearance in high-grade chondrosarcoma of proximal femoral
shaft. B, Hemorrhagic necrosis and extension into soft tissue in high-grade chondrosarcoma of shaft of humerus. C and D, Bisected
distal femur shows tumor necrosis and soft tissue invasion by high-grade chondrosarcoma. (A and B, Courtesy Dr. A.G. Ayala,
Houston.)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 487

A B

C D

FIGURE 7-13  ■  Conventional chondrosarcoma: microscopic features. A-C, Intermediate photomicrographs showing low-grade chon-
drosarcoma with hyaline cartilage matrix and high cellularity. Malignant cartilage cells are residing in lacunar and surrounding
hyaline matrix. D, High power photomicrographs showing malignant cartilage cells with irregularities in size and shape of nuclei.
(A-C, ×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
488 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-14  ■  Conventional chondrosarcoma: microscopic features. A and B, Low power photomicrographs showing aggressive
growth pattern of chondrosarcoma with permeation of intertrabecular spaces within the medullary cavity. C and D, Advancing
edge of chondrosarcoma infiltrating the intratrabecular spaces within the medullary cavity. (A and B, ×10; C and D, ×20)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 489

A B

C D
FIGURE 7-15  ■  Conventional chondrosarcoma: microscopic features. A and B, Low power photomicrographs showing permeation
of intertrabecular spaces by myxoid chondrosarcoma. Note thickened trabeculae of bone in A and advancing edge of the tumor in
B. C, and D, Intermediate power photomicrographs showing myxoid matrix and immature cartilage cells of myxoid chondrosarcoma
in C and advancing edge of the tumor in D. (A and B, ×10; C and D, ×100) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
490 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-16  ■  Conventional chondrosarcoma: microscopic features. A-D, Low power photomicrographs showing aggressive growth
pattern on chondrosarcoma with permeation of the cortex. (A-D, ×10) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 491

A B

C D
FIGURE 7-17  ■  Conventional chondrosarcoma: microscopic features. A and B, Low and intermediate power photomicrographs
showing grade 2 chondrosarcoma with pronounced hypercellularity and irregularities in size and shape of nuclei. The matrix in
this tumor is predominantly myxoid. C and D, Low and intermediate power photomicrographs of another area in the same
tumor showing hypercellularity and clustering of cells within predominantly myxoid matrix. (A and C, ×100; B and D, ×200)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
492 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-18  ■  Conventional chondrosarcoma: microscopic features. A-D, Low and intermediate power photomicrographs of grade
2 chondrosarcoma with myxoid matrix. Note hypercellularity and variations of size and shape of nuclei. (A and C, ×100; B and
D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 493

A B

C D
FIGURE 7-19  ■  Conventional chondrosarcoma: microscopic features. A-D, Low and intermediate power photomicrographs of
grade 2 chondrosarcoma. Note high degree of hypercellularity and nuclear atypia of cartilage cells. (A and C, ×100; B and D, ×200)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
494 7  Malignant Cartilage Tumors

clinical and radiographic data. For example, lesions in in grade.55 That usually happens in grade 2 lesions, which
certain anatomic locations, such as the ribs, sternum, and may recur as grade 3 chondrosarcomas.
flat bones, nearly always behave in an aggressive manner.
By contrast, cartilage lesions located distal to the wrist Special Techniques
and ankle joints are nearly always clinically benign.
However, it must be mentioned that enchondromas do Immunohistochemically chondrosarcoma express
occur in the axial skeleton, and well-documented exam- markers of cartilage lineage delineation such as SOX9
ples of benign cartilage lesions in the ribs, sternum, and and S-100 and as such have overlapping immunohisto-
flat bones have also been reported. Pain is an important chemical profiles with other cartilage lesions including
symptom of cartilage malignancy and is believed to be enchondroma, chondroblastoma, and chondromyxoid
related to an infiltrative growth pattern. Radiographic fibroma.95 Therefore, the cartilage lineage expression
data are indispensable in the evaluation of cartilage profile is of minimal use in differential diagnosis of
lesions. The large size of the mass indicates a continuous various types of benign and malignant cartilage lesions. It
growth potential and a clinically aggressive lesion. Radio- can be of help in those rare instances when the cartilage
graphic evidence of bone contour expansion, cortical nature of the tumor in question is not evident on conven-
thinning, endosteal scalloping, and the presence of solid tional histologic preparations. Chondrosarcoma is usually
periosteal new bone formation with cortical thickening easy to identify on conventional hematoxylin-eosin–
in the vicinity of the tumor all indicate a clinically aggres- stained sections; immunohistochemical stains are rarely
sive lesion.43 Grade 1 chondrosarcoma is a slowly growing, needed to identify the cartilaginous nature of the lesion.
locally aggressive tumor with an indolent course and Typically S-100 protein is uniformly strongly positive,
recurrent growth potential. Metastatic spread is not a but in grade 3 lesions, it can be focally negative, especially
feature of grade 1 chondrosarcoma, but uncontrolled in less differentiated areas.95 The issue of coexpression of
local recurrence can lead to a fatal outcome. epithelial markers by some cartilage lesions in the central
axis (skull and spine) and their significance for differential
Grade 2 Chondrosarcoma.  Grade 2 chondrosarcoma diagnosis of chordoma, chondroid chordoma, and chon-
is characterized by a definite and uniformly increased drosarcoma is discussed in Chapter 18.
cellularity. The cells can be more or less evenly distrib- Ultrastructurally, grade 1 chondrosarcomas are similar
uted in the cartilaginous matrix, or they can form loose to enchondromas.30 The cells of grade 2 chondrosarco-
clusters that vary in size (Figs. 7-17 to 7-19). The carti- mas have enlarged round or oval nuclei with prominent
lage cells are plump and have definitely enlarged nuclei nucleoli. The cytoplasm of cartilage cells shows promi-
with an open chromatin pattern; distinct nucleoli are nent rough cytoplasmic reticulum, mitochondria, and
present in the majority of cells (Fig. 7-20). Binucleated large amounts of glycogen (Fig. 7-24). Myxoid chondro-
cells are frequent and occasionally, multinucleated cells sarcomas are characterized by the presence of loose gran-
with highly atypical nuclei can be seen. Foci of myxoid ular extracellular matrix and stellate mesenchymal cells
change are frequently present. Myxoid tumors are classi- (Fig. 7-25). The cytoplasm of these cells has enlarged
fied as grade 2 chondrosarcomas, even when the cellular- (dilated) endoplasmic reticulum that forms multiple vac-
ity of the lesion is relatively low (i.e., similar to that of uoles. Grade 3 chondrosarcomas show prominent cellu-
grade 1 chondrosarcoma) (Fig. 7-21). Grade 2 chondro- lar and nuclear pleomorphism of cartilage cells and
sarcomas are locally aggressive tumors with a great focally have undifferentiated mesenchymal cells. Ultra-
potential for local recurrence. Approximately 10% to structural features are of limited value in the differential
15% of such tumors metastasize. The most frequent sites diagnosis of chondrosarcoma and are almost never
of metastasis are lung, regional lymph nodes, and liver. required to support the diagnosis.
Local recurrence with multiple soft tissue nodules at the DNA ploidy measurements have shown that virtually
site of prior excision is typical for this tumor. The recur- all grade 1 chondrosarcomas are diploid. Conversely,
rences of grade 1 chondrosarcomas sometimes show an nearly all grade 3 and some grade 2 tumors are aneuploid.
increase in grade sufficient to be classified as grade 2. Aneuploidy in chondrosarcoma correlates with aggres-
sive clinical behavior (i.e., an increased recurrence rate
Grade 3 Chondrosarcoma.  High-grade chondrosarco- and high propensity for metastasis).1,2,49,54,60 Similarly, an
mas are rare and make up approximately 5% to 10% of increased proportion of cells in the S phase (>14%) is
all chondrosarcomas. These tumors are characterized by usually seen in high-grade tumors and correlates with a
high cellularity, prominent nuclear atypia, and the pres- high propensity for metastases.2 Some preliminary data
ence of mitoses (Figs. 7-22 and 7-23). These features can suggest that the morphometric analysis of nuclear volume
be uniformly present throughout the tumor or can be can be helpful in the differential diagnosis of enchon-
seen only focally within a tumor that has overall morpho- droma versus grade 1 chondrosarcoma.12,49
logic features similar to those of grade 2 chondrosar- Chromosomal analysis shows that chondrosarcomas,
coma. In general, lesions that are cytologically grade 2 especially of high histologic grade, have complex aberra-
chondrosarcomas and are found to have more than tions with nonreciprocal translocations and deletions of
one mitosis per high-power field in focal areas should numerous chromosomes.57,69 It appears that rearrange-
be classified as grade 3 chondrosarcoma. Grade 3 chon- ment of 1p and possibly 4, 5, 9, and 20 are nonrandom
drosarcomas are highly aggressive, rapidly growing and may play a role in the biology of these neoplasms.
lesions with definite metastatic potential. It is estimated The accumulation of p53 protein that results from the
that more than 50% of these lesions eventually metasta- mutation of its coding gene preferentially occurs in the
size. Chondrosarcomas that recur can show an increase Text continued on p. 501

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 495

A B

C D
FIGURE 7-20  ■  Conventional chondrosarcoma: microscopic features. A-D, Intermediate and high power photomicrographs of grade
2 chondrosarcoma showing hypercellularity and nuclear atypia with open chromatin architecture. Note the presence of prominent
nucleoli. The cytoplasm of the cartilage cells is oval, densely eosinophilic with occasional vacuoles displacing the nucleus eccentri-
cally. (A and C, ×200; B and D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
496 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-21  ■  Conventional chondrosarcoma: microscopic features. A-D, Grade 2 chondrosarcoma with myxoid features. Note
prominent myxoid stroma with dispersed immature cartilage cells. Some areas of the tumor contain malignant cartilage cells with
vacuolated cytoplasm. (A-C, ×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 497

A B

C D

FIGURE 7-22  ■  Conventional chondrosarcoma: microscopic features. A-D, Examples of grade 3 chondrosarcoma with pronounced
cellularity, cellular pleomorphism, and nuclear atypia. Inset, High power photomicrograph showing malignant cartilage cells with
dense eosinophilic cytoplasm and nuclear atypia. (A-D, ×200; inset, ×400) (A-D and inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
498 7  Malignant Cartilage Tumors

FIGURE 7-23  ■  Conventional chondrosarcoma: microscopic features. A, Grade 1 chondrosarcoma. B, Grade 2 chondrosarcoma. C and
Insets, Grade 3 chondrosarcoma. Note the increasing cellularity that parallels the progression from grades 1 through 3 with pro-
nounced cellular pleomorphism and nuclear atypia in grade 3 chondrosarcoma. (A-C and insets, ×200) (A-C and insets,
hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 499

N1

GC

N2

FIGURE 7-24  ■  Conventional chondrosarcoma: ultrastructural features. A, Tumor cells with prominent rough endoplasmic reticulum
and abundant glycogen (G). Note loose fibrillar stroma and calcifications (right). B, Cluster of three chondrosarcoma cells. Inset,
Binuclear (N1 and N2) tumor cell with prominent perinuclear Golgi center (GC). (A, ×4000; B, ×3500; inset, ×18,000).

ERRNVPHGLFRVRUJ
500 7  Malignant Cartilage Tumors

DC

DC

B
FIGURE 7-25  ■  Myxoid chondrosarcoma: ultrastructural features. A, Stellate mesenchymal cells in amorphous granular stroma of
low density. B, High magnification of A shows tumor cell with dilated channels of endoplasmic reticulum (DC). (A, ×2000; B, ×4000)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 501

high-grade lesions and may be an indicator of poor prog- the metastasizing potential of a primary bone sarcoma
nosis in chondrosarcoma.26 Loss of heterozygosity (LOH) when small amounts of tumor bone or osteoid formation
of 5q, 9p, 11p, 13q, and 19q in chondrosarcoma and go undetected because of sampling problems. Careful
chondroblastoma suggest the involvement of genes such attention to clinical and radiologic correlation helps
as TP53, RB1, CDKN2/p16, ERC, and XRCC in the devel- prevent these mistakes. Chondrosarcomas are more
opment of these tumors.70 Genomic profiling using array common in patients beyond the fifth decade of life
comparative hybridization supports the notion that poly- and are very rare in adolescents. Osteosarcomas with
ploidization of initially hyperhaploid/hypodiploid cell abundant cartilage matrix formation usually show radio-
populations is a common mechanism of chondrosarcoma logic features more consistent with a bone-forming
progression.40 Such studies also suggest the involvement tumor, including cloudlike radiodensity and prominent
of ribosomal protein S6 and cyclin-dependent kinase 4 in periosteal new bone formation in response to cortical
these tumors.77 disruption. The latter often takes the form of a sunburst
The presence of IDH1 and IDH2 mutations in approx- or “hair-on-end” appearance that is rarely seen in
imately 50% of central conventional chondrosarcomas chondrosarcomas.
suggests the important role of these genes in the develop- Enchondromas and low-grade conventional chondro-
ment of chondrosarcoma. The identification of similar sarcomas can be almost indistinguishable on cytologic
mutations in solitary enchondromas and the multiple grounds, and attention must be paid to the clinical setting
chondromas of Ollier’s disease and Maffucci’s syndrome and radiologic response of the surrounding bone, as well
as well as periosteal chondromas provides a unifying as the presence or absence of pain, the age of the patient,
concept for the pathogenesis of these diverse conditions.3 and whether the lesion was detected as an incidental
Interestingly, similar mutations of IDH1 and IDH2 genes finding. The last becomes a factor when asymptomatic
were detected in central nervous system gliomas, acute enchondromas are discovered only because radiographs
myelocytic leukemia, and several other solid human are obtained after an injury or bone isotope scans reveal
tumors.51,71 For central nervous system gliomas and acute the presence of an undiagnosed central cartilage tumor.
myelocytic leukemia IDH mutations identify a distinct Histologically, enchondromas tend to be less cellular, the
subset of patients with prognostic implications.5,23,52,65,74,88,97 chondroid matrix tends to be uniformly hyaline, and the
No such relationship between IDH mutations and prog- matrix calcification may be abundant. The nuclei of the
nosis has been reported for chondrosarcomas. The IDH benign cartilage tumor cells are small, uniform, and
mutations cause the accumulation of oncometabolite (R)- round and have homogeneous chromatin density, in con-
2-hydroxyglutarate affecting cell differentiation, survival, trast to the larger nuclei with open chromatin patterns
and proliferation.94 A detailed description of IDH biology seen in chondrosarcomas. Multinucleation of chondro-
in cartilage neoplasia is provided in Chapter 6, in the sarcoma cells is far more frequent than it is in enchon-
section describing Ollier’s disease. dromas. The matrix in chondrosarcomas may show
Several oncogenic pathways affecting the key cellular prominent myxoid change, and the tumor tends to infil-
functions such as migration, cell-stroma interaction, dif- trate the intertrabecular spaces and the haversian canals
ferentiation, and proliferation essential for aggressive rather than showing the discrete lobules bordered by
behavior of cartilage cells have been implicated in the lamellar bone associated with enchondromas.
biology of chondrosarcoma.9 Integrin activation with The cellular dysplastic cartilage seen in enchondroma-
upregulation of matrix metalloproteinases and extracel- tosis (Ollier’s disease) may present serious problems in the
lular matrix degradation leading to increased cell migra- differential diagnosis because of its high cellularity and
tion appears to be dependent on phosphoinositide-3 more frequent binucleation of chondrocytes. The diffi-
kinase and MEK-extracellular signal-regulated kinase culty in distinguishing between enchondromatosis and
(ERK) signaling.11,84 Additionally, chondrosarcoma cell low-grade chondrosarcoma is further complicated by the
proliferation and matrix degradation are controlled fact that highly eccentric dyschrondroplastic lesions and
by peroxisome proliferator-activated receptor-gamma even subperiosteal ones can simulate extraosseous exten-
(PPAR-γ) activity.11 Ephrin-A5 (EFNA5) downregulation sion of chondrosarcoma. Radiologic demonstration of
is a consistent finding in chondrosarcoma and may con- the typical pattern of polyostotic lesions of Ollier’s disease
tribute to its progression associated with altered cell facilitates this distinction.
adhesion.53 Chondrosarcoma cells are also dependent on Fibrous dysplasia with abundant cartilage differentiation
such well known regulators of cartilage differentiation (fibrocartilaginous dysplasia) may be difficult to distin-
and proliferation as hedgehog, p53, insulin-like growth guish from chondrosarcoma, particularly with limited
factor, cyclin-dependent kinase 4, hypoxia-inducible biopsy samples having only the cartilage component. The
factor, SRC, and AKT.14,91 All these pathways are impli- radiographic features of fibrous dysplasia, especially when
cated as potential therapeutic targets for chondrosar- multiple skeletal sites are affected, offer a good opportu-
coma, but no effective targeted therapy is available at the nity to make this distinction. The cartilage in a lesion of
time of this writing.19,50,85 fibrous dysplasia may also show microscopic evidence
of an orderly enchondral ossification sequence that
Differential Diagnosis mimics a growth plate. This is usually not found in
chondrosarcomas.
Because of its predominantly cartilaginous features, chon- Fracture callus and the exuberant reparative changes
droblastic osteosarcoma may be mistaken for chondrosar- associated with pubic osteolysis can contain an abun-
coma. This mimicry can lead to the error of underestimating dance of proliferating cartilage tissue that sometimes

ERRNVPHGLFRVRUJ
502 7  Malignant Cartilage Tumors

suggests cartilage neoplasia. These can usually be rec- in nature and not part of a reactive or metaplastic
ognized as nonneoplastic conditions when all of the clini- condition (pubic osteolysis), fibrous dysplasia, or enchon-
cal and radiographic data, in addition to the absence of dromatosis (Ollier’s disease) should be presumptively
true microscopic anaplasia of the reparative tissue, are considered to be malignant until proven otherwise. The
considered. flat bones have relatively narrow medullary cavities, and
Synovial chondromatosis, particularly when it is associ- cortical disruption occurs at these sites much earlier than
ated with bone erosion, has occasionally led to the mis- in the long tubular bones. Virtually all chondrosarcomas
diagnosis of chondrosarcoma. This uncommon situation of flat bones show features of cortical disruption and
arises most often in connection with synovial chondro- extensive soft tissue involvement at the time of diagnosis.
matosis of the hip and of the temporomandibular joint, Typically they present as larger masses than those occur-
where extension into bone by cortical erosion occurs ring in long bones (Figs. 7-8 and 7-9). Their prognosis
more often. Clinical and radiologic suspicion of the syno- is significantly worse than that for tumors occurring in
vial origin of the cartilaginous nodular tissue can often long bones because the stage at diagnosis tends to be
be confirmed microscopically. higher and their location renders complete surgical
removal more complicated. Exceptional cases of pelvic
Conventional Chondrosarcoma chondrosarcoma are those that originate in the periace-
in Different Anatomic Sites tabular region. These are often diagnosed more promptly
because early onset of pain is related to the proximity of
Similar to other bone sarcomas, conventional chondro- the tumor to the hip joint.
sarcoma has identical microscopic features and biologic
potential regardless of its anatomic location. However, it Chondrosarcoma of Ribs and Sternum.  The ribs and
has a unique predilection to involve certain anatomic sites sternum are frequent sites (~12% of all cases).15,27,28,61
and is extremely rare in some parts of the skeleton. In Enchondromas at these sites are extremely rare and
general, the overall anatomic distribution of chondrosar- are typically small, asymptomatic lesions (2 cm in diam-
coma differs significantly from that of enchondroma, eter). Chondrosarcomas of the ribs and sternum may
although some overlap exists. Moreover, the clinical sig- occasionally be discovered incidentally on chest radio-
nificance, technical feasibility of complete removal, and graphs, and the peak age incidence is lower than that for
chance for cure differ in relation to the anatomic site. For conventional chondrosarcoma at other sites. Typically
this reason, separate descriptions of chondrosarcoma and an expansile lucent lesion is seen with punctate calcifica-
its differential diagnosis in various anatomic sites are tions that are better visualized on CT and MRI (Figs.
provided. 7-26 and 7-27). Wide en bloc excision, including of
the surrounding chest wall, is required to obtain local
Chondrosarcoma of Long Tubular Bones.  Approxi- control of these tumors. The histologic differentiation
mately 40% of chondrosarcomas occur in the long tubular is usually grade 1 or 2 in rib and sternal tumors. The
bones of the extremities.6,17,37,38,42 The proximal femur is clinical differences associated with tumors that arise in
the most frequently affected site in the appendicular skel- this particular location suggest that they may repre-
eton (~12% of cases), followed by the distal femur (8%), sent a pathogenetically distinct group of chondrosarco-
proximal humerus (8%), and proximal tibia (5%). It is mas. For practical purposes, all cartilage lesions of the
important to remember that benign intramedullary car- ribs and sternum with enchondroma-like morphologic
tilage lesions of long bones are typically small and clini- features that exceed 2 to 3 cm in diameter should be
cally asymptomatic and are often discovered as incidental considered potential chondrosarcomas. Other benign
findings on radiographs. On the other hand, chondrosar- cartilage-containing lesions that should be considered in
comas of long bones are symptomatic calcified intramed- the differential diagnosis of chondrosarcoma at these sites
ullary tumors that alter the adjacent trabecular and are cartilage-containing reactive lesions such as fracture
cortical bone as they infiltrate the marrow cavity and callus, fibrous dysplasia with foci of cartilage, and costo-
provoke cortical bone thickening (Figs. 7-6 and 7-7). chondritis. Clinical and radiographic evidence of trauma
Intramedullary cartilage lesions should be suspected of can be extremely helpful in the differential diagnosis. The
being grade 1 chondrosarcoma if the following features periosteal location of the lesion with swelling at the costo-
are present: foci of increased cellularity with plump nuclei chondral junction also helps identify the benign reactive
and open nuclear chromatin structure; an infiltrative or process. It should be remembered that fibrous dysplasia
destructive growth pattern; size that exceeds several cen- is the lesion occurring most frequently in the ribs. In this
timeters in greatest dimension; pain, particularly at rest; condition, massive amounts of cartilage may be present.
and radiographic features of destructive growth. The cartilage can be confused with chondrosarcoma.

Chondrosarcoma of Flat Bones.  The pelvis is the Chondrosarcoma of Craniofacial Bones.  Chondro-
single most frequent site involved by chondrosarcoma sarcomas of the craniofacial bones are rare (2% of all
(~25% of all cases).27,38,62 The ilium is the most frequently cases). Typically they occur at the base of the skull (Fig.
involved bone (~15% of all cases), followed by the pubis 7-28), but they may present clinically as orbital, nasal,
and ischium (9%). A second flat bone frequently involved sinus, or pharyngeal masses.7,13,41,44,48,56,63,78,86,89 They tend
is the scapula (5%). Benign enchondromas are extremely to be histologically of low grade with prominent punctate
rare at all of these sites. Therefore for practical purposes, calcifications best demonstrated on CT or MRI (Fig.
any cartilage lesion identified at these sites as neoplastic 7-28). The cartilage-containing lesions of the maxilla and

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 503

B C

E G H
FIGURE 7-26  ■  Chondrosarcoma of ribs and sternum: radiographic and gross features. A, Axial computed tomogram (CT) showing
a destructive mass involving the head and shaft of the vertebra and the rib. B, Fat-saturated T1-weighted axial magnetic resonance
image (MRI) with contrast of case in A showing irregular signal enhancement in a chondrosarcoma of the rib. C, Bisected specimen
of the case shown in A and B with extensive cartilaginous mass involving the head and shaft. D, Axial CT showing destructive
sternal mass with punctate calcifications. E, Bisected sternal resection specimen showing a cartilaginous mass involving the body
of the sternum. F and G, Fat-saturated T1-weighted axial MRIs with contrast showing a destructive sternal mass with focal calcifica-
tions. H, Axially bisected resection sternal specimen showing a cartilaginous intramedullary mass involving the sternal body.

ERRNVPHGLFRVRUJ
504 7  Malignant Cartilage Tumors

B
FIGURE 7-27  ■  Chondrosarcoma of rib. A, Anteroposterior radiograph of chest shows expansile mass in anterior end of rib in an
18-year-old man who had noted palpable mass for several months. B, Bisected specimen shows eccentric growth of intramedullary
grade 1 chondrosarcoma near junction with costal cartilage. Tumor expands through convex surface into adjacent soft tissue external
to rib cage.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 505

A B

C D

FIGURE 7-28  ■  Chondrosarcoma of skull base: radiographic features. A and B, T1-weighted coronal magnetic resonance images
(MRIs) with contrast of two skull base chondrosarcomas showing the right sided parasellar masses. C and D, Coronal and axial
T1-weighted MRIs with contrast showing two examples of maxillary chondrosarcomas.

ERRNVPHGLFRVRUJ
506 7  Malignant Cartilage Tumors

mandible most frequently represent chondroblastic apply to high-grade chondrosarcomas. Consequently the
osteosarcoma rather than true chondrosarcomas.7 It lesions located in anatomic sites where wide local excision
should be emphasized that in chondroblastic osteosar- is technically possible have better prognoses than those
coma the cartilaginous component can dominate the located at sites where complete removal is not possible.
lesion, and microscopic evidence of direct osteoid pro- The overall prognosis is related to the size of the lesion,
duction by sarcomatous tumor cells can be inconspicuous anatomic location, and histologic grade.17,29,31,33,42 In
and difficult to document in limited biopsy material. general, lesions of the appendicular skeleton have better
Chondrosarcomas of the base of the skull should also be prognoses than those of the axial skeleton. Data from the
differentiated from chondroid chordomas.48 A detailed University of Texas M.D. Anderson Cancer Center indi-
description of this entity is provided in Chapter 18. Syno- cate that the survival rate is strictly related to the histo-
vial chondromatosis may involve the temporomandibular logic grade of the tumor,31 with 5-year survival rates of
joint and can erode the adjacent bone, presenting as a 90% for grade 1, 81% for grade 2, and 29% for grade 3
cartilaginous mass at the base of the skull, mimicking a tumors. It is evident that the most significant difference
chondrosarcoma. in clinical behavior is between grade 2 and 3 chondrosar-
comas. Grade 1 and 2 lesions behave clinically in a some-
Chondrosarcoma of Vertebral Column and Sacrum.  what similar manner. The overall 5-year survival rate
Chondrosarcomas of the vertebral column and sacrum varies between 48% and 60%. Recurrences in chondro-
are relatively infrequent (~5% of all cases). Typically they sarcoma typically occur 5 to 10 years or more after
originate in a vertebral body and extend into the adjacent surgery. In some instances, recurrence can be associated
soft tissue and spinal canal.73,87,90 Chondrosarcoma has with an increase in histologic grade and more aggressive
some predilection for the lower thoracic and lumbar clinical behavior than that of the primary neoplasm.55
segment of the spine and for the sacrum. It is somewhat Distant metastases are most often observed in grade 3
less common in the upper thoracic and cervical spine. chondrosarcoma (66%), and the lungs are the most fre-
Radiologically chondrosarcoma presents as a radiolucent quent site. Grade 2 lesions sometimes give rise to distant
defect. If typical punctate calcifications are present, the metastases (10%). Chondrosarcoma metastasizes to the
cartilaginous nature of the lesion can be suspected lymph nodes more frequently than other bone sarcomas.
on radiographs. The extent of involvement is best dem- Other less frequent sites of metastasis are the liver,
onstrated by CT and MRI. Radiographically, chondro- kidneys, and brain. Grade 1 chondrosarcomas do not
sarcoma of the spine must be differentiated from metastasize, but a fatal outcome in 10% of cases results
osteosarcoma, giant cell tumor, osteoblastoma, and meta- from local uncontrollable growth. In addition, in rare
static tumors. Plain radiographs may reveal only a com- cases, low-grade chondrosarcoma can recur as a high-
pression fracture of the body of a vertebra. grade pleomorphic (dedifferentiated) neoplasm. Although
remote, the possibility of dedifferentiation should be
Chondrosarcoma of Small Bones of Hands and Feet.  considered if conservative management of a cartilage
The majority of cartilaginous lesions of the small bones lesion is planned.
of the hands and feet are clinically benign and represent Tumors that are not amenable to complete excision
enchondromas. On average, enchondromas in this loca- because of their location, such as the skull base, are
tion are more cellular and have more nuclear atypia than treated by conventional radiotherapy for limited local
in other parts of the skeleton. Pathologic fractures are control in the postoperative setting or in advanced inop-
often present, and reparative change may complicate the erable cases with definitive intent. The use of proton
microscopic interpretation of enchondromas. Although therapy after maximal surgical resection in such locations
very rare (1% of all cases), chondrosarcomas do occur in offers increased probability of a long-term cure with rela-
the acral skeleton (Figs. 7-29 to 7-31). The diagnosis of tively low risk of significant complications.4,67
rare cases of chondrosarcoma distal to wrist and ankle
joints is based on strict correlation between the radio- Personal Comments
graphic and pathologic data.22,92 A solitary cartilaginous
lesion of the acral skeleton should be suspected of being The most common diagnostic problem in chondrosar-
a chondrosarcoma if it shows increased cellularity micro- coma is the evaluation of an intramedullary cartilage
scopically with multiple plump cells exhibiting an open tumor, most often in a long bone of an extremity that
nuclear structure and atypia. The cartilage lesions at shows equivocal histologic features and is borderline in
these sites should be considered chondrosarcomas if an size. The presence of radiologic changes in the adjacent
infiltrative destructive growth pattern is documented cortex and surrounding cancellous bone and the clinical
microscopically and is confirmed by radiographic imaging history regarding the events that led to the discovery of
(Figs. 7-30 and 7-31). the cartilage lesion are of paramount importance in the
recognition of a low-grade chondrosarcoma. The wide-
spread use of radioisotope scanning with bone-seeking
Treatment and Behavior
isotopes and MRI, both of which are extremely sensitive
The successful eradication of a malignant cartilage tumor in demonstrating focally increased mineral turnover and
depends on complete wide excision, if this is technically marrow alterations produced by small asymptomatic
feasible. Surgery is the primary and most effective treat- lesions, have led to more frequent identification of
ment for chondrosarcoma. Other modalities, such as irra- cartilage tumors. The use of these techniques has multi-
diation and chemotherapy, play a minor role and only plied the frequency with which these cartilaginous masses

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 507

B
FIGURE 7-29  ■  Conventional chondrosarcoma: radiographic and gross features. A, Lateral radiograph of foot shows radiodense,
heavily calcified chondrosarcoma in medullary cavity of calcaneus. B, Sagittally cut specimen from below-knee amputation shows
grade 1 chondrosarcoma of calcaneus with extension into soft tissue. Note ivory-like areas corresponding to zones of enchondral
ossification in hyaline cartilage of tumor.

ERRNVPHGLFRVRUJ
508 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-30  ■  Conventional chondrosarcoma of phalanx. A and B, Frontal and oblique radiographs of chondrosarcoma involving
proximal phalanx in third finger of a 72-year-old man. Tumor has ill-defined margins, permeative growth pattern, and matrix calci-
fication. C, Ray amputation specimen cut sagittally to show expanded contour of phalanx and extension of tumor into soft tissue.
D, Intermediate-power photomicrograph shows features of grade 2 chondrosarcoma. (D, ×200) (D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 509

A B C

D E

FIGURE 7-31  ■  Conventional chondrosarcoma of thumb. A, Lateral radiographic image showing a destructive lesion involving the
proximal phalanx of the thumb. Note a lesion with punctuate calcifications involving the distal phalanx corresponding to an enchon-
droma. B, Fat-saturated T2-weighted sagittal magnetic resonance image showing a signal intensity lesion of the proximal phalanx
with extension to the paraosseous soft tissue. C, Bisected resection specimen showing a diffusely growing intramedullary lesion
with the cortical breakthrough to the soft tissue (arrow) involving the proximal phalanx. D and E, Low power photomicrographs
showing aggressive growth pattern of a chondrosarcoma permeating the medullary cavity of the thumb. Inset, Intermediate power
photomicrograph documenting the high cellularity of this chondrosarcoma present universally throughout the lesion. (D and E, ×20;
inset, ×100) (D, E, and inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
510 7  Malignant Cartilage Tumors

must be evaluated histologically. In general, a borderline


lesion should be suspected to be a chondrosarcoma if Dedifferentiated Chondrosarcoma
radiologically it provokes a sclerotic reaction with thick- Definition
ening of the adjacent cortex, if there is an alteration in
the trabecular pattern in the surrounding cancellous bone The phenomenon of the transformation of a low-grade
(usually loss of trabeculae), and if there is associated pain, (grade 1 or 2) chondrosarcoma into a high-grade sarcoma
particularly at rest. Enchondromas can certainly produce that most frequently exhibits features of malignant fibrous
endosteal scalloping in the adjacent cortex, especially in histiocytoma or osteosarcoma is referred to as dedifferen-
small tubular bones, but in large lesions of long bones, tiation. Described in 1971 by Dahlin and Beabout,109
this must always be regarded as a radiologic sign of dedifferentiated chondrosarcoma is a prototype for all
growth. dedifferentiated tumors. More generally, the term dedif-
In enchondromatosis (Ollier’s disease), the recognition ferentiation is used when a high-grade sarcoma develops
of secondary malignant change may be extremely difficult in association with a preexisting locally aggressive low-
if the pathologist is not familiar with the spectrum of grade tumor. In addition to chondrosarcoma in bone,
radiologic appearances of this disorder because the diag- dedifferentiation occurs in giant cell tumor, low-grade
nosis of secondary malignancy often cannot be based on intramedullary and surface (parosteal) osteosarcomas,
histologic criteria alone. This results from the fact that and some soft tissue neoplasms such as liposarcoma.
in Ollier’s disease, the dysplastic cartilage can be highly Dedifferentiation also occurs in extraskeletal chondrosar-
cellular and show mild to moderate atypia and prominent coma and chondrosarcomas arising in preexisting condi-
myxoid changes. Extension of cellular cartilaginous tissue tions (secondary chondrosarcomas).100,101,104,126
into the adjacent soft tissue and a clinical history of a Microscopically, dedifferentiated chondrosarcoma is
newly discovered mass and associated pain are the most defined as a lesion in which the two components—a high-
consistent signals of malignant transformation. grade sarcoma with or without heterologous elements
Synovial chondromatosis is a lesion of soft tissue or (e.g., malignant fibrous histiocytoma, osteosarcoma,
joints that offers many interpretive pitfalls to the unsus- and rhabdomyosarcoma) and a low-grade cartilaginous
pecting pathologist, particularly when it involves unusual lesion—coexist in one tumor and there is abrupt clear
sites, such as the temporomandibular joint and spinal demarcation between them.106,109,110,114,119,124,139 The onset
facet joints. Prominent nuclear pleomorphism and chon- of dedifferentiation is signified clinically by a sudden
drocyte multinucleation should not be regarded as evi- increase in aggressiveness, usually with lethal conse-
dence of malignancy in this synovial metaplastic condition. quences. On the other hand, a gradual increase in the
When bone erosion occurs in long-standing synovial grade of malignant cartilage tumors in itself does not
chondromatosis, it can be mistaken for chondrosarcoma. warrant the use of the term dedifferentiation.
In these cases, it is essential to look for the presence
of affected synovium to distinguish this lesion from Incidence and Location
chondrosarcoma.
Chondrosarcoma also may be mimicked in two other In all major series, dedifferentiated chondrosarcoma rep-
nonneoplastic conditions: pubic osteolysis and topha- resents less than 10% of all chondrosarcomas. The SEER
ceous pseudogout. In bulky soft tissue deposits of calcium data analysis, based on 2757 cases of chondrosarcoma,
pyrophosphate dihydrate (pseudogout), erosion into shows that only 5.3% of these tumors fall into the cate-
bone (hip joint, temporomandibular joint) can occur, and gory of dedifferentiated chondrosarcoma. Therefore, it
the chondroid metaplasia frequently found in the con- can be anticipated that dedifferentiation theoretically
nective tissue surrounding the tophaceous pseudogout may occur in approximately 1 of 10 to 20 untreated chon-
deposit can be misinterpreted as chondrosarcoma. This drosarcomas.93,109,110,114,119,124 The anatomic distribution of
error can be avoided by the recognition of the specific dedifferentiated chondrosarcoma is similar to that of
nature of the pseudogout crystalline deposit, which is not conventional chondrosarcoma and supports the relation-
seen in the calcifications of chondrosarcoma. ship between the two conditions (i.e., the development
Pubic osteolysis is a reparative nonneoplastic condi- of a high-grade sarcoma in association with a preexisting
tion that is often rich in cellular cartilage associated with conventional chondrosarcoma). The femur is the most
pelvic insufficiency fractures. When the fracture callus frequently involved bone, where nearly 30% of all cases
produces a significant soft tissue mass, it can raise suspi- of dedifferentiated chondrosarcoma are diagnosed. It is
cion of a pubic chondrosarcoma. If a pathologist is followed by the pelvis (20%), humerus (16%), ribs (7%),
presented with a small biopsy sample from such a mass, and scapula (7%). The age range is wide, but the majority
it is all too easy to make an erroneous diagnosis of of tumors occur in patients older than age 50 years. The
chondrosarcoma unless radiologic correlation is pursued mean age is approximately 60 years. The peak age inci-
scrupulously. dence and most common skeletal sites are shown in
Cartilaginous lesions of the testes, ovaries, and perito- Figure 7-32.
neal or mediastinal soft parts are typically teratomatous
in nature. In such instances, meticulous search for other Clinical Symptoms
components of germ cell neoplasia is advised before the
lesion is classified as chondrosarcoma in these unusual Pain is usually the presenting complaint. Occasionally, a
sites. clinical history of prolonged local discomfort that had

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 511

Peak
incidence
50
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 7-32  ■  Dedifferentiated chondrosarcoma. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites
are indicated by solid black arrows.

changed more recently with the development of a rapidly indicate that a high-grade lesion is present. When these
enlarging mass can be obtained. Dedifferentiation of a features are present in association with an otherwise
preexisting low-grade lesion can result in pathologic frac- radiographically typical cartilaginous lesion, they suggest
ture, and almost 50% of the patients initially have this dedifferentiation.
complication. Computed tomography and MRI may be used for
more accurate resolution of smaller lytic areas of dedif-
ferentiation or the presence of a residual calcified tumor
Radiographic Imaging
within large lytic areas. These imaging techniques also
The existence of the two components (low- and high- help in evaluating the extent and relationships of any
grade) of dedifferentiated chondrosarcoma frequently associated soft tissue component (Figs. 7-33 and 7-34).
can be recognized radiographically.106,109-111,114,119,124,136 They are also useful in guiding needle biopsies to
The typical radiographic presentation of dedifferentiated obtain material from areas suspected of representing
chondrosarcoma is an area of punctate opacities sur- dedifferentiation.136
rounded by purely lytic areas that exhibit a complete loss
of bony architecture or a permeative growth pattern. Gross Findings
Complete cortical disruption and extension into soft
tissue, with or without a displaced pathologic fracture, The two components of the tumor—a low-grade carti-
are almost always present (Figs. 7-33 and 7-34). The laginous lesion and a high-grade dedifferentiated
high-grade component can dominate the lesion, and only sarcoma—can often be identified grossly. The precursor
small areas of calcified cartilaginous tumor may be identi- lesion has the typical gross appearance of a lobulated
fied on radiographs. On the other hand, in the initial cartilaginous mass that is present at the periphery of or
stages of dedifferentiation, a relatively minor component within a sarcomatous fleshy mass (Figs. 7-33 to 7-36).
of dedifferentiated sarcoma may not be identifiable on The high-grade sarcomatous component may show
radiographs. As previously mentioned, features such as hemorrhage and necrosis. Cortical disruption and
early cortical disruption in a relatively small lesion or the extension into soft tissue are frequently present. The
presence of lytic areas with a destructive growth pattern Text continued on p. 516

ERRNVPHGLFRVRUJ
512 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-33  ■  Dedifferentiated chondrosarcoma: radiographic and gross features. A, Frontal radiograph of right proximal humerus
of a 69-year-old man with destructive tumor in humeral head and proximal shaft. Punctate calcifications are present within lytic
area proximally, and cortex is absent medially. B, T1-weighted coronal magnetic resonance image of tumor in A reveals tumor mass
of low-density signal; tumor is invading soft tissue. C, Resected specimen of tumor shown in A. Lobulated cartilage can be seen in
proximal and mid-portion surrounded by fleshy hemorrhagic tumor that erodes cortex. D, Specimen radiograph of C shows calcified
chondroid matrix.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 513

A B

C D
FIGURE 7-34  ■  Dedifferentiated chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph of the proximal
humerus showing a destructive lytic process involving the head and neck and a calcified lesion corresponding to a preexisting low
grade chondrosarcoma (arrows). B, Fat-saturated T2-weighted coronal magnetic resonance image showing biphasic tumor with a
high signal intensity component corresponding to a dedifferentiated high-grade sarcoma with soft tissue extension medially. The
low-grade intramedullary chondrosarcoma shows intermediate signal intensity and corresponds to calcified component shown on
plain radiograph in A. C, Bisected resection specimen shows an intramedullary cartilaginous component distally and a high-grade
destructive mass involving the head and neck with soft tissue extension medially. D, Closer view of C showing cortical breakthrough
and soft tissue extension.

ERRNVPHGLFRVRUJ
514 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-35  ■  Dedifferentiated chondrosarcoma: gross features. A, Coronal section through distal femoral tumor in a 74-year-old
woman. Lobulated, peripherally calcified, cartilaginous tumor is seen distally, and fleshy high-grade sarcoma is seen proximally,
extending into adjacent soft tissue. B, Coronal section through proximal femoral tumor. Lobulated cartilaginous intramedullary
tumor is seen distally, and hemorrhagic necrotic high-grade sarcoma involves neck area proximally. C, Intramedullary low-grade
cartilaginous component is seen on both sides of pathologic fracture in proximal femoral shaft. High-grade sarcoma with hemor-
rhage and necrosis is present at site of pathologic fracture. D, Pathologic fracture at site of dedifferentiation in tumor involving distal
femur. (C and D, courtesy Dr. A.G. Ayala, Houston.)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 515

A B

C D
FIGURE 7-36  ■  Dedifferentiated chondrosarcoma. A, Anteroposterior radiograph of right hip of a 72-year-old woman with 2-month
history of pain. Central area of dense calcification is bordered by large zone of lucency medially. B, Coronally sectioned proximal
femur shown in A after radical en bloc excision. Note separate appearances of two components of tumor with fleshy sarcomatous
portion above and medial to calcified cartilage. C, Low power photomicrographs show sharp transition between low-grade chon-
drosarcoma component and high-grade spindle-cell tumor above (×100). D, Higher power view of dedifferentiated component with
features of malignant fibrous histiocytoma. (C, ×100; D, ×200) (C and D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
516 7  Malignant Cartilage Tumors

preexisting cartilage tumor can be engulfed and partially are extremely rare. Divergent epithelial differentiation
destroyed by the high-grade component. In such within a dedifferentiated component is extremely rare.112
instances, the low-grade tumor can be reduced to several It must be mentioned that several distinct pathways of
scattered residual foci that can be easily overlooked on divergent differentiation (i.e., smooth muscle, rhabdo-
gross examination. If a pathologic fracture is present, myoblastic, epithelial) can be observed within one tumor.
extensive hemorrhage can obscure the gross details of the In the past, lesions that exhibited these features were
lesion. Careful sampling of the intramedullary compo- classified as malignant mesenchymomas. Currently this
nent can reveal the microscopic foci of the precursor diagnostic designation is rarely used in reference to these
cartilage lesion. lesions.

Microscopic Findings Special Techniques


The hallmark of dedifferentiated chondrosarcoma is the The two components of dedifferentiated chondrosarcoma
coexistence of its two components: a low-grade cartilagi- have distinct immunohistochemical profiles.112,120,124,143,144
nous precursor lesion and a high-grade sarcoma.106,190- The low-grade cartilage precursor lesion recapitulates
111,114,119,123,124,126
Typically, there is an abrupt zone where the immunohistochemical profile of enchondroma/
a low-grade cartilage lesion changes to a high-grade conventional chondrosarcoma and expresses the markers
sarcoma. If the precursor lesion was partially destroyed, of cartilage lineage differentiation such as SOX9 and
it can be present in the form of several separate nodules S-100. The dedifferentiated component typically loses
measuring from less than 1 cm to several centimeters. the phenotype of a well differentiated cartilage lesion
The low-grade cartilage lesion is sharply demarcated and has immunohistochemical features overlapping with
from the surrounding dedifferentiated component high-grade spindle-cell sarcoma or malignant fibrous
(Figs. 7-36 to 7-41). The absence of any gradual transi- histiocytoma. The immunohistochemical features of
tion between the cartilage lesion and the high-grade cartilaginous differentiation may be occasionally posi-
sarcoma is an important feature diagnostically. It is tive within the high-grade component, especially in the
useful in distinguishing dedifferentiated chondrosarcoma areas of divergent cartilaginous differentiation. Diver-
from other sarcomas that can contain malignant cartilage gent differentiation in the high-grade component can be
and that may show gradual transition to a spindle-cell documented by appropriate smooth and skeletal muscle
morphology. and epithelial markers such as SMA, desmin, myoglobin,
The precursor cartilage lesion is typically located keratin, and epithelial membrane antigen.128,129,143,144
within the intramedullary portion of the tumor. The rel- Ultrastructural features of the precursor cartilage
atively small volume of the low-grade chondrosarcoma lesion are identical to those seen in grades 1 and 2 chon-
component may require intensive sampling to establish drosarcoma. The dedifferentiated component consists of
its presence. Most frequently, the precursor cartilagi- highly pleomorphic mesenchymal cells that may show
nous lesion has features of a grade 1 chondrosarcoma, or features of divergent differentiation (i.e., osteoblastic,
so-called borderline cartilage lesion, with an enchondroma- chondroblastic, smooth muscle, or rhabdomyoblastic)
like appearance (Fig. 7-41). Sometimes the underlying (Fig. 7-44).99,118,143
cartilage tumor is a grade 2 chondrosarcoma that may DNA ploidy measurements have shown that the low-
have myxoid features. In general, the precursor carti- grade precursor cartilage lesion is diploid and that the
lage tumor in which dedifferentiation occurs is a low- dedifferentiated component is almost always aneuploid,
to intermediate-grade (grade 1 or 2) chondrosarcoma usually having multiple clones of cells with an abnormal
(Figs. 7-36 to 7-40). Grade 3 chondrosarcoma is a DNA content.103,122,145 Multiple genetic and molecular
rare precursor lesion of dedifferentiated chondrosar- studies provide strong evidence that the dedifferenti-
coma. The dedifferentiated component most frequently ated component represents a clonal evolution from the
exhibits features of a high-grade spindle-cell sarcoma low-grade cartilaginous component.102,105,122,131,140-142
that has features of malignant fibrous histiocytoma or These studies also indicate that the so-called collision
osteosarcoma (Figs. 7-36 to 7-41). The dedifferenti- concept of dedifferentiated chondrosarcoma is no longer
ated osteosarcomatous component may show features valid and is only of historical interest. Molecular studies
of telangiectatic osteosarcoma.130,132 More rarely, dedif- disclose complex chromosomal alterations with involve-
ferentiation may occur as a high-grade sarcoma rich in ment of multiple molecular pathways in the develop-
giant cells that resembles a giant cell variant of malignant ment of dedifferentiation. These include the genetic and
fibrous histiocytoma (Figs. 7-42 and 7-43). Such tumors epigenetic alterations of TP53, RB1, H-ras, cell-cycle
can be mistaken for giant cell tumors.113 Rhabdomyo- regulatory pathway (p21WAF1, p16INK4, p14ARF),
blastic differentiation is less frequent, but occasionally apoptosis (DAPK, FHIT), DNA repair (hMLH1), and
the dedifferentiated component can have microscopic, cell adhesion (E-cadherin).107,108,116,125,134,135,137,140,142 Acti-
immunohistochemical, and ultrastructural features of vation of enzymes that participate in the destruction of
rhabdomyosarcoma of the pleomorphic type (Fig. the intercellular matrix and their interacting molecules is
7-44).121,129,133 Features of smooth muscle differentiation a common event in a high-grade sarcomatous component
can also be present.112,128 The dedifferentiated compo- of the dedifferentiated chondrosarcoma and may play
nent is typically of high histologic grade regardless of its a contributory role to the aggressive behavior of these
phenotype. Examples of dedifferentiation with features malignancies.117,138
of a low-grade fibroblastic osteosarcoma or fibrosarcoma Text continued on p. 525

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 517

A B

C D
FIGURE 7-37  ■  Dedifferentiated chondrosarcoma: microscopic features. A, Abrupt demarcation between low-grade (grade 1) cartilage
precursor lesion and high-grade dedifferentiated component. B, Two components of dedifferentiated osteosarcoma. Note osteoid
deposition within dedifferentiated tumor. C, Somewhat irregular but still abrupt demarcation between low-grade cartilaginous
and dedifferentiated components. D, Grade 2 myxoid chondrosarcoma as precursor lesion in dedifferentiated chondrosarcoma.
(A-D, ×200; A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
518 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-38  ■  Dedifferentiated chondrosarcoma: microscopic features. A, Low power photomicrograph shows overall architecture
of dedifferentiated chondrosarcoma with abrupt demarcation between low-grade cartilage precursor lesion (lower part) and high-
grade dedifferentiated component (upper part). B, High power photomicrograph of A shows cytoarchitectural details of a high-grade
spindle-cell component. C, Osteoid production in other areas of the same tumor, disclosing osteoblastic lineage differentiation in
dedifferentiated component. D, Highly pleomorphic features in other parts of the same tumor. Inset, Nuclear details of the pleomor-
phic component of the tumor. (A and C, ×100; B and D, ×200; inset, ×400) (A-D and inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 519

A B

C D
FIGURE 7-39  ■  Dedifferentiated chondrosarcoma: microscopic features. A and B, Less clear demarcation between dedifferentiated
high-grade sarcomatoid component of the tumor and cartilaginous precursor lesion. C and D, Intermediate and high power photo-
micrographs showing pleomorphic features of a high-grade sarcomatoid component of the same tumor shown in A and B. (A and
B, ×100; C, ×200; D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
520 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-40  ■  Dedifferentiated chondrosarcoma: microscopic features. A, Sharp demarcation between the cartilaginous lesion (lower
part) and high-grade sarcomatoid component of the tumor (upper part). B, Intermediate photomicrograph showing the morphology
of the cartilage precursor lesion in the tumor shown in A. C and D, Intermediate and high power photomicrographs showing extreme
pleomorphism and bizarre nuclear features within a dedifferentiated sarcomatoid component of the tumor shown in A and B.
(A, ×100; B and C, ×200; D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 521

A B

C D
FIGURE 7-41  ■  Dedifferentiated chondrosarcoma: microscopic features. A, Sharp demarcation between cartilaginous (lower part) and
dedifferentiated (upper part) components of the tumor (×100). B, Higher magnification showing spindle-cell component of the tumor
shown in A (×200). C, Higher magnification showing low cellularity and bland nuclear features of the cartilaginous precursor lesion
(×200). D, Lobular growth pattern with trabeculae of bone embracing cartilage islands within the cartilaginous precursor component
of the tumor. The bland nuclear features and the lobular growth pattern throughout the entire cartilaginous precursor lesion implicate
that the precursor lesion in this particular instance may in fact represent an enchondroma rather than a low-grade chondrosarcoma.
(A, ×100; B and C, ×200; D, ×50) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
522 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-42  ■  Dedifferentiated chondrosarcoma mimicking giant cell tumor: radiographic and microscopic features. A, Frontal
radiograph of left proximal humerus with destructive tumor in humeral head and proximal metaphyseal area. B, Fat-saturated
T2-weighted coronal magnetic resonance image (MRI) of tumor reveals heterogeneous signal intensity with low signal intensity
areas composed of cartilage lobules. Note multifocal cortical destruction. C, T2-weighted coronal MRI with lobulated focally
confluent high signal intensities corresponding to the cartilaginous component of the tumor. D, Low power photomicrograph
showing typical pattern of dedifferentiated chondrosarcoma with sharp transition between low-grade chondrosarcoma and non-
chondroid dedifferentiated component. (D, ×50) (D, hematoxylin-eosin) (Reprinted with permission from Estrada EG, et al: Ann Diagn
Pathol 6:159-163, 2002.)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 523

A B

C D

FIGURE 7-43  ■  Dedifferentiated chondrosarcoma mimicking giant cell tumor: microscopic features. A, Photomicrograph showing
low-grade chondrosarcoma juxtaposed to the nonchondroid giant cell-rich lesion (×100). B, View of nonchondroid component with
many multinucleated osteoclastic giant cells mimicking conventional giant cell tumor of bone (×200). C, High power view of dedif-
ferentiated component devoid of giant cells and composed predominantly of mononuclear cells (×200). D, Area of dedifferentiated
component with stromal spindle cells showing atypia. Inset, Atypical nuclei within dedifferentiated component. (A, ×100; B-D, ×200;
inset, ×400) (A-D and inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
524 7  Malignant Cartilage Tumors

FIGURE 7-44  ■  Dedifferentiated chondrosarcoma: ultrastructural features. A, Low power magnification of cartilaginous precursor
lesion exhibits features of low-grade conventional chondrosarcoma. Note cartilage cell with abundant glycogen surrounded by
fibrillar matrix. B, Dedifferentiated component exhibits features of divergent differentiation ranging from smooth muscle (inset: top)
to rhabdomyoblastic with well-developed sarcomeric structures (inset: bottom). (A, ×5000; B, ×4000; insets, ×13,000)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 525

Differential Diagnosis dedifferentiated sarcoma. The possibility of this highly


lethal phenomenon should be taken into account when
High-grade conventional chondrosarcoma may show spin- conservative management of relatively indolent low-
dling of the tumor cells at the periphery of tumor lobules, grade chondrosarcoma is considered.
but the transition from malignant chondrocytes is a
gradual one. The distinct biphasic character of dediffer-
entiated chondrosarcoma with sharp demarcation of the Clear Cell Chondrosarcoma
two components is lacking. Mesenchymal chondrosarcoma Definition
also shows a more gradual transition between the chon-
droid elements and the undifferentiated round-cell or Clear cell chondrosarcoma is defined as a low-grade
spindle-cell component than that seen in dedifferentiated malignant cartilaginous tumor in which the tumor cells
chondrosarcoma. The latter occurs generally in a much contain a significant amount of glycogen, which is respon-
older population of patients than mesenchymal chondro- sible for the clear cell appearance of their cytoplasm.
sarcoma. Because the most common form of dedifferen- The hallmark of this tumor is its overall mimicry of
tiation in chondrosarcoma is that of malignant fibrous chondroblastoma.
histiocytoma, small biopsy samples may be misinterpreted
as malignant fibrous histiocytoma when the cartilaginous Incidence and Location
elements are not found. Primary malignant fibrous his-
tiocytoma in bone has a better prognosis than dediffer- Clear cell chondrosarcoma is a rare tumor that accounts
entiated chondrosarcoma; hence, the differential diagnosis for less than 5% of all chondrosarcomas. The SEER data
is important in predicting the outcome of therapy. Atten- indicate that only 36 (1.3%) of all 2757 chondrosarcomas
tion to the radiographic finding of cartilage matrix calci- were classified as clear cell chondrosarcomas. Therefore,
fication is essential in recognizing the true nature of the clear cell chondrosarcoma is among the rarest forms of
tumor in such cases and in guiding the selection of a cartilage tumors. The limited experience with these
biopsy site. Fibrous dysplasia containing abundant cartilage lesions indicates that the peak incidence is during the
(fibrocartilaginous dysplasia) may be mistaken for dedif- third and fourth decades of life, but individual cases are
ferentiated chondrosarcoma, but the bland appearance of scattered from the second to ninth decades.149,159,163,174,176
the cells in the fibrous component and the lack of cellular There is a clear male predominance with a male-to-
atypia in the cartilaginous lobules, as well as the nonag- female ratio of 2.4 : 1. Clear cell chondrosarcoma has a
gressive radiologic features of this lesion, are helpful in predilection for the ends of long tubular bones and
recognizing this rare benign entity. extends to the articular cartilage. The proximal end of
Metastatic high-grade spindle-cell sarcomas in bone, the femur is the most frequently affected site, where
including leiomyosarcoma and rhabdomyosarcoma, can approximately 45% of the cases occur. The second most
suggest the diagnosis of dedifferentiated chondrosar- frequently affected site is the proximal end of the humerus.
coma. In such cases, a careful search for a precursor The remaining cases are relatively uniformly scattered
cartilaginous tumor is essential. throughout the skeleton. The trunk bones, including the
spine, as well as craniofacial bones, including extraskel-
etal sites such as the larynx, can also be sporadically
Treatment and Behavior
involved.156,165,170 The most common skeletal sites and
Dedifferentiated chondrosarcoma is highly lethal, and peak age incidence are shown in Figure 7-45.
20% of patients have metastasis at the time of initial
diagnosis.115,127 The initial prognosis was dismal with less Clinical Symptoms
than a 10% survival rate after 1 year. Widespread hema-
togeneous metastases are the rule, and the response to Pain in the affected area is usually the presenting
chemotherapy is poor. In general, the dedifferentiated symptom. Clear cell chondrosarcoma is a slow-growing
component is more resistant to chemotherapy and radia- lesion, and frequently there is a history of pain or minor
tion therapy than de novo malignant fibrous histiocytoma regional discomfort of several years’ duration.
or osteosarcoma. Widespread metastases occur early
despite surgery. Modern chemotherapy protocols seem Radiographic Imaging
to be more effective than older regimens, and the survival
rates of recently treated cases appear to be improv- Clear cell chondrosarcoma typically involves the ends of
ing.115,127 However, the overall long-term prognosis is long tubular bones and extends to the epiphysis and the
still dismal, with practically no long-term survivors. articular cartilage.151,159,169,173 It produces lytic defects that
Recent analysis of 266 patients with dedifferentiated may resemble chondroblastoma. The lesion can be radio-
chondrosarcoma by a European group provide a more lucent or heavily mineralized. The defect is usually
optimistic scenario and indicated that the overall survival sharply demarcated and can have sclerotic margins (Figs.
of patients with dedifferentiated chondrosarcoma treated 7-46 to 7-49). More advanced lesions produce expansion
with modern chemotherapy protocols was 28% at 10 of the end of a bone. Secondary aneurysmal bone cyst
years.115 The poor prognostic factors were the presence may be responsible for blowout features. Clear cell chon-
of a pathologic fracture at diagnosis, a pelvic location, drosarcomas are relatively slowly growing lesions that
and being older than age 60 years.115 A low-grade expand the bone contour (Fig. 7-49). Cortical disruption
chondrosarcoma rarely recurs as a highly aggressive is not a typical feature of this tumor. Because of the

ERRNVPHGLFRVRUJ
526 7  Malignant Cartilage Tumors

Peak
incidence
20 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 7-45  ■  Clear cell chondrosarcoma. Peak age incidence and frequent sites of skeletal involvement. Most frequent site is indi-
cated by solid black arrow.

involvement of the ends of long tubular bones, it must of the tumor cell embedded in a loose cartilaginous matrix.
be differentiated from chondroblastoma and giant cell The cells are large with an oval contour and distinct cell
tumor on radiographs. boundaries (Fig. 7-50). The centrally located nuclei have
prominent nucleoli. Multinucleated osteoclast-like giant
cells are typically present. Scattered trabeculae of bone
Gross Findings
between groups of clear cells are frequently seen (Figs.
The cartilaginous nature of the lesion is difficult to rec- 7-51 and 7-52). Foci of coarse calcification are present, and
ognize on gross examination. The lesion is typically well occasionally large areas of the tumor can be heavily calci-
circumscribed and is composed of soft, gray tissue that fied. Large solid areas with more conventional appearance
can be focally calcified (Figs. 7-47 to 7-49). The presence and well developed hyaline cartilage matrix can be present
of multiple trabeculae of bone within the tumor and (Fig. 7-53). The cellularity of the lesion is very high, but
cartilage calcification may impart a gritty consistency to the nuclei of the tumor cells are relatively uniform and
the tissue. Prominent cystic change and hemorrhage have a low mitotic rate. The lesion has an overall lobu-
can be present. Small lesions are confined to the bone lated architecture accentuated by thin fibrovascular cores.
and are typically well demarcated. The larger masses Although it is grossly and radiographically well demar-
expand the bone contour, erode the cortex, and promote cated, microscopically the periphery of the lesion dis-
periosteal new bone formation (Fig. 7-49). The tumor closes an infiltrative growth pattern similar to that seen in
can occasionally cause disruption of the bony end plate conventional chondrosarcomas. Although this tumor has
and extend to involve joint tissues. overlapping clinical and radiographic features with chon-
droblastoma, its microscopic appearance is distinct from
chondroblastoma. The latter does not contain prominent
Microscopic Findings
clear cells or trabeculae of bone. These trabeculae seem
This tumor exhibits one of the most distinctive histologic to be a part of the lesion; their nature (i.e., tumor bone
appearances of all bone tumors.147,153,155,158,162,169,171,173,174,176,177 versus reactive bone) is unclear and remains the subject of
The characteristic feature is the abundant clear cytoplasm Text continued on p. 535

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 527

A B

FIGURE 7-46  ■  Clear cell chondrosarcoma: radiographic features. A and B, Anteroposterior and lateral radiographs showing a lesion
with calcified matrix involving the distal femoral end. C and D, T2-weighted sagittal magnetic resonance images (MRIs) showing an
intermediate signal intensity lesion involving the anterior aspect of the femoral head. Inset, Fat-saturated T2-weighted axial MRI
showing signal enhancement in the lesion involving the medial femoral condyle.

ERRNVPHGLFRVRUJ
528 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-47  ■  Clear cell chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph of left hip shows tumor
involving femoral head and neck in a 35-year-old man. Note punctate and ringlike calcifications in central portion of lesion.
B, Radiograph of hip shows calcified tumor occupying femoral head of a 27-year-old man. C, Anteroposterior radiograph of right
shoulder of a 35-year-old man with tumor in proximal end of humerus containing punctate calcifications. D, Anteroposterior radio-
graph of lytic tumor with sclerotic margins involving right femoral neck area. Inset, Resected femoral head shows variegated appear-
ance of tumor with gritty hemorrhagic and chalklike foci of calcification.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 529

FIGURE 7-48  ■  Clear cell chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph of pelvis shows a tumor
with punctuate calcification occupying the left femoral head. B, T1-weighted coronal magnetic resonance image showing a tumor
of low signal intensity involving the femoral head. Note punctate signal voids to tumor matrix calcifications. C, Bisected resection
specimen shows a mineralized tumor involving the femoral head. Inset, Closer view documenting variegated appearance of the
tumor.

ERRNVPHGLFRVRUJ
530 7  Malignant Cartilage Tumors

A B

C D

F G

FIGURE 7-49  ■  Clear cell chondrosarcoma: radiographic and gross features. A, Anteroposterior (AP) radiograph showing a well
demarcated lytic lesion of the femoral head. B, Axial computed tomogram showing well demarcated lesion of the femoral head
with punctate calcifications. C, Fat-saturated T2-weighted coronal magnetic resonance image (MRI) showing well demarcated lesion
of the femoral head with high signal intensity. D, Bisected resection specimen showing a well demarcated lesion within the femoral
head with variegated mineralized pattern. E, AP radiograph showing a lesion expanding the proximal fibular end with delicate
punctate calcifications. F, Fat-saturated T2-weighted coronal MRI of the lesion shown in A documenting high signal intensity in the
clear cell chondrosarcoma of the proximal fibular end. G, Bisected resection specimen of the lesion shown in A and F shows var-
iegated mineralization pattern in the clear cell chondrosarcoma expanding the proximal end of the fibula.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 531

A B

C D

FIGURE 7-50  ■  Clear-cell chondrosarcoma: microscopic features. A and B, Intermediate and high power photomicrographs show
sheets of clear cells with discrete focal woven trabeculae of bone, typically present in clear cell chondrosarcoma. C and D, Interme-
diate and high power photomicrographs show solid proliferation of clear cells with centrally located nuclei. (A and C, ×200; B and
D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
532 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-51  ■  Clear-cell chondrosarcoma: microscopic features. A-D, Solid proliferation of clear tumor cells with interspaced well
developed woven bone in clear cell chondrosarcoma. This feature can lead to problems in differential diagnosis with osteosarcoma.
(A and C, ×200; B and D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 533

A B

C D
FIGURE 7-52  ■  Clear-cell chondrosarcoma: microscopic features. A, Areas of conventional hyaline cartilage (lower part) and prominent
woven trabeculae of bone (upper part) in clear cell chondrosarcoma. B, A higher magnification of A showing gradual transition from
cartilage matrix to bony matrix production. C and D, Intermediate and high power photomicrographs showing prominent woven
trabeculae in clear cell chondrosarcoma. Again, these features can present differential diagnostic problems with osteosarcoma, in
particular of chondroblastic type. (A, ×100; B and C, ×200 D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
534 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-53  ■  Clear-cell chondrosarcoma: microscopic features. A-D, Low and intermediate power photomicrographs showing solid
areas of hyaline cartilage matrix and its gradual transition to osteoid appearing deposition. Such deposits are referred to as osteo-
chondroid matrix. (A-C, ×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 535

debate. Similar to conventional osteosarcoma, clear cell absence of malignant osteoblastic components excludes
chondrosarcoma can dedifferentiate.158 Dedifferentiation osteosarcoma.
in the form of high-grade undifferentiated sarcoma can Conventional chondrosarcomas may show prominent vac-
occur in a primary tumor at the time of initial diagnosis uolization of the malignant cartilage cells, even in low-
or can present as a recurrence or as metastatic disease at grade tumors. This artifact can usually be distinguished
distant sites.158 from the clear cell morphologic features of this variant.
The absence of the other common microscopic features
Special Techniques of prominent osteoclast-like giant cells and trabeculae of
reactive bone in conventional chondrosarcoma helps in
Ultrastructurally, the tumor cells contain a large amount the differential diagnosis.
of glycogen, which is responsible for the clear cell appear- Metastatic clear cell carcinoma in bone and other clear
ance of their cytoplasm and which can be documented cell lesions involving bone can be distinguished by the
histochemically by the demonstration of large amounts absence of a chondroid matrix and by the immunohisto-
of material positive for periodic acid-Schiff stain, which chemical demonstration of S-100 protein, which is
is diastase sensitive in the cytoplasm of these cells (Fig. intensely positive in the characteristic cells of clear cell
7-54).147,150,155,161,162,167 Matrix composition studies identify chondrosarcoma.
the expression of cartilage collagen type II and proteogly-
cans, confirming the cartilaginous nature of tumor cells
Treatment and Behavior
in clear cell chondrosarcoma.146,150,153 Consistent with
this impression, clear cell chondrosarcomas are strongly Clear cell chondrosarcomas are low-grade malignant
positive for S-100 protein.146,175-177 Our personal experi- tumors, but they have definite metastatic poten-
ence with these tumors indicates that they also express tial.148,149,152,154,157,163 Because of their relatively indolent
SOX9. The cartilaginous nature of the tumor is further clinical course and radiologic overlap with chondroblas-
confirmed by the expression of parathyroid hormone- toma, some of these tumors have been initially inade-
related peptide (PTHrP) and PTH/PTHrP receptor, quately treated. Marginal excision or curettage usually
which are involved in the biology of cartilage.160 The results in recurrence. The overall recurrence rate is 16%.
presence of well developed trabeculae of bone in these Complete en bloc excision should be performed if cure
tumors is considered to be an integral part of the lesion is expected in the treatment of clear cell chondrosarcoma.
and implicates the bone-forming activity of tumor cells. In nearly all larger series of clear cell chondrosarcoma,
Consistent with this concept is the expression of the there are cases in which the tumor metastasized. Of the
so-called bone forming proteins, which contribute to the reported cases, approximately 10% of patients died of
discussion on the possible histogenesis and nature of tumor-related causes. Seven of eight cases in which the
these enigmatic tumors.150 More recent studies confirm tumors metastasized had recurrences and then spread to
frequent expression of epithelial markers, with the most the lungs. Brain and bone metastases have also occurred.
frequent expression of AE1/AE3 keratins in the majority Recurrences have been noted up to 13 years after incom-
of clear cell chondrosarcomas and occasional expressions plete removal. Patients in whom metastases developed
of CK7, CK8, CK18, and CK20.164 Clear cell chondro- died an average of 7 years after initial therapy. Although
sarcomas frequently show overexpression of p53 protein there are insufficient data to evaluate the effectiveness of
but without associated mutations.168 Limited cytogenetic chemotherapy, it appears to provide some lengthening of
studies provide information on clonal chromosomal survival. Several patients lived for years with known
abnormalities with considerable case-to-case heterogene- pulmonary metastases while receiving adjuvant chemo-
ity. Extra copies of chromosome 20 and rearrangements therapy. In view of the fact that clear cell chondrosarcoma
of 9p appear to be recurrent.166,172 occurs so frequently at the ends of the long bones of the
extremities, en bloc resection with joint replacement may
provide ideal surgical treatment by combining radical
Differential Diagnosis
excision and limb preservation. More recent follow-up
Because of its location in the epiphyseal ends of long studies confirm these trends of clinical behavior.157 It
bones, its cartilaginous matrix with calcification, and appears that the overall recurrence rate is in the range of
its occasional occurrence beyond the second and third 20% with a median of 1.7 years after the initial diagnosis.
decades of life, benign chondroblastoma may present dif- The rate of distant metastasis is similar (approximately
ferential diagnostic problems with clear cell chondro- 20%), with a median time to original diagnosis of approx-
sarcoma. Benign chondroblastoma can be recognized by imately 8 years.157
the absence of clear cell morphologic features or the
reactive trabeculae of bone that are characteristic of
clear cell chondrosarcoma. Furthermore, chondroblas- Mesenchymal Chondrosarcoma
tomas almost invariably manifest a self-limited indolent Definition
growth pattern. The presence of trabeculae of bone
with osteoblastic and osteoclastic rimming in clear cell Mesenchymal chondrosarcoma is a rare, distinct malig-
chondrosarcoma may lead to the misdiagnosis of osteo- nant neoplasm characterized by the presence of solid,
blastoma or even osteosarcoma. The presence of a promi- highly cellular areas composed of round or slightly spin-
nent cartilage matrix and clear cell morphologic features dled primitive mesenchymal cells with foci of cartilagi-
militates against the diagnosis of osteoblastoma, and the nous differentiation. The noncartilaginous elements

ERRNVPHGLFRVRUJ
536 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-54  ■  Clear-cell chondrosarcoma: microscopic features. A, Solid areas of tumor cells with clear cytoplasm, granular cyto-
plasm, or both and centrally located nuclei. Woven trabeculae of bone are interspersed among tumor cells. B, Tumor cells are
strongly positive for S-100 protein. C, Tumor cells are strongly positive for periodic acid-Schiff stain. D, High magnification shows
abundance of cytoplasmic granules positive for periodic acid-Schiff stain. (A-C, ×200; D, ×800.) (A, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 537

Peak
incidence
10 30
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 7-55  ■  Mesenchymal chondrosarcoma. Peak age incidence and frequent sites of skeletal involvement.

usually predominate, and such lesions can be confused organs were also described.197,209 The peak age incidence
histologically with Ewing’s sarcoma or hemangiopericy- and sites most frequently involved are shown in Figure
toma. Mesenchymal chondrosarcoma was described by 7-55.
Lichtenstein and Bernstein200 in 1959. It is clinically,
radiographically, and microscopically distinct from con- Clinical Symptoms
ventional and dedifferentiated chondrosarcoma.
Pain and swelling of the affected area are the primary
initial symptoms. The symptoms may persist for more
Incidence and Location
than 1 year. Mesenchymal chondrosarcoma can present
Mesenchymal chondrosarcoma is extremely rare and as a soft tissue mass.
accounts for less than 5% of all chondrosarco-
mas.179,191,192,195,205,213,219 The SEER data indicates that Radiographic Imaging
only 98 (3.6%) out of 2757 chondrosarcomas were clas-
sified as mesenchymal chondrosarcoma. The limited Mesenchymal chondrosarcoma appears radiographically
experience with these lesions indicates that they usually as a radiolucent lesion with varying degrees of
affect young adults and teenagers. The peak incidence is matrix calcification. Lesions involving craniofacial bones
during the third decade of life. Male and female patients frequently represent masses at the base of the skull or
are almost equally affected. The maxilla and mandible are maxillary tumor masses with a destructive growth pattern
the frequent sites of involvement.198,210,216,220 The other (Figs. 7-56 and 7-57). In long tubular bones the lesions
frequent sites are the vertebrae, ribs, pelvis, and humerus. are often eccentrically located (Fig. 7-58). In extraskeletal
The involvement of the central nervous system both locations, the lesions are well demarcated and frequently
cranial and spinal is typical for this tumor.212,214,221 The affect the paraosseous soft tissue (Figs. 7-57 and 7-58). It
bones of the appendicular skeleton are rarely involved. is estimated that in nearly half stippled calcifications are
Approximately 30% of mesenchymal chondrosarcomas frequently present and may coalesce to form large, dis-
present as extraskeletal soft tissue lesions. Rare examples crete opacities (Figs. 7-60 to 7-62). Occasionally, mesen-
of mesenchymal chondrosarcoma arising in visceral chymal chondrosarcomas present as heavily calcified

ERRNVPHGLFRVRUJ
538 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-56  ■  Mesenchymal chondrosarcoma of the craniofacial bones: radiographic features. A, Fat-saturated T1-weighted coronal
magnetic resonance image (MRI) with contrast showing high signal intensity lesion involving the orbit and the anterior cranial fossa.
B, Fat-saturated T2-weighted axial MRI showing intermediate signal intensity mass involving the sphenoid sinus. C and D, Fat-
saturated T1-weighted axial MRIs with contrast showing tumors of variegated signal intensity involving the left and right maxillary
sinus, respectively. Note extension to the interior cranial fossa in C.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 539

A B

C D

FIGURE 7-57  ■  Mesenchymal chondrosarcoma: radiographic features. A and B, Reformatted coronal bone and soft tissue computed
tomographic (CT) images of skull of a 16-year-old boy with calcified tumor involving the right ethmoid and maxillary sinuses and
extending into orbit and anterior cranial fossa. C and D, Anteroposterior radiographic image and axial CT image of a paraspinal
mass in a 55-year-old woman. Heavy calcified eccentric tumor at junction of rib and vertebral transverse process is present.

ERRNVPHGLFRVRUJ
540 7  Malignant Cartilage Tumors

A B

C D

FIGURE 7-58  ■  Mesenchymal chondrosarcoma: radiographic features. A, Lytic lesion in body of mandible, which is devoid of matrix
calcification. B, Heavily mineralized chest wall mass in a 10-year-old boy. C, Soft tissue mass with prominent punctate calcifications
adjacent to proximal femoral shaft in a 48-year-old woman. D, Eccentric lucent tumor involving anterior surface of distal tibial shaft.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 541

masses. Stippled calcifications disclosing the cartilagi- chondrosarcoma. The difference is in the mesenchymal
nous nature of the lesion are also frequently present in component, which shows rhabdomyoblastic or lipoblas-
the soft tissue lesions.193,196,216 Prominent extensions into tic differentiation either focally or diffusely distributed
soft tissue are seen in about 50% of cases (Figs. 7-59 to in the undifferentiated mesenchyme surrounding the
7-61). The overall growth pattern is that of a highly cartilage (Fig. 7-71). The two patterns (i.e., focal and
aggressive lesion with permeative or moth-eaten radio- diffuse) can coexist in one lesion. There are very few
graphic appearance (Figs. 7-59 and 7-60). Surprisingly, cases described in the literature, and this limited experi-
this highly aggressive tumor may occasionally have a ence does not permit a definitive statement as to whether
sharply demarcated sclerotic margin. these lesions represent a distinct entity or whether some
of them are variants of mesenchymal chondrosarcoma.
Gross Findings The tumor cells in the cartilaginous areas of these lesions
stain positively for S-100 protein, and the cells exhibiting
The tissue is gray to pink with foci of calcification. The rhabdomyoblastic differentiation are strongly positive for
level of mineralization can vary, and occasionally large desmin (Fig. 7-72). Lipoblastic differentiation, if present,
areas of heavily calcified tissue can be present. The tumor is associated with positivity for S-100 protein.
may have an overall lobulated architecture and is typically
well demarcated from the surrounding bone and soft
Special Techniques
tissue (Figs. 7-60 to 7-62). Disruption of the cortex and
extension into soft tissue are frequently seen. Larger Ultrastructurally, mesenchymal chondrosarcoma is com-
areas of cartilaginous differentiation can sometimes be posed of oval or plump elongated primitive mesenchymal
grossly identified. Zones of necrosis and hemorrhage can cells.202,217 Foci of cartilaginous differentiation show more
be present. mature cells with well-developed cytoplasm containing
glycogen, rough endoplasmic reticulum, and numerous
mitochondria (Fig. 7-73). The cells in these foci are sur-
Microscopic Findings
rounded by a granular low-density matrix consistent with
Mesenchymal chondrosarcoma is composed of solid areas chondroid. Mesenchymal chondrosarcomas are distinct
of primitive round or slightly spindle-shaped mesenchymal from the rest of conventional chondrosarcomas and do
cells (Figs. 7-63 to 7-69). The primitive mesenchymal not carry IDH mutations. The immunohistochemical
component frequently exhibits hemangiopericytoma- staining pattern is nonspecific and implicates cartilage
like features with multiple endothelial-lined vascular lineage differentiation.182,185
spaces. Islands of cartilage are present within these solid The small cell component of mesenchymal chondro-
areas.179,191,192,195,205,213,216,219 The level of cartilaginous sarcoma has a phenotype of chondroprogenitor cells and
differentiation can vary from small foci with a loose shows strong nuclear expression of SOX9 protein.225
arrangement to large areas of mature cartilage with a These cells also express p30/34 (MIC2) protein.194
relatively well-differentiated appearance (Figs. 7-63 to Cartilaginous areas continue to express SOX9 and
7-67). The cartilaginous component frequently contains gain the expression of S-100 protein.186,204,207,218,225 The
foci of coarse calcification. Enchondral ossification or latter is typically negative in the small cell component
even the formation of bone with marrow spaces contain- of the tumor. The small cells of mesenchymal chon-
ing hematopoietic elements can be seen (Fig. 7-72). The drosarcoma frequently show divergent differentiation.
proportion of cartilaginous and primitive elements can Scattered positivity of isolated small cells for desmin is a
vary among the tumors and in different areas of the same relatively common finding.187,190,194 In rare instances, the
lesion. At one end of the spectrum are tumors composed small cell component of mesenchymal chondrosarcoma
predominantly of primitive mesenchymal cells, which may show diffuse rhabdomyoblastic differentiation that
require differentiation from other round-cell malignan- is associated with positivity for skeletal muscle differ-
cies such as Ewing’s sarcoma and small cell osteosarcoma. entiation and expression of such markers as desmin and
On the opposite side of the spectrum are predominantly myogenin.190,194
cartilaginous lesions with a relatively benign appearance, SOX9 is a valuable marker of mesenchymal chon­
in which the presence of a minor primitive mesenchymal drosarcoma, differentiating it from other small cell
component can be overlooked (Fig. 7-64). The carti- malignancies. It is not however entirely specific because
laginous foci contain cells that show strong reactivity its expression is common in all cartilage lineage tumors,
for S-100 protein, whereas the undifferentiated cells are both benign and malignant. It is also expressed in other
negative for this marker (Fig. 7-70). Pleuropulmonary blas- soft tissue tumors such as synovial sarcoma.183 A wide
toma, or the so-called pulmonary blastoma of childhood, is range of solid tumors including carcinomas can express
a tumor that occurs in the soft tissue of the chest wall SOX9 as a part of the phenomenon referred to as epithe-
and lungs in young children.178,180,188,196,199,201,211,215,222,223 lial to mesenchymal transition. In addition, the lack of
These neoplasms are characterized by the presence of expression of FLI1 can be used as a biomarker in the
small primitive cells with blastomatous qualities and differential diagnosis of mesenchymal chondrosarcoma
may exhibit divergent rhabdomyoblastic, cartilaginous, with Ewing’s sarcoma.199
and lipoblastic differentiation. The prognosis for these Recent identification of HEY1-NCOA2 and IRF2BP2-
patients is grave because more than 50% die within 2 CDX1 gene fusions in mesenchymal chondrosarcoma is
years after diagnosis. At least some of these tumors have of potential major importance for differential diagnosis
microscopic features similar to those of mesenchymal Text continued on p. 557

ERRNVPHGLFRVRUJ
542 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-59  ■  Mesenchymal chondrosarcoma: radiographic features. A, Lateral radiograph showing a destructive tumor with anterior
extension involving the distal femoral metaphysis. B, T1-weighted sagittal magnetic resonance image (MRI) showing a low signal
intensity intramedullary tumor with anterior cortical penetration and soft tissue extension. C, Fat-saturated T2-weighted sagittal MRI
showing increased signal intensity in the intramedullary tumor involving the distal femoral metaphysis. D, Fat-saturated T2-weighted
axial MRI showing high signal intensity of the intramedullary tumor with anterior cortical penetration and semi-circumferential
extension to the paraosseous soft tissue anteriorly.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 543

A B

C D

FIGURE 7-60  ■  Mesenchymal chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph showing a calcified
destructive tumor mass involving proximal humerus. Note circumferential cortical destruction and extension to soft tissue with
radiating mineralized spicules. Such a mineralization pattern can be radiographically misinterpreted as an osteosarcoma. B, Fat-
saturated T2-weighted axial magnetic resonance image showing high signal intensity tumor with circumferential extension to
paraosseous soft tissue. C and D, Gross photographs of the bisected resection specimen in A and B showing fleshy tumor mass
involving medullary cavity of proximal humerus with circumferential soft tissue extension.

ERRNVPHGLFRVRUJ
544 7  Malignant Cartilage Tumors

C
FIGURE 7-61  ■  Mesenchymal chondrosarcoma: radiographic and gross features. A and B, Oblique and anteroposterior radiographs
showing a calcified tumor involving the scapula. C, Resected specimen shows a tan lobulated tumor extending anteriorly and pos-
teriorly to the periscapular soft tissue.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 545

C
FIGURE 7-62  ■  Mesenchymal chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph shows a calcified
mass in the right paraspinal region. B, Axial computed tomogram demonstrates a calcified mass in the soft tissue of the right
paraspinal region. C, Resection specimen shows a well demarcated soft tissue mass.

ERRNVPHGLFRVRUJ
546 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-63  ■  Mesenchymal chondrosarcoma: microscopic features. A, Low power photomicrograph showing small cell tumor with
discreet ill-defined patches of early cartilaginous differentiation shown as less cellular areas. B, Interface between an island of atypi-
cal cartilage and small cell undifferentiated components of the tumor. C, Small cell proliferation with prominent open vascular
channels of various sizes and shapes (×100). D, Higher magnification showing solid proliferation of undifferentiated small cells. (A-C,
×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 547

A B

C D

FIGURE 7-64  ■  Mesenchymal chondrosarcoma: microscopic features. A and B, Biphasic nature of mesenchymal chondrosarcoma
exemplified by ill-defined islands of cartilaginous differentiation and poorly differentiated small cell component. C, Large area of
cartilaginous differentiation with well developed hyaline cartilage matrix and lacunar pattern of cartilage cells. D, Higher magnifica-
tion of poorly differentiated somewhat spindle-cell component of the tumor. (A-C, ×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
548 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-65  ■  Mesenchymal chondrosarcoma: microscopic features. A and B, Interface between the cartilaginous areas and a small
cell poorly differentiated component of the tumor. Note ill-defined irregular interface between the hyaline cartilage areas and undif-
ferentiated component. C, Large cartilaginous area of the tumor with well developed hyaline cartilage matrix. Note atypia of cartilage
cells. D, Intermediate power view of poorly differentiated small cell component of the tumor. (A and B, ×100; C and D, ×200)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 549

A B

C D
FIGURE 7-66  ■  Mesenchymal chondrosarcoma: microscopic features. A, Low power photomicrograph of the central portion of a large
cartilaginous area showing vascular ingrowth and enchondral ossification of the calcified matrix. B, More cellular, poorly differenti-
ated component of the tumor. C and D, Variations of morphology within the poorly differentiated component of the tumor showing
spindle (C) and round (D) cell variants within the same tumor. Inset, Higher magnification showing nuclear details of the round cell
component of the tumor. (A, ×50; B, ×100; C and D, ×200; inset, ×400) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
550 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-67  ■  Mesenchymal chondrosarcoma: microscopic features. A and B, Low power photomicrographs showing dense prolif-
eration of undifferentiated tumor cells with prominent capillary vasculature. C and D, Low and intermediate power photomicrographs
showing ill-defined islands of cartilaginous differentiation. (A-C, ×100; D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 551

A B

C D
FIGURE 7-68  ■  Mesenchymal chondrosarcoma: microscopic features. A and B, Low and intermediate power photomicrographs
showing spindle cell variant of mesenchymal chondrosarcoma. Tumors with such features can be confused with synovial sarcoma.
C and D, Low and intermediate power photomicrographs showing mesenchymal chondrosarcoma with prominent vasculature.
(A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
552 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-69  ■  Mesenchymal chondrosarcoma: microscopic features. A-D, Intermediate and high power photomicrographs
of mesenchymal chondrosarcoma showing proliferation of short plump spindle cells. (A and D, ×200; B, ×400; C, ×100)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 553

A B

C D
FIGURE 7-70  ■  Mesenchymal chondrosarcoma: microscopic features. A-D, Low and high power photomicrographs of mesenchymal
chondrosarcoma show intense immunoreactivity for S-100 protein in cartilaginous areas and absence of staining in primitive mes-
enchymal cells. (A and C, ×150; B and D, ×300)

ERRNVPHGLFRVRUJ
554 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-71  ■  Thoracopulmonary blastoma: microscopic features. A and B, Low and medium power photomicrographs of S-100
protein immunoreactivity confined to cells in chondroid component of thoracoblastoma. C and D, Same tumor as in A and B dem-
onstrates strongly positive desmin stain in primitive spindle-cell elements of this unique tumor. (A and C, ×100; B and D, ×200)
(Courtesy Dr. J.Y. Ro, Houston.)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 555

B
FIGURE 7-72  ■  Mesenchymal chondrosarcoma: microscopic features. A, Low power photomicrograph of mesenchymal chondrosar-
coma of rib. Tumor contained radiographic evidence of marked calcification. Note coarse foci of calcification of cartilaginous
component. B, Higher power magnification of A shows vascular ingrowth and endochondral ossification of calcified matrix.
Note osteoclastic resorptive activity. Inset, Detail of matrix calcification. (A, ×100; B, and inset, ×200) (A, B, and inset,
hematoxylin-eosin)

ERRNVPHGLFRVRUJ
556 7  Malignant Cartilage Tumors

B
FIGURE 7-73  ■  Mesenchymal chondrosarcoma: ultrastructural features. A, Focus of cartilaginous differentiation shows tumor cells
surrounded by fibrillar matrix. B, Undifferentiated mesenchymal cells have scanty cytoplasm and sparse organelles. Inset, High
magnification of area exhibiting early chondroid differentiation shows loose substance with fibrillary elements. (A, ×4000; B, ×3000;
inset, ×30,000)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 557

NCOA2
Chr 8
23.3 23.2
23.1
22
4 3 2 1
21.3
21.1 21.2
12 23 22 21 20 19 18 17 16 15 14 13 12 11 10 9 8 7 6 5
11.2
11.1 breakpoint
11.1
11.22
11.21
11.23 HEY1
12
13 NCOA2 Interstitial deletion
or
21.1 5 4 32 1
21.2 HEY1 t(8;8)(q13;q21) translocation breakpoint
21.3
22.1 NCOA2 HEY1
22.2
22.3
23

24.1
23 22 2120 19 1817 16 15 14 13 4 3 2 1
24.2
24.3
breakpoint
A 148364022 bp NCOA2 portion HEY1 portion
HEY1
308 aa

NCOA2
1464 aa
Fusion protein
HEY1 NCOA2
706 aa

breakpoint bHLH – basic helix-loop-helix motif


PAS – Per Arndt-Sim motif
HEY1 portion NCOA2 portion MYC-TYPE – Myc-type, basic, helix-loop-helix (bHLH) domain
MOT01-04 – LXXLL motif
NID – nuclear receptor interaction domain
AD1/CID – transcriptional activation domain I/CBP/p300 interaction domain
MOTLXX – LLXXLXXXL motif
Q-rich – glutamine rich region
B AD2 – transcriptional activation domain 2
FIGURE 7-74  ■  Molecular structure of HEY1-NCOA2 fusion gene and its chimeric protein in mesenchymal chondrosarcoma.
A, Chromosomal locations and exon-intron structures of NCOA2 and HEY1 genes. Solid black arrows indicate the locations of the
breakpoints within introns. The molecular structure of the resulting chimeric gene includes coding sequences of the segment of
NCOA2 and segment of HEY1. B, Functional domains of the two proteins. The HEY1 protein contains MYC-type domain at the
N-terminus part of the protein. The NCOA2 protein contains multiple functional domains throughout the protein. In the chimeric
protein the C-terminus transcriptional activation domains (AD1/CID and AD2) of NCOA2 partner genes are present and are respon-
sible for the transcriptional upregulation of the target genes.

and may offer new therapeutic targets for these aggres- protein causes global transcriptional activation of the
sive malignancies.208,224 genes involved in cell proliferation.
The HEY1 and NCOA2 genes are about 10 Mb apart, The second fusion gene identified in mesenchymal
mapping to 8q21.1 and 8q13.3, and the HEY1-NCOA2 chondrosarcoma involves IRF2PB2-CDX1, resulting
fusion most likely results from a cryptic interstitial dele- from cytogentically detectable reciprocal translocation
tion of del(8)(q13.3q21.1) (Fig. 7-74). The NCOA2 gene t(1;5)(q42;q32) (Fig. 7-75). The 3′ partner of the fusion,
is a 3′ fusion partner and belongs to the p160 nuclear CDX1, belongs to the homeobox gene family, which
hormone receptor transcriptional coactivator family. share their homeobox domain encoding a DNA binding
NCOA2 binds the ligand-bound nuclear receptor through protein functioning as a transcription factor. The CDX1
its nuclear receptor interaction domain (NID) and its gene is an upstream regulator of Hoxgene expression,
C-terminal transcriptional activation domains (AD1/ implicated in the development of leukemias and some
CID and AD2). The complex recruits histone acetyl- solid human tumors such as colon cancer. The 5′ end
transferases and methyltransferases, inducing chromatin partner, IRF2BP2, encodes two isoform proteins result-
remodeling and transcriptional activation of target genes. ing from alternative splicing of the gene. Both isoforms
The 5′ partner of the fusion, HEY1, is a downstream contain a Zinc finger motif at their N-terminus involved
effector of the Notch pathway, which activates its expres- in DNA binding. The direct involvement of IRF2BP2
sion. It is hypothesized that the HEY1-NCOA2 chimeric in human carcinogenesis has not been reported but its

ERRNVPHGLFRVRUJ
558 7  Malignant Cartilage Tumors

Chr1 Chr5 der(1) der(5)


36.3 15.3 36.3 15.3
36.2 36.2
15.2 15.2
36.1 15.1 36.1 15.1
35 14 35 14
34.3 34.3
34.1
34.2 13.3 13.2 34.1
34.2 13.3 13.2
IRF2BP2 1q42.3
33 13.1 33 13.1
32.3 12 32.3 12
32.1 32.2 11 11.1 32.1 32.2 11 11.1 Isoform A
31.3 11.2 31.3 11.2
31.2 31.2
12 12 Isoform B
31.1 13.1 31.1 13.1
13.3 13.2 13.3 13.2 1 2
22.3
22.2
22.3
22.2
breakpoint
22.1 14 22.1 14 breakpoint
21
15
21
15 CDX1 5q32
21 21
13.3 13.3
13.2 22 13.2 22
13.1 12 23.1 13.1 12 23.1
11 23.2 11 23.2 1 2 3
11 23.3 11 23.3
12 31.1 12 31.1
31.2 31.2
31.3 CDX1 31.3
21.1 21.1 41
21.2 33.1 32 21.2 silent
21.3 21.3
22 33.3 33.2 22
23 34 23
35.1 35.2
24 24
25
35.3
25 IRF2BP2 CDX1 Isoform A

31 31
1 2 3 Isoform B
32.1 32.1
32.2 32.2 32.3 breakpoint
32.3
IRF2BP2 IRF2BP2 portion CDX1 portion
41 IRF2BP2 41
42.1 CDX1
42.2 42.3 fusion gene Fusion transcripts
43
44 B
t(1;5)(q42;q32)

IRF2BP2
587 aa
CDX1
Fusion proteins 265 aa
IRF2BP2 CDX1
466 aa

450 aa

breakpoint Interferon regulatory factor 2-binding protein 1&2,


Zink finger domain
IFR2BP2 portion CDX1 portion Caudal-like activation region
C Homeodomain
FIGURE 7-75  ■  Molecular structure of reciprocol translocation t(1;5)(q42;q32) and IRF2BP2-CDX1 fusion genes in mesenchymal chon-
drosarcoma. A, Chromosomal locations of the IRF2BP2 and CDX1 genes and the diagram representing the reciprocal translocation
with IRF2BP2-CDX1 fusion on der(1). The breakpoint on der(5) is silent. B, Exon-intron structures of the IRF2BP2 and CDX1 genes.
Solid black arrows indicate the locations of the breakpoints within introns. The molecular structure of the resulting chimeric gene
includes coding sequences of the N-terminal domain of IRF2BP2 and the homeodomain with the C-terminal segment of CDX1.
C, Functional domains of the two proteins. The IRF2BP2 protein contains a Zinc finger motif at the N-terminus involved in DNA
binding. The CDX1 protein contains caudal-like activation region and homeodomain. In the chimeric protein the C-terminus of the
IRF2BP2 protein is linked to the N-terminus of the CDX1 protein. The resulting hybrid protein contains a Zinc finger domain from
IRF2BP2 and homeodomain from CDX1 and is anticipated to transcriptionally upregulate multiple target genes involved in
self-proliferation.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 559

interacting proteins involve such well-known tumor sup- absence of their respective skeletal muscle and lymphoid
pressor genes as TP53 and the oncogene IRF2. Similar to biomarkers. The complementary cytogenetic and molec-
other chimeric proteins, the IFR2PB2-CDX1 hybrid ular studies for specific chromosomal translocation and
protein is anticipated to induce transcriptional upregula- fusion genes are used to further classify the tumors.
tion of multiple target genes involved in cell prolifera- Ewing’s sarcomas typically are positive for the break-
tion. Additional chromosome translocations and sole points and fusion genes involving EWSR1. It can be
cytogenetic abnormalities were identified and imply anticipated that the newly discovered HEY1-NCOA2
genetic diversity of the underlying mechanisms.189,206 and IRF2BP2-CDX1 fusion genes will be used in the
The limited experience with both fusion genes nearest future to confirm the diagnosis of mesenchymal
does not permit a definite statement concerning their chondrosarcoma in difficult cases. In rare instances, other
frequency of involvement and specificity for differential round-cell tumors such as small cell variant of synovial
diagnosis of mesenchymal chondrosarcoma, but extrap- sarcoma can be confused with extraskeletal mesenchymal
olating the usefulness of similar gene fusions in other chondrosarcomas. The diagnostic difficulties can be
human malignancies one can anticipate major use of further amplified by the frequent positivity of synovial
these findings in the differential diagnosis of small cell sarcoma for SOX9. In such instances molecular testing
malignancies. Additional findings suggest involvement of for SYT breakpoints or fusion transcripts typical for
MYC and mTOR pathways.181,203 synovial sarcoma can solve the diagnostic dilemma. The
diagnostic criteria for small cell osteosarcoma and its dif-
Differential Diagnosis ferential diagnosis are described in Chapter 5.
The distinct problems in differential diagnosis of mes-
The distinction between mesenchymal chondrosarcoma enchymal chondrosarcoma are created by the presence
and dedifferentiated chondrosarcoma usually is not difficult of extensive areas with relatively mature cartilage and
because of the abrupt boundaries seen histologically minimal atypia. In such lesions, the small cell compo-
between the cartilaginous components of dedifferenti- nent can be obliterated by the cartilaginous areas and is
ated chondrosarcomas and the high-grade, anaplastic not as striking to the observer on microscopic inspec-
component. In mesenchymal chondrosarcoma, the two tion as in typical cases. Such lesions can be mistaken
elements of this biphasic tumor blend more gradually. for benign cartilage conditions or can be misclassified
Furthermore, the distinctive round-cell or hemangio- as conventional chondrosarcomas or chondroblastic
pericytomatous appearance of the nonchondroid element osteosarcomas. Familiarity with the spectrum of dif-
facilitates the recognition of mesenchymal chondro- ferent presentations of mesenchymal chondrosarcoma
sarcoma. The rather striking age differences between as well as attention to the presence of undifferentiated
these two variants of chondrosarcoma are also useful. small cell or spindle cell components among larger
Mesenchymal chondrosarcoma is a tumor of the young, areas of cartilaginous differentiation is helpful to avoid
whereas dedifferentiated chondrosarcoma almost always misdiagnosis.
occurs in elderly patients. Dedifferentiated chondrosar- Hemangiopericytomas and other primary spindle-cell
comas are most common in the appendicular skeleton, malignancies that arise in bone do not show the chon-
whereas mesenchymal chondrosarcomas often affect the droid components that are present in mesenchymal
axial and craniofacial skeleton. Histologically, dediffer- chondrosarcomas.
entiated chondrosarcomas most frequently present as
malignant fibrous histiocytomas or as high-grade osteo-
Treatment and Behavior
sarcomas complicating low-grade chondrosarcoma; they
do not manifest the undifferentiated round-cell–like or Surgery is the primary treatment in mesenchymal chon-
hemangiopericytoma-like pattern characteristic of mes- drosarcoma.213 Some suggest that tumors with so-called
enchymal chondrosarcoma. Ewing-like microscopic features respond somewhat
The resemblance to a malignant round-cell neoplasm better to combination chemotherapy than those with
in the nonchondroid parts of a mesenchymal chondro- spindle-cell and hemangiopericytoma-like areas.195 How­
sarcoma causes confusion in some cases with Ewing’s ever, this concept is not universally accepted, and the
sarcoma of bone or its related tumors (e.g., primitive neu- clinical data that justify this approach are not convincing.
roectodermal tumors, Askin’s tumor) as well as with The limited experience with these rare tumors indicates
embryonal rhabdomyosarcoma and malignant lymphoma that mesenchymal chondrosarcomas are clinically aggres-
involving bone. Similarly, small cell osteosarcoma could sive, highly lethal lesions that have a high propensity to
conceivably be confused with mesenchymal chondrosar- metastasize. The overall 10-year survival rate is approxi-
coma. The differential diagnosis of these tumors, gener- mately 28%. More than 50% of affected patients die
ally referred to as small cell malignancies, is a complex within 5 years. More recent studies with long-term
process that involves a combination of microscopic follow-up confirm this initial impression and indicate
features and biomarker profiles revealed by immunohis- that for patients who are treated with complete surgical
tochemistry or histochemistry, complemented by cytoge- excision and chemotherapy, the 10-year disease-free sur-
netic and molecular studies. The expression of SOX9 and vival is 76%. In patients who cannot have their tumors
the absence of FLI1 are the features of mesenchymal completely excised due to anatomic location or advanced
chondrosarcoma. In contrast, Ewing’s sarcomas are typi- state of disease and cannot successfully complete chemo-
cally FLI1 positive and SOX9 negative. Rhabdomyosar- therapy protocols, the 10-year disease-free survival drops
comas and lymphomas are typically excluded by the to 17%.184

ERRNVPHGLFRVRUJ
560 7  Malignant Cartilage Tumors

intramedullary chondrosarcoma. The typical lesion pres-


JUXTACORTICAL CHONDROSARCOMA ents as a large mass that measures more than 5 cm in
Definition diameter.

Juxtacortical chondrosarcoma (periosteal chondrosar- Microscopic Findings


coma) is a type of conventional chondrosarcoma that
develops on the surface of bone. It may show cortical Microscopic features are those of conventional chondro-
erosion, but the tumor is extramedullary. sarcoma. Most frequently, juxtacortical chondrosarcoma
is a low- to intermediate-grade (grade 1 or 2) lesion (Figs.
7-77 and 7-79). The high-grade cartilaginous lesions of
Incidence and Location
the bone surface should be differentiated from periosteal
Juxtacortical chondrosarcoma is a very rare lesion, with osteosarcoma, which typically contains prominent carti-
only individual cases reported in the major series of bone laginous components, and its bone-forming component
tumors.226,229,232-234,236 In the current SEER data, only 26 may not be represented in limited biopsy material. The
(0.9%) of 2757 chondrosarcomas were classified as juxta- most important features differentiating juxtacortical
cortical chondrosarcomas.227,229,236,237 This data indicates chondrosarcoma from juxtacortical chondroma are the
that juxtacortical chondrosarcoma is among the rarest presence of nuclear atypia throughout the lesion and the
forms of all chondrosarcomas. This lesion typically aggressive infiltrative growth into the underlying cortex.
involves the surface of the diaphysis or metaphysis of Rare examples of dedifferentiated juxtacortical chondro-
major long tubular bones, such as the femur or tibia. sarcoma have been described.230
Single cases involving the flat bones and craniofacial
bones have also been reported.228,235 Most patients with
Differential Diagnosis
this tumor are adults in the third or fourth decade of life;
the male-to-female ratio is 1 : 1. The most important lesions that present differential
diagnostic problems for this type of chondrosarcoma are
periosteal chondroma (juxtacortical chondroma) and periosteal
Clinical Symptoms
osteosarcoma.
A firm, bulky mass adherent to the surface of bone is a Periosteal chondromas seldom exceed 2 to 3 cm in size
typical presenting sign of juxtacortical chondrosarcoma. and usually show radiographic features of solid periosteal
The patient may also report pain, discomfort, and ten- bone buttresses. Histologically, they show less overall
derness over the affected area. If the lesion is near a joint, cellularity than juxtacortical chondrosarcomas and lack
limitation of motion can be present. The duration of the cytologic atypicality of chondrosarcoma. Periosteal
symptoms is usually long. In some instances, regional osteosarcoma is a malignant surface lesion of bone with
lymph node metastasis can be present at the time of cells that produce a predominantly chondroid matrix;
original diagnosis.231 however, tumor osteoid and bone production by tumor
cells can always be found in spindle-cell areas between
the cartilage lobules. It is this finding that allows this
Radiographic Imaging
low- to intermediate-grade osteosarcoma to be distin-
Juxtacortical chondrosarcoma radiographically pres- guished from surface chondrosarcoma.
ents as a radiolucent surface bone lesion (Figs. 7-76 to Less frequent surface lesions such as florid reactive peri-
7-78).226,227,232-234,236,237 If the typical punctate matrix cal- ostitis, subungual exostosis, and bizarre parosteal osteocarti-
cifications are present, its cartilaginous nature may be laginous proliferation (Nora’s lesion) have been mistaken for
recognized (Fig. 7-76). The subperiosteal location of the surface chondrosarcomas because of their often cellular,
lesion, which is separated from the medullary cavity by a proliferative, nonneoplastic cartilage components. The
rim of cortical bone and covered by an elevated perios- usual anatomic site (acral parts) and lack of true cytologic
teum, is best documented by CT and MRI (Fig. 7-76). atypia, as well as the clinical and radiologic features of
Erosion of the underlying outer cortical surface is usually these lesions, are helpful in separating them from the
present. The lesion is usually well demarcated from the neoplastic group.
adjacent soft tissue and from the underlying cortex. Cod-
man’s triangles are not seen. Elevation of the periosteum
with multilayered periosteal new bone formation can be SECONDARY CHONDROSARCOMAS
present on the bone surface adjacent to the lesion. The
prominent perpendicular new bone formation with a hazy Secondary chondrosarcomas are morphologically and
lesional-cortical border seen in periosteal osteosarcoma radiographically similar to conventional chondrosar-
is typically not present in juxtacortical chondrosarcoma. coma and cannot be distinguished microscopically
without other supportive data, such as clinical and radio-
graphic findings. Secondary chondrosarcomas are dis-
Gross Findings
tinguished by their development in association with a
A lobulated mass adherent to the bone surface and sur- predisposing condition that can be identified clinically,
rounded by a fibrous capsule is easily identified as car- radiographically, or occasionally microscopically. A
tilaginous on gross examination. The gross appearance list of predisposing conditions for the development
of the tumor tissue is identical to that of conventional Text continued on p. 565

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 561

C D
FIGURE 7-76  ■  Juxtacortical chondrosarcoma: radiographic features. A and B, Anteroposterior and lateral radiographs of juxtacortical
focally calcified tumor on anterior surface of distal femoral shaft in a 25-year-old woman (arrows). C and D, T1-weighted sagittal
magnetic resonance images (MRIs) with and withour contrast of juxtacortical chondrosarcoma of proximal tibial metaphysis in a
13-year-old boy. Inset, Fat-saturated T2-weighted axial MRI demonstrates high-intensity signal in surface cartilage tumor. Histologi-
cally, this tumor showed prominent myxoid features and microscopic evidence of marrow invasion.

ERRNVPHGLFRVRUJ
562 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-77  ■  Juxtacortical chondrosarcoma: radiographic, gross, and microscopic features. A, Lateral radiograph showing bone
surface lesion on the posterior aspect of femoral metaphysis. Note cortical erosions and its thickening beneath the lesion. B, Bisected
resection specimen showing cartilaginous bone surface lesion. C, Closer view of specimen shown in A and B depicting cartilaginous
nature of the lesion and erosions of the underlying cortex. D, Low power photomicrograph showing well differentiated cartilaginous
tumor within increased cellularity and nuclear atypical consistent with grade 1 chondrosarcoma . (D, ×100) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 563

A B

C D

FIGURE 7-78  ■  Juxtacortical chondrosarcoma: radiographic and gross features. A, Anteroposterior radiograph showing a calcified
bone surface lesion involving the proximal humerus. B, Bisected resection specimen showing cartilaginous bone surface mass.
C, Closer view of specimen shown in B depicting globular cartilaginous structure of the tumor with chalky calcifications. Note infil-
tration of the underlying cortex. D, Gross photograph of the bisected regional lymph node with metastatic chondrosarcoma. Inset,
Whole-mount specimen showing a focus of metastatic chondrosarcoma almost completely replacing the involved lymph node.

ERRNVPHGLFRVRUJ
564 7  Malignant Cartilage Tumors

A B

C D
FIGURE 7-79  ■  Juxtacortical chondrosarcoma: microscopic features. A, Base of the lesion showing infiltrative growth pattern into the
underlying cortex. Inset, Higher magnification showing hypercellularity (×100). B, Interface between the tumor and underlying cortex.
C, Deeper portion of cortex showing permeative growth pattern of the tumor. D, Medullary cavity beneath the juxtacortical chon-
drosarcoma showing aggressive tumor invasion. This is the same case as presented in Figure 7-78. Note that cytoarchitectual
features including nuclear atypia are not impressive and convincingly diagnostic of chondrosarcoma. In contrast, the infiltrative
growth pattern into the cortex and underlying medullary cavity discloses the aggressive nature of the lesion. In addition, the pres-
ence of axillary lymph node metastasis shown in Figure 7-78 is an ultimate proof for the aggressive nature of the lesion. (A-D, ×50;
inset, ×100) (A-D and inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 565

20. Cleton-Jansen AM, Timmerman MC, van de Vijver MJ, et al:


TABLE 7-1 Predisposing Conditions in A distinct phenotype characterizes tumors from a putative genetic
Secondary Chondrosarcoma trait involving chondrosarcoma and breast cancer occurring in the
same patient. Lab Invest 84:191–202, 2004.
Ollier’s disease and Maffucci’s syndrome 21. Crim JR, Seeger LL: Diagnosis of low-grade chondrosarcoma.
Multiple hereditary exostoses Radiology 189:503–504, 1993.
Solitary osteochondroma 22. Dahlin DC, Salvador AH: Chondrosarcomas of bone of the hands
Solitary enchondroma and feet: a study of 30 cases. Cancer 34:755–760, 1974.
Paget’s disease 23. Dang L, Jin S, Su SM: IDH mutations in glioma and acute myeloid
Radiation injury leukemia. Trends Mol Med 16:387–397, 2010.
24. Dekker PJ, Andrew AC: Laryngeal chondrosarcoma: an unusual
presentation. J Laryngol Otol 106:450–452, 1992.
of chondrosarcoma is shown in Table 7-1. A detailed 25. Demetrick DJ, Kneafsey PD, Hwang WS: Signet-ring chondro-
sarcoma: a new morphologic entity. Hum Pathol 22:1175–1179,
description of secondary chondrosarcoma is provided in 1991.
chapters describing those predisposing conditions. 26. Dobashi Y, Sugimura H, Sato A, et al: Possible association of p53
overexpression and mutation with high-grade chondrosarcoma.
REFERENCES Diagn Mol Pathol 2:257–263, 1993.
Intramedullary Chondrosarcoma 27. Dorfman HD, Czerniak B: Epidemiology of bone tumors:
SEER population-based data, 1973-1987. Cancer 75:223–227,
Conventional Chondrosarcoma 1995.
1. Alho A, Connor JF, Mankin HJ, et al: Assessment of malignancy 28. Eng J, Sabanathan S, Pradhan GN, et al: Primary bony chest wall
of cartilage tumors using flow cytometry: a preliminary report. tumours. J R Coll Surg Edinb 35:44–47, 1990.
J Bone Joint Surg 65A:779–785, 1983. 29. Eriksson AI, Schiller A, Mankin HJ: The management of chon-
2. Alho A, Skjeldal S, Melvik JE, et al: The clinical importance of drosarcoma of bone. Clin Orthop 153:44–66, 1980.
DNA synthesis and aneuploidy in bone and soft tissue tumours. 30. Erlandson RA, Huvos AG: Chondrosarcoma: a light and electron
Anticancer Res 13:2383–2387, 1993. microscopic study. Cancer 34:1642–1674, 1974.
3. Amary MF, Bacsi K, Maggiani F, et al: IDH1 and IDH2 mutations 31. Evans HL, Ayala AG, Romsdahl MM: Prognostic factors in chon-
are frequent events in central chondrosarcoma and central and drosarcoma of bone: a clinicopathologic analysis with emphasis on
periosteal chondromas but not in other mesenchymal tumours. histologic grading. Cancer 40:818–831, 1977.
J Pathol 224:334–343, 2011. 32. Exner GU, von Hochstetter AR, Augustiny N, et al: Magnetic
4. Amichetti M, Amelio D, Cianchetti M, et al: A systematic review resonance imaging in malignant bone tumours. Int Orthop 14:49–
of proton therapy in the treatment of chondrosarcoma of the skull 55, 1990.
base. Neurosurg Rev 33:155–165, 2010. 33. Feaux de Lacroix W, Dietlein M, Schmidt J, et al: Histological
5. Andronesi OC, Kim GS, Gerstner E, et al: Detection of investigation for comparison of cartilaginous tumors of unknown
2-hydroxyglutarate in IDH-mutated glioma patients by in vivo biological course with unequivocal chondrosarcomas. Zentralbl
spectral editing and 2D correlation magnetic resonance spectros- Pathol 138:339–343, 1992.
copy. Sci Transl Med 4:116, 2012. 34. Ferrandez L, Ramos L, Usabiaga J, et al: Low grade chon-
6. Barnes R, Catto M: Chondrosarcoma of bone. J Bone Joint Surg drosarcoma occurring in unusual sites. Int Orthop 16:392–397,
Br 48:729–764, 1966. 1992.
7. Batsakis JG: Osteogenic and chondrogenic sarcomas of the jaws. 35. Fletcher CDM, Bridge JA, Pancras CW, et al, editors: WHO clas-
Ann Otol Rhinol Laryngol 96:474–475, 1987. sification of tumors: soft tissue and bone, Lyon, 2013, International
8. Bjornsson J, McLeod RA, Unni KK, et al: Primary chondrosar- Agency for Research on Cancer.
coma of long bones and limb girdles. Cancer 83:2105–2119, 1998. 36. Geirnaerdt MJ, Hogendoorn PC, Bloem JL, et al: Cartilaginous
9. Bloch O, Sughrue ME, Mills SA, et al: Signaling pathways in tumors: fast contrast-enhanced MR imaging. Radiology 214:539–
cranial chondrosarcoma: potential molecular targets for directed 546, 2000.
chemotherapy. J Clin Neurosci 18:881–885, 2011. 37. Gitelis S, Bertoni F, Picci P, et al: Chondrosarcoma of bone: the
10. Bloch OG, Jian BJ, Yang I, et al: A systematic review of intracra- experience at the Istituto Ortopedico Rizzoli. J Bone Joint Surg
nial chondrosarcoma and survival. J Clin Neurosci 16:1547–1551, 63A:1248–1257, 1981.
2009. 38. Giuffrida AY, Burgueno JE, Koniaris LG, et al: Chondrosarcoma
11. Boeuf S, Kunz P, Hennig T, et al: A chondrogenic gene expression in the United States (1973 to 2003): an analysis of 2890 cases
signature in mesenchymal stem cells is a classifier of conventional from the SEER database. J Bone Joint Surg Am 91:1063–1072,
central chondrosarcoma. J Pathol 216:158–166, 2008. 2009.
12. Bohm G, Salzer-Kuntschik M, Lintner F: Morphometric analysis 39. Hachitanda Y, Tsuneyoshi M, Daimaru Y, et al: Extraskeletal
of cartilaginous tumors. Pathol Res Pract 188:570–575, 1992. myxoid chondrosarcoma in young children. Cancer 61:2521–2526,
13. Bourgouin PM, Tampieri D, Robitaille Y, et al: Low-grade 1988.
myxoid chondrosarcoma of the base of the skull: CT, MR, and 40. Hallor KH, Staaf J, Bovee JV, et al: Genomic profiling of chon-
histopathology. J Comput Assist Tomogr 16:268–273, 1992. drosarcoma: chromosomal patterns in central and peripheral
14. Bovee JV, Hogendoorn PC, Wunder JS, et al: Cartilage tumors tumors. Clin Cancer Res 15:2685–2694, 2009.
and bone development: molecular pathology and possible thera- 41. Hassounah M, Al-Mefty O, Akhtar M, et al: Primary cranial and
peutic targets. Nature Rev Cancer 10:481–488, 2010. intracranial chondrosarcoma. Acta Neurol 78:123–132, 1985.
15. Burt M, Fulton M, Wessner-Dunlap S, et al: Primary bony and 42. Henderson ED, Dahlin DC: Chondrosarcoma of bone: a study of
cartilaginous sarcomas of chest wall: results of therapy. Ann Thor 288 cases. J Bone Joint Surg 45A:1450–1458, 1963.
Surg 54:226–232, 1992. 43. Hickey M, Farrokhyar F, Deheshi B, et al: A systematic review
16. Buse S, Behnisch W, Kulozik A, et al: Primary chondrosarcoma and meta-analysis of intralesional versus wide resection for intra-
of the kidney: case report and review of the literature. Urol Int medullary grade I chondrosarcoma of the extremities. Ann Surg
83:116–118, 2009. Oncol 18:1705–1709, 2011.
17. Campanacci M, Guernelli N, Leonessa C, et al: Chondrosarcoma: 44. Hong P, Taylor SM, Trites JR, et al: Chondrosarcoma of the head
a study of 133 cases, 80 with long-term follow-up. Ital J Orthop and neck: report of 11 cases and literature review. J Otolaryngol
Traumatol 1:387–414, 1975. Head Neck Surg 38:279–285, 2009.
18. Casadei R, Ricci M, Ruggieri P, et al: Chondrosarcoma of the soft 45. Hudson TM, Manaster BJ, Springfield DS, et al: Radiology of
tissues: two different subgroups. J Bone Joint Surg Br 73:162–168, medullary chondrosarcoma: preoperative treatment planning. Skel
1991. Radiol 10:69–78, 1983.
19. Clark JC, Dass CR, Choong PF: Development of chondrosar- 46. Huvos AG, Marcove RC: Chondrosarcoma in the young: a clini-
coma animal models for assessment of adjuvant therapy. ANZ J copathologic analysis of 79 patients younger than 21 years of age.
Surg 79:327–336, 2009. Am J Surg Pathol 11:930–942, 1987.

ERRNVPHGLFRVRUJ
566 7  Malignant Cartilage Tumors

47. Insabato L: Chondrosarcoma with intranuclear cytoplasmic vacu- invited by the Euro-CNS Research Committee. Clin Neuropathol
oles. Hum Pathol 23:1088, 1992. (letter; comment). 30:217–230, 2011.
48. Ishida T, Dorfman HD: Chondroid chordoma versus low-grade 72. Pritchard DJ, Lunke RJ, Taylor WF, et al: Chondrosarcoma:
chondrosarcoma of the base of the skull: can immunohistochem- a clinicopathologic and statistical analysis. Cancer 45:149–157,
istry resolve the controversy? J Neuro Oncol 18:199–206, 1994. 1980.
49. Ishida T, Kikuchi F, Machinami R: Histological grading and mor- 73. Quiriny M, Gebhart M: Chondrosarcoma of the spine: a report
phometric analysis of cartilaginous tumours. Virchows Arch [A] of three cases and literature review. Acta Orthop Belg 74:885–890,
418:149–155, 1991. 2008.
50. Jamil N, Howie S, Salter DM: Therapeutic molecular targets in 74. Ravandi F, Patel K, Kuthra R, et al: Prognostic significance of
human chondrosarcoma. Int J Exper Pathol 91:387–393, 2010. alterations in IDH enzyme isoforms in patients with AML treated
51. Juratli TA, Kirsch M, Robel K, et al: IDH mutations as an early with high-dose cytarabine and idarubicin. Cancer 118:2665–2673,
and consistent marker in low-grade astrocytomas WHO grade II 2012.
and their consecutive secondary high-grade gliomas. J Neuro- 75. Rizzo S, Strinati F, Longari F, et al: Chondrosarcoma of the
Oncol 108:403–410, 2012. larynx: presentation of a case and review of the literature. Tumori
52. Kalinina J, Carroll A, Wang L, et al: Detection of “oncometabo- 94:864–868, 2008.
lite” 2-hydroxyglutarate by magnetic resonance analysis as a 76. Rosenthal DI, Schiller AL, Mankin HJ: Chondrosarcoma: cor-
biomarker of IDH1/2 mutations in glioma. J Mol Med (Berl) relation of radiological and histological grade. Radiology 150:21–
90:1161–1171, 2012. 26, 1984.
53. Kalinski T, Ropke A, Sel S, et al: Down-regulation of ephrin-A5, 77. Rozeman LB, Szuhai K, Schrage YM, et al: Array-comparative
a gene product of normal cartilage, in chondrosarcoma. Hum genomic hybridization of central chondrosarcoma: identification
Pathol 40:1679–1685, 2009. of ribosomal protein S6 and cyclin-dependent kinase 4 as a can-
54. Kreicbergs A, Boquist L, Borssén B, et al: Prognostic factors in didate target genes for genomic aberrations. Cancer 107:380–388,
chondrosarcoma: a comparative study of cellular DNA content 2006.
and clinicopathologic features. Cancer 50:577–583, 1982. 78. Ruark DS, Schlehaider UK, Shah JP: Chondrosarcomas of the
55. Kristensen IB, Sunde LM, Jensen OM: Chondrosarcoma: increas- head and neck. World J Surg 16:1010–1016, 1992.
ing grade of malignancy in local recurrence. Acta Pathol Microbiol 79. Saki N, Akhlagh SN, Mostofi NE, et al: Chondrosarcoma of the
Immunol Scand [A] 94:73–77, 1986. hyoid bone: imaging, surgical, and histopathologic correlation.
56. Lloyd GA, Phelps PD, Michaels L: The imaging characteristics Laryngoscope 118:1211–1213, 2008.
of naso-sinus chondrosarcoma. Clin Radiol 46:189–192, 1992. 80. Saleh G, Evans HL, Ro JY, et al: Extraskeletal myxoid chondro-
57. Mandahl N, Heim S, Arheden K, et al: Chromosomal rearrange- sarcoma: a clinicopathologic study of 10 patients with long-term
ments in chondromatous tumors. Cancer 65:242–248, 1990. follow-up. Cancer 70:2827–2830, 1992.
58. Mankin HJ, Cantley KP, Lippiello L, et al: The biology of human 81. Sanerkin NG: The diagnosis and grading of chondrosarcoma of
chondrosarcoma. I. Description of the cases, grading, and bio- bone: a combined cytologic and histologic approach. Cancer
chemical analyzes. J Bone Joint Surg 62A:160–176, 1980. 45:582–594, 1980.
59. Mankin HJ, Cantley KP, Schiller AL, et al: The biology of human 82. Sanerkin NG, Gallagher P: A review of the behavior of chondro-
chondrosarcoma. II. Variation in chemical composition among sarcoma of bone. J Bone Joint Surg Br 61:395–400, 1979.
types and subtypes of benign and malignant cartilage tumors. 83. Schiller AL: Diagnosis of borderline cartilage lesions of bone.
J Bone Joint Surg 62A:176–188, 1980. Semin Diagn Pathol 2:42–62, 1985.
60. Mankin HJ, Connor JF, Schiller AL, et al: Grading of bone 84. Schrage YM, Lam S, Jochemsen AG, et al: Central chondrosar-
tumors by analysis of nuclear DNA content using flow cytometry. coma progression is associated with pRb pathway alterations:
J Bone Joint Surg 67A:404–413, 1985. CDK4 down-regulation and p16 overexpression inhibit cell
61. Marcove RC, Huvos AG: Cartilaginous tumors of the ribs. Cancer growth in vitro. J Cell Mol Med 13:2843–2852, 2009.
27:794–801, 1971. 85. Schwab JH, Boland PJ, Agaram NP, et al: Chordoma and chon-
62. Marcove RC, Miké V, Hutter RV, et al: Chondrosarcoma of the drosarcoma gene profile: implications for immunotherapy. Cancer
pelvis and upper end of the femur: an analysis of factors influenc- Immunol Immunother 58:339–349, 2009.
ing survival time in one hundred and thirteen cases. J Bone Joint 86. Sen CN, Sekhar LN, Schramm VL, et al: Chordoma and chon-
Surg 54A:561–572, 1972. drosarcoma of the cranial base: an 8-year experience. Neurosurgery
63. Mark RJ, Tran LM, Sercarz J, et al: Chondrosarcoma of the head 931–941, 1989.
and neck: the UCLA experience, 1955-88. Am J Clin Oncol 87. Shives TC, McLeod RA, Unni KK, et al: Chondrosarcoma of the
16:232–237, 1993. spine. J Bone Joint Surg 71A:1158–1165, 1989.
64. Meachim G: Histological grading of chondrosarcomata. J Bone 88. SongTao Q, Lei Y, Si G, et al: IDH mutations predict longer
Joint Surg Br 61:393–394, 1979. survival and response to temozolomide in secondary glioblastoma.
65. Metellus P, Coulibaly B, Colin C, et al: Absence of IDH mutation Cancer Sci 103:269–273, 2012.
identifies a novel radiologic and molecular subtype of WHO 89. Stapleton SR, Wilkins PR, Archer DJ, et al: Chondrosarcoma of
grade II gliomas with dismal prognosis. Acta Neuropathol 120:719– the skull base: a series of eight cases. Neurosurgery 32:348–356,
729, 2010. 1993.
66. Namizato CS, Muriel-Cueto P, Baez-Perez JM, et al: Chondro- 90. Strike SA, McCarthy EF: Chondrosarcoma of the spine: a series
sarcoma of the uterus: case report and literature review. Arch Gyn of 16 cases and a review of the literature. Iowa Orthop J 31:154–
Obstet 278:369–372, 2008. 159, 2011.
67. Nikoghosyan AV, Rauch G, Munter MW, et al: Randomised trial 91. Tiet TD, Hopyan S, Nadesan P, et al: Constitutive hedgehog
of proton vs. carbon ion radiation therapy in patients with low and signaling in chondrosarcoma up-regulates tumor cell prolifera-
intermediate grade chondrosarcoma of the skull base, clinical tion. Am J Pathol 168:321–330, 2006.
phase III study. BMC Cancer 10:606, 2010. 92. Tos P, Artiaco S, Linari A, et al: Chondrosarcoma in the distal
68. Odink AE, van Asperen CJ, Vandenbroucke JP, et al: An phalanx of index finger: clinical report and literature review. Chir
association between cartilaginous tumours and breast cancer de la Main 28:265–269, 2009.
in the national pathology registration in The Netherlands 93. Unni KK, Inwards CY: Dahlin’s bone tumors: General aspects and
points towards a possible genetic trait. J Pathol 193:191–192, data on 10,165 cases, ed 6, Philadelphia, 2009, Lippincott Williams
2001. & Wilkins.
69. Ozisik YY, Meloni AM, Peier A, et al: Cytogenetic findings in 19 94. Ward PS, Cross JR, Lu C, et al: Identification of additional IDH
malignant bone tumors. Cancer 74:2268–2275, 1994. mutations associated with oncometabolite (R)-2-hydroxyglutarate
70. Papachristou DJ, Goodman MA, Cieply K, et al: Comparison of production. Oncogene 31:2491–2498, 2012.
allelic losses in chondroblastoma and primary chondrosarcoma of 95. Weiss A-PC, Dorfman HD: S-100 protein in human cartilage
bone and correlation with fluorescence in situ hybridization analy- lesions. J Bone Joint Surg 68A:521–526, 1986.
sis. Hum Pathol 37:890–898, 2006. 96. Welkerling H, Dreyer T, Delling G: Morphological typing of
71. Preusser M, Capper D, Hartmann C, et al: IDH testing in diag- chondrosarcoma: a study of 94 cases. Virchows Arch [A] 418:419–
nostic neuropathology: review and practical guideline article 425, 1991.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 567

97. Yen KE, Bittinger MA, Su SM, et al: Cancer-associated IDH histochemical study of six cases. Arch Anat Cytol Pathol 36:218–225,
mutations: biomarker and therapeutic opportunities. Oncogene 1988.
29:6409–6417, 2010. 121. Lamovec J, Zidar A, Bracko M, et al: Primary bone sarcoma with
98. Young CL, Sim FH, Unni KK, et al: Chondrosarcoma of bone in rhabdomyosarcomatous component. Pathol Res Pract 190:51–60,
children. Cancer 66:1641–1648, 1990. 1994.
122. Mandahl N, Gustafson P, Mertens F, et al: Cytogenetic aberra-
Dedifferentiated Chondrosarcoma tions and their prognostic impact in chondrosarcoma. Genes Chro-
99. Abenoza P, Neumann P, Manivel JC, et al: Dedifferentiated mosomes Cancer 33:188–200, 2002.
chondrosarcoma: an ultrastructural study of two cases, with 123. Mavrogenis AF, Ruggieri P, Mercuri M, et al: Dedifferenti-
immunocytochemical correlations. Ultrastruct Pathol 10:529–538, ated chondrosarcoma revisited. J Surg Orthop Adv 20:106–111,
1986. 2011.
100. Akahane T, Shimizu T, Isobe K, et al: Dedifferentiated chondro- 124. McCarthy EF, Dorfman HD: Chondrosarcoma of bone with
sarcoma arising in a solitary osteochondroma with leiomyosarco- dedifferentiation: a study of 18 cases. Hum Pathol 13:36–40,
matous component: a case report. Arch Orthop Trauma Surg 1982.
128:951–953, 2008. 125. Meijer D, de Jong D, Pansuriya TC, et al: Genetic characteriza-
101. Bertoni F, Present D, Bacchini P, et al: Dedifferentiated periph- tion of mesenchymal, clear cell, and dedifferentiated chondrosar-
eral chondrosarcomas: a report of seven cases. Cancer 63:2054– coma. Gene Chromosome Cancer 51:899–909, 2012.
2059, 1989. 126. Mirra JM, Marcove RC: Fibrosarcomatous dedifferentiation of
102. Bovee JV, Cleton-Jansen AM, Rosenberg C, et al: Molecular primary and secondary chondrosarcoma. J Bone Joint Surg
genetic characterization of both components of a dedifferentiated 56A:285–296, 1974.
chondrosarcoma, with implications for its histogenesis. J Pathol 127. Mitchell AD, Ayoub K, Mangham DC, et al: Experience in the
189:454–462, 1999. treatment of dedifferentiated chondrosarcoma. J Bone Joint Surg
103. Bovee JV, Sciot R, Dal Cin P, et al: Chromosome 9 alterations and 82:55–61, 2000.
trisomy 22 in central chondrosarcoma: a cytogenetic and DNA 128. Munk PL, Connell DG, Quenville NF: Dedifferentiated chon-
flow cytometric analysis of chondrosarcoma subtypes. Diagn Mol drosarcoma of bone with leiomyosarcomatous mesenchymal com-
Pathol 10:228–235, 2001. ponent: a case report. Can Assoc Radiol J 39:218–220, 1988.
104. Brandwein M, Moore S, Som P, et al: Laryngeal chondrosarco- 129. Niezabitowski A, Edel G, Roessner A, et al: Rhabdomyosarcoma-
mas: a clinicopathologic study of 11 cases, including two tous component in dedifferentiated chondrosarcoma. Pathol Res
“dedifferentiated” chondrosarcomas. Laryngoscope 102:858–867, Pract 182:275–282, 1987.
1992. 130. Okada K, Hasegawa T, Tateishi U, et al: Dedifferentiated chon-
105. Bridge JA, DeBoer J, Travis J, et al: Simultaneous interphase cyto- drosarcoma with telangiectatic osteosarcoma-like features. J Clin
genetic analysis and fluorescence immunophenotyping of dedif- Pathol 59:1200–1202, 2006.
ferentiated chondrosarcoma: implications for histopathogenesis. 131. O’Malley DP, Opheim KE, Barry TS, et al: Chromosomal
Am J Pathol 144:215–220, 1994. changes in a dedifferentiated chondrosarcoma: a case report and
106. Capanna R, Bertoni F, Bettelli G, et al: Dedifferentiated chondro- review of the literature. Cancer Genet Cytogenet 124:105–111,
sarcoma. J Bone Joint Surg 70A:60–69, 1988. 2001.
107. Casorzo L, Chiecchio L, Pisacane A, et al: Cytogenetic findings 132. Radhi JM, Loewy J: Dedifferentiated chondrosarcoma with fea-
in a case of dedifferentiated chondrosarcoma. Cancer Genet Cyto- tures of telangiectatic osteosarcoma. Pathology 31:428–430, 1999.
genet 144:61–64, 2003. 133. Reith JD, Bauer TW, Fischler DF, et al: Dedifferentiated chon-
108. Coughlan B, Feliz A, Ishida T, et al: p53 expression in cartilage drosarcoma with rhabdomyosarcomatous differentiation. Am J
lesions. Human Pathol 26:620–624, 1995. Surg Pathol 20:293–298, 1996.
109. Dahlin DC, Beabout JW: Differentiation of low-grade chondro- 134. Ropke M, Boltze C, Neumann HW, et al: Genetic and epigenetic
sarcomas. Cancer 28:461–466, 1971. alterations in tumor progression in a dedifferentiated chondrosar-
110. Daly PJ, Sim FH, Wold LE: Dedifferentiated chondrosarcoma of coma. Pathol Res Pract 199:437–444, 2003.
bone (clinical conference). Orthopedics 12:763–767, 1989. 135. Rozeman LB, de Bruijn IH, Bacchini P, et al: Dedifferentiated
111. de Lange EE, Pope TL, Jr, Fechner RE: Dedifferentiated peripheral chondrosarcomas: regulation of EXT-downstream
chondrosarcoma: radiographic features. Radiology 161:489–492, molecules and differentiation-related genes. Mod Pathol 22:1489–
1986. 1498, 2009.
112. Dervan PA, O’Loughlin J, Hurson BJ: Dedifferentiated chondro- 136. Saifuddin A, Mann BS, Mahroof S, et al: Dedifferentiated chon-
sarcoma with muscle and cytokeratin differentiation in the ana- drosarcoma: use of MRI to guide needle biopsy. Clin Radiol
plastic component. Histopathology 12:517–526, 1988. 59:268–272, 2004.
113. Estrada EG, Ayala AG, Lewis V, et al: Dedifferentiated chondro- 137. Sakamoto A, Oda Y, Adachi T, et al: H-ras oncogene mutation in
sarcoma with a noncartilaginous component mimicking a conven- dedifferentiated chondrosarcoma: polymerase chain reaction-
tional giant cell tumor of bone. Ann Diagn Pathol 6:159–163, restriction fragment length polymorphism analysis. Mod Pathol
2002. 14:343–349, 2001.
114. Frassica FJ, Unni KK, Beabout JW, et al: Dedifferentiated chon- 138. Sakamoto A, Oda Y, Iwamoto Y, et al: Expression of membrane
drosarcoma: a report of the clinicopathological features and treat- type 1 matrix metalloproteinase, matrix metalloproteinase 2 and
ment of 78 cases. J Bone Joint Surg 68A:1197–1205, 1986. tissue inhibitor of metalloproteinase 2 in human cartilaginous
115. Grimer RJ, Gosheger G, Taminiau A, et al: Dedifferentiated tumors with special emphasis on mesenchymal and dedifferenti-
chondrosarcoma: prognostic factors and outcome from a Euro- ated chondrosarcoma. J Cancer Res Clin Oncol 125:541–548,
pean group. Eur J Cancer 43:2060–2065, 2007. 1999.
116. Grote HJ, Schneider-Stock R, Neumann W, et al: Mutation of 139. Sanerkin NG, Woods CG: Fibrosarcomata and malignant fibrous
p53 with loss of heterozygosity in the osteosarcomatous compo- histiocytomata arising in relation to enchondromata. J Bone Joint
nent of a dedifferentiated chondrosarcoma. Virchows Archiv 436: Surg 61B:366–372, 1979.
494–497, 2000. 140. Simms W, Ordóñez N, Johnston D, et al: p53 expression in dedif-
117. Hackel C, Czerniak B, Ayala AG, et al: Expression of plasminogen ferentiated chondrosarcoma. Cancer 76:223–227, 1995.
activators and plasminogen activator inhibitor 1 in dedifferenti- 141. Swarts SJ, Neff JR, Johansson SL, et al: Cytogenetic analysis of
ated chondrosarcoma. Cancer 79:53–58, 1997. dedifferentiated chondrosarcoma. Cancer Genet Cytogenet 89:49–
118. Jaworski RC: Dedifferentiated chondrosarcoma: an ultrastructural 51, 1996.
study. Cancer 53:2674–2678, 1984. 142. Tarkkanen M, Wiklund T, Virolainen M, et al: Dedifferentiated
119. Johnson S, Tetu B, Ayala AG, et al: Chondrosarcoma with addi- chondrosarcoma with t(9;22) (q34;q11-12). Genes Chromosomes
tional mesenchymal component (dedifferentiated chondrosar- Cancer 9:136–140, 1994.
coma). I. A clinicopathologic study of 26 cases. Cancer 58:278–286, 143. Têtu B, Ordóñez NG, Ayala AG, et al: Chondrosarcoma with
1986. additional mesenchymal component (dedifferentiated chondro-
120. Karabela-Bouropoulou V, Markaki S, Kypparidou L, et al: Dedif- sarcoma). II. An immunohistochemical and electron microscopic
ferentiated chondrosarcoma: a clinicopathological and immuno- study. Cancer 58:287–298, 1986.

ERRNVPHGLFRVRUJ
568 7  Malignant Cartilage Tumors

144. Wick MR, Siegal GP, Mills SE, et al: Dedifferentiated chon­ 170. Ryu KM, Myung NH, Seo PW: Clear cell chondrosarcoma
drosarcoma of bone: an immunohistochemical and lectin- arising from the sternum: a rare tumor in an uncommon location.
histochemical study. Virchows Arch [A] 411:23–32, 1987. Ann Thorac Surg 93:1725–1727, 2012.
145. Zalupski MM, Ensley JF, Ryan J, et al: A common cytogenetic 171. Singh S, Ng KC, Tan SK: Clear cell chondrosarcoma: a report of
abnormality and DNA content alterations in dedifferentiated 2 cases. Singapore Med J 32:154–157, 1991.
chondrosarcoma. Cancer 66:1176–1182, 1990. 172. Sreekantaiah C, Leong SP, Davis JR, et al: Cytogenetic and flow
cytometric analysis of a clear cell chondrosarcoma. Cancer Genet
Clear Cell Chondrosarcoma Cytogenet 52:193–199, 1991.
146. Aigner T, Dertinger S, Belke J, et al: Chondrocytic cell differen- 173. Taconis WK: Clear cell chondrosarcoma: a report of three cases
tiation in clear cell chondrosarcoma. Hum Pathol 27:1301–1305, and review of the literature. Diagn Imag Clin Med 55:219–227,
1996. 1986.
147. Angervall L, Kindblom LG: Clear-cell chondrosarcoma: a light- 174. Unni KK, Dahlin DC, Beabout JW, et al: Chondrosarcoma: clear-
and electron-microscopic and histochemical study of two cases. cell variant—a report of 16 cases. J Bone Joint Surg 58A:676–683,
Virchows Arch 389A:27–41, 1980. 1976.
148. Bagley L, Kneeland JB, Dalinka MK, et al: Unusual behavior of 175. Wang LT, Liu TC: Clear cell chondrosarcoma of bone: a report
clear cell chondrosarcoma. Skeletal Radiol 22:279–282, 1993. of three cases with immunohistochemical and affinity histochemi-
149. Bjornsson J, Unni KK, Dahlin DC, et al: Clear cell chondrosar- cal observations. Pathol Res Pract 189:411–415, 1993.
coma of bone: observations in 47 cases. Am J Surg Pathol 8:223– 176. Weiss A-PC, Dorfman HD: Clear-cell chondrosarcoma: a report
230, 1984. of 10 cases and review of the literature. Surg Pathol 1:123–129,
150. Bosse A, Ueda Y, Wuisman P, et al: Histogenesis of clear cell 1988.
chondrosarcoma: an immunohistochemical study with osteonec- 177. Yamaguchi H, Isu K, Ubayama Y, et al: Clear cell chondrosar-
tin, a non-collagenous structure protein. J Cancer Res Clin Oncol coma: a report of two cases and review of the literature. Acta Pathol
117:43–49, 1991. Jpn 36:1577–1585, 1986.
151. Collins MS, Koyama T, Swee RG, et al: Clear cell chondrosar-
coma: radiographic, computed tomographic, and magnetic reso- Mesenchymal Chondrosarcoma
nance findings in 34 patients with pathologic correlation. Skeletal 178. Ashworth TG: Pulmonary blastoma: a true congenital neoplasm.
Radiol 32:687–694, 2003. Histopathology 7:585–594, 1983.
152. Corradi D, Bacchini P, Campanini N, et al: Aggressive clear cell 179. Bertoni F, Picci P, Bacchini P: Mesenchymal chondrosarcoma of
chondrosarcomas: do distinctive characteristics exist?: a report of bone and soft tissues. Cancer 52:533–541, 1983.
4 cases. Arch Pathol Lab Med 130:1673–1679, 2006. 180. Boldrini R, Devito R, Diomedi-Camassei F, et al: Pulmonary blas-
153. Ding J, Hashimoto H, Tsuneyoshi M, et al: Clear cell chondro- tomas of childhood: histologic, immunohistochemical, ultrastruc-
sarcoma: a case report with topographic analysis. Acta Pathol Jpn tural aspects and therapeutic considerations. Ultrastruct Pathol
39:533–538, 1989. 29:493–501, 2005.
154. Donati D, Yin JQ, Colangeli M, et al: Clear cell chondrosarcoma 181. Brown RE: Morphoproteomic portrait of the mTOR pathway in
of bone: long time follow-up of 18 cases. Arch Orthop Trauma Surg mesenchymal chondrosarcoma. Ann Clin Lab Sci 34:397–399,
128:137–142, 2008. 2004.
155. Faraggiana T, Sender B, Glicksman P: Light- and electron- 182. Brown RE, Boyle JL: Mesenchymal chondrosarcoma: molecular
microscopic study of clear cell chondrosarcoma. Am J Clin Pathol characterization by a proteomic approach, with morphogenic
75:117–121, 1981. and therapeutic implications. Ann Clin Lab Sci 33:131–141,
156. Hsu W, McCarthy E, Gokaslan ZL, et al: Clear-cell chondrosar- 2003.
coma of the lumbar spine: case report and review of the literature. 183. Cajaiba MM, Jianhua L, Goodman MA, et al: Sox9 expression is
Neurosurgery 68:1160–1164, 2011. not limited to chondroid neoplasms: variable occurrence in other
157. Itala A, Leerapun T, Inwards C, et al: An institutional review of soft tissue and bone tumors with frequent expression by synovial
clear cell chondrosarcoma. Clin Orthop Rel Res 440:209–212, 2005. sarcomas. Int J Surg Pathol 18:319–323, 2010.
158. Kalil RK, Inwards CY, Unni KK, et al: Dedifferentiated clear cell 184. Cesari M, Bertoni F, Bacchini P, et al: Mesenchymal chondrosar-
chondrosarcoma. Am J Surg Pathol 24:1079–1086, 2000. coma. An analysis of patients treated at single institution. Tumori
159. Kumar R, David R, Cierney G, III: Clear cell chondrosarcoma. 93:423–427, 2007.
Radiology 154:45–48, 1985. 185. Damato S, Alorjiani M, Bonar F, et al: IDH1 mutations are not
160. Kunisada T, Moseley JM, Slavin JL, et al: Co-expression of para- found in cartilaginous tumours other than central and periosteal
thyroid hormone-related protein (PTHrP) and PTH/PTHrP chondrosarcomas and enchondromas. Histopathology 60:363–365,
receptor in cartilaginous tumours: a marker for malignancy? 2012.
Pathology 34:133–137, 2002. 186. Fanburg-Smith JC, Auerbach A, Marwaha JS, et al: Reappraisal
161. Kuroda N, Sogoh T, Ohara M, et al: Clear cell chondrosarcoma: of mesenchymal chondrosarcoma: novel morphologic observa-
an ultrastructural study. Med Mol Morphol 42:185–188, 2009. tions of the hyaline cartilage and endochondral ossification and
162. Le Charpentier Y, Forest M, Postel M, et al: Clear-cell chondro- beta-catenin, Sox9, and osteocalcin immunostaining of 22 cases.
sarcoma: a report of five cases including ultrastructural study. Hum Pathol 41:653–662, 2010.
Cancer 44:622–629, 1979. 187. Fanburg-Smith JC, Auerbach A, Marwaha JS, et al: Immunopro-
163. Leggon RE, Jr, Unni KK, Beabout JW, et al: Clear-cell chondro- file of mesenchymal chondrosarcoma: aberrant desmin and EMA
sarcoma. Orthopaedics 13:593–596, 1990. expression, retention of INI1, and negative estrogen receptor in
164. Matsuura S, Ishii T, Endo M, et al: Epithelial and cartilaginous 22 female-predominant central nervous system and musculoskel-
differentiation in clear cell chondrosarcoma. Hum Pathol 44:237– etal cases. Ann Diagn Pathol 14:8–14, 2010.
243, 2013. 188. Fosdal MB: Pleuropulmonary blastoma. J Pediatr Oncol Nurs
165. Mokhtari S, Mirafsharieh A: Clear cell chondrosarcoma of the 25:295–302, 2008.
head and neck. Head Neck Oncol 4:13, 2012. 189. Gatter KM, Olson S, Lawce H, et al: Trisomy 8 as the sole cyto-
166. Nishio J, Reith JD, Ogose A, et al: Cytogenetic findings in genetic abnormality in a case of extraskeletal mesenchymal chon-
clear cell chondrosarcoma. Cancer Genet Cytogenet 162:74–77, drosarcoma. Cancer Genet Cytogenet 159:151–154, 2005.
2005. 190. Gengler C, Letovanec I, Taminelli L, et al: Desmin and myogenin
167. Ohno T, Park P, Oguro K, et al: Ultrastructural study of a clear reactivity in mesenchymal chondrosarcoma: a potential diagnostic
cell chondrosarcoma. Ultrastruct Pathol 10:321–330, 1986. pitfall. Histopathology 48:201–203, 2006.
168. Park YK, Park HR, Chi SG, et al: Overexpression of p53 and 191. Goldman RL: “Mesenchymal” chondrosarcoma: a rare malignant
absent genetic mutation in clear cell chondrosarcoma. Int J Oncol chondroid tumor usually primary in bone. Cancer 20:1494–1498,
19:353–357, 2001. 1967.
169. Present D, Bacchini P, Pignatti G, et al: Clear cell chondrosar- 192. Harwood AR, Krajbich JI, Fornasier VL: Mesenchymal
coma of bone: a report of eight cases. Skel Radiol 20:187–191, chondrosarcoma: a report of 17 cases. Clin Orthop 158:144–148,
1991. 1981.

ERRNVPHGLFRVRUJ
7  Malignant Cartilage Tumors 569

193. Hashimoto N, Ueda T, Joyama S, et al: Extraskeletal mesenchy- 215. Schultz KA, Pacheco MC, Yang J, et al: Ovarian sex cord-stromal
mal chondrosarcoma: an imaging review of ten new patients. Skel tumors, pleuropulmonary blastoma and DICER1 mutations:
Radiol 34:785–792, 2005. a report from the International Pleuropulmonary Blastoma Reg-
194. Hoang MP, Suarez PA, Donner LRY, et al: Mesenchymal chon- istry. Gynecol Oncol 122:246–250, 2011.
drosarcoma: a small cell neoplasm with polyphenotypic differen- 216. Shakked RJ, Geller DS, Gorlick R, et al: Mesenchymal chondro-
tiation. Int J Surg Pathol 8:291–301, 2000. sarcoma: clinicopathologic study of 20 cases. Arch Pathol Lab Med
195. Huvos AG, Rosen G, Dabska M, et al: Mesenchymal chondrosar- 136:61–75, 2012.
coma: a clinicopathologic analysis of 35 patients with emphasis on 217. Steiner GC, Mirra JM, Bullough PG: Mesenchymal chondrosar-
treatment. Cancer 51:1230–1237, 1983. coma: a study of the ultrastructure. Cancer 32:926–939, 1973.
196. Jambhekar NA, Desai SS, Aggarwal MG, et al: Mesenchymal 218. Swanson PE, Lillemoe TJ, Manivel JC, et al: Mesenchymal chon-
chondrosarcoma: a series of 23 cases. Ind J Pathol Microbiol drosarcoma: an immunohistochemical study. Arch Pathol Lab Med
47:491–493, 2004. 114:943–948, 1990.
197. Kaneko T, Suzuki Y, Takata R, et al: Extraskeletal mesenchymal 219. Takahashi K, Sato K, Kanazawa H, et al: Mesenchymal chondro-
chondrosarcoma of the kidney. Int J Urol 13:285–286, 2006. sarcoma of the jaw: report of a case and review of 41 cases in the
198. Knott PD, Gannon FH, Thompson LD: Mesenchymal chondro- literature. Head Neck 15:459–464, 1993.
sarcoma of the sinonasal tract: a clinicopathological study of 13 220. Taori K, Patil P, Attarde V, et al: Primary retroperitoneal extraskel-
cases with a review of the literature. Laryngoscope 113:783–790, etal mesenchymal chondrosarcoma: a computed tomography
2003. diagnosis. Br J Radiol 80:268–270, 2007.
199. Lee AF, Hayes MM, Lebrun D, et al: FLI-1 distinguishes Ewing 221. Tuncer S, Kebudi R, Peksayar G, et al: Congenital mesenchymal
sarcoma from small cell osteosarcoma and mesenchymal chondro- chondrosarcoma of the orbit: case report and review of the litera-
sarcoma. Applied Immunohistochem Mol Morphol 19:233–238, 2011. ture. Ophthalmology 111:1016–1022, 2004.
200. Lichtenstein L, Bernstein D: Unusual benign and malignant 222. Valderrama E, Saluja G, Shende A, et al: Pulmonary blastoma:
chondroid tumors of bone. Cancer 12:1142–1157, 1959. report of two cases in children. Am J Surg Pathol 2:415–422, 1978.
201. Manivel JC, Priest JR, Watterson J, et al: Pleuropulmonary blas- 223. Venkatramani R, Malogolowkin MH, Wang L, et al: Pleuro-
toma: the so-called pulmonary blastoma of childhood. Cancer pulmonary blastoma: a single-institution experience. J Pediatr
62:1516–1526, 1988. Hematol Oncol 34:e182–e185, 2012.
202. Martinez-Tello FJ, Navas-Palacios JJ: Ultrastructural study of 224. Wang L, Motoi T, Khanin R, et al: Identification of a novel,
conventional chondrosarcomas and myxoid- and mesenchymal- recurrent HEY1-NCOA2 fusion in mesenchymal chondrosarcoma
chondrosarcomas. Virchows Arch 396A:197–211, 1982. based on a genome-wide screen of exon-level expression data.
203. Morrison C, Radmacher M, Mohammed N, et al: MYC amplifi- Gene Chromosome Cancer 51:127–139, 2012.
cation and polysomy 8 in chondrosarcoma: array comparative 225. Wehrli BM, Huang W, DeCrombrugghe B, et al: Sox9, a master
genomic hybridization, fluorescent in situ hybridization, and asso- regulator of chondrogenesis, distinguishes mesenchymal chon-
ciation with outcome. J Clin Oncol 23:9369–9376, 2005. drosarcoma from other small blue round cell tumors. Hum Pathol
204. Nakamura Y, Becker LE, Marks A: S-100 protein in tumors of 34:263–269, 2003.
cartilage and bone: an immunohistochemical study. Cancer
52:1820–1824, 1983. Juxtacortical Chondrosarcoma
205. Nakashima Y, Unni KK, Shives TC, et al: Mesenchymal chon- 226. Bertoni F, Boriani S, Laus M, et al: Periosteal chondrosarcoma
drosarcoma of bone and soft tissue: a review of 111 cases. Cancer and periosteal osteosarcoma: two distinct entities. J Bone Joint
57:2444–2453, 1986. Surg Br 64:370–376, 1982.
206. Naumann S, Krallman PA, Unni KK, et al: Translocation 227. Chaabane S, Bouaziz MC, Drissi C, et al: Periosteal chondrosar-
der(13;21)(q10;q10) in skeletal and extraskeletal mesenchymal coma. AJR Am J Roentgenol 192:1–6, 2009.
chondrosarcoma. Mod Pathol 15:572–576, 2002. 228. Costa EM, Lucas BL, Silva MR, et al: Mandibular periosteal (jux-
207. Nussbeck W, Neureiter D, Soder S, et al: Mesenchymal chondro- tacortical) chondrosarcoma. Brazil Dent J 20:428–433, 2009.
sarcoma: an immunohistochemical study of 10 cases examining 229. Huvos AG: Malignant surface lesions of bone. Curr Diagn Pathol
prognostic significance of proliferative activity and cellular dif- 7:247–250, 2001.
ferentiation. Pathology 36:230–233, 2004. 230. Kumta SM, Griffith JF, Chow LT, et al: Primary juxtacortical
208. Nyquist KB, Panagopoulos I, Thorsen J, et al: Whole- chondrosarcoma dedifferentiating after 20 years. Skel Radiol
transciptome sequencing identifies novel IRF2BP2-CDX1 fusion 27:569–573, 1998.
gene brought about by translocation t(1;5)(q42;q32) in mesenchy- 231. Matsumoto K, Hukuda S, Ishizawa M, et al: Parosteal (juxtacorti-
mal chondrosarcoma. PLoS ONE 7:e49705, 2012. cal) chondrosarcoma of the humerus associated with regional
209. Oh BG, Han YH, Lee BH, et al: Primary extraskeletal mesenchy- lymph node metastasis. A case report. Clin Orthop Rel Res 290:168–
mal chondrosarcoma arising from the pancreas. Korean J Radiol 173, 1993.
8:541–544, 2007. 232. Mirra JM: Bone tumors: clinical, radiologic, and pathologic correlations,
210. Pellitteri PK, Ferlito A, Fagan JJ, et al: Mesenchymal chondro- Philadelphia, 1989, Lea & Febiger.
sarcoma of the head and neck. Oral Oncol 43:970–975, 2007. 233. Nojima T, Unni KK, McLeod RA, et al: Periosteal chondroma
211. Priest JR, McDermott MB, Bhatia S, et al: Pleuropulmonary blas- and periosteal chondrosarcoma. Am J Surg Pathol 9:666–677,
toma: a clinicopathologic study of 50 cases. Cancer 80:147–161, 1985.
1997. 234. Schajowicz F: Juxtacortical chondrosarcoma. J Bone Joint Surg
212. Ranjan A, Chacko G, Joseph T, et al: Intraspinal mesenchymal 59B:473–480, 1977.
chondrosarcoma: case report. J Neurosurg 80:928–930, 1994. 235. Van Damme PA, de Wilde PC, Koot RA, et al: Juxtacortical chon-
213. Salvador AH, Beabout JW, Dahlin DC: Mesenchymal chondro- drosarcoma of the mandible: report of a unique case and review
sarcoma: observation on 30 new cases. Cancer 28:605–615, of the literature. Int J Oral Maxillofacial Surg 34:94–98, 2005.
1971. 236. Vanel D, De Paolis M, Monti C, et al: Radiological features of 24
214. Salvati M, Caroli E, Frati A, et al: Central nervous system mes- periosteal chondrosarcomas. Skel Radiol 30:208–212, 2001.
enchymal chondrosarcoma. J Exper Clin Cancer Res 24:317–324, 237. Zenmyo M, Komiya S, Nakashima M, et al: Giant juxtacortical
2005. chondrosarcoma of the humerus. Orthopedics 23:497–498, 2000.

ERRNVPHGLFRVRUJ
C H A P T E R 8 

Fibrous Dysplasia and


Related Lesions
CHAPTER OUTLINE

FIBROUS DYSPLASIA FIBROCARTILAGINOUS DYSPLASIA


Fibrous Dysplasia in Different Anatomic Sites

FIBROUS DYSPLASIA of multiple endocrinopathies of the Albright-McCune


syndrome and fibrous dysplasia.2,11,15,35,43,48,50,55,84-86,101,103
This disorder was recognized as a clinical syndrome of However, not all cases of fibrous dysplasia or Albright-
disseminated skeletal deformities long before it was given McCune syndrome can be linked to mutations of G pro-
the name fibrous dysplasia by Lichtenstein and Jaffe in two teins, indicating that other molecular mechanisms may be
classic publications that appeared in 1938 and 1942.41,42 involved.31,40,49 G proteins form heterotrimeric complexes
In older literature, it had been discussed under the term and bind guanine nucleotides.13,107 They couple a large
osteitis fibrosa or generalized fibrocystic disease of bone, which number of cell surface receptors to their corresponding
also included such conditions as renal osteodystrophy and intracellular signal transduction pathways. Mutations of
hyperparathyroidism. The craniofacial lesions were clas- G proteins alter these pathways and may disturb the
sified as leontiasis ossea or cherubism. They were linked tissue differentiation process (Fig. 8-1).81 The mutations
to fibrous dysplasia as an underlying condition by Pugh65 in fibrous dysplasia involve the α chain of the heterotri-
in 1945. Fibrous dysplasia as a solitary lesion was last to meric G subunit and result in the substitution of histidine,
be linked to a disseminated polyostotic form of the cysteine, or serine for arginine at position 201.2,30 These
disease. Albright-McCune syndrome was independently substitutions are caused by single nucleotide mutations
described by two groups in 1937, and the skeletal abnor- involving predominantly exon 8 and less frequently exon
malities in this condition were originally classified as 9 of the gene encoding for Gsα (GNAS1) located on the
osteitis fibrosa.1,45 They were reclassified as polyostotic long arm of chromosome 20 at 20q13.2-3. These muta-
fibrous dysplasia in 1942 by Lichtenstein and Jaffe.42 In tions disrupt the inherent guanosine triphosphate activity
1967, Mazabraud et al.52 described an association of of the α chain, stimulate adenyl cyclase, and result in
fibrous dysplasia with soft tissue myxomas.94 independent cell proliferations.40,51 Identical mutations
Fibrous dysplasia is best explained if it is considered are also present in pituitary and other endocrine tumors
as a dysplastic anomaly of bone-forming mesenchymal associated with Albright-McCune syndrome and in the
tissue. The hallmark of this disease is a solitary focal or soft tissue myxomas associated with fibrous dysplasia such
generalized multifocal inability of bone-forming tissue to as Mazabraud’s syndrome.60 They can be also detected in
produce mature lamellar bone. It is arrested at the level solitary soft tissue myxomas unrelated to fibrous dyspla-
of woven bone. Even if a large amount of osteoid is pro- sia.60,100 The osteoblastic cells expressing mutant Gsα show
duced, it cannot mature to lamellar bone. The remark- increased proliferations and inappropriate differentiation
able consistency of this feature in various clinical forms that results in overproduction of a disorganized fibrotic
of fibrous dysplasia ranging from solitary asymptomatic bone lesion. In animal studies the implantation of human
lesions to severe generalized conditions suggests that the marrow progenitor cells with mutant Gsα admixed with
underlying molecular mechanism involves the fundamen- nonmutant cells results in the formation of abnormal
tal cell differentiation process. The differences in the ectopic ossicle recapitulating fibrous dysplasia.6,75 The
extent of skeletal involvement and the coexistence of mutations of the GNAS1 gene have also been explored
extraskeletal abnormalities can be related to somatic as potential biomarkers in the differential diagnosis of
mosaicism of the defect. The disease is evolutionary con- fibrous dysplasia and other fibroosseous lesions, includ-
served; recent evidence indicates that Homo sapiens’ ances- ing well-differentiated fibroblastic osteosarcoma.10,63,77,88
tors such as Neanderthals were affected by a similar Unfortunately, the presence of GNAS1 mutations in
disorder.57 well-differentiated fibroblastic osteosarcoma, both paros-
The involvement of a gene or genes playing a funda- teal and intramedullary, limits the diagnostic applications
mental role in modulation of cellular differentiation into of those mutations in the differential diagnosis of fibrous
mature osteocytes capable of producing lamellar bone can dysplasia and low-grade fibroblastic osteosarcoma.12,63
be postulated. In fact, mutations of signal-transducing The increased levels of the c-fos and c-jun oncoproteins
G proteins have been implicated in the development have been documented in lesions of fibrous dysplasia,
570
ERRNVPHGLFRVRUJ
Receptor Effector

o o
CH3O-Cys-S HN-Gly Cys-S
C

 GDP

A
GTP
A
GDP

Receptor Effector

o o B
CH3O-Cys-S HN-Gly Cys-S
Second
 Messenger/

Response
A 

Gs- splice variants:


Gs- long ± Ser,
Gs- short ± Ser
meth

Chr 20 Maternal NESP XLs 1A Gs- +/- +/-


C
allele 2 4 5 6-13
1 1 1 1 3 A
G

13
12 BMSCs: BMSCs: R201 mutation Exon 8
11.2 XLas biallelic Gs- biallelic, asymmetry paternal or maternal allele
11.1 11.1
11.2 meth meth
12
13.1 Paternal NESP XLs 1A Gs- +/-
+/-
C
allele 2 4 5 6-13
13.2 3 A
GNAS 1 1 1 1 G

13.3

Gs- imprinted:
C kidney, thyroid, pituitary, ovary

GNAS Exon 8
p.R201G p.R201S Exon 9
p.R201C p.R201P p.Q227L p.Q227K
p.R201H p.R201L p.Q227R p.Q227H

1 394 aa

P-loop containing nucleoside triphosphate hydrolase, 33-64


G protein alpha subunit, helical insertion, 58-202
D P-loop containing nucleoside triphosphate hydrolase, 198-393
FIGURE 8-1  ■  Biologic effects of the G protein signaling pathway. A, The heterotrimeric guanine nucleotide-binding regulatory pro-
teins (G proteins) are localized to the inner surface of the cell membrane. They transduce signals from transmembrane receptors.
In the inactive state (top diagram) the G protein exists as a trimer containing α, β, and γ subunits. Interaction of the G protein complex
with agonist-bound receptor stimulates the activation of GTP to GDP on the α subunit and the β and γ subunits from the active α
subunit (bottom diagram). The GTP-bound α subunit and the free β-γ dimer can independently stimulate effectors and second mes-
senger response. The hydrolysis of GTP to GDP deactivates the α subunit, allowing the reassociation of the α, β, and γ trimer and
the recoupling of the complex to the receptor. (A, Modified from Casey PJ: Curr Opin Cell Biol 6:219-225, 1994.) B, Molecular model
of the heterotrimeric complex containing α, β, and γ subunits highlighting regions that are known from biochemical experiments to
contact receptors (purple). (B, Reprinted with permission from Lambright DG, et al: Nature 379:311-319, 1996.) C, The human GNAS
gene is located in an imprinted gene locus at 20q13.2-3 and encodes several transcripts and alternative isoforms of the G1α protein.
The GNAS chromosomal region with maternal and paternal alleles showing promoters (solid boxes) and first exons specific for
each transcript (NESP55, SLαs, 1A, and Gs-α) as white boxes and exons common to all GNAS transcripts as gray boxes (exons 2-13)
are depicted. NESP55 is expressed from the maternal allele (red), whereas SLαs and 1A are expressed from the paternal allele (blue)
due to methylation patterns (meth). In clonogenic bone marrow stromal cells derived from either normal or fibrous dysplasia tissue,
NESP55 is restricted to a subset of clonal cells and is expressed from the maternal allele. However, unlike in other tissues, SLαs is
expressed from both the maternal and paternal alleles in normal and fibrous dysplasia bone marrow stromal cells (dashed arrows).
Gs-α is expressed from both alleles in most (but not all) normal tissues; four different splice variants have been reported: Gs-α-1
(short, without exon 3), GS-α-3 (long form with CAG immediately upstream of exon 4) (codon CAG), and Gs-α-4 (short form with
CAG). The R201 fibrous dysplasia mutation site is in exon 8. In kidney, thyroid, pituitary, and ovarian tissues, GS-α is only expressed
from the paternal allele (black arrow), but in normal and fibrous dysplasia bone marrow stromal cells, overall, it is expressed from
both alleles (dashed arrows), although a great deal of asymmetry of expression is noted in clonal bone marrow stromal cell strains.
The mutations predominantly involving exon 8 of the gene result in substitutions of arginine (A) for glycine (G), cysteine (C), histidine
(H), serine (S), proline (P), and leucine (L) at position 201 and less frequently in exon 9 at position 227 resulting in substitution of
glutamine (G) for leucine (L), arginine (R), lysine (K), and histidine (H). These mutations interfere with the formation of the tight
complex among α, β, and γ subunits and promote their dissociated active state, causing hyperproliferation and abnormal maturation
of bone forming cells. They also affect proliferations of cells in multiple neuroendocrine organs, skin melanocytes, and mesenchymal
cells. (C, Modified and reprinted with permission from Riminucci M, et al: J Mol Endocrinol 45:355-364, 2010.) D, Structure of the GWAS
ERRNVPHGLFRVRUJ
protein and its functional domains. The positions of most frequent mutations in exons 8 and 9 are indicated.
572 8  Fibrous Dysplasia and Related Lesions

but their role in the pathogenesis of this disorder is in which trabeculae of immature woven bone may be
unclear.9,44,76 The alterations of Gsα affect downstream present. The latter are typically not bordered by palisad-
regulatory pathways, playing an important role in the ing osteoblasts.
proliferation of cells involved in the development of the
skeleton, neuroendocrine sites, and skin melanocytes as Clinical Symptoms
the most frequently affected tissues. Transcriptional acti-
vation of the c-fos- and c-jun protooncogenes as well as Fibrous dysplasia can occur in several distinct clinical
Runx2 and Wnt/β-catenin, which play important roles in settings (Table 8-1). It has two basic clinical forms:
skeletal development, is caused by activating mutations of monostotic and polyostotic (Figs. 8-2 and 8-3).46,69 It can
Gsα protein.30,67,72 The human GNAS locus is maternally be associated with extraskeletal symptoms, which occur
and paternally imprinted and encodes multiple transcripts more frequently in patients with the polyostotic form.
and alternative isoforms. The genetic imprinting of both The most common extraskeletal symptom is hyperpig-
maternal and paternal alleles and of different alternative mentation of the skin (café au lait spots)(Fig. 8-4).4,70
isoforms produces a mosaic of transcripts with restricted More severe forms of polyostotic fibrous dysplasia may
expressions to specific tissues. This complexity, coupled be associated with endocrine abnormalities, such as pre-
with a background of somatic mutations and a mixture cocious puberty in female subjects (Albright-McCune
of mutant and nonmutant alleles, is responsible for the syndrome).1,45,53 Other less frequent endocrine dysfunc-
variable phenotypic presentations of GNAS related dis- tions include acromegaly, hyperthyroidism, hyperpara-
orders. In individual skeletal progenitors, the transcrip- thyroidism, and Cushing’s syndrome.5,20,26 Association
tional activity of two Gsα alleles can be different and each of fibrous dysplasia, typically of the polyostotic form,
allele can be selectively expressed or completely silenced. with soft tissue myxomas (Mazabraud’s syndrome) is
This is believed to be responsible for the highly variable the rarest form of clinical manifestation of this disease
clinical manifestations of GNAS related disorders ranging (Fig. 8-5).3,52,64
from a single focus to widespread skeletal involvement Monostotic fibrous dysplasia occurs more frequently
as well as distinctive alterations in predominantly neu- than the polyostotic variant. The ratio between the
roendocrine sites and skin melanocytes but also affect- monostotic and polyostotic forms varies among series
ing soft tissue, including cardiac muscle, among others. from 8 : 1 to 10 : 1. Monostotic fibrous dysplasia affects a
The presence of clonal structural alterations involving single bone, and typically one focus of involvement is
chromosomes 3, 8, 10, 12, and 15 suggests that fibrous identified. Polyostotic fibrous dysplasia is characterized
dysplasia may represent a neoplastic condition with a by multiple foci involving several bones. The polyostotic
predisposition to somatic mutations of skeleton-forming form is further subdivided into monomelic and polymelic
mesenchymal tissue.16,19,54 Extremely rare cases of familial subtypes. The lesions in polyostotic fibrous dysplasia
association between multiple endocrine neoplasia type 1 tend to be unilateral and to involve the bones of one
and Albright-McCune syndrome have been described, extremity (monomelic polyostotic fibrous dysplasia).
and this suggests a possible pathogenetic link between More severe forms can exhibit widespread skeletal
these two conditions.59 Deletions involving chromosome involvement (polymelic polyostotic fibrous dysplasia).
9q37.3 were found in several individuals with Albright- The disease usually manifests during the first three
McCune syndrome, suggesting the possible involvement decades of life (approximately 70% of cases). Monostotic
of this chromosome region in the development of the fibrous dysplasia may be asymptomatic and is discovered
syndrome.62 Rare familial transmission of fibrous dyspla- incidentally on radiographs obtained for other reasons.
sia has also been reported.40 The most common clinical symptom is swelling or
deformity of the affected site. In fact, severe deformi-
ties of the affected sites represent a major problem in
Definition
the clinical management of fibrous dysplasia (Fig. 8-6).
Fibrous dysplasia represents a dysplastic disorder of bone The lesion may occasionally be heralded by mild to
characterized by solitary or multifocal polyostotic intra- moderate pain of long duration. Pathologic fracture, par-
medullary lesions composed of proliferations of fibroblast- ticularly in the long tubular bones, may also be a present-
like spindle cells with a characteristic whorled pattern ing symptom. Often, the growth of the lesion stabilizes

TABLE 8-1 Summary of Clinicoradiographic Features of Fibrous Dysplasia


Variant of Fibrous Dysplasia Distribution of Lesions and Clinical Features
Monostotic Single focus in one bone
Polyostotic Multiple foci in several bones
  Monomelic Predominant or exclusive unilateral involvement of one extremity and of homolateral pelvis
or scapula
  Polymelic Widespread skeletal involvement
Albright-McCune syndrome Polyostotic (typically polymelic) fibrous dysplasia with endocrine abnormalities (most
frequently precocious puberty in female patients); less frequent anomalies include
acromegaly, hyperthyroidism, hyperparathyroidism, and Cushing’s syndrome
Mazabraud’s syndrome Polyostotic fibrous dysplasia associated with soft tissue myxomas

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 573

Polyostotic

Monostotic Monomelic Polymelic

FIGURE 8-2  ■  Fibrous dysplasia: sites of involvement. Typical sites of skeletal involvement in various forms of fibrous dysplasia.

at a certain point. This usually occurs during puberty. enlargement and premature development of pubic and
Skin pigmentations may correspond to sites of skeletal axillary hair. The café au lait spots are usually present at
involvement. birth or develop shortly thereafter. They are classically
The more extensive the disease, the earlier the onset described as having jagged outlines referred to as a coast
of symptoms. In the monostotic form, the most frequent of Maine outline. They show some respect to the midline
sites of involvement are the ribs, craniofacial bones, prox- but there are frequent exceptions to this rule. The devel-
imal femur, and tibia. The monomelic variant of the opment of café au lait spots with midline lateralism in the
polyostotic form frequently affects the lower extremity adjacent skin regions may produce a harlequin pattern
and the homolateral hemipelvis. The polymelic form (the of hyperpigmentation. The prevalence of major clinical
most severe generalized variant) shows widespread findings in fibrous dysplasia/Albright-McCune syndrome
involvement of both extremities, the trunk, and craniofa- is described in Table 8-2. Less common clinical find-
cial bones. Even in this form, the disease may predomi- ings are summarized in Table 8-3. Two thirds of patients
nantly involve one side. Involvement of the vertebral show involvement of the thyroid with hyperparathy-
column is extremely rare in fibrous dysplasia.58 roidism. Some patients with extensive fibrous dysplasia
Albright-McCune syndrome is clinically defined by may have hypophosphatemia due to the overproduction
the triad of fibrous dysplasia, café au lait spots, and of a circulating phosphaturic hormone caused by over-
precocious puberty.17,24,78 Clinical manifestations of production of FGF23 in the fibrous dysplasia tissue.17,18
Albright-McCune syndrome relative to age and their Overproduction of growth hormone is caused by the
clinical behavior are summarized in Figure 8-7. It is a presence of GNAS mutations in the anterior pituitary
rare syndrome with estimated prevalence of 1/100,000 causing hyperplasia manifesting as adenomas. Cushing’s
to 1/1,000,000. Typically, the signs of precocious puberty syndrome is one of the rarest endocrine abnormali-
or symptoms related to skeletal involvement by fibrous ties found in Albright-McCune syndrome and typically
dysplasia as well as early onset of café au lait spots are the occurs in the neonatal period. Other extraskeletal mani-
initial presentations. In girls, precocious puberty usually festations include gastrointestinal reflux, gastrointestinal
presents as vaginal bleeding or spotting accompanied by polyps, pancreatitis, and cardiac abnormalities potentially
the premature development of breast tissue and pubic resulting in tachycardia and sudden death.
hair. In boys, it is associated with testicular and penile Text continued on p. 578

ERRNVPHGLFRVRUJ
574 8  Fibrous Dysplasia and Related Lesions

A B
FIGURE 8-3  ■  Fibrous dysplasia: radionucleotide scanning with technetium-99. A, Skeletal survey reveals a single lesion involving the
left proximal femur proved histologically to be fibrous dysplasia. B, Polyostotic fibrous dysplasia showing multifocal involvements
of predominantly left lower extremity. Note less prominent involvement of the controlateral (right lower) extremity.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 575

B D E
FIGURE 8-4  ■  Representative café au lait spots seen in Albright-McCune syndrome. A and B, A spectrum of café au lait spots is seen
that both respect the midline and display “coast of Maine” borders. C, A very unusual café au lait spot seen in a child with Albright-
McCune syndrome and neonatal Cushing’s syndrome. While the spot respects the midline, the borders are smooth and the spots
alternate from left to right in a harlequin pattern. D, A very large café au lait spot with somewhat smooth borders in a patient with
relatively little fibrous dysplasia. E, A café au lait spot that clearly does not respect the midline. (Reprinted with permission from Collins
MT, et al: Orphanet J Rare Dis 7:54-67, 2010.)

ERRNVPHGLFRVRUJ
576 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-5  ■  Mazabraud’s syndrome: radiographic features. A, Plain radiograph of right forearm shows extensive polyostotic fibrous
dysplasia and soft tissue mass. B and D, T1- and T2-weighted magnetic resonance images (MRIs) show well-circumscribed soft
tissue mass and multiple low signal intensity (T1) intramedullary lesion of radius. Note uniform high signal intensity on T2-weighted
image of soft tissue mass. C, Gross appearance of excised soft tissue myxoma from same case.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 577

A B
FIGURE 8-6  ■  Fibrous dysplasia: clinical manifestations. A and B, Severe facial deformity in fibrous dysplasia of craniofacial bones.

Fibrous dysplasia
Café-au-lait
Precocious Puberty
Thyroid
Phosphaturia
Growth Hormone Excess
Cushings
0 5 10 15 20 30 50
Age
Pre-clinical Persistent abnormal menses
Clinically evident Spontaneous resolution possible
FIGURE 8-7  ■  Clinical manifestations relative to age in Albright-McCune syndrome. The relative age at which any given aspect of
the disease is preclinical or clinically evident is depicted. The possibility of spontaneous resolution of the symptoms in a fraction
of patients is also provided. The period of time during which abnormal menstruation can be expected is also depicted. (Reprinted
with permission from Collins MT, et al: Orphanet J Rare Dis 7:54-67, 2010.)

ERRNVPHGLFRVRUJ
578 8  Fibrous Dysplasia and Related Lesions

(Fig. 8-12). Predominantly radiopaque lesions occur more


TABLE 8-2 Prevalence of Major Findings
frequently in the craniofacial bones than in other parts of
in the NIH Cohort of Patients
the skeleton. This is related to the particular prevalence
with Fibrous Dysplasia/
of well-mineralized bone trabeculae in fibrous dyspla-
Albright-McCune Syndrome
sia of craniofacial bones. The bone contour is usually
Clinical Finding % Patients* expanded. This feature is particularly evident in bones
with small diameter (ribs and fibulas) and in flat bones but
Fibrous dysplasia 98
Café au lait spots 66 can also be seen in the major long tubular bones (Figs.
Gonadal abnormalities 8-8 and 8-9). In the femur, tibia, or humerus, fibrous
  Male: (ultrasound)† 70 dysplasia may cause expansion of the bone contour with
  Female: precocious puberty 50 cortical thinning and endosteal scalloping (Figs. 8-9 and
Thyroid abnormalities
  Abnormal ultrasound (U/S) 66
8-13). In the long tubular bones, the shaft is typically
  Hyperthyroid + abnormal U/S 28 involved, but the metaphyses are also frequently affected.
Renal phosphate wasting 43 Lesions extending to the epiphyseal end of a bone are
  Hypophosphatemia 10 unusual. Foci of punctate and ringlike calcification may
Growth hormone excess 21 be present and represent the areas of cartilaginous differ-
Cushing’s syndrome 4
entiation that may be found in fibrous dysplasia.79 When
*N = 140; 58 males, 82 females cartilaginous differentiation is extensive, the radiographic

Detected on ultrasound. appearance is similar to that seen in cartilage lesions.
Collins MT, et al: Orphanet J Rare Dis 7:S4, 2012. This variant of fibrous dysplasia is sometimes referred to
as fibrocartilaginous dysplasia.
The lesions in fibrous dysplasia are often well circum-
TABLE 8-3 Prevalence of Less Common scribed and are typically surrounded by a relatively wide
Findings in the NIH Cohort of rim of sclerotic bone sometimes referred to as a rind
Patients with Fibrous Dysplasia/ (Figs. 8-10, 8-11 and 8-12). This feature is often seen in
Albright-McCune Syndrome long-standing asymptomatic solitary lesions. Even the
expansile lesions are usually delineated by a thin rim of
Other Clinical Findings % Patientsa newly formed bone. Bony deformities, particularly of the
Gastrointestinal 7 weight-bearing bones, are associated with more extensive
  History of hepatitisb 4 involvement (Figs. 8-9 and 8-14 to 8-16).7,9,21 An example
  Refluxb 5 is the shepherd’s crook deformity of the proximal femur
  Pancreatitisb 3 (Figs. 8-14 and 8-15). In extremely rare cases, fibrous
  Polypsc 5
Cardiac 6 dysplasia can form an exophytic extraosseous mass or can
  Tachycardiad 4 grow on the surface of bone (Figs. 8-17 and 8-18). This
  Aortic root dilatation (GH excess)e 2 rare form typically occurs in smaller bones, such as the
Hematologic 1 ribs or short tubular bones of the hands and feet. These
  Platelet dysfunction 1
Cancer 4
lesions are sometimes attached to the adjacent bone by a
  Thyroidf 1 bony stalk or form exophytic sessile masses. This unusual
  Breastf 2 variant of fibrous dysplasia is also referred to as fibrous
  Bonef 1 dysplasia protuberans.23
  Testicularf 1
Hyperparathyroid 1
Neuropsychiatric 9 Gross Findings
a
b
N = 140; 58 males, 82 females. When a focus of fibrous dysplasia is examined in situ,
Appeared in childhood, common causes excluded. as in a resected rib or fibula, it presents as a centrally
c
Atypical, upper GI tract polyps, myxomatoid pathology more
common. located lesion composed of fibrous tan to gray gritty
d
Unexplained/not associated with hyperthyroidism. tissue (Fig. 8-19). Lesions that are more heavily ossified
e
Only seen in patients with growth hormone excess. may be yellow to white focally. Fibrous dysplasia usually
f
All tumors bear GNAS1 mutation, adjacent normal tissue mutation has sharp borders, and the bone contour is expanded
negative.
Collins MT, et al: Orphanet J Rare Dis 7:S4, 2012.
with a thinned cortex. Areas of cartilaginous tissue are
sometimes recognized as discrete translucent blue-white
nodules. Hemorrhage and cystic change may be present,
Radiographic Imaging and areas of prominent xanthogranulomatous reaction
have a yellow appearance. Some lesions may develop
On radiographs, fibrous dysplasia appears as a medul- extensive cystic change. Hemorrhagic, blowout areas
lary lesion with the so-called ground-glass appearance usually signify secondary aneurysmal bone cyst formation.
(Figs. 8-8 and 8-9).7-9 The radiographic density of the
lesion varies and depends on the relative proportions of Microscopic Findings
bone and fibrous elements. Lesions with abundant bone
elements are more radiopaque (Figs. 8-10 and 8-11). On Fibrous dysplasia is composed of spindle cells that have
the other hand, the predominance of fibrous tissue is a whorled or storiform arrangement with interspersed
associated with a more lucent radiographic appearance Text continued on p. 591

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 579

A B
FIGURE 8-8  ■  Fibrous dysplasia: radiographic features. A, Multifocal fibrous dysplasia of fibula. B, Enlargement of lesions shown in
A. Note central (intramedullary) location of lesions and their ground-glass appearance.

ERRNVPHGLFRVRUJ
580 8  Fibrous Dysplasia and Related Lesions

A B C
FIGURE 8-9  ■  Fibrous dysplasia: radiographic features. A, Shepherd’s crook deformity of proximal femoral shaft with external bowing
in patient with polyostotic fibrous dysplasia. B, Opposite femur of patient shown in A demonstrates anterior bowing and multiple
healed fractures and pseudarthrosis of proximal diaphysis. C, Expansile lesion in left humeral diaphysis of same patient. Note
ground-glass density of lesional tissue in A to C.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 581

C
FIGURE 8-10  ■  Fibrous dysplasia: radiographic features. A, Anteroposterior radiograph of humerus shows intramedullary lesion with
central opacities. B, T1-weighted magnetic resonance image shows low signal lesion with irregular signal void in heavily mineral-
ized areas, same case as in A. C, A lateral radiograph of left hip shows isolated fibrous dysplasia in intertrochanteric region of
femoral shaft. Note ringlike sclerosis of bone at periphery, which is characteristic of fibrous dysplasia in long bones.

ERRNVPHGLFRVRUJ
582 8  Fibrous Dysplasia and Related Lesions

B
FIGURE 8-11  ■  Fibrous dysplasia: radiographic features. A, Anteroposterior radiograph of proximal femur shows intramedullary lesion
with ill-defined opacities. B, T1-weighted magnetic resonance image shows low signal lesion involving the intertrochanteric region
and extending to the femoral neck.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 583

B C
FIGURE 8-12  ■  Fibrous dysplasia: radiographic features. A, Lateral radiograph shows radiolucent lesion with sclerotic margins involv-
ing calcaneus. B, T1-weighted magnetic resonance image shows low signal lesion of the calcaneus, same lesion as in A. C, Fat
suppression sequence reveals signal irregularities within the lesion as well as well-defined low signal intensity (sclerotic) margin,
same lesion as shown in A and B.

ERRNVPHGLFRVRUJ
584 8  Fibrous Dysplasia and Related Lesions

A B
FIGURE 8-13  ■  Polyostotic fibrous dysplasia: radiographic features. A and B, Plain radiographs of humerus and tibia of same patient
show extensive intramedullary involvement of multiple bones with cortical thinning and scalloping.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 585

FIGURE 8-14  ■  Fibrous dysplasia: radiographic features. A, Anteroposterior (AP) radiograph of pelvis and proximal femora shows
intramedullary lesion of left proximal femur with shepherd’s crook deformity. B, AP radiograph of pelvis and proximal femora in a
28-year-old woman with polyostotic fibrous dysplasia and Albright-McCune syndrome. There is asymmetric involvement with more
severe changes on right side (monomelic form). Note malunited femoral shaft fracture on right (arrows).

ERRNVPHGLFRVRUJ
586 8  Fibrous Dysplasia and Related Lesions

B C
FIGURE 8-15  ■  Fibrous dysplasia: radiographic features. A, Extensive skull involvement in polyostotic fibrous dysplasia with Albright-
McCune syndrome. Note marked thickening with ground-glass opacity. B, Pathologic fracture of femoral shaft in fibrous dysplasia,
same case as in A. C, Bowing deformity with transverse fractures on convex side of femur in fibrous dysplasia.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 587

B C D
FIGURE 8-16  ■  Polyostotic fibrous dysplasia: radiographic features (Albright-McCune syndrome). A, Anteroposterior view of pelvis
and proximal femora shows shepherd’s crook deformity of proximal femur on left. B-D, Expansile diaphyseal lesions in humerus,
femur, and second metatarsal, respectively, in same patient.

ERRNVPHGLFRVRUJ
588 8  Fibrous Dysplasia and Related Lesions

A C
FIGURE 8-17  ■  Exophytic fibrous dysplasia (fibrous dysplasia protuberans). A, Lateral radiograph shows sclerotic, well-delineated
lesion at distal end of tibia with protrusion posteriorly. B and C, T1- and T2-weighted magnetic resonance images of same lesion
demonstrate exophytic nature of lesion.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 589

A B

C
FIGURE 8-18  ■  Exophytic fibrous dysplasia: radiographic features. A, Exophytic, sharply circumscribed mass in shaft of right tibia is
seen in this axial computed tomogram (CT). Note sclerotic margin on medullary aspect. B, Plain radiograph of chest shows heavily
calcified, pedunculated mass attached to surface of left sixth rib. C, CT of chest shows parosteal mass with matrix calcification.
(From Dorfman HD, Ishida T, Tyuneyoshi M: Hum Pathol 25:1234-1237, 1994.)

ERRNVPHGLFRVRUJ
590 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-19  ■  Fibrous dysplasia of rib: gross features. A and B, Rib resections show fusiform expansion of anterior portion of rib
with gritty gray-tan solid fibrous tissue in medullary cavity. C, Rib resection shows expansile fusiform lesion with gritty gray-tan
fibrous tissue. D, Rib resection shows fusiform expansion by fibrous lesion with brown areas of old hemorrhage.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 591

trabeculae of immature woven bone devoid of rimming Differential Diagnosis


osteoblasts (Fig. 8-20).36,66,71,98 The number and distribu-
tion of bone trabeculae and their level of maturation may Fibrous dysplasia is most frequently confused histologi-
vary among different lesions and in different areas of cally with other benign fibroosseous lesions, in particular
the same lesion (Figs. 8-21 to 8-23). The most common with osteofibrous dysplasia. This entity is composed of a
and characteristic pattern of bone produced in fibrous mixture of fibrous tissue and mature bone trabeculae that
dysplasia consists of slender, curved, and branching tra- exhibit classic osteoblastic rimming. Osteofibrous dyspla-
beculae of bone without surface osteoblasts. These are sia is an intracortical lesion that occurs in children,
frequently described as resembling Chinese characters whereas fibrous dysplasia tends to be more centrally
(Fig. 8-22), although no such characters actually exist in located in the medullary cavity and is often first diag-
written Chinese. In some cases, the trabeculae of bone nosed in adults.47 It affects the tibia and fibula almost
are sparsely distributed with a predominance of cellular exclusively, whereas fibrous dysplasia can occur in any
fibrous tissue. At the other end of the spectrum are heavily bone. Desmoplastic fibroma, because it can mimic the
ossified lesions in which relatively mature bone trabec- fibrous component of fibrous dysplasia, is sometimes
ulae overshadow the fibrous component of the lesion considered when a small biopsy sample does not include
(Figs. 8-23 and 8-24). The lesions with abundant bony the osteoid and woven bone elements found in fibrous
components frequently contain foci of matrix mineraliza- dysplasia. Reactive bone formation in desmoplastic
tion that resemble cementoid bodies (Fig. 8-24). These fibroma at the edge of the lesion can usually be recog-
concentrically laminated calcified structures are most fre- nized by the prominent osteoblastic rimming. It is most
quently seen in fibrous dysplasia involving craniofacial important to distinguish between fibrous dysplasia and
bones but may also be present in other sites.89,92,99 In the low-grade intramedullary osteosarcoma. The latter lesion
past, lesions with prominent cementoid structures were can be difficult to diagnose when the bone trabeculae of
designated, especially in the oral pathology literature, as the tumor forms the branching or interconnected pattern
fibrocementomas or cementifying fibromas. usually associated with fibrous dysplasia. However, the
Examination under polarized light reveals a very char- spindle-cell areas in this entity usually show nuclear
acteristic and diagnostically important feature: All bone atypia and larger nuclei with a coarser chromatin pattern
trabeculae in fibrous dysplasia, despite their mature than those found in fibrous dysplasia. As opposed to the
appearance, show a polarization pattern that forms dis- circumscribed expansile growth pattern of fibrous dyspla-
organized, haphazard deposits characteristic of woven sia with peripheral bone sclerosis, low-grade intramedul-
bone (Fig. 8-25). Secondary changes in fibrous dysplasia lary osteosarcoma permeates cancellous bone trabeculae
may alter the typical microscopic appearance and make with an infiltrative growth pattern. Preliminary evidence
it difficult to diagnose. Hemorrhage and secondary fibro- indicates that the presence of mutations involving a gene
histiocytic reaction may be present focally. Occasionally coding for Gsα subunit in fibrous dysplasia may be a useful
a diffuse infiltrate of foamy histiocytes may obscure the marker in differential diagnosis of this disorder and other
characteristic microscopic features of the lesion. Such fibroosseous lesions, including well-differentiated fibroblas-
lesions may be incorrectly designated as xanthomas of tic osteosarcoma.11,63,77
bone. Intralesional hemorrhage in fibrous dysplasia can Polyostotic fibrous dysplasia may mimic enchondroma-
provoke extensive giant cell reaction, which leads to con- tosis on radiographs because of the multiplicity of lesions
fusion with giant cell tumor (Figs. 8-26 and 8-27). Myxoid in both conditions, but enchondromatosis tends to show
change of stromal tissue can be focal or quite extensive a much greater unilateral predominance. Histologically,
in some lesions. These secondary changes are more likely these lesions may be confused when fibrous dysplasia
to be found in the older lesions of fibrous dysplasia. contains an abundance of cartilage. In these cases, atten-
Reactive bone with prominent osteoblastic rimming tion to the radiographic features and careful study of the
may be seen focally in fibrous dysplasia complicated by histologic material for evidence of fibroosseous elements
pathologic fracture. Small fractures of the thinned cortex usually resolves the dilemma. In more florid examples of
may not be clinically and radiographically evident; there- massive chondroid differentiation in fibrous dysplasia,
fore the stimulus to reactive bone formation remains there is also the danger of confusion with conventional
unrecognized. Secondary aneurysmal bone cyst with chondrosarcoma. This can be excluded by the absence of
radiographic evidence of an expansile lesion is a common chondrocyte atypia and the presence of enchondral ossi-
complication (Figs. 8-26 to 8-28).91,96 Cystic change with fication at the borders of the dysplastic chondroid foci.
accumulation of serous fluid in fibrous dysplastic tissue is
a distinct phenomenon and should not be automatically Treatment and Behavior
interpreted as a secondary aneurysmal bone cyst. The
underlying fibrous dysplasia can be almost completely In fibrous dysplasia the multifocal skeletal lesions develop
obliterated by either type of secondary cystic change. more or less synchronously. Therefore the initially diag-
Microscopic foci of cartilaginous differentiation are fre- nosed monostotic forms typically do not progress to
quently present in fibrous dysplasia, but in rare cases the polyostotic generalized forms. However, additional foci
cartilage matrix may dominate the microscopic and of fibrous dysplasia occasionally develop in an affected
radiographic pictures. These rare cases are referred to as bone.9,36 A solitary focus may enlarge in size, and the
fibrous dysplasia with massive cartilaginous differentiation or extent of involvement can progress over time. In rare
fibrocartilaginous dysplasia. This peculiar variant of fibrous instances, such locally progressive lesions may expand
dysplasia is separately described in further detail. Text continued on p. 601

ERRNVPHGLFRVRUJ
592 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-20  ■  Fibrous dysplasia: microscopic features. A-D, Different patterns of branching and focally anastomosing trabeculae of
woven bone in fibrous stroma (A-C, ×100; D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 593

A B

C D
FIGURE 8-21  ■  Fibrous dysplasia: microscopic features. A-D, Different patterns of branching and anastomosing trabeculae of imma-
ture woven bone in fibrous stroma (A-C, ×200; D, ×100). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
594 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-22  ■  Fibrous dysplasia: microscopic features. A-D, Mineralized trabeculae of woven bone in fibrous stroma
(A-D, ×100) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 595

A B

C D
FIGURE 8-23  ■  Fibrous dysplasia: microscopic features. A-D, Different patterns of branching and anastomosing trabeculae of woven
bone in moderately cellular fibrous stroma (A-D, ×100) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
596 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-24  ■  Fibrous dysplasia: microscopic features. A-D, Different patterns of cementum-like, round, calcified spherules of woven
bone in otherwise typical fibrous dysplasia. This finding is noted in a minority of cases in extragnathic sites. They may be numerous
or few and arranged in clusters (A and B, ×50; C and D, ×100). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
A

FIGURE 8-25  ■  Fibrous dysplasia: microscopic features. A, A pattern of lamellar mineralization in cortical bone overlaying a focus of
fibrous dysplasia shown in B. Inset, Higher magnification of A with clear lamellar mineralization pattern showing parallel lines of
mineralization under polarized light. B, Focus of fibrous dysplasia showing well delineated trabeculae of woven bone in cellular
fibrous stroma. Inset, Haphazardous woven bone mineralization pattern under polarized light (A and B, ×100; insets, ×200).
(A, B, and insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
598 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-26  ■  Fibrous dysplasia: microscopic features. A and B, Different patterns of woven bone trabeculae with anastomosing
features in fibrous stroma. C and D, Areas of fresh hemorrhage and giant cell reaction consistent with secondary aneurysmal bone
cyst in the same case of fibrous dysplasia (A-D, ×100) (A-D, hemotoxylin-eosin).

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 599

A B

C D
FIGURE 8-27  ■  Fibrous dysplasia with secondary changes: microscopic features. A, Typical pattern of immature bone trabeculae in
fibrous dysplasia of rib. B, Extensive giant cell reaction associated with intralesional hemorrhage. Different lesion of case shown in
A. C, Higher magnification of B shows prominent giant cell and spindle-cell reactions. D, Features of aneurysmal bone cyst super-
imposed on fibrous dysplasia (same case as in A and B) (A, B and D, ×100; C, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
600 8  Fibrous Dysplasia and Related Lesions

C
FIGURE 8-28  ■  Fibrous dysplasia: radiographic features. A, Anteroposterior radiograph shows fluid level in expanded cystic mandibu-
lar lesion. B, Panorex radiograph of jaw shows expanded lucent lesion at left angle of mandible. C, Computed tomogram shows
multiloculation in lesion of fibrous dysplasia with extensive secondary aneurysmal bone cyst formation.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 601

beyond the bone contour into the soft tissue.39 This microscopic features. For these reasons, separate descrip-
occurs most frequently during the skeletal growth period. tions of the disorder in various anatomic sites are
The lesions have a tendency to stabilize after puberty, provided.
and new foci rarely develop during adulthood. The
enlargement of lesions of fibrous dysplasia after puberty Fibrous Dysplasia of Craniofacial Bones.  Craniofacial
is more frequently related to secondary aneurysmal bone bones are the most frequent sites of involvement by soli-
cyst and simple cystic degeneration rather than to actual tary fibrous dysplasia (Figs. 8-28 and 8-29).27,28,37,87,104
proliferative activity of the lesion. Superimposition of They may also be involved in the polyostotic generalized
aneurysmal bone cyst and rapid enlargement of the form (Fig. 8-30). The most frequently affected bone is
lesion may prompt surgical intervention.106 Pregnancy the maxilla, followed by the bones of the cranial vault and
seems to promote enlargement of the lesions, but the the mandible. Clinically the disease is manifested by
mechanism of this phenomenon is unclear. It is unknown malar enlargement (cherubism) or unilateral facial hyper-
whether it is related to increased proliferative activity trophy. The most typical radiographic presentation of
or simply to secondary changes such as hyperemia and fibrous dysplasia in the craniofacial bones is in the form
hemorrhage. of a cloudy opacity of the maxillary bone. The lesions of
Bone deformities in fibrous dysplasia are treated by the mandible and of the cranial bones produce fusiform
corrective surgery. A solitary focus that is prone to patho- expansion of the bone contour. Computed tomography
logic fracture, especially in the long bones, is treated by and magnetic resonance imaging are particularly helpful
curettage and bone grafting with or without internal fixa- in delineating the extent of involvement in this region
tion. Lesions of small-diameter expendable bones, such and in showing the expansile lesion surrounded by a
as the ribs or fibula, are usually treated by segmental thinned and scalloped cortex that is intact (Fig. 8-29).14,97
resection. In severe polyostotic disease, extensive involvement of
The development of secondary sarcoma in fibrous dys- the craniofacial bones can produce pronounced disfigur-
plasia is an extremely rare but well-established phenom- ing deformities (Figs. 8-6, 8-15, and 8-29).
enon.9,22,25,29,32,38,56,61,68,73,74,82,83,90,93,95,105 It occurs in less Microscopically, fibrous dysplasia of the craniofacial
than 1% of patients during adult life in long-standing bones tends to produce more bone trabeculae than in
lesions. Lesions treated with radiation therapy are more other skeletal sites.27 The bone production is usually pro-
likely to develop sarcomatous transformation, but rare nounced, with formation of an anastomosing network of
cases of spontaneous malignant transformation also have well-mineralized woven bone arranged in broader tra-
been reported.34 Malignant transformation is related to beculae. Sometimes these trabeculae approach lamellar
the extent of involvement, and patients with polyostotic maturation. The calcification pattern may include con-
fibrous dysplasia have a greater risk of the development centric spherical structures designated as cementoid bodies
of sarcomatous transformation than those with mono- (Fig. 8-24). Such lesions, especially in the oral pathology
stotic forms. The average lag between the diagnosis and literature, have been designated as fibrocementomas with
the development of a malignancy is 13.5 years, and the implication that they are pathogenetically distinct
patients are generally in the third and fourth decades of from fibrous dysplasia. The current trend is to consider
life. The types of sarcoma that complicate fibrous dyspla- them as examples of fibrous dysplasia with a peculiar
sia are most frequently malignant fibrous histiocytoma pattern of mineralization rather than as a distinct entity.
(fibrosarcoma) or osteosarcoma of high histologic grade. The presence of similar cementum-like structures in
Chondrosarcoma that develops in association with fibrous fibrous dysplasia involving other skeletal sites supports
dysplasia has also been reported.22,32 An extremely rare the concept that they are not specific for dental cemen-
case of association between desmoplastic fibroma and toid differentiation.
fibrous dysplasia was documented in one report.102 The
craniofacial skeleton and femur are the most common Fibrous Dysplasia of Ribs.  Ribs are frequently involved
sites of malignant transformation. This generally corre- by a solitary focus of fibrous dysplasia.33,80,98 In some
lates with the frequency of distribution of bones involved studies, they represent the most frequently affected site.
in fibrous dysplasia. The prognosis for patients who Involvement of the ribs can also occur in polyostotic
develop sarcomatous transformation is poor. Pulmonary forms. The lesion usually presents as an asymptomatic,
metastasis develops in most patients, and the mean sur- palpable, fusiform enlargement of the rib. Radiographi-
vival period is 3.4 years. Alterations in the clinical course cally, it is a lucent mass of ground-glass density expanding
in patients older than age 40 years with known fibrous the rib contour (Figs. 8-31 and 8-32). Clinically and
dysplasia, such as pain and swelling, should be carefully radiographically, fibrous dysplasia must be differentiated
evaluated. from cartilage lesions (chondrosarcoma), especially if the
mass is growing and is associated with pain. In this site,
Fibrous Dysplasia in Different fibrous dysplasia is treated by segmental excision. The
lesions in the ribs frequently show minimal bone produc-
Anatomic Sites tion and are predominantly fibrous. Secondary changes
The biologic potential of fibrous dysplasia is not related (myxoid, xanthogranulomatous, giant cell, and reactive
to the site of involvement. Differential diagnosis, clinical bone formation) frequently occur in the rib lesions.
presentation, and treatment may vary in different ana- These changes are probably the result of repeated
tomic sites. Moreover, fibrous dysplasia in certain loca- mechanical stress related to respiratory movements and
tions, such as the craniofacial bones, has some distinct Text continued on p. 606

ERRNVPHGLFRVRUJ
602 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-29  ■  Fibrous dysplasia: radiographic features. A, Computed tomogram (CT) of maxilla shows extensive involvement by
fibrous dysplasia with obliteration of left maxillary sinus and nasal cavity. B, CT of skull shows expansile lesion of fibrous dysplasia
in left frontal bone. C, Severe craniofacial distortion by polyostotic fibrous dysplasia in a 35-year-old woman. Note extensive involve-
ment of mandible, maxilla, and frontal region of skull. D, Three-dimensional CT of skull shown in B. Bulging lesion depresses roof
of orbit and expands frontal bone.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 603

A B

C D
FIGURE 8-30  ■  Polyostotic fibrous dysplasia: radionuclide scanning. A-D, Technetium-99 radionuclide scans in a 22-year-old woman
who had right arm pain associated with humeral shaft lesion proved histologically to be fibrous dysplasia. Skeletal survey revealed
lesions in contralateral humerus (A), base of skull (B), left femur (C), and left ilium (D).

ERRNVPHGLFRVRUJ
604 8  Fibrous Dysplasia and Related Lesions

C D
FIGURE 8-31  ■  Fibrous dysplasia: radiographic features. A, Radiograph of thorax shows expanded anterior portion of right upper rib
involved by isolated lesion of fibrous dysplasia. Inset, Computed tomogram (CT) shows fusiform expansion of rib anteriorly with
hazy lucency in medullary cavity and intact shell of cortical bone. B, Anteroposterior radiograph of chest shows fusiform expansion
produced by single focus of fibrous dysplasia (arrows). C, Technetium 99 radionuclide scan shows lesion involving second left rib.
D, CT documents presence of fusiform lesion in rib with cortical thinning.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 605

C D
FIGURE 8-32  ■  Fibrous dysplasia: radiographic features. A and B, Anteroposterior radiographs of left and right hands of patient with
polyostotic fibrous dysplasia show bilateral involvement of metacarpal shafts and phalanges. Note broadening of diaphysis with
ground-glass medullary opacity. C, Expansile lytic lesion of rib incidentally discovered on chest radiograph. D, Low-power photo-
micrograph shows network of woven bone in fibrous stroma (D, ×25, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
606 8  Fibrous Dysplasia and Related Lesions

may overshadow the diagnostic features of fibrous dys- lesion that is generally confined to the cortex. Fibrous
plasia. Consequently, some fibrous dysplasia lesions in dysplasia is almost always an intramedullary lesion.
the ribs cannot be recognized with certainty and are des-
ignated as unclassified fibroosseous lesions.
FIBROCARTILAGINOUS DYSPLASIA
Fibrous Dysplasia of Extremity Bones.  The long
tubular bones of the appendicular skeleton are frequently In 1984, Dahlin et al.111 described a rare lesion of bone
involved in the monostotic and polyostotic forms of that contained fibroosseous components and extensive
fibrous dysplasia. Involvement of the short tubular bones areas of cartilage as fibrocartilaginous mesenchymoma of
of the hands and feet is extremely rare. It typically occurs bone and believed that it had a low-grade malignant
in the polyostotic form with associated involvement of potential. Several additional similar lesions were described
other bones of the same extremity, widespread skeletal under the name of fibrocartilaginous mesenchymoma of
involvement, or both (Figs. 8-13, 8-14, 8-16, and 8-32). bone.108,113,114 Our own experience with eight cases of
In monostotic fibrous dysplasia, the proximal femoral similar lesions indicates that it should be considered as a
shaft is involved in approximately 40% of patients, fol- variant of fibrous dysplasia rather than as a separate and
lowed by the tibia (15%), humerus (5%), and radius (5%). distinct neoplasm. Moreover, the long-term clinical
In severe polyostotic fibrous dysplasia, the femur is follow-up data indicate no evidence of malignant behav-
usually involved. The tibia is involved in approximately ior.108 For all these reasons, we designate these lesions as
50% of patients, followed by the humerus (30%). Severe fibrous dysplasia with massive cartilaginous differentiation or
deformities (shepherd’s crook deformity), pathologic fibrocartilaginous dysplasia.115 The main significance of this
fractures, or both (Figs. 8-9, 8-14, and 8-16) may develop condition is that it may be misinterpreted as a cartilage
in long tubular bones, especially the weight-bearing neoplasm. The lesion is extremely rare; the Mayo Clinic
bones. recently reported only 12 cases of this entity from their
files.73 Several additional individual cases of fibrocarti-
Fibrous Dysplasia of Flat Bones.  Monostotic fibrous laginous dysplasia have been described in the litera-
dysplasia of the flat bones (pelvis or scapula) is rare. ture.110,112,117,118,120 A case of what is probably the same
Involvement of the flat bones is typical in polyostotic entity was originally reported by Telford119 in 1930.
fibrous dysplasia. In these patients, the flat bones are A series of eight additional cases was reported by
involved in addition to the corresponding bones of the Choi et al.109
appendicular skeleton (i.e., the pelvis and femur or
scapula and humerus). Multifocal involvement of the Incidence and Location
pelvic bones is present in approximately 50% of patients
with severe polyostotic fibrous dysplasia. Massive chondroid differentiation in fibrous dysplasia is
extremely rare, and eight cases of this entity were found
in our consultation files. The clinical data for these eight
Personal Comments
cases are summarized in Table 8-4. The age of patients
This dysplastic anomaly of bone formation commonly ranged from 4 to 26 years, with an average age of 17.5
presents as a solitary lesion in a rib, the maxilla, or a major years. Four patients were male, and four were female.
long bone, but it is very rare in its polyostotic form. Much Two patients had an associated polyostotic form of
of the diagnostic confusion with regard to fibrous dyspla- fibrous dysplasia. One patient had a history of Wilms’
sia is related to the variable clinical presentations and the tumor of the right kidney at age 4 years and had under-
variety of histologic patterns associated with this condi- gone nephrectomy, chemotherapy, and radiation therapy.
tion. It has been known since the initial pathologic This patient also had fibromatosis of the buttock at age
description by Lichtenstein41 that, in addition to the 14 years.
fibrous and osseous components, cartilage differentiation The proximal portion of the femur is the most common
is frequently found and may be voluminous, causing con- site of fibrocartilaginous dysplasia. Five of the eight cases
fusion with cartilaginous neoplasms in the skeleton. In involved the femoral neck or proximal shaft of the femur.
addition, extensive intralesional hemorrhage with giant Tibial involvement was seen in one case and involvement
cell reaction can lead to a misdiagnosis of giant cell tumor of the midfemur and distal femur was seen in another
or giant cell reparative granuloma. We have seen exam- case. This distribution is distinct from that seen by the
ples with extensive xanthomatous reactions interpreted Mayo Clinic, which reported a majority of their cases to
pathologically as fibroxanthomas or as Erdheim-Chester be localized in the tibia.108
disease. We have also seen rare examples of highly eccen-
tric and even exophytic fibrous dysplasia producing pro- Clinical Symptoms
tuberant lesions that could be mistaken on radiographs
for osteochondromas. The symptoms of fibrocartilaginous dysplasia vary. The
In the past, there has been much emphasis on the dif- presenting symptoms are usually pain with or without
ficulty in distinguishing histologically between osteofi- swelling. In our series, two patients had swelling or a
brous dysplasia (formerly called ossifying fibroma of long mass, and two patients were asymptomatic. Three patients
bones) and fibrous dysplasia. This distinction should not had pain, and one patient had a pathologic fracture.
be difficult, however, since osteofibrous dysplasia occurs The duration of symptoms ranged from a few weeks to
exclusively in the tibia and fibula of children and is a 15 years.

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 607

TABLE 8-4 Summary of Clinical Data of Fibrocartilaginous Dysplasia


Age Duration of Calcification Preconsultation
Case (yr)/sex Location Type Symptoms Symptoms on Radiograph Diagnosis Other Findings
1 20/M Femur Polyostotic Mass 15 yr + Chondrosarcoma −
(proximal)
2 23/F Tibia Polyostotic − − + FD with cartilage Ovarian
(proximal) dysgenesis
3 8/F Femur Monostotic Pain 7 mo + Cartilage tumor −
(proximal)
4 20/M Femur Monostotic Pain ? − FD with cartilage −
(proximal)
5 26/F Ischiopubic Monostotic Mass 8 yr + Chondrosarcoma −
bone
6 14/F Femur Monostotic − − + Cartilage tumor Wilms’ tumor,
(proximal) fibromatosis
7 25/M Femur Monostotic Pain, 2 yr (pain) + Enchondroma, R/O −
(proximal) fracture chondrosarcoma
8 4/M Femur Monostotic Limp ? −* ? −
(proximal)

*Chondroid tumor suggested by magnetic resonance imaging.


FD, fibrous dysplasia; R/O, rule out.
From Ishida T, Dorfman HD: Am J Surg Pathol 17:924-930, 1993.

Radiographic Imaging myxoid change of the cartilaginous matrix was seen in


only one case. The lobules of cartilage are sharply demar-
On radiographs the lesions are generally well demarcated cated from the surrounding fibroosseous tissue. A more
and show ground-glass opacity or have an osteolytic irregular pattern of cartilage matrix deposition is fre-
appearance (Figs. 8-33 and 8-34). When the shaft of the quently present. Enchondral ossification is commonly
femur or tibia is involved, cortical expansion can be seen; seen in the peripheral regions of the cartilage islands
however, the cortex is always intact (Figs. 8-33 and 8-34). (Figs. 8-36 to 8-38). Focally, hypertrophic chondrocytes
In one case a typical shepherd’s crook deformity was are arranged in columns with calcification of the matrix
found in the femoral neck. In six cases, stippled or ring- and formation of irregular columns of enchondral ossifi-
like calcifications suggesting chondroid elements were cation similar to that seen in the epiphyseal growth plate.
evident on plain radiographs (Figs. 8-33 and 8-34). In one Prominent capillaries between the vertical columns of the
case, magnetic resonance imaging showed a low-intensity areas mimic primary spongiosa, and osteoclastic resorp-
signal in T1-weighted images and a high intensity signal tion is present. Active enchondral ossification is seen
with lobulation in T2-weighted images, suggesting a focally in the majority of cases. Enchondrally ossified
chondroid lesion with a lobular architecture (Fig. 8-34). trabeculae merge into the irregularly shaped bone tra-
beculae in the surrounding fibroosseous lesion (Figs.
Gross Findings 8-37 and 8-38). Although the fibroosseous lesion shows
the typical histologic appearance of fibrous dysplasia,
When curettage is performed, the gross appearance of bone trabeculae around the cartilage islands are focally
the lesion is similar to that of enchondroma or low-grade surrounded by osteoblasts, which represent the process
chondrosarcoma and consists of fragments or irregular of enchondral ossification. Prominent immature osteoid
masses with an obvious cartilaginous appearance. In cases formation can be seen focally. Larger areas of predomi-
with less prominent cartilaginous components, the lesion nantly fibrous tissue show more typical features of fibrous
can have a fibrous, soft, or gritty appearance similar to dysplasia. Fibrous septa separating sharply demarcated
that seen in typical fibrous dysplasia. islands of cartilage may exhibit hypercellularity with
prominent plump fibroblastic cells.
Microscopic Findings
Differential Diagnosis
A large amount of hyaline cartilage and a disproportional
amount of cartilage versus fibroosseous elements are Although cartilaginous differentiation in fibrous dyspla-
troublesome findings. Cartilaginous differentiation in sia is usually inconspicuous, its presence is well known,
this variant of fibrous dysplasia is usually in the form of and all textbook descriptions of fibrous dysplasia include
large lobules surrounded by fibroosseous tissue that con- at least a passing reference to the occasional presence of
tains features typical of fibrous dysplasia (Fig. 8-35). The cartilage islands. In rare instances, there may be massive
cartilage in this condition has microscopic features of amounts of cartilage elements that reach several cen-
immaturity (i.e., increased cellularity and mild nuclear timeters in diameter.80 In these cases, large dysplastic
atypia) (Figs. 8-36 to 8-38). The level of matrix matura- cartilage islands may be misinterpreted as benign or even
tion can approach that seen in hyaline cartilage, but foci malignant cartilaginous neoplasms. It is of the utmost
of myxoid change can also be seen. More extensive Text continued on p. 614

ERRNVPHGLFRVRUJ
608 8  Fibrous Dysplasia and Related Lesions

B C
FIGURE 8-33  ■  Fibrous dysplasia with cartilage differentiation (fibrocartilaginous dysplasia): radiographic features. A, Femoral neck
lesion shows stippled calcification in well-demarcated lytic area. B, Ringlike, punctate, and curvilinear arclike calcifications in lytic
lesion occupying upper femoral shaft and femoral neck. C, Slightly expansile lytic lesion occupying proximal two thirds of tibial
shaft. Note overall ground-glass appearance with punctate and ringlike calcifications corresponding to cartilage matrix in proximal
part of lesion. (From Ishida T, Dorfman HD: Am J Surg Pathol 17:924-930, 1993.)

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 609

A B C

D
FIGURE 8-34  ■  Fibrous dysplasia with cartilage (fibrocartilaginous dysplasia). A, Anteroposterior radiograph of femur of a 4-year-old
boy shows expanded contour of diaphysis with well-circumscribed borders delineating lobulated, lucent intramedullary lesion.
B, T1-weighted magnetic resonance image (MRI) shows low signal area with sharp borders. C, T2-weighted MRI shows high signal
area with lobulated pattern corresponding to high water content of cartilage lobules. D, Expansile lytic lesion of pubic bone contain-
ing predominantly punctate calcification. (From Ishida T, Dorfman HD: Am J Surg Pathol 17:924-930, 1993.)

ERRNVPHGLFRVRUJ
610 8  Fibrous Dysplasia and Related Lesions

B C
FIGURE 8-35  ■  Fibrous dysplasia with cartilage differentiation (fibrocartilaginous dysplasia): microscopic features. A, Low power
view, showing islands of hyalinized cartilage (left) and fibrous stroma with trabeculae of woven bone characteristic of fibrous
dysplasia. B, Higher magnification of A showing an interface between cartilage islands and fibrous stroma. Note an
irregular contour of the cartilage islands and the trabeculae of woven bone within the fibrous stroma. C, Higher magnification of
another area from A showing trabeculae of woven bone in fibrous stroma characteristic of fibrous dysplasia (A, ×50; B and C, ×100).
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 611

B
FIGURE 8-36  ■  Fibrous dysplasia with cartilage differentiation (fibrocartilaginous dysplasia): microscopic features. A and B, Low
and intermediate power view of the interface between large areas of hyaline cartilage and fibrous stroma. Note irregular ragged
border of the cartilage island with enchondral ossification characteristic of fibrocartilaginous dysplasia (A, ×50; B, ×100). (A and
B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
612 8  Fibrous Dysplasia and Related Lesions

A B

C D
FIGURE 8-37  ■  Fibrous dysplasia with cartilage differentiation (fibrocartilaginous dysplasia): microscopic features. A-D, Large area
of hyaline cartilage and intervening stromal fibrous tissue showing irregular outline of cartilage and features of enchondral ossifica-
tion (A and B, ×50; C and D, ×100). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 613

A B

C D

FIGURE 8-38  ■  Fibrous dysplasia with cartilage differentiation (fibrocartilaginous dysplasia): microscopic features. A-C, Cartilage
island and fibrous tissue interface showing columnar arrangement of chondrocytes and endochondral ossification mimicking
epiphyseal growth plate frequently seen in fibrocartilaginous dysplasia. D and Inset, Higher magnification of cartilage island showing
enlarged atypical chondrocytes (A-C, ×100; D, ×200; inset, ×400.)

ERRNVPHGLFRVRUJ
614 8  Fibrous Dysplasia and Related Lesions

importance to be aware of the possibility of finding car- 3. Aoke T, Kouho H, Hisaoka M, et al: Intramuscular myxoma with
tilaginous predominance in an otherwise typical lesion of fibrous dysplasia: a report of two cases with review of literature.
Pathol Int 45:165–171, 1995.
monostotic or polyostotic fibrous dysplasia and to avoid 4. Benedict PH, Szabo G, Fitzpatrick TB, et al: Melanotic macules
the risk of diagnosing cartilage neoplasia. in Albright’s syndrome and in neurofibromatosis. JAMA 205(9):
Although the chondrocytes in fibrocartilaginous dys- 618–626, 1968.
plasia are generally small with condensed nuclei, they 5. Benjamin DR, McRoberts J: Polyostotic fibrous dysplasia associ-
ated with Cushing syndrome. Arch Pathol 96(3):175–178, 1973.
may also show atypical findings such as hypercellularity, 6. Bianco P, Kuznetsov SA, Riminucci M, et al: Reproduction of
binucleate cells, and enlargement in focal areas.115 Large human fibrous dysplasia of bone in immunocompromised mice by
amounts of atypical cartilage are diagnostic pitfalls in the transplanted mosaics of normal and Gsα-mutated skeletal progeni-
histologic diagnosis. The presence of a fibroosseous tor cells. J Clin Invest 101(8):1737–1744, 1998.
lesion typical of fibrous dysplasia adjacent to the cartilage 7. Bohay RN, Daley T: Osteosarcoma and fibrous dysplasia: radio-
graphic features in the differential diagnosis: a case report. J Can
islands is the most important diagnostic feature that dif- Dent Assoc 59(11):931–934, 1993.
ferentiates this entity from other cartilaginous neoplasms 8. Bousson V, Rey-Jouvin C, Laredo JD, et al: Fibrous dysplasia and
such as enchondroma and chondrosarcoma. McCune-Albright syndrome: imaging for positive and differential
The cartilage islands become calcified peripherally and diagnoses, prognosis, and follow-up guidelines. Eur J Radiol 83:
1828–1842, 2014.
show enchondral ossification merging with the surround- 9. Campanacci M: Bone and soft tissue tumors, Vienna, 1990, Springer.
ing fibroosseous lesion. Such prominent enchondral ossi- 10. Candeliere GA, Glorieux FH, Prud’homme J, et al: Increased
fication is not usually found in conventional chondroid expression of the c-fos proto-oncogene in bone from patients with
neoplasms. This is another characteristic finding of fibro- fibrous dysplasia. N Engl J Med 332(23):1546–1551, 1995.
cartilaginous dysplasia and is helpful in distinguishing 11. Candeliere GA, Roughley PJ, Glorieux FH: Polymerase chain
reaction-based technique for the selective enrichment and analysis
these lesions from true cartilage neoplasms. of mosaic arg201 mutations in G-α-s from patients with fibrous
In addition to enchondroma and conventional chon- dysplasia of bone. Bone 21:201–206, 1997.
drosarcoma, the differential diagnosis also includes dedif- 12. Carter JM, Inwards CY, Jin L, et al: Activating GNAS mutations
ferentiated chondrosarcoma. Fibrous areas within fibrous in parosteal osteosarcoma. Am J Surg Pathol 38:402–409, 2014.
13. Casey PJ: Lipid modifications of G proteins. Curr Opin Cell Biol
dysplasia sometimes show high cellularity. In such cases, 6:219–225, 1994.
dedifferentiated chondrosarcoma must be considered. 14. Casselman JW, De Jonge I, Neyt L, et al: MRI in craniofacial
However, careful microscopic examination of this tumor fibrous dysplasia. Neuroradiology 35(3):234–237, 1993.
reveals malignant cartilage and anaplastic dedifferentiated 15. Clementi E, Malgaretti N, Meldolesi J, et al: A new constitutively
sarcomatous elements. It is not difficult to distinguish activating mutation of the Gs protein α subunit-gsp oncogene
is found in human pituitary tumours. Oncogene 5:1059–1061, 1990.
dedifferentiated chondrosarcoma from fibrocartilaginous 16. Cohen MM: Fibrous dysplasia is a neoplasm. Am J Med Genet
dysplasia when the radiologic features are considered. 98(4):290–293, 2001.
Furthermore, the absence of nuclear anaplasia in the 17. Collins MT, Singer FR, Eugster E: McCune-Albright syndrome
spindle-cell elements is crucial in recognizing fibrocar- and the extraskeletal manifestations of fibrous dysplasia. Orphanet
J Rare Dis 7(Suppl 1):S4, 2012.
tilaginous dysplasia. 18. Collins MT, Chebli C, Jones J, et al: Renal phosphate wasting in
fibrous dysplasia of bone is part of a generalized renal tubular
dysfunction similar to that seen in tumor-induced osteomalacia.
Clinical Behavior J Bone Miner Res 16:806–813, 2001.
Foci of well-developed hyaline cartilage nodules are 19. Dal Cin P, Sciot R, Brys P, et al: Recurrent chromosome aberra-
tions in fibrous dysplasia of the bone: a report of the CHAMP
present in a small percentage of fibrous dysplasia and they study group. Cancer Genet Cytogenet 122:30–32, 2000.
vary from small microscopic foci to large, grossly evident 20. Danon M, Robboy SJ, Kim S, et al: Cushing syndrome, sexual
masses. The appellation of fibrocartilaginous dysplasia is precocity, and polyostotic fibrous dysplasia (Albright syndrome)
used for those cases in which the cartilage is abandoned in infancy. J Pediatr 87(6 Pt 1):917–921, 1975.
21. DePalma A, Ahmad I: Fibrous dysplasia associated with shepherd’s
and may overshadow the underlying fibroosseous process. crook deformity of humerus. Clin Orthop 97:38–39, 1973.
The clinical experience with this lesion is extremely 22. DeSmet AA, Travers H, Neff JR: Chondrosarcoma occurring in
limited but it is evident that foci of abundant cartilage a patient with polyostotic fibrous dysplasia. Skeletal Radiol 7:197–
can develop in polyostotic fibrous dysplasia and may 201, 1981.
affect only one or several affected sites. The clinical 23. Dorfman HD, Ishida T, Tsuneyoshi M: Exophytic variant of
fibrous dysplasia (fibrous dysplasia protuberans). Hum Pathol
symptomatology indicates an indolent protracted disease. 25(11):1234–1237, 1994.
The single case observations, based on many years of 24. Dumitrescu CE, Collins MT: McCune-Albright syndrome.
follow-up, indicate benign but continuous growth that Orphanet J Rare Dis 3:12, 2008.
may cause considerable disfiguring deformation necessi- 25. Eguchi K, Ishi S, Sugiura H, et al: Angiosarcoma of the chest wall
in a patient with fibrous dysplasia. Eur J Cardiothorac Surg 22(4):
tating ablative surgery.116 654–655, 2002.
26. Ehrig U, Wilson DR: Fibrous dysplasia of bone and primary
REFERENCES hyperparathyroidism. Ann Intern Med 77(2):234–238, 1972.
27. Fechner RE: Problematic lesions of the craniofacial bones. Am J
Fibrous Dysplasia Surg Pathol 13(Suppl 1):17–30, 1989.
1. Albright F, Butler A, Hampton A, et al: Syndrome characterized 28. Ferguson BJ: Fibrous dysplasia of the paranasal sinuses. Am J
by osteitis fibrosa disseminata, areas of pigmentation and endo- Otolaryngol 15(3):227–230, 1994.
crine dysfunction, with precocious puberty in females: report of 29. Fukuroku J, Kusuzaki K, Murata H, et al: Two cases of secondary
five cases. N Engl J Med 216:727–746, 1937. angiosarcoma arising from fibrous dysplasia. Anticancer Res 19(5C):
2. Alman BA, Greel D, Wolfe HJ: Activating mutations of Gs 4451–4457, 1999.
protein in monostotic fibrous lesions of bone. J Orthop Res 14:311– 30. Gaiddon C, Boutillier AL, Monnier D, et al: Genomic effects of
315, 1996. the putative oncogene G-α-s: chronic transcriptional activation of

ERRNVPHGLFRVRUJ
8  Fibrous Dysplasia and Related Lesions 615

the c-fos proto-oncogene in endocrine cells. J Biol Chem 269: 55. Michou L, Brown JP: Genetics of bone diseases: Paget’s disease,
22663–22671, 1994. fibrous dysplasia, osteopetrosis, and osteogenesis imperfecta. Joint
31. Gessl A, Freissmuth M, Czech T, et al: Growth hormone- Bone Spine Rev Rhumatisme 78:252–258, 2011.
prolactin-thyrotropin-secreting pituitary adenoma in atypical 56. Milgram JW: Malignant degeneration of polyostotic fibrous dys-
McCune-Albright syndrome with functionally normal Gsα protein. plasia of bone. Bull Hosp Joint Dis 36(2):137–149, 1975.
J Clin Endocrinol Metab 79(4):1128–1134, 1994. 57. Monge J, Kricun M, Radovcic J, et al: Fibrous dysplasia in a
32. Halawa M, Aziz AA: Chondrosarcoma in fibrous dysplasia of the 120,000+ year old Neanderthal from Krapina, Croatia. PLoS ONE
pelvis. A case report and review of the literature. J Bone Joint Surg 8:64539, 2013.
Br 66(5):760–764, 1984. 58. Nabarro MN, Giblin PE: Monostotic fibrous dysplasia of the
33. Henry A: Monostotic fibrous dysplasia. J Bone Joint Surg Br thoracic spine. Spine 19(4):463–465, 1994.
51(2):300–306, 1969. 59. O’Halloran DJ, Shalet SM: A family pedigree exhibiting features
34. Huvos AG, Higinbotham NL, Miller TR: Bone sarcomas arising of both multiple endocrine neoplasia type 1 and McCune-Albright
in fibrous dysplasia. J Bone Joint Surg Am 54(5):1047–1056, syndromes. J Clin Endocrinol Metab 78(3):523–525, 1994.
1972. 60. Okamoto S, Hisaoka M, Ushijima M, et al: Activating Gsα muta-
35. Idowu BD, Al-Adnani M, O’Donnell P, et al: A sensitive mutation- tion intramuscular myxomas with and without fibrous dysplasia of
specific screening technique for GNAS1 mutations in cases of bone. Virchows Arch 437:133–137, 2000.
fibrous dysplasia: the first report of a codon 227 mutation in bone. 61. Perkinson NG, Higinbotham NL: Osteogenic sarcoma arising in
Histopathology 50:691–704, 2007. polyostotic fibrous dysplasia: report of a case. Cancer 8(2):396–
36. Jaffe HL: Tumors and tumorous conditions of bones and joints, Phila- 402, 1955.
delphia, 1958, Lea & Febiger. 62. Phelan MC, Rogers RC, Clarkson KB, et al: Albright hereditary
37. Jan M, Dweik A, Destrieux C, et al: Fronto-orbital sphenoidal osteodystrophy and del(2) (q37.3) in four unrelated individuals.
fibrous dysplasia. Neurosurgery 34(3):544–547, 1994. Am J Med Genet 58(1):1–7, 1995.
38. Jhala DN, Eltoum I, Carroll AJ, et al: Osteosarcoma in a patient 63. Pollandt K, Engels C, Kaiser E, et al: Gsα gene mutations in
with McCune-Albright syndrome and Mazabraud’s syndrome: a monostotic fibrous dysplasia of bone and fibrous dysplasia-like
case report emphasizing the cytological and cytogenetic findings. low-grade central osteosarcoma. Virchows Arch 439:170–175,
Hum Pathol 34(12):1354–1357, 2003. 2001.
39. Kashima TG, Gamage NM, Ye H, et al: Locally aggressive fibrous 64. Prayson MA, Leeson MC: Soft-tissue myxomas and fibrous dys-
dysplasia. Virchows Arch 463:79–84, 2013. plasia of bone. A case report and review of the literature. Clin
40. Landis CA, Masters SB, Spada A, et al: GTPase inhibiting muta- Orthop 291:222–228, 1993.
tions activate the α chain of Gs, and stimulate adenylyl cyclase in 65. Pugh DG: Fibrous dysplasia of skull: a probable explanation for
human pituitary tumours. Nature 340:692–696, 1989. leontiasis ossea. Radiology 44:548–555, 1945.
41. Lichtenstein L: Polyostotic fibrous dysplasia. Arch Surg 36:874– 66. Reed RJ: Fibrous dysplasia of bone. A review of 25 cases. Arch
898, 1938. Pathol 75:480–495, 1963.
42. Lichtenstein L, Jaffe HL: Fibrous dysplasia of a bone: condition 67. Regard JB, Cherman N, Palmer D, et al: Wnt/β-catenin signaling
affecting one, several or many bones, graver cases of which may is differentially regulated by Gα proteins and contributes to
present abnormal pigmentation of skin, premature sexual develop- fibrous dysplasia. PNAS 108:20101–20106, 2011.
ment, hyperthyroidism or still other extraskeletal abnormalities. 68. Riddell DM: Malignant change in fibrous dysplasia; report of a
Arch Pathol 33:777, 1942. case. J Bone Joint Surg Br 46:251–255, 1964.
43. Lietman SA, Levine MA: Fibrous dysplasia. Pediatr Endocrinol Rev 69. Riddle ND, Bui MM: Fibrous dysplasia. Arch Pathol Lab Med
2:389–396, 2013. 137:134–138, 2013.
44. Machwate M, Jullienne A, Moukhtar M, et al: Temporal variation 70. Rieger E, Kofler R, Borkenstein M, et al: Melanotic macules fol-
of c-fos proto-oncogene expression during osteoblast differentia- lowing Blaschko’s lines in McCune-Albright syndrome. Br J Der-
tion, and osteogenesis in developing rat bone. J Cell Biochem matol 130(2):215–220, 1994.
57:62–70, 1995. 71. Riminucci M, Fisher LW, Shenker A, et al: Fibrous dysplasia of
45. MacMahon HE: Albright’s syndrome—30 years later. (polyostotic bone in the McCune-Albright syndrome: abnormalities in bone
fibrous dysplasia). Pathol Annu 6:81–146, 1971. formation. Am J Pathol 151:1587–1600, 1997.
46. Madaro A: Polyostotic fibrous dysplasia. Arch Radiol 30:303–319, 72. Riminucci M, Robey PG, Saggio I, et al: Skeletal progenitors and
1955. the GNAS gene: fibrous dysplasia of bone read through stem cells.
47. Maki M, Saitoh K, Horiuchi H, et al: Comparative study of J Mol Endocrinol 45:355–364, 2010.
fibrous dysplasia and osteofibrous dysplasia: histopathological, 73. Ruggieri P, Sim FH, Bond JR, et al: Malignancies in fibrous dys-
immunohistochemical, argyrophilic nucleolar organizer region plasia. Cancer 73(5):1411–1424, 1994.
and DNA ploidy analysis. Pathol Int 51(8):603–611, 2001. 74. Ruggieri P, Sim FH, Bond HR, et al: Osteosarcoma in a patient
48. Malchoff CD, Reardon G, MacGillivray DC, et al: An unusual with polyostotic fibrous dysplasia and Albright’s syndrome. Ortho-
presentation of McCune-Albright syndrome confirmed by an pedics 18(1):71–75, 1995.
activating mutation of the Gsα subunit from a bone lesion. J Clin 75. Saggio I, Remoli C, Spica E, et al: Constitutive expression of Gsα
Endocrinol Metab 78:803–806, 1994. (R201C) in mice produces a heritable, direct replica of human
49. Mangion J, Edkins S, Goss AN, et al: Familial craniofacial fibrous fibrous dysplasia bone pathology and demonstrates its natural
dysplasia: absence of linkage to GNAS1 and the gene for history. J Bone Miner Res 29:2357–2368, 2014.
cherubism. J Med Genet 37(11):E37, 2000. 76. Sakamoto A, Oda Y, Iwamoto Y, et al: A comparative study of
50. Marie PJ: Cellular and molecular basis of fibrous dysplasia. Histol fibrous dysplasia and osteofibrous dysplasia with regard to expres-
Histopathol 16:981–988, 2001. sions of c-fos and c-jun products and bone matrix proteins: a clini-
51. Marie PJ, de Pollak C, Chanson P, et al: Increased proliferation copathological review and immunohistochemical study of c-fos,
of osteoblastic cells expressing the activating Gsα mutation in c-jun, type I collagen, osteonectin, osteopontin and osteocalcin.
monostotic and polyostotic fibrous dysplasia. Am J Pathol 150: Human Pathol 30:1418–1426, 1999.
1059–1069, 1997. 77. Sakamoto A, Oda Y, Iwamoto Y, et al: A comparative study of
52. Mazabraud A, Semat P, Roze R: Apropos of the association of fibrous dysplasia and osteofibrous dysplasia with regard to Gsα
fibromyxomas of the soft tissues with fibrous dysplasia of the mutation at the Arg201 codon. J Mol Diagn 2(2):67–72, 2000.
bones. Presse Med 75(44):2223–2228, 1967. 78. Salpea P, Stratakis CA: Carney complex and McCune Albright
53. McCune DJ, Bruch H: Osteodystrophia fibrosa: report of case in syndrome: an overview of clinical manifestations and human
which condition was combined with precocious puberty, patho- molecular genetics. Mol Cell Endocrinol 386:85–91, 2014.
logic pigmentation of skin, and hyperparathyroidism. Am J Dis 79. Sanerkin NG, Watt I: Enchondromata with annular calcification
Child 54:806–848, 1937. in association with fibrous dysplasia. Br J Radiol 54(648):1027–
54. Mertens F, Albert A, Heim S, et al: Clonal structural chromosome 1033, 1981.
aberrations in fibrous dysplasia. Gene Chromosome Cancer 11(4): 80. Schlumberger HF: Fibrous dysplasia of single bones. Mil Surg
271–272, 1994. 99:504–527, 1946.

ERRNVPHGLFRVRUJ
616 8  Fibrous Dysplasia and Related Lesions

81. Schnabel P, Bohm M: Mutations of signal-transducing G proteins 100. Walther I, Walther BM, Chen Y, et al: Analysis of GNAS1 muta-
in human disease. J Mol Med 73(5):221–228, 1995. tions in myxoid soft tissue and bone tumors. Pathol Res Pract
82. Schneider LC, Dolinsky HB, Grodjesk JE, et al: Malignant 210:1–4, 2014.
spindle cell tumor arising in the mandible of a patient with florid 101. Weinstein LS, Shenker A, Gejman PV, et al: Activating mutations
osseous dysplasia. Oral Surg Oral Med Oral Pathol Oral Radiol of the stimulatory G protein in the McCune-Albright syndrome.
Endod 88(1):69–73, 1999. N Engl J Med 325:1688–1695, 1991.
83. Schwartz DT, Alpert M: The malignant transformation of fibrous 102. West R, Huvos AG, Lane JM: Desmoplastic fibroma of bone
dysplasia. Am J Med Sci 247:1–20, 1964. arising in fibrous dysplasia. Am J Clin Pathol 79(5):630–633, 1983.
84. Schwindinger WF, Francomano CA, Levine MA: Identification of 103. Wilson LC, Oude Luttikhuis ME, Clayton PT, et al: Parental
a mutation in the gene encoding the alpha subunit of the stimula- origin of Gsα gene mutations in Albright’s hereditary osteodystro-
tory G protein of adenylyl cyclase in McCune-Albright syndrome. phy. J Med Genet 31(11):835–839, 1994.
Proc Natl Acad Sci 89:5152–5156, 1992. 104. Wojno KJ, McCarthy EF: Fibro-osseous lesions of the face and
85. Shenker A, Weinstein LS, Moran A, et al: Severe endocrine and skull with aneurysmal bone cyst formation. Skeletal Radiol 23(1):
nonendocrine manifestations of the McCune-Albright syndrome 15–18, 1994.
associated with activating mutations of stimulatory G protein GS. 105. Yabut SM, Kenan S: Malignant transformation of fibrous dyspla-
J Pediatr 123(4):509–518, 1993. sia. A case report and review of the literature. Clin Orthop 228:
86. Shenker A, Weinstein LS, Sweet DE, et al: An activating Gsα 281–289, 1988.
mutation is present in fibrous dysplasia of bone in the McCune- 106. Yuen VH, Jordan DR, Jabi M, et al: Aneurysmal bone cyst associ-
Albright syndrome. J Clin Endocrinol Metab 79(3):750–755, ated with fibrous dysplasia. Ophthal Plast Reconstr Surg 18(6):471–
1994. 474, 2002.
87. Sherman NH, Rao VM, Brennan RE, et al: Fibrous dysplasia 107. Lambright DG, Sondek J, Bohm A, et al: The 2.0 A crystal struc-
of the facial bones and mandible. Skeletal Radiol 8(2):141–143, ture of a heterotrimeric G protein. Nature 379:311–319, 1996.
1982.
88. Shi RR, Li XF, Zhang R, et al: GNAS mutational analysis in dif- Fibrocartilaginous Dysplasia
ferentiating fibrous dysplasia and ossifying fibroma of the jaw. Mod 108. Bulychova IV, Unni KK, Bertoni F, et al: Fibrocartilagenous mes-
Pathol 26:1023–1031, 2013. enchymoma of bone. Am J Surg Pathol 17(8):830–836, 1993.
89. Sissons HA, Steiner GC, Dorfman HD: Calcified spherules in 109. Choi IH, Kim CJ, Cho TJ, et al: Focal fibrocartilaginous dysplasia
fibro-osseous lesions of bone. Arch Pathol Lab Med 117(3):284– of long bones: report of eight additional cases and literature
290, 1993. review. J Pediatr Orthop 20(4):421–427, 2000.
90. Slow IN, Friedman EW: Osteogenic sarcoma arising in a preexist- 110. Cockshott WP, Martin R, Friedman L, et al: Focal fibrocartilagi-
ing fibrous dysplasia: report of case. J Oral Surg 29(2):126–129, nous dysplasia and tibia vara: a case report. Skeletal Radiol 23(5):
1971. 333–335, 1994.
91. Stewart MJ, Gilmer WS, Edmonson AS: Fibrous dysplasia of 111. Dahlin DC, Bertoni F, Beabout JW, et al: Fibrocartilaginous mes-
bone. J Bone Joint Surg Br 44-B:302–318, 1962. enchymoma with low-grade malignancy. Skeletal Radiol 12(4):263–
92. Su L, Weathers DR, Waldron CA: Distinguishing features of focal 269, 1984.
cemento-osseous dysplasias and cemento-ossifying fibromas: I. A 112. Drolshagen LF, Reynolds WA, Marcus NW: Fibrocartilaginous
pathologic spectrum of 316 cases. Oral Surg Oral Med Oral Pathol dysplasia of bone. Radiology 156(1):32, 1985.
Oral Radiol Endod 84(3):301–309, 1997. 113. Gedikoglu G, Aksoy MC, Ruaccan S: Fibrocartilaginous mesech-
93. Sun TT, Tao XF, Shi HM: Spontaneous osteosarcoma in cranio- moma of the distal femur: case report and literature review. Pathol
maxillofacial fibrous dysplasia: clinical and computed tomographic Int 51:638–642, 2001.
features in 8 cases. Oral Surg Oral Med Oral Pathol Oral Radiol 114. Gibson JNA, Reid R, McMaster MJ: Fibrocartilaginous mesen-
118:24–31, 2014. chymoma of the fifth lumbar vertebra treated by vertebrectomy.
94. Szendroi M, Rahoty P, Antal I, et al: Fibrous dysplasia associated Spine 19:1992–1997, 1994.
with intramuscular myxoma (Mazabraud’s syndrome): a long-term 115. Ishida T, Dorfman HD: Massive chondroid differentiation in
follow-up of three cases. J Cancer Res Clin Oncol 124(7):401–406, fibrous dysplasia of bone (fibrocartilaginous dysplasia). Am J Surg
1998. Pathol 17(9):924–930, 1993.
95. Taconis WK: Osteosarcoma in fibrous dysplasia. Skeletal Radiol 116. Kyriakos M, McDonald DJ, Sundaram M: Fibrous dysplasia with
17(3):163–170, 1988. cartilaginous differentiation (“fibrocartilaginous dysplasia”): a
96. Tresserra F, Rami R, Salas A, et al: Polyostotic coexisting fibrous review, with an illustrative case followed for 18 years. Skeletal
dysplasia and aneurysmal bone cyst of the chest wall. Thorac Car- Radiol 33:51–62, 2004.
diovasc Surg 41(5):321–324, 1993. 117. Pelzmann KS, Nagel DZ, Salyer WR: Case report 114. Skeletal
97. Tsuyuguchi N, Ohata K, Morino M, et al: Magnetic resonance Radiol 5(2):116–118, 1980.
imaging and (11C)methyl-l-methionine positron emission to- 118. Sumner TE, Ward WG, Kilpatrick SE, et al: Fibrocartilaginous
mography of fibrous dysplasia—two case reports. Neurol Med Chir mesenchymoma of bone: case report and review of the literature.
42(8):341–345, 2002. Pediatr Radiol 30:315–317, 2000.
98. Valls J, Schajowicz F: Fibrous dysplasia of bone. Rev Orthop Trau- 119. Telford ED: A case of osteitis fibrosa (with formation of hyaline
matol 20:83–119, 1950. cartilage). Br J Surg 18:409–414, 1930.
99. Voytek TM, Ro JY, Edeiken J, et al: Fibrous dysplasia and 120. Vargas-Gonzalez R, Sanchez-Sosa S: Fibrocartilaginous dysplasia
cemento-ossifying fibroma. A histologic spectrum. Am J Surg (fibrous dysplasia with extensive cartilaginous differentiation).
Pathol 19(7):775–781, 1995. Pathol Oncol Res 12:111–114, 2006.

ERRNVPHGLFRVRUJ
C H A P T E R 9 

Fibrous and
Fibrohistiocytic Lesions
CHAPTER OUTLINE

NONOSSIFYING FIBROMA (FIBROUS SKELETAL FIBROMATOSES


CORTICAL DEFECT) Desmoplastic Fibroma
Multifocal Nonossifying Fibroma Associated Juvenile Multifocal Myofibromatosis
with Clinical Syndromes Congenital Fibromatosis–like Fibrosarcoma
BENIGN FIBROUS HISTIOCYTOMA (Infantile Fibrosarcoma)

FIBROMYXOMA MALIGNANT FIBROUS HISTIOCYTOMA


ANGIOMATOID FIBROUS HISTIOCYTOMA FIBROSARCOMA

NONOSSIFYING FIBROMA involve the diaphysis. Its long axis is parallel to the
long axis of the affected bone. The radiographic findings
(FIBROUS CORTICAL DEFECT) are specific, and the diagnosis can be made with certainty
Definition if the lesion is present in a typical skeletal site and appro-
priate age group. On this basis, such a lesion can be
The terms nonossifying fibroma (nonosteogenic fibroma), observed untreated. The rarity of both fibrous cortical
fibrous cortical defect, and metaphyseal fibrous defect have defects and nonossifying fibroma on radiographs of
been applied to the same metaphyseal fibrohistiocytic adults is indirect evidence of a natural history character-
process. The location of the lesion in the growing por- ized by spontaneous regression of most of these lesions
tions of long tubular bones of skeletally immature patients over time.
and its frequent spontaneous resolution indicate that it
represents a self-limiting process most likely related to Clinical Symptoms
incomplete ossification.
Most nonossifying fibromas are asymptomatic. The
lesion is usually incidentally identified on radiographs
Incidence and Location
obtained for other reasons such as trauma. Larger lesions
The peak age incidence and the most frequent sites of can cause pain or expansion of the affected area. In
skeletal involvement are shown in Figure 9-1. Radio- some instances, pathologic fracture can be a presenting
graphic evidence of fibrous cortical defect of long tubular symptom.3,10,29 Superimposed aneurysmal bone cyst with
bones can be found in approximately 30% of young indi- the development of an expansive, rapidly enlarging mass
viduals with unfused growth plates.7,16,17,34,41 The majority can also call attention to the lesion.
of lesions are asymptomatic and are incidental radio-
graphic findings. A small percentage of these lesions can Radiographic Imaging
produce larger lytic defects that can become symptom-
atic. The larger ones are prone to pathologic fracture Metaphyseal fibrous defect produces an eccentric cortical-
and may require prophylactic curettage.3,10,29 The peak based lytic lesion with distinct and frequently scalloped,
incidence is in the second decade of life. The distal sclerotic margins (Figs. 9-2 to 9-6).4,7,13,16,20,32 The cortex
femoral metaphysis is the most common site. The second overlying the lesion is usually minimally to moderately
most frequent site is the proximal tibial metaphysis expanded and markedly thinned but intact (Figs. 9-2 to
followed by the distal tibial metaphysis. Nonossifying 9-5). There is no matrix mineralization, but in older
fibroma occurs less commonly in the upper extremity, patients evidence of bone sclerosis can accompany healing
where it may be found in the proximal humerus and of the lesion. The distance from the growth plate usually
distal radius. Occasionally the lesions may be multifocal, increases with age and in older adolescents.30,39 The
simultaneously involving several symmetric metaphyseal lesion appears to migrate in a shaftward direction as a
sites, most frequently in the long bones of the lower result of skeletal growth. The lesions typically are
extremity.27 The lesion is metaphyseal but can also lytic. Incomplete trabeculation is frequently seen and is
617
ERRNVPHGLFRVRUJ
618 9  Fibrous and Fibrohistiocytic Lesions

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 9-1  ■  Nonossifying fibroma. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites of involve-
ment are indicated by black solid arrows.

accentuated in the peripheral parts of the defect (Figs. Microscopic Findings


9-3 and 9-4). Older lesions may show prominent mar-
ginal or intralesional sclerosis (Figs. 9-4 and 9-5). In The nonossifying fibroma is composed of fibroblastic,
smaller bones, such as the fibula, an eccentric location is usually highly cellular, stromal tissue with a prominent
less common, and the lesion may appear as a central, storiform pattern (Fig. 9-9). In this stroma, there are a
expansile trabeculated lytic defect (Fig. 9-2). Pathologic few multinucleated giant cells and foci of xanthomatous
fracture is a frequent presenting sign (Fig. 9-7). Com- reaction with foamy histiocytes present (Figs. 9-9 to 9-11
puted tomography (CT), magnetic resonance imaging and 9-16). Usually, there is a substantial amount of hemo-
(MRI), or both modalities can precisely document the siderin pigment clearly visible in the stromal cells (Fig.
relationship between the lesion, the medullary cavity, the 9-16). Minimal numbers of mononuclear inflammatory
cortex, and the periosteum (Figs. 9-4 to 9-6).15,18,31 These cells may be present. Fresh hemorrhage and cystic change
imaging techniques show that the lesion represents an similar to that seen in aneurysmal bone cyst may be
eccentric defect delineated from the medullary cavity by present. Secondary aneurysmal bone cyst formation can
a rim of sclerotic bone and covered by a thinned, intact lead to rapid enlargement and a “blowout” expansion into
cortical bone and periosteum (see Figs. 9-4 to 9-6). the soft tissue. Occasionally multinucleated giant cells
dominate, but they are usually focally distributed (Fig.
Gross Findings 9-14). With low power magnification, it is possible to
observe the localization of prominent giant cell reaction
When evaluated intact in its setting, nonossifying fibroma to areas of hemorrhage. Ghosts of giant cells and patterns
is an eccentric and cortically based lesion with an inner demonstrating the storiform architecture of the lesion
boundary that is scalloped and distinctly demarcated. may be perceived in areas of necrosis. Necrosis, as well
The cortex can be markedly thinned and expanded but as the presence of occasional normal mitotic figures, are
intact. The tissue is fibrous, fleshy, and yellow or tan- not worrisome features in otherwise typical nonossifying
brown (Fig. 9-8). There may be evidence of cystic change, fibroma and should not be considered as microscopic fea-
hemorrhage, or extensive necrosis, particularly in asso- tures of malignancy. Atypical mitoses are absent. Rarely
ciation with pathologic fracture of larger lesions. Text continued on p. 629

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 619

A B

C D
FIGURE 9-2  ■  Nonossifying fibroma: radiographic features. A and B, Anteroposterior (AP) radiographs showing two examples of
nonossifying fibroma of distal radial metaphysis in adolescents. Thinning of slightly expanded overlying cortex is present in both.
C, AP radiograph showing slightly expansile, well-marginated lytic lesion of proximal fibular shaft in teenage patient. D, AP radio-
graph of distal metaphyseal lesion with trabeculated pattern, expanded bone contour, and sharp demarcation. In fibula, nonossifying
fibroma often involves the entire medullary cavity and does not show eccentric features.

ERRNVPHGLFRVRUJ
620 9  Fibrous and Fibrohistiocytic Lesions

A B

C D

FIGURE 9-3  ■  Nonossifying fibroma: radiographic features. A and B, Anteroposterior (AP) radiographs showing large bilateral, eccen-
tric, lucent lesions in proximal tibial metaphyses of a 12-year-old girl. Scalloped borders and incomplete trabeculation are typical
of nonossifying fibroma. Note expanded and scalloped but intact cortex. C and D, AP and lateral radiographs of ankle region show
large, well-demarcated lucent lesion in distal tibial diametaphysis. Note slightly expansile contour and narrow zone of transition to
normal bone.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 621

A B

C D

FIGURE 9-4  ■  Nonossifying fibroma: radiographic features. A and B, Anteroposterior (AP) and lateral radiographs showing an eccen-
tric lesion in the tibial metaphysis with scalloped sclerotic margins and coarse trabeculation characteristic of nonossifying fibroma.
C, AP radiograph of a large nonossifying fibroma in the distal femoral metaphysis. Note sclerotic margins and an expanded cortex
overlaying the lesion. D, Coronal magnetic resonance image (MRI) showing an eccentric well demarcated lesion in the distal femoral
metaphysis. Same case as shown in C. Inset, Axial CT scan showing an eccentric lytic lesion with scalloped well demarcated margins.

ERRNVPHGLFRVRUJ
622 9  Fibrous and Fibrohistiocytic Lesions

A C

D E
FIGURE 9-5  ■  Nonossifying fibroma: radiographic features. A, Anteroposterior radiograph showing an eccentric lesion in the distal
tibial diaphysis with sclerotic margins and expanded overlying cortex. B and C, Axial computed tomography (CT) scans showing
eccentric lesion of the tibia. Note well demarcated sclerotic medullary margin. D and E, Sagittal and coronal T1-weighted magnetic
resonance images showing low density eccentric lesion of distal tibial metaphysis with well demarcated sclerotic scalloped margins.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 623

A B

D E
FIGURE 9-6  ■  Nonossifying fibroma: radiographic and microscopic features. A, Anteroposterior radiograph showing an eccentric lytic
lesion of the proximal tibial metaphysis with fine trabeculations and expanded overlying cortex. B, Fat-saturated T2-weighted coronal
magnetic resonance image (MRI) of the lesion shown in A shows signal enhancement in an eccentric tibial metaphyseal lesion.
C and D, Axial fat-saturated T2-weighted and T-weighted MRIs showing an eccentric well demarcated lesion involving the proximal
tibia. E, Low power microphotograph of nonossifying fibroma showing fibrohistiocytic proliferation with storiform pattern (E, ×100)
(E, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
624 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-7  ■  Nonossifying fibroma: radiographic features. A and B, Nonossifying fibroma in distal shaft of radius with pathologic
fracture shown in anteroposterior and lateral views of forearm. C and D, Fracture healing with normal callus formation several
months later in patient shown in A and B.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 625

A B

C D
FIGURE 9-8  ■  Nonossifying fibroma: gross features. A, Bisected fibular segment shows two separate cortically oriented lesions, one
containing abundant lipid and the other with extensive hemosiderin deposits. B, Whole-mount section of fibular shaft shows cortical
orientation of nonossifying fibroma and circumscription by a narrow zone of sclerotic bone on medullary aspect. C, Brown, gelati-
nous tissue excised from proximal tibial metaphysis reflects high content of hemosiderin pigment. D, Segmental resection of fibula
contains fibrous lesion with focal brown discoloration in medullary cavity.

ERRNVPHGLFRVRUJ
626 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-9  ■  Nonossifying fibroma: microscopic features. A and B, Low and intermediate power photomicrographs show bundles
of spindle cells with rare interspersed multinucleated giant cells. C and D, Low and intermediate power photomicrographs showing
spindle cell proliferations with interspersed multinucleated giant cells. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 627

A B

C D
FIGURE 9-10  ■  Nonossifying fibroma: microscopic features. A-D, Low and intermediate power photomicrographs showing fibrohis-
ticytic proliferation with storiform pattern and rare interspersed multinucleated giant cells and histiocytes. (A and C, ×100; B and
D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
628 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-11  ■  Nonossifying fibroma: microscopic features. A and B, Low and intermediate power photomicrographs showing a
proliferation of histiocytic cells with occasional vacuolated foamy cytoplasm and prominent multinucleated giant cells. C and
D, Low and intermediate power photomicrographs showing spindle cells growing in a storiform pattern showing scattered multi-
nucleated giant cells. Note an angulated cytoplasmic outline of multinucleated giant cells and their somewhat pyknotic nuclei, a
frequent feature of giant cells in nonossifying fibroma. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 629

nonossifying fibroma can be markedly hypercellular and cell elements or storiform pattern of nonossifying
is composed of plump fibrohistiocytic cells with enlarged fibroma.
nuclei (Figs. 9-12 to 9-14). Such lesions may raise sus- Benign fibrous histiocytoma is a term applied to lesions
picion of malignancy but the true cytologic atypia and that are histologically identical with nonossifying fibroma
atypical mitosis are absent. In addition, correlation with but that occur in unusual locations such as the flat bones,
radiographic features showing typical radiographic pre- ribs, or vertebrae.
sentation of nonossifying fibroma with the absence of
imaging features indicating an aggressive growth pattern,
Treatment and Behavior
confirm the benign nature of such lesions. Focal myxoid
change as well as areas of reactive bone formations As stated, nonossifying fibromas are frequently asymp-
can be present (Fig. 9-15). In summary, the lesion has tomatic, self-healing lesions that do not require treat-
microscopic features that overlap with those of the fibro- ment. The larger lesions that expand the bone, develop
histiocytic reactive lesions commonly seen in extraskel- secondary aneurysmal bone cysts, or pose a danger of
etal sites and with the reactive changes superimposed fracture are treated by curettage and bone grafting.35,40,42
on various preexisting skeletal conditions.15 The best Most lesions stabilize at puberty and show signs of healing
example of this is giant cell tumor of bone, which may by peripheral sclerosis or solid opacification.7,30,32 In rare
contain a substantial component of reactive fibrohistio- instances nonossifying fibroma may continue to grow and
cytic changes resembling nonossifying fibroma or benign show considerable increase in size over time.42
fibrous histiocytoma. As a consequence, nonossifying In one study of 23 cases of pathologic fractures through
fibroma can be confused with many other conditions if nonossifying fibromas, it was found that the distal tibia
its microscopic appearance is evaluated without clinico- was most frequently involved. Lesions that occupied
radiologic correlation.16,17 more than 50% of the transverse diameter of the involved
bone were likely to fracture. Pathologic fractures through
these lesions can be treated by cast bracing until the
Special Techniques
fracture heals; then curettage with or without bone graft-
Ultrastructural studies suggest the histiocytic rather than ing can be done.3,10,11
fibroblastic origin of nonossifying fibroma.14,16,36 This Cases in which a malignant tumor was present inde-
observation is consistent with the strong positivity pendently in a bone that also contained a nonossifying
of nonossifying fibroma for CD68. Cytogenetically, fibroma have been reported.19,22 The malignancies have
nonossifying fibromas have a near-diploid chromosomal all been osteosarcomas.
complement.39 Several clonal chromosomal aberrations, Several rare cases of apparent malignant transforma-
including translocation t(1;4)(p31;q34), suggest the true tion of nonossifying fibroma have been described.5,25,38 In
neoplastic nature of at least a subset of nonossifying all of these cases, the original radiographic presentation
fibromas.6,28,39 of the lesion was not characteristic of nonossifying
fibroma. Bone production and periosteal reaction were
present in the first instance and loss of anterior cortex in
Differential Diagnosis
the second. In the third example, the lesion involved the
The presence of multinucleated giant cells in a spindle- mandible, which is a very unlikely location for nonossify-
cell background without the formation of bone or carti- ing fibroma. This raises the possibility that the original
lage matrix may raise the question of giant cell tumor in lesion in all of those instances was most likely malignant
the microscopic differential diagnosis of nonossifying from the time of presentation. Furthermore, benign-
fibroma. The distinction is easily made on clinical and appearing, whorled fibrous tissue and foam cells, which
radiologic grounds because giant cell tumor occurs almost microscopically characterize nonossifying fibroma, can
exclusively in adult patients at the ends of long bones, not be seen focally in several bone malignancies.
in the metaphyses.
Fibrous dysplasia may be considered when small corti- Personal Comments
cal fractures occur in nonossifying fibroma and lead to
reactive bone formation. Biopsy samples from such areas Surprisingly, nonossifying fibromas are still occasionally
show parallel arrays of immature bone trabeculae with diagnosed histologically as giant cell tumors, even though
bordering osteoblasts. Although a storiform pattern is review of the clinical and radiologic features would
sometimes present in fibrous dysplasia, the multinucle- suffice to exclude this diagnosis when considering a
ated giant cells seen in nonossifying fibroma are not a metaphyseal lesion in a preadolescent patient. Radiologic
common feature of fibrous dysplasia. Furthermore, the correlation would also serve to explain the presence of
radiologic eccentricity of nonossifying fibroma is not reactive bone in curetted material when a recent or old
characteristic of fibrous dysplasia. pathologic fracture is present in the thin cortex of such a
Desmoplastic fibroma may be mimicked by the more lesion. Nonossifying fibromas produce eccentrically ori-
heavily collagenized areas of nonossifying fibromas ented lucencies, whereas a focus of fibrous dysplasia is
undergoing involutional changes. Eccentric radiolucent indicative of a central intramedullary lesion. Knowledge
lesions of the medial supracondylar side of the femur of the latter helps prevent the less frequent error of diag-
in the cortical irregularity syndrome can have overlapping nosing a nonossifying fibroma with reactive bone as
features with fibrous cortical defects, but this avulsive fibrous dysplasia.
lesion usually does not show the multinucleated giant Text continued on p. 634

ERRNVPHGLFRVRUJ
630 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-12  ■  Nonossifying fibroma: microscopic features. A-D, Low and intermediate power photomicrographs showing highly
cellular variant of nonossifying fibroma composed of plump mononuclear cells with somewhat hyperchromatic nuclei. Such features
can raise suspicion of malignancy. In such instances, correlation with a radiographic presentation of the lesion is helpful to avoid
misclassification of hypercellular variants of nonossifying fibroma. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 631

A B

C D
FIGURE 9-13  ■  Nonossifying fibroma: microscopic features. A and B, Low and intermediate power photomicrographs showing
hypercellular nonossifying fibroma. Inset shows details of plump mononuclear cells with hyperchomatic nuclei. C and D, Low
and intermediate power photomicrographs showing more typical presentation of nonossifying fibroma composed of fibro-
histiocytic proliferation with intracytoplasmic deposition of hemosiderin. (A and C, ×100; B and D, ×200; Inset, ×400).)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
632 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-14  ■  Nonossifying fibroma: microscopic features. A-D, Low and intermediate power photomicrographs showing
storiform fibrohistiocytic formations with prominent scattered multinucleated giant cells. (A and C, ×100; B and D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 633

A B

C D
FIGURE 9-15  ■  Nonossifying fibroma: microscopic features. A, Low power photomicrograph showing storiform fibrohistiocytic pro-
liferation. B, Low power photomicrograph showing irregular hypocellular areas with stromal myxoid change. C and D, Low and
intermediate power photomicrographs showing an area of reactive woven bone formation at the periphery of the nonossifying
fibroma. (A, B, and D, ×100; C, ×50.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
634 9  Fibrous and Fibrohistiocytic Lesions

Multifocal Nonossifying Fibroma the histologic picture, the diagnosis of xanthoma, xantho-
Associated with Clinical Syndromes fibroma, or fibrous xanthoma of bone may be suggested.
Microscopically, it is similar to the more common benign
Multifocal nonossifying fibromas can occur in several fibrous histiocytoma of soft tissue. In bone, it is micro-
extremely rare clinical settings. The first is familial mul- scopically indistinguishable from nonossifying fibroma or
tifocal nonossifying fibromas. Evans and Park12 reported from the prominent fibrohistiocytic reaction found in
three patients in the same family who had multiple non- some giant cell tumors. There is controversy about
ossifying fibromas in long bones with many associated whether this is a distinct benign bone neoplasm or merely
abnormalities. Patients with neurofibromatosis (von a reactive lesion superimposed on a preexisting condition
Recklinghausen’s disease) have a greater likelihood of that cannot be easily identified.
developing multifocal nonossifying fibromas in the typical
long bone sites.23,33 Moser et al.27 found that 5% of 900
Incidence and Location
patients with multifocal nonossifying fibromas had
underlying neurofibromatosis. In both of these clinical Benign fibrous histiocytoma is a very rare entity with
syndromes, the nonossifying fibromas are found in the strict radiologic and microscopic criteria.44-46,51 Lesions
usual sites (i.e., metaphyses of long bones), and they may with the clinical and radiographic features of nonossify-
be bilateral and symmetric. ing fibroma or giant cell tumor should be excluded from
Jaffe-Campanacci syndrome is an uncommon, congeni- the category of benign fibrous histiocytoma. Patient age
tal disorder in which multifocal nonossifying fibromas of has a wide range from 5 to 75 years, and there is no clear
bone are associated with café au lait pigmentation, various sex predilection. The ilium and ribs are the most fre-
nonskeletal anomalies, and mental retardation.1,2,8,21,24,26,37 quently involved sites. Individual cases have been reported
Originally, it was thought that neurofibromatosis is not a in flat bones, including the craniofacial bones, vertebrae,
component of this syndrome. More recent observations and sacrum.43,47,49,50,52-58,65,66 Occasionally, benign fibrous
indicate, however, that Jaffe-Campanacci syndrome may histiocytoma may present as a central intramedullary
represent a unique presentation of neurofibromatosis diaphyseal lesion of the long tubular bones.63 Examples
type 1 because some of these patients may have other of benign fibrous histiocytoma involving the acral skel-
stigmata of neurofibromatosis, such as cutaneous neuro- eton have also been described.60 The age distribution
fibromas.9 The nonossifying fibromas in Jaffe-Campanacci and typical sites of skeletal involvement are shown in
syndrome tend to be more pronounced in the metaphy- Figure 9-19.
seal areas of the long tubular bones, but they also form
separate diaphyseal lesions. Widespread, more or less Clinical Symptoms
symmetric involvement of the long tubular bones of the
extremities is typical in this syndrome (Figs. 9-17 and The most frequent symptom is pain, ranging in duration
9-18). In Jaffe-Campanacci syndrome, the trunk, axial from months to several years. Pathologic fracture is rarely
skeleton, and craniofacial bones are also typically involved. a presenting symptom.
Each individual lesion has the radiographic appearance
of a nonossifying fibroma, but occasionally each focus Radiographic Imaging
coalesces, especially in the metaphyseal parts, and forms
a larger trabeculated lesion. Pathologic fracture fre- On radiographs, benign fibrous histiocytoma appears as
quently occurs in this syndrome, especially in younger a purely lytic, sharply demarcated lesion. A sclerotic rim
patients. Similar to ordinary nonossifying fibroma, and fine trabeculations may be present (Figs. 9-20 and
skeletal lesions in Jaffe-Campanacci syndrome have a 9-21). The tumor can disrupt the cortex and expand into
tendency to stabilize and heal after puberty. The nonskel- soft tissue, but usually it is delineated by a thin rim of
etal anomalies in this syndrome are hypogonadism, newly formed bone. In the long bones, the diaphysis and
cryptorchidism, ocular anomalies, and cardiovascular metaphysis are usually involved. The reported cases
malformations, including mitral insufficiency and aortic located in the ends of long tubular bones are controver-
stenosis. In some cases, precocious puberty and kypho- sial because it is difficult, or perhaps even impossible, to
scoliosis have been reported. These patients appear to exclude a giant cell tumor with a prominent fibrohistio-
have a generalized defect of mesenchymal tissue. Rarely, cytic component.59
familial distribution has been reported. Nonossifying
fibromas in Jaffe-Campanacci syndrome have micro- Gross Findings
scopic features identical to those seen in ordinary nonos-
sifying fibromas. The lesion evaluated in situ represents an intramedul-
lary, well-demarcated lesion that is tan-gray with yellow
and brown areas related to the presence of lipids and
BENIGN FIBROUS HISTIOCYTOMA hemorrhage.
Definition
Microscopic Findings
Benign fibrous histiocytoma of bone is a spindle-cell lesion
composed of fibroblast-like cells with a storiform pattern. The spindle-shaped, fibroblast-like cells have elongated
Foamy histiocytes and multinucleated giant cells are plump nuclei and small prominent nucleoli.44,45,61,62 A sto-
usually present. When xanthomatous changes dominate riform pattern is typically present, at least focally. Foamy,

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 635

A B

C D
FIGURE 9-16  ■  Nonossifying fibroma: microscopic features. A and B, Intermediate power photomicrographs show fibrohistiocytic
lesion with storiform pattern. Note hemosiderin deposits in B. C, Multinucleated giant cells concentrated in area of fresh hemor-
rhage. D, Clusters of xanthoma cells (A-D, ×200) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
636 9  Fibrous and Fibrohistiocytic Lesions

A B
FIGURE 9-17  ■  Jaffe-Campanacci syndrome: radiographic features. A and B, Anteroposterior radiographs of bilateral, multifocal
nonossifying fibromas of humeri. (Courtesy Dr. M. Campanacci, Bologna, Italy.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 637

A B
FIGURE 9-18  ■  Jaffe-Campanacci syndrome: radiographic features. A and B, Multiple, bilateral, lower-extremity long bone nonossify-
ing fibromas in an 11-year-old boy with mental retardation, leg-length discrepancies, and skin pigmentation. (Courtesy Dr. M. Cam-
panacci, Bologna, Italy.)

ERRNVPHGLFRVRUJ
638 9  Fibrous and Fibrohistiocytic Lesions

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 9-19  ■  Benign fibrous histiocytoma. Age distribution and frequent sites of skeletal involvement. Most frequent sites of
involvement are indicated by solid black arrows.

lipid-filled macrophages are interspersed among spindle abundant lipids within histiocytes, and reactive bone for-
cells and occasionally form larger solid areas (Fig. 9-20). mation. These changes can be very extensive or only
Multinucleated giant cells are almost always present and focal. Such areas sampled in small biopsy fragments
are sparsely distributed among the other elements. Fea- may be interpreted as benign fibrous histiocytoma.
tures of fresh and old hemorrhage with hemosiderin The radiographic features of such cases are usually diag-
deposits can be found. Cystic changes can frequently be nostic for giant cell tumor of bone. Indeed, adequate
seen microscopically. Occasional mitotic figures can be sampling of a lytic tumor in the end of a long bone of an
present, but atypical mitoses are absent. Overall, the cel- adult patient that shows “benign fibrous histiocytoma”
lularity can be high with occasional hyperchromatic features usually reveals small areas of classic giant cell
nuclei, but true nuclear atypia is absent. Similar to nonos- tumor.
sifying fibroma, ultrastructural studies confirmed the his- The diagnosis of benign fibrous histiocytoma of bone
tiocytic nature of cells in benign fibrous histiocytoma, is made by exclusion of other entities (eosinophilic granu-
which are CD68 positive.62 loma, fibrous dysplasia, giant cell tumor, and giant cell
reparative granuloma) that may exhibit prominent fibro-
histiocytic reactions.
Differential Diagnosis
Benign fibrous histiocytoma is histologically indistin- Treatment and Behavior
guishable from nonossifying fibroma, differing only in its
clinical and radiographic features. In the ribs, pelvis, Benign fibrous histiocytoma should be treated by curet-
skull, scapula, and vertebral column, such lesions may tage and bone grafting. Lesions that can be resected (i.e.,
contain large amounts of lipids and may be referred to as rib, fibula) because of their anatomic site should be
xanthofibromas or xanthogranulomas of bone. Indeed, such treated by en bloc excision. Recurrences can occur in
lipid-filled lesions in ribs have been mistaken for local- the bone after curettage, but local aggressive behavior
ized lesions of Erdheim-Chester disease.48 and particularly distant metastasis indicate that the
Giant cell tumor of bone may be characterized by fibro- diagnosis of benign fibrous histiocytoma was inappropri-
histiocytic elements with a prominent storiform pattern, ate. These cases, in our opinion, were in all probability

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 639

A B

C D

FIGURE 9-20  ■  Benign fibrous histiocytoma (xanthofibroma) of bone: radiographic and microscopic features. A, Well-marginated,
radiolucent, asymptomatic lesion discovered on intravenous pyelogram of a 13-year-old girl. She had nephrectomy for a renal tumor
at age 8, and follow-up studies revealed this lesion, samples of which were taken at subsequent biopsy. B, Fibrohistiocytic prolifera-
tion with scattered multinucleated giant cells. C and D, Xanthomatous and spindle-cell features without cytologic atypia. Note the
overall similarity of the lesion to a nonossifying fibroma. (B, C, ×100; D, ×200.) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
640 9  Fibrous and Fibrohistiocytic Lesions

B C
FIGURE 9-21  ■  Benign fibrous histiocytoma: radiographic features. A, Oblique radiograph of foot of a 16-year-old boy with pathologic
fracture through well-demarcated lytic lesion in calcaneus. B and C, Axial computed tomograms show lytic lesion with sclerotic
borders and cortical infarction. Histologic features of benign fibrous histiocytoma closely resembling nonossifying fibroma were
present in curettage specimen.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 641

misdiagnosed examples of giant cell tumor or low-grade patient has local pain, tenderness, and expansion of bone
malignant fibrous histiocytoma of bone.64 contour. The characteristic and unique feature is the
frequent presence of general symptoms such as fever,
Personal Comments anemia, hypogammaglobulinemia, and weight loss. Local
lymphadenopathy was also present in two of our cases
This controversial entity is undoubtedly overdiagnosed that presented as primary bone lesions.
when the designation is applied to giant cell tumors con-
taining extensive areas of fibrohistiocytic reaction.59 A
Radiographic Imaging
thorough histologic search for foci of conventional giant
cell tumor usually reveals the true nature of the lesion, All lesions presented as expansile lytic lesions with eccen-
but sometimes all the available material is composed of tric cortical disruption and extension into soft tissue
fibrohistiocytic reaction. In such cases, the characteristic (Figs. 9-23 and 9-24). One lesion was originally diag-
radiographic features of a lytic lesion at the end of a long nosed as nonspecific inflammatory reaction and recurred
bone in a skeletally mature individual allow the inference 3 months after initial curettage. Compterized imaging
to be drawn that the lesion is a giant cell tumor with techniques can document the multilocular cystic nature
secondary changes. This is analogous to making a diag- of the lesion.90
nosis of giant cell tumor or any other bone lesion in
which secondary aneurysmal bone cyst formation is so Gross Findings
profuse that it obscures the precursor lesion.
It is best to reserve the term benign fibrous histiocytoma The curetting available for gross examination consisted
for lesions that are indistinguishable from nonossifying of irregular tan-gray and hemorrhagic fragments of soft
fibroma but are located in unusual skeletal sites or present tissue.
in older patients.
Microscopic Findings
FIBROMYXOMA The microscopic appearance of angiomatoid fibrous his-
tiocytoma of bone is similar to that described for soft
Fibromyxoma is a rare lesion that bears certain simi­ tissue lesions. The tumor is composed of solid areas of
larities to chondromyxoid fibroma, but the few cases undifferentiated mesenchymal cells with a histiocytoid
described in the literature occurred in older individu- appearance (Figs. 9-24 and 9-25). Multiple blood-filled
als.67-75 In addition, such lesions lack the lobular architec- cystic (telangiectatic) spaces are present within these areas
ture and the chondroid matrix that are characteristic of (Figs. 9-24 and 9-26). In fact, some lesions can be domi-
chondromyxoid fibroma. Radiographically, fibromyxoma nated by extensive multilocular cystic change that may
appears as a lytic defect with scalloped margins (Fig. obscure the details of the underlying lesion.82 The overall
9-22). Microscopically, fibromyxoma is uniformly myxoid microscopic appearance mimics aneurysmal bone cyst or
with low to intermediate cellularity (Fig. 9-22). Some telangiectatic osteosarcoma, and the lesion in bone must
investigators believe that these lesions represent variants be distinguished from these entities. In fact, one of our
of chondromyxoid fibroma. cases was initially classified as telangiectatic osteosar-
coma. In addition, the lesion can be confused with heman-
giopericytoma or glomus tumor of bone. Solid areas with
ANGIOMATOID FIBROUS histiocytoid cells with foci of inflammatory cells are typi-
HISTIOCYTOMA cally present (Fig. 9-25). Areas of old and fresh hemor-
rhage and focal myxoid and xanthomatous changes also
In 1979, Enzinger81 described a distinct tumor predomi- can be seen. The lesion is typically surrounded (especially
nantly occurring in the subcutaneous soft tissue of chil- in the area of extension into soft tissue) by a thick fibrous
dren and young adults that is composed of solid nests of capsule. Prominent lymphoplasmocytic infiltration can
histiocytoid cells with multiple blood-filled cystic spaces. cause confusion with chronic osteomyelitis (Fig. 9-25).
He proposed the term angiomatoid malignant fibrous his- Some lesions may focally show a whorllike arrangement
tiocytoma. This lesion has been renamed angiomatoid of cells and psammoma bodies similar to those commonly
fibrous histiocytoma to reflect the relative rarity of metas- seen in menigiomas. An unusual intraosseous angioma-
tasis and the overall favorable prognosis.78 The originally toid fibrous histiocytoma with sclerotic features mimick-
suggested fibrohistiocytic origin of the lesion is question- ing an osteosarcoma has also been described.88 Soft tissue
able.81,87,103 We have seen five examples of this lesion lesions may have extensive areas of pleomorphic cells, but
arising in bone. Additional single case report of angioma- such variants of angiomatoid fibrous histiocytoma have
toid fibrous histiocytoma involving bone have been dis- not been observed in bone.77,89,101
cussed in the literature.88,90,94
Special Techniques
Clinical Symptoms
Ultrastructurally, the histiocytoid cells of angiomatoid
Angiomatoid fibrous histiocytoma occurs primarily in fibrous histiocytoma represent undifferentiated mesen-
children and young adults. It is extremely rare in patients chymal cells.86,96 Immunohistochemically, they are positive
older than age 40 years. When it occurs in bone, the Text continued on p. 647

ERRNVPHGLFRVRUJ
642 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-22  ■  Fibromyxoma: radiographic and microscopic features. A, Anteroposterior (AP) radiograph of proximal femur showing
a lytic lesion of intertrochanteric area with scalloped sclerotic margin. Note similarity of this radiographic presentation with more
conventional chondromyxoid fibroma. B, AP radiograph showing a lytic, well demarcated defect of ilium with sclerotic margins and
trabeculations. Note similarity between this lesion and more conventional chondromyxoid fibroma at this site. C and D, Uniformly
myxoid lesion with low cellularity. Note absence of lobulated architecture with septa containing chondroblasts and giant cells seen
in typical chondromyxoid fibroma. (C and D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 643

A B
FIGURE 9-23  ■  Angiomatoid fibrous histiocytoma of bone: radiographic features. A and B, Internal rotation and anteroposterior views
of humerus of 8-year-old boy show expansile lytic lesion with blowout features and prominent periosteal reaction. There was associ-
ated axillary and supraclavicular lymphadenopathy, high fever, episodic nausea and vomiting, and a 6-pound weight loss in 6 weeks.
Lesion was initially treated by curettage with recurrences 3 and 9 months after original curettage. Finally, patient was treated with
proximal humeral resection.

ERRNVPHGLFRVRUJ
644 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-24  ■  Angiomatoid fibrous histiocytoma: radiographic and microscopic features. A, Axial computed tomogram of skull (bone
window) shows right-sided mandibular destruction and large associated soft tumor mass in a 7-year-old girl. There is no evidence
of matrix mineralization. B, Multicystic hemorrhage in tumor tissue producing pseudoangiomatous low power appearance.
C, Higher magnification shows pseudoangiomatoid architecture. D, High power photomicrograph shows bland nuclear features in
round-to-oval tumor cells and two foci of calcification. (B, ×50; C, ×100; D, ×400). (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 645

A B

C D
FIGURE 9-25  ■  Angiomatoid fibrous histiocytoma: microscopic features. A, Low power photomicrograph showing prominent band
of inflammatory cells at the periphery of the lesion. B, Intermediate power photomicrograph shows sheets of plump spindle and
histiocytoid cells. C and D, Proliferation of plump histiocytoid cells with concentric whorllike arrangement of cells. (A-C, ×100;
D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
646 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-26  ■  Angiomatoid fibrous histiocytoma: microscopic features. A and B, Low power photomicrographs showing multicystic
architecture in tumor tissue producing pseudoangiomatous appearance. C and D, Solid proliferations of indistinct plump histiocytoid
cells. (A and B, ×25; C and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 647

for vimentin and negative for histiocytic and endothelial and biologic potential occurring in different anatomic
markers.83,91,100 The presence of scattered CD68 (KP-1)– sites and occasionally involving the skeleton. The term
positive cells can be documented in approximately 50% fibromatosis covers a broad spectrum of fibroblastic pro-
of cases.96 They most likely represent true macrophages liferations that are intermediate in their biologic behavior
attracted to the lesion by hemorrhage. In some cases, the between benign and malignant lesions; that is, they exhibit
expression of desmin suggests muscle differentiation, but indolent local destructive growth but do not metastasize.
other muscle markers are typically negative.83,91 Angio- Soft tissue fibromatoses are subdivided into several major
matoid fibrous histiocytomas are diploid by cytometric groups based on their site and organ involvement. Their
measurements.80 In general, the immunohistochemi- description is beyond the scope of this book. Skeletal
cal profile is not specific. The hallmark of angiomatoid fibromatoses occur less frequently than their soft tissue
fibrous histiocytoma is the presence of three distinct counterparts and are found in several basic clinical
chromosomal translocations resulting in the presence of settings—as the solitary lesion referred to as desmoplastic
chimeric genes: EWSR1-ATF1 t(12;22)(q13;q12) (72%), fibroma6 and even more rarely seen forms designated
EWSR1-CREB1 t(2;22)(q34;q12) (21%), and FUS-ATF1 infantile multifocal skeletal myofibromatosis and congenital
t(12;22)(q13;q12) (7%).76,79,84,85,92,93,102 The first two trans- fibromatosis–like fibrosarcoma (infantile fibrosarcoma).
locations are the most frequent and involve the EWSR1
gene, a prototypic gene of the translocations in Ewing’s
sarcoma and several other soft tissue tumors. The EWSR1 Desmoplastic Fibroma
gene located on chromosome 22 is a 5′ fusion partner for Definition
the two main translocations, while the 3′ partners involve
most frequently ATF1, from chromosome 12, and less Desmoplastic fibroma is a rare, locally aggressive, solitary
frequently CREB1, from chromosome 2 (Fig. 9-27). In tumor microscopically composed of well-differentiated
the chimeric proteins, EWSR1 contributes primarily its myofibroblasts with abundant dense collagen deposition.
strong promoter and its TAD-transactivation N-terminal Microscopically, it is identical to soft tissue fibromatosis.
domain. Both of the 3′ fusion partners (i.e., ATF1 and Jaffe16 first used this term in 1958 to describe five cases
CREB1) provide leucine zipper C-terminal domains in of a previously unclassified central fibrous tumor histo-
the chimeric protein (Fig. 9-28). The less frequent trans- logically similar to desmoid tumor of the abdominal wall.
location involves the FUS gene on chromosome 16 and The term periosteal desmoid was initially introduced for
again ATF1 on chromosome 12 (Fig. 9-29). The FUS a process that produces a cortical defect in the postero-
gene is the 5′ fusion partner in this translocation while medial aspect of the distal femoral diaphysis at the inser-
the ATF1 gene is a 3′ fusion partner. In the chimeric tion of the tendon of the adductor magnus muscle. This
protein, the FUS gene provides the N-terminal QGSY- benign, avulsive, self-limited process is discussed in
rich domain while ATF1, similarly to the main two further detail in Chapter 23 in the section on cortical
translocations, contributes the leucine zipper C-terminal irregularity syndrome.
domain. These translocations are well documented in the
soft tissue variant of angiomatoid fibrous histiocytoma; Incidence and Location
however, their presence in the lesions affecting bone is
very preliminary. They represent potential therapeutic Desmoplastic fibroma is an extremely rare tumor
targets and are useful in differential diagnosis of angio- that accounts for less than 0.1% of all bone tumors,
matoid fibrous histiocytoma.97-99 and the vast majority of reports present single
cases.113,114,118,122,123,126,136,137,140 Reports documenting larger
series are rare and typically come from major reference
Treatment and Behavior
centers.105,109-111,117,125,133-135 The lesions have been reported
The soft tissue lesions have a potential for locally aggres- from age 20 months to the seventh decade of life, but
sive behavior but they are typically successfully managed typically they affect adolescents and young adults. There
by complete local excision, which results in the reversal appears to be no sex predilection. Any bone may be
of systemic symptoms. Resection of three of the five bone affected, but the long bones and mandible are most com-
lesions resulted in the prompt reversal of the systemic monly involved. In the long bones, the lesion is usually
symptoms and signs (fever, anemia, and weight loss). In metaphyseal but may also involve the epiphysis. The
two of these cases treated by simple curettage, the tumor most characteristic site is in the mental region of the
recurred within 3 to 4 months and displayed radiographic mandible, but the femur, tibia, and humerus are also
features of a destructive growth pattern, which prompted frequent sites of involvement. In general, it appears that
more aggressive resections with prosthetic replacement. the craniofacial bones are most frequently involved. The
But long-term follow-up and the ultimate outcome is not peak age incidence and most frequent sites of skeletal
available for these cases. involvement are shown in Figure 9-30.

Clinical Symptoms
SKELETAL FIBROMATOSES
Pain and swelling of the affected area are the most
The term skeletal fibromatosis, as well as the pathogenetic common symptoms. Pathologic fracture or deformity
concept of fibromatosis, was proposed by Stout.138,139 It of the affected bone can occasionally be a presenting
encompasses lesions with similar microscopic appearance symptom.

ERRNVPHGLFRVRUJ
648 9  Fibrous and Fibrohistiocytic Lesions

der(2) Chr2
25.3 25.3
25.1 25.2 25.2
25.1
24 24
23 23
22 22
21 21
16 16
Chr12
15 15 13.3
14 14 13.1 13.2 der(22)
13 12.3 13
13 12.2 12.1
12 12 11.2 12 der(12)
11 11.1 11.2
11.2 11.2 11.1 13.3
11.1 11.1 der(22) 11.1 Chr22 13.1
12 11.1
13.1 13.2
11.1 11.2
11.2
13 11.2 13 ATF1 EWSR1-ATF1 12.2
12.3
12 12 13.3 13.2 chimeric gene 12.1
13 12 12 11.2
13 14 11.1
14.1 11.2 11.1 14.1 11.2 11.1 11
14.2 11.1 11.2 14.2 11.1 15 12
14.3 14.3 11.2 21.1 13.1
21.2 21.1 EWSR1-CREB1 21.2 21.1 EWSR1 EWSR1 21.2 Silent
21.3 34
chimeric gene 21.3 12.1 12.2 21.3
breakpoint
22 22 12.3 13.1 22
13.2
23 23 13.3 23
24.1 24.1 24.1
24.2 24.2
24.3 24.3 24.2
24.31 24.32
31 31 24.33
32.1 32.1
32.2 32.2
32.3 32.3
33 33
34 Silent 34 CREB1
breakpoint 35
36
37.1
37.2
37.3

A
E10
E11

E12
E13
E14
E15
E16
E17
E1

E2
E3
E4

E5

E6

E7
E8

E9

EWSR1

E4

E5
E6

E7
E1

E2

E3
ATF1

Type 1 (50%) Exon 8 Exon 4

breakpoint
B EWSR1-ATF1 t(12;22)(q13;q12) EWSR1 portion ATF1 portion
Chimeric transcripts
E10
E11

E12
E13
E14
E15
E16
E17
E1

E2
E3
E4

E5

E6

E7

E8

E9

EWSR1
E1

E2

E3
E4

E5
E6

E7
E8

E9

CREB1

Exon 7 Exon 7

breakpoint
EWSR1-CREB1 t(2;22)(q34;q12) EWSR1 portion CREB1 portion
C Chimeric transcripts

FIGURE 9-27  ■  Angiomatoid fibrous histiocytoma: chromosomal translocation. A, Chromosomal translocations involving chromo-
somes 2:22 and 12:22 resulting in EWSR1-CREB1 t(2;22)(q34;q12) and EWSR1-ATF1 t(12;22)(q13;q12). B, Exon-intron structures of
EWSR1 and ATF1. Solid black arrows indicate the locations of the breakpoints within the introns. The molecular structure of the
resulting chimeric gene, which includes coding sequences of the N-terminal domain of EWSR1 and the C-terminal segment of ATF1,
is shown. C, Exon-intron structures of EWSR1 and CREB1. The molecular structure of the resulting chimeric gene, which includes
coding sequences of the N-terminal domain of EWSR1 and the C-terminal segment of CREB1, are shown.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 649

EWSR1
1 656 aa

breakpoint
TAD - transactivation (glycine/glutamine/serine/tyrosine “SYGQ” rich) domain 1-285 aa RRM - RNA recognition motif domain 361-447 aa
SF1int - interaction with SF1 domain 228-264 aa RG2 - arginine/glycine/proline rich domain 454-513 aa
IQ - binds calmodulin domain 256-285 aa ZF - zinc finger (RanBP2 type) domain 518-549 aa
RG1 - arginine/glycine rich domain 300-340 aa

ATF1
1 271 aa

Coactivator CBP, pKID domain 31-90 aa breakpoint


Basic leucine zipper domain 211-268 aa

CREB1
1 341 aa

Coactivator CBP, pKID domain 101-160 aa breakpoint


Basic leucine zipper domain 281-339 aa

EWSR1-ATF1 chimeric protein t(2;22)(q33;q12)


1 431 aa

EWSR1-CREB1 chimeric protein t(12;22)(q13;q12)


1 426 aa

B
FIGURE 9-28  ■  Angiomatoid fibrous histiocytoma: functional domains and chimeric proteins involved in translocations. A, The func-
tional domains of EWSR1, ATF1, and CREB1. B, Chimeric EWSR1-ATF1 and EWSR1-CREB1 proteins. The EWSR1-ATF1 chimeric
protein contains the N-terminal of TAD-transactivation domain of EWSR1 and leucine zipper C-terminal domain of ATF1. The EWSR1-
CREB1 chimeric protein again contains the N-Terminal of TAD-transactivation domain of EWSR1 and leucine zipper C-terminal
domain of CREB1.

Radiographic Imaging Gross Findings


The radiographic appearance is usually that of an expans- The tumors typically range in size from 3 to 10 cm, but
ile, lucent lesion with well-defined margins (Figs. 9-31 tumors up to 20 cm have been reported. They are gray-
and 9-32).120,128,141,145 There is often destruction of the white, fibrous, solid lesions that resemble desmoid lesions
overlying cortex with extension into the soft tissue (Fig. of soft tissue.
9-32). Features of a more destructive growth pattern with
irregular, ill-defined margins and pathologic fracture may Microscopic Findings
be present (Fig. 9-31). Honeycombed or moth-eaten pat-
terns have been described, but the radiographic presenta- Microscopically, the lesion is similar to soft tissue fibro-
tion is nonspecific. Desmoplastic fibroma can at least be matosis, consisting of spindle-shaped fibroblasts within
suspected if the lesion is completely lytic, is present in a an abundant, dense matrix of collagen (Figs. 9-34 and
young patient, and involves a typical site (i.e., the 9-35). Mitotic figures are rare. The cellularity varies in
mandible) (Fig. 9-33). Desmoplastic fibroma rarely pres- different areas of the lesion. It can be focally high, with
ents as a surface lesion. In such instances, a secondary less intercellular collagen gradually merging with pre-
involvement of bone by the more common soft tissue dominantly fibrous (scarlike) areas composed of dense
fibromatosis should be considered in the differential eosinophilic material with sparsely distributed fibroblas-
diagnosis. CT and MRI are useful techniques to delineate tic cells. The lesion exhibits an infiltrative destructive
the lesion and its relationship to the adjacent structures, growth pattern with permeation of bone marrow spaces
especially in anatomically complex locations such as cra- and haversian canals. The borders of the tumor, espe-
niofacial bones (Fig. 9-32).145 MRI typically shows a cially in soft tissue, are irregular, with fingerlike projec-
lesion with lobulated architecture and strong T2 signal tions infiltrating adipose tissue and skeletal muscle. At the
enhancement.115 periphery, it is occasionally difficult to distinguish the

ERRNVPHGLFRVRUJ
650 9  Fibrous and Fibrohistiocytic Lesions

Chr12 Chr16 der(12) der(16)


13.3 13.3 13.3 24
13.1 13.2 13.2 13.1 13.2 23
12.3 13.1 12.3
12.2 12.2 22
12.1 12 12.1
11.2 11.2 21
11.1 11.2 FUS 11.1 13
11 11.1 11 12.2 12.1
12 11.1 12
11.2 11.2 11.1
13.1 ATF1 12.1 13.1 FUS
13.3 12.2 11.2
11.1 FUS-ATF1
13.2 13
14 21
12
13.3 chimeric gene
13.1 13.2
15 22 13.2
21.1 14
23 13.3 15
21.2
21.3 24 21.1
21.2
22 21.3
23 22
24.1 23
24.2 24.1
24.31 24.32
24.33 24.2
24.31 24.32
24.33
A

FUS

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14

Exon 15
Exon 1

Exon 2
Exon 3

Exon 4
Exon 5

Exon 6

Exon 7

Exon 8

Exon 9
ATF1
Exon 1

Exon 2

Exon 3

Exon 4

Exon 5
Exon 6

Exon 7
Exon 1

Exon 2
Exon 3

Exon 4

Exon 5

Exon 5
Exon 6

breakpoint Exon 7
FUS-ATF1 t(12;16)(q13;p11) FUS portion ATF1 portion
B Chimeric transcripts

Chimeric gene
FUS
265 388
1 1 526 aa
415 454 501
165 285 371
breakpoint
QGSY - rich domain RNA recognition motif domain
RGG - Arginine glycine box domain Zinc finger, RanBP2 - type domain
Nucleotide - binding, alpha-beta plait domain PY-NLS - proline-tyrosine nuclear localization signal domain

ATF1
1 271 aa
31 90 211 268
breakpoint
Coactivator CBP, pKID domain
Basic leucine zipper domain
C

FUS-ATF1 chimeric protein


1 342 aa

D
FIGURE 9-29  ■  Angiomatoid fibrous histiocytoma: chromosomal translocation. A, Chromosomal translocation t(12;16)(q13;p11)
resulting in FUS-ATF1 chimeric gene. B, Exon-intron structures of FUS and ATF1. Solid black arrows indicate the locations of the
breakpoints within the introns. The molecular structure of the resulting chimeric gene, which includes coding sequences of the
N-terminal domain of FUS and the C-terminal segment of ATF1, is shown. C, Functional domains of FUS and ATF1 proteins are
shown. D, FUS-ATF1 chimeric protein contains the N-terminal QGSY-rich domain of FUS and coactivator CBP, pKID as well as leucine
zipper domains of the C-terminal portion of ATF1.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 651

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 9-30  ■  Desmoplastic fibroma. Peak age incidence and most frequent skeletal sites of involvement. Most frequent sites of
involvement are indicated by solid black arrows.

tumor tissue from secondary reactive fibrosis. Rare exam- action of the multiprotein complex to phosphorylate
ples of desmoplastic fibroma with extensive cartilaginous β-catenin. Unphosphorylated β-catenin is stable and
metaplasia have been described.108 Desmoplastic fibro- after translocation to the nucleus activates transcrip-
mas associated with other conditions such as fibrous dys- tion by binding to Tcf-Lef family proteins. Mutations
plasia melorheostosis and Paget’s disease have also been of β-catenin are found in numerous tumors. In fibroma-
described.122,126,144 toses, they typically cluster in the sequences encoding
the N-terminal component of the protein, essentially
Special Techniques affecting codons 41 and 45. Clustering of these muta-
tions at threonine 41 (T41A) and serine 45 (S45F and
Ultrastructural and immunohistochemical studies reveal S45P) is consistent with their role as phosphorylation
prominent myofibroblastic differentiation.124,130 Most targets. Functionally, these mutations prevent phos-
desmoplastic fibromas show at least focal positivity for phorylation and stabilize β-catenin, which upregulates
smooth muscle markers such as smooth muscle actin nuclear transcription. As a consequence, myofibroblastic
(SMA). The hallmark of sporadic extraskeletal fibromato- cells in fibromatoses frequently contain mutated stable
ses arising in deep soft tissue is the mutation of β-catenin β-catenin protein that can be detected immunohisto-
(Fig. 9-36).104,107,127 β-catenin plays an important role in chemically in the nucleus of tumor cells.106,112 In soft
the embryonic development of mesenchymal cells affect- tissue, the presence of mutations involving β-catenin is
ing proliferation, motility, and invasiveness. It mediates associated with more aggressive growth pattern with a
the canonical Wnt (wingless) signaling pathway.119,129,132,142 higher rate of local recurrences as compared to those
The quiescent status of the Wnt pathway promotes lesions that do not carry mutations of β-catenin.116,124,131
interaction between a multiprotein complex, includ- Such associations are not, however, available for skeletal
ing adenomatous polyposis coli (APC), β-catenin, axin, lesions. Moreover, preliminary data indicate that muta-
and glycogen synthase kinase-3β and serine and thero- tions of β-catenin (CTNNB1) gene are not present in
nine residues in the N-terminals of β-catenin, inducing skeletal fibromatoses.121 Desmoplastic fibromas show;
their phosphorylation. The phosphorylated protein is a however, occasionally strong positive nuclear staining
target for ubiquitination and degradation. The activa- for β-catenin.121 The mechanisms stabilizing β-catenin in
tion of the Wnt signaling pathway inhibits the inter- Text continued on p. 657

ERRNVPHGLFRVRUJ
652 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-31  ■  Desmoplastic fibroma: radiographic features. A, Anteroposterior (AP) radiograph showing a pathologic fracture
through lytic lesion in shaft of distal radius in a 33-year-old woman. Curettage showed fibrous lesion with benign cytologic features.
The lesion was bone grafted and healing was achieved. B, One year later, lesion shown in A recurred, and recurrent desmoplastic
fibroma had broken through cortex on ulnar side of radius. Patient was clinically asymptomatic 2 years after second curettage and
bone grafting. C and D, AP and lateral radiographs of knee in a 20-year-old woman with expanded lytic lesion involving proximal
end of tibia. Note trabeculated appearance and absence of periosteal reaction.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 653

A B

C D
FIGURE 9-32  ■  Desmoplastic fibroma: radiographic features. A and B, Anteroposterior (AP) and lateral radiographs of a destructive
lytic lesion involving the proximal fibula. C, Fat-saturated T2-weighted coronal magnetic resonance image showing signal enhance-
ment and overall macroglobular arrangement of desmoplastic fibroma. Same case as shown in A and B. D, AP radiograph showing
a recurrent desmoplastic fibroma surrounding an intramedullary metallic rod of the femur.

ERRNVPHGLFRVRUJ
654 9  Fibrous and Fibrohistiocytic Lesions

A B

D
FIGURE 9-33  ■  Desmoplastic fibroma of mandible: clinical, radiographic, and microscopic features. A, Clinical photograph of
a 29-year-old woman with palpable mass in submental region. B and C, Radiographs show well-circumscribed lytic lesion of
mandible in same patient. D, Low-power photomicrograph shows spindle-cell fibrous lesion. Note involvement of skeletal muscle
(×50; hematoxylin-eosin).

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 655

A B

C D

FIGURE 9-34  ■  Desmoplastic fibroma: microscopic features. A-D, Low and intermediate power photomicrographs showing bundles
of spindle cells with abundant intercellular collagen. There is no osteoid or cartilage matrix seen. Inset, Nuclear details of spindle
cells. (A and C, ×100; B and D, ×200; inset, ×400.) (A-D, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
656 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-35  ■  Desmoplastic fibroma: microscopic features. A and B, Low and intermediate power photomicrographs showing
uniform spindle cells with abundant collagen. Inset shows strong positive nuclear staining for β-catenin (CTNNB1). C and D, Low
and intermediate power photomicrographs showing somewhat hypercellular areas of desmoplastic fibroma with proliferation of
plump spindle cells. (A and C, ×100; B and D, and inset, ×200.) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 657

CTNNB1
Chr 3 B

Exon 10

Exon 11
Exon 12
Exon 13

Exon 14

Exon 15

Exon 16
26

Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6

Exon 7

Exon 8
Exon 9
25
24.3 24.2
24.1

41240941

41281939
23
22
21.3
21.2
CTNNB1
21.1
14.3 14.2
14.1
13
12
11.1 11.2 CTNNB1 S45P, S45F
11.2 11.1
12
13.1
13.2
13.3

T41A
395
15
21

692
22
23

49

18
10
5

13
261
417
24

8
7

9
78

874
25.1

36
12

16

90
434
25.2

4
19

13
14
14
12
25.3

7
4
4
6
8
9
6

5
6
8
6
9
26.1
26.2
26.3
27
28
29
A 198022430 bp SYLDSCIHSGATTTAPSLSG
Phosphorylation by GSK-3
Missense substitution
Nonsense substitution - Stop
Insertion inframe
Deletion inframe
Insertion frameshift
13
4

6
4
Deletion frameshift
Complex - deletion in frame
Unknown 1 781 aa
Substitution - coding silent -catenin FIBROMATOSIS Armadilo-type fold domain 134-664
Complex - frameshift C
FIGURE 9-36  ■  Mutations of β-catenin (CTNNB1) gene in extraskeletal soft tissue fibromatoses. A, Chromosomal localization of
β-catenin gene on the short arm of chromosome 3. B, Exon-intron structure of the β-catenin gene. C, Distribution of mutations in
relation to functional domains of β-catenin. Note clustering of mutations corresponding to the phosphorylation sites by GSK3β,
including T41A, S45P, and S45F, frequently found in fibromatoses.

desmoplastic fibromas are uncertain. Therefore, fibro- Furthermore, engulfed trabeculae of lamellar bone in
matoses arising in bone may develop without mutations desmoplastic fibroma can be misinterpreted as fibrous
involving B-catenin and other genetic mechanisms have dysplastic bone. Attention to its lamellar composition is
been postulated.121,143 important in avoiding this error.

Differential Diagnosis Treatment and Behavior


Soft tissue fibromatosis has microscopic features that overlap Desmoplastic fibroma exhibits locally aggressive behav-
with desmoplastic fibroma and can be difficult to distin- ior without the capacity to metastasize. Earlier reports
guish in cases in which the soft tissue lesion invades bone indicate that simple curettage and bone grafting resulted
sufficiently deeply to produce defects that are visible on in a recurrence rate as high as 40%. Wide resection is the
radiograph. More important is the distinction from low- preferable mode of treatment, although recurrences have
grade fibrosarcoma of bone. This lesion is rarely so well differ- also been reported with this mode of treatment. Rare
entiated that it does not show, in adequate biopsy samples, association of desmoplastic fibroma with other lesions
some nuclear irregularity and hyperchromatism, as well such as fibrous dysplasia or melorheostosis has been
as a degree of mitotic activity not present in desmoplastic reported.64,75
fibroma. Conversely, some low-grade fibrosarcomas show
heavy collagenization and deceptively benign-appearing, Personal Comments
small dark nuclei in focal areas. Furthermore, the radio-
logic features may be paradoxical, in that fibrosarcoma Desmoplastic fibroma is among the rarest of bone tumors,
can be well circumscribed and appear to be noninvasive, although it is sometimes diagnosed inappropriately on
whereas desmoplastic fibroma not only shows aggressive histologic grounds when the biopsy sample in fibrous
radiologic features, but also grows in an invasive, locally dysplasia is from a heavily collagenized area devoid of
infiltrative manner. Hence, the distinction in the border woven bone spicules. True desmoplastic fibroma is a
zone of these two lesions, both of which exhibit local locally aggressive, intraosseous lesion that histologically
aggressiveness, may not be a clinically significant one. reproduces the pattern of fibromatosis in soft tissue. Its
Fibrous dysplasia is sometimes mistaken for desmoplas- clinical behavior can closely resemble that of aggressive
tic fibroma when the sample does not contain histologic fibromatosis of soft tissue in that the lesion can infiltrate
evidence of osteoid or delicate woven bone trabeculae. locally but does not metastasize. Whether it differs

ERRNVPHGLFRVRUJ
658 9  Fibrous and Fibrohistiocytic Lesions

essentially from low-grade fibrosarcoma seems academic discovered during the first 3 months of life.174,182,209 Most
because both lesions have the same biologic potential. often the lesion involves the soft tissue of the limb but
Both should be treated by wide local excision. We are not can also involve the soft tissue of the trunk and the head
aware of any report indicating the transformation of a and neck area.195,207,208,210,211 The bone lesions occasionally
desmoplastic fibroma to a fully malignant metastasizing become manifest much later in life and have been diag-
sarcomatous tumor. nosed several years after birth.
Microscopically, congenital fibrosarcoma is more cel-
lular than desmoplastic fibroma and is composed of less
Juvenile Multifocal Myofibromatosis mature fibroblasts with minimal deposition of collagen
Juvenile multifocal myofibromatosis was first described (Fig. 9-38). Mitoses and scattered inflammatory cells are
by Stout165 in 1954 as congenital generalized fibromatosis. frequently present. Overall, the microscopic picture is
The lesions are typically multifocal, but occasionally the similar to adult fibrosarcoma.
disorder can present as a solitary focus.147-152,154-157,162,166,168 Data on clinical behavior are available only in refer-
It has two basic clinical forms. Somatic involvement is ence to the soft tissue lesions. Originally, congenital
limited to the subcutaneous, muscular, or skeletal tissue; fibrosarcoma was considered to be a tumor with no meta-
in the combined somaticovisceral form, in addition to static potential, but analysis of larger series indicated that
soft tissue, viscera can be involved. This separation is the risk of metastasis ranges from 5% to 10%.210 A spon-
prognostically valid. The somatic form has a benign taneous regression similar to that seen in congenital
course that leads to the resolution of cutaneous soft tissue fibromatosis has been reported.190,196,199 On the other
and skeletal lesions within a few months to a few years. hand, progression to a high-grade sarcoma with malig-
Some authors postulate that spontaneous apoptosis can nant fibrous histiocytoma–like features has also been
be a putative molecular mechanism causing regression of reported.201 A few cases of congenital fibrosarcoma, with
these lesions.153 The combined somaticovisceral form has the primary tumor in bone, have recurred after local exci-
a poor prognosis, with death occurring in the first months sion and required ablative surgery to control the disease.
of life. Skeletal involvement can occur in both forms. A Our personal experience is confined to one case seen in
few cases with extensive primary involvement of the skel- consultation. This patient was seen at age 3 years with a
eton have been reported.148,151,152,155,163,167 Extensive destructive lytic lesion in the parietooccipital region of
involvement of an extremity by multifocal myofibroma- the skull. After 17 years the fatal outcome was the result
tosis may cause length discrepancy.160 of multiple recurrences with uncontrolled local growth,
The disease has a predilection for the metaphyses of eventually involving vital central nervous system struc-
the long tubular bones. It presents radiographically as tures (Fig. 9-39). There are too few cases to evaluate the
multiple, lytic, sharply demarcated defects that tend to metastatic potential of skeletal lesions. The available data
be eccentric. Flat bones (pelvis, scapula, ribs, and skull) support the notion that congenital fibrosarcoma is more
can also be involved. Microscopically, the lesions are aggressive than conventional fibromatosis. In terms of its
similar to conventional fibromatosis but overall are biologic potential, it is probably intermediate between
more cellular and less fibrous. The cells are arranged in conventional fibromatosis and adult fibrosarcoma.
interlacing bundles with a prominent network of thin- Various nonrandom cytogenetic abnormalities distinct
walled sinusoidal vascular channels that produce a from those seen in cases of adult fibrosarcoma or malig-
hemangiopericytoma-like pattern (Fig. 9-37). The pres- nant fibrous histiocytoma have been reported in con-
ence of a hemangiopericytoma-like pattern is a distinctive genital juvenile fibrosarcoma.169,171,175,183,202 The described
microscopic feature of infantile myofibromatosis.159 It is anomalies range from trisomy 11 and other trisomies
found at least focally and is typically most prominent in to nonrandom translocations involving chromosomes
the central portion of the lesion. The ultrastructural 12 and 13.206 The distinctive molecular abnormality
studies, as well as immunohistochemical stains, reveal of fibromatosis-like fibrosarcoma comprises t(12;15)
prominent myofibroblastic differentiation. In fact, some (p13;q25) translocations resulting in ETV6-NTRK3 gene
studies suggest that there is a continuous spectrum of fusion (Fig. 9-40).170,186,187,205 The 5′ partner of the fusion
lesions ranging from typical fibromatosis to infantile is the ETV6 gene located on chromosome 12 and the 3′
hemangiopericytoma.158 Cytogenetic reports document partner is NTRK3 located on chromosome 15. The ETV6
interstitial deletion of 6q, del(6)(q12;q15), and unbal- fusion genes participate in translocations of several solid
anced translocation of 9q resulting in 9q monosomy and and hematologic malignancies.184 In the chimeric protein
trisomy of 16q.161,164 Infantile fibromatosis is negative for the N-terminal of the protein comprises SAM/PNT
t(12;15)(p12;q25) translocation and the ETV6-NTRK3 (Sterile alpha motif/PNT pointed domain from ETV6)
gene fusion.146 while the C-terminal contains the cysteine-rich flank-
ing region. This translocation has been well documented
Congenital Fibromatosis–like in the more frequent soft tissue variant of the tumor.
The preliminary evidence indicates that it may also be
Fibrosarcoma (Infantile Fibrosarcoma) present in the rare instances of infantile fibromatosis–
This rare tumor typically occurs in the soft like fibrosarcoma involving bone. The presence of this
tissue.172,173,178-181,185,188,191,194,197,200,203,204,211 It is even more translocation is a useful tool in the differential diagnosis
rare as a primary lesion in bone, with only a few single of this rare pediatric malignancy and may also represent
case reports of its occurrence.96,102 Typically the tumor is its potential therapeutic target.176,177,189,192,193,198,205
congenital and clinically evident at birth, or it is Text continued on p. 663

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 659

B C
FIGURE 9-37  ■  Juvenile fibromatosis of tibia: microscopic features. A, Low power photomicrograph shows spindle-cell proliferation
with hemangiopericytoma-like pattern. Note increasing cellularity in central portion of lesion. B and C, Intermediate and high power
photomicrographs of the central portion of the lesion showing proliferations of plump myofibroblastic cells. Note prominent vas-
culature mimicking hemangiopericytoma. (A, ×50; B, ×100; C, ×200.) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
660 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-38  ■  Infantile fibromatosis–like fibrosarcoma: microscopic features. A-D, Low and intermediate power photomicrographs
show cellular spindle-cell fibroblast-like lesion. Note uniformly higher level of cellularity compared with more typical desmoplastic
fibroma. Inset, Bland nuclear features with finely dispersed chromatin in plump spindle cells. (A and C, ×100; B and D, ×200; inset,
×400.) (A-D and inset, hematoxylin-eosin.) (Courtesy Dr. H. Sonobe, Kochi, Japan.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 661

A B

C D
FIGURE 9-39  ■  Infantile fibromatosis–like fibrosarcoma: radiographic features. A, Lateral plain radiograph shows destructive lesion
in parietooccipital area. B, Axial computed tomogram shows destructive lesion on the right. C, Same patient at age 12 years with
recurrent tumor in same area. D, Uncontrolled local growth of tumor extensively involving cranial bone. (Courtesy Dr. H. Sonobe,
Kochi, Japan.)

ERRNVPHGLFRVRUJ
662 9  Fibrous and Fibrohistiocytic Lesions

Chr12 Chr15 der(12) der(15)


26.3 26.2
13.3 13.2 13 13
13.1 12 26.1 NTRK3-ETV6 12
ETV6 11.2 no transcription
12.2 12.3 11.2
11.1 11.1 12.2 12.3 11.1
11.2 12.1 11.2 11.1 11.2
12 11.2 12.1 12
11 11.1 13 11.1 13
14 11 14
12 12
15 15
13.1 21.1 13.1 21.1
13.3 13.2 21.2 13.2 21.2
21.3 13.3 21.3
14 22.1 14 22.1
22.2 22.2
15 22.3 15 22.3
21.1 23 23
21.2 21.1 24
24 21.2
21.3 21.3 ETV6-NTRK3
22
25 NTRK3 25
chimeric gene
26.1 22 13.2
23 26.2 13.1
26.3 23 13.3
24.1 24.1
24.2 24.2
24.31 24.32 24.31
24.33 24.32
24.33

A
NTRK3

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14

Exon 15

Exon 16
Exon 17

Exon 18
Exon 19
Exon 1
Exon 3
Exon 4
Exon 5

Exon 6
Exon 7
Exon 8
Exon 9
Exon 2

ETV6
Exon 1

Exon 2

Exon 3

Exon 4

Exon 5

Exon 6
Exon 7
Exon 8

Exon 5 Exon 15

breakpoint
ETV6-NTRK3 t(12;15)(p13;q25) ETV6 portion NTRK3 portion
B Chimeric transcripts

ETV6 NTRK3
1 452 aa 1 839 aa
18 127 339 420 31 63 160 208 309 321 399 538 828

breakpoint breakpoint
SAM/PNT- Sterile alpha motif/PNT pointend domain Leucine-rich repeat-containing N-terminal domain
Winged helix-turn-helix DNA - binding/Ets domain Cysteine-rich flanking region, C-terminal domain
Immunoglobulin-like fold domain
Protein kinase-like domain

ETV6-NTRK3 chimeric protein


1 648 aa

C
FIGURE 9-40  ■  Infantile fibromatosis–like fibrosarcoma: chromosomal translocation. A, Schematic diagram of t(12;15)(p13;q25) trans-
location. B, Exon-intron structures of the NTRK3 and ETV6 genes. Solid black arrows indicate the locations of the breakpoints. The
molecular structure of the resulting chimeric gene, which includes coding sequence of the N-terminal domain of ETV6 and the
C-terminal segment of NTRK3, are shown. C, The functional domains of the two proteins are depicted. The chimeric protein contains
SAM dimerization domain of ETV6 fused to the protein tyrosine kinase (PTK) domain of NTRK3.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 663

MALIGNANT FIBROUS HISTIOCYTOMA osteoblastic subtypes.239,241,257,265 On the other hand, gene


expression profiling studies disclose the mesenchymal
The terms malignant histiocytoma and malignant fibrous stem cell-like phenotype and the involvement of the Wnt
xanthoma were coined by Stout and his coworkers in the pathway in malignant fibrous histicytoma, supporting the
early 1960s.249,251 The concept that these tumors are stem cell origin of these neoplasms.228,230,239
derived from fibrohistiocytic cells was based on tissue Modern classifications, including the recent WHO
culture observations. These experiments have shown that Classifications of Tumours of Soft Tissue and Bone,
the cells derived from these lesions look and behave propose the term undifferentiated high-grade pleomorphic
in culture as fibroblasts.227,251 In the initial stages of sarcoma instead of malignant fibrous histiocytoma.226 In
culture, they exhibit features of ameboid movement fact, this term is widely used in diagnostic practice
and phagocytosis. These observations, together with the but does not solve all of the problems because some of
ultrastructural data, led to the use of the term fibrous these tumors are not necessarily pleomorphic and many
histiocytoma. This is a somewhat misleading term because have predominantly spindle, rhabdoid, or epithelioid cell
it implies that the origin is from macrophage-monocyte morphology.
lineage. We now understand that these tumors are not
derived from macrophage-monocyte precursors. The Definition
phenotypic features of these lesions are of primitive
mesenchymal derivation that exhibit, at least in part, Malignant fibrous histiocytoma is a malignant neoplasm
fibroblastic or more often myofibroblastic differentia- characterized by a mixture of spindle and pleomorphic
tion.212,227,237,243,253-255 The controversy about the histogen- cells with a prominent storiform arrangement of the
esis of this tumor persists. Moreover, it is still not clear spindle cells. The latter exhibit focal myofibroblastic dif-
whether this tumor is a specific entity with uniform ferentiation. These neoplasms in bone formerly were
pathogenesis or simply represents a microscopic pattern classified as high-grade, pleomorphic, undifferentiated
common to a heterogeneous group of undifferentiated or osteosarcomas or high-grade fibrosarcomas.
poorly differentiated sarcomas of different origins. The
latter view is supported by its frequent occurrence as a Incidence and Location
secondary sarcoma complicating various benign precur-
sors. Moreover, tumors with morphologic features indis- Malignant fibrous histiocytoma of bone is relatively
tinguishable from de novo malignant fibrous histiocytoma rare and makes up less than 2% of all primary malig-
frequently arise as dedifferentiated components of low- nant bone tumors. The peak age incidence and frequent
grade, locally aggressive tumors in bone and soft tissue. sites of skeletal involvement are shown in Figure 9-41.
In fact, sarcomatoid carcinomas developing in many The age-related incidence rate and frequency distribu-
organs in association with a preexisting epithelial neo- tion show a gradual increase, with the peak incidence
plasm share many similarities with malignant fibrous in patients older than age 50 years, but there is a wide
histiocytoma. distribution from the second to the seventh decades of
In spite of these controversies, the term malignant life (Fig. 9-42). Approximately 10% of cases are diag-
fibrous histiocytoma is widely used in diagnostic pathology nosed in patients younger than age 20 years. Malignant
and defines a group of lesions that may not be histoge- fibrous histiocytoma has a predilection for the major
netically and pathogenetically uniform but that have long tubular bones, and the femur is most frequently
some common features defining them as a distinct clini- involved. Approximately 30% of cases occur in the knee
copathologic group.220,223,225,229,232,240,242,246,249,259,260 The area with involvement of the distal femoral and proxi-
identification of malignant fibrous histiocytoma in bone mal tibial metaphyses. The pelvis is the most frequently
as a distinct entity in the 1970s resulted in a sharp decline involved trunk bone. Individual cases are reported in
in the use of other diagnostic terms (predominantly osteo- other parts of the skeleton, and almost any bone can be
lytic osteosarcoma and fibrosarcoma) to describe non–bone- involved.221,256,259,260,261,264 Extremely rare cases of mul-
forming sarcomatous neoplasms of bone. tifocal and familial skeletal malignant fibrous histio-
The investigations of malignant fibrous histiocytoma cytoma have been described.216,218,224,250 About 20% of
during the past two decades have revolved around two malignant fibrous histiocytomas in bone occur as a result
major themes. The first one and most common postulates of underlying conditions. Malignant fibrous histiocy-
that these tumors represent a common pattern of pro- toma is a well known complication of such conditions
gression similar to many mesenchymal neoplasms irre- as Paget’s disease, bone infarct, and radiation damage.
spective of their original lineage differentiation. The It also arises as a dedifferentiation of such low-grade
second theme postulates that malignant fibrous histiocy- preexisting bone tumors as chondrosarcoma, paraosteal
toma is not a result of dedifferentiation but develops de osteosarcoma, low-grade intraosseous osteosarcoma, and
novo by transformation of mesenchymal stem cells.238 giant cell tumor. In addition, a rare autosomal dominant
Extensive use of novel differentiation markers and molec- familial syndrome known as diaphyseal medullary steno-
ular testing provide evidence of diverse differentiation sis predisposes to the development of malignant fibrous
in a subset of malignant fibrous histiocytoma. Using histiocytoma. It is estimated that approximately 30% of
these approaches several mesenchymal differentiation patients affected by this syndrome develop malignant
patterns can at least be focally detected in many malig- fibrous histiocytoma. Radiographically, it is characterized
nant fibrous histiocytomas to subclassify them as myo- by diffuse diaphyseal medullary stenosis with the overly-
genic, myoepithelial, myofibroblastic, lipoblastic, and ing cortex showing extensive thickening. There are also

ERRNVPHGLFRVRUJ
664 9  Fibrous and Fibrohistiocytic Lesions

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 9-41  ■  Malignant fibrous histiocytoma. Peak age incidence and frequent sites of skeletal involvement in fibrous histiocytoma.
Most frequent sites of involvement are indicated by solid black arrows.

1 25

0.8 20
(cases/100,000 persons)

Age distribution (%)


Incidence rate

0.6 15

0.4 10

0.2 5

0 0
0-4

5-9

10-14

15-19

20-24

25-29

30-34

35-39

40-44

45-49

50-54

55-59

60-64

65-69

70-74

75-79

80-84

85+

Age at diagnosis
FIGURE 9-42  ■  Epidemiology of malignant fibrous histiocytoma. National Cancer Institute Surveillance Epidemiology, and End Result
Program, 1973-2005. Age-adjusted incidence rate and age-specific frequency, all races, both sexes, 1277 cases.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 665

metaphyseal striations, scattered foci of infarctions, and are descriptive and do not necessarily imply distinct
diffuse sclerosis involving the long bones.213,231,236,244,248 pathogeneses or differences in clinical behavior.
The storiform-pleomorphic variant represents a pro-
Radiographic Imaging totype pattern of malignant fibrous histiocytoma. The
spindle cells resemble fibroblasts and are arranged in
Radiographically the tumor presents as a purely lytic, storiform patterns. Areas with less differentiated spindle
ill-defined intramedullary defect. The lesion most fre- cells exhibit prominent nuclear atypia and brisk mitotic
quently involves the metaphyseal parts of long tubular activity. Mononuclear cells with abundant cytoplasm
bones and may extend into the epiphysis.240,245,247 Diaphy- that resemble histiocytes containing pleomorphic, highly
seal locations are less common. The tumor exhibits a atypical nuclei represent undifferentiated mesenchymal
geographically destructive growth pattern, but moth- components. Multiple atypical mitoses are common.
eaten or permeative patterns also have been described. Giant cells of both the malignant type and the benign
Early cortical disruption and extension into soft tissue osteoclastic type are present. The cytoplasm of these cells
occur (Figs. 9-43 and 9-44). Typically, there is little or no is usually densely eosinophilic and can be responsible for
periosteal new bone formation associated with the tumor. superficial mimicry of pleomorphic rhabdomyoblasts. On
The radiographic features are not specific, and the exact the other hand, vacuolar change of the cytoplasm can
diagnosis cannot be established from findings on radio- mimic lipoblastic differentiation. Foci of necrosis, scat-
graphs (Figs. 9-43 to 9-46). Pathologic fracture is fre- tered foamy histiocytes, and inflammatory cells are fre-
quent, especially in the weight-bearing bones. quently present. The overall pattern is characteristic and
easy to recognize.
Gross Findings The classic pattern of malignant fibrous histiocytoma
is rarely confused with benign conditions, but occasion-
The gross appearance of malignant fibrous histiocytoma ally the tumor is composed predominantly of mature
is highly variable (see Figs. 9-46, 9-47, and 9-48). The fibroblastic cells with inconspicuous atypia (Fig. 9-57).
consistency varies from soft to fibrous. Most tumors are The presence of numerous multinucleated, osteoclast-
tan-gray with fleshy yellow areas. The tumor has irregu- like giant cells is a frequent feature of malignant fibrous
lar, ill-defined borders. Areas of necrosis and hemorrhage histiocytoma of bone. These cells occasionally dominate
are frequently present. Cortical disruption with poorly the picture, and giant cell–rich malignant fibrous histio-
demarcated infiltration of the adjacent soft tissue is fre- cytoma can be confused with giant cell tumor or other
quently seen. benign giant cell–containing conditions of bone (Fig.
9-51). Inflammatory cells are frequently present in malig-
Microscopic Findings nant fibrous histiocytoma of bone, but a distinct inflam-
matory variant of malignant fibrous histiocytoma is rarely
The mixture of spindle cells in storiform arrangement identified in bone (Fig. 9-57).
and pleomorphic cellular areas is a hallmark of malignant Numeric grading of malignant fibrous histiocytoma
fibrous histiocytoma (Figs. 9-49 to 9-52).217,219,220,221,232,247 that applies a conventional Broders’ system remains con-
The microscopic diversity of this tumor is broad, and the troversial. Many authors simply divide these tumors into
range of patterns varies from almost exclusively spindle low- and high-grade variants. The data suggest that pre-
cell through hemangiopericytoma-like, epithelioid to dominantly fibroblastic tumors with minimal atypia have
highly pleomorphic lesions (see Figs. 9-52 to 9-58). a better prognosis than highly atypical lesions. Convinc-
The amount of collagen also varies. In more differenti- ing clinical data supporting the relationship between
ated spindle-cell areas, the tumor can form thick clinical behavior and complex histologic grading in these
fibrillar deposits surrounding individual cells that mimic tumors are lacking for bone lesions. Nevertheless, modern
osteoid. This feature accounts for earlier inclusion of combined-modality treatment protocols require subdivi-
these tumors in the group of osteosarcomas. The most sion of these tumors and other sarcomas into low and
distinct feature of these tumors is the storiform arrange- high grades.
ment of spindle cells, which is present focally in most
cases. Malignant fibrous histiocytoma of soft tissue has
Special Techniques
been subclassified into several types: storiform-
pleomorphic, myxoid, giant cell–rich, and inflammatory. Immunohistochemistry plays little direct role in the diag-
All these histologic subtypes may be found in primary nosis of malignant fibrous histiocytoma. It is used pre-
malignant fibrous histiocytoma of bone (Figs. 9-49, 9-51, dominantly to exclude other malignant neoplasms
9-56, and 9-58). considered in the differential diagnosis. The tumor cells
Angiomatoid malignant fibrous histiocytoma has are strongly and uniformly positive for vimentin. Weak
recently been reclassified and is no longer considered a positivity for SMA is a frequent finding. Other markers,
variant of malignant fibrous histiocytoma on the basis of such as keratin, S-100 protein, and desmin, are predomi-
its distinct pathologic and clinical features. For this nantly used to rule out other primary and metastatic
reason, a separate description of this entity is provided. neoplasms of bone. The immunohistochemical markers
The most frequent types of malignant fibrous histio- for macrophage-monocyte lineage, such as KP-1 (CD68),
cytoma in bone are the storiform-pleomorphic and giant have not proved useful in the diagnosis of malignant
cell variants. Rare cases of myxoid and inflammatory sub- fibrous histiocytoma because they are not expressed in
types have been reported (Fig. 9-56). These designations Text continued on p. 682

ERRNVPHGLFRVRUJ
666 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-43  ■  Malignant fibrous histiocytoma: radiographic features. A, Lateral radiograph of distal femur of a 28-year-old man with
large lytic tumor in shaft with wide zone of transition and anterior cortical destruction. B, T2-weighted sagittal magnetic resonance
image (MRI) shows cortical breakthrough and overlying soft tissue component of tumor. Inset, Axial computed tomogram shows
permeative bone destruction and extension into soft tissue. Calcified material within the tumor represents residual bone. C, Lateral
radiograph of distal femur in a 20-year-old woman. Ill-defined permeative destruction is present in distal metaphysis without associ-
ated cortical destruction or periosteal reaction. D, T2-weighted sagittal MRI of tumor shown in C reveals greater extent of tumor
proximally than can be appreciated from plain film. Note that scar of growth plate delimits tumor distally.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 667

A B

C
FIGURE 9-44  ■  Malignant fibrous histiocytoma: radiographic features. A, Anteroposterior radiograph showing a destructive perme-
ative lesion involving periacetabular region extending to ischium and pubic ramus. B, Axial computed tomogram of same case as
A showing a destructive lesion of the left pelvis with soft tissue extension. C, T1-weighted axial magnetic resonance image of case
in A and B showing extensive low density tumor involving the left pelvis and adjacent soft tissue.

ERRNVPHGLFRVRUJ
668 9  Fibrous and Fibrohistiocytic Lesions

A C

D E
FIGURE 9-45  ■  Malignant fibrous histiocytoma: radiographic features. A, Anteroposterior (AP) radiograph of pelvis of a 39-year-old
man with large destructive tumor in left sacroiliac region of pelvis. B, Axial computed tomogram (CT) (bone window) shows moth-
eaten destruction of right ilium and sacral wing with cortical breakthrough and soft tissue mass. C, Soft tissue window CT shows
extent of large soft tissue mass. D, AP radiograph of thoracic spine and ribs shows destructive changes in posterior rib and adjacent
vertebral body in a 27-year-old man. E, Axial CT reveals completely lytic tumor expanding posterior part of rib (vertebral end) and
involving adjacent thoracic vertebra.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 669

A B

C D

FIGURE 9-46  ■  Malignant fibrous histiocytoma: radiographic and gross features. A, Anteroposterior radiograph showing a destructive
moth-eaten lesion of the proximal humerus. B, Fat-saturated T2-weighted coronal magnetic resonance image showing a high signal
intensity lesion of the proximal humerus. C and D, Gross photomicrographs of bisected resection specimen showing an intramedul-
lary tumor of the proximal humerus with mixed fleshy and mucinous appearance.

ERRNVPHGLFRVRUJ
670 9  Fibrous and Fibrohistiocytic Lesions

A B

C D

FIGURE 9-47  ■  Malignant fibrous histiocytoma: gross features. A, Fleshy intramedullary tumor of distal femur. B, Intramedullary
fleshy tumor of proximal humerus with soft tissue extension and elevation of periosteum seen medially. C, Fleshy intramedullary
tumor of the proximal tibia fused with the overlaying cortex. Note relatively sharp demarcation of the intramedullary component
of the tumor. D, Fleshy mass of proximal tibia with areas of hemorrhage and cystic change. Note cortical disruption.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 671

A B

C D
FIGURE 9-48  ■  Malignant fibrous histiocytoma: gross features. A, Fleshy intramedullary tumor of distal femur with cystic changes
and hemorrhage proximally. B, Circumferential soft tissue extension of distal femur with extensive necrosis and multifocal hemor-
rhage grossly mimicking telangiectatic osteosarcoma. C, Fleshy tumor with soft tissue extension involving distal end of femur.
D, Destructive fleshy mass with cortical disruption and soft tissue extension of distal tibia end.

ERRNVPHGLFRVRUJ
672 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-49  ■  Malignant fibrous histiocytoma: microscopic features. A and B, Low and intermediate power views showing plump
malignant spindle cells with brisk mitotic activity. C and D, Low and intermediate power views of pleomorphic spindle cells. (A and
C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 673

A B

C D
FIGURE 9-50  ■  Malignant fibrous histiocytoma: microscopic features. A and B, Low power views showing malignant spindle cells
with atypical nuclei. Inset shows atypical nuclei with prominent nucleoli. C and D, Intermediate and high power views showing
atypical plump cells. (A, ×50; B and C, ×100; D and inset, ×200.) (A-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
674 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-51  ■  Malignant fibrous histiocytoma: microscopic features. A and B, Intermediate and high power views of pleomorphic
tumor with scattered multinucleated giant cells with osteoclast-like features. C and D, Low and intermediate power views of a
spindle-cell tumor with a somewhat parallel arrangement of cells. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 675

A B

C D

FIGURE 9-52  ■  Malignant fibrous histiocytoma: microscopic features. A and B, Low and intermediate power views of the advancing
edge of malignant fibrous histiocytoma permeating the marrow spaces. C and D, Low and intermediate power views of tumor
in A and B showing pleomorphic features of tumor cells. (A, ×10; B, ×50; C, ×100; D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
676 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-53  ■  Malignant fibrous histiocytoma: microscopic features. A-D, Low and intermediate power views of a tumor composed
of plump atypical cells with prominent eosinophylic cytoplasm. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 677

A B

C D
FIGURE 9-54  ■  Malignant fibrous histiocytoma: microscopic features. A-D, Intermediate and high power views of a tumor composed
of atypical somewhat epithelioid cells with dense eosinophilic cytoplasm. Tumors with such features can be confused with
rhabdomyosarcoma. All markers of skeletal muscle differentiation were negative in this case. (A and C, ×100; B and D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
678 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-55  ■  Malignant fibrous histiocytoma: microscopic features. A and B, Pleomorphic tumor with prominent hemangiopericytoma-
like vasculature. C and D, More solid components of the same tumor with prominent pleomorphism and nuclear atypia. (A and
C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 679

A B

C D
FIGURE 9-56  ■  Malignant fibrous histiocytoma: microscopic features. A-D and Inset, Malignant fibrous histiocytoma with
prominent component of inflammatory cells admixed with foamy histiocytic cells. (A and C, ×100; B and D, and inset, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
680 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-57  ■  Malignant fibrous histiocytoma: microscopic features. A-D, Intermediate and high power views of tumor composed
of short, plump, somewhat epithelioid cells with prominent collagen matrix depostion. (A and C, ×100; B and D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 681

A B

C D
FIGURE 9-58  ■  Malignant fibrous histiocytoma: microscopic features. A, Anaplastic spindle cells in malignant fibrous histiocytoma.
B, Pleomorphic cells with some epithelioid features. C, Giant cell variant of malignant fibrous histiocytoma (×200). D, Myxoid variant
of malignant fibrous histiocytoma (A, ×100; B-D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
682 9  Fibrous and Fibrohistiocytic Lesions

the pleomorphic tumor cells, but rather show positivity The differential diagnostic features of metastatic car-
in attracted, nontumorous, inflammatory cells. Bone cinomas with spindle-cell features and solitary metastases
morphogenetic protein markers are found in sarcomas of of spindle-cell sarcomas are discussed in the section on
bone of various origins. Therefore these markers cannot fibrosarcoma.
be used in distinguishing malignant fibrous histiocytomas
from osteosarcoma.268 Ultrastructural studies show that
Treatment and Behavior
malignant fibrous histiocytoma in bone exhibits similar
features to its soft tissue counterpart.220,222,229,237,266 Data from the National Cancer Institute’s Surveillance,
Extensive use of lineage differentiation markers in Epidemiology, and End Results (SEER) Program (1973-
several recent studies have shown that some malignant 2005) containing more than 1200 cases indicate that
fibrous histiocytomas can be subclassified into several there has been improvement of the 5-year survival rate
categories based on their myogenic, myoepithelial, myo- for patients with malignant fibrous histiocytoma of bone
fibroblastic, lipoblastic, and osteoblastic phenotypes.241 over the past decade. The national 5-year survival rates
At least focal positivity for smooth muscle markers is seen in the 1980s and 1990s were in the range of 30% to
in approximately 50% of these tumors. There is no evi- 40%. The establishment of modern combined surgery
dence however that such subclassification segregates and chemotherapy protocols have resulted in improve-
malignant fibrous histiocytomas into clinical categories ment of the 5-year survival rate to 67%. This represents
with distinct behavior or unique therapeutic response. a substantial improvement when compared to the preche-
Cytogenetically, malignant fibrous histiocytomas motherapy era, when the survival rates of patients treated
show complex chromosomal complements with numer- with surgery alone were less than 20%.214 In general, the
ous structural aberrations and marker chromosomes. survival rates of patients with malignant fibrous histio-
Microarray-based studies disclosed a similar disarray of cytoma of bone and soft tissue are similar. In addition,
genome-wide copy number variations with preferential there is significant overlap between the survival rates
amplifications of 8q24 and overexpression of MYC.258,262 and chemosensitivity between malignant fibrous histiocy-
Surprisingly, the rate of mutation of TP53 and MDM2 toma of bone and osteosarcoma.233 Tumors that arise in
amplification is relatively low and is seen in less than 20% preexisting conditions (e.g., secondary malignant fibrous
of these tumors.234,263 Deletions of 9p21-22 are present histiocytoma) behave in a more aggressive manner than
in both sporatic malignant fibrous histiocytoma and de novo lesions. In this group, a majority of patients
malignant fibrous histiocytoma associated with diaphy- die within 1 to 2 years after diagnosis, and there are no
seal medullary stenosis, but CDKN1A is not a target long-term survivors. In general, malignant fibrous histio-
gene.235,258 cytoma of bone is a highly aggressive tumor with a high
propensity for metastasis. Metastases typically present
in the lungs; it appears that nearly 50% of patients with
Personal Comments
malignant fibrous histiocytoma develop this serious com-
The problem of deciding whether a high-grade pleomor- plication. Metastatic spread to regional lymph nodes
phic spindle-cell sarcoma primary in bone represents an is very unusual. Combined multiple modality treat-
“undifferentiated osteolytic sarcoma” or a malignant ment with chemotherapy, surgery, and adjuvant radio-
fibrous histiocytoma is approached differently on the therapy is a standard approach in the management of
basis of one’s own preferences with regard to the inter- these lesions.214-216 Similar to osteosarcoma, the propor-
pretation of foci of coarse collagenous bundles that tion of necrosis after chemotherapy is a strong prog-
resemble tumor osteoid. Some pathologists tend to nostic factor in the management of malignant fibrous
regard any high-grade pleomorphic spindle-cell sarcoma histiocytoma.252,267
in bone as an undifferentiated osteosarcoma, regardless
of whether tumor bone or osteoid production can be
found. We regard the finding of even small amounts of
definite tumor osteogenesis as tantamount to a diagnosis FIBROSARCOMA
of osteosarcoma. Definition
Fibrosarcoma is a malignant spindle-cell neoplasm that
Differential Diagnosis
exhibits myofibroblastic differentiation. It is distinguished
The same entities discussed in the differential diagnosis from malignant fibrous histiocytoma and fibroblastic
of fibrosarcoma of bone are relevant to this entity, namely osteosarcoma by the absence of osteoid production and
osteosarcoma, fibrosarcoma, desmoplastic fibroma, and meta- by its uniform spindle-cell myofibroblastic features. With
static tumors with spindle-cell features. the recognition in the 1970s that malignant fibrous his-
Fibrosarcoma, which exhibits a herringbone pattern as tiocytoma arising in bone was a separate entity, there was
well as more uniformity of cells, is now largely replaced a sharp decline in the use of this diagnostic designation.
as a diagnostic category by malignant fibrous histiocy- There is some controversy regarding the specificity of the
toma of bone. Fibroblastic osteosarcoma and high-grade term fibrosarcoma of bone, and many authors now tend to
pleomorphic variants of osteosarcoma are distinguished classify these tumors as variants of malignant fibrous his-
from malignant fibrous histiocytoma only by the finding tiocytoma. We retain the description of fibrosarcoma in
of microscopic evidence of unequivocal osteoid and bone in this new edition, but the readers should be aware
tumor bone. that this diagnostic designation is rarely used to define

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 683

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 9-59  ■  Fibrosarcoma. Peak age incidence and frequent sites of skeletal involvement.

spindle-cell malignancies in bone. Modern reports on cell tumor, Paget’s disease, irradiation damage, chronic
fibrosarcoma of bone are extremely rare, and typically osteomyelitis, fibrous dysplasia, and bone infarct. More
they are combined with malignant fibrous histiocytoma recently, secondary sarcomas complicating these condi-
of bone.219,257,276 tions that do not exhibit bone-forming features are
usually classified as malignant fibrous histiocytomas.
Similar to malignant fibrous histiocytoma, rare cases of
Incidence and Location
multifocal skeletal fibrosarcoma were reported.273 The
It is difficult to establish the frequency of this tumor peak age incidence and frequent sites of skeletal involve-
because of different criteria used to establish the diagno- ment are shown in Figure 9-59.
sis. Data from the SEER Program indicates that although
fibrosarcoma was more frequently diagnosed in the past, Clinical Symptoms
it is now exceedingly rarely used as a diagnostic designa-
tion for bone sarcomas. This trend suggests that this Pain is the most common initial symptom of fibrosar-
tumor is combined either with fibroblastic osteosarcoma coma. Swelling and tenderness of the affected area may
or malignant fibrous histiocytoma. It most frequently be present. Pathologic fracture may also be a presenting
involves the metaphyseal portions of long tubular symptom.
bones.269-272,274,277 Similar to conventional osteosarcoma,
nearly 50% of cases occur in the distal femoral and proxi-
Radiographic Imaging
mal tibial metaphyses, but every bone of the skeleton can
be affected.278,279 It is rarely diagnosed in the first decade The lesion is purely lytic and exhibits a destructive growth
of life and is widely distributed from the second through pattern with ill-defined irregular borders (Fig. 9-60).281
seventh decades with a bell-shaped curve. The peak inci- Permeative or moth-eaten patterns of bone destruction
dence is in the fifth decade of life. There is no clear sex may be present. Features of early, sometimes multifo-
predilection, but male patients seem to be slightly more cal, cortical disruption and extension into soft tissue are
frequently affected than female patients. In the past, it present. Periosteal elevation with Codman’s triangles
was reported as a secondary sarcoma complicating giant and periosteal new bone formation also may be present.

ERRNVPHGLFRVRUJ
684 9  Fibrous and Fibrohistiocytic Lesions

A B

C D
FIGURE 9-60  ■  Fibrosarcoma: radiographic and microscopic features. A and B, Anteroposterior and lateral radiographs show
destructive lytic lesion of proximal tibia. C and D, Anaplastic spindle cells arranged in interlacing bundles in high-grade fibrosarcoma.
Note overlapping microscopic features with spindle-cell variant of malignant fibrous histiocytoma. (C and D, ×100.) (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 685

The lesion is most frequently located in the metaphysis. herringbone pattern with few giant cells, either osteoclast
In skeletally immature patients, the growth plate forms or tumor, are designated as fibrosarcomas.
a barrier to extension into the epiphysis, which may If more than a few areas of coarsely hyalinized collagen
be penetrated in more advanced lesions. In skeletally are present, especially exhibiting evidence of mineraliza-
mature patients, the tumor frequently extends into the tion, the tumors are interpreted as fibroblastic osteosarcoma.
epiphysis. At the better-differentiated end of the spectrum of
non–bone-forming spindle-cell tumors, those with little
or no nuclear atypia, absence of mitotic activity, and
Gross Findings
coarser collagen bundles overlap with desmoplastic fibroma.
Fibrosarcoma has a fleshy, fibrous appearance with indis- When well-differentiated cartilage islands are present
tinct irregular borders and foci of tan to gray soft tissue. in what otherwise resembles a fibrosarcoma of bone, it is
Foci with a more myxoid appearance, necrosis, or hemor- important to recognize that the tumor represents a dedif-
rhage can be present. ferentiated chondrosarcoma and not a primary fibrosarcoma
of bone. As with sarcomas that complicate Paget’s disease,
dedifferentiated chondrosarcomas have a poorer progno-
Microscopic Findings
sis than de novo fibrosarcomas, and they tend to occur in
Fibrosarcoma is characterized by the presence of older patients. Similarly, fibrosarcomas can be found in
uniform fibroblast-like spindle cells arranged in parallel association with bone infarcts, and this association should
bundles (Fig. 9-60). The spectrum ranges from well- be verified by radiologic correlation. The underlying
differentiated, heavily collagenized lesions to highly lesion (e.g., infarct) may not be present in histologic
cellular tumors with obvious nuclear atypia and brisk samples and may be detected only on accompanying
mitotic activity.269-272,274,277-282 Microscopic evidence of radiographs.
bone and cartilage matrix production is totally absent Metastatic carcinoma with spindle-cell features can simu-
from these tumors. The hallmark of fibrosarcoma and late fibrosarcoma. The former usually presents in
its most striking feature is the herringbone arrangement an appropriate clinical setting and is associated with mul-
of bundles of spindle cells that are present focally. The tifocal lesions. Diagnosis is facilitated by the use of
tumor may exhibit focal myxoid features as well. Tumors immunohistochemical techniques to demonstrate the
that are predominantly myxoid are sometimes referred to fundamental epithelial nature of the lesion. Occasionally,
as myxofibrosarcoma or myxoid fibrosarcoma of bone. Tradi- a solitary metastatic focus of a spindle-cell sarcoma (e.g.,
tionally, fibrosarcomas have been graded numerically on leiomyosarcoma) can simulate a primary fibrosarcoma of
their degree of pleomorphism and mitotic rates. More bone. The use of appropriate tumor marker techniques
recently, the trend among bone tumor pathologists has can help the clinician avoid this pitfall.
been to avoid the use of the term fibrosarcoma for high-
grade pleomorphic tumors. It is typically reserved for
Treatment and Behavior
better-differentiated lesions with features of grade 1 or
2 fibrosarcoma. The high-grade pleomorphic lesions are In general, purely spindle-cell lesions with overall low-
now almost universally classified as malignant fibrous grade features are considered to be less aggressive than
histiocytoma. Ultrastructurally, these lesions uniformly malignant fibrous histiocytoma but in comparison with
exhibit myofibroblastic differentiation with variable extra- fibromatosis they have definite metastatic potential.275
cellular collagen production.280 The potential overlap of Data from the SEER Program show, however, a signifi-
microscopic features in fibrosarcoma with malignant cant overlap in terms of clinical behavior for patients with
fibrous histiocytoma of bone was mentioned by several fibrosarcoma and malignant fibrous histiocytoma of
investigators early during the development of diagnostic bone. Similar to malignant fibrous histiocytoma, there
concepts for non–bone-forming sarcomas.257,280,282 Immu- has been an improvement of the 5-year survival rate,
nohistochemically, the cytoplasm of the tumor cells is from 37% in the 1970s to 54% during the past decade.
strongly positive for vimentin and exhibits weak positiv- The long-term survival rates for patients with exclusively
ity for smooth muscle markers, such as SMA and muscle low-grade fibrosarcoma are more optimistic; it is esti-
common actin. mated that nearly 80% of them are alive after 10 years.276
There is a tendency to treat low-grade tumors by radical
excision (limb-salvage procedure) or amputation. The
Differential Diagnosis
treatment of high-grade fibrosarcomas does not differ
Malignant fibrous histiocytoma, fibroblastic osteosarcoma, and from that of malignant fibrous histiocytoma.276
desmoplastic fibroma are the three entities that are consid-
ered most often in the differential diagnosis of fibrosar- REFERENCES
coma of bone. In many cases, particularly those diagnosed Nonossifying Fibroma
in recent years, the distinctions between malignant 1. Abdel-Wanis ME, Kawahara N: Skeletal disorders associated with
fibrous histiocytoma and fibrosarcoma have been based skin pigmentation: a role of melatonin? Med Hypotheses 61:640–
on arbitrary criteria rather than on distinct morphologic 642, 2003.
differences. Tumors with a well-defined storiform pattern, 2. Al-Rikabi AC, Ramaswamy JC, Bhat VV: Jaffe-Campanacci syn-
drome. Saudi Med J 26:104–106, 2005.
an admixture of large round histiocytic cells with vacu- 3. Arata MA, Peterson HA, Dahlin DC: Pathological fractures
olated cytoplasm, and bizarre tumor giant cells have been through nonossifying fibromas: review of the Mayo Clinic experi-
termed malignant fibrous histiocytoma. Those exhibiting a ence. J Bone Joint Surg 63A:980–988, 1981.

ERRNVPHGLFRVRUJ
686 9  Fibrous and Fibrohistiocytic Lesions

4. Betsy M, Kupersmith LM, Springfield DS: Metaphyseal fibrous 33. Schwartz AM, Ramos RM: Neurofibromatosis and multiple non-
defects. J Am Acad Orthop Surg 12:89–95, 2004. ossifying fibromas. AJR Am J Roentgenol 135:617–619, 1980.
5. Bhagwandeen SB: Malignant transformation of a non-osteogenic 34. Selby S: Metaphyseal cortical defects in the tubular bones of
fibroma of bone. J Pathol Bacteriol 92:562–564, 1966. growing children. J Bone Joint Surg 43:395–400, 1961.
6. Brassesco MS, Valera ET, Engel EE, et al: Clonal complex chro- 35. Sethi A, Agarwal K, Sethi S, et al: Allograft in the treatment of
mosome averration in non-ossifying fibroma. Pedatr Blood Cancer benign cystic lesions of bone. Arch Orthop Trauma Surg 112:167–
54:764–767, 2010. 170, 1993.
7. Caffey J: On fibrous defects in cortical walls of growing tubular 36. Steiner GC: Fibrous cortical defect and nonossifying fibroma
bones: their radiologic appearance, structure, prevalence, natural of bone. A study of the ultrastructure. Arch Pathol 97:205–210,
course and diagnostic significance. Adv Pediatr 7:13–51, 1955. 1974.
8. Campanacci M, Laus M, Boriani S: Multiple nonossifying fibro- 37. Steinmetz JC, Pilon VA, Lee JK: Jaffe-Campanacci syndrome.
mata with extraskeletal anomalies: a new syndrome? J Bone Joint J Pediatr Orthop 8:602–604, 1988.
Surg 65B:627–632, 1983. 38. Tanaka T, Kobayashi T, Lino M: Transformation of benign
9. Colby RS, Saul RA: Is Jaffe-Campanacci syndrome just a mani- fibrous histiocytoma into malignant fibrous histiocytoma in the
festation of neurofibromatosis type 1? Am J Med Genet 123A:60– mandible: case report. J Oral Maxillofac Surg 69:285–290, 2011.
63, 2003. 39. Tarkkanen M, Kaipainen A, Karaharju E, et al: Cytogenetic study
10. Drennan DB, Maylahn DJ, Fahey JJ: Fractures through large of 249 consecutive patients examined for a bone tumor. Cancer
nonossifying fibromas. Clin Orthop 103:82–88, 1974. Genet Cytogenet 68:1–21, 1993.
11. Easley ME, Kneisl JS: Pathologic fractures through nonossifying 40. Tiedeman JJ, Huurman WW, Connolly JF, et al: Healing of a
fibromas: is prophylactic treatment warranted? J Pediatr Orthop large nonossifying fibroma after grafting with bone matrix and
17:808–813, 1997. marrow: a case report. Clin Orthop 265:302–305, 1991.
12. Evans GA, Park WM: Familial multiple non-osteogenic fibro- 41. Unni KK, Inwards CY: Dahlin’s bone tumors: general aspects and data
mata. J Bone Joint Surg 60B:416–419, 1978. on 11,087 cases, ed 6, Philadelphia, 2010, Lippincott Williams &
13. Glockenberg A, Sobel E, Noel JF: Nonossifying fibroma. Four Wilkins.
cases and review of the literature. J Am Podiatr Med Assoc 87:66– 42. Young JW, Levine AM, Dorfman HD: Case report 293: diagnosis
69, 1997. of nonossifying fibroma (NOF) of the upper tibial diametaphysis,
14. Herrera GA, Reimann BE, Scully TJ, et al: Nonossifying fibroma. with considerable increase in size over a three-year period. Skeletal
Electron microscopic examination of two cases supporting a his- Radiol 12:294–297, 1984.
tiocytic rather than a fibroblastic origin. Clin Orthop Relat Res
167:269–276, 1982. Benign Fibrous Histiocytoma
15. Iagaru A, Henderson R: PET/CT follow-up in nonossifying 43. Balasubramanian C, Rajaraman G, Singh CS, et al: Benign fibrous
fibroma. AJR Am J Roentgenol 187:830–832, 2006. histiocytoma of the sacrum—diagnostic difficulties facing this rare
16. Jaffe HL: Tumors and tumorous conditions of the bones and joints, bone tumor. Pediatr Neurosurg 41:253–257, 2005.
Philadelphia, 1958, Lea & Febiger. 44. Bertoni F, Calderoni P, Bacchini P, et al: Benign fibrous histiocy-
17. Jaffe HL, Lichtenstein L: Non-osteogenic fibroma of bone. Am toma of bone. J Bone Joint Surg 68A:1225–1230, 1986.
J Pathol 28:205–221, 1942. 45. Bertoni F, Unni KK, McLeod RA, et al: Xanthoma of bone. Am
18. Jee WH, Choe BY, Kang HS, et al: Nonossifying fibroma: char- J Clin Pathol 90:377–384, 1988.
acteristics at MR imaging with pathologic correlation. Radiology 46. Ceroni D, Dayer R, De Coulon G, et al: Benign fibrous histiocy-
209:197–202, 1998. toma of bone in a pediatric population: a report of 6 cases. Mus-
19. Katz JF, Marek FM: Case of coexistent benign and malignant bone culoskel Surg 95:107–114, 2011.
tumors. J Mt Sinai Hosp 17:187, 1950. 47. Clark TD, Stelling CB, Fechner RE: Case report 328: benign
20. Klein MH, Rosenberg ZS, Lehman WB: Nonossifying fibroma fibrous histiocytoma of the left 8th rib. Skeletal Radiol 14:149–151,
of bone: a case report. Bull Hosp Joint Dis 50:64–69, 1990. 1985.
21. Kourelis TV, Manola A: Interesting medical images: the Jaffe- 48. Dalinka MK, Turner ML, Thompson JJ, et al: Lipid granuloma-
Campanacci syndrome. Conn Med 76:279–281, 2012. tosis of the ribs: focal Erdheim-Chester disease. Radiology 142:
22. Kyriakos M, Murphy WA: Concurrence of metaphyseal fibrous 297–299, 1982.
defect and osteosarcoma: report of a case and review of the litera- 49. Demiralp B, Kose O, Oguz E, et al: Benign fibrous histiocytoma
ture. Skeletal Radiol 6:179–186, 1981. of the lumbar vertebrae. Skeletal Radiol 38:187–191, 2009.
23. Mandell GA, Dalinka MK, Coleman BG: Fibrous lesions in the 50. Destouet JM, Kyriakos M, Gilula LA: Fibrous histiocytoma
lower extremities in neurofibromatosis. AJR Am J Roentgenol (fibroxanthoma) of a cervical vertebra: a report with a review of
133:1135–1138, 1979. the literature. Skeletal Radiol 5:241–246, 1980.
24. Mankin HJ, Trahan CA, Fondren G, et al: Non-ossifying fibroma, 51. Grohs JG, Nicolakis M, Kainberger F, et al: Benign fibrous his-
fibrous cortical defect and Jaffe-Campanacci syndrome: a biologic tiocytoma of bone: a report of 10 cases and review of literature.
and clinical review. Chir Degli Organi Movimento 93:1–7, 2009. Wiener Linische Wochenschrift 114:56–63, 2002.
25. Mirra JM: Case records of the Massachusetts General Hospital: 52. Heo MS, Cho JH, Kwon KJ, et al: Benign fibrous histiocytoma
case 1-1986. N Engl J Med 314:105–113, 1986. in the mandible. Oral Surg Oral Med Oral Pathol Oral Radiol
26. Mirra JM, Gold RH, Rand F: Disseminated nonossifying fibromas Endodont 97:276–280, 2004.
in association with café-au-lait spots (Jaffe-Campanacci syn- 53. Hermann G, Steiner GC, Sherry HH: Case report 465: benign
drome). Clin Orthop 168:192–205, 1982. fibrous histiocytoma (BFH). Skeletal Radiol 17:195–198, 1988.
27. Moser RP, Jr, Sweet DE, Haseman DB, et al: Multiple skeletal 54. Ideguchi M, Kajiwara K, Yoshikawa K, et al: Benign fibrous his-
fibroxanthomas: radiologic-pathologic correlation of 72 cases. tiocytoma of the skull with increased intracranial pressure caused
Skeletal Radiol 16:353–359, 1987. by cerebral venous sinus occlusion. J Neurosurg 111:504–508,
28. Nelson M, Perry D, Ginsburg G, et al: Translocation (1;4) 2009.
(p31;q34) in nonossifying fibroma. Cancer Genet Cytogenet 142: 55. Katagiri W, Nakazawa M, Kishino M: Benign fibrous histiocy-
142–144, 2003. toma in the condylar process of the mandible: case report. Br J
29. Peterson HA, Fitzgerald EM: Fractures through nonossifying Oral Maxillofacial Surg 46:e1–e2, 2008.
fibromata in children. Minn Med 63:139–144, 1980. 56. Kishino M, Murakami S, Toyosawa S, et al: Benign fibrous
30. Ponseti IV, Friedman B: Evolution of metaphyseal fibrous defects. histioctyoma of the mandible. J Oral Pathol Med 34:190–192,
J Bone Joint Surg 31:582, 1949. 2005.
31. Ritschl P, Hajek PC, Pechmann U: Fibrous metaphyseal defects: 57. Kuruvath S, O’Donovan DG, Aspoas AR, et al: Benign fibrous
magnetic resonance imaging appearances. Skeletal Radiol 18:253– histiocytoma of the thoracic spine: case report and review of the
259, 1989. literature. J Neurosurg Spine 4:260–264, 2006.
32. Ritschl P, Karnel F, Hajek P: Fibrous metaphyseal defects: deter- 58. Magliato HJ, Nastasi A: Non-osteogenic fibroma occurring in
mination of their origin and natural history using a radiomorpho- the ilium: report of a case. J Bone Joint Surg 49A:384–386,
logical study. Skeletal Radiol 17:8–15, 1988. 1967.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 687

59. Matsuno T: Benign fibrous histiocytoma involving the ends of 84. Hallor KH, Mertens F, Jin Y, et al: Fusion of the EWSR1 and
long bone. Skeletal Radiol 19:561–566, 1990. ATF1 genes without expression of the MITF-M transcript in
60. Onovo OD, Eyesan SU, Abdulkareen FB: Benign fibrous histio- angiomatoid fibrous histiocytoma. Gene Chromosome Cancer 44:
cytoma of a metacarpal bone. BMJ Case Reports 2011 [Epub ahead 97–102, 2005.
of print]. 85. Jain S, Xu R, Prieto VG, et al: Molecular classification of soft
61. Roessner A, Immenkamp M, Weidner A, et al: Benign fibrous tissue sarcomas and its clinical applications. Int J Clin Exp Pathol
histiocytoma of bone: light- and electron-microscopic observa- 3:416–429, 2010.
tions. J Cancer Res Clin Oncol 101:191–202, 1981. 86. Kay S: Angiomatoid malignant fibrous histiocytoma: report of two
62. Saito R, Caines MJ: Atypical fibrous histiocytoma of the humerus: cases with ultrastructural observations of one case. Arch Pathol Lab
a light and electron microscopic study. Am J Clin Pathol 68:409– Med 109:934–937, 1985.
415, 1977. 87. Leu HJ, Makek M: Angiomatoid malignant fibrous histiocytoma.
63. Sanatkumar S, Rajagoplan N, Mallikarjunaswamy B, et al: Benign Arch Pathol Lab Med 110:466–467, 1986. (letter).
fibrous histioctyoma of the distal radius with congenital disloca- 88. Mangham DC, Williams A, Lalam RK, et al: Angiomatoid fibrous
tion of the radial head: a case report. J Orthop Surg 13:83–87, histiocytoma of bone: a calcifying sclerosing variant mimicking
2005. osteosarcoma. Am J Surg Pathol 34:279–285, 2010.
64. Tanaka T, Kobayashi T, Iino M: Transformation of benign 89. Matsumura T, Yamaguchi T, Tochigi N, et al: Angiomatoid
fibrous histiocytoma into malignant fibrous histioctyoma in the fibrous histiocytoma including cases with pleomorphic features
mandible: case report. J Oral Maxillofacial Surg 69:e285–e290, analyzed by fluorescence in situ hybridization. J Clin Pathol
2011. 63:124–128, 2010.
65. Tubbs RS, Kelly DR, Pugh JL, et al: Benign fibrous histiocy- 90. Petrey WB, LeGallo RD, Fox MG, et al: Imaging characteristics
toma of the skull base. Case report, J Neurosurg 106:65–67, of angiomatoid fibrous histiocytoma of bone. Skeletal Radiol 40:
2007. 233–237, 2011.
66. van Giffen NH, van Rhijn LW, van Ooij A, et al: Benign fibrous 91. Pettinato G, Manivel JC, De Rosa G, et al: Angiomatoid malig-
histiocytoma of the posterior arch of C1 in a 6-year-old boy: nant fibrous histiocytoma: cytologic, immunohistochemical, ultra-
a case report. Spine 28:E359–E363, 2003. structural, and flow cytometric study of 20 cases. Mod Pathol 3:
479–487, 1990.
Fibromyxoma 92. Rossi S, Szuhai K, Ijszenga M, et al: EWSR1-CREB1 and EWSR1-
67. Abdelwahab IF, Hermann G, Klein MJ, et al: Fibromyxoma of ATF1 fusion genes in angiomatoid fibrous histiocytoma. Clin
bone. Skeletal Radiol 20:95–98, 1991. Cancer Res 13:7322–7328, 2007.
68. Caballes RL: Fibromyxoma of bone. Radiology 130:97–99, 1979. 93. Shao L, Singh V, Cooley L: Angiomatoid fibrous histiocytoma
69. Hardes J, Scheil-Bertram S, Gosheger G, et al: Fibromyxoma of with t(2,22)(q33;q12.2) and EWSR1 gene rearrangement. Pediatr
bone: a case report and review of the literature. Acta Orthop Belg Develop Pathol 12:143–146, 2009.
72:100–104, 2006. 94. Song JY, Lee SK, Kim SG, et al: Angiomatoid fibrous histiocy-
70. Infante-Cossio P, Martinez-de-Fuentes R, Garcia-Perla-Garcia A, toma on the hard palate: case report. Oral Maxillofacial Surg
et al: Myxofibroma of the maxilla. Reconstruction with iliac crest 16:237–242, 2012.
graft and dental implants after tumor resection. Med Oral, Patol 95. Smith ME, Costa MJ, Weiss SW: Evaluation of CD68 and other
Oral Cir Bucal 16:e532–e536, 2011. histiocytic antigens in angiomatoid malignant fibrous histiocy-
71. Kuhne CA, Engelhorn T, Homann M, et al: Fibromyxoma of the toma. Am J Surg Pathol 15:757–763, 1991.
iliac wing. Skeletal Radiol 32:170–173, 2003. 96. Sun CC, Jr, Toker C, Breitenecker R: An ultrastructural study of
72. Marcove RC, Kambolis C, Bullough PG, et al: Fibromyxoma of angiomatoid fibrous histiocytoma. Cancer 49:2103–2111, 1982.
bone: a report of three cases. Cancer 17:1209–1213, 1964. 97. Tanas MR, Rubin BP, Montgomery EA, et al: Utility of FISH in
73. Marcove RC, Lindeque BG, Huvos AG: Fibromyxoma of the the diagnosis of angiomatoid fibrous histioctyoma: a series of 18
bone. Surg Gynecol Obstet 169:115–118, 1989. cases. Mod Pathol 23:93–97, 2010.
74. Mavrogenis AF, Casadei R, Gambarotti M, et al: Fibromyxoma of 98. Thway K, Stefanaki K, Papadakis V, et al: Metastatic angiomatoid
the axis. Orthop 35:e1133–e1135, 2012. fibrous histiocytoma of the scalp, with EWSR1-CREB1 gene
75. McClure DK, Dahlin DC: Myxoma of bone: report of three cases. fusion in primary tumor and nodal metastasis. Hum Pathol 44:289–
Mayo Clin Proc 52:249–253, 1977. 293, 2013.
99. Thway K, Fisher C: Tumors with EWSR1-CREB1 and EWSR1-
Angiomatoid Fibrous Histiocytoma ATF1 fusions: the current status. Am J Surg Pathol 36:e1–e11,
76. Antonescu CR, Dal Cin P, Nafa K, et al: EWSR1-CREB1 is the 2012.
predominant gene fusion in angiomatoid fibrous histioctyoma. 100. Thway K: Angiomatoid fibrous histiocytoma. Arch Pathol Lab Med
Gene Chromosome Cancer 46:1051–1060, 2007. 132:273–277, 2008.
77. Chen G, Folpe AL, Colby TV, et al: Angiomatoid fibrous histio- 101. Tornoczky T, Bogner B, Krausz T, et al: Angiomatoid fibrous
cytoma: unusual sites and unusual morphology. Mod Pathol 24: histiocytoma: pleomorphic variant associated with multiplication
1560–1570, 2011. of EWSR1-CREB1 fusion gene. Pathol Oncol Res 18:545–548,
78. Costa MJ, Weiss SW: Angiomatoid malignant fibrous histiocy- 2012.
toma: a follow-up study of 108 cases with evaluation of possible 102. Waters BL, Panagopoulos I, Allen EF: Genetic characterization
histologic predictors of outcome. Am J Surg Pathol 14:1126–1132, of angiomatoid fibrous histiocytoma identifies fusion of the FUS
1990. and ATF-1 genes induced by a chromosomal translocation involv-
79. Dunham C, Husson J, Seiff M, et al: Primary intracerebral angio- ing bands 12q13 and 16p11. Cancer Genet Cytogenet 121:109–116,
matoid fibrous histiocytoma: report of a case with t(12;22) 2000.
(q13;q12) causing type 1 fusion of the EWS and ATF-1 genes. Am 103. Wegmann W, Heitz PU: Angiomatoid malignant fibrous histio-
J Surg Pathol 32:478–484, 2008. cytoma: evidence for the histiocytic origin of tumor cells. Virchows
80. El-Naggar AK, Ro JY, Ayala AG, et al: Angiomatoid malignant Arch [Pathol Anat] 406:59–66, 1985.
fibrous histiocytoma: flow cytometric DNA analysis of six cases.
J Surg Oncol 40:201–204, 1989. Skeletal Fibromatoses
81. Enzinger FM: Angiomatoid malignant fibrous histiocytoma: a
distinct fibrohistiocytic tumor of children and young adults simu- Desmoplastic Fibroma
lating a vascular neoplasm. Cancer 44:2147–2157, 1979. 104. Amary MF, Pauwels P, Meulemans E, et al: Detection of beta-
82. Filingeri V, Gravante G, Marino B, et al: A rare case of cystic catenin mutations in paraffin-embedded sporadic desmoids-type
variety of angiomatoid fibrous histiocytoma. Eur Rev Med Phar- fibromatosis by mutation-specific restriction enzyme digestion
malog Sci 14:887–890, 2010. (MSRED): an ancillary diagnostic tool. Am J Surg Pathol 31:1299–
83. Fletcher CD: Angiomatoid “malignant fibrous histiocytoma”: an 1309, 2007.
immunohistochemical study indicative of myoid differentiation. 105. Bertoni F, Calderoni P, Bacchini P, et al: Desmoplastic fibroma of
Hum Pathol 22:563–568, 1991. bone: a report of six cases. J Bone Joint Surg Br 66:265–268, 1984.

ERRNVPHGLFRVRUJ
688 9  Fibrous and Fibrohistiocytic Lesions

106. Bhattacharya B, Dilworth HP, Iacobuzio-Donahue C, et al: 132. Lips DJ, Barker N, Clevers H, et al: The role of APC and beta-
Nuclear beta-catenin expression distinguishes deep fibromatosis catenin in the aetiology of aggressive fibromatosis (desmoids
from other benign and malignant fibroblastic and myofibroblastic tumors). Eur J SurgOncol 35:3–10, 2009.
lesions. Am J Surg Pathol 29:653–659, 2005. 133. Makek M, Lello GE: Desmoplastic fibroma of the mandible: lit-
107. Bo N, Wang D, Wu B, et al: Analysis of catenin expression and erature review and report of three cases. J Oral Maxillofac Surg
exon 3 mutations in pediatric sporadic aggressive fibromatosis. 44:385–391, 1986.
Pediatr Devel Pathol 15:173–178, 2012. 134. Marks KE, Bauer TW: Fibrous tumors of bone. Orthop Clin North
108. Callahan KS, Eberhardt SC, Fechner RE, et al: Desmoplastic Am 20:377–393, 1989.
fibroma of bone with extensive cartilaginous metaplasia. Ann 135. Rabhan WN, Rosai J: Desmoplastic fibroma: report of ten
Diagn Pathol 10:3–346, 2006. cases and review of the literature. J Bone Joint Surg 50A:487–502,
109. Cohen P, Goldenberg RR: Desmoplastic fibroma of bone. J Bone 1968.
Joint Surg 47A:1620–1625, 1965. 136. Selfa-Moreno S, Arana-Fernández E, Fernández-Latorre F, et al:
110. Cranin AN, Gallo L, Madan S: Desmoplastic fibroma: a rare oral Desmoplastic fibroma of the skull—case report. J Neurosurg
tumor in children. NY State Dent J 60:34–39, 1994. 82:119–120, 1995.
111. Dahlin DC, Hoover NW: Desmoplastic fibroma of bone. JAMA 137. Sleeman DJ, Paterson A, Eveson JW: Desmoplastic fibroma of
188:685–687, 1964. the maxillary alveolus. Eur J Cancer Oral Oncol 29B:151–152,
112. Domont J, Salas S, Lacroix L, et al: High frequency of beta- 1993.
catenin heterozygous mutations in extra-abdominal fibromatosis: 138. Stout AP: Fibrous tumors of the soft tissues. Minn Med 43:455,
a potential molecular tool for disease management. Br J Cancer 1960.
102:1032–1036, 2010. 139. Stout AP: The fibromatoses. Clin Orthop 19:11, 1961.
113. Dong PR, Seeger LL, Eckardt JJ, et al: Case report 847: juxtacor- 140. Sugiura I: Desmoplastic fibroma: case report and review of the
tical aggressive fibromatosis (desmoplastic fibroma) of the forearm. literature. J Bone Joint Surg 58A:126–130, 1976.
Skeletal Radiol 23:560–563, 1994. 141. Taconis WK, Schutte HE, van der Heul RO: Desmoplastic
114. Eisen MZ, Butler HE: Desmoplastic fibroma of the maxilla: fibroma of bone: a report of 18 cases. Skeletal Radiol 23:283–288,
report of a case. J Am Dent Assoc 108:608–609, 1984. 1994.
115. Frick MA, Sundaram M, Unni KK, et al: Imaging findings in 142. Tjandra SS, Hsu C, Goh YI, et al: IFN-beta signaling positively
desmoplastic fibroma of bone: distinctive T2 characteristics. Am regulates tumorigenesis in aggressive fibromatosis, potentially by
J Roentgenol 184:1762–1767, 2005. modulating mesenchymal progenitors. Cancer Res 67:7124–7131,
116. Gebert C, Hardes J, Kersting C, et al: Expression of beta-catenin 2007.
and p53 are prognostic factors in deep aggressive fibromatosis. 143. Trombetta D, Macchia G, Mandahl N, et al: Molecular genetic
Histopathology 50:491–497, 2007. characterization of the 11q13 breakpoint in a desmoplastic
117. Gebhardt MC, Campbell CJ, Schiller AL, et al: Desmoplastic fibroma of bone. Cancer Genet 205:410–403, 2012.
fibroma of bone: a report of eight cases and review of the litera- 144. West R, Huvos AG, Lane JM: Desmoplastic fibroma of bone
ture. J Bone Joint Surg 67A:732–747, 1985. arising in fibrous dysplasia. Am J Clin Pathol 79:630–633, 1983.
118. Greenspan A, Unni KK: Case report 787: desmoplastic fibroma. 145. Young JW, Aisner SC, Levine AM, et al: Computed tomography
Skeletal Radiol 22:296–299, 1993. of desmoid tumors of bone: desmoplastic fibroma. Skeletal Radiol
119. Grigoryan T, Wend P, Klaus A, et al: Deciphering the function of 17:333–337, 1988.
canonical Wnt signals in development and disease: conditional
loss- and gain-of-function mutations of beta-catenin in mice. Juvenile Multifocal Myofibromatosis
Genes Devel 22:2308–2341, 2008. 146. Alaggio R, Barisanni D, Ninfo V, et al: Morphologic overlap
120. Haney J, Olson PN, Griffiths HJ: Radiologic case study: the clini- between infantile myofibromatosis and infantile fibrosarcoma: a
cal and radiologic features of desmoplastic fibroma of bone. Ortho- pitfall in diagnosis. Pediatr Dev Pathol 11:355–362, 2008.
pedics 17:77, 80-85, 88, 1994. 147. Arcangeli F, Calista D: Congenital myofibromatosis in two sib-
121. Hauben EI, Jundt G, Cleton-Jansen AM, et al: Desmoplastic lings. Eur J Dermatol 16:181–183, 2006.
fibroma of bone: an immunohistochemical study including beta- 148. Asirvatham R, Moreau PG, Antonius JI: Solitary infantile myofi-
catenin expression and mutational analysis for beta-catenin. Hum bromatosis of axis: a case report. Spine 19:80–82, 1994.
Pathol 36:1025–1030, 2005. 149. Bolano LE, Yngve DA, Altshuler G: Solitary fibromatosis of bone:
122. Hillmann JS, Mesgarzadeh M, Tang CK, et al: Case report 481: a rare variant of congenital generalized fibromatosis. Clin Orthop
benign intraosseous fibroma (desmoplastic fibroma) associated 263:238–241, 1991.
with Paget disease of the iliac bone. Skeletal Radiol 17:356–359, 150. Coffin CM, Dehner LP: Fibroblastic-myofibroblastic tumors in
1988. children and adolescents: a clinicopathologic study of 108 exam-
123. Hinds EC, Kent JN, Fechner RE: Desmoplastic fibroma of the ples in 103 patients. Pediatr Pathol 11:569–588, 1991.
mandible: report of a case. J Oral Surg 27:271–274, 1969. 151. Davies RS, Carty H, Pierro A: Infantile myofibromatosis: a review.
124. Huang PW, Tzen CY: Prognostic factors in desmoid-type fibro- Br J Radiol 67:619–623, 1994.
matosis: a clinicopathological and immunohistochemical analysis 152. Disler DG, Alexander AA, Mankin HJ, et al: Multicentric fibro-
of 46 cases. Pathology 42:147–150, 2010. matosis with metaphyseal dysplasia. Radiology 187:489–492, 1993.
125. Inwards CY, Unni KK, Beabout JW, et al: Desmoplastic fibroma 153. Fukasawa Y, Ishikura H, Takada A, et al: Massive apoptosis in
of bone. Cancer 68:1978–1983, 1991. infantile myofibromatosis: a putative mechanism of tumor regres-
126. Ippolito V, Mirra JM, Motta C, et al: Case report 771: melorhe- sion. Am J Pathol 144:480–485, 1994.
ostosis in association with desmoid tumor. Skeletal Radiol 22:284– 154. Hartig G, Koopmann C, Jr, Esclamado R: Infantile myofibroma-
288, 1993. tosis: a commonly misdiagnosed entity. Otolaryngol Head Neck
127. Jilong Y, Jian W, Xiaoyan Z, et al: Analysis of APC/beta-catenin Surg 109:753–757, 1993.
genes mutations and Wnt signaling pathway in desmoid-type 155. Heiple KG, Perrin E, Aikawa M: Congenital generalized fibro-
fibromatosis. Pathology 39:319–325, 2007. matosis: a case limited to osseous lesions. J Bone Joint Surg
128. Kumar R, Madewell JE, Lindell MM, et al: Fibrous lesions of 54A:663–669, 1972.
bones. Radiographics 10:237–256, 1990. 156. Kindblom LG, Termen G, Save-Soderbergh J, et al: Congenital
129. Lacroix-Triki M, Geyer FC, Lambros MB, et al: β-catenin/Wnt solitary fibromatosis of soft tissues, a variant of congenital general-
signaling pathway in fibromatosis, metaplastic carcinomas and ized fibromatosis: two case reports. Acta Pathol Microbiol Scand
phyllodes tumours of the breast. Mod Pathol 23:1438–1448, 2010. 85:640, 1977.
130. Lagace R, Delage C, Bouchard HL, et al: Desmoplastic fibroma 157. Liew S, Haynes M: Localized form of congenital generalized
of bone: an ultrastructural study. Am J Surg Pathol 3:423–430, fibromatosis: a report of three cases with myofibroblasts. Pathology
1979. 13:257–266, 1981.
131. Lazar AJ, Tuvin D, Hajibashi S, et al: Specific mutations in the 158. Lin JJ, Svoboda DJ: Multiple congenital mesenchymal tumors:
beta-catenin gene (CTNNB1) correlate with local recurrence in multiple vascular leiomyomas in several organs of a newborn.
sporadic desmoids tumors. Am J Pathol 173:1518–1527, 2008. Cancer 28:1046–1053, 1971.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 689

159. Mentzel T, Calonje E, Nascimento AG, et al: Infantile hemangio- 184. De Braekeleer E, Douet-Guilbert N, Morel F, et al: ETV6 fusion
pericytoma versus infantile myofibromatosis: study of a series sug- genes in hematological malignancies: A review. Leukemia Res 36:
gesting a continuous spectrum of infantile myofibroblastic lesions. 945–961, 2012.
Am J Surg Pathol 18:922–930, 1994. 185. Ferrari A, Orbach D, Sultan I, et al: Neonatal soft tissue sarcomas.
160. Orozco-Covarrubias L, Soriano-Hernandez Y, Duran-McKinster Semin Fetal Neonat Med 17:231–238, 2012.
C, et al: Infantile myofibromatosis: a cause of leg length discrep- 186. Fisher C: Soft tissue sarcomas with non-EWS translocations:
ancy. Pediatr Dermatol 19:520–522, 2002. molecular genetic features and pathologic and clinical correla-
161. Sirvent N, Perrin C, Lacour JP, et al: Monosomy 9q and trisomy tions. Virchows Arch 456:153–166, 2010.
16q in a case of congenital solitary infantile myofibromatosis. 187. Knezevich SR, McFadden DE, Tao W, et al: A novel ETV6-
Virchows Arch 445:537–540, 2004. NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet
162. Sonoda T, Itami S, Seguchi S, et al: Infantile myofibromatosis: 18:184–187, 1998.
report of two cases. J Dermatol 21:508–513, 1994. 188. Kynaston JA, Malcolm AJ, Craft AW, et al: Chemotherapy in the
163. Spadola L, Anooshiravani M, Sayegh Y, et al: Generalized infan- management of infantile fibrosarcoma. Med Pediatr Oncol 21:488–
tile myofibromatosis with intracranial involvement: imaging find- 493, 1993.
ings in a newborn. Pediatr Radiol 32:872–874, 2002. 189. Lannon CL, Sorensen PHB: ETV6-NTRK3: a chimeric protein
164. Stenman G, Nadal N, Persson S, et al: del(6)(q12;q15) as the sole tyrosine kinase with transformation activity in multiple cell lin-
cytogenetic anomaly in a case of solitary infantile myofibromato- eages. Semin Cancer Biol 15:215–223, 2005.
sis. Oncol Rep 6:1101–1104, 1999. 190. Madden NP, Spicer RD, Allibone EB, et al: Spontaneous regres-
165. Stout AP: Juvenile fibromatoses. Cancer 7:953, 1954. sion of neonatal fibrosarcoma. Br J Cancer Suppl 18:S72–S75,
166. Walts AE, Asch M, Raj C: Solitary lesion of congenital fibroma- 1992.
tosis: a rare cause of neonatal intestinal obstruction. Am J Surg 191. Matev I, Stoytscheff K: Congenital sarcoma in the forearm: a long
Pathol 6:255–260, 1982. term follow-up of a case. J Hand Surg 16B:346–348, 1991.
167. Wright C, Corbally MT, Hayes R, et al: Multi-focal infantile 192. McCahon E, Sorensen PH, Davis JH, et al: Non-resectable
myofibromatosis and generalized fibromuscular dysplasia in a congenital tumors with the ETV6-NTRK3 gene fusion are
child: evidence for a common pathologic process. Pediatr Devel highly responsive to chemotherapy. Med Pediatr Oncol 40:288–
Pathol 7:385–390, 2004. 292, 2003.
168. Zhou DB, Zhao JZ, Zhang D, et al: Multicentric infantile myofi- 193. Miettinen M: From morphological to molecular diagnosis of soft
bromatosis: a rare disorder of the calvarium. Acta Neurochir tissue tumors. Adv Experl Med Biol 587:99–113, 2006.
151:641–646, 2009. 194. Mnif H, Zrig M, Maazoun K, et al: Congenital infantile fibrosar-
coma of the forearm. Chir Main 30:148–151, 2011.
Congenital Fibromatosis–like Fibrosarcoma 195. Muzumdar D, Michaud J, Ventureyra EC: Primary giant congeni-
(Infantile Fibrosarcoma) tal infantile fibrosarcoma of the scalp: case report and review of
169. Adam LR, Davison EV, Malcolm AJ, et al: Cytogenetic analysis literature. Childs Nervous System 22:300–304, 2006.
of a congenital fibrosarcoma. Cancer Genet Cytogenet 52:37–41, 196. Orbach D, Rey A, Cecchetto G, et al: Infantile fibrosarcoma:
1991. management based on the European experience. J Clin Oncol
170. Argani P, Fritsch M, Kadkol SS, et al: Detection of the ETV6- 28:318–323, 2010.
NTRK3 chimeric RNA of infantile fibrosarcoma/cellular congeni- 197. Pinto A, Dold OR, Mueller D, et al: Pathological cases of
tal mesoblastic nephroma in paraffin-embedded tissue: application the month: infantile fibrosarcoma. Am J Dis Child 147:691–692,
to challenging pediatric renal stromal tumors. Mod Pathol 13:29– 1993.
36, 2000. 198. Rizkalla H, Wildgrove H, Quinn F, et al: Congenital fibrosarcoma
171. Argyle JC, Tomlinson GE, Stewart D, et al: Ultrastructural of the ileum: case report with molecular confirmation and litera-
immunocytochemical, and cytogenetic characterization of a large ture review. Fetal Pediatr Pathol 30:156–160, 2011.
congenital fibrosarcoma. Arch Pathol Lab Med 116:972–975, 1992. 199. Robinson W, Crawford AH: Infantile fibrosarcoma: report of a
172. Balsaver AM, Butler JJ, Martin RG: Congenital fibrosarcoma. case with long-term follow-up. J Bone Joint Surg Am 72:291–294,
Cancer 20:1607–1616, 1967. 1990.
173. Ben Arush MW, Ben Arie Y, Bialik V, et al: Limb congenital 200. Sachdev R, Singhal N, Mandal AK: Congenital fibrosarcoma of
fibrosarcoma: report of two cases and review of the literature. the upper extremity—a case report and review of the literature.
Pediatr Hematol Oncol 10:357–361, 1993. Ind J Pathol Microbiol 48:474–476, 2005.
174. Bernado L, Admella C, Lucaya J, et al: Infantile fibrosarcoma of 201. Salloum E, Caillaud JM, Flamant F, et al: Poor prognosis infantile
femur. Pediatr Pathol 7:201–207, 1987. fibrosarcoma with pathologic features of malignant fibrous his-
175. Bernstein R, Zeltzer PM, Lin F, et al: Trisomy 11 and other tiocytoma after local recurrence. Med Pediatr Oncol 18:295–298,
nonrandom trisomies in congenital fibrosarcoma. Cancer Genet 1990.
Cytogenet 78:82–86, 1994. 202. Sankary S, Dickman PS, Wiener E, et al: Consistent numerical
176. Bourgeois JM, Knezevich SR, Mathers JA, et al: Molecular detec- chromosome aberrations in congenital fibrosarcoma. Cancer Genet
tion of the ETV6-NTRK3 gene fusion differentiates congenital Cytogenet 65:152–156, 1993.
fibrosarcoma from other childhood spindle cell tumors. Am J Surg 203. Schofield DE, Fletcher JA, Grier HE, et al: Fibrosarcoma in
Pathol 24:937–946, 2000. infants and children: application of new techniques. Am J Surg
177. Chi HT, Ly BT, Kano Y, et al: ETV6-NTRK3 as a therapeutic Pathol 18:12–24, 1994.
target of small molecule inhibitor PKC412. Biochem Biophys Res 204. Soule EH, Pritchard DJ: Fibrosarcoma in infants and children: a
Comm 429:87–92, 2012. review of 110 cases. Cancer 40:1711–1721, 1977.
178. Chung EB, Enzinger FM: Infantile fibrosarcoma. Cancer 38:729– 205. Steelman C, Katzenstein H, Parham D, et al: Unusual presenta-
739, 1976. tion of congenital infantile fibrosarcoma in seven infants with
179. Coffin CM, Dehner LP: Soft tissue tumors in first year of life: molecular-genetic analysis. Fetal Pediatr Pathol 30:329–337, 2011.
a report of 190 cases. Pediatr Pathol 10:509–526, 1990. 206. Strehl S, Ladenstein R, Wrba F, et al: Translocation (12;13) in a
180. Coffin CM, Jaszcz W, O’Shea PA, et al: So-called congenital- case of infantile fibrosarcoma. Cancer Genet Cytogenet 71:94–96,
infantile fibrosarcoma: does it exist and what is it? Pediatr Pathol 1993.
14:133–150, 1994. 207. Tarsitano BF: Oral infantile fibrosarcoma: report of a case. J Oral
181. Corsi A, Boldrini R, Bosman C: Congenital-infantile fibrosar- Maxillofac Surg 48:1228–1230, 1990.
coma: study of two cases and review of the literature. Tumori 208. Ummat S, Nasser JG: Fibrosarcoma of the infratemporal fossa
80:392–400, 1994. in childhood: a challenging problem. J Otolaryngol 21:441–446,
182. Dahlin DC: Case report 189: infantile fibrosarcoma (congenital 1992.
fibrosarcoma–like fibromatosis). Skeletal Radiol 8:77–78, 1982. 209. Vinnicombe SJ, Hall CM: Infantile fibrosarcoma: radiological and
183. Dal Cin P, Brock P, Casteels-Van Daele M, et al: Cytogenetic clinical features. Skeletal Radiol 23:337–341, 1994.
characterization of congenital or infantile fibrosarcoma. Eur J 210. Weiss S, Goldblum JR: Enzinger and Weiss’s soft tissue tumors,
Pediatr 150:579–581, 1991. ed 4, St. Louis, 2001, Mosby, Inc.

ERRNVPHGLFRVRUJ
690 9  Fibrous and Fibrohistiocytic Lesions

211. Wong CH, Dietrich RB, Pais MJ, et al: Pediatric case of the day: ity at chromosome bands 9p21-22—evidence for a common
congenital, undifferentiated retroperitoneal fibrosarcoma. Radio- genetic defect. Gene Chromosome Cancer 27:191–195, 2000.
graphics 14:1428–1430, 1994. 236. Martignetti JA, Desnick RJ, Aliprandis E, et al: Diaphyseal med-
ullary stenosis with malignant fibrous histiocytoma: a hereditary
Malignant Fibrous Histiocytoma bone dysplasia/cancer syndrome maps to 9p21-22. Am J Hum
212. Antonescu CR, Erlandson RA, Huvos AG: Primary fibrosarcoma Genet 64:801–807, 1999.
and malignant fibrous histiocytoma of bone—a comparative ultra- 237. Martorell M, Calabuig C, Peydro-Olaya A, et al: Fibroblast and
structural study: evidence of a spectrum of fibroblastic differentia- myofibroblast participation in malignant fibrous histiocytoma
tion. Ultrastruct Pathol 24:83–91, 2000. (MFH) of bone: ultrastructural study of eight cases with immu-
213. Arnold WH: Hereditary bone dysplasia with sarcomatous degen- nohistochemical support. Pathol Res Pract 184:582–590, 1989.
eration. Study of a family. Ann Intern Med 78:902–906, 1973. 238. Matushansky I, Charytonowicz E, Mills J, et al: MFH classifica-
214. Bacci G, Springfield D, Capanna R, et al: Adjuvant chemotherapy tion: differentiating undifferentiated pleomorphic sarcoma in the
for malignant fibrous histiocytoma in the femur and tibia. J Bone 21st century. Exp Rev Anticancer Ther 9:1135–1144, 2009.
Joint Surg 67A:620–625, 1985. 239. Matushansky I, Hernando E, Socci ND, et al: Derivation of sar-
215. Bielack SS, Schroeders A, Fuchs N, et al: Malignant fibrous his- comas from mesenchymal stem cells via inactivation of the Wnt
tiocytoma of bone: a retrospective EMSOS study of 125 cases. pathway. J Clin Invest 117:3248–3257, 2007.
European Musculo-Skeletal Oncology Society. Acta Orthop Scand 240. McCarthy EF, Matsuno T, Dorfman HD: Malignant fibrous his-
70:353–360, 1999. tiocytoma of bone: a study of 35 cases. Hum Pathol 10:57–70,
216. Bramwell VH, Steward WP, Nooij M, et al: Neoadjuvant chemo- 1979.
therapy with doxorubicin and cisplatin in malignant fibrous his- 241. Mertens F, Romeo S, Bovee JVMG, et al: Reclassification
tiocytoma of bone: A European Osteosarcoma Intergroup study. and subtyping of so-called malignant fibrous histiocytoma of
J Clin Oncol 17:3260–3269, 1999. bone: comparison with cytogenetic features. Clin Sarcoma Res
217. Capanna R, Bertoni F, Bacchini P, et al: Malignant fibrous histio- 1:10, 2011.
cytoma of bone: the experience at the Rizzoli Institute: report of 242. Molenaar WM, van den Berg E, Veth RP, et al: Tumor progres-
90 cases. Cancer 54:177–187, 1984. sion in a giant-cell type malignant fibrous histiocytoma of bone:
218. Chen KT: Multiple fibroxanthosarcoma of bone. Cancer 42:770– clinical, radiologic, histologic, and cytogenetic evidence. Gene
773, 1978. Chromosome Cancer 10:66–70, 1994.
219. Collini P, Sorensen PH, Patel S, et al: Sarcomas with spindle cell 243. Montgomery E, Fisher C: Myofibroblastic differentiation in
morphology. Semin Oncol 36:324–337, 2009. malignant fibrous histiocytoma (pleomorphic myofibrosarcoma):
220. Dahlin DC, Unni KK, Matsuno T: Malignant (fibrous) histiocy- a clinicopathological study. Histopathology 38:499–509, 2001.
toma of bone: fact or fancy? Cancer 39:1508–1516, 1977. 244. Muroya K, Nishimura G, Douya H, et al: Diaphyseal medullary
221. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210, stenosis with malignant fibrous histiocytoma: further evidence for
1995. loss of heterozygosity involving 9p21-22 in tumor tissue. Gene
222. Erlandson RA, Antonescu CR: The rise and fall of malignant Chromosome Cancer 33:326–328, 2002.
fibrous histiocytoma. Ultrastruct Pathol 28:283–289, 2004. 245. Murphey MD, Gross TM, Rosenthal HG: From the archives of
223. Feldman F, Norman D: Intra- and extraosseous malignant histio- the AFIP: musculoskeletal malignant fibrous histiocytoma—
cytoma (malignant fibrous xanthoma). Radiology 104:497–508, radiologic-pathologic correlation. Radiographics 14:807–826,
1972. 1994.
224. Finci R, Gunhan O, Ucmakli E, et al: Multiple and familial malig- 246. Newland RC, Harrison MA, Wright RG: Fibroxanthosarcoma of
nant fibrous histiocytoma of bone: a report of two cases. J Bone bone. Pathology 7:203–208, 1975.
Joint Surg 72A:295–298, 1990. 247. Nishida J, Sim FH, Wenger DE, et al: Malignant fibrous histio-
225. Fletcher CD, Gustafson P, Rydholm A, et al: Clinicopathologic cytoma of bone. A clinicopathologic study of 81 patients. Cancer
re-evaluation of 100 malignant fibrous histiocytomas: prognostic 79:482–493, 1997.
relevance of subclassification. J Clin Oncol 19:3045–3050, 2001. 248. Norton KI, Wagreich JM, Granowetter L, et al: Diaphyseal med-
226. Fletcher CDM, Bridge JA, Hogendoorn PCW, et al, editors: ullary stenosis (sclerosis) with bone malignancy (malignant fibrous
WHO classification of tumours of soft tissue and bone, Lyon, 2013, histiocytoma): Hardcastle syndrome. Pediatr Radiol 26:675–677,
IARC. 1996.
227. Fu Y-S, Gabbiani G, Kaye GI, et al: Malignant soft tissue tumors 249. O’Brien JE, Stout AP: Malignant fibrous xanthomas. Cancer 17:
of probable histiocytic origin (malignant fibrous histiocytoma): 1445, 1964.
general considerations and electron microscopic and tissue culture 250. Ozaki T, Taguchi K, Sugihara S, et al: Multiple malignant fibrous
studies. Cancer 35:176–198, 1975. histiocytoma of bone: a case report. Acta Orthop Scand 65:209–211,
228. Gazziola C, Cordani N, Wasserman B, et al: Malignant fibrous 1994.
histiocytoma: a proposed cellular origin and identification of its 251. Ozzello L, Stout AP, Murray MR: Cultural characteristics of
characterizing gene transcripts. Int J Oncol 23:343–351, 2003. malignant histiocytomas and fibrous xanthomas. Cancer 16:331,
229. Ghandur-Mnaymneh L, Zych G, Mnaymneh W: Primary malig- 1963.
nant fibrous histiocytoma of bone: report of six cases with ultra- 252. Papagelopoulos PJ, Galanis EC, Sim FH, et al: Clinicopathologic
structural study and analysis of the literature. Cancer 49:698–707, features, diagnosis, and treatment of malignant fibrous histiocy-
1982. toma of bone. Orthopedics 23:59–65, 2000.
230. Guillou L, Aurias A: Soft tissue sarcomas with complex genomic 253. Roholl PJ, Kleyne J, Elbers J, et al: Characterization of tumor
profiles. Virchows Arch 456:201–217, 2010. cells in malignant fibrous histiocytomas and other soft tissue
231. Hardcastle P, Nade S, Arnold W: Hereditary bone dysplasia with tumours in comparison with malignant histiocytes. I. Immu-
malignant change. Report of three families. J Bone Joint Surg Am nohistochemical study on paraffin sections. J Pathol 147:87–95,
68:1079–1089, 1986. 1985.
232. Huvos AG, Heilweil M, Bretsky SS: The pathology of malignant 254. Roholl PJM, Kleyne J, Van Unnik JAM: Characterization of
fibrous histiocytoma of bone: a study of 130 patients. Am J Surg tumor cells in malignant fibrous histiocytomas and other soft
Pathol 9:853–871, 1985. tissue tumors, in comparison with malignant histiocytes. II.
233. Jeon DG, Song WS, Kong CB, et al: MFH of bone and osteosar- Immunoperoxidase study on cryostat sections. Am J Pathol 121:
coma show similar survival and chemosensitivity. Clin Orthop Relat 269–274, 1985.
Res 469:584–590, 2011. 255. Roessner A, Vassallo J, Vollmer E, et al: Biological characteriza-
234. Kawaguchi K, Oda Y, Sakamoto A, et al: Molecular analysis of tion of human bone tumors X. The proliferation behaviour of
p53, MDM2, and H-ras genes in osteosarcoma and malignant macrophages as compared to fibroblastic cells in malignant fibrous
fibrous histiocytoma of bone in patients older than 40 years. Mod histiocytoma and giant-cell tumor of bone. J Cancer Res Clin Oncol
Pathol 15:878–888, 2002. 113:559–562, 1987.
235. Martignetti JA, Gelb BD, Pierce H, et al: Malignant fibrous his- 256. Roger DJ, Uhl RL, Carl A: Malignant fibrous histiocytoma of the
tiocytoma: inherited and sporadic forms have loss of heterozygos- patella. Orthopedics 17:189–192, 1994.

ERRNVPHGLFRVRUJ
9  Fibrous and Fibrohistiocytic Lesions 691

257. Romeo S, Bovee JV, Kroon HM, et al: Malignant fibrous histio- Fibrosarcoma
cytoma and fibrosarcoma of bone: a re-assessment in the light of 269. Bertoni F, Capanna R, Calderoni P, et al: Primary central (medul-
currently employed morphological, immunohistochemical and lary) fibrosarcoma of bone. Semin Diagn Pathol 1:185–198, 1984.
molecular approaches. Virchows Arch 461:561–570, 2012. 270. Campannacci M, Olmi R: Fibrosarcoma of bone: a study of 114
258. Simons A, Schepens M, Jeuken J, et al: Frequent loss of cases. Ital J Orthop Traumatol 3:199–206, 1977.
9p21 (p16(INK4A)) and other genomic imbalances in human 271. Dahlin DC, Ivins JC: Fibrosarcoma of bone: a study of 114 cases.
malignant fibrous histioctyoma. Cancer Genet Cytogenet 118:89–98, Cancer 23:35–41, 1969.
2000. 272. Eyre-Brook AL, Price CH: Fibrosarcoma of bone: review of fifty
259. Spanier SS: Malignant fibrous histiocytoma of bone. Orthop Clin consecutive cases from the British Bone Tumour Registry. J Bone
North Am 8:947–961, 1977. Joint Surg 51B:20–37, 1969.
260. Spanier SS, Enneking WF, Enriquez P: Primary malignant fibrous 273. Hernandez FJ, Fernandez BB: Multiple diffuse fibrosarcoma of
histiocytoma of bone. Cancer 36:2084–2098, 1975. bone. Cancer 37:939–945, 1976.
261. Sundaresan N, Rosen G, Boriani S: Primary malignant tumors of 274. Huvos AG, Higinbotham NL: Primary fibrosarcoma of bone: a
the spine. Orthop Clin No Am 40:21–36, 2009. clinicopathologic study of 130 patients. Cancer 35:837–847, 1975.
262. Tarkkanen M, Larramendy ML, Bohling T, et al: Malignant 275. Jeffree GM, Price CH: Metastatic spread of fibrosarcoma of bone:
fibrous histiocytoma of bone: analysis of genomic imbalances by a report on forty-nine cases, and a comparison with osteosarcoma.
comparative genomic hybridization and C-MYC expression by J Bone Joint Surg 58B:418–425, 1976.
immunohistochemistry. Eur J Cancer 42:1172–1180, 2006. 276. Papagelopoulos PJ, Galanis EC, Trantafyllidis P, et al: Clinico-
263. Taubert H, Berger D, Hinze R, et al: How is the mutational status pathologic features, diagnosis, and treatment of fibrosarcoma of
for tumor suppressors p53 and p16(INK4A) in MFH of the bone? bone. Am J Orthop 31:253–257, 2002.
Cancer Lett 123:147–151, 1998. 277. Pritchard DJ, Soule EH, Taylor WF, et al: Fibrosarcoma: a clini-
264. Teng H, Xinghai Y, Wei H, et al: Malignant fibrous histiocytoma copathologic and statistical study of 199 tumors of the soft tissues
of the spine: a series of 13 clinical case reports and review of 17 of the extremities and trunk. Cancer 33:888–897, 1974.
published cases. Spine 36:E1453–E1462, 2011. 278. Sadoff RS, Rubin MM: Fibrosarcoma of the mandible: a case
265. Ueda T, Araki N, Mano M, et al: Frequent expression of smooth report. J Am Dent Assoc 121:247–248, 1990.
muscle markers in malignant fibrous histiocytoma of bone. J Clin 279. Shrode LW, Buehler MT, Millican MD: Fibrosarcoma of the
Pathol 55:853–858, 2002. cervical spine. J Manipulative Physiol Ther 15:195–198, 1992.
266. Wigger HJ, Mitsudo SM: Fibrous histiocytoma simulating con- 280. Suh CH, Ordóñez NG, Mackay B: Fibrosarcoma: observations on
genital fibromatosis: a light-, electron microscopic and tissue the ultrastructure. Ultrastructural Pathol 17:221–229, 1993.
culture study. Virchows Arch 370:255–266, 1976. 281. Taconis WK, Mulder JD: Fibrosarcoma and malignant fibrous
267. Yokoyama R, Tsuneyoshi M, Enjoji M, et al: Prognostic factors of histiocytoma of long bones: radiographic features and grading.
malignant fibrous histiocytoma of bone. A clinical and histopatho- Skeletal Radiol 11:237–245, 1984.
logic analysis of 34 cases. Cancer 72:1902–1908, 1993. 282. Taconis WK, van Rijssel TG: Fibrosarcoma of long bones: a study
268. Yoshikawa H, Rettig WJ, Lane JM, et al: Immunohistochemical of the significance of areas of malignant fibrous histiocytoma.
detection of bone morphogenetic proteins in bone and soft-tissue J Bone Joint Surg 67B:111–116, 1985.
sarcomas. Cancer 74:842–847, 1994.

ERRNVPHGLFRVRUJ
C H A P T E R 1 0 

Giant Cell Lesions


CHAPTER OUTLINE

CONVENTIONAL GIANT CELL TUMOR MALIGNANT GIANT CELL TUMOR


OF BONE Secondary Malignant Giant Cell Tumor
Multifocal Conventional Giant Cell Tumor Primary Malignant Giant Cell Tumor
Conventional Giant Cell Tumor with
GIANT CELL REPARATIVE GRANULOMA
Pulmonary Metastases
CHERUBISM AND RELATED SYNDROMES
GIANT CELL TUMOR IN PAGET’S DISEASE

Giant cell lesions form a group of entities with diverse is composed of proliferating mononuclear histiocytic/
clinical presentations and behaviors unified by the pres- macrophage cells and multinucleated osteoclast-like
ence of a prominent population of multinucleated giant giant cells. It frequently undergoes secondary changes
cells. They have been, by convention, divided into two that complicate its classic morphology, and its diagnosis
major groups: those that were considered to be truly can be challenging. In addition, a small proportion of
neoplastic and those that were perceived as being reactive giant cell tumors may be de novo malignant or may
in nature. The prototypic example of neoplastic giant cell develop secondary malignant transformation. In addi-
lesions is a giant cell tumor of bone; giant cell reparative tion, the so-called conventional giant cell tumor may give
granuloma was traditionally considered as a reactive rise, in extremely rare instances, to distant metastases,
process. Recent investigations appear to confirm the neo- typically in the lung. The vast majority of giant cell
plastic nature of a giant cell tumor. In contrast, the condi- tumors are solitary lesions, but occasionally they can
tions characterized by overlapping microscopic features present as multifocal metachronous and synchronous
in general referred to as giant cell reparative granuloma are lesions. A unique form of multifocal giant cell tumor is
subdivided into several categories based on their unique associated with Paget’s disease.
clinical presentation and genetic background. These two
groups of conditions are characterized by somewhat dis-
tinct but occasionally overlapping microscopic features,
and careful correlation with their clinical and radio-
CONVENTIONAL GIANT CELL
graphic presentations is required for correct classification TUMOR OF BONE
of these lesions. Definition
In addition, the ubiquitous presence of multinucleated
giant cells in many unrelated lesions of bone further Giant cell tumor of bone is a distinct, locally aggressive
complicates their classification. Moreover, two giant cell– neoplasm composed of oval or plump, spindled mono-
containing lesions—neoplastic and reactive—frequently nuclear cells and uniformly distributed multinucleated
coexist, and the latter can overshadow the underlying giant cells. The tumor most frequently involves the ends
neoplasm. Neoplastic giant cell lesions can be benign or of long bones in skeletally mature individuals. In other
malignant. Malignant giant cell tumors can arise de novo skeletal sites, it is almost invariably located in epiphyseal
or through transformation of a preexisting benign condi- or epiphyseal-equivalent portions of bone.
tion. Secondary reactive changes in a benign lesion some-
times mimic malignant transformation, and a great deal Incidence and Localization
of expertise may be needed to recognize its benign nature.
On the other hand, features of malignancy can be focal Giant cell tumor of bone accounts for approximately 4%
and inconspicuous, requiring careful examination of mul- of all primary bone tumors. Most patients are between
tiple sections. ages 20 and 55 years, and the peak age incidence is in the
Clinicopathologic correlation on the basis of careful third decade of life. Approximately 70% of cases are
consideration of the radiologic features is of paramount diagnosed in patients between ages 20 and 40 years, and
importance in reaching a correct diagnosis in this diverse it is very unusual for giant cell tumor to occur in patients
group of histologically overlapping entities. younger than age 20 years or older than age 55 years.
Giant cell tumor is a prototypic giant cell–containing Although the diagnosis of giant cell tumor in these age
neoplastic process that shows locally aggressive behavior groups should be treated with skepticism, the unusual
and is referred to as a conventional giant cell tumor. It occurrence of giant cell tumor in the first two decades of
692
ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 693

life, as well as its occasional presentation in patients older Radiographic Imaging


than age 55 years, has been reported.14,15,64-66,110 During
the second decade of life, giant cell tumor usually occurs The radiographic picture of a giant cell tumor is quite
in female patients with fused growth plates.80 characteristic and diagnostic if present in the specific
Giant cell tumor is found most often in the epiphyseal anatomic site of skeletally mature patients. Radiographs
ends of long tubular bones, with the distal end of the reveal a well-defined, eccentric, lytic subchondral lucency
femur, proximal end of the tibia, and the distal end of the that involves the former epiphysis and metaphysis (Figs.
radius being the most frequent sites. It is uncommon in 10-2 and 10-3). The cortex is frequently expanded and,
short tubular bones of the hands and feet. It is extremely at least focally, destroyed. In a small percentage of cases,
rare in the vertebral bodies, but the sacrum is the most there can be minimal periosteal reaction when the cortex
common site in the axial skeleton. Giant cell tumor is is breached. The borders, although well defined, usually
unusual in flat bones and ribs and very rarely affects cra- do not show marginal sclerosis, and trabeculation usually
niofacial bones in the absence of Paget’s disease. The is not present. Occasionally, particularly in a weight-
association of giant cell tumor with Goltz syndrome bearing bone, a giant cell tumor can show marked tra-
(focal dermal hypoplasia), a rare condition in which there beculation and marginal sclerosis that produces the
are multiple congenital anomalies of skin, teeth, and nontypical “soap bubble” appearance (Fig. 10-4). These
bone, has been reported.107 Its presence in two cases sug- changes can be correlated with the microscopic findings
gests that it may represent an integral element of the of extensive reactive fibrohistiocytic features (see later
spectrum of anomalies observed in this very rare condi- section). Pathologic fracture is found in 5% to 10% of
tion. Giant cell tumor may occur more frequently in giant cell tumors.37 It may be the presenting clinical
Chinese people than in people who live in Western feature for patients with such tumors in weight-bearing
countries. The estimated rate in the Chinese population bones (Figs. 10-5 and 10-6).
has been reported to account for about 20% of all Some observers have proposed a radiologically based
primary tumors of bone.106 The peak age incidence and classification defining three main types of giant cell
the most common skeletal sites of involvement are shown tumor: quiescent (type I), active (type II), and aggressive
in Figure 10-1. Text continued on p. 699

Peak
incidence
20 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 10-1  ■  Giant cell tumor. Peak age incidence and frequent sites of skeletal involvement in giant cell tumor. Most frequent sites
are indicated by solid black arrows.

ERRNVPHGLFRVRUJ
694 10  Giant Cell Lesions

A B

C D
FIGURE 10-2  ■  Giant cell tumor in a skeletally mature 16-year-old girl: radiographic features. A and B, Anteroposterior and lateral
plain radiographs show involvement of lateral condyle. C, T2-weighted coronal magnetic resonance image (MRI) shows high signal
in tumor. D, Axial T1-weighted (upper) and T2-weighted (lower) MRIs show signal characteristics of tumor in lateral condyle.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 695

A B

C
FIGURE 10-3  ■  Giant cell tumor of proximal end of tibia: radiographic and microscopic features. A, Radiograph of knee of an 18-year-
old skeletally mature woman with giant cell tumor involving lateral tibial plateau and extending into metaphysis. B, Photomicrograph
of curetted tibial lesions shows even distribution of giant cells in spindle-cell stroma (×100). C, Higher magnification of tumor shown
in B (×200) (B and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
696 10  Giant Cell Lesions

A B

C D

FIGURE 10-4  ■  Giant cell tumor with sclerotic margins and trabeculation: radiographic features. A and B, Anteroposterior (AP) and
lateral views of giant cell tumor in distal femur of a 27-year-old man. Note expanded contour of femur. C, AP radiograph of giant
cell tumor in distal femur of a 52-year-old woman with marginal sclerosis. D, Computed tomogram of tumor shown in C clearly
demonstrates dense cortexlike rim surrounding tumor. This finding is unusual in giant cell tumor.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 697

A B

C D
FIGURE 10-5  ■  Giant cell tumor with pathologic fracture: radiographic features. A and B, Radiographs of right knee of a 31-year-old
man who fell and sustained displaced pathologic fracture through medial cortex as well as midarticular surface of femur. C and
D, Anteroposterior and lateral radiographs of distal femoral giant cell tumor with impacted pathologic fracture. Patient was a 40-year-
old man who noticed aching pain and slight swelling above his knee for 1 year. While involved in strenuous activity, he felt his knee
give way.

ERRNVPHGLFRVRUJ
698 10  Giant Cell Lesions

A B

C D
FIGURE 10-6  ■  Neglected pathologic fracture through giant cell tumor: radiographic and gross features. A, Anteroposterior radio-
graph of distal femoral giant cell tumor with impacted pathologic fracture. B, Pathologic fracture associated with giant cell tumor
of proximal humerus. C, Radiograph of left knee of a 54-year-old woman who had pins placed in distal femoral fracture 1 year previ-
ously. Fact that giant cell tumor was present was not appreciated, and tumor grew into soft tissue around femur. D, Coronally
bisected amputation specimen of case shown in C with giant cell tumor within fractured distal femur and adjacent soft tissue.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 699

(type III).14,15,29 These three varieties were referred to by evidence of hemorrhage, cyst formation, and necrosis.
Enneking29 as surgical stages. The quiescent type is char- All these changes can be present in one tumor. There
acterized by a lytic defect limited to the medullary cavity also may be evidence of pathologic fracture, most fre-
of the bone with minimal or no involvement of the corti- quently involving the subchondral bony endplate.
cal bone. Usually the defect is surrounded by a more or Hemorrhage and necrosis are particularly frequent and
less clear rim of sclerosis, and fine trabeculation can be extensive in the weight-bearing bones (Fig. 10-10). They
present. The active radiologic type is characterized by a may be present with or without associated pathologic
thinned, expanded cortex and somewhat unclear margins. fracture.
The aggressive radiologic type is characterized by a lytic
defect with ill-defined margins, invasion of the cortex,
Microscopic Findings
and extension of the tumor into surrounding soft tissue.
This radiologic classification of giant cell tumor has Histologically, the basic pattern of giant cell tumor is that
limited usefulness. So-called radiologic features of aggres- of a moderately vascularized stroma with oval or plump,
siveness do not necessarily correlate with microscopic spindle-shaped mononuclear cells uniformly interspersed
criteria of malignancy or the ultimate clinical behavior of with multinucleated giant cells (Figs. 10-12 to 10-14).
the tumor. In the authors’ shared opinion, the terms The latter can have nuclei sometimes numbering 50 to
quiescent, active, or aggressive should not be used even in 100 or even more, which are clustered in the middle of
the descriptive sense in reference to radiographic presen- the cell (Figs. 10-14 and 10-15). The nuclei of stromal
tations of giant cell tumors. These are clinically irrelevant and giant cells are similar. They are round or oval with
and misleading terms because they overlap with terms regular outlines, evenly distributed fine chromatin, and
used to describe histologic features. Radiologic features a prominent nucleolus. In addition to these two cell
of aggressiveness with huge destructive lesions, massive populations, occasional binucleated and trinucleated cells
invasion into the cortex, and extension of the tumor into can be seen, suggesting a gradual transition of some
surrounding soft tissue are merely reflections of the stage mononuclear cells into multinucleated giant cells. This
of the disease. They do not necessarily correlate with phenomenon can be seen particularly well in cytologic
aggressive histologic features in our experience. preparations in which whole intact cells are analyzed.
Overlap in radiologic presentation with fibrosarcoma, Occasionally, stromal and giant cells may show pyknotic
malignant fibrous histiocytoma, multiple myeloma, and changes of the nuclei. Giant cells usually have dense
other destructive lesions can occur. Consequently, in eosinophilic cytoplasm with clear, sharp, oval borders.
many cases, the exact radiologic diagnosis of giant cell Occasionally, cells can be irregular in shape and vacuo-
tumor cannot be made. lated (Figs. 10-14 and 10-15).
In very unusual instances, giant cell tumor may occur Morphologically, several types of mononuclear stromal
in the metaphyseal portion of bone without involving the cell can be distinguished. The stroma can be composed
epiphysis (Fig. 10-7). This may occur in young patients of small or intermediate-sized oval cells. The spindle cells
whose epiphyseal plates are still open. Nonepiphyseal can be short and plump or elongated and fibroblast-like
localization of the lesion sometimes occurs in skeletally (Figs. 10-16 and 10-17). All these types of cells can be
mature patients. So-called nonepiphyseal giant cell tumor present in one tumor, sometimes accounting for a super-
represents a medical curiosity and is extremely rare. ficial impression of pleomorphism. True pleomorphism,
Marked bone expansion and secondary aneurysmal not related to the presence of various types of stromal
bone cyst are frequent secondary changes seen in giant cells, should not be seen in a conventional giant cell
cell tumor that are best assessed by computed tomog­ tumor. The mitotic rate of mononuclear stromal cells can
raphy and magnetic resonance imaging (Figs. 10-8 be quite high, but atypical mitoses are not present. There
and 10-9). is no mitotic activity within multinucleated giant cells.
No bone or cartilage matrix production by tumor cells is
Gross Findings seen. The classic account of the histologic criteria for the
diagnosis of giant cell tumors by Jaffe et al.47 emphasized
Giant cell tumor of bone in its typical location and intact the relationship between clinical behavior and histologic
in its setting is soft, friable, fleshy, and red-brown with evidence of spindle-cell predominance, focal attenuation
yellowish areas (Figs. 10-10 and 10-11). The tumor tissue of giant cells, and increased mitotic activity. This early
is well demarcated and usually extends to the articu- histologic description included a numeric grading system
lar cartilage. The articular cartilage is unlikely to be (grades 1 through 3) that was designed to predict recur-
invaded or perforated. The lesion usually occupies an rent behavior and metastatic potential. Subsequent expe-
eccentric position in the epiphyseal end of the bone. Its rience has shown that the use of numeric histologic
shaftward portion (i.e., advancing toward the medullary grading lacks predictive value.
cavity) is usually more centrally located. It is delimited The classic microscopic pattern of giant cell tumor
in its periphery by a thin layer of fibrous and reactive is frequently modified by a secondary reactive prolifera-
bone tissue. The cortical bone may not be involved, tion of fibrohistiocytic tissue, hemorrhage, necrosis, and
and the original contour of the bone may be preserved. aneurysmal bone cyst formation. Reactive fibrous tissue
However, more frequently, the original cortex has been with a prominent storiform pattern and xanthogranulo-
destroyed, and the bone contour has been expanded. A matous reaction can be documented, at least focally,
thin shell of subperiosteal bone surrounding the periph- in nearly all appropriately sampled lesions. In some
ery of the lesion is sometimes seen. Often there is gross Text continued on p. 711

ERRNVPHGLFRVRUJ
700 10  Giant Cell Lesions

A B

C D

FIGURE 10-7  ■  Nonepiphyseal giant cell tumor: radiographic features. A, Anteroposterior radiograph of giant cell tumor in skeletally
immature patient shows no involvement of epiphysis. B, Lateral view of tumor shown in A shows no extension to secondary growth
center. Patient was a 14-year-old girl with knee pain. C, Lateral radiograph of distal femur of a 27-year-old woman with pain in
lower thigh. Giant cell tumor is confined to metaphysis. D, T1-weighted magnetic resonance image shows that giant cell tumor seen
in C is limited to shaft.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 701

A B

D E
FIGURE 10-8  ■  Giant cell tumor of distal radius: radiographic and gross morphologic features. A, Anteroposterior radiograph of distal
radial giant cell tumor. B, T2-weighted coronal magnetic resonance image (MRI) shows expansile distal radial tumor with multilocular
architecture and variegated signal enhancement. C and D, Radial T1-weighted MRI and T2-weighted MRI with contrast with fluid
levels. E, Bisected distal radial resection specimen showing an expansile multilocular giant cell tumor composed of dark and light
tan tissue with focal yellowish areas.

ERRNVPHGLFRVRUJ
702 10  Giant Cell Lesions

A B

C E
FIGURE 10-9  ■  Giant cell tumor of proximal fibula: radiographic and gross morphologic features. A, Anteroposterior radiograph
showing an expansile lytic lesion of the proximal fibula with fine trabecular pattern. B, Bisected proximal fibula resection specimen
showing an expansile giant cell tumor with multilocular cystic architecture. C, T2-weighted coronal magnetic resonance image (MRI)
shows lobular signal enhancement within the expansile lesion involving the proximal fibula. D and E, Radial T2-weighted MRI and
T2-weighted MRI with contrast show multilocular cystic lesion in the fibula.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 703

A B

C D
FIGURE 10-10  ■  Giant cell tumor: gross mophologic features. A, Bisected distal femoral resection specimen shows light and dark tan
giant cell tumor with marbling fibrous tissue. Mild expansion of the bone contour with intact cortex and sharp demarcation of the
tumor from the surrounding medullary bone is present. B, Expansile giant cell tumor of distal femur composed of light and dark
tan tissue with central fibronecrotic areas. Impacted pathologic fracture is present. C, Giant cell tumor of tibia extending to the
articular plate and expanding the bone contour laterally. The lesion is composed of light and dark tan tissue with areas of fibrosis.
Note small hemorrhagic cystic spaces. D, A large expansile giant cell tumor of the proximal humerus composed of light and dark
tan tissue. Large areas of yellowish necrotic tissue in the central portion of the lesion are present.

ERRNVPHGLFRVRUJ
704 10  Giant Cell Lesions

A B

C D
FIGURE 10-11  ■  Giant cell tumor: gross morphologic features. A, Bisected distal segment of femur with well-demarcated tumor tissue.
Note destroyed cortex and expansion of tumor into soft tissue. B, Posterior aspect of specimen shown in A shows destroyed cortex
and expansion of tumor into popliteal fossa. C, Bisected distal end of radius with well-demarcated eccentric tumor mass expanding
bone contour. Tumor tissue is red-brown with fine yellowish septations. D, Specimen radiograph of tumor shown in C. Note focal
destruction of cortex.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 705

A B

C D
FIGURE 10-12  ■  Giant cell tumor: microscopic features. A, Classic microscopic pattern of giant cell tumor with oval or plump spindle
mononuclear cells uniformly interspersed with multinucleated giant cells. B, Higher power view of A shows multinucleated giant
cells and oval or plump mononuclear cells. C, Classic microscopic features of giant cell tumor composed of mononuclear histiocytic
cells with evenly distributed multinucleated giant cells. D, Higher magnification of C shows mononuclear histiocytic cells and mul-
tinucleated giant cells with hemosiderin deposits. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
706 10  Giant Cell Lesions

A B

C D
FIGURE 10-13  ■  Giant cell tumor: microscopic features. A, Classic microscopic pattern of giant cell tumor with oval or plump spindle
mononuclear cells uniformly interspersed with multinucleated giant cells. B, Higher power view of A shows multinucleated giant
cells and oval or plump mononuclear cells. C, Classic microscopic features of giant cell tumor composed of mononuclear histiocytic
cells with evenly distributed multinucleated giant cells. D, Higher magnification of C shows mononuclear histiocytic cells and mul-
tinucleated giant cells with hemosiderin deposits. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 707

A B

C D
FIGURE 10-14  ■  Giant cell tumor: microscopic features. A, Variant of giant cell tumor with numerous multinucleated giant cells. Some
of the cells have more than 100 nuclei. B, Higher power view of A shows multinucleated giant cells with more than 100 nuclei and
dense eosinophilic cytoplasm. C, Conventional giant cell tumor with spindling of mononuclear cells and scattered multinucleated
giant cells with ragged contours of their cytoplasm. D, Higher magnification of C showing multinucleated giant cells with large
irregular cytoplasm. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
708 10  Giant Cell Lesions

A B

C D

E F

G H
FIGURE 10-15  ■  Giant cell tumor: microscopic features. A-H, Spectrum of multinucleated giant cells frequently seen in giant
cell tumors ranging in size of their cytoplasm and the number of nuclei from a few to several hundred. (A-H, ×200)
(A-H, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 709

A B

C D
FIGURE 10-16  ■  Giant cell tumor with fibrohistiocytic changes: microscopic features. A, Xanthogranulomatous reaction in giant cell
tumor obliterating its classical cytoarchitectural features. B, Higher magnification of A showing foamy histiocytic infiltrate. C, Fibro-
histiocytic reaction in giant cell tumor. Ill-defined bands of spindle-cell proliferations interspaced with histiocytic infiltrate are present.
D, Higher magnification of C showing mixed fibrous and histiocytic infiltrate with occasional atypical cells. (A and C, ×100; B and
D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
710 10  Giant Cell Lesions

A B

C D
FIGURE 10-17  ■  Giant cell tumor with atypia: microscopic features. A-D, Giant cell tumor exhibiting enlargement and nuclear atypia
of the mononuclear histiocytic cells. This type change is present focally in many giant cell tumors and is not indicative of malignant
transformation. (A-D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 711

instances, reactive bone formation can be associated with (Fig. 10-21). The overall similarity of these proliferations
these changes. Occasionally, fibrohistiocytic reaction to nodular fasciitis is helpful to distinguish such benign
massively replaces the underlying tumor so that it mimics reactive processes from sarcomatoid transformation.
lesions such as nonossifying fibroma or benign fibrous
histiocytoma. In such instances, the analysis of radio-
Cytology
graphic data and additional sampling of the tumor are
usually sufficient to document the existence of an under- Fine needle aspirates of giant cell tumor typically show
lying giant cell tumor. The presence of prominent, reac- two populations of cells with predominating mononu-
tive fibroblastic tissue and xanthogranulomatous reaction clear cells and less abundant multinucleated giant cells
correlates with radiographically prominent sclerotic (Fig. 10-22). Mononuclear cells, which show a histiocyt-
margins and coarse trabeculations (Fig. 10-18). oid appearance, are singly dispersed or may be arranged
Prominent focal reactive bone sometimes can be cor- in small three-dimensional clusters (Fig. 10-22). Mul-
related with the presence of small cortical infractions. A tinucleated giant cells are similar to osteoclasts but
shell of reactive bone usually is seen at the periphery of usually contain many more nuclei. Characteristically,
the lesion (Fig. 10-19). Although there is usually no peri- the nuclei of mononuclear histiocytoid cells are identi-
osteal reactive bone seen on radiographs, a thin rim of cal to nuclei of giant osteoclast-like cells. This is a dis-
periosteal new bone formation is nearly always present tinct cytologic feature of giant cell tumor of bone. The
microscopically. This peculiar capacity of giant cell tumor cytologic features of giant cell tumor of bone are often
to induce reactive peripheral ossification is maintained in obscured by secondary changes, such as proliferation of
recurrences in soft tissues, in pulmonary implants, and fibrous tissue accompanied by foamy histiocytes. In such
even in the transplanted fragments of tumor tissue to instances, correlation of cytologic findings with clinical
athymic nude mice.6 In approximately 30% of resected and radiologic data may help to establish the correct
giant cell tumors, intravascular invasion of tumor tissue diagnosis.
can be observed in the adjacent soft tissue. This inciden-
tal finding does not appear to correlate with local aggres-
Differential Diagnosis
siveness or the development of pulmonary implants.
Hemorrhage, necrosis, or both usually result from Giant cell tumor should be differentiated from giant
fracture or mechanical compression. Old and fresh hem- cell reparative granuloma and other reactive giant cell–
orrhage, as well as necrosis, can be present without any containing lesions, such as the brown tumor of hyper-
obvious cause and can be quite extensive. For unknown parathyroidism. Less frequently, it can be confused
reasons, the mononuclear stromal cells usually develop with nonossifying fibroma, chondroblastoma, chondromyxoid
the recognizable features of necrosis first. It is common fibroma, and the solid areas of aneurysmal bone cysts. A more
to observe well-preserved giant cells in a completely substantial problem arises in separating this lesion from
necrotic stroma. Early necrotic changes associated with malignant fibrous histiocytoma and giant cell–rich osteosar-
nuclear pyknosis and increased nuclear variability can coma. Bone erosion in pigmented villonodular synovitis can
mimic malignant change (Figs. 10-20 and 10-21). The sometimes present difficulties in differential diagnosis.
focal nature of this necrosis-related atypia in an otherwise Giant cell reparative granuloma most frequently
conventional giant cell tumor and the absence of atypical occurs in the jaws, where true giant cell tumors do not
mitoses are helpful in avoiding a misdiagnosis of malig- arise in the absence of underlying Paget’s disease. The
nant change. In general, focal necrosis is a common most useful histologic criterion in making this distinction
finding in giant cell tumor and its presence is not indica- is the uniformity of distribution of the multinucleated
tive of aggressive behavior or malignant change. giant cells and the absence of reactive bone formation
Microscopic foci of aneurysmal bone cyst can be fre- and stromal collagenization in unaltered giant cell tumor.
quently documented if appropriate samples are available. Brown tumor of hyperparathyroidism, which represents
The presence of such foci does not necessarily correlate a giant cell reparative granuloma of known etiology, can
with radiographic and gross features of a fully developed be recognized by the characteristic biochemical find-
aneurysmal bone cyst. Typical radiographic and gross ings of hypercalcemia, hypophosphatemia, and elevated
features of an expansile lesion with honeycombed blood- parathormone levels. The absence of chondroid matrix
filled spaces can develop in some giant cell tumors (Figs. and the characteristic plump, spindle-shaped appear-
10-8 and 10-9). A giant cell tumor is reported to be an ance of the mononuclear cell component are impor-
underlying condition in 10% of secondary aneurysmal tant in excluding a giant cell–rich chondroblastoma or
bone cysts. On the other hand, solid areas containing chondromyxoid fibroma. The exclusion of nonossifying
numerous multinucleated giant cells in a spindle-cell fibroma should offer no substantial difficulty if atten-
stroma are frequently present in an aneurysmal bone cyst tion is paid to radiologic signs of skeletal immaturity
and can be readily misinterpreted as an underlying giant and metaphyseal location. Irregular distribution of com-
cell tumor. This finding should be interpreted only with pressed and attenuated multinucleated giant cells in a
reference to appropriate radiographic features and clini- more fibroblastic background is also characteristic of
cal setting to avoid misdiagnosis of giant cell tumor. nonossifying fibroma. Giant cell–rich osteosarcoma and
In rare instances, florid proliferation of spindle cells malignant fibrous histiocytoma are differentiated on
that sometimes mimic so-called tissue culture fibroblastic the basis of nuclear anaplasia, abnormal mitotic figures,
proliferations can be present in giant cell tumors, further and neoplastic osteoid production, which are present
complicating the microscopic presentation of the lesion Text continued on p. 717

ERRNVPHGLFRVRUJ
712 10  Giant Cell Lesions

A B

C D
FIGURE 10-18  ■  Giant cell tumor with prominent fibrohistiocytic reaction: radiographic and microsocpic features. A, Anteroposterior
radiograph shows marginally sclerotic giant cell tumor in proximal end of tibia. B, Computed tomogram shows no cortical break-
through and prominent sclerosis of surrounding bone. C, Fibroxanthomatous reaction in giant cell tumor with conventional tumor
tissue in upper right corner. D, Higher power photomicrograph of fibroxanthomatous component that is commonly seen in giant
cell tumors with reactive sclerosis and trabeculation. (C, x100; D, ×200). (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 713

A B

C
FIGURE 10-19  ■  Giant cell tumor: microscopic features. A, Giant cell tumor with engorgement of stromal vessels and cytoplasmic
vacularization. B, Higher magnification of B shows multinucleated giant cells and dialated stromal vessels. C, Giant cell tumor
extending to soft tissue. The periphery of the tumor is delineated by a shell of reactive bone. (A and C, ×100; B, ×200).
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
714 10  Giant Cell Lesions

A B

C D
FIGURE 10-20  ■  Necrosis in giant cell tumor: microscopic features. A, A focus of viable giant cell tumor in a background of diffuse
necrosis. B, Higher magnification of A showing the so-called anoxic atypia affecting predominantly the mononuclear cells. C, Area
of extensive necrosis with spindle-cell proliferation. D, Higher power view of C shows florid spindle-cell proliferation at the border
of necrosis and viable tumor. (A and C, ×100; B, ×400; C, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 715

A B

C D
FIGURE 10-21  ■  Necrosis in giant cell tumor: microscopic features. A, Low power view of an interface between necrosis on the left
and viable tumor on the right. B, Higher power of A showing hyperchromasia of cells in the interface between necrosis and viable
tumor. C, Interface between necrotic area and viable tumor showing a loose texture and nuclear hyperchromasia. D, Complete
necrosis. Note well preserved multinucleated giant cells. (A, ×100; B-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
716 10  Giant Cell Lesions

A B C

E F

D G
FIGURE 10-22  ■  Giant cell tumor of proximal tibia: radiographic, gross, and aspiration cytology features. A, Anteroposterior view of
giant cell tumor in plain radiograph involving epiphysis of proximal tibial. Note expanded contour of tibia. B, Gross photograph of
resection specimen shown in A with expansile red-brown tumor containing fine yellowish septations. The cortex overlying tumor
is destroyed and expanded bone contour is delineated by thin fibrous capsule. C, High magnification showing two multinucleated
giant cells within the mononuclear stroma. Note that the nuclei of mononuclear stromal cells and of multinucleated giant cells look
similar. D, Histologic appearance of the same tumor showing scattered multinucleated giant cells within mononuclear stromal cells
resembling histiocytes. E and F, Fine needle aspirates containing multiple mononuclear stromal cells and multinucleated osteoclastic
cell. G, Higher power demonstrating mononuclear stromal cells with oval nuclei and discrete nucleoli. Inset, An oval histiocytic cell
with two nuclei and densely eosinophilic cytoplasm. (C, E, and F, ×400; D, ×200; G and inset, ×600.) (C-G, hematoxylin-eosin.)
(C-G and inset, reprinted with permission from Czerniak B, et al: In Koss LG, Melamed MR, editors: Koss’ diagnostic cytology and its his-
topathologic bases, ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 717

FIGURE 10-23  ■  Giant cell tumor. Secondary phenomena observed in conventional giant cell tumor.

in the former. The summary of secondary changes surgical treatment of the primary lesion. In some unusual
complicating the cytoarchitectural features of conven- instances, it may occur many years after the removal of
tional giant cell tumors and various clinical phenomena the primary tumor.36,67 Recurrence can be identified on
observed during its clinical course are summarized in radiographs by a prominent shell of reactive bone sur-
Figure 10-23. rounding its periphery (Fig. 10-25). Simple excision is
usually sufficient to control the disease. In the past, radia-
Treatment and Behavior tion therapy was frequently used to control the disease
locally and has been proved to be effective in preventing
Approximately 25% of conventional giant cell tumors are local recurrences. Because the majority of malignant
considered to be locally aggressive on clinical or radio- transformations in giant cell tumor are linked to prior
logic grounds.5 These tumors show extensive bone radiation, radiotherapy is no longer recommended as a
destruction, cortical expansion, and invasion into soft primary mode of treatment.21 It is still, however, used to
tissue.93 These features cannot be correlated with any control the disease in anatomic sites, such as the spine, for
specific histologic findings. Approximately 25% to 35% which it is technically difficult to perform en bloc exci-
of giant cell tumors recur after simple curettage.14,64-66,93 sion or complete curettage.2,17,96,100,103 Vasoocclusive
Recurrence typically occurs within 3 years of curettage.64-66 therapy by arterial embolization may be considered as an
In unusual instances, the lesion may recur many years or alternative approach in anatomic sites for which it is tech-
even decades after removal of the primary tumor. The nically difficult to remove the tumor mass surgically.19
recurrent lesion usually has the same morphologic fea- Pulmonary metastases, or so-called benign pulmonary
tures as the primary tumor (Fig. 10-24). Curettage sup- implants, may develop in approximately 1% to 2% of
plemented by cryotherapy is used in some centers to patients with conventional giant cell tumors. Typically
reduce the rate of local recurrence. However, it is not the pulmonary nodules grow slowly and are amenable to
universally accepted as a primary mode of treatment. surgical excision with a prospect for cure.10,11,24,25,51,62 Still,
Wide excision with allograft or prosthetic replacement some patients may die of disease as a result of pulmonary
significantly reduces, but does not completely eliminate, miliary spread or progressive growth of multiple lung
recurrences and is performed when appropriate and tech- lesions.21,24
nically feasible.14,15,65 The preferred mode of primary In general, surgery is the main treatment modality for
treatment of a conventional giant cell tumor is still thor- giant cell tumor and is occasionally complemented by
ough curettage and bone grafting.14,15,65,96 This general radiation therapy.74,103,108,110,113 Recent advances in molec-
approach is modified in relation to the anatomic site and ular biology of giant cell tumor has provided new
clinical stage of the disease. Recurrent lesions are usually treatment options, with the RANKL inhibitor (deno-
adequately treated by a second curettage. Recurrence in sumab) and bisphosphonates, which inhibit osteoclast
soft tissue is a rare local complication of a conventional driven bone resorption.2,27,42,103,109,113 These two treatment
giant cell tumor (Fig. 10-25). Typically, it is a result of options are most frequently used for primary tumors
tumor implantation into soft tissue at the time of surgical that cannot be resected or for metastatic disease that
treatment.30 It usually occurs within the first 3 years after cannot be surgically controlled. The reported partial and

ERRNVPHGLFRVRUJ
718 10  Giant Cell Lesions

A B

C D
FIGURE 10-24  ■  Recurrence of conventional giant cell tumor: radiographic and microscopic features. A, Radiograph of knee of 17-year-
old skeletally mature girl with a 6-month history of knee pain whose giant cell tumor involved lateral half of tibial plateau; subchon-
dral bone was curetted and bone grafted. B, Histologic appearance of original tumor shows conventional giant cell tumor.
C, Radiograph of same knee 5.5 years later when symptoms recurred. Note that radiolucency is larger in extent and bony septum
is present. D, Histologic appearance of recurrent giant cell tumor is identical to primary neoplasm. (B and D, ×200) (B and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 719

A B C

D E
FIGURE 10-25  ■  Recurrence of giant cell tumor of bone in soft tissue: radiographic and microscopic features. A, Radiograph of knee
of a 27-year-old woman shows eccentric lytic tumor on medial side of tibial plateau. Two years later, recurrence was curetted and
bone grafted. B, Eighteen months later patient returned with palpable nodule in soft tissue beneath surgical scar (arrows) with
peripheral calcification seen on radiograph. C, Specimen radiograph film of excised recurrence (implant) in soft tissue. Note streaks
of mineralization peripherally and traversing nodule. D, Histologic appearance of primary tumor shown in A. E, Photomicrograph
of recurrent tumor nodule with peripheral shell of reactive bone. (D, ×200; E, ×50) (D and E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
720 10  Giant Cell Lesions

complete responses to anti-RANKL therapy are causing ubiquitously overexpressed in giant cell tumor (approxi-
an increased interest in this treatment modality. mately 70% of the cases) and can be used in the differential
Development of sarcoma in conventional giant cell diagnosis of giant cell-containing lesions.56 More recent
tumor is the most serious complication but fortunately is investigations implicate that there is a subpopulation of
rare. As mentioned previously, the majority of secondary stromal cells in giant cell tumors that are in fact osteo-
sarcomas that arise in association with conventional giant blastic in nature which have clonogenic potential and
cell tumor are linked to prior radiation therapy.21,40 A produce osteoclast differentiation signals.3,27,31,54,70,109,118
primary (de novo) malignant giant cell tumor is a well- One of such signaling pathways involves a ligand for
known but extremely unusual phenomenon. With the receptor activator of nuclear factor κB (RANKL).4,45,54
decline in the use of therapeutic irradiation for giant cell In giant cell tumors, neoplastic stromal cells that express
tumors, malignant transformation has become exceed- RANKL are thought to stimulate giant cell formation
ingly rare. from the monocytic cells that express receptor activator of
NFκB (Fig. 10-26).109 The mechanisms of overexpression
of RANKL by stromal cells are not well understood, but
Special Techniques
it is postulated that parathyroid hormone-related protein
It appears that several cell types that belong to the (PTHrP), also expressed by stromal cells in giant cell
macrophage/osteoclastic and osteoblastic lineages con- tumors, may play a role in the stimulation of RANKL.20,73
tribute to the development of giant cell tumors. Ultra- PTHrP is in turn regulated by microRNA-126-5p, which
structurally, the cytoplasm of mononuclear cells contains has been shown to play a role as the upstream regulator
abundant rough endoplasmic reticulum, moderate num­ in the PTHrP-RANKL axis.119 Surprisingly, mutations of
bers of mitochondria, a few lysosome-like bodies, and the isocitrate dehydrogenase (IDH) genes, also frequently
occasionally multiple lipid vacuoles.105 The cytoplasm of seen in gliomas, acute myeloid leukemias, and cartilagi-
the multinucleated giant cell contains numerous mito- nous lesions, are often found in giant cell tumors.50 The
chondria, few lysosomes, and sparse, rough endoplasmic mutations involve the IDH2-R172S gene and can be
reticulum and appears to originate from mononuclear documented in nearly 80% of giant cell tumors.50 The
cells. Occasionally, filamentous viruslike intranuclear mutations of the histone H3.3 encoding gene exclusively
inclusions that are morphologically identical to those involving H3F3A and leading to a p.Gly34Trp change in
found in Paget’s disease of bone are present.1,22,32,68,98 the majority of cases were found in approximately 90% of
Unlike the inclusions seen in the osteoclasts of Paget’s giant cell tumors.7 Surprisingly, mutations of the histone
disease and in giant cell tumors that arise in a setting of H3.0 encoding gene were also found in a large propor-
Paget’s disease, these intranuclear inclusions can be found tion of chondroblastoma.7 Both of these mutations (i.e.,
only after prolonged and careful survey of great numbers those involving IDH2 and H2F3A) because of their high
of cells. In summary, the ultrastructure is of little help frequency, may represent driver mutations for giant cell
to elucidate various dilemmas related to the origin of a tumor of bone but their exact biologic role in the devel-
giant cell tumor. It suggests, however, that the mononu- opment of this tumor is yet unknown.
clear cells have some ultrastructural similarities with cells Little is known about the factors governing local
of histiocytic lineage, macrophage lineage, or both. In aggressive behavior, recurrence rate, and metastatic
fact, some of the mononuclear cells express the receptor potential of conventional giant cell tumors. DNA ploidy
for the immunoglobulin G crystallizable fragment and measurements show that a high proportion of these
differentiation antigens associated with a macrophage- lesions are aneuploid. This does not correlate with the
monocyte lineage.13,52,57 Both mononuclear and multi- clinical behavior of the lesion and cannot be used as a
nucleated giant cells are also positive for α1-antitrypsin.57 reliable factor for predicting recurrence or pulmonary
The other major population of mononuclear cells in giant metastases.53,95,97 A majority of giant cell tumors show
cell tumor does not express antigens of monocyte lineage, random or clonal chromosomal aberrations, with telo-
is strongly positive for tartrate-resistant acid phospha- meric fusion being the most striking abnormality.12,99 It
tase, and expresses parathormone and calcitonin recep- has been suggested that this unique chromosomal aber-
tors, indicating that the cells are undergoing osteoclastic ration may correlate with aggressive local behavior, a
differentiation (Fig. 10-26).13,52 These cells also are func- high recurrence rate, or the metastatic potential of a
tionally dependent on parathormone and calcitonin and conventional giant cell tumor.12 This observation is based
increase their level of cyclic adenosine monophosphate on analysis of a limited number of cases, and its true
on exposure to these hormones.38,39,52 An intermediate clinical value remains to be elucidated. Allelic losses of
population of mononuclear cells expressing phenotypic 1p, 9q, and 19q are frequent in giant cell tumors but do
features of both monocyte-macrophage and osteoclas- not correlate with local recurrence or metastatic poten-
tic lineages can also be found. The cells of monocyte- tial.83 In contrast, loss of genetic material on 17p in prox-
macrophage lineage do not proliferate well in vivo and imity to the p53 locus and on 9p appear to correlate with
are usually eliminated from tissue culture explants.39 In the metastatic potential of giant cell tumors.83 This
contrast, the osteoclastic cells proliferate well in tissue observation is in concert with the identification of TP53
culture, forming multinucleated giant cells. In summary, mutations and deletions that parallel the malignant trans-
the main population of cells in giant cell tumor have formation of giant cell tumors.76,77,92 The pathways that
phenotypic features of both macrophage-like and osteo- may play a role in the aggressive behavior of giant cell
clastic cells.26,60 Recent cDNA microarray studies have tumors include the upregulation of p53 signaling, osteo-
identified that the TP63 gene encoding the p63 protein is clast differentiation, and Wnt signaling. Among these

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 721

A B

C D
FIGURE 10-26  ■  Giant cell tumors: immunohistochemical features. A, Classic microscopic pattern of giant cell tumors. B, Immuno-
histochemical stains for CD-68 showing strong positive cytoplasmic stain of multinucleated giant cells and of scattered mononuclear
cells. Note that not all mononuclear cells are showing a phenotype of macrophage cells. C, Immunohistochemical stain showing
strong expression of RANKL among mononuclear stromal cells. Note that multinucleated giant cells do not express RANKL. D, High
proliferation rate documented by positive immunohistochemical staining for Ki67. Note the proliferation is restricted to mononuclear
cells. (A-D, ×200) (A, hematoxylin-eosin; C and D, immunohistochemical stains, DAB.)

ERRNVPHGLFRVRUJ
722 10  Giant Cell Lesions

pathways the upregulation of IGF1, MDM2, STAT1, anatomic location. However, in different anatomic sites
and RAC1 appear to be most significantly associated and age groups, it may cause various diagnostic dilem-
with a recurrence, while genomic amplifications involv- mas. The clinical significance, the technical feasibility of
ing CCND1 and MET were found in association with complete removal, and consequently the chance for cure
malignant transformation.18,91 Similar to many other can also significantly differ in relation to the anatomic
tumors, giant cell tumor of bone, both primary and meta- site. For this reason, it is necessary to provide separate
static, may ectopically upregulate beta-human chorionic descriptions of giant cell tumor and its differential diag-
gonadotropin.55 nosis in various anatomic sites.

Giant Cell Tumor of Long Tubular Bones.  The


Personal Comments
tumor is most often found in the epiphyseal ends of long
Giant cell lesions in bone are the most frequently encoun- tubular bones, with the distal end of the femur and proxi-
tered diagnostic problems in consultation material. The mal end of the tibia being the most frequent sites (Fig.
ubiquitous distribution of multinucleated giant cells of 10-1 to Fig. 10-6). Approximately 60% of cases occur
osteoclastic type accounts for this morphologic overlap around the knee. Giant cell tumor occurs approximately
and for difficulties in segregating true giant cell tumors 4 times more frequently in the lower extremities than in
from unrelated giant cell–containing lesions. The key to the upper extremities. The other common locations of
distinguishing these lesions is in the unswerving adher- giant cell tumor in the epiphyses of long tubular bones
ence to clinicoradiologic correlation to arrive at a are the distal radius, proximal femur, humerus, and proxi-
diagnosis. mal fibula (Fig. 10-27). Giant cell tumor occurs less com-
Generally, a diagnosis of giant cell tumor is suggested monly in the distal tibia, very rarely in the distal ulna and
by the presence of a radiolucent lesion in the end of a fibula, and very uncommonly around the elbow joint.
long bone or an equivalent epiphyseal site in a skeletally When it occurs in the elbow joint, it is seen more fre-
mature individual. Other common locations include the quently in the distal humerus and very rarely in the proxi-
sacrum and “epiphyseoid” bones, such as the carpal and mal ulna and the radius.
tarsal bones and the patella. The true giant cell tumor, for In the epiphyses of the long tubular bones, a giant cell
practical purposes, does not arise in the craniofacial skel- tumor has radiologic features of an eccentric epiphyseal
eton, and it very rarely develops in nonepiphyseal loca- or metaphyseal defect with well-defined margins and an
tions. The short tubular bones of the hands and feet expanded cortex that is very frequently, at least focally,
present a particular problem because of the morphologic destroyed. Usually, there is no radiologically recogniz-
overlap with giant cell reparative granuloma, which has a able periosteal reaction. In a small percentage of cases,
predilection for this skeletal site. In this situation, atten- periosteal reaction can be present and is usually minimal.
tion to the specific site of involvement with respect to In thinner bones, such as the distal ulna or fibula,
epiphyseal location and skeletal maturity is particularly the lesion is usually centrally located with marked expan-
important. Perhaps the most important problem in histo- sion of the bone contour. Although hemorrhage, necro-
logic recognition of true giant cell tumor is created by sis, and a fibrohistiocytic xanthogranulomatous reaction
the tendency for this tumor to undergo fibrohistiocytic can develop in any giant cell tumor, regardless of its
reactive changes that can simulate benign or malignant location, such effects are seen much more frequently
primary tumors of fibrohistiocytic origin. Such changes and are much more extensive in the weight-bearing
can largely or even completely obscure the classic histo- bones of the lower extremities. Radiologic evidence of
logic appearance of giant cell tumor. It is this tendency fracture may be present. In this location, it is common
that has led to the misapplication of the term benign or for the tumor to be nearly completely replaced by necro-
malignant fibrous histiocytoma of bone to some examples sis and secondary fibrohistiocytic xanthogranulomatous
of altered giant cell tumor. Strict adherence to the use of reaction.
clinicoradiologic correlation and thorough sampling of In the long tubular bones, giant cell tumor should be
the tumor tissue avoid most of these errors in diagnosis. differentiated radiologically from chondroblastoma, nonos-
Another question that frequently arises is the extent sifying fibroma, chondromyxoid fibroma, aneurysmal bone cyst,
to which the aggressiveness of a giant cell tumor can be pigmented villonodular synovitis, and osteogenic sarcoma.
predicted on the basis of histologic criteria. Whether to Occasionally, fibroblastic tumors, such as desmoplastic
assign numeric grades or to use adjectival modifiers in fibroma, fibrosarcoma, and malignant fibrous histiocytoma,
designating local aggressiveness or metastasizing poten- may enter into the differential diagnosis.
tial has been debated extensively. Our experience indi- Chondroblastoma occurs in children and adolescents
cates that the use of such devices is without merit, except who have open epiphyses and typically has radiologically
to designate giant cell tumor as conventional or malig- detectable calcifications. Chondroblastomas at the ends
nant (either primary or secondary) on the basis of the of long bone seldom cause expansion of the contour
presence or absence of frankly sarcomatous features. unless they contain secondary aneurysmal bone cysts, and
cortical disruption is usually not present. Nonossifying
Conventional Giant Cell Tumor in Different fibroma is radiologically an eccentric metaphyseal-
Anatomic Sites diaphyseal lesion with well-developed scalloped sclerotic
margins in skeletally immature patients. These features
The conventional giant cell tumor has identical micro- are particularly helpful in distinguishing a giant cell
scopic features and biologic potential regardless of its tumor with an extensive secondary fibrohistiocytic

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 723

A B

C D
FIGURE 10-27  ■  Giant cell tumor involving long bones of extremities: radiographic features. A, Radiograph of distal end of radius
with disruption of cortex by expansile giant cell tumor. This is third most common site after femoral condyles and tibial plateau.
B, Radiograph of wrist shows eccentric tumor involving distal end of ulna. Note expanded contour, marginal sclerosis, and trabecu-
lation. C, Eccentric, totally lytic, distal tibial tumor. D, Giant cell tumor involving head of fibula.

ERRNVPHGLFRVRUJ
724 10  Giant Cell Lesions

reaction from a nonossifying fibroma. Chondromyxoid the difficulty in completely removing the tumor rather
fibroma is an eccentric metaphyseal lesion with scalloped than because of an intrinsically aggressive nature.
margins, fine trabeculations, and intact cortex. Its histo- A giant cell tumor of small tubular bones should
logic features are so characteristic that only in the most be differentiated mainly from a giant cell reparative gran-
unusual circumstances can it be confused with a giant cell uloma. Occasionally, these two entities may have very
tumor. similar microscopic features. If this diagnostic dilemma
De novo aneurysmal bone cysts produce lytic, eccen- occurs, radiologic evidence of involvement of the epiphy-
trically expansile defects in long bones that are easily sis in a skeletally mature patient should favor the diagnosis
distinguished from giant cell tumors by their predilection of a giant cell tumor. On the contrary, lack of epiphy-
for the diaphysis or metaphysis. Conversely, it is usual for seal involvement and the presence of unfused epiphy-
secondary aneurysmal bone cysts to occur in association seal plates should be considered as radiologic features in
with giant cell tumors of long bones. This combination favor of a giant cell reparative granuloma. Enchondroma
usually alters the radiologic appearance by accentuating is seldom confused with giant cell tumor because of its
the expansile and destructive features of the giant cell location in the shaft and its characteristic calcification
tumor. For this reason, aneurysmal bone cysts that occur pattern. Pigmented villonodular synovitis arising in tendon
in the ends of long bones of adults should always raise the sheaths of the hands and feet can erode short tubular
possibility of an underlying giant cell tumor. bones and thus simulate giant cell tumors.
Pigmented villonodular synovitis of major joints
sometimes invades the ends of long bones and produces Giant Cell Tumor of Vertebral Column.  Giant cell
lytic defects that can superficially resemble a giant cell tumor of the vertebral column exclusive of the sacrum is
tumor. This mimicry is complicated by the presence of extremely rare.25,48,94 In fact, many of the lesions previ-
many giant cells in the histologic sample of such lesions, ously classified as giant cell tumor in this site probably
but the clinical and radiologic evidence of bone defects represent other giant cell–containing entities. On the
in more than one bone bordering a joint can obviate this other hand, the sacrum is one of the more frequently
diagnostic problem. involved skeletal sites (Figs. 10-29 and 10-30). Patients
High-grade osteogenic sarcomas may occasionally with giant cell tumors of the vertebral column have a
produce osteolytic defects that mimic giant cell tumor. tendency to be younger than patients with lesions of long
Usually the patient’s age, the metaphyseal-diaphyseal tubular bones. When giant cell tumor affects a vertebral
location of the lesion, its ill-defined margins and features body, it usually occurs in a skeletally immature patient.
of massive cortical destruction, and the presence of A high proportion of giant cell tumors that involve the
prominent periosteal reaction are radiologic features vertebral column occur in women during the second
favoring malignancy. Fibroblastic tumors, such as desmo- decade of life.94 The bodies of vertebrae are usually the
plastic fibroma, fibrosarcoma, and malignant fibrous his- site of origin, and it almost never arises in the posterior
tiocytoma, can radiologically mimic giant cell tumor. arch or in the vertebral processes (Fig. 10-31). Radiologi-
However, they are typically diaphyseal in location and cally, the tumor presents as a well-circumscribed defect
show massive destruction of bone and cortex. Their his- best demonstrated by computed tomography and mag-
tologic features are usually diagnostic, but occasionally it netic resonance imaging. Plain radiographs may reveal
is difficult to distinguish a de novo malignant fibrous only compression fracture of the body or an abnormal
histiocytoma from secondary malignant change in an contour and deformed or missing pedicles. The radio-
underlying giant cell tumor. In such instances, the clinical logic features are nonspecific, and the exact diagnosis
history, radiographs, and careful sampling of the tumor usually cannot be made before biopsy. The clinical symp-
are very helpful. toms are related to pathologic fracture and compression
of the cord and nerve roots. Complete removal of the
Giant Cell Tumor of Small Bones of Hands and tumor in the spine and sacrum is technically difficult to
Feet.  Approximately 3% to 4% of all giant cell tumors perform. For this reason, radiotherapy and other alterna-
are found in the small bones of the hands and feet. Giant tive modalities, such as arterial embolization, are used to
cell tumors in younger patients appear to occur in these control the disease in the axial skeleton.
locations more frequently than lesions in the long The differential diagnosis of a giant cell tumor of
bones.115 These tumors probably occur more frequently the vertebral column should radiologically include aneu-
in the metacarpal and metatarsal bones than in other rysmal bone cyst, osteoblastoma, and giant cell reparative
bones of the hands and feet. In the small bones, it is granuloma. Rarely, eosinophilic granuloma in skeletally
unusual to see the eccentric epiphyseal origin unless the immature patients may present a diagnostic problem.
lesion is detected very early. The majority of lesions have Giant cell reparative granuloma of the vertebrae can
radiologic features of a central epiphyseal-metaphyseal mimic a giant cell tumor both radiologically and histo-
lytic defect with prominent enlargement of bone contours logically. Aneurysmal bone cyst and osteoblastoma are
(Fig. 10-28). Involvement of the epiphysis is almost a rule, more likely to arise in the posterior neural arch in con-
although in small bones a significant portion of the shaft trast to giant cell tumor and giant cell reparative granu-
or even the entire bone can be involved. A giant cell tumor loma, which characteristically involve the vertebral body.
of the acral skeleton may show somewhat more aggressive The distinctive anatomic locations of these lesions are
behavior than one in the long tubular bones (i.e., a higher best demonstrated by computed tomography and mag-
recurrence rate and a tendency to invade soft tissue), netic resonance imaging. The rapidly evolving, waferlike
probably more because of its location in small bones and collapse of vertebral bodies involved by eosinophilic

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 725

A B C

D E

F
FIGURE 10-28  ■  Giant cell tumor involving bones of hands and feet: radiographic features. A, Radiograph of thumb shows lytic lesion
in base of proximal phalanx of a 37-year-old woman (arrows). B, Giant cell tumor in metacarpal head of a 27-year-old woman.
C, Radiograph of same tumor shown in B 1 year later shows progressive enlargement and further cortical destruction. D, Giant
cell tumor of medial cuneiform of a 12-year-old girl (arrows). E, Computed tomogram of giant cell tumor of cuneiform bone shown
in D. F, Giant cell tumor in calcaneus of an adult woman.

ERRNVPHGLFRVRUJ
726 10  Giant Cell Lesions

A B

C D
FIGURE 10-29  ■  Giant cell tumor of the coccyx: radiographic and gross morphologic features. A, Sagittal computed tomogram (CT)
showing a large presacral tumor originating in the coccyx and filling the lower pelvic space and impinging on the rectum. B, Tran-
sected sacral resection specimen of the same case as shown in A showing a soft tan and partially hemorrhagic presacral tumor that
originates in the coccyx. Insert. Axial CT shows a sacrococcygeal mass with peripheral calcifications. C, T1-weighted sagittal mag-
netic resonance image (MRI) showing presacral low signal mass. D, Post-contrast T2-weighted MRI showing moderate signal
enhancement within the lesion.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 727

A B C

D E
FIGURE 10-30  ■  Giant cell tumor of sacrum: radiographic features. A, Anteroposterior radiograph of lumbosacral spine of a 16-year-
old girl with pain in lower back for 3 months. A lytic tumor was found in the sacrum. B, T2-weighted sagittal magnetic resonance
image (MRI) of sacral tumor shows high signal. C, Photomicrograph of conventional giant cell tumor. D and E, CT and T1-weighted
axial MRI demonstrate location of giant cell tumor in body of sacrum. (C, ×100) (C, ×100, hematoxylin-eosin).

granuloma helps distinguish this lesion from rare verte- flat bones, such as the pelvis, because of the nonspecificity
bral giant cell tumor of skeletally immature patients. of its imaging characteristics in this site. Because the
pelvis is one of the more common sites of involvement
Giant Cell Tumor of Flat Bones.  Giant cell tumor by Paget’s disease, when a giant cell tumor occurs in the
rarely occurs in flat bones. The pelvis is the most frequent pelvis, it is important to rule out this underlying condi-
site, accounting for approximately 4% of all giant cell tion, particularly in patients older than age 50 years.
tumors. Nearly half of the cases involving flat bones
occur in the ilium and pubis, with the iliac crest and the Giant Cell Tumor of Craniofacial Bones.  Giant cell
acetabulum being the most frequent sites. Individual tumor of the craniofacial bones is extremely rare in the
cases have been reported in the scapula, sternum, and absence of Paget’s disease; therefore, all cases of giant cell
ribs. In the flat bones, giant cell tumor has radiologic tumor in this location should be carefully examined for
features of large lytic defects with markedly expanded evidence of underlying Paget’s disease. The few cases that
contours of bone and a large soft tissue component. In fulfill the criteria of de novo giant cell tumor of craniofa-
these anatomic sites, the extent of the disease and its cial bones are most frequently located in the sphenoid
relationship to adjacent structures are best evaluated by bone or other anatomic locations within the base of the
computed tomography and magnetic resonance imaging. skull such as the clivus90,116 (Fig. 10-32). The extent of the
The radiographic features are nonspecific and overlap disease and its relation to other structures are best evalu-
with those of chondroblastoma, fibrosarcoma, and malig- ated by computed tomography and magnetic resonance
nant fibrous histiocytoma. The diagnosis of giant cell imaging. It usually extends into the orbit, superior nasal
tumor is less likely to be made on radiologic grounds in cavity, or anterior cranial fossa, causing a variety of clinical

ERRNVPHGLFRVRUJ
728 10  Giant Cell Lesions

A B

C D
FIGURE 10-31  ■  Giant cell tumor of vertebral column: radiographic and microscopic features. A, Anteroposterior radiograph of lumbar
spine in a 18-year-old woman shows radiolucency and partial collapse of L4. B, Lateral view of giant cell tumor involving body
of fourth lumbar vertebra. C, Computed tomogram of L4 shows radiolucent expansile tumor of vertebral body that is beginning
to compress dural sac. D, Photomicrograph of conventional giant cell tumor shown in previous images (D, ×200) (D, ×200,
hematoxylin-eosin).

symptoms related to the involvement of the adjacent logically, but they do not share their biologic characteristics.
structures.116 An exact radiologic diagnosis is usually not Pigmented villonodular synovitis of the temporomandib-
possible because the radiographic features are indistin- ular joint occasionally produces temporal bone destruc-
guishable from more frequent primary bone lesions such tion that can be mistaken for a true giant cell tumor.
as fibrous dysplasia, brown tumor of hyperparathyroid-
ism, and giant cell reparative granuloma. These lesions Multifocal Conventional Giant
often have been inappropriately classified as giant cell
tumors, especially in oral pathology and ophthalmic lit-
Cell Tumor
erature. Giant cell reparative granulomas are reactive Multifocal giant cell tumors are extremely rare without
fibroosseous lesions with prominent giant cell compo- underlying Paget’s disease. However, there are well-
nents that may closely resemble giant cell tumors histo- documented cases of concurrent involvement of more

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 729

C
FIGURE 10-32  ■  Giant cell tumor involving sphenoid bone of a 19-year-old man with headaches and blurred vision: radiographic
features. A and B, Computed tomograms (CTs) show lytic expansile lesion involving wall of sphenoid sinus. C, Sagittal reconstruc-
tion CT demonstrates posterior placement of tumor in sphenoid bone.

ERRNVPHGLFRVRUJ
730 10  Giant Cell Lesions

than one, usually two or three, anatomic sites without any giant cell tumor of bone. They have classic radiologic
evidence of underlying Paget’s disease.23,28,34,43,44,101,102,104 A features of lung metastasis but may be partly calcified.
multifocal giant cell tumor often involves its typical ana- Usually, there are few nodules measuring 1 to 2 cm in
tomic sites, such as the epiphyses of long tubular bones. diameter. Pulmonary metastasis can also occur as a soli-
Sometimes both sides of the knee joint can be involved tary metastatic lung tumor. Occasionally, the patient may
(i.e., separate lesions can be present in the distal femur have multiple miliary-type small nodules that measure
and the proximal tibia of the same extremity). In these several millimeters in diameter. The nodules have micro-
rare cases, pigmented villonodular synovitis enters into the scopic features of conventional giant cell tumor. The
differential diagnosis. Lesions can also be present in periphery of the nodule has a thick rim of reactive fibrous
several diverse anatomic sites such as separate bones of tissue and, at least focally, a shell of reactive bone. The
one extremity or in extremity bones on one side of the latter may be identifiable on plain radiographs or com-
body.89 However, in some cases the lesions are distributed puted tomograms of the chest. There is nothing charac-
without any apparent anatomic order. The behavior of teristic in the radiologic or histologic findings of the
multifocal giant cell tumors does not differ from that of primary lesion, including the presence of blood vessel
a unifocal lesion.79 In these cases, it is essential to rule out invasion that can reliably predict the development of pul-
multifocal giant cell reparative granuloma and underlying monary implants in a patient with conventional giant cell
brown tumor of hyperparathyroidism. tumor. The reported development of pulmonary implants
in a male patient who tested positive for the human immu-
Conventional Giant Cell Tumor nodeficiency virus and who had a conventional giant cell
tumor and in an immunocompromised patient who had
with Pulmonary Metastases had an allograft bone marrow transplant suggest that the
A number of well-documented cases of conventional host’s immune response may play an important role in
giant cell tumors have metastasized to the lungs (Figs. controlling the clinical behavior of these neoplasms.58
10-33 to 10-35).10,51,62,63,69,86,88,114 This phenomenon is rare
and probably occurs in not more than 1% to 2% of
patients with conventional giant cell tumors. The pulmo- GIANT CELL TUMOR
nary metastases, or so-called pulmonary implants, in his- IN PAGET’S DISEASE
tologically benign giant cell tumor should be distinguished
from the true metastases observed in malignant giant cell Giant cell tumors may complicate Paget’s disease of bone
tumors. They have distinct radiologic and pathologic but do so much less frequently than osteosarcoma or
features, self-limited growth potential, and a relatively malignant fibrous histiocytoma (Figs. 10-36 and 10-37).41
good prognosis.114 Some tumors may even spontaneously When they do, they are morphologically indistinguish-
regress. Therefore such lesions should be resected when able from a conventional giant cell tumor in patients
it is technically feasible with a good prospect of cure. without underlying Paget’s disease (Fig. 10-36). Giant
However, some patients may die as a result of progressive cell tumor that occurs with Paget’s disease may be mul-
growth of multiple inoperable lung lesions. The clinical tifocal and has a predilection for the craniofacial bones
behavior of pulmonary implants in conventional giant and other anatomic sites frequently involved in Paget’s
cell tumor is therefore unpredictable. Although the disease.33,41,43,49,71,81 Unlike with conventional giant cell
overall prognosis of pulmonary implants is much better tumor, when it occurs with Paget’s disease it shows no
than that of conventional pulmonary metastatic disease, predilection for the epiphyses of long tubular bones. The
approximately 25% of patients eventually die of the neoplastic nature of giant cell tumor in Paget’s disease
disease. Pulmonary metastases in histologically benign and its true relationship to conventional giant cell tumor
giant cell tumor occur typically within 3 years after the are questionable. There is usually no recurrence after
removal of the primary lesion in bone. In some instances, simple curettage, and it can be controlled by systemic
pulmonary metastases may appear many years or even treatment with steroids. Subsequent multifocal lesions
decades after removal of the primary lesion.117 It is con- may develop in the vicinity of the primary tumor or in
ceivable that they are the result of curettage and mechan- other skeletal sites involved by Paget’s disease.78 A giant
ical seeding of blood vessels or lymphatics with small cell tumor arising in association with Paget’s disease may
fragments of tumor tissue that are subsequently implanted represent a reactive process that only superficially mimics
in the lung. They are, however, occasionally observed in a neoplasm. This is further supported by the fact that
patients without prior surgery at the time of diagnosis of patients with Paget’s disease may develop a giant cell
the primary bone tumor. In such instances, pulmonary lesion that is microscopically more similar to a giant
metastasis is most likely related to invasion by a giant cell cell reparative granuloma than to a conventional giant
tumor into blood vessels, a finding that can be docu- cell tumor.112 A familial variant of Paget’s disease has been
mented in approximately 30% of conventional giant cell described in several families living in New Jersey whose
tumor resection specimens. In extremely rare cases the members had multifocal giant cell tumors involving the
so-called histologically benign giant cell tumor of bone craniofacial bones and the vertebral bodies. Ancestors
can metastasize to other sites (e.g., lymph nodes, bone, of these families were traced to the southern Italian city
and skin).82,111,117 of Avellino in the Campania region.46,85 Subsequently, a
Initially, the pulmonary metastases are asymptomatic high prevelance of Paget’s disease with unique familial
and are usually an incidental finding on chest x-ray films clustering was identified in the entire Campania region.84
taken during follow-up of the patient with a history of Text continued on p. 736

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 731

A B

C D
FIGURE 10-33  ■  Conventional giant cell tumor with pulmonary implants: microscopic features. A, Metastatic conventional giant
cell tumor in lung. B, Higher magnification of A showing conventional (nonmalignant) cytoarchitectural features of giant cell tumor
in pulmonary implant. C, Formation of reactive osteoid associated with pulmonary implants of conventional giant cell tumor.
D, Pulmonary implant of giant cell tumor delineated by a reactive shell of bone and peripheral scarring. (A and D, ×25; B, ×100;
C, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
732 10  Giant Cell Lesions

A C

D E

FIGURE 10-34  ■  Conventional giant cell tumor with pulmonary implants: microscopic features. A, Small (miliary) pulmonary implants
of conventional giant cell tumor in lung. B and C, Higher magnification of A showing well defined nodules of conventional
giant cell tumor with peripheral scarring in lung. D, Small (miliary) implant of conventional giant cell tumor in subpleural
lung associated with prominent reactive bone forming an incomplete peripheral shell. E, Higher magnification of D shows a pulmo-
nary implant predominantly composed of mononuclear cells with prominent reactive bone. (A and D, ×25; B, C, and E, ×50.)
(A-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 733

A B

C D
FIGURE 10-35  ■  Conventional giant cell tumor with pulmonary implants: microscopic features. A and B, Pulmonary implants of
conventional giant cell tumor with prominent reactive bone. C and D, Higher magnifications of A and B showing prominent
interconnecting bone trabeculae. The tumor is composed predominantly of mononuclear histiocytic cells with stromal fibrosis
obscuring, together with reactive bone, the classic microscopic architecture of giant cell tumor. (A and B, ×25; C and D, ×50.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
734 10  Giant Cell Lesions

A B

C D
FIGURE 10-36  ■  Giant cell tumor of vertebral column complicating Paget’s disease of bone: radiographic features. A, Lateral radio-
graph of skull of a 69-year-old woman with an 8-year history of Paget’s disease. Note “cotton-wool” densities and thickened cal-
varium. B, Radiograph of lumbar spine shows “picture frame” vertebral characteristics of involvement by Paget’s disease (arrows).
C, Oblique view of thoracic spine with partial collapse of bodies of T5 and T6 at site of giant cell tumor (arrows). D, T1-weighted
sagittal magnetic resonance image shows tumor (arrows) with low signal compressing dural sac.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 735

A B

C D
FIGURE 10-37  ■  Giant cell tumor complicating Paget’s disease of bone: microscopic features. A, Photomicrograph shows pagetoid
changes in vertebral bone of patient shown in Figure 10-28. Note mosaic pattern of cement lines, as well as prominent osteoclastic
resorption and osteoblastic rimming. B, Conventional giant cell tumor arising in thoracic vertebra involved by Paget’s disease.
C, Electron micrograph of nucleus of giant cell containing viruslike filamentous inclusion. D, Higher magnification of filamentous
inclusion body resembling paramyxovirus particles. Inset, Inclusions could be identified by light microscopy as eosinophilic bodies
within giant cell nuclei. (A, ×50; B, ×150; C, ×1400; D, ×30,000; inset, ×200).

ERRNVPHGLFRVRUJ
736 10  Giant Cell Lesions

It appears that Paget’s disease in these patients shows a documented if the tumor is carefully sampled. The key
higher clinical severity and greater frequency of neoplas- microscopic features in diagnosing malignant change in
tic transformations including giant cell tumor of bone.84 giant cell tumor are the presence of atypical mitoses and
Some of the patients from this group responded well to true nuclear pleomorphism in a spindle-cell component
treatment with high doses of dexamethasone. A large that is next to or intermixed with a conventional giant
pedigree-based analysis of giant cell tumor associated cell tumor. These features are easily recognizable in high-
with familial Paget’s disease failed to identify a single grade tumors. However, a distinction between low-grade
predisposing locus within the human genome.35 Giant fibrosarcoma or malignant fibrous histiocytoma and reac-
cell tumors in Paget’s disease frequently contain intra- tive fibrohistiocytic proliferation is not always easy. This
nuclear inclusions, the presence of which can sometimes is a particularly frequent diagnostic problem in small
be distinguished in conventional histologic sections as biopsy samples in which the architecture of the tissue in
intranuclear eosinophilic bodies. They are best docu- question and especially its interaction with other compo-
mented by electron microscopy, which reveals 10-nm nents of the lesion cannot be evaluated. In such instances,
thick filamentous intranuclear inclusions as well as occa- an examination of additional biopsy material may be
sional cytoplasmic bodies (Fig. 10-37). They resemble required to solve this diagnostic dilemma. Occasionally,
paramyxovirus inclusions, but the precise nature of these a florid reactive proliferation of fibroblasts with high
structures remains to be determined.1,16,61 Attempts to mitotic activity may mimic fibrosarcoma. In such instances,
identify measles virus in giant cell tumor of Paget’s attention should be directed toward the gradual blending
disease and in Paget’s disease itself with polymerase chain of such suspicious areas with clearly benign reactive tissue
reaction (PCR)–based techniques failed to document ele- to avoid a false diagnosis of sarcomatous change. In addi-
ments of the measles genome.75 tion, it is sometimes helpful to note the resemblance of
such areas to a monolayer culture of proliferating fibro-
blasts. Bone production is not a feature of a conventional
MALIGNANT GIANT CELL TUMOR giant cell tumor, and there is no direct production of
osteoid by tumor cells. However, reactive bone is nearly
Giant cell tumor has a tendency to progress to a high- always present at the periphery of the lesion and at times
grade malignancy, which may develop within the preex- can extend within it for a considerable distance. The
isting conventional giant cell tumor typically after several diagnosis of osteosarcoma complicating a conventional
recurrences.5,8,9 Such tumors are referred to as secondary giant cell tumor requires the presence of a malignant
malignant giant cell tumors. A primary de novo malig- bone-forming tumor (i.e., the presence of pleomorphic
nant giant cell tumor is extremely rare. cells with atypical mitoses that produce tumor osteoid).

Secondary Malignant Giant Cell Tumor Primary Malignant Giant Cell Tumor
Malignant change typically occurs after multiple local As mentioned, a primary de novo malignant giant cell
recurrences of a conventional giant cell tumor. It tumor is extremely rare and most frequently occurs in
can also happen after a single local recurrence.8,9,36,40,93 typical anatomic sites involved by a conventional giant
Occasionally, malignant change can be identified in the cell tumor.67 The microscopic pattern is that of high-
primary conventional giant cell tumor at the time of its grade pleomorphic sarcoma with occasional telan-
initial diagnosis. Sarcomatous change is more likely to giectatic features. There is no collagen and no direct
happen in a previously irradiated lesion, usually 5 years osteoid production by tumor cells. Reactive bone can be
or more after the initial radiation exposure. Sarcomatous present focally and should be appropriately identified as
change can also develop many years or even decades after not being an integral part of the lesion. There is high
removal of the primary lesion.36,72,87 A giant cell tumor mitotic activity of both stromal and tumor giant cells,
with malignant change has radiologic features of a with multiple atypical mitoses present. These features
large destructive lesion with cortical destruction and can be uniformly present in the entire lesion or can be
prominent invasion into soft tissue (Fig. 10-38). However, focal. In reference to focal changes, it may be difficult
these features are frequently observed in a locally in some cases to distinguish between primary malignant
advanced conventional giant cell tumor. On the other giant cell tumor and secondary malignant change in a
hand, a sarcomatous component can be present in a giant conventional giant cell tumor. In cases with prominent
cell tumor that has no radiologic features of aggressive- atypia and numerous atypical mitoses, the diagnosis of
ness (Fig. 10-40). Therefore as previously mentioned, malignancy is easy. However, as pointed out originally by
radiologic features of aggressiveness in giant cell tumors Jaffe et al.,47 there is a continuum of microscopic change
do not correlate well with histologic criteria of malig- between clearly benign (conventional) and malignant
nancy or with metastatic potential. giant cell tumors. The degree of biologic aggressiveness
Microscopically, malignant change in giant cell tumor increases as stromal cells become more prominent and
may be in the form of fibrosarcoma, malignant fibrous his- show an increase in mitotic activity. This is accompanied
tiocytoma, osteosarcoma, or undifferentiated high-grade pleo- by a decrease in the number of giant cells. According to
morphic sarcoma (Figs. 10-39 to 10-42). In extremely rare this concept, Jaffe et al. divided giant cell tumors into
cases, a phenotypic switch to epithelial malignancy with three grades. Grade 1 corresponds to a conventional,
squamous differentiation has been reported.59 A focus microscopically benign giant cell tumor, and grade 3 is
of conventional benign giant cell tumor usually can be Text continued on p. 742

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 737

A B

C D
FIGURE 10-38  ■  Secondary malignant giant cell tumor: radiographic and microscopic features. A, Anteroposterior radiograph of a
recurrent giant cell tumor presenting as a large expansile lesion, which destroyed the cortex of distal femoral epiphysis. B, Micro-
scopic features of the tumor in A showing a conventional component of this giant cell tumor. C and D, High-grade undifferentiated
sarcomatoid component of the lesion shown in A and B with pronounced atypia and brisk mitotic activity (B-D, ×200.)
(B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
738 10  Giant Cell Lesions

A B

C D
FIGURE 10-39  ■  Secondary malignant giant cell tumor: microscopic features. A, A component of the tumor showing cytoarchitectural
features consistent with a conventional giant cell tumor. B, Higher magnification of A showing mononuclear histiocytic cells and
scattered multinucleated giant cells. C, Sarcomatoid component of the tumor showing proliferation of atypical spindle cells. Note
the absence of multinucleated giant cells. D, Higher magnification of C showing disorganized proliferation of undifferentiated sar-
comatoid cells with pronounced atypia. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 739

A B

C D
FIGURE 10-40  ■  Secondary malignant giant cell tumor: radiographic and microscopic features. A, Eccentric lytic lesion of distal
femoral epiphysis. B, Computed tomogram of lesion in A shows eccentric, sharply demarcated defect with focally destroyed
cortex. C and D, Areas of typical osteosarcoma with prominent tumor osteoid were present in addition to conventional giant cell
tumor. (C and D, ×100.) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
740 10  Giant Cell Lesions

A B

C D
FIGURE 10-41  ■  Primary malignant giant cell tumor: microscopic features. A, A component of the tumor with features resembling
conventional giant cell tumor. Note the dominance of mononuclear cells with a few multinucleated giant cells. B, Sarcomatoid
component of the tumor composed of undifferentiated mesenchymal cells in confluent growth. C, Higher magnification of
B showing sarcomatoid plump, spindle, and oval undifferentiated mesenchymal cells. D, Sarcomatoid cells growing in a dense col-
lagenous stroma mimicking osteoid. (A and B, ×100; C and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 741

A B

C D
FIGURE 10-42  ■  Primary malignant giant cell tumor: microscopic features. A, Undifferentiated pleomorphic sarcoma of the distal
femeral epiphysis. B, Higher magnification of A showing pronounced atypia and mitotic activity in sarcomatoid cells. C, Undifferenti-
ated sarcomatoid cells with pronounced atypia and focal clear cell change in another area of the tumor shown in A and B. D, Higher
magnification of C showing pronounced atypia and focal clearing of the cytoplasm in sarcomatoid cells. (A and C, ×100; B and
D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
742 10  Giant Cell Lesions

a clearly histologically malignant lesion with metastatic giant cell–containing fibrous appearance in the short
potential. Grade 2 corresponds to an intermediate tumor tubular bones of hands and feet. Subsequently, micro-
with increased stromal cellularity, high mitotic activity, scopically similar lesions of vertebrae and ethmoid bones
and more than usual local aggressiveness. Grade 3 tumors were described by Sanerkin et al.194 under the name solid
frequently behave in a malignant fashion. However, it is variant of aneurysmal bone cyst. All such lesions are micro-
very difficult to predict the behavior of grade 1 and 2 scopically similar and were perceived as representing a
lesions in terms of their local growth potential and ten- similar process that merely presents in different skeletal
dency for recurrence. For this reason, Jaffe himself, along sites.153,154,158,189,194,207,209-211 Modern data implies that giant
with many others, discontinued the numeric grading of cell reparative granulomas in different anatomic sites
giant cell tumors. The abandoned grading system and the have distinct pathogenesis and unique genetic back-
overall approach to giant cell tumors have alerted clini- grounds but are characterized microscopically by over-
cians and pathologists to a spectrum of biologic behaviors lapping features with the presence of multinucleated
in these lesions. giant cells that form unevenly distributed clusters within
fibrous stroma that contains areas of reactive bone
formation.
GIANT CELL REPARATIVE GRANULOMA Initially, giant cell reparative granulomas were per-
ceived as a peculiar reaction linked to intraosseous hem-
A group of lesions microscopically designated as giant orrhage, trauma, or both that may develop in normal
cell reparative granuloma is pathogenetically heteroge- bone or in preexisting lesions, such as fibrous dyspla-
neous and represents a range of conditions with overlap- sia, hyperparathyroid bone disease, or rarely Paget’s
ping microscopic presentations. These can be separated disease.126,142,144,198,204 The relationship to aneurysmal
into several distinct clinicopathologic entities on the basis bone cyst was postulated because solid components of
of their unique presentation, associated disorders, and aneurysmal bone cysts carry many microscopic similari-
genetic background. Giant cell reparative granulomas ties with giant cell reparative granulomas. In fact, giant
that are associated with primary or secondary hyperpara- cell reparative granulomas were considered as an initial
thyroidism are truly reactive conditions and are referred stage in the spectrum of intraosseous responses that,
to as the brown tumor of hyperparathyroidism. However, in some instances, can culminate as rapidly expanding
giant cell reparative granulomas of the appendicular skel- destructive multicystic lesions.137,142,150,151,199,202,207,209 Fur-
eton, especially those involving the bones of the hands thermore, in nearly 30% of giant cell reparative granu-
and feet, are related to a solid aneurysmal bone cyst and lomas, there are microscopic foci with multiple cavities
frequently carry the USP6 gene rearrangement.121 Giant filled with blood resembling aneurysmal bone cysts.199
cell reparative granulomas affecting the craniofacial
bones are pathogenetically distinct from those in the Incidence and Location
appendicular skeleton and typically do not carry the
USP6 gene rearrangement.121 Most giant cell reparative The peak age incidence of giant cell reparative granu-
granulomas are solitary lesions except those that are asso- loma is in the second decade of life, and most patients
ciated with hyperparathyroidism or Paget’s disease, which are between ages 10 and 25 years.124,138 This lesion
are often multifocal. Multifocal giant cell reparative gran- usually occurs in the craniofacial bones, such as the
ulomas of the jaw bones are associated with a unique mandible and maxilla.122,124,137,154,162,180 A second common
genetic abnormality most often involving the SH3BP2 location is in the small bones of the hands and
gene and are clinically referred to as cherubism. Giant feet.128,131,145,148,153,177,181,183,187,189 It is extremely rare in long
cell–containing lesions resembling giant cell reparative tubular bones and vertebrae.142,152,155,200 Single cases have
granulomas involving both bones and soft tissue are an also been reported in the flat bones such as the pelvis. It
integral part of Noonan’s syndrome, characterized by appears that patients with giant cell reparative granuloma
multiple developmental abnormalities in a background of of the long tubular bones and the vertebrae are somewhat
heterogeneous mutations involving SOS1, RAF1, NRAS, older; the peak age incidence in these anatomic sites is in
SHOC2, CBL, and NF1.201 The involvement of the RAS- the third decade of life. Most giant cell reparative granu-
MAPK pathway and mutations of PTPN11 appear as a lomas that are not associated with hyperparathyroidism
dominant theme in Noonan’s syndrome. or Paget’s disease and are not a part of cherubism or
Noonan’s syndrome are solitary lesions. Only in rare
Definition instances do they involve multiple bones (e.g., of the acral
skeleton).136 The peak age incidence and the most fre-
Giant cell reparative granulomas are uncommon benign quent sites of involvement are shown in Figure 10-43.
lesions with a predilection for the craniofacial bones.
Since Jaffe’s original description in 1953,162 multiple Radiographic Imaging
reports of this entity in craniofacial bones and in other
skeletal sites have appeared in the literature. In 1962, In craniofacial bones, giant cell reparative granuloma
Ackerman and Spjut120 described the first two cases produces a round or oval area of lucency with occasional
involving the small tubular bones of the hands and des- fine trabeculations (Fig. 10-44). It has distinct borders
ignated the lesions as giant cell reactions. In 1980 the term with minimal reactive sclerosis. The contour of bone can
giant cell reparative granuloma was proposed by Lorenzo be expanded with markedly thinned but usually intact
and Dorfman176 in reference to reactive lesions with a cortex and no periosteal reaction.132

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 743

Peak
incidence
10 25
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 10-43  ■  Giant cell reparative granuloma. Peak incidence and frequent sites of skeletal involvement. Most frequent site is
indicated by solid black arrow.

In the small tubular bones of the hands and feet, the amounts of collagen. The cellularity of the stromal tissue
lesion may be metaphyseal, diaphyseal, or both and does is moderate to high. Normal mitoses are rarely found in
not cross the unfused growth plate (Fig. 10-45). In skel- fibroblasts. The key feature is the presence of multinucle-
etally mature patients, the lesion can involve the epiphy- ated giant cells that are unevenly distributed and form
sis or even the entire bone. Epiphyseal involvement is clearly recognizable clusters separated by bundles of scar-
secondary to progression from the metaphysis, and there like stromal tissue (Figs. 10-48 and 10-49). The giant
is always a significant metaphyseal and sometimes a cells and their clustering are particularly evident in areas
diaphyseal component. In the small tubular bones, the of hemorrhage. Microscopic evidence of fresh and old
lesion is centrally located with markedly expanded hemorrhage is almost always present. Usually, there are
contour of the bone and a thinned but intact cortex. few mononuclear inflammatory cells and trabeculae of
A giant cell reparative granuloma of the long tubular newly formed reactive bone rimmed by osteoblasts. The
bones produces an eccentric metaphyseal or diaphyseal reactive bone components sometimes are quite extensive
lytic defect (Fig. 10-46). The contour of bone can be and obscure other elements of the lesion (Figs. 10-49 and
eccentrically expanded with a thinned intact cortex. In 10-50). Microscopic evidence of hemorrhagic cyst forma-
vertebrae, it presents as an expansile lytic lesion of the tion resembling an aneurysmal bone cyst is frequently
body (Fig. 10-47). present. On the other hand, some giant cell reparative
granulomas may be hypercellular, showing florid prolif-
eration of plump fibrohistiocytic cells. Such lesions
Gross Findings
should not be confused with malignancy (Fig. 10-51).
The lesion is composed of friable, gritty tissue that is
tan-gray or brown. There may be evidence of cystic Special Techniques
change and hemorrhage.
Ultrastructurally, multinucleated giant cells of giant cell
reparative granuloma are similar to those present in giant
Microscopic Findings
cell tumor and express features of osteoclasts.124,143,161,174,195
Giant cell reparative granuloma is composed of fibrous On the other hand, the surrounding stromal spindle
stromal tissue with spindle-shaped fibroblasts and varying cells typically show myofibroblastic differentiation.143 In

ERRNVPHGLFRVRUJ
744 10  Giant Cell Lesions

C
FIGURE 10-44  ■  Giant cell reparative granuloma of craniofacial bones: radiographic features. A, Lytic lesion in left maxilla (arrows).
B, Lytic focus in right maxilla. C, Axial computed tomogram showing destructive lesion in right mandible.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 745

A B C

D E F
FIGURE 10-45  ■  Giant cell reparative granuloma of short tubular bones: radiographic features. A, Lytic lesion of fourth metacarpal
bone in a 14-year-old boy. Note that lesion is adjacent to but does not cross physis. B, Typical appearance of giant cell reparative
granuloma of fifth metacarpal bone in a skeletally mature 15-year-old boy with involvement of epiphysis. C, Expansile eccentric
lesion involving distal phalanx of fifth finger. D, Expansile lytic lesion of a 7-year-old boy. Note that lesion is diaphyseal and does
not cross physis. E, A seventeen-year-old boy with pathologic fracture through expanded lytic diaphyseal lesion involving shaft of
first metatarsal bone. F, Extensive lytic lesion involving almost entire first metatarsal bone in a skeletally mature 17-year-old girl.

ERRNVPHGLFRVRUJ
746 10  Giant Cell Lesions

A B

C D
FIGURE 10-46  ■  Giant cell reparative granuloma of long bones: radiographic features. A, Lytic metaphyseal lesion of proximal fibula
in a skeletally immature 15-year-old boy. Note marked expansion of bone contour. B, T1-weighted coronal magnetic resonance
image of lesion in A shows high signal in tumor. Note that tumor does not cross physis. C, Sharply demarcated lytic lesion of distal
fibular shaft in a skeletally mature 16-year-old girl. D, Lytic lesion with marginal sclerosis in distal femoral shaft (metaphysis).

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 747

A B C
FIGURE 10-47  ■  Giant cell reparative granuloma of vertebral column: radiographic features. A, Lateral view of lytic lesion involving
body of C2. B and C, T2-weighted coronal magnetic resonance imageshows high signal in lesion involving body of C2.

contrast with giant cell tumor, mononuclear stromal cells Giant cell reparative granuloma can be microscopi-
with a macrophage-like ultrastructure are not present. cally confused with giant cell tumor. The clustering
Cytogenetic testing performed on a few cases of giant cell of giant cells and the absence of typical mononuclear
reparative granuloma show some clonal rearrangement stromal cells are features of a giant cell reparative gran-
involving different chromosomes, including t(1;17;18) uloma. Conversely, the uniform distribution of multi-
and t(X;4)(q22; q31.3), suggesting that although tradi- nuclear giant cells in mononuclear stromal cells favors
tionally perceived as reactive proliferations, they may in giant cell tumor. Sometimes a secondary fibrohistio-
fact represent clonally expanding peculiar neoplastic pro- cytic reaction in giant cell tumor can be microscopically
cesses.178 Some studies have shown upregulation of SRC indistinguishable from giant cell reparative granuloma.
and WWOX genes in giant cell reparative granulomas of In such instances, the presence of foci of conventional
the jaw bones.123,165 Genetic profiling of a single case of giant cell tumor and its typical radiographic features are
giant cell reparative granuloma using microarray tech- helpful in making the diagnosis. The metaphyseal loca-
nology has shown an upregulation of cell cycle and sig- tion, diaphyseal location, or both of the lesion and the
naling transduction genes.133 presence of unfused growth plates are features favor-
ing giant cell reparative granuloma. On the other hand,
Differential Diagnosis the involvement of the epiphyseal end of the bone in
a skeletally mature patient should favor a giant cell
Giant cell reparative granuloma should be differenti- tumor.
ated from brown tumor of hyperparathyroidism, giant cell Nonossifying fibroma, especially with a prominent
tumor, nonossifying fibroma, and pigmented villonodular multinucleated giant cell reaction, can occasionally be
tenosynovitis. confused with a giant cell reparative granuloma. Charac-
Microscopically, giant cell reparative granuloma is teristic microscopic features of nonossifying fibroma
indistinguishable from a brown tumor of hyperparathy- consist of a fibrohistiocytic stroma with a storiform
roidism. Therefore in every case of giant cell reparative pattern and a prominent xanthogranulomatous reaction.
granuloma, it is important to establish the serum calcium Radiographically, nonossifying fibroma produces an
and phosphorus levels and to evaluate radiographic and eccentric metaphyseal lytic defect with scalloped and
clinical data to rule out underlying hyperparathyroidism. sclerotic margins and occurs predominantly in the long
In fact, the multifocality of giant cell reparative granu- tubular bones of the lower extremities in skeletally imma-
loma can provide a clue that it may be associated with a ture patients. This anatomic site is very unusual for a
metabolic disorder or is a part of a genetic predisposing giant cell reparative granuloma.
syndrome.163 Text continued on p. 752

ERRNVPHGLFRVRUJ
748 10  Giant Cell Lesions

A B

C D
FIGURE 10-48  ■  Giant cell reparative granuloma: microscopic features. A, Scattered multinucleated giant cells in a background of
hemorrhagic stroma. B, Low power view showing an area of hemorrhage with multinucleated giant cells juxtaposed with fibrous
areas containing reactive bone formation. C, Higher magnification of B showing multinucleated giant cells in a background of fresh
hemorrhage with adjacent fibrous area containing reactive bone formation. D, Reactive bone formation with prominent osteoblastic
rims. (A, C, and D, ×100; B, ×50) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 749

A B

C D
FIGURE 10-49  ■  Giant cell reparative granuloma: microscopic features. A, Low power view of giant cell reparative granuloma showing
an even distribution of giant cells, which cluster in the areas of hemorrhage. B, Higher magnification of A showing an area of hem-
orrhage with a cluster of multinucleated giant cells. C, Low power view showing fibrous area with reactive bone formation. D, Higher
magnification of C showing reactive osteoid with prominent osteoblastic rims within loose fibrous stromal tissue. (A and C, ×50;
B and D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
750 10  Giant Cell Lesions

A B

C D
FIGURE 10-50  ■  Giant cell reparative granuloma: microscopic features. A, Irregular distribution of multinucleated giant cells. Note
that the multinucleated giant cells cluster in the area of hemorrhage. B, Higher power view of A showing multinucleated giant cells
in a background of fresh hemorrhage. C and D, Higher magnification showing multinucleated giant cells with hemosiderin granules
within their cytoplasm (A, ×50; B, ×100; C and D, ×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 751

A B

C D
FIGURE 10-51  ■  Giant cell reparative granuloma: microscopic features. A, Irregular distribution of multinucleated giant cells in spindle-
cell stroma associated with hemorrhage and inflammatory cell infiltrate. B, Higher magnification of A showing an aggregate of
unevenly distributed multinucleated giant cells associated with fresh hemorrhage. C, Irregular distribution of multinucleated giant
cells in fibrous stroma. D, Higher magnification of C showing a cluster of multinucleated giant cells associated with hemorrhage
and inflammatory cell infiltrate. Florid proliferations with nuclear atypia in some giant cell reparative granuloma may raise suspicion
of malignancy. (A and C, ×25; B and D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
752 10  Giant Cell Lesions

Pigmented villonodular synovitis can be problematic, rearrangement of the same USP6 gene. This gene is not
particularly if it involves unusual articular sites such as involved in lesions developing in the craniofacial bones.
the temporomandibular and spinal facet joints and teno- Most examples of this lesion occur in the jaw bones
synovial structures of the hands and feet with secondary and nonepiphyseal sites of short tubular bones of the
involvement of bone. Radiographically, it can produce hands and feet, where the cause of disease is more obscure.
eccentric bone erosions that result from the proliferating Although these two groups of lesions have overlapping
juxtaarticular synovial masses. Microscopically, identifi- pathologic features, the current evidence indicates that
cation of synovial membrane containing the characteris- they may evolve via distinct pathologic mechanisms. The
tic histiocytic and giant cell infiltrates serves to distinguish most difficult differential diagnosis between giant cell
pigmented villonodular synovitis from giant cell repara- tumor and giant cell reparative granuloma involves those
tive granuloma. Radiographic involvement of bone on lesions that develop in the epiphyseal ends of phalanges,
both sides of the joint favors the diagnosis of pigmented metacarpals, and metatarsal bones, where the distinction
villonodular synovitis with secondary bone erosion. often cannot be made on histologic grounds. The diffi-
culty is further complicated by the fact that giant cell
Treatment and Behavior reparative granuloma in short tubular bones has a ten-
dency to recur after curettage and bone grafting in a
Giant cell reparative granulomas of the jaws or short tu- significant proportion of cases.
bular bones are generally adequately treated by curettage
with or without bone grafting.130,151,166,176,189,190,192,205,211,212
Giant cell reparative granuloma of the maxilla has been CHERUBISM AND
reported to show good, although transient, response to RELATED SYNDROMES
steroid, calcitonin, and interferon alfa-2b treatment.129,140
In phalangeal, metacarpal, or metatarsal lesions that Cherubism is an autosomal dominant inherited syn-
destroy most or all of the bone of origin, digital or ray drome characterized by extensive bone remodeling of
amputation may be necessary, particularly in recurrent the jaw bones followed by the development of giant
cases. Recurrence does occur in a significant proportion cell–containing fibrous lesions microcopically similar to
of cases, ranging from 33% to 50% that are treated by more common sporadic giant cell reparative granuloma
curettage and bone grafting.176,189 No recurrences have (Figs. 10-52 and 10-53).146,156,179,184,188,196,197 The onset
been reported after resection or ray amputation, and of cherubism is early, typical age 2 to 5 years, at
malignant transformation has not been observed. Pulmo- which time patients develop facial swelling and cervi-
nary implantation, as seen in conventional giant cell cal lymphadenopathy. The facial swelling is due to the
tumor, does not occur in giant cell reparative granuloma. development of bilateral giant cell reparative granulo-
Recurrence usually occurs within 15 months of curettage, mas of the maxilla and mandible (Fig. 10-52). Radio-
with a range of 3 months to 4 years. Most patients are graphically cherubism is characterized by symmetric jaw
adequately treated by second curettage and bone graft- involvement that is confined to the maxilla and mandi-
ing.179,189 Some giant cell granulomas show a progressive ble. Imaging data typically shows expansile involvement
locally destructive growth pattern and may have a high of the maxillary and mandibular bones, with thinning
propensity for recurrence.160 No radiologic or histologic of the cortex and multilocular radiolucencies with
features of giant cell reparative granuloma predict its coarse trabeculations.125 The lesions and facial swelling
local aggressive behavior, response to therapy, or ten- substantially regress after puberty but bone deformi-
dency to recur. ties and osseous defects can be detected in old age.172,206
Some cases may show a persistent destructive growth
pattern after puberty associated with major facial dis-
Personal Comments
figurement.208 The mutations encoding c-Abl-binding
Although originally giant cell reparative granuloma was protein SH3BP2 mapped to the 4-16.3 region cause
considered a reactive condition, current studies indicate cherubism (Fig. 10-52).134,135,139,141,149,157,159,168,169,175,182,203
that many of them are associated with unique mutations The gene is a critical regulator of osteoclast function
that are clonal and may represent driver alterations and its mutations hyperactivate osteoclastogenesis,
playing an important role in the development of some of which is responsible for the excessive bone remodeling
these lesions. Giant cell reparative granulomas associated in cherubism.171
with hyperparathyroidism are still best understood if Multiple giant cell reparative granulomas of the
considered as reactive because they regress when the facial bones can also be associated with Noonan’s syn-
underlying metabolic disorder is corrected or cured. As drome, a dysmorphic condition characterized by hyper-
a reactive process the lesion develops in relation to telorism, short stature, congenital heart defects, short
intraosseous hemorrhage, either in normal bone or within and webbed neck, skeletal anomalies, and bleeding dia-
certain preexisting conditions such as fibrous dysplasia or thesis.132,167,170,173,185 Germline mutations that affect com-
hyperparathyroid bone disease. At times and in certain ponents of the RAS-MAPK (mitogen-activated protein
specific locations, it can be difficult to distinguish such kinase) pathway are implicated in the pathogenesis
lesions from true giant cell tumors. It is evident that of Noonan’s syndrome, with the missense mutations
giant cell reparative granulomas, especially of the small of PTPN11 (protein tyrosine phosphatase, nonrecep-
bones of the hand and feet, are clearly related to the tor type 11), a gene mapped to chromosome 12q24.1,
so-called solid aneurysmal bone cyst and they carry the appearing to be most frequent.127,147,164,186,191 Noonan’s

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 753

C
FIGURE 10-52  ■  Cherubism: clinical and radiographic features. A, Individual with cherubism at age 6 years. The facial swelling in this
severe case affects the upper and lower jaws. The right side is somewhat more prominent than the left. B, Computed tomography
(CT) three dimensional reconstruction image shows extensive bone destruction and replacement by soft tissue masses. Insert,
Coronal CT reveals marked expansion of the mandible, which is filled with soft tissue density mass and delineated by thinned,
focally disrupted cortex. C, Panoramic radiograph of the jaw shows multilocular osteolytic lesions, bilaterally in the mandible, with
centrally dislocated teeth.

ERRNVPHGLFRVRUJ
754 10  Giant Cell Lesions

16.1 kb

9 10 11 12
1 2 3 45 67 8 13

561 aa
1 25 133 201 210 453 555
PH Domain SH3 Binding SH2 Domain
Domain

415 418 420


H L QR S P P DGQ S F
D
C
FIGURE 10-53  ■  Cherubism: microscopic features and molecular mechanism. A, Microscopic features of giant cell reparative granu-
loma from a patient shown in Figure 10-50. Note irregular distribution of multinucleated giant cells in fibrous stroma. B, Higher
magnification of A showing a cluster of giant cells and mononuclear histiocytic cells. C, Higher magnification of B showing a pro-
liferation of mononuclear histiocytic cells. D, Gene structure and functional domains of SH3BP2. The gene consists of 13 exons
extending over 16.1 kilobases. The 561-amino acid (aa) protein consists of three modular domains (pleckstrin homology [PH] domain,
SH3-binding domain, and SH2 domain). All mutations found are located within a 6-aa sequence 33-38 aa upstream of the SH2
domain. (A, ×25; B, ×50; C, ×100) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 755

syndrome has a somewhat heterogeneous genetic back- 24. Dominkus M, Ruggieri P, Bertoni F, et al: Histologically verified
ground, and multifocal giant cell reparative granulomas lung metastases in benign giant cell tumours—14 cases from a
single institution. Int Orthop 30:499–504, 2006.
involving both bone and soft tissue have been reported 25. Donthineni R, Boriani L, Ofluoglu O, et al: Metastatic behaviour
in association with a wide range of mutations involv- of giant cell tumour of the spine. Int Orthop 33:497–501, 2009.
ing the SOS1, RAF1, NRAS, SHOC2, CBL, and NF1 26. Doussis IA, Puddle B, Athanasou NA: Immunophenotype of mul-
genes.186,191,193 tinucleated and mononuclear cells in giant cell lesions of bone and
soft tissue. J Clin Pathol 45:398–404, 1992.
27. Dufresne A, Derbel O, Cassier P, et al: Giant-cell tumor of bone,
REFERENCES anti-RANKL therapy. Bonekey Rep 1:149, 2012.
28. Dumford K, Moore TE, Walker CW, et al: Multifocal, metachro-
Conventional Giant-Cell Tumor of Bone nous, giant cell tumor of the lower limb. Skeletal Radiol 32(3):147–
1. Abelanet R: Virus-like filamentous intranuclear inclusions in a 150, 2003.
giant-cell tumor, not associated with Paget’s disease: a case report. 29. Enneking WF: Staging benign lesions. In Enneking WF, editor:
J Bone Joint Surg 68A:952, 1986. (letter). Musculoskeletal tumor surgery, vol 1, New York, 1983, Churchill
2. Amanatullah DF, Clark TR, Lopez MJ, et al: Giant cell tumor of Livingstone.
bone. Orthopedics 37:112–120, 2014. 30. Exarchou E, Maris J, Assimakopoulos A: Soft tissue recurrence of
3. Atkins GJ, Haynes DR, Graves SE, et al: Expression of osteoclast osteoclastoma. J Bone Joint Surg Br 71:432–433, 1989.
differentiation signals by stromal elements of giant cell tumors. 31. Fellenberg J, Sahr H, Liu L, et al: Rescue of silenced UCHL1
J Bone Miner Res 15:640–649, 2000. and IGFBP4 expression suppresses clonogenicity of giant cell
4. Atkins GJ, Kostakis P, Vincent C, et al: RANK expression as a cell tumor-derived stromal cells. Cancer Lett 336:61–67, 2013.
surface marker of human osteoclast precursors in peripheral 32. Fornasier VL, Flores L, Hastings D, et al: Virus-like filamentous
blood, bone marrow, and giant cell tumors of bone. J Bone Miner intranuclear inclusions in a giant-cell tumor, not associated with
Res 21:1339–1349, 2006. Paget’s disease of bone: a case report. J Bone Joint Surg 67A:333–
5. Balke M, Schremper L, Gebert C, et al: Giant cell tumor of bone: 336, 1985.
treatment and outcome of 214 cases. J Cancer Res Clin Oncol 33. Gamage NM, Kashima TG, McNally MA, et al: Giant-cell-rich
134:969–978, 2008. pseudotumour in Paget’s disease. Skeletal Radiol 42:595–599,
6. Bauer FC, Urist MR: The reaction of giant cell tumors of bone 2013.
to transplantation into athymic nude mice: including observations 34. Ghostine B, Sebaaly A, Ghanem I: Multifocal metachronous giant
on osteoinduction in the host bed. Clin Orthop 159:257–264, 1981. cell tumor: case report and review of the literature. Case Rep Med
7. Behjati S, Tarpey PS, Presneau N, et al: Distinct H3F3A and 2014 [epub ahead of print].
H3F3B driver mutations define chondroblastoma and giant cell 35. Gianfrancesco F, Rendina D, Merlotti D, et al: Giant cell tumor
tumor of bone. Nat Genet 45:1479–1482, 2013. occurring in familial Paget’s disease of bone: report of clinical
8. Bertoni F, Bacchini P, Staals EL: Malignancy in giant cell tumor. characteristics and linkage analysis of a large pedigree. J Bone
Skeletal Radiol 32:143–146, 2003. Miner Res 28:341–350, 2013.
9. Bertoni F, Bacchini P, Staals EL: Malignancy in giant cell tumor 36. Gitelis S, Wang JW, Quast M, et al: Recurrence of a giant-cell
of bone. Cancer 97:2520–2529, 2003. tumor with malignant transformation to a fibrosarcoma 25 years
10. Bertoni F, Present D, Enneking WF: Giant-cell tumor of bone after primary treatment: a case report. J Bone Joint Surg 71A:757–
with pulmonary metastases. J Bone Joint Surg 67A:890–900, 1985. 761, 1989.
11. Bertoni F, Present D, Sudanese A, et al: Giant-cell tumor of bone 37. Goldenberg RR, Campbell CJ, Bonfiglio M: Giant cell tumor of
with pulmonary metastases: six case reports and a review of the bone: an analysis of 218 cases. J Bone Joint Surg 52A:619–664,
literature. Clin Orthop 237:275–285, 1988. 1970.
12. Bridge JA, Neff JR, Bhatia PS, et al: Cytogenetic findings and 38. Goldring SR, Dayer JM, Rosenblatt M: Factors regulating the
biologic behavior of giant cell tumors of bone. Cancer 65:2697– response of cells cultured from human giant cell tumors of bone
2703, 1990. to parathyroid hormone. J Clin Endocrinol Metab 53:295–300,
13. Burmester GR, Winchester RJ, Dimitriu-Bona A, et al: Delinea- 1981.
tion of four cell types comprising the giant cell tumor of bone: 39. Goldring SR, Schiller AL, Mankin HJ, et al: Characterization of
expression of Ia and monocyte-macrophage lineage antigens. cells from human giant cell tumors of bone. Clin Orthop 204:59–
J Clin Invest 71:1633–1648, 1983. 75, 1986.
14. Campanacci M: Giant cell tumor. In Campanacci M, editor: Bone 40. Grote HJ, Braun M, Kalinski T, et al: Spontaneous malignant
and soft tissue tumors, Vienna, 1991, Springer-Verlag. transformation of conventional giant cell tumor. Skeletal Radiol
15. Campanacci M, Baldini N, Boriani S, et al: Giant-cell tumor of 33:169–175, 2004.
bone. J Bone Joint Surg 69A:106–114, 1987. 41. Haibach H, Farrell C, Dittrich FJ: Neoplasms arising in Paget’s
16. Carles D, Rivel J, Devars F, et al: Giant cell tumors developing in disease of bone: a study of 82 cases. Am J Clin Pathol 83:594–600,
Paget’s disease: presentation of 2 cases with an ultrastructural 1985.
study. Ann Pathol 9:47–53, 1989. 42. Hakozaki M, Tajino T, Yamada H, et al: Radiological and patho-
17. Chakarun CJ, Forrester DM, Gottsegen CJ, et al: Giant cell logical characteristics of giant cell tumor of bone treated with
tumor of bone: review, mimics, and new developments in treat- denosumab. Diagn Pathol 9:111, 2014.
ments. Radiographics 33:197–211, 2013. 43. Hoch B, Hermann G, Klein MJ, et al: Giant cell tumor complicat-
18. Chen S, Li C, Wu B, et al: Identification of differentially expressed ing Paget disease of long bone. Skeletal Radiol 36:973–978,
genes and their subpathways in recurrent versus primary bone 2007.
giant cell tumors. Int J Oncol 45(3):1133–1142, 2014. 44. Hoch B, Inwards C, Sundaram M, et al: Multicentric giant cell
19. Chuang VP, Soo CS, Wallace S, et al: Arterial occlusion: manage- tumor of bone. Clinicopathologic analysis of thirty cases. J Bone
ment of giant cell tumor and aneurysmal bone cyst. AJR Am J Joint Surg Am 88:1998–2008, 2006.
Roentgenol 136:1127–1130, 1981. 45. Huang L, Xu J, Wood DJ, et al: Gene expression of osteoprote-
20. Cowan RW, Singh G, Ghert M: PTHrP increases RANKL gerin ligand, osteoprotegerin, and receptor activator of NF-kappaB
expression by stromal cells from giant cell tumor of bone. J Orthop in giant cell tumor of bone: possible involvement in tumor cell-
Res 30:877–884, 2012. induced osteoclast-like cell formation. Am J Pathol 156:761–767,
21. Dahlin DC: Caldwell lecture: giant cell tumor of bone—highlights 2000.
of 407 cases. AJR Am J Roentgenol 144:955–960, 1985. 46. Jacobs TP, Michelsen J, Polay JS, et al: Giant cell tumor in Paget’s
22. De Chiara A, Apice G, Fazioli F, et al: Multicentric giant cell disease of bone: familial and geographic clustering. Cancer 44:742–
tumor with viral-like inclusions associated Paget’s disease of bone: 747, 1979.
a case treated by steroid therapy. Oncol Rep 5:317–320, 1998. 47. Jaffe HL, Lichtenstein L, Portis RB: Giant cell tumor of bone: its
23. Dhillon MS, Prasad P: Multicentric giant cell tumour of bone. pathologic appearance, grading, supposed variants and treatment.
Acta Orthop Belg 73:289–299, 2007. Arch Pathol 30:993–1031, 1940.

ERRNVPHGLFRVRUJ
756 10  Giant Cell Lesions

48. Junming M, Cheng Y, Dong C, et al: Giant cell tumor of the 72. Mori Y, Tsuchiya H, Karita M, et al: Malignant transformation of
cervical spine: a series of 22 cases and outcomes. Spine 33:280–288, a giant cell tumor 25 years after initial treatment. Clin Orthop Relat
2008. Res 381:185–191, 2000.
49. Karakida K, Ota Y, Aoki T, et al: Multiple giant cell tumors in 73. Ng PK, Tsui SK, Lau CP, et al: CCAAT/enhancer binding protein
maximlla and skull complicating Paget’s disease of bone. Takai J beta is up-regulated in giant cell tumor of bone and regulates
Exp Clin Med 35:112–117, 2010. RANKL expression. J Cell Biochem 110:438–446, 2010.
50. Kato Kaneko M, Liu X, Oki H, et al: Isocitrate dehydrogenase 74. Ng VY, Davidson DJ, Kim EY, et al: The multidisciplinary man-
mutation is frequently observed in giant cell tumor of bone. Cancer agement of giant cell tumor of bone. Expert Rev Anticancer Ther
Sci 105:744–748, 2014. 14:783–790, 2014.
51. Katz E, Nyska M, Okon E, et al: Growth rate analysis of lung 75. Nuovo MA, Nuovo GJ, MacConnell P, et al: In situ analysis of
metastases from histologically benign giant cell tumor of bone. Paget’s disease of bone for measles-specific PCR-amplified cDNA.
Cancer 59:1831–1836, 1987. Diagn Mol Pathol 1:256–265, 1992.
52. Komiya S, Sasaguri Y, Inoue A, et al: Characterization of cells 76. Oda Y, Sakamoto A, Saito T, et al: Secondary malignant giant-cell
cultured from human giant-cell tumors of bone: phenotypic rela- tumour of bone: molecular abnormalities of p53 and H-ras gene
tionship to the monocyte-macrophage and osteoclast. Clin Orthop correlated with malignant transformation. Histopathology 39:629–
258:304–309, 1990. 637, 2001.
53. Ladanyi M, Traganos F, Huvos AG: Benign metastasizing giant 77. Okubo T, Saito T, Mitomi H, et al: p53 mutations may be involved
cell tumors of bone: a DNA flow cytometric study. Cancer 64: in malignant transformation of giant cell tumor of bone through
1521–1526, 1989. interaction with GPX1. Virchows Arch 463:67–77, 2013.
54. Lau CP, Huang L, Tsui SK, et al: Pamidronate, farnesyl transfer- 78. Pathak HJ, Nardi PM, Thornhill B: Multiple giant cell tumors
ase, and geranylgeranyl transferase-I inhibitors affects cell prolif- complicating Paget’s disease. AJR Am J Roentgenol 172:1696–1697,
eration, apoptosis, and OPG/RANKL mRNA expression in 1999.
stromal cells of giant cell tumor of bone. J Orthop Res 29:403–413, 79. Peimer CA, Schiller AL, Mankin HJ, et al: Multicentric giant-cell
2011. tumor of bone. J Bone Joint Surg 62A:652–656, 1980.
55. Lawless ME, Jour G, Hoch BL, et al: Beta-human chorionic 80. Picci P, Manfrini M, Zucchi V, et al: Giant-cell tumor of bone in
gonadotropin expression in recurrent and metastatic giant cell skeletally immature patients. J Bone Joint Surg 65A:486–490,
tumors of bone: a potential mimicker of germ cell tumor. Int J 1983.
Surg Pathol 22(7):617–622, 2014. 81. Potter HG, Schneider R, Ghelman B, et al: Multiple giant cell
56. Lee CH, Espinosa I, Jensen KC, et al: Gene expression profiling tumors and Paget disease of bone: radiographic and clinical cor-
identifies p63 as a diagnostic marker for giant cell tumor of the relations. Radiology 180:261–264, 1991.
bone. Mod Pathol 21:531–539, 2008. 82. Qureshi SS, Puri A, Agarwal M, et al: Recurrent giant cell tumor
57. Ling L, Klein MJ, Sissons HA, et al: Expression of Ia and of bone with simultaneous regional lymph node and pulmonary
monocyte-macrophage lineage antigens in giant cell tumor of metastases. Skeletal Radiol 34:225–228, 2005.
bone and related lesions. Arch Pathol Lab Med 112:65–69, 1988. 83. Rao UN, Goodman M, Chung WW, et al: Molecular analysis of
58. List AF: Metastatic giant-cell bone tumor in a man positive for primary and recurrent giant cell tumors of bone. Cancer Genet
HIV. N Engl J Med 318:517, 1988. (letter). Cytogenet 158:126–136, 2005.
59. Machinami R, Nishida K, Ishida T, et al: Carcinosarcomatous 84. Rendina D, Gennari L, De Filippo G, et al: Evidence for increased
malignancy, osteosarcoma and squamous cell carcinoma, in giant clinical severity of familial and sporadic Paget’s disease of bone in
cell tumor of the right distal femur. Pathol Res Pract 204:583–588, Campania, southern Italy. J Bone Miner Res 21:1828–1835, 2006.
2008. 85. Rendina D, Mossetti G, Soscia E, et al: Giant cell tumor and
60. Maggiani F, Forsyth R, Hogendoorn PC, et al: The immunophe- Paget’s disease of bone in one family: geographic clustering. Clin
notype of osteoclasts and macrophage polykaryons. J Clin Pathol Orthop Relat Res 421:218–224, 2004.
64:701–705, 2011. 86. Rock MG, Pritchard DJ, Unni KK: Metastases from histologically
61. Magitsky S, Lipton JF, Reidy J, et al: Ultrastructural features of benign giant-cell tumor of bone. J Bone Joint Surg 66A:269–274,
giant cell tumors in Paget’s disease. Clin Orthop Relat Res 402:213– 1984.
219, 2002. 87. Rock MG, Sim FH, Unni KK, et al: Secondary malignant giant-
62. Makis W, Alabed YZ, Nahal A, et al: Giant cell tumor pulmonary cell tumor of bone: clinicopathological assessment of nineteen
metastases mimic primary malignant pulmonary nodules on patients. J Bone Joint Surg 68A:1073–1079, 1986.
18
F-FDG PET/CT. Nucl Med Mol Imaging 46:134–137, 2012. 88. Ropars M, Siret P, Kaila R, et al: Recurrent primary giant cell
63. Maloney WJ, Vaughan LM, Jones HH, et al: Benign metastasiz- tumour of the proximal radius with pulmonary metastases. Joint
ing giant-cell tumor of bone: report of three cases and review of Bone Spine 75:740–741, 2008.
the literature. Clin Orthop 243:208–215, 1989. 89. Rousseau MA, Handra-Luca A, Lazennec JY, et al: Metachronous
64. McCarthy EF: Giant-cell tumor of bone: an historical perspective. multicentric giant-cell tumor of the bone in the lower limb. Case
Clin Orthop 153:14–25, 1980. report and Ki-67 immunohistochemistry study. Virchows Arch
65. McDonald DJ, Sim FH, McLeod RA, et al: Giant-cell tumor of 445:79–82, 2004.
bone. J Bone Joint Surg 68A:235–242, 1986. 90. Roy S, Joshi NP, Sigamani E, et al: Clival giant cell tumor
66. McGrath PJ: Giant cell tumour of bone: an analysis of 52 cases. presenting with isolated trigeminal nerve involvement. Eur Arch
J Bone Joint Surg Br 54:216–229, 1972. Otorhinolaryngol 270(3):1167–1171, 2013.
67. Merad M, Mesurolle B, Guinebretière JM, et al: Recurrence of a 91. Saada E, Peoc’h M, Decouvelaere AV, et al: CCND1 and Met
primary malignant giant cell tumour of bone 14 years after initial genomic amplification during malignant transformation of a giant
surgery. Eur Radiol 11:1483–1486, 2001. cell tumor of bone. J Clin Oncol 29:86–89, 2011.
68. Mirra JM, Bauer FC, Grant TT: Giant cell tumor with viral-like 92. Saito T, Mitomi H, Suehara Y, et al: A case of de novo secondary
intranuclear inclusions associated with Paget’s disease. Clin Orthop malignant giant-cell tumor of bone with loss of heterozygosity of
158:243–251, 1981. p53 gene that transformed within a short-term follow-up. Pathol
69. Mirra JM, Ulich T, Magidson J, et al: A case of probable benign Res Pract 207:664–669, 2011.
pulmonary “metastases” or implants arising from a giant cell 93. Sanerkin NG: Malignancy, aggressiveness, and recurrence in giant
tumor of bone. Clin Orthop 162:245–254, 1982. cell tumor of bone. Cancer 46:1641–1649, 1980.
70. Miyamoto N, Higuchi Y, Tajima M, et al: Spindle-shaped cells 94. Sanjay BK, Sim FH, Unni KK, et al: Giant-cell tumours of the
derived from giant-cell tumor of bone support differentiation of spine. J Bone Joint Surg Br 75:148–154, 1993.
blood monocytes to osteoclast-like cells. J Orthop Res 18:647–654, 95. Sara AS, Ayala AG, el-Naggar A, et al: Giant cell tumor of bone:
2000. a clinicopathologic and DNA flow cytometric analysis. Cancer
71. Mooney WW, Bridger GP, Baldwin M, et al: Recurrent giant 66:2186–2190, 1990.
cell tumour of the maxilla associated with both Paget’s disease 96. Savini R, Gherlinzoni F, Morandi M, et al: Surgical treatment of
and primary hyperparathyroidism. ANZ J Surg 73:863–864, giant-cell tumor of the spine: the experience at the Istituto Orto-
2003. pedico Rizzoli. J Bone Joint Surg 65A:1283–1289, 1983.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 757

97. Schajowicz F, Cabrini RL, Gimenez I: Microspectrophotometric findings in giant cell granulomas of the jaws. J Dent Res Dent Clin
quantitation of DNA in bone tumors with giant cells (osteoclas- Dent Prospects 7:225–229, 2013.
toma, osteosarcoma and chondroblastoma). Clin Orthop 156:91– 123. Amaral FR, Diniz MG, Bernardes VF, et al: WWOX expression
97, 1981. in giant cell lesions of the jaw. Oral Surg Oral Med Oral Pathol
98. Schajowicz F, Ubios AM, Araujo ES, et al: Virus-like intranuclear Oral Radiol 116:210–213, 2013.
inclusions in giant cell tumor of bone. Clin Orthop 201:247–250, 124. Auclair PL, Cuenin P, Kratochvil FJ, et al: A clinical and histo-
1985. morphologic comparison of the central giant cell granuloma and
99. Schwartz HS, Jenkins RB, Dahl RJ, et al: Cytogenetic analyses on the giant cell tumor. Oral Surg Oral Med Oral Pathol 66:197–208,
giant-cell tumors of bone. Clin Orthop 240:250–260, 1989. 1988.
100. Schwimer SR, Bassett LW, Mancuso AA, et al: Giant cell tumor 125. Beaman FD, Bancroft LW, Peterson JJ, et al: Imaging character-
of the cervicothoracic spine. AJR Am J Roentgenol 136:63–67, istics of cherubism. AJR Am J Roentgenol 182:1051–1054, 2004.
1981. 126. Bedard CH, Nichols RD: Osteitis fibrosa (brown tumor) of the
101. Shekhar A, Murgod G, Korlhalli S: Synchronous multicentric maxilla. Laryngoscope 84:2093–2100, 1974.
giant cell tumour (GCT)—a rare case report. J Clin Diagn Res 127. Beneteau C, Cavé H, Moncla A, et al: SOS1 and PTPN11 muta-
8:185–186, 2014. tions in five cases of Noonan syndrome with multiple giant cell
102. Sim FH, Dahlin DC, Beabout JW: Multicentric giant-cell tumor lesions. Eur J Hum Genet 2009 [epub ahead of print].
of bone. J Bone Joint Surg 59(A):1052–1060, 1977. 128. Bertheussen KJ, Holck S, Schiodt T: Giant cell lesion of bone of
103. Skubitz KM: Giant cell tumor of bone: current treatment options. the hand with particular emphasis on giant cell reparative granu-
Curr Treat Options Oncol 15(3):507–518, 2014. loma. J Hand Surg 8A:46–49, 1983.
104. Stratil PG, Stacy GS: Multifocal metachronous giant cell tumor 129. Body JJ, Jortay AM, de Jager R, et al: Treatment with steroids
in a 15-year-old boy. Pediatr Radiol 35:444–448, 2005. of a giant cell granuloma of the maxilla. J Surg Oncol 16:7–13,
105. Steiner GC, Ghosh L, Dorfman HD: Ultrastructure of giant cell 1981.
tumours of bone. Hum Pathol 3:569–586, 1972. 130. Borges BB, Fornazieri MA, Bezerra AP, et al: Giant cell bone
106. Sung HW, Kuo DP, Shu WP, et al: Giant-cell tumor of bone: lesions in the craniofacial region: a diagnostic and therapeutic
analysis of 208 cases in Chinese patients. J Bone Joint Surg challenge. Int Forum Allergy Rhinol 2:501–506, 2012.
64A:755–761, 1982. 131. Breitling G, Holthusen W, Vogler E, et al: Die riesenzellige reak-
107. Tanaka H, Yasui N, Kuriskaki E, et al: The Goltz syndrome asso- tion des os metatarsale. Fortschr Röntgenstr 133:101–102, 1980.
ciated with giant cell tumour of bone: a case report. Int Orthop 132. Cancino CM, Gaião L, Sant’Ana Filho M, et al: Giant cell lesions
14:179–181, 1990. with a Noonan-like phenotype: a case report. J Contemp Dent Pract
108. Taraz-Jamshidi MH, Gharadaghi M, Mazloumi SM, et al: Clinical 8:67–73, 2007.
outcome of en-block resection and reconstruction with nonvascu- 133. Carinci F, Piattelli A, Martinelli M, et al: Genetic profiling of
larized fibular autograft for the treatment of giant cell tumor of central giant cell granuloma of the jaws. J Craniofac Surg 16:399–
distal radius. J Res Med Sci 19:117–121, 2014. 407, 2005.
109. Thomas DM: RANKL, denosumab, and giant cell tumor of bone. 134. Carvalho VM, Perdigão PF, Amaral FR, et al: Novel mutations in
Curr Opin Oncol 24:397–403, 2012. the SH3BP2 gene associated with sporadic central giant cell
110. Tunn PU, Schlag PM: Giant cell tumor of bone. An evaluation of lesions and cherubism. Oral Dis 15:106–110, 2009.
87 patients]. Z Orthop Ihre Grenzgeb 141:690–698, 2003. 135. Carvalho VM, Perdigão PF, Pimenta FJ, et al: A novel mutation
111. Tyler W, Barrett T, Frassica F, et al: Skin metastasis from conven- of the SH3BP2 gene in an aggressive case of cherubism. Oral Oncol
tional giant cell tumor of bone: conceptual significance. Skeletal 44:153–155, 2008.
Radiol 31:166–170, 2002. 136. Caskey PM, Wolf MD, Fechner RE: Multicentric giant cell repar-
112. Upchurch KS, Simon LS, Schiller AL, et al: Giant cell reparative ative granuloma of the small bones of the hand: case report and
granuloma of Paget’s disease of bone: a unique clinical entity. Ann review of the literature. Clin Orthop 193:199–205, 1985.
Intern Med 98:35–40, 1983. 137. Chuong R, Kaban LB, Kozakewich H, et al: Central giant cell
113. van der Heijden L, Dijkstra PD, van de Sande MA, et al: The lesions of the jaws: a clinicopathologic study. J Oral Maxillofacial
clinical approach toward giant cell tumor of bone. Oncologist Surg 44:708–713, 1986.
19:550–561, 2014. 138. D’Alonzo RT, Pitcock JA, Milford LW: Giant-cell reaction of
114. Vanel D, Contesso G, Rebibo G, et al: Benign giant-cell tumours bone: report of two cases. J Bone Joint Surg 54A:1267–1271, 1972.
of bone with pulmonary metastasis and favorable prognosis. 139. de Lange J, van den Akker HP, van den Berg H, et al: Limited
Skeletal Radiol 10:221–226, 1983. regression of central giant cell granuloma by interferon alpha after
115. Wold LE, Swee RG: Giant cell tumor of the small bones of the failed calcitonin therapy: a report of 2 cases. Int J Oral Maxillofac
hands and feet. Semin Diagn Pathol 1:173–184, 1984. Surg 35:865–869, 2006.
116. Wolfe JT, III, Scheithauer BW, Dahlin DC: Giant-cell tumor of 140. de Lange J, van den Akker HP, Veldhuijzen van Zanten GO, et al:
the sphenoid bone: review of 10 cases. J Neurosurg 59:322–327, Calcitonin therapy in central giant cell granuloma of the jaw: a
1983. randomized double-blind placebo-controlled study. Int J Oral
117. Wray CC, Macdonald AW, Richardson RA: Benign giant cell Maxillofac Surg 35:791–795, 2006.
tumour with metastases to bone and lung: one case studied over 141. de Lange J, van Maarle MC, van den Akker HP, et al: A new
20 years. J Bone Joint Surg Br 72:486–489, 1990. mutation in the SH3BP2 gene showing reduced penetrance in a
118. Zheng MH, Robbins P, Xu J, et al: The histogenesis of giant cell family affected with cherubism. Oral Surg Oral Med Oral Pathol
tumour of bone: a model of interaction between neoplastic cells Oral Radiol Endod 103:378–381, 2007.
and osteoclasts. Histol Histopathol 16:297–307, 2001. 142. De Smet AA, Travers H, Neff JR: Case report 207: giant cell
119. Zhou W, Yin H, Wang T, et al: MIR-126-5p regulates osteolysis reparative granuloma of the left femur arising in polyostotic
formation and stromal cells proliferation in giant cell tumor fibrous dysplasia. Skeletal Radiol 8:314–318, 1982.
through inhibition of PTHrP. Bone 66:267–276, 2014. 143. Desai P, Steiner GC: Ultrastructure of brown tumor of hyperpara-
thyroidism. Ultrastruct Pathol 14:505–511, 1990.
Giant-Cell Reparative Granuloma 144. Ehrlich GW, Genant HK, Kolb FO: Secondary hyperparathy-
120. Ackerman LV, Spjut HJ: Giant cell reaction in tumors of bone and roidism and brown tumors in a patient with gluten enteropathy.
cartilage. In Spjut HJ, Dorfman HD, Fechner RE, et al, editors: AJR Am J Roentgenol 141:381–383, 1983.
Atlas for tumor pathology, Washington, DC, 1962, Armed Forces 145. Fechner RE: Problematic lesions of the craniofacial bones. Am J
Institute of Pathology. section II, fascicle 4, 1st series. Surg Pathol 13:17–30, 1989.
121. Agaram NP, LeLoarer FV, Zhang L, et al: USP6 gene rearrange- 146. Font RL, Blanco G, Soparkar CN, et al: Giant cell reparative
ments occur preferentially in giant cell reparative granulomas granuloma of the orbit associated with cherubism. Ophthalmology
of the hands and feet but not in gnathic location. Hum Pathol 110:1846–1849, 2003.
45:1147–1152, 2014. 147. Gelb BD, Tartaglia M: Noonan syndrome and related disorders:
122. Aghbali A, Sina M, Vahid Pakdel SM, et al: Correlation of histo- dysregulated RAS-mitogen activated protein kinase signal trans-
pathologic features with demographic, gross and radiographic duction. Hum Mol Genet 15(Spec2):R220–R226, 2006.

ERRNVPHGLFRVRUJ
758 10  Giant Cell Lesions

148. Glass TA, Mills SE, Fechner RE, et al: Giant cell reparative gran- 174. Liu B, Yu SF, Li TJ: Multinucleated giant cells in various forms
uloma of the hands and feet. Radiology 149:65–68, 1983. of giant cell containing lesions of the jaws express features of
149. Gleason BC, Kleinman PK, Debelenko LV, et al: Novel karyo- osteoclasts. J Oral Pathol Med 32:367–375, 2003.
types in giant cell-rich lesions of bone. Am J Surg Pathol 31:926– 175. Lo B, Faiyaz-Ul-Haque M, Kennedy S, et al: Novel mutation in
932, 2007. the gene encoding c-Abl-binding protein SH3BP2 causes
150. Gouin F, Grimaud E, Redini F, et al: Metatarsal giant cell tumors cherubism. Am J Med Genet 121A:37–40, 2003.
and giant cell reparative granuloma are similar entities. Clin 176. Lorenzo JC, Dorfman HD: Giant-cell reparative granuloma of
Orthop Relat Res 416:278–284, 2003. the short tubular bones of the hands and feet. Am J Surg Pathol
151. Güngörmüs M, Akgül HM: Central giant cell granuloma of the 4:551–563, 1980.
jaws: a clinical and radiologic study. J Contemp Dent Pract 4:87–97, 177. MacDonald DF, Binhammer PA, Rubenstein JD, et al: Giant cell
2003. reparative granuloma of the hand: case report and review of giant
152. Hermann G, Abdelwahab IF, Klein MJ, et al: Case report 603: cell lesions of hands and feet. Can J Surg 46:471–473, 2003.
giant cell reparative granuloma of the distal end of the right femur. 178. Manor E, Kachko L, Brennan PA, et al: Cytogenetics of central
Skeletal Radiol 19:367–369, 1990. giant cell granuloma of the mandible. J Oral Maxillofac Surg
153. Hirschl S, Katz A: Giant cell reparative granuloma outside the jaw 71:1541–1544, 2013.
bone. Human Pathol 5:171–181, 1974. 179. Meng XM, Yu SF, Yu GY: Clinicopathologic study of 24 cases of
154. Hoopes PC, Anderson RL, Blodi FC: Giant cell (reparative) cherubism. Int J Oral Maxillofac Surg 34:350–356, 2005.
granuloma of the orbit. Ophthalmology 88:1361–1366, 1981. 180. Menge M, Maier W, Feuerhake F, et al: Giant cell reparative
155. Hori T, Kanamori M, Ohmori K, et al: Giant cell reparative granuloma of the temporal bone. Acta Neurochir (Wien) 151:397–
granuloma of the proximal tibia: a case report. J Orthop Surg (Hong 399, 2009.
Kong) 16:364–367, 2008. 181. Merkow RL, Bansal M, Inglis AE: Giant cell reparative granuloma
156. Hyckel P, Berndt A, Schleier P, et al: Cherubism—new hypotheses in the hand: report of three cases and review of the literature.
on pathogenesis and therapeutic consequences. J Craniomaxillofac J Hand Surg 10:733–739, 1985.
Surg 33:61–68, 2005. 182. Miah SM, Hatani T, Qu X, et al: Point mutations of 3BP2 identi-
157. Idowu BD, Thomas G, Frow R, et al: Mutations in SH3BP2, the fied in human-inherited disease cherubism result in the loss of
cherubism gene, were not detected in central or peripheral giant function. Genes Cell 9:993–1004, 2004.
cell tumours of the jaw. Br J Oral Maxillofac Surg 46:229–230, 183. Morris JM, Lane JI, Witte RJ, et al: Giant cell reparative granu-
2008. loma of the nasal cavity. AJNR Am J Neuroradiol 25:1263–1265,
158. Ilaslan H, Sundaram M, Unni KK: Solid variant of aneurysmal 2004.
bone cysts in long tubular bones: giant cell reparative granuloma. 184. Mortellaro C, Bello L, Lucchina AG, et al: Diagnosis and treat-
AJR Am J Roentgenol 180:1681–1687, 2003. ment of familial cherubism characterized by early onset and rapid
159. Imai Y, Kanno K, Moriya T, et al: A missense mutation in the development. J Craniofac Surg 20:116–120, 2009.
SH3BP2 gene on chromosome 4p16.3 found in a case of nonfa- 185. Neumann TE, Allanson J, Kavamura I, et al: Multiple giant cell
milial cherubism. Cleft Palate Craniofac J 40:632–638, 2003. lesions in patients with Noonan syndrome and cardio-facio-
160. Ishinaga H, Otsu K, Mouri G, et al: Aggressive giant cell repara- cutaneous syndrome. Eur J Hum Genet 17:420–425, 2009.
tive granuloma of the nasal cavity. Case Rep Otolaryngol 690194, 186. Niihori T, Aoki Y, Narumi Y, et al: Germline KRAS and BRAF
2013. mutations in cardio-facio-cutaneous syndrome. Nat Genet 38:294–
161. Itonaga I, Schulze E, Burge PD, et al: Phenotypic characterization 296, 2006.
of mononuclear and multinucleated cells of giant cell reparative 187. Picci P, Baldini N, Sudanese A, et al: Giant cell reparative granu-
granuloma of small bones. J Pathol 198:30–36, 2002. loma and other giant cell lesions of the bones of the hands and
162. Jaffe HL: Giant cell reparative granuloma, traumatic bone cyst feet. Skeletal Radiol 15:415–421, 1986.
and fibrous (fibro-osseous) dysplasia of the jaw bones. Oral Surg 188. Pontes FS, Ferreira AC, Kato AM, et al: Aggressive case of
6:159–175, 1953. cherubism: 17-year follow-up. Int J Pediatr Otorhinolaryngol
163. Jebasingh F, Jacob JJ, Shah A, et al: Bilateral maxillary brown 71:831–835, 2007.
tumours as the first presentation of primary hyperparathyroidism. 189. Ratner V, Dorfman HD: Giant-cell reparative granuloma of the
Oral Maxillofac Surg 12:97–100, 2008. hand and foot bones. Clin Orthop 260:251–258, 1990.
164. Jorge AA, Malaquias AC, Arnhold IJ, et al: Noonan syndrome 190. Rawashdeh MA, Bataineh AB, Al-Khateeb T: Long-term clinical
and related disorders: a review of clinical features and mutations and radiological outcomes of surgical management of central giant
in genes of the RAS/MAPK pathway. Horm Res 71:185–193, cell granuloma of the maxilla. Int J Oral Maxillofac Surg 35:60–66,
2009. 2006.
165. Khiavi MM, Aghbali AA, Halimi M, et al: Immunohistochemical 191. Roberts AE, Araki T, Swanson KD, et al: Germline gain-of-
expression of Src protein in peripheral and central giant cell function mutations in SOS1 cause Noonan syndrome. Nat
granulomas of the jaws. J Oral Maxillofac Pathol 17:358–362, Genet 39:70–74, 2007.
2013. 192. Rubio-Correa I, Manzano-Solo de Zaldivar D, Gonzalez-Garcia
166. Kryst L, Piekarczyk J: Giant-cell tumour-like changes in the R, et al: Giant cell granuloma of the maxilla. Global management,
maxilla. J Maxillofacial Surg 8:138–142, 1980. review of the literature and case report. J Clin Exp Dent 4:129–
167. Lee SM, Cooper JC: Noonan syndrome with giant cell lesions. 131, 2012.
Int J Paediatr Dent 15:140–145, 2005. 193. Ruggieri M, Pavone V, Polizzi A, et al: Unusual form of recurrent
168. Li CY, Yu SF: A novel mutation in the SH3BP2 gene causes giant cell granuloma of the mandible and lower extremities in a
cherubism: case report. BMC Med Genet 7:84, 2006. patient with neurofibromatosis type 1. Oral Surg Oral Med Oral
169. Lietman SA, Kalinchinko N, Deng X, et al: Identification of a Pathol Oral Radiol Endod 87:67–72, 1999.
novel mutation of SH3BP2 in cherubism and demonstration that 194. Sanerkin NG, Mott MG, Roylance J: An unusual intraosseous
SH3BP2 mutations lead to increased NFAT activation. Hum lesion with fibroblastic, osteoclastic, osteoblastic, aneurysmal and
Mutat 27:717–718, 2006. fibromyxoid elements: “solid” variant of aneurysmal bone cyst.
170. Lietman SA, Prescott NL, Hicks DG, et al: SH3BP2 is rarely Cancer 51:2278–2286, 1983.
mutated in exon 9 in giant cell lesions outside cherubism. Clin 195. Sapp JP: An ultrastructural study of nuclear and centriolar con-
Orthop Relat Res 459:22–27, 2007. figurations in multinucleated giant cells. Lab Invest 34:109–114,
171. Lietman SA, Yin L, Levine MA: SH3BP2 is an activator of NFAT 1976.
activity and osteoclastogenesis. Biochem Biophys Res Commun 196. Schultze-Mosgau S, Holbach LM, Wiltfang J: Cherubism: clinical
371:644–648, 2008. evidence and therapy. J Craniofac Surg 14:201–208, 2003.
172. Lima Gde M, Almeida JD, Cabral LA: Cherubism: clinicoradio- 197. Silva GC, Gomez RS, Vieira TC, et al: Cherubism: long-term
graphic features and treatment. J Oral Maxillofac Res 1:2, 2010. follow-up of 2 patients in whom it regressed without treatment.
173. Limal JM, Parfait B, Cabrol S, et al: Noonan syndrome: relation- Br J Oral Maxillofac Surg 45:567–570, 2007.
ships between genotype, growth, and growth factors. J Clin Endo- 198. Smith GA, Ward PH: Giant-cell lesions of the facial skeleton.
crinol Metab 91:300–306, 2006. Arch Otolaryngol 104:186–190, 1978.

ERRNVPHGLFRVRUJ
10  Giant Cell Lesions 759

199. Struthers PJ, Shear M: Aneurysmal bone cyst of the jaws. II. 206. Von Wowern N: Cherubism: a 36-year long-term follow-up of 2
Int J Oral Surg 13:92–100, 1984. generations in different families and review of the literature. Oral
200. Subasi M, Kapukaya A, Buyukbayram H, et al: Giant-cell Surg Oral Med Oral Pathol Oral Radiol Endod 90:765–772, 2000.
reparative granuloma of the tibia. Acta Orthop Belg 69:363–367, 207. Waldron CA, Shafer WG: The central giant cell reparative granu-
2003. loma of the jaws: an analysis of 38 cases. Am J Clin Pathol 45:437–
201. Tartaglia M, Zampino G, Gelb BD: Noonan syndrome: clini- 447, 1966.
cal aspects and molecular pathogenesis. Mol Syndromol 1:2–26, 208. Wang CN, Song YL, Peng B, et al: The aggressive form of
2010. cherubism: report of two cases in unrelated families. Br J Oral
202. Tillman BP, Dahlin DC, Lipscomb PR, et al: Aneurysmal bone Maxillofac Surg 44:322–324, 2006.
cyst: an analysis of 95 cases. Mayo Clinic Proc 43:478–495, 209. Wold LE, Dobyns JH, Swee RG, et al: Giant cell reaction (giant
1968. cell reparative granuloma) of the small bones of the hands and
203. Ueki Y, Tiziani V, Santanna C, et al: Mutations in the gene encod- feet. Am J Surg Pathol 10:491–496, 1986.
ing c-Abl-binding protein SH3BP2 cause cherubism. Nat Genet 210. Yamaguchi T, Dorfman HD: Giant cell reparative granuloma:
28:125–126, 2001. a comparative clinicopathologic study of lesions in gnathic and
204. Upchurch KS, Simon LA, Schiller AL, et al: Giant cell reparative extragnathic sites. Int J Surg Pathol 9:189–200, 2001.
granuloma of Paget’s disease of bone: a unique clinical entity. Ann 211. Yamaguchi T, Dorfman HD, Eisig S: Cherubism: clinicopatho-
Intern Med 98:35–40, 1983. logic features. Skeletal Radiol 28:350–353, 1999.
205. Vered M, Buchner A, Dayan D: Central giant cell granuloma of 212. Yoshida T, Sakamoto A, Tanaka K, et al: Alternative surgical treat-
the jawbones—new insights into molecular biology with clinical ment for giant-cell reparative granuloma in the metacarpal, using
implications on treatment approaches. Histol Histopathol 23:1151– phenol and ethanol adjuvant therapy. J Hand Surg [Am] 32:887–
1160, 2008. 892, 2007.

ERRNVPHGLFRVRUJ
C H A P T E R 1 1 

Ewing’s Sarcoma and


Related Tumors
CHAPTER OUTLINE

CHROMOSOMAL TRANSLOCATIONS ASKIN’S TUMOR


UNIFIED PATHOGENETIC CONCEPT OF EWING’S IMMUNOHISTOCHEMICAL FEATURES OF
SARCOMA AND RELATED TUMORS EWING’S SARCOMA AND PNET
EWING’S SARCOMA DIFFERENTIAL DIAGNOSIS OF SMALL ROUND-
CELL TUMORS OF BONE
PRIMITIVE NEUROECTODERMAL TUMOR

In the early 1920s, James Ewing identified a peculiar their specific chimeric genes to over 15 distinct transloca-
round-cell tumor that he called diffuse endothelioma of bone.1 tions, many of which have multiple variants (Fig. 11-1).
The tumor predominantly affected the long tubular bones This data has expanded our current understanding of
of young patients. This lesion was distinguished from the complex biology of these tumors and is the basis
the general category of unclassified round-cell sarcomas of an emerging novel classification of Ewing’s sarcoma
and lymphohematopoietic tumors based on its clinico- and related tumors. There is also hope that the evolv-
pathologic features (i.e., small, round-cell morphology), ing molecular data can guide us to more effective tar-
a predilection for the long tubular bones of skeletally geted therapies for these tumors. From a diagnostic point
immature patients, and the absence of a lymphoma-like of view, Ewing’s sarcoma and related tumors serve as a
or leukemia-like clinical picture.1,2 Although many of prototype for a modern diagnostic approach in which
the original pathogenetic theories proposed by Ewing clinical, radiographic, microscopic, and molecular data
and other early investigators have been significantly contribute to the diagnosis. The emergence of molecular
modified, Ewing’s sarcoma remains recognized as a dis- techniques as a significant aid in the diagnosis of this
tinct entity and represents a prototype of nonhemato- group of malignancies should be particularly emphasized.
logic round-cell malignancy.10 The delineation of this In this chapter conventional descriptions of Ewing’s
entity also serves as an example of the brilliant corre- sarcoma, PNET, and Askin’s tumors are retained, but the
lations of clinical, radiographic, and microscopic find- reader should understand that in view of modern molecu-
ings that set us on a quest to understand the biology of lar data, these tumors are considered as variants of the
this unique tumor, providing the foundation for its same entity. Moreover, recent genomic data has expanded
diagnosis and treatment. Several additional lesions with this group of tumors exponentially by the inclusion of
clinicopathologic features different from classical Ewing’s tumors deviating somewhat from the classical Ewing’s
sarcoma have subsequently been described in the soft sarcoma morphology; these are referred to as Ewing’s
tissue and specific anatomic regions; these are referred sarcoma-like and are characterized by chromosomal
to as Askin’s tumor and primitive neuroectodermal tumor translocations and gene fusions differing from those orig-
(PNET). Tumors with microscopic features similar to inally identified. The description of molecular features of
classical Ewing’s sarcoma or PNET have also been this group of tumors is focused on the nature of translo-
described in parenchymal and reproductive organs. The cations and their genes as potential biomarkers in clas-
consistent presence of a chromosomal abnormality— sification and differential diagnosis. The interested reader
the reciprocal translocation of chromosomes 11 and is referred to Chapter 3, where the biologic effects of
22, which involves bands q24 and q12 of the chromo- signature hybrid proteins in this group of tumors are
somes, respectively—is a unique finding in this group of described.
tumors.6,64,65 This finding is essential in understanding
these malignancies, collectively referred to as the Ewing’s
sarcoma/PNET family of tumors. During the past three CHROMOSOMAL TRANSLOCATIONS
decades, molecular studies have elucidated the structure
of the chromosomal breakpoint and have identified the Breaks in chromosomes may result in the abnormal
fusion partners of the chimeric gene. Subsequent inves- assembly of genetic material, resulting in the transfer of
tigations and recent genome-based sequencing studies one fragment of a chromosome to another. These, in
have expanded the list of chromosomal translocations and turn, can fuse together the coding sequences of genes
760
ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 761

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

19 20 21 22 X Y
A

Chr 11 Chr 22 der Chr 11 der Chr 22


15.5 15.4 13 15.5 15.4 13
15.3 15.2 12 15.3 15.2 11.2 12
15.1 11.2 15.1 11.1 11.1
11.1
14 11.1 11.2 14 chimeric 11.2
breakpoint EWSR1 EWSR1
13 13 gene FLI1
12 12.1 12.2 12
11.2 12.3 13.1 11.2 25
11.11 11.12 11.11 11.12
11 13.2 13.3 11
12 13.1 12 13.1
13.2 13.3 13.2 13.3
13.4 13.5 13.4 13.5
14.1 14.2 14.1 14.2
14.3 21 14.3 21
22.1 22.2 22.1 22.2
22.3 23.1 22.3 23.1
23.2 23.2
23.3 23.3
24
breakpoint FLI1
24 silent
25 breakpoint

11 11

22
22

breakpoint breakpoint silent Chimeric


breakpoint EWSR1/FLI1
gene

C
FIGURE 11-1  ■  Representative karyotype and diagram of reciprocal translocation t(11;22)(q24;q12) in Ewing’s sarcoma. A, Female
karyotype showing t(11;22) and t(7;16) translocations. B, Schematic diagram of the t(11;22)(q24;q12) translocation characteristic of
Ewing’s sarcoma. C, Magnified view of the t(11;22) translocation. This genetic abnormality can be detected in nearly 90% of Ewing’s
sarcomas/primitive neuroectodermal tumors. The translocation results in one active chimeric gene and one silent breakpoint where
an active chimeric gene is not formed. (Modified and reprinted with permission from Lazar A, et al: Arch Pathol Lab Med 130:1199-1207,
2006.)

ERRNVPHGLFRVRUJ
762 11  Ewing’s Sarcoma and Related Tumors

that, in normal genomes, function as separate indepen-


TABLE 11-1 Translocation Involving EWSR1 in
dent coding units.21 Such fusions create abnormal hybrid
Bone and Soft Tissue Sarcomas
mRNAs that contain coding sequences of both genes and
result in the expression of a new protein, which can have Tumor Type Cytogenetics Genes Involved
profound effects on cell biology, contributing to malig-
Angiomatoid fibrous t(2;22)(q34;q12) EWSR1-CREB1
nant transformation.32 histiocytoma t(12;22)(q13;q12) EWSR1-ATF1
The unifying feature of the group of tumors described Clear cell sarcoma t(12;22)(q13;q12) EWSR1-ATF1
in this chapter is the presence of distinct chromosomal t(2;22)(q34;q12) EWSR1-CREB1
translocations that are clonal and result in the expression Desmoplastic small t(11;22)(p13;q12) EWSR1-WT1
round-cell tumor
of new proteins that do not exist in normal cells. These Extraskeletal myxoid t(9;22)(q22;q12) EWSR1-NR4A3
translocations drive the biology of tumors, are diagnos­ chondrosarcoma
tically specific, and may represent future therapeutic Myoepithelioma, t(6;22)(p21;q12) EWSR1-POU5F1
targets. In fact, the Ewing’s sarcoma/PNET family of soft tissue t(19;22)(q13;q12) EWSR1-ZNF444
tumors carry a unique abnormality, t(11;22)(q24;q12), t(1;22)(q23;q12) EWSR1-PBX1
Myoepithelial tumor t(6;22)(p21;q12) EWSR1-POU5F1
resulting in the expression of a chimeric protein, EWSR1- of bone t(19;22)(q13;q12) EWSR1-ZNF444
FLI1, which represents a prototypic chromosomal abnor- t(1;22)(q23;q12) EWSR1-PBX1
mality in soft tissue and bone tumors. In addition to this Myxoid/round-cell t(12;22)(q13;q12) EWSR1-DDIT3
prototypic translocation found in the majority of Ewing’s liposarcoma
sarcoma family of tumors, over 15 distinct chromosomal
translocations and hybrid genes have been identified in
this group of tumors, providing new unifying concepts
for their diagnosis and treatment.39,52 tions in soft tissue and bone tumors. It is involved in
all the alternative translocations identified in Ewing’s
Prototypic Chromosomal Translocations sarcoma/PNET, as well as in distinct translocations in
desmoplastic small round-cell tumor, clear cell sarcoma
Involving EWSR1 and ETS Partners (melanoma of soft parts), some cases of extraskeletal
Multiple chromosomal translocations resulting in hybrid myxoid chondrosarcoma, angiomatoid fibrous histiocy-
genes identified in the Ewing’s family of tumors can be toma, myxoid liposarcoma, and myoepithelial tumors of
divided into two main groups: those containing hybrid soft tissue and bone (Table 11-1).22,34,41,63,68 The EWSR1
genes with fusion partners that belong to the E-26 trans- gene has 17 exons and spans approximately 40 kb.45 It
formation specific (ETS) transcription factors and those encodes a protein with homology to a putative RNA-
that contain hybrid genes with fusion partners that do binding site of RNA polymerase II. Nearly 80% of the
not belong to this family, referred to as non-ETS part- breakpoints found in Ewing’s sarcoma/PNET are located
ners.12,41 The ETS family represents one of the largest within introns 7 and 8.31 A breakpoint in these introns
groups of transcription factors, which are evolutionary results in a chimeric transcript that includes exons 1
highly conserved and play important roles in cell devel- through 7 of the EWSR1 gene. Regardless of whether the
opment and can have major transforming effects contrib- breakpoint is within intron 7 or 8, the exon 8 sequence
uting to tumor development.26 There are 29 members in is spliced out of the chimeric transcript. The second
this group in humans, with FLI1 and ERG representing component in the most common fusion variant is the
the most frequently involved ETS members in the human homolog of the murine Fli1 gene located on chro-
Ewing’s sarcoma/PNET family of tumors. mosome 11q24.38 The murine Fli1 gene contains a ret-
rovirus integration site for the Friend murine leukemia
Translocation t(11;22)(q24;q12) with virus and is rearranged in erythroleukemias induced by
EWSR1-FLI1 Chimeric Gene this virus.46 Both human and murine variants of the FLI1
gene belong to a family of DNA-binding proteins.8 The
Ewing’s sarcoma and related tumors exhibit a unique human FLI1 protein shares approximately 70% sequence
karyotypic abnormality, t(11;22)(q24;q12), that results in homology with the protein encoded by the ETS1 gene
the expression of the chimeric protein EWSR1-FLI1(Fig. (which is expressed during cranial neural crest migration
11-2).13,14,17,58,64 This genetic abnormality occurs in nearly and possibly during vasculogenesis) and seems to func-
90% of Ewing’s sarcoma family tumors.22,51,65,67 The tion primarily as a transcription factor.7,13,14 In the chime-
reciprocal translocation of portions of chromosomes 11 ric protein, the RNA-binding domain of the protein
and 22, involving bands (q24;q12), in Ewing’s sarcoma/ encoded by the EWSR1 gene is substituted for the DNA-
PNET represents a prototypic chromosomal abnormal- binding domain of the FLI1 gene product. The chimeric
ity in soft tissue and bone tumors. EWSR1-FLI1 gene is transcribed under control of the
An initial molecular analysis showed that the chromo- EWSR1 promoter.9 Expression of both of these individual
some 22 breakpoint is located within the EWSR1 gene, genes is virtually ubiquitous in normal human tissues.
whereas the chromosome 11 breakpoint is located The molecular downstream targets for chimeric proteins
within the FLI1 gene.9,38,69,70 The reciprocal translocation are still poorly understood.4 It has been reported that the
between these two chromosomes creates a new chimeric cyclin E, protein tyrosine phosphatase (PTLP1), cyclin-
protein, consisting of portions of the EWSR1 and FLI1 dependent kinase inhibitor 1A (p21/WAF1/CIP1), and
proteins.70 The EWSR1 gene located within 22q12 plays the LAMB3 gene encoding the β3 chain basement mem-
a recurrent major role in several chromosomal transloca- brane laminin 5 are molecular targets for EWSR1-ETS

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 763

EWSR1 22q12

Exon 10
Exon 11
Exon 12

Exon 13

Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 1

Exon 2
Exon 3

Exon 4
Exon 5

Exon 6

Exon 7

Exon 8

Exon 9
cds
cen tel

FLI1 11q24 22q12

Exon 1

Exon 2

Exon 3

Exon 4

Exon 5

Exon 6
Exon 7
Exon 8
Exon 9
cds
cen tel

EWSR1-FLI1 fusion gene


Exon 1

Exon 2
Exon 3

Exon 4
Exon 5

Exon 6

Exon 7
Exon 6

Exon 7

Exon 8

Exon 9
cen tel

breakpoint
EWSR1 portion FLI1 portion
A

EWSR1 protein
1 656 aa

TAD - transactivation domain, 1-285 aa RNA recognition motif, 361-447 aa


Interaction with SF1 domain, 228-264 aa Arginine/glycine/proline rich domain, 454-513 aa
IQ domain, binds calmodulin, 256-285 aa Zinc finger (RanBP2 type) domain, 518-549 aa
Arginine/glycine rich domain, 300-340 aa Arginine/glycine rich domain, 559-640 aa
FLI1 protein
1 452 aa

Sterile alpha motif -pointed domain, 99-105 aa


Winged helix-turn-helix DNA binding, Ets domain, 251-383 aa

EWSR1-FLI fusion protein


1 497 aa

breakpoint
EWSR1 portion FLI portion

C
FIGURE 11-2  ■  Molecular structure associated with translocation t(11;22)(q24;q12) resulting in the EWSR1-FLI1 fusion gene. A, Exon-
intron structures of the EWSR1 and FLI1 genes. Large solid arrows indicate the most frequent locations of the breakpoints within
introns, and the open arrows indicate less frequent variants. The molecular structure of one of the most frequent (type 1) chimeric
genes, which includes coding sequences for the N-terminal domain of the EWSR1 gene and the DNA-binding domain of the FLI1
gene, is illustrated. B, Functional domains of the two proteins involved in the translocation are depicted. The C-terminus end of the
EWSR1 protein contains an RNA-binding domain; the FLI1 protein’s C-terminus contains a DNA-binding domain. The ETS domain
defines a broad family of transcription factors; half of all EWSR1 chimeric partners in the Ewing’s sarcoma/PNET family of tumors
are members of this group. C, The structure of the chimeric protein encoded by the two genes is shown. In the chimeric protein,
the amino terminus of the EWSR1-encoded protein is combined with the DNA-binding domain of the FLI1-encoded protein; the
expression of the chimeric fusion gene is driven by the EWSR1 promoter. In concert with additional cofactors, it acts as a transcrip-
tion factor, resulting in aberrant activation of genetic targets that drive the pathogenesis of this tumor.

ERRNVPHGLFRVRUJ
764 11  Ewing’s Sarcoma and Related Tumors

A B C

D
FIGURE 11-3  ■  Complex karyotype of Ewing’s sarcoma using spectral karyotyping (SKY) to demonstrate the t(11;22) (q24;q12).
A, Chromosomal complement of a cultured Ewing’s sarcoma metaphase cell. B, The same chromosomes hybridized with the SKY
probes. C, Computer processed pseudocolored image of the karyogram of the case shown in A and B. D, The analysis of SKY clearly
demonstrates a translocation involving the long arms of chromosomes 11 and 22 as well as polyploidization of chromosomes 4, 5,
and 8. (Courtesy of Dr. L. Abruzzo, Cytogenetics Laboratory, The University of Texas M. D. Anderson Cancer Center, Houston, TX.)

fusion proteins.3,24,33,40 The expression of the EWSR1- transcription polymerase chain reaction (RT-PCR) and
FLI1 fusion protein upregulates genes associated DNA sequencing, which identify the specific fusion tran-
with neural crest development and is responsible for scripts.11,43 More detailed description of these techniques
the phenotype of Ewing’s sarcoma/PNET family of is provided in Chapter 3.
tumors.23,56 In general, the expression signature of these
tumors is characterized by upregulation of the genes Translocation t(21;22)(q22;q12) with
that are the targets of the ETS family of transcription EWSR1-ERG Chimeric Gene
factors.
This and other translocations in Ewing’s sarcoma It has been shown that EWSR1 may be translocated to
and related tumors can be detected by a variety of cyto- other locations and can form chimeric genes with alterna-
genetic and molecular techniques.5,18,37,51,55 In typical tive partners (Table 11-2). A second alternative cytoge-
cases, conventional cytogenetics identifies the recipro- netic abnormality, t(21;22)(q22;q12), is present in
cal rearrangements between chromosomes 11 and 22 approximately 10% of Ewing’s sarcomas and PNETs
characteristic of Ewing’s sarcoma. However, when such (Fig. 11-4). This translocation fuses the portion of the
a rearrangement occurs in the background of complex EWSR1 gene from 22q12 to the ERG gene located on
chromosomal abnormalities, its conventional cytogenetic 21q22.19,54 In some instances, complex multistep rear-
identification may be problematic. In such instances, rangements of chromosomes 19, 21, and 22 are involved
spectral karyotyping (SKY) is useful to demonstrate the in a reciprocal inversion-insertion mechanism that
specific genetic abnormality (Fig. 11-3). Other tech- results in the EWS1-ERG fusion.19,36 The ERG gene
niques that are commonly used to identify the transloca- encodes a protein that belongs to the ETS family of
tions identify the presence of breakpoints within the gene transcription factors, which, in cell physiology, regulates
using fluorescence in situ hybridization (FISH) with the embryonic development, cell proliferation, differentia-
so-called break-apart or fusion probes. Both cytogenetic tion, angiogenesis, inflammation, and apoptosis. The
and FISH techniques can be complemented by reverse major source of lethal effects of loss of function of

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 765

Biologically, they represent functional substitutes for the


TABLE 11-2 Genetic Aberrations in Ewing’s
two main EWSR1 fusion partners, FLI1 and ERG,
Sarcoma/PNET Tumors
described previously in this chapter.29 These fusions
Neoplasm Translocation Involved gene(s) develop in the absence of the common translocations
such as EWSR1-FLI1 and EWSR1-ERG. They appear to
Ewing’s sarcoma/ t(11;22)(q24;q12) EWSR1-FLI1
PNET with t(21;22)(q22;q12) EWSR1-ERG functionally replace these more common translocations
involvement of t(7;22)(p22;q12) EWSR1-ETV1 in oncogenic transformation.
ETS t(17;22)(q12;q12) EWSR1ETV4
t(2;22)(q33;q12) EWSR1-FEV
t(16;21)(p11;q22) FUS-ERG Translocations with Non-ETS
t(2;16)(q35;p11) FUS-FEV Fusion Partners
Ewing’s sarcoma- t(6;22)(p21;q12) EWSR1-POU5F1
like tumors Inv(22)(q12) EWSR1-ZSG Multiple translocations with fusion partners that do
without ETS (ZNF278) not belong to the ETS family of transcriptional factors
involvement t(4;22)(q31;q12) EWSR1-SMARCA have been identified in Ewing’s sarcoma/PNET family
t(2;22)(q31;q12) EWSR1-SP3
t(1;22)(p36.1;q12) EWSR1-ZNF278
of tumors (Fig. 11-7). They can be divided into two
Complex ring EWSR1-NFATC2 main groups: those that involve EWSR1 as the 5′ partner
chromosome fused to a non-ETS 3′ partner and those that involve
t(4;19)(q35;q13) CIC-DUX4 a completely new 5′ partner such as CIC or BCOR
Inversion involving BCOR-CCNB3 combined with a non-ETS 3′ partner. The first group
Xp114; Xp11.22 CIC-FOXO4
t(X;19) (q13;q13.3) comprises the following translocations: t(6;22)(p21;q12)
EWRS1-POU5F1, Inv(22)(q12) EWSR1-ZSG(ZNF278),
t(4;22)(q31;q12) EWSR1-SMARCA, t(2,22)(q31;q12)
EWSR1-SP3, t(1;22)(p36.1;q12) EWSR1-ZNF278, ring
chromosome with amplification and translocations result-
ing in EWSR1-NFATC2.15,50,60,61,66 The second group of
ERG in mutagenesis animal experiments is related to tumors characterized by involvement of non-ETS genes
hematopoietic stem cell defects. In addition to Ewing’s fused to non-EWSR1 5′ partners include: t(4;19)(q35;q13)
sarcoma, this gene is a common fusion partner of hybrid CIC-DUX4 and an inversion involving Xp11.4; Xp11.22
genes in prostate cancer such as TMPRSS2-ERG and resulting in BCOR-CCNB3.25,30,44,47,53,57 Preliminary data
NDRG1-ERG. Its biologic effects and potential contribu- implicates small cell sarcomas of bone and soft tissue
tion to Ewing’s sarcoma depends on its similarity to FLI1. characterized by these abnormalities that have a differ-
ERG and FLI1 have similar structures and are likely to ent gene expression profile from the group of Ewing’s
have similar biologic effects in chimeric fusions with sarcomas containing fusions with ETS partners.57
EWSR1. ERG shares 68% of the amino acid sequence The 3′ fusion partners in this group of Ewing’s/PNET
identity with FLI1. Moreover, the ETS common domain sarcomas represent a diverse group of transcription
required for sequence specific binding to DNA shows factors with distinct DNA sequence binding domains.
98% identity with FLI1. The formation of the EWSR1- The POU5F1 gene contains a POU homeodomain that
ERG chimeric gene develops in the absence of the most plays a key role in embryonic development and stem
frequent EWSR1-FLI1 fusion and is likely to be able to cell pluripotency. The ZSG gene encodes an A-T hook
replace EWSR1-FLI1 in the oncogenic pathway contrib- DNA binding motif, which plays an important role in
uting to the development of Ewing’s sarcoma. Consider- chromatin remodeling and transcriptional regulation.
ing the similarities between these two fusion partners, it It also contains the Poz domain, which functions as a
is very likely that both chimeras use similar mechanisms site of protein-to-protein interactions and transcriptional
to orchestrate an overlapping repertoire of target genes. repression, as well as the zinc-fingers DNA binding
domain. The SMARCA1 gene encodes a member of the
SWI/SNF family of proteins with helicase and ATPase
Translocations with Alternative
activities that regulate chromatin structure around the
ETS Fusion Partners
target genes. The SP3 gene encodes a protein that regu-
Several additional translocations with alternative ETS lates transcription by binding to a consensus GC- and
fusion partners have been identified in the Ewing’s GT-box regulatory element in target genes. It contains
sarcoma/PNET family of tumors. Three of them involve several other DNA binding domains, including a zinc
EWSR1 combined with ETV1, ETV4, or FEV: t(7;22) finger. The ZNF278 gene encodes a protein that contains
(p22;q12) EWRS1-ETV1, t(17:22)(q12;q12) EWRS1- several transcriptional factor domains, including an A-T
ETV4, and t(2;22)(q33;q12) EWSR1-FEV.16,27,49,66 In hook DNA binding motif that plays an important role in
addition, two translocations of ERG or FEV with FUS chromatin remodeling and transcription regulation. The
were identified: t(16;21)(p11;q22) FUS-ERG and t(2;16) NFATC2 gene is a major transcription factor of activated
(q35;p11) FUS-FEV.16,194 All of these genes belong to the T cells and has several DNA binding domains with REL-
ETS family of transcription factors and contain the ETS and NFAT-homology regions.
domain required for sequence specific binding to DNA The two recently identified fusion genes, CIC-DUX4
in the regulatory elements of the target genes. Overall, and BCOR-CCNB3, are unique because each represents
they have high sequence homology and are nearly identi- a fusion of two genes with distinct transcription factor
cal in terms of the ETS sequence domain (Fig. 11-7). activities (Figs 11-5 and 11-6).25,30,44,47,53,57 The 5′ partner

ERRNVPHGLFRVRUJ
766 11  Ewing’s Sarcoma and Related Tumors

EWSR1
EWSR1 - ERG Translocation t(21;22)(q24;q12)

Exon 10
Exon 11
Exon 12

Exon 13

Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 1

Exon 2
Exon 3
Exon 4
Exon 5

Exon 6

Exon 7

Exon 8

Exon 9
cds
Chr 21 der(21) der(19)
13 13.3 cen tel
12
11.2 13.2
11.1 11.1 13.1
11.2 12 breakpoint
21 11 11
12
22.1
13.2 der(22) ERG
22.2 ERG 13.3
22.3

Exon 12

Exon 11
Exon 10
13.4

Exon 9
Exon 8
Exon 7
Exon 6

Exon 5

Exon 4
Exon 3

Exon 2

Exon 1
Chr 22

cds
Chr 19 EWSR1
Chr 21
ERG
Chr 22 der(22) 13.3 cen tel
13 13.2 Chr 19
12 13.1
12 breakpoint
11.2 11.1 11 11
11.1 11.2 12
EWSR1
12.1 EWSR1 13.1
12.3 12.2 ERG 13.2
inversion
13.1
13.2
13.4
13.3 EWSR1-ERG fusion gene
13.3

Exon 10
Exon 11

Exon 12
A
Exon 1

Exon 2
Exon 3
Exon 4
Exon 5

Exon 6

Exon 2

Exon 3
Exon 4

Exon 5

Exon 6
Exon 7
Exon 8
Exon 9
cen tel

breakpoint
B EWSR1 portion ERG portion

EWSR1 protein
1 656 aa

TAD - transactivation domain, 1-285 aa RNA recognition motif, 361-447 aa


Interaction with SF1 domain, 228-264 aa Arginine/glycine/proline rich domain, 454-513 aa
IQ domain, binds calmodulin, 256-285 aa Zinc finger (RanBP2 type) domain, 518-549 aa
Arginine/glycine rich domain, 300-340 aa Arginine/glycine rich domain, 559-640 aa

ERG protein
1 486 aa

Sterile alpha motif -pointed domain, 117-216 aa


Winged helix-turn-helix DNA binding, Ets domain, 287-418 aa
C

EWSR1-ERG fusion protein


1 518 aa

breakpoint
D EWSR1 portion ERG portion

FIGURE 11-4  ■  Complex rearrangement of chromosomes 19, 21, and 22 with reciprocal inversion-insertion with the EWS-ERG fusion
gene. A, Schematic diagram of the complex translocation of t(21;22)(q13;q12) resulting from a two-step insertion-inversion also
involving chromosome 19 and resulting in EWSR1-ERG fusion on the der(22). B, Exon-intron structures of the EWSR1 and ERG
genes are shown. Arrows indicate the frequent locations of the breakpoints within the introns. The molecular structure of the result-
ing chimeric gene, which contains the coding sequences of EWSR1 at the 5′ end and the ERG coding sequences at the 3′ end of the
chimeric gene, is shown. C, Functional domains of the two proteins involved in the translocations are depicted. Note that the ERG
protein contains the ETS domain similar to that present in the FLI1 protein. D, The structure of the chimeric protein encoded by the
two genes. In the chimeric EWSR1-ERG protein, the N-terminus portion contains sequences encoded by EWSR1 and the C-terminus
contains the ETS DNA binding domain encoded by ERG.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 767

der(4)
16
15.3 CIC
Chr 4 15.2

Exon 10

Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 19
Exon 20
15.1

Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8
Exon 9
16 14

cds
13
15.3 12
15.2 Chr 19 11
11 der(19)
15.1 12
14 13.3 13.1 13.3 cen tel
13 13.3 13.2 13.2
12 13.2 21.1
11 13.1 21.2 13.1
11 12 21.3 12 breakpoint
12 11 11 11
13.1 11 22 12
13.2 12 23 13.1

Exon 1
13.3 24
21.1 13.1 CIC-DUX4
21.2 25
DUX4

cds
21.3 13.2 CIC chimera
26
22 13.3
23 13.4 27
24 cen tel
25 28
26 31.1
27 31.2 breakpoint
31.3
28 32
31.1 33 CIC-DUX4 fusion gene
31.2 34
31.3 35

Exon 10

Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 19
Exon 20
32

Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8
Exon 9

Exon 1
33
34
35
DUX4
cen tel
A
B breakpoint
CIC portion DUX4 portion

CIC protein DUX4 protein


1 1608 aa 1 424 aa

HMG - High-mobility group domain HD1, HD2 - Homeodomain


CHD2, CHD1 - Capicua homology domains
NLS - Nuclear localization sequence domain
C
CIC-DUX4 fusion protein
1 1644 aa

breakpoint
D CIC portion DUX4 portion

FIGURE 11-5  ■  Molecular structure associated with translocation t(4:19)(q35;q13) resulting in the CIC-DUX4 fusion gene. A, Sche-
matic diagram of the reciprocal translocation between the long arms of chromosomes 4 and 19 resulting in CIC-DUX4 fusion on
the der(19). B, Exon-intron structures of the CIC and DUX4 genes. Arrows indicate the frequent locations of the breakpoints. The
molecular structure of the resulting chimeric gene, which contains almost the entire coding sequence of the CIC at the 5′ end and
a portion of the encoding exon of DUX4 at the 3′ end. C, Functional domains of the two proteins involved in the translocation.
D, The structure of the chimeric protein encoded by the two fused genes. It contains almost the entire amino acid sequence of CIC
with its three functional domains at the 5′ end and a portion of exon 1 encoding sequence of DUX4 at the 3′ end.

of the first fusion, the CIC gene, is a human homolog UNIFIED PATHOGENETIC CONCEPT
of a high mobility group (HMG) box transcription
factor that has been shown to mediate c-ErbB signaling. OF EWING’S SARCOMA AND
The 3′ partner in this fusion is the DUX4 gene, RELATED TUMORS
which encodes to two homeoboxes, the repeat-array
and ORF. The protein functions as a transcriptional acti- When t(11;22)(q24;q12) was cytogenetically identified, it
vator. In the second fusion, the 5′ partner is BCOR, represented, until the elucidation of the molecular struc-
encoding the BCL6 corepressor and the 3′ partner, ture of the breakpoint, the only specific finding in Ewing’s
CCNB3, is encoding the testis-specific cyclin B3 and sarcoma. However, the tumors were still mostly classified
plays an essential role in the control of cell cycle. The as Ewing’s sarcoma on the basis of their small cell mor-
group of Ewing’s sarcoma-like tumors is consistently phology and clinicoradiologic features (i.e., a predilection
expanding; a new CIC-FOXO4 gene fusion has been for long tubular bones of young individuals and the
recently identified.59 Moreover, new cytogenetic abnor- absence of a lymphoma-like or leukemia-like picture
malities continue to be identified, as well as complex clinically). Therefore several lesions with clinicopatho-
chromosomal abnormalities with still uncertain clinical logic features distinct from those of classic Ewing’s
significance.28,42,48 sarcoma, such as the presence of neural differentiation
Preliminary data indicate that the diversity of molecu- microscopically or a predilection for the thoracopulmo-
lar abnormalities in Ewing’s sarcoma and related lesions nary region as in Askin’s tumor, were not originally linked
has an impact on their biologic behavior and may repre- to the Ewing’s sarcoma family of tumors.71,85,89,96-98 Sub-
sent clinically useful prognostic factors.4,11,20,42,68,70 sequent investigations showed that t(11;12)(q24;q12) was

ERRNVPHGLFRVRUJ
768 11  Ewing’s Sarcoma and Related Tumors

BCOR

Exon 10

Exon 11

Exon 12
Exon 13
Exon 14

Exon 15
Exon 1

Exon 2
Exon 3

Exon 4

Exon 5
Exon 6

Exon 7
Exon 8
Exon 9
cds
Chr X Chr X
22.3 22.3 tel cen
22.2 22.2
22.1 22.1
21.3 21.3
21.2 21.1 21.2 21.1
11.4 BCOR 11.4 CCNB3
11.23 11.3 CCNB3-CCNB3

Exon 10
Exon 11
Exon 12
CCNB3

Exon 1
Exon 2
Exon 3

Exon 4

Exon 5
Exon 6
Exon 7

Exon 8

Exon 9
11.21 11.22 11.21 11.22
11.1 11.1 11.1 11.1

cds
12 11.2 12 11.2
21.1 13 21.1 13
21.2 21.2 cen tel
21.3 21.3
22.1 22.1
22.2 22.3 22.2 22.3
23 23 breakpoint
24 24
25 25
26 26 BCOR-CCNB3 fusion gene
27 27
28 28

Exon 10

Exon 11

Exon 12
Exon 13
Exon 14

Exon 15

Exon 10
Exon 11
Exon 12
A
Exon 1

Exon 2
Exon 3

Exon 4

Exon 5
Exon 6

Exon 7
Exon 8
Exon 9

Exon 5
Exon 6
Exon 7

Exon 8

Exon 9
cen tel

breakpoint
B BCOR portion CCNB3 portion

BCOR protein
1 1755 aa
Ankyrin domains
CCNB3 protein
1 1395 aa

Cyclin-like domains
C

BCOR-CCNB3 fusion protein


1 3001 aa

breakpoint
BCOR portion CCNB3 portion
D
FIGURE 11-6  ■  Molecular structure of the inversion involving Xp11.4;Xp11.22 with the BCOR-CCNB3 fusion gene. A, Schematic
diagram showing the cryptic inversion within the short arm of chromosome X, which results in BCOR-CCNB3 fusion. B, Exon-intron
structures of the BCOR and CCNB3 genes. Arrow shows an example of a breakpoint within CCNB3. The molecular structure of the
resulting chimeric gene, which contains the entire coding sequence of BCOR at the 5′ end and exons 5-12 of CCNB3 at the 3′ end.
C, Functional domains of the two proteins involved. D, The structure of the chimeric protein encoded by the two fused genes. It
contains the entire sequence of amino acid encoded by BCOR at N-terminus and a portion of the amino acid sequence of CCNB3,
including its cyclinlike domain at the C-terminus.

a consistent cytogenetic abnormality in these lesions; this neural crest lineage differentiation opens the possibility
contributed to the current understanding of these condi- that, occasionally, pathways other than neural differentia-
tions as variants of Ewing’s sarcoma62,77,85,87,92,93 (Fig. tion can occur in a tumor with basic small round-cell
11-8). Although our current understanding is that these morphologic features.
tumors are related pathogenetic entities, there is still a Finally, the concept of pluripotential differentiation
clinical tendency to analyze them separately.72,94 Further in tumors with small round-cell components similar to
investigations revealed that the same cytogenetic abnor- those of Ewing’s sarcoma, such as seen in extremely rare
mality can be identified in at least some of the tumors examples of lesions designated as primitive pluripotential
classified as small cell osteosarcomas.76,78,90 Most likely sarcoma, polyhistioma, or ectomesenchymoma, has been
some of the tumors diagnosed in the past as small cell postulated.80,82,83,91,99 Review of a large series of small cell
osteosarcomas would be classified today as variants of tumors disclosed the presence of rare cases of ill-defined
Ewing’s sarcoma.73,79,88 The production of tumor bone in primitive sarcomas.127 Neural, muscular, and epithelial
a lesion with microscopic, immunohistochemical, and differentiation was documented immunohistochemically
molecular features of Ewing’s sarcoma should be consid- in some of these lesions and was confirmed by ultrastruc-
ered as divergent differentiation rather than a feature of tural studies. It seems that the tendency for pluripotential
a distinct pathologic entity. This is not, however, to deny differentiation is more often seen in lesions involving
the existence of small cell osteosarcoma distinct from the thoracopulmonary region. Moreover, the presence of
Ewing’s sarcoma. MIC2 gene expression confined to primitive round-cell
The concept that Ewing’s sarcoma originates from areas has been shown in some of these lesions.99 The pres-
undifferentiated mesenchymal cells with a propensity for ence of an EWSR1-FLI1 fusion gene has been identified

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 769

FLI1

ERG

FEV

ETV4

ETV1

Same amino acid


A Similar amino acids (K and Q, N and S, A and S, and Q and R)
B

POU5F1 1 360 aa 100


ETS region
Lambda repressor-like, DNA-binding domain Whole protein

Sequence homology (%)


POU-specific domain 80
Homeodomain-like

ZGS 1 687 aa 60
BTB/POZ fold
Zinc finger, C2H2-like domain
40
SMARCA 1 954 aa
HARP domain 20
P-loop containing nucleoside triphosphate hydrolase domain
Helicase superfamily 1/2, ATP-binding domain
SNF2-related domain
P-loop containing nucleoside triphosphate hydrolase domain 0
Helicase, C-terminal domain
ERG FEV ETV4 ETV1
C
SP3 1 781aa
Zinc finger C2H2-type/integrase DNA-binding domain
Zinc finger, C2H2-like domain

NFATC2 1 925 aa
p53-like transcription factor, DNA-binding domain
Immunoglobulin E-set domain
Immunoglobulin-like fold domain

DUX4 1 424 aa
HD1, HD2 - Homeodomain

CCNB3 1 1395 aa
Cyclin-like domains

D
FIGURE 11-7  ■  Structure and functional domains of ETS and non-ETS fusion partners involved in chromosomal translocations of
Ewing’s sarcoma/PNET tumors. A, Molecular structure and the complementary DNA binding sequence of the FLI1 gene and its ETS
domain. It binds to a central GGA(A/T) DNA sequence in regulatory elements of the target genes. B, Sequence homology of the ETS
domains within the genes involved in translocations of Ewing’s sarcoma/primitive neuroectodermal tumors (PNETs), which belong
to the ETS transcription factor family. Note the high degree of similarity of the ETS amino acid sequence of ERG, FEV, ETV4, and
ETV1 as compared to FLI1. C, The analysis of sequence homology of the ETS domains for genes involved in Ewing’s sarcoma/PNET
translocations. D, Protein structures and their functional domains encoded by the genes involved in translocations of Ewing’s
sarcoma/PNET tumors, which form a heterogeneous group defined as non-ETS members. Note that each of these genes has com-
pletely distinct amino acid composition as well as functional domains. (A, Reprinted with permission from Liang H, et al: Nature Struc-
tural Biology 1:871–876, 1994.)

in biphenotypic sarcoma with divergent myogenic/neural of novel emerging molecular classifications of this family
differentiation and in tumors exhibiting adamantinoma- of tumors (Fig. 11-8). It appears that using this infor-
like cytoarchitecture, providing objective evidence that at mation, the Ewing’s sarcoma/PNET family and related
least some of these lesions may have a common patho- tumors can be divided into the following groups: (1)
genesis with the Ewing’s sarcoma group.74,75,81,84,86,95,100 classical Ewing’s sarcoma/PNET group with most fre-
The identification of multiple variants of reciprocal quent fusion genes (EWSR1-FLI1 and EWSR1-ERG), (2)
translocations with the EWSR1 gene and other fusion classical Ewing’s sarcoma/PNET group with alternative
partners belonging to the ETS and non-ETS tran- 3′ ETS fusion partners (EWSR1-EVT1, EWSR1-EVT4,
scriptional factor families as well as translocations with EWSR1-FEV, FUS-ERG, and FUS-FEV), and (3) Ewing’s
gene fusions in which the 5′ partners are different than sarcoma-like tumors with non-ETS 3′ fusion partners
ESWR1, such as FUS, CIC, and BCOR, is the foundation (EWSR1-POU5F1, EWSR1-ZNF287, EWSR1-SMARCA,

ERRNVPHGLFRVRUJ
770 11  Ewing’s Sarcoma and Related Tumors

Relative frequency (%-log)


1 10 100

PNET 1

containing fusions with ETS


Ewing’s sarcoma/PNET
Neural 2
differentiation 3

6
Ewing’s sarcoma 7

containing fusions with non-ETS


Ewing’s sarcoma/PNET
9
? 10
Askin’s tumor Small cell
osteosarcoma 11
(ES/PNET of
thoracopulmonary 12
region) Primitive 13
pluripotential
sarcoma 14
Adamantinoma-like ES
A B
FIGURE 11-8  ■  Unified concept of Ewing’s sarcoma/PNET tumors and related entities. A, Ewing’s sarcoma is considered the prototype
tumor of group. Primitive neuroectodermal tumors (PNETs) represent a variety of closely related lesions distinguished on the basis
of an arbitrarily defined degree of neural differentiation. Askin’s tumor represents either classic Ewing’s sarcoma or PNET involving
thoracopulmonary region. t(11;22)(q24;q12) has been also identified in some small cell osteosarcomas. Link between Ewing’s
sarcoma and primitive sarcoma with pluripotential differentiation has been postulated but is not well established. B, Tentative
molecular classifications of Ewing’s sarcoma/PNETs based on the type of fusion genes. The tumors can be classified in two major
groups containing fusion genes with ETS and non-ETS partners.

EWSR1-SP3, EWSR1-NFATC2 , CIC-DUX4, and BCOR- genetic abnormality can be documented in approximately
CCNB3). The emerging data suggests that all fusions 90% of tumors morphologically classified as Ewing’s sar-
with the ETS members are functionally similar and that comas. In a small proportion of Ewing’s sarcomas, alter-
tumors with such characteristics form a uniform group native chromosomal translocations involving EWSR1 and
with similar gene expression profiles. On the other hand, other genes can occur. In general, the expanded family of
the group containing fusions with non-ETS partners is Ewing’s sarcoma tumors can be separated into two major
heterogeneous due to the involvement of transcriptional groups containing chimeric genes with ETS and non-ETS
factors belonging to different families and to targeting fusion partners.
different sets of genes. Such evidence is available for
at least some of the distinct fusion genes, such as CIC- Incidence and Location
DUX4 and BCOR-CCNB3.47,57 Most interestingly, these
tumors appear to have microscopic features somewhat The peak age incidence and frequent skeletal sites of
different from those of classical Ewing’s sarcoma/PNET, involvement are shown in Figure 11-9. Ewing’s sarcoma
with a tendency to fall into the atypical category, and may accounts for 6% to 8% of all primary malignant tumors
be clinically distinct, exhibiting a propensity for lymph of bone. It is the most frequently occurring primary
node metastasis rather than the lung metastasis charac- sarcoma of bone after osteosarcoma and chondrosar-
teristic of classical Ewing’s sarcoma/PNET.47,57 coma. It has a peculiar predilection for whites and practi-
cally never occurs in blacks. In the 1973 to 2010
Surveillance, Epidemiology, and End Results (SEER)
program series, only approximately 3.5% of Ewing’s
EWING’S SARCOMA sarcoma cases occurred in black patients. This striking
Definition difference in the incidence of Ewing’s sarcoma, which
shows a tenfold higher incidence in the white population
Ewing’s sarcoma is a distinct, round-cell sarcoma of compared to the black population, is potentially of major
bone that occurs predominantly in the long bones of pathogenetic significance and may provide important
skeletally immature patients. The tumor is composed clues to the genetic predisposition for this unusual malig-
of undifferentiated, round, mesenchymal cells that are nancy.114 The peak incidence is during the second decade
rich in glycogen and exhibit a karyotypic abnormality of life, and there is a male predilection (male-to-female
representing reciprocal t(11;22)(q24;q12) and resulting in ratio, 1.3 : 1 to 1.4 : 1) (Figs. 11-10 and 11-11). Approxi-
the expression of a chimeric EWSR1-FLI1 protein. This mately 80% of all patients are younger than age 20 years,

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 771

Askin’s
tumor
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 11-9  ■  Ewing’s sarcoma: peak age incidence and frequent sites of skeletal involvement. Most frequent sites are indicated
by solid black arrows.

1 30

0.9
25
0.8
(cases/100,000 persons)

0.7
20
Incidence Rate

0.6
Frequency (%)
0.5 15

0.4
10
0.3

0.2
5
0.1

0 0
0

+
1-

5-

-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80

Age Group
FIGURE 11-10  ■  Epidemiology of Ewing’s srcoma. Age-specific incidence rates and frequency distribution pattern of 1142 cases. All
races, both sexes, SEER data for 1973 to 2010.

ERRNVPHGLFRVRUJ
772 11  Ewing’s Sarcoma and Related Tumors

0.7

0.6 White Male


(cases per 100,000 persons)

White Female
0.5 Black Male
Incidence Rate

Black Female
0.4

0.3

0.2

0.1

0
0

+
1-

5-

-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80
Age Distribution
FIGURE 11-11  ■  Epidemiology of Ewing’s sarcoma. Age-specific incidence rates by sex and race; 1142 cases from SEER data for 1973
to 2010.

35 metastatic neuroblastoma or acute leukemia should also


be ruled out. These entities are traditionally listed as
30 potential differential diagnostic problems with Ewing’s
sarcoma and other round-cell malignancies. With the
25 advent of molecular probes and immunohistochemical
Frequency (%)

techniques, differentiation among these tumors rarely


20
presents a diagnostic problem. Ewing’s sarcoma has a
15
tendency to involve the shafts of long tubular bones, the
pelvis, and ribs, but practically every bone can be affected,
10 including the acral skeleton, craniofacial bones, and the
clavicle (Fig. 11-12).
5
Radiographic Imaging
0
Typically Ewing’s sarcoma presents as an ill-defined, per-
l

m r
es
ia

s
e

m r
is

es
tre pe

tre w e
ib
in
ac

meative or focally moth-eaten, destructive intramedul-


lv
iti

iti
R

Ex Up
Sp

Pe

Ex L o
of

lary lesion that affects the diaphysis of a major long


ni
ra

tubular bone. The presence of a large, ill-defined density


C

FIGURE 11-12  ■  Distribution of Ewing’s sarcoma by skeletal in soft tissue in addition to the permeative intramedullary
sites. Distribution of Ewing’s sarcoma in major sites of skeleton lesion is a frequent feature (Figs. 11-13 to 11-18). The
as shown by 1142 cases from SEER data for 1973 to 2010.
lesion is often accompanied by a prominent multilayered
periosteal reaction (onion skin) (Figs. 11-13 and 11-15).
The perpendicular “sunburst” type of periosteal new
bone formation can be present but is less common com-
and 50% are affected in the second decade of life. The pared with its occurrence in osteosarcoma. The cortex
remaining cases are diagnosed in patients between ages overlying the lesion can exhibit diffuse sclerosis and
20 and 30 years. Ewing’s sarcoma is extremely rare in thickening (Fig. 11-19). The radiographic features are
patients older than age 30 years. When confronted with nonspecific, but with the clinical features, the diagnosis
the possibility of Ewing’s sarcoma in patients older than of Ewing’s sarcoma is correctly suspected in the majority
age 30 years, the clinician must first eliminate small cell of cases. The lesion involves the medullary cavity and
carcinoma and large cell lymphoma from the differential soft tissue more extensively than is usually documented
diagnosis. In very young patients, the possibility of Text continued on p. 780

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 773

C
FIGURE 11-13  ■  Ewing’s sarcoma of bone: radiographic features. A, Plain radiograph of forearm of a 19-year-old woman with large
soft tissue mass surrounding intramedullary tumor of proximal radial shaft. No periosteal bone formation is present, and cortex
appears intact. B and C, T1- and T2-weighted magnetic resonance images show extensive intramedullary tumor involving radial
shaft and including head of radius. Bulky soft tissue mass developed by tumor permeating through cortex without leaving gross
evidence of destruction in plain films.

ERRNVPHGLFRVRUJ
774 11  Ewing’s Sarcoma and Related Tumors

A B

C
FIGURE 11-14  ■  Ewing’s sarcoma: radiographic features. A and B, Lateral and anteroposterior radiographs of the femur in a 20-year-
old woman show a destructive intramedullary tumor with lamellated periosteal reations; margins are ill defined. C, Computed
tomogram (soft tissue window) of tumor shown in A and B. Image on left shows intramedullary tumor and large extraosseous mass
in posterior muscle group.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 775

A
FIGURE 11-15  ■  Ewing’s sarcoma of bone: radiographic features. A, Plain radiograph of humerus of a 19-year-old man shows large
area of diaphyseal destruction with permeative features. Note lamellated pattern of periosteal reaction in areas of cortical break-
through. B, T1-weighted magnetic resonance image of tumor shown in A reveals large extraosseous mass and intramedullary tumor.

ERRNVPHGLFRVRUJ
776 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-16  ■  Ewing’s sarcoma of bone: radiographic features. A, Anteroposterior radiograph shows a destructive lytic lesion
involving the distal fibula. B, Magnetic resonance image (MRI) shows a high signal intensity mass originating in the distal fibula
with circumferential soft tissue extension. C and D, Axial MRI showing different signal characteristics in T1-weighted (C) and
T2-weighted (D) images.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 777

A B

C D
FIGURE 11-17  ■  Ewing’s sarcoma of bone: radiographic features. A, Anteroposterior radiograph shows a permeative lesion of the
distal femoral metaphysis with soft tissue extension and elevation of periosteum (Codman’s triangle). B, Coronal magnetic resonance
image of the lesion in A shows high signal intensity in an intramedullary tumor with extensive involvement of the parosteal soft
tissue. C, Radioisotope bone scan of the same lesion shows increased uptake in the right distal femur. D, Positron emission
tomogram/computed tomogram of the same tumor identifies an fluorodeoxyglucose (FDG)-avid tumor in the distal femur. Note
particularly high activity in the extraosseus component of the tumor.

ERRNVPHGLFRVRUJ
778 11  Ewing’s Sarcoma and Related Tumors

A B

C D E
FIGURE 11-18  ■  Ewing’s sarcoma: radiographic and gross features. A, A-P radiograph showing a permeative lesion of femoral shaft
with extension into soft tissue. Note radiating periosteal new bone formation and concave cortical defect (saucerization). B, Magnetic
resonance image (MRI) shows abnormal medullary density and lateral subperiosteal mass; impression of bone surface causes
concave defect (saucerization). C, A-P radiograph showing a permeative lesion of femoral shaft. Note thickening of the overlying
cortex and concave cortical defect. D, MRI shows abnormal medullary density and subperiosteal mass filling the concave cortical
defect. E, Gross photograph of the same case shown in C and D shows intramedullary mass permeating overlying thickened cortex
with subperiosteal mass.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 779

A B
FIGURE 11-19  ■  Ewing’s sarcoma: radiographic features. A, Permeative lesion of proximal femoral shaft associated with cortical
thickening. B, Magnetic resonance image (MRI) shows low-signal intramedullary tumor involving long segment of tibial shaft and
extending into intertrochanteric area. Note that involvement documented by MRI extends beyond anticipated area demonstrated
on plain film in A.

ERRNVPHGLFRVRUJ
780 11  Ewing’s Sarcoma and Related Tumors

on plain radiographs. Magnetic resonance imaging and cells with dark condensed nuclei). The latter represent
computed tomography permit more accurate preopera- tumor cells undergoing apoptosis. These types of cells
tive assessment of the lesion than plain radiographs. are sometimes referred to as principal (light) and secondary
They typically demonstrate intramedullary lesions that (dark) cells, respectively. The ratio between these two
involve large segments of the intramedullary cavity and types of cells varies from tumor to tumor and in different
extend beyond the area shown to be involved on plain areas of the same lesion. In some tumors, cords or clus-
radiographs. Ewing’s sarcoma can produce a concave ters of dark apoptotic cells form an interconnecting
defect on the bone surface that is referred to as sauceriza- network of irregular patches that create a pseudoorgan-
tion (Figs. 11-18 and 11-20). This type of radiographic oid pattern. Necrosis is a frequent finding in Ewing’s
appearance is typically seen in predominantly subperios- sarcoma and varies from small patchy foci to large irregu-
teal lesions. More often, saucerization is seen in intra- lar geographic areas (Fig. 11-31).
medullary lesions in which the cortex is permeated and a Tumor cells occasionally form rosettelike structures
secondary subperiosteal and soft tissue mass that com- (Figs. 11-25 and 11-30). Several types of rosettes can be
presses the outer surface of the cortex is formed (Fig. present in Ewing’s sarcoma. In general, less than 20% of
11-18). The majority of Ewing’s sarcomas are purely lytic tumor tissue contains these structures. The presence of
lesions (Figs. 11-20 to 11-24). Occasionally, Ewing’s rosettes in more than 20% of tumor tissue is considered
sarcoma can induce a prominent diffuse thickening of the by some authors to be diagnostic of PNET. For a specific
preexisting bone and may present radiographically as a description of these structures, the reader is referred to
sclerotic lesion, or it may contain focal cloudy opacities. the section on PNET in this chapter.
The sclerotic features or focal opacities are more often Typically, a minimal amount of stromal elements or
seen in lesions involving the metaphyseal portions of long none at all is found among tumor cells that fill medullary
bones or in flat bones and are practically never seen in intertrabecular spaces and replace normal marrow ele-
diaphyseal lesions. On the other hand, diaphyseal lesions ments (Fig. 11-32). The tumor cells permeate the cortex,
of long bones are often associated with diffuse sclerosis invading its haversian and Volkmann’s canals. Complete
and thickening of the overlying cortex. Presentation permeation of the cortex and its eventual disruption are
at an advanced stage with a large mass is a recurring associated with the formation of a mass that initially
theme for patients with Ewing’s sarcoma (Figs. 11-25 and forms subperiosteally and then extends into soft tissue
11-26). (parosteal). Penetration of the cortex and disturbance of
the periosteum are associated with new bone formation.
Gross Findings The proportion of stromal reticular elements and blood
vessels is increased near the advancing edge of the tumor
Intact in its setting, Ewing’s sarcoma presents as a gray compared with its more central intramedullary portion.
to white intramedullary mass with distinct borders. The A more dispersed pattern of growth, including inter-
tumor can involve large segments of the medullary cavity spersed collagen bundles, strands of stromal tissue, and
without causing cortical disruption. Although complete the presence of infiltrated skeletal muscle is characteristic
cortical disruption is not a characteristic feature, the of soft tissue involvement (Fig. 11-33). Multilayered
cortex is typically permeated by small nests of the tumor. periosteal new bone formation, corresponding to an
These finally penetrate the cortex completely and grow identifiable onion-skin appearance on radiographs, can
subperiosteally. Early involvement of adjacent soft tissue accompany the advancing tumor cells. Periosteal bone
is a constant feature of most Ewing’s sarcomas (Figs. formation is associated with plump reactive osteoblasts
11-27 and 11-28). Formation of a large extraosseous and multinucleated osteoclast-like giant cells and may
mass, which often exceeds the size of the intraosseous have foci of cartilage metaplasia. Peripheral areas of the
component, is frequently seen. A large proportion of tumor within soft tissue may show more dispersed tumor
Ewing’s sarcomas have a large circumferential soft tissue cells invading collagenized stroma, adipose tissue, and
mass at presentation that is fusiform in shape. skeletal muscle. Occasionally, a distinct growth pattern in
the form of larger lobules or nests composed of compact
Microscopic Findings tumor cells and separated by fibrous septa can be seen.
This is usually present within the extraosseous compo-
Ewing’s sarcoma is the prototype of a nonhematologic, nent and is rarely seen within the intramedullary portion
small round-cell tumor characterized by a proliferation of the lesion. The absence of inflammatory cell infiltrates
of undifferentiated mesenchymal cells (Fig. 11-29). The and the paucity of stromal elements are constant features
cells grow in solid, densely packed sheets and nests filling of Ewing’s sarcoma. Vascular formation in the central
intertrabecular spaces. They have round, centrally located portion is inconspicuous and is represented by slitlike
nuclei with indistinct cytoplasmic features (Fig. 11-30). capillaries with fine endothelial cells among tumor
The nuclear chromatin is finely granular, and there cells. Larger, thick-walled vessels can be seen within
are usually one to three clearly identifiable small- to stromal bands separating tumor cells. Examination of
intermediate-sized nucleoli. The presence of a prominent cytologic preparations obtained by fine-needle aspira-
nucleolus is generally not a feature of Ewing’s sarcoma. tions, imprints, and scrapings are important elements of
The cytoplasm is indistinct and forms an ill-defined the microscopic examination of Ewing’s sarcoma and
narrow rim around the nucleus. Often a biphasic pattern other small cell malignancies. The morphologic features
is simulated by the presence of so-called dark cells and of cells, and especially the details of the nuclei, are often
light cells (i.e., cells with an open chromatin structure and Text continued on p. 795

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 781

B C
FIGURE 11-20  ■  Ewing’s sarcoma: radiographic features. A Computed tomogram shows intramedullary tumor with cortical disruption
posteriorly and extension into soft tissue. B and C, Anteroposterior and lateral plain radiographs show permeative lesion in metaphy-
seal region; so-called cortical saucerization (concave defect) can be seen on posterior surface in C.

ERRNVPHGLFRVRUJ
782 11  Ewing’s Sarcoma and Related Tumors

B
FIGURE 11-21  ■  Ewing’s sarcoma of bone: radiographic features. A, Large destructive tumor of body and glenoid regions of scapula
with associated soft tissue mass. B, Soap-bubble expansion of proximal phalanx of second toe in young adult. This atypical plain
radiographic appearance was interpreted initially as probable vascular tumor. C, Expansile tumor with moth-eaten pattern of bone
destruction in distal end of fibula in young adult.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 783

C
FIGURE 11-22  ■  Ewing’s sarcoma of acral skeleton: radiographic features. A, Destructive lesion of second metatarsal bone with eleva-
tion of periosteum. B, Radiograph of amputation specimen shows destruction of proximal plate of great toe and ill-defined soft
tissue mass. C, Permeative destructive lesion involving entire shaft of proximal phalanx of finger is shown in this oblique
radiograph.

ERRNVPHGLFRVRUJ
784 11  Ewing’s Sarcoma and Related Tumors

B C
FIGURE 11-23  ■  Ewing’s sarcoma of bone: radiographic features. A, Lateral radiograph of ankle and foot of young adult shows rar-
efaction of posterior part of os calcis. B, Anteroposterior radiograph shows permeative destruction of cancellous bone with ill-defined
border. C, Technetium 99 bone scan shows high uptake of isotope in os calcis, which is more intense posteriorly. Although distal
tibia showed increased isotope uptake, tumor was not present in this site.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 785

A B
FIGURE 11-24  ■  Ewing’s sarcoma: radiographic features. A and B, Plain radiographs show destructive lytic mass of proximal tibia
with increase of new periosteal bone formation.

ERRNVPHGLFRVRUJ
786 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-25  ■  Ewing’s sarcoma: radiographic features. A, A large destruction lesion of the right iliac bone. B, Coronal magnetic
resonance image (MRI) showing a large pelvic tumor. C and D, Axial MRIs showing a large pelvic tumor with low signal intensity.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 787

A B

C D
FIGURE 11-26  ■  Askin’s tumor (Ewing’s sarcoma of the thoracopulmonary region): radiographic features. A and B, Coronal and sagit-
tal computed tomograms showing a large tumor involving the upper portions of the left thoracic wall and pulmonary cavity. C and
D, Axial and sagittal magnetic resonance images showing a large low signal intensity tumor involving the left paraspinal region
and thoracic wall.

ERRNVPHGLFRVRUJ
788 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-27  ■  Ewing’s sarcoma: gross features. A, Ewing’s sarcoma of humeral diaphysis. Note intramedullary tumor involving
large segment of femoral shaft. Fusiform extension into soft tissue with elevation of periosteum is evident. B, Posttreatment Ewing’s
sarcoma of proximal femur. Tumor is completely necrotic. Note prominent cortical thickening, a frequent feature of diaphyseal
lesions. C, Ewing’s sarcoma of proximal humerus. Proximal circumferential soft tissue extension with elevation of periosteum is
evident. D, Ewing’s sarcoma of fibula. Note large subperiosteal lesion with concave cortical surface; defect is referred to as
saucerization.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 789

A B
FIGURE 11-28  ■  Ewing’s sarcoma: gross features. A, Distal femoral resection specimen. Intramedullary tan-gray tumor with posterior
subperiosteal and soft tissue extension associated with concave cortical defect. B, Ewing’s sarcoma of distal tibia with circumferential
subperiosteal and soft tissue extension. C, Closer view of specimen shown in B shows intramedullary tumor with cortical permeation
and periosteal elevation.

ERRNVPHGLFRVRUJ
790 11  Ewing’s Sarcoma and Related Tumors

A B

C D

FIGURE 11-29  ■  Ewing’s sarcoma of bone: microscopic features. A, Low power photomicrograph shows typical patterns in Ewing’s
sarcoma that include solid areas of uniform undifferentiated round cells with minimal amount of cytoplasm. B, High power photo-
micrograph of the case shown in A. Note uniformity of nuclei and absence of brisk mitotic activity. Stroma is minimal and confined
to few delicate fibrous tissue strands and blood vessels. C, Low power photomicrograph shows prominent Homer Wright rosettes.
D, Higher magnification of C shows uniform tumor cells with minimal amount of cytoplasm. Inset, High power photomicrograph of
Homer Wright rosette consisting of a central fibrillar core bounded by concentrically arranged tumor cells. (A and C, ×100; B and
D, ×200; Inset, ×400). (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 791

A B

C D

FIGURE 11-30  ■  Ewing’s sarcoma: microscopic features. A, Low power photomicrograph shows typical pattern in Ewing’s sarcoma.
B, High power photomicrograph showing clear cell change frequently observed in Ewing’s sarcoma. C, Low power photomicrograph
with clear cell change accentuating the cytoplasm and cell membranes of Ewing’s sarcoma cells. D, High power photomicrograph
of Ewing’s sarcoma showing rosette-like arrangement. Inset, Homer Wright rosette with central fibrillar core. (A and C, ×100; B and
D, ×200; Inset, ×400). (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
792 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-31  ■  Ewing’s sarcoma: microscopic features. A, Typical pattern in Ewing’s sarcoma composed of uniform cells showing
focal clear cell change. B, Biphasic pattern in Ewing’s sarcoma produced by scattered pyknotic (dark) cells. C, Extensive geographic
necrosis of Ewing’s sarcoma. Note the presence of apoptotic dark cells at the interphase of viable and necrotic tumor. D, Higher
magnification of the interphase between viable and necrotic tumor tissue showing prominent dark apoptotic cells. (A, B and D, ×200;
C, ×100). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 793

A B

C D
FIGURE 11-32  ■  Ewing’s sarcoma: microscopic features. A, Typical pattern of Ewing’s sarcoma. B-D, Minor variations of the patterns
seen in various areas of the same tumor. (A-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
794 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-33  ■  Ewing’s sarcoma: microscopic features. A and B, Somewhat less compact arrangement of tumor cells in Ewing’s
sarcoma. C and D, Low and high power magnifications showing Ewing’s sarcoma cells infiltrating the skeletal muscle. (A, B, and
D, ×200; C, ×100.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 795

easier to evaluate in cytologic preparations than in smaller Electron Microscopic Findings


biopsy specimens. Therefore in many centers, fine-needle
aspiration is used as the initial diagnostic approach In the past, ultrastructural studies provided essential
when Ewing’s sarcoma is suspected from radiographic support for the diagnosis of Ewing’s sarcoma and
findings. were frequently used to categorize small round-cell
These classic morphologic features are seen in a tumors.128,130,131,134 They have, at best, marginal use in
majority of Ewing’s sarcomas. Microscopic cystic changes differential diagnosis of Ewing’s sarcoma today. The
with blood-filled spaces, as well as changes resembling description of ultrastructural features of Ewing’s sarcoma
aneurysmal bone cyst, can be superimposed on Ewing’s is retained here as they explain the light microscopic
sarcoma. In a small number of tumors, the microscopic appearance of this tumor and provide support for the
appearance of tumor cells may deviate from the so-called primitive undifferentiated nature of tumor cells.
classic pattern. Tumors that deviate from the usual pattern Classic Ewing’s sarcoma is composed of primitive oval
are referred to as atypical Ewing’s sarcomas.108,119,122,126,133,145 to round mesenchymal cells. The cellularity is high, and
In some cases, the tumor cells are larger than those seen the tumor cells are densely packed with almost nonexis-
in conventional tumors, may show cytoplasmic vacuoliza- tent stroma. Occasionally, tight and desmosome-like
tion, and may exhibit greater heterogeneity. These fea- junctions can be present. Two types of cells as defined by
tures are more often seen in recurrent and treated lesions light microscopy—a principal type (light with open intact
but can also be present in primary tumors. In addition, chromatin) and a secondary type (dark with condensed
some lesions may show focal spindle-cell change in the chromatin)—can also be recognized at the ultrastructural
tumor cells. In such areas the tumor cells have short, level. Minimal amounts of stromal elements associated
plump, oval nuclei. The deviation from the classic mor- with endothelial-lined capillaries and occasional larger
phologic features in atypical variants of Ewing’s sarcoma vessels are seen focally. In conventional Ewing’s sarcoma,
is rather minimal and is not striking at low-power mag- the average size of tumor cells is approximately 14 µm,
nification. The large cells are only slightly increased in and the cells are rather uniform. Centrally located nuclei
size (20 to 24 µm), and they do not have features observed are oval to round and have outlines with occasional
in pleomorphic or large cell sarcomatoid or epithelial indentations of the nuclear membrane. The chromatin is
neoplasms.128-130 Similarly, spindle-cell change represents fine, granular, and evenly distributed. Typically, there are
a rather minimal deviation from classic Ewing’s sarcoma, one to three medium-sized nucleoli. The cytoplasm is
and the tumor cells do not appear to be the type of cells primitive, with sparse inconspicuous organelles (Fig.
seen in typical spindle-cell lesions such as malignant 11-35). Mitochondria are few and rather small. Some
fibrous histiocytoma or fibrosarcoma. Molecularly veri- intracytoplasmic reticulum and poorly developed small
fied cases of Ewing’s sarcoma-like tumors with significant Golgi centers are present. The most relevant ultrastruc-
deviation from classical morphology have been published; tural feature, with an overall primitive mesenchymal
they include lesions with the presence of desmoplastic appearance of tumor cells, is the abundance of glycogen
stroma, organoid and glandular patterns, as well as arranged in rosettelike deposits (Fig. 11-35). This feature
adamantinoma- or Merkel cell carcinoma-like fea- is present in appropriately fixed and processed specimens
tures.35,74,75,81,84,86,100,180 Such tumors can mimic a wide (i.e., in nonaqueous solutions). Inappropriately fixed and
range of epithelial and nonepithelial neoplasms, and their processed samples, especially those retrieved from paraf-
diagnosis can be very challenging. fin blocks, may show washed-out areas within the cyto-
plasm, which are devoid of organelles (Fig. 11-36).
Ultrastructurally, there is a continuous transition from
Cytology
intact principal cells to dark apoptotic cells, and in some
Aspirates from Ewing’s sarcoma/PNET are usually very areas, the so-called dark cells can predominate. The
cellular and contain a population of dispersed small ultrastructure of the dark cells may show all the features
undifferentiated mesenchymal cells (Fig. 11-34). A deli- of nuclear condensation and segmentation described for
cate, finely granular chromatin pattern and clearly iden- apoptosis. Fully developed apoptotic bodies seem to be
tifiable small to medium nucleoli are characteristic. The the final destiny of dark cells.
cytoplasm is indistinct and forms only a narrow rim
around the nucleus. Aspirates from Ewing’s sarcoma are Treatment and Behavior
important elements in the diagnostic workup.116 The
morphologic features of cells, and especially the details Ewing’s sarcoma belongs to the category of the most
of the nuclei, are often easier to evaluate on cytologic aggressive tumors and has a high propensity for local
preparations than in small biopsy specimens. Therefore, recurrences and distant metastases, which occur predom-
fine-needle aspiration is used as the initial diagnostic inantly in the lungs. Before the advent of chemotherapy,
approach when Ewing’s sarcoma is suspected from radio- the prognosis was dismal, with a 5-year survival rate of
graphic findings. Immunohistochemical and molecular less than 20%. The use of multimodality treatment plans
study allowing differential diagnosis with other small of irradiation and multidrug chemotherapy plus surgery
cell malignances may be performed on material obtained has significantly changed this survival rate. Those patients
for cytologic preparations.148 Overall Ewing’s sarcoma/ who initially have localized, resectable disease and are
PNET represent a primary bone sarcoma that can be treated with multidrug chemotherapy in addition to
definitively diagnosed by aspiration cytology in most surgery have a 5-year survival rate of approximately
cases. 70%.75,101-103,125,151 For patients with disseminated disease

ERRNVPHGLFRVRUJ
796 11  Ewing’s Sarcoma and Related Tumors

A B

C D

FIGURE 11-34  ■  Ewing’s sarcoma: fine-needle aspiration cytology. A-D, Fine-needle aspirate containing undifferentiated round
cells with occasional nucleoli and indistinct cytoplasm, occasionally forming rosette-like structures (arrows). Inset, Rosettelike struc-
ture formed by circumferential arrangement of tumor cells around central core containing delicate fibrillar cytoplasmic material.
(A-D, ×300; Inset, ×400). (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 797

B
FIGURE 11-35  ■  Ewing’s sarcoma of bone: ultrastructural features. A and B, Undifferentiated mesenchymal cells containing sparse
cytoplasmic organelles and prominent deposits of glycogen. Note fine, evenly dispersed, granular chromatin of tumor cell nuclei
(A, ×5000; B, ×14,000).

ERRNVPHGLFRVRUJ
798 11  Ewing’s Sarcoma and Related Tumors

B
FIGURE 11-36  ■  Ewing’s sarcoma: ultrastructural features. A, Undifferentiated tumor cells containing sparse cytoplasmic organelles
and regular round nuclei with freely dispersed chromatin on occasional nucleoli. B, Tumor cell with prominent washed-out glycogen
pools (A, ×2500; B, ×6000).

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 799

at diagnosis, the 5-year survival rate is significantly lower, latter sites are significantly larger at presentation and
in the range of 30%. Similar to osteosarcoma, it has been show extensive soft tissue involvement. The tumor
shown that surgical removal of the resectable lung metas- volume estimated from radiographs also correlates with
tases improves survival. Thus the presence of metastases survival.101,124,138 Patients who have lesions smaller than
is the single most important factor adversely affecting the 100 ml have an 80% 3-year survival compared with 32%
survival of patients with Ewing’s sarcoma. The incidence for patients who have larger tumors.108 The relationship
of disseminated disease at the time of diagnosis is high, between tumor volume and survival has been confirmed
and approximately 15% to 28% of patients initially have by other studies, although the cutoff point for small-
metastatic disease.109,114,115,117,119,121,124,125,132,141,146 versus large-volume lesions was higher (i.e., 200 ml).101
A variety of clinical factors, such as age and sex, site Several studies have shown that some clinical labo-
of primary tumor and its size, and some clinical labora- ratory parameters (increased levels of serum lactate
tory parameters, have been postulated to be predictive of dehydrogenase, increased sedimentation rate, and ele-
the prognosis.58,104,105,118,123,140,146,149 The relationship vated white blood cell count) are adverse prognostic
between survival, gender, and age appears to be inconsis- factors most likely related to more advanced stages of
tent among the published reports; the consensus is that, presentation.117,120,124,125
most likely, these parameters do not significantly affect Some microscopic features are implicated as predic-
the prognosis.103,109,113,120,124,125,135,137 Similarly, SEER data tors of more aggressive behavior. Extensive spontaneous
implies that female patients show more favorable survival necrosis of untreated lesions is a predictor of more
rates as compared to males, but the data are not consis- aggressive clinical behavior and is linked to lower survival
tent in time intervals and are not statistically significant. rates.111,113,129 Other studies have shown that the presence
SEER data show significant improvement of national of dark (apoptotic) cells and a so-called filigree pattern are
survival rates in the three 5-year intervals spanning 1973 more predictive of a poor prognosis than necrosis.132 The
to 1988, which parallel the introduction of modern neo- association between poor outcome and the filigree pattern
adjuvant chemotherapy protocols (Fig. 11-37). The was originally recognized in a large series of interinstitu-
Ewing’s sarcoma/PNET family of tumors are obvious tional cases.127 This peculiar pattern of Ewing’s sarcoma
candidates for targeted therapy which is under experi- is typically seen in the soft tissue component and is prac-
mental testing, but the potential benefits of this novel tically never observed in the central intramedullary
therapeutic modality are not yet reflected in the portion of the tumor. Its presence is considered by some
population-based survival statistics. authors to be synonymous with invasion into soft tissue
The site of the primary tumor is another prognostic and may signify a higher stage and volume of lesions.128
factor. Patients who have resectable lesions of the extrem- It is of importance that in patients with localized disease,
ity bones have a better survival rate than those who have the presence of this pattern has been associated with poor
lesions affecting the trunk bones, such as the pelvis and prognosis regardless of differences in morphology and
the thoracopulmonary region. In addition, lesions in the phenotypic differentiation.146

80
1973-1977 1989-1994
70
1978-1982 1995-2000
Relative Survival Rate (%)

1983-1988 2001-2005
60

50

40

30

20

10

0
Both Sexes Male Female
FIGURE 11-37  ■  Ewing’s sarcoma: survival rates. Five-year survival rate of 1142 patients with Ewing’s sarcoma by sex and time periods
for all races from SEER data for 1973 to 2005.

ERRNVPHGLFRVRUJ
800 11  Ewing’s Sarcoma and Related Tumors

A correlation between a high mitotic index (one or STAG2 and TP53 mutations is associated with poor out-
more mitoses per high-power field) and poor survival was comes, and they define a clinically aggressive variant of
found in one study.111 Conversely, the presence of glyco- Ewing’s sarcoma.147
gen, the degree of tumor tissue vascularization, and the
presence of vascular invasion were shown to have no rela-
tion to clinical behavior in patients treated with modern
multidrug protocols.129,139 PRIMITIVE NEUROECTODERMAL TUMOR
Postoperative evaluation of necrosis induced by Definition
therapy similar to that used for osteosarcoma appears to
be an important factor in predicting the length of local PNET occurs less frequently than conventional Ewing’s
control and disease-free time.123,136,141-144,149 The technical sarcoma. In addition to Ewing’s sarcoma-like features,
aspects of histologic mapping of therapeutic responses it exhibits primitive neural differentiation. Neural dif-
in Ewing’s sarcoma are identical to those described ferentiation in PNET can be identified by light micros-
for osteosarcoma (see Chapters 1 and 5). Favorable copy, which reveals the formation of unequivocal Homer
response, which is defined as total or subtotal (90% Wright rosettes, and immunohistochemically by the
to 100%) necrosis, appears to be a strong predictor expression of neural markers. Electron microscopy is
of long-term survival. The link between favorable prog- considered by many authors as the most reliable tech-
nosis and good chemotherapy response has been consis- nique to document the neural differentiation of PNET.
tently shown in several independent studies. Moreover, Like Ewing’s sarcoma, PNET exhibits reciprocal
the degree of postchemotherapy necrosis seems to cor- t(11;22)(q24;q12), resulting in expression of the chimeric
relate with the rate of disease-free survival. In a study EWSR1/FLI1 protein.152,154,158,165 This genetic abnormal-
from the Rizzoli Institute, the 5-year disease-free survival ity can be identified in a majority of tumors morphologi-
rate was 90% for patients with complete necrosis, 53% cally classified as PNET. Most authors consider Ewing’s
for those with microscopic residual tumors, and 32% sarcoma and PNET to be related entities within a con-
for those whose lesions had gross evidence of residual tinuous spectrum of minimal to prominent primitive
tumor.137 neural differentiation. Similar to classical Ewing’s
Alterations of several oncogenes such as c-erb-B2, sarcoma, alternative translocations involving EWSR1 and
TP53, and MDM2 occur very infrequently and do not other genes can occur; the PNET group can be divided
seem to play a major role in either the pathogenesis or into tumors characterized by chimeric genes containing
prognosis of Ewing’s sarcoma. Immunohistochemical fusions with ETS and non-ETS partners.
features of overexpression of the genes involved in the
development of drug resistance, such as P glycoprotein, Incidence and Location
show some promising results, but too few cases have been
studied to assess the practical application of these If strict criteria are applied to the diagnosis (i.e., more
findings.140,186 than one unequivocal feature of neural differentiation
More than ten different fusion transcripts containing is required), PNET is a relatively rare lesion. At The
exons of EWSR1 and FLI1 have been identified in Ewing’s University of Texas M.D. Anderson Cancer Center,
sarcoma.69,70 In addition, other translocations in this where virtually all Ewing’s sarcoma-like tumors were, in
group of tumors continue to have multiple variants due the past, examined by electron microscopy in addition to
to the mobile positions of breakpoints, which creates other techniques, PNET represented approximately 10%
incomprehensive diversity of fusion proteins and poten- of the lesions classified by light microscopy as Ewing’s
tially different biologic effects. It is uncertain whether sarcoma–like tumors. The peak age incidence and skele-
this diversity is of clinical significance, but several reports tal distribution patterns and the radiographic features of
indicate that type I fusion transcript for the EWSRI- PNET are identical to those of Ewing’s sarcoma.
FLI1 chimera is associated with better survival when
compared with type II fusion transcripts independent of Microscopic Findings
tumor site and stage.42,112,150 Moreover, Ewing’s sarcomas
with secondary genetic changes revealed by a compara- Microscopically, the tumor has all the features of Ewing’s
tive genomic hybridization have high propensity for sarcoma. It is composed of small, round, primitive, mes-
distant metastasis.106 Ewing’s sarcomas carrying fusion enchymal cells.152-154,156 In a typical case, the presence of
genes with non-ETS members may be biologically and unequivocal rosettes of Homer Wright type is a promi-
clinically different from conventional Ewing’s sarcomas nent and easily recognizable feature (Fig. 11-38). It is
carrying fusion genes with ETS members. proposed that approximately 20% or more of the tumor
A novel group of prognostic factors is emerging from tissue should bear clearly recognizable Homer Wright
genome profiling studies. These studies documented that rosettes to be considered as morphologic evidence of
Ewing’s sarcoma has one of the lowest mutational neural differentiation and to permit the designation of a
burdens. It is not unusual to see gene fusions as sole tumor as PNET on the basis of light microscopic evi-
structural genomic abnormalities. A small fraction of dence alone. In our experience, PNETs with this micro-
Ewing’s sarcoma carries the somatic mutations in the scopic feature, prominent Homer Wright rosettes in
cohesin complex subunit STAG2, which are mutually more than 20% of tumor tissue, are exceedingly rare.
exclusive with CDKN2A mutations.107 They frequently Several types of rosettes can be present in PNET. This
coexist with TP53 mutations.107,110 The presence of structure represents a clearly recognizable circular

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 801

FIGURE 11-38  ■  PNET: microscopic features. A and B, Prominent rosette formations in Ewing’s sarcoma cells. Inset, Enlarged image
of Homer Wright rosette (A and B, ×100; Inset, ×300). (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
802 11  Ewing’s Sarcoma and Related Tumors

arrangement of cells with processes that form a fibrillar classic Ewing’s sarcoma had a 60% survival rate after 7 1 2
material oriented toward the center of the structure. years of follow-up compared with a 45% rate for PNET.
These types of rosettes are identical to those seen in Similar results have been documented in the study
neuroblastoma (Fig. 11-29). Less mature, smaller (primi- from the National Cancer Institute, which showed that
tive) rosettelike arrangements formed by several (6 to 10) patients with Ewing’s sarcoma are more likely to have
cells oriented around a corelike space without clearly localized disease than patients with PNET. The presence
recognizable fibrillar centers can also be present (Fig. of neural differentiation in Ewing’s sarcoma may have
11-30). These types of rosettes should not be used as sole prognostic significance only if stringent criteria for clas-
microscopic evidence of neural differentiation. Flexner- sification of tumors as PNET are applied. In summary,
Wintersteiner rosettes containing a clear, sharply delin- from diagnostic and therapeutic points of view, PNET
eated, central lumen, similar to that described in and Ewing’s sarcoma can be considered one group of
ependymoma and retinoblastoma, can be present but are tumors.
exceptionally rare in PNET of bone.

Electron Microscopic Findings ASKIN’S TUMOR


It is generally accepted that ultrastructural identification In 1979, Askin et al. described a small round-cell tumor
of neural differentiation within a tumor that has the of the thoracopulmonary region that affected children.
overall morphology of Ewing’s sarcoma is the most reli- In the original report, the authors postulated that this
able way to identify PNET. Ultrastructurally, the neural lesion had a distinct pathogenesis and apparently arose in
differentiation is rather discrete and typically focal in a the ribs, predominantly in the periosteum, but could also
tumor otherwise consistent with Ewing’s sarcoma (Fig. arise in the soft tissue, and possibly within the lung.71
11-39).152,153,159-161,164 Several ultrastructural features of Microscopically, the tumor is indistinguishable from
neural differentiation are usually present. Sparse neuro- the group of tumors that include Ewing’s sarcoma and
secretory granules are associated with both developing PNET. Since the original publication, several subsequent
Golgi centers and cell processes. Neurosecretory gran- studies have focused on the issue of whether the lesion
ules are 100 to 140 nm in size and represent electron- represents a distinct clinicopathologic entity or is a
dense, round or oval structures composed of a central variant of Ewing’s sarcoma and PNET involving the tho-
dense core surrounded by a membrane (Fig. 11-40). racopulmonary region.152,157,168,170,171 The lesion may show
These granules can be finely distributed in the cytoplasm, classic features of Ewing’s sarcoma or may exhibit light
but in more mature PNETs, they have a tendency to microscopic, immunohistochemical, and ultrastructural
group together to form loose clusters. The formation features of primitive neural differentiation as seen in
of cytoplasmic projections is a frequent feature of PNET. Atypical variants of the tumor that resemble
PNET. Occasionally, these projections are organized and those described for Ewing’s sarcoma and PNET have also
form typical Homer Wright rosettes in which the cyto- been described.153
plasmic projections are oriented toward the central Immunohistochemical studies have shown phenotypic
core (Fig. 11-39). The outer perimeter of the cells features similar or identical to those of Ewing’s sarcoma
forming the rosette can sometimes be delineated by an and PNET. The lesions are frequently positive for one
incomplete basal lamina–like material. The cytoplasm of or several of the so-called neural markers, including
better-differentiated cells may contain filaments or even neuron-specific enolase and neurofilaments of 70 kD,
neurotubules, which represent the ultrastructural fea- and may also express neuroendocrine markers such as
tures of the neuralxcc differentiation (Fig. 11-40). chromogranin.152,164,168 Occasionally, these tumors can be
focally positive for low-molecular-weight keratins, and
Treatment and Behavior the staining for the MIC2 gene product is similar to that
for conventional Ewing’s sarcoma.130,152,199 Moreover,
The treatment plan for PNET is identical to that for cytogenetic analysis has revealed the same t(11;22)
Ewing’s sarcoma. The presence of neural differentiation (q24;q12) as seen in Ewing’s sarcoma and PNET.166 In
in Ewing’s sarcoma has been reported by several studies general, genetic abnormalities and especially the gene
to be associated with more aggressive behavior, but that fusions overlap with those of Ewing’s sarcoma.
association is not supported by other studies.155-157,162,163 The current consensus is that Askin’s tumor is a variant
It appears that the diagnostic criteria for classifying a of Ewing’s sarcoma and PNET that involves the thora-
tumor as a PNET differ among the series, and these dif- copulmonary region. The unique features seem to be
ferences may be responsible for the discrepancies. In a frequent primitive neural or neuroendocrine differentia-
recent interinstitutional study involving several centers in tion, as well as focal epithelial differentiation. Although
the United States and Europe, the analysis of 315 cases the lesion is considered to be a variant of Ewing’s sarcoma
showed no association between neural differentiation and and PNET, it has a higher propensity for pluripotential
more aggressive behavior.146 In contrast, a study based on divergent differentiation than does conventional Ewing’s
the German tumor registry in Kiel showed that neural sarcoma.
differentiation can be associated with more aggressive Review of a large series of this tumor indicates the
behavior if stringent diagnostic criteria for PNET are aggressive nature of Askin’s tumor, with a 15% five year
used (i.e., if the presence of strong positivity for at least survival.167 The patients with Askin’s tumor frequently
two neural markers can be documented).162 In this study, present with locally advanced disease and large tumors; a

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 803

B
FIGURE 11-39  ■  PNET: ultrastructural features. A, Tumor cells with sparse cytoplasmic organelles as seen on low power magnifica-
tion. Axonal differentiation is focally present (arrow). B, Centrally placed cytoplasmic processes correspond to core of rosette
(asterisk). Inset, Cross section of cytoplasmic processes of tumor cells. (A, ×1500; B, ×3000; Inset, ×8000)

ERRNVPHGLFRVRUJ
804 11  Ewing’s Sarcoma and Related Tumors

D
FIGURE 11-40  ■  PNET of bone: ultrastructural features. A, Axonal differentiation of tumor cells with formation of interconnecting
cytoplasmic processes (arrows). B, Neurosecretory granules (arrows) in tumor cell cytoplasm. C, Interdigitalizing cytoplasmic pro-
cesses with neurosecretory granules (arrows). D, Neurotubules with the cytoplasmic process of tumor cell (asterisk). (A, ×3500;
B, ×9000; C, ×24,000; D, ×35,000)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 805

Ewing’s
PNET PNETs.174,179,191,203 In evaluating the positivity of a lesion
sarcoma for the MIC2 gene product, it is important to remember
Light microscopic
neural differentiation
that it is not specific for Ewing’s sarcoma and related
(Homer Wright rosettes) conditions. It is also positive in pediatric lymphomas,
20% of tumor tissue lymphocytic lymphoma, and occasionally in rhabdomyo-
Ultrastructural sarcoma and even synovial sarcoma.174,179,203,205
neural differentiation Vimentin is the most consistently positive marker and
can be documented in 80% to 90% of Ewing’s sarcomas
Expression of
neural markers and PNETs (Fig. 11-42).198-200 Features of neural dif-
ferentiation are frequently seen in Ewing’s sarcoma and
PNET. On one end of the spectrum are the so-called
MIC2 expression classic examples of Ewing’s sarcoma, which are posi-
tive for vimentin only, express the MIC2 gene product,
Vimentin contain large amounts of diastase-sensitive material
(glycogen) positive for periodic acid–Schiff, and are
FIGURE 11-41  ■  Ewing’s sarcoma versus PNET: phenotypic dif- completely negative for any neural markers. A strong
ferences. Ewing’s sarcoma and primitive neuroectodermal positivity for periodic acid–Schiff is present in at least
tumor (PNET) are related entities. Distinguishing feature is
degree of neural differentiation. Generally, two or more major
70% of Ewing’s sarcomas and PNETs. On the opposite
features of neural differentiation are needed for diagnosis of end of the spectrum are PNETs with obvious micro-
PNET. scopic neural differentiation (Homer Wright rosettes in
20% or more of tumor tissue), strong diffuse immuno-
histochemical expression of several neural markers, and
ultrastructural features supportive of neural differentia-
significant proportion of them have evidence of meta- tion (see the section on electron microscopic findings in
static disease at presentation.167,169,172,173 PNET).130,199,202,207,208 Between these two extremes is a
continuous spectrum of lesions that exhibit focal features
of neural differentiation as revealed by the expression of
IMMUNOHISTOCHEMICAL FEATURES one or another neural marker. It is generally accepted
OF EWING’S SARCOMA AND PNET that more than one obvious feature of neural differentia-
tion is required to classify a lesion as PNET.
It is unnecessary to provide separate descriptions of Positivity for neuron-specific enolase is generally dis-
immunophenotypic features for Ewing’s sarcoma and regarded as a specific marker of neural differentiation.
PNET because most authors consider these lesions to be On the other hand, if its expression can be correlated
related entities within a continuum of overlapping immu- with other features of neural differentiation, it provides
nophenotypic features. As a consequence, specific desig- a valuable tool with which to assess the degree of neural
nation of a lesion as either Ewing’s sarcoma or PNET is phenotypic expression.129,177 We agree with the authors
based on subjective, arbitrary, and/or semiquantitative who defend the applicability of using neuron-specific
assessment of the amount of neural differentiation enolase for the assessment of the neural phenotype seen
(Fig. 11-41). in Ewing’s sarcoma and PNET in conjunction with other
Expression of the MIC2 gene product (CD99) in a features that support neural differentiation.130,177 Patterns
majority of Ewing’s sarcomas and PNETs is a diagnosti- of expression similar to those for neuron-specific enolase
cally important feature of these lesions.174 Although it is can be documented with other neural markers in Ewing’s
not specific for Ewing’s sarcoma and PNET, it provides sarcoma and PNET (Fig. 11-44). These neural markers
great support for the diagnosis of these tumors if used are Leu-7, HNK-1, protein gene product 9.5 (PGP9.5),
with other features (Figs. 11-42 and 11-43). Understand- neurofilaments of 200 kD, glial fibrillary acidic protein,
ing the biology of this gene in normal tissue and its and S-100 protein.176,177,185,187,193,197,204
pattern of expression in Ewing’s sarcoma, PNET, and Neuroendocrine differentiation can be revealed by
other tumors is helpful for its application as a diagnostic ultrastructural studies for the presence of neurosecretory
tool. MIC2 is a gene located in the X-Y pairing region granules or by immunohistochemical testing for the pres-
and is mapped to the Xp22,32p-ter region of the X chro- ence of the chromogranin family of markers such as CgA,
mosome.183 This region corresponds to the euchromatin CgB, and CgC-SgII. It has been shown that Ewing’s
region p11.2p-ter on the Y chromosome. It codes for a sarcoma and PNET are typically strongly positive for
30-kD cell adhesion-membrane glycoprotein (p30/32) a distinct class of chromogranin proteins, designated as
involved in the proliferation and migratory ability of SgII and are negative for CgA and CgB.202 Neuroblasto-
Ewing’s sarcoma cells.182,189,190 The product was originally mas are typically positive for CgA and CgB components.
known as the antigen involved in the T-cell adhesion Interesting clues for phenotypic differentiation and of
process and recognized by the 12E7 antibody. The potential significance for the biology of Ewing’s sarcoma
HBA-71 antibody was raised against Ewing’s sarcoma and PNET were recently provided by studies of the Trk
cells. These two antibodies identify the antigen in family of tyrosine kinases, which belong to the superfam-
formalin-fixed, paraffin-embedded tissue.179,184,192 Several ily of transmembrane receptors.178,201 These proteins are
studies have shown that the MIC2 (CD99) product can essential for neural differentiation and are molecular
be identified in almost 90% of Ewing’s sarcomas and receptors for nerve growth factors.175,196 The presence of

ERRNVPHGLFRVRUJ
806 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-42  ■  Ewing’s sarcoma: microscopic features. A, Intermediate power view of small round-cell tumor with sparse stromal
elements. B, Strong diffused positivity of tumor cells for vimentin. C and D, Low and high power views of strong membranous posi-
tivity of tumor cells for MIC2 (CD99) gene expression. (A, B, and D, ×200; C, ×100.) (A, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 807

A B

C D
FIGURE 11-43  ■  Ewing’s sarcoma: microscopic features. A, Ewing’s sarcoma composed of small round cells. B. Diffuse cytoplasmic
expression of the MIC2 gene. C and D, Tumor cell exhibits strong positivity for periodic acid–Schiff stain, which is diastase sensitive
(A-D, ×400). (A, Hematoxylin-eosin; B, DAB; C, PAS; D, PAS after digestion with diastase.)

ERRNVPHGLFRVRUJ
808 11  Ewing’s Sarcoma and Related Tumors

A B

C D
FIGURE 11-44  ■  Ewing’s sarcoma and PNET: immunohistochemical features. A, Focal positivity for synaptophysin. B, Cytoplasmic
positivity for neuron-specific enolase. C, Strong diffuse cytoplasmic positivity for MIC2 gene product revealed with HBA-71 antibody.
D, Strong, diffuse cytoplasmic positivity for vimentin (A-D, ×400).

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 809

TrkB and TrkC receptors seems to be a feature of Ewing’s interphase cytogenetics, which identifies genetic abnormali-
sarcoma without any additional neural differentiation, ties in nondividing cells and can be applied to formalin-
whereas the expression of TrkA predominantly occurs fixed and paraffin-embedded tissues. The most popular
with other neural differentiation features.201 technique is FISH, with a breakapart probe documenting
Ewing’s sarcoma and PNET are typically negative for rearrangement of the EWSR1 gene. This test is positive
epithelial markers. Scattered positivity of individual regardless of the type of EWSR1 3′ fusion partner and
tumor cells for keratins can be seen in approximately can detect the vast majority of Ewing’s sarcoma/PNET
10% of these tumors.181,209 In extremely rare instances the tumors. These cytogenetic techniques can be performed
diffuse strong positivity of tumor cells in otherwise typical on both conventional cytologic preparations and
Ewing’s sarcoma for keratin can be identified.209 It is formalin-fixed paraffin-embedded histologic sections. It
uncertain whether lesions with such phenotype carry the should be emphasized that a negative test for EWSR1
genetic abnormality characteristic of Ewing’s sarcoma/ rearrangement with the breakapart probe does not abso-
PNET family of tumors. On the other hand, several cases lutely disqualify the diagnosis of Ewing’s sarcoma/PNET
of unusual tumors with obvious epithelial differentiation tumor; there are rare examples in which the 5′ fusion
and adamantinoma- or Merkel cell carcinoma-like fea- partners may involve genes other than EWSR1, such as
tures carrying the EWSR1/FLI1 chimeric gene have been FUS, CIC, or BCOR.
described (Fig. 11-45).74,81,84,86,100,180 The other frequently applied technology is the
Modern immunohistochemistry offers an opportunity RT-PCR detection of the fusion transcripts. Such tests
to identify at least some of the fusion partners.191 Strong are typically designed with multiple primers and are
expression of FLI1 suggests that the tumor may contain capable of detecting several variants of fusion transcripts.
the EWSR1-FLI1 hybrid gene, in which the overexpres- However, this approach is not absolutely safe because
sion of FLI1 is driven by the EWSR1 promoter. Simi- the technique can generate false-positive results due to
larly, the strong positivity for ERG is a useful surrogate primer annealing to alternative nonspecific sequences.
suggesting the presence of the EWSR1-ERG hybrid The test is highly effective in detecting fusion transcripts
gene.210 It has also been shown that the presence of in RNA extracted from formalin-fixed paraffin-embedded
newly identified hybrid genes such as BCOR-CCNB3 can tissue. Sequencing of RT-PCR fusion transcripts pro-
be suggested immunohistochemically by the strong over- vides confirmation of the specificity of the test by iden-
expression of CCNB3 protein.44,47 Morphoproteomic tifying the sequences of the genes involved in the fusion.
profiling with multiple markers is being explored to Novel emerging approaches that are not yet widely
identify the upregulation of potentially therapeutic path- available are based on genome sequencing technologies
ways such as mTOR and Ras/Raf/ERK in this group of and are designed to identify all possible fusions using a
malignancies.206 panel of primers and massive genomic sequencing. Such
tests are capable of detecting all translocations in sarco-
mas and may dominate future molecular testing of sarco-
DIFFERENTIAL DIAGNOSIS OF SMALL mas. These technologies also offer the genome-wide
ROUND-CELL TUMORS OF BONE identification of mutations in therapeutically targetable
genes.
Diagnostic features specific for Ewing’s sarcoma/PNET Neuroblastoma typically presents in infancy and child-
tumors should be evident from the preceding descrip- hood.188,194,231 The majority of cases occur in children
tions of these entities. In general, the differential diagno- during the first 5 years of life. The tumor frequently
sis of small round-cell tumors of bone include not only metastasizes to bone, liver, lymph nodes, and skin. Meta-
the entities described in this chapter, but also a variety of static disease can be a presenting sign. The elevated level
mesenchymal and epithelial (primary and metastatic) of catecholamines and their metabolites in the urine is an
tumors that may occur in children, adolescents, and important, diagnostically helpful feature. Histologically,
adults. the presence of neuropil and ganglionic differentiation is
Ewing’s sarcoma and PNET must be differentiated in helpful in distinguishing the tumor from Ewing’s sarcoma
children and adolescents from metastatic neuroblastoma, and PNET, but in small biopsy specimens, neuroblas-
small-cell osteosarcoma, mesenchymal chondrosarcoma, toma can be indistinguishable from other small-cell
rhabdomyosarcoma, and hematopoietic malignan- tumors. Many neural markers (i.e., chromogranin and
cies.211,212,215,216,220 Rare examples of Ewing’s sarcoma and glial fibrillary acidic protein) that overlap with the neural
PNET in older patients require differentiation primarily differentiation of Ewing’s sarcoma and PNET can be
from metastatic small-cell carcinoma and lymphoma. In positive in all three types of lesions. Even glycogen-rich
addition, Ewing’s sarcoma/PNET tumors can also rarely forms of neuroblastoma that closely resemble Ewing’s
originate in the internal organs. It is recommended that sarcoma have been described.232 On the other hand,
rendering such a diagnosis in an unusual clinical setting neural differentiation in Ewing’s sarcoma and PNET
should be verified by molecular testing. can be confused with neuroblastoma.227 The marker
The diagnostic molecular verification of Ewing’s that seems to be extremely helpful is the MIC2 gene,
sarcoma may be accomplished using several technolo- the expression of which is notably absent in neuroblas-
gies.213,214,217,218,224,225,230 Conventional cytogenetics is still toma. Moreover, the typical t(11;22)(q24;q12) and the
being used to identify the chromosomal translocations, alternative EWSR1 gene translocations are absent in
but because it requires fresh tumor tissue and tissue neuroblastoma. The typical cytogenetic abnormalities of
culture, it has been largely replaced by the so-called neuroblastoma consist of chromosome 1 deletion distal

ERRNVPHGLFRVRUJ
810 11  Ewing’s Sarcoma and Related Tumors

A B

D
FIGURE 11-45  ■  Adamantinoma-like Ewing’s sarcoma. A and B, Plain radiograph and magnetic resonance image showing permeative
tumor diffusely involving the fibula with prominent cortical thickening. C, Clusters of epithelial cells resembling adamantinoma of
long bone. Inset, The presence of EWSR1/FLI1 chimeric gene documented by the EWSR1/FLI1 fluorescent in situ hybridization fusion
probe. Arrow indicates the fused hybridization signal. The presence of EWSR1/FLI1 chimeric gene was confirmed by the DNA
sequencing of the fusion transcript. D, High power photomicrograph of an epithelial cluster shown in C. E, Strong diffuse staining
for keratin in tumor cells. Insets, Desmosome (upper right) and tonofilaments (lower left) confirming epithelial differentiation in
these unusual tumors. (C, ×100; D, ×400; E, ×200; Insets, ×40,000) (C and D, hematoxylin-eosin.) (Modified with permission from Bridge
J, et al: Am J Surg Pathol 23:159–165, 1999.)

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 811

to 1p32 (present in 70% to 80% of these tumors). The lymphoma are entities that frequently involve the skele-
second consistent abnormality is the presence of homo- ton in patients older than age 40 years and can be diag-
geneously stained chromosomal regions and the presence nosed with the support of appropriate epithelial and
of double-minute chromosomes. These represent a mor- lymphoma marker studies. We believe that the diagnosis
phologic manifestation of the N-myc gene amplification. of Ewing’s sarcoma and PNET in an unusual clinical
From this description, the reader may get an impression setting such as older patients should be confirmed by
that the distinction between neuroblastoma and Ewing’s appropriate molecular studies.218
sarcoma is extremely difficult and often impossible. On When it is not clear whether the lesion in question is
the contrary, if the entire clinical presentation (i.e., the a primary soft tissue or bone tumor, several additional
presence of suprarenal mass, elevated levels of catechol- conditions, such as high-grade soft tissue myxoid chon-
amines, and pattern of skeletal involvement) is consid- drosarcoma and poorly differentiated synovial sarcoma,
ered, the majority of cases can be easily distinguished must also be ruled out. Occasionally, sclerosing epitheli-
from Ewing’s sarcoma. oid fibrosarcoma, malignant melanoma, and even vil­
Small cell osteosarcoma may be confused with Ewing’s lonodular tenosynovitis can mimic a small round-cell
sarcoma and PNET.211,215,219,223,226,228 We believe that the tumor.215,220
lesions showing microscopically direct bone production
by tumor cells and exhibiting immunohistochemical and REFERENCES
molecular characteristics of Ewing’s sarcoma, the pres-
1. Ewing J: Diffuse endothelioma of bone. Proc NY Pathol Soc 21:17–
ence of t(11;22)(q24;q12) or characteristic fusion tran- 24, 1921.
scripts, should be classified as Ewing’s sarcoma rather 2. Oberling CH: Le reticulo-sarcome et le reticulo endothelio-
than small cell osteosarcoma. Expression of galectin sarcoma de la moelle osseuse (sarcomas d’Ewing). Bull Cancer
GAL-1) may be useful in the differential diagnosis (Paris) 17:259–296, 1928.
of small cell osteosarcoma and Ewing’s sarcoma. It is Chromosomal Translocations
frequently expressed in osteoblastic tumors, including 3. Abaan OD, Levonson A, Khan O, et al: PTPL1 is a direct tran-
small cell osteosarcoma and is expressed in less than 10% scriptional target of EWS-FLI1 and modulates Ewing’s sarcoma
of Ewing’s sarcomas.195 tumorigenesis. Oncogene 24:2715–2722, 2005.
Mesenchymal chondrosarcoma can represent a differ- 4. Aman P: Fusion oncogenes in tumor development. Semin Cancer
Biol 15:236–243, 2005.
ential diagnostic problem if the cartilaginous component 5. Antonescu CR, Dal Cin P: Promiscuous genes involved in recur-
is not present in the biopsy specimen or is inconspic- rent chromosomal translocations in soft tissue tumours. Pathology
uous. Microscopically, the presence of a prominent 46:105–112, 2014.
hemangiopericytoma-like pattern and foci of atypical 6. Aurias A, Rimbaut C, Buffe D, et al: Translocation involving chro-
cartilage that stain positive for S-100 protein are dis- mosome 22 in Ewing’s sarcoma: a cytogenetic study of four fresh
tumors. Cancer Genet Cytogenet 12:21–25, 1984.
tinguishing features.212,221,229 The expression of a master 7. Baud V, Lipinski M, Rassart E, et al: The human homolog of the
regulator of chondrogenesis, Sox9 protein, in primitive mouse common viral integration region, FLI-1, maps to 11q23-
mesechymal cells is consistent with their chondroprogen- q24. Genomics 11:223–224, 1991.
itor nature distinguishing mesenchymal chondrosarcoma 8. Ben-David Y, Giddens EB, Letwin K, et al: Erythroleukemia
induction by friend murine leukemia virus: insertional activation
from other small round-cell tumors.234 Identification of of a new member of the ETS gene family, FLI-1, closely linked to
two distinct chromosomal translocations, t(1;5)(q42;q32) c-ETS-1. Genes Dev 5:908–918, 1991.
with the IRF2PB2-CDX1 fusion gene and a cryptic inter- 9. Bhagirath T, Abe S, Nojima T, et al: Molecular analysis of a
stitial deletion of del(8)(q13.3q21.1) with the HEY1- t(11;22) translocation junction in a case of Ewing’s sarcoma. Genes
NCOA2 fusion gene, in mesenchymal chondrosarcoma Chromosomes Cancer 13:126–132, 1995.
10. Borden EC, Baker LH, Bell RS, et al: Soft tissue sarcomas of
provides new diagnostic tools in the differential diagnosis adults: state of the translational science. Clin Cancer Res 9:1941–
of small cell malignancies.222,233 1956, 2003.
Rhabdomyosarcoma of embryonal type, especially the alve- 11. Burchill SA: Ewing’s sarcoma: diagnostic, prognostic, and thera-
olar variant, belongs to the category of small round-cell peutic implications of molecular abnormalities. J Clin Pathol
56:96–102, 2003.
tumors and should be differentiated from Ewing’s 12. Cooper CD, Newman JA, Gileadi O: Recent advances in the
sarcoma, especially in soft tissue lesions found in young structural molecular biology of Ets transcription factors: interac-
children.215,220 The confusion between embryonal rhab- tions, interfaces and inhibition. Biochem Soc Trans 42:130–138,
domyosarcoma and Ewing’s sarcoma occurs most often 2014.
when analyzing frozen-section specimens. Examination 13. Delattre O, Zucman J, Melot T, et al: The Ewing family of
tumors—a subgroup of small-round-cell tumors defined by spe-
of paraffin-embedded material typically reveals rhabdo- cific chimeric transcripts. N Engl J Med 331:294–299, 1994.
myoblastic features in most embryonal rhabdomyosarco- 14. Delattre O, Zucman J, Plougastel B, et al: Gene fusion with an
mas. On the other hand, alveolar rhabdomyosarcoma is ETS DNA-binding domain caused by chromosome translocation
a true round-cell tumor and may present the diagnostic in human tumours. Nature 359:162–165, 1992.
15. Deng FM, Galvan K, de la Roza G, et al: Molecular characteriza-
problem. Positivity of the tumor cells for muscle markers tion of an EWSR1-POU5F1 fusion associated with a t(6;22) in an
and the presence of nesting and alveolar patterns are undifferentiated soft tissue sarcoma. Cancer Genet 204:423–429,
distinguishing features. The presence of t(2;13)(q34;q14) 2011.
or t(1;13)(p36;q14) and their respective FKHR-PAX3 and 16. Dormann D, Haass C: Fused in sarcoma (FUS): an oncogene
FKHR-PAX7 fusion genes are the molecular characteris- goes awry in neurodegeneration. Mol Cell Neurosci 56:475–486,
2013.
tics of alveolar rhabdomyosarcoma. 17. Douglass EC: Chromosomal rearrangements in Ewing’s sarcoma
Small round-cell tumors in older patients are very unlikely and peripheral neuroectodermal tumor (PNET). Semin Devel Biol
to represent Ewing’s sarcoma. Metastatic carcinoma and 1:393–396, 1990.

ERRNVPHGLFRVRUJ
812 11  Ewing’s Sarcoma and Related Tumors

18. Downing JR, Head DR, Parham DM, et al: Detection of the 42. Oliveira AM, Fletcher CD: Molecular prognostication for soft
(11;22)(q24;q12) translocation of Ewing’s sarcoma and peripheral tissue sarcomas: are we ready yet? J Clin Oncol 22:4031–4034,
neuroectodermal tumor by reverse transcription polymerase chain 2004.
reaction. Am J Pathol 143:1294–1300, 1993. 43. Pellin A, Boix J, Blesa JR, et al: EWS/FLI-1 rearrangement in
19. Dunn T, Praissman L, Hagag N, et al: ERG gene is translocated small round cell sarcomas of bone and soft tissue detected by
in an Ewing’s sarcoma cell line. Cancer Genet Cytogenet 76:19–22, reverse transcriptase polymerase chain reaction amplification. Eur
1994. J Cancer 30A:827–831, 1994.
20. Feng FY, Brenner JC, Hussain M, et al: Molecular path ways: 44. Pierron G, Tirode F, Lucchesi C, et al: A new subtype of bone
targeting ETS gene fusions in cancer. Clin Cancer Res 20:4442– sarcoma defined by BCOR-CCNB3 gene fusion. Nat Genet 44:461–
4448, 2014. 466, 2012.
21. Frohling S, Dohner H: Molecular origins of cancer: chromo- 45. Plougastel B, Zucman J, Peter M, et al: Genomic structure
somal abnormalities in cancer. N Engl J Med 359:722–734, of the EWS gene and its relationship to EWSR1, a site of
2008. tumor-associated chromosome translocation. Genomics 18:609–
22. Helman LJ, Meltzer P: Mechanisms of sarcoma development. Nat 615, 1993.
Rev Cancer 3:685–694, 2003. 46. Prasad DD, Rao VN, Reddy ES: Structure and expression of
23. Hu-Lieskovan S, Zhang J, Wu L, et al: EWS-FLI1 fusion protein human FLI-1 gene. Cancer Res 52:5833–5837, 1992.
up-regulates critical genes in neural crest development and is 47. Puls F, Niblett A, Marland G, et al: BCOR-CCNB3 (Ewing-like)
responsible for the observed phenotype of Ewing’s family of sarcoma: a clinicopathologic analysis of 10 cases, in comparison
tumors. Cancer Res 65:4633–4644, 2005. with conventional Ewing sarcoma. Am J Surg Pathol 38:1307–
24. Irifune H, Nishimori H, Watanabe G, et al: Aberrant laminin β3 1318, 2014.
isoforms downstream of EWS-ETS fusion genes in Ewing family 48. Riccardi GF, Stein C, de la Roza G, et al: Newly described trans-
tumors. Cancer Biol Ther 4:449–455, 2005. location (18;19)(q23;q13.2) in abdominal wall soft-tissue tumor
25. Italiano A, Sung YS, Zhang L, et al: High prevalence of CIC resembling Ewing sarcoma/primitive neuroectodermal tumor.
fusion with double-homeobox (DUX4) transcription factors in Cancer Genet Cytogenet 201:1–5, 2010.
EWSR1-negative undifferentiated small blue round cell sarcomas. 49. Rougemont AL, Bouron-Dal Soglio D, Patey-Mariaud de Serre
Genes Chromosomes Cancer 51:207–218, 2012. N, et al: A t(17;22)(q21;q12) with partial ETV4 deletion in a soft
26. Janknecht R: EWS-ETS oncoproteins: the linchpins of Ewing tissue Ewing sarcoma. Cancer Genet 205:55–60, 2012.
tumors. Gene 363:1–14, 2005. 50. Sadri N, Barroeta J, Pack SD, et al: Malignant round cell tumor
27. Jeon I-S, Davis JN, Braun BS, et al: A variant Ewing’s sarcoma of bone with EWSR1-NFATC2 gene fusion. Virchows Arch 465:
translocation (7;22) fuses the EWS gene to the ETS gene ETV-1. 233–239, 2014.
Oncogene 10:1229–1234, 1995. 51. Sandberg AA, Bridge JA: Updates on cytogenetics and molecular
28. Jinawath N, Morsberger L, Norris-Kirby A, et al: Complex rear- genetics of bone and soft tissue tumors: Ewing sarcoma and
rangement of chromosomes 1, 7, 21, 22 in Ewing sarcoma. Cancer peripheral primitive neuroectodermal tumors. Cancer Genet Cyto-
Genet Cytogenet 201:42–47, 2010. genet 123:1–26, 2000.
29. Kar A, Gutierrez-Hartmann A: Molecular mechanisms of ETS 52. Sankar S, Lessnick SL: Promiscuous partnerships in Ewing’s
transcription factor-mediated tumorigenesis. Crit Rev Biochem Mol sarcoma. Cancer Genet 204:351–365, 2011.
Biol 48:522–543, 2013. 53. Smith SC, Buehler D, Choi EYK, et al: CIC-DUX sarcomas dem-
30. Kawamura-Saito M, Yamazaki Y, Kaneko K, et al: Fusion between onstrate frequent MYC amplification and ETS-family transcrip-
CIC and DUX4 up-regulates PEA3 family genes in Ewing-like tion factor expression. Mod Pathol 1–12:2014.
sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet 54. Sorensen PH, Lessnick SL, Lopez-Terrada D, et al: A second
15:2125–2137, 2006. Ewing’s sarcoma translocation, t(21;22), fuses the EWS gene to
31. Khoury JD: Ewing sarcoma family of tumors. Adv Anat Pathol another ETS-family transcription factor, ERG. Nat Genet 6:146–
12:212–220, 2005. 151, 1994.
32. Lazar A, Abruzzo LV, Pollock RE, et al: Molecular diagnosis of 55. Sorensen PH, Liu XF, Delattre O, et al: Reverse transcriptase
sarcomas: chromosomal translocations in sarcomas. Arch Pathol PCR amplification of EWS/FLI-1 fusion transcripts as a diagnos-
Lab Med 130:1199–1207, 2006. tic test for peripheral primitive neuroectodermal tumors of child-
33. Li X, Tanaka K, Nakatani F, et al: Transactivation of cyclin E gene hood. Diagn Mol Pathol 2:147–157, 1993.
by EWS-Fli1 and antitumor effects of cyclin dependent kinase 56. Spahn L, Siligan C, Bachmaier R, et al: Homotypic and hetero-
inhibitor on Ewing’s family tumor cells. Int J Cancer 116:385–394, typic interactions of EWS, FLI1 and their oncogenic fusion
2005. protein. Oncogene 22:6819–6829, 2003.
34. Lopez-Terrada D: Molecular genetics of small round cell tumors. 57. Specht K, Sung YS, Zhang L, et al: Distinct transcriptional sig-
Semin Diagn Pathol 13:242–249, 1996. nature and immunoprofile of CIC-DUX4 fusion-positive round
35. Machado I, Noguera R, Mateos EA, et al: The many faces of cell tumors compared to EWSR1-rearranged Ewing sarcomas:
atypical Ewing’s sarcoma. A true entity mimicking sarcomas, car- further evidence toward distinct pathologic entities. Genes Chro-
cinomas and lymphomas. Virchows Arch 458:281–290, 2011. mosomes Cancer 53:622–633, 2014.
36. Maire G, Brown CW, Bayani J, et al: Complex rearrangement of 58. Stephenson CF, Bridge JA, Sandberg AA: Cytogenetic and patho-
chromosomes 19, 21, and 22 in Ewing sarcoma involving a novel logic aspects of Ewing’s sarcoma and neuroectodermal tumors.
reciprocal inversion-insertion mechanism of EWS-ERG fusion Hum Pathol 23:1270–1277, 1992.
gene formation: a case analysis and literature review. Cancer Genet 59. Sugita S, Arai Y, Tonooka A, et al: A novel CIC-FOXO4 gene
Cytogenet 181:81–92, 2008. fusion in undifferentiated small round cell sarcoma: a genetically
37. Mangham DC, Williams A, McMullan DJ, et al: Ewing’s sarcoma distinct variant of Ewing-like sarcoma. Am J Surg Pathol 38:1571–
of bone: the detection of specific transcripts in a large, consecutive 1576, 2014.
series of formalin-fixed, decalcified, paraffin-embedded tissue 60. Sumegi J, Nishio J, Nelson M, et al: A novel t(4;22)(q31;q12)
samples using the reverse transcriptase-polymerase chain reaction. produces an EWSR1-SMARCA5 fusion in extraskeletal Ewing
Histopathology 48:363–376, 2006. sarcoma/primitive neuroectodermal tumor. Mod Pathol 24:333–
38. May WA, Gishizky ML, Lessnick SL, et al: Ewing sarcoma 11;22 342, 2011.
translocation produces a chimeric transcription factor that 61. Szuhai K, Ijszenga M, de Jong D, et al: The NFATc2 gene is
requires the DNA-binding domain encoded by FLI1 for transfor- involved in a novel cloned translocation in a Ewing sarcoma
mation. Proc Natl Acad Sci USA 90:5752–5756, 1993. variant that couples its function in immunology to oncology. Clin
39. Moore DD, Haydon RC: Ewing’s sarcoma of bone. Cancer Treat Cancer Res 15:2259–2268, 2009.
Res 162:93–115, 2014. 62. Terrier-Lacombe MJ, Guillou L, Chibon F, et al: Superficial
40. Nakatani F, Tanaka K, Sakimura R, et al: Identification of primitive Ewing’s sarcoma: a clinicopathologic and molecular
p21WAF1/CIP1 as a direct target of EWS-Fli1 oncogenic fusion cytogenetic analysis of 14 cases. Mod Pathol 22:87–94, 2009.
protein. J Biol Chem 278:15105–15115, 2003. 63. Thway K, Fisher C: Tumors with EWSR1-CREB1 and EWSR1-
41. Oikawa T, Yamada T: Molecular biology of the Ets family of ATF1 fusions: the current status. Am J Surg Pathol 36:e1–e11,
transcription factors. Gene 303:11–34, 2003. 2012.

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 813

64. Turc-Carel C, Aurias A, Mugneret F, et al: Chromosomes in 87. Marino-Enriquez A, Fletcher CD: Round cell sarcomas—
Ewing’s sarcoma. I. An evaluation of 85 cases and remarkable biologically important refinements in subclassification. Int J
consistency of t(11;22)(q24;q12). Cancer Genet Cytogenet 32:229– Biochem Cell Biol 53:493–504, 2014.
238, 1988. 88. Nakajima H, Sim FH, Bond JR, et al: Small cell osteosarcoma of
65. Turc-Carel C, Philip I, Berger M-P, et al: Chromosome study of bone. Review of 72 cases. Cancer 79:2095–2106, 1997.
Ewing’s sarcoma (ES) cell lines: consistency of a reciprocal trans- 89. Nesbitt KA, Vidone RA: Primitive neuroectodermal tumor (neu-
location t(11;22)(q24;q12). Cancer Genet Cytogenet 12:1–19, 1984. roblastoma) arising in the sciatic nerve of a child. Cancer 37:1562–
66. Wang L, Bhargava R, Zheng T, et al: Undifferentiated small 1570, 1976.
round cell sarcomas with rare EWS gene fusions: identification of 90. Noguera R, Navarro S, Triche TJ: Translocation (11;22) in small
a novel EWS-SP3 fusion and of additional cases with the EWS- cell osteosarcoma. Cancer Genet Cytogenet 45:121–124, 1990.
ETV1 and EWS-FEV fusions. J Mol Diagn 9:498–509, 2007. 91. Pearson JM, Harris M, Eyden BD, et al: Divergent differentiation
67. Whang-Peng J, Triche TJ, Knutsen T, et al: Cytogenetic charac- in small round-cell tumors of the soft tissue with neural features:
terization of selected round cell tumors of childhood. Cancer Genet an analysis of 10 cases. Histopathology 23:1–9, 1993.
Cytogenet 21:185–208, 1986. 92. Roessner A: Jürgens H: Round cell tumours of bone. Pathol Res
68. Xia SJ, Barr FG: Chromosome translocations in sarcomas and the Pract 189:111–136, 1993.
emergence of oncogenic transcription factors. Eur J Cancer 41: 93. Seemayer TA, Vekemans M, de Chadarevian JP: Histological and
2513–2527, 2005. cytogenetic findings in a malignant tumor of the chest wall and
69. Zucman J, Delattre O, Desmaze C, et al: Cloning and character- lung (Askin tumor). Virchows Arch A Pathol Anat Histopathol
ization of the Ewing’s sarcoma and peripheral neuroepithelioma 408:289–296, 1985.
t(11;22) translocation breakpoints. Genes Chromosomes Cancer 94. Sirivella S, Gielchinsky I: Treatment outcomes in 23 thoracic
5:271–277, 1992. primitive neuroectodermal tumours: a retrospective study. Interact
70. Zucman J, Melot T, Desmaze C, et al: Combinational generation Cardiovasc Thorac Surg 17:273–279, 2013.
of variable fusion proteins in the Ewing family of tumours. EMBO 95. Sorensen PH, Shimada H, Liu XF, et al: Biphenotypic sarcomas
J 12:4481–4487, 1993. with myogenic and neural differentiation express the Ewing’s
sarcoma EWS/FLI1 fusion gene. Cancer Res 55:1385–1392, 1995.
Unified Pathogenetic Concept of Ewing’s Sarcoma 96. Stout AP: A tumor of the ulnar nerve. Proc NY Pathol Soc 12:2–12,
and Related Tumors 1918.
71. Askin FB, Rosai J, Sibley RK, et al: Malignant small cell tumor of 97. Stout AP, Murray MR: Neuroepithelioma of the radial nerve with
the thoracopulmonary region in childhood: a distinctive clinico- study of its behavior in vitro. Rev Cancer Biol 1:651–659, 1942.
pathological entity of uncertain histogenesis. Cancer 43:2438– 98. Tefft M, Vawter GF, Mitus A: Paravertebral “round cell” tumors
2451, 1979. in children. Radiology 92:1501–1509, 1969.
72. Basharkhah A, Pansy J, Urban C, et al: Outcomes after interdis- 99. Tsokos M, Miser J, Horowitz M, et al: Primitive round cell
ciplinary management of 7 patients with Askin tumor. Pediatr Surg sarcoma of bone: a group of tumors resembling Ewing’s sarcoma
Int 29:431–436, 2013. by light microscopy, but with distinctive ultrastructural appear-
73. Bishop JA, Shum CH, Sheth S, et al: Small cell osteosarcoma: ance. Lab Invest 96A:570, 1993.
cytopathologic characteristics and differential diagnosis. Am J 100. van Haelst UJ, de Haas van Dorsser AH: A perplexing malignant
Clin Pathol 133:756–761, 2010. bone tumor. Highly malignant so-called adamantinoma or non-
74. Bridge JA, Fidler ME, Neff JR, et al: Adamantinoma-like Ewing’s typical Ewing’s sarcoma. Virchows Arch A Pathol Anat Histol
sarcoma: genomic confirmation, phenotypic drift. Am J Surg 365:63–74, 1975.
Pathol 23:159–165, 1999.
75. Bridge JA, Fidler ME, Neff JR, et al: Adamantinoma-like Ewing’s Ewing’s Sarcoma
sarcoma: genomic confirmation, phenotypic drift. Am J Surg 101. Ahrens S, Jabar S, Heinecke A, et al: Prognostic significance of
Pathol 24:322–323, 2000. tumor volume in a volume adapted treatment strategy for local-
76. Debelenko LV, McGregor LM, Shivakumar BR, et al: A novel ized Ewing’s sarcoma of bone. Med Pediatr Oncol 23(221):P130,
EWSR1-CREB3L1 fusion transcript in a case of small cell osteo- 1994.
sarcoma. Genes Chromosomes Cancer 50:1054–1062, 2011. 102. Bacci G, Picci P, Gitelis S, et al: The treatment of localized
77. Delattre O, Zucman J, Melot T, et al: The Ewing family of Ewing’s sarcoma: the experience of the Instituto Ortopedico
tumors: a subgroup of small round cell tumors defined by specific Rizzoli in 163 cases treated with and without adjuvant chemo-
chimeric transcripts. N Engl J Med 331:294–299, 1994. therapy. Cancer 49:1561–1570, 1982.
78. Dragoescu E, Jackson-Cook C, Domson G, et al: Small cell osteo- 103. Bacci G, Toni A, Avella M, et al: Long-term results in 144 local-
sarcoma with Ewing sarcoma breakpoint region 1 gene rearrange- ized Ewing’s sarcoma patients treated with combined therapy.
ment detected by interphase fluorescence in situ hybridization. Cancer 63:1477–1486, 1989.
Ann Diagn Pathol 17:377–382, 2013. 104. Biswas B, Rastogi S, Khan SA, et al: Outcomes and prognostic
79. Edeiken J, Raymond AK, Ayala AG, et al: Small-cell osteosar- factors for Ewing-family tumors of the extremities. J Bone Joint
coma. Skeletal Radiol 16:621–628, 1987. Surg 96A:841–849, 2014.
80. Frydman CP, Klein MJ, Abdelwahab IF, et al: Primitive multipo- 105. Biswas B, Rastogi S, Khan SA, et al: Developing a prognostic
tential primary sarcoma of bone: a case report and immunohisto- model for localized Ewing sarcoma family of tumors: a single
chemical study. Mod Pathol 6:768–772, 1991. institutional experience of 224 cases treated with uniform chemo-
81. Hauben E, van den Broek LC, Van Marck E, et al: Adamantinoma- therapy protocol. J Surg Oncol 2014. [epub ahead of print].
like Ewing’s sarcoma and Ewing’s-like adamantinoma. The 106. Brisset S, Schleiermacher G, Peter M, et al: CGH analysis of
t(11;22), t(21;22) status. J Pathol 195:218–221, 2001. secondary genetic changes in Ewing tumors: correlation with
82. Hutter RV, Foote FW, Jr, Francis KC, et al: Primitive multipo- metastatic disease in a series of 43 cases. Cancer Genet Cytogenet
tential primary sarcoma of bone. Cancer 19:1–25, 1966. 130:57–61, 2001.
83. Kawamoto EH, Weidner N, Agostini RM, Jr, et al: Malignant 107. Brohl AS, Solomon DA, Chang W, et al: The genomic landscape
ectomesenchymoma of soft tissue: report of two cases and review of the Ewing sarcoma family of tumors reveals recurrent STAG2
of the literature. Cancer 59:1791–1802, 1987. mutation. PLoS Genet 10:e1004475, 2014.
84. Kikuchi Y, Kishimoto T, Ota S, et al: Adamantinoma-like Ewing 108. Callaghan RC, Carda C, Peydro-Olaya A, et al: Small round cell
family tumor of soft tissue associated with the vagus nerve: a case tumors of bone and soft tissue: a morphometric and stereometric
report and review of the literature. Am J Surg Pathol 37:772–779, comparative analysis of 119 cases. Anal Quant Cytol Histol 17:374–
2013. 382, 1995.
85. Ladanyi M: The emerging molecular genetics of sarcoma trans- 109. Cangir A, Vietti TJ, Gehan EA, et al: Ewing’s sarcoma metastatic
locations. Diagn Mol Pathol 4:162–173, 1995. at diagnosis: results and comparisons of two intergroup Ewing’s
86. Lezcano C, Clarke MR, Zhang L, et al: Adamantinoma-like sarcoma studies. Cancer 66:887–893, 1990.
Ewing sarcoma mimicking basal cell adenocarcinoma of the 110. Crompton BD, Stewart C, Taylor-Weiner A, et al: The genomic
parotid gland: a case report and review of the literature. Head Neck landscape of pediatric Ewing sarcoma. Cancer Discov 4:1326–1341,
Pathol 2014. [epub ahead of print]. 2014.

ERRNVPHGLFRVRUJ
814 11  Ewing’s Sarcoma and Related Tumors

111. Daugaard S, Kamby C, Sunde LM, et al: Ewing’s sarcoma: a 135. Nesbit ME, Jr, Gehan EA, Burgert EO, Jr, et al: Multimodal
retrospective study of histological and immunohistochemical therapy for the management of primary, non-metastatic Ewing’s
factors and their relation to prognosis. Virchows Arch A Pathol Anat sarcoma of bone: a long-term follow-up of the First Intergroup
Histopathol 414:243–251, 1989. Study. J Clin Oncol 8:1664–1674, 1990.
112. de Alava E, Kawai A, Healey JH, et al: EWS-FLI1 fusion tran- 136. Oberlin O, Patte C, Demeocq F, et al: The response to initial
script structure is an indepdent determinant of prognosis in chemotherapy as a prognostic factor in localized Ewing’s sarcoma.
Ewing’s sarcoma. J Clin Oncol 16:1248–1255, 1998. Eur J Cancer Clin Oncol 21:463–467, 1985.
113. de Stefani E, Carzoglio J, Deneo Pellegrini H, et al: Ewing’s 137. Oberlin O, Zucker JM, Brunat-Mentigny M: Prognostic factors
sarcoma: value of tumor necrosis as a predictive factor. Bull Cancer in localized Ewing’s sarcoma. a study of the French Society of
(Paris) 71:16–21, 1984. Pediatric Oncology (SFOP). Med Pediatr Oncol 23:573, 1993.
114. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210, 138. Orr WS, Denbo JW, Billups CA, et al: Analysis of prognostic
1995. factors in extraosseous Ewing sarcoma family of tumors: review
115. Fizazi K, Dohollou N, Spielmann M, et al: Adults with Ewing’s of St. Jude Children’s Research Hospital experience. Ann Surg
sarcoma: a retrospective study of 146 cases. Eur J Cancer 31A:250S, Oncol 19:3816–3822, 2012.
1995. 139. Picci P, Rougraff BT, Bacci G, et al: Prognostic significance of
116. Frostad B, Tani E, Brosjo O, et al: Fine needle aspiration cytology histopathologic response to chemotherapy in nonmetastatic
in the diagnosis and management of children and adolescents with Ewing’s sarcoma of the extremities. J Clin Oncol 11:1763–1769,
Ewing sarcoma and peripheral primitive neuroectodermal tumor. 1993.
Med Pediatr Oncol 38:33–40, 2002. 140. Roessner A, Ueda Y, Bockhorn-Dworniczak B, et al: Prognostic
117. Glaubiger DL, Makuch R, Schwarz J, et al: Determination of implication of immuno-detection of P-glycoprotein in Ewing’s
prognostic factors and their influence on therapeutic results in sarcoma. J Cancer Res Clin Oncol 119:185–189, 1993.
patients with Ewing’s sarcoma. Cancer 45:2213–2219, 1980. 141. Rosen G, Caparros B, Nirenberg A, et al: Ewing’s sarcoma: ten-
118. Hamilton SN, Carlson R, Hasan H, et al: Long-term outcomes year experience with adjuvant chemotherapy. Cancer 47:2204–
and complications in pediatric Ewing sarcoma. Am J Clin Oncol 2213, 1981.
2015. [epub ahead of print]. 142. Rosenberg AE, Amstalden EMI, Nielsen GP, et al: Ewing’s
119. Hartman KR, Triche TJ, Kinsella TJ, et al: Prognostic value of sarcoma/PNET: preoperative chemotherapy induced necrosis and
histopathology in Ewing’s sarcoma: long-term follow-up of distal outcome, United States and Canadian Academy of Pathology,
extremity primary tumors. Cancer 67:163–171, 1991. annual meeting. Lab Invest 74(13A):53, 1996.
120. Hayes FA, Thompson EI, Meyer WH, et al: Therapy for localized 143. Sailer SL, Harmon DC, Mankin HJ, et al: Ewing’s sarcoma: surgi-
Ewing’s sarcoma of bone. J Clin Oncol 7:208–213, 1989. cal resection as a prognostic factor. Int J Radiat Oncol Biol Phys
121. Hayes FA, Thompson EI, Parvey L, et al: Metastatic Ewing’s 15:43–52, 1988.
sarcoma: remission induction and survival. J Clin Oncol 5:1199– 144. Schoedel K, Dickman PS, Krailo M: Histologic response to che-
1204, 1987. motherapy and prognosis in osseous Ewing’s sarcoma/primitive
122. Hutington RW, Henkelmann CR, Franklin R: Atypical Ewing’s neuroectodermal tumor (ES/PNET), Twentieth International
tumor in the sister of a patient with Ewing’s tumor. J Bone Joint Congress of the International Academy of Pathology. Int J Pathol
Surg 4A:572–574, 1961. 2S:443, 1995.
123. Jurgens H: Ewing’s sarcoma and peripheral primitive neuroecto- 145. Soule EH, Newton W, Jr, Moon TE, et al: Extraskeletal Ewing’s
dermal tumor. Curr Opin Oncol 6:391–396, 1994. sarcoma: a preliminary review of 26 cases encountered in the
124. Jurgens H, Exner U, Gadner H, et al: Multidisciplinary treatment Intergroup Rhabdomyosarcoma Study. Cancer 42:259–264, 1978.
of primary Ewing’s sarcoma of bone: a 6-year experience of a 146. Terrier P, Llombart-Bosch A, Contesso G: Small round blue cell
European Cooperative Trial. Cancer 61:23–32, 1988. tumors in bone: prognostic factors correlated to Ewing’s sarcoma
125. Kinsella TJ, Miser JS, Waller B, et al: Long-term follow-up of and neuroectodermal tumor. Semin Diagn Pathol 13:250–257,
Ewing’s sarcoma of bone treated with combined modality therapy. 1996.
Int J Radiat Oncol Biol Phys 20:389–395, 1991. 147. Tirode F, Surdez D, Ma X, et al: Genomic landscape of Ewing
126. Kiss R, Larsimont D, Huvos AG: Digital cell image analysis of sarcoma defines an aggressive subtype with co-association of
Ewing’s sarcoma. Anal Quant Cytol Histol 13:356–362, 1991. STAG2 and TP53 mutations.Cancer Discov 4:1342–1353, 2014.
127. Kissane JM, Askin FB, Foulkes M, et al: Ewing’s sarcoma of bone: 148. Udayakumar AM, Sundareshan TS, Goud TM, et al: Cytogenetic
clinicopathologic aspects of 303 cases from the Intergroup Ewing’s characterization of Ewing tumors using fine needle aspiration
Sarcoma Study. Hum Pathol 14:773–779, 1983. samples. A 10-year experience and review of the literature. Cancer
128. Llombart-Bosch A, Blache R, Peydro-Olaya A: Round cell sarco- Genet Cytogenet 127:42–48, 2001.
mas of bone and their differential diagnosis (with particular 149. Wilkins RM, Pritchard DJ, Burgert EO, Jr, et al: Ewing’s sarcoma
emphasis on Ewing’s sarcoma and reticulosarcoma): a study of 233 of bone: experience with 140 patients. Cancer 58:2551–2555, 1986.
tumors with optical and electron microscopic techniques. Pathol 150. Zoubek A, Dockhorn-Dworniczak B, Delattre O, et al: Does
Annu 17:113–145, 1982. expression of different EWS chimeric transcripts define clinically
129. Llombart-Bosch A, Contesso G, Henry-Amar M, et al: Histo- distinct risk groups of Ewing tumor patients? J Clin Oncol
pathological predictive factors in Ewing’s sarcoma of bone 14:1245–1251, 1996.
and clinicopathological correlations: a retrospective study of 151. Zucker JM, Henry-Amar M, Sarrazin D, et al: Intensive systemic
261 cases. Virchows Arch A Pathol Anat Histopathol 409:627–640, chemotherapy in localized Ewing’s sarcoma in childhood: a his-
1986. torical trial. Cancer 52:415–423, 1983.
130. Llombart-Bosch A, Contesso G, Peydro-Olaya A: Histology,
immunohistochemistry, and electron microscopy of small round Primitive Neuroectodermal Tumor
cell tumors of bone. Semin Diagn Pathol 13:153–170, 1996. 152. Contesso G, Llombart-Bosch A, Terrier P, et al: Does malignant
131. Llombart-Bosch A, Peydro-Olaya A: Scanning and transmission small round cell tumor of the thoracopulmonary region (Askin
electron microscopy of Ewing’s sarcoma of bone: typical and tumor) constitute a clinicopathologic entity? An analysis of 30
atypical variants—an analysis of nine cases. Virchows Arch A Pathol cases with immunohistochemical and electron-microscopic
Anat Histopathol 398:329–346, 1983. support treated at the Institute Gustave Roussy. Cancer 69:1012–
132. Mamegham H, Fisher RJ, O’Gorman-Hughes D, et al: Ewing’s 1020, 1992.
sarcoma: long-term follow-up in 49 patients treated from 1967 to 153. Hasegawa T, Hirose T, Kudo E, et al: Atypical primitive neuro-
1989. Int J Radiat Oncol Biol Phys 25:431–438, 1993. ectodermal tumors: comparative light and electron microscopic
133. Nascimento AG, Unni KK, Pritchard DJ, et al: A clinico- and immunohistochemical studies on peripheral neuroepitheli-
pathologic study of 20 cases of large-cell (atypical) Ewing’s oma and Ewing’s sarcoma. Acta Pathol Jpn 41:444–454, 1991.
sarcoma of bone. Am J Surg Pathol 4:29–36, 1980. 154. Hashimoto H, Enjoji M, Nakajima T, et al: Malignant neuroepi-
134. Navarro S, Gonzalez-Devesa M, Ferrandez-Izquierdo A, et al: thelioma (peripheral neuroblastoma): a clinicopathologic study of
Scanning electron microscopic evidence for neural differentiation 15 cases. Am J Surg Pathol 7:309–318, 1983.
in Ewing’s sarcoma cell lines. Virchows Arch A Pathol Anat Histo- 155. Hijazi Y, Tsokos M, Steinberg S, et al: Neuroectodermal differ-
pathol 416:383–391, 1990. entiation does not play a role in the prognosis of Ewing’s sarcoma

ERRNVPHGLFRVRUJ
11  Ewing’s Sarcoma and Related Tumors 815

(ES) versus primitive neuroectodermal tumor (PNET), United 176. Caillaud JM, Benjelloun S, Bosq J, et al: HNK-1–defined antigen
States and Canadian Academy of Pathology, annual meeting. Lab detected in paraffin-embedded neuroectodermal tumors and those
Invest 74(7A):21, 1996. derived from cells of the amino precursor uptake and decarboxyl-
156. Jaffe R, Santamaria M, Yunis EJ, et al: The neuroectodermal ation system. Cancer Res 44:4432–4439, 1984.
tumor of bone. Am J Surg Pathol 8:885–898, 1984. 177. Carter RL, al-Sams SZ, Corbett RP, et al: A comparative study
157. Kushner BH, Hajdu SI, Gulati SC, et al: Extracranial primitive of immunohistochemical staining for neuron-specific enolase,
neuroectodermal tumors: the Memorial Sloan-Kettering Cancer protein gene product 9.5 and S-100 protein in neuroblastoma,
Center experience. Cancer 67:1825–1829, 1991. Ewing’s sarcoma and other round cell tumours in children. Histo-
158. Landanyi M, Heinemann FS, Huvos AG, et al: Neural differentia- pathology 16:461–467, 1990.
tion in small round cell tumors of bone and soft tissue with the 178. Donovan MJ, Hempstead BL, Horvath C, et al: Immunohisto-
translocation t(11;22)(q24;12): an immunohistochemical study of chemical localization of Trk receptor protein in pediatric small
11 cases. Hum Pathol 21:1245–1251, 1990. round blue cell tumors. Am J Pathol 143:1560–1567, 1993.
159. Llombart-Bosch A, Lacombe MJ, Contesso G, et al: Small round 179. Fellinger EJ, Garin-Chesa P, Triche TJ, et al: Immunohistochem-
blue cell sarcoma of bone mimicking atypical Ewing’s sarcoma ical analysis of Ewing’s sarcoma cell surface antigen p30/32MIC2.
with neuroectodermal features: an analysis of five cases with Am J Pathol 139:317–325, 1991.
immunohistochemical and electron microscopic support. Cancer 180. Fernandez-Flores A, Suarez-Penaranda JM, Alonso S: Study
60:1570–1582, 1987. of EWS/FLI1 rearrangement in 18 cases of CK20+/CM2B4
160. Llombart-Bosch A, Lacombe JM, Peydro-Olaya A, et al: Malig- Merkel cell carcinoma using FISH and correlation to the
nant peripheral neuroectodermal tumors of bone other than Askin differential diagnosis of Ewing sarcoma/peripheral neuroecto-
neoplasms: characterization of 14 new cases with immunohisto- dermal tumor. Appl Immunohistochem Mol Morphol 21:379–385,
chemistry and electron microscopy. Virchows Arch A Pathol Anat 2013.
Histopathol 412:421–430, 1988. 181. Folpe AL, Goldblum JR, Rubin BP, et al: Morphologic and immu-
161. Papierz W, Alwasiak J, Kolasa P, et al: Primitive neuroectodermal nophenotypic diversity in Ewing’s family tumors: a study of 66
tumors: ultrastructural and immunohistochemical studies. Ultra- genetically confirmed cases. Am J Surg Pathol 29:1025–1033,
struct Pathol 19:147–166, 1995. 2005.
162. Schmidt D, Herrmann C, Jurgens H, et al: Malignant peripheral 182. Gelin C, Aubrit F, Phalipon A, et al: The E2 antigen, a 32kd
neuroectodermal tumor and its necessary distinction from Ewing’s glycoprotein involved in T-cell adhesion processes, is the MIC2
sarcoma: a report from the Kiel Pediatric Tumor Registry. Cancer gene product. EMBO J 8:3253–3259, 1989.
68:2251–2259, 1991. 183. Goodfellow P, Banting G, Sheer D, et al: Genetic evidence that a
163. Terrier P, Henry-Amar M, Triche TJ, et al: Is neuroectodermal Y-linked gene in man is homologous to a gene on the X chromo-
differentiation of Ewing’s sarcoma of bone associated with an some. Nature 302:346–349, 1983.
unfavorable prognosis? Eur J Cancer 31A:307–314, 1995. 184. Hamilton G, Fellinger EJ, Schratter I, et al: Characterization of
164. Ushigome S, Shimoda T, Takaki K, et al: Immunocytochemical a human endocrine tissue and tumor-associated Ewing’s sarcoma
and ultrastructural studies of the histogenesis of Ewing’s sarcoma antigen. Cancer Res 48:6127–6131, 1988.
and putatively related tumors. Cancer 64:52–62, 1989. 185. Harris MD, Moore IE, Steart PV, et al: Protein gene product
165. Whang-Peng J, Triche TJ, Knutsen T, et al: Chromosome trans- (PGP) 9.5 as a reliable marker in primitive neuroectodermal
location in peripheral neuroepithelioma. N Engl J Med 311:584– tumours: an immunohistochemical study of 21 childhood cases.
585, 1984. Histopathology 16:271–277, 1990.
186. Hijazi YM, Axiotis CA, Navarro S, et al: Immunohistochemical
Askin’s Tumor detection of P-glyprotein in Ewing’s sarcoma and peripheral
166. Füzesi L, Heller R, Schreiber H, et al: Cytogenetics of Askin’s primitive neuroectodermal tumors before and after chemother-
tumor: case report and review of the literature. Pathol Res Pract apy. Am J Clin Pathol 102:61–67, 1994.
189:235–241, 1993. 187. Kawaguchi K, Koike M: Neuron-specific enolase and Leu-7
167. Laskar S, Nair C, Mallik S, et al: Prognostic factors and outcome immunoreactive small round cell neoplasms: the relationship to
in Askin-Rosai tumor: a review of 104 patients. Int J Radiat Oncol Ewing’s sarcoma in bone and soft tissues. Am J Clin Pathol 86:79–
Biol Phys 79:202–207, 2011. 83, 1986.
168. Linnoila RI, Tsokos M, Triche TJ, et al: Evidence for neural 188. Kiyonari S, Kadomatsu K: Neuroblastoma models for insights
origin and PAS-positive variants of the malignant small cell tumor into tumorigenesis and new therapies. Expert Opin Drug Discov
of thoracopulmonary region (“Askin tumor”). Am J Surg Pathol 10:53–62, 2015.
10:124–133, 1986. 189. Kreppel M, Aryee DN, Schaefer KL, et al: Suppression of
169. Liu Z, Zou W, Ma G, et al: Primitive chest wall neuroectodermal KCMF1 by constitutive high CD99 expression is involved in the
tumor in a pediatric patient. Interact Cardiovasc Thorac Surg 13: migratory ability of Ewing’s sarcoma cells. Oncogene 25:2795–
440–441, 2011. 2800, 2006.
170. Seemayer TA, Vekemans M, de Chadarevian JP: Histological and 190. Levy R, Dilley J, Fox RI, et al: A human thymus leukemia antigen
cytological findings in a malignant tumor of the chest wall and defined by hybridoma monoclonal antibodies. Proc Natl Acad Sci
lung (Askin tumor). Virchows Arch A Pathol Anat Histopathol USA 76:6552–6556, 1979.
408:289–296, 1985. 191. Li Q, Cui W, Abulajiang G, et al: Application of immunohisto-
171. Shamberger RC, Grier HE, Weinstein HJ, et al: Chest wall chemistry in the diagnosis of small round blue-cell tumors of soft
tumors in infancy and childhood. Cancer 63:774–785, 1989. tissue. Clin Lab 60:1383–1392, 2014.
172. van den Berg H, van Rijn RR, Merks JH: Management of tumors 192. Lipinski M, Braham K, Caillaud JM, et al: HNK-1 antibody
of the chest wall in childhood: a review. J Pediatr Hematol Oncol detects an antigen expressed on neuroectodermal cells. J Exp Med
30:214–221, 2008. 158:1775–1780, 1983.
173. Xu Q, Xu K, Yang C, et al: Askin tumor: four case reports 193. Lizard-Nacol S, Lizard G, Justrabo E, et al: Immunologic char-
and a review of the literature. Cancer Imaging 11:184–188, acterizaton of Ewing’s sarcoma using mesenchymal and neural
2011. markers. Am J Pathol 135:847–855, 1989.
194. Louis CU, Shohet JM: Neuroblastoma: molecular pathogenesis
Immunohistochemical Features of Ewing’s and therapy. Annu Rev Med 66:49–63, 2015.
Sarcoma and PNET 195. Machado I, Lopez-Guerrero JA, Navarro S, et al: Galectin-1
174. Ambros IM, Ambros PF, Strehl S, et al: MIC2 is a specific marker (GAL-1) expression is a useful tool to differentiate between small
for Ewing’s sarcoma and peripheral primitive neuroectodermal cell osteosarcoma and Ewing sarcoma. Virchows Arch 462:665–
tumors: evidence for a common histogenesis of Ewing’s sarcoma 671, 2013.
and peripheral primitive neuroectodermal tumors from MIC2 196. Martin-Zanca D, Oskam R, Mitra G, et al: Molecular and bio-
expression and specific chromosome aberrations. Cancer 67:1886– chemical characterization of the trk proto-oncogene. Mol Cell Biol
1893, 1991. 9:24–33, 1989.
175. Barbacid M: The Trk family of neurotrophin receptors. J Neuro- 197. Miettinen M, Lehto VP, Virianen I: Histogenesis of Ewing’s
biol 25:1386–1403, 1994. sarcoma: an evaluation of intermediate filaments and endothelial

ERRNVPHGLFRVRUJ
816 11  Ewing’s Sarcoma and Related Tumors

cell markers. Virchows Arch B Cell Pathol Incl Mol Pathol 41:277– 215. D’Amore ES, Ninfo V: Soft tissue small round cell tumors: mor-
284, 1982. phological parameters. Semin Diagn Pathol 13:184–203, 1996.
198. Moll R, Lee I, Gould VE, et al: Immunohistochemical analysis of 216. Devaney K, Vinh TN, Sweet DE: Small cell osteosarcoma of
Ewing’s tumors: patterns of intermediate filaments and desmo- bone: an immunohistochemical study with differential diagnostic
somal proteins indicate cell type heterogeneity and phenopoten- considerations. Hum Pathol 24:1211–1225, 1993.
tial diferentiation. Am J Pathol 127:288–304, 1987. 217. Gamberi G, Cocchi S, Benini S, et al: Molecular diagnosis in
199. Navarro S, Cavazzana AO, Llombart-Bosch A, et al: Comparison Ewing family tumors: the Rizzoli experience—222 consecutive
of Ewing’s sarcoma of bone and peripheral neuroepithelioma: an cases in four years. J Mol Diagn 13:313–324, 2011.
immunocytochemical and ultrastructural analysis of two primitive 218. Hill DA, O’Sullivan MJ, Zhu X, et al: Practical application of
neuroectodermal neoplasms. Arch Pathol Lab Med 118:608–615, molecular genetic testing as an aid to the surgical pathologic
1994. diagnosis of sarcomas: a prospective study. Am J Surg Pathol
200. Navas Palacios JJ, Aparicio-Duque R, Valdes MD: On the histo- 26:965–977, 2002.
genesis of Ewing’s sarcoma: an ultrastructural, immunohisto- 219. Martin SE, Dwyer A, Kissane SM, et al: Small-cell osteosarcoma.
chemical and cytochemical study. Cancer 53:1882–1901, 1984. Cancer 50:990–996, 1982.
201. Nogueira E, Navarro S, Pellin A, et al: Activation of Trk genes in 220. Meis-Kindblom JM, Stenman G, Kindblom LG: Differential
Ewing’s sarcoma: Trk A receptor expression is linked to neural diagnosis of small round cell tumors. Semin Diagn Pathol 13:213–
differentiation. Diagn Mol Pathol 6:10–16, 1997. 241, 1996.
202. Pagani A, Fischer-Colbrie R, Eder U, et al: Neural and mesen- 221. Nakashima Y, Unni KK, Shives TC, et al: Mesenchymal chon-
chymal differentiations in Ewing’s sarcoma cell lines: morphologi- drosarcoma of bone and soft tissue: a review of 111 cases. Cancer
cal, immunophenotypic, molecular biological and cytogenetic 57:2444–2453, 1986.
evidence. Int J Cancer 63:738–743, 1995. 222. Nyquist KB, Panagopoulos I, Thorsen J, et al: Whole-
203. Perlman EJ, Dickman PS, Askin FB, et al: Ewing’s sarcoma: transciptome sequencing identifies novel IRF2BP2-CDX1 fusion
routine diagnostic utilization of MIC2 analysis—a Pediatric gene brought about by translocation t(1;5)(q42;q32) in mesenchy-
Oncology Group/Children’s Cancer Group intergroup study. mal chondrosarcoma. PLoS ONE 7:e49705, 2012.
Hum Pathol 25:304–307, 1994. 223. Papadimitiou JC, Drachenberg CB: Ultrastructural features of the
204. Pinto A, Grant LH, Hayes FA, et al: Immunohistochemical matrix of small cell osteosarcoma. Hum Pathol 25:430–431, 1994.
expression of neuro-specific enolase and Leu-7 in Ewing’s sarcoma 224. Ponce-Castaneda MV, Garcia-Chequer AJ, Equia Aguilar P, et al:
of bone. Cancer 64:1266–1273, 1989. Detection of common chromosomal translocations in small round
205. Riopel M, Dickman PS, Link MP, et al: MIC2 analysis in pediatric blue cell pediatric tumors. Arch Med Res 45:143–151, 2014.
lymphomas and leukemias. Hum Pathol 25:396–399, 1994. 225. Puls F, Niblett AJ, Mangham DC: Molecular pathology of
206. Subbiah V, Brown RE, Jiang Y, et al: Morphoproteomic profiling bone tumours: diagnostic implications. Histopathology 64:461–476,
of the mammalian target of rapamycin (mTOR) signaling pathway 2014.
in desmoplastic small round cell tumor (EWS/WT1), Ewing’s 226. Ringus JC, Ridell RH: Small cell osteosarcoma: ultrastructural
sarcoma (EWS/FLI1) and Wilms tumor (WT1). PLoS ONE description and differentiation from atypical Ewing’s sarcoma. Lab
8(7):e68985, 2013. Invest 44:55A, 1981.
207. Tsuneyoshi M, Yokoyama R, Hashimoto H, et al: Comparative 227. Schmidt D, Mackay B, Ayala AG: Ewing’s sarcoma of bone
study of neuroectodermal tumor and Ewing’s sarcoma of the bone: with neuroblastoma-like features. Ultrastruct Pathol 3:143–151,
histopathologic, immunohistochemical and ultrastructural fea- 1982.
tures. Acta Pathol Jpn 39:573–581, 1989. 228. Sim FH, Unni KK, Beabout JW, et al: Osteosarcoma with small
208. Ushigome S, Shimoda T, Nikaido T, et al: Primitive neuroecto- cells simulating Ewing’s tumor. J Bone Joint Surg 61A:207–215,
dermal tumors of bone and soft tissue. With reference to histo- 1979.
logic differrentiation in primary metstatic foci. Acta Pathol Jpn 229. Steiner GC, Mirra JM, Bullough PG: Mesenchymal chondrosar-
42:483–493, 1992. coma: a study of the ultrastructure. Cancer 32:926–939, 1973.
209. Vakar-Lopez F, Ayala AG, Raymond AK, et al: Epithelial pheno- 230. Szuhai K, Cleton-Jansen AM, Hogendoorn PC, et al: Molecular
type in Ewing’s sarcoma and primitive neuroectodermal tumor. pathology and its diagnostic use in bone tumors. Cancer Genet
Int J Surg Pathol 8:59–65, 2000. 205:193–204, 2012.
210. Wang WL, Patel NR, Caragea M, et al: Expression of ERG, an 231. Triche TJ, Askin FB: Neuroblastomas and the differential diag-
Ets family transcription factor, identifies ERG-rearranged Ewing nosis of small round blue cell tumors. Hum Pathol 14:569–595,
sarcoma. Mod Pathol 25:1378–1383, 2012. 1983.
232. Triche TJ, Ross WE: Glycogen containing neuroblastoma with
Differential Diagnosis of Small Round-Cell Tumors of Bone clinical and histological features of Ewing’s sarcoma. Cancer 41:
211. Ayala AG, Ro JY, Raymond AK, et al: Small cell osteosarcoma: 1425–1432, 1978.
a clinicopathologic study of 27 cases. Cancer 64:2162–2173, 1989. 233. Wang L, Motoi T, Khanin R, et al: Identification of a novel,
212. Bertoni F, Picci P, Bacchini P, et al: Mesenchymal chondrosar- recurrent HEY1-NCOA2 fusion in mesenchymal chondrosarcoma
coma of bone and soft tissue. Cancer 52:533–541, 1983. based on a genome-wide screen of exon-level expression data.
213. Cantile M, Marra L, Franco R, et al: Molecular detection and Genes Chromosomes Cancer 51:127–139, 2012.
targeting of EWSR1 fusion transcripts in soft tissue tumors. Med 234. Wehrli BM, Huang W, de Crombrugghe B, et al: Sox9, a master
Oncol 30:412, 2013. regulator of chondrogenesis, distinguishes mesenchymal chon-
214. Choi EY, Gardner JM, Lucas DR, et al: Ewing sarcoma. Semin drosarcoma from other small blue round cell tumors. Hum Pathol
Diagn Pathol 31:39–47, 2014. 34:263–269, 2003.

ERRNVPHGLFRVRUJ
C H A P T E R 1 2 

Hematopoietic Tumors
April Ewton, MD

CHAPTER OUTLINE

CLASSIFICATION OF HEMATOPOIETIC TUMORS HISTIOCYTIC AND DENDRITIC CELL


PROLIFERATIONS
PLASMA CELL MYELOMA AND PLASMA CELL
NEOPLASMS Langerhans Cell Histiocytosis
Amyloidosis Associated with Multiple Erdheim-Chester Disease
Myeloma Sinus Histiocytosis with Massive
Distinct Forms of Myeloma Lymphadenopathy (Rosai-Dorfman
Disease)
NON-HODGKIN’S LYMPHOMA
MYELOID SARCOMA
Diffuse Large B-cell Lymphoma
Adult T-cell Lymphoma/Leukemia MASTOCYTOSIS
CLASSICAL HODGKIN’S LYMPHOMA

This chapter addresses some of the issues pertaining to lines of differentiation, a megakaryocyte/erythroid line,
skeletal manifestations of hematopoietic tumors, espe- and a granulocyte/monocyte lineage.
cially when they present as primary tumors in bone. In The second model, the myeloid-based model2,3 (Fig.
such instances, a bone pathologist or even a general 12-1), suggests that there is a common precursor that
pathologist, rather than a hematopathologist, is more maintains the capacity to produce both myeloid and lym-
likely to be confronted with diagnostic challenges. For phoid progeny. Experimental evidence that the myeloid-
these reasons, familiarity with skeletal manifestations of based model is more accurate than the myeloid-lymphoid
hematopoietic lesions should be of general interest. This model continues to accumulate; however, at present there
chapter also discusses plasma cell myeloma and other is no universal agreement. The myeloid-based model of
plasma cell neoplasms, non-Hodgkin lymphomas, leuke- lineage differentiation is also supported by examples of
mias, histiocytic and dentritic cell proliferations, and neoplasms exhibiting lineage plasticity such as acute leu-
mastocytosis, with particular reference to their involve- kemias of ambiguous lineage with both myeloid and lym-
ment of skeletal tissue. phoid features.5 There are even reported cases of
Langerhans cell histiocytosis with clonally rearranged
T-cell or B-cell receptors1 and Langerhans cell histiocy-
tosis occurring with clonally related T-cell acute lympho-
CLASSIFICATION OF blastic leukemia4 or acute leukemia of ambiguous lineage.6
HEMATOPOIETIC TUMORS T lymphocytes, B lymphocytes (and later plasma
cells), and natural killer cells arise from the common
In general, the approach to classifying hematopoietic lymphoid precursor. Dendritic cells arise from a common
tumors parallels the current knowledge of lineage dif- macrophage/dendritic precursor. Normal Langerhans
ferentiation, naming the diseases in terms of their postu- cells arise from fetal macrophages that differentiate
lated normal cell counterparts (Fig. 12-1). The earliest after migrating to the skin. Neoplastic Langerhans cells,
hematopoietic stem cells are capable of producing however, are thought to arise from a myeloid precursor.
cells of all blood lineages. Although there is still contro- Currently, the most widely accepted classification is
versy regarding the degree and timing of lineage com- the World Health Organization Classification of Hematopoi-
mitment, two prevailing models are considered here. etic and Lymphoid Tissues, first published in 2001 and
Both models begin with a long-lived uncommitted hema- revised in 2008.5 There is a trend toward classification of
topoietic stem cell that then differentiates into a short- distinct neoplasms by their molecular and genetic fea-
lived multipotent hematopoietic stem cell. At this point, tures when warranted, within the context of clinical and
the models diverge. morphologic features. As targeted therapy becomes more
The first model, the classical myeloid-lymphoid and more prevalent, the molecular alteration of pathways
model, postulates that the first step occurs with lineage that drive the proliferation and survival of these hemato-
commitment to myeloid or lymphoid differentiation. poietic neoplasms is becoming important in the descrip-
The common myeloid precursor then gives rise to two tion of these entities.
817
ERRNVPHGLFRVRUJ
818 12  Hematopoietic Tumors

Plasma cell myeloma


Lymphoma
B cell
ce

Lymphoma
CLP
T cell

Myeloid sarcoma
Granulocyte
Granulocyte
l
Sinus Histiocytosis with
massive lymphadenopathy
LMPP Erdheim-Chester disease
Monocyte Macrophage
Macr
rop
oph
pha
hage
g Myeloid sarcoma

Langerhans cell histiocytosis

HSC GMP MDP


Dendritic cell

F l
Fetal Langerhans
CMP macrophage cell

Erythrocytes
Myeloid sarcoma

MkEP

Platelets
FIGURE 12-1  ■  Classification of hematopoietic and lymphoid tumors in the context of lineage commitment and postulated cell of
origin. Uncommited hematopoietic stem cells give rise to cells with progressive commitment to development of various cell lineages.
Hematopoietic neoplasms arise from cells of specific committed lineages or their precursors. HSC, hematopoietic stem cell;
LMPP, lymphoid-primed multipotent progenitor; CMP, common myeloid progenitor; CLP, common lymphoid progenitor;
GMP, granulocyte/monocyte progenitor; MkEP, megakaryocyte/erythroid progenitor; MDP, myeloid/dendritic progenitor. (Based on
and adapted from Luc S, et al: Delineating the cellular pathways of hematopoietic lineage commitment. Semin Immunol 20:213–220, 2008.)

PLASMA CELL MYELOMA AND PLASMA most common hematopoietic malignancy, accounting for
approximately 10% of hematopoietic neoplasms, and the
CELL NEOPLASMS most frequent neoplasm presenting as skeletal lesions.30,40,58
Definition The skull, vertebral bodies, pelvis, and proximal parts of
the major long tubular bones are most frequently involved
Plasma cell myeloma is a monoclonal, neoplastic prolif- (Fig. 12-2). More than 95% of cases are diagnosed in
eration of plasma cells that involves the bone marrow and patients older than age 40 years, with a peak incidence
occasionally involves extraskeletal sites. Lytic bone lesions between ages 65 and 74 years (Fig. 12-3).57 Plasma cell
are a prominent feature of typical cases. myeloma is more common in the black population (Fig.
12-4).57 Plasma cell myeloma is always preceded by
monoclonal gammopathy of undetermined significance
Clinical Features
(MGUS), but MGUS may not be detected in this early
Plasma cell myeloma is one of the most frequently occur- subclinical phase of disease. Patients with MGUS prog-
ring hematopoietic neoplasms and accounts for approxi- ress to plasma cell myeloma at the rate of approximately
mately 1% of all malignant neoplasms in white patients 1% per year.39 The incidence of plasma cell myeloma is
and approximately 2% in black patients. It is the second increasing, and there is currently no definitive cure;

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 819

55
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 12-2  ■  Plasma cell myeloma. Peak age incidence and most frequently involved skeletal sites are indicated by solid black
arrows. Less frequently involved sites are indicated by clear arrows.

30 9

8
25
(cases/100,000 persons)

7
Age distribution (%)

20 6
Incidence rate

5
15
4
10 3

2
5
1
0 0
<20 20-34 35-44 45-54 55-64 65-74 75-84 >84
e

es

es

es
e
al

al

al

al

al

al
m

m
fe

Age at diagnosis
s,

fe

fe
te

k
e

s,

ac
ac

hi

te

k
e

ac
Bl
W
ac

hi

FIGURE 12-3  ■  Age distribution of multiple myeloma cases. The


lr

Bl
W
Al

lr

peak age of incidence is 65 to 74 years. SEER Data, 1973-2010.


Al

FIGURE 12-4  ■  Distribution of plasma cell myeloma by race and


sex. The most commonly affected group is black males. SEER
however, with greater understanding of the pathogenesis, Data, 1973-2010.
patient survival is improving.
Plasma cell myeloma usually presents with multifocal a history of recurrent infections. In the majority of
osteolytic lesions, proliferation of plasma cells, and patients, increased levels of monoclonal immunoglobu-
monoclonal gammopathy. Typically, patients have multi- lins (M-protein) can be detected by serum or urine elec-
focal bone pain (especially in the weight-bearing sites), trophoresis. Recommended laboratory testing includes
anemia, hypercalcemia, renal failure, proteinuria, and complete blood count, serum creatinine, serum calcium,

ERRNVPHGLFRVRUJ
820 12  Hematopoietic Tumors

serum protein electrophoresis with immunofixation,


TABLE 12-1 Laboratory Tests for
serum free light chain assay, and urine protein analysis
Multiple Myeloma
(Table 12-1).18
History and physical examination Diagnostic criteria used to subclassify plasma cell neo-
Complete blood count and differential; peripheral blood plasms include laboratory and clinical features. MGUS is
smear defined as the presence of an M-protein at levels below
Chemistry screen, including calcium and creatinine
Serum protein electrophoresis, immunofixation 3 g/dL in the absence of any other defining features of
Nephelometric quantification of serum immunoglobulins plasma cell myeloma. Two broad categories of plasma cell
Routine urinalysis, 24-hour urine collection for myeloma are designated smoldering (asymptomatic) myeloma
electrophoresis and immunofixation and symptomatic myeloma. By the 2008 WHO criteria, a
Bone marrow aspirate or biopsy
Cytogenetics (metaphase karyotype and fluorescence in
diagnosis of asymptomatic plasma cell myeloma may be
situ hybridization [FISH]) made if there are more than 10% clonal plasma cells in
Radiologic skeletal bone survey, including spine, pelvis, the bone marrow or the serum M-protein is greater than
skull, humerus, and femur; magnetic resonance imaging 3 g/dL. A diagnosis of symptomatic plasma cell myeloma
in certain circumstances requires the presence of a clonal plasma cell population
Serum β2-microglobulin and lactate dehydrogenase
Measurement of serum-free light chains of any quantity (usually with associated detectable
M-protein) and end-organ damage, as represented by the
Adapted from Dimopoulos M, et al: Consensus acronym CRAB (hyperCalcemia, Renal insufficiency,
recommendations for standard investigative workup: report Anemia, lytic Bone lesions) (Table 12-2).18,38,40,42
of the International Myeloma Workshop Consensus Panel 3.
Blood 117:4701-4705, 2011.
The main prognostic factors are related to cytogenetic
findings, extent of disease, patient age, and renal func-
tion. Factors that can be evaluated in the laboratory

TABLE 12-2 Diagnostic Criteria for Plasma Cell Disorders


Disorder/ Criteria Comment
Monoclonal gammopathy Serum monoclonal protein <3 g/dL All three criteria must
of undetermined Clonal bone marrow plasma cells <10% be met
significance (MGUS) Absence of end-organ damage, such as hypercalcemia, renal
insufficiency, anemia, or bone lesions
Smoldering Serum monoclonal protein (IgG or IgA) ≥3 g/dL or clonal bone marrow Both criteria must be
(asymptomatic) plasma cells ≥10% met
multiple myeloma Absence of end-organ damage, such as lytic bone lesions, anemia,
hypercalcemia, or renal failure
Symptomatic multiple Clonal bone marrow plasma cells ≥10% All three criteria must
myeloma Presence of serum or urinary monoclonal protein (except in patients with be met except as
nonsecretory multiple myeloma) noted
Evidence of end-organ damage (CRAB):
HyperCalcemia: serum calcium ≥11.5 mg/dL
Renal insufficiency: serum creatinine >2 mg/dL
Anemia: normochromic, normocytic with a hemoglobin value of >2 g/dL
below the lower limit of normal, or a hemoglobin value <10 g/dL
Bone lesions: lytic lesions, severe osteopenia, or pathologic fractures
Nonsecretory myeloma >10% clonal plasma cells confirmed by biopsy
<0.5 mg/dL serum protein by SPEP
<200 mg/24 hrs of light chain proteinuria by UPEP
Unquantifiable free light chain measurement
Solitary plasmacytoma of Minimal or absent M-protein in serum or urine
bone Single area of bone destruction due to clonal plasma cells
Bone marrow <10% clonal plasma cells
Normal skeletal survey (and MRI if done)
No related organ or tissue impairment (CRAB) other than solitary bone
lesion
Extramedullary Minimal or absent M-protein in serum or urine
plasmacytoma Extramedullary tumor of clonal plasma cells
Bone marrow <5% clonal plasma cells
Normal skeletal survey
No related organ or tissue impairment (end- organ damage, including
bone lesions)

Adapted from Kyle RA, et al: Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report
of the International Myeloma Working Group. Br J Haematol 121:749-757, 2003 and Kilciksiz S, et al: A review for solitary
plasmacytoma of bone and extramedullary plasmacytoma. Scientific World Journal 2012:1-6, 2012.
SPEP, serum protein electrophoresis; UPEP, urine protein electrophoresis.
Adapted from Dimopoulos M, et al: Consensus recommendations for standard investigative workup: report of the International
Myeloma Workshop Consensus Panel 3. Blood 117:4701-4705, 2011 and Kilciksiz S, et al: A review for solitary plasmacytoma of
bone and extramedullary plasmacytoma. Scientific World Journal 2012:1-6, 2012.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 821

40

35

Relative Survival (%) 30

25

20

15

10

0
73

75

77

79

81

83

85

87

89

91

93

95

97

99

01

03

05
19

19

19

19

19

19

19

19

19

19

19

19

19

19

20

20

20
FIGURE 12-5  ■  Plasma cell myeloma: 5-year survival. Five-year survival for multiple myeloma from 1973 through 2006. Survival has
been improving with newer therapies. SEER Data, 1973-2006.

include cytogenetic status, proliferation index, neoplastic currently cytoreductive therapy followed by transplant
plasma cells as a percentage of total plasma cells, free is the standard of care; however, treatment with novel
light chain ratio, serum lactate dehydrogenase, and β2- agents may replace this approach in the future. Com-
microglobulin level.40 monly used novel agents are immunomodulatory drugs
With current therapeutic approaches, the progression- such as lenolidamide or thalidomide, and the proteasome
free survival and overall survival for patients with plasma inhibitor bortezomib. Commonly used chemotherapeu-
cell myeloma has improved (Fig. 12-5).57 Before 1962, in tic drugs include melphalan and cyclophosphamide.44
the absence of any known treatment, the median survival
for newly diagnosed myeloma patients was less than 1 Radiographic Imaging
year.37 Although there has been steady improvement in
survival over the years, median overall survival in the The International Myeloma Working Group (IMWG)
early 2000s was still only 2 to 3 years. In the current era consensus guidelines for imaging studies were last updated
of novel therapeutic agents, overall median survival is in 2009. A skeletal survey, consisting of a series of plain
now approximately 8 years.44 radiographs, is recommended for every patient. If the
Patient prognosis and choice of therapy are most com- skeletal survey detects no lesions, or if the initial diagno-
monly determined by cytogenetic risk factors and age. An sis is solitary plasmacytoma of bone, magnetic resonance
example of risk stratification as delineated by the Mayo imaging (MRI) should be performed to detect possible
group identifies high risk, intermediate risk, and standard occult bone lesions. Computed tomography (CT) scan or
risk groups based on cytogenetic features or gene expres- MRI are required for any patient with suspected spinal
sion profiling in the context of response to currently cord compression. Positron emission tomography (PET)
available therapies. High risk features are deletion of 17p, scan is not routinely recommended.19
t(14;16), t(14;20), or a high risk signature by gene expres- Distinctive radiographically detectable changes are
sion profiling. Intermediate risk features are t(4;14), dele- present in approximately 80% of patients with multiple
tion of 13, hypodiploidy, and a plasma cell labeling index myeloma. The axial skeleton and the trunk bones are
greater than 2. Standard risk patients have none of the preferentially involved. The most pronounced and earli-
preceding features and may have t(11;14) or t (6;14).44 est changes are typically seen in the skull, vertebrae, ribs,
The current median overall survival for high risk, inter- and pelvis and correspond to the predominance of hema-
mediate risk, and standard risk is 3 years, 4 to 5 years, topoietic marrow at these sites in patients older than age
and 8 to 10 years, respectively.44 50 years.15 They represent multifocal, sharply demar-
The most widely used therapeutic regimens include cated, lytic foci and are often called punched-out lesions
dexamethasone or prednisone and one or more novel (Figs. 12-6 to 12-8). Erosions of the cortex are frequently
agents, in some cases augmented with alkylating agents seen, and prominent periosteal new bone formation is
or anthracyclines. Patients under age 70 years are poten- typically not present. Bones with a smaller diameter, such
tially eligible for autologous stem cell transplant, and as the ribs, can exhibit expanded contours. Pathologic

ERRNVPHGLFRVRUJ
822 12  Hematopoietic Tumors

B C D E
FIGURE 12-6  ■  Plasma cell myeloma: radiographic features. A, Lateral radiograph showing extensive multiple lytic lesions of cal-
varium. B and C, Anteroposterior (AP) radiographs of extensive multiple lytic lesions of both humeri. D and E, AP radiographs of
multiple lytic lesions of bilateral femora and fibulae. All images are from the same patient showing extensive involvement of virtu-
ally the entire skeleton.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 823

A B C

D E
FIGURE 12-7  ■  Plasma cell myeloma: radiographic features. A, Anteroposterior (AP) radiograph of humerus showing a destructive
lytic lesion centered in the shaft with involvement of the surrounding soft tissue. B, AP radiograph of lytic lesion of the humeral
shaft. C, Lateral radiograph of the calvarium showing multiple lytic lesions. D, AP radiograph of femoral head and neck showing
a destructive lytic lesion. E, Radioisotopic bone scan showing increased uptake in the left proximal humerus. Same case as
shown in D.

ERRNVPHGLFRVRUJ
824 12  Hematopoietic Tumors

A B

C D
FIGURE 12-8  ■  Plasma cell myeloma: radiographic features. A, Lateral radiograph of skull showing large expansile lytic lesion in
vertex of skull with multiple, small, punched-out foci of lucency in parietal and occipital areas. B, Radioisotope scan (technetium
99) showing increased uptake in region of destructive skull lesions. Note photon deficit in center of large lesion at vertex. C, Magnetic
resonance image of head and neck shown in A reveals skull defect and tumor at vertex impinging on cerebral hemisphere. D, Pho-
tomicrograph showing features of atypical plasma cells (×200, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 825

fractures are frequent and can be the presenting sign. In hematoxylin-eosin–stained sections, and deeply baso-
fact, collapse of the vertebral body is frequently a pre- philic in Wright-Giemsa or Diff-Quik (Romanovsky’s)
senting symptom (Fig. 12-9). The disease typically first stained smears. As described by Bartl et al in 1987, the
appears in the axial skeleton and trunk bones, but with plasma cells may vary considerably in cytomorphology
progression of the process, extensive involvement of (Fig. 12-13).11 The morphologic range includes cells with
the appendicular skeleton and multiple pathologic frac- polymorphous or asynchronous features, as well as forms
tures can develop. Surprisingly, despite extensive bone resembling normal mature plasma cells, small lympho-
destruction, the results of radioisotopic bone scans are cytes, blasts, or lymphocytes with cleaved nuclei. These
frequently negative in patients with multiple myeloma. morphologic variants may pose diagnostic challenges,
This is explained by the predominant bone destructive raising the differential diagnosis of lymphoma, leukemia
activity of myeloma cells with typically minimal new or other blastic neoplasms, or even carcinoma in cases
bone production. A small number of patients do not with marked pleomorphism.
have lytic punched-out lesions at presentation and may The degree of immaturity of the myeloma cells is a
show generalized osteoporosis that is sometimes quite prognostic factor.26 Loss of the morphologic plasma
pronounced. In some instances, the lesions in multiple cell phenotype with the presence of forms resembling
myeloma may appear sclerotic on radiographs (Figs. blasts is associated with more aggressive behavior and a
12-10 and 12-11). Sclerotic lesions are typical for POEMS poorer prognosis. In most myelomas, clearly recogniz-
syndrome (discussed in the section on osteosclerotic able features of plasma cells can be found at least
myeloma in this chapter).28 focally.
Numerous Russell bodies representing cytoplasmic
spherical structures of polymerized immunoglobulins can
Gross Findings
be present. The accumulation of cytoplasmic immuno-
Antemortem gross examination is typically restricted globulins can be present in the form of morular or
to small biopsy or curettage samples from the site of Mott cells, representing grapelike cytoplasmic structures.
pathologic fracture and show fragments of tan-gray soft These structures are best seen in Giemsa stained cyto-
tissue. At autopsy the principal growth patterns can be logic preparations. Although Russell bodies are frequently
appreciated: diffuse involvement of bone marrow and seen in reactive plasma cells, intranuclear inclusions, or
growth in the form of discrete nodules (Fig. 12-12). Most Dutcher bodies, almost always indicate neoplastic plasma
likely, the diffuse involvement represents a more advanced cells (Fig. 12-14). The cytoplasm of myeloma cells has
stage and is produced by the confluence of individual ultrastructural features of maturity with well-developed
nodules. On the other hand, some patients have general- rough endoplasmic reticulum and a prominent Golgi
ized osteoporosis at presentation, and nodular lytic region (Fig. 12-16). The latter can be seen by light
changes develop later in the course of the disease. The microscopy as a perinuclear clearing.
individual nodules are best seen in the vertebral bodies. A
more advanced process is associated with the collapse of Immunohistochemistry and
one or several vertebral bodies. The ribs typically show Differential Diagnosis
multiple extended foci. The long bones in the appendicu-
lar skeleton show nodules or diffuse involvement with Plasma cells represent the end stage of B-cell differen-
cortical thinning and disruption. Pathologic fractures are tiation, but they typically do not express common pan–
frequent, especially at the weight-bearing sites. The prox- B-cell antigen CD20. They are also typically negative
imal parts of the appendicular skeleton are initially pref- for most T-cell markers. A summary of microscopic ultra-
erentially involved. structural and immunophenotypic features of myeloma
cells is provided in Table 12-3. Myeloma cells are of
the plasma cell lineage and have many of the same fea-
Microscopic Findings
tures. The most characteristic feature of myeloma cells
Plasma cell myeloma consists of a proliferation of cells is their monotypic expression of κ or λ immunoglobu-
that usually include at least a subset of cells that resemble lin. They typically brightly express normal plasma cell
normal plasma cells. The cells form clusters or sheets markers CD38, CD138, and MUM1.41 In the majority
within the bone marrow of involved tissue. These cells of cases, the cells show aberrant expression of CD56,
are round or oval and have an eccentric nucleus and an adhesion molecule, which may be lost in plasma
a clumped “clockface” chromatin pattern with the chro- cell leukemia and extramedullary plasmacytomas. The
matin clustered at the periphery of the nucleus near the second most common aberrantly expressed marker is
nuclear membrane. The plasma cell cytoplasm has a CD117.
prominent perinuclear hof, corresponding ultrastructur- The differential diagnosis includes reactive plasmacy-
ally to a prominent Golgi region, and rough endoplastic tosis, reactive inflammatory conditions with a prominent
reticulum appropriate for active immunoglobulin pro- plasma cell component, and other neoplasms. Normal
duction and packaging. bone marrow plasma cells are located primarily in a peri-
When multiple myeloma is suspected, smears should vascular distribution, in contrast to myeloma plasma cells,
always be prepared because the plasma cell nature of which form sheets and clusters in a predominantly inter-
the round-cell infiltrate is quite often more evident in stitial pattern. Binucleate plasma cells without nuclear
cytologic preparations than in conventional histologic atypia can frequently be found in reactive infiltrations.
sections. The cytoplasm is dense and eosinophilic in Text continued on p. 832

ERRNVPHGLFRVRUJ
826 12  Hematopoietic Tumors

A B

C D

FIGURE 12-9  ■  Plasma cell myeloma: radiographic features. A, Anteroposterior radiograph of right shoulder shows destructive lesion
in axillary border of scapula with soap-bubble appearance and small lucent foci in proximal humeral metaphysis. B and C, Lateral
radiographs of lumbar spine show osteopenia and partial collapse of vertebral bodies. D, Lytic lesion of humeral shaft with patho-
logic fracture.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 827

B C
FIGURE 12-10  ■  Sclerosing plasma cell myeloma: radiographic features. A, Lateral radiograph of skull shows multiple foci of radioden-
sity varying from a few millimeters to more than a centimeter in diameter. B, Right side of pelvis and proximal femur seen in this
anteroposterior radiograph contain small focal densities with diameters ranging up to several centimeters. C, Lateral radiograph of
thoracic spine shows osteopenia and focus of radiodensity within upper border of vertebral body.

ERRNVPHGLFRVRUJ
828 12  Hematopoietic Tumors

B
FIGURE 12-11  ■  Sclerosing plasma cell myeloma: radiographic features. A, Radiograph of pelvis shows multiple radiodense foci.
B, Sclerotic lesions of thoracic and lumbar vertebral bodies. C, Proximal humerus with several radiodense foci.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 829

A B

C D
FIGURE 12-12  ■  Plasma cell myeloma: radiographic, gross, and microscopic features. A, Radiograph of skull shows multiple lytic
punched-out lesions. B, Gross photograph of autopsy specimen shows multiple lytic foci composed of soft gray tissue (same case
as in A). C, Whole-mount specimen of perpendicular section of parietal bone shows nodular growth pattern of multiple myeloma.
D, Higher magnification of C shows nodules to be composed of sheets of plasma cells (×200, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
830 12  Hematopoietic Tumors

A B

C D
FIGURE 12-13  ■  Plasma cell myeloma: cytologic features. A, Blastoid plasma cells with high nuclear cytoplasmic ratio and prominent
nucleoli. B, Mature plasma cells with feathered cytoplasmic borders. C, Pleomorphic plasma cells, immature forms, and multinucle-
ated forms. D, Small lymphocyte-like variant of multiple myeloma with small nuclei and scant cytoplasm (A-D, ×1000) (A-D, Wright-
Geimsa stain).

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 831

A B

C D

FIGURE 12-14  ■  Plasma cell myeloma: microscopic features. A, Intranuclear inclusions of immunoglobulin (Dutcher bodies) (×1000).
B, Small lymphocyte-like variant resembling lymphoma (×400). C, Blastic variant with prominent nucleoli resembling acute leukemia
(×400). D, Pleomorphic variant with markedly atypical nuclei and multinucleated plasma cells (×400). Inset, Higher magnification of
Wright-Geimsa stain of same case as D (×1000). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
832 12  Hematopoietic Tumors

infiltrates may be composed by up to 50% of plasma


TABLE 12-3 Summary of Microscopic and
cells.51 The polyclonal nature of the plasma cells may
Immunophenotypic Features
be demonstrated by immunohistochemical stains, in
of Myeloma Cells
situ hybridization, or polymerase chain reaction (PCR)
Microscopic studies for immunoglobulin clonality.
Cytoplasm Neuroendocrine tumors and melanoma may have
Dense eosinophilic to amphophilic cytoplasm plasmacytoid cytomorphology but can be differentiated
Perinuclear hof from plasma cell myeloma by staining for neuroendo-
Variable immunoglobulin inclusions: Russell bodies, Mott crine markers such as synaptophysin and chromogranin
cells, rare crystals/rods or granules (CD56 will not resolve this diagnostic dilemma). The
Nucleus majority of melanomas express MUM1, and a subset
Clumped “clock face” chromatin pattern of melanoma cases may show variable expression of
Variable features: Prominent nucleoli, open blastic CD138.27,48 Most plasma cell myelomas are strongly and
chromatin uniformly positive for CD138 and MUM1, while the
Variable immunoglobulin inclusions: Dutcher bodies
staining is typically more variable in melanoma. Κ and λ
Ultrastructure immunohistochemical stains will show monotypic light
Clumped chromatin chain expression in the majority of plasma cell myeloma
Prominent Golgi cases, whereas melanoma is negative for light chain
Immunophenotypic expression and positive for S-100 in the majority of cases.
Cytoplasm HMB-45 or Melan-A may stain unusual S-100– negative
CD138+, CD79a+, CD56+ (majority) cases of melanoma.
CD19−, CD20−, CD45− or dim (majority) Pleomorphic variants of plasma cell myeloma may
κ or λ light chain restriction by immunohistochemistry or resemble carcinoma or lymphoma histologically. Care
in situ hybridization should be taken in selecting and interpreting immuno-
Variable: Small lymphocyte-like variant may express CD20
histochemical stains in tumors with pleomorphic or ana-
Nucleus plastic morphology. Several types of carcinomas may
MUM1+ show variable expression of CD138 and MUM1.47,55
Variable: Subset of cases show overexpression of cyclin Again, staining for CD138 and MUM1 is typically
D-1 uniform in plasma cell myeloma and more variable in
Variable: Small lymphocyte-like variant may express PAX5
carcinomas. Κ and λ immunohistochemical stains will
Pitfalls show monotypic light chain expression in the majority of
Various carcinomas and occasional melanomas may be plasma cell myeloma cases. Although carcinoma and
positive for CD138 plasma cell myeloma may be positive for EMA, a variety
The majority of melanomas are positive for MUM1
of cytokeratin stains will be positive in the majority of
carcinomas and negative in plasma cell myeloma. Most
T-cell lymphomas will express some combination of
T-cell lineage markers (CD2, CD3, CD4, CD5, CD7,
CD8), which are absent in plasma cell myeloma. Inter-
Immunohistochemical stains or in situ hybridization for pretation of B-cell lineage markers is more problematic,
immunoglobulin κ and λ are very helpful in distinguish- because the majority of markers expressed by plasma cell
ing reactive polytypic plasma cell infiltrates from plasma myeloma may also be expressed in various subtypes
cell myeloma. The normal κ-to-λ ratio is approximately of B-cell lymphomas. Plasma cell myeloma is typically
2 or 3 : 1. Increased polytypic plasma cells may be seen negative for CD20, but a subset of myelomas, including
in a number of reactive conditions, including viral and the small lymphocyte-like variant, may express CD20
bacterial infections, autoimmune disease, cirrhosis, and and PAX5 (Fig. 12-15). In difficult cases, staining for
solid tumors.23 CD56, CD117, or uniform strong staining for cyclin D1
The presence of other inflammatory cells together support a diagnosis of plasma cell myeloma. Clinical
with associated fibrosis and a prominent vasculature workup for myeloma may help distinguish B-cell lym-
favors a reactive process. The clinical and radiographic phoma from plasma cell myeloma in cases with overlap-
data in such cases with the absence of bone marrow plas- ping features.43
macytosis, a lack of multifocal skeletal changes, and the Finally, the small lymphocyte-like variant of plasma
absence of the M component are additional general clini- cell myeloma has many morphologic and immunohisto-
cal, radiologic, and laboratory features that support the chemical similarities with mature B-cell lymphomas with
diagnosis of a benign disorder. small cell cytomorphology.29 In particular, this variant
An unusual pattern of immunoproliferation, including may be misdiagnosed as mantle cell lymphoma on the
numerous polyclonal plasma cells and reactive immu- basis of staining with CD20 and cyclin D1 (Fig. 12-15).
noblasts, may mimic plasma cell myeloma. This prolif- Mantle cell lymphoma will be positive for CD5 in the
eration has been termed systemic polyclonal immunoblastic majority of cases, and cyclin D1, while positive in the
proliferation and may present with systemic symptoms and majority of cells, will show variable intensity of staining
plasma cell infiltrates mimicking plasma cell myeloma. from cell to cell. Plasma cell myeloma will be negative
The proliferation involves blood and bone marrow with for CD5 and typically shows strong homogeneous stain-
variable involvement of other organs and tissues. The ing of the myeloma cells.43

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 833

C D
FIGURE 12-15  ■  Plasma cell myeloma, small lymphocyte-like variant: microscopic and immunohistochemical features. A, Multiple
myeloma small lymphocyte-like variant involving bone marrow (×200). Inset, Small lymphocyte-like variant of multiple myeloma
with small nuclei and scant cytoplasm (×1000). B, Cyclin D1 immunohistochemical stain of small lymphocyte-like plasma cells. Note
bright homogeneous nuclear staining (×200). C, PAX5 immunohistochemical stain of small lymphocyte-like plasma cells (×200).
D, CD20 immunohistochemical stain of small lymphocyte-like plasma cells (×200). (A-D, hematoxylin-eosin; inset, Wright-Geimsa
stain)

ERRNVPHGLFRVRUJ
834 12  Hematopoietic Tumors

A B

C D
FIGURE 12-16  ■  Plasma cell myeloma: microscopic and ultrastructural features. A, Intermediate power photomicrograph shows
proliferation of atypical plasma cells (×200). B, Amorphous eosinophilic extracellular deposition of amyloid associated with myeloma
(×200). C, Higher power photomicrograph of B showing amyloid associated with myeloma (×400). D, Ultrastructural features of
myeloma cells. Note resemblance to normal plasma cells. Cells have eccentric nuclei with prominent nucleolus and peripherally
clumped chromatin. Cytoplasm contains abundant rough endoplasmic reticulum (×3500). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 835

Initiation Progression
Germinal Center Bone marrow Peripheral blood

Post-germinal- Smoldering Plasma cell


MGUS Myeloma
center B cell myeloma leukemia

Inherited variants
Primary genetic events:
• IGH @ translocations
• Hyperdiploidy
Secondary genetic events:
• Copy number abnormalities
• DNA hypomethylation
• Acquired mutations

Competition selection for bone marrow niche Migration and


founder effect

Clonal advantage
g

Tumor cell diversity

Genetic lesions

FIGURE 12-17  ■  Initiation and progression of plasma cell myeloma. Monoclonal gammopathy of undetermined significance (MGUS)
is an indolent, asymptomatic condition that transforms to myeloma at a rate of 1% per annum. Smoldering myeloma lacks clinical
features; by contrast, symptomatic myeloma has various clinical features that are collectively referred to as calcium, renal, anemia,
and bone abnormalities (CRAB), which provide an indication that treatment is required. Later in the disease progression, the
myeloma plasma cells are no longer restrained to growth within the bone marrow and can be found at extramedullary sites and as
circulating leukemic cells. It is thought that transition through these different states requires the acquisition of genetic abnormalities
that lead to the development of the biologic hallmarks of myeloma. The initial deregulated cell belongs to the MGUS clone; however,
subsequent to the development of sufficient genetic abnormalities, it acquires a clonal advantage, expands, and evolves. This clonal
evolution is through the branching pathways that are typically associated with Darwin’s explanation of the origin of species. During
the evolution of MGUS to myeloma, these processes lead to the development of numerous ecosystems, which correspond to the
clinically recognized phases of disease. At the end of this evolutionary process, at the stage of plasma cell leukemia (PCL), the clone
is proliferative and no longer confined to the bone marrow; the clone expands rapidly and leads to patient death. Cells at this stage
are substantially altered genetically, and the precursor subclones will be present at low levels because of competition for access to
the stromal niches in the bone marrow: these clones may be eradicated by more aggressive clones. In evolutionary terms, this
phase of disease could be considered to be initiated by a migration and founder effect whereby a cell that is able to survive
and grow in the peripheral blood is faced with no competition, thus limiting its clonal expansion. IGH@, immunoglobulin heavy
chain locus. (Modified and reprinted with permission from Morgan G, et al: The genetic architecture of multiple myeloma. Nature Rev
12:335-348, 2012.)

Genetic Features and Pathogenesis natural selection, resulting in survival and proliferation
of the best adapted clone or clones, at this stage repre-
The normal cell counterpart is a postgerminal center senting plasma cell myeloma and in some cases plasma
plasma cell with clonal immunoglobulin gene rearrange- cell leukemia (Fig. 12-17).46
ments and somatic hypermutation of immunoglobulin Plasma cell myeloma can be broadly divided into two
genes, typically secreting antibody. Following early major molecular categories: hyperdiploid and nonhyper-
molecular events, the abnormal plasma cells migrate to diploid. The majority of both hyperdiploid and non-
the bone marrow, where the cells are supported by their hyperdiploid cases show features of dysregulation of D
interactions with constituents of the marrow microenvi- cyclins, driving cell growth. Overall, hyperdiploid cases
ronment. Subclones with slightly different mutations have a better prognosis. Hyperdiploidy in plasma cell
compete for survival within the marrow microenviron- myeloma is the result of trisomies of the odd-numbered
ment. This stage of disease represents MGUS. Random chromosomes 3, 5, 7, 9, 11, 15, 19, and 21. Nonhy-
mutations result in progression of disease and clonal evo- perdiploid cases are associated with reciprocal transloca-
lution of multiple subclones. These subclones undergo tions involving immunoglobulin genes. Hyperdiploidy

ERRNVPHGLFRVRUJ
836 12  Hematopoietic Tumors

TABLE 12-4 Genetic Events Underlying the Initiation and Progression of Myeloma to Plasma
Cell Leukemia
Inherited Variations Molecular Hallmarks
Single-Nucleotide Polymorphisms Immortalization
2p: DTNB and DNMT3A G1/S abnormality: CDKN2C, RB1 (3%), CCND1 (3%), and
3p: ULK4 and TRAK1 CDKN2A
7p: DNAH11 and CDCA7L Proliferation: NRAS (21%), KRAS (28%), BRAF (5%), and MYC
(1%)
Primary Genetic Events (% of tumors) Resistance to apoptosis: PI3K and AKT
IGH@ Translocations and Genes Affected NFĸB pathway: TRAF3 (3%), CYLD (3%), and I-ĸB
Abnormal localization and bone disease: DKK1, FRZB, and
t(4;14): FGFR3 and MMSET (11%) DNAH5 (8%)
t(6;14): CCND3 (<1%) Abnormal plasma cell differentiation: (XBP1 (3%), BLIMP1
t(11:14): CCND1 (14%) (also known as PRDM1) (6%), and IRF4 (5%)
t(14;16): MAF (3%) Abnormal DNA repair: TP53 (6%), MRE11A (1%), and PARP1
t(14;20): MAFB (1.5%) RNA editing: DIS3 (13%), FAM46C (10%), and LRRK2 (5%)
Hyperdiploidy (57%) Epigenetic abnormalities: KDM6A (also known as UTX) (10%),
MLL (1%), MMSET (8%), HOXA9, and KDM6B
Trisomies of chromosomes 3, 5, 7, 9, 11, 15, 19, and 21 Abnormal immune surveillance
Secondary Genetic Events (% of tumors) Abnormal energy metabolism and ADME events
Gains Epigenetic Events
1q: CKS1B and ANP32E (40%) Global hypomethylation (MGUS to myeloma)
12p: LTBR Gene-specific hypermethylation (myeloma to plasma cell
17q: NIK leukemia)
Secondary Translocations
t(8;14): MYC
Other non-IGH@ translocations
Deletions
1p: CDKN2C, FAF1, and FAM46C (30%)
6q (33%)
8p (25%)
11q: BIRC2 and BIRC3 (7%)
13: RB1 and DIS3 (45%)
14q: TRAF3 (38%)
16q: CYLD and WWOX (35%)
17p: TP53 (8%)

Reprinted with permission from Morgan GJ, Walker BA, Davies FE: The genetic architecture of multiple myeloma, Nature
Reviews 12:335-348, 2012..
ADME, absorption, distribution, metabolism, and excretion; ANP32E, acidic (leucine-rich) nuclear phosphoprotein 32 family, member E;
BIRC, baculoviral IAP repeat-containing protein; BLIMP1, B lymphocyte-induced maturation protein 1; CCND, cyclin D; CDCA7L, cell
division cycle-associated 7-like; CDKN, cyclin-dependent kinase inhibitor; CKS1B, CDC28 protein kinase regulatory subunit 1B;
CYLD, cylindromatosis; DNAH, dynein, axonemal, heavy chain; DNMT3A, DNA methyltransferase 3α; DTNB, dystrobrevin, beta;
FAF1, FAS-associated factor 1; FAM46C, family with sequence similarity 46, member C; FGFR3, fibroblast growth factor receptor 3;
HOXA9, Homeobox A9; IGH@, immunoglobulin heavy chain locus; IRF4, interferon regulatory factor 4; I-ĸB, inhibitor of nuclear factor
ĸB, KDM, lysine demethylase; LRRK2, leucine-rich repeat kinase 2; LTBR, lymphotoxin beta receptor; MGUS, monoclonal gammopathy
of undetermined significance; MLL, mixed-lineage leukemia; MMSET, multiple myeloma SET domain; MRE11A, meiotic recombination
11A; NFĸB, nuclear factor ĸB; PARP1, poly (ADP-ribose) polymerase 1; TP53, TRAF3, TNF receptor-associated factor 3;
TRAK1, trafficking protein, kinesin binding 1; ULK4, unc-51 like kinase 4;WWOX, WW domain-containing oxidoreductase; XBP1, X
box-binding protein 1.

and immunoglobin gene translocations represent early deaminase (AID). As a result of these translocations,
molecular events in plasma cell neoplasia, and the major- expression of partner genes is placed under the control
ity of cases of MGUS harbor one of these abnormalities. of the immunoglobulin gene enhancer, with associated
In addition to these major molecular categories, inher- enhanced expression of the partner gene product. The
ited single nucleotide polymorphisms and monosomy or most frequent gene partners in these translocations are
partial deletion of chromosome 13 are recognized as ini- CCND1, CCND3, MMSET, FGFR3, MAF, MAFB, and
tiating events (Table 12-4 and Fig. 12-18).36 MMSET/FGFR (Table 12-4 and Figs. 12-19 and 12-20).
Early chromosomal translocations in MGUS and CCND genes encode D cyclins involved in cell cycle
plasma cell myeloma most frequently involve the immu- progression. Overall, these translocations are associated
noglobulin heavy chain gene IGH@ on chromosome 14, with worse prognosis, with the exception of t(11;14)
and less frequently involve lambda light chain gene IGL@ IGH@/CCND1 (Fig. 12-19), which confers a better
on chromosome 22. These translocations are thought to prognosis.46
arise from aberrant class switch recombinations or Mutations associated with disease progression and
somatic hypermutations mediated by activation-induced proliferation are additional genetic gains and losses,

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 837

Rare de novo MM

Germinal center Intramedullary Extramedullary


MGUS
B cell myeloma myeloma

Karyotype and epigenetic abnormalities


Nonhyperdiploid
1ry Ig tx
Secondary (Ig) TLC
11q13
6p21 NF-κB - activating mutations
16q23
PIsK/AKT dysregulation: DEPTOR, PTEN, PIK3CA
20q11
4p16
other

DEL 13

Protein translation/RNA processing


Trisomy FAM46C, DIS3, XBP1, LRRK2 mutation
3, 5, 7,
9, 11,15, N-RAS
Hyperdiploid

19, 21 K-RAS, FGFR3, BRAF


MYC (Ig) rearrangement
↑MYC RNA P18, RB inactivation
Cyclin D dysregulation ± MYC rearrangement p53 inactivation
? BLIMP1, IRF4 mutation

Definite
Uncertain TR1 TR2
FIGURE 12-18  ■  Model for molecular pathogenesis of monoclonal gammopathy of undetermined significance and plasma cell
myeloma. The initial transition (TR1) to a recognizable tumor involves two mostly nonoverlapping pathways (IGH translocations
versus multiple trisomies) that include primary events associated with disregulated cyclin D expression in monoclonal gammopathy
of undetermined significance (MGUS) and multiple myeloma (MM). The transition from MGUS to MM (TR2) is associated with
increased MYC expression and sometimes with activating mutations of K-RAS or chromosome 13 deletion. Early and late progres-
sion events for symptomatic MM tumors are shown. (Modified and reprinted with permission from Kuehl WM, et al: Molecular patho-
genesis of multiple myeloma and its premalignant precursor. J Clin Invest 122:3456-3463, 2012.)

secondary translocations involving immunoglobulin microRNAs 15a and 16 are downregulated in myeloma.
genes, translocations involving MYC, epigenetic changes, When present at normal levels, microRNAs 15a and 16
and mutations resulting in dysregulation of various sig- are thought to decrease BCL-2 expression, inhibit the
naling pathways (Table 12-4 and Fig. 12-18). NFκB is an NFkB pathway, and inhibit angiogenesis.54 Levels of cir-
example of a key pathway that is constitutively activated culating microRNAs have also been shown to have prog-
as a result of both activating and inactivating mutations nostic and theragnostic significance in myeloma.7
of the regulators of this pathway. NFκB activation is In addition to being secreted, microRNAs, cytokines,
important for cell survival.8 and other proteins can also be directly transferred from
Genome sequencing of 38 cases of plasma cell myeloma bone marrow stromal cells and internalized by myeloma
detected 23 translocations and 35 point mutations that cells via exosomes, small membrane bound vesicles. In
resulted in an amino acid changes. These mutations were particular, a recent study found that bone marrow stromal
detected in several classes of genes with various functions cells in plasma cell myeloma produced exosomes contain-
and included genes known to be mutated in plasma cell ing increased interleukin-6 (IL-6) and decreased microR-
myeloma as well as several genes in which mutations had NA15a relative to normal bone marrow stromal cells.
not previously been detected. These mutations included The study demonstrated transfer of these altered exo-
oncogenes and genes involved in RNA processing, somes from the mesenchymal cells to the myeloma cells.53
protein homeostasis, histone modification, and cell sig- The mechanism by which microRNA 15a is decreased is
naling. Frequently mutated genes included TP53, NRAS, unknown; however, a recent sequencing study detected
KRAS, and genes involved in the NFκB signaling pathway. mutations of DIS3, an exosome-based RNase, in approxi-
Although not common, mutations of BRAF were also mately 10% of myeloma samples.59
detected in 4% of patients.13 The neoplastic plasma cells in plasma cell myeloma
Several microRNAs are upregulated or downregulated share complex interactions with constituents of the
in myeloma and other neoplasms.17,194 For example, bone marrow microenvironment, including bone marrow

ERRNVPHGLFRVRUJ
838 12  Hematopoietic Tumors

Chr 11 Chr 14 Chr der(11) Chr der(14)


15.5 13 Reciprocal
15.4 12 product
15.3
15.1 15.2 11.2
11.1 11.1
14 11.2
13 12
12 13
11.2 11.12
11.11 21
11
12 13.1 22
13.2 CCND1 23
24.1
13.4 13.3 24.2
14.1 13.5 24.3
14.3 14.2 31
22.1 21 32.1 IgH Enhancer
22.2 32.2 32.3
22.3
23.2 23.1 CCND1
IgH REGION
23.3 Fusion
24
25 product

CCND1

Exon 1

Exon 2
Exon 3

Exon 4

Exon 5
69455872

69469242
Chr 11
cen tel
B 11q13 breakpoints

IgH REGION
Constant JH ∼20kb DH ∼50kb VH ∼900kb
region IgH Enhancer (1-4) (1-12) (1,2,...,n)
Chr 14
14q32 breakpoint
Exon 1

Exon 2
Exon 3

Exon 4

Constant Exon 5
region IgH Enhancer

cen tel

Fusion product t(11;14)(q13;q32)


C
FIGURE 12-19  ■  Translocation t(11; 14)(q13;q32). A, Chromosomal diagrams depicting reciprocal translocation involving fragments
of the q arm of chromosome 11 with a breakpoint at q13 and a small fragment of q arm of chromosome 14 with a breakpoint at
q32. The breakpoint on chromosome 11 involves the CCND1 locus and juxtaposes this gene in front of the IgH on chromosome 14.
B, The genomic structure of the involved genes with the position of the most frequent breakpoints. The breakpoints with CCND1
occur centromerically to exon 1; the breakpoints within the IgH region involve the region telomeric to the IgH enhancer. C, The
resulting hybrid gene contains the entire reading frame of CCND1 regulated by the strong IgH enhancer.

matrix, stromal cells, endothelial cells, T cells, dendritic and survival, such as IL-6, insulin growth factor-1, B-cell
cells, and osteoclasts and osteoblasts and their precursors activating factor, and a proliferation inducing ligand.36
(Fig. 12-21). These interactions can be categorized as IL-6 is one of the key cytokines produced by both
adhesion-mediated and cytokine-mediated. myeloma cells and the cells of the bone marrow microen-
Adhesion molecules play a role in homing and local- vironment. IL-6 promotes proliferation and blocks apop-
ization of myeloma cells to the bone marrow and mediate tosis of myeloma cells. IL-6 is secreted by bone marrow
the direct cell interactions between myeloma cells and stromal cells, osteoclasts, and osteoblasts. The normal
the constituents of the bone marrow microenvironment. homeostasis of bone formation and resorption is disrupted
Two adhesion molecules expressed by myeloma cells are in plasma cell myeloma and is mediated by cytokines such
syndecan 1 (CD138) and neural cell adhesion molecule as macrophage inflammatory protein-1α (MIP-1α) pro-
(CD56). Myeloma cells that are bound to bone marrow duced by plasma cells and receptor activator of nuclear
constituents demonstrate resistance to chemotherapy.31 factor κB ligand (RANKL) produced by marrow stromal
The binding of plasma cells to the stromal cells via adhe- cells. Increased MIP-1α and RANKL promote osteo-
sion molecules results in upregulation of the secretion of clast differentiation and activation. Increased Dickkopf-
multiple cytokines that enhance plasma cell proliferation related protein 1 (DKK1) produced by myeloma cells and

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 839

FGFR3

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17

Exon 18
Exon 1
Exon 2

Exon 3
Exon 4

Exon 5
Exon 6
Exon 7

Exon 8

Exon 9
Chr 4
Chr 14
MMSET FGFR3
13
16 12
15.3 11.2
15.2
15.1
11.1 11.1 FGFR3
11.2
14 12 1 806 aa
13 13 39 112 151 248 253 358 461 777
12 11
11 21
12 Immunoglobuline-like domain Protein kinase, catalytic domain
13.1 22
13.3 13.2 23
21.1 24.1
21.2 24.2
21.3 24.3
22 31
23 32.1
24 32.2
25 32.3
26 IgH REGION
27
MMSET
28 t(4;14)

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17
Exon 18

Exon 19
Exon 20
Exon 21
Exon 22
31.1

Exon 2
Exon 3

Exon 4
Exon 5

Exon 6
Exon 7
Exon 8
Exon 9
Exon 1
31.2
31.3
32
33
34
35
A MMSET
1 1365 aa
219 287 433 522 632 713 816 891 943 1011 1062 1186 1205 1296

PWWP (Pro-Trp-Trp-Pro) AWS domain Zinc Finger domain


HMG (High Mobility Group) SET domain
B

P P P P P*
Chr14q32 Em Im Sm Sg g 3’Ea Chr4p16 FGFR3 1 3 MMSET

Switch recombination
t(4;14)
P DJ P P P P*
Chr14q32 Em Im Sm Im Ig Sg g 3’Ea der(4) Em Im Sm 3 MMSET

C P P
der(14) FGFR3 1 Sg g 3’Ea

Eµ - IgH enhancer P - promoter


Eα - 3’IgH enhancer P* - cryptic promoter
D
FIGURE 12-20  ■  Translocation t(4;14)(p16.3;q32.2) in plasma cell myeloma involving the IgH locus. A, Chromosomal diagrams depict-
ing reciprocal translocation involving small fragments of the p arm of chromosome 4 with a breakpoint at p16.3 and a fragment of
q arm of chromosome 14 with a breakpoint at q32.2. The translocation is undetectable by conventional cytogenetic analysis.
B, Genomic and protein structures of the FGFR and MMSET genes involved t(4 : 14) and mapping to 4p16.3. The breakpoints on
chromosome 4 at p16.3 involve two alternative genes. They can occur in the region mapping to the FGFR3 or MMSET genes.
C, Translocations involving an immunoglobulin (Ig) locus—an early event that dysregulates an oncogene by juxtaposing it near a
strong Ig enhancer—are important in many B-cell neoplasms. The translocation breakpoints in the non-Ig chromosome (chr.) are
usually mediated by unknown mechanisms, but the location of the breakpoints in the Ig loci indicates involvement of three B-cell
mechanisms that generate double-stranded DNA breaks while remodeling the Ig loci during development. VDJ recombinase break-
points occur close to sites that are involved in VDJ recombination, somatic hypermutation breakpoints occur in the mutation-
susceptible region downstream of Ig promoters, and Ig heavy chain (IgH) switch breakpoints occur within or very near repetitive
switch regions that are located upstream of each IgH constant region. Breakpoints that are mediated by VDJ recombination or
somatic hypermutation occur telomeric to the intronic IgH enhancer (Eµ), which remains associated with the 3′ IgH enhancer(s) (Eα)
located downstream of one or both of the α-IgH genes on the derivative of chromosome 14 [der(14) ]. By contrast, breakpoints that
are mediated by switch recombination dissociate Eµ and Eα, so that distinct genes can be dysregulated by an IgH enhancer on each
derivative chromosome. During normal IgH switch recombination from µ (line 2) to γ, there is formation of a hybrid µ/γ switch region,
with deletion of intervening sequences (line 1). D, Similarly, a translocation that is mediated by an error in IgH switch recombination
generates separate double-stranded DNA breaks in µ and γ switch regions. These two ends are joined to the two ends created by
a third double-stranded break in another chromosome (4p16.3 in line 3), again with loss of intervening sequences. In this way, the
intronic enhancer (Eµ) on der(4) becomes juxtaposed to MMSET, which encodes a nuclear SET domain protein. This results in hybrid
mRNA transcripts that initiate from the JH and Iµ promoters, but also from a cryptic promoter (P*) on 4p16. The 3′ enhancer (3′ Eα)
on der(14) dysregulates the expression of FGFR3 (fibroblast growth factor receptor 3), and sometimes results in reciprocal hybrid
mRNA transcripts between the noncoding exons of MMSET and a constant region (γ). (Based on and adapted from data from Kalff A,
et al: The t4;14) translocation and the FGFR3 overexpression in multiple myeloma: prognostic implications and current clinical strategies.
Blood Cancer J 2, e89; doi:10.1038/bcj.2012.37, 2012 and Dalton WS, et al: Multiple myeloma. Hematology Am Soc Hematol Educ Program
157–177, 2001. Kuehl WM, et al: Multiple myeloma: evolving genetic events and host interactions. Nature Reviews Cancer 2:175–187,
2002.)

ERRNVPHGLFRVRUJ
840 12  Hematopoietic Tumors

osteoblast OPG osteoclast

MIP-1

DKK1
IL-6
IL-6
6
RANKL
RANK
RANKL
IL-3

adhesion m
Dysregulated ole
cu
les
MYC
Hypoxia HIF-1α
IGF-1
MM
IL-6
IL 6

BAFF/APRIL
cell
T cell

RANKL stromal cell


VEGF IGF-1

Endothelial cell
Endothelial
FIGURE 12-21  ■  Interactions of plasma cell myeloma tumor cells with the bone marrow microenvironment. Five kinds of cells in the
bone marrow (BM) microenvironment are depicted, as well as a few of the complex interactions among these cells and myeloma
plasma cells. Some critical survival and growth factors, such as interleukin-6 (IL-6), are made by more than one kind of BM cell.
External stimuli such as hypoxia and internal signals resulting from dysregulated MYC, stimulate HIF-1α and VEGF secretion, which
in turn stimulate endothelial cells to secrete insulin-like growth factor-1 (IGF-1). The hallmark uncoupling of bone remodeling is
partially explained by an increase in osteoclast activity (mediated by RANKL/RANK interactions, decreased osteoprotegerin [OPG],
and increased MIP-1α) and a decrease in osteoblast activity (mediated by DKK1 and IL-3). The resultant increase in osteoclast activity
stimulates the survival and growth of MM cells, at least partially by increased IL-6. Potential therapeutic agents that directly inhibit
some of these interactions include bisphosphonates (which inhibit osteoclast function), anti-RANKL antibody, anti-DKK1 antibody,
and exogenous OPG. (Modified and reprinted with permission from Kuehl WM, et al: Molecular pathogenesis of multiple myeloma and its
premalignant precursor. J Clin Invest 122:3456–3463, 2012.)

decreased osteoprotegerin result in decreased osteoblast chain fragments. AL amyloid is most frequently depos-
differentiation and activation. This disruption results in ited in the bone marrow, kidneys, gastrointestinal tract,
net loss of bone and the characteristic osteolytic lesions liver, spleen, muscle (e.g., tongue), skin, and nervous
seen in plasma cell myeloma.36 system. The involvement of the kidneys, the heart, or
Increased vessel density supports tumor growth and is both is most ominous. Involvement of the heart muscle
associated with poor prognosis in plasma cell myeloma. occurs in 60% of primary amyloidosis, and patients with
Vascular endothelial growth factor (VEGF) is produced amyloidosis-related congestive heart failure have a
by myeloma cells, marrow stromal cells, and vascular median survival of approximately 6 months, compared to
endothelial cells. VEGF promotes vascular proliferation a median survival of 2 years for primary amyloidosis as a
and also upregulates secretion of IL-6 from marrow group.39 The t(11;14) is more common in primary amy-
stromal cells. VEGF secretion is upregulated by MYC loidosis than in plasma cell myeloma as a group.22
expression and hypoxia inducible factor-1α (HIF-1α).36
Distinct Forms of Myeloma
Amyloidosis Associated with
The majority of myelomas present with a triad of
Multiple Myeloma obvious radiologic, laboratory, and pathologic findings,
Amyloidosis with significant impairment of the functions but in a small percentage of cases, the presentation may
of involved organs is present in approximately 10% of significantly deviate from this pattern. These distinct
patients with plasma cell myeloma.39 These cases repre- forms of myeloma are designated as solitary plasmacy-
sent primary amyloidosis (also known as AL or immuno- toma of bone, extramedullary plasmacytoma, nonsecre-
globulin light chain amyloidosis), in which clonal plasma tory myeloma, plasma cell leukemia, and osteosclerotic
cells secrete AL amyloid consisting of light chains or light myeloma (POEMS).

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 841

A B C
FIGURE 12-22  ■  Solitary plasma cell myeloma. A, Lateral radiograph of tibia shows large, expansile lytic lesion in diaphysis with
pathologic fracture of midshaft. B and C, T1- and T2-weighted magnetic resonance images show replacement of marrow by tumor
mass and fracture line extending from anterior cortex to posterior surface.

Solitary Plasmacytoma of Bone.  Solitary plasmacy- bone have longer overall survival than plasma cell
toma of bone is a localized form of neoplastic plasma cell myeloma patients with typical presentation.60
proliferation accounting for less than 5% of plasma cell The treatment of solitary plasmacytoma of bone is
neoplasms.60 It represents a clonal neoplastic prolifera- primarily local and consists of radiation therapy with
tion of plasma cells that produces a solitary destructive surgical stabilization when needed.25
bone lesion as seen on a radiologic skeletal survey (Fig.
12-22) but is otherwise without bone marrow involve- Extramedullary Plasmacytoma.  Extramedullary plas-
ment or features of end-organ damage (CRAB) (Table macytoma is a localized neoplastic plasma cell prolifera-
12-2).38 The natural history of this disease is somewhat tion otherwise without bone marrow involvement or
unpredictable but is distinct from ordinary multiple features of end-organ damage (CRAB) (Table 12-2). It
myeloma. Patients usually have levels of monoclonal accounts for less than 5% of plasma cell neoplasms.60 The
gammopathy lower than 3 g/dL. Approximately 25% of majority of extramedullary plasmacytomas develop in the
patients with solitary myeloma have no M component in upper respiratory tract but have been described in virtu-
the serum by electrophoresis. An abnormal serum free ally every organ.25,33,35,50
light chain ratio can be detected in more than half of the Extramedullary plasmacytoma has a better prognosis
patients with no M component by electrophoresis. than solitary plasmacytoma of bone. Head and neck plas-
Solitary plasmacytoma of bone most frequently affects macytomas have a more favorable prognosis. The recur-
vertebral bodies (approximately 50% of cases). The other rence rate is less than 10%. From 30% to 50% of patients
typical sites of involvement are the pelvis, femur, and will progress to plasma cell myeloma but have longer
humerus.62 Radiographically, the lesion presents as a survival (50-80% 10-year survival) than plasma cell
lytic and destructive process with minimal or no blastic myeloma patients with typical presentation.34
reaction.
From 65% to 84% of patients with solitary plasmacy- Nonsecretory Myeloma.  In approximately 5% of
toma of bone will progress to plasma cell myeloma within patients with multiple myeloma, no monoclonal protein
5 years, and 65% to100% will progress to myeloma in 10 can be demonstrated in the serum or urine by electro-
years.34 After progression to plasma cell myeloma, patients phoresis. By serum free light chain assay, less than 3% of
with the initial presentation of solitary plasmacytoma of patients have undetectable monoclonal protein.42 These

ERRNVPHGLFRVRUJ
842 12  Hematopoietic Tumors

myelomas are referred to as nonsecretory myelomas.21 In the Histologically, the bone lesions are composed of sheets
majority of these cases, immunohistochemical studies of plasma cells similar to those seen in typical myeloma.
reveal monoclonal immunoglobulin synthesis restricted In contrast to typical plasma cell myeloma, the nonle-
to the light or heavy chains.32 This indicates that the sional bone marrow in POEMS syndrome typically con-
tumor cells retain the ability to synthesize immunoglobu- tains less than 10% plasma cells and may be completely
lin but the products cannot be secreted extracellularly.42 normal in approximately 10% of cases. The bone marrow
Rare cases (approximately 15%) show no evidence of in the majority of cases shows megakaryocyte clustering.
immunoglobulin production by immunohistochemis- In approximately half of the cases, the marrow shows
try.38 In one nonsecretory myeloma patient, a κ light megakaryocyte hyperplasia or lymphoid aggregates sur-
chain frameshift mutation was detected that prevented rounded by light chain restricted, usually λ-expressing,
linking of the immunoglobulin chains.14 Some series have plasma cells.16 The immunohistochemical features are
reported better prognosis in nonsecretory myeloma, identical to those described for plasma cell myeloma.
while others report statistics similar to conventional The cell of origin is typically a λ light chain-expressing
plasma cell myeloma.56 These discrepancies may be due plasma cell. The majority of the clinical manifestations
to varying cytogenetic findings that were not investigated are thought to be mediated by cytokines. VEGF pro-
specifically. In one series of patients with IgM or IgD duced by platelets and the clonal plasma cells is elevated
nonsecretory myeloma, 83% harbored t(11;14), which in all cases of POEMS syndrome. IL-6, tumor necrosis
may account in part for the better prognosis reported in factor-alpha (TNFα), and IL-1β are also elevated.
some series.9,42 POEMS syndrome is treated with radiation or chemo-
therapy. Radiation therapy at the site of bone lesions is
Plasma Cell Leukemia.  Plasma cell leukemia is diag- the treatment of choice for patients with two or fewer
nosed when more than 20% of the white blood cells in bone lesions without other clonal bone marrow plasma-
the peripheral blood represent neoplastic plasma cells or cytosis. Systemic chemotherapy is given to patients with
the absolute number of tumor plasma cells exceeds 2 × three or more bone lesions and to any patient with clonal
109/L.52 Plasma cell leukemia typically presents as a late bone marrow plasmacytosis. The median overall survival
complication of advanced multiple myeloma. It is seen in is approximately 14 years.20
approximately 2% to 5% of terminal multiple myeloma
cases.52 In extremely rare cases, plasma cell leukemia may
be present de novo.43 In all instances, plasma cell leuke-
mia heralds a particularly aggressive form of myeloma NON-HODGKIN’S LYMPHOMA
that is associated with massive replacement of the bone Definition
marrow and poor survival (usually <6 months).18,24,49,63
Plasma cell leukemia is more frequently observed in Primary non-Hodgkin’s lymphoma of bone is a prolifera-
IgE, IgD, and light chain only myelomas.55,61 Plasma tion of malignant lymphocytes classified according to its
cell leukemia cells often exhibit an immature blastic extraskeletal counterparts.68,73 Although there is no uni-
morphology.12 versal definition, by convention, an interval of 4 to 6
months between skeletal manifestation of the lesion and
Osteosclerotic Myeloma (POEMS).  POEMS syn- the development of extraskeletal disease is required for
drome is a paraneoplastic syndrome caused by a clonal the lesion to be considered a primary tumor in bone. In
plasma cell population. Historically, the clinical manifes- every case of lymphoma presenting as a bone lesion, the
tations, as represented by the acronym, were polyneu- appropriate staging procedure is required to rule out the
ropathy, organomegaly, endocrinopathy, monoclonal presence of extraskeletal disease.
protein in serum or urine, and skin changes.10,28 However, Historically, the description of lymphoma as a primary
the original components of the syndrome as listed do lesion in bone is credited to Oberling, although he did
not always manifest. Mandatory major criteria required not make the distinction between true lymphomas and
for the diagnosis of POEMS are polyneuropathy and Ewing’s sarcoma.97 The recognition of primary lym-
the presence of a clonal plasma cell proliferation. Λ phoma of bone as a clinicopathologic entity distinct from
light chain is expressed in more than 95% of cases. A other lymphomas and especially from Ewing’s sarcoma
definitive diagnosis requires meeting at least one addi- was made by Parker and Jackson in 1939.99 Two parallel
tional major criterion (Castleman disease, one or more classifications based on the functional and phenotypic
osterosclerotic lesion, or vascular endothelial growth features of tumor cells as delineated by marker studies
factor [VEGF] elevation) and one minor criterion were proposed by Lukes and Collins in the United States
(organomegaly, extravascular volume overload, endocri- and by Lennert in Germany (Kiel classification) in 1975
nopathy, skin changes, papilledema, or thrombocytosis and 1974, respectively.112,115 The simplified Revised
and polycythemia).20 Working Formulation105,106 was published in 1982 in an
Greater than 95% of patients with POEMS syndrome effort to bridge the gap between morphologic and clinical
have one or more bone lesion, typically sclerotic, but aspects of lymphoma classification, combining some pre-
often mixed lytic and sclerotic or lytic with a sclerotic rim vious descriptive terms, mainly from the Rappaport
as visualized by conventional radiographs. Some authors system, with the biologic concepts and terminology pro-
consider 18F-fluorodeoxyglucose (FDG) PET/CT to be posed by Lukes and Collins.
a better method for visualizing osteosclerotic lesions and The Revised European-American Classification System
assessing their degree of activity.45 of Lymphoid Neoplasms (REAL Classification) proposed

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 843

Incidence

40

1 2 3 4 5 6 7 8
Age in decades

FIGURE 12-23  ■  Epidemiology of non-Hodgkin’s lymphoma of bone. Peak age incidence and frequent skeletal sites of involvement.
Most frequent sites are indicated by solid black arrows.

by the International Lymphoma Study Group74,85 was The peak age incidence and most frequent sites of
published in 1994 and represented a consensus effort to primary bone lymphoma are shown in Figure 12-23. The
define the known distinct hematolymphoid neoplasms incidence of primary bone lymphoma is rising (Fig.
in terms of clinical, morphologic, immunophenotypic, 12-24). More than 50% of primary lymphomas in bone
and molecular features. The REAL classification was the occur in patients older than age 60 years (Fig. 12-25)87,102
starting point for 7 years of efforts to build the now There is a slight male predominance in several major
generally accepted consensus classification, the World series, with a male-to-female ratio of approximately 1.2 : 1
Health Organization (WHO) Classification of Tumours (Fig. 12-26).87 The vertebrae, femur, and pelvis are the
of Haematopoietic and Lymphoid Tissues.111 most frequently involved bones, accounting for approxi-
mately 29%, 12%, and 13% of lesions, respectively. The
humerus, ribs, and skull are next in frequency, and each
Diffuse Large B-cell Lymphoma is involved in approximately 10% of cases.102 In general,
non-Hodgkin’s lymphomas have a predilection for the
Clinical Features
trunk bones, including the ribs, sternum, and clavicle.
Primary lymphoma of bone is rare and accounts for 3% The major long tubular bones, such as the femur and
to 7% of bone tumors and less than 2% of adult lympho- humerus, are the most frequently involved sites in the
mas overall.79,88 Because of the infrequency of primary appendicular skeleton. Lymphomas of bone often present
bone lymphoma and the disagreement as to its clinical as multifocal lesions. The multifocality can be in the form
definition, there have been few published series docu- of several foci within one bone, or several bones can be
menting patient outcomes, clinical characteristics, and simultaneously affected. The rate of multifocal lesions is
biological aspects. The following discussion focuses on approximately 15%.87
diffuse large B-cell lymphoma and is based primarily on Pain is the most common symptom. Other symptoms,
three published large series of primary lymphoma of such as nerve compression, are related to the location of
bone including several histologic subtypes; however, in the tumor. Local tenderness, redness, and swelling may
these series, at least 70%, 79%, and 83% of the cases be present. Pathologic fracture can be the initial symptom.
were classified as diffuse large B-cell lymphoma.66,87,102 In contrast with symptoms of Ewing’s sarcoma and

ERRNVPHGLFRVRUJ
844 12  Hematopoietic Tumors

0.2

0.15
(cases/100,000 persons)
Incidence rate

0.1

0.05

0
73

75

77

79

81

83

85

87

89

91

93

95

97

99

01

03

05
19

19

19

19

19

19

19

19

19

19

19

19

19

19

20

20

20
FIGURE 12-24  ■  Incidence of non-Hodgkin’s lymphoma of bone. A general increase in incidence of cases from 1973 to 2006, similar
to the increased incidence of lymphoma of all sites over this time period. SEER Data, 1973-2006.

12

10
Age distribution (%)

0
9

4
9

+
1

-4

-5
0-

1-

5-

-1

-1

-2

-2

-3

-3

-4

-5

-6

-6

-7

-7

-8

85
50
10

15

20

25

30

35

40

45

55

60

65

70

75

80

FIGURE 12-25  ■  Age distribution of non-Hodgkin’s lymphoma of bone. The peak age of incidence is age 75 to 79 years. There is a
significant increase in incidence after age 45. SEER Data, 1973-2010.

generalized lymphoma, fever is uncommon. Features of localized bone involvement. Conflicting results have
generalized systemic disease, such as lymphadenopathy been published, with no clear advantage to radiation
and hepatosplenomegaly, are not present at presentation versus chemotheraphy alone versus combined chemo-
by definition. therapy and radiation therapy.66
Radiation therapy and chemotherapy are the primary The most important prognostic factors for primary
treatments for non-Hodgkin’s lymphoma of bone.69,78,104 lymphoma of bone are age at diagnosis and stage of
There is no consensus as to the optimal therapy for disease. Patients under age 30 years have high rates of

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 845

0.25

0.2

(cases/100,000 persons)
Incidence rate
0.15

0.1

0.05

es

es

es

es
al

al

al

al

al

al
,m

em

m
fe

fe
es

,f

te

k
ac
es
ac

hi

te

k
ac
Bl
W
ac

hi
lr

Bl
W
Al

lr
Al
FIGURE 12-26  ■  Non-Hodgkin’s lymphoma of bone. Distribution of non-Hodgkin’s lymphoma by race and sex. The most commonly
affected group is white males. SEER Data, 1973-2010.

remission, with approximately 75% survival at 400 PET/CT is useful in staging and confirming disease
months versus 0% survival in patients age 60 years or limited to the bone.96
older. Patients with lymphoma limited to the bone have
better survival than systemic lymphoma. The 5- and Gross Findings
10-year survival rates for primary bone lymphoma are
65% and 53%, compared with 53% and 43% for patients On gross examination, diffuse large B-cell lymphoma of
with systemic lymphoma in addition to bone masses.87 bone resembles its nodal counterpart and presents as a
pink-tan or gray-white and fleshy lesion (Fig. 12-29).
Areas of necrosis, hemorrhage, and cystic degeneration
Radiographic Imaging
may be present. The cortex and the bone at its periphery
Radiographic features of non-Hodgkin’s lymphoma are show patchy erosions and permeation of the medullary
not specific and overlap with other small cell tumors. cavity. Complete cortical disruption and extension into
They usually do not permit a specific diagnosis. Typically, soft tissue are frequently present. Large masses may show
non-Hodgkin’s lymphoma presents as a lytic destructive sharp demarcation at their peripheries. Reactive sclerosis
lesion with a permeative or “moth-eaten” pattern (Fig. presents as firm ivory-like areas and may occasionally
12-27).76,101 Prominent periosteal new bone formation occupy a significant portion of the tumor.
with multiple concentric or so-called onion-skin layers
may be present but is usually less evident than in Ewing’s Microscopic Findings
sarcoma. In long bones, the shaft is preferentially
involved, and disease may extend to the end of the bone. The conventional diffuse growth pattern with solid pro-
A lytic lesion with periosteal reaction at or near the end liferation of round tumor cells permeating the bone
of a long bone, while not specific, raises the index of marrow spaces and haversian canals is most frequently
suspicion for lymphoma (Fig. 12-28).89 Non-Hodgkin’s seen. Sclerosing diffuse growth is often focally present in
lymphoma is usually lytic, but in some cases it provokes bone. In this pattern, the tumor cells are separated by
bone sclerosis and presents as a blastic lesion. Rarely, dense fibrous bands and can form nests, cords, or organ-
plain radiographs may show little or no abnormality.96 oid structures. Occasionally, they may have spindle cell
Computed tomography or MRI are helpful in evaluat- morphology and grow in storiform structures (Fig.
ing the extent of involvement within bone, to detect 12-30). Compressed tumor cells may also develop a spin-
cortical erosion and soft tissue extension of tumor, as well dled morphology. The presence of reactive lymphocytes
as to evaluate the extent of involvement of vertebral and can alter the appearance of these lesions. Deviations from
paraspinous lesions. MRI is particularly useful for detect- the classic lymphoma morphology can lead to an errone-
ing marrow involvement, which is characteristic in bone ous diagnosis of an inflammatory condition or nonlym-
lymphoma. Bone marrow involvement may be detected phoid tumor, as in the case of the rare signet ring cell
by T1-weighted (low signal) or T2-weighted (bright variant of diffuse large B-cell lymphoma (Fig. 12-30).
signal) magnetic resonance images.96 A clue to the diag-
nosis is prominent marrow and soft tissue involvement
Immunohistochemical Stains and
with relatively minor cortical bone involvement.89
Differential Diagnosis
Radionuclide scintigraphy is helpful in documenting
additional foci of bone involvement, a common feature A variety of special studies may be used to supplement
of non-Hodgkin’s lymphomas in the skeleton. FDG the diagnosis of lymphoma of bone, including flow

ERRNVPHGLFRVRUJ
846 12  Hematopoietic Tumors

A B C
FIGURE 12-27  ■  Non-Hodgkin’s lymphoma of bone: radiographic features. A, Lateral radiograph of humerus of adult who complained
of pain that had been present for several months. Permeative bone destruction with slight expansion of contour is noted over long
segment of humeral diaphysis. Borders of lesion are poorly demarcated. B and C, Anteroposterior and lateral radiographs of lower
tibia and fibula of middle-aged adult show ill-defined destructive lesion in lower tibial shaft. Lesion has moth-eaten appearance.
Rectangular defect seen in B represents previous biopsy site. Biopsy material removed several months earlier was erroneously
interpreted as chronic osteomyelitis.

cytometry, classical cytogenetics, fluorescence in situ rhabdomyosarcoma, and metastatic small cell and poorly dif-
hybridization (FISH) studies for common translocations, ferentiated non–small cell carcinoma. The use of nonlym-
PCR for detection of translocations, and clonal immuno- phoma markers, such as cytokeratins, desmin, smooth
globulin gene rearrangement. Gene expression profiling muscle actin, and FLI1 (Ewing’s sarcoma), can help rule
is useful for subclassifying diffuse large B-cell lymphoma out other round-cell tumors.
but is still not widely used in clinical practice. Exome or A minimalist approach to suspected diffuse large B-cell
whole genome sequencing is becoming more widely lymphoma is initial evaluation of CD20 immunohisto-
available and likely will be one of the major tools used to chemical stain; however, more often a panel including
identify actionable genetic alterations, allowing design CD45 (leukocyte common antigen), CD20 (pan–B-cell
and application of regimens, including specific targeted marker), and CD3 (T-cell marker) is ordered, in addition
therapeutic agents. to other lineage markers if the differential diagnosis is
Immunohistochemistry remains a useful tool in the broad. The majority of untreated diffuse large B-cell
diagnosis and subclassification of diffuse large B-cell lym- lymphomas are positive for CD20. In cases of CD20-
phoma. Lymphoma must be distinguished from other negative diffuse large B-cell lymphoma, PAX5 or CD79
hematopoietic neoplasms, such as plasma cell myeloma, will usually be positive. Once a diagnosis of diffuse large
myeloid sarcoma, Langerhans cell histiocytosis, and masto- B-cell lymphoma is established, additional prognostic
cytosis. The differential diagnosis also includes nonhe- studies may be considered depending on the needs of the
matologic round-cell tumors such as Ewing’s sarcoma, patient and treating physicians. A typical follow-up panel

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 847

A B

C D
FIGURE 12-28  ■  Non-Hodgkin’s lymphoma of bone: radiographic features. A, Lateral radiograph of humerus showing a destructive
mass with extensive involvement of the adjacent soft tissue. B, Coronal magnetic resonance image (MRI) of same case as A showing
a destructive lesion involving the proximal humerus with extensive involvement of the adjacent soft tissue. C, Oblique radiograph
showing a destructive lytic lesion with soft tissue extension of the proximal ulna. D, Axial MRI of the same case as in C showing a
lesion of variable signal intensity involving the proximal ulna and adjacent soft tissue.

ERRNVPHGLFRVRUJ
848 12  Hematopoietic Tumors

A B

FIGURE 12-29  ■  Non-Hodgkin’s lymphona of bone: gross features. A and B, Sagittally bisected tibia shows extensive gray-tan mass
with extensive involvement of tibial shaft and adjacent soft tissue.

includes Ki-67 to evaluate proliferation rate, and CD10, scoring system, patients with at least 70% of lymphoma
BCL6, and MUM1 (with or without additional markers) cells positive for BCL2 and at least 40% of lymphoma
to subclassify diffuse large B-cell lymphoma as either cells positive for MYC had a worse prognosis when
germinal center subtype or activated B-cell subtype (also treated with conventional chemotherapy.82
see the discussion on genetic features and pathogenesis) Hematologic neoplasms are frequently included in the
(Figs. 12-31 and 12-32). In addition, Epstein-Barr encod- differential diagnosis of diffuse large B-cell lymphoma.
ing region (EBER) in situ hybridization may be consid- Multiple myeloma, especially the pleomorphic type, can
ered in patients older than age 50 years to confirm or resemble diffuse large B-cell lymphoma. Review of serum
exclude a diagnosis of Epstein-Barr virus (EBV)-positive and urine protein electrophoresis, if available, may help
diffuse large B-cell lymphoma of the elderly. distinguish myeloma from lymphoma. In addition, immu-
If the Ki-67 proliferation index is above 80%, some nohistochemical stains for κ and λ immunoglobulin
recommend FISH testing for translocations involving chains are negative in most diffuse large B-cell lympho-
BCL-2, BCL-6, and MYC, to identify possible “double mas and positive in the vast majority of plasma cell
hit” or “triple hit” lymphomas, which have BCL2 and/or myeloma cases.
BCL-6 translocations in addition to MYC translocations. Myeloid sarcoma may mimic diffuse large B-cell lym-
Double hit and triple hit lymphomas are particularly phoma. The most sensitive immunohistochemical stains
aggressive lymphomas with poor prognosis and poor for myeloid sarcoma are CD43 and lysozyme. The major-
response to conventional chemotherapy. Others question ity of diffuse large B-cell lymphomas are also positive for
the use of a Ki-67 proliferation limit as a guide to con- CD43 but are negative for lysozyme, CD34, and myelo-
sider FISH testing.92 An immunohistochemical score for peroxidase. Eosinophilic myeloblasts, when present, are
double hit lymphomas has also been described. In this a clue to the diagnosis of myeloid sarcoma. The myeloid

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 849

A B

C D
FIGURE 12-30  ■  Diffuse large B-cell lymphoma of bone: microscopic features. A, The tumor is composed of large lymphocytes with
variably prominent nucleoli, with inflammatory response and bone destruction (×200). B, The rare signet ring cell variant has lym-
phocytes with prominent cytoplasmic vacuoles and peripheral displacement and indentation of the nuclei (×400). C, Diffuse large
B-cell lymphoma showing large lymphocytes with one to multiple prominent nucleoli (×400). D, Diffuse large B-cell lymphoma with
bone destruction (×400). (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
850 12  Hematopoietic Tumors

A B

C D
FIGURE 12-31  ■  Diffuse large B-cell lymphoma of bone, germinal center type: microscopic and immunohistochemical features.
A, Photomicrograph of diffuse large B-cell lymphoma showing centroblastic and plasmacytoid features (×400). B, Photomicrograph
of diffuse large B-cell lymphoma stained for CD10 showing diffuse membranous staining (×200). C, Photomicrograph of diffuse large
B cell lymphoma stained for BCL-6 showing nuclear staining (×200). D, Photomicrograph of diffuse large B cell lymphoma stained
for MUM-1 showing fewer cells with nuclear staining (×200).

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 851

GCB involvement by lymphoma. The use of appropriate


Non-GCB
markers to document the epithelial nature of cells and
+ + frequent neuroendocrine differentiation, as demonstrated
CD10 MUM-1 by positivity for CD56, synaptophysin, crhomogranin,
and TTF1, in small cell carcinomas are helpful distin-
− + − GCB guishing features.
BCL-6 Non-Hodgkin’s lymphoma of bone is frequently mis-
− diagnosed as chronic osteomyelitis. This error can be
Non-GCB
avoided if attention is paid to the clinicoradiographic
FIGURE 12-32  ■  Hans classifier for diffuse large B-cell lymphoma features. The involvement of the shaft of long tubular
subtyping. Three immunohistochemical stains approximate bones in patients older than age 40 years is rarely a
gene expression profile-based subtyping of diffuse large B-cell feature of chronic osteomyelitis. The presence of reactive
lymphoma into germinal center subtype and non-germinal lymphocytes in malignant lymphoma is frequently
center subtype. GCB, germinal center B-cell subtype; non-GC,
non-germinal center B-cell subtype. (Modified and reprinted with responsible for this error. In this instance, the recognition
permission from Hans CP, et al: Confirmation of the molecular clas- of atypical lymphoid cells and the identification of the
sification of diffuse large B-cell lymphoma by immunohistochemis- phenotypic features consistent with lymphoma are the
try using a tissue microarray. Blood 103:275-282, 2004.) keys to the correct diagnosis.

Genetic Features and Pathogenesis


nature of the cells in question is often easier to recognize
on touch preparations stained with Wright-Giemsa. The normal cell counterpart is a mature B cell that has
Chloracetate esterase is inactivated by acid decalcifica- been exposed to antigen and undergone somatic hyper-
tion, and stains performed on decalcified tissue can mutation of immunoglobulin genes in the germinal
provide false-negative results. Immunohistochemical center. The normal cell counterpart may be a germinal
stains for lysozyme myeloperoxidase are useful in identi- center B cell (GCB) or post-germinal center B cell.
fying the myeloid nature of the tumor cells. The post-germinal center subtype of diffuse large B-cell
Langerhans cell histiocytosis can be easily distinguished lymphoma is commonly referred to as activated B cell
from lymphoma by the presence of a mixture of histio- (ABC) subtype. The neoplastic B cells show clonal rear-
cytic cells with a prominent eosinophilic infiltrate. Occa- rangement of immunoglobulin genes and show somatic
sionally, when Langerhans cells predominate, it can be hypermutation of immunoglobulin variable chain genes,
difficult to distinguish Langerhans cell histiocytosis from as is seen in normal germinal center and post-germinal
diffuse large B-cell lymphoma. The strong positivity of center B cells.
Langerhans cells for S-100, the younger age of the The molecular profile of diffuse large B-cell lym-
patients, and radiographic features usually help distin- phoma of bone is similar to that for the diffuse large
guish this disorder from lymphoma. B-cell lymphoma occurring at other sites. In 2000, Aliza-
Mastocytosis can be suspected if the entire clinical pre- deh et al. divided diffuse large B-cell lymphoma into two
sentation, the presence of skin lesions and systemic symp- subtypes on the basis of gene expression profiles and
toms, is taken into consideration. The mast cell nature of showed that diffuse large B-cell lymphoma of GCB has
the cells in question can be suspected if a round-cell a better prognosis than ABC type.67 The prognostic sig-
infiltrate is negative for common leukocyte and epithelial nificance of these subtypes has been confirmed by some
markers. Mast cells are positive for CD117 and mast cell studies and have been challenged by authors of other
tryptase, and their granules can be revealed in prepara- studies.86,107 In 2004, Hans et al. described a simple
tions stained with Giemsa, toluidine, or alcian blue. immunohistochemical panel (CD10, BCL6, and MUM1),
Diffuse large B-cell lymphoma may be differentiated now commonly named the Hans classifier (Fig. 12-32) that
from pediatric small round blue cell tumors such as loosely reproduced cDNA microarray-based subtyping
Ewing’s sarcoma/primitive neuroectodermal tumor (ES/ with a positive predictive value of 87% for GCB subtype
PNET) by evaluating an immunohistochemical panel, and 73% for ABCsubtype.84 The panel also predicted
including CD99, FLI1, and cytokeratin. CD99 is positive better survival for the lymphoma classified as GCB
in ES/PNET; however, 55% of myeloid sarcomas may subtype, similar to the gene expression profile classifica-
also be positive for CD99.118 ES/PNETs are negative for tion. Subsequently, several groups have proposed algo-
CD43 and lysozyme, positive for FLI1, and show cyto- rithms based on more extensive immunohistochemical
keratin expression in a subset of cases. Ewing’s sarcoma panels that slightly improve correlation between sub-
is extremely rare in patients older than age 40 years, an grouping based on gene expression-based and immuno-
age when most non-Hodgkin’s lymphomas are diagnosed. histochemical features.75,95
Multifocal skeletal lesions are extremely rare in Ewing’s There is a tremendous degree of genetic variation
sarcoma. On the other hand, multifocality is a frequent among diffuse large B-cell lymphomas, resulting in highly
feature of skeletal lymphomas. In addition, detection of variable response to current treatment regimens. It is
t(11;12) (q24,q12) by karyotyping, PCR, or FISH con- estimated that an individual case of diffuse large B-cell
firms a diagnosis of ES/PNET. lymphoma harbors between 30 and 100 different muta-
Metastatic small cell carcinoma occasionally can be very tions.100 A comprehensive discussion of molecular genet-
difficult to distinguish from lymphoma because it may ics in diffuse large B-cell lymphoma is clearly beyond the
have similar features, especially with sclerotic bone scope of this chapter. Table 12-5 lists common genetic

ERRNVPHGLFRVRUJ
852 12  Hematopoietic Tumors

DNA breakage that occurs during normal B-cell develop-


TABLE 12-5 Diffuse Large B-Cell Lymphoma:
ment. The mechanisms that produce immunoglobulin
Common Genetic Abnormalities
diversity via DNA breakage and reassembly include
Germinal Center Type Activated B cell Type variable, diversity, and joining [V(D)J] immunoglobulin
segment recombination, class switch recombination, and
BCL2 translocations TNFAIP3 mutations/deletions
[e.g., t(14;18)] MYD88 mutations somatic hypermutation of immunoglobulin genes. The
MYC translocations CD79A/CD79B mutations mediators of these processes, such as recombination acti-
EZH2 mutations CARD11 mutations vating genes (RAG, which is required for V(D)J recom-
MEF2B mutations BCL2 amplifications bination) and activation induced cytidine deaminase
BCL6 translocations/ BLIMP1 mutations/deletions
mutations CREBBP/EP300 mutations
(AID, which initiates somatic hypermutation and class
PTEN deletions MLL2/MLL3 mutations switch recombination) have been implicated in many of
CREBBP/EP300 mutations B2M mutations/deletions the translocations and point mutations seen in B-cell
MLL2/MLL3 mutations CD58 mutations/deletions lymphoma.83
B2M mutations/deletions BCL6 translocations The most common translocations in diffuse large
CD58 mutations/deletions
B-cell lymphoma involve the B-cell lymphoma 6 (BCL6)
Adapted from Pasqualucci L, et al: The genetic basis of diffuse gene on the long arm of chromosome three at 3q27 (Fig.
large B-cell lymphoma. Curr Opin Hematol 20:336-344, 2013. 12-33).98 Translocations involving BCL6 are present in
B2M, Beta2 microglobulin; BCL2, B cell CLL/lymphoma 2; BCL6, B 30% to 40% of diffuse large B-cell lymphomas and are
cell CLL/lymphoma 6; BLIMP1, B lymphocyte-induced protein 1;
CARD11, caspase recruitment domain-containing protein more common in the ABC type.100 In normal B cells,
11;CREBBP, cAMP response element-binding protein; EP300, BCL6 is expressed only during the time the B cell is
E1A binding protein p300; EZH2, enhancer of zeste 2 polycomb within the germinal center. BCL6 is a transcriptional
repressive complex 2 subunit; MEF2B, myocyte enhancer factor repressor that maintains germinal center B-cell functions
2B; MLL2/3 = mixed lineage leukemia 2/3; MYD88, myeloid
differentiation primary response 80; PTEN, phosphatase and
until the cells mature into post-germinal center memory
tensin homolog; TNFAIP3, tumor necrosis factor, alpha-induced B cells or plasma cells. BCL6 protein also represses TP53,
protein 3. which is a checkpoint gene and activator of DNA repair
mechanisms. The absence of TP53 promotes genetic
instability, allowing somatic hypermutation of immuno-
abnormalities in diffuse large B-cell lymphoma. In one globulin variable segment genes.91 In normal B-cell mat-
large study, exome sequencing of diffuse large B-cell lym- uration, somatic hypermutation produces mature memory
phomas detected recurrent mutations in 322 genes. The B cells and plasma cells that produce high affinity immu-
types of mutations included copy number alterations, noglobulin specific for a given antigen. In the setting of
intergenic mutations, and mutations of coding and regu- lymphoma, aberrant somatic mutation may be seen as a
latory regions of various genes. The types of genes primary event or may be associated with transformation
mutated included genes implicated in apoptosis, cell to a more aggressive lymphoma.
adhesion, cell cycle, cell differentiation, metabolism, epi- BCL6 can rearrange with more than 30 different gene
genetic modification, DNA repair, immune response, partners. The translocation partner is an immunoglobu-
membrane transport, protein modification, signal trans- lin gene in more than half of cases, with the remaining
duction, and ubiquitin cycle.117 translocation partners represented by nonimmunoglobu-
Frequent mutations are being discovered in genes pre- lin genes.64,91 The majority of the breakpoints for BCL6
viously not known to be affected in lymphomas. These gene rearrangement are located in major translocation
genes include epigenetic modifiers, genes involved in clusters, including exon 1 and an intronic sequence
immune surveillance, and genes that regulate signaling between exon 1 and exon 2 (Fig. 12-33). The result is
pathways (Table 12-5). Mixed lineage leukemia 2 (MLL2) translocation of a nearly complete BCL6 gene under the
is an example of a gene encoding a histone methyltrans- control of the promotors of several different gene part-
ferase that is mutated in more than 30% of diffuse large ners. The BCL6 breakpoints have sequences that are tar-
B-cell lymphomas. Inactivating mutations of β2 micro- geted by AID in normal immunoglobulin class switch
globulin (B2M) are seen in approximately 30% of diffuse recombination, and it is thought that these translocations
large B-cell lymphomas, resulting in the inability of cyto- occur in the germinal center under the influence of AID.
toxic T cells to recognize lymphoma cells.100 In addition, the BCL6 gene in normal B cells and lym-
Constitutive activation of nuclear factor κB (NFκB) is phoma undergoes somatic hypermutation, also thought
a frequent finding in diffuse large B- cell lymphoma, to be mediated by AID.91,100
particularly the ABC subtype.90 Mutations of several Another common translocation, seen in up to 40% of
regulatory genes may be implicated in the constitutive diffuse large B-cell lymphomas, is t(14;18)(q32;q21) (Fig.
activation of NFκB, including myeloid differentiation 12-34).116 This translocation involves the immunoglobu-
primary response 88 (MYD88, mutated in 30% of ABC lin heavy chain gene (IGH) on chromosome 14 and B-cell
diffuse large B-cell lymphomas), TNFα-induced protein lymphoma 2 (BCL2) on chromosome 18, resulting in
3 (TNFAIP3/A20, mutated in approximately 30% of ABC overexpression of BCL2 under the control of the IgH
diffuse large B-cell lymphomas), and caspase recruitment enhancer (Fig. 12-34). Increased expression of BCL2
domain-containing protein 11 (CARD11, mutated in 9% increases cell survival by inhibiting apoptosis. The trans-
of ABC diffuse large B-cell lymphomas).100 location alone is not sufficient to cause lymphoma. The
B-cell lymphomas are thought to harbor many muta- t(14;18) is present in small numbers of B cells in normal
tions at least in part due to mistargeting of physiologic germinal center B cells.109

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 853

Chr 3 BCL6
26
25

Exon 7

Exon 6

Exon 5

Exon 4

Exon 3

Exon 2

Exon 1
24.3 24.2
24.1 23

187442728

187452695
22
21.3
21.2 21.1
14.3 14.2 cen tel
14.1
13
12 BCL6
11.1 11.2

4
11.2 11.1
12
13.1 13.2
13.3 1 706 aa
21
22
23 Missense substitution
24 BTB/POZ fold domain
25.1 Nonsense substitution - Stop
25.2 25.3 Zinc finger, C2H2-like domain
26.1 Insertion frameshift
26.2 Zinc finger C2H2-type/integrase DNA-binding domain
26.3 27 Deletion frameshift
28 BCL6
29 Substitution - coding silent
A

MTC
Chr 3 5’ 1 2
3’
BCL6
1kb
S
breakpoint
hyper-cluster Xh S

200bp
B
FIGURE 12-33  ■  B-cell lymphoma 6 (BCL6): gene, protein, mutations, and gene rearrangement breakpoints in B-cell lymphoma.
A, Location of BCL6 gene on chromosome 3 at 3q27. BCL6 gene includes 6 exons. BCL6 protein functional domains include Bric-a-
brac tamtrack broad complex/poxvirus and Zinc finger (BTB/POZ) fold domain, Zinc finger C2H2-like domain, and Zinc finger C2H2-
type/integrase DNA binding domain. Various types of mutations are depicted. B, BCL6 rearrangement breakpoints occur most
frequently within the Major translocation cluster (MTC), including exon 1 and 3′ intronic sequence between exon 1 and exon 2. An
enlarged MTC is depicted and shows the breakpoint hypercluster. Red lines represent breakpoints for immunoglobulin/BCL6 rear-
rangements. Blue lines represent breakpoints for non-immunoglobulin/BCL6 rearrangements. Green bars represent deletions. (Modi-
fied and based on Akasaka H,et al: Cancer Res 60:2341-2355, 2000.)

The t(14;18) IGH breakpoints are within the joining that of endemic HTLV-1 prevalence, most common in
segments (JH). Approximately 50% of the BCL2 break- the Caribbean, Central America, South America, and
points are clustered within the major breakpoint region, Japan. The virus is transmitted via blood, sexual inter-
with the majority of the remaining breakpoints within the course, or breast milk. Although up to 40% of the popu-
minor cluster region (25%) and intermediate cluster lation in these areas have been infected by HTLV-1, only
region.65,77 The breakpoints in the BCL2 major break- 2% to 3% percentage of these individuals go on to
point cluster show an overrepresentation of sequences develop lymphoma/leukemia. The average age at diagno-
that are targeted by AID, and it is thought that t(14;18) sis is 55 years.114
occurs in bone marrow B-cell progenitors during VDJ The four clinical forms of adult T-cell lymphoma/
recombination under the influence of AID.77 leukemia are smoldering, chronic, lymphomatous, and
acute. These forms vary in sites of involvement and
numbers of HTLV-infected T cells in the peripheral
Adult T-Cell Lymphoma/Leukemia blood. In the smoldering form, there are circulating
polyclonal, oligoclonal, or monoclonal HTLV-infected
Definition
T cells at levels insufficient to produce absolute lympho-
Adult T-cell lymphoma/leukemia is a malignancy of cytosis. There is no evidence of involvement of other
mature CD4-positive T cells caused by infection with organs, other than limited skin or lung involvement. In
human T-cell lymphotrophic virus-1 (HTLV-1). It is the chronic form, there is absolute lymphocytosis with a
characterized by the presence of numerous, mostly non- monoclonal T-cell population, skin rash and papules,
recurrent, genetic abnormalities and atypical cytologic mild lactate dehydrogenase elevation, and mild hepato-
features with marked nuclear irregularity. Lytic bone splenomegaly and lymphadenopathy.
lesions and hypercalcemia are common.113 The acute and lymphomatous forms represent aggres-
sive disease, in which bone masses may be present. The
acute form accounts for 60% of cases and presents
Clinical Features
with lymphadenopathy, a leukemic picture, and a mono-
The geographic distribution of adult T-cell lymphoma/ clonal absolute T lymphocytosis as well as atypical
leukemia is uncommon in North America and parallels morphology. The lymphomatous form, accounting for

ERRNVPHGLFRVRUJ
854 12  Hematopoietic Tumors

Chr 14 Chr 18 der(14) der(18)


13 11.32 13 11.32 11.31
12 11.31 12
11.2 11.2 11.2
11.2 11.1 11.1
11.1 11.1 11.1 11.1 11.1 11.1
11.2 11.2 11.2 11.2
12.1 12 12.1
12 12.2 12.2
13 12.3 13 12.3
21.1 21.1
21 21.2 21 21.2 21.3
21.3
22 22 BCL2 22
23 23
24.1 23 24.1
24.2 24.2
24.3 24.3
31 32.1 31 32.1 IgH
32.2 32.3 32.2 32.3 REGION
IgH REGION BCL2
A

Exon 3

Exon 2
Exon 1
BCL2
Chr 18
cen tel
Major breakpoint region
BCL2
1 239 aa
BH4 BH3 BH1 BH2 TM
Apoptosis regulator, Bcl-2, motif Transmembrane region
B

IgH REGION
Constant JH ∼20kb DH ∼50kb VH ∼900kb
region IgH Enhancer (1-6 ) (1-12) (1,2,...,n)
Chr 14
cen tel
IgH breakpoints

der(14)t(14;18)
Exon 3

Exon 2
Constant Exon 1
region IgH Enhancer JH6

C
FIGURE 12-34  ■  t(14;18)(q32.3:q21.3) in diffuse large B-cell lymphoma. A, Chromosomal diagrams depicting reciprocal translocation
involving the IGH locus on chromosome 14 (Chr14) at 14q32.3 and the BCL2 locus on chromosome 18 (Chr18) at 18q21.3. The
translocation results in two derivative chromosomes, chr der(14) and chr der(18). B, The BCL2 gene consists of three exons and an
intron of approximately 250 kb (upper diagram). Approximately 50% of BCL2 translocation breakpoints occur within the major
breakpoint region in the 5′ untranslated region of exon 3. The BCL2 protein is 239 amino acids in length and includes BCL-2 Homol-
ogy motifs 1, 2, 3, and 4 (BH1-4) and a transmembrane region (TM) (lower diagram). The BH4 domain of BCL-2 is necessary for the
antiapoptotic function of BCL-2. C, The IGH breakpoints are located in the joining segments of the immunoglobulin heavy chain
(JH). The resulting derivative chromosome 14, der(14)t(14;18), places BCL2 gene expression under the control of the IgH enhancer,
resulting in expression of BCL-2 protein under the control of the IgH enhancer. (Based on data from Godon A, et al: Is t(14;18)(q32;q21)
a constant finding in follicular lymphoma? An interphase FISH study on 63 patients. Leukemia 17:255-259, 2003.)

20% of cases, presents without lymphocytosis but with a clinical trial comparing front-line antiviral therapy with
monoclonal proliferation of atypical T cells replacing chemotherapy for the acute form of adult T-cell
lymph nodes. Both of these forms are associated with lymphoma/leukemia resulted in a 5-year survival rate of
elevated lactate dehydrogenase and may typically involve 28% for the antiviral arm versus 10% for the chemo-
the liver, spleen, skin, or bone.70,110,111 Bone involvement therapy arm. Antiviral treatment of the chronic form
is most commonly seen in the acute form of adult T-cell resulted in a 100% 5-year survival rate compared with
lymphoma/leukemia. Hypercalcemia is a characteristic less than 20% with no treatment in another study.70,72 In
clinical finding, present in 70% of patients with the acute contrast to leukemic forms of adult T-cell lymphoma/
form, not all of whom have lytic bone lesions.70 leukemia, patients with the lymphomatous form respond
Chemotherapy has had little success in the treatment better to chemotherapy.70
of adult T- cell lymphoma/leukemia, with low response
rates and high rates of relapse. Recent studies have shown Radiographic Findings
that the acute, smoldering, and chronic forms have a
better response to antiviral therapy with interferon Patients with the acute form typically have multiple lytic
alfa-2b and zidovudine than to chemotherapy. A recent bone lesions involving both the axial and appendicular

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 855

skeleton.80 PET scan will show increased uptake in loca- multiple cell-cycle mediators and signal transduction
tions of lytic bone lesions and in involved lymph nodes pathways.94,119 TP53 deletion is associated with progres-
and organs (Fig. 12-35). sion of disease.108
Clonally rearranged T-cell receptor genes can be dem-
onstrated in the vast majority of cases. Virtually all cases
Microscopic Findings
are aneuploid.93 As a result of TAX-mediated genomic
Adult T-cell lymphoma/leukemia may show significant instability, numerous genetic defects have been detected
variation in morphology from case to case. The chronic in adult T-cell lymphoma/leukemia by cytogenetic studies
and smoldering forms are usually composed of small lym- and comparative genomic hybridization. The majority of
phocytes with mild pleomorphism. Acute and lympho- these are nonrecurrent abnormalities.
matous forms may display small lymphocyte morphology The hypercalcemia associated with adult T-cell
but are more likely to show marked pleomorphism. The lymphoma/leukemia is a result of increased osteoclast
acute form typically shows marked pleomorphism, with activity. Adult T-cell lymphoma/leukemia cells overexpress
circulating T cells with characteristic deeply lobulated Wnt5, which has been shown to transform granulocyte/
nuclei known as flower cells. Lytic bone lesions show monocyte precursors into osteoclasts. It is thought that
resorption of bone by osteoclasts (Fig. 12-36). There may activation of Wnt5a/Ror (receptor tyrosine kinase-like
or may not be an associated lymphomatous infiltrate. orphan receptor) β-catenin independent noncanonical sig-
When present, the infiltrate frequently consists of large naling results in a shift toward osteoclastic bone resorption
pleomorphic lymphocytes (Fig. 12-36). and decreased osteoblastic bone formation.71

Immunohistochemical Stains and


Differential Diagnosis CLASSICAL HODGKIN’S LYMPHOMA
The neoplastic T cells are CD4 positive, CD8 negative, Definition
and show bright expression of CD25 (Fig. 12-36). In the
majority of cases, the T cells show a loss of CD7 expres- Classical Hodgkin’s lymphoma is a proliferation of large
sion and maintain expression of CD2 and CD5. atypical B lymphocytes in a characteristic microenviron-
The differential diagnosis includes diffuse large B-cell ment of mixed benign inflammatory cells. The classifica-
lymphoma, other T-cell lymphomas, and myeloid sarcoma. tion includes four types of classical Hodgkin’s lymphoma:
Adult T-cell lymphoma/leukemia has a characteristic nodular sclerosis, mixed cellularity, lymphocyte-depleted,
although not specific immunophenotype. The T cells are and lymphocyte-rich.145 A separate disease entity, nodular
positive for CD4 and strongly positive for CD25. Dem- lymphocyte predominant Hodgkin’s lymphoma, has fea-
onstration of HTLV-1 seropositivity confirms the diag- tures more closely resembling B-cell lymphoma and will
nosis. B-cell markers, such as CD20, will be negative. not be discussed further here, as it very rarely presents as
Myeloid markers such as lysozyme, CD34, and myelo- a bone mass.146
peroxidase will be negative. Other T-cell lymphomas,
particularly peripheral T-cell lymphoma not otherwise speci- Clinical Features
fied are also considered in the differential diagnosis and
may have the same immunophenotype. Confirmation of Classical Hodgkin’s lymphoma commonly involves lymph
seropositivity for HTLV-1 is needed for a definitive diag- nodes, with an incidence of 2.8 per 100,000 persons. The
nosis of adult T-cell lymphoma/leukemia. peak age of incidence is in young adults ages 20 to 34.
White males are the most commonly affected group. The
5-year survival rate is 85%, with most disease-related
Genetic Features and Pathogenesis
deaths occurring in patients over age 55 years.144
The normal cell counterpart is an HTLV-1-infected Bone involvement in Hodgkin’s lymphoma is seen
mature T cell with features of a regulatory T cell (Treg). radiographically as detectable changes in only 1% to 3%
Direct infection of the T cells by HTLV-1 is required for of patients. Most frequently, patients with demonstrable
malignant transformation. HTLV-1 viral DNA is clonally bone lesions have end-stage (stages IIIB or IV) disease
integrated into the T cell DNA.70 with clinically apparent lymph node involvement. In one
Transformation is largely mediated by HTLV-1 TAX series of 25 patients with osseous classical Hodgkin’s
protein. Examples of the myriad effects of TAX include lymphoma, 3 patients (12%) had unifocal or multifocal
dysregulation of cyclins involved in cell-cycle progres- disease limited to bone.139 Bone involvement can occur
sion, impairment of DNA repair mechanism via inactiva- early or late in the disease. Overall, with modern treat-
tion of p53, upregulation of cytokines IL-2 and IL-15, ment, radiographically identifiable bone involvement
and constitutive activation of NFκB. Upregulation of is extremely unusual during the course of the disease
cytokines IL-2 and IL-15 promotes tumor cell growth in because most patients (90%) are disease-free for a very
an autocrine fashion. Constitutive activation of NFκB long time and are often considered cured.
results in decrease in apoptotic activity and transcription Several rare case reports of apparently primary
of several genes.70,81,103 TAX protein expression is fre- Hodgkin’s lymphoma of bone have been pub-
quently decreased in late stages of disease, when HTLV-1 lished.124,125,133,135,136,138 If a patient presents with skeletal
basic leucine-zipper (bZIP) transcription factor (HBZ) is changes, it is very likely that there is also extraskeletal
thought to maintain cellular proliferation via effects on disease. The age and sex distributions of patients with

ERRNVPHGLFRVRUJ
856 12  Hematopoietic Tumors

A B

C D
FIGURE 12-35  ■  Adult T-cell lymphoma of bone: radiographic features. A, Anteroposterior (AP) radiograph of femur in an adult
showing involvement of femoral neck, greater trochanter, and proximal femur by adult T-cell lymphoma. B, Radioisotopic bone
scan showing multiple foci of increased uptake throughout the skeleton. C, AP radiograph of humerus in an adult showing lytic
lesions of adult T-cell lymphoma. D, Axial computed tomography scan showing a destructive lesion involving the thoracic vertebral
body.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 857

A B

FIGURE 12-36  ■  Adult T-cell lymphoma of bone: microscopic and immunophenotypic features. A, Photomicrograph of adult T-cell
lymphoma showing lytic bone lesion with osteoclasts (×200). B, Higher power photomicrograph of case seen in A (×400). C, Photo-
micrograph of adult T-cell lymphoma showing lymphocytes with pleomorphic nuclei (×200). Inset; Higher magnification of adult
T-cell lymphoma shown in C (×1000). D, Photomicrograph of adult T-cell lymphoma with immunohistochemical IMM stain showing
positive CD4 (×1000). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
858 12  Hematopoietic Tumors

skeletal changes are identical to those for nodal Hodg-


TABLE 12-6 Histopathologic and
kin’s lymphoma.122,126,127,131,134 Hodgkin’s lymphoma has a
Immunophenotypic Features of
predilection for the trunk bones.113,123 The vertebral
Classical Hodgkin’s Lymphoma*
bodies (lower thoracic and lumbar) are preferentially
involved. The involvement of the pelvis is next in fre- Cell Type Phenotypic Features
quency. In the appendicular skeleton, the proximal parts
Reed-Sternberg Cells CD15+ CD45−
of major tubular bones, such as the femur and humerus,
Large vesicular nuclei with CD30+ T cell−
are preferentially involved.139 macronucleoli
Bone pain, especially in weight-bearing sites, is fre- May have one, two, or multiple PAX5+ B cell−
quently a symptom in Hodgkin’s lymphoma and may be nuclei (variable)
present without radiographically detectable changes. For EMA−
unknown reasons, the pain is frequently precipitated by Cellular Environment
alcohol consumption. Pathologic fracture can occur as an T cells (Th2) CD3+, CD4+
initial symptom. Eosinophils CD15+
Histiocytes CD68+
Plasma cells (variable) CD138
Radiographic Imaging Neutrophils (variable) CD15+
Bone lesions in Hodgkin’s lymphoma are typically mul- *The immunophenotype of the Reed-Sternberg cells is the same
tiple. The changes can be lytic, blastic, or mixed.139 Scle- in all subtypes of classical Hodgkin’s lymphoma. The
rotic changes of the vertebral bodies with an “ivory” immunophenotype of the cellular environment listed above is
appearance are frequent findings. Periosteal new bone characteristic of nodular sclerosis and mixed cellularity
subtypes, which represent the majority of cases. In lymphocyte-
formation, although not very prominent, is often seen.125 rich classical Hodgkin’s lymphoma, the cellular environment
Lesions in the long tubular bones have a tendency to be consists predominantly of mantle zone B cells. In lymphocyte-
more lytic and may have a permeative or moth-eaten depleted classical Hodgkin’s lymphoma, the cellular
appearance. Blastic sclerotic changes can also be present environment consists predominately of histiocytes.
in the appendicular skeleton (Fig. 12-37). Associated
local soft tissue lesions may be seen in up to 50% of
patients.139
phenotype is not considered a feature of conventional
Microscopic Findings Reed-Sternberg cells, occasionally the cells may show
focal variable positivity for CD20 and other B-cell
By conventional definition, the diagnosis of Hodgkin’s markers. In the background, CD4-positive T cells pre-
lymphoma requires the identification of diagnostic Reed- dominate. Reed-Sternberg cells are uncommon or absent
Sternberg cells in the appropriate cellular microenviron- in lymphocyte predominant Hodgkin’s lymphoma in
ment (Fig. 12-38).120 Diagnostic Reed-Sternberg cells in which large neoplastic cells express B-cell antigen
bone have the same morphologic characteristics as those CD20, are positive for CD45, and often coexpress EMA.
that occur in the lymph nodes (Fig. 12-38). They are Typically, they are negative for the two most common
large cells with round or lobulated nuclei, and they are antigens expressed by conventional Reed-Sternberg cells:
frequently binucleate or multinucleate. The striking CD15 and CD30. The background lymphocytes in this
feature is the presence of homogenous acidophilic, variant of Hodgkin’s lymphoma represent a mixture of B
inclusion-like nucleoli that approximate 25% of the size and T cells.
of the nucleus. The classic architectural features of variants of Hodg-
The cellular microenvironment includes T cells, kin’s lymphoma are difficult to evaluate in bone biopsies.
histiocytes, plasma cells, eosinophils, neutrophils, and The types that are frequently found in bone have features
fibroblasts. The classical lymphocyte-rich variant is of nodular sclerosis and mixed cellularity subtypes. The
the exception and is associated with a background of predominance of reactive fibrosis is frequently seen in
mantle zone B lymphocytes and variable numbers of his- bone lesions regardless of the variant of Hodgkin’s lym-
tiocytes and T cells. Some cases are associated with sig- phoma. In addition, the background cells may often be
nificant fibrosis, resulting in distortion of the histology polymorphous, with plasma cells and eosinophils causing
(Fig. 12-39). frequent confusion with osteomyelitis.
The microscopic and phenotypic features of major The complex differential diagnosis of Hodgkin’s lym-
types of Hodgkin’s lymphoma are summarized in phoma and other lymphohematopoietic lesions contain-
Table 12-6. ing Reed-Sternberg-like cells is beyond the scope of this
book. To avoid confusion with reactive or inflammatory
Immunohistochemical Stains and disorders such as osteomyelitis, it is important to con-
Differential Diagnosis sider the entire clinicopathologic picture. The correct
diagnosis is based on identification of Reed-Sternberg
The typical Reed-Sternberg cells in conventional Hodg- cells or variants and demonstration of the appropriate
kin’s lymphoma are positive for CD15 and CD30 and are immunophenotype by immunohistochemical stains.120
negative for CD45 and B- and T-cell markers.123,129,141,147 The morphologic recognition of these cells can be sup-
Reed-Sternberg cells are also variably positive for ported by the identification of their phenotypic features
PAX5 (Fig. 12-39). Although the expression of B-cell in the appropriate background cells.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 859

A B

C D
FIGURE 12-37  ■  Hodgkin’s lymphoma of bone: radiographic features. A, Sagittal magnetic resonance image (MRI) showing multifocal
involvement of lumbar vertebra by Hodgkin’s lymphoma. B, Axial MRI of the same case as in A showing diffuse involvement of the
vertebral body by Hodgkin’s lymphoma. C, Sagittal MRI showing involvement of a lumbar vertebra and sacrum by Hodgkin’s lym-
phoma. D, Axial MRI of the same case as in C showing diffuse involvement of lumbar vertebra by Hodgkin’s lymphoma.

ERRNVPHGLFRVRUJ
860 12  Hematopoietic Tumors

A B

C D
FIGURE 12-38  ■  Hodgkin’s lymphoma of bone: radiographic and microscopic features. A, Lytic lesions in patient previously
treated for Hodgkin’s lymphoma. B, Photomicrograph of Hodgkin’s lymphoma showing classic binucleate Reed-Sternberg cell,
mononuclear Hodgkin’s cell, and a background of small lymphocytes and histiocytes (×400). C, Higher power photomicrograph of
case in A (×1000). D, Photomicrograph of lacunar cells with cytoplasmic retraction artifact and many background eosinophils (×400).
(B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 861

A B

C D
FIGURE 12-39  ■  Hodgkin’s lymphoma: microscopic and immunohistochemical features. A, Photomicrograph of lacunar cells with
cytoplasmic retraction artifact and many background eosinophils. B and C, Photomicrograph of Hodgkin’s lymphoma showing a
fibrotic background with rare classic binucleate Reed-Sternberg cell, and a background of lymphocytes and histiocytes. D, Photo-
micrograph of immunohistochemical stains showing Reed-Sternberg cells positive for CD30 (top), PAX5 (middle), and CD15
(bottom). Note dim PAX5 staining of Reed-Sternberg cells compared to bright staining of normal small B cells (A-C, ×400; D, ×200).
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
862 12  Hematopoietic Tumors

Genetic Features and Pathogenesis CD40 on the surface of Reed-Sternberg cells, resulting
in activation of the NFκB signaling pathway.132 Increased
The Reed-Sternberg cell is thought to arise from a ger- macrophages in the cellular background of classical
minal center B cell with mutations resulting in loss of Hodgkin’s lymphoma, as demonstrated by gene expres-
functional immunoglobulin. Discovery of genetic altera- sion profiling or CD68 immunohistochemical stain,
tions in Hodgkin’s lymphoma has lagged somewhat have been associated with worse disease-specific sur-
behind that of other lymphomas. Most studies require vival.121,130,143 Increased histiocytes have also been associ-
microdissection of Reed-Sternberg cells from the cellular ated with EBV-positive classical Hodgkin’s lymphoma.
microenvironment. In addition to translocations involv- EBV positivity confers a worse overall survival in patients
ing immunoglobulin genes common to many B-cell lym- older than age 45 years.128 Gene expression profiling pre-
phomas, Reed-Sternberg cells have been found to have dicts worse overall survival in patients with a profile
complex genetic abnormalities, including aneuploidy, suggesting T helper 1 (Th1) reaction, including upregu-
gains and losses of multiple chromosome loci, and mul- lated expression of interferon gamma (IFNG) and IFNG-
tiple translocations and mutations (Table 12-7). regulated genes CXCL11, IRF1, STAT1, TNFSF10, and
Some of these genetic abnormalities result in de­ major histocompatibility class I (MHC I) genes.142
regulation of crucial signaling pathways. Constitutive
activation of NFκB and JAK/STAT pathways has been
implicated as a key driver of cell proliferation and sur- HISTIOCYTIC AND DENDRITIC
vival, while mutations of FAS are associated with in- CELL PROLIFERATIONS
creased cell survival via inhibition of apoptosis.137 Genetic
alterations contributing to NFκB activation are gains of The mononuclear phagocytic lineage consists of mono-
c-REL and NFκB-inducing kinase (NIK, also known as cytes, macrophage/histiocytes, and dendritic cells. Mono-
MAP3K14), deletions of TRAF, and inactivating muta- cytes circulate in the peripheral blood and can be localized
tions of TNFα-induced protein 3 and NFκB inhibitors to tissues, especially lymph nodes, spleen, and bone
A and E.132,140 marrow. Macrophage/histiocytes and dendritic cells are
Reed-Sternberg cell proliferation and survival are also tissue based. Macrophages ingest cells, cellular debris,
driven by their interactions with the cells of the micro- and microorganisms and secrete inflammatory cytokines.
environment of classical Hodgkin’s lymphoma. Reed- Dendritic cells ingest antigens and present antigen to T
Sternberg cells have direct and indirect interactions with cells. Bone marrow macrophage/dendritic precursors
the surrounding cells and secrete various cytokines and give rise to monocytes and common dendritic precursors.
chemokines that attract the various constituents of the Circulating monocytes give rise to macrophage/
cellular microenvironment. For example, CD40 ligand histiocytes and a subset of dendritic cells. The majority
on a subset of helper T cells (Th2 cells) binds directly to of dendritic cells arise from common dendritic precur-
sors.160,165,201 Dendritic cells of the skin (Langerhans cells)
are also derived from a myeloid precursor.207 Primitive
macrophages migrate to the mesoderm during embry-
TABLE 12-7 Classical Hodgkin’s Lymphoma:
onic development and differentiate into mature Langer-
Common Genetic Abnormalities
hans cells only under the influence of the microenvironment
Loss of function immunoglobulin gene mutations of their final location in the skin.185 Immunohistochemi-
Immunoglobulin gene translocations cal features of normal and pathologic histiocytic and den-
FAS mutations dritic cells are summarized in Table 12-8.
JAK2 translocations and and copy number gains
NFKBIA/NFKBIE mutations
Proliferations of monocytic/histiocytic cells range
SOCS1 mutations from solitary benign lesions to indolent multifocal disor-
TP53 mutations ders to aggressive systemic diseases. Identification of the
CIITA translocations functional status of monocytic/histiocytic cells and of
TNFAIP3 mutations their interaction with other elements of the immunohe-
TRAF3 deletion
MAP3K14 copy number gains matopoietic system provided a basis for modern classifi-
REL copy number gains cation of this group of disorders. This discussion is
Aneuploidy limited to the histiocytic/dendritic cell proliferations that
involve the skeleton: Langerhans cell histiocytosis,
CIITA, Class II MHC transactivator;JAK2, Janus kinase 2;
MAP3K14, nuclear factor κ B-inducing kinase kinase kinase 14; Erdheim-Chester disease, and Rosai-Dorfman disease
NFKBIA/NFKBIE, nuclear factor of κ light polypeptide gene (sinus histiocytosis with massive lymphadenopathy).
enhancer in B cells B inhibitor α and ε; SOCS1, suppressor of These three diseases have distinctive clinical and mor-
cytokine signaling 1; TNFAIP3, tumor necrosis factor, alpha- phologic findings, but a subset of patients may have more
induced protein 3; TP53, tumor protein 53; TRAF3, tumor
necrosis factor receptor-associated factor 3.
than one histiocytosis.175,183,195
Adapted from Otto C, et al: Genetic lesions of the TRAF3 and
MAP3K14 genes in classical Hodgkin lymphoma. Br J
Haematol 157:702-708, 2012; Kuppers R: New insights in the Langerhans Cell Histiocytosis
biology of Hodgkin lymphoma. Hematology Am Soc
Hematol Educ 2012:328-334, 2012; Shaffer AL, et al: Definition
Pathogenesis of human B cell lymphomas. Annu Rev
Immunol 30:565-610, 2012; and Kuppers R, et al: Hodgkin Langerhans cell histiocytosis is a proliferation of cells
lymphoma. J Clin Invest 122:3439-3447, 2012. with features of Langerhans cells, characterized by

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 863

TABLE 12-8 Immunophenotypic Features of TABLE 12-10 Langerhans Cell Histiocytosis


Histiocytes and Dendritic Cells Frequency of Involvement of
Various Organ Systems
Normal Erdheim- Langerhans
Marker Histiocyte SHML* Chester Cell Bone 77%
CD1a − − − + Multisystem disease 44%
CD4 + + + + Craniofacial lesions 43%
CD68 + + + +/− Skin 39%
CD123 − − − − Vault 21%
CD163 + + + − Ears 20%
Langerin − − − + Lymph nodes 19%
Lysozyme + Weak +/− + +/− Liver 16%
S100 +/− + Weak +/− + Bone marrow 13%
CD21 − − − − Spleen 13%
CD35 − − − − Eyes 13%
Oral 13%
*Sinus histiocytosis with massive lymphadenopathy. Cervical nodes 13%
Adapted from Swerdlow SH, et al, eds: World Health Lung 10%
Organization classification of tumours of haematopoietic Gastrointestinal tract 4%
and lymphoid tissues, Lyon, 2008, IARC Press. Genital mucosa 3%
Skull base 2%

Adapted from Grois N, et al: Risk factors for diabetes insipidus


in Langerhans cell histiocytosis. Pediatr Blood Cancer
TABLE 12-9 Classification of Langerhans 46:228-233, 2006.
Cell Proliferations
Langerhans Cell Histiocytosis
Single organ
Multifocal bone often resulting in Christian’s triad (lytic skull lesions,
Multisystem exophthalmos, and diabetes insipidus). Letterer-Siwe
Pulmonary Langerhans Cell Histiocytosis disease was described as an aggressive systemic form of
Langerhans cell histiocytosis involving multiple organs
Langerhans Cell Sarcoma and systems with associated functional impairment of the
affected sites. Frequent sites of involvement in Letterer-
Siwe disease were lymph nodes, liver spleen, lung, and
skin.207
expression of CD1a and langerin (CD207) by immuno-
histochemistry and the presence of Birbeck granules by Clinical Features
electron microscopy. Normal Langerhans cells have den-
dritic processes, and pathologic Langerhans cells have Langerhans cell histiocytosis primarily affects young
rounded cytoplasmic borders without classical dendritic individuals during the first three decades of life (Fig.
cell morphology. 12-40). Approximately 80% of cases are diagnosed in
A broad classification of Langerhans cell proliferations patients younger than age 30 years, and 50% of patients
is listed in Table 12-9.179,203 Langerhans cell histiocytosis are children younger than age 10 years. Multisystem
may present as a unifocal lesion, multifocal bone involve- forms of Langerhans cell histiocytosis predominntly
ment, or multisystem disease. Pulmonary Langerhans occur during the first 2 years of life (Fig. 12-41). The
cell histiocytosis occurs in smokers and is clinically dis- disease more commonly affects whites and is slightly
tinct from other forms of Langerhans cell histiocytosis. more common in males (Fig. 12-42).175
Langerhans cell sarcoma is a tumor composed of mor- Bone is the most common site of involvement, with
phologically malignant Langerhans cells.179,203 Particu- 77% of patients having bone lesions.172 The craniofacial
larly fulminant forms of Langerhans cell histiocytosis, bones are most frequently affected by this disorder, and
sometimes in association with malignant lymphoma or the cranial vault is the most frequently affected site.
leukemia, have also been described.155,170,192 Other common sites include the mandible, vertebral
The previous designation of histiocytosis X, eosino- bodies, ribs, pelvis, and femur. These sites are most fre-
philic granuloma, and the clinical syndromes Hand- quently involved as solitary lesions but are also sites of
Schuller Christian and Letterer-Siwe are no longer used predilection in multisystem disease. In the appendicular
for classification but are useful in describing the organ skeleton, the major long tubular bones are most fre-
systems most commonly involved. Of the three terms, quently affected. The acral skeleton (i.e., the bones of the
eosinophilic granuloma is still relatively commonly used to hands and feet) are rarely involved. In the long bones, the
refer to unifocal or multifocal bone involvement by lesions are predominantly diaphyseal or metaphyseal and
Langerhans cell histiocytosis, usually without involve- almost never extend to the end of a bone. Rarely, epiphy-
ment of other organ systems. The term Hand-Schüller- seal lesions are seen in young children (Fig. 12-40). Other
Christian disease referred predominantly to disease in common sites of involvement are skin, ears, lymph nodes,
young patients with extensive multifocal involvement of and liver, with other organs involved less frequently
the skeleton, predominantly of the craniofacial bones, (Table 12-10).172

ERRNVPHGLFRVRUJ
864 12  Hematopoietic Tumors

Disseminated
aggressive forms
predominantly occur
during first 2 years
of life
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 12-40  ■  Langerhans cell histiocytosis: skeletal distribution. Most frequent sites of involvement are indicated by solid black
arrows.

Treatment and prognosis of Langerhans cell histiocy- organs may benefit from intensified chemotherapy.169
tosis depend largely on age at presentation and the Cladribine (2-chlorodeoxyadenosine; 2CdA) has been
site(s) and extent of involvement. Virtually all patients used to treat adults with Langerhans cell histiocytosis
with unifocal bone involvement are cured. Features asso- bone lesions. Cytarabine (cytosine arabinoside; ara-C)
ciated with inferior event-free survival and overall sur- may be superior.157
vival are age at diagnosis less than 1 year, multisystem
involvement, and the presence of multiple bone lesions.179 Radiographic Imaging
The group of patients with the worst overall survival are
neonates with multisystem disease, who have a 5-year The lesions are lytic and sharply demarcated, “punched-
survival rate of only 57%.187 out” intramedullary defects (Figs. 12-43 to 12-46). They
In pediatric clinical trials, patients have been stratified are rarely intracortical. The intracortical lesions are
into those with single system involvement, multisystem almost exclusively seen in larger-diameter bones, such as
involvement of low risk organs, and multisystem involve- the major long tubular bones. The size of a typical lesion
ment including risk organs (spleen, liver, or hematopoi- is within 1 to 2 cm. Sometimes a thin sclerotic rim can
etic system). Patients with craniofacial or vertebral bone be seen. The larger lesions (4 to 5 cm) can erode or even
lesions are also considered higher risk with regard to completely disrupt the cortex and expand into the adja-
central nervous system (CNS) involvement.186 cent soft tissue. In the vertebral column, the lesion rep-
Disease limited to unifocal or multifocal non-CNS resents a lytic involvement of the vertebral body with
risk bone involvement may be treated with observation frequent collapse (vertebra plana) (Fig. 12-45).150 Typi-
or curettage followed by steroid injection. If bone sta- cally, there is minimal or no periosteal new bone forma-
bilization is needed, radiotherapy may be considered. tion, but an expanded thin rim of elevated periosteum
Patients with multisystem disease, risk organ involve- may be seen in younger patients (Fig. 12-46). In rare
ment, or bone lesions conferring CNS risk should be instances, especially in small-diameter bones such as ribs,
treated with chemotherapy.186 Patients who present Langerhans cell histiocytosis can produce lesions with a
before age 2 years or who have involvement of risk permeative or moth-eaten appearance (Fig. 12-47). This

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 865

35

30

25
Age distribution (%)

20

15

10

0
1

+
0-

1-

5-

-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80
Age at diagnosis
FIGURE 12-41  ■  Langerhans cell histiocytosis. Data shows the peak incidence of Langerhans cell histiocytosis is in children under
age 5 years. SEER Data, 1973-2010.

0.035

0.03
(cases/100000 persons)

0.025
Incidence rate

0.02

0.015

0.01

0.005

0
e

es

es

es
e
al

al

al

al

al

al
m

m
fe
,

fe

fe
es

te

k
,

ac
es
ac

hi

te

k
ac
Bl
W
ac

hi
lr

Bl
W
Al

lr
Al

FIGURE 12-42  ■  Langerhans cell histiocytosis. Distribution of Langerhans cell histiocytosis by race and sex. The most commonly
affected group is white males. SEER Data, 1973-2010.

feature is seen more often in young children and does not demonstrating the activity of disease, the response to
preclude more aggressive behavior, compared with lesions treatment, and reactivation of disease.181
that have more classic presentations. The extent of skel-
etal involvement ranges from a solitary focus to a dis- Gross Findings
seminated multifocal skeletal and extraskeletal disorder.
CT is useful in delineating the extent and exact location Solitary lesions appear as sharply demarcated gray-tan
of a given bone lesion. MRI is more instructive in delin- lytic foci with frequent cortical disruption. Foci of tan or
eating soft tissue extension.150 FDG PET/CT is useful in Text continued on p. 871

ERRNVPHGLFRVRUJ
866 12  Hematopoietic Tumors

B
FIGURE 12-43  ■  Langerhans cell histiocytosis: radiographic features. A and B, Calvarial destructive lesions in two cases of eosinophilic
granuloma in children. Lesions involve full thickness of skull and are sharply circumscribed.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 867

A B

C D

FIGURE 12-44  ■  Langerhans cell histiocytosis: radiographic features. A, Lytic lesion of cranial bone. B, Lytic punched-out lesion of
scapula. C, Lytic destructive lesion of humoral shaft. D, Destructive lesion of ulna with moth-eaten pattern.

ERRNVPHGLFRVRUJ
868 12  Hematopoietic Tumors

A B

C D

FIGURE 12-45  ■  Langerhans cell histiocytosis: radiographic features. A, Lateral radiograph of cervical spine of a teenage boy with
neck pain. Body of third cervical vertebra shows partial collapse because of involvement by Langerhans cell histiocytosis (eosino-
philic granuloma). B, Two weeks later, same vertebra shown in A is further flattened as a result of continued bone destruction
(vertebra plana). C, Lateral radiograph of humerus of a young child with Langerhans cell histiocytosis involving midshaft of humerus.
Note permeative pattern of destruction with poorly demarcated borders and prominent periosteal new bone formation. D, Antero-
posterior radiograph of Langerhans cell histiocytosis shown in C. Note periosteal reaction.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 869

A B

C D
FIGURE 12-46  ■  Langerhans cell histiocytosis: radiographic features. A, Anteroposterior (AP) radiograph of proximal femur showing
a lytic lesion with cortical destruction. B, Coronal magnetic resonance image (MRI) of case shown in A showing a high signal density
lesion of the proximal femur. C, AP radiograph of humerus showing a lytic lesion involving shaft. D, Coronal MRI of the same case
as in C showing a high signal intensity lesion with cortical disruption of the humeral shaft.

ERRNVPHGLFRVRUJ
870 12  Hematopoietic Tumors

A C

FIGURE 12-47  ■  Langerhans cell histiocytosis: radiographic features. A, Left hip of young adult with lytic lesions in supraacetabular
portion of ilium and femoral neck. B, Chest radiograph shows diffuse bilateral infiltrates in lung fields. C, Technetium-99 bone scan
of pelvis shows increased uptake in region of supraacetabular destruction shown in A.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 871

yellowish tissue and fibrosis can be present in lesions osteomyelitis. Typically, intact foci of Langerhans cell
overgrown with ordinary histiocytes and in lesions under- histiocytosis are seen focally. The surrounding bone may
going regression. show prominent resorptive osteoclastic activity, which is
mediated by cytokines produced by the Langerhans cells
Microscopic Findings and T cells.161 Reactive bone trabeculae with prominent
osteoblastic rimming are usually associated with patho-
Langerhans cell histiocytosis is composed of two basic logic fracture.
cell types, eosinophils and Langerhans cells, but only the
Langerhans cells are pathognomonic (Fig. 12-48). In Immunohistochemical Stains, Special
addition to these two basic types of cells, an admixture of Techniques, and Differential Diagnosis
other inflammatory cells may be present. The proportion
of Langerhans cells and inflammatory cells, especially Ultrastructurally, Langerhans cells contain prominent
eosinophils, can vary among different lesions and in indentations of the nuclear membrane (nuclear grooves)
various areas of the same lesion. and are distinguished from ordinary histiocytes by their
In a typical case, Langerhans cells represent mono- low phagocytic activity and the presence of Birbeck gran-
nuclear histiocyte-like cells with oval nuclei and clearly ules. Birbeck granules are distinctive organelles with a
demarcated round or oval cytoplasm. The majority of tubular tennis racket–shaped membranous structure sur-
nuclei show a prominent nuclear groove parallel to the rounded by a unit membrane and containing a dense
long axis of the nucleus (coffee-bean nuclei) (Fig. 12-48). homogenous core (Fig. 12-50).
It represents a deep longitudinal indentation of the Immunophenotypically, they have features consistent
nuclear membrane that is best seen in electron micro- with Langerhans cells. The microscopic and phenotypic
graphs. Some of the nuclei may have oval or completely features of Langerhans cells are summarized in Table
round nucleoli. The variability in nuclear shapes and sizes 12-8. By immunohistochemical staining, Langerhans
is, to some extent, related to the plane of sectioning of cells are strongly positive for S-100, CD1a, and langerin
the individual nucleus. In a typical case, there is little to (CD207). These stains help distinguish Langerhans cells
no nuclear atypia. Mitotic activity is typically low, and from normal histiocytes, which are negative for all three
usually fewer than five mitoses can be found per 10 high- stains (Table 12-8). Langerin is also expressed by a limited
power fields. Atypical mitoses are not present. Occasion- subset of non-Langerhans dendritic cells. Langerin is a
ally, mild to moderate atypia of Langerhans cells can be lectin that can bind multiple types of antigens, including
seen. Atypical Langerhans cells have hyperchromatic viruses, fungi, and possibly malignant tumor cells.204 It
enlarged nuclei, an increased nuclear-to-cytoplasmic has been shown that binding of surface langerin is fol-
ratio, and medium-sized nuclei. Occasionally, multinu- lowed by internalization of langerin and localization to
cleated giant cells may be present and can have some Birbeck granules, which cannot form in the absence of
nuclear features typical of Langerhans cells or can resem- langerin. Although the exact function of Birbeck granules
ble ordinary osteoclasts. In cases which show a high is uncertain, they have been shown to play a role in
degree of nuclear atypia, Langerhans cell sarcoma should antigen processing.185 Neoplastic Langerhans cells are
be considered. also positive for p53 (Fig. 12-48). VE1, an antibody spe-
The accompanying inflammatory cells may both aid cific for the BRAF V600E mutation, is positive in cases
and hinder the diagnosis. Ordinary histiocytes may be bearing this mutation.
present and are distinguished microscopically by their Langerhans cell histiocytosis must be distinguished
high phagocytic activity and the absence of nuclear principally from inflammatory conditions, such as osteo-
grooves. They may appear as lipid-laden foam cells or myelitis, granulomatous inflammation, and Hodgkin’s lym-
may contain nuclear debris or fragments of degenerated phoma and non-Hodgkin’s lymphoma. In contrast to
eosinophils, such as Charcot-Leyden crystals. The prom- Langerhans cell histiocytosis, osteomyelitis rarely affects
inent reaction of foamy histiocytes is often seen in chronic craniofacial bones, but the radiographic features can
lesions. On the other hand, Langerhans cells may also overlap in some cases. Microscopically, the presence of
contain phagocytized material. In general, the presence polymorphic inflammatory cell infiltrates with neutro-
of prominent phagocytosis can alter the classic features phils and the presence of necrotic bone trabeculae favor
of Langerhans cell histiocytosis, making its microscopic osteomyelitis.
diagnosis very difficult. Granulomatous inflammation can be confused with
Other inflammatory cells, especially eosinophils, Langerhans cell histiocytosis in cases in which granulo-
may predominate (Fig. 12-49). Occasionally, they form mas are ill defined. The presence of necrosis surrounded
aggregates with necrosis (eosinophilic abscesses). Lang- by palisading histiocytes favors an inflammatory process.
erhans cells are usually seen at the periphery of such The identification of acid-fast bacilli or fungi by use of
abscesses. A predominance of other inflammatory cells, appropriate special stains help rule out Langerhans his-
lymphocytes, and plasma cells, especially with associated tiocytosis; however, the sensitivity of these stains is low.
fibrosis, can lead to a misdiagnosis of osteomyelitis. Indi- In addition, radiographs of patients with Langerhans cell
vidual lesions can undergo spontaneous regression that histiocytosis almost never show collapsed intervertebral
is associated with progressive fibrosis and foamy histio- disk spaces or contiguous involvement of adjacent verte-
cytic infiltrates. Regressive changes in Langerhans cell brae, a feature frequently seen in granulomatous inflam-
histiocytosis simulate nonossifying fibroma or chronic mation involving the spine.

ERRNVPHGLFRVRUJ
872 12  Hematopoietic Tumors

A B

C F
FIGURE 12-48  ■  Langerhans cell histiocytosis: microscopic features. A, Low power photomicrograph of Langerhans cell histiocytosis
with many intermixed lymphocytes and eosinophils (×100). B, Photomicrograph of Langerhans cell histiocytosis with prominent
eosinophils (×200). C, Higher magnification of case in B shows Langerhans cells with indented bean-shaped nuclei and nuclear
grooves (×400). D, Langerhans cells positive for CD1a (×100). E, Langerhans cells positive for S-100 (×100). F. Langerhans cells with
overexpression of p53 (×100). (A-C, hematoxylin-eosin; D, DAB.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 873

B
FIGURE 12-49  ■  Langerhans cell histiocytosis: ultrastructural features. A and B, Electron micrographs show eosinophilic leukocytes
containing characteristic granules and portions of cytoplasmic borders of Langerhans cells. Note paucity of lysosomal structures
and irregular cytoplasmic projections in Langerhans cells. Inset shows features of cytoplasmic granules of eosinophilic leukocytes.
(A and B, ×3500; inset, ×15,000.)

ERRNVPHGLFRVRUJ
874 12  Hematopoietic Tumors

D
FIGURE 12-50  ■  Langerhans cell histiocytosis: ultrastructural features. A-D, Birbeck granules are pentalaminar rod-shaped cytoplas-
mic organelles (A-D, ×45,000).

Classical Hodgkin’s lymphoma almost never presents as Genetic Features and Pathogenesis
a primary skeletal lesion, and patients usually have
extraskeletal disease. The presence of diagnostic Reed- The normal cell counterpart is a cell with morphologic,
Sternberg cells, associated fibrosis in the appropriate cel- immunophenotypic, and ultrastructural features of Lang-
lular background, and the so-called lacunar cells in the erhans cells. Normal Langerhans cells in mice are derived
nodular-sclerotic variant is a typical microscopic feature from early yolk sac–derived monocytic/macrophage pre-
of classical Hodgkin’s lymphoma. The diagnosis may be cursors that migrate to the early mesoderm destined to
made by identification of Reed-Sternberg cells with char- become dermis, where they differentiate and form
acteristic immunophenotypic features. Birbeck granules after birth.185,197 Normal human Lang-
Langerhans cell histiocytosis can be confused with a erhans cells are thought to also derive from early myeloid
large-cell non-Hodgkin’s lymphoma. Most primary skeletal precursors found in fetal dermis around week 9 of devel-
non-Hodgkin’s lymphomas are diagnosed in patients opment.202 Pathologic human Langerhans cells were pre-
older than age 40 years. In this age group, Langerhans viously thought to be derived from mature epidermal
cell histiocytosis is extremely rare. The misdiagnosis of Langerhans cells.193 The current prevailing view is that
Langerhans cell histiocytosis as non-Hodgkin’s lym- they are derived from a myeloid dendritic cell precur-
phoma is usually caused by a dominance of Langerhans sor.148 The normal cell counterpart may be a more imma-
cells. The use of appropriate markers and the identifica- ture myeloid precursor in aggressive forms of disease and
tion of phenotypic features of Langerhans cells help to a more mature differentiated cell in localized forms of
avoid this error. disease.152

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 875

Chr 7 BRAF
22

Exon 18
Exon 17

Exon 16
Exon 15
Exon 14

Exon 13
Exon 12
Exon 11
Exon 10
21

Exon 9

Exon 8
Exon 7
Exon 6
Exon 5
Exon 4

Exon 3

Exon 2

Exon 1
15.3 15.2
15.1

140433812

140624564
14
13
12
11.2
11.1 11.1 cen tel
11.22 11.21
11.23
21.1
21.2
21.3
22

44
31.1
31.2
31.3

5
32
33
BRAF
35 34

21
36
BRAF

5
5
6

17

4
7

34.285
5

5
4
12
4

110

138
4

5
21

51

12

67
19

95

10
8

4
4

6
6
4

4
4

4
4
7

7
1 766 aa

V600E
Missense substitution Raf-like Ras-binding domain
Nonsense substitution - Stop Protein Kinase C-like, phorbol ester/diacylglycerol binding domain
Insertion inframe Protein Kinase-like domain
Deletion inframe
Insertion frameshift
Deletion frameshift
Complex - deletion inframe
Unknown
Substitution - coding silent
Complex - frameshift
Complex - insertion inframe
Nonstop extension
Complex - compound substitution
Complex
FIGURE 12-51  ■  BRAF V600E mutation in Langerhans cell histiocytosis and Erdheim-Chester disease. The chromosomal location and
the exonal structure of the BRAF gene are shown. The structure of the encoded protein with its functional domain with superimposed
positions of mutations identified in various tumors and available in the COSMIC database is also shown. The V600E mutation,
representing the most common genetic abnormality detected to date in both Langerhans cell histiocytosis and Erdheim- Chester
disease, is highlighted. BRAF V600E is an activating missense mutation involving the substitution from valine to glutamate at protein
kinase domain at codon 600, resulting in upregulation of the RAF/MET/ERK signaling pathway. (Based on data from Haroche J, et al:
High prevalence of BRAF V600E mutations in Erdheim-Chester disease but not in other non-Langerhans cell histiocytoses. Blood 120:2700-
2703, 2012.)

Clonality can be demonstrated in the majority of soli- as peripheral blood monocytes and dendritic cells. In
tary and multisystem disease, and in a significant minority contrast, patients with limited disease have BRAF V600E
(29%) of isolated adult lung Langerhans cell histiocyto- only in lesional Langerhans cells.152 This appears to at
sis.208,209 A recent sequencing study of 61 pediatric cases least partially explain the variable presentations of Lang-
of Langerhans cell histiocytosis demonstrated recurrent erhans cell histiocytosis, with the more aggressive forms
mutations in neoplastic Langerhans cells. The most of disease arising from a bone marrow myeloid dendritic
common of these is BRAF V600E mutation, detected in cell precursor and the low risk disorders representing a
57% of Langerhans cell histiocytosis (Fig. 12-51).151 The clonal expansion of more differentiated monocytic or
same study detected mutations of TP53 and MET dendritic cells.
in single samples. The protein p53 is overexpressed
in all cases of Langerhans histiocytosis; however, the
mechanism in most cases is unknown and is not the result Erdheim-Chester Disease
of a mutation involving TP53. Definition
Patients with high risk and multisystem Langerhans
cell histiocytosis have recently been shown to have BRAF Erdheim-Chester disease is a non-Langerhans cell histio-
V600E mutation not only in Langerhans cells within cytosis. In 1930, Erdheim and Chester described two
lesions, but also within small numbers of bone marrow patients who had distinct skeletal lesions and histiocytic
CD34-positive myeloid dendritic cell precursors as well infiltrates of bone marrow that they believed to be

ERRNVPHGLFRVRUJ
876 12  Hematopoietic Tumors

Symmetric involvement
of major long tubular bones
Incidence

40

1 2 3 4 5 6 7 8
Age in decades

FIGURE 12-52  ■  Erdheim-Chester disease. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites are
indicated by solid black arrows.

distinct from Langerhans cell histiocytosis.159 Jaffe standing of its clinical features. Approximately 500 cases
reported another case and also suggested that this disor- have been described in the literature.175 Greater than
der was distinct from what is currently considered Lang- 80% of patients present after age 40 years (Fig. 12-52),
erhans cell histiocytosis.178 The distinction was based on with a mean age at diagnosis of 55, and a male-to-female
the peculiar radiographic appearance of the lesion (i.e., ratio of approximately 3 : 1.149,158 Occasionally other his-
diffuse sclerotic lesions symmetrically involving diaphy- tiocytoses, including Langerhans cell histiocytosis or
seal and metaphyseal parts of the major long bones) and Rosai-Dorfman disease, may occur in patients with
the presence of lipid-laden histiocytes. Erdheim-Chester disease.175
Erdheim-Chester disease is best described as a mul- The most frequent symptom at presentation is bone
tisystem proliferative histiocytic disorder that almost pain, with diabetes insipidus, neurologic symptoms, and
always involves bone, producing diffuse symmetric scle- constitutional symptoms also commonly seen. Although
rotic lesions of major long bones in classic cases. Micro- bone involvement can be demonstrated in at least 95%
scopically, it is characterized by the presence of ordinary of cases, only about 50% of patients will experience bone
(foam cell) histiocytic infiltrate with small microscopic pain.175 The most distinctive finding is diffuse symmetric
foci of histiocytes confirmed by immunohistochemical involvement of the major long bones (Fig. 12-53). Bone
stains. The histiocytes are positive for CD68, negative lesions at other sites such as ribs, sacrum, craniofacial
for langerin, and usually negative for S-100. There are bones, and lumbar vertebrae can also be seen. Involve-
variable numbers of intermixed T cells, predominately ment of craniofacial bones may be associated with diabe-
Th1 cells.149 BRAF V600E mutation is present in up to tes insipidus. Greater than 90% of patients have one
100% of patients using sensitive assays.156 extrasekeletal manifestation.149 Although virtually any
organ system may be affected, commonly involved
extraskeletal sites include the cardiovascular system,
Clinical Features
central nervous system (CNS), kidneys, and lungs.175
Erdheim-Chester disease is extremely rare but is being Involvement may result in encasement and compression
recognized with increasing frequency and greater under- of structures such as the aorta or ureters.175

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 877

B D
FIGURE 12-53  ■  Erdheim-Chester disease: radiographic features. A and B, Anteroposterior radiographs of knees and lower legs of
same case showing diffuse symmetric sclerosis of both tibia and distal left femur. C, Radioisotopic bone scan showing an increased
uptake in both tibia and left distal humerus. D, Positron emission tomography/computed tomography fused image showing
fluorodeoxyglucose-avid lesion within the medullary cavity of both tibiae.

ERRNVPHGLFRVRUJ
878 12  Hematopoietic Tumors

A B C
FIGURE 12-54  ■  Erdheim-Chester disease: radiographic features. A, Early changes of sclerosis seen in midshaft of tibia and fibula of
middle-aged man. B and C, Radiographs taken 1 year later show progression of mixed diaphyseal lysis and sclerosis of tibia and
fibula.

Treatment and Behavior a thickened trabecular pattern and endosteal cortical


thickening. Up to 30% of patients may also have lytic
Erdheim-Chester disease is typically a slowly progressive bone lesions.196 CT scan of the chest and abdomen is
disease, with a subset of cases showing rapid progression useful in detecting the characteristic distribution of
and multisystem involvement. Current treatment of disease such as encasement of the aorta (“coated aorta”)
Erdheim-Chester disease is largely immunomodulatory, and involvement of the perirenal fascia (“hairy kidney”).
with interferon alpha. Prior to the use of interferon alpha, MRI is most useful for detecting cardiac and CNS
mean patient survival was 19.2 months.175 The current involvement. PET/CT is recommended for evaluation of
5-year survival rate is approximately 68%.149 Three the initial extent of disease and for monitoring response
patients with V600E BRAF mutated Erdheim-Chester to treatment.175,190
(two also with Langerhans cell histiocytosis) treated with
the BRAF inhibitor, vemurafenib, showed significant Microscopic Findings
clinical improvement.176
The most typical pattern is that of foamy histiocytic infil-
trates with associated fibrosis and thickened bone tra-
Radiographic Imaging
beculae. The majority of cells have microscopic features
The typical and virtually diagnostic changes are seen in of ordinary histiocytes. No nuclear grooves are seen (Fig.
the major long tubular bones of the appendicular skele- 12-57). Ultrastructurally, they have features of ordinary
ton (Fig. 12-53) by radiography or bone scintigraphy. histiocytes with numerous lysosomes and lipid vacuoles.
Diffuse bone sclerosis is present in the diaphyseal and No Birbeck granules are present ultrastructurally. Occa-
metaphyseal parts of the long bones (Figs. 12-53 to sionally multinucleated (Touton) giant cells are present.
12-55). Additional foci can be seen in the trunk and Proliferation of histiocytic cells is accompanied by thick-
craniofacial bones (Fig. 12-56). Sclerosis is produced by ened bone trabeculae (Fig. 12-57).

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 879

A B
FIGURE 12-55  ■  Erdheim-Chester disease: radiographic features. A and B, Anteroposterior and lateral radiographs of femur show
mixed sclerosis and lysis of diaphysis that spares bone ends. Coarse trabeculation confined to diaphysis is characteristic of marrow-
infiltrative process.

Immunohistochemical Stains and Erdheim-Chester disease must be distinguished from


Differential Diagnosis other histiocytic cell proliferations, such as conventional
Langerhans cell histiocytosis; lysosomal storage disorders,
Immunophenotypically, the majority of histiocytes in predominantly Gaucher’s disease; bone infarcts; and second-
Erdheim-Chester disease are positive for histiocyte anti- ary foamy histiocytic reactions superimposed on other bone
gens such as CD68, CD163, and lysozyme and negative lesions such as fibrous dysplasia. Correlation with the
for markers of Langerhans cells (S-100, CD1a, and very unique radiologic presentation of Erdheim-Chester
langerin/CD207). However, 20% of cases may show dim disease is the best way to diagnose this entity correctly.
S-100 expression.175 A subset of cases may have scattered In the absence of bone involvement, the diagnosis may
Langerhans cells or small clusters of Langerhans cells be quite difficult. Characteristic patterns of involvement
within the dominant proliferation of histiocytes. In these at various sites support this diagnosis, as do typical his-
cases, small clusters (usually 10 to 100 cells) of Langer- tologic and immunophenotypic findings.
hans cells with nuclear grooves that are positive for S-100 Although not specific, the presence of a BRAF V600E
protein and CD1a can be seen (Fig. 12-58). VE1, an mutation in the absence of other features of Langerhans
antibody specific for BRAF V600E mutation, is positive cell histiocytosis would also support the diagnosis while
in a subset of the histiocytes.156 simultaneously identifying a potential target for therapy.

ERRNVPHGLFRVRUJ
880 12  Hematopoietic Tumors

C
FIGURE 12-56  ■  Erdheim-Chester disease: radiographic features. A, Anteroposterior radiograph of ankle shows coarse trabeculation
in diaphysis of tibia and sparing of bone end. B, Lateral radiograph of skull shows lytic lesion in occipital region. C, Lateral radio-
graph of ankle shows mixed lucency and sclerosis in distal tibial shaft.

Genetic Features and Pathogenesis cases by conventional assays and in up to 100% of cases
by ultrasensitive assays.156,177 The BRAF mutation can
The normal cell counterpart is a histiocyte. The litera- also be demonstrated in a small number of circulating
ture reflects disagreement as to whether Erdheim-Chester monocytes in patients with Erdheim-Chester disease.156
disease represents a clonal disorder or a reactive process. As a result of the BRAF V600E mutation, the mitogen
While some investigators have demonstrated clonality activated protein kinase (MAPK) signaling pathway is
by HUMARA assay and cytogenetic abnormalities, activated, resulting in increased proliferation. Dysregula-
others have found no clonal populations.171,205,208 These tion of multiple chemokines and cytokines results in the
discordant results may be due to the variability of the recruitment of Th1 cells and nonmutated histiocytes
number of pathologic histiocytes in lesions containing to Erdheim-Chester lesions.149 A similar cytokine/
numerous recruited nonpathologic histiocytes. BRAF chemokine profile has been demonstrated in oncogene-
V600E mutation has been detected in more than 50% of induced senescence, an inflammatory protective process

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 881

A B

C D
FIGURE 12-57  ■  Erdheim-Chester disease: microscopic features. A, Histiocytic infiltrate within intertrabecular spaces. Note thickened
bone trabeculae (×100). B, Foamy histiocytes with occasional multinucleated giant cells (×200). C, Higher magnification shows his-
tiocytic infiltrate within marrow (×200). D, Histiocytic infiltrate in dermis (extraskeletal involvement) in patient with Erdheim-Chester
disease (×200). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
882 12  Hematopoietic Tumors

A B

C D
FIGURE 12-58  ■  Erdheim-Chester disease: immunohistochemical features. A and B, Low and intermediate power views of a dominant
proliferation of histiocytes negative for S-100, immunohistochemical stain IMM with a small cluster of Langerhans cells positive for
S-100. Note that the Langerhans cells comprise a minor population relative to histiocytes. C and D, Low and intermediate power
views of similar clusters positive for immunohistochemical stain IMM CD1a. Note presence of nuclear grooves. (A and C, ×100;
B and D, ×400.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 883

that prevents malignant transformation despite the pres- have other extranodal involvement, such as involvement
ence of an oncogenic mutation. It has been proposed that in the upper respiratory or gastrointestinal tract. The
the variety of cells seen in Erdheim-Chester lesions may mean age of onset for patients with skeletal lesions is
be the result of this proinflammatory response.156 approximately 23 years.
Fever and massive cervical lymphadenopathy are
the most frequent symptoms at presentation. Other
Sinus Histiocytosis with symptoms include weight loss, malaise, and night sweats.
Bone involvement in the absence of lymphadenopathy is
Massive Lymphadenopathy rare, reported in only 2% of cases.173 Approximately 25%
(Rosai-Dorfman Disease) of patients have evidence of immune disorders, most fre-
quently antibodies against red blood cells, joint disease
Definition
ranging from mild arthralgia to severe arthritis, and glo-
Sinus histiocytosis with massive lymphadenopathy (Rosai- merulonephritis.168 Some patients may have unusually
Dorfman disease) is a rare, proliferative, histiocytic dis- severe infections, most frequently pneumonia. Quite
order that was described in 1965 by Destombes and often the disease fully manifests after a short period
recognized as a clinicopathologic entity in 1969 by Rosai of a nonspecific fever and pharyngitis. To date, no evi-
and Dorfman.163,198 It is characterized by the enlargement dence of a viral or other infectious etiology has been
of lymph node sinuses caused by an aggregation of his- demonstrated.
tiocytic cells that exhibit marked emperipolesis; that is, Rosai-Dorfman disease is considered a histologically
numerous intact cells, predominately lymphocytes, are benign, proliferative, histiocytic disorder with a variable,
present in the cytoplasm of the histocytic cell. The disease but occasionally fatal, outcome. The majority of patients
is often associated with marked gross enlargement of the have indolent regressive or clinically stable disease
lymph nodes, predominantly in the head and neck area, after several years of follow-up.180 The most important
and primarily affects teenagers and young adults.199 prognostic factors consist of the number and extent of
Primary or secondary involvement of extranodal sites, involvement of extranodal sites, clinically progressive
including the skeleton, is frequent. disease, and the presence of associated autoimmune dis-
orders, predominantly of autoimmune hemolytic anemia.
In the Sinus Histiocytosis with Massive Lymphadenopa-
Clinical Features
thy registry data, 14 of 423 patients died of or with clini-
Rosai-Dorfman disease is rare but occurs more frequently cally active disease.166,167 It appears that patients who died
than was originally thought. By 1990, 423 cases had been of sinus histiocytosis with massive lymphadenopathy have
reported to the registry at Yale University.167 The epide- a tendency to have multiple and more extensive sites of
miologic data, frequency of various sites of involvement, nodal and extranodal involvement. Among the extranodal
and clinical behavior discussed here are based on the sites, involvement of the kidneys, liver, and lungs seems
series of data from the Sinus Histiocytosis with Massive to be particularly ominous because approximately 30%
Lymphadenopathy registry published in 1990 by Foucar of patients with disease at these sites die of or with the
et al167 and are supplemented with current data from this disease. Approximately 10% of patients with bone
registry as provided by Dr. Juan Rosai (Memorial Sloan- involvement die of the disease. The fatal outcome in
Kettering Cancer Center, New York). The youngest these cases is related to the frequent involvement of other
patient had congenital disease, and the oldest was in his extraskeletal sites such as the lungs and kidneys.166
eighth decade of life. The majority of patients are teenag-
ers and young adults. The mean age at onset is 20 years. Radiographic Imaging
The disease is slightly more common in blacks and
males.167 Nearly 80% of patients had cervical lymphade- Skeletal involvement manifests by the presence of solitary
nopathy. Other frequently involved groups of lymph or multifocal lytic lesions with poorly or well-demarcated
nodes, in order of frequency, are the axillary, inguinal, borders (Fig. 12-59).162 The lesions are intramedullary
and mediastinal nodes. Involvement of extranodal sites and are associated with cortical erosion, complete cortical
occurs quite frequently and is seen in approximately 25% disruption, elevation of the periosteum, or a combination
to 40% of patients. The most frequent extranodal sites of these features. Radiographic manifestations and clini-
involved are various structures of the head and neck, cal symptoms suggest an inflammatory disorder, such as
the upper respiratory tract airway, and skin. However, osteomyelitis.
virtually every organ, including the CNS, has been
reported to be involved. Skeletal involvement is relatively Microscopic Findings
uncommon and has been documented in only 25 of 423
cases reported to the Sinus Histiocytosis with Massive In typical cases, the sinuses of lymph nodes are filled with
Lymphadenopathy registry.166 Skeletal lesions can be soli- histiocytes.167,184,191 These cells have prominent pale
tary or multifocal.174,182,200,206 They may be located in any eosinophilic cytoplasm, indistinct borders, and round or
bone of the skeleton. In the long tubular bones, they may oval nuclei with a very fine chromatin pattern and a single
be diaphyseal, metaphyseal, or epiphyseal. The involve- small nucleolus. Nuclear grooves are not present, and
ment of all these sites may be present in one patient. The some of these cells may have several nucleoli. Occasional
disease rarely presents as a primary skeletal lesion or cells with multilobulated nuclei may be present. Mitotic
lesions. The majority of patients with skeletal disease also figures are rare but can be easily identified. Atypical

ERRNVPHGLFRVRUJ
884 12  Hematopoietic Tumors

A B

C D
FIGURE 12-59  ■  Rosai-Dorfman disease: radiographic and microscopic features. A, Lateral radiograph of elbow of teenage boy shows
well-circumscribed, lytic, 2-cm intramedullary focus in lower humeral shaft. B and C, Mixed inflammatory cell infiltrates with promi-
nent components of multinucleated histiocytes as well as lymphocytes and plasma cells. Inset and D, Higher power magnification
shows a mixture of lymphocytes and large histiocytes with foamy cytoplasm. Some of the latter cells contain intracytoplasmic
lymphocytes (emperipolesis). (B and C, ×100; inset and D, ×200.) (B-D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 885

mitoses are not present. Aside from the distinct appear- Langerhans cell histiocytosis detected genomic gains and
ance of the histiocytic cells, the most striking and diag- losses in the Langerhans cells but detected no abnormalt-
nostically important feature of these cells is prominent ies in the histiocytes in areas with features of sinus his-
emperipolesis or lymphophagocytosis (i.e., the presence tiocytosis with massive lymphadenopathy.195
of well-preserved lymphocytes within their cytoplasm)
(Fig. 12-59). In addition to lymphocytes, a smaller
number of phagocytized plasma cells, neutrophils, and
red blood cells are also present. Prominent neutrophilic MYELOID SARCOMA
infiltrates with the formation of microabscesses may be Definition
found in some cases. The lymph node capsule is typically
fibrotic and thickened, and the lymph node shows Myeloid sarcoma is a localized tumor composed of
reactive hyperplasia of follicular centers. Extranodal myeloblasts and variable numbers of more mature
disease has all these features except that histiocytic myeloid cells. The first description of this lesion was in
cells, instead of growing in sinuses, form irregular geo- 1853 by King, who used the term chloroma to describe
graphic areas separated by other inflammatory cells. the green color of the gross mass due to production of
Involvement of the skeleton has the same features. Ini- myeloperoxidase.221
tially the lesions may not have fully developed micro-
scopic diagnostic features; that is, they may show Clinical Features
nonspecific inflammatory cell infiltrates only. The diag-
nosis of Rosai-Dorfman disease can be more difficult in Myeloid sarcoma can occur in three different clinical
extranodal sites, including the skeleton. In general, extra- settings: (1) as a solitary mass in patients without prior or
nodal disease exhibits less prominent emperipolesis, is concurrent leukemia; (2) in association with myelodys-
accompanied by extensive fibrosis, and has fewer histio- plastic disorder or myeloproliferative neoplasm; and (3)
cytic cells. Therefore the likelihood that it can be misdi- in association with acute myelogenous leukemia, either
agnosed is much higher in extranodal sites than in nodal concurrent with leukemia, or at relapse, in which setting
sites, especially in cervical disease. The histiocytic cells it may be the only site of relapse.
in Rosai-Dorfman disease are positive for S-100 protein Myeloid sarcoma is considered a form of acute myeloid
and negative for CD1a, but they coexpress markers of leukemia. Not all patients present with leukemic involve-
ordinary histiocytes.153,164 ment of the blood and bone marrow; however, the major-
ity go on to develop frank acute myeloid leukemia with
a lag time ranging from 1 month to 4 years.229 There are
Genetic Features and Pathogenesis
exceptional cases of spontaneous regression of myeloid
The normal cell counterpart is a histiocyte. The etiology sarcomas and patients who do not develop acute leukemia
remains uncertain. Some cases follow upper respiratory even at 16 years follow-up.227
infection, but no infectious etiology has been confirmed. Involvement of the skin and subcutaneous tissue is
No genetic abnormalities have been demonstrated in typical.223,236 Other sites of involvement in order of
sporadic Rosai-Dorfman disease. decreasing frequency are lymph nodes, testis, intestine,
At least some cases previously thought to represent bone, and CNS.210,224,226,230,234,235 Approximately 3% of
familial Rosai-Dorfman disease have been shown to have myeloid sarcomas involve bone.230 Common sites of
mutations of SLC29A3.189 SLC29A3 encodes human bone involvement include the orbit, sacrum, sinus, ver-
equilibrative nucleoside transporter 3, which may trans- tebrae, sternum, and ribs; however, any bone may be
port nucleosides between lysosomal, cytoplasmic, and involved.214,218,231,238
mitochondrial nucleoside pools. The term H syndrome A diagnosis of myeloid sarcoma is considered synony-
has been proposed to unify multiple disorders with muta- mous with a diagnosis of acute myeloid leukemia, and
tions of SLC29A3, encompassing Faisalabad histiocytosis as such should be treated with systemic chemotherapy,
and at least some cases of familial Rosai-Dorfman followed by stem cell transplant in eligible patients.
disease.154 Patients with H syndrome harbor biallelic Both chemotherapy and transplant improve event-free
mutations and may present with lymphadenopathy with survival.213
histologic features similar to those seen in Rosai-Dorfman
disease. One of 79 patients with this disorder was reported Radiographic Imaging
to have lytic bone lesions; however, histologic features
were not reported.154 The clinical features of H syndrome Radiographic presentation is nonspecific. However, the
vary from sporadic Rosai-Dorfman disease. The most diagnosis can be suspected if a destructive lytic bone
common feature of H syndrome is skin hyperpigmenta- lesion develops in a patient with a history of acute myeloid
tion with hypertrichosis, followed by flexion contractures leukemia, a myelodysplastic disorder, or a myeloprolif-
of the fingers and toes.188 erative neoplasm (Fig. 12-60). In the absence of a history
Notably BRAF V600E mutation has not been detected of a myeloid neoplasm, the radiographic findings may
in sporadic Rosai-Dorfman disease, as has been seen in overlap with a variety of reactive and neoplastic pro-
subsets of patients with Langerhans histiocytosis and cesses. By CT and MRI, myeloid sarcoma is isointense
Erdheim-Chester disease. One study of comparative to bone marrow, and by CT it is isointense to muscle.219
genomic hybridization in patients with concurrent FDG PET is more sensitive than a CT scan in detecting
sinus histiocytosis with massive lymphadenopathy and early disease.220

ERRNVPHGLFRVRUJ
886 12  Hematopoietic Tumors

A B

C D
FIGURE 12-60  ■  Myeloid sarcoma: radiographic features. A, Lateral radiograph of distal femur showing a destructive lytic lesion with
moth-eaten pattern. B, Sagittal magnetic resonance image of the same case as shown in A with destructive lesion of the distal femur
of intermediate signal intensity. C, Anteroposterior radiograph of the proximal femur showing a destructive lytic lesion involving
head, neck, and intertrochanteric region. D, Radioisotopic scan showing diffuse involvement of the axial and proximal appendicular
skeleton with a high signal intensity corresponding to the destructive lesion of the left proximal femur. Same case as shown in C.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 887

Microscopic Findings Rare cases of myeloid sarcoma may exhibit erythroid


differentiation, variably positive for E-cadherin, gly-
Microscopically the lesion represents a space-occupying cophorin A, or hemoglobin A.216 Those rare cases with
proliferation of atypical myeloid cells that efface the megakaryocytic differentiation may be identified by vari-
normal architecture of the involved tissue (Fig. 12-61). able staining with CD61, CD41, CD31, or Factor VIII.
The cellular composition ranges from a homogeneous Markers of lymphoid lineage are generally negative.
population of blasts to a mixed population of blasts and CD20, PAX5, and CD79a are helpful in ruling out B-cell
more mature myeloid precursors, ranging from promy- lymphoma or leukemia. However, in myeloid sarcoma
elocytes to neutrophils. Occasional cases may show more with t(8;21) the blasts frequently express CD19 and
than one line of myeloid differentiation, erythroid dif- PAX5. T cell markers (CD1a, CD3, CD5, CD8) should
ferentiation, or megakaryoblastic features.230 Prominent be negative; however, a subset of myeloid leukemias may
monocytic features are common, with more abundant be positive for CD7, and occasional cases of acute pro-
cytoplasm or folded nuclei.217 When present, immature myelocytic leukemia with t(15;17) are positive for CD2.
eosinophil myelocytes (Fig. 12-62) are a clue to the Although monocytic leukemias frequently express CD4,
diagnosis. myeloid sarcomas are less commonly positive, seen in
only 1% of cases in a large study and 22% of cases in one
small study.230,232
Immunohistochemical Stains and
Other neoplasms with blastic features, such as acute
Differential Diagnosis
lymphoblastic lymphoma and blastic mantle cell lymphoma,
If the diagnosis is suspected, fresh tissue may be submit- must be ruled out. Mantle cell lymphoma expresses B cell
ted for flow cytometric immunophenotyping, cytogenetic markers such as CD20, and nearly every case will show
studies, and molecular studies. If fresh tissue is not overexpression of cyclin D1. Lymphoblastic lymphoma
reserved, immunohistochemical studies will confirm the expresses PAX5 and CD19 and is negative for myeloper-
diagnosis in most cases. FISH for common leukemic oxidase and lysozyme. Blastic plasmacytoid dendritic cell
rearrangements should also be pursued if there is suffi- tumor is positive for CD123, CD4, TCL1, and CD56
cient tissue available. and is negative for myeloperoxidase and lysozyme.232
The list of differential diagnostic possibilities is long. Myeloid sarcoma may be differentiated from pediatric
The diagnosis is aided by history in cases of known pre- small round blue cell tumors such as Ewing’s sarcoma/
ceding acute myeloid leukemia. The diagnosis of primary primitive neuroectodermal tumor (ES/PNET) by evaluating
myeloid sarcoma, however, may be quite difficult, with an immuohiostochemical panel that includes CD99,
misdiagnosis rates ranging from 25% to 100%.213,228 The FLI-1, and cytokeratin. CD99 is positive in ES/PNET;
most common incorrect diagnoses are diffuse large Bcell however, 55% of myeloid sarcomas may also be positive
lymphoma and lymphoblastic lymphoma. for CD99.240 ES/PNETs are negative for CD43 and lyso-
Flow cytometric immunophenotyping, including lym- zyme, positive for FLI-1, and show cytokeratin expres-
phoid, myeloid, and blast markers, is performed in sus- sion in a subset of cases.
pected cases. Often, however, a diagnosis of myeloid Extramedullary hematopoiesis may be a diagnostic con-
sarcoma is not considered at the time of biopsy. A gener- sideration; however, myeloid sarcoma will form a mass,
ous panel of immunohistochemical stains is needed to whereas the cells of extramedullary hematopoiesis will be
confirm the diagnosis and rule out other diagnostic present within normal or slightly expanded spaces of the
possibilities. Lysozyme and CD68 are positive in the preexisting architecture of the involved organ.
majority of cases. Although CD45 is often among the In summary, the most sensitive immunohistochemical
first round of stains when evaluating a “small round stains for myeloid sarcoma are CD43 and lysozyme;
blue cell tumor” or poorly differentiated tumor, the however, these stains are by no means specific, and a full
majority of myeloid sarcomas are negative for CD45 by panel of stains should be performed in cases in which this
immunohistochemical staining. Leukocyte origin is con- diagnosis is suspected.
firmed by CD43 positivity in the vast majority of cases.
Useful stains for blasts include CD34, CD117, and occa-
Genetic Findings and Pathogenesis
sionally TdT. The absence of CD34 staining does not
rule out myeloid sarcoma, because more than half of cases The normal cell counterpart is a myeloid blast, usually
may be negative for CD34 (Fig. 12-62).230 with monocytic or granulocytic features, and occasionally
The myeloid nature of the blasts can be confirmed by with megakaryocytic or erythroid features. The blasts
staining with lysozyme, myeloperoxidase, or CD33; may show maturation to more mature myeloid forms.
however, many cases of myeloid sarcoma have monocytic The reasons for blast homing to extramedullary tissues
or myelomonocytic features, and myeloperoxidase may are uncertain; however, interactions between adhesion
be negative or variably positive. Monocytic markers molecules have been suggested as a possible mechanism.
include CD68 and CD163. CD68 was reported to be In addition, matrix metalloproteinase 9 (MMP9), which
positive in 100% of cases of myeloid sarcoma in a series breaks down extracellular matrix, may play a role in extra-
of 92 patients ; however, occasional cases negative for medullary localization.237
CD68 have also been reported.230,232 The KP-1 clone of Cytogenetic and FISH studies show no specific cyto-
CD68 is more sensitive than the PG-M1 clone.230 Non- genetic or molecular abnormality in myeloid sarcomas,
specific esterase cytochemical stain is also useful for dem- but rather these lesions share features commonly seen in
onstrating monocytic differentiation (Fig. 12-62). otherwise typical leukemias (Table 12-11). Common

ERRNVPHGLFRVRUJ
888 12  Hematopoietic Tumors

A B

C D
FIGURE 12-61  ■  Myeloid sarcoma: microscopic, immunohistochemical and ultrastructural features. A, Myeloid sarcoma with poorly
differentiated myeloblasts (×200). B, Higher power photomicrograph of myeloid sarcoma in A, showing myeloblasts with scant
cytoplasm, open nuclear chromatin, and small nucleoli (×400). C, Photomicrograph of myeloid sarcoma with granulocytic differentia-
tion, showing myeloblasts intermixed with more mature granulocytes, including neutrophils and eosinophil precursors (×400).
D, Ultrastructure of myeloblasts, which exhibits cytoplasmic granules consistent with granulocytic myeloid differentiation (×3500).
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 889

A B

C D

FIGURE 12-62  ■  Myeloid sarcoma: microscopic, immunohistochemical and cytochemical features. A, Photomicrograph of myeloid
sarcoma showing myeloblasts with prominent nucleoli and intermixed eosinophil myelocytes (×200). B, Higher power photomicro-
graph of case seen in A (×400). C, Immunohistochemical stains show myeloblasts positive for CD68 (top) and majority negative
for CD34 (bottom), with rare blasts positive for CD34 (arrow) (×200). D, Photomicrograph of cytochemical stain for naphthol
AS-D chloroacetate esterase showing cytoplasmic staining of myeloblasts indicating monocytic differentiation (×200). (A and
B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
890 12  Hematopoietic Tumors

TABLE 12-11 Myeloid Sarcoma: Common TABLE 12-12 Classification of Mastocytosis


Genetic Findings
Cutaneous Mastocytosis
Trisomy 8 Urticaria pigmentosa/maculopapular cutaneous
Monosomy 7 mastocytosis
t(8;21)(q22;q22); RUNX1-RUNX1T1 Diffuse cutaneous mastocytosis
inv(16)(p13.1;q22) or t(16;16)(p13.1;q22); CBFB-MYH11 Solitary mastocytoma of skin
11q23 rearrangements; MLL and multiple partner genes
t(9;22)(q34;q11.2); BCR-ABL1 Systemic Mastocytosis
NPM mutations Indolent Systemic Mastocytosis
FLT3 internal tandem duplications
Bone marrow mastocytosis
Smoldering systemic mastocytosis
Adapted from Alvarez P, et al: Granulocytic sarcoma of the
small bowel, greater omentum and peritoneum associated Systemic Mastocytosis with Associated Clonal
with a CBFβ/MYH11 fusion and inv(16)(p13q22): a case Hematologic Non-Mast Cell Lineage Disease
report. Int Arch Med 4:3, 2011; McKenna M, et al: Myeloid
sarcoma of the small bowel associated with a CBFβ/MYH11 Aggressive Systemic Mastocytosis
fusion and inv(16)(p13q22): a case report. J Clin Pathol Lymphadenopathic masotycotis with eosinophilia
62:757-759, 2009; Pileri SA, et al: Myeloid sarcoma: clinic-
pathologic, phenotypic and cytogenetic analysis for 92 adult Mast Cell Leukemia
patients. Leukemia 21:340-350, 2007; Schwyzer R, et al: Mast Cell Sarcoma
Granulocytic sarcoma in children with acute myeloblastic Extracutaneous Mastocytoma
leukemia and t(8:21). Med Pediatr Oncol 31:144-149, 1998;
Xavier SG, et al: Granulocytic sarcoma of the small Adapted from Swerdlow SH, et al, eds: World Health
intestine with CBFβ/MYH11 fusion gene: report of an Organization classification of tumours of haematopoietic
aleukaemic case and review of the literature. Leuk Res and lymphoid tissues, Lyon, 2008, IARC Press.
27:1063-1066, 2003.

findings are t(8;21) RUNX1/RUNX1T1 and MLL (also systemic mastocytosis, aggressive systemic mastocytosis,
known as KMT2A) rearrangements (Fig. 12-63).230 The mast cell leukemia, and systemic mastocytosis associated
t(8;21) RUNX1/RUNX1T1 is common in acute myeloid with a hematologic non-mast cell disorder, which may be
leukemia (AML); in myeloid sarcoma, it is associated with chronic or acute, myeloid or lymphoid in nature. Skeletal
orbital masses in pediatric patients.222,233 MLL rearrange- involvement occurs in systemic mastocytosis and not in
ments are common in AML with monocytic differentia- localized cutaneous mastocytosis.
tion, which involves skin more frequently than other
subtypes of AML. Chronic myelogenous leukemia may Definition
progress to blast crisis as a granulocytic sarcoma. In such
cases, t(9;22) BCR-ABL is present. In addition, inv(16) Systemic mastocytosis involves one or more organs in
and various trisomies and monosomies have been reported addition to the skin. A diagnosis of systemic mastocytosis
in myeloid sarcoma.230 Inv(16) CBFB-MYH11 myeloid requires the presence of mast cell aggregates of greater
sarcoma shows characteristic involvement of intestine than or equal to 15 cells within the bone marrow or other
and mesentery and less commonly breast and genitouri- noncutaneous site (major criterion) and the presence
nary sites.211,225,239,241 Rarely, t(15;17) PML-RARA acute of one additional minor criterion. In the absence of
promyelocytic leukemia has been reported to present as mast cell aggregates, the diagnosis may be made by
a primary myeloid sarcoma.231 meeting three minor criteria, which include the follow-
NPM mutations and FLT3 internal tandem duplica- ing: greater than 25% of the mast cells have spindle or
tions have each been detected in approximately 15% of atypical cell morphology, detection of KIT D816V muta-
cases.212,217 A limited study of myeloid sarcomas by array tion, aberrant mast cell expression of CD2 or CD25, and
comparative genomic hybridization showed multiple serum tryptase greater than 20 ng/mL.265 If a KIT muta-
unbalanced chromosomal abnormalities in all seven cases tion is detected in a patient with mast cell–mediated
studied, with numeric abnormalities of chromosome 8 symptoms but criteria are not met for systemic mastocy-
being the most common finding.215 tosis, a diagnosis of primary mast cell activation syn-
drome may be made.267

MASTOCYTOSIS Clinical Features


Mast cell proliferations are broadly classified as cutane- Systemic mast cell disease is rare. Skeletal changes occur
ous mastocytosis and systemic mastocytosis. The World in up to 80% of cases. Bone changes can be the primary
Health Organization classification of mast cell diseases is or only manifestations of the disorder and may play a
provided in Table 12-12.265 Cutaneous mastocytosis critical role in the initial diagnosis.244,257 In contrast to
refers to localized involvement limited to skin lesions, pure cutaneous mastocytosis, which is seen more fre-
almost exclusively seen in the pediatric age group and quently in children, systemic mast cell disease is usually
commonly associated with spontaneous regression. Sys- diagnosed in patients older than age 30 years. The male-
temic mastocytosis is further subclassified as indolent to-female ratio is approximately 1 : 1. The trunk bones,

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 891

Chr 8 Chr 21 der(8) der(21)


23.3 23.2 13
23.1 12
22 11.2 11.1
21.3 21.2 11.1
21.1 11.2
12 21
11.2 11.1 Break point
11.1 RUNX1
11.22 11.21 22.2 22.1
12 11.23 22.3
13
21.1
21.2 RUNX1T1
21.3 RUNX1T1
22.1 RUNX1
22.2
22.3
23
24.1
24.2
A 24.3 RUNX1
Exon 1
Exon 2

Exon 3

Exon 4

Exon 5

Exon 6

Exon 7

Exon 8

Exon 9
tel cen
RUNX1T1

Exon 10
Exon 11

Exon 12
Exon 1

Exon 3
Exon 4
Exon 5

Exon 8
Exon 9
Exon 6
Exon 2

Exon 7
B tel
cen

RUNX1-RUNX1T1 fusion gene

Exon 12
Exon 10

Exon 11
Exon 3
Exon 4
Exon 5

Exon 6

Exon 7

Exon 8
Exon 9
Exon 1
Exon 2

Exon 3

Exon 4

Exon 5

tel cen
Fusion transcript
breakpoint

RUNX1 portion RUNX1T1 portion


C
RUNX1
1 453 aa

177
NRDBn/NRHn: negative regulatory region for DNA binding in N-terminal / TE1, TE2 and TE3: subelements within domain AD
negative regulatory region for heterodimerization domain 1-49 NMTS: nuclear matrix targeting signal domain 351-381
RUNT: p53/RUNT-type transcripton factor, DNA-binding domain 50-170 ID: transcription inhibition domain 371-411
NLS: nuclear localization signal domain 167-183 NRHc: negative regulatory region for heterodimerization
NRDBc: negative regulatory region for DNA binding in C-terminal domain 184-291 in C-terminal domain 372-453
AD: transcription activation domain 243-371 VWRPY: domain 450-453

29
RUNX1T1
1 604 aa

TAFH/NHR1: TATA-box-associated factor 110 / Nervy homology region 1 domain 120-215


NHR2-like: Nervy homology region 2 (promotes the formation RUNX1/RUNX1T1 homodimers) domain 337-403
Zink finger: MYND-type (myeloid, Nervy, and DEAF-1) domain 515-551

RUNX1-RUNX1T1 fusion protein


1 752 aa

breakpoint
RUNX1 portion RUNX1T1 portion
D
FIGURE 12-63  ■  RUNX1-RUNX1T1 fusion in t(8;21) acute myeloid leukemia and myeloid sarcoma. A, The RUNX1 gene is located on
the long arm of chromosome 21. The RUNX1T1 gene is located on the long arm of chromosome 8. As a result of this translocation,
two derivative chromosomes are formed. B, The RUNX1 breakpoint is between exons 5 and 6 at q22. The RUNX1 breakpoint results
in loss of transcription activation domains. The RUNX1T1 breakpoint is between exons 2 and 3 at q22. The RUNX1T1 breakpoint
maintains an almost complete RUNX1T1 gene. C, The resulting hybrid gene typically contains exons 1 through 5 of RUNX1 and
almost the entire reading frame of RUNX1T1. The resulting fusion transcript is transcribed from derivative chromosome 21. D, The
RUNX1-RUNX1T1 fusion protein conserves the RUNT domain and the negative regulatory regions for DNA binding and heterodi-
merization domains of RUNX1 and fuses to an almost complete RUNX1T1 gene. (Based on data from Licht JD: AML1 and the AML1-
ETO fusion protein in the pathogenesis of t(8;21) AML. Oncogene 20:5660-5676, 2001.)

ERRNVPHGLFRVRUJ
892 12  Hematopoietic Tumors

Incidence

30

1 2 3 4 5 6 7 8
Age in decades

FIGURE 12-64  ■  Mastocytosis: peak age incidence and skeletal distribution. Most frequent sites are indicated by solid black arrows.

especially the axial skeleton and the proximal parts of the mast cell mediators. Bisphosphonates may be given for
major long tubular bones, are preferentially involved osteoporosis. Anaphylaxis is treated with epinephrine.
(Fig. 12-64). The most common treatment regimen includes inter-
Anaphylaxis, constitutional flushing, syncope, hypo- feron alpha-2b, with or without corticosteroids. Treat-
tension, dyspnea, tachycardia, and headache are manifes- ment with interferon alfa-2b has been shown to improve
tations of systemic mastocytosis that are mediated by symptoms secondary to mast cell mediator release, as well
substances released by mast cells, such as histamine, as increasing bone density. Cladribine may be used in
heparin, platelet activating factor, TNF, leukotrienes, cases with bulky disease.254,268
prostaglandins, cytokines, and growth factors. In addition Tyrosine kinase inhibitors have been used with occa-
to these general, nonspecific symptoms, most patients sional success but have not been shown to improve overall
with indolent forms of mast cell disease have skin changes survival in the majority of patients. Most patients do not
typical of urticaria pigmentosa. In addition, gastrointes- respond to imatinib because the KIT D816V mutation
tinal symptoms such as abdominal pain, diarrhea, vomit- most commonly seen is resistant to imatinib. Other KIT
ing, and steatorrhea are present in approximately 20% of inhibitors, such as dasatinib and midostaurin, are cur-
patients.255 rently being investigated and appear to be effective in a
The prognosis of systemic mastocytosis varies with subset of patients. Mastocytosis patients with associated
subtype. Patients with indolent systemic mastocytosis non-mast cell neoplasms are treated as appropriate for
have overall survival similar to age-matched controls, the associated neoplasm.254
with a median survival of 198 months. The median sur-
vival times of aggressive systemic mastocytosis, mast cell Radiographic Imaging
leukemia, and systemic mastocytosis associated with a
hematologic non-mast cell disorder are 41 months, 2 The skeletal changes are variable and range from gener-
months, and 24 months, respectively. Advanced age, alized osteopenia or osteosclerosis to focal mixed lytic/
cytopenias, and hypoalbuminemia are predictors of sclerotic changes (Fig. 12-65).260-262,264 Mast cells secrete
poorer prognosis.250 several substances that may play a role as mediators of
Treatment is multimodal and includes avoidance of bone changes. Histamine, the major mast cell product,
mast cell degranulation triggers and drugs that block stimulates bone formation and may be responsible, at

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 893

A B
FIGURE 12-65  ■  Mastocytosis involving bone: radiographic features. A, Anteroposterior radiograph of hand shows generalized osteo-
penia and small punched-out erosions of phalanges and metacarpals. B, Radiograph of left hip of an elderly woman with nonunited
fracture of femoral neck. Generalized osteopenia was caused by mastocytosis involving the skeleton.

least in part, for blastic changes in mast cell disease. Two composed of sheets of mast cells with spindle cell mor-
other products, heparin and prostaglandins, can induce phology, often associated with fibrosis (Fig. 12-66).
bone destruction. Nodular lesions may be paratrabecular or perivascular.
Tryptase levels have been shown to correlate with The nodules may be composed predominantly of mast
the degree of osteoporosis, and biomarkers of bone turn- cells or may be accompanied by a significant number of
over are also elevated.246 In addition to generalized inflammatory cells, including lymphocytes, eosinophils,
skeletal changes, localized destructive, sharply circum- and histiocytes. Distinct targetoid zonation of cell types
scribed lesions have also been described. The skeletal may be seen. Nodules of mast cells may be intermixed
manifestations of indolent mast cell disease have a with eosinophils and surrounded by a rim of small
tendency to be in a form of generalized sclerotic or lymphocytes (Figs. 12-66 and 12-67), or nodules of lym-
osteopenic changes. In aggressive or malignant mast phocytes may be surrounded by mast cells.
cell disease, in addition to generalized skeletal changes,
there is a tendency for focal destructive lesions, which
Immunohistochemical Stains, Special
may result in the development of pathologic fracture.258
Studies, and Differential Diagnosis
The degree of skeletal involvement and tumor burden
can, to some extent, be obtained from the scintigraphic Mast cell granules are best documented by metachro-
patterns of skeletal involvement and urinary histamine matic staining with toluidine blue (Fig. 12-67) or
levels.259,262 Giemsa.247,248,252,256 The granules of mast cells are best
documented by electron microscopy.249,251 Normal and
neoplastic mast cells are positive for mast cell tryptase
Microscopic Findings
and CD117 by immunohistochemical staining. Neoplas-
Mast cells can have a highly variable microscopic appear- tic mast cells may show aberrant expression of CD2 or
ance. The microscopic features range from spindled to CD25. Interpretation of these stains may be difficult in
epithelioid, or they may have a “fried egg” appearance cases in which there are many T lymphocytes that are
with abundant clear cytoplasm (Fig. 12-66). Decreased positive for CD2 with a subset of T cells also positive for
cytoplasmic granules and spindle cell morphology are CD25. CD30 is strongly expressed in 85% of aggressive
features of neoplastic mast cells. systemic mastocytosis, with weaker or no expression of
Mast cell lesions in systemic mastocytosis may be these aberrant markers in more indolent forms of the
diffuse or nodular. Diffuse infiltrates typically are disease.263

ERRNVPHGLFRVRUJ
894 12  Hematopoietic Tumors

A B

C D
FIGURE 12-66  ■  Mastocytosis: microscopic features. A, Low power photomicrograph showing mastocytosis with patchy infiltrates
hugging trabeculae (×100). B, Higher power photomicrograph of A showing some mast cells with spindle cell morphology
and associated fibrosis (×200). C, Mast cell lesion with central aggregate of mast cells surrounded by lymphocytes and eosinophils
(×200). D, Higher power magnification of C showing mast cells with fried egg appearance with abundant clear cytoplasm (×400).
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 895

A B

C D
FIGURE 12-67  ■  Mastocytosis: microscopic and cytochemical features. A and B, Low-power photomicrographs of skin in urticaria
pigmentosa show dermal infiltration of mast cells containing metachromatic granules positive for Geimsa stain. C and D, Intramed-
ullary infiltrates within bone of mast cells admixed with inflammatory cells and fibroblasts. (A-D, ×100.) (C and D, hematoxylin-
eosin.)

ERRNVPHGLFRVRUJ
896 12  Hematopoietic Tumors

KIT

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17

Exon 18
Exon 19
Exon 20
Exon 21
Exon 1

Exon 2

Exon 3
Exon 4

Exon 5

Exon 6

Exon 7

Exon 8

Exon 9
Chr 4
16
15.3
15.2
15.1
14
13
12
11 11 KIT
KIT 5
13.1 12
13.3 13.2
21.2 21.1 1 977 aa
21.3
22 28 115 118 206 212 310 322 409 425 505 551 937
23
24 D816V
25
26 Synonymous substitution Immunoglobulin-like domain
27 Missense substitution Protein kinase, catalytic domain
28
Duplication
31.1
31.2 Insertion inframe
31.3 Deletion inframe
32
33
34
35

FIGURE 12-68  ■  Mastocytosis KITD816V mutation. The chromosomal location and exonal structure of the KIT gene are shown. The
encoded protein structure with its functional domain is also shown. The distribution of mutations detected in various tumors avail-
able in the COSMIC data base is also shown. The most common mutation detected in mastocytosis, D816V, is highlighted. This is
an activating missense mutation involving the substitution from aspartatis acid to valine at amino acid position 816, resulting in a
conformational change of the enzymatic pocket of the activation loop of the catalytic domain of KIT. This mutation causes constitu-
tive activation of KIT and the downstream signaling pathway, resulting in increased proliferation and decreased apoptosis of mast
cells. (Based on data from Verstovsek S: Advanced systemic mastocytosis: the impact of KIT mutations in diagnosis, treatment, and pro-
gression. Eur J Heamat 90:89–98, 2012.)

3. Luc S, Buza-Vidas N, Jacobsen SEW: Delineating the cellular


Genetic Features and Pathogenesis pathways of hematopoietic lineage commitment. Semin Immunol
The normal cell counterpart is a mast cell. Mutation of 20:213–220, 2008.
4. Rodig SJ, Payne EG, Degar BA, et al: Aggressive Langerhans cell
KIT is present in 90% of cases of systemic mastocytosis, histiocytosis following T-ALL: clonally related neoplasms with
seen in both indolent and aggressive forms,242 and 76% persistent expression of constitutively active NOTCH1. Am J
of cutaneous mastocytosis cases also have KIT muta- Hematol 83:116–1121, 2008.
tions.243 The most common mutation in both cutaneous 5. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health
Organization classification of tumours of haematopoietic and lymphoid
and systemic mastocytosis is point mutation D816V tissues, Lyon, 2008, IARC Press.
involving the tyrosine kinase domain in exon 17 (Fig. 6. Yohe SL, Chenault CB, Torlakovic EE, et al: Langerhans cell
12-68).253,268 Other KIT mutations have also been identi- histiocytosis in acute leukemias of ambiguous or myeloid lineage
fied in systemic and cutaneous mastocytosis. KIT in in adult patients: support for a possible clonal relationship. Mod
normal mast cells is activated by binding of stem Pathol 27:651–656:2013.
cell factor (SCF). The KIT D816V mutation causes con- Plasma Cell Myeloma and Plasma Cell Neoplasms
stitutive activation of KIT, resulting in activation of 7. Ahmad N, Haider S, Jagannathan S, et al: MicroRNA theragnos-
downstream signaling pathways including PI3-K/AKT/ tics for the clinical management of multiple myeloma. Leukemia
mTOR, JAK2/STAT1/5, and SFK pathways. Activation 28:732–738, 2014.
8. Annunziata CM, Davis RE, Demchenko Y, et al: Frequent
of KIT results in increased proliferation and survival of engagement of the classical and alternative NF-κB pathways by
mast cells and also plays a role in terminal differentiation, diverse genetic abnormalities in multiple myeloma. Cancer Cell
adhesion, and release of mast cell mediators.243 12:115–130, 2007.
Mutations in addition to KIT mutations are associated 9. Avet-Loiseau H, Garand R, Lode L, et al: Intergroupe Franco-
with aggressive forms of mastocytosis. TET2 mutations phone du Myelome: translocation t(11;14)(q13;q32) is the hall-
mark of IgM, IgE, and nonsecretory multiple myeloma variants.
are seen in approximately 29% of cases of systemic mas- Blood 101:1570–1571, 2003.
tocytosis and confer a worse prognosis.266 Other rela- 10. Bardwick PA, Zvaifler NJ, Gill GN, et al: Plasma cell dyscrasia
tively common mutations involve ASXL1 and SRSF2.243 with polyneuropathy, organomegaly, endocrinopathy, M protein
Mutations in LYN and BTK are less common, each occur- and skin changes: the POEMS syndrome—report on two cases
and a review of the literature. Medicine 59:311–322, 1980.
ring in less than 10% of cases.245 Other mutations char- 11. Bartl R, Frisch B, Fateh-Moghadam A, et al: Histologic classifica-
acteristic of other myeloid neoplasms are seen in systemic tion and staging of multiple myeloma: a retrospective and pro-
mastocytosis associated with these neoplasms. spective study of 674 cases. Am J Clin Pathol 87:342–355, 1987.
12. Bernier GM, Berman JH, Fanger MW: Plasma cell leukemia with
REFERENCES excretion of half-molecules of immunoglobulin A (α1 λ1). Ann
Intern Med 86:572–575, 1977.
Classification of Hematopoietic Tumors 13. Chapman MA, Lawrence MS, Keats JJ, et al: Initial genome
1. Chen W, Wang J, Wang E, et al: Detection of clonal lymphoid sequencing and analysis of multiple myeloma. Nature 471:467–
receptor gene rearrangements in Langerhans cell histiocytosis. 472, 2011.
Am J Surg Pathol 34:1049–1057, 2010. 14. Coriu D, Weaver K, Schell M, et al: A molecular basis for non-
2. Kawamoto H, Wada H, Katsura Y: A revised scheme for develop- secretory myeloma. Blood 104:829–831, 2004.
mental pathways of hematopoietic cells: the myeloid-based model. 15. Dahlin DC, Unni KK: Bone tumors: general aspects and data on 8,542
Int Immunol 22:65–70, 2010. cases, ed 4, Springfield, Ill, 1986, Charles C Thomas.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 897

16. Dao LN, Hanson CA, Dispenzieri A, et al: Bone marrow histo- 41. Ling NR, MacLennan ICM, Mason DY: B-cell and plasma cell
pathology in POEMS syndrome: a distinctive combination of antigens: new and previously defined clusters. In McMichael AJ,
plasma cell, lymphoid, and myeloid findings in 87 patients. Blood Beverley PCL, Cobbold S, editors: Leukocyte typing III: white cell
117:6438–6444, 2011. differentiation antigens, Oxford, England, 1987, Oxford University
17. Dimopoulos K, Gimsing P, Gronbaek K: Aberrant microRNA Press.
expression in multiple myeloma. Eur J Haematol 91:95–105, 2013. 42. Lonial S, Kaufman JL: Non-secretory myeloma: a clinician’s
18. Dimopoulos M, Kyle R, Fermand JP, et al: Consensus recom- guide. Oncology 27:924–928, 2013.
mendations for standard investigative workup: report of the 43. Lorsbach RB, His ED, Dogan A, et al: Plasma cell myeloma and
International Myeloma Workshop Consensus Panel 3. Blood 117: related neoplasms. Am J Clin Pathol 136:168–182, 2011.
4701–4705, 2011. 44. Mikhael JR, Dingli D, Roy V, et al: Mayo Clinic: management
19. Dimopoulos M, Terpos E, Comenzo RL, et al: International of newly diagnosed symptomatic multiple myeloma: updated
Myeloma Working Group consensus statement and guidelines Mayo stratification of myeloma and risk-adapted therapy
regarding the current role of imaging techniques in the diagnosis (mSMART) consensus guidelines 2013. Mayo Clin Proc 88:360–
and monitoring of multiple myeloma. Leukemia 1–12, 2009. 376, 2013.
20. Dispenzieri A: POEMS syndrome: 2014 update on diagnosis, 45. Minarik J, Scudla V, Bacovsky J, et al: Comparison of imaging
risk-stratification and management. Am J Hematol 89:214–223, methoids in POEMS syndrome. Biomed Pap Med Fac Univ Palacky
2014. Olomouc Czech Repub 156:52–57, 2012.
21. Driecer R, Alexanian R: Non-secretory multiple myeloma. Am J 46. Morgan GJ, Walker BA, Davies FE: The genetic architecture of
Hematol 13:313–318, 1982. multiple myeloma. Nature Rev 12:335–348, 2012.
22. Fonseca R, Barlogie B, Bataille R, et al: Genetics and cytogenetics 47. Natkunam Y, Warnke RA, Montgomery K, et al: Analysis of
of multiple myeloma: a workshop report. Cancer Res 64:1546– MUM1/IRF4 protein expression using tissue microarrays and
1558, 2004. immunohistochemistry. Mod Pathol 14:686–694, 2001.
23. Foucar K, Stocker J, Wagner B: Non-neoplastic lymphoid and 48. O’Connell FP, Pinkus JL, Pinkus GS: CD138 (syndecan-1), a
plasma cell disorders. In King D, Gardner W, Sobin L, et al, plasma cell marker immunohistochemical profile in hematopoietic
editors: Non-neoplastic disorders of bone marrow (APJP/fascicle), and nonhematopoietic neoplasms. Am J Clin Pathol 121:254–263,
Washington, DC, 2008, American Registry of Pathology, 2004.
pp 249–282. 49. Osanto S, Müller HP, Schuit HR, et al: Primary plasma cell leu-
24. Gailani S, Seon BK, Henderson ES: Plasma cell leukemia: response kemia: a case report and a review of the literature. Acta Haematol
to conventional myeloma therapy. J Med 8:403–414, 1977. 70:122–129, 1983.
25. Greenberg P, Parker RG, Fu YS, et al: The treatment of solitary 50. Papadimitriou CS, Schwarze EW: Extramedullary non-
plasmacytoma of bone and extramedullary plasmacytoma. Am J gastrointestinal plasmacytoma: an immunohistochemical study of
Clin Oncol 10:199–204, 1987. 16 cases. Pathol Res Pract 176:306–312, 1983.
26. Greipp PR, Raymond NM, Kyle RA, et al: Multiple myeloma: 51. Peterson LC, Kueck B, Arthur DC, et al: Systemic polyclonal
significance of plasmablastic subtype in morphological classifica- immunoblastic proliferations. Cancer 61:1350–1358, 1988.
tion. Blood 65:305–310, 1985. 52. Preud’homme JL, Hurez D, Danon F, et al: Intracytoplasmic and
27. Gualco G, Weiss LM, Bacchi CE: MUM1/IRF4: a review. Appl surface-bound immunoglobulins in nonsecretory and Bence-
Immunohistochem Mol Morphol 18:301–310, 2010. Jones myeloma. Clin Exp Immunol 25:428–436, 1976.
28. Hall FM, Gore SM: Osteosclerotic myeloma variants. Skeletal 53. Roccaro AM, Sacco A, Maiso P, et al: BM mesenchymal stromal
Radiol 17:101–105, 1988. cell-derived exosomes facilitate multiple myeloma progression.
29. Heerema-McKenney A, Waldron J, Hughes S, et al: Clinical, J Clin Invest 123:1542–1555, 2013.
immunophenotypic, and genetic characterization of small 54. Roccaro AM, Sacco A, Thompson B, et al: MicroRNAs 15a and
lymphocyte-like plasma cell myeloma. Am J Clin Pathol 133:265– 16 regulate tumor proliferation in multiple myeloma. Blood
270, 2010. 113:6669–6680, 2009.
30. Hernandez JA, Land KJ, McKenna RW: Leukemias, myeloma, 55. Saqi A, Yun SS, Yu GH, et al: Utility of CD138 (syndecan-1) in
and other lymphoreticular neoplasms. Cancer 75(Suppl):381–394, distinguishing carcinomas from mesotheliomas. Diagn Cytopathol
1995. 33:65–70, 2005.
31. Hideshima T, Mitsiades C, Tonon G, et al: Understanding mul- 56. Smith DB, Harris M, Gowland E, et al: Non-secretory multiple
tiple myeloma pathogenesis in the bone marrow to identify new myeloma: a report of 13 cases with a review of the literature.
therapeutic targets. Nat Rev Cancer 8:585–598, 2007. Hematol Oncol 4:307–313, 1986.
32. Hurez D, Preud’homme JL, Seligmann M: Intracellular “mono- 57. Surveillance, Epidemiology and End Results Program, National
clonal” immunoglobulin in nonsecretory human myeloma. Cancer Institute. http://seer.cancer.gov/data/citation.html.
J Immunol 104:263–264, 1970. 58. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health
33. Kapadia SB, Desai U, Cheng VS: Extramedullary plasmacytoma Organization classification of tumours of haematopoietic and lymphoid
of the head and neck: a clinicopathologic study of 20 cases. Medi- tissues, Lyon, 2008, IARC Press.
cine 61:317–329, 1982. 59. Tomecki R, Drazkowska K, Kucinski I, et al: Multiple myeloma-
34. Kilciksiz S, Karakoyun-Celik O, Agaoglu FY, et al: A review for associated hDIS3 mutations cause perturbations in cellular RNA
solitary plasmacytoma of bone and extramedullary plasmacytoma. metabolism and suggest hDIS3 PIN domain as a potential drug
ScientificWorldJournal 1-6, 2012. target. Nucleic Acid Res 42:1270–1290, 2014.
35. Knowling MA, Harwood AR, Bergsagel DE: Comparison of 60. Weber DM: Solitary bone and extramedullary plasmacytoma.
extramedullary plasmacytomas with solitary and multiple plasma Hematology Am Soc Hematol Educ Program 373–376, 2005.
cell tumors of the bone. J Clin Oncol 1:255–262, 1983. 61. West NC, Smith AM, Ward R: IgE myeloma associated with
36. Kuehl WM, Bergsagel PL: Molecular pathogenesis of multiple plasma cell leukemia. Postgrad Med J 59:784–785, 1983.
myeloma and its premalignant precursor. J Clin Investig 122:3456– 62. Wiltshaw E: The natural history of extramedullary plasmacytoma
3463, 2012. and its relation to solitary myeloma of bone and myelomatosis.
37. Kumar SK, Rajkumar SV, Dispenzieri A, et al: Improved survival Medicine 55:217–238, 1976.
in multiple myeloma and the impact of novel therapies. Blood 63. Woodruff RK, Malpas JS, Paxton AM, et al: Plasma cell leukemia
111:2516–2520, 2008. (PCL): a report on 15 patients. Blood 52:839–845, 1978.
38. Kyle RA, International Myeloma Working Group: Criteria for the
classification of monoclonal gammopathies, multiple myeloma Non-Hodgkin’s Lymphoma
and related disorders: a report of the International Myeloma 64. Akasaka H, Akasaka T, Kurata M, et al: Molecular anatomy of
Working Group. Br J Haematol 121:749–757, 2003. BCL6 translocations revealed by long-distance polymerase chain
39. Kyle RA, Gertz MA: Primary systemic amyloidosis: clinical reaction-based assays. Cancer Res 60:2341–2355, 2000.
and laboratory features in 474 cases. Semin Hematol 32:45–59, 65. Akasaka T, Akasaka H, Yonetani N, et al: Refinement of the
1995. BCL2/immunoglobulin heavy chain fusion gene in t(14;18)
40. Kyle RA, Rajkumar SV: Multiple myeloma. Blood 111:2962–2972, (q32;q21) by polymerase chain reaction amplification for long
2008. targets. Genes Chromosomes Cancer 21:17–29, 1998.

ERRNVPHGLFRVRUJ
898 12  Hematopoietic Tumors

66. Alencar A, Pitcher D, Byrne G, et al: Primary bone lymphoma— 89. Krishnan A, Shirkhoda A, Tehranzadeh J, et al: Primary bone
the University of Miami experience. Leuk Lymphoma 51:39–49, lymphoma: radiographic-MR imaging correlation. Radiographics
2010. 23:1371–1383, 2003.
67. Alizadeh AA, Eisen MB, Davis RE, et al: Distinct types of diffuse 90. Lim KH, Yang Y, Staudt LM: Pathogenetic importance and thera-
large B-cell lymphoma identified by gene expression profiling. peutic implications of NF-κB in lymphoid malignancies. Immunol
Nature 403:503–511, 2000. Rev 246:359–378, 2012.
68. Baar J, Burkes RL, Bell R, et al: Primary non-Hodgkin’s lym- 91. Lu Z, Tsai AG, Akasaka T, et al: BCL6 breaks occur at different
phoma of bone: a clinicopathologic study. Cancer 73:1194–1199, AID sequence motifs in Ig-BCL6 and non-Ig-BCL6 rearrange-
1994. ments. Blood 121:4451–4454, 2013.
69. Bacci G, Jaffe N, Emiliani E, et al: Therapy for primary non- 92. Lynnhtun K, Renthawa J, Varikatt W: Detection of MYC rear-
Hodgkin’s lymphoma of bone and a comparison of results with rangement in high grade B cell lymphomas: correlation of MYC
Ewing’s sarcoma: 10 years’ experience at the Istituto Ortopedico immunohistochemistry and FISH analysis. Pathology 46:211–215,
Rizzoli. Cancer 57:1468–1472, 1986. 2014.
70. Bazarbachi A, Suarez F, Fields P, et al: How I treat adult T-cell 93. Marriott SJ, Lemoine FJ, Jeang KT: Damaged DNA and mis-
leukemia/lymphoma. Blood 118:1736–1745, 2011. counted chromosomes: human T cell leukemia virus type I tax
71. Bellon M, Ko NL, Lee MJ, et al: Adult T-cell leukemia cells oncoprotein and genetic lesions in transformed cells. J Biomed Sci
overexpress Wnt5a and promote osteoclast differentiation. Blood 9:292–298, 2002.
121:5045–5054, 2013. 94. Matsuoka M, Yasanaga J: Human T-cell leukemia virus type 1:
72. Bittencourt AL, da Gracas Vieira M, Brites CR, et al: Adult replication, proliferation and propagation by Tax and HTLV-1
T-cell leukemia/lymphoma in Bahia, Brazil: analysis of prognostic bZIP factor. Curr Opin Virol 3:684–691, 2013.
factors in a group of 70 patients. Am J Clin Pathol 128:875–882, 95. Meyer PN, Fu K, Greiner TC, et al: Immunohistochemical
2007. methods for predicting cell of origin and survival in patients with
73. Boston HC, Jr, Dahlin DC, Ivins JC, et al: Malignant lymphoma diffuse large B-cell lymphoma treated with rituximab. J Clin Oncol
(so-called reticulum cell sarcoma) of bone. Cancer 34:1131–1137, 29:200–207, 2011.
1974. 96. Mikhaeel NG: Primary bone lymphoma. Clin Oncol (R Coll Radiol)
74. Chan JKC, Banks PM, Cleary ML, et al: A revised European- 24:366–370, 2012.
American classification of lymphoid neoplasms proposed by the 97. Oberling C: Les reticulosarcomes et les reticuloendotheliosar-
International Lymphoma Study Group: a summary version. Am J comes de la moelle osseuse (sarcomes d’Ewing). Bull Assoc Fr
Clin Pathol 103:543–560, 1995. Cancer 17:259–296, 1928.
75. Choi WWL, Weisenburger DD, Greiner TC, et al: A new immu- 98. Ohno H: Pathogenetic and clinical implications of non-
nostain algorithm classifies diffuse large B-cell lymphoma into immunoglobulin BCL6 translocations in B-cell non-Hodgkin’s
molecular subtypes with high accuracy. Clin Cancer Res 15:5494– lymphoma. J Clin Exp Hematol 46:43–53, 2006.
5502, 2009. 99. Parker F, Jr, Jackson H, Jr: Primary reticulum cell sarcoma of
76. Clayton F, Butler JJ, Ayala AG, et al: Non-Hodgkin’s lymphoma bone. Surg Gynecol Obstet 68:45–53, 1939.
of bone: pathologic and radiologic features with clinical correlates. 100. Pasqualucci L: The genetic basis of diffuse large B-cell lymphoma.
Cancer 60:2494–2501, 1987. Curr Opin Hematol 20:336–344, 2013.
77. Cui X, Lu Z, Kurosawa A, et al: Both CpG methylation and 101. Phillips WC, Kattapuram SV, Dosoretz DE, et al: Primary lym-
activation-induced deaminase are required for the fragility of the phoma of bone: relationship of radiographic appearance and prog-
human BCL-2 major breakpoint region: implications of the timing nosis. Radiology 144:285–290, 1982.
of the breaks in the t(14;18) translocation. Mol Cell Biol 33:947– 102. Ramadan KM, Shenkier T, Sehn LH, et al: A clinicopathological
957, 2013. retrospective study of 131 patients with primary bone lymphoma:
78. Dosoretz DE, Murphy GF, Raymond AK, et al: Radiation therapy a population-based study of successively treated cohorts from
for primary lymphoma of bone. Cancer 51:44–46, 1983. the British Columbia Cancer Agency. Ann Oncol 18:129–135,
79. Dubey P, Ha CS, Besa PC, et al: Localized primary malignant 2007.
lymphoma of bone. Int J Radiat Oncol Biol Phys 37:1087–1093, 103. Reid RL, Lindholm PF, Mireskandari A, et al: Stabilization of
1997. wild-type p53 human T-lymphocytes transformed by HTLV-I.
80. George CD, Wilson AG, Philpott NJ, et al: The radiological Oncogene 8:3029–3036, 1993.
features of adult T-cell leukaemia/lymphoma. Clin Radiol 49:83– 104. Reimer RR, Chabner BA, Young RC, et al: Lymphoma presenting
88, 1994. in bone: results of histopathology, staging, and therapy. Ann Intern
81. Grassmann R, Aboud M, Jeang KT: Molecular mechanisms of Med 87:50–55, 1977.
cellular transformation by HTLV-1 tax. Oncogene 24:5976–5985, 105. Rosenberg SA: Classification of lymphoid neoplasms. Blood
2005. 84:1359–1360, 1994.
82. Green TM, Young KH, Visco C, et al: Immunohistochemical 106. Rosenberg SA, Berard CW, Brown BW, Jr, et al: National Cancer
double-hit score is a strong predictor of outcome in patients with Institute sponsored study of classifications of non-Hodgkin’s lym-
diffuse large B-cell lymphoma treated with rituximab plus cyclo- phomas: summary and description of a working formulation for
phosphamide, doxorubicin, vincristine, and prednisone. J Clin clinical usage. Cancer 49:2112–2135, 1982.
Oncol 30:3460–3467, 2012. 107. Rosenwald A, Wright G, Chan WC, et al: The use of molecular
83. Gu X, Shivarov V, Strout MP: The role of activation-induced profiling to predict survival after chemotherapy for diffuse large-
cytidine deaminase in lymphomagenesis. Curr Opin Hematol B-cell lymphoma. N Engl J Med 346:1937–1947, 2002.
19:292–298, 2012. 108. Sakashita A, Hattori T, Miller CW, et al: Mutaitons of the p53
84. Hans CP, Weisenburger DD, Greiner TC, et al: Confirmation of gene in adult T-cell leukemia. Blood 79:477–480, 1992.
the molecular classification of diffuse large B-cell lymphoma by 109. Shen HM, Peters A, Baron B, et al: Mutation of BCL-6 gene in
immunohistochemistry using a tissue microarray. Blood 103:275– noarmal B cells by the process of somatic hypermutations of Ig
282, 2004. genes. Science 280:1750–1752, 1998.
85. Harris NL, Jaffe ES, Stein H, et al: A revised European-American 110. Shimoyama M: Diagnostic criteria and classification of clinical
classification of lymphoid neoplasms: a proposal from the subtypes of adult T-cell leukaemia-lymphoma. A report from the
International Lymphoma Study Group. Blood 84:1361–1392, Lymphoma Study Group (1984-87). Br J Haematol 79(3):428–437,
1994. 1991.
86. Hong F, Kahl BS, Gray R: Incremental value in outcome predic- 111. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health
tion with gene expression-based signatures in diffuse large B-cell Organization classification of tumours of haematopoietic and lymphoid
lymphoma. Blood 121:156–158, 2013. tissues, Lyon, 2008, IARC Press.
87. Jawad MU, Schneiderbauer MM, Min ES, et al: Primary lym- 112. Stansfeld AG, Diebold J, Noel H, et al: Updated Kiel classifica-
phoma of bone in adult patients. Cancer 116:871–879, 2010. tion for lymphomas. Lancet 1:292–293, 1988.
88. Kitsoulis P, Vlychou M, Papoudou-Bai A, et al: Primary lympho- 113. Tsukasaki K, Hermine O, Bazarbachi A, et al: Definition, prog-
mas of bone. Anticancer Res 26:325–338, 2006. nostic factors, treatment, and response criteria of adult T-cell

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 899

leukemia-lymphoma: a proposal from an International Consensus 140. Otto C, Giefing M, Massow A, et al: Genetic lesions of the
Meeting. J Clin Oncol 27:453–459, 2009. TRAF3 and MAP3K14 genes in classical Hodgkin lymphoma.
114. Vose JM: Peripheral T-cell non-Hodgkin’s lymphoma. Hematol Br J Haematol 157:702–708, 2012.
Oncol Clin North Am 22:997–1005, 2008. 141. Pinkus GS, Thomas P, Said JW: Leu-M1: A marker for Reed-
115. Warnke RA, Weiss LM, Chan JKC, et al: Rosai J, editor: Atlas Sternberg cells in Hodgkin’s diease: an immunoperoxidase
of tumor pathology: tumors of the lymph nodes and spleen, 3rd study of paraffin-embedded tissues. Am J Pathol 119:244–252,
series, Washington, DC, 1994, Armed Forces Institute of 1985.
Pathology. 142. Scott DW, Chan FC, Hong G, et al: Gene expression-based
116. Xu Y, McKenna R, Doolitle J, et al: The t(14;18) in diffuse large model using formalin-fixed paraffin-embedded biopsies predicts
b-cell lymphoma: correlation with germinal center-associated overall survival in advanced-stage classical Hodgkin lymphoma.
markers and clinical features. Appl Immunohistochem Mol Morphol J Clin Oncol 31:692–700, 2013.
13:116–123, 2005. 143. Steidl C, Lee T, Shah SP: Tumor-associated macrophages and
117. Zhang J, Grubor V, Love CL, et al: Genetic heterogeneity of survival in classic Hodgkin’s lymphoma. N Engl J Med 362:875–
diffuse large B-cell lymphoma. Proc Natl Acad Sci 110:1398–1403, 885, 2010.
2013. 144. Surveillance, Epidemiology and End Results Program, National
118. Zhang PJ, Barcos M, Stewart CC, et al: Immunoreactivity of Cancer Institute. http://seer.cancer.gov/data/citation.html.
MIC2 (CD99) in acute myelogenous leukemia and related dis- 145. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health
eases. Mod Pathol 13:452–458, 2000. Organization Classification of tumours of haematopoietic and lymphoid
119. Zhao T, Matsuoka M: HBZ and its role in HTLV-1 oncogenesis. tissues, Lyon, 2008, IARC Press.
Front Microbiol 3:247, 2012. 146. Zhao FX: Nodular lymphocyte-predominant Hodgkin lymphoma
or T-cell/histiocyte rich large B-cell lymphoma: the problem
Classical Hodgkin’s Lymphoma in “grey zone” lymphomas. Int J Clin Exp Pathol 1:300–305,
120. Burke JS: Hodgkin’s disease: histopathology and differential diag- 2008.
nosis. In Knowles DM, editor: Neoplastic hematopathology, Balti- 147. Zukerberg LR, Collins AB, Ferry JA, et al: Coexpression of CD15
more, 1992, Williams & Wilkins. and CD20 by Reed-Sternberg cells in Hodgkin’s disease. Am J
121. Chetaille B, Bertucci F, Finetti P: Molecular profiling of classical Pathol 139:475–483, 1991.
Hodgkin lymphoma tissues uncovers variations in the tumor
microenvironment and correlations with EBV infection and Histiocytic and Dendritic Cell Proliferations
outcome. Blood 113:2765–2775, 2009. 148. Allen CE, Li L, Peters TL, et al: Cell-specific gene expression in
122. Correa P, O’Conor GT: Epidemiologic patterns of Hodgkin’s Langerhans cell histiocytosis lesions reveals a distinct profile com-
disease. Int J Cancer 8:192–201, 1971. pared to epidermal Langerhans cells1. J Immunol 184:4557–4567,
123. Frierson HF, Jr, Innes DJ, Jr: Sensitivity of anti–Leu-M1 as a 2010.
marker in Hodgkin’s disease. Arch Pathol Lab Med 109:1024–1028, 149. Arnaud L, Gorochov G, Charlotte F, et al: Systemic perturbation
1985. of cytokine and chemokine networks in Erdheim-Chester disease:
124. Gold RH, Mirra JM: Case report 101: primary Hodgkin disease a single-center series of 37 patients. Blood 117:2783–2790, 2011.
of humerus. Skeletal Radiol 4:233–235, 1979. 150. Azouz EM, Saigal G, Rodriguez MM, et al: Langerhans’ cell his-
125. Granger W, Whitaker R: Hodgkin’s disease in bone, with tiocytosis: pathology, imaging and treatment of skeletal involve-
special reference to periosteal reaction. Br J Radiol 40:939–948, ment. Pediatr Radiol 35:103–115, 2008.
1967. 151. Badalian-Very G, Vergilio JA, Degar BA, et al: Recurrent BRAF
126. Grufferman S, Delzell E: Epidemiology of Hodgkin’s disease. mutations in Langerhans cell histiocytosis. Blood 116:1919–1923,
Epidemiol Rev 6:76–106, 1984. 2010.
127. Gutensohn N, Cole P: Childhood social environment and Hodg- 152. Berres ML, Lim KP, Peters T, et al: BRAF-V600E expression in
kin’s disease. N Engl J Med 304:135–140, 1981. precursor versus differentiated dendritic cells defines clinically
128. Jarrett RF, Stark GL, White J: Impact of tumor Epstein-Barr virus distinct LCH risk groups. J Exp Med 211:669–683, 2014.
status on presenting features and outcome in age-defined sub- 153. Bonetti F, Chilosi M, Menestrina F, et al: Immunohistological
groups of patients with classic Hodgkin lymphoma: a population- analysis of Rosai-Dorfman histiocytosis: a disease of S-100+CD11
based study. Blood 106:2444–2451, 2005. histiocytes. Virchows Arch A 411:129–135, 1987.
129. Kadin ME: Hodgkin’s disease: immunobiology and pathogenesis. 154. Bolze A, Abhyankar A, Grant AV, et al: A mild form of SLC29A3
In Knowles DM, editor: Neoplastic hematopathology, Baltimore, disorder: a frameshift deletion leads to the paradoxical translation
1992, Williams & Wilkins. of an otherwise noncoding mRNA splice variant. PLoS ONE
130. Kamper P, Bendix K, Hamilton-Dutoit S, et al: Tumor-infiltrating 7:e29708, 2012.
macrophages correlate with adverse prognosis and Epstein-Barr 155. Burns BF, Colby TV, Dorfman RF: Langerhans cell granuloma-
virus status in classical Hodgkin’s lymphoma. Haematologica tosis (histiocytosis X) associated with malignant lymphomas. Am
96:269–276, 2011. J Surg Pathol 7:529–533, 1983.
131. Kaplan HS: Hodgkin’s disease, ed 2, Cambridge, 1980, Harvard 156. Cangi MG, Biavasco R, Cavalli G, et al: BRAF V600E mutation
University Press. is invariably present and associated with oncogene-induced
132. Kuppers R, Engert A, Hansmann ML: Hodgkin lymphoma. J Clin senescence in Erdheim-Chester disease. Ann Rheum Dis 2014.
Invest 122:3439–3447, 2012. doi: 10.1136/annrheumdis-2013-204924. Mar 26, [Epub ahead of
133. Li Y, Wang XB, Tian XY, et al: Unusual primary osseous Hodgkin print].
lymphoma in rib with associated soft tissue mass: a case report and 157. Cantu MA, Lupo PJ, Bilgi M, et al: Optimal therapy for adults
review of literature. Diagn Pathol 7:64, 2012. with Langerhans cell histiocytosis bone lesions. PLoS ONE 7:
134. MacMahon B: Epidemiological evidence on the nature of Hodg- e43257, 2012.
kin’s disease. Cancer 10:1045–1054, 1957. 158. Cavalli G, Guglielmi B, Berti A, et al: The multifaceted clinical
135. Mangala gouri SR, Rao SG: Hodgkin’s lymphoma of bone—a rare presentations and manifestations of Erdheim-Chester disease:
case report. J Clin Diagn Res 8:160–161, 2014. comprehensive review of the literature and of 10 cases. Ann Rheum
136. Mills SE, Sloop FB, Jr, Thiele AL, et al: Case report 251: hodgkin Dis 72:1691–1695, 2013.
disease, nodular sclerosing variant, primary form. Skeletal Radiol 159. Chester W: Über Lipoidgranulomatose. Virchows Arch A 279:561–
10:287–289, 1983. 602, 1930.
137. Muschen M, Re D, Brauninger A, et al: Somatic mutations of the 160. Chow A, Brown BD, Merad M: Studying the mononuclear
CD95 gene in Hodgkin and Reed-Sternberg cells. Cancer Res phagocyte system in the molecular age. Nature Rev 11:788–798,
60:5640–5643, 2000. 2011.
138. Newcomer LN, Silverstein MB, Cadman EC, et al: Bone involve- 161. da Costa CE, Annels NE, Faaij CM, et al: Presence of osteoclast-
ment in Hodgkin’s disease. Cancer 49:338–342, 1982. like multinucleated giant cells in the bone and nonostotic
139. Ostrowski ML, Inwards CY, Strickler JG, et al: Osseous Hodgkin lesions of Langerhans cell histiocytosis. J Exp Med 201:687–693,
disease. Cancer 85:1166–1178, 1999. 2005.

ERRNVPHGLFRVRUJ
900 12  Hematopoietic Tumors

162. Demicco EG, Rosenberg AE, Bjornsson J, et al: Primary Rosai- 187. Minkov M, Prosch H, Steiner M, et al: Langerhans cell histiocy-
Dorfman disease of bone: a clinicopathologic study of 15 cases. tosis in neonates. Pediatr Blood Cancer 45:802–807, 2005.
Am J Surg Pathol 34:1324–1333, 2010. 188. Molho-Pessach V, Ramot Y, Camille F, et al: H syndrome: the first
163. Destombes P: Adenitis with lipid excess in children or young 79 patients. J Am Acad Dermatol 70:80–88, 2014.
adults, seen in the Antilles and in Mali (4 cases). Bull Soc Pathol 189. Morgan NV, Morris MR, Cangul H, et al: Mutations, SLC29A3,
Exot Filiales 58:1169–1175, 1965. encoding an equilibrative nucleoside transporter ENT3, cause
164. Eisen RN, Buckley PJ, Rosai J: Immunophenotypic characteriza- a familial histiocytosis syndrome (Faisalabad histiocytosis)
tion of sinus histiocytosis with massive lymphadenopathy (Rosai- and familial Rosai-Dorfman disease. PLoS Genet 6:e1000833,
Dorfman disease). Semin Diagn Pathol 7:74–82, 1990. 2010.
165. Foucar K, Foucar E: The mononuclear phagocyte and immuno- 190. Moulis G, Sailler L, Bonneville F, et al: Imaging in Erdheim-
regulatory effector (M-PIRE) system: evolving concepts. Semin Chester disease: classic features and new insights. Clin Exp Rheu-
Diagn Pathol 7:4–18, 1990. matol 32:410–414, 2014.
166. Foucar E, Rosai J, Dorfman RF: Sinus histiocytosis with massive 191. Nawroz IM, Wilson-Storey D: Sinus histiocytosis with massive
lymphadenopathy: an analysis of 14 deaths occurring in a patient lymphadenopathy (Rosai-Dorfman disease). Histopathology 14:91–
registry. Cancer 54:1834–1840, 1984. 99, 1989.
167. Foucar E, Rosai J, Dorfman RF: Sinus histiocytosis with massive 192. Neumann MP, Frizzera G: The coexistence of Langerhans cell
lymphadenopathy (Rosai-Dorfman disease): review of the entity. granulomatosis and malignant lymphoma may take different
Semin Diagn Pathol 7:19–73, 1990. forms: report of seven cases with a review of the literature. Hum
168. Foucar E, Rosai J, Dorfman RF, et al: Immunological abnormali- Pathol 17:1060–1065, 1986.
ties and their significance in sinus histiocytosis with massive 193. Nezelof C, Basset F, Rousseau MF: Histiocytosis X: histogenetic
lymphadenopathy. Am J Clin Pathol 82:515–525, 1984. arguments for a Langerhans cell origin. Biomedicine 18:365–371,
169. Gadner H, Grois N, Potschger U: Improved outcome in multi- 1973.
system Langerhans cell histiocytosis is associated with therapy 194. O’Connell RM, Rao DS, Chaudhuri AA, et al: Physiological and
intensification. Blood 111:2556–2562, 2008. pathological roles for microRNAs in the immune system. Nature
170. Goldner MG, Volk BW: Fulminant normocholeteremic xantho- Rev 10:111–122, 2010.
matosis (histiocytosis X). Arch Intern Med 689–698, 1953. 195. O’Malley DP, Duong A, Barry TS, et al: Co-occurrence of Lang-
171. Gong L, He XL, Li YH, et al: Clonal status and clinicopathologi- erhans cell histiocytosis and Rosai-Dorfman disease: possible rela-
cal feature of Erdheim-Chester disease. Pathol Res Pract 205:601, tionship of two histiocytic disorders in rare cases. Mod Pathol
2009. 23:1616–1623, 2010.
172. Grois N, Potschger U, Prosch H, et al: DALHX- and LCH I and 196. Oweity T, Schneithauer BW, Ching HS, et al: Multiple system
II Study Committee: risk factors for diabetes insipidus in Lang- Erdheim-Chester disease with massive hypothalamic-sellar
erhans cell histiocytosis. Pediatr Blood Cancer 46:228–233, 2006. involvement and hypopituitarisms. J Neurosurg 96:344–351,
173. Gupta P, Babyn P: Sinus histiocytosis with massive lymphade- 2002.
nopathy (Rosai-Dorfman disease): a clinicoradiological profile of 197. Romani N, Clausen BE, Stoitzner P: Langerhans cells and more:
three cases including two with skeletal disease. Pediatr Radiol langerin-expressing dendritic cell subsets in the skin. Immunol Rev
38:721–728, 2008. 234:120–121, 2010.
174. Hamels J, Fiasse L, Thiery J: Atypical lymphohistiocytic bone 198. Rosai J, Dorfman RF: Sinus histiocytosis with massive lymphade-
tumour (osseous variant of Rosai-Dorfman disease?). Virchows nopathy: a newly recognized benign clinicopathological entity.
Arch A 408:183–189, 1985. Arch Pathol 87:63–70, 1969.
175. Haroche J, Arnaud L, Cohen-Aubart F, et al: Erdheim-Chester 199. Rosai J, Dorfman RF: Sinus histiocytosis with massive lymphade-
disease. Rheum Dis Clin N Am 39:299–311, 2013. nopathy: a pseudolymphomatous benign disorder—analysis of 34
176. Haroche J, Cohen-Aubart F, Emile JF, et al: Dramatic efficacy of cases. Cancer 30:1174–1188, 1972.
vemurafenib in both multisystemic and refractory Erdheim- 200. Sartoris DJ, Resnick D: Osseous involvement in sinus histiocytosis
Chester disease and Langerhans cell histiocytosis harboring the with massive lymphadenopathy (Rosai-Dorfman disease). Eur J
BRAF V600 mutation. Blood 121:1495–1500, 2013. Pediatr 145:238–240, 1986. (letter).
177. Haroche J, Charlotte F, Arnaud L, et al: High prevalence of BRAF 201. Satpathy AT, Wu X, Albring JC, et al: Re(de)fining the dendritic
V600E mutations in Erdheim-Chester disease but not in other cell image. Nature Rev 13:1145–1154, 2012.
non-Langerhans cell histiocytoses. Blood 120:2700–2703, 2012. 202. Schuster C, Vaculik C, Fiala C, et al: HLA-DR leukocytes acquire
178. Jaffe HL: Lipid (cholesterol) granulomatosis. In Metabolic, degen- CD1 antigens in embryonic and fetal human skin and contain
erative and inflammatory diseases of the bone and joints, Philadelphia, functional antigen-presenting cells. JEM 206:169, 2009.
1972, Lea & Febiger. 203. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health
179. Jubran RF, Marachelian A, Dorey F, et al: Predictors of outcome Organization classification of tumours of haematopoietic and lymphoid
in children with Langerhans cell histiocytosis. Pediatr Blood Cancer tissues, Lyon, 2008, IARC Press.
45:37–42, 2005. 204. van der Vlist M, Geijtenbeek BH: Langerin functions as a antiviral
180. Komp DM: The treatment of sinus histiocytosis with massive receptor on Langerhans cells. Immunol Cell Biol 88:410–415,
lymphadenopathy (Rosai-Dorfman disease). Semin Diagn Pathol 2010.
7:83–86, 1990. 205. Vencio EF, Jenkins RB, Schiller JL, et al: Clonal cytogenetic
181. Lee HJ, Ahn BC, Lee SW, et al: The usefulness of F-18 fluoro- abnormalities in Erdheim-Chester disease. Am J Surg Pathol
deoxyglucose positron emission tomography/computed tomogra- 31:319, 2007.
phy in patients with Langerhans cell histiocytosis. Ann Nucl Med 206. Walker PD, Rosai J, Dorfman RF: The osseous manifestations of
26:730–737, 2012. sinus histiocytosis with massive lymphadenopathy. Am J Clin
182. Lewin JR, Das SK, Blumenthal BI, et al: Osseous pseudotumor: Pathol 75:131–139, 1981.
the sole manifestation of sinus histiocytosis with massive lymph- 207. Weiss LM: Histiocytic and dendritic cell proliferations. In
adenopathy. Am J Clin Pathol 84:547–550, 1985. Knowles DM, editor: Neoplastic hematopathology, Baltimore, 1992,
183. Liu L, Perry AM, Cao W, et al: Relationship between Rosai- Williams & Wilkins.
Dorfman disease and IgG4-related disease: study of 32 cases. Am 208. Willman CL, Busque L, Griffith BB, et al: Langerhans’-cell his-
J Clin Pathol 140:395–402, 2013. tiocytosis (histiocytosis X)—a clonal proliferative disease. N Engl
184. McAlister WH, Herman T, Dehner LP: Sinus histiocytosis with J Med 331:154–160, 1994.
massive lymphadenopathy (Rosai-Dorfman disease). Pediatr Radiol 209. Yousem SA, Colby TV, Chen YY, et al: Pulmonary Langerhans’
20:425–432, 1990. cell histiocytosis: molecular analysis of clonality. Am J Surg Pathol
185. Merad M, Ginhoux F, Collin M: Origin, homeostasis and function 25:630–636, 2001.
of Langerhans cells and other langerin-expressing dendritic cells.
Nature Rev 8:935–947, 2008. Myeloid Sarcoma
186. Minkov M: Multisystem Langerhans cell histiocytosis in children: 210. Abeler V, Kjørstad KE, Langholm R, et al: Granulocytic sarcoma
current treatment and future directions. Paediatr Drugs 13:75–86, (chloroma) of the uterine cervix: report of two cases. Int J Gynecol
2011. Pathol 2:88–92, 1983.

ERRNVPHGLFRVRUJ
12  Hematopoietic Tumors 901

211. Alvarez P, Navascues CA, Ordieres C, et al: Granulocytic sarcoma 236. Sun NC, Ellis R: Granulocytic sarcoma of the skin. Arch Dermatol
of the small bowel, greater omentum and peritoneum associated 116:800–802, 1980.
with a CBFβ/MYH11 fusion and inv(16)(p13q22): a case report. 237. Stefanidakis M, Karjalainen K, Jaalouk DE, et al: Role of leukemia
Int Arch Med 4:3, 2011. cell invadosome in extramedullary infiltration. Blood 114:3008–
212. Ansari-Lari MA, Yang CF, Tinawi-Aljundi R, et al: FLT3 3017, 2009.
mutations in myeloid sarcoma. Br J Haematol 126:785–791, 238. Takagi M, Ishikawa G, Kamiyama R: Granulocytic sarcoma of the
2004. jaw. Bull Tokyo Med Dent Univ 30:1–7, 1983.
213. Antic D, Elezovic I, Milic N, et al: Is there a “gold” standard 239. Xavier SG, Fagundes EM, Hassan R, et al: Granulocytic sarcoma
treatment for patients with isolated myeloid sarcoma? Biomed of the small intestine with CBFβ/MYH11 fusion gene: report of
Pharmacother 67:72–77, 2013. an aleukaemic case and review of the literature. Leuk Res 27:1063–
214. Conran MJ, Keohane C, Kearney PJ: Chloroma of the mandible: 1066, 2003.
a problem of diagnosis and management. Acta Paediatr Scand 240. Zhang PJ, Barcos M, Stewart CC, et al: Immunoreactivity of
71:1041–1043, 1982. MIC2 (CD99) in acute myelogenous leukemia and related dis-
215. Deeb G, Baer MR, Gaile DP, et al: Genomic profiling of myeloid eases. Mod Pathol 13:452–458, 2000.
sarcoma by array comparative genomic hybridization. Genes Chro- 241. Zhang XH, Zhang R, Li Y: Granulocytic sarcoma of abdomen in
mosomes Cancer 44:373–383, 2005. acute myeloid leukemia patient with inv(16)t(6;17) abnormal
216. Dong HY, Wilkes S, Yang H: CD71 is selectively and ubiquitously chromosome: case report and review of the literature. Leukem Res
expressed at high levels in erythroid precursors of all maturation 34:958–961, 2010.
stages: a comparative immunochemical study with glycophorin A
and hemoglobin A. Am J Surg Pathol 35:723–732, 2011. Mastocytosis
217. Falini B, Lenze D, Hasserjian R, et al: Cytoplasmic mutated 242. Akin C, Metcalfe DD: The biology of Kit in disease and the
nucleophosmin (NPM) defines the molecular status of a signifi- application of pharmacogenetics. J Allergy Clin Immunol 114:13–
cant fraction of myeloid sarcomas. Leukemia 21:1566–1570, 19, 2004.
2007. 243. Bibi S, Langfield F, Jeanningros S, et al: Molecular defects in
218. Fritz J, Vogel W, Bares R, et al: Radiologic spectrum of extramed- mastocytosis: KIT and beyond KIT. Immunol Allergy Clin N Am
ullary relapse of myelogenous leukemia in adults. AJR Am J Roent- 34:239–262, 2014.
genol 189:209–218, 2007. 244. Brunning RD, McKenna RW, Rosai J, et al: Systemic mastocyto-
219. Guermazi A, Feger C, Rousselot P, et al: Granulocytic sarcoma sis: extracutaneous manifestations. Am J Surg Pathol 7:425–438,
(chloroma): imaging findings in adults and children. AJR Am J 1983.
Roentgenol 178:319–325, 2002. 245. Gleixner KV, Mayerhofer M, Cerny-Reiterer S, et al: KIT-D816-
220. Karlin L, Itti E, Pautas Cm Rachid M, et al: PET-imaging as a independent oncogenic signaling in neoplastic cells in systemic
useful tool for early detection of the relapse site in the manage- mastocytosis: role of Lyn and Btk activation and disruption by
ment of primary myeloid sarcoma. Haematologica 91(2 Supl): dasatinib and bosutinib. Blood 118:1885–1898, 2011.
ECR54, 2006. 246. Guillaume N, Desoutter J, Chandesris O, et al: Bone complica-
221. King A: A case of chloroma. Monthly J Med Soc 17:17, 1853. tions of mastocytosis: a link between clinical and biological char-
222. Licht JD: AML1 and the AML1-ETO fusion protein in the acteristics. Am J Med 126:75, 2013.
pathogenesis of t(8;21) AML. Oncogene 20:5660–5679, 2001. 247. Horton MA, O’Brien HA: Characterization of human mast cells
223. Long JC, Mihm MC: Multiple granulocytic tumors of the skin: in long-term culture. Blood 62:1251–1260, 1983.
report of six cases of myelogenous leukemia with initial manifesta- 248. Kaliner MA: The mast cell: a fascinating riddle. N Engl J Med
tions in the skin. Cancer 39:2004–2016, 1977. 301:498–499, 1979.
224. McCarty KS, Jr, Wortman J, Daly J, et al: Chloroma (granulocytic 249. Kobayasi T, Midtgard K, Asboe-Hansen G: Ultrastructure of
sarcoma) without evidence of leukemia: facilitated light micro- human mast cell granules. J Ultrastr Res 23:153–165, 1968.
scopic diagnosis. Blood 56:104–108, 1980. 250. Lim KH, Tefferi A, Lasho TL, et al: Systemic mastocytosis in 342
225. McKenna M, Arnold C, Catherwood MA, et al: Myeloid sarcoma consecutive adults: survival studies and prognostic factors. Blood
of the small bowel associated with a CBFβ/MYH11 fusion 113:5727–5736, 2009.
and inv(16)(p13q22): a case report. J Clin Pathol 62:757–759, 251. Ludatscher RM, Haim S, Gellei B, et al: A comparative ultrastruc-
2009. tural study of mast cells in mastocytoma and mastocytosis. Der-
226. McLeod AJ, Lewis E, Cox D: Chloroma of the urinary bladder: matol 155:80–89, 1977.
sonographic findings. J Clin Ultrasound 12:434–435, 1984. 252. Mikhail GR, Miller-Milinska A: Mast cell population in human
227. Meis JM, Butler JJ, Osborne BM, et al: Granulocytic sarcoma and skin. J Invest Dermatol 43:245–249, 1964.
nonleukemic patients. Cancer 58:2697–2709, 1986. 253. Nagata H, Worobec AS, Oh CD, et al: Identification of a point
228. Menasce LP, Banerjee SS, Beckett E, et al: Extra-medullary mutation in the catalytic domain of the protooncogene c-kit in
myeloid tumour (granulocytic sarcoma) is often misdiagnosed: a peripheral blood mononuclear cells of patients who have masto-
study of 26 cases. Histopathology 34:391–398, 1999. cytosis with an associated hematologic disorder. PNAS 92:10560–
229. Neiman RS, Barcos M, Berard C, et al: Granulocytic sarcoma: a 10564, 1995.
clinicopathologic study of 61 biopsied cases. Cancer 48:1426–1437, 254. Pardanani A: Systemic mastocytosis in adults: 2013 update on
1981. diagnosis, risk stratification, and management. Am J Hematol
230. Pileri SA, Ascani S, Cox MC, et al: Myeloid sarcoma: clinic- 88:612–624, 2013.
pathologic, phenotypic and cytogenetic analysis of 92 adult 255. Parwaresch MR, Horny H-P, Bodewadt-Radzun S: Mast cell
patients. Leukemia 21:340–350, 2007. disease. In Knowles DM, editor: Neoplastic hematopathology,
231. Reichart PA, von Roemeling RV, Krech R: Mandibular myelo- Baltimore, 1992, Williams & Wilkins.
sarcoma (chloroma): primary oral manifestation of promyelo- 256. Parwaresch MR, Horny H-P, Lennert K: Tissue mast cells in
cytic leukemia. Oral Surg Oral Med Oral Pathol 58:424–427, health and disease. Pathol Res Pract 179:439–461, 1985.
1984. 257. Pinto-Lopez P, Fonseca FA, Silva R, et al: Indolent systemic mas-
232. Sangle NA, Schmidt RL, Patel JL, et al: Optimized immunohis- tocytosis limited to the bone: a case report and review of the lit-
tochemical panel to differentiate myeloid sarcoma from blastic erature. Sao Paulo Med J 131:198–204, 2013.
plasmacytoid dendritic cell neoplasm. Mod Pathol 27:1137–1143, 258. Rafii M, Firooznia H, Golimbu C, et al: Pathologic fracture in
2014. systemic mastocytosis: radiographic spectrum and review of the
233. Schwyzer R, Sherman GG, Cohn RJ, et al: Granulocytic sarcoma literature. Clin Orthop 180:260–267, 1983.
in children with acute myeloblastic leukemia and t(8;21). Med 259. Rosenbaum RC, Frieri M, Metcalfe DD: Patterns of skeletal scin-
Pediatr Oncol 31:144–149, 1998. tigraphy and their relationship to plasma and urinary histamine
234. Seo IN, Hull MT, Pak HY: Granulocytic sarcoma of the cervix as levels in systemic mastocytosis. J Nucl Med 25:859–864, 1984.
a primary manifestation: case without overt leukemic features for 260. Sahger F, Cohen C, Schorr S: Concomitant bone changes in
26 months. Cancer 40:3030–3037, 1977. urticaria pigmentosa. J Invest Dermatol 18:425–432, 1952.
235. Singh T, Jayaram G, Gupta AK: Cytologic diagnosis of myeloid 261. Schwartz LB, Austen KF: Structure and function of the chemical
sarcoma. Am J Ophthalmol 99:496–497, 1985. mediators of mast cells. Prog Allergy 34:271–321, 1984.

ERRNVPHGLFRVRUJ
902 12  Hematopoietic Tumors

262. Sostre S, Handler HL: Bony lesions in systemic mastocytosis: 266. Tefferi A, Pardanani A: Systemic mastocytosis: current concepts
scintigraphic evaluation. Arch Dermatol 113:1245–1247, 1977. and treatment advances. Curr Hematol Rep 3:197–202, 2004.
263. Sotlar K, Cerny-Reiterer S, Petat-Dutter K, et al: Aberrant 267. Valent P, Akin C, Arock M, et al: Definitions, criteria and global
expression of CD30 in neoplastic mast cells in high-grade masto- classification of mast cell disorders with special reference to mast
cytosis. Mod Pathol 24:585–595, 2011. cell activation syndromes: a consensus proposal. Int Arch Allergy
264. Stark E, Van Buskirk FW, Daly JF: Radiologic and pathologic Immunol 157:215–225, 2012.
bone changes associated with urticaria pigmentosa. Arch Pathol 268. Verstovsek S: Advanced systemic mastocytosis: the impact of KIT
62:143–148, 1956. mutations in diagnosis, treatment, and progression. Eur J Haema-
265. Swerdlow SH, Campo E, Harris NL, et al, editors: World Health tol 90:89–98, 2013.
Organization classification of tumours of haematopoietic and lymphoid
tissues, Lyon, 2008, IARC Press.

ERRNVPHGLFRVRUJ
C H A P T E R 1 3 

Vascular Lesions

CHAPTER OUTLINE

BENIGN VASCULAR LESIONS Angiosarcoma


Hemangioma Hemangiopericytoma
Lymphangioma and Lymphangiomatosis Kaposi’s Sarcoma
Angiomatoses REACTIVE VASCULAR LESIONS
Glomus Tumor Bacillary Angiomatosis of Bone
MALIGNANT VASCULAR TUMORS
Epithelioid Hemangioendothelioma
Spindle-cell Hemangioendothelioma

The complex architecture of the human body requires an of bone occur less frequently than hemangiomas. Their
efficient life-support system that transports gases, liquids, behavior ranges from that of indolent, low-grade tumors,
and other nutrients as well as circulating cells that play such as epithelioid hemangioendothelioma, to that of
important roles in body defense systems. These interact- lethal, high-grade angiosarcomas or hemangiopericyto-
ing tasks are carried out by a system that is ubiquitous to mas. The classification of malignant vascular lesions is
all vertebrates and comprises two treelike branched still in a state of flux, and controversy continues about
systems of tubules known as blood and lymphatic vessels the biologic potential of those tumors currently desig-
lined by highly specialized endothelial cells. These two nated as low-grade or borderline endothelial tumors. The
tubular systems are connected through a large lymphatic situation has been further complicated by the human
vessel, which drains the lymph fluid to the blood circula- immunodeficiency virus (HIV) pandemic and its associ-
tion and is referred to as the thoracic duct. The dysfunc- ated vascular lesions in bone, such as bacillary angioma-
tions and malformations of these systems contribute to tosis and Kaposi’s sarcoma.
the pathogenesis of many human diseases and frequently The most significant developments contributing to
give rise to tumors and tumorlike malformations in the our knowledge of vascular lesions stem from molecu-
skin, soft tissue, and viscera. In bone, the nutrient arteries lar biology, which provides new information on the
penetrate the cortex and branch into an abundant network molecular mechanisms controlling vascular growth and
of small arteries and capillaries. The rich capillary differentiation.1,3,14
network in the medullary cavity is drained to efferent In brief, vascular endothelial growth factor-A (VEGF-
venules and veins. The periosteal vessels supply the outer A), which is a part of a large superfamily of growth
portion of the cortex. Communication between the peri- factors, is required for differentiation of endothelial pre-
osteal and medullary vasculature is provided by anasto- cursor cells.8 The sprouting of endothelial cells is regu-
motic vessels within Volkmann’s and haversian canals. A lated by Notch signaling receptors and their Delta-like 4
rich network of periosteal lymphatics is present. The (DLL4) ligand. Precurser lymphatic endothelial cells
presence of medullary lymphatics can be best docu- form a distinctive cluster in mid-gestation embryos on
mented in abnormal conditions of lymph stasis. Despite the dorsal side of the jugular vein. The prospero related
the rich vascularity of bone, skeletal vascular lesions are homeobox-1 (PROX1) transcriptional factor and vascular
rare, and consequently knowledge of their clinical and endothelial growth factor receptor-3 (VEGFR3) are
pathologic features is still limited. responsible for their differentiation into lymphatic endo-
Solitary hemangiomas are the most common vascular thelial cells.5,9,17 Although endothelial cells of blood
lesions of bone. Angiomatoses occur less frequently than vessels and lymphatics are somewhat similar, the lym-
solitary hemangiomas. The clinical behavior and the phatics lack a continuous basement membrane and are
extent of skeletal involvement by angiomatoses provide a not tightly sealed by intercellular junctions, permitting
means of descriptively dividing them into regional versus the free access of interstitial tissue fluid into the vessel.
disseminated and nonaggressive versus aggressive forms. Each component of both vascular and lymphatic systems
Skeletal angiomatoses typically present in two distinct has its own expression signature. For example, the arterial
clinical settings: disseminated skeletal angiomatosis, gene expression signature involves VEGFA, VEGFR2,
sometimes referred to as cystic angiomatosis, and massive neuropilin-1 (NRPI), forkhead box protein C1 or C2
osteolysis (Gorham’s disease). Malignant vascular tumors (FOXC1/2), Notch signaling, and ephrin-B2; the venous
903
ERRNVPHGLFRVRUJ
904 13  Vascular Lesions

expression signature includes the COUP transcription Lectin proteins such as Ulex europaeus, frequently used
factor 2 (COUP-TFII), Notch signaling, and ephrin-B4. in the past, are rarely used as markers of endothelial cells
From the diagnostic point of view, these investigations in modern diagnostics.
have provided us with a new generation of markers iden- Recently, genome sequencing has identified a new
tifying endothelial phenotypes useful in differential diag- class of chromosomal translocation (1;3)(p36;q25) result-
nosis of vascular lesions. The standard vascular markers ing in a WWTR1-CAMTA1 gene fusion that appears to
used in pathologic differential diagnosis of vascular be specific for epithelioid hemangioendothelioma regard-
conditions include von Willebrand factor (vWF), factor less of its site of origin, including those that arise in the
VIII-associated protein, platelet endothelial cell adhesion skeleton.115,129
molecule-1 (CD31), human hematopoietic progenitor
cell antigen (CD34), v-ets avian erythroblastosis virus
E26 oncogene homolog (ERG), human herpes virus 8 BENIGN VASCULAR LESIONS
(HHV-8), latency-associated nuclear antigen (LANA1),
podoplanin (D2-40), and PROX1. Hemangioma
Factor VIII-associated protein is historically the first Definition
immunohistochemical marker of endothelial cells. vWF
is least sensitive of the vascular markers and is present in Hemangioma is a benign solitary tumor composed of
only 50% to 70% of vascular tumors.2 newly formed vessels of capillary or cavernous types.
The CD31 marker, an adhesion molecule for platelets,
is one of the newer markers of endothelial cell differen- Incidence and Location
tiation and is considered to be among the most specific
and sensitive markers in the differential diagnosis of vas- The peak age incidence and most common sites of skel-
cular tumors.11,15 It is believed to be expressed in more etal involvement are shown in Figure 13-1. Hemangio-
than 90% of vascular tumors. It can occasionally be mas of bone are rare lesions, accounting for less than 1%
expressed in carcinomas but the expression is very weak of all primary tumors of bone.15,19,28,34,36 The identification
and focal. It is consistently expressed in high levels in of skeletal hemangiomas of vertebral bodies in more than
tumor macrophages. 10% of autopsies, however, indicates that they occur
The transmembrane glycoprotein (CD34) is not more frequently. A significant number of solitary skeletal
entirely specific for endothelial cells; it is also expressed on hemangiomas are asymptomatic and are never diagnosed
hematopoietic cells, the interstitial cells of Cajal, dermal during life. It should be anticipated that increasing the
dendritic cells, and nerve sheaths.18 It is also present use of magnetic resonance imaging, particularly for
in several soft tissue tumors such as dermatofibrosar- symptoms of back pain, should increase the frequency of
coma protuberans, solitary fibrous tumor, gastrointestinal incidentally diagnosed hemangiomas of bone. Heman-
stromal tumor, and epithelioid sarcoma among others. giomas have a wide age distribution, ranging from the
The ETS family of transcription factors play an first to eighth decades of life, with nearly 70% of cases
important role in the pathogenesis of several bone and diagnosed in patients between ages 30 and 60 years.
soft tissue tumors as fusion partners of chimeric genes.10 Occasionally, hemangiomas become clinically evident
Ewing’s sarcoma is a prototypic tumor exhibiting a during the first decade of life. There is no clear sex pre-
translocation resulting in the EWSR1-FLI1 fusion gene dilection, but in some series, female patients are slightly
present in nearly 80% of these tumors. The ERG gene is more frequently affected than male patients.
a rare partner of this fusion, seen in less than 10% of Hemangiomas frequently occur in the craniofacial
Ewing’s sarcomas. It has been recently shown that it is a bones, predominantly in the calvarium, and in some
major partner in the TMPRSS2-ERG fusion gene, present series nearly 50% of the lesions occur at this site. The
in approximately 50% of prostatic adenocarcinomas. spine is involved in approximately 20% of cases. On the
Both FLI1 and ERG are expressed by endothelial cells other hand, if autopsy data on asymptomatic lesions are
and are positive in nearly 95% of vascular tumors of all included, the vertebral bodies represent the most fre-
types. ERG is currently considered one of the best quent site of involvement by hemangioma. The major
markers of endothelial differentiation. long tubular bones are the bones of the appendicular
The identification of the specific herpes virus HHV-8 skeleton most frequently involved. Hemangiomas can be
and its associated antigens as markers of Kaposi’s sarcoma multifocal. The small multifocal lesions are most fre-
has facilitated their use in the differential diagnosis of quently identified in the vertebral column involving adja-
vascular endothelial proliferations.4,6,13,16 LANA1 is par- cent vertebral bodies.
ticularly useful because it is present as a nuclear protein
in all forms of Kaposi’s sarcoma.4,16
Radiographic Imaging
Podoplanin (D2-40) is a transmembrane glycoprotein
expressed by lymphatic endothelial cells. It is also Radiographically, hemangiomas present as lucent, well-
expressed in glomerular podocytes, choroid plexus epi- demarcated defects.28,59 In flat bones, they markedly
thelium, type 1 alveolar cells, osteoblasts, and mesothelial expand the bone contour and produce rarefaction with
cells. Its expression is regulated by PROX1, and both radially oriented striations (Fig. 13-2). The vascular
proteins are considered to be markers of lymphatic endo- nature of the lesion is often suggested by its bubbly or
thelial cells, but as with many other markers, they are not honeycomb, trabeculated appearance (Figs. 13-3 to 13-5).
fully specific for lymphatic endothelial cells.7,12 The overlying cortex is expanded and thinned, but

ERRNVPHGLFRVRUJ
13  Vascular Lesions 905

30 60
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-1  ■  Hemangioma. Peak age incidence and most frequent sites of skeletal involvement are indicated by solid black arrows.

A B
FIGURE 13-2  ■  Hemangioma of bone: radiographic and gross features. A, Hemangioma of frontal bone with prominent striations of
bone trabeculae traversing lesion. B, Clinical photograph of lesion shown in A.

ERRNVPHGLFRVRUJ
906 13  Vascular Lesions

A B C

D E

FIGURE 13-3  ■  Hemangiomas of rib and nasal bones: radiographic, gross, and microscopic features. A, Expansile lesion of vertebral
end of rib has honeycomb appearance and spiculated reactive bone of periosteal origin. Lesion had been asymptomatic and was
diagnosed incidentally. B, Specimen radiograph shows radiating spicules of periosteal new bone and coarse trabeculation. C, Gross
photograph of resected rib shows radiating spicules of periosteal new bone and prominent trabeculation. D, Lateral plain radiograph
of face shows honeycomb appearance of nasal bone produced by cavernous hemangioma. E, Low power photomicrograph of nasal
bone hemangioma shown in D composed of cavernous vascular channels. Note prominent trabeculation throughout the lesion.
(E, ×50) (E, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
13  Vascular Lesions 907

A B
FIGURE 13-4  ■  Hemangioma of tibia: radiographic features. A and B, Anteroposterior and lateral radiographs of knee of young woman
who sustained pathologic fracture through large, previously asymptomatic honeycomb lesion occupying proximal half of tibia.
Radiating spicules of periosteal reactive bone are prominent, especially on lateral aspect, and suggest preoperative diagnosis of
primary malignant bone tumor.

complete cortical disruption and invasion into soft tissue subperiosteal lesion (Fig. 13-8). Magnetic resonance
are not present. The characteristic sunburst appearance imaging of hemangiomas generally reveals a low signal
of hemangioma is seen in skull lesions (Fig. 13-2) and is on T1-weighted images and a high signal on T2-weighted
produced by fine spicules of reactive new bone in the images. The latter is explained by the fluid content of
periosteum radiating outward from the center of the tumor vessels. In vertebral body hemangiomas, the loss
lesion. The sunburst type of periosteal new bone forma- of hematopoietic cells in the interstices of hemangiomas
tion should not be confused with that seen in association and apparent increase in fat can produce a high signal in
with osteosarcoma of long bone. Smaller lesions may T1-weighted images.
present as intracortical rarefactions with or without a
honeycomb appearance and adjacent sclerosis (Fig. 13-6). Gross Findings
They can mimic benign osteoblastic lesions (osteoid
osteoma, osteoblastoma) radiographically. Some heman- Hemangioma presents as a brown-red or dark red, well-
giomas are purely lytic lesions with sharply demarcated demarcated, medullary lesion. Occasionally, it can be
borders and should be differentiated from cystic lesions intracortical or subperiosteal (Fig. 13-8). It may have a
(Fig. 13-7). Rarely a hemangioma may present as a honeycomb appearance with sclerotic bone trabeculae

ERRNVPHGLFRVRUJ
908 13  Vascular Lesions

A B
FIGURE 13-5  ■  Hemangioma of tibia: radiographic and microscopic features. A, Anteroposterior radiograph shows ex­pansible tra-
beculated lucent lesion of distal tibial shaft. B, Photomicrograph of biopsy material of same lesion shows engorged capillary-sized
vessels (×50; hematoxylin-eosin).

interspersed among hemorrhagic cavities. Some heman- tissue is composed of loose connective tissue that may
giomas provoke an abundance of reactive bone and exhibit myxoid change. The cancellous bone in the
appear more sclerotic (Fig. 13-6). Some lesions are com- affected area can be completely resorbed. In such cases,
posed exclusively of vascular tissue and contain no bone. the lesion is composed of vessels and stromal tissue, but
Perpendicular periosteal new bone formation and the more often, some residual trabeculae of cancellous bone
radiating spicules can be seen on the surface of some of are present. These trabeculae are usually thickened. Some
the resected hemangiomas (Fig. 13-3). hemangiomas may cause pronounced sclerosis of the
intralesional and adjacent bone. The vascular channels of
hemangioma are complete, are separate, and do not show
Microscopic Findings
an anastomosing pattern. In vertebral body hemangio-
Hemangiomas are composed of a conglomerate of thin- mas, the blood-forming elements may be lost completely
walled blood vessels.28,59 The vessels can have dilated in the interstices, leading to an unmasking of adipose
open channels (cavernous hemangioma) or, less fre- tissue. This apparent increase in fat in the interstitial
quently, may be composed of capillary-sized vessels (cap- tissue of vertebral hemangiomas can be quite prominent.
illary hemangioma) (Figs. 13-9 to 13-12). A majority of Hemangiomas have a characteristic microscopic
bone hemangiomas are of cavernous or mixed types (cav- appearance and in the majority of cases are easy to
ernous and capillary). Predominantly or exclusively capil- recognize. However, secondary changes may alter this
lary hemangiomas are exceedingly rare in bone. The size classic pattern and make the diagnosis difficult. Hem-
of cavernous vessels may vary, but occasionally hemangi- angiomas of any type, but especially those with large
oma can be composed of vessels that are relatively uniform cavernous vessels, occasionally develop thromboses with
in size (Fig. 13-9). The vascular channels are lined by a calcifications. This in turn can promote the develop-
single layer of flat endothelial cells (Figs. 13-7 and 13-9). ment of papillary endothelial hyperplasia (Fig. 13-11).
Occasionally, these cells can have plump cytoplasm and The presence of this change can alter the pattern of
even epithelioid features (Fig. 13-10). The intercellular vascular channels and lead to the misdiagnosis of a

ERRNVPHGLFRVRUJ
13  Vascular Lesions 909

A C
FIGURE 13-6  ■  Hemangioma of tibia: radiographic, gross, and microscopic features. A, Intracortical lucency with trabeculation in
tibia of patient who had pain for 8 months. Initial impression was that of osteoid osteoma. B, Specimen slices from intracortical
lesion shown in A. Note dark, spongy tissue, which is well circumscribed from surrounding thick cortex. C, Photomicrograph
of resected intracortical hemangioma demonstrates engorged vessels and trabeculated bony architecture of the lesion. (C, ×50)
(C, hematoxylin-eosin.)

malignant vascular tumor.20,22,23,25 Papillary endothelial central fibrin or hyaline cores typically are seen. Micro-
hyperplasia was described by Masson in 1923 under the scopic examination at low power is extremely important
term hemangioendotheliome vegetant intravasculaire.45 It can because this magnification allows visualization of the
occur in any part of the body involving blood vessels papillary structures within preexisting vascular spaces and
or soft tissue hemangioma or can be superimposed on the pattern of progression toward the center of the vessel.
a preexisting vascular tumor.22,23,25,29,35,40,49,52,56 Most fre- Similar orientation can be seen if the lesion develops in a
quently, it presents as a small mass in the skin and super- focus of hemorrhage. Under high power magnification,
ficial soft tissue.20,33,41,49 It is less common in vascular cross-sectioned papillary structures appear to be floating
lesions of bone. Papillary endothelial hyperplasia may in the vascular lumina. The spaces between them can
be so abundant that it tends to obscure the underlying form a network that simulates anastomosing channels,
hemangioma, or it may be found only in focal areas. which together with the presence of plump endothelial
Papillary proliferations of plump endothelial cells with cells, can be misinterpreted as the vascular pattern of

ERRNVPHGLFRVRUJ
910 13  Vascular Lesions

B
FIGURE 13-7  ■  Calcaneus hemangioma: radiographic and microscopic features. A, Lateral radiograph shows well-circumscribed,
lucent lesion occupying anterior half of calcaneus. Clinical diagnosis of solitary bone cyst was suggested until biopsy was performed.
B, Photomicrograph of curetted material from calcaneal lesion shows hemangioma characterized by vascular spaces lined by flat
endothelial cells (B, ×50) (B, hematoxylin-eosin.)

angiosarcoma. Fibrin with calcified cores may be present. similarity to corresponding lesions occurring in soft
Calcified spherical structures sometimes mimic psam- tissue has been recognized.30,37,43,46 Its biologic potential
moma body–like structures or cementoid bodies. Various is still not entirely clear, as indicated by a recently reported
phases of organization in the thrombotic material also case with regional lymph node involvement.32 Moreover,
are present and are associated with an inflammatory cell a study of 17 vascular tumors of bone30 suggested that
infiltrate. In rare instances, aneurysmal bone cyst can be epithelioid hemangioma could not be separated from epi-
superimposed on a preexisting hemangioma. thelioid hemangioendothelioma, and that rather than
representing a distinct benign entity, epithelioid heman-
gioma is part of the spectrum of hemangioendothelioma
Epithelioid Hemangioma
of bone. Clearly, as of this writing, the place of epithelioid
This variant of hemangioma affecting bone has been hemangioma in the nosology of vascular tumors, espe-
more frequently diagnosed in recent years, and its cially those involving the skeleton, remains controversial.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 911

A C
FIGURE 13-8  ■  Subperiosteal hemangioma: radiographic and gross features. A, Anteroposterior radiograph shows lytic subperiosteal
lesion of tibial shaft. B, Computed tomography shows subperiosteal lesion with cortical erosion delineated by elevated periosteum.
C, Resected specimen showing perpendicular sections through intracortical hemangioma.

The recent identification of the t(1;3)(p36;q25) and not present.21,26,34,46,47 The presence of prominent spindle-
WWTR1-CAMTA1 fusion in epithelioid hemangioendo- cell elements and hemorrhage rarely complicate the his-
thelioma should help to resolve this controversy but the tologic diagnosis.37,53
experience with bone lesions is limited.115,129 Epithelioid hemangioma as a distinctive neoplasm
An epithelioid change of endothelial cells lining the was originally described as angiolymphoid hyperpla-
vessels of hemangioma (epithelioid hemangioma) can sia with eosinophilia and subsequently as inflammatory
cause this benign vascular lesion to be misdiagnosed as angiomatoid nodule, pseudopyogenic granuloma, or his-
an epithelioid hemangioendothelioma. Epithelioid hem- tiocytoid hemangioma.47,50,58 It is somewhat similar to
angioma is basically a benign hemangioma with plump but pathogenetically distinct from the lesion designated
and somewhat more active endothelial cells (Fig. 13-10). as Kimura’s disease.38,39,42 Some epithelioid hemangiomas
Other features, such as solid or cordlike proliferations of are rather reactive and are seen in association with an
endothelial cells and the anastomosing vascular pattern injured vessel.31 Hemangiomas with epithelioid change
characteristic of epithelioid hemangioendothelioma, are in bone have no apparent relationship to angiolymphoid

ERRNVPHGLFRVRUJ
912 13  Vascular Lesions

A B

C D
FIGURE 13-9  ■  Hemangioma of bone: microscopic features. A and B, Hemangioma engorged vessels occupying intertrabecular
marrow spaces. Thin-walled blood vessels are lined by flat endothelial cells. C and D, Hemangioma composed of capillary vessels
in fibrous stroma. (A-D, ×50) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 913

A B

C D
FIGURE 13-10  ■  Epithelioid hemangioma of bone: microscopic features. A and B, Angiomatous lesion composed of capillary-sized
vessels with thicker wall lined by plump endothelial cells that project into lumen in tombstone fashion. C and D, Larger vessels lined
by plump endothelial cells with regular vesicular nuclei. (A-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
914 13  Vascular Lesions

A B

FIGURE 13-11  ■  Papillary endothelial hyperplasia (Masson’s type) in cavernous hemangioma. A, Plain radiograph of hemangioma of
distal humerus in adult. B, Papillary endothelial proliferation representing organized thrombotic vegetations in benign cavernous
hemangioma of humerus shown in A. C, Higher power photomicrograph shows cavernous hemangioma with papillary (vegetant)
endothelial proliferations. (B, ×60; C, ×150) (B and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 915

B
FIGURE 13-12  ■  Hemangioma of sphenoid bone: radiographic features. A and B, Axial and coronal computed tomograms show
expansile tumor at base of skull involving sphenoid bone. C, Arteriogram shows tumor blush.

hyperplasia with eosinophilia or Kimura’s disease. There- cords of endothelial cells and an anastomosing pattern of
fore the term epithelioid hemangioma in bone should be irregular vascular channels—are not present in a benign
used in a descriptive sense only. The radiologic features hemangioma with epithelioid change.
of bone hemangiomas and epithelioid hemangiomas
often overlap, and their radiographic differentiation is, Treatment and Behavior
in such instances, not possible.43,57
Asymptomatic small hemangiomas require no treatment;
Differential Diagnosis some may even undergo spontaneous regression. Symp-
tomatic lesions or those that are large and may cause
The flat endothelial cells lining the capillary or cavernous pathologic fracture or vertebral collapse require treat-
vascular channels of hemangiomas can be easily distin- ment. Curettage and bone grafting usually is sufficient.
guished from the plump, enlarged endothelial cells Symptomatic vertebral lesions are treated by decompres-
of hemangioendotheliomas. Individual lesions of skeletal sion laminectomy; radiotherapy may be added to curet-
angiomatosis are histologically identical to those of solitary tage. Surgically inaccessible hemangiomas may be treated
bone hemangiomas; the distinction is based on the by radiotherapy alone. Transarterial embolization, verte-
mult-centric distribution of angiomatosis. Epithelioid brectomy, and vertebroplasty have become widely used
hemangiomas can be confused with a low-grade epithelioid in the management of symptomatic vertebral hemangio-
hemangioendothelioma. However, other diagnostic features mas.19 Hemangiomas of the cranial vault and those of
of epithelioid hemangioendothelioma—solid nest or the facial bones are usually treated by en bloc excision.

ERRNVPHGLFRVRUJ
916 13  Vascular Lesions

Hemangiomas in Different Anatomic Sites apparent increase of fat. Magnetic resonance imaging
occasionally reveals evidence of associated involvement
Hemangiomas involving different parts of the skeleton of soft tissue. T2-weighted images show an area of high
present unique radiographic and pathologic characteris- signal. An interesting relationship has been noted between
tics. In addition, the lesions in different skeletal sites pregnancy and the development of clinical signs of spinal
present distinct diagnostic problems and can present with cord compression by vertebral hemangiomas.24 The
different clinical features. For these reasons, separate mechanism of the phenomenon is unclear and can be
descriptions of hemangiomas in various anatomic sites related to increased venous pressure in the paravertebral
are provided. plexus or can be the result of an increased growth rate of
hemangiomas during pregnancy. Symptomatic heman-
Hemangiomas of Craniofacial Bones.  The craniofa- giomas of the vertebral column are treated by surgery
cial bones are frequent sites of involvement by heman- (spinal cord decompression), radiotherapy, or both.
giomas,27,36,44,48,51,54,55 which typically occur in the frontal
area of the cranial vault (Fig. 13-2). Hemangiomas at Hemangiomas of Other Skeletal Sites.  Hemangiomas
this site present with a sunburst effect of radiating bone that involve bones other than the vertebrae or skull are
spicules that extend from the center of a lucent defect.27,28 very rare and usually occur in the bones of the lower
The shape of the defect is circular in an en face view and extremity and ribs; however, any bone can be involved
ellipsoid in a lateral view. Occasionally the typical honey- (Figs. 13-3 to 13-8). Hemangiomas that involve long
comb appearance can be seen in craniofacial lesions (Fig. bones can reach considerable size before they become
13-3, D). Hemangiomas of the skull produce a convex symptomatic (Figs. 13-4 and 13-5). Consequently, lesions
surface bulge of the cortex externally. Expansion of the of the long tubular bones are usually larger than those of
inner concave surface with signs of increased intracra- the skull and vertebral bodies. They involve the metaphy-
nial pressure is rare but was observed in two cases in seal and diaphyseal parts of long tubular bones and present
our series. Hemangiomas very rarely involve the base radiographically as lytic areas with bone trabeculation
of the skull (Fig. 13-12). Microscopically, hemangiomas creating a soap-bubble or honeycomb appearance as a
of the craniofacial bones, especially those of the skull, result of the expansive proliferation of engorged vessels
are cavernous and show more abundant interstitial con- and thickened, remodeled bone trabeculae. Expansion of
nective tissue than vertebral hemangiomas. The most the bone contour with thinning of the cortex is frequently
striking feature is the presence of prominent trabecular seen. Spiculated periosteal reaction may be present. Long
bone between the engorged vascular elements. Rarely, bone hemangiomas are rarely diagnosed correctly preop-
aneurysmal bone cyst can be superimposed on a preexist- eratively, and they are often misdiagnosed radiologically
ing calvarial hemangioma. as malignant lesions. Hemangiomas that involve the flat
bones, such as the pelvis and scapula, are rare and have
Hemangiomas of Vertebral Column.  The involve- the same sunburst appearance as the calvarial lesions. In
ment of vertebral bodies, especially of the lower thoracic extremely rare cases, hemangioma can present radio-
and lumbar regions, is typical for hemangiomas (Figs. graphically as an intracortical lesion and mimic other
13-13 and 13-14).19,24,28 There is a disparity between intracortical lesions that occur more frequently, such as
the frequency of finding vertebral hemangiomas during osteoid osteoma and abscess (Fig. 13-6). Hemangioma
autopsies and their relatively rare clinical significance. can also present as a subperiosteal lesion (Fig. 13-8).
Many vertebral hemangiomas are asymptomatic. They
either are too small or do not disrupt the trabecular archi-
Personal Comments
tecture sufficiently to be visualized on plain radiographs.
Symptomatic hemangiomas of the vertebral column are The diagnosis of craniofacial and vertebral hemangiomas
usually diagnosed in patients older than age 40 years. on the basis of clinical and radiographic features usually
Vertebral hemangiomas that produce clinical symp- presents little difficulty, but large hemangiomas of long
toms are usually associated with back pain and muscle bones may raise the suspicion of a malignant bone tumor
spasm followed by an acute episode of severe pain and because of the prominent perpendicular pattern of reac-
neurologic symptoms of spinal cord compression. The tive new bone in the overlying periosteum. Intracortical
lesion is usually associated with collapse of the vertebral hemangiomas in long bones have been mistaken for
body or bulging cortex that compresses the spinal cord osteoid osteoma and even for adamantinoma before
or nerve roots. biopsy because of their features on radiographs. Benign
Vertebral hemangiomas appear on radiographs as areas hemangiomas of bone with secondary changes (epitheli-
of rarefaction traversed by the thickened vertical bone oid or superimposed papillary endothelial hyperplasia)
trabeculae, which give them a striated or corduroy-like are frequently overdiagnosed as malignant vascular neo-
appearance (Fig. 13-13). The majority of lesions in this plasms. Vertebral hemangiomas can present diagnostic
location measure less than 1 cm. Large lesions can occupy problems when both the centrum and posterior elements
the entire vertebral body and expand its contour. Findings are involved and there is associated soft tissue involve-
on axial computed tomograms are very characteristic ment, with extradural impingement.
because of the polka-dot appearance of thickened vertical Although borderline vascular tumors involving bone
trabeculae seen on end (Fig. 13-13). T1-weighted mag- are relatively rare, few clinicopathologic entities have
netic resonance imaging clearly demonstrates the lesion engendered more controversy in the literature than epi-
as a well-demarcated area of high signal as a result of the thelioid hemangioendothelioma of bone and epithelioid

ERRNVPHGLFRVRUJ
13  Vascular Lesions 917

A B

C D
FIGURE 13-13  ■  Hemangioma of vertebra: radiographic and microscopic features. A, Lateral view of computed tomogram (CT) of
upper lumbar spine shows corduroy-like appearance of vertebral body produced by thickening of vertical bone trabeculae traversing
hemangioma. B, Anteroposterior radiograph of the vertebral body shown in A showing coarse trabeculation of vertebral body caused
by hemangioma. C, Axial CT of thoracic vertebra with hemangioma of vertebral body. Cross sections of thickened vertebral (weight-
bearing) trabeculae produce polka-dot appearance typical of this lesion. D, Biopsy material of the lesion shown in A-C documenting
enlarged vascular channels filled with blood and reactive bone trabeculae. (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
918 13  Vascular Lesions

A B

FIGURE 13-14  ■  Hemangioma of vertebra: radiographic, gross, and microscopic features. A, T2-weighted magnetic resonance image
showing a high signal intensity lesion. B, Gross photograph of vertebral hemangioma identified as an incidental finding at autopsy.
Note well-delineated hemorrhagic lesion involving the vertebral body. C, Low power photomicrograph of hemangioma shown in
B composed of well-developed cavernous vessels. (C, ×50) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 919

hemangioma. As stated before, the identification of the


TABLE 13-1  Skeletal Angiomatoses
specific translocation in epithelioid hemangioendotheli-
oma should help to resolve this long-lasting controversy Type of Angiomatosis Anatomic Distribution
as preliminary data indicates that epithelioid hemangio-
Nonaggressive
mas are negative for this abnormality. However, valida-
Regional Involvement of one anatomic
tion of this concept in a significant number of bone cases region (one or several bones)
is not available in the literature at the time of this writing. Extraskeletal angiomatosis may
be present
Disseminated (cystic Disseminated multifocal
Lymphangioma and Lymphangiomatosis angiomatosis) involvement predominantly
affecting trunk bones
Lymphangioma represents a benign lesion composed of Extraskeletal angiomatosis (soft
newly formed lymph vessels, in the form of dilated cystic tissue or visceral) may be
spaces. They are usually diffuse or multiple and most present
likely represent hamartomatous malformations rather Aggressive
than true neoplasms.63,65,68,70 Solitary lymphangiomas Massive osteolysis Regional involvement
occur less frequently, and only single cases have been (Gorham’s disease) predominantly affecting trunk
reported in the literature.60,61,66,69 Dilated intraosseous bones
lymphatics can also present in association with lymph Aggressive clinical behavior with
stasis. Multiple lymphangiomas of the ribs can be associ- progressive bone destruction
ated with chylothorax or chylopericardium.62,63 Lymph-
angiomatosis of bone can be associated with soft tissue
(extraskeletal) lymphangioma.67
Microscopically, lymphangioma or lymphangiomato- (aggressive versus nonaggressive) and patterns of skeletal
sis consists of dilated, thin-walled vessels filled with pro- involvement (regional versus disseminated) (Table 13-1).
teinaceous fluid that has no blood elements (Fig. 13-15). Regional angiomatosis involves one or several bones of
It may be difficult to distinguish between blood and other the same anatomic region (e.g., spine, shoulder, knee)
lymphatic spaces when the contents have been lost during (Figs. 13-13 to 13-18). Disseminated generalized angio-
tissue processing. Often a mixture of lymphatics and matosis, also referred to as cystic angiomatosis, is a general-
blood vessels can be present in one lesion. Immunohis- ized multifocal disorder that predominantly involves the
tochemical stains may help to identify the lymphatic trunk bones. Aggressive angiomatosis, or massive oste-
nature of vessels in both lymphangioma and lymphangi- olysis, is a peculiar form of angiomatosis that progres-
omatosis. The putative markers of lymphatic endothelial sively destroys (lyses) the affected bone or bones.
differentiation are: VEGFR-3, podoplanin (D2-40), and
PROX1. However, none of these markers is entire spe-
Cystic Angiomatosis
cific for lymphatic endothelial cells and are expressed to
some degree in nonlymphatic vascular tumors as well as The term cystic angiomatosis is applied to extremely rare
in tumors of nonvascular origin. Lymphangiomatosis conditions of disseminated multifocal hemangiomas in
predominantly occurs in children and rarely becomes the skeleton.71,78,81–83,85 The bone involvement may be
manifest after age 20 years. Diagnosis at birth is uncom- accompanied by soft tissue and visceral hemangiomas.
mon; a latent period is required for lesions to achieve The sites of extraskeletal involvement include soft tissue,
sufficient size to become symptomatic. The lesion can lung, liver, and particularly spleen.78,84,86,88 The disease is
also occasionally present as a pathologic fracture, espe- probably congenital and sometimes is clinically evident
cially in the long bones. during the first decade of life. Most cases are recognized
On radiographs lymphangiomatosis can show pro- in the first three decades of life. The condition can be
nounced expansion of large affected areas of the entire asymptomatic and is often detected incidentally in radio-
bone with sunburst periosteal reaction (Figs. 13-16 and graphs obtained for other reasons. The typical age of
13-17). The lesions seem to be composed of multiple diagnosis and most frequent skeletal sites of involvement
coalescent lytic areas separated by thin sclerotic zones. are presented in Figure 13-19.
The use of lymphangiography helps establish the nature The disorder preferentially affects the trunk and occa-
of the lesion before biopsy. The overall prognosis is good, sionally cranial bones (Figs. 13-20 to 13-23). Radiolucent
unless there is significant visceral involvement.64 The lesions with a soap-bubble or honeycomb appearance are
lesions eventually stabilize and sclerose, but the profound present in the skull, spine, ribs, and pelvis. The disease
bone deformity, if present, persists. Some cases of lymph- tends to spare the acral appendicular skeleton. As men-
angiomatosis of bone can pursue an aggressive clinical tioned, the lesions are typically radiolucent but rarely
course with progressive bone loss (massive osteolysis) and sclerotic. In rare instances, the disorder is manifested on
may have radiographic features of disseminated general- radiographs as disseminated osteoblastic lesions that
ized disease (cystic angiomatosis). mimic osteoblastic metastases.81,89
Microscopically, the lesions consist of hemangiomas
that are predominantly cavernous (Fig. 13-22). Usually,
Angiomatoses an admixture of dilated lymphatics is also present (Fig.
Classification of skeletal angiomatoses into distinct clini- 13-24). In fact, some of the foci may be composed entirely
copathologic entities is based on their clinical behavior of lymphatics. Foci composed of small capillary vessels

ERRNVPHGLFRVRUJ
920 13  Vascular Lesions

A B

C D
FIGURE 13-15  ■  Lymphangiomatosis: microscopic features. A and B, Lymphangiomatosis of the rib showing cortical bone eroded by
medullary lymphangiomatous process composed of dilated, endothelial-lined channels. C and D, Dilated lymphatic channels of
varying sizes forming a spongy architecture within the medullary bone. (A and B, ×25; C and D, ×100.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 921

A B

C D
FIGURE 13-16  ■  Lymphangioma of fibula: radiographic, gross, and microscopic features. A, Radiograph of fibula shows area of corti-
cal expansion with coarse trabeculation. B, Gross photograph of bivalved resection segment of fibular shaft shows localized area
of honeycomb appearance located eccentrically in cortex. C and D, Low and intermediate power photomicrographs of lymphangioma
of bone show dilated, endothelial-lined channels with delicate walls. Lymphocytic infiltrate is seen focally in septa. (C, ×25; D, ×50.)
(C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
922 13  Vascular Lesions

C B
FIGURE 13-17  ■  Lmphangiomatosis of bone: radiographic features. A, Chest radiograph of a 7-year-old boy with lymphangiomatosis
of multiple right ribs and right-sided chylous effusion. B, Coarse trabeculation of multiple ribs on right side was produced by lymph-
angiomatosis. C, Computed tomogram of chest shows chylous effusion and rarefaction of posterior portion of rib.

can also be found. Some foci can be accompanied by Massive Osteolysis


prominent new reactive bone trabeculae that occasionally
can dominate the lesion (Fig. 13-24). In the sclerosing Massive osteolysis, also referred to as disappearing or
variant of skeletal angiomatosis, individual vessels can be phantom bone disease, was described in 1954 by Gorham
surrounded by thick sclerotic bone (Fig. 13-25). et al.76 One year later, Gorham and Stout77 established
On radiographs, lytic lesions may be seen to contain the fact that a peculiar clinically aggressive form of angio-
some residual lamellar bone and foci of reactive woven matosis was responsible for this syndrome. The peak age
bone. Blastic lesions contain prominent, thickened bone incidence and most frequent sites of skeletal involvement
trabeculae of mature lamellar bone with areas of woven are shown in Figure 13-26. The disease usually affects
bone rimmed by osteoblasts. Features of osteoclastic children or young adults.72–75,78–80 It is sporadic, without
resorption can be present in lytic and blastic lesions. It evidence of hereditary transmission, and has no sex pre-
has been noted that the sclerotic lesions occur most often dilection. Usually, it involves the shoulder and hip areas
in older patients. Evaluation of serial radiographs in this and starts in the trunk bones, and it may separately
disorder has shown that increasing osteosclerosis can be involve several bones in the same region. In the appen-
an age-related phenomenon in angiomatous foci that are dicular skeleton, the proximal parts of the extremity
originally lytic. It may be that the lesions spontaneously bones—the proximal humerus and femur—are typically
heal or regress and that this process is accompanied by involved.
sclerosis. The skeletal lesions in this disorder are usually Microscopically, the vascular changes in Gorham’s
stable or may even regress. Fatal complications (i.e., disease represent hemangiomas, lymphangiomas, or a
internal bleeding) are caused by visceral lesions. Rare combination. They are indistinguishable from ordinary,
cases of angiosarcoma arising from skeletal angiomatosis nonaggressive angiomatoses and represent a complex
have been described.90 Text continued on p. 931

ERRNVPHGLFRVRUJ
13  Vascular Lesions 923

A B

C D E

FIGURE 13-18  ■  Skeletal angiomatosis (regional): radiographic features. A, Angiomatosis of spine involving four lumbar vertebral
bodies. Note radiolucent lesions with longitudinal, parallel trabeculations. B, Multiple lucent areas with coarse trabeculation involv-
ing patella, proximal tibial metaphysis and epiphysis, and proximal fibular shaft in teenaged patient. C, Lucent areas with peripheral
sclerosis involving humerus. D, Lytic lesion with peripheral sclerotic margins involving the femur. Note pronounced bowing defor-
mity. E, Multilocular well-demarcated lucent lesions involving proximal humerus.

ERRNVPHGLFRVRUJ
924 13  Vascular Lesions

Predominantly
trunk bones
Age at diagnosis

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-19  ■  Cystic angiomatosis. Most frequent age at diagnosis and frequent sites of skeletal involvement.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 925

B
FIGURE 13-20  ■  Cystic angiomatosis of bone: radiographic features. A, Computed tomogram of pelvis and sacrum of 60-year-old
man with sclerosing variant of cystic angiomatosis. B, Radionuclide bone scan shows increased uptake in portions of pelvis showing
sclerotic lesions in A and in left second and fifth ribs.

ERRNVPHGLFRVRUJ
926 13  Vascular Lesions

FIGURE 13-21  ■  Cystic angiomatosis of bone (sclerotic variant): radiographic features. Lateral radiograph of lumbar spine shows
sclerotic lesions in vertebral bodies.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 927

B C

FIGURE 13-22  ■  Cystic angiomatosis of bone (sclerotic variant): radiologic and microscopic features. A, Sclerotic variant of cystic
angiomatosis involving both left and right pelvic bones. B, Sclerotic lesions mimicking osteoblastic metastases adjacent to sacroiliac
joint and iliac crest. C, Low power photomicrograph shows vascular lesion overgrown by well-developed thickened trabeculae of
bone (C, ×25) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
928 13  Vascular Lesions

FIGURE 13-23  ■  Cystic angiomatosis of bone (sclerotic variant): radiologic features. Sclerotic variant of angiomatosis involving both
left and right pelvic bones. Note multifocal sclerotic lesions mimicking osteoblastic metastasis adjacent to sacroiliac joints.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 929

B
FIGURE 13-24  ■  Skeletal angiomatosis: microscopic features. A, Low power photomicrograph shows dilated, thin-walled vessels in
marrow spaces of cancellous bone with delicate trabeculae of reactive woven bone surrounding angiomatosis elements. B, Medium
power photomicrograph shows thin-walled vascular channels in marrow spaces bordered by thickened mature bone trabeculae
(A and B, ×50; hematoxylin-eosin).

ERRNVPHGLFRVRUJ
930 13  Vascular Lesions

B
FIGURE 13-25  ■  Cystic angiomatosis of bone (sclerosing variant): microscopic features. A, Low power photomicrograph of sclerotic
focus in angiomatosis. Note vascular channels in marrow spaces surrounded by thick trabeculae of lamellar bone resembling corti-
cal bone (×50). B, Medium power photomicrograph of A shows osteoclastic resorption cavity (right) and embedded vascular struc-
tures (×100). (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 931

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-26  ■  Massive osteolysis. Peak age incidence and most frequent sites of skeletal involvement are indicated by solid black
arrows.

network of dilated, thin-walled capillaries growing in the Vascular Malformations


marrow spaces. The active lesions are purely vascular, and
the osteoclastic resorptive activity can be seen near the Vascular malformations are developmental in nature and
advancing edge. Stabilization of the lesion is associated typically grow proportionally with the host. They are
with an increased amount of intervening connective classified similarly to vascular neoplasms as of arterial,
tissue. On radiographs, massive osteolysis appears as lytic venous, or capillary in nature. They often manifest arte-
lesions that progressively involve the affected bones and riovenous shunting. They are frequently complicated
eventually obliterate the cortical and cancellous architec- by vascular ectasia and thrombosis. They range from
ture (Figs. 13-27 and 13-28). The dilated cystic spaces can superficial varicosities to large, deep masses that infiltrate
be seen grossly (Fig. 13-29). Late lesions can be predomi- soft tissue and may involve large portions of the body
nantly fibrous. The process tends to stabilize spontane- (Fig. 13-30). They are predominantly soft tissue lesions
ously. In some cases, extensive involvement of the chest and their involvement of the skeleton is rather secondary.
wall (ribs) is associated with pleural effusion and severe These conditions are best diagnosed in correlation with
pulmonary compromise, which can lead to a fatal outcome. clinical presentation and radiographic imaging data.
Because the clinical course of massive osteolysis is
variable and unpredictable, the choice of a particular
method of therapy is fraught with uncertainty. In fact,
Glomus Tumor
any histologically benign angiomatosis of bone or soft The glomus body is a form of arteriovenous anastomosis
tissue may occasionally behave clinically as a slowly pro- that plays a role in thermal regulation. It is distributed
gressive disorder that eventually compromises the func- throughout the body in the reticular dermis but is most
tion of the affected organ. In such instances, major frequently seen in the subungual region. The normal
resection procedures including amputation may be glomus body consists of an afferent arteriole that branches
required in order to control the process. Surgical resec- into two to four connecting arterioles. The arterioles
tion, embolization, radiation therapy, steroids, and more have thick segments surrounded by concentric layers
recently, interferon alfa have all been used in cases of modified perivascular smooth muscle cells. These
showing progression.87 segments are called Sucquet-Hoyer canals and consist of

ERRNVPHGLFRVRUJ
932 13  Vascular Lesions

single most common site is the subungual region of the


fingers. Other common sites include the pelvis, wrist,
forearm, and foot. They also can occur in the tip of the
coccyx, where they arise from the glomus coccygeum.102
In rare instances, they may originate in the deep organs
without apparent association with normal glomus bodies.92
The subungual lesions typically erode the underlying
distal phalanges.95,100,103,106,108 The second most frequent
location is in the subcutaneous tissue.91,97 Intraosseous
glomus tumors are extremely rare, and only single cases
have been reported in the literature.93,94,96,103,104,109–111
Most of these reports describe lesions that were com-
pletely embedded in the distal phalanges. Others have
been described as periosteal lesions in long bones.98,101
In our opinion, the possibility that some of these lesions
secondarily erode the bone cannot be completely ruled
out. Exceptional cases of glomus tumor arising in other
bones such as the proximal phalanx, tibia, ulna, and pelvis
have also been reported.92,94,103,108,110,111 The peak age inci-
dence and most frequent sites of skeletal involvement are
shown in Figure 13-33.

Clinical Symptoms
Glomus tumors are extremely painful, and symptoms are
often out of proportion with the size of the tumor. Par-
oxysms of pain in the affected area can be triggered by
changes of temperature or even by minor mechanical
disturbance of the affected area. The paroxysms of pain
may also appear spontaneously with no apparent cause.
Intraosseous lesions that cause these symptoms raise
clinical suspicion of osteoid osteoma. In contrast to
osteoid osteoma, pain from a glomus tumor is not pre-
FIGURE 13-27  ■  Massive osteolysis (disappearing bone disease) dominantly nocturnal, has a paroxysmal pattern, and does
involving humerus. Proximal two thirds of right humerus in a not respond to salicylates.
young adult has completely disappeared, and distal portion of
the shaft shows typical “sucked candy” attenuation. Fibrous
remnant showed capillary angiomatosis process in biopsy Radiographic Imaging
samples. (Courtesy Dr. T. Bauer, Cleveland.)
The subungual glomus tumor erodes the underlying
distal phalanx93,103,105 (Fig. 13-34). Occasionally, it can be
multifocal, involving the phalanges bilaterally.104 Intraos-
seous lesions present as well-demarcated lytic defects that
arteriovenous anastomosing vessels connected with effer- measure less than 1 cm in diameter (Fig. 13-35). In addi-
ent veins (Fig. 13-31). The glomus tumor was described tion to osteoid osteoma, intraosseous epidermal inclusion
in 1924 by Masson,105 who linked this peculiar neoplasm cysts and invasive subungual keratoacanthomas of the
to a normal glomus body and proposed that in some cases nail bed can produce lucent, painful lesions in the distal
it may represent a hyperplasia of perivascular cells rather phalanx that must be distinguished from intraosseous
than a true neoplasm. Normal glomus bodies can be glomus tumor. The lesions in bones other than phalanges
found within the bones and can be the source of an are intracortical, expand the cortex, and promote mild
intraosseous glomus tumor (Fig. 13-32). sclerosis in the adjacent bone. In summary, together with
its clinical symptoms, the lesion resembles osteoid
Definition osteoma on radiographs. In one of our cases the tumor
eroded the cortex of the ulna and extended into the sur-
Glomus tumor is a rare, benign distinctive neoplasm rounding soft tissues.108
composed of vascular channels and modified perivas-
cular muscle cells that recapitulate the structure of a Gross Findings
glomus body.
Glomus tumor in its typical location—the subungual
region—represents a well-circumscribed, soft, tan-gray
Incidence and Location
mass that can be separated easily during curettage from
Glomus tumors are extremely rare lesions, and most are the surrounding soft tissue or bone (Fig. 13-34). The cut
diagnosed in patients between ages 20 and 40 years. The Text continued on p. 939

ERRNVPHGLFRVRUJ
13  Vascular Lesions 933

B
FIGURE 13-28  ■  Massive osteolysis (disappearing bone disease) involving ribs. A, Plain radiograph of chest of young man with disap-
pearance of posterior portions of two upper ribs on left. Biopsy revealed capillary angiomatosis consistent with Gorham’s disease.
B, Enlargement of radiograph shown in A with disappearance of posterior portions of two left ribs. C, Radionuclide scan shows
absence of uptake in corresponding two ribs on left.

ERRNVPHGLFRVRUJ
934 13  Vascular Lesions

B
FIGURE 13-29  ■  Massive osteolysis: gross features. A, Bisected clavicle and portion of sternum. Note multiple cystic spaces and
replacement of bone by fibrous vascularized tissue. B, Higher magnification of A shows bisected claviculomanubrial joint. Note
multiple cystic changes in clavicle.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 935

A B

C
FIGURE 13-30  ■  Vascular malformation of the lower extremity: gross and radiographic features. A, Amputation specimen showing
large vascular structures representing tortuous thick-walled vasculature with cavernous dilations extensively involving the soft tissue
of the lower extremity. Inset, Higher magnification of specimen shown in A. B, T2-weighted magnetic resonance image showing
grapelike high signal intensities corresponding to soft tissue vascular lesions. C, Higher power gross photograph of the foot showing
extensive cystlike vascular lesions involving the soft tissue and eroding bones.

ERRNVPHGLFRVRUJ
936 13  Vascular Lesions

B
FIGURE 13-31  ■  Glomus body: microscopic features. A, Low power view of normal glomus coccygeum. B, Higher magnification of
A shows cluster of perpendicularly sectioned glomic arterioles of Sucquet-Hoyer canals (arrows) surrounded by thick layer of glomus
cells. Inset shows glomic arteriole surrounded by glomus cells and peripheral collecting vessel (denoted by asterisk). (A, ×50; B and
Inset, ×100) (A, B, and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 937

A B
FIGURE 13-32  ■  Glomus body: microscopic features. A, Nail bed containing normal glomus bodies (×50). B, Incidentally discovered
intraosseous normal glomus body. (A and B, ×100) (A and B, hematoxylin-eosin.)

20 40
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-33  ■  Glomus tumor. Peak age incidence and most frequent sites of skeletal involvement are indicated by solid black arrows.

ERRNVPHGLFRVRUJ
938 13  Vascular Lesions

A B

C D
FIGURE 13-34  ■  Glomus tumor: radiographic, gross, and microscopic features. A and B, Radiographs of distal phalanges show pres-
sure erosion of bone by subungual glomus tumors. C, Gross photograph of enucleated glomus tumor on fingernail. Exposed con-
cavity of tumor bed in distal phalanx can be seen just proximal to base of nail. D, Photomicrograph of glomus tumor of subungual
region shows endothelial-lined vessels and perivascular glomus cells in small nests and sheets (D, ×50) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 939

B
FIGURE 13-35  ■  Intraosseous glomus tumor of ulna: radiographic features. A, Anteroposterior view of elbow in a young woman who
had severe pain. A 1-cm lytic lesion (arrow) is seen in proximal ulna. B, Lateral view of elbow shows ovoid lytic lesion in ulna
(arrow). Sharp circumscription but no sclerosis is present.

surface is usually tan but can be hemorrhagic. Older Glomangiomyomas occur with equal frequency in the
lesions may have a fibrous appearance. upper and lower extremities. All the lesions reported as
primary bone tumors have been of the glomus tumor
proper category. The microscopic features are very char-
Microscopic Findings
acteristic and easy to recognize. The lesion has a rich
Extraosseous glomus tumors are divided into three vascular network surrounded by cuffs of epithelioid
microscopically distinct subtypes: glomus tumor proper, glomus cells (Fig. 13-36). These structures can be densely
glomangioma, and glomangiomyoma. The classification packed, forming highly cellular solid areas, or can be
is based on differing proportions of vascular, smooth separated by strands of loose connective tissue (compare
muscle, and epithelioid glomus cells. Glomus tumor Figs. 13-36 and 13-37). More solid lesions can mimic
proper occurs most frequently in the fingers. Glomangio- epithelial neoplasms. The rich, branching vascular
mas are more often seen in the soft tissue of the forearm. network can create a hemangiopericytoma-like pattern.

ERRNVPHGLFRVRUJ
940 13  Vascular Lesions

A B

FIGURE 13-36  ■  Glomus tumor: microscopic features. A and B, Common pattern within glomus tumor is composed of solid sheets
of cells with interspaced vessels. C, Low power photomicrograph shows large cystic spaces within solid sheets of tumor cells.
(A, ×60; B, ×100; C, ×50) (A-C, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
13  Vascular Lesions 941

A B

C D
FIGURE 13-37  ■  Glomus tumor: microscopic patterns. A, Periphery of glomus tumor shows tumor cells in fibrous stroma.
B, Cluster of glomus cells separated by fibrous stroma. C, Solid area of tumor with less prominent stromal tissue.
D, Higher magnification of C shows tumor cells with eosinophilic cytoplasm and round or oval nuclei. (A and C, ×200; B and D, ×400)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
942 13  Vascular Lesions

Ultrastructurally the glomus tumor cells represent modi- Definition


fied perivascular smooth muscle cells.99,107 There are
many deviations from this most frequent pattern, but Epithelioid hemangioendothelioma is a unique, well-
their description is beyond the scope of this book. differentiated endothelial vascular neoplasm with
an epithelioid appearance of its endothelial cells and a
Treatment and Behavior tendency to be multifocal.

Glomus tumors are benign, and simple excision of both


Incidence and Location
the soft tissue and intraosseous forms is curative. Malig-
nant glomus tumor is extremely rare in soft tissue and has Epithelioid hemangioendotheliomas of bone are ex-
not been described in bone. tremely rare. The limited experience with these lesions
indicates that they most frequently occur during the
second and third decades of life and there is a definite
MALIGNANT VASCULAR TUMORS predominance in male patients. Bones of the lower
extremities are preferentially involved. The tibia is the
Epithelioid Hemangioendothelioma most frequently involved, but any bone can be affected.
Most patients initially have multifocal lesions with a ten-
In 1979, Rosai et al.50 proposed a unifying concept dency to involve bones of the same region (e.g., bones of
of vascular tumors in which endothelial cells with abun- one extremity). Synchronous involvement of paired
dant cytoplasm and prominent vacuoles resembled his- bones, most frequently the tibia and fibula, is common,
tiocytes. These tumors involved a variety of tissues, but in some instances, completely separate synchronous
including bone, and Rosai et al. called them histiocyt- foci are present in distant anatomic sites such as the
oid hemangiomas. Later, in 1982, Weiss and Enzinger131 clavicle and lumbar vertebrae. The peak age incidence
proposed the term epithelioid hemangioendothelioma to and most frequent sites of skeletal involvement are shown
describe an unusual vascular tumor of soft tissue in in Figure 13-39.
which endothelial cells had an epithelioid appearance.
Epithelioid hemangioendotheliomas are not unique to
Radiographic Imaging
the soft tissue and occur in deep organs, most frequently
the lungs and liver.112,113,119 They also arise within The radiographic appearance of the lesion is not specific.
bone.114,116,117,121,122,124,125,130 Epithelioid hemangioendo- Epithelioid hemangioendotheliomas present as purely
theliomas in different sites are morphologically and bio- lytic lesions with varying degrees of peripheral sclerosis
logically similar and pursue a clinical course between (Figs. 13-40 to 13-43). The margins are well demarcated,
that of a hemangioma and a conventional high-grade and cortical erosion or complete disruption may be seen
angiosarcoma. They are classified as low-grade malig- (Figs. 13-44 and 13-45). Occasionally a more aggressive
nancies and have an indolent clinical behavior.114,117,120,123 growth pattern, such as a moth-eaten pattern, can be
We believe that epithelioid hemangioma is a variant of seen. Invasion into soft tissue is rare, and periosteal new
benign hemangioma and should not be confused with a bone formation is not present. The diagnosis of epithe-
low-grade epithelioid hemangioendothelioma (also see lioid hemangioendothelioma of bone can be suspected
the section on hemangioma). from findings on radiographs because of the multifocality
Recent transcriptomic sequencing combined with con- of the lytic defects and the peculiar tendency to involve
ventional cytogenetics identified the translocation involv- bones of one extremity (Figs. 13-45 and 13-46). In
ing (1;3)(p36;q25) resulting in the WWTR1-CAMTA1 unusual circumstances, the epithelioid hemangioendo-
chimeric gene in epithelioid hemangioendothelioma thelioma can provoke sclerosis and may present as a
(Fig. 13-38).115,129 It involves the WW domain-containing blastic lesion on radiographs.
transcription regulator 1 (WWTR1) mapping to 3q25 and
calmodulin-binding transcription activator 1 (CAMTA1)
Gross Findings
mapping to 1p36. The WWTR1-CAMTA1 chimeric gene
is under the transcription control of WWTR1 promoter. Typically epithelioid hemangioendotheliomas are well
The new protein contains the amino terminus compo- demarcated with irregular scalloped borders and a bright
nent of WWTR1 and a large portion of CAMTA1 at the red hemorrhagic and sometimes gelatinous appearance
carboxyl terminus. The transcription factor WWTR1 (Figs. 13-47 and 13-48). Erosions of cortex with complete
is physiologically highly expressed in endothelial cells, cortical disruption and extension into soft tissue can be
whereas CAMTA1 is normally expressed only in the central present. Multifocal lesions involving one bone or several
nervous system. The fusion with WWTR1 upregulates bones of an affected extremity are frequently seen.
CAMTA1, causing its aberrant expression in endothelial
cells of epithelioid hemangioendothelioma. Preliminary
Microscopic Findings
data indicate that this fusion is specific for epithelioid
hemangioendothelioma regardless of its site of origin, The tumor usually consists of solid nests or anastomosing
including those that develop in bone and can be used cords of epithelioid cells that occasionally form narrow
in the differential diagnosis of vascular conditions with vascular channels (Figs. 13-49 and 13-50).117,120,124,130
epithelioid features.115,129 Text continued on p. 956

ERRNVPHGLFRVRUJ
13  Vascular Lesions 943

Chr 1
Chr 3

cds
26
WWTR1 36.3
CAMTA1 36.2
25 36.1
24.3 35
24.1
24.2 cen tel 34.3
23 34.2
34.1
22 12 3 4 5 6 7 33
21.3 32.3
32.2
32.1
21.2 31.3
21.1

cds
31.2
14.3
14.1
14.2 CAMTA1 31.1
13 22.3
22.2
12 22.1
11.1 11.2 21
11.1
11.2 12 13.3
13.1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 13.1 13.2
13.2 12
11
13.3 11
21 12
22
23 21.1
21.2 21.3
24 WWTR1
25.1 22
25.2 23
25.3
26.1 24
26.2 25
26.3
27
28 1 2 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 31
29
32.1
706 aa 32.2 32.3
41
A 42.1
42.2 42.3
43
44

WWTR1
WW Q-rich CC PDZ

breakpoint
CAMTA1
CG-1 TIG ANK IQ

breakpoint
Fusion protein
WW TIG ANK IQ 1455 aa

breakpoint
WW – WW domain
WWTR1 portion CAMTA1 portion CC – Coiled-coil domain
PDZ – PDZ binding motif
CG-1 – CG-1 DNA binding domain
TIG – Transcription factor immunoglobulin domain
ANK – Ankyrin repeats
IQ – IQ calmodulin-binding motifs
Q-rich – Glutamine rich region
B
FIGURE 13-38  ■  Molecular structure and breakpoints associated with t(1;3)(p36;q25) resulting in WWTR1-CAMTA1 gene fusion in
epithelioid hemangioendothelioma. A, Chromosomal diagrams depicting the t(1;3)(p36;q25) resulting in WWTR1-CAMTA1 fusion.
The WWTR1 gene is located on the long arm of chromosome 3. The CANTA1 is located on the short arm of chromosome 1. As a
result of translocation two derivatives chromosomes are formed. The WWTR1-CAMTA1 fusion is transcribed from der(1). B, Exon-
intron structures of the WWTR1 and CAMTA1 genes are shown. The molecular structure of the hybrid gene, which includes the
coding sequences of the amino-terminus domain WW of WWTR1 and several transcriptionally active domains (TIG, ANK, and IQ)
of CAMTA1 at the carboxyl terminus is shown. In this fusion, the transcriptional domains of CAMTA1 are driven by the strong
promotor of WWTR1 and are overexpressed in cells of epithelioid hemangioendothelioma. C, The structures of the WWTR1 and
CAMTA1 proteins as well as the structure and functional domains of the fusion protein are shown. Solid arrows indicate the posi-
tion of breakpoints.

ERRNVPHGLFRVRUJ
944 13  Vascular Lesions

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-39  ■  Epithelioid hemangioendothelioma. Peak age incidence and most frequent sites of skeletal involvement are indicated
by solid black arrows.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 945

A B

C E
FIGURE 13-40  ■  Epithelioid hemangioendothelioma: radiographic features. A and B, Lytic sharply demarcated lesion of distal humerus
with expansion of bone contour medially and posteriorly. C, Lytic focus in the tibial shaft with diffuse sclerosis of adjacent bone.
D, Destructive lytic lesions in distal femoral shaft. Note motheaten pattern of bone destruction. E, Computed tomogram of lesion
in D shows low signal intramedullary lesion with multiple erosions of cortex.

ERRNVPHGLFRVRUJ
946 13  Vascular Lesions

C
FIGURE 13-41  ■  Epithelioid hemangioendothelioma: radiographic features. A, Multiple lytic lesions of right pelvis. B and C, Computed
tomograms of lesions shown in A document multiple sharply demarcated lytic lesions of ilium.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 947

A B

C D
FIGURE 13-42  ■  Epithelioid hemangioendothelioma: radiographic features. A-C, Extensive involvement of metatarsal bones with
punched-out lytic lesions involving cortical bone and medullary cavities. D, Specimen radiograph of resected distal metatarsal
segment containing lytic focus of epithelioid hemangioendothelioma.

ERRNVPHGLFRVRUJ
948 13  Vascular Lesions

A
FIGURE 13-43  ■  Multicentric epithelioid hemangioendothelioma of bone: radiographic features. A, Anteroposterior radiograph of foot
of an 18-year-old man with multiple lytic lesions of metatarsal and tarsal bones. B, Opposite foot of patient in A shows pathologic
fracture of fourth metatarsal through focus of epithelioid hemangioendothelioma.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 949

FIGURE 13-44  ■  Multicentric epithelioid hemangioendothelioma: radiographic features. Computed tomograms of right and left tarsal
bones show multicentric, sharply demarcated lesions on right side.

ERRNVPHGLFRVRUJ
950 13  Vascular Lesions

B
FIGURE 13-45  ■  Multicentric epithelioid hemangioendothelioma: radiographic features. A and B, Anteroposterior and lateral radio-
graphs of foot of young man show multiple lytic foci without reactive sclerosis or periosteal new bone formation involving meta-
tarsals, phalanges, tarsal bones, and distal tibial shaft.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 951

A B

C D
FIGURE 13-46  ■  Multicentric epithelioid hemangioendothelioma of bone: radiographic features. A and B, Lateral and anteroposterior
radiographs of leg of a 29-year-old woman show multiple small, sharply circumscribed lytic lesions of right tibia and fibula.
C, Radionuclide scan of patient in A and B shows increased isotope uptake in multiple tarsal bones and distal tibia. D, Lateral radio-
graph of foot and ankle shows multiple lytic lesion in calcaneus and cuneiform bone.

ERRNVPHGLFRVRUJ
952 13  Vascular Lesions

C D
FIGURE 13-47  ■  Epithelioid hemangioendothelioma: radiographic and gross features. A, Lateral radiograph of leg show multiple
sharply demarcated lytic lesions of tibia and tarsal bones. B, Magnetic resonance image showing multifocal well-demarcated low
signal lesions involving distant tibia and tarsal bones. C, Gross photograph of bisected amputation specimen showing multifocal
well-demarcated hemorrhagic lesions involving distal tibia and tarsal bones. D, Higher magnification of the specimen shown in C.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 953

A B

C D
FIGURE 13-48  ■  Epithelioid hemangioendothelioma: gross features. A, Cut surface of distal segment of metatarsal bone containing
intramedullary focus of epithelioid hemangioendothelioma eroding cortex in neck region. B, Whole-mount section of lesion shown
in A. Note sharp circumscription of tumor tissue. C, Resected head and shaft of proximal radius containing gelatinous, dark red
lobulated tumor expanding into soft tissue. D, Hemangioendothelioma involving medullary cavity of proximal ulna.

ERRNVPHGLFRVRUJ
954 13  Vascular Lesions

A B

C D
FIGURE 13-49  ■  Epithelioid hemangioendothelioma: microscopic features. A, Solid nests of epithelioid endothelial cells and irregular
open vascular channels. B, Higher magnification of A shows irregular vascular channel lined by epithelioid endothelial cells.
Note multiple layers of endothelial cells with micropapillary structures. C, Irregular anastomosing vascular spaces within epithelioid
cells. D, Cords and small nests of epithelioid endothelial cells occasionally forming primitive vascular channels. (A, ×200; B-D, ×400)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 955

A B

C D

FIGURE 13-50  ■  Epithelioid hemangioendothelioma: microscopic features. A, Irregular anastomosing cords of epithelioid endothelial
cells. B, Higher magnification of A shows epithelial endothelial cells forming ill-defined cords and nests. C, Irregular anastomosing
vessels and cords of epithelioid endothelial cells. D, Higher magnification of C showing epithelioid endothelial cells lining vascular
channels and forming cords. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
956 13  Vascular Lesions

Scattered, more solid clusters of epithelioid cells also may of angiogenesis can be documented ultrastructurally as
be present. The supporting connective tissue may exhibit well (Fig. 13-58).128,130
myxoid or chondroid change (Fig. 13-51). Focally, indi-
vidual epithelioid cells are embedded in a basophilic Differential Diagnosis
matrix that mimics hyaline cartilage. Individual cells are
round or polygonal. Cellular pleomorphism is not con- The epithelioid features of this tumor, with clusters of
spicuous. Mitotic activity is low, within the range of 1 to large endothelial cells as well as solid nests and cords,
2 mitoses per 10 high-power fields. Tumors showing an can mimic metastatic carcinoma in bone. Immunohisto-
abundance of mitoses, marked nuclear pleomorphism, chemical demonstration of keratin and epithelial mem-
and atypical, multipolar mitotic figures should be classi- brane antigen usually identifies the adenocarcinoma cells
fied as angiosarcomas, with or without epithelioid fea- in the absence of reactivity for endothelial markers such
tures. The cytoplasm of the epithelioid cells may be as factor VIII-related antigen, CD31, CD34, and ERG.
densely eosinophilic (Fig. 13-52). The nuclei are On the other hand, keratin reactivity in epithelioid
round with prominent nucleoli. The cytoplasm of the hemangioendothelioma has been reported and signifies
epithelioid cells characteristically shows vacuolization, the need for the use of a panel of multiple antibodies to
and sometimes, a signet-ring–like appearance is produced distinguish these tumors.
by a large vacuole that distorts the cell (Fig. 13-53). The Composite tumors containing areas of epithelioid
large cytoplasmic vacuoles probably signify attempts of hemangioendothelioma and hemangioma30,127 as well as
the tumor to form primitive vascular lumina. epitheloid angiosarcomas arising in hemangioendotheli-
Vascular channels in varying stages of angiogenesis oma have been described.118
usually are present. Some channels have only single cells The distinction from angiosarcoma is based on the
with intracytoplasmic lumina, whereas others consist of uniformity of the proliferating endothelial cells, the low
several cells with a compact, solid arrangement. Solid mitotic rate, and the minimal nuclear pleomorphism
nests of epithelioid cells that merge with capillary-sized exhibited by the endothelial cells in these tumors. Angio-
vessels mimic vessels of granulation tissue. Some cases sarcoma shows a much higher level of nuclear atypicality
can be exclusively composed of anastomosing or branch- and prominent spindle-cell differentiation. Intraluminal
ing cords of epithelioid cells in a background of chon- budding of neoplastic endothelial cells with a high
droid and myxoid matrix. The individual epithelioid cell nuclear/cytoplasmic ratio is commonly seen in angiosar-
cytoplasmic lumina may contain red blood cells, which coma but not in epithelioid hemangioendothelioma. The
can also be found within the cords of epithelioid cells myxoid and chondroid matrices, which may be found
that resemble primitive vessels. An inflammatory infil- in epithelioid hemangioendotheliomas, can sometimes
trate of varying degree is often present and consists of suggest a cartilage neoplasm in biopsy material. Their pres-
eosinophils, lymphocytes, and plasma cells. In some ence in areas of cells that contain cytoplasmic luminal
cases, the eosinophilic infiltrate can be very pronounced vacuoles can add to the resemblance to cartilage lesions,
and may obscure the endothelial tumor elements. The but these cells often show immunoreactivity for factor
lesion may have an overall nodular architecture com- VIII-related antigen CD31, CD34, and ERG. They are
posed of areas of solid epithelioid endothelial cells with typically negative for cartilage markers. It is expected that
poorly developed irregular anastomosing vascular chan- molecular tests for WWTR1-CAMTA1 fusion will play a
nels (Figs. 13-54 and 13-55). Foci of hemorrhage and major role in differential diagnosis as preliminary evi-
necrosis may be present. Rarely, the stroma may contain dence indicates high specificity of this abnormality for
an abundance of osteoclast-like giant cells.132 The vacu- epithelioid hemangioendothelioma.
oles of epithelioid cells are negative in periodic acid–
Schiff and alcian blue stains. Silver stains show reticulin Treatment and Behavior
fibers encircling individual epithelioid cells and outline
their small nest or cords. Epithelioid cells express a full Epithelioid hemangioendotheliomas are low-grade, well-
roster of endothelial markers and are at least focally differentiated borderline tumors with locally destructive
positive for factor VIII-related antigen, CD31, CD34, indolent behavior. There is no unanimity as to treatment.
and ERG (Fig. 13-56).30,120,230 Less frequently, the coex- Some lesions have been treated with en bloc excision or
pression of epithelial markers with focal positivity for curettage only. In some cases, especially with reference
epithelial membrane antigen and keratin, particularly to multifocal lesions, radiation therapy is used. Of seven
low-molecular-weight keratin, can be present.57 Ultra- patients who had multicentric tumors and who were
structurally, the epithelioid cells are identified as endo- available for follow-up, all were alive for 1 month to 10
thelial cells containing Weibel-Palade bodies. Reflecting years (mean, 4.3 years) after treatment. Four patients who
the light microscopic appearance, epithelioid endothe- had solitary lesions and for whom follow-up data were
lial cells are seen lining the vascular spaces, sometimes available were alive 9 months to 4 years (mean, 1.8 years)
occluding them and showing abundant cytoplasm with after surgical treatment. In one patient in this group, a
irregular free cytoplasmic borders with pinocytotic ves- nodule of the abdominal wall was excised, but the tumor
icles (Fig. 13-57). Abundant cytoplasmic aggregates of recurred 1 year after treatment. These data indicate that
intermediate filaments with a thickness of 10 nm can epithelioid hemangioendotheliomas of bone pursue an
be correlated with the epithelioid light microscopic indolent course and can be controlled by limited surgery,
appearance (Fig. 13-58). The formation of primitive radiofrequency ablation,126 and radiotherapy.130
intracellular vascular lumina representing early stages Text continued on p. 965

ERRNVPHGLFRVRUJ
13  Vascular Lesions 957

A B

C D
FIGURE 13-51  ■  Epithelioid hemangioendothelioma: microscopic features. A, Epithelioid endothelial cells forming poorly developed
vascular channels, cords, and nests in a chondroid-like stroma. B, Higher magnification of A shows epithelioid endothelial cells
forming cords and nests in chondroid-like stroma. C, Epithelioid endothelial cells forming solid sheets with slit vascular spaces.
D, Higher magnification of C shows solid sheets of epithelioid endothelial cells with poorly developed vascular spaces. (A and
C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
958 13  Vascular Lesions

A B

C D
FIGURE 13-52  ■  Epithelioid hemangioendothelioma: microscopic features. A and B, Low power photomicrographs showing solid
sheets and small nests of epithelioid endothelial cells. C and D, Higher power photomicrographs showing solid sheets and nests
of epithelioid endothelial cells occasionally forming poorly developed small vascular spaces. (A and B, ×100; C and D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 959

B
FIGURE 13-53  ■  Epithelioid hemangioendothelioma of bone: microscopic features. A, Low power magnification of pattern of plump,
epithelioid cells arranged in cords in basophilic myxoid stroma. B, Higher magnification shows characteristic intracytoplasmic
vacuoles in epithelioid endothelial cells. Matrix has chondroid aura. Inset shows various-sized cytoplasmic vacuoles and dense
cytoplasm in same cells. Note signet-ring appearance of some endothelial cells. (A, ×100; B, ×200; Inset, ×400) (A, B, and
Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
960 13  Vascular Lesions

B
FIGURE 13-54  ■  Epithelioid hemangioendothelioma: microscopic features. A and B, Low power photomicrographs show overall
nodular architecture of this epithelioid hemangioendothelioma. (A, ×25; B, ×50.) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 961

B
FIGURE 13-55  ■  Epithelioid hemangioendothelioma: microscopic features. A, Solid nests of epithelioid endothelial cells with anasto-
mosing vascular pattern. B, Higher magnification of A shows densely packed epithelioid endothelial cells with distinct cytoplasmic
borders. (A, ×200; B, ×400) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
962 13  Vascular Lesions

A B

C D
FIGURE 13-56  ■  Epithelioid hemangioendothelioma: immunohistochemical features. A, B, and D, Strong reactivity of epithelial endo-
thelioid cells with Ulex europaeus lectin. C, Strong positivity of epithelial endothelioid cells for factor VIII-related antigen. (A, B, and
D, ×400; C, ×200.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 963

B
FIGURE 13-57  ■  Epithelioid hemangioendothelioma: light microscopic and ultrastructural features. A, Anastomosing pattern of
vessels lined by prominent epithelial endothelial cells. B, Ultrastructure of crowded epithelioid endothelial cells that line vascular
channel. (A, ×200; B, ×4000) (A, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
964 13  Vascular Lesions

B
FIGURE 13-58  ■  Epithelioid hemangioendothelioma: ultrastructural features. A, Perpendicularly cut cord of endothelial cells in fibrous
stroma. B, Higher magnification of A shows cytoplasms of two endothelial cells connected by tight junctions. Note prominent base-
ment membrane surrounding cord of endothelial cells. Inset documents densely packed intermediate filaments consistent with
vimentin within cytoplasm of endothelial cell. (A, ×3,000; B, ×6,500; Inset, ×15,000)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 965

Spindle-cell Hemangioendothelioma who received thorium dioxide (Thorotrast).172 A single


case of malignant papillary angioendothelioma (Dabska
Spindle-cell hemangioendothelioma was described in tumor) arising in bone has been reported, which was
1986 by Weiss and Enzinger142 as a peculiar vascular thought to have arisen in a hemangioma of the femur.168
tumor that exhibited features of cavernous hemangioma Primary angiosarcoma of bone is very rare and accounts
and contained foci of spindle-cell proliferation with fea- for less than 1% of all angiosarcomas.158 In bone, this
tures similar to Kaposi’s sarcoma. This rare tumor occurs term is reserved for malignant vascular endothelial neo-
predominantly in the superficial soft tissue (subcutis and plasms of intermediate to high histologic grade. In the
dermis) of distal parts of the extremities, particularly in past, such terms as malignant hemangioendothelioma and
the hand.137,140–142 In soft tissue, it grows locally in a mul- hemangioendothelial sarcoma of bone were used interchange-
tinodular pattern. The low power microscopic features ably with angiosarcoma.114,117,160,173–177 This created some
include the presence of large vascular spaces with throm- ambiguity; therefore, we recommend that the term
bosis and adjacent proliferation of blunt spindle cells. hemangioendothelioma of bone be used to designate very
The spindle cells also can merge with cavernous vessels low-grade to borderline endothelial vascular bone tumors
that proliferate within the thickened intervascular septa. while the term angiosarcoma in bone should be used to
We have seen only two examples of spindle-cell heman- define intermediate to high-grade tumors with definitive
gioendothelioma in bone. One was in the form of two metastatic potential.
separate foci in the calvarium, and the second involved
the body of a vertebra (Fig. 13-59). The two basic pat- Definition
terns of spindle-cell proliferation described in soft tissue
lesions were observed in these cases (Fig. 13-60). In addi- Angiosarcoma is an intermediate- to high-grade malig-
tion, the cranial lesions also contained foci of epithelioid nant tumor composed of atypical endothelial cells that
endothelial cells. Originally, spindle-cell hemangioendo- have vasoformative features.
theliomas were considered to be low-grade malignant
tumors.137,140,142 In recent reported series, the follow-up Incidence and Location
data of these tumors indicate that there is no evidence of
malignant behavior after excision.134,135 The exact bio- Angiosarcoma is an extremely rare tumor. Data from the
logic potential of these tumors in bone is unknown. The Surveillance, Epidemiology, and End Results study indi-
two patients in our series who had spindle-cell heman- cate that less than 2% of primary bone sarcomas were
gioendothelioma of bone are free of disease 5 and 7 years classified as angiosarcomas. These tumors usually occur
after curettage. We are not completely convinced that in long tubular bones. Like low-grade vascular tumors,
this lesion represents a distinct form of vascular neo- they can be multicentric, involving multiple bones of the
plasm. In our opinion, the possibility that the spindle-cell lower extremity. More typically, angiosarcoma presents as
component may represent a secondary reactive change in a solitary lesion. This may explain the early observation
a preexisting vascular lesion cannot be completely ruled that multicentric malignant vascular tumors of bone seem
out. In fact, several recent studies from different institu- to have a better prognosis. The disparity may be more
tions also advanced this concept.133,136 Additional reported related to better differentiation than to multicentricity.
cases affecting the skeleton follow benign clinical behav- Angiosarcomas of bone have been observed to occur as
ior.138,143 The current consensus is that, in the spectrum secondary malignancies in bone infarcts,144,150 in fibrous
of tumors designated as hemangioendothelioma, the dysplasia,164 and after exposure to external irradiation.178
spindle-cell variant is very likely a benign condition.139
Radiographic Imaging
Angiosarcoma The radiographic presentation of angiosarcoma of bone
The term angiosarcoma is applied to a wide range of is not specific. A solitary lesion presents as a destructive
malignant endothelial vascular neoplasms that affect a lytic mass with irregular borders. High-grade lesions
variety of extraskeletal sites. Angiosarcomas arising at exhibit features of complete cortical destruction and
different sites and in different organs have some distinct extension into soft tissue (Figs. 13-61 and 13-62). The
features and are regarded as separate clinicopathologic vascular nature of the lesion can be suspected on radio-
entities. They occur most frequently in the dermis and graphs if synchronous multicentric involvement of several
subcutis, especially in the head and neck region.147,153 bones of one extremity or of one anatomic region is
Other frequent anatomic sites of involvement are the present.
breast, liver, and spleen.146,161,162 These neoplasms may
develop as complications of a preexisting condition.
Microscopic Findings
Chronic lymphatic stasis is a widely recognized predis-
posing condition for angiosarcoma of the dermis and soft The tumor is composed of atypical endothelial cells
tissue.148,149,151,157,159,163,167,170 Angiosarcomas can also be and exhibits vasoformative features. In a typical case,
induced by radiation exposure152,155,165,178 and are linked vascular differentiation is easy to recognize. The vessels
to various chemical carcinogens such as arsenic trioxide– usually produce an anastomosing system of irregular
containing insecticides, vinyl chloride,169,172 and long- channels lined by plump endothelial cells (Figs. 13-63
term treatment with androgenic anabolic steroids.161 to 13-67). High-grade tumors exhibit prominent nuclear
Nearly 25% of hepatic angiosarcomas arise in patients Text continued on p. 975

ERRNVPHGLFRVRUJ
966 13  Vascular Lesions

B C
FIGURE 13-59  ■  Spindle-cell hemangioendothelioma of skull: radiographic features. A, Lateral radiograph of skull of a 19-year-old
woman with multifocal lytic lesions of frontal and parietal regions. B, Computed tomogram shows expansile lytic tumor of parietal
region. C, Axial magnetic resonance image with gadolinium enhancement shows both frontal and parietal lesions.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 967

A B

C D
FIGURE 13-60  ■  Spindle-cell hemangioendothelioma of bone: microscopic features. A, Low powered view shows two components
of tumor: vascular and solid spindle cells. B, Higher magnification of solid spindle-cell area composed of interlacing bundles of
plump spindle cells. Note hemosiderin deposits and scattered inflammatory cells. C, Higher magnification of vascular component.
D, Interference between vascular and spindle-cell components shows spindle-cell proliferation in septa of vascular spaces. (A, ×25;
B-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
968 13  Vascular Lesions

FIGURE 13-61  ■  Angiosarcoma of bone: radiographic features. Destruction by tumor of distal tibial shaft with cortical breakthrough
laterally and slight periosteal new bone formation. Tumor shows wide transition zone and is completely lytic in appearance.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 969

A C
FIGURE 13-62  ■  Angiosarcoma of bone: radiographic features. A, Sagittal magnetic resonance image (MRI) of spine shows low density
destructive tumor of sacrum. Note metastases involving multiple vertebral bodies. B and C, Coronal MRIs showing widespread
metastasis involving pelvic and femoral bones.

ERRNVPHGLFRVRUJ
970 13  Vascular Lesions

A B

C D
FIGURE 13-63  ■  Angiosarcoma of bone: microscopic features. A and B, Low and higher power photomicrographs showing irregular
anastomosing vascular channels lined by highly atypical endothelial cells. C and D, Irregular anastomosing vascular spaces and
solid areas of highly atypical spin cell proliferations. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 971

A B

C D
FIGURE 13-64  ■  Angiosarcoma of bone: microscopic features. A and B, Low and high power photomicrographs showing anastomos-
ing vascular channels lined by highly atypical endothelial cells. C and D, Low and high power photomicrographs showing irregular
anastomosing channels and solid glomeruloid proliferations of highly atypical endothelial cells inside of vascular spaces. (A and
C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
972 13  Vascular Lesions

A B

C D
FIGURE 13-65  ■  Angiosarcoma of bone: microscopic features. A and B, Low and intermediate power photomicrographs show poorly
developed anastomosing vascular spaces and solid areas of spindle cell proliferations. C and D, Intermediate and high power pho-
tomicrographs showing irregular anastomosing vascular spaces lined by highly atypical endothelial cells. (A, ×60; B and C, ×100;
D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 973

A B

C D
FIGURE 13-66  ■  Angiosarcoma of bone: microscopic features. A and B, Intermediate and high power photomicrographs showing
vascular spaces lined by highly atypical epithelioid endothelial cells forming occasionally solid nodules. C and D, High power
photomicrographs showing vascular channels lined by highly atypical epithelioid endothelial cells forming loosely arranged
nodules. Note single epithelioid cells dispersed in stromal tissue mimicking metastatic carcinoma. (A, ×100; B and C, ×200; D, ×400.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
974 13  Vascular Lesions

A B

C D
FIGURE 13-67  ■  Angiosarcoma of bone: microscopic features. A and B, Low and intermediate power photomicrographs showing
irregular cystlike vascular spaces and proliferations of atypical endothelial cells forming septations and cords. C and D, High power
photomicrograph showing highly atypical epithelioid cells forming solid nests, cords, and septations. (A, ×60; B, ×100; C, ×200;
D, ×400.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 975

atypia and brisk mitotic activity with atypical mitotic pattern.195,196,199,200,203,206 On the other hand, the so-called
figures. There is a marked tendency toward intralu- hemangiopericytoma-like pattern frequently occurs in
minal budding of endothelial cells (Fig. 13-64). Large a wide range of spindle-cell neoplasms—mesenchymal
solid areas of endothelial cells that mimic an epithelial chondrosarcoma, endometrial stromal sarcoma, malignant
neoplasm can be present. Spindle-cell differentiation fibrous histiocytoma, synovial sarcoma, and osteosar-
with pleomorphic features can also be prominent (Fig. coma, among others. Therefore it is frequently stated that
13-63). Intermediate-grade, well-differentiated angio- the diagnosis of hemangiopericytoma requires exclusion
sarcomas may have better developed vascular channels of other spindle-cell neoplasms in which this pattern can
and exhibit less pronounced atypia of endothelial cells. be seen.198,208 As discussed in Chapter 22, bone and soft
Immunohistochemically, cells that exhibit vasoforma- tissue tumors with a hemangiopericytomatous pattern are
tive features usually show reactivity for endothelial sometimes associated with the paraneoplastc syndrome
markers (factor VIII-related antigens, CD31, CD34, and of oncogenic osteomalacia (phosphaturic mesenchy-
ERG).11,18,144,145,151,154,156,166,171 Accentuation of reactiv- mal tumors).186,187 The modern concept unifies solitary
ity can be seen at the luminal border of these cells for fibrous tumors and hemangiopericytomas as represent-
cytoplasmic antogens. ERG belongs to the family of ing microscopic variants of a spectrum of tumors with
transcription factors and typically shows strong nuclear similar pathogenesis. They range from lesions displaying
staining. In less differentiated areas lacking obvious vaso- bland spindle cells separated by prominent bands of ropy
formative features, endothelial marker reactivity can be collagen and diffuse positive staining for CD34 to more
very focal or absent. In contrast with low-grade endo- classical hemangiopericytomas in which the stromal col-
thelial bone neoplasms, epithelial markers (epithelial lagen is less prominent and the lesion has an overall cel-
membrane antigen and cytokeratins) are less frequently lular appearance with high vascularity and some mitotic
present.156,166,171 activity. It is not unusual to have those two components,
less cellular and more cellular, admixed in one lesion.
The cellular variants of hemangiopericytoma have a ten-
Differential Diagnosis
dency to be less strongly positive for CD34, which is
Vasoformative features may be inconspicuous in higher- typically restricted to vasculature, but there are some
grade angiosarcomas of bone, and such tumors may focal areas of positivity within the hypercellular areas.
show only weak reactivity for endothelial immunohisto- The lesions seen in bone typically fall into the category
chemical markers. The absence of cytokeratin reactivity of hypercellular classical hemangiopericytomas, and the
and strong reactions to antibodies for vimentin helps to so-called fibrous variants are less frequent as primary in
distinguish these tumors from carcinomas of the spindle-cell extracranial sites in the skeleton. The unified concept
type. Distinguishing lower-grade angiosarcomas from epi- is further confirmed by the presence of the same NAB2-
thelioid hemangioendothelioma is discussed in the section STAT6 gene fusion in these tumors.183,201,202 The fusion
on the differential diagnosis for that neoplasm, but can be detected by fluorescent in situ hybridization in
coexistence of epithelioid hemangioendothelioma and conventional histologic preparations. A convenient
epithelioid angiosarcoma in the same lesion is possible. surrogate of molecular testing is immunohistochemis-
try documenting the overexpression of STAT6 protein
driven by the NAB2 promoter in the solitary fibrous/
Treatment and Behavior
hemangiopericytoma family of tumors.202
High-grade angiosarcomas exhibit extremely aggressive
behavior with rapid local growth and early disseminated Definition
metastases. Radical en bloc excision or amputation is
indicated. Some lesions have a less aggressive clinical Hemangiopericytomas are malignant neoplasms com-
course, but the prognosis in a group of patients with posed of pericytic cells that grow in solid sheets and nests
high-grade angiosarcomas is poor because these tumors around irregular branching vascular spaces. These chan-
have a high propensity for metastases. Combinations of nels are lined by a single layer of endothelial cells.
radiation therapy and chemotherapy have been used as
adjuvant methods of treatment, but significant data of Incidence and Location
their effectiveness are lacking.
Hemangiopericytoma is an extremely rare neoplasm
Hemangiopericytoma in bone, and several series consisting of fewer than
10 cases have been reported from major bone tumor
Hemangiopericytoma was described by Stout and centers.180,182,194,206 On the basis of published data, it seems
Murray in 1942.205 The authors postulated that the tumor that this tumor usually involves the long bones of the
is composed of cells related to blood vessels similar to extremities, particularly of the leg, but any bone can
those described by Zimmerman in 1923 and designated be affected.184,188,208 Hemangiopericytomas are relatively
as pericytes.209 Pericytes are smooth muscle–like perivas- frequent in the head and neck region. Some lesions in
cular cells with contractile potential. They do not have this region exhibiting involvement of bone are prob-
readily identifiable or specific phenotypic features and ably not primary bone tumors and may originate in the
are difficult to distinguish from other mesenchymal adjacent meningothelial or soft tissue. Most reported
spindle cells. For that reason, the diagnosis of this neo- cases are in adults. It is well known that any apparent
plasm is based on the recognition of its architectural primary hemangiopericytomas in bone may prove to be

ERRNVPHGLFRVRUJ
976 13  Vascular Lesions

metastatic from extraskeletal neoplasms. The hemangio- channels. The size of the vessels ranges from narrow
pericytoma of meninges is the most frequent source of capillaries to large, open cavernous vessels (Fig. 13-69).
these late metastases in bone (see Chapter 14).179,191–193 Characteristically, hemangiopericytoma shows branching
These tumors may rarely be associated with oncogenic open vascular channels with antler or staghorn configura-
rickets and osteomalacia (see Chapter 22). Hemangio- tions. The solid perivascular areas are composed of
pericytomas show clonal chromosomal aberrations, and plump, densely packed spindle cells that lack the fascicu-
some of them are aneuploid by flow cytometric mea- lar arrangement seen in fibrosarcoma or synovial sarcoma.
surements.185,204 Similar clonal translocations involving The level of cellularity can vary in different tumors and
chromosomes 12 and 19 were seen in hemangioperi- even in different areas of the same lesion. It can range
cytomas in different anatomic sites (intracranial and from predominantly fibrous areas with gradual transition
extracranial).190 This suggests that hemangiopericytomas to more cellular foci. In soft tissue tumors the level
involving different organs are related and form a patho- of mitotic activity, necrosis, and pronounced nuclear
genetically distinct group. These earlier observations are atypia correlate to some extent with clinical behavior.
in keeping with the modern concept postulating a unified The value of these features in predicting the behavior of
pathogenesis for the family of solitary fibrous tumors/ hemangiopericytoma in bone has not yet been estab-
hemangiopericytomas at different anatomic sites based lished. Ultrastructurally, hemangiopericytoma cells have
on the presence of NAB2-STAT6 gene fusion in these some features of perivascular cells with smooth muscle
tumors.183,201,202 differentiation.180,181,189,197

Radiographic Imaging Differential Diagnosis


The radiographic presentation of hemangiopericytoma It is essential to exclude other neoplasms that may exhibit
is not specific and shows a purely lytic intramedullary patterns of anastomosing branching vascular lumens that
mass with a destructive growth pattern (Fig. 13-68). Cor- suggest hemangiopericytoma in focal areas. These include
tical disruption and extension into soft tissue may be osteosarcoma, malignant fibrous histiocytoma, mesenchymal
present. chondrosarcoma, and synovial sarcoma. In our experience,
many tumors with indubitable features of hemangio-
pericytoma prove to be metastases from other sites,
Microscopic Findings
most frequently the meningeal hemangiopericytoma or
The tumor is composed of compact areas of spindle cells gastrointestinal stromal tumors. The frequent immu-
that surround thin-walled, endothelial-lined vascular nohistochemical positivity of hemangiopericytoma and

FIGURE 13-68  ■  Hemangiopericytoma: radiographic features. Computed tomogram shows destructive lesion involving body of tho-
racic vertebra and adjacent rib.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 977

B C
FIGURE 13-69  ■  Hemangiopericytoma of bone: microscopic features. A, Intermediate power photomicrograph of spindle-cell tumor
with branching staghorn pattern of vascular spaces. B, Low power magnification shows spindle-cell tumor with branching staghorn
pattern of vascular spaces. C, Intermediate power photomicrograph shows bland nuclei in spindle cells. Vascular channels are lined
by flat, inconspicuous endothelial cells. (A and C, ×100; B, ×50) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
978 13  Vascular Lesions

gastrointestinal stromal tumors for CD34 can be another virtually all cases of Kaposi’s sarcoma, regardless of its
confusing feature. A panel of immunohistochemical type, and serves as the most efficient biomarker for this
and molecular markers is recommended; gastrointesti- condition.4,16,236
nal stromal tumors are typically positive for c-Kit and
DOG-1. In addition, they carry mutations of the c-Kit Incidence and Location
and PDGFR genes. Patients with meningeal hemangio-
pericytoma metastatic to the extracranial skeletal sites Involvement of bones has been reported in all four forms
typically have a history of primary meningeal tumor. of Kaposi’s sarcoma.210,213,218,219,222,225,227,233,237 The bone
More recent reappraisal of small series of cases previ- lesions develop during the more advanced stages of the
ously diagnosed as hemangiopericytoma of bone with a clinical course. A patient who presents with the lesion in
panel of immunohistochemical and molecular markers a bone invariably has extraskeletal disease. In the classic
indicates that some of these cases can be reclassified as chronic form, the skeletal involvement usually occurs in
synovial sarcomas or solitary fibrous tumors.207 the region affected by the skin and soft tissue disease—
in the small bones of the feet, often by direct extension.
In other forms, the site of skeletal involvement is unpre-
Treatment and Behavior
dictable and may appear in any bone. In view of the
En bloc resection or amputation is performed. The epidemic proportions of HIV infection and the large
behavior of hemangiopericytoma is difficult to predict, number of Kaposi’s sarcomas diagnosed in extraskeletal
but the prognosis seems to be more favorable than that sites, bone involvement in this far from rare disease
of high-grade angiosarcoma. The tumor frequently is relatively uncommon. Our personal experience is
recurs locally, and distant metastases may occur. limited to several cases of both classic and HIV-related
Kaposi’s sarcomas involving bone. The peak age inci-
dence and typical sites of skeletal involvement for
Kaposi’s Sarcoma classic (non–HIV-related) Kaposi’s sarcoma are shown in
Kaposi’s sarcoma occurs in four clinical forms: chronic, Figure 13-70.
lymphadenopathic, transplantation associated, and HIV
related.226,210 Radiographic Imaging
The chronic or classic form primarily affects men
older than age 50 years. It is most frequent in the On radiographs, Kaposi’s sarcoma presents as a destruc-
Eastern European/Mediterranean region (Italy) and tive lytic focus in bone. More advanced lesions show
Central Africa.211,226 This form of Kaposi’s sarcoma is complete cortical disruption and extension into soft
associated with a second malignancy, usually of the lym- tissue. Pathologic fracture can be present. The nature of
phoreticular or hematopoietic system.234 This form of the lesion can be suspected on radiographs if it occurs in
Kaposi’s sarcoma usually presents with skin involvement the appropriate clinical setting; otherwise, the radio-
of distal parts of the lower extremities. The lesions slowly graphic features are not specific. The involvement of
progress proximally, coalesce, and may even ulcerate. metatarsal bones by direct extension in a classic (non–
The lymphadenopathic form predominantly involves HIV-related) Kaposi’s sarcoma is shown in Figure 13-71.
young African children, who have cervical, inguinal, or Figure 13-72 depicts a destructive lytic lesion of the
mediastinal lymphadenopathy.215,223,228,229,232 The course rib in an HIV-positive man with disseminated Kaposi’s
of the disease is aggressive, with early involvement of sarcoma.
extranodal organs.
Transplantation-associated Kaposi’s sarcoma affects Microscopic Findings
fewer than 0.5% of patients who receive renal trans-
plants, but it has been reported in association with There is no difference in microscopic appearance among
other organ transplants and with immunosuppressive the clinical forms of Kaposi’s sarcoma. The early changes
therapy.216,217,220,221,224,230,231 The clinical course of this referred to as the patch and the plaque phases have not
form of Kaposi’s sarcoma is more aggressive than that of been described in bone. All lesions reported in bone are
the classic form. The disease usually develops in the diagnosed when the lesion is a well-established nodule
retroperitoneal lymph nodes or in some instances at with bone destruction sufficient to cause symptoms. The
the site of previous surgery. The fatal outcome is usually lesion is composed of intersecting arcs of bland spindle
due to widespread involvement of the gastrointestinal cells similar to the pattern seen in well-differentiated
tract. fibrosarcoma. Multiple slitlike spaces separate spindle
HIV-related Kaposi’s sarcoma affects young patients cells and occasionally contain intracytoplasmic erythro-
and may clinically manifest in any location.212,235 The cytes (Fig. 13-73). Better-developed vascular channels
disease has a tendency to involve deep lymph nodes and with open spaces are present at the periphery of the
the gastrointestinal tract in addition to the skin. lesion. Features of hemorrhage with hemosiderin depos-
The identification of a novel type of herpesvirus its and scattered inflammatory cells are present. Intracy-
(HHV-8) as a causative agent for Kaposi’s sarcoma made toplasmic and extracytoplasmic, diastase-resistant hyaline
possible the targeting of its viral antigens as diagnostic globules positive for periodic acid–Schiff and measuring
markers for this condition.4,6,16,214 The latency-associated 1 to 7 µm are present. They are very characteristic of
nuclear antigen (LANA1) that mediates the anchoring of Kaposi’s sarcoma and most likely represent residues of
the viral DNA to the host protein is upregulated in extravasated degenerated erythrocytes. The lesion has an

ERRNVPHGLFRVRUJ
13  Vascular Lesions 979

Above 50
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 13-70  ■  Kaposi’s sarcoma. Peak age incidence and most frequent site of skeletal involvement in classic (non–HIV-related)
Kaposi’s sarcoma are indicated by a solid black arrow.

overall bland appearance and minimal pleomorphism. Treatment and Behavior


Rare mitotic figures may be present.
Kaposi’s sarcoma cells typically express a full roster The behavior of Kaposi’s sarcoma varies in its different
of endothelial markers. CD31, CD34, and ERG are clinical forms. The mortality rate for the classic form
expressed in both the spindle and endothelial cell com- ranges from 10% to 20% after a prolonged (approxi-
ponents of the lesion (Fig. 13-74). Kaposi’s sarcoma cells mately 10-year) indolent clinical course. The mortality
also express VEGFR-3 and podoplanin (D2-40), which rate of the HIV-related form is much higher, within the
are generally considered as markers of lymphatic endo- range of 40%, and is affected by additional risk factors
thelial cells. The HHV-8 (LANA1) antigen shows strong such as opportunistic infection and secondary malignancy.
nuclear positivity in virtually all cases of Kaposi’s sarcoma Treatment of these lesions may involve curettage, en bloc
and serves as a useful marker differentiating this tumor excision, and internal fixation of pathologic fractures.
from other spindle-cell neoplastic lesions, including Radiotherapy is also useful to control the disease locally.
spindle- cell variants of angiosarcoma (Fig. 13-74).

Differential Diagnosis REACTIVE VASCULAR LESIONS


The separation from other forms of vascular sarcoma is Bacillary Angiomatosis of Bone
based on the distinctive microscopic appearance and
invariable association with extraskeletal disease. For Bacillary angiomatosis of bone occurs in immunocom-
example, we have seen rib lesions of HIV-related Kaposi’s promised patients, particularly those infected with
sarcoma in a patient who already had pathologically HIV.239,240,242,248,249 The development of lesions is linked
documented skin and gastrointestinal lesions. Direct to gram-negative rickettsial organisms of Rochalimaea
extension from overlying skin and involvement of soft (Bartonella) species (predominantly Bartonella hense-
tissue in the classic form facilitate the diagnosis of this lae).238,244 In some cases, Bartonella quintana, known as the
type of bone lesion. agent of cat-scratch fever, was also isolated.245,247 These

ERRNVPHGLFRVRUJ
980 13  Vascular Lesions

FIGURE 13-71  ■  Classic (non–HIV-related) Kaposi’s sarcoma involving bone: radiographic features. Multiple lytic lesions involving
first and second metatarsal bones in elderly man with long history of Kaposi’s sarcoma involving overlying skin and contiguous
soft tissue.

organisms have been isolated from lesions of skin, periph- capillary or arteriole size, lined by a single layer of plump
eral blood, and bone. The cutaneous lesions appear as endothelial cells (Fig. 13-75). Multiple concentric layers
protuberant, nonerythematous, firm, dry lesions with a of proliferating endothelial cells that form cuffs around
collar of hyperkeratotic skin. They can also form larger luminal spaces can be present focally. The nuclei of
lobulated vascularized masses. The bone involvement can the endothelial cells are enlarged and may show small
be associated with the skin lesions, but it can also be the nucleoli. Mitotic figures are present, but atypical mitoses
presenting sign of the disease.241,243,246,248 It must be men- are absent. The vessels can have a branching pattern
tioned that bacillary angiomatosis is extremely rare in focally. The intervening hypercellular stromal tissue
bone, and only individual cases have been reported in the shows an inflammatory cell infiltrate. In general, the
literature. This infectious, nonneoplastic process can microscopic features mimic those of low-grade vascular
mimic a low-grade vascular endothelial bone tumor both endothelial neoplasms. The infectious agent can some-
radiographically and histologically. times be identified by silver stains (e.g., Warthin-Starry)
On radiographs, bacillary angiomatosis of bone pres- and confirmed by bacteriologic cultures of peripheral
ents as a lytic defect that may disrupt the cortex and blood and fresh lesional tissue. The most sensitive tech-
extend into the soft tissue. Pain and tenderness in soft niques for identifying Bartonella species in biopsy mate-
tissue areas are usually the presenting symptoms. The rial are based on the polymerase chain reaction, which
radiographic findings and clinical symptoms are sugges- can be performed on DNA extracted from fresh or
tive of acute osteomyelitis. Microscopically, the lesion is paraffin-embedded tissues.238,245,247
composed of newly formed, well-developed vessels of Text continued on p. 985

ERRNVPHGLFRVRUJ
13  Vascular Lesions 981

B
FIGURE 13-72  ■  HIV-related Kaposi’s sarcoma involving bone: microscopic features. A, Kaposi’s sarcoma involving rib in HIV-positive
38-year-old man. Skin and gastrointestinal tract lesions were also present. Ill-defined destructive lesion is present with eccentric
expansion and periosteal new bone formation. B, Computed tomogram of abdomen in patient shown in A reveals destructive lesion
in lower right rib.

ERRNVPHGLFRVRUJ
982 13  Vascular Lesions

A B

C D
FIGURE 13-73  ■  Kaposi’s sarcoma in bone: microscopic features. A, Medium power photomicrograph shows arrays of bland spindle
cells. Narrow slitlike vascular spaces are seen. B, Higher magnification shows details of endothelial-lined vascular slits and bland
nuclei in surrounding spindle cells. C, Medium power photomicrograph shows proliferation of spindle cells. D, Higher magnification
of C shows nuclei of spindle cells with some atypia and slitlike vascular spaces with red blood cells. (A and C, ×100; B and C, ×200)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 983

A B

C D
FIGURE 13-74  ■  Kaposi’s sarcoma in bone: microscopic features. A, Low power photomicrograph shows arrays of bland spindle cells
and an occasional better-developed vascular structure. B, Strong nuclear positivity for HHV-8 LANA1 antigen in the majority of nuclei
of Kaposi’s sarcoma cells. C, Strong nuclear positivity of Kaposi’s sarcoma cells for ERG. D, Strong cytoplasmic expression of CD34
in Kaposi’s sarcoma (A-D, ×100) (A, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
984 13  Vascular Lesions

B
FIGURE 13-75  ■  Bacillary angiomatosis: microscopic features. A, Lytic lesion of maxillary bone is composed of proliferating vessels
with plump endothelial cell. B, Proliferating endothelial cells form cuffs obliterating vascular lumina. Note prominent inflammatory
infiltrate. Inset (top), Rickettsial organism documented by Warthin-Starry stain. Inset (bottom), Vessels with proliferating endothelial
cells forming concentric layers and obliterating vascular lumen. (Courtesy J. G. Batsakis, Houston.)

ERRNVPHGLFRVRUJ
13  Vascular Lesions 985

REFERENCES 23. Chen KT: Extravascular papillary endothelial hyperplasia. J Surg


Oncol 36:52–54, 1987.
1. Adams RH, Alitalo K: Molecular regulation of angiogenesis and 24. Chi JH, Manley GT, Chou D: Pregnancy-related vertebral hem-
lymphangiogenesis. Nat Rev Mol Cell Biol 8:464–478, 2007. angioma. Case report, review of the literature, and management
2. Burgdorf WH, Mukai K, Rosai J: Immunohistochemical identifi- algorithm. Neurosurg Focus 19(3):E7, 2005.
cation of factor VIII-related antigen in endothelial cells of cutane- 25. Clearkin KP, Enzinger FM: Intravascular papillary endothelial
ous lesions of alleged vascular nature. Am J Clin Pathol 75:167–171, hyperplasia. Arch Pathol Lab Med 100:441–444, 1976.
1981. 26. Cone RO, Hudkins P, Nguyen V, et al: Histiocytoid hemangioma
3. Carmeliet P, Jain RK: Molecular mechanisms and clinical applica- of bone: a benign lesion which may mimic angiosarcoma. Skeletal
tions of angiogenesis. Nature 473:298–307, 2011. Radiol 10:165–169, 1983.
4. Cheuk W, Wong KO, Wong CS, et al: Immunostaining for 27. Curtin HD, Jensen JE, Barnes L, Jr, et al: “Ossifying” hemangio-
human herpesvirus 8 latent nuclear antigen-1 helps distinguish mas of the temporal bone: evaluation with CT. Radiology 164:831–
Kaposi sarcoma from its mimickers. Am J Clin Path 121:335–342, 835, 1987.
2004. 28. Dorfman HD, Steiner GC, Jaffe HL: Vascular tumors of bone.
5. Folpe AL, Veikkola T, Valtola R, et al: Vascular endothelial Hum Pathol 2:349–376, 1971.
growth factor receptor-3 (VEGFR-3): a marker of vascular tumors 29. Eusebi V, Fanti PA, Fedeli F, et al: Masson’s intravascular vegetant
with presumed lymphatic differentiation, including Kaposi’s hemangioendothelioma. Tumori 66:489–498, 1980.
sarcoma, kaposiform and Dabska-type hemangioendotheliomas, 30. Evans HL, Raymond AK, Ayala AG: Vascular tumors of bone: a
and a subset of angiosarcomas. Mod Pathol 13:180–185, 2000. study of 17 cases other than ordinary hemangioma, with an evalu-
6. Ghandi M, Greenblatt RM: Human herpesvirus 8, Karposi’s ation of the relationship of hemangioendothelioma of bone to
sarcoma, and associated conditions. Clin Lab Med 22:883–910, epithelioid hemangioma, epithelioid hemangioendothelioma, and
2002. high-grade angiosarcoma. Hum Pathol 34(7):680–689, 2003.
7. Hong YK, Harvey N, Noh YH, et al: Prox1 is a master control 31. Fetsch JF, Weiss SW: Observations concerning the pathogenesis
gene in the program specifying lymphatic endothelial cell fate. of epithelioid hemangioma (angiolymphoid hyperplasia). Mod
Dev Dyn 225:351–357, 2002. Pathol 4:449–455, 1991.
8. Johnson KE, Wilgus TA: Vascular endothelial growth factor and 32. Floris G, Deraedt K, Samson I, et al: Epithelioid hemangioma of
angiogenesis in the regulation of cutaneous wound repair. Adv bone: a potentially metastasizing tumor? Int J Surg Pathol 14(1):9–
Wound Care 3:647–661, 2014. 15, 2006.
9. Karpanen T, Alitalo K: Molecular biology and pathology of lym- 33. Hashimoto H, Daimaru Y, Enjoji M: Intravascular papillary endo-
phangiogenesis. Annu Rev Pathol 3:367–397, 2008. thelial hyperplasia: a clinicopathologic study of 91 cases. Am J
10. Meadows SM, Myers CT, Krieg PA: Regulation of endothelial cell Dermatopathol 5:539–546, 1983.
development by ETS transcription factors. Semin Cell Dev Biol 34. Henry PG, Burnett JW: Angiolymphoid hyperplasia with eosino-
22:976–984, 2011. philia. Arch Dermatol 114:1168–1172, 1978.
11. Miettinen M, Lindenmayer AE, Chaubal A: Endothelial cell 35. Izukawa D, Lach B, Benoit B: Intravascular papillary endothelial
markers CD31, CD34, and BNH9, antibody to H- and hyperplasia in an intracranial cavernous hemangioma. Neurosur-
Y-antigens—evaluation of their specificity and sensitivity in the gery 21:939–941, 1987.
diagnosis of vascular tumors and comparison with von Willebrand 36. James JN: Cavernous hemangioma of the mandible. Proc R Soc
factor. Mod Pathol 7:82–90, 1994. Med 57:797–799, 1964.
12. Pan Y, Wang WD, Yago T: Transcriptional regulations of podo- 37. Keel SB, Rosenberg AE: Hemorrhagic epithelioid and spindle cell
planin expression by Prox1 in lymphatic endothelial cells. Micro- hemangioma: a newly recognized, unique vascular tumor of bone.
vasc Res 94:96–102, 2014. Cancer 85(9):1966–1972, 1999.
13. Patel RM, Goldblum JR, His ED: Immunohistochemical detec- 38. Kimura T, Yoshimura S, Ishikawa E: Unusual granulation com-
tion of human herpes virus-8 latent nuclear antigen-A is useful in bined with hyperplastic change of lymphatic tissue. Trans Soc
the diagnosis of Kaposi sarcoma. Mod Pathol 17:456–460, 2004. Pathol Jpn 37:179, 1948.
14. Pries AR, Secomb TW: Making microvascular networks work: 39. Kung ITM, Gibson JB, Bannatyne PM: Kimura’s disease: a clini-
angiogenesis, remodeling, and pruning. Physiology 29:446–455, copathological study of 21 cases and its distinction from angio-
2014. lymphoid hyperplasia with eosinophilia. Pathology 16:39–44, 1984.
15. Pusztaszeri MP, Seelentag W, Bosman FT: Immunohistchemical 40. Kuo T, Gomez LG: Papillary endothelial proliferation in cystic
expression of endothelial markers CD31, CD34, von Willebrand lymphangiomas: a lymphatic vessel counterpart of Masson’s veg-
factor, and Fli-1 in normal human tissues. J Histochem Cytochem etant intravascular hemangioendothelioma. Arch Pathol Lab Med
54:385–395, 2006. 103:306–308, 1979.
16. Robin YM, Guillou L, Michels JJ, et al: Human herpesvirus 8 41. Kuo T, Sayers CP, Rosai J: Masson’s “vegetant intravascular he-
immunostaining: a sensitive and specific method for diagnosing mangioendothelioma”: lesion often mistaken for angiosarcoma—
Kaposi sarcoma in paraffin-embedded sections. Am J Clin Pathol study of 17 cases located in the skin and soft tissues. Cancer
121:330–334, 2004. 38:1227–1236, 1976.
17. Tammela T, Alitalo K: Lymphaniogenesis: molecular mechanisms 42. Kuo TT, Shih L-Y, Chan H-L: Kimura’s disease: involvement of
and future promise. Cell 140:460–476, 2010. regional lymph nodes and distinction from angiolymphoid hyper-
18. van de Rijn M, Rouse R: CD34: a review. Appl Immunohistochem plasia with eosinophilia. Am J Surg Pathol 12:843–854, 1988.
2:71–80, 1994. 43. Ling S, Rafii M, Klein M: Epithelioid hemangioma of bone.
Benign Vascular Lesions Skeletal Radiol 30(4):226–229, 2001.
44. Lund BA, Dahlin DC: Hemangiomas of the mandible and maxilla.
Hemangioma J Oral Surg 22:234–242, 1964.
19. Acosta FL, Jr, Dowd CF, Chin C, et al: Current treatment strate- 45. Masson P: Hemangioendotheliome vegetant intravasculaire. Bull
gies and outcomes in the management of symptomatic vertebral Soc Anat 93:517–523, 1923.
hemangiomas. Neurosurgery 58(2):287–295, 2006. 46. O’Connell JX, Kattapuram SV, Mankin HJ, et al: Epithelioid
20. Amerigo J, Berry CL: Intravascular papillary endothelial hyper- hemangioma of bone. Am J Surg Pathol 17:610–617, 1993.
plasia in the skin and subcutaneous tissue. Virchows Arch [A] 47. Ose D, Vollmer R, Shelburne J, et al: Histiocytoid hemangioma
387:81–90, 1980. of the skin and scapula: a case report with electron microscopy
21. Angervall L, Kindblom LG, Karlsson K, et al: Atypical heman- and immunohistochemistry. Cancer 51:1656–1662, 1983.
gioendothelioma of venous origin: a clinicopathologic, angio- 48. Rimondini P, Ferri GR: Hemangiomas of the nasal bone. Otori-
graphic, immunohistochemical, and ultrastructural study of two nolaring Ital 37:210–225, 1969.
endothelial tumors within the concept of histiocytoid hemangi- 49. Rosai J, Ackerman LR: Intravenous atypical vascular proliferation:
oma. Am J Surg Pathol 9:504–516, 1985. a cutaneous lesion simulating a malignant blood vessel tumor.
22. Barr RJ, Graham JH, Sherwin LA: Intravascular papillary endo- Arch Dermatol 109:714–717, 1974.
thelial hyperplasia: a benign lesion mimicking angiosarcoma. Arch 50. Rosai J, Gold J, Landy R: The histiocytoid hemangiomas: a unify-
Dermatol 114:723–726, 1978. ing concept embracing several previously described entities of

ERRNVPHGLFRVRUJ
986 13  Vascular Lesions

skin, soft tissue, large vessels, bone, and heart. Hum Pathol 10:707– 80. Hambach R, Pujman J, Maly V: Massive osteolysis due to heman-
730, 1979. giomatosis: report of a case of Gorham’s disease with autopsy.
51. Siegelman SS, Frankel TN, Lewin ML: Hemangioma of the nasal Radiology 71:43–47, 1958.
bone: report of a case. Arch Otolaryngol 88:269–272, 1968. 81. Ishida T, Dorfman HD, Steiner GC, et al: Cystic angiomatosis of
52. Stewart M, Smoller BR: Multiple lesions of intavascular papillary bone with sclerotic changes mimicking osteoblastic metastases.
endothelial hyperplasia (Masson’s lesion). Arch Pathol Lab Med Skeletal Radiol 23(4):247–252, 1994.
118:315–316, 1994. 82. Karlin CA, Brower AC: Multiple primary hemangiomas of bone.
53. Suneja R, Davies AM, Deshmukh NS, et al: Metachronous haem- AJR Am J Roentgenol 129:162–164, 1977.
orrhagic epithelioid and spindle cell haemangioma of bone. Skel- 83. Moseley JE, Starobin SG: Cystic angiomatosis of bone, manifesta-
etal Radiol 31(8):475–478, 2002. tion of a hamartomatous disease entity. AJR Am J Roentgenol
54. Topazian RG: Central hemangioma of the mandible: report of a 91:1114–1120, 1964.
case. Oral Surg 18:1–6, 1964. 84. Ritchie G, Zeier FG: Hemangiomatosis of the skeleton and
55. Walker EA, Jr, McHenry LC: Primary hemangioma of the spleen. J Bone Joint Surg 38A:115–122, 1956.
zygoma. Arch Otolaryngol 81:199–203, 1965. 85. Schajowicz F, Aiello CL, Francone MV, et al: Cystic angiomatosis
56. Weber FL, Babel J: Intravascular papillary endothelial hyperplasia (hamartomatous haemolymphangiomatosis) of bone: a clinico-
of the orbit. Br J Ophthalmol 65:18–22, 1981. pathological study of three cases. J Bone Joint Surg Br 60:100–106,
57. Wenger DE, Wold LE: Benign vascular lesions of bone: 1978.
radiologic and pathologic features. Skeletal Radiol 29(2):63–74, 86. Spjut HJ, Lindbom A: Skeletal angiomatosis: report of two cases.
2000. Acta Pathol Microbiol Scand 55:49–58, 1962.
58. Wells GC, Whimster IW: Subcutaneous angiolymphoid hyper- 87. Takahashi A, Ogawa C, Kanazawa T, et al: Remission induced by
plasia with eosinophilia. Br J Dermatol 81:1–14, 1969. interferon alfa in a patient with massive osteolysis and extension
59. Wold LE, Swee RG, Sim FH: Vascular lesions of bone. Pathol of lymph-hemangiomatosis: a severe case of Gorham-Stout syn-
Annu 20:101–137, 1985. drome. J Pediatr Surg 40(3):E47–E50, 2005.
88. Waldron RL, II, Zeller JA: Diffuse skeletal hemangiomatosis with
Lymphangioma and Lymphangiomatosis visceral involvement. J Can Assoc Radiol 20:119–123, 1969.
60. Bickel WH, Broders AC: Primary lymphangioma of the ilium. 89. Winterberger AR: Radiographic diagnosis of lymphangiomatosis
J Bone Joint Surg 29:517–522, 1947. of bone. Radiology 102:321–324, 1972.
61. Bullough PG, Goodfellow JW: Solitary lymphangioma of bone: a 90. Yamamoto T, Iwasaki Y, Kurosaka M, et al: Angiosarcoma arising
case report. J Bone Joint Surg 58A:418–419, 1976. from skeletal hemangiomatosis in an atomic bomb survivor. J Clin
62. Ducharme JC, Belanger R, Simard P, et al: Chylothorax, chylo- Pathol 54(9):716–717, 2001.
pericardium with multiple lymphangioma of bone. J Pediatr Surg
17:365–367, 1982. Glomus Tumor
63. Goldstein MR, Benchimol A, Cornell W, et al: Chylopericardium 91. Anagnostou GD, Papademetriou DG, Toumazani MN: Subcuta-
with multiple lymphangioma of bone. N Engl J Med 280:1034– neous glomus tumors. Surg Gynecol Obstet 136:945–950, 1973.
1037, 1969. 92. Apfelberg DB, Teasley JL: Unusual locations and manifestations
64. Gomez CS, Calonje E, Ferrar DW, et al: Lymphangiomatosis of of glomus tumors (glomangiomas). Am J Surg 116:62–64, 1968.
the limbs. Clinicopathologic analysis of a series with a good prog- 93. Bahk WJ, Mirra JM, Anders KH: Intraosseous glomus tumor of
nosis. Am J Surg Pathol 19(2):125–133, 1995. the fibula. Skeletal Radiol 29(12):708–712, 2000.
65. Harris R, Prandoni AG: Generalized primary lymphangiomas of 94. Bjorkengren AG, Resnick D, Haghighi P, et al: Intraosseous
bone. Ann Intern Med 33:1302–1313, 1950. glomus tumor: report of a case and review of the literature. AJR
66. Jumbelic M, Feuerstein I, Dorfman HD: Solitary intraosseous Am J Roentgenol 147:739–741, 1986.
lymphangioma. J Bone Joint Surg 66A:1479–1481, 1984. 95. Camirand P, Giroux JM: Subungual glomus tumor: radiological
67. Najman E, Fabecic-Sabadi V, Temmer B: Lymphangioma in the manifestations. Arch Dermatol 102:677–679, 1970.
inguinal region with cystic lymphangiomatosis of bone. J Pediatr 96. Chan CW: Intraosseous glomus tumor: a case report. J Hand Surg
71:561–566, 1967. 6A:368–369, 1981.
68. Nixon GW: Lymphangiomatosis of bone demonstrated by lym- 97. Freier DT, Lindenauer SM: Subcutaneous glomus tumor. Am J
phangiography. Am J Roentgen Rad Ther Nucl Med 110:582–586, Surg 120:359–362, 1970.
1970. 98. Gonzalez-Llanos F, Lopez-Barea F, Isla A, et al: Periosteal glomus
69. Rosenquist CJ, Wolfe DC: Lymphangioma of bone. J Bone Joint tumor of the femur: a case report. Clin Orthop Relat Res 380:199–
Surg 50A:158–162, 1968. 203, 2000.
70. Steiner GM, Farman J, Lawson JP: Lymphangiomatosis of bone. 99. Harris M: Ultrastructure of a glomus tumor. J Clin Pathol 24:520–
Radiology 93:1093–1098, 1969. 523, 1971.
100. Harris WR: Erosion of bone produced by glomus tumor. Can Med
Angiomatoses Assoc J 70:684–685, 1954.
71. Boyle WJ: Cystic angiomatosis of bone. J Bone Joint Surg Br 101. Hermann G, Klein MJ, Springfield D, et al: Glomus tumor of the
54:626–636, 1972. thigh: confluent with the periosteum of the femur. Skeletal Radiol
72. Campbell J, Almond HG, Johnson R: Massive osteolysis of the 34(2):116–120, 2005.
humerus with spontaneous recovery. J Bone Joint Surg Br 57:238– 102. Ho KL, Pak MS: Glomus tumor of the coccygeal region: case
240, 1975. report. J Bone Joint Surg 62A:141–142, 1980.
73. Cannon SR: Massive osteolysis: a review of seven cases. J Bone 103. Mackenzie DH: Intraosseous glomus tumors: report of two cases.
Joint Surg Br 68:24–28, 1986. J Bone Joint Surg Br 44:648–651, 1962.
74. Choma ND, Biscotti CV, Bauer TW, et al: Gorham’s syndrome: 104. Maley ED, MacDonald CJ: Bilateral subungual glomus tumors.
a case report and review of the literature. Am J Med 83:1151– Plast Reconstr Surg 55:488–489, 1975.
1156, 1987. 105. Masson PL: Neuromyoarterial des regions tactiles et ses tumeurs.
75. Fornasier VL: Haemangiomatosis with massive osteolysis (angio- Lyon Chir 21:257–280, 1924.
matosis). J Bone Joint Surg Br 52:444–451, 1970. 106. Mullis WF, Rosato FE, Rosato EF, et al: The glomus tumor. Surg
76. Gorham LW, Schultz AW, Maxon FC: Disappearing bones: a rare Gynecol Obstet 135:705–707, 1972.
form of osteolysis: report of two cases, one with autopsy findings. 107. Murad TM, Von Haam E, Murthy MS: The ultrastructure of
Am J Med 17:674–682, 1954. hemangiopericytoma and a glomus tumor. Cancer 22:1239–1249,
77. Gorham LW, Stout AP: Massive osteolysis (acute spontaneous 1968.
absorption of bone, phantom bone, disappearing bone): relation 108. Rozmaryn LM, Sadler AH, Dorfman HD: Intraosseous glomus
to hemangiomatosis. J Bone Joint Surg 37A:985–1004, 1955. tumor in the ulna: a case report. Clin Orthop 220:126–129, 1987.
78. Gutierrez RM, Spjut HJ: Skeletal angiomatosis: report of three 109. Shugart RR, Soule EH, Johnson EW: Glomus tumor. Surg Gynecol
cases and review of the literature. Clin Orthop 85:82–97, 1972. Obstet 117:334–340, 1963.
79. Halliday DR, Dahlin DC, Pugh DG, et al: Massive osteolysis and 110. Siegel MW: Intraosseous glomus tumor: a case report. Am J
angiomatosis. Radiology 82:637–644, 1964. Orthop 9:68–69, 1967.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 987

111. Sugiura I: Intra-osseous glomus tumor: a case report. J Bone Joint 135. Fletcher CD, Beham A, Schmid C: Spindle cell haemangioendo-
Surg Br 58:245–247, 1976. thelioma: a clinicopathological and immunohistochemical study
indicative of a non-neoplastic lesion. Histopathology 18:291–301,
Malignant Vascular Tumors 1991.
136. Imayama S, Murakamai Y, Hashimoto H, et al: Spindle cell
Epithelioid Hemangioendothelioma hemangioendothelioma exhibits the ultrastructural features of
112. Azumi N, Churg A: Intravascular and sclerosing bronchioloalveo- reactive vascular proliferation rather than of angiosarcoma. Am J
lar tumor: a pulmonary sarcoma of probable vascular origin. Am Clin Pathol 97:279–287, 1992.
J Surg Pathol 5:587–596, 1981. 137. Lai FM, Allen PW, Yuen PM, et al: Locally metastasizing vascular
113. Bhagavan BS, Dorfman HD, Murthy MS, et al: Intravascular tumor: spindle cell, epithelioid, or unclassified hemangioendothe-
bronchiolo-alveolar tumor (IVBAT): a low grade sclerosing epi- lioma? Am J Clin Pathol 96:660–663, 1991.
thelioid angiosarcoma of lung. Am J Surg Pathol 6:41–52, 1982. 138. Ma J, Shi QL, Jiang SJ, et al: Primary kaposiform heangioendo-
114. Campanacci M, Boriani S, Giunti A: Hemangioendothelioma of thelioma of a long bone: two cases in unusual locations with
bone: a study of 29 cases. Cancer 46:804–814, 1980. long-term follow up. Pathol Int 61:382–386, 2011.
115. Errani C, Zhang L, Sung YS, et al: A novel WWTR1-CAMTA1 139. Requena L, Kutzner H: Hemangioendothelioma. Semin Diagn
gene fusion is a consistent abnormality in epithelioid hemangio- Pathol 30:29–44, 2013.
endothelioma of different anatomic sites. Genes Chromosomes 140. Terashi H, Itami S, Kurata S, et al: Spindle cell hemangioendo-
Cancer 50:644–653, 2011. thelioma: report of three cases. J Dermatol 18:104–111, 1991.
116. Fienberg R, Baehr FH: Hemangioendothelioma of tibia with 141. Tsang WY, Chan JK, Fletcher CD: Recently characterized vascu-
metastasis to the popliteal artery. Arch Pathol 31:811–818, 1941. lar tumors of skin and soft tissues. Histopathology 19:489–501,
117. Hartmann WH, Stewart FW: Hemangioendothelioma of bone: 1991.
unusual tumor characterized by indolent course. Cancer 15:846– 142. Weiss SW, Enzinger FM: Spindle cell hemangioendothelioma: a
854, 1962. low-grade angiosarcoma resembling a cavernous hemangioma and
118. Hisaoka M, Okamoto S, Aoki T, et al: Spinal epithelioid heman- Kaposi’s sarcoma. Am J Surg Pathol 10:521–530, 1986.
gioendothelioma with epithelioid angiosarcomatous areas. Skeletal 143. Winter A, Siu A, Jamshidi A, et al: Spindle cell hemangioendo-
Radiol 34(11):745–749, 2005. thelioma of the sacrum: case report. J Neurosurg Spine 21:275–278,
119. Ishak KG, Sesterhenn IA, Goodman MZD, et al: Epithelioid 2014.
hemangioendothelioma of the liver: a clinicopathologic and
follow-up study of 32 cases. Hum Pathol 15:839–852, 1984. Angiosarcoma
120. Kleer CG, Unni KK, McLeod RA: Epithelioid hemangioendo- 144. Abdelwahab IF, Klein MJ, Hermann G, et al: Angiosarcomas
thelioma of bone. Am J Surg Pathol 20(11):1301–1311, 1996. associated with bone infarcts. Skeletal Radiol 27(10):546–551,
121. Maruyama N, Kumagai Y, Ishida Y, et al: Epithelioid haemangio- 1998.
endothelioma of the bone tissue. Virchows Arch [A] 407:159–165, 145. Alles JU, Bosslet K: Immunocytochemistry of angiosarcomas: a
1985. study of 19 cases with especial emphasis on the applicability of
122. Mirra JM, Kameda N: Myxoid angioblastomatosis of bones: a case endothelial cell specific markers to routinely prepared tissues. Am
report of a rare, multifocal entity with light, ultramicroscopic, and J Clin Pathol 89:463–471, 1988.
immunopathologic correlation. Am J Surg Pathol 9:450–458, 146. Alrenga DP: Primary angiosarcoma of the liver: review article. Int
1985. Surg 60:198–203, 1975.
123. Morgenstern P, Westing SW: Malignant hemangioendothelioma 147. Bardwil JM, Mocega EE, Butler JJ, et al: Angiosarcomas of the
of bone: fourteen-year follow-up in a case treated with radiation head and neck region. Am J Surg 116:548–553, 1968.
alone. Cancer 23:221–224, 1969. 148. Barnett WO, Hardy JD, Hendrix JH: Lymphangiosarcoma fol-
124. O’Connell JX, Nielsen GP, Rosenberg AE: Epithelioid vascular lowing postmastectomy lymphedema. Ann Surg 169:960–968,
tumors of bone: a review and proposal of a classification scheme. 1969.
Adv Anat Pathol 8(2):74–82, 2001. 149. Bowers WF, Shear EW, LeGolvan PC: Lymphangiosarcoma in
125. Reed RJ: Malignant myxoid angioblastoma of bone. Am J Surg post-mastectomy lymphedematous arm. Am J Surg 90:682, 1955.
Pathol 6:159–163, 1982. 150. Cerilli LA, Fechner RE: Angiosarcoma arising in a bone infarct.
126. Rosenthal DI, Treat ME, Mankin HJ, et al: Treatment of epithe- Ann Diagn Pathol 3(6):370–373, 1999.
lioid hemangioendothelioma of bone using novel combined 151. Chen KT, Gilbert EF: Angiosarcoma complicating generalized
approach. Skeletal Radiol 30(4):219–222, 2001. lymphangiectasia. Arch Pathol Lab Med 103:86–88, 1979.
127. Shah ZK, Peh WC, Shek TW, et al: Hemangioendothelioma with 152. Chen KT, Hoffman KD, Hendricks EJ: Angiosarcoma following
an epithelioid phenotype arising in hemangioma of fibula. Skeletal therapeutic irradiation. Cancer 44:2044–2048, 1979.
Radiol 34(11):750–754, 2005. 153. Chow RW, Wilson CB, Olsen ER: Angiosarcoma of the skull:
128. Steiner GC, Dorfman HD: Ultrastructure of hemangioendothe- report of a case and review of the literature. Cancer 25:902–906,
lial sarcoma of bone. Cancer 29:122–135, 1972. 1970.
129. Tanas MR, Sboner A, Oliveira AM, et al: Identification of a 154. Cohen PR, Rapini RP, Farhood AI: Expression of the human
disease-defining gene fusion in epithelioid hemangioendotheli- hematopoietic progenitor cell antigen CD34 in vascular and
oma. Cancer 3(98):1–9, 2011. spindle cell tumors. J Cutan Pathol 20:15–20, 1993.
130. Tsuneyoshi M, Dorfman HD, Bauer TW: Epithelioid hemangio- 155. Davies JD, Rees GJ, Mera SL: Angiosarcoma in irradiated post-
endothelioma of bone: a clinicopathologic, ultrastructural, and mastectomy chest wall. Histopathology 7:947–956, 1983.
immunohistochemical study. Am J Surg Pathol 10:754–764, 1986. 156. Deshpande V, Rosenberg AE, O’Connell JX, et al: Epithelioid
131. Weiss SW, Enzinger FM: Epithelioid hemangioendothelioma: a angiosarcoma of bone: a series of 10 cases. Am J Surg Pathol
vascular tumor often mistaken for a carcinoma. Cancer 50:970– 27(6):709–716, 2003.
981, 1982. 157. Devi KR, Bahuleyan CK: Lymphangiosarcoma of the lower
132. Williams SB, Butler BC, Gilkey FW, et al: Epithelioid hemangio- extremity associated with chronic lymphedema of filarial origin.
endothelioma with osteoclastlike giant cells. Arch Pathol Lab Med Ind J Cancer 14:176–178, 1977.
117(3):315–318, 1993. 158. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210,
1995.
Spindle-Cell Hemangioendothelioma 159. Dubin HV, Creehan EP, Headington JT: Lymphangiosarcoma
133. Battocchio S, Facchetti F, Brisigotti M: Spindle cell haemangio- and congenital lymphedema of the extremity. Arch Dermatol
endothelioma: further evidence against its proposed neoplastic 110:608–614, 1974.
nature. Histopathology 22:296–298, 1993. 160. Dunlop J: Malignant hemangioendothelioma of bone: case report
134. Ding J, Hashimoto H, Imayama S, et al: Spindle cell haemangio- of en bloc resection and prosthetic hip replacement. J Bone Joint
endothelioma: probably a benign vascular lesion not a low-grade Surg 59A:832–834, 1977.
angiosarcoma—clinicopathological, ultrastructural and immuno- 161. Falk H, Thomas LB, Popper H, et al: Hepatic angiosarcoma
histochemical study. Virchows Arch A Pathol Anat Histopathol associated with androgenic-anabolic steroids. Lancet 2:1120–1123,
420:77–85, 1992. 1979.

ERRNVPHGLFRVRUJ
988 13  Vascular Lesions

162. Falk S, Krishnan J, Meis JM: Primary angiosarcoma of the spleen: 188. Gunhan O, Yildiz FR, Selmanpakoglu N: Mandibular hemangio-
a clinicopathologic study of 40 cases. Am J Surg Pathol 17:959– pericytoma: report of a case and review of the literature. J Oral
970, 1993. Maxillofac Surg 53:704–707, 1995.
163. Francis KC, Lindquist HD: Lymphangiosarcoma of the lower 189. Hahn MJ, Dawson R, Esterly JA, et al: Hemangiopericytoma: an
extremity involved with chronic lymphedema. Am J Surg 10:617, ultrastructural study. Cancer 31:255–261, 1973.
1967. 190. Henn W, Wullich B, Thonnes M, et al: Recurrent t(12;19)
164. Fukuroku J, Kusuzaki K, Murata H, et al: Two cases of second- (q13;q13.3) in intracranial and extracranial hemangiopericytoma.
ary angiosarcoma arising from fibrous dysplasia. Anticancer Res Cancer Genet Cytogenet 71:151–154, 1993.
19(5C):4451–4457, 1999. 191. Hoshi M, Araki N, Naka N, et al: Bone metastasis of intracra-
165. Goette DK, Detlefs RL: Postirradiation angiosarcoma. J Am Acad nial meningeal heangiopericytoma. Int J Clin Oncol 10:208–213,
Dermatol 12:922–926, 1985. 2005.
166. Hasegawa T, Fujii Y, Seki K, et al: Epithelioid angiosarcoma of 192. Kabus D, Sidhu GS, Wieczorek RL, et al: Metastatic meningi-
bone. Hum Pathol 28(8):985–989, 1997. oma: hemangiopericytoma or angioblastic meningioma. Am J
167. Huey GR, Steham FB, Roth LM, et al: Lymphangiosarcoma of Surg Pathol 17:1144–1150, 1993.
the edematous thigh after radiation therapy for carcinoma of the 193. Klein EA: Metastatic meningioma: hemangiopericytoma or
vulva. Gynecol Oncol 20:394–401, 1985. angioblastic meningioma? Am J Surg Pathol 18:1071–1072, 1994.
168. McCarthy EF, Lietman S, Argani P, et al: Endovascular papillary 194. Legre G, Payan H, Aubert M: Osseous hemangiopericytoma. Rev
angioendothelioma (Dabska tumor) of bone. Skeletal Radiol Chir Orthop 52:551–559, 1966.
28(2):100–103, 1999. 195. McMaster MJ, Soule EH, Ivins JC: Hemangiopericytoma: a clini-
169. Mark L, Delmore F, Creech JL, Jr, et al: Clinical and morphologic copathologic study and long-term follow-up of 60 patients. Cancer
features of hepatic angiosarcoma in vinyl chloride workers. Cancer 36:2232–2244, 1975.
37(149):163, 1976. 196. Mena H, Ribas JL, Pezeshkpour GH, et al: Hemangiopericytoma
170. Merrick TA, Erlandson RA, Hajdu SI: Lymphangiosarcoma of of the central nervous system: a review of 94 cases. Hum Pathol
a congenitally lymphedematous arm. Arch Pathol 91:365–371, 22:84–91, 1991.
1971. 197. Murad TM, von Haam E, Murthy MS: The ultrastructure of a
171. Mitsuhashi T, Shimizu Y, Ban S, et al: Multicentric contigous hemangiopericytoma and a glumus tumor. Cancer 22:1239–1249,
variant of epithelioid angiosarcoma of the bone. A rare variant 1968.
showing angiotropic spread. Ann Diagn Pathol 9(1):33–37, 198. Nappi O, Ritter JH, Pettinato G, et al: Hemangiopericytoma:
2005. histological pattern or clinicopathologic entity. Semin Diagn
172. Popper H, Thomas LB, Telles NC, et al: Development of hepatic Pathol 12(3):221–232, 1995.
angiosarcoma in man induced by vinyl chloride, Thorotrast, and 199. Radley MG, McDonald JV: Meningeal hemangiopericytoma of
arsenic: comparison with cases of unknown etiology. Am J Pathol the posterior fossa and thoracic spinal epidural space: case report.
92:349–369, 1978. Neurosurgery 30:446–452, 1992.
173. Stjernvall L: Vertebral angiosarcoma: a case report. Acta Orthop 200. Rao CR, Naresh KN, Pattabhiraman V, et al: Hemangiopericy-
Scand 41:165–168, 1970. toma of bone: a case report. Ind J Cancer 31:264–267, 1994.
174. Sweterlitsch PR, Torg JS, Watts H: Malignant hemangioendothe- 201. Robinson DR, Wu YM, Kalyana-Sundaram S, et al: Identification
lioma of the cervical spine: a case report. J Bone Joint Surg 52A: of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor
805–808, 1970. by integrative sequencing. Nat Genet 45:180–187, 2013.
175. Volpe R, Mazabraud A: Hemangioendothelioma (angiosarcoma) 202. Schweizer L, Koelsche C, Sahm F, et al: Meningeal hemangio-
of bone: a distinct pathologic entity with an unpredictable course? pericytoma and solitary fibrous tumors carry the NAB2-STAT6
Cancer 49:727–736, 1982. fusion and can be diagnosed by nuclear expression of STAT6
176. Wenger DE, Wold LE: Malignant vascular lesions of bone: protein. Acta Neuropathol 125:651–668, 2013.
radiologic and pathologic features. Skeletal Radiol 29(11):619–631, 203. Sellke FW, Laszewski MJ, Robinson RA, et al: Hemangiopericy-
2000. toma of the sternum. Arch Pathol Lab Med 115:242–244, 1991.
177. Wold LE, Unni KK, Beabout JW, et al: Hemangioendothelial 204. Sreekantaiah C, Bridge JA, Rao UN, et al: Clonal chromosomal
sarcoma of bone. Am J Surg Pathol 6:59–70, 1982. abnormalities in hemangiopericytoma. Cancer Genet Cytogenet
178. Yamamoto T, Iwasaki Y, Kurosaka M, et al: Angiosarcoma arising 54:173–181, 1991.
from skeletal hemangiomatosis in an atomic bomb survivor. J Clin 205. Stout AP, Murray MR: Hemangiopericytoma: a vascular tumor
Pathol 54(9):716–717, 2001. featuring Zimmermann’s pericytes. Ann Surg 116:26–33, 1942.
206. Unni KK, Ivins JC, Beabout JW, et al: Hemangioma, hemangio-
Hemangiopericytoma pericytoma and hemangioendothelioma (angiosarcoma) of bone.
179. Anderson C, Rorabeck CH: Skeletal metastases of an intracranial Cancer 27:1403–1414, 1971.
malignant hemangiopericytoma. J Bone Joint Surg 62A:145–148, 207. Verbeke SL, Fletcher CD, Alberghini M, et al: A reappraisal of
1980. hemangiopericytoma of bone; analysis of cases reclassified as
180. Battifora H: Hemangiopericytoma: ultrastructural study of five synovial sarcoma and solitary fibrous tumor of bone. Am J Surg
cases. Cancer 31:1418–1432, 1973. Pathol 34:777–783, 2010.
181. Batsakis JG, Jacobs JB, Templeton AC: Hemangiopericytoma of 208. Warraich I, Dunn DM, Oliver JW: Solitary fibrous tumor of the
the nasal cavity: electron-optic study and clinical correlations. orbit with epithelioid features. Arch Pathol Lab Med 130(7):1039–
J Laryngol Otol 97:361–368, 1983. 1041, 2006.
182. Chin LS, Rabb CH, Hinton DR, et al: Hemangiopericytoma of 209. Zimmerman KW: Der feinere Bau der Blutcapillaren. Z Anat
the temporal bone presenting as a retroauricular mass. Neurosur- Entwicklungsg 68:29, 1923.
gery 33:728–731, 1993.
183. Chmielecki J, Crago AM, Rosenberg M, et al: Whole-exome Kaposi’s Sarcoma
sequencing identifies a recurrent NAB2-STAT6 fusion in solitary 210. Biviji AA, Paiement GD: Steinbach: Musculoskeletal manifesta-
fibrous tumors. Nat Genet 45:131–132, 2013. tions of human immunodeficiency virus infection. J Am Acad
184. Fathie K: Hemangiopericytoma of the thoracic spine: case report. Orthop Surg 10(5):312–320, 2002.
J Neurosurg 32:371–374, 1970. 211. Chor PJ, Santa Cruz DJ: Kaposi’s sarcoma: a clinicopathologic
185. Finn WG, Goolsby CL, Rao MS: DNA flow cytometric analysis review and differential diagnosis. J Cutan Pathol 19:6–20, 1992.
of hemangiopericytoma. Am J Clin Pathol 101:181–185, 1994. 212. Di Bella S, Capone A, Olearo F, et al: Vertebral lesions from
186. Folpe AL, Fanburg-Smith JC, Billings SD, et al: Most AIDS-related Kaposi’s sarcoma. Curr HIV Res 9:270–275, 2011.
osteomalacia-associated mesenchymal tumors are a single histo- 213. Gorham LW: Kaposi’s sarcoma involving bone with particular
pathologic entity: an analysis of 32 cases and a comprehensive attention to angiomatous components of the tumor in relation to
review of the literature. Am J Surg Pathol 28(1):1–30, 2004. osteolysis. Arch Pathol 76:456–463, 1963.
187. Fuentealba C, Pinto D, Ballesteros F, et al: Oncogenic hypophos- 214. Gramolelli S, Schulz TF: The role of Kaposi sarcoma-associated
phatemic osteomalacia associated with a nasal hemangiopericy- herpesvirus in the pathogenesis of Kaposi sarcoma. J Pathol 235:
toma. J Clin Rheumatol 9(6):373–379, 2003. 368–380, 2014.

ERRNVPHGLFRVRUJ
13  Vascular Lesions 989

215. Grave GF: Kaposi sarcoma in African children. Ann R Coll Surg 234. Safai B, Mike V, Giraldo G, et al: Association of Kaposi’s sarcoma
Engl 54:270–271, 1974. with second primary malignancies: possible etiopathogenic impli-
216. Hardy MA, Goldfarb P, Levine S, et al: De novo Kaposi’s sarcoma cations. Cancer 45:1472–1479, 1980.
in renal transplantation: case report and brief review. Cancer 235. Scully PA, Steinman HK, Kennedy C, et al: AIDS-related
38:144–148, 1976. Kaposi’s sarcoma displays differential expression of endothelial
217. Harwood AR, Osoba D, Hofstader SL, et al: Kaposi’s sarcoma in surface antigens. Am J Pathol 130:244–251, 1988.
recipients of renal transplants. Am J Med 67:759–765, 1979. 236. Sun Q, Tsurimoto T, Juillard F, et al: Kaposi’s sarcoma-associated
218. Horusitzky A, Cariou D, Chicheportiche V, et al: Kaposi’s sarcoma herpesvirus LANA recruits the DNA polymerase clamp loader
involving bone in a patient with AIDS. AIDS 9:206–208, 1995. to mediate efficient replication and virus persistence. PNAS
219. Isenbarger DW, Aronson NE: Lytic vertebral lesions: an unusual 111:11816–11821, 2014.
manifestation of AIDS-associated Kaposi’s sarcoma. Clin Infec Dis 237. Tehranzadeh J, Ter-Oganesyan RR, Steinbach LS: Musculoskel-
19:751–755, 1994. etal disorders associated with HIV infection and AIDS. Part I:
220. Juhlin L: Kaposi’s sarcoma after immunosuppressant therapy. Acta infectious musculoskeletal conditions. Skeletal Radiol 33(5):249–
Dermatol Venereol 53:238, 1973. 259, 2004.
221. Klepp O, Dahl O, Stenwig JT: Association of Kaposi’s sarcoma
and prior immunosuppressive therapy: 5 year material of Kaposi’s Reactive Vascular Lesions
sarcoma in Norway. Cancer 42:2626–2630, 1978.
222. Langford A, Pohle HD, Reichart P: Primary intraosseous AIDS- Bacillary Angiomatosis of Bone
associated Kaposi’s sarcoma: report of two cases with initial jaw 238. Birtles RJ, Harrison TG, Taylor AG: The causative agent of bacil-
involvement. Int J Oral Maxillofac Surg 20:366–368, 1991. lary angiomatosis. N Engl J Med 325:1447–1448, 1991.
223. Lee SCH, Moore OS: Kaposi’s sarcoma of lymph nodes. Arch 239. Glick M, Cleveland DB: Oral mucosal bacillary epithelioid angio-
Pathol 80:651–654, 1965. matosis in a patient with AIDS associated with rapid alveolar bone
224. Lopez-Rubio F, Anguita M, Arizon JM, et al: Visceral Kaposi’s loss: case report. J Oral Pathol Med 22:235–239, 1993.
sarcoma without mucocutaneous involvement in a heart trans- 240. Goodman P, Balachandran S: Bacillary angiomatosis in a patient
plant recipient. J Heart Lung Transplant 13:913–915, 1994. with HIV infection. AJR Am J Roentgenol 160:207–208, 1993.
225. Martinez-Sapina MJ, Mosquera J, Castro JM, et al: Kaposi 241. Herts BR, Rafii M, Spiegel G: Soft-tissue and osseous lesions
sarcoma involving bone in an HIV negative patient. Eur J Radiol caused by bacillary angiomatosis: unusual manifestations of cat-
15:200–202, 1992. scratch fever in patients with AIDS. AJR Am J Roentgenol 157:
226. Mitsuyasu RT: Clinical variants and staging of Kaposi’s sarcoma. 1249–1251, 1991.
Semin Oncol 14:13–18, 1987. 242. Myers SA, Prose NS, Garcia JA, et al: Bacillary angiomatosis in a
227. Nichols CM, Flaitz CM, Hicks MJ: Primary intraosseous Kaposi’s child undergoing chemotherapy. J Pediatr 121:574–578, 1992.
sarcoma of the maxilla in human immunodeficiency virus infec- 243. Omarini LP, Loes SK, Garcia J, et al: Case report 860: bacillary
tion: review of literature and report of case. J Oral Maxillofac Surg angiomatosis of the calcaneus. Skeletal Radiol 23:478–481, 1994.
53:325–329, 1995. 244. Reed JA, Brigati DJ, Flynn SD, et al: Immunocytochemical iden-
228. O’Connell KM: Kaposi’s sarcoma in lymph nodes: histological tification of Rochalimaea henselae in bacillary (epithelioid) angio-
study of lesions from 16 cases in Malawi. J Clin Pathol 30:696–703, matosis, parenchymal bacillary peliosis, and persistent fever with
1977. bacteremia. Am J Surg Pathol 16:650–677, 1992.
229. Olweny CL, Kaddumukasa A, Atine I, et al: Childhood Kaposi’s 245. Relman DA: Bacillary angiomatosis and Rochalimaea species. Curr
sarcoma: clinical features and therapy. Br J Cancer 33:555–560, Clin Top Infect Dis 14:205–219, 1994.
1976. 246. Standiford KN, Emery CD, Schiffman RJ: Case report 865: bacil-
230. Penn I: Kaposi’s sarcoma in organ transplant recipients: report of lary angiomatosis. Skeletal Radiol 23:569–571, 1994.
20 cases. Transplantation 27:8–11, 1979. 247. Taylor AG, Birtles R, Harrison TG: Cat-scratch, Kaposi’s sarcoma,
231. Qunibi WY, Barri Y, Alfurayh O, et al: Kaposi’s sarcoma in renal and bacillary angiomatosis. Lancet 342:686, 1993.
transplant recipients: a report of 26 cases from a single institution. 248. Tsang WY, Chan JK: Bacillary angiomatosis: a “new” disease with
Transplant Proc 25:1402–1405, 1993. a broadening clinicopathologic spectrum. Histol Histopathol 7:143–
232. Ramos CV, Taylor HB, Hernandez BA, et al: Primary Kaposi’s 152, 1992.
sarcoma of lymph nodes. Am J Clin Pathol 66:998–1003, 1976. 249. Webster GF, Cockerell CJ, Friedman-Kien AE: The clinical
233. Ritz-Quillac L, Machet L, Machet MC, et al: Bone involvement spectrum of bacillary angiomatosis. Br J Dermatol 126:535–541,
in a case of Kaposi’s sarcoma. Dermatology 198:73–74, 1999. 1992.

ERRNVPHGLFRVRUJ
C H A P T E R 1 4 

Neurogenous Tumors and


Neurofibromatosis Affecting Bone
Gregory N. Fuller, MD, PhD

CHAPTER OUTLINE

MENINGIOMA Neurofibromatosis Type 2


SOLITARY FIBROUS TUMOR/ Neurofibromatosis Type 3
(Schwannomatosis)
HEMANGIOPERICYTOMA FAMILY TUMORS
VON HIPPEL-LINDAU DISEASE
MYXOPAPILLARY EPENDYMOMA
Hemangioblastoma
INTRAOSSEOUS SCHWANNOMA Papillary Endolymphatic Sac Tumor
NEUROGENIC TUMOR PREDISPOSITION
SYNDROMES
Neurofibromatosis Type 1

Neurogenous tumors arising within bone are exceedingly have a unique propensity to involve the contiguous bone
rare. Neural lesions frequently exert secondary effects and may mimic a primary bone tumor.4,6,7,13,14 Involve-
on bones, eroding them from the surface and deforming ment of bone is not indicative of malignant behavior per
and expanding neural canals and foramina because of se. Meningiomas may alter adjacent bones by direct inva-
the pressure of expansile growth, as in the case of neuro- sion or may cause a hyperostotic reaction.15,18,19,26,31 In the
fibromatosis. On the other hand, some of the most latter case, there is no true invasion of bone. This neo-
common tumors of the central nervous system, namely plasm provokes bone overgrowth of periosteal origin,
meningiomas and ependymomas, have a tendency to usually from the inner table of the cranial bones.
invade bone and may provoke striking reactive changes in Most meningiomas become clinically evident in
affected bone that mimic primary bone neoplasms. The patients older than age 40 years and are rare in children.
rarest of primary neural tumors arising in bone are the Meningiomas occur more frequently in women. The
neurilemmomas (schwannomas) and malignant periph- male-to-female ratio is 2 : 3 for intracranial lesions;
eral nerve sheath tumors. Neurofibromatosis, separated for intraspinal meningiomas, the male-to-female ratio
into three distinct clinical syndromes referred to as types is 1 : 10.
1 through 3, is primarily a disorder of the soft tissues, but Meningiomas are slow-growing lesions, but their
it can affect many other organs and the skeleton. Von indolent growth rate is increased during pregnancy. This
Hippel-Lindau disease and its hallmark tumor, heman- is partially explained by the presence of steroid receptors
gioblastoma, is also included in this chapter, with the in these tumors. Theoretically, meningiomas can develop
discussion focusing on the skeletal manifestations of the anywhere in the meninges; however, they appear fre-
disease with the development of a unique papillary endo- quently in certain anatomic sites. The parasagittal area is
lymphatic sac tumor involving the petrous portion of the the most frequent location for cranial meningiomas.
temporal bone. Other common locations include the lateral aspects of the
cranial vault (over the cerebral convexities), the wing of
sphenoid bone (sphenoid ridge meningioma), the sella
MENINGIOMA turcica area (suprasellar meningioma), the cribriform
plate (olfactory groove meningioma), and the area of the
Meningiomas are not generally considered to be neural foramen magnum and the optic nerve. In addition to the
tumors of bone, but they are in fact the most common common locations within the central nervous system,
tumors of the central nervous system to erode bone. meningiomas can present as extracranial lesions in many
Meningiomas are common neoplasms that originate sites.17,23,25,36 The soft tissues of the head and neck are
from meningothelial cells. Although meningiomas may the most frequent sites of involvement external to the
invade the brain, they most often affect the central central nervous system.8,9,17,22,30 Rare examples of menin-
nervous system structures by a pushing, expansile growth giomas in the lung, the mediastinum, and even the fingers
rather than by invasion. On the other hand, meningiomas have been described. We have seen in consultation two
990
ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 991

examples of apparently primary meningiomas involving meningiomas associated with neurofibromatosis type 2.
the pubic ramus. Skull lesions may have the same or The somatic mutations of the NF2 gene are also present in
similar radiographic features as metastatic carcinoma or, the majority of sporadic meningiomas not associated with
less frequently, hemangioma or osteosarcoma. neurofibromatosis type 2. Sporadic meningiomas appear
Meningiomas usually occur sporadically but it is esti- to have unique genetic alterations that are exclusive of
mated that approximately 5% are familial and fewer than the NF2 gene mutations. A recent genomic analysis of
10% of individuals have multiple lesions. The environ- meningiomas has identified that sporadic meningiomas
mental risk factors, except for radiation, are uncertain. not associated with neurofibromatosis type 2 are charac-
An increased risk for the development of meningiomas terized by mutations in TRAF7, a proapoptotic E3 ubiq-
has been associated with several tumor susceptibility syn- uitin ligase, in approximately 25% of tumors (Fig. 14-1).2
dromes such as neurofibromatosis type 2, Cowden syn- These mutations are concurrent with the mutations in
drome, and Werner syndrome. These syndromes have KLF4, a transcription factor inducing pluripotency, or
unique genetic backgrounds and are caused by mutations AKT1 activating the PI3K pathway and SMO mutations
in the NF2, PTEN, and RECQL2 genes, respectively.1 that activate Hedgehog signaling (Fig. 14-1).2 Extremely
The mutations of the tumor suppressor gene NF2 are rare cases of familial meningiomas without neurofibro-
present in more than 70% of meningiomas developing matosis type 2 were shown to be associated with muta-
in individuals affected by neurofibromatosis type 2. The tions involving the SWI/SNF chromatin-remodeling
loss of the wild type NF2 allele is present in almost all complex subunit genes SMARCE1 or SMARCB1.24,28

TRAF7 KLF4
Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 20
Exon 19
Exon 21
Chr 16 Chr 9
Exon 1
Exon 2

Exon 3
Exon 4
Exon 5
Exon 6
Exon 7
Exon 8
Exon 9

Exon 1

Exon 2

Exon 3

Exon 4

Exon 5
13.3 TRAF7 24
13.2

110247132

110252047
13.1 23
2205798

2228130

22
12 21
11.2 13
11.1 11.1 tel cen tel cen
12
11.2 11.1 11.1
12.1
13 12.2 TRAF7 12 KLF4
21 13
22 21.1
23 21.2
21.3
24 1 671 aa 22.1 1 594 aa
22.2
22.3
Zinc finger domain 31 KLF4
32 Zinc finger C2H2-type/integrase
TRAF like domain 33 DNA-binding domain
34.1
SIAH type domain 34.2 34.3 Synonymous substitution
Zinc Finger, TRAF type domain
Missense substitution
TRAF like domain
Insertion frameshift
W D40/YVTN repeat-like-containing domain B
Synonymous substitution
Missense substitution
Insertion frameshift
A

AKT1 SMO
Exon 10
Exon 11
Exon 12
Exon 13
Exon 14

Exon 10
Exon 11
Exon 12
Exon 1

Exon 2

Exon 3

Exon 4
Exon 5
Exon 6
Exon 7
Exon 8
Exon 9

Exon 1

Exon 2

Exon 3
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8
Exon 9

Chr 14 Chr 7
13 22
12
128828712

128853385
105235686

105262080

11.2 21
11.1 11.1 15.3
11.2 15.1 15.2
12 tel cen 14 cen tel
13 13
21 AKT1 11.2 12 SMO
167

11.1 11.1
10

22 11.22 11.21
23 11.23
24.1
24.2
24.3 1 480 aa 21.1 21.2 1 788 aa
31 32.1 21.3
32.2 32.3 Pleckstrin homology-like domain 22 Frizzled domain
AKT1 Protein kinase-like domain 31.1 GPCR, family 2-like domain
31.2
AGC-kinase, C-terminal domain 31.3
SMO Synonymous substitution
33 32
Synonymous substitution 34 Missense substitution
35
Missense substitution 36 Nonsense substitution - Stop
C Insertion inframe
Deletion frameshift
D
FIGURE 14-1  ■  Mutations of genes in sporadic meningioma. A, Chromosomal location, exonal structure, functional domains, and the
distribution of mutations in the TRAF7 gene. B, Chromosomal location, exonal structure, functional domains, and the distribution
of mutations in the KLF4 gene. C, Chromosomal location, exonal structure, functional domains, and the distribution of mutations
in the AKT1 gene. D, Chromosomal location, exonal structure, functional domains, and the distributions of mutations in the SMO
gene.

ERRNVPHGLFRVRUJ
992 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

SMARCB1

Exon 1

Exon 2
Exon 3

Exon 4

Exon 5

Exon 6

Exon 7

Exon 8
Exon 9
24129149

24176705
Chr 22
13 cen tel
12
11.2
11.1
11.1 SMARCB1
SMARCB1
12.1

19
11.2

14
12.3 12.2

5
6

11

15
28
13.1

6
13.2
13.3
1 366 aa
Synonymous substitution
Missense substitution
Nonsense substitution - Stop
Insertion inframe
Deletion inframe
SMARCE1

Exon 10

Exon 11
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8
Exon 9
Exon 1

Exon 2
Exon 3
Insertion frameshift
A Deletion frameshift
Chr 17

38783975

38804103
13
12 tel cen
11.2 11.1
11.1 11.2 SMARCE1
12 SMARCE1
21.2
21.1
22 21.3 1 412 aa
23 High mobility group (HMG) box
24
25 Synonymous substitution
Missense substitution
B Nonsense substitution - Stop

SUFU
Exon 10

Exon 11
Exon 12
Exon 1
Exon 2

Exon 3
Exon 4
Exon 5
Exon 6
Exon 7
Exon 8

Exon 9

Chr 10
15
46919235

46945289

14
13
12.2 12.3
12.1 cen tel
11.2
11.1 11.1
11.2
SUFU
21.1
21.2
21.3 1 594 aa
22.1
22.2 Suppressor of fused domain
22.3
23.1 23.2 Suppressor of fused C-terminal domain
23.3 SUFU
24.2 24.1 Synonymous substitution
25.1 24.3
25.3 25.2 Missense substitution
26.2 26.1 Nonsense substitution - Stop
26.3
Insertion inframe
Deletion frameshift
C
FIGURE 14-2  ■  Mutations of genes in familial meningioma. A, Chromosomal location, exonal structure, functional domains, and the
distribution of mutations in the SMARCB1 gene. B, Chromosomal location, exonal structure, functional domains, and the distribution
of mutations in the SMARCE1 gene. C, Chromosomal location, exonal structure, functional domains, and the distribution of muta-
tions in the SUFU gene.

Loss of SUFU, suppressor of fused homolog of Drosoph- anterior cranial fossa mass centered in the midline (olfac-
ila, dysregulating Hedgehog signaling was also identified tory groove meningioma) (Fig. 14-3). Meningioma may
in some families affected by multiple meningiomas also present as a purely intraosseous tumor. In such
(Fig. 14-2).1 instances, it may be mistaken for a different type of
primary bone lesion; for example, intraosseous menin-
gioma of the sphenoid bone may be mistaken for fibrous
Radiologic Features
dysplasia. In addition, some histologic subtypes of menin-
The classic radiologic presentation of meningioma is that gioma may contribute to misdiagnosis; for example,
of a homogeneously contrast enhancing dura-based mass intraosseous secretory meningioma may be mistaken for
with small peripheral extensions referred to as the dural metastatic carcinoma (Fig. 14-4).
tail (Fig. 14-3). Other radiographic presentations include Some meningiomas show involvement of three ana-
a flattened extensive lesion referred to as en plaque menin- tomic compartments: cranial bone, intracranial exten-
gioma or an intraventricular (choroid plexus) meningi- sion, and extracranial extension into the scalp (Fig. 14-5).
oma. In addition, classic presentation is as a very large Often, meningiomas induce prominent periosteal new

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 993

A B

C D
FIGURE 14-3  ■  Meningioma: radiographic features. A, T1-weighted axial postcontrast magnetic resonance image (MRI) showing a
dura-based meningioma with uniform signal enhancement and dural tail sign. B, T1-weighted coronal postcontrast MRI showing
en plaque meningioma with uniform signal enhancement (arrow). C, T1-weighted coronal postcontrast MRI showing left intraven-
tricular (glomus chorideum) meningioma with uniform signal enhancement. D, T1-weighted sagittal postcontrast MRI showing
olfactory groove meningioma with uniform signal enhancement.

ERRNVPHGLFRVRUJ
994 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-4  ■  Meningioma: radiographic and microscopic features. A, Axial computed tomogram showing thickening of the
sphenoid bone caused by intraosseous meningioma (arrows). B, T1-weighted axial magnetic resonance image (MRI) showing
thickening of the sphenoid bone caused by intraosseous meningioma (arrows). Same case as A. C, Low power photomicrograph
showing meningioma infiltrating bone (×100). D, High power photomicrograph showing secretory meningioma infiltrating fibrous
tissue (×200). Inset, Expression of carcinoembryonic antigen in meningioma cells revealed by immunohistochemistry (×400). (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 995

A B

C D
FIGURE 14-5  ■  Meningioma: radiographic features. A, Axial computed tomogram showing intraosseous meningioma of the frontal
bone (arrow). B, T1-weighted axial postcontrast magnetic resonance image (MRI) showing intraosseous meningioma. C, T1-weighted
axial postcontrast MRI showing hyperostotic meningioma of the frontal bone with extension intracranially and extracranially into
the soft tissues of scalp. D, T1-weighted coronal postcontrast MRI showing a markedly hyperostotic meningioma of the frontal bone
with intracranial and extracranial extension.

ERRNVPHGLFRVRUJ
996 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

bone formation in the form of perpendicular striations fibrillogranular material surrounded by unit membrane
that radiate from the lesion on the inner and outer sur- with microvilli (Fig. 14-9).
faces of the affected bone (Fig. 14-6). Meningiomas Lymphoplasmacyte rich meningioma is the least common,
must therefore be differentiated radiographically from least well characterized, and most controversial menin-
other primary and metastatic bone lesions. Higher grade gioma subtype. Many examples previously reported in
meningiomas frequently exhibit atypical radiographic the literature are now recognized as reactive or inflam-
features as well, including a blurred or diffuse interface matory lesions, rather than meningiomas. Nevertheless,
with the subjacent brain parenchyma, or a multilobulated there are legitimate examples of meningiomas with
appearance (mushroom sign). In addition, brain invasive prominent intratumoral lymphoplasmacytic infiltrates.
meningiomas are often accompanied by marked brain The significance awaits further investigation (Fig. 14-9).
edema (Fig. 14-7). Microcystic meningioma exhibits a distinctive architec-
ture of spidery cell processes that form variously sized
Microscopic Findings microcysts to the extent that, to the uninitiated, the
tumor may not be recognized as a meningioma. Closer
Microscopically, meningiomas represent a group of neo- inspection will reveal typical bland nuclei. Microcystic
plasms with various heterogenous features linked by focal meningioma often shows morphologic features that
similarities to normal meningothelial cells (Table 14-1).10 overlap with those of angiomatous meningioma (rich
The nuclei are round or oval with small nucleoli and fine, vasculature) and secretory meningioma (occasional
evenly dispersed chromatin. Intranuclear inclusions are pseudopsammoma bodies) (Fig. 14-10). All three of
frequently present. The four classic histologic subtypes these meningioma variants—angiomatous, secretory, and
of meningioma are syncytial, fibroblastic, transitional, microcystic—frequently display “degenerative” nuclear
and psammomatous (Fig. 14-8). atypia, and in some tumors the morphologic features are
Syncytial meningioma is characterized by patternless so overlapping that any of the three appellations might
architecture, without other distinctive features. The lack be applied. In the absence of high-grade features (such as
of a distinct cytoplasmic border (i.e., the syncytial appear- elevated mitotic activity or brain invasion), all three are
ance of meningothelial cells) is explained at the ultra- benign, WHO grade I, subtypes with no prognostic
structural level by the complex interdigitating cytoplasmic significance.
processes. Chordoid meningioma is characterized by cords of
Fibroblastic or spindle-cell meningioma is characterized meningothelial cells typically surrounded by a rich
by the presence of elongated spindle cells arranged in myxoid matrix (Fig. 14-11).
fascicles with a focal storiform pattern. Focal syncytial Clear cell meningioma is characterized by prominent
features with a whorled pattern and psammoma bodies cytoplasmic clearing (Fig. 14-11).
are present in some fibroblastic meningiomas. Papillary meningioma represents a distinct variant of
Transitional meningioma shows the presence of clearly meningioma. Recognition of this entity has clinical sig-
recognizable syncytial areas and fibroblastic features. nificance because these neoplasms exhibit locally aggres-
Cellular whorls are typical for this type, which is also sive growth and are associated with late metastases. These
known as mixed meningioma. neoplasms are composed of meningothelial cells arranged
Psammomatous meningioma is characterized by promi- in papillary structures with central fibrovascular cores
nent numbers of calcifications with a concentrically (perivascular pseudorosettes) (Fig. 14-11).
layered architecture referred to as psammoma bodies. Rhabdoid meningioma is another meningioma subtype
Scattered psammoma bodies can be seen in virtually any with potential for aggressive clinical behavior. Tumor
subtype of meningioma; however, they are, by definition, cells exhibit characteristic rhabdoid morphology, often
particularly abundant and densely packed in the psam- lacking other typical architectural and cytologic features
momatous variant. of meningioma.
Angiomatous meningioma is characterized by prominent Atypical and anaplastic meningiomas comprise the higher
vascularity. The outdated term angioblastic meningioma grade categories of meningiomas. Atypical meningiomas
encompassed at least three different types of tumor, (WHO grade II) exhibit at least three of the following
including angiomatous meningioma, hemangiopericy- five histologic features: foci of micronecrosis, prominent
toma, and hemangioblastoma, which are considered dis- nucleoli, sheeting architecture, small cell formation, or
tinctly different neoplasms today.12 The morphologic hypercellularity (Figs. 14-12). Anaplastic meningioma
features frequently overlap with those of microcystic (WHO grade III) exhibits morphologic features resem-
meningioma (Fig. 14-9). bling sarcoma, carcinoma, or melanoma. These are
Metaplastic meningioma exhibits focal areas of metapla- among the rarest meningioma variants.
sia, which may differentiate along cartilaginous, osseous, Brain invasive meningiomas exhibit increased potential
or lipomatous lines. There is no prognostic significance for early recurrence and are therefore classified as WHO
(Fig. 14-9). grade II. In cases in which it is uncertain whether the
Secretory meningioma has microscopic features similar invaded tissue is brain parenchyma or reactive leptomen-
to those of syncytial or transitional meningioma. In ingeal tissue, immunostaining for GFAP or S-100 protein
addition, individual tumor cells show discrete intracy- can be performed (Fig. 14-13).
toplasmic inclusion bodies that are strongly positive for Grading of meningiomas is based on levels of mitotic
carcinoembryonic antigen. Ultrastructurally, these inclu- activity (Table 14-2).10 Upgrading of any meningioma
sions are intracytoplasmic and represent deposits of dense histologic subtype is warranted when increased mitotic

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 997

A B

C D
FIGURE 14-6  ■  Meningioma: radiographic, gross, and microscopic features. A, Axial computed tomogram showing characteristic
“hair on end” or “starburst” sign in a hyperostotic frontal meningioma. B, Whole-mount gross photograph of hyperostotic frontal
bone shown in A. C, Low power photomicrograph of meningioma with reactive bone corresponding to the radiographic “hair on
end” sign (×100). D, Intermediate power photomicrograph of the characteristic bone spicules extending into subcutaneous tissue
in a hyperostotic meningioma (×200). (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
998 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-7  ■  Meningioma: atypical radiographic features. A, T1-weighted axial postcontrast magnetic resonance image (MRI)
showing a large falcine signal-enhancing tumor in the anterior cranial fossa. Note blurring of the tumor-brain interface in this atypi-
cal meningioma. B, T1-weighted coronal postcontrast MRI showing a signal-enhancing meningioma located in the right parietooc-
cipital region with lobulated contour (“mushrooming” sign) in this atypical meningioma. C, T1-weighted axial postcontrast MRI
showing a signal-enhancing meningioma with lobulated contour located in right frontal region in this anaplastic meningioma.
D, T1-weighted sagittal postcontrast MRI showing frontal signal-enhancing meningioma surrounded by marked cerebral edema.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 999

TABLE 14-1  Microscopic Variants of Meningioma


Type Microscopic Features
WHO Grade I
Meningothelial (syncytial) Syncytium-like histology (cell borders are not easily discernible).
Fibroblastic (fibrous) Compact fascicles of highly spindled cells (fibroblast-like) alternating with areas of syncytial
appearance, giving an alternating “on-edge” and “en face” feeling. Some tumors have
prominent collagen bands, which may calcify in a linear, spiculated fashion (distinct from
spherical psammoma bodies).
Transitional A swirling together of the syncytial and fibroblastic patterns, with resultant prominent cell whorls
that constitute the hallmark feature of this subtype. The whorls may calcify, beginning in the
center, creating a psammoma body.
Psammomatous Essentially a transitional meningioma in which a large percentage of the meningothelial cell
whorls have calcified (formed psammoma bodies). There is no specified number, percentage,
or density of psammoma bodies that is required to “qualify” as a psammomatous meningioma.
Angiomatous Meningioma in which the vasculature is especially dense and prominent. Frequently shows
degenerative nuclear atypia. An older term, angioblastic, is inaccurate and obsolete.
Metaplastic Displays focal areas of bone, cartilage of fatty metaplasia. As with the other grade I meningioma
subtypes, there is no prognostic significance.
Secretory The hallmark morphologic feature is the presence of pseudopsammoma bodies, which are
brightly eosinophilic intracystoplasmic globular inclusions (proteinaceous secretions) of varying
number and size. Pseudopsammoma bodies are strongly positive for carcinoembryonic antigen,
and the cells that produce them are strongly positive for epithelial membrane antigen and
keratin.
Lymphoplasmacyte-rich Among the rarest of meningioma subtypes, this exhibits prominent collections of chronic
inflammatory cells (lymphocytes and plasma cells).
Microcystic Characterized by prominent microcystic architecture throughout the tumor; may not otherwise
bear much resemblance to traditional meningioma morphology. Frequently displays prominent,
often hyalinized, vasculature and degenerative atypia, thus overlapping with the angiomatous
subtype.

WHO Grade II
Chordoid Cords of tumor cells separated by myxoid matrix.
Clear cell Cytoplasmic clearing in a majority of tumor cells can obscure the meningothelial nature of the
tumor; whorls can often be highlighted by immunostaining for vimentin or epithelial membrane
antigen.

WHO Grade III


Rhabdoid Rhabdoid morphology (small epithelioid cells with globular eosinophilic cytoplasm and eccentric
nuclei) predominates in this variant, which, like the microcystic subtype, may not be instantly
recognizable as meningothelial in origin by those not familiar with the entity. Mitotic activity is
typically elevated.
Papillary Tumor cells form perivascular pseudorosettes (similar to those seen in ependymoma), which may
artifactually dehisce, giving a papillary (pseudopapillary) appearance. As with the rhabdoid
subtype, mitotic activity is usually elevated.

activity is present. Specifically, meningiomas with four or for S-100 protein. Keratin is typically not present in
more mitoses per 10 contiguous high-power fields are meningioma, but it can be focally positive, especially in
classified as WHO grade II, and meningiomas with 20 or secretory meningioma.
more mitoses per 10 contiguous high-power fields are
classified as WHO grade III (Fig. 14-13).
Clinical Behavior
Meningiomas that invade osseous structures can
provoke prominent reactive bone formation. Sometimes The vast majority of meningiomas are clinically benign
reactive new bone forms parallel striations. This orienta- and their eradication depends on the size of the lesion,
tion is best seen on specimen radiographs but can specific topographic involvement, as well as involvement
also be demonstrated during low power microscopic of the vital structures of the central nervous system.
examination. Reactive bone formation may also take the A small proportion of meningiomas exhibit locally
form of haphazardly arranged bone trabeculae. Occasion- aggressive behavior. These tumors rarely may exhibit
ally, the bone formation is so abundant that it overshad- metastatic potential with involvement of extracranial
ows underlying tumor cells. osseous sites.16,20,27,29 Both grading of meningiomas and
Immunohistochemically, meningiomas are positive for their molecular characteristics correlate to some degree
epithelial membrane antigen.5,11,21 In the setting of cranial with clinical behaviors. In general, grade I meningiomas
lesions, this feature is very helpful in distinguishing behave as benign tumors, and grade II are locally aggres-
meningiomas from schwannomas and tumors of glial cell sive, whereas grade III meningiomas have some degree
origin. As in all cells of mesenchymal origin, vimentin of metastatic potential. Meningiomas characterized by
is strongly positive in meningiothelial cells. Approxi- mutations of the TRAF7, KLF4, and SMO genes are
mately 20% of meningiomas show at least focal positivity Text continued on p. 1006

ERRNVPHGLFRVRUJ
1000 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-8  ■  Meningioma: microscopic features. A, Intermediate power photomicrograph showing transitional (WHO grade I)
subtype meningioma with prominent whorl formation. B, Intermediate power photomicrograph showing syncytial (WHO grade I)
subtype of meningioma. C, Intermediate power photomicrograph showing fibrous (fibroblastic) (WHO grade I) subtype of menin-
gioma. D, Intermediate power photomicrograph showing psammomatous (WHO grade I) subtype of meningioma (A-D, ×200,
hematoxylin-eosin).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1001

A B

C D
FIGURE 14-9  ■  Meningioma: microscopic features. A, Intermediate power photomicrograph showing angiomatous (WHO grade I)
subtype of meningioma with rich vasculature. B, Intermediate power photomicrograph showing metaplastic (WHO grade I) subtype
of meningioma with areas of cartilage formation. C, Intermediate power photomicrograph showing secretory (WHO grade I)
subtype of meningioma with prominent pseudopsammoma body formation. D, Intermediate power photomicrograph showing
lymphoplasmacyte-rich (WHO grade I) subtype of meningioma with prominent lymphoplasmacytic infiltrate between meningioma
whorls (A-D, ×200, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1002 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-10  ■  Meningioma: microscopic features. A, Low power photomicrograph of a microcystic subtype of meningioma, in which
the architecture is dominated by a microcyst formation (×100). B, Intermediate power photomicrograph of microcystic meningioma
showing the often spider web–like morphology of the microcystic architecture (×200). C, Low power photomicrograph of a menin-
gioma with mixed features of the angiomatous, microcystic, and secretory variants (×100). D, High power photomicrograph of a
meningioma with mixed angiomatous, microcystic, and secretory features. Meningiomas with this combination of features are not
uncommon, and they often display prominent degenerative (“aging”) nuclear atypia, as seen in this case. Note the pseudopsam-
moma bodies (secretory globules) in the center of the field (×400) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1003

A B

C D
FIGURE 14-11  ■  Meningioma: microscopic features. A, Intermediate power photomicrograph showing chordoid (WHO grade II)
subtype of meningioma with nests and cords of meningioma cells in a myxoid background. B, Intermediate power photomicrograph
showing clear cell (WHO grade II) subtype of meningioma. C, Intermediate power photomicrograph showing papillary (WHO grade
III) subtype of meningioma with prominent perivascular pseudorosette formation. D, Intermediate power photomicrograph showing
rhabdoid (WHO grade III) meningioma (A-D, ×200, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1004 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-12  ■  Meningioma: microscopic features. A, Intermediate power photomicrograph showing a focus of micronecrosis in an
atypical (WHO grade II) meningioma. B, Intermediate power photomicrograph showing prominent nucleoli in an atypical (WHO
grade II) meningioma. C, Intermediate power photomicrograph showing “sheeting” architecture in an atypical (WHO grade II)
meningioma. D, Intermediate power photomicrograph showing small cell foci in an atypical (WHO grade II) meningioma (A-D, ×200,
hematoxylin-eosin).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1005

A B

C D
FIGURE 14-13  ■  Meningioma: microscopic features. A, Low power photomicrograph showing invasion of brain by meningioma (×100).
B, Low power photomicrograph confirming brain invasion by expression of GFAP in invaded brain tissue (×100). C, Intermediate
power photomicrograph showing brisk mitotic activity in this anaplastic (WHO grade III) meningioma (×200). D, High power photo-
micrograph of mitotic figure identification using immunohistochemistry demonstrating expression of phosphohistone H3 in M-phase
cells (×400). (A and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1006 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

hemangiopericytoma (also see Chapter 13). Intracranial


TABLE 14-2 World Health Organization (WHO)
hemangiopericytoma has a high propensity for extracra-
Grading of Meningiomas
nial skeletal metastases. In fact, most bone hemangioperi-
WHO Grade I cytomas are metastatic lesions, and a significant proportion
Benign Meningioma of them represent distant metastases from meningeal
Histologic variant other than clear cell, chordoid, papillary, hemangiopericytoma.12,16,20 Extracranial skeletal metasta-
or rhabdoid ses also occasionally occur in conventional (i.e., transi-
Lacks criteria of grades II and III meningiomas tional) meningiomas with or without features of cytologic
WHO Grade II
atypia.20,27,29 His­torically, central nervous system heman-
giopericytomas were lumped in with angiomatous menin-
Atypical Meningioma
giomas in a now discredited category of tumor termed
Mitotic index ≥4 per 10 high-power fields
or angioblastic meningioma.
At least 3 of 5 parameters:
Sheeting architecture (loss of whorling or fascicles)
Small cell formation (high N/C* ratio) Radiologic Features
Macronucleoli
Hypercellularity
The radiologic features of solitary fibrous tumor of the
Spontaneous necrosis (i.e., not induced by embolization central nervous system typically mimic those of menin-
or radiation) gioma; that is, they are dura-based circumscribed masses
or that show enhancement after contrast administration
Brain Invasion (Fig. 14-14). At the more aggressive end of the spectrum,
or hemangiopericytoma (cellular solitary fibrous tumor) has
a tendency for early recurrence and metastasis, including
Clear Cell Meningioma a high propensity for extracranial skeletal metastases.
or
Chordoid Meningioma Gross Findings
WHO Grade III Grossly the lesions are usually well delineated, firm, and
Anaplastic (Malignant) Meningioma nodular, with an overall fibrous appearance. On cut
Mitotic index ≥20 per 10 high-power fields section, the nodular nature of the lesion is evident and
or the great white fibrous mass may have foci of myxoid
Frank anaplasia (sarcoma, carcinoma, or melanoma-like
histology) change or hemorrhage.
or
Papillary Meningioma Microscopic Findings
or
Solitary fibrous tumors are composed of spindle cells
Rhabdoid Meningioma arranged in short ill-defined fascicles, described as a pat-
ternless pattern. A characteristic feature is the presence
*N/C, nuclear/cytoplasmic.
of striking hyalinization. The vasculature varies from
narrow vascular channels to prominent open branching
vessels referred to as hemangiopericytoma-like vasculature.
distinct and nearly always show a benign clinical course The cellular variants of solitary fibrous tumors have over-
with chromosomal stability and have a tendency to origi- lapping features with soft tissue hemangiopericytoma.
nate in the medial skull base. Meningiomas containing The solitary fibrous tumor family comprises a spectrum
the mutant NF2 gene frequently exhibit loss of chromo- of tumors ranging from relatively indolent solitary fibrous
some 22, are genomically unstable, and are more likely tumor at one end to aggressive hemangiopericytoma (cel-
atypical. These tumors have a tendency to originate over lular solitary fibrous tumor) at the other (Fig. 14-14).
the cerebral and cerebellar convexities. The microscopic features of solitary fibrous tumor in the
central nervous system overlap with those of the more
common ones that affect the soft tissue, predominantly
SOLITARY FIBROUS TUMOR/ the pleura.34,37,39,41-44,46,48 At the two ends of the spectrum,
the tumors exhibit distinctive classical morphologic fea-
HEMANGIOPERICYTOMA tures, with solitary fibrous tumor displaying bland spindle
FAMILY TUMORS cells separated by prominent bands of ropy collagen and
strong diffuse immunoreactivity for CD34, whereas clas-
The solitary fibrous tumor, considered by most authors sical hemangiopericytoma is a highly vascular, densely
as a separate entity, is also designated as intracranial cellular, mitotically active neoplasm in which CD34 reac-
hemangiopericytoma.32,33,35,38,40,47,49 Although the meningo- tivity is confined to the vasculature (Fig. 14-13). However,
thelial origin of these neoplasms is questionable, they it was recognized early on that many hemangiopericyto-
have age-distribution and site-predilection patterns mas display at least focal CD34 reactivity, and in fact the
within the central nervous system that are similar to those spectrum of solitary fibrous tumor embraces a wide range
of conventional meningiomas. The overall microscopic of transitional forms that blend in various proportions
appearance of this neoplasm is identical to that of the morphologic, immunophenotypic and proliferative

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1007

FIGURE 14-14  ■  Solitary fibrous tumor/hemangiopericytoma: radiographic and microscopic features. A, T1-weighted sagittal post-
contrast magnetic resonance image (MRI) showing heterogeneously signal enhancing tumor of parietal region. B, T1-weighted axial
postcontrast MRI showing signal enhancing mass in the right cavernous sinus region. C, Low power photomicrograph of the tumor
in A showing bland fibroblastic cells separated by dense bands of collagen characteristic of solitary fibrous tumor (×100). Upper
Inset, Strong diffuse expression of CD34 in solitary fibrous tumor revealed by immunohistochemistry (×200), Lower Inset, Nuclear
localization of STAT6 revealed by immunohistochemistry (×200). D, Intermediate power photomicrograph of the tumor in C showing
uniform hypercellular undifferentiated mesenchymal cells with brisk mitotic activity characteristic of hemangiopericytoma (cellular
solitary fibrous tumor) (×200). Upper Inset, In contrast to solitary fibrous tumor, only the blood vessels stain with CD34; the
tumor cells are negative (×200). Lower Inset, Reflecting the same underlying molecular driver as solitary fibrous tumor (NAB2-
STAT6 translocation and fusion), strong nuclear localization of STAT6 is also seen in hemangiopericytoma (x200). (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1008 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

features of classical hemangiopericytoma and solitary of all ependymomas, including the myxopapillary variant,
fibrous tumor. Recognition of the kinship between soli- is their microscopically recognizable dualistic phenotype
tary fibrous tumor and hemangiopericytoma was finally (i.e., glial and epithelial). However, immunohistochemi-
conclusively demonstrated beyond doubt by discovery of cal coexpression of epithelial and glial markers such
a shared underlying genetic signature. as glial fibrillary acidic proteins, epithelial membrane
Solitary fibrous tumors are characterized by the pres- antigen, and keratin is variable. Most ependymomas are
ence of a NAB2-STAT6 gene fusion in the vast majority positive for glial fibrillary acidic protein and often are
of both soft tissue and central nervous system tumors focally positive for epithelial membrane antigen.54,70
(Fig. 14-15).36,44,45 The fusion is caused by an inversion Myxopapillary ependymomas are slow-growing
within chromosome 12 resulting in the juxtaposition of tumors with a tendency for local recurrence. Lesions that
NAB2 and STAT6 neighbor genes. The breakpoint within are completely excised are associated with an approxi-
STAT6 is conserved, and typically the fusion variants mately 20-year mean survival rate. Typically, myxopapil-
contain almost the entire STAT6 reading frame. There lary ependymoma exhibits slow, local destructive growth.
are shifting breakpoints within the NAB2 gene, but all of Eventually the lesion disrupts the bone and extends to
the variants retain the early growth response protein 1 the perisacral tissue.57,60-63,65,68,69 Recurrences are typically
binding domain (EBD). The transcriptional activation multiple nodules that may coalesce. Local dissemination
domain (TAD) of the STAT6 fusion partner is most likely with multiple metastatic or satellite nodules is frequently
responsible for the proliferation inducing activity of the seen in terminal stages of the disease.55,59,64,73 Long-term
chimeric NAB2-STAT6 protein. The nuclear localization lesions may grow to a considerable size and compress
of STAT6 protein driven by the NAB2 promoter in soli- pelvic and abdominal organs.
tary fibrous tumors can be used as an auxiliary test in their
differential diagnosis.
INTRAOSSEOUS SCHWANNOMA
MYXOPAPILLARY EPENDYMOMA Intraosseous schwannoma (neurilemmoma) is extremely
rare. We have seen only a few examples of this lesion in
Myxopapillary ependymoma is a tumor that almost exclu- bone, and fewer than 200 examples have been described
sively occurs in the region corresponding to the filum in the literature.74-76,82,83 Intraosseous schwannoma is
terminale and cauda equina.53,62,66,70-72 Microscopically, similar to more common soft tissue schwannomas. It
myxopapillary ependymoma resembles embryonal struc- accounts for less than 0.2% of all primary bone tumors.
tures of this region. In fact, some myxopapillary ependy- Schwannoma can involve bone by three mechanisms:
momas may originate from the so-called ependymal (1) secondary erosion by an extraosseous tumor; (2)
myxopapillary rests that occasionally are identified in tumor arising from a nerve coursing through a canal in
the sacrococcygeal region.67 Similar to meningiomas, a bone and causing erosion of the bone, creating a
myxopapillary ependymomas can destroy bone and dumbbell-shaped configuration; or (3) tumor arising
present with features suggesting a primary bone tumor centrally (intramedullary) in a bone. The first two mecha-
(Fig. 14-16). Rare examples of these lesions have been nisms occur most frequently. A true intramedullary origin
described in the presacral or postsacral soft tissue. Lesions is exceptional.
located in extradural soft tissue are frequently associated
with spina bifida, but myxopapillary ependymomas that Definition
occur in soft tissue have also been reported without
any associated anatomic anomaly in the sacrolumbar Schwannoma (neurilemmoma) is a benign peripheral
region.50,52,56,58,60 They can present as lytic bone lesions nerve sheath tumor composed of Schwann cells that are
that involve the lumbosacral region.51-53,61,65 frequently arranged in hypercellular and hypocellular
As the name implies, myxopapillary ependymoma is areas, referred to as Antoni A and B tissue, respectively.
composed of ependymal cells arranged in papillary struc- Schwannomas are encapsulated and typically arise from
tures that show prominent stromal myxoid change. In a recognizable nerve trunk.
general, the architecture of this variant of ependymoma
mimics the normal embryonal structures of the filum Incidence and Location
terminale and cauda equina. The cells are arranged in a
fernlike pattern around a central fibrovascular core (Fig. Intraosseous schwannomas arise most frequently in
14-10). The cytoplasmic processes of neoplastic cells the mandible and sacrum.76,77,79,82,83,88,89 They have also
form radiating, rosette-like structures around vessels. been reported in a number of other locations, includ-
The papillary architecture is accentuated by the collarlike ing the vertebral bodies, ulna, humerus, femur, tibia,
arrangement of ependymal cells around the vessels. A patella, scapula, rib, maxilla, and small bones of the
mucoid substance is often present in the form of vesicular hands.77-80,85,91,92 Intraosseous schwannomas were also
intercytoplasmic structures of varying sizes that separate described in a setting of neurofibromatosis type 3
tumor cells (Fig. 14-17). Distinctive, peculiar structures (schawannomatosis).84
seen in myxopapillary ependymoma are eosinophilic
globoid bodies positive for periodic acid-Schiff, alcian Clinical Symptoms
blue, Masson, and reticulin stains. Ultrastructurally, they
are composed of a collagen-rich substance surrounded by The most common symptoms are pain and localized
tumor cell processes resting on basal lamina. A hallmark swelling over the affected area.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1009

NAB2

Exon 1

Exon 2

Exon 3

Exon 4
Exon 5
Exon 6

Exon 7
Chr 12
13.3
13.2
13.1 cen tel
12.3
12.2
12.1 STAT6

Exon 22
Exon 21
Exon 20

Exon 19
Exon 18
Exon 17
Exon 16
Exon 15
Exon 14

Exon 13
Exon 12

Exon 11
Exon 10
11.2

Exon 9
Exon 8
Exon 7
Exon 6
Exon 5
Exon 4
Exon 3
Exon 2

Exon 1
11.1
11
12

13.1
13.2 NAB2 cen tel
13.3
STAT6 invert
14 cen tel

15

Exon 10
Exon 11

Exon 12
Exon 13

Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 19

Exon 20
Exon 21
Exon 22
Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6
Exon 7
Exon 8
Exon 9
21.1
21.2
21.3

22

23

24.1
24.2
24.31 cen tel
24.32 Fusion transcript
24.33
breakpoint
A NAB2 portion STAT portion

NAB2 STAT6
EBD NCD2 RD CCD1 DBD SH2 TAD
1 525 aa 1 847 aa
EBD - Early growth response protein 1 CCD1 - Coiled-coil domain
binding domain DBD - DNA-binding domain
NCD2 - NAB2 conserved domain
SH2 - Src (sarcoma) homology 2 domain
RD - Transcriptional repressor domain
TAD - Transcriptional activator domain
EBD NCD2 RD TAD
SFT-18 1 706 aa
EBD NCD2 RD SH2 TAD
SFT-3 1 779 aa
EBD NCD2 RD SH2 TAD
SFT-28 1 741 aa
EBD NCD2 RD CCD1 DBD SH2 TAD
SFT-31 1 1307 aa
EBD NCD2 RD CCD1 DBD SH2 TAD
SFT-40 1 1222 aa
EBD NCD2 RD SH2 TAD
SFT-44/10 1 755 aa
B
FIGURE 14-15  ■  Molecular structure of NAB2-STAT6 fusion gene and its chimeric protein in solitary fibrous tumor. A, Exon-intron
structures of NAB2 and STAT6 genes. The molecular structure of the resulting chimeric gene, which includes the entire STAT6 gene
after inversion and 3′ segment of NAB2, is shown. B, Functional domains of the two proteins are depicted. C, The structural variants
of the chimeric protein resulting from the alternative breakpoints of the two genes. Note that all variants contain the EBD (early
growth response protein 1 binding domain) of the NAB2 gene and TAD (transcriptional activator domain) of the STAT6 gene. (Adapted
from Robinson DR, et al: Identification of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor by integrative sequencing. Nature
Genetics 45:180–187, 2013).

ERRNVPHGLFRVRUJ
1010 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-16  ■  Myxopapillary ependymoma: radiographic features. A, T2-weighted sagittal magnetic resonance image (MRI) showing
signal enhancing circumscribed (“boxcar shaped”) mass in the lumbar cistern, characteristic of myxopapillary ependymoma.
B, T1-weighted sagittal postcontrast MRI showing signal enhancing circumscribed (“boxcar shaped”) mass in the lumbar cistern,
characteristic of myxopapillary ependymoma. C, T1-weighted sagittal MRI showing sacral myxopapillary ependymoma. D, T1-
weighted sagittal postcontrast MRI showing sacral myxopapillary ependymoma.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1011

A B

C D
FIGURE 14-17  ■  Myxopapillary ependymoma: microscopic features. A, Intermediate power photomicrograph of cytologic smear
preparation showing fibrillary cytoplasmic processes and myxoid material characteristic of myxopapillary ependymoma (×200).
B, Intermediate power photomicrograph showing prominent perivascular myxoid cuffing and myxoid microcysts (×200). C, Inter-
mediate power photomicrograph showing papillary architecture of sacral myxopapillary ependymoma (×200). D, High power pho-
tomicrograph showing dotlike and ringlike expression of epithelial membrane antigen as revealed by immunohistochemistry (×400).
(B and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1012 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

Radiographic Imaging Differential Diagnosis


Radiographically, the lesions are lytic, but a narrow scle- Microscopically, intraosseous schwannoma should be dis-
rotic zone may be present at the periphery (Fig. 14-18). tinguished from desmoplastic fibroma, well-differentiated
The bone contour may be expanded with no periosteal fibrosarcoma, fibrous dysplasia, and nonossifying fibroma.
reaction. The cortex may be thinned, but no true cortical In distinguishing cellular schwannoma from a malig-
destruction or extension into soft tissue is present. nant spindle-cell neoplasm, it is important to remem-
Although the lesions are primarily lytic, they may have ber that atypical mitoses are never present in benign
incomplete internal trabeculation or lobulation (Fig. schwannoma. Atypia in benign schwannoma is of the
14-18). The radiographic appearance is usually inter- degenerative type. Moreover, a hypercellular tumor with
preted as benign, but a specific diagnosis cannot be recognizable phenotypic features of Schwann cells is more
made from radiographic findings alone. It is difficult to likely to represent a benign schwannoma than a malig-
differentiate intraosseous schwannoma on radiographs nant spindle-cell neoplasm (also see the section on dif-
from other conditions that have similar radiographic ferential diagnosis of malignant peripheral nerve sheath
features, such as solitary bone cyst, chondroblastoma, tumor).92 The presence of specific radiographic and
chondromyxoid fibroma, and giant cell tumor.74-76,78,80,86 microscopic features usually distinguishes schwannoma
CT scans show sharp delineation of the tumor and may from other benign intraosseous conditions such as nonos-
disclose the dumbbell nature of the lesion (Figs. 14-19 sifying fibroma and fibrous dysplasia.
and 14-20). On MRI, schwannomas appear as uniformly In addition to its microscopic features, desmoplastic
contrast enhancing well-delineated lesions with an ana- fibroma can be ruled out by the absence of positivity for
tomic relationship to the adjacent nerve structures S-100 protein in fibroblastic cells.
(Figs.14-19 and 14-20).
Treatment and Behavior
Gross Findings
Intraosseous schwannomas are benign, and excision is
Intraosseous schwannomas are soft, tan-gray lesions with curative. Excision can be accomplished by enucleation or
frequent yellowish patchy areas. Hemorrhage, myxoid, curettage.
and cystic changes are frequently present. When the
lesion is present in a neural canal or foramen, a capsule
may be discernible. NEUROGENIC TUMOR
PREDISPOSITION SYNDROMES
Microscopic Findings
A comprehensive description of all aspects of neurogenic
The microscopic features of intraosseous schwannoma tumor predisposition syndromes is beyond the scope of
are similar to those of their more common soft tissue this chapter. Our description is restricted to the basic
counterparts.77,79 The tumors have two basic patterns: clinical and pathologic features with emphasis on the
hypercellular (Antoni A) and hypocellular (Antoni B) osseous manifestations. The clinical diagnostic criteria
(Fig. 14-21). In Antoni A areas of schwannoma cells that for the three forms of neurofibromatosis, referred to as
are compactly arranged form focally palisading structures types 1 to 3, are summarized in Table 14-3. These three
(Verocay bodies). In Antoni B areas, schwannoma cells forms are clinically different and have distinct predispos-
are widely separated by a loose intervening collagenous ing genetic mechanisms.163,177,178,188 We also include in
matrix. Vessels with thick hyalinized walls are present this section description of the skeletal manifestations of
focally. Variable numbers of histiocytes, macrophages, von Hippel-Lindau disease with its hallmark neoplasm,
lymphocytes, and mast cells may be present. Some hemangioblastoma.148,161,206
intraosseous schwannomas are hypercellular and may
have less distinguishable Antoni A and B areas. These Neurofibromatosis Type 1
lesions may exhibit some nuclear atypia, and occasional
mitoses may be present. These tumors are compatible Neurofibromatosis type 1, traditionally referred to as
with so-called cellular schwannomas and should be care- von Recklinghausen’s disease, is a congenital and familial
fully differentiated from malignant neoplasms.92 disorder with multiple manifestations that can involve
virtually any organ. Multiple soft tissue and cutaneous
neurofibromas and café au lait spots constitute the hall-
Special Techniques
mark dermatologic features of this condition. Although
Ultrastructural features of intraosseous schwannomas are neurofibromatosis type 1 primarily involves tissues of
the same as those described for soft tissue schwanno- neural crest origin, it also affects the mesoderm and
mas.77,81,87,90 Spindle-shaped tumor cells with abundant endoderm.103,135,137,177,178,185,196 Neurofibromatosis type 1 is
cytoplasmic processes are surrounded by bands of diagnosed if two or more features listed in Table 14-3 are
basal lamina (Fig. 14-22). Occasionally, desmosome-like present. Neurofibromatosis type 1 has an autosomal
junctions may be present. Multivesicular bodies and dominant pattern of inheritance with 50% pene-
long-spaced collagen fibers (Luse bodies) are also seen. trance.114,115,118 Roughly half of neurofibromatosis type 1
Immunohistochemically, the tumor cells are positive for patients have a documented family history of the disease,
vimentin and S-100 protein (Fig. 14-21). Text continued on p. 1018

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1013

A B

C D
FIGURE 14-18  ■  Intraosseous schwannoma: radiographic features. A, Lateral radiograph of mandible with lytic lesion produced by
expanding nerve sheath tumor arising in association with mandibular nerve. Note close relationship to mandibular canal and erosion
of root of molar. B, Radiograph of expansile tumor of pubic ramus in young adult with long history of groin pain. C, Anteroposterior
(AP) radiograph of sacrum showing expansile multiloculated lucent lesion. D, AP radiograph of sacrum showing expansile multi-
loculated lesion.

ERRNVPHGLFRVRUJ
1014 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-19  ■  Schwannoma: radiographic features. A, T1-weighted sagittal postcontrast magnetic resonance image (MRI) showing
supraorbital mass with uniform signal enhancement (arrow). Inset, Sagittal computed tomogram showing thinning of superior
orbital bone by schwannoma (arrow). B, T2-weighted coronal MRI showing brachial plexus schwannoma. C, T1-weighted axial
postcontrast MRI showing right intracanalicular schwannoma (arrow). D, T1-weighted axial postcontrast MRI showing right cerbel-
lopontine angle schwannoma.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1015

B C

D E
FIGURE 14-20  ■  Schwannoma: radiographic features. A, Axial computed tomogram (CT) showing dumbbell schwannoma with widen-
ing of the intervertebral foramen. B, T1-weighted axial postcontrast magnetic resonance image (MRI) showing uniform signal
enhancing dumbbell schwannoma. Same case as in A. C, CT 3D reconstruction showing widening of the L2–L3 intervertebral foramen
(arrow). Same case as in A and B. D, Sagittal CT showing sacral schwannoma. E, T1-weighted sagittal postcontrast MRI showing
signal enhancement in sacral schwannoma. Same case as in D.

ERRNVPHGLFRVRUJ
1016 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-21  ■  Schwannoma: microscopic features. A, Low power photomicrograph showing densely cellular (Antoni type A) and
hypocellular (Antoni type B) components of schwannoma (×100). B, Low power photomicrograph showing plexiform schwannoma
(×50). C, Intermediate power photomicrograph showing nuclear palisades (Verocay bodies) in schwannoma (×200). D, Expression
of S-100 protein in schwannoma (Verocay bodies) revealed by immunohistochemistry (×200). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1017

B
FIGURE 14-22  ■  Schwannoma: ultrastructural features. A, Spindle cells with abundant cytoplasmic processes surrounded by basal
lamina (×4000). Inset, Long-spaced collagen (Luse bodies) in intercellular matrix (×28,000). B, Higher magnification of interdigitating
cytoplasmic processes with basal lamina. Note relationship of long-spaced collagen to basal lamina (×8,000).

ERRNVPHGLFRVRUJ
1018 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

TABLE 14-3  Neurofibromatoses: Clinical Diagnostic Criteria


Neurofibromatosis Type 1*
Six or more café au lait macules (over 5 mm in greatest diameter in prepubertal individuals or over 15 mm in greatest
diameter in postpubertal individuals)
Two or more neurofibromas of any type OR one plexiform neurofibroma
Freckling in the axillary or inguinal region
Optic glioma (pilocytic astrocytoma of the optic pathway)
Two or more Lisch nodules (iris hamartomas)
A distinctive osseous lesion, such as sphenoid dysplasia or tibial pseudarthrosis
A first-degree relative (parent, sibling, or offspring) with NF1 as defined by the above criteria

Neurofibromatosis Type 2†
Confirmed (Definite) NF2
Bilateral vestibular schwannoma (“acoustic neuroma”)
Presumptive (Probable) NF2
Family history of NF2
Unilateral vestibular schwannoma OR any 2 of the following: meningioma, glioma, schwannoma, juvenile posterior
subcapsular lenticular opacity, juvenile cortical cataract
Suspicious for NF2 (evaluation of the patient is needed if either of the conditions are present)
Unilateral vestibular schwannoma AND at least 2 of any of the following: meningioma, glioma, schwannoma, juvenile
posterior subcapsular lenticular opacities/juvenile cortical cataract
Two or more meningiomas AND either unilateral vestibular schwannoma or any 2 of the following: glioma, schwannoma,
juvenile posterior subcapsular lenticular opacities/juvenile cortical cataract

Neurofibromatosis Type 3‡
Confirmed (Definite) Schwannomatosis
Two or more nonintradermal, nonvestibular schwannomas (at least 1 with histologic confirmation) in a patient older than age
30 years
One histologically confirmed schwannoma AND a first-degree relative who meets the above criterion for definite
schwannomatosis (regardless of patient age)
Possible Schwannomatosis
• Patients younger than age 30 years: 2 or more nonintradermal, nonvestibular (confirmed by imaging) schwannomas (at least
1 with histologic confirmation)
• Patients older than age 45 years: 2 or more nonintradermal, nonvestibular schwannomas (at least 1 with histologic
confirmation), with no evidence of eighth nerve dysfunction (but magnetic resonance imaging confirmation of absence of
vestibular schwannoma is not required)
• Radiographic (imaging) evidence of a nonintradermal, nonvestibular schwannoma AND a first-degree relative meeting the
criteria for definite schwannomatosis
Segmental Schwannomatosis
Meets criteria for either definite or possible schwannomatosis, but the schwannomas are limited to one limb or to five or
fewer contiguous segments of the spine

*NIH diagnostic criteria for NF1 are met in patients who exhibit at least two of the seven features.
†Clinical diagnostic criteria for confirmed and presumptive NF2, as well as features suspicious for NF2 that warrant additional clinical
investigation.
‡Patients diagnosed with schwannomatosis must not meet any of the criteria for definite or probable NF2, and thus must not have
evidence of a vestibular schwannoma (confirmed by imaging), a constitutional NF2 mutation, or a first-degree relative with NF2.

and the remaining half are the result of new mutations. suppressor factor.154,208 NF1 encoded protein shows
Neurofibromatosis type 1 is linked to a very wide variety extensive sequence similarities to two Saccharomyces cere-
of alterations (deletions, insertions, and mutations) as visiae (yeast) genes encoding IRA1and IRA2 proteins that
well as alternate splicing of the neurofibromatosis 1 are activating GTPase for yeast Ras and mammalian
(NF1) gene located in the pericentromeric region of chro- Ras-GAP proteins.154 The NF1 mutations result in hyper-
mosome 17 (Fig. 14-23).96,115,124,127,139,149,156,172,181,182,184,204 It activation of the Ras pathway similar to that caused by
is one of the largest genes of the human genome, span- Ras gene mutations (Fig. 14-23).93,191 It has been shown
ning a distance of 300 megabases (mb) and having a that neurofibromas in this syndrome may develop because
complex exon-intron structure.158 The product of this of additional secondary hits within the NF1 locus, such
gene, neurofibromin, is a structural component of the as a somatic deletion, that inactivate the normal allele of
microtubular system and is ubiquitously expressed in all the gene.115 It appears that the Schwann cell precursors
human tissues.114 Neurofibromin represents a class of are the primary targets of loss of NF1 function. The
guanosine triphosphatase–activating proteins that play a somatic loss of heterozygosity (LOH) in these cells
role in signal transduction, modulation of cell differentia- orchestrate the development of histologically complex
tion, and proliferation programs. NF1 cooperates with growth by inducing the co-proliferation of various cell
Ras genes and has the overall biologic activity of a tumor types involved in neural development by paracrine

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1019

NF1

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
Exon 17
Exon 18
Exon 19
Exon 20
Exon 22
Exon 23
Exon 24
Exon 25
Exon 26
Exon 27
Exon 28
Exon 29
Exon 30
Exon 31
Exon 32
Exon 33
Exon 34
Exon 35
Exon 36
Exon 37
Exon 38
Exon 39
Exon 40
Exon 41
Exon 42
Exon 43
Exon 44
Exon 45
Exon 46
Exon 47
Exon 48
Exon 49
Exon 50
Exon 51
Exon 52
Exon 53
Exon 54
Exon 55
Exon 56
Exon 57
Exon 58
Exon 1

Exon 2
Exon 3
Exon 4
Exon 5
Exon 6
Exon 7
Exon 8
Exon 9

Exon 1
Chr 17
13
12
11.2 11.1
11.1 NF1 NF1-NEUROFIBROMIN
12 11.2
21.2 21.1 1 2839 aa
21.3
22
23 Armadilo-type fold domain 278-2677
24
CSRD: cystein/serine-rich domain 543-909
25
TBD: tubulin-binding domain 1095-1194
GRD: GAP-related domain 1203-1549
Sec14/PH: Sec14-homologous domain 1560-1818
A NLS: nuclear localization sequence 2205-2785

INACTIVE
Ras stimulatory
Pi signal

GDP GEF

GAP blockage GTP


caused by
GDP
oncogenic
mutation

Ras

GTP

ACTIVE

+/-
Nf1
∆54 A1281 P1395 F1434 K1436 Schwann
E1264 cell
R1276 Q1426
N1430 LOH
K1423
C +/- -/- +/+
K1391 Nf1 Nf1 Nf1
mast Schwann mast
cell cell cell
α5c
N
R1413
NF91 L1425
recruitment of
G1404
fibroblasts and normal nerve
S1468 A1396
P1400
K1419
endothelial cells sheaths
ATCHSLLNKATVKEKKENKKS
Type II insert
B C neurofibroma

FIGURE 14-23  ■  Molecular mechanisms of neurofibromatosis Type 1. A, Chromosomal location, exonal structure, and functional
domains of the NF1 gene. B, The role of neurofibromin in the Ras signaling cycle. The Ras protein progresses the cycle, in which
it is activated by a guanine nucleotide exchange factor (GEF) and inactivated by a GTPase-activating protein (Ras-GAP). Neurofibro-
min (NF1) is one of the main Ras-GAPs that can stimulate Ras activity more than 1000-fold. The structure of the NF1 domain that
interacts with Ras, referred to as the arginine finger, is depicted. It carries an arginine (R1276) residue that is critical for the interac-
tion with the GTPase cleft of Ras. Mutations in the region that directly affect the R1276 residue and change the conformational status
of the domain activate the Ras pathway similar to oncogenic mutations directly affecting the Ras protein. (A and B, Modified and
reprinted with permission from Scheffzek K, et al: EMBO J 17:4313–4327, 1988; Weinberg RA: The biology of cancer, ed 2, New York,
2014, Garland Science.) C, The animal model data suggests that mast cells from bone marrow play an important role in the develop-
ing spinal cord where they contribute to the morphogenesis of the peripheral nerve. The absence of one copy of the NF1 gene
inactivated by the mutation makes them hypersensitive to signals released by the Schwann cells with NF1−/− genotype. This creates
a hypersensitive environment with aberrant morphogenesis of nerves and hyperproliferation of both fibroblasts and endothelial
cells. (C, Modified and reprinted with permission from Weinberg RA: The biology of cancer, ed 2, New York, 2014, Garland Science.)

ERRNVPHGLFRVRUJ
1020 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

signaling. The loss of NF1 function (NF1−/− genotype) alveolitis, megacolon, lipoma, neuroblastoma, carcinoid
is in the background of the NF1+/− genotype, which tumor, and Wilms’ tumor can occasionally be associated
might create a stimulatory hyperresponsive environment with neurofibromatosis type 1.
(Fig. 14-23).214 It has been shown that the recruitment of Variants of neurofibromatosis type 1 with incomplete
marrow cells, such as mast cells, plays a critical role in or aberrant features in clinical presentation include
the formation of plexiform neurofibromas.208,214 The segmental manifestation, manifestation restricted to gas-
development of malignant peripheral nerve sheath tumors trointestinal or spinal involvement, and the so-called
is associated with additional genomic alterations such as “familial” café au lait spots.
deletions of chromosome 17q and alterations of tumor
suppressor genes such as TP53.171,184 Molecular data Café Au Lait Spots.  Café au lait spots typically develop
support the clonal origin of malignant peripheral nerve first. They usually involve unexposed areas of the skin.
sheath tumors in neurofibromatosis.132 Similar to neuro- Initially, they are small and resemble freckles. They grad-
fibromas, malignant peripheral nerve sheath tumors ually increase in size and become darker. Common loca-
exhibit somatic deletions of the NF1 gene.156 These and tions are the trunk as well as the axillary and inguinal
earlier data support the clinicopathologic observation regions (Fig. 14-17). Eventually, more than 30% of
that malignant transformation in neurofibromatosis patients with neurofibromatosis type 1 develop these
typically occurs in a preexisting neurofibroma; that is, lesions. The number and size of café au lait spots cor-
malignant cells represent a subclone of an underlying respond to some extent to the severity of the disease.
neurofibroma.132 These insights still do not address many Patients with less prominent skin pigmentation may have
of the questions surrounding the formation of tumors a later or less severe onset of neurofibromatosis (i.e.,
associated with neurofibromatosis type 1. palpable neurofibromas). The changes in these patients
may be restricted to one major anatomic region. Some
patients may prove to have neurofibromatosis type 2.
Clinical Symptoms
The borders of the pigmented areas are less irregular
Clinical presentation of neurofibromatosis type 1 is than those associated with Albright-McCune syndrome
highly variable, but a combination of cardinal signs, listed (“coast of California” rather than “coast of Maine”
in Table 14-3, must be present to establish the diagnosis contours). They occur most often over the trunk and
of this syndrome. The severity and extent of the involve- axillary regions. They do not become confluent to the
ment varies dramatically from patient to patient and extent that skin lesions in Albright-McCune syndrome
across families (Figs. 14-24 and 14-25). Although myriads do. Rarely, the macular pigmentations may have jagged
of genetic alterations have been discovered involving the borders and become confluent. Microscopically, café au
NF1 gene, mysteriously, the type of alteration cannot be lait spots are characterized by an increased level of pig-
used to predict the various clinical manifestations of the mentation in the basal and peribasal layers of epidermis.
disorder. Typically, the disease becomes evident during The presence of giant melanosomes (macromelano-
the first few years of life and is diagnosed by the presence somes) is suggested by some authors to be pathogno-
of dermatologic or ophthalmic lesions when café au lait monic for neurofibromatosis.146 However, they are not
spots and Lisch nodules develop.116,145 The Lisch nodules always present, and their absence does not help to rule
are pigmented hamartomas of the iris, asymptomatic out neurofibromatosis in the differential diagnosis of skin
lesions that are helpful for establishing the diagnosis. Ini- pigmentations.194
tially, the skin lesions resemble freckles, but they enlarge
with age and become darker, predominantly involving the Neurofibroma.  The presence of multiple neurofibro-
areas unexposed to sun. The typical presenting sign is the mas is a hallmark of neurofibromatosis. Several types of
so-called axillary freckle sign. The presence of multiple neurofibromas may be present in neurofibromatosis.
skin neurofibromas is the hallmark of fibromatosis type 1 Multiple localized neurofibromas that form discrete
and they typically develop during childhood after the café nodules in the soft tissue are the most common lesions in
au lait spots become evident. Some of these tumors are well-developed cases of neurofibromatosis type 1. These
evident at birth; others continue to develop through late are frequently present in the subcutaneous tissue and
adulthood. They involve both superficial and deep nerves, dermis (Fig. 14-24). In fully developed cases, crops of
and the symptoms may be related to the involvement of these tumors ranging from small nodules that measure
nerves in various organs and anatomic sites. In addition several millimeters to several centimeters can be seen
to neurofibromas, patients with neurofibromatosis also (Fig. 14-25). Nodular or localized neurofibromas do not
develop central nervous system tumors, including optic differ from solitary neurofibromas unassociated with
nerve glioma and astrocytoma (Fig. 14-27). Nearly half neurofibromatosis. They are therefore the least charac-
of patients may have associated developmental skeletal teristic lesions and are not absolutely pathognomonic for
abnormalities varying from vertebral scalloping through neurofibromatosis. Nodular localized neurofibromas in
focal dysplasia to major disfiguring abnormalities (Figs. neurofibromatosis are usually larger than solitary neuro-
14-26, 14-27, 14-39, and 14-40). Gynecomastia may fibromas unassociated with neurofibromatosis.
develop in male patients. The association of neurofi- Radiographically, especially on CT and MRI, neuro-
bromatosis type 1 with schwannoma, pheochromocy- fibromas are well delineated signal-enhancing lesions
toma, ganglioneuroma, nephroblastoma, gastrointestinal that may show association with the major neighboring
stromal tumor, and leukemia has been reported. In addi- nerve structure (Figs. 14-26 and 14-27).
tion, such conditions as vascular aneurysms, fibrosing Text continued on p. 1025

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1021

B
FIGURE 14-24  ■  Neurofibromatosis type 1: clinical features. A, Facial cutaneous and subcutaneous neurofibromas in young man with
relatively few skin lesions. B, Café au lait skin pigmentation in discrete, nonconfluent distribution over trunk. Note relatively smooth
borders around pigmented areas.

ERRNVPHGLFRVRUJ
1022 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

B
FIGURE 14-25  ■  Neurofibromatosis type 1: clinical features. A, Elderly woman with extensive cutaneous neurofibromas and malignant
peripheral nerve sheath tumor arising in left brachial plexus (arrow). B, Posterior trunk showing extensive cutaneous neurofibro-
matous involvement.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1023

A B

C D
FIGURE 14-26  ■  Neurofibromatosis type 1: radiographic features. A, T1-weighted axial magnetic resonance image (MRI) showing a
plexiform neurofibroma involving the soft tissues of the head, sphenoid bone, and orbit. Note the characteristic “targetoid” pattern
of plexiform neuriofibroma. B, Anteroposterior radiograph of the head showing sphenoorbital dysplasia, which can occur alone or
in association with orbital neurofibroma. Same case as A. C, T1-weighted sagittal MRI showing characteristic scalloping of the
posterior aspects of the lumbosacral vertebral bodies that can be seen in NF1. D, Axial computed tomogram showing lumbar ver-
tebral body scalloping.

ERRNVPHGLFRVRUJ
1024 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-27  ■  Neurofibromatosis type 1: radiographic features. A, T1-weighted axial magnetic resonance image (MRI) showing foci
of abnormal T2 signal hyperintensity (“unidentified bright objects”) that are commonly seen in NF1. Bilateral basal ganglia lesions,
as illustrated here, are very common. B, T1-weighted axial postcontrast MRI showing bilateral gliomas of the optic pathway (optic
glioma). Optic gliomas are pilocytic astrocytomas. These can be unilateral or bilateral and can affect any part of the pregeniculate
optic pathway, including the optic nerves, chiasm, or tracts, or all of these components as seen here (double midbrain sign).
C, T2-weighted sagittal MRI showing multiple spinal neurofibromas arising at virtually every level of the spinal cord. D, T1-weighted
axial postcontrast MRI showing a biopsy-proven cerebellar glioblastoma. Diffuse gliomas are part of the spectrum of nervous system
tumors that can be seen in NF1.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1025

Grossly, these lesions are encapsulated and tan-gray. have neurofibromatosis. Enzinger and Weiss128 estimate
Neurofibromas growing within major nerves produce a that at least 10% of patients with diffuse neurofibroma
fusiform expansion. On longitudinal sections, a nerve and have neurofibromatosis. On the other hand, large diffuse
its structures pass through the lesion, and this mass is lesions of this type involving the head region in chil-
delineated by a perineurium. dren are almost always associated with neurofibroma-
Microscopically, the neurofibroma is composed of tosis. Microscopically, the tumor cells grow in a diffuse,
haphazardly arranged spindle cells with an abundance of infiltrative pattern along fibrous septa and various
extracellular stroma representing collagen and mucin adnexal skin structures (Fig. 14-31). The adnexal dermal
(Fig. 14-28). The cells are associated with strands of col- and subcutaneous structures are not destroyed by the
lagen separated by myxoid matrix. The overall architec- tumor.
ture of neurofibroma is frequently described as being
patternless as opposed to neurilemmoma, which is com- Schwannoma.  Schwannomas (neurilemmomas) are
posed of bundles of palisaded cells. Various inflammatory almost always solitary lesions. They rarely occur in asso-
cells, such as mast cells, lymphocytes, and xanthomatous ciation with neurofibromatosis type 1. In his review,
histiocytes, are frequently present. Myxoid change may Stout200 states that approximately 18% of patients with
be prominent, especially in larger lesions (Fig. 14-29). neurilemmoma also have neurofibromatosis type 1. The
Microscopically, these lesions may resemble soft tissue highly significant association of schwannoma and neuro-
myxoma. Schwann cells are usually present in neurofi- fibromatosis is also confirmed by observations made by
bromas and predominate focally in some tumors. Neuro- Enzinger and Weiss.128 Rarely (approximately 5% of
fibromas may contain heterologous elements, such as cases), schwannoma may exhibit a plexiform or multi-
glandular epithelial structures of various types, or even nodular growth pattern.95,128,213 This variant, however, is
foci resembling rhabdomyoma. Myelinated axons may be not associated with neurofibromatosis.95,130,212 These
demonstrated by neurofilament protein immunostaining associations were established before the current under-
or Luxol fast blue myelin staining. standing of the diversity of three forms of neurofibroma-
Immunohistochemically, neurofibroma cells are posi- tosis was fully clinically developed. A more contemporary
tive for vimentin and show variable focal positivity view is primarily related to the clear separation of neu-
for S-100 protein. Cells positive for S-100 protein may rofibromatosis type 2 and 3 with firm establishment of
in fact represent scattered individual Schwann cells. the concept that neurofibromatosis type 3, also referred
Uniform, strong positivity of tumor cells for S-100 to as schwannomatosis, is clinically distinct from neuro-
protein, typical in schwannoma, is not a feature of fibromatosis type 2. The large population based studies
neurofibroma. of schwannomas indicate that nearly 90% of schwanno-
Ultrastructurally, the majority of cells are consistent mas are solitary sporadic tumors and only 3% are associ-
with fibroblasts. Occasionally, Schwann cells surrounded ated with neurofibromatosis type 2. A distinct minority,
by basal lamina are present and are typically associated approximately 2% of schwannomas, are associated with
with the axonal processes that traverse the lesion neurofibromatosis type 3. Overall, 5% of schwannomas
(Fig. 14-32).90,110,155 occur together with meningiomas. This last association
Plexiform neurofibroma is pathognomonic for neurofi- is seen in patients with and without neurofibromatosis
bromatosis if stringent gross criteria are applied to its type 2. The alteration of the NF2 gene is a dominant
diagnosis. The plexiform variant of neurofibroma usually genetic abnormality in schwannomas regardless of
develops in early childhood. It affects large segments of whether they are sporadic or are associated with neuro-
a nerve, distorting its normal anatomic structures with fibromatosis type 2.
multiple nodules frequently described as “a bag of worms”
(Fig. 14-30). The identification of plexiform neurofi- Malignant Peripheral Nerve Sheath Tumor.  Malig-
broma for the diagnosis of neurofibromatosis should nant peripheral nerve sheath tumor represents the most
be confirmed by gross examination. The microscopic serious and often fatal complication of neurofibromato-
features of a plexiform growth pattern alone are not sis. This term encompasses previously used designations
pathognomonic for neurofibromatosis. Fully developed such as malignant schwannoma, neurofibrosarcoma, and neu-
plexiform neurofibromas are large lesions that involve rogenic sarcoma. These terms imply that the tumors reca-
one anatomic region or an entire extremity (elephantiasis pitulate or originate from specific cells of the nerve
neuromatosa). The skin overlying the affected region is sheath, such as Schwann cells, perineural fibroblasts, or
loose, redundant, and hyperpigmented. fibroblasts. Although this is true in some cases, many of
Microscopically, plexiform neurofibroma consists of these neoplasms show only focal specific differentiation,
tortuous nerve branches expanded by nodules (Fig. such as Schwann cells, whereas the majority of tumor
14-30). Nuclear pleomorphism and atypia with hyper- cells represent undifferentiated mesenchymal cells (Fig.
chromatism are often seen in neurofibroma, especially in 14-32). Phenotypic variation (divergent differentiation)
larger, older lesions. These features do not indicate is frequently seen in these tumors, which can exhibit dif-
malignant transformation. In plexiform lesions, individ- ferentiation toward nonneural elements such as rhabdo-
ual nodules are delineated by perineural cells that are myoblasts, smooth muscle, adipose tissues, cartilage,
positive for epithelial membrane antigen. bone, and epithelial glandlike structures. In fact, many of
Diffuse neurofibroma is the rarest form of neurofi- these lesions do not show neural differentiation and are
broma. It is not pathognomonic for neurofibromatosis; diagnosed as sarcomas of neural origin because of their
that is, not all patients with this form of neurofibroma Text continued on p. 1031

ERRNVPHGLFRVRUJ
1026 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-28  ■  Neurofibromatosis type 1: imaging, gross and microscopic features. A, Positron emission tomogram/computed
tomogram showing an FDG-avid tumor in the proximal upper arm. B, Gross photograph of neurofibroma resected en toto showing
prominent myxoid content as revealed by transillumination. C, Whole-mount cross-section of neurofibroma. D, Intermediate power
photomicrograph showing bland fusiform cells in myxoid background (×200). (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1027

A B

C D
FIGURE 14-29  ■  Neurofibroma: microscopic features. A, Low power photomicrograph of neurofibroma showing prominent myxoid
content (×100). B, Intermediate power photomicrograph showing concentration of axons in the center of neurofibroma, same case
as A (×200). C, High power photomicrograph showing spindle cells and wavy collagen of neurofibroma with inconspicuous inter-
spersed axons (×400). D, High power photomicrograph highlighting entrapped myelinated axons within neurofibroma as revealed
with a Luxol fast blue stain (×400). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1028 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C
FIGURE 14-30  ■  Plexiform neurofibroma: microscopic features. A, Low power photomicrograph showing tangled mass of enlarged
nerves (“bag of worms”) characteristic of plexiform neurofibroma (×100). B, Intermediate power photomicrograph showing plexi-
form neurofibroma with prominent myxoid background and central concentration of axons, same case as A (×200). C, High power
photomicrograph showing centrally preserved axons, same case as A (×400). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1029

A B

C D
FIGURE 14-31  ■  Cutaneous (intradermal) neurofibroma: microscopic features. A, Low power photomicrograph of diffuse form of
cutaneous (intradermal) neurofibroma (×100). B, Intermediate power photomicrograph showing characteristic spindle cells of neu-
rofibroma (×200). C, Low power photomicrograph of cutaneous plexiform neurofibroma (×100). D, Intermediate power photomicro-
graph showing whorl formation in a cutaneous plexiform neurofibroma (×200). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1030 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

FIGURE 14-32  ■  Neurofibroma: ultrastructural features. A, Low magnification showing spindle cells in collagenous stroma (×3500).
B, Higher magnification of Schwann cell processes invested with basal lamina. Cytoplasm of Schwann cell contains myelin sheath
(dark ring) surrounding an axon (×10,500). Inset shows spindle cell with fibroblastic features at higher magnification. Note absence
of basal lamina and prominent rough endoplasmic reticulum (×7200).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1031

association with major nerves or neurofibromatosis. For and hypercellular fascicles may be present. In addition,
all these reasons, the term malignant peripheral nerve various peculiar organizational patterns that most likely
sheath tumor is preferred for the designation of a unique reflect an attempt to recapitulate some neural structures
set of tumors that arise in major nerves or occur in a may be seen (Fig. 14-36). They include nodular, whorled,
clinical setting of neurofibromatosis type 1. and palisading structures. Focal divergent differentiation
Approximately 50% of all malignant peripheral nerve is present in 10% to 15% of these tumors.125,126,128 Islands
sheath tumors are associated with neurofibromatosis type of mature cartilage or bone are the most frequent
1. The early concepts regarding the relationship between examples of this phenomenon. On rare occasions, foci
these tumors and neurofibromatosis were complicated by of skeletal muscle differentiation (malignant Triton
different sets of diagnostic criteria used by different tumors) or glandular mucin-secreting structures can be
authors.142 The current consensus is that patients with seen.117,131,174,212 Focal epithelioid features are frequently
neurofibromatosis type 1 have a smaller risk of develop- seen, but a predominantly epithelioid appearance is a rare
ing malignant peripheral nerve sheath tumors than the phenomenon in these tumors.160 The spectrum of malig-
24% risk originally estimated. It is now believed that less nant peripheral nerve sheath tumors ranges from tumors
than 5% of patients with neurofibromatosis type 1 even- with deceptively benign appearances that resemble neu-
tually develop malignant peripheral nerve sheath tumors. rofibroma, except that they are hypercellular and show
This serious complication is typically seen in patients mitotic activity, to highly pleomorphic tumors whose
with long-term, fully developed disease (10 to 20 years) neural origin cannot be identified microscopically. The
and is occasionally multifocal. Malignant peripheral latter are diagnosed as malignant peripheral nerve sheath
nerve sheath tumors typically occur in patients between tumors because they arise in major nerves or occur in a
ages 20 and 50 years. Malignant peripheral nerve sheath setting of neurofibromatosis.
tumors, both solitary and associated with neurofibroma- Immunohistochemically, malignant peripheral nerve
tosis, are rare in children.3,169 The mean age of patients sheath tumors are focally positive for neural markers
diagnosed with these tumors in association with neurofi- such as S-100 protein, Leu-7, neurofilament protein,
bromatosis is lower than the mean age of patients who and myelin basic protein.121,128,140,147,160,168,209,211 Although
have sporadic malignant peripheral nerve sheath tumors. immunohistochemical evidence of neural differentiation
The peak incidence of malignant peripheral nerve sheath can be documented in more than 50% of patients, it is
tumors in neurofibromatosis is during the third decade typically only focal. S-100 protein is the marker most
of life. The peak incidence of sporadic malignant periph- often used. Neuron-specific enolase cannot be used alone
eral nerve sheath tumors is during the fifth decade of life. as evidence of neural differentiation because it is fre-
The male-to-female ratio of malignant peripheral nerve quently positive in many other lesions. S-100 protein is
sheath tumors associated with neurofibromatosis is 4 : 1; usually only focally positive, and strong uniform positiv-
80% occur in neurofibromatosis-affected men. Sporadic ity for this marker is rare in malignant peripheral nerve
malignant peripheral nerve sheath tumors are almost sheath tumors. In cases of strong uniform positivity for
equally distributed between the sexes. S-100 protein, other spindle-cell neoplasms (primarily
Most malignant peripheral nerve sheath tumors arise cellular schwannoma) should be excluded before the
in association with major nerves. The most frequently diagnosis of malignant peripheral nerve sheath tumor is
affected nerves include the sciatic nerve and the sacral rendered.
and brachial plexus. Consequently, most cases are located Ultrastructurally, malignant peripheral nerve sheath
within the soft tissue of proximal parts of the extremities, tumors have some of the features seen in benign periph-
in the soft tissues of the buttocks, and in the shoulder eral nerve sheath tumors (Fig. 14-37).7,110-112,129,140,202
region.3,143,197 Skeletal involvement usually stems from However, the ultrastructural examination of these lesions
secondary invasion into the adjacent bone (Figs. 14-33 often is disappointing and shows undifferentiated mesen-
and 14-34).108 Primary malignant peripheral nerve sheath chymal cells. Ultrastructural features suggestive of neural
tumors in bone are extremely rare. Bullock et al.104 differentiation include long, branching, and interdigitat-
reported such a primary tumor of the distal femur. After ing cytoplasmic processes that contain microtubules and
reviewing the literature, they found only 18 examples in neurofilaments. The extracellular matrix contains basal
bone, 15 of which occurred in the mandible or maxilla. lamina (frequently incomplete), collagen with focal fea-
In view of these data, primary intraosseous malignant tures of long-spaced collagen, and amianthoid fibers.
peripheral nerve sheath tumor is one of the rarest neo- Malignant peripheral nerve sheath tumors are highly
plasms of bone. aggressive and have a high propensity to metastasize to
Microscopically, a typical malignant peripheral nerve distant sites, predominantly the lung, liver, and oddly
sheath tumor is a spindle-cell sarcoma that resembles a enough, bone. In fact, most malignant peripheral nerve
fibrosarcoma in regard to its overall cellular structure and sheath tumors that involve the skeleton either are meta-
architectural organization. In contrast to typical fibrosar- static or represent secondary involvement of bone by a
coma, most malignant peripheral nerve sheath tumors are soft tissue lesion. Regional lymph node metastases are
histologically of high grade and show prominent nuclear extremely unusual in malignant peripheral nerve sheath
atypia. On longitudinal sections, the tumor cells are spin- tumors. Several studies have shown that malignant
dled, blunt ended, or comma shaped (Fig. 14-35). Per- peripheral nerve sheath tumors associated with neurofi-
pendicularly cut nuclei are oval or round. Similar to bromatosis are more aggressive than their sporadic coun-
fibrosarcoma, the cells are arranged in fascicles with an terparts not associated with neurofibromatosis, but others
occasional herringbone pattern. Alternating hypocellular believe that the clinical behavior of both variants is

ERRNVPHGLFRVRUJ
1032 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C
FIGURE 14-33  ■  Malignant peripheral nerve sheath tumor: radiographic features. A, T1-weighted coronal magnetic resonance image
(MRI) showing large malignant peripheral nerve sheath tumor of low signal intensity in brachial plexus. B, T1-weighted coronal
postcontrast MRI showing malignant peripheral nerve sheath tumor with heterogeneous signal enhancement in brachial plexus.
C, Axial computed tomogram showing large sacral malignant peripheral nerve sheath tumor.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1033

neurofibromatosis (approximately 20% to 30% 5-year


survival).100,119,120,122,125,126,133,136,166,192,198,199,207,210

Skeletal Anomalies.  The skeleton is affected in approx-


imately 80% of patients with neurofibromatosis. Skeletal
changes range from minor erosive or pressure defects
through localized dysplasia to major disfiguring anoma-
lies caused by abnormal bone patterning (Figs. 14-19,
14-27, 14-38, and 14-40).106,107,141,144,150,157,179 The erosive
defects are caused by soft tissue neurofibromas develop-
ing within nerves pressing through bone foramina or
involving nerves in the vicinity of bone.107 These types of
abnormalities are typically seen in the vertebral column
(enlargement of intervertebral foramina, pressure defects
of vertebral bodies, or both conditions) and in the ribs
(Figs. 14-26 and 14-27).106,107 Such abnormalities may
also be present in other parts of the skeleton. Scoliosis is
a frequent axial anomaly and is present in approximately
50% of patients. Scoliosis in neurofibromatosis typically
involves the lower thoracic region and is associated with
sharp angulation that can cause paraplegia. Frequently, it
is associated with kyphosis predominantly affecting the
cervical segment.
The most frequent skeletal anomaly is macrocrania,
which can be documented in nearly 90% of children with
neurofibromatosis. More significant changes in these
regions include the congenital absence of a portion of the
orbit or sphenoid bone or a defect involving the cranial
vault. Congenital calvarial defects adjacent to the lamb-
doidal suture are most frequent and often involve the left
side (Fig. 14-38). Another frequent site of this anomaly
is the posterior aspect of the sagittal suture.
Gracile or twisted-ribbon bones are the typical anom-
alies observed in the long bones (Fig. 14-39). Other
anomalies of the appendicular skeleton include pseudar-
throsis, focal overgrowth (gigantism), or dwarfism. The
anomalies of the long tubular bones may be caused by
abnormal appositional growth.176 The periosteum seems
to show abnormal attachment to the bone surface. In
response to trauma and subperiosteal hemorrhage, it is
easily detached from the underlying bone, causing large
subperiosteal hematomas. Poor callus formation and fre-
quent pseudarthrosis of long tubular bones complicating
fractures can also be related to these peculiar features
of malfunctioning periosteum in neurofibromatosis. The
mechanism of pseudarthrosis in neurofibromatosis,
however, is not clear and cannot be related only to trauma
because many of these changes represent congenital
defects (Fig. 14-40). Congenital pseudarthrosis usually
occurs in the tibia and in the bones of the forearm.
A B Patients with neurofibromatosis tend to develop mul-
tiple nonossifying fibromas that predominantly affect
FIGURE 14-34  ■  Malignant peripheral nerve sheath tumor: radio-
graphic features. A and B, Anteroposterior and lateral radio-
the metaphyseal parts of the long tubular bones (Fig.
graphs of tibia show erosion caused by malignant peripheral 14-41).165,176,192 Originally, Jaffe-Campanacci syndrome,
nerve sheath tumor in the overlying soft tissue. characterized by multifocal extensive nonossifying fibro-
mas that may involve the mandible and vertebrae and
are associated with café au lait pigmentation, various
similar. In several reports, the survival rates of patients nonskeletal anomalies, and mental retardation, was con-
with solitary malignant peripheral nerve sheath tumors sidered to be distinct from neurofibromatosis. More
were approximately twice as high (approximately 40% recent clinical data, however, indicate that patients with
to 50% 5-year survival) as those for patients with malig- Jaffe-Campanacci syndrome may have other stigmata of
nant peripheral nerve sheath tumors associated with Text continued on p. 1038

ERRNVPHGLFRVRUJ
1034 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-35  ■  Malignant peripheral nerve sheath tumor: microscopic features. A, Intermediate power photomicrograph showing
spindle-cell variant of malignant peripheral nerve sheath tumor (×200). B, High power photomicrograph showing compact fascicles
of malignant spindle cells with prominent mitotic activity (×400). C, Intermediate power photomicrograph showing epithelioid
malignant peripheral nerve sheath tumor (×200). D, High power photomicrograph showing epithelioid variant of malignant peripheral
nerve sheath tumor (×400) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1035

A B

C D
FIGURE 14-36  ■  Triton variant of malignant peripheral nerve sheath tumor: microscopic features. A, Low power photomicrograph
showing pleomorphic malignant peripheral nerve sheath tumor (×100). B, Intermediate power photomicrograph showing pleomor-
phic malignant peripheral nerve sheath tumor with spindle and epithelioid components, same case as A (×200). C, Intermediate
power photomicrograph showing strong expression of Myo-D as revealed by immunohistochemistry (×200). D, Intermediate power
photomicrograph showing strong expression of desmin (×200). (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1036 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

B
FIGURE 14-37  ■  Malignant peripheral nerve sheath tumor: ultrastructural features. A, Cytoplasm of malignant Schwann cell contain-
ing arrays of interdigitating cytoplasmic processes; (×7500). B, Higher magnification of A showing interdigitating cytoplasmic pro-
cesses. (×18,000).

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1037

B
FIGURE 14-38  ■  Neurofibromatosis: radiographic features. A, Lateral radiograph of skull in young adult shows geographic areas of
radiolucency in occipital region. B, Anteroposterior radiograph of skull shows large frontoparietal area of sharply defined radiolu-
cency representing absence of calvarial bone.

ERRNVPHGLFRVRUJ
1038 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B
FIGURE 14-39  ■  Neurofibromatosis: radiographic features. A and B, Anteroposterior and lateral radiographs of leg of child with con-
genital pseudarthrosis of tibia and lateral bowing deformity of fibula.

neurofibromatosis type 1.105,175 It may well be that this fied: neurofibroma, meningioma, glioma, schwannoma,
syndrome represents a unique, previously unrecognized, ocular juvenile posterior subcapsular ventricular opacity,
manifestation of neurofibromatosis. (See Chapter 9.) or cerebral calcifications (Table 14-3).
The typical clinical presentation is the onset of tin-
Neurofibromatosis Type 2 nitus or hearing loss during adolescence or early adult
life because of the development of bilateral acoustic
Neurofibromatosis type 2 occurs less frequently than schwannomas. They typically involve the vestibular
type 1, making up approximately 10% of all neurofibro- portion of the eighth cranial nerve. Café au lait spots and
matosis patients. It has an autosomal dominant pattern neurofibromas are rare and less developed or even com-
of transmission with a 95% penetrance.151,167 The gene pletely absent in neurofibromatosis type 2. Patients
that predisposes to the condition is located on chromo- affected with this syndrome also have an increased risk
some 22 and codes for a product designated merlin for other central nervous system tumors. They include
(moesin-ezrin-radixin–like protein).94,151,201 The protein schwannomas of other cranial nerves and cutaneous
plays a role in binding of the cytoskeleton matrix to the tissue, meningioma, ependymoma, and glioma (Figs.
cell membrane and may have the overall biologic activity 14-43 and 14-44). Schwannomas in neurofibromatosis
of a growth suppressor.151,162,190 Neurofibromatosis type 2 type 2 are similar to the more common solitary sporadic
is linked to mutations that inactivate the NF2 gene (Fig. schwannomas.
14-42).94,101,102,152,203 Neurofibromatosis type 2 is diag-
nosed if an individual has bilateral masses in the eighth Neurofibromatosis Type 3
cranial nerve (Fig. 14-43).98,123,134,153,163,164,173,187 It may also
be diagnosed if a first-degree relative has neurofibroma-
(Schwannomatosis)
tosis type 2 and a unilateral vestibular nerve mass is Neurofibromatosis type 3 is the third recognized
present, or if two of the following lesions can be identi- form of neurofibromatosis characterized by multiple

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1039

A B
FIGURE 14-40  ■  Neurofibromatosis: radiographic features. A and B, Severe bowing deformity of radius and remnant of proximal ulna
in patient with congenital pseudarthrosis of forearm. C, Clinical photograph showing deformity of left forearm in patient with
neurofibromatosis.

nonvestibular, nonintradermal schwannomas.97,109,180 The alleles (hits 2 and 3). The somatic mutation of the remain-
schwannomas may involve the spinal nerves (most ing NF2 allele (hit 4) is the concluding third step that
common), the peripheral soft tissues, or the nonvestibu- causes the loss of function of the two involved genes. The
lar cranial nerves (least common). Patients who exhibit a four hit, three step mechanism of SMARCB1 and NF2
SMARCB1 mutation may also develop meningiomas in inactivation is considered to be the dominant genetic
addition to schwannomas.170,189 In sharp contrast to the mechanism causing neurofibromatosis type 3.
other two, far more common, neurofibromatosis sub- Neurofibromatosis type 3 criteria are categorized as
types, the vast majority of neurofibromatosis type 3 cases either definitive or possible, with an additional set of cri-
result from new mutations, with only approximately 15% teria for segmental schwannomatosis (Table 14-3).97,98,180
having a verified familial history. The inactivation of two A diagnosis of definitive schwannomatosis requires
genes on chromosome 22, the NF2 gene and its neighbor either two (or more) nonintradermal schwannomas (one
gene SMARCB1, by four hits in three steps has been of which must be histologically confirmed) in a patient
proposed as a causative mechanism of neurofibromatosis older than age 30 years, or one histologically confirmed
type 3 (Fig. 14-45).193 The predisposing event is a germ- schwannoma plus a first-degree relative with defini-
line mutation in SMARCB1 (hit 1), which represents the tive schannomatosis (by the first set of criteria). Pos-
first step of the sequence and is followed by a somatic loss sible schwannomatosis can be diagnosed if a patient
of a portion of chromosome 22 (step 2) that contains the meets any one of three sets of criteria: (1) two (or more)
second SMARCB1 allele and one of the neighboring NF2 nonintradermal schwannomas (one of which must be

ERRNVPHGLFRVRUJ
1040 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A
FIGURE 14-41  ■  Neurofibromatosis: radiographic features. A, Plain radiograph of both knees of an 11-year-old girl with neurofibro-
matosis. Multiple radiolucent lesions in metaphyses of femora and tibiae bilaterally represent nonossifying fibromas. B, Lateral
radiograph of left knee of patient shown in A emphasizes eccentric location of nonossifying fibromas.

NF2
Exon 10

Exon 11
Exon 12
Exon 13
Exon 14

Exon 15
Exon 16
Exon 1
Exon 2
Exon 3
Exon 4

Exon 5
Exon 6
Exon 7
Exon 8

Exon 9

Chr 22
29999544

30079904

13
12
11.2 11.1 cen tel
11.1 11.2
12.1 NF2
12.3 12.2 NF2-MERLIN
13.2 13.1
13.3 1 595 aa
Unique N-terminal domain
FERM domain
Ezrin / radixin / moesin, C-terminal domain
FIGURE 14-42  ■  Molecular mechanisms of neurofibromatosis type 2. Chromosomal location, exonal structure, and functional domains
of the NF2 gene.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1041

A B

C D
FIGURE 14-43  ■  Neurofibromatosis type 2: radiographic features. A, T1-weighted axial postcontrast magnetic resonance image (MRI)
showing the definitive bilateral vestibular schwannomas of neurofibromatosis type 2. Additional tumors seen in this image are a
middle fossa meningioma and a contralateral optic nerve sheath meningioma. B, T1-weighted coronal postcontrast MRI showing
multiple meningiomas, which are frequently seen in neurofibromatosis type 2. C, T1-weighted axial postcontrast MRI showing a
spinal cord ependymoma. Spinal cord ependymoma is the most common intramedullary spinal cord tumor in neurofibromatosis
type 2. D, T2-weighted axial MRI showing metallic artifact associated with a cochlear implant commonly seen in MRI of neurofibro-
matosis type 2 patients. The signature lesions of neurofibromatosis type 2, bilateral vestibular schwannomas, often lead to hearing
loss that can be treated via cochlear implants.

ERRNVPHGLFRVRUJ
1042 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-44  ■  Neurofibromatosis type 2: radiographic features. A, T1-weighted sagittal postcontrast magnetic resonance image
(MRI) showing multiple spinal meningiomas. B, T2-weighted sagittal MRI of the same patient as A, highlighting the dura-based
origin of spinal meningiomas. C, T1-weighted sagittal postcontrast MRI showing multiple spinal schwannomas. D, T1-weighted
coronal postcontrast MRI, same patient as C, highlighting the bilateral nature of the peripheral nerve sheath tumors in this patient.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1043

cen
24129149 Germline Somatic
Exon 1
Exon 2
Exon 3 Hit 2
Exon 4
Exon 5 m – m

SMARCB1
Loss 22 NF2 mutation
Exon 6
Hit 3 Hit 4
Exon 7
Hit 1
Chr 22 + – m1
13 Exon 8
m +
12 Exon 9
11.2 11.1 24176705
11.1 SMARCB1 tel

6 Mb
12.1 11.2
12.3 NF2 cen
12.2 29999544 Schwannoma 1
13.2 13.1 Exon 1
13.3
Exon 2
Exon 3 + +
Exon 4
m – m
Exon 5
Exon 6 Loss 22 NF2 mutation
NF2

Exon 7
Exon 8
Exon 9
Exon 10
Exon 11 + – m2
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16
30079904 Schwannoma 2
A tel
B
FIGURE 14-45  ■  Molecular mechanisms of schwannomatosis (neurofibromatosis type 3). A, Chromosomal location and exonal struc-
tures of the vicinal SMARCB1 and NF2 genes on chromosome 22 involved in neurofibromatosis type 3. B, Four-hit mechanism
involving germline mutations of the SMARCB1 gene followed by somatic deletions of the remaining SMARCB1 copy and mutations
of NF2, combined with loss of the remaining NF2 allele, resulting in the complete inactivation of the two genes in multifocal
schwannomas.

histologically confirmed) in a patient younger than age Lindau disease.113 In 1904, von Hippel described similar
30 years; (2) two (or more) nonintradermal schwanno- patients, and finally in 1927 Lindau proposed the sys-
mas (one of which must be histologically confirmed) in a temic nature of the disorder and provided the link
patient older than age 45 years; or (3) imaging evidence of between the hereditary retinal angiomas and central
a schwannoma plus a first-degree relative with definitive nervous system hemangioblastomas.159,205 Von Hippel-
schwannomatosis. A diagnosis of segmental schwanno- Lindau disease is an autosomal dominant disorder char-
matosis is appropriate when a patient meets the criteria acterized by germline mutations of the VHL gene on
for either definitive or possible schwannomatosis, but the chromosome 3p25-26 (Fig. 14-47).148,186,206 Based on the
schwannomas are confined to a single limb or to five or pattern of mutations in the VHL gene, the disease is
fewer contiguous spine segmental levels. subdivided into von Hippel-Lindau disease subtype 1 and
Clinically the disease affects men and women with 2, characterized by deletions associated with nonsense
equal frequency and presents with multiple, often painful, and missense mutations and pure missense mutations,
schwannomas involving skin and soft tissue. Some patients respectively. Type 2 is subdivided into A, B, and C forms
may present with a striking segmental involvement. based on the patterns of missense mutations. The indi-
Although the development of multiple skin and soft tissue vidual subtypes have different risk levels for tumor devel-
schwannomas is a hallmark of the disease, some patients opment at various topographic sites. Physiologically, the
may develop tumors in the central cranial and spinal VHL gene encodes two alternative proteins via different
nerves (Fig. 14-46). inframe start codons. The longer form contains 213
amino acids and the short form consists of amino acid
residues 54 to 213 and lacks the acidic repeat domain.
VON HIPPEL-LINDAU DISEASE The three dimensional structure of the VHL protein
reveals the presence of two structural subdomains termed
In 1894, Collins reported two siblings who, in retrospect, α and β. The α domain binds and activates elongin C,
were affected by a disorder now termed von Hippel- cullin 2, elongin B, and Rbx1. The β domain is important

ERRNVPHGLFRVRUJ
1044 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-46  ■  Schwannomatosis (neurofibromatosis type 3): radiographic features. A-D, T1-weighted sagittal postcontrast mag-
netic resonance images showing spinal schwannomas at multiple vertebral levels in schwannomatosis.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1045

A
Chr 3 VHL

Exon 1

Exon 2

Exon 3
26 25
24.3 VHL
24.2 10183318

10195354
24.1
22 23
21.3
21.2
21.1 VHL
14.3 14.2
14.1 1 213 aa
13
von Hippel-Lindau disease tumor suppressor, beta/alpha domain
12
11.1 11.2
11.2 11.1
12
13.1
13.2
13.3
21
B Normoxia PHD OH
22 VHL HIF
23 O2 OH
24
25.2
25.1
25.3 EloC Proteasome
26.1 Ubiquitylation
26.2
26.3 O2 HIF
27
28
29 VEGF
HIF PDGF
PHD VHL TGF
HIF EPO
Hypoxia EloC and others

Endothelial cells
C
Pericyte

PDGF

TGF-
VEGF, TGF- Blood vessel
?
?
EPO Nuclear HIF VHL+/-
tumor cell

MMPs ?
TIMPs SDF-1
FIGURE 14-47  ■  Molecular mechanisms of von Hippel-Lindau disease. A, Chromosomal location, exonal structure, and functional
domains of the VHL gene. B, Role of pVHL in the hypoxia inducible factor (HIF) pathway. The VHL protein is an essential part of an
ubiquitin ligase protein complex that includes elongin C (EloC) and other partners. Under normal oxygenation, HIFα is hydroxylated
on two proline residues by prolyl hydroxylase 2 (PHD2), then conjugates to pHVL. It targets the complex for ubiquitination and
proteasomal degradation. Under hypoxic conditions, HIFα is stabilized, binds to HIF1β, and stimulates the expression of downstream
targets such as vascular endothelial growth factor (VEGF), platelet-derived growth factor β (PDGFβ), transforming growth factor α
(TGFα), and erythropoietin (EPO). The VHL mutations block the degradation pathway and mimic the effects of the hypoxic condi-
tions, stimulating the expression of multiple growth factors. (A and B, Modified and reprinted with permission from Richard S, et al:
Semin Cancer Biol 23:26–37, 2013.) C, Altered expression of HIF-responsive growth factors and proteases in pVHL-defective cells. The
VHL mutations block the degradation pathway and mimic the effects of the hypoxic conditions, stimulating the expression of multiple
growth factors. (C, Modified and reprinted with permission from Kaelin WG Jr: Annu Rev Pathol Mech Dis 2:145–173, 2007.)

ERRNVPHGLFRVRUJ
1046 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

for binding to hypoxia-inducible factor 1α (HIF1α). The blood vessels are occupied by lipidized stromal cells,
most significant binding substraits of the VHL protein which constitute the neoplastic element of the tumor
are the alpha subunits (HIF1-3α) of the heterodimeric (Fig. 14-49). This component of the tumor typically
transcription factor HIF (Fig. 14-47). The deregulation exhibits reactivity for inhibin-α, which is often used as a
of the HIF pathway appears to be a major functionally distinguishing marker to separate hemangioblastoma
significant target of VHL mutations, but there are many from metastatic renal cell carcinoma, which is also a
alternative interacting proteins, referred to as the pVHL- highly vascular tumor with greatly increased incidence in
associated proteins, that play contributory roles to its von Hippel-Lindau disease (Fig. 14-49).
biologic effects (Fig. 14-47).
Von Hippel-Lindau disease is characterized by the
frequent development of specific tumors in selected topo-
Papillary Endolymphatic Sac Tumor
graphic sites. Affected individuals develop hemangioblas- As the name implies, the endolymphatic sac tumor, also
tomas in the central nervous system, clear cell carcinomas referred to as Heffner tumor or aggressive middle ear papil-
of the kidney, pheochromocytomas of the adrenal gland, lary tumor, originates in close association with the endo-
extraadrenal paragangliomas, endolymphatic sac tumors, lymphatic sac, which, embryologically, is a neural crest
pancreatic microcystic adenomas, and pancreatic neuro- derivative and is located subjacent to the posteromedial
endocrine tumors. This description is restricted to skel- surface of the temporal bone.99,183,195 These two features
etal manifestations of von Hippel-Lindau disease. The explain many of the most important aspects of endolym-
hallmark neoplasm is hemangioblastoma. In addition, phatic sac tumor, such as clinical presentation, radio-
another characteristic von Hippel-Lindau disease tumor, graphic features, and immunophenotype. Endolymphatic
papillary endolymphatic sac tumor, originates within the sac tumors are locally aggressive, bone erosive tumors
petrous portion of the temporal bone; hence the rel- and, by virtue of their origin in immediate proximity to
evance of von Hippel-Lindau disease to the field of bone the inner ear structures, most commonly present with
pathology.138 hearing loss, tinnitus, and vertigo.

Hemangioblastoma Radiographic Imaging


Hemangioblastoma is the hallmark tumor of von Hippel- Papillary endolymphatic sac tumor, which originates
Lindau disease and most commonly arises in the cerebel- within the petrous portion of the temporal bone, erodes
lum, spinal cord, and brain stem. They are significantly the bone as it grows. Individual tumors may expand in
less common in the supratentorial compartment (such as one or in several directions. One classical presentation is
the cerebral hemispheres and sellar region). Very rarely, erosive growth out of the temporal bone into the medi-
they may arise in extraneural sites. ally located cerebellopontine angle (Fig. 14-50). The dif-
ferential diagnosis of cerebello-pontine angle mass always
includes endolymphatic sac tumor.
Radiographic Imaging
The characteristic imaging presentation is that of a Histologic Findings
spherical, avidly contrast-enhancing mass. Size varies
from minute, punctate lesions, most commonly seen in Endolymphatic sac tumors typically exhibit a papil-
the cerebellum, spinal cord, and spinal cord nerve root- lary and glandular architecture, with a columnar epi-
lets, to large tumors that are often accompanied by an thelium covering a fibrovascular stromal core.99,195 The
even larger cyst formation (Fig. 14-48). In the latter architecture is similar to that of choroid plexus papil-
instance, it is often the enlargement of the cyst that loma, which can also present as a cerebellopontine angle
incites clinical symptoms, secondary to mass effect com- mass, leading to potential misdiagnosis of papillary
pression of adjacent structures, and leads the patient to endolymphatic sac tumor as choroid plexus papilloma
seek clinical attention. (Fig. 14-50). Moreover, because papillary endolymphatic
In all anatomic locations, hemangioblastomas invari- sac tumor originates from the neural crest–derived endo-
ably closely approach the pial surface at some point lymphatic sac, the tumor can express neural antigens,
along their circumference, and small tumors are invari- such as glial fibrillary acidic protein (GFAP), similar
ably located very superficially, immediately subjacent to to choroid plexus papilloma. Resolution of this dif-
the pia. ferential diagnosis is quite easy, however, and requires
only the examination of the preoperative CT and MRI
studies, which will show erosion of the temporal bone
Histologic Findings
in endolymphatic sac tumor, but not in choroid plexus
Hemangioblastomas are highly vascular tumors that typi- papilloma.
cally display the full range of blood vessel caliber, from
large to medium to small arteries and veins. However, Treatment and Outcome
the signature vessel is the capillary (hence capillary heman-
gioblastoma), and all hemangioblastomas show a very rich The treatment for endolymphatic sac tumor is surgical
capillary bed. The relative prominence of the capillary resection, which can result in a cure in many cases. Local
network varies depending on the degree of engorgement recurrence can be treated with radiation therapy, with a
present in tissue sections. The intervening spaces between 50% cure rate.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1047

A B

C D
FIGURE 14-48  ■  Hemangioblastoma: radiographic and microscopic features. A, T1-weighted axial postcontrast magnetic resonance
image (MRI) showing small hemangioblastoma of the right temporal lobe (arrow). B, T1-weighted coronal postcontrast MRI showing
classical “cyst with enhancing mural nodule” appearance of cerebellar hemangioblastoma. C, T1-weighted sagittal postcontrast MRI
showing multiple hemangioblastomas located in the sellar region, cerebellum, and brain stem. D, T1-weighted sagittal postcontrast
MRI showing multiple minute hemangioblastomas of the spinal cord.

ERRNVPHGLFRVRUJ
1048 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

A B

C D
FIGURE 14-49  ■  Hemangioblastoma: microscopic features. A, Low power photomicrograph showing the characteristic highly vascular
architecture of hemangioblastoma with blood vessels of all sizes (×100). B, Intermediate power photomicrograph showing the dense
cellularity of hemangioblastoma (×200). C, High power photomicrograph showing characteristic foamy stromal cells interspersed
between a rich capillary network (×400). D, High power photomicrograph showing strong expression of inhibin-α by stromal cells
as revealed by immunohistochemistry (×400). (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1049

A B

C D
FIGURE 14-50  ■  Papillary endolymphatic sac tumor: radiographic and microscopic features. A, T1-weighted axial postcontrast mag-
netic resonance image (MRI) showing invasive tumor of the left sphenoid bone. B, Axial computed tomogram showing extensive
destruction of the left sphenoid bone. C, Intermediate power photomicrograph of endolymphatic sac tumor showing commonly
occurring papillary architecture (×100). D, Higher magnification photomicrographs showing expression of glial fibrillary acidic protein
(GFAP) in papillary endolymphatic sac tumor as revealed by immunohistochemistry (×200). (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1050 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

REFERENCES oma of the sphenoid wings. Am J Neuroradiol 15:555–560,


1994.
Meningioma 27. Tominaga T, Koshu K, Narita N, et al: Metastatic meningioma to
1. Aavikko M, Li SP, Saarinen S, et al: Loss of SUFU function in the second cervical vertebral body: a case report. Neurosurgery
familial multiple meningioma. Am J Hum Genet 91:520–526, 34:538–539, 1994.
2012. 28. van den Munckhof P, Christiaans Im Kenter SB, Baas F, et al:
2. Clark VE, Erson-Omay EZ, Serin A, et al: Genomic analysis of Germline SMARCB1 mutation predisposes to multiple meningio-
Non-NF2 meningiomas reveals mutations in TRAF7, KLF4, mas and schwannomas with preferential location of cranial menin-
AKT1, and SMO. Science 339:1077–1080, 2013. giomas at the falx cerebri. Neurogenetics 13:1–7, 2012.
3. Coffin CM, Dehner LP: Peripheral neurogenic tumors of the soft 29. Wasserkrug R, Peyser E, Lichtig C: Extracranial bone metas-
tissues in children and adolescents: a clinicopathologic study of tases from intracranial meningiomas. Surg Neurol 12:480–484,
139 cases. Pediatr Pathol 9:387–407, 1989. 1979.
4. Crawford TS, Kleinschmidt-DeMasters BK, Lillehei KO: Primary 30. Willen R, Gad A, Willen H, et al: Extracranial meningioma pre-
intraosseous meningioma: case report. J Neurosurg 83:912–915, senting as a nasal polyp. J Otorhinolaryngol Relat Spec 41:234–239,
1995. 1979.
5. Cruz-Sanchez FF, Miquel R, Rossi ML, et al: Clinicopathological 31. Younis G, Sawaya R: Intracranial osteolytic malignant meningio-
correlations in meningiomas: a DNA and immunohistochemical mas appearing as extracranial soft tissue masses. Neurosurgery
study. Histol Histopathol 8:1–8, 1993. 30:932–935, 1992.
6. Fujita T, Nakagawa H, Tsuruzono K, et al: Extradural temporal
meningioma directly extended to cervical bone: case report. Solitary Fibrous Tumor/Hemangiopericytoma Family Tumors
Neurol Med Chir 33:458–462, 1993. 32. Bisceglia M, Dimitri L, Giannatempo G, et al: Solitary fibrous
7. Ghobashy A, Tobler W: Intraosseous calvarial meningioma of the tumor of the central nervous system: report of an additional 5
skull presenting as a solitary osteolytic skull lesion: case report and cases with comprehensive literature review. Int J Surg Pathol
review of the literature. Acta Neurochir 129:105–108, 1994 19:476–486, 2011.
(review). 33. Bisceglia M, Galliani C, Giannatempo G, et al: Solitary fibrous
8. Kuroda R, Nakatani J, Yorimae A, et al: Clinical experience of tumor of the central nervous system: a 15-year literature survey
intraorbital optic nerve sheath meningioma: report of eight cases. of 220 cases. Adv Anat Pathol 18:356–392, 2011.
Neurol Med Chir 30:468–475, 1990. 34. Cardillo G, Lococo F, Carleo F, et al: Solitary fibrous tumors of
9. Landini G, Kitano M: Meningioma of the mandible. Cancer the pleura. Curr Opin Pulm Med 18:339–346, 2012.
69:2917–2920, 1992. 35. Chen H, Zeng XW, Wu JS, et al: Solitary fibrous tumor of the
10. Louis DN, Ohgaki H, Wiestler OD, et al: WHO classification of central nervous system: a clinicopathologic study of 24 cases. Acta
tumours of the central nervous system. In World Health Organiza- Neurochir 154:237–248, 2012.
tion Classification of Tumours, ed 4, Lyon, 2007, International 36. Chmielecki J, Crago AM, Rosenberg M, et al: Whole-exome
Agency for Research on Cancer. sequencing identifies a recurrent NAB2-STAT6 fusion in solitary
11. Meis JM, Ordóñez NG, Bruner JM: Meningiomas: an immuno- fibrous tumors. Nature Genet 45:131–132, 2013.
histochemical study of 50 cases. Arch Pathol Lab Med 110:934–937, 37. Demicco EG, Park MS, Araujo DM, et al: Solitary fibrous tumor:
1986. clinicopathological study of 110 cases and proposed risk assess-
12. Morild I, Bang G, Mork S: Hemangiopericytic meningioma with ment model. Mod Pathol 25:1298–1306, 2012.
extracranial spread to multiple sites without intracranial recur- 38. Fargen KM, Opalach KJ, Wakefield D, et al: The central nervous
rence. Clin Neuropathol 3:128–130, 1984. system solitary fibrous tumor: a review of clinical, imaging and
13. Nager GT, Heroy J: Hoeplinger M: Meningiomas invading pathologic findings among all reported cases from 1996 to 2010.
the temporal bone with extension to the neck. Am J Otolaryngol Clin Neurol Neurosurg 113:703–710, 2011.
4:297–324, 1983. 39. Gengler C, Guillou L: Solitary fibrous tumour and haemangio-
14. Oka K, Hirakawa K, Yoshida S, et al: Primary calvarial meningio- pericytoma: evolution of a concept. Histopathology 48:63–74,
mas. Surg Neurol 32:304–310, 1989 (review). 2006.
15. Olmsted WW, McGee TP: Prognosis in meningioma through 40. Hayashi Y, Uchiyama N, Hayashi Y, et al: A reevaluation of
evaluation of skull bone patterns. Radiology 123:375–377, 1977. the primary diagnosis of hemangiopericytoma and the clinical
16. Palmer JD, Cook PL, Ellison DW: Extracranial osseous metasta- importance of differential diagnosis from solitary fibrous tumor
ses from intracranial meningioma. Br J Neurosurg 8:215–218, of the central nervous system. Clin Neurol Neurosurg 111:34–38,
1994 (review). 2009.
17. Papini M, Chiantelli A, Cantini R, et al: Nasal meningioma: 41. Musyoki FN, Nahal A, Powell TI: Solitary fibrous tumor: an
report of a case. Acta Otorhinolaryngol Belg 43:335–338, 1989. update on the spectrum of extrapleural manifestations. Skeletal
18. Persky MS, Som ML: Olfactory groove meningioma with para- Radiol 41:5–13, 2012.
nasal sinus and nasal cavity extension: a combined approach. Oto- 42. Park MS, Araujo DM: New insights into the hemangiopericytoma/
laryngology 86:ORL-714–ORL-720, 1978. solitary fibrous tumor spectrum of tumors. Curr Opin Oncol 21:
19. Pompili A, Derome PJ, Visot A, et al: Hyperostosing meningio- 327–331, 2009.
mas of the sphenoid ridge: clinical features, surgical therapy, and 43. Penel N, Amela EY, Decanter G, et al: Solitary fibrous tumors
long-term observations—review of 49 cases. Surg Neurol 17:411– and so-called hemangiopericytoma. Sarcoma 2012 (e-pub ahead of
416, 1982. print).
20. Russell T, Moss T: Metastasizing meningioma. Neurosurgery 44. Robinson DR, Wu YM, Kalyana-Sundaram S, et al: Identification
19:1028–1030, 1986. of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor
21. Schnitt SJ, Vogel H: Meningiomas: diagnostic value of immuno- by integrative sequencing. Nature Genet 45:180–187, 2013.
peroxidase staining for epithelial membrane antigen. Am J Surg 45. Schweizer L, Koelsche C, Sahm F, et al: Meningeal hemangio-
Pathol 10:640–649, 1986. pericytoma and solitary fibrous tumors carry the NAB2-STAT6
22. Simpson MT, Sneddon KJ: Extracranial meningioma of the oral fusion and can be diagnosed by nuclear expression of STAT6
cavity. Br J Oral Maxillofac Surg 25:520–525, 1987. protein. Acta Neuropathol 125:651–658, 2013.
23. Singh RV, Yeh JS, Broome JC, et al: Primary ectopic intramuscu- 46. Travis WD: Sarcomatoid neoplasms of the lung and pleura. Arch
lar meningioma of the thigh. Clin Neurol Neurosurg 95:245–247, Pathol Lab Med 134:1645–1658, 2010.
1993. 47. Wang XQ, Zhou Q, Li ST, et al: Solitary fibrous tumors of the
24. Smith MJ, O’Sullivan J, Bhaskar SS, et al: Loss-of-function muta- central nervous system: clinical features and imaging findings in
tions in SMARCE1 cause an inherited disorder of multiple spinal 22 patients. J Comput Assisted Tomogr 37:658–665, 2013.
meningiomas. Nature Genet 45:295–299, 2013. 48. Wignall OJ, Moskovic EC, Thway K, et al: Solitary fibrous
25. Spittaler PJ, Johnston IH: Multiple extracranial meningiomas in tumors of the soft tissues: review of the imaging and clinical fea-
a child. Childs Nerv Syst 8:144–146, 1992. tures with histopathologic correlation. AJR 195:55–62, 2010.
26. Terstegge K, Schorner W, Henkes H, et al: Hyperostosis in 49. Yilmaz C, Kabatas S, Ozen OI, et al: Solitary fibrous tumor. J Clin
meningiomas: MR findings in patients with recurrent meningi- Neurosci 16:1578–1581, 2009.

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1051

Myxopapillary Ependymoma 77. De La Monte SM, Dorfman HD, Chandra R, et al: Intraosseous
50. Bale PM: Sacrococcygeal developmental abnormalities and schwannoma: histologic features, ultrastructure, and review of the
tumors in children Perspect Pediatr Pathol 8:9–56, 1984 (review). literature. Hum Pathol 15:551–558, 1984.
51. Biagini R, Demitri S, Orsini U, et al: Osteolytic extra-axial sacral 78. Dickson JH, Waltz TA, Fechner RE: Intraosseous neurilemmoma
myxopapillary ependymoma. Skeletal Radiol 28:584–589, 1999. of the third lumbar vertebra. J Bone Joint Surg Am 53:349–355,
52. Chan HS, Becker LE, Hoffman HJ, et al: Myxopapillary ependy- 1971.
moma of the filum terminale and cauda equina in childhood: 79. Fawcett KJ, Dahlin DC: Neurilemmoma of bone. Am J Clin
report of seven cases and review of the literature. Neurosurgery Pathol 47:759–766, 1967.
14:204–210, 1984. 80. Gordon EJ: Solitary intraosseous neurilemmoma of the tibia:
53. Ciapetta P, Raco A: Intrasacral ependymoma: report of 2 cases and review of intraosseous neurilemmoma and neurofibroma. Clin
review of the literature. Zentralbl Neurochir 45:326–328, 1984. Orthop 117:271–282, 1976.
54. Cruz-Sanchez FF, Rossi ML, Hughes JT, et al: An immunohisto- 81. Inoue S, Kikuchi I, Sakanashi Y: An electron microscopic study
logical study of 66 ependymomas. Histopathology 13:443–454, on neurilemmoma. Kumamoto Med J 26:120–125, 1973.
1988. 82. Jones HM: Neurilemmoma of bone. Br J Surg 41:63–65, 1953.
55. Davis C, Barnard RO: Malignant behavior of myxopapillary epen- 83. Junnarker RV: Neurilemmoma of bone. J Ind Med Assoc 59:374–
dymoma: report of three cases. J Neurosurg 62:925–929, 1985. 375, 1968.
56. Gerston KF, Suprun H, Cohen H, et al: Presacral myxopapillary 84. Kashima TG, Gibbons MRJP, Whitwell D, et al: Intraosseous
ependymoma presenting as an abdominal mass in a child. J Pediatr schwannoma in schwannomatosis. Skeletal Radiol 42:1665–1671,
Surg 20:276–278, 1985. 2013.
57. Gupta R, Rishi A, Suri V, et al: Sacral myxopapillary ependymoma 85. Polkey CE: Intraosseous neurilemmoma of the cervical spine
with extensive osteolysis. J Neurooncol 86:349–352, 2008. causing paraparesis and treated by resection and grafting. J Neurol
58. Helwig EB, Stern JB: Subcutaneous sacrococcygeal myxopapillary Neurosurg Psychiatr 38:776–781, 1975.
ependymoma: a clinicopathologic study of 32 cases. Am J Clin 86. Samter TG, Vellios F, Shafer WG: Neurilemmoma of bone:
Pathol 81:156–161, 1984. report of three cases with review of the literature. Radiology
59. Kindblom LG, Lodding P, Hagmar B, et al: Metastasizing myxo- 75:215–222, 1960.
papillary ependymoma of the sacrococcygeal region: a clinico- 87. Sian CS, Ryan SF: The ultrastructure of neurilemoma with
pathologic, light- and electron microscopic, immunohistochemical, emphasis on Antoni B tissue. Hum Pathol 12:145–160, 1981.
tissue culture, and cytogenetic analysis of a case. APMIS 94:79–90, 88. Sun Z, Sun L, Li T, et al: Intraosseous trigeminal schwannoma of
1986. mandible with intracranial extension. J Laryngol Otol 125:418–
60. Kumar ND, Misra K: Fine needle aspiration cytodiagnosis of 422, 2011.
subcutaneous sacrococcygeal myxopapillary ependymoma: a case 89. Verma A, Banerjee K, Verma A, et al: Maxillary neurilemmoma—
report. Acta Cytol 34:851–854, 1990. rarest of the rare tumour: report of 2 cases. Int J Surg Case Rep
61. Lemberger A, Stein M, Doron J, et al: Sacrococcygeal extradural 4:1044–1047, 2013.
ependymoma. Cancer 64:1156–1159, 1989. 90. Waggener JD: Ultrastructure of benign peripheral nerve sheath
62. Maiorana A, Fante R, Fano RA: Myxopapillary ependymoma of tumors. Cancer 19:699–709, 1966.
the sacrococcygeal region: report of a case. Pathologica 81:471– 91. Wang XJ, Hartley K, Holt GE, et al: Intracortical schwannoma
476, 1989. of the femur. Skeletal Radiol 2013 (epub ahead of print).
63. Metcalf JS, Balentine JD, Maize JC: Myxopapillary ependymoma 92. White W, Shiu MH, Rosenblum MK, et al: Cellular schwannoma:
of the cauda equina with extension to the skin of the sacrococ- a clinicopathologic study of 57 patients and 58 tumors. Cancer
cygeal region. Am J Dermatopathol 6:249–257, 1984. 66:1266–1275, 1990.
64. Miralbell R, Louis DN, O’Keeffe D, et al: Metastatic ependy- Neurogenic Tumor Predisposition Syndromes
moma of the sacrum. Cancer 65:2353–2355, 1990.
65. Moelleken SM, Seeger LL, Eckardt JJ, et al: Myxopapillary epen- 93. Abernathy CR, Colman SD, Kousseff BG, et al: Two NF1 muta-
dymoma with extensive sacral destruction: CT and MR findings. tions: frameshift in the GAP-related domain, and loss of two
J Comput Assist Tomogr 16:164–166, 1992. codons toward the 38 end of the gene. Hum Mutat 3:347–352,
66. Prins BA, Landsman JN: Sacrococcygeal myxopapillary ependy- 1994.
moma. Neth J Surg 41:47–48, 1989 (review). 94. Arai E, Tokino T, Imai T, et al: Mapping the breakpoint of a
67. Pulitzer DR, Martin PC, Collins PC, et al: Subcutaneous sacro- constitutional translocation on chromosome 22 in a patient with
coccygeal (“myxopapillary”) ependymal rests. Am J Surg Pathol NF2. Genes Chromosomes Cancer 6:235–238, 1993.
12:672–677, 1988. 95. Barbosa J, Hansen LS: Solitary multilobular schwannoma of the
68. Quraishi NA, Wolinsky JP, Bydon A, et al: Giant destructive oral cavity. J Oral Med 39:232–235, 1984.
myxopapillary ependymomas of the sacrum. J Neurosurg Spine 96. Barker D, Wright E, Nguyen K, et al: Gene for von Recklinghau-
12:154–159, 2010. sen neurofibromatosis is in the pericentromeric region of chromo-
69. Shors SM, Jones TA, Jhaveri MD, et al: Best cases from the AFIP: some 17. Science 236:1100–1102, 1987.
Myxopapillary ependymoma of the sacrum. Spine Imag Intervent 97. Baser ME, Friedman JM, Evans GR: Increasing the specificity of
26:S111–S116, 2006. diagnostic criteria for schwannomatosis. Neurology 66:730–732,
70. Sonneland PR, Scheithauer BW, Onofrio BM: Myxopapillary 2006.
ependymoma: a clinicopathologic and immunocytochemical study 98. Baser ME, Friedman JM, Joe H, et al: Empirical development of
of 77 cases. Cancer 56:883–893, 1985. improved diagnostic criteria for neurofibromatosis 2. Genet Med
71. Specht CS, Smith TW, DeGirolami U, et al: Myxopapillary epen- 13:576–581, 2011.
dymoma of the filum terminale: a light and electron microscopic 99. Bell D, Gidley P, Levine N, et al: Endolymphatic sac tumor
study. Cancer 58:310–317, 1986. (aggressive papillary tumor of middle ear and temporal bone): sine
72. Wani NA, Mir F, Qayum A, et al: Sacrococcygeal extraspinal qua non radiology-pathology and the University of Texas MD
ependymoma. Acta Neurochir 152:917–918, 2010. Anderson Cancer Center experience. Ann Diagn Pathol 15:117–
73. Woyke S, Czerniak B: Fine needle aspiration cytology of meta- 123, 2011.
static myxopapillary ependymoma. Acta Cytol 22:312–315, 1978. 100. Bojsen-Moller M, Myrhe-Jensen O: A consecutive series of 30
malignant schwannomas: survival in relation to clinicopathologi-
cal parameters and treatment. Acta Pathol Microbiol Immuno Scand
Intraosseous Schwannoma (A) 92:147–155, 1984.
74. Agha FP, Lilienfeld RM: Roentgen features of osseous neurilem- 101. Bourn D, Carter SA, Mason S, et al: Germline mutations in the
moma. Radiology 102:325–326, 1972. neurofibromatosis type 2 tumour suppressor gene. Hum Mol Genet
75. Benazzo F, Marullo M, Rossi SMP, et al: Giant intraosseous 3:813–816, 1994.
schwannoma of the ileopubic ramus. Orthopedics 36:e982–e985, 102. Bourn D, Evans G, Mason S, et al: Eleven novel mutations in the
2013. NF2 tumour suppressor gene. Hum Genet 95:572–574, 1995.
76. Conley AH, Miller DS: Neurilemmoma of bone: a case report. 103. Brasfield RD, Das Gupta TK: Von Recklinghausen’s disease: a
J Bone Joint Surg 24A:684–689, 1962. clinicopathological study. Ann Surg 175:86–104, 1972.

ERRNVPHGLFRVRUJ
1052 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

104. Bullock MJ, Bedard YC, Bell RS, et al: Intraosseous malignant 131. Foraker AG: Glandlike elements in a peripheral neurosarcoma.
peripheral nerve sheath tumor: report of a case and review of the Cancer 1:286–290, 1948.
literature. Arch Pathol Lab Med 119:367–370, 1995. 132. Friedman JM, Fialkow PJ, Greene CL, et al: Probable clonal
105. Campanacci M, Laus M, Boriani S: Multiple non-ossifying fibro- origin of neurofibrosarcoma in a patient with hereditary neurofi-
mata with extraskeletal anomalies: a new syndrome? J Bone Joint bromatosis. J Natl Cancer Inst 69:1289–1292, 1982.
Surg Br 65:627–632, 1983. 133. Ghosh BC, Ghosh L, Huvos AG, et al: Malignant schwannoma:
106. Casselman ES, Mandell GA: Vertebral scalloping in neurofibro- a clinicopathologic study. Cancer 31:184–190, 1973.
matosis. Radiology 131:89–94, 1979. 134. Gonzalvo A, Fowler A, Cook RJ, et al: Schwannomatosis,
107. Casselman ES, Miller WT, Shu Ren Lin, et al: Von Recklinghau- sporadic schwannomatosis, and familial schwannomatosis: a surgi-
sen’s disease: incidence of roentgenographic findings with a clini- cal series with long-term follow-up. J Neurosurg 114:756–762,
cal review of the literature. Crit Rev Diagn Imaging 9:387–419, 2011.
1977. 135. Goodnough CP, Kuhlmann RP, Stark E: Von Recklinghausen’s
108. Charache H: Multiple neurofibroma with sarcomatous transfor- neurofibromatosis. NY State J Med 75:2407–2409, 1975.
mation and skeletal involvement. Arch Dermatol Syph 40:185–191, 136. Guccion JG, Enzinger FM: Malignant schwannoma associated
1939. with von Recklinghausen’s neurofibromatosis. Virchows Arch
109. Chen S, Liu C, Liu B, et al: Schwannomatosis: a new member of (Pathol Anat) 383:43–57, 1979.
neurofibromatosis family. Chin Med J 126:2656–2660, 2013. 137. Gutmann DH, Collins FS: Recent progress toward understanding
110. Chino F, Tsuruhara T: Electron microscopic study of von Reck- the molecular biology of von Recklinghausen’s neurofibromatosis.
linghausen’s disease. Jpn J Med Sci 21:249–257, 1968. Ann Neurol 31:555–561, 1992.
111. Chitale AR, Dickersin GR: Electron microscopy in the diagnosis 138. Hamazaki S, Yoshida M, Yao M, et al: Mutation of von Hippel-
of malignant schwannomas: a report of six cases. Cancer 51:1448– Lindau tumor suppressor gene in a sporadic endolymphatic sac
1461, 1983. tumor. Hum Pathol 32:1272–1276, 2001.
112. Chiu HF, Troster M: Ultrastructure of malignant schwannomas. 139. Heim RA, Kam-Morgan LN, Binnie CG, et al: Distribution of
Lab Invest 40A:246–250, 1979. 13 truncating mutations in the neurofibromatosis 1 gene. Hum
113. Collins ET: Intra-ocular growths (two cases, brother and sister, Mol Genet 4:975–981, 1995.
with peculiar vascular new growth, probably retinal, affecting both 140. Herrera GA, de Moraes HP: Neurogenic sarcomas in patients
eyes). Trans Ophthalmol Soc 14:141–149, 1894. with neurofibromatosis (von Recklinghausen’s disease): light, elec-
114. Colman SD, Wallace MR: Neurofibromatosis type I. Eur J Cancer tron microscopy and immunohistochemistry study. Virchows Arch
30:1974–1981, 1994. (Pathol Anat) 403:361–376, 1984.
115. Colman SD, Williams CA, Wallace MR: Benign neurofibromas 141. Holt JF, Wright EM: The radiologic features of neurofibromato-
in type 1 neurofibromatosis (NF1) show somatic deletions of the sis. Radiology 51:647–663, 1948.
NF1 gene. Nature Genet 11:90–92, 1995. 142. Hosoi K: Multiple neurofibromatosis (von Recklinghausen’s
116. Crawford AH: Neurofibromatosis in children. Am Fam Physician disease) with special reference to malignant transformation. Arch
17:163–170, 1978. Surg 22:258–281, 1931.
117. Cross PA, Clarke NW: Malignant nerve sheath tumor with epi- 143. Hruban RH, Shiu MH, Senie RT, et al: Malignant peripheral
thelial elements. Histopathology 12:547–549, 1988. nerve sheath tumors of the buttock and lower extremity: a study
118. Crowe FW, Schull WJ, Neel JV: A clinical, pathological, and genetic of 43 cases. Cancer 66:1253–1265, 1990.
study of multiple neurofibromatosis, Springfield, Ill, 1956, Charles C. 144. Hunt JC, Pugh DG: Skeletal lesions in neurofibromatosis. Radiol-
Thomas. ogy 76:1–20, 1961.
119. D’Agostino AN, Soule EH, Miller RH: Primary malignant neo- 145. Izumi AK, Rosato FE, Wood MG: Von Recklinghausen’s disease
plasm of nerves (malignant neurilemomas) in patients without associated with multiple neurilemomas. Arch Dermatol 104:172–
manifestations of multiple neurofibromatosis (von Recklinghau- 176, 1971.
sen’s disease). Cancer 16:1003–1014, 1963. 146. Jimbow K, Szabo G, Fitzpatrick TB: Ultrastructure of giant pig-
120. D’Agostino AN, Soule EH, Miller RH: Sarcomas of the periph- mented granules (macromelanosomes) in the cutaneous pig-
eral nerves and somatic soft tissues associated with multiple mented macules of neurofibromatosis. J Invest Dermatol 61:
neurofibromatosis (von Recklinghausen’s disease). Cancer 16: 300–309, 1973.
1015–1027, 1963. 147. Johnson MD, Glick AD, Davis BW: Immunohistochemical evalu-
121. Daimaru Y, Hashimoto H, Enjoji M: Malignant peripheral nerve ation of Leu-7, myelin basic protein, S-100 protein, glial fibrillary
sheath tumors (malignant schwannomas): an immunohistochemi- acidic protein, and LN3 immunoreactivity in nerve sheath tumors
cal study of 29 cases. Am J Surg Pathol 9:434–444, 1985. and sarcomas. Arch Pathol Lab Med 112:155–160, 1988.
122. Das Gupta TK, Brasfield RD: Solitary malignant schwannoma. 148. Kaelin WG, Jr: von Hippel-Lindau disease. Annu Rev Pathol Mech
Ann Surg 171:419–428, 1970. Dis 2:145–173, 2007.
123. Dirks MS, Butman JA, Kim HJ, et al: Long-term natural history 149. Kayes LM, Burke W, Riccardi VM, et al: Deletions spanning the
of neurofibromatosis type 2-associated intracranial tumors. J Neu- neurofibromatosis 1 gene: identification and phenotype of five
rosurg 117:109–117, 2012. patients. Am J Hum Genet 54:424–436, 1994.
124. Dublin S, Riccardi VM, Stephens K: Methods for rapid detection 150. Klatte EC, Franken EA, Smith JA: The radiographic spectrum in
of a recurrent nonsense mutation and documentation of pheno- neurofibromatosis. Semin Roentgenol 11:17–33, 1976.
typic features in neurofibromatosis type 1 patients. Hum Mutat 151. Kley N, Whaley J, Seizinger BR: Neurofibromatosis type 2 and
5:81–85, 1995. von Hippel–Lindau disease: from gene cloning to function. Glia
125. Ducatman BS, Scheithauer BW: Malignant peripheral nerve 15:297–307, 1995 (review).
sheath tumors with divergent differentiation. Cancer 54:1049– 152. Kluwe L, Mautner VF: A missense mutation in the NF2 gene
1057, 1984. results in moderate and mild clinical phenotypes of neurofibro-
126. Ducatman BS, Scheithauer BW, Piepgras DG, et al: Malignant matosis type 2. Hum Genet 97:224–227, 1996.
peripheral nerve sheath tumors: a clinicopathologic study of 120 153. Koontz NA, Wiens AL, Agarwal A, et al: Schwannomatosis:
cases. Cancer 57:2006–2021, 1986. The overlooked neurofibromatosis? Neuroradiology/Head and Neck
127. Eisenbarth I, Hoffmeyer S, Kaufmann D, et al: Analysis of an Imaging 200:W646–W653, 2013.
alternatively spliced exon of the neurofibromatosis type I gene in 154. Largaespada DA, Brannan CI, Jenkins NA, et al: Nf1 deficiency
cultured melanocytes from patients with neurofibromatosis 1. causes Ras-mediated granulocyte/macrophage colony stimulating
Arch Dermatol Res 287:413–416, 1995. factor hypersensitivity and chronic myeloid leukaemia. Nat Genet
128. Enzinger FM, Weiss SW: Soft tissue tumors, ed 3, St Louis, 1995, 12:137–143, 1996.
Mosby. 155. Lassmann H, Jurecka W, Lassmann G, et al: Different types of
129. Erlandson RA, Woodruff JM: Peripheral nerve sheath tumors: an benign nerve sheath tumors: light microscopy, electron micros-
electron microscopic study of 43 cases. Cancer 49:273–287, 1982. copy, and autoradiography. Virchows Arch (Pathol Anat) 375:197–
130. Fletcher CDM, Davies SE: Benign plexiform (multinodular) 210, 1977.
schwannoma: a rare tumor unassociated with neurofibromatosis. 156. Legius E, Marchuk DA, Collins FS, et al: Somatic deletion of the
Histopathology 10:971–980, 1986. neurofibromatosis type 1 gene in a neurofibrosarcoma supports a

ERRNVPHGLFRVRUJ
14  Neurogenous Tumors and Neurofibromatosis Affecting Bone 1053

tumour suppressor gene hypothesis. Nature Genet 3:122–126, 183. Rao Q, Zhou J, Wang JD, et al: Endolymphatic sac tumor with
1993. von Hippel-Lindau desase: report of a case with analysis of von
157. Levene LJ: Bone changes in neurofibromatosis. Arch Intern Med Hippel-Lindau gene and review. Ann Diagn Pathol 14:361–364,
103:570–580, 1959. 2010.
158. Li Y, O’Connell P, Breidenbach HH, et al: Genomic organiza- 184. Reynolds JE, Fletcher JA, Lytle CH, et al: Molecular character-
tion of the neurofibromatosis 1 gene (NF1). Genomics 25:9–18, ization of a 17q11.2 translocation in a malignant schwannoma cell
1995. line. Hum Genet 90:450–456, 1992.
159. Lindau A: Zur Frage der Angiomatosis Retinae und Ihrer Hirn- 185. Riccardi VM: Von Recklinghausen neurofibromatosis. N Engl J
complikation. Acta Ophthalmol 4:193–226, 1927. Med 305:1617–1627, 1981.
160. Lodding P, Kindblom LG, Angervall L: Epthelioid malignant 186. Richard S, Gardie B, Couve S, et al: von Hippel-Lindau: How a
schwannoma: a study of 14 cases. Virchows Arch (Pathol Anat) rare disease illuminates cancer biology. Semin Cancer Biol 23:26–
409:433–451, 1986. 37, 2013.
161. Lonser RR, Glenn GM, Walther M, et al: von Hippel-Lindau 187. Rodriguez HA, Berthrong M: Multiple primary intracranial
disease. Lancet 361:2059–2067, 2003. tumors in von Recklinghausen’s neurofibromatosis. Arch Neurol
162. Lutchman M, Rouleau GA: The neurofibromatosis type 2 gene 14:467–475, 1966.
product, schwannomin, suppresses growth of NIH 3T3 cells. 188. Rodriguez FJ, Stratakis CA, Evans DG: Genetic predisposition to
Cancer Res 55:2270–2274, 1995. peripheral nerve neoplasia: diagnostic criteria and pathogenesis of
163. MacCollin M, Chiocca EA, Evans DG, et al: Diagnostic criteria neurofibromatoses, Carney complex, and related syndromes. Acta
for schwannomatosis. Neurology 64:1838–1845, 2005. Neuropathol 123:349–367, 2012.
164. MacCollin M, Woodfin W, Kronn D, et al: Schwannomatosis: a 189. Rosseau G, Noguchi T, Bourdon V, et al: SMARCB1/INI1 germ-
clinical and pathologic study. Neurology 46:1072–1079, 1996. line mutations contribute to 10% of sporadic schwannomatosis.
165. Mandell GA, Dalinka MK, Coleman BG: Fibrous lesions in the BMC Neurol 11:1–3, 2011.
lower extremities in neurofibromatosis. Am J Roentgenol 133:1135– 190. Rouleau GA, Merel P, Lutchman M, et al: Alterations in a new
1138, 1979. gene encoding a putative membrane-organizing protein causes
166. Mannarino E, Watts JW: Malignant tumors arising from periph- neuro-fibromatosis type 2. Nature 363:515–521, 1993.
eral nerves. J Int Coll Surg 37:550–553, 1962. 191. Scheffzek K, Ahmadian MR, Wiesmuller L, et al: Structural anal-
167. Martuza RL, Eldridge R: Neurofibromatosis 2 (bilateral acoustic ysis of the GAP-related domain from neurofibromin and implica-
neurofibromatosis). N Engl J Med 318:684–688, 1988. tions. EMBO J 17:4313–4327, 1998.
168. Matsunou H, Shimoda T, Kakimoto S, et al: Histopathologic and 192. Schwartz AM, Ramos RM: Neurofibromatosis and multiple non-
immunohistochemical study of malignant tumors of peripheral ossifying fibromas. Am J Roentgenol 135:617–619, 1980.
nerve sheath (malignant schwannoma). Cancer 56:2269–2279, 193. Sestini R, Bacci C, Provenzano A, et al: Evidence of a four-hit
1985. mechanism involving SMARCB1 and NF2 in schwannomatosis-
169. Meis JM, Enzinger FM, Martz KL, et al: Malignant peripheral associated schwannomas. Hum Mutat 29:227–231, 2008.
nerve sheath tumors (malignant schwannoma) in children. Am J 194. Silvers DN, Greenwood RS, Helwig EB: Café-au-lait spots
Surg Pathol 16:694–707, 1992. without giant pigment granules: occurrence in suspected neurofi-
170. Melean G, Velasco A, Hernandez-Imaz E, et al: RNA-based bromatosis. Arch Dermatol 110:87–88, 1974.
analysis of two SMARCB1 mutations associated with familial 195. Skalova A, Sima R, Bohus P, et al: Endolymphatic sac tumor
schwannomatosis with meningiomas. Neurogenetics 13:267–274, (aggressive papillary tumor of middle ear and temporal bone).
2012. Report of two cases with analysis of the VHL gene. Pathol Res
171. Menon AG, Anderson KM, Riccardi VM, et al: Chromosome 17p Pract 204:599–606, 2008.
deletions and p53 gene mutations associated with the formation 196. Snyder SH: Nerve growth in neurofibromatosis. N Engl J Med
of malignant neurofibrosarcomas in von Recklinghausen’s neuro- 290:626–627, 1974 (letter).
fibromatosis. Proc Natl Acad Sci USA 87:5435–5439, 1990. 197. Sordillo PP, Helson L, Hajdu SI, et al: Malignant schwannoma:
172. Merel P, Khe HX, Sanson M, et al: Screening for germ-line muta- clinical characteristics, surgery, and response to therapy. Cancer
tions in the NF2 gene. Genes Chromosomes Cancer 12:117–127, 47:2503–2509, 1981.
1995. 198. Sorensen SA, Mulvihill JJ, Nielsen A: Long-term follow-up of von
173. Merker VL, Esparza S, Smith MJ, et al: Clinical features of Recklinghausen neurofibromatosis: survival and malignant neo-
schwannomatosis: a retrospective analysis of 87 patients. Oncologist plasms. N Engl J Med 314:1010–1015, 1986.
17:1317–1322, 2012. 199. Stewart FW, Copeland MM: Neurogenic sarcoma. Am J Cancer
174. Michel SL: Epithelial elements in a malignant neurogenic tumor 15:1235–1237, 1931.
of the tibial nerve. Am J Surg 113:404–408, 1967. 200. Stout AP: The peripheral manifestations of specific nerve sheath
175. Mirra JM, Gold RH, Rand F: Disseminated nonossifying fibromas tumor (neurilemoma). Am J Cancer 24:751–796, 1935.
in association with café-au-lait spots (Jaffe-Campanacci syn- 201. Takeshima H, Izawa I, Lee PS, et al: Detection of cellular proteins
drome). Clin Orthop 168:192–205, 1982. that interact with the NF2 tumor suppressor gene product. Onco-
176. Moser RP, Jr, Sweet DE, Haseman DB, et al: Multiple skeletal gene 9:2135–2144, 1994.
fibroxanthomas: radiologic-pathologic correlation of 72 cases. 202. Tsuneyoshi M, Enjoji M: Primary malignant peripheral nerve
Skeletal Radiol 16:353–359, 1987. tumors (malignant schwannomas): a clinicopathologic and elec-
177. Mulvihill JJ, Parry DM, Sherman JL, et al: Neurofibromatosis 1 tron microscopic study. Acta Pathol Jpn 29:363–375, 1979.
(Recklinghausen disease) and neurofibromatosis 2 (bilateral 203. Twist EC, Ruttledge MH, Rousseau M, et al: The neurofibroma-
acoustic neurofibromatosis): an update. Ann Intern Med 113:39– tosis type 2 gene is inactivated in schwannomas. Hum Mol Genet
52, 1990. 3:147–151, 1994.
178. National Institutes of Health: Neurofibromatosis review: 204. Upadhyaya M, Shen M, Cherryson A, et al: Analysis of mutations
Bethesda, Md., US Department of Health and Human Services, at the neurofibromatosis 1 (NF1) locus. Hum Mol Genet 1:735–
July 13-15. Natl Inst Health Consens Dev Conf Consens Statement 740, 1992.
6(12):1–9, 1987. 205. von Hippel E: Ueber eine sehr seltene Erkrankung der Nethaut.
179. Pitt MJ, Mosher JF, Edeiken J: Abnormal periosteum and bone in Graefe Arch Ophthalmol 59:83–106, 1904.
neurofibromatosis. Radiology 103:143–146, 1972. 206. Vortmeyer AO, Falke EA, Glasker S, et al: Nervous system
180. Plotkin SR, Blakeley JO, Evans DG, et al: Update from the 2011 involvement in von Hippel-Lindau disease: pathology and mecha-
international schwannomatosis workshop: from genetics to diag- nisms. Acta Neuropathol 125:333–350, 2013.
nostic criteria. Am J Med Genet Part A 161A:405–416, 2012. 207. Wanebo JE, Malik JM, Vandenberg SR, et al: Malignant periph-
181. Purandare SM, Lanyon WG, Arngrimsson R, et al: Characterisa- eral nerve sheath tumors: a clinicopathologic study of 28 cases.
tion of a novel splice donor mutation affecting position +1 in Cancer 71:1247–1253, 1993.
intron 18 of the NF-1 gene. Hum Mol Genet 4:767–768, 1995. 208. Weinberg RA: The biology of cancer, ed 2, New York, 2014, Garland
182. Purandare SM, Lanyon WG, Connor JM: Characterisation of Science.
inherited and sporadic mutations in neurofibromatosis type-1. 209. Weiss SW, Langloss JM, Enzinger FM: Value of S-100 protein in
Hum Mol Genet 3:1109–1115, 1994. the diagnosis of soft tissue tumors with particular reference to

ERRNVPHGLFRVRUJ
1054 14  Neurogenous Tumors and Neurofibromatosis Affecting Bone

benign and malignant Schwann cell tumors. Lab Invest 49:299– 213. Woodruff JM, Marshall ML, Godwin TA, et al: Plexiform (mul-
308, 1983. tinodular) schwannoma: a tumor simulating the plexiform neuro-
210. White HR, Jr: Survival in malignant schwannoma: an 18-year fibroma. Am J Surg Pathol 7:691–697, 1983.
study. Cancer 27:720–729, 1971. 214. Yang FC, Ingram DA, Chen S, et al: Nf1-dependent tumors
211. Wick MR, Swanson PE, Scheithauer BW, et al: Malignant periph- require a microenvironment containing Nf1+/− and c-kit-
eral nerve sheath tumor: an immunohistochemical study of 62 dependent bone marrow. Cell 135:437–448, 2008.
cases. Am J Clin Pathol 87:425–433, 1987.
212. Woodruff JM, Chernik NL, Smith MC, et al: Peripheral nerve
tumors with rhabdomyosarcomatous differentiation (malignant
“Triton” tumors). Cancer 32:426–439, 1973.

ERRNVPHGLFRVRUJ
C H A P T E R 1 5 

Cystic Lesions

CHAPTER OUTLINE

ANEURYSMAL BONE CYST INTRAOSSEOUS GANGLION


Primary Aneurysmal Bone Cyst EPIDERMAL INCLUSION CYST AND
Secondary Aneurysmal Bone Cyst DERMOID CYST
Soft Tissue Aneurysmal Bone Cyst
SOLITARY BONE CYST

Cystic lesions of bone represent a heterogeneous group stage of the process (i.e., are aneurysmal bone cysts in
of entities in which the common feature is de novo for- statu nascendi). Both benign and malignant bone lesions
mation of unilocular or multilocular cavities filled with are prone to develop aneurysmal bone cysts as second-
blood, serous fluid, mucinous content, or keratin debris. ary phenomena superimposed on preexisting conditions.
Secondary cystic changes in preexisting conditions, such These lesions are referred to as secondary aneurysmal
as in chondroblastoma, fibrous dysplasia, and giant-cell bone cysts.33,36
tumor, are discussed in conjunction with those underly- Cytogenetic studies demonstrating clonal rearrange-
ing conditions. The development of secondary aneurys- ments of chromosomal bands 16q22 and 17p13 in at least
mal bone cyst engrafted on other lesions is also discussed some aneurysmal bone cysts have raised doubts about the
in this chapter. reactive nature of these lesions.5,23,48,57,69,71 In fact, recent
molecular studies have shown that t(16;17) (q22;p13)
creates a chimeric gene in which a promoter region of
ANEURYSMAL BONE CYST the osteoblast cadherin (CDH11) gene mapping to 16q22
is juxtaposed to the ubiquitin-specific protease (USP6)
The term aneurysmal bone cyst was introduced by Jaffe and gene mapping to 17p13 (Fig. 15-1). Several variants of
Lichtenstein.26,27,35 In their words “the term aneurysmal CDH11-USP6 fusion transcripts as well as alternative
relates to a sort of blowout distention of the contour rearrangements of these genes were identified in a subset
of the affected bone and the term bone cyst relates to of primary aneurysmal bone cysts but not in secondary
the fact that it represents mainly a blood-filled cavity.” aneurysmal bone cysts.44 The chimeric CDH11-USP6
They defined this process in bone as a distinct lesion gene consists of the noncoding exon 1 and 2 of CDH11
separate from giant-cell tumor and other entities con- fused to the entire USP6 coding sequence. The fusion
taining giant cells. breakpoints in various splicing variants are after the tran-
Aneurysmal bone cyst is a multiloculated cystic lesion scription initiation site of the CDH11 gene. The break-
that almost always arises in bone and is also, although point of the second fusion partner is before the USP6
rarely, observed as a secondary phenomenon in certain ATG transcription initiation signal, which results in
soft tissue lesions.6,50,68 Most likely, it is initiated by up-regulation of USP6 ubiquitin-specific protease. USP6
an “injury” to the rich capillary network of the pre- has a high degree of homology to TBCID3 and USP32
cursor lesion, culminating in an expansile destructive genes, from which it evolved.37,49 It has also been docu-
process caused by capillary pressure of the extravasated mented that the presence of TBC1D3 and TBC domains
blood.11,30,34,35 Measurements of blood pressure in the is critical for the transforming activity of the USP6
cysts, the absence of clotting in the venules, and veno- protein.43 Several additional rearrangements of the
graphic studies indicate that the cysts are actively con- CDH11 gene with alternative fusion partners were sub-
nected with the host capillary network.7,11,30 It has been sequently identified.18,29,45-47 They involve t(1;17)(p34.1-
postulated that intraosseous hemorrhage can provoke 34.3;p13) TRAP150-USP6; t(3;17)(q21;p13) ZNF9-USP6;
either a solid reactive lesion in the form of a giant-cell t(9;17)(q22;p13) OMD-USP6; and t(17;17)(p13;q21)
reparative granuloma or an aneurysmal bone cyst. Hence, COL1A1-USP6. The mechanisms responsible for the
some have proposed the term solid aneurysmal bone cyst oncogenic effects of these fusion genes are similar to
for lesions that have large areas of solid tissue with fea- those seen with CDH11-USP6. In all of these chimeric
tures of giant-cell reparative granuloma and occur in sites genes, the USP6 coding sequences are juxtaposed to the
typical for aneurysmal bone cyst.16,28,55 Lesions that do promoter regions of the alternative translocation part-
not develop a blowout component may represent an early ners. In addition, rearrangements of the USP6 gene with
1055
ERRNVPHGLFRVRUJ
1056 15  Cystic Lesions

16q22 CDH11
1 2

3 45 6 12 13
7 8 9 10 11

1 2 17p13 USP6
1 2

3 89 13 14 23 24 25 26 30
4 5 6 7 10 11 12 15 16 17 18 19 20 21 22 27 28 29

1 2

promoter

1 23 89 13 14 23 24 25 26 30
4 5 6 7 10 11 12 15 16 17 18 19 20 21 22 27 28 29

CDH11 portion USP6 portion

CDH11 protein CDH11-USP6

PFAM: Type 1 1 1 TBC UBP


CA CA CA CA CA Cadherin-C
Type 2 1 TBC UBP
Cadherin Cadherin-C
terminal Type 3 1 2 1 TBC UBP
USP6 protein Type 4 1 2 TBC UBP
TBC-RabGAP UCH-2-3 Type 5 1 2 1 TBC UBP

TBC/RabGAP Ubiquitin carboxyl-terminal hydrolases

B C
FIGURE 15-1  ■  Molecular structure of breakpoints associated with t(16;17) (q22; p13). A, Exon-intron structure of the CDH11 and
USP6 genes. Large solid arrows indicate the most frequent locations of the breakpoints within introns. The molecular structure of
the chimeric CDH11-USP6 gene, which include the promoter region and untranslated exon 1 and 2 of the CDH11 gene and the entire
coding sequence of the USP6 gene. The number indicates exon number. B, Functional domains of the two proteins involved in the
translocation. The CDH11 gene contributes its promoter region, resulting in transcriptional up-regulation of its fusion partner, the
USP6 gene. The USP6 protein contains TBC/GAP domain involved in Rab/Ypt GTPase signaling and the UBP domain with high
degree of homology to its ancestor genes, TBC1D3(PRC17) and USP32, respectively. C, Predicted structures of fusion transcripts
encoded by the chimeric CDH11-USP6 genes.

SS18 typically involved in the development of synovial loose, fibroconnective tissue with prominent giant-cell
sarcoma have also been reported in solid variants of aneu- reaction and focal reactive bone formation. In primary,
rysmal bone cysts.20 Some of these translocations modu- or de novo, aneurysmal bone cysts, no underlying condi-
late the production of matrix metalloproteinases and tion can be identified radiographically or microscopically.
upregulate NFκB. These findings clearly indicate that at
least some aneurysmal bone cysts are true neoplasms with Incidence and Location
identifiable oncogene and promoter gene mechanisms of
development. 5,57 Aneurysmal bone cyst is relatively rare, accounting for
approximately 2.5% of all primary bone tumors. The
male-to-female sex ratio is approximately 1 : 1. The age
Primary Aneurysmal Bone Cyst distribution is strikingly different from that of giant-cell
tumor, and almost 80% of the lesions occur in skeletally
Definition
immature patients who are younger than age 20 years
This term is used to describe a peculiar lesion of bone that (Fig. 15-2). The peak incidence of this lesion is during
is characterized by the presence of spongy or multilocu- the second decade of life. An aneurysmal bone cyst may
lar cystic tissue filled with blood. The process is benign affect any bone with similar frequency. Hence, its almost
in nature, but it is locally destructive and has a high uniform skeletal distribution is a unique feature among
propensity for recurrence. Microscopically, cystic spaces bone tumors.8,13,14,21,30 Aneurysmal bone cyst has a slight
are bordered by septa composed of a well-vascularized, predilection for the craniofacial bones and vertebral

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1057

45

40 Male 46
Female 0400
Total 86
35
Primary

Distribution (%)
30
Secondary
25

20

15

10

0
1 2 3 4 5 6 7 8 9
Age in decades
FIGURE 15-2  ■  Age-related distribution of primary and secondary aneurysmal bone cyst. Note that peak incidence of both types of
aneurysmal bone cyst is during second decade of life. Lesions in patients older than age 50 years are extremely rare.

column, which are involved in 18% and 24% of cases, suggested on the basis of x-ray features.9,12,13,14,22,54,58,60,64,67
respectively. The major long tubular bones of the upper The most characteristic feature is a blowout or ballooned
and lower extremities account for 20% of the cases. distention of the periosteum outlined by a paper-thin
Approximately 12% of cases are located in the small shell of subperiosteal bone. The blowout or expanded
bones of the hands and feet. Tarsal bones are more fre- component is found overlying an area of cortical disrup-
quently involved than carpal bones. The flat bones (the tion. The lesion is lytic and may have a “soap-bubble,”
pelvis and scapula) are also well-known locations for multiloculated pattern (Figs. 15-4 and 15-5). The radio-
aneurysmal bone cyst. In the long tubular bones, the graphic features are best understood if the lesion is envi-
metaphyseal areas are preferentially involved. Lesions sioned as an expansile, multilocular balloon disrupting
that involve the ends and midshaft areas of long bones the adjacent bone and elevating the periosteum. The
occur less frequently. The typical sites of skeletal involve- blowout character of the lesion is usually most strikingly
ment are presented in Figure 15-3. demonstrated in the long bones, but it is also evident in
Rare examples of multiple metachronous aneurysmal other locations (Figs. 15-6 to 15-8). Although rare, reac-
bone cysts in as many as five different skeletal sites tive bone in the lesion can be documented radiographi-
have been reported.56,62 Recent observation of a familial cally as lesional calcifications.59 The lesion is typically
incidence of aneurysmal bone cysts have suggested a eccentric and involves the metaphysis (Figs. 15-7 and
hereditary factor in the pathogenesis of primary aneurys- 15-8). Involvement of the end of a bone and midshaft
mal bone cyst.17,32 location is less frequent (Figs. 15-8 to 15-10). Aneurys-
mal bone cysts can cross joints and involve several adja-
cent bones. This typically occurs in spinal lesions, where
Clinical Symptoms
several adjacent vertebrae and ribs can be affected. Aneu-
Pain and swelling (occasionally very pronounced) are the rysmal bone cyst of the vertebral column typically origi-
typical symptoms, which may vary in duration from nates in the posterior neural arch and expands unilaterally
weeks to several years. Occasionally the patient has a to produce an eccentric paravertebral blowout lesion. In
relatively short history of pain and swelling that progres- some cases, destruction of vertebral bodies with partial
sively developed over a few weeks. The vertebral lesions or complete collapse occurs (Fig. 15-8, A).
typically present with a full gamut of signs and symptoms Computed tomography and magnetic resonance
related to compression of the spinal cord and nerves. imaging are extremely valuable tools in the preoperative
evaluation of the lesion because they document the mul-
tilocular cystic nature of the lesion and the fluid-fluid
Radiographic Imaging
levels (Figs. 15-4 and 15-5). Magnetic resonance imaging
The radiographic presentation is very distinctive, and in demonstrates the expansile nature of the lesion, which is
most cases the diagnosis of aneurysmal bone cyst is Text continued on p. 1066

ERRNVPHGLFRVRUJ
1058 15  Cystic Lesions

10 20
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 15-3  ■  Aneurysmal bone cyst: peak age incidence and frequent sites of skeletal involvement. Most frequent sites are indicated
by solid black arrows.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1059

B C

D
FIGURE 15-4  ■  Aneurysmal bone cyst: radiographic features. A, Anteroposterior radiograph of chest of a 5-year-old boy with aneu-
rysmal bone cyst of thoracic spine. Note blowout lesion extending from right side of spinal column. B, Computed tomogram of
lesion shown in A. Large expansile cystic lesion extends from vertebral body and lateral elements to involve adjacent rib. C, Mag-
netic resonance image (MRI) of a 14-year-old girl with aneurysmal bone cyst of posterior neural arch of upper thoracic vertebra.
Note fluid level. D, Sagittal MRI of lesion shown in C reveals multiloculation and partial collapse of vertebral body.

ERRNVPHGLFRVRUJ
1060 15  Cystic Lesions

A B

C D
FIGURE 15-5  ■  Aneurysmal bone cyst: radiographic features. A and B, Anteroposterior and lateral radiographs of distal femoral
aneurysmal bone cyst in an 18-year-old woman shows ill-defined lucent area in distal shaft. Note posterior expansion of bone
contour in B. C, Axial T1-weighted magnetic resonance image (MRI) shows fluid levels within cystic lesion. D, Sagittal MRI demon-
strates loculation in T2-weighted image and posterior expansion into adjacent soft tissue.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1061

A B

C D
FIGURE 15-6  ■  Aneurysmal bone cyst: radiographic features. A and B, Anteroposterior and lateral radiographs of eccentric lytic lesion
in proximal tibial metaphysis of female adolescent. Completely lytic lesion shows medial expansion and is limited proximally by
growth plate. C, Plain radiograph of shoulder of a 40-year-old woman shows aneurysmal bone cyst of scapula. Note that multilocu-
lated lytic lesion involves glenoid region of scapula. D, Computed tomogram of the lesion in C shows expansile radiolucent lesion
in scapula.

ERRNVPHGLFRVRUJ
1062 15  Cystic Lesions

A B

C
FIGURE 15-7  ■  Aneurysmal bone cyst: radiographic features. A and B, Oblique and anteroposterior radiographs of aneurysmal bone
cyst of proximal ulnar shaft. Note blowout expansion with partially delimiting shell of periosteal bone. C, Computed tomogram of
lesion shown in A and B. Note cortical disruption and large soft tissue component.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1063

A B

C D
FIGURE 15-8  ■  Aneurysmal bone cyst: radiographic features. A, Radiograph of lumbar spine in adult with aneurysmal bone cyst that
led to partial collapse of vertebral body. B, Lateral radiograph of distal femur and knee of a 19-year-old woman with huge aneurys-
mal bone cyst replacing entire distal end of femur. Note narrow zone of transition proximally and inclusion of femoral shaft end in
expansile mass (“finger-in-the-balloon” sign). C, Subperiosteal aneurysmal bone cyst of ulnar shaft that developed 4 months after
direct trauma. D, Subperiosteal aneurysmal bone cyst of tibia.

ERRNVPHGLFRVRUJ
1064 15  Cystic Lesions

A B

C D E
FIGURE 15-9  ■  Aneurysmal bone cyst in short tubular bones: radiographic features. A, Aneurysmal bone cyst involving metacarpal
bone in child. B, Aneurysmal bone cyst involving metacarpal bone in adult. C, Aneurysmal bone cyst involving phalanx in child.
Note that unfused growth plate in A and C acts as barrier to metaphyseal lesion. In contrast, metacarpal lesion B in adult extends
to bone end. D and E, Metacarpal aneurysmal bone cyst shown in T1- and T2-weighted magnetic resonance images. Bright signal
in T2-weighted image shows multiloculation to better advantage.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1065

A B

C D
FIGURE 15-10  ■  Aneurysmal bone cyst: radiographic features. A, Lateral plain radiograph of ankle of a 12-year-old girl shows lucent
lesion of talus. B and C, Sagittal and coronal T1-weighted magnetic resonance images (MRIs) of aneurysmal bone cyst of talus show
low signal intensity, sharp circumscription, and lateral eccentric expansion. D, Sagittal T2-weighted MRI of aneurysmal bone cyst
of talus shows high signal intensity.

ERRNVPHGLFRVRUJ
1066 15  Cystic Lesions

encircled by a thin rim of periosteal bone of low signal Differential Diagnosis


intensity.
The overall diagnostic approach should be to rule out an
Gross Findings underlying condition (i.e., secondary aneurysmal bone
cyst). The lesion must be sampled extensively. Any solid
Aneurysmal bone cyst examined in situ appears as a areas should be submitted completely for microscopic
spongy or multilocular cystic lesion filled with blood examination because they are likely to contain elements
(Figs. 15-11 to 15-13). The size of the cystic spaces ranges of a precursor condition.
from less than 1 mm (spongy areas) to large cavities that Distinction from telangiectatic osteosarcoma is the most
measure several centimeters. The lesion contains only a important diagnostic problem and may be difficult
small amount of spongy, red-brown soft tissue or thin because these two conditions have overlapping clinical
membranous septa. The operating surgeon frequently and radiographic features. The problem is further ampli-
encounters what appears to be a hole containing blood. fied by the fact that an aggressive appearance on
The lesion has irregular, lobulated, but sharply demar- radiographs may suggest the incorrect diagnosis of malig-
cated borders. The more solid or spongy tissue is usually nancy. The distinction between aneurysmal bone cyst
found peripherally within the intramedullary component. and telangiectatic osteosarcoma is ultimately based on
The central portion and especially the blowout extra- microscopic features. The presence of highly anaplastic
medullary component are typically composed of large sarcomatous cells with atypical mitoses producing tumor
cystic spaces and blood. The extramedullary component osteoid is highly diagnostic of osteosarcoma. Some clini-
is sharply delineated from the surrounding soft tissue by coradiographic clues that are helpful in distinguishing
an elevated and expanded periosteum that has a thin shell these two conditions are as follows:
of reactive bone. 1. Telangiectatic osteosarcoma is uncommon and very
rarely involves the vertebral column, small bones of
Microscopic Findings the hands or feet, or craniofacial bones, where
aneurysmal bone cyst is common.
The microscopic features of aneurysmal bone cyst closely 2. Aneurysmal bone cyst typically has a blowout
parallel the gross findings. The multilocular cystic archi- appearance on radiographs. Even expansile, highly
tecture of the lesion is evident under low-power examina- destructive lesions usually show an eggshell-like
tion (Figs. 15-14 to 15-16). When curetted material is rim of reactive periosteum.
examined, the collapsed membranous septa of distended Conventional osteosarcoma rarely exhibits focal fea-
cystic spaces and irregular spongy fragments of tissue are tures of telangiectatic osteosarcoma. Moreover, a second-
present in a background of hemorrhagic material. The ary aneurysmal bone cyst may be superimposed on a
septa and more solid areas are composed of loose, fibrous conventional osteosarcoma. In such instances, pathologic-
tissue that has numerous capillary channels, multinucle- radiologic correlation is extremely helpful to avoid a mis-
ated giant cells, inflammatory cells, and extravasated red diagnosis. In the majority of cases, radiographs show a
blood cells. In general, it mimics young granulation bone-forming, radiodense lesion in addition to a lytic or
tissue. Occasionally, larger, better formed vascular chan- blowout component. The radiographs may also disclose
nels (feeding vessels) that run parallel to the long axis of a discrepancy between the radiographic presentation of
the septum can be identified. The cystic spaces typically the lesion and its microscopic appearance. In such
do not have any clearly recognizable lining, but flattened instances, another biopsy can be performed to sample a
endothelial-like cells can be present focally.3 Focal pres- portion of the tumor that shows the worrisome radio-
ence of endothelial cells can be documented immunohis- graphic features.
tochemically.4 However, ultrastructural studies do not The more solid areas of an aneurysmal bone cyst,
confirm the presence of an endothelial lining in the cystic because they contain numerous multinucleated giant
spaces.61 Giant osteoclast-like cells can dominate the cells, may be confused with a giant cell tumor. An aneu-
picture and form irregular clusters that are usually located rysmal bone cyst can also be engrafted on a preexisting
in the area of extravasated red blood cells. Reactive bone giant-cell tumor. Giant cell tumors may also cause an
formation is almost always present focally and occasion- aneurysmal bone cyst to develop as a secondary phenom-
ally can be very prominent. Areas of immature reactive enon. Distinction between a giant cell tumor with a sec-
osteoid with prominent plump osteoblasts often raise the ondary aneurysmal bone cyst and a primary aneurysmal
suspicion of a malignant, bone-forming tumor. Osteoid bone cyst on the basis of microscopic features alone can
is frequently deposited in the form of long strands that be difficult. In such instances, involvement of the end of
may be partly mineralized; these strands are oriented the long bone, especially in the knee area of a skeletally
parallel to the long axis of the septum. At the periphery, mature patient, suggests giant cell tumor. Epiphyseal
especially within the medullary canal, solid areas of tissue involvement by a similar lesion in a skeletally immature
with a prominent vasculature can be present. The sur- patient suggests a chondroblastoma with a secondary
rounding cancellous or cortical bone shows features of aneurysmal bone cyst superimposed.
resorption with prominent osteoclastic activity. The Giant cell reparative granuloma is considered by some
blowout soft tissue component is delineated by a rim of authors as a common precursor lesion of aneurysmal
fibrous tissue representing elevated periosteum. On its bone cyst that is synonymous with its solid variant. Giant
inner surface, a shell of reactive bone with prominent cell reparative granuloma is predominantly solid and
osteoblastic activity is typically present. Text continued on p. 1073

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1067

A B

C D
FIGURE 15-11  ■  Aneurysmal bone cyst: radiographic and gross features. A, Specimen radiograph of expansive lesion of the proximal
fibula. B, Bisected proximal segment of fibula shows aneurysmal bone cysts with hemorrhagic spongelike architecture. Same case
as shown in A. C, Enlarged photograph of resection specimen shown in B discloses multilocular cystic lesion. D, Low power pho-
tomicrograph shows hemorrhagic cysts and fibrous septations. (D, ×25) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1068 15  Cystic Lesions

A B

C D
FIGURE 15-12  ■  Aneurysmal bone cyst: gross features. A, Expansile hemorrhagic lesion of outer end of clavicle. Note blood-filled
cystic spaces in cut surface. B, Bisected aneurysmal bone cyst of ulna. Inner surfaces of cystic locules are discolored by old hemor-
rhage (hemosiderin). C, Resected segment of rib containing aneurysmal bone cyst that shows hemorrhagic, spongelike cut surface.
D, Specimen radiograph of lesion shown in C demonstrates ossification in septa.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1069

A B

C D
FIGURE 15-13  ■  Aneurysmal bone cyst: gross and radiographic features. A, Specimen radiograph of humeral resection. Cyst is radio-
lucent, and shell of periosteal bone encloses soft tissue extension. B, Coronally sectioned proximal end of humerus containing
aneurysmal bone cyst in metaphysis and shaft shown in A. Note hemorrhagic intramedullary lesional tissue with destruction of
medial cortex and blowout expansion into soft tissue. C, Radiograph of right hip region showing a large, multiloculated blowout of
ischial ramus of a young adult; this lesion developed over 4 months. D, Gross specimen of lesion resected from ischial ramus shown
in C consisting of multiloculated fibrous sac with hemorrhagic staining of its walls.

ERRNVPHGLFRVRUJ
1070 15  Cystic Lesions

A B

C D
FIGURE 15-14  ■  Aneurysmal bone cyst: microscopic features. A, Parallel arrays of osteoid trabeculae along septum in typical aneu-
rysmal bone cyst. B, Higher power photomicrograph shows focus of osteoid formation in septum. C and D, Irregularly oriented
osteoid trabeculae in thickened septum of aneurysmal bone cyst. This early ossification is reactive in type. (A, ×100; B, ×200; C and
D, ×400.) (A-C, hematoxylin-eosin; D, trichrome.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1071

A B

C D
FIGURE 15-15  ■  Aneurysmal bone cyst: microscopic features. A-D, Examples of bone formation in septa of aneurysmal bone cysts.
Osteoid and woven bone trabeculae often are oriented along long axes of septa. Random orientation, however, is not uncommon
(A, ×50; B, ×100; C, ×100; D, ×200) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1072 15  Cystic Lesions

A B

C D
FIGURE 15-16  ■  Aneurysmal bone cyst: microscopic features. A, Low power photomicrograph shows irregular cystic space filled with
blood and bordered by thick septa. B and C, Higher magnification shows septa with spindle cells and numerous multinucleated
giant cells. D, Solid component of aneurysmal bone cyst with numerous multinucleated giant cells. Such areas of aneurysmal bone
cyst can be misdiagnosed as giant cell tumor. (A ×50, B-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1073

frequently contains microscopic foci of cystic change that inducing shrinkage and healing.19,52 Adjuvant radiation
are consistent with those seen in an aneurysmal bone cyst; therapy is reserved for treatment of lesions that cannot
however, the features of an expansile blowout lesion be completely excised because of their location and to
are not seen on radiographs. The identification of USP6 prevent damage to functionally important structures,
gene rearrangement in giant cell reparative granulomas such as the spinal cord. Rare cases of high-grade sarcoma
of the appendicular skeleton, especially those involving (osteosarcoma and malignant fibrous histiocytoma)
the bones of hands or the feet, further confirms the developing at the site of a previously treated aneurysmal
relationship between these two entities. However, the bone cyst have been described.2,31 Most of these patients
pathogenesis of giant cell reparative granulomas appears received radiation therapy. Examples of spontaneous
to be heterogeneous; those affecting the craniofacial malignant transformation without previous radiation
bones are distinct from those in the appendicular skele- therapy also have been described.25,31
ton and typically do not carry the USP6 gene rearrange-
ment. More complex description of the molecular
background of giant cell reparative granulomas is pro- Secondary Aneurysmal Bone Cyst
vided in Chapter 10. In general, the identification of the Definition
clonal USP6 gene rearrangement in aneurysmal bone
cyst, giant cell reparative granulomas, and myositis ossi- The term secondary aneurysmal bone cyst is applied to aneu-
ficans provides important clues to the pathogenesis of rysmal bone cysts that are superimposed on a preexisting
these conditions and may be useful in their differential condition. The prognosis and behavior of these lesions
diagnosis. are generally determined by the biologic potential of the
A solitary bone cyst may occasionally be confused with underlying condition.
an aneurysmal bone cyst in biopsy material. Foci of hem-
orrhage associated with a pathologic fracture of a solitary Incidence and Location
cyst may contain multinucleated giant cells and reactive
bone, which can closely simulate aneurysmal bone cyst. More than 50% of all aneurysmal bone cysts are
In such instances, a review of radiographs is extremely superimposed on a recognizable precursor condi-
helpful. In addition, the mentioned giant cell–containing tion.1,13,14,33,36,40,54,64,66,67,70,71 The age-related distribution
areas are typically focal, and the remaining material patterns of primary and secondary aneurysmal bone cysts
shows the bland-looking fibrous membrane of a solitary are similar and show a single peak during the second
bone cyst. decade of life (Fig. 15-2). Both variants of aneurysmal
bone cyst are rare in patients older than age 30 years.
This indicates that young individuals, especially those
Treatment and Behavior
who are skeletally immature, are more likely to develop
Aneurysmal bone cysts are locally destructive lesions this process, either as a primary or secondary phenome-
that may produce severe deformity and functional non, compared with older individuals.16 In contrast to
impairment.53,54,64,67 Lesions located in the vertebral primary aneurysmal bone cyst, secondary aneurysmal
column or in the craniofacial bones can compress vital bone cyst is more frequently found in weight-bearing
structures and cause serious disability or death, which bones. On the other hand, there is no difference in pre-
is unusual.12,24,41,51,65,70 The overall prognosis is good; dilection of primary versus secondary aneurysmal bone
approximately 90% of patients have satisfactory long- cyst in non–weight-bearing sites.
term treatment results. The kinetics of the lesion can vary
from case to case. Some lesions do not increase in size Radiographic Imaging
and may develop radiographic features of intralesional
sclerosis.39 Most lesions, however, increase in size and The radiographic appearance of secondary aneurysmal
eventually require surgical intervention, other therapeu- bone cyst reflects the superimposition of a blowout lesion
tic measures, or a combination of treatments to stabilize on the radiographic features of the preexisting condition
the process and to prevent further damage. (Figs. 15-17 and 15-18). Quite frequently, the underlying
The most effective treatment is complete surgical condition can be identified only microscopically. Because
excision of the lesion, but in many instances such an of the anatomic location, involvement of specific areas of
approach may produce a major functional impairment. bone, the age of the patient, and other factors, certain
Therefore most lesions are treated by curettage and bone preexisting conditions can be anticipated.13,14,21,33,36,41,63 In
grafting. Unfortunately, this type of treatment is associ- the long tubular bones, involvement of the ends of bone
ated with a high recurrence rate that ranges from 20% in skeletally immature patients and blowout lesions of
to 70% in different series.10,11,54,64 Recurrence typically tarsal bones suggest that chondroblastoma could be the
occurs within 6 months after treatment and almost never predisposing condition. Chondroblastoma is almost
occurs after 2 years.64 Cryotherapy has reduced the rate invariably a source of a blowout lesion that develops in
of recurrence to 8% in one report.7 In some instances, in the calcaneus. It also should be suspected in the differen-
situ transcatheter embolization can be performed.42 tial diagnosis of blowout lesions of the acetabulum. In
However, the clinical value of such treatment has not skeletally mature patients, giant cell tumor is the frequent
been established in larger series. Percutaneous injection underlying condition of the blowout lesion in the end of
of calcitonin, methylprednisolone, and sclerosing agents long bones, especially if it involves the knee area. Giant
such as Ethibloc have been reported to be effective in cell reparative granuloma is a frequent predisposing

ERRNVPHGLFRVRUJ
1074 15  Cystic Lesions

FIGURE 15-17  ■  Secondary aneurysmal bone cyst: radiographic features. A, Blowout lesion of proximal humeral end represents
secondary aneurysmal bone cyst superimposed on chondroblastoma. B, Expansile lytic lesion of intertrochanteric region of femur
represents secondary aneurysmal bone cyst superimposed on nonossifying fibroma. C, Expansile lesion involves distal end of
second metacarpal bone and is secondary aneurysmal bone cyst superimposed on giant cell reparative granuloma.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1075

A B

C D
FIGURE 15-18  ■  Secondary aneurysmal bone cyst: gross, radiographic, and microscopic features. A, Bisected rib with large secondary
aneurysmal bone cyst engrafted on fibrous dysplasia. B, Specimen radiograph of A shows ground-glass appearance of more
solid area at top, which contains fibrous dysplasia, and lucent area of aneurysmal bone cyst below. C, Radiograph of chondroblas-
toma of glenoid region of scapula with superimposed aneurysmal bone cyst. D, Low power photomicrograph of curetted material
from C shows thickened septum of aneurysmal bone cyst with focus of chondroid matrix and sheets of chondroblasts. (D, ×100)
(D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1076 15  Cystic Lesions

superimposed on a giant cell tumor may have all or some


TABLE 15-1 Precursor Conditions for Secondary
of the complications related to both conditions.
Aneurysmal Bone Cyst
Giant cell tumor Eosinophilic granuloma Personal Comments
Chondroblastoma Hemangioma
Osteoblastoma Myositis ossificans A useful radiologic feature of aneurysmal bone cyst, the
Nonossifying fibroma Osteosarcoma
Fibrous dysplasia Malignant fibrous histiocytoma
“finger-in-the-balloon” sign, has been observed by Dr.
Giant cell reparative Metastatic carcinoma Robert Freiberger (personal communication). This is the
granuloma preservation of a cortical bone cuff that penetrates for a
Chondromyxoid Miscellaneous lesions containing short distance into the expanded area of destructive
fibroma rich vascular network blowout. This sign is not present in telangiectatic osteo-
(hamartoma)
Fibrous histiocytoma sarcoma or other aggressive malignant tumors of bone.
Solitary bone cyst The most frequent histologic obstacle in the recogni-
tion of aneurysmal bone cyst is the presence of significant
amounts of reactive bone and osteoid within the septa
and even in solid areas of the lesion. Delicate osteoid
seams may be abundant in an aneurysmal bone cyst. They
are usually oriented along the septa, unlike the random
condition for the development of secondary aneurysmal orientation of osteoid and tumor bone trabeculae of
bone cyst in the small bones of the hands and feet and in osteosarcomas.
the mandible. Fibrous dysplasia should be considered in Microscopic features of an aneurysmal bone cysts are
reference to lesions located in the shafts of long bones often combined with those of giant cell reparative granu-
of the extremities, in the ribs, and in the craniofacial loma, especially in lesions found in the short tubular
region. In the metaphyseal parts of the long tubular bones of the hands and feet and in vertebral lesions. The
bones, especially in a lower extremity, lesions such as composite nature of these lesions suggests that aneurys-
nonossifying fibroma and, less frequently, chondromyx- mal bone cyst and giant cell reparative granuloma may
oid fibroma should be considered. The most frequent represent closely related but distinct reactions to intraos-
underlying condition for secondary aneurysmal bone cyst seous hemorrhage after trauma.
of the vertebral column is osteoblastoma. Osteoblastoma
should also be anticipated in sites such as the mandible
and the maxilla.63 An expansile, fluid-filled lesion of the
Soft Tissue Aneurysmal Bone Cyst
sacrum typically represents an aneurysmal bone cyst Aneurysmal bone cysts very rarely arise in extraosseous
superimposed on a giant cell tumor. The most frequent sites, including the somatic soft tissue of the extremities,
underlying conditions for secondary aneurysmal bone most often in the thigh and shoulder regions.6,68 It has
cyst are listed in Table 15-1. It must be remembered that also been reported in the larynx and in arterial walls.34,50
aneurysmal bone cyst is rarely superimposed on primary Some of these appear to have arisen within the hemor-
or metastatic malignant tumors.15 All bone lesions rhagic central zones of myositis ossificans lesions.6
containing components with rich vasculature may occa-
sionally develop aneurysmal bone cysts as secondary phe-
nomena. Therefore it can be seen even in such conditions SOLITARY BONE CYST
as vascular cartilaginous hamartoma.38
Similar to aneurysmal bone cyst, solitary bone cyst is not
Microscopic Findings a true neoplasm, but it is usually included in the discus-
sion of bone tumors. Sometimes, it is also referred to as
Microscopic features of secondary aneurysmal bone cyst a simple or unicameral bone cyst. However, the term uni-
do not differ from those of primary aneurysmal bone cameral is misleading because some of these lesions may
cyst. In addition, residual foci with microscopic features consist of several multilocular cavities.
of an underlying condition—chondroblastoma, giant
cell tumor, giant cell reparative granuloma—are present
Definition
(Figs. 15-19 and 15-20).
Solitary bone cyst is typically a unilocular cystic lesion
Differential Diagnosis that does not have a lining and is filled with serous fluid.
It arises most frequently in the metaphyseal portion of
Differential diagnosis of a secondary aneurysmal bone the major long bones, such as the humerus or femur, in
cyst is similar to that of a primary aneurysmal bone cyst. skeletally immature patients.

Treatment and Behavior Incidence and Location


The treatment and behavior of secondary and primary Solitary bone cyst is typically diagnosed during the first
aneurysmal bone cysts are similar. The prognosis is two decades of life.72,76,77,81,87 The peak incidence is
related to the biology of the underlying condition. For between ages 3 and 14 years, when nearly 80% of cases
example, the patient with an aneurysmal bone cyst are diagnosed. In several major series, the solitary bone

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1077

A B

C D
FIGURE 15-19  ■  Secondary aneurysmal bone cyst engrafted on chondroblastoma: microscopic features. A and B, Low and intermedi-
ate power photomicrographs show aneurysmal cystic component of lesion. C and D, Low and intermediate power photomicrographs
show solid component of lesion consistent with chondroblastoma. Note focus of cartilaginous differentiation better seen in D.
(A and C, ×200; B and D, ×400) (A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
1078 15  Cystic Lesions

A B

C D
FIGURE 15-20  ■  Secondary aneurysmal bone cyst: microscopic features engrafted on osteoblastoma. A and B, Low power photomi-
crographs show aneurysmal cystic component of lesion. C, Low power photomicrograph shows osteoblastic lesion in wall of
cyst. D, Anastomosing pattern of bone trabeculae rimmed by osteoblasts in fibrovascular stroma consistent with osteoblastoma.
(A, B, and D, ×200; C, ×20.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1079

5 15
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 15-21  ■  Solitary bone cyst. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites are indicated
by solid black arrows.

cyst had a predilection for male patients; the overall Radiographic Imaging
male-to-female sex ratio is approximately 2 : 1.
Solitary bone cyst is relatively uncommon in patients Solitary bone cyst presents as a central lucency within
older than age 20 years. It has been noted that the lesion the medullary cavity of the shaft of the major long
preferentially involves the major long tubular bones, such bones.77,82,89,96,98,111,113 Fine trabeculation and scalloped
as the humerus and femur, during the first two decades sclerotic borders are frequently seen (Figs. 15-22 to
of life. Nearly 80% of cases are diagnosed in these ana- 15-26). Central involvement of the medullary cavity and
tomic sites. On the other hand, rare cases diagnosed in extension to the growth plate are typical (Fig. 15-26). The
older patients usually involve the ilium, talus, and calca- cortex overlying the lesion is thinned and scalloped with
neus.72,75,87,88,103,107 In rare instances, a solitary bone cyst a distended bone contour. It is important to note that the
may involve other anatomic sites, such as the spine, cortex is never completely disrupted and that the lesion
sacrum, craniofacial bones, forearm, and acral skele- does not extend into soft tissue. Occasionally, it is possible
ton.83,91-93,97,100,110 Peak age incidence and the most fre- to document on serial radiographs that the epiphysis
quent sites of skeletal involvement are presented in grows away from the cyst. Pathologic fracture is fre-
Figure 15-21. quently the presenting symptom. Such fracture ranges
from a small cortical infarction to a complete fracture
with displacement. A fragment of displaced cortex can
Clinical Symptoms
drop to the distal part of the cyst and produce the so-called
Pain in the area of the lesion is a common symptom. fallen-fragment sign (Fig. 15-22).108 A shift in the position
Sometimes the lesion is discovered after pathologic of the fragment in the cyst confirms the nonsolid character
fracture. Swelling or deformity of the affected area of the bone defect. Computed tomography and magnetic
can also be a presenting symptom.72,75,87,107,114 Calcaneal resonance imaging accurately reveal the extent of the
cysts, usually found in older patients, are usually asymp- lesion and disclose its cystic nature and water density
tomatic; however, the cyst’s large size in this weight- content. Occasionally, these scans reveal that several cystic
bearing bone is associated with a significant risk of spaces are present (multilocular cyst). More typically, a
fracture. Text continued on p. 1085

ERRNVPHGLFRVRUJ
1080 15  Cystic Lesions

A B
FIGURE 15-22  ■  Solitary bone cyst: radiographic features. A and B, External rotation view of proximal humerus of child shows patho-
logic fracture through large solitary bone cyst. Linear fragments of comminuted cortical bone (fallen-fragment sign) can be seen in
lower end of cyst in both views (arrows).

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1081

A B

C
FIGURE 15-23  ■  Solitary bone cyst: radiographic features. A and B, Well-demarcated lucent lesion in proximal femoral shaft of child.
Upper border of cyst is close to growth plate of greater trochanter. Parallel periosteal reaction on the cortical surface distally reflects
presence of pathologic fracture (arrow). C, Same lesion 5 years later. Note increased size with enlargement and involvement of
femoral neck.

ERRNVPHGLFRVRUJ
1082 15  Cystic Lesions

B C
FIGURE 15-24  ■  Solitary bone cyst: radiographic features. A, Anteroposterior (AP) radiograph of pelvis shows cystic lesion of superior
pubic ramus and acetabular area in adult. B, AP radiograph of right side of pelvis shows very large cyst occupying entire ilium of
adult and extending into both superior and inferior rami. Pelvic cysts of this type occur almost exclusively in adults. C, AP film of
foot of adult shows solitary bone cyst in very uncommon location in shaft of second metatarsal bone. Note extension into epiphysis,
which typically occurs in skeletally mature patients.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1083

A B
FIGURE 15-25  ■  Solitary bone cyst: radiographic features. A and B, Anteroposterior and lateral radiographs show lytic lesion with
expansion of bone contour that involves distal end of fibula. Note prominent trabeculation.

ERRNVPHGLFRVRUJ
1084 15  Cystic Lesions

B C

FIGURE 15-26  ■  Solitary bone cyst: radiographic features. A, Lateral radiograph of ankle in young adult shows solitary bone cyst in
typical location within anterior half of calcaneus. B, Distal fibular lesion in young adult occupies diametaphyseal area of fibula with
expansion of bone contour. C, Bivalved solitary bone cyst of distal fibula in B shows single cavity with ridging of inner surface and
focal hemosiderin deposition.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1085

single, large cavity filled with fluid the density of water grafting is frequently used to treat enlarging cysts of
is present. Extension to the end of bone is rare and, if weight-bearing bones.74,78,80,84,85,87,96,98,102,105 Other types
present, it is usually seen in skeletally mature patients. of treatment, including subtotal resection, allografting,
and packing with various synthetic materials; percutane-
ous injection of demineralized bone matrix; and autoge-
Gross Findings
nous bone marrow transplant, have also been repor
Evaluation of an intact solitary bone cyst is rarely possible ted.74,79,80,94,96,98,101,106,112 The recurrence rate after both
(Fig. 15-26). In such instances, it presents as a large injection and curettage is approximately 20%. Repeat
intramedullary cavity filled with a clear or yellowish fluid injection or curettage is often necessary. Epiphyseal
that has a low viscosity. In general, it is similar to serous involvement by solitary bone cysts is occasionally observed,
effusions of body cavities. The lesion is usually composed and these patients may exhibit growth arrest.99,109,112 Rare
of a single cyst, but occasionally septations divide it into instances of secondary malignancy in solitary bone cysts,
several cavities. A spongy component composed of mul- including a chondrosarcoma, have been reported.86,90
tiple smaller cysts can be present focally. This component
is usually located peripherally within the medullary cavity
and is attached to the wall of the cyst. The wall is com- INTRAOSSEOUS GANGLION
posed of a paper-thin, tan-yellow fibrous tissue with mul- Definition
tiple bony ridges. Recent or old fractures create focal
nodularities related to the healing process. An intraosseous ganglion cyst is identical to its soft tissue
counterpart. It is composed of a small cavity without
lining that is filled with mucoid viscous material. Typi-
Microscopic Findings
cally, it occurs in the subchondral parts of bones near
The wall of the cyst is composed of a thin layer of fibrous joint surfaces. Similar to other cystic conditions discussed
tissue without any lining (Figs. 15-27 to 15-29). Dilated in this chapter, it is not a true neoplasm. It probably
vessels, scattered inflammatory cells, and multinucleated represents a degenerative change with development of a
giant cells are commonly present. Hemosiderin pigment cavity filled with mucoid material. To some extent, it is
and foamy macrophages are seen focally. Fibrin deposits similar to the so-called subchondral cyst frequently seen
in the wall of the cyst can become mineralized and produce in degenerative joint disease. In fact, the degenerative
concentric lamellar structures that resemble cementum mechanism that leads to the development of these condi-
(Figs. 15-29). Fibrous membranes curetted from the tions could be similar. A lesion should be classified as an
bone cysts containing these cementoid bodies can be intraosseous ganglion if it occurs as a solitary change that
confused with fibrous dysplasia or even an odontogenic is not associated with degenerative joint disease. By con-
tumor.73,95,104,110 This accounts for reports of some cemen- vention, subchondral cystic lesions that occur in the pres-
tomas of long bones in older literature. Prominent giant ence of osteoarthritis should be considered as an integral
cell reaction in the wall can occasionally be responsible element of a degenerative disorder.122,123,124,126
for its being confused with a giant cell lesion. Bone resorp-
tion with osteoclastic activities is seen in the tissue sur- Incidence and Location
rounding the lesion. In addition, reactive bone formation
with prominent osteoblasts can be present, corresponding The true incidence of this condition is unknown because
to a site of pathologic fracture. Occasionally, septa sepa- many of these lesions are asymptomatic and are inciden-
rating individual cysts can be seen and can have micro- tally discovered on radiographs obtained for other reasons.
scopic features similar to those of aneurysmal bone cysts. There are very few reported series of more than 10 cases
of intraosseous ganglion. On the basis of information from
Differential Diagnosis the major reported series, it is possible to conclude that
the lesion is typically identified in skeletally mature patients
The problems in identifying a solitary bone cyst are pre- older than age 20 years (approximately 80% of cases). The
dominantly related to the lack of radiologic and clinical peak incidence is probably in the fourth or fifth decade of
data—the pathologist is not aware of the cystic nature of life. In most series, there is a slight male predominance,
the lesion in question. Therefore the microscopic phe- with a male-to-female ratio of approximately 1.2 : 1. Char-
nomena observed in the curetted fibrous membrane are acteristic sites are the juxtaarticular subchondral locations,
interpreted to be an integral component of the solid most frequently in the long tubular bones. The hip, knee,
lesion. Moreover, although the cystic nature of the lesion and ankle regions are most frequently affected.155-117,120,123,
125,127-129
can be recognized, it is usually interpreted to be a second- Other typical sites include the shoulder and wrist
ary phenomenon (i.e., cystic degeneration of a preexist- regions. The joints of the lower extremities are more fre-
ing solid lesion). Thus the subsequent misdiagnosis of quently involved than those of the upper extremities. The
solitary bone cyst as fibrous dysplasia, odontogenic tumor, or peak age incidence and the most frequent sites of skeletal
giant cell lesion is a consequence of these two major errors. involvement are presented in Figure 15-30.

Treatment and Behavior Clinical Symptoms


In addition to dual-needle aspiration of the cyst and instil- Some intraosseous ganglion cysts are asymptomatic, and
lation of methylprednisolone, which is now the treatment the lesions are discovered incidentally when a radiograph
of choice, the traditional method of curettage with bone is obtained for other reasons. The lesion is symptomatic

ERRNVPHGLFRVRUJ
1086 15  Cystic Lesions

A B

C D
FIGURE 15-27  ■  Solitary bone cyst: microscopic features. A and B, Fibrous membrane without lining surrounding the solitary bone
cyst cavity. C, Delicate fibrous membrane showing sparse cellularity and deposits of eosinophilic fibrinlike material. D, Reactive
bone formation in the wall of solitary bone cyst (A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1087

A B

C D
FIGURE 15-28  ■  Solitary bone cyst: microscopic features. A, Fibrous membrane with sparse cellularity showing myxoid change and
vascular proliferation. B, Fibrous membrane with eosinophilic fibrinlike deposits. C and D, Medium and higher power magnifications
of reactive bone formation in the wall of solitary bone cyst. (A-C, ×100; D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1088 15  Cystic Lesions

A B

C D
FIGURE 15-29  ■  Solitary bone cyst: microscopic features. A and B, Low power photomicrographs show multinucleated giant cell
reaction and osteoid formation in wall of cyst. C and D, Calcified fibrin deposits may undergo ossification, as shown in these pho-
tomicrographs of thickened cyst wall. (A and B, ×100; C and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1089

40 60
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 15-30  ■  Intraosseous ganglion. Peak age incidence and frequent sites of involvement. Most frequent sites are indicated by
solid black arrows.

in approximately 60% of cases. Pain of several months’ examination of the lesion is based on curetted material.
to several years’ duration is the most common symptom. In such instances, it represents a disrupted fibrous mem-
Symptoms can be aggravated by standing or exercise. brane mixed with gelatinous fluid.

Radiographic Imaging Microscopic Findings


Radiographs reveal a well-defined lytic area with sclerotic As mentioned, the microscopic features of intraosseous
margins located eccentrically in the subchondral bone ganglion cysts are identical to the those of their soft tissue
(Fig. 15-31).115,116,120,122,127,128 The average size of the counterparts. The wall of the cyst is composed of flat-
lesion is 1 cm in diameter. Lesions larger than 2 cm in tened fibrous elements without lining (Fig. 15-32). The
diameter are extremely rare, but a few intraosseous gan- capsule can have two distinct layers. The outer, or periph-
glion cysts as large as 5 cm in diameter have been reported eral, layer is composed of more compact fibrous elements
(Fig. 15-31).115,118,124 The lesion is not connected with a (Fig. 15-32). In contrast, the inner portion is loose and
joint or adjacent synovial-lined structure. In typical cases, shows myxoid change and stellate-shaped cells. Areas of
the bone contours are not altered. Lesions exceeding myxoid change within the capsule can also be seen focally.
2 cm in diameter can expand into the adjacent cortex and Scattered inflammatory cells and foamy macrophages
distort the articular cartilage. Rare cases of subperiosteal may be present. The gelatinous material represents
ganglion cysts have also been reported.119 amorphous granular or membranous material with occa-
sional inflammatory and red blood cells (Fig. 15-33). The
surrounding tissue may contain reactive bone with osteo-
Gross Findings
blastic rimming, focally resorptive changes with osteo-
Intraosseous ganglion cysts typically have a relatively clastic activity, or a combination of these features.
small cavity (less than 1 cm) and are surrounded by a thin,
fibrous capsule filled with gelatinous or mucoid fluid. Differential Diagnosis
The bone and articular cartilage in the vicinity of the
lesion are not altered. Occasionally a thin sclerotic rim If the lesion is present in a typical location, it rarely causes
of bone surrounds the cyst. In a majority of cases, gross diagnostic problems. The diagnosis is usually established

ERRNVPHGLFRVRUJ
1090 15  Cystic Lesions

B C D
FIGURE 15-31  ■  Intraosseous ganglion: radiographic features. A, Anteroposterior radiograph shows well-circumscribed, lucent lesion
in medial malleolus of tibia in young adult. Narrow zone of bone sclerosis forms margin of this loculated cyst. B, Radiograph shows
well demarcated area of lucency in distal portion of scaphoid. C and D, Unusually large ganglion cyst involves proximal end of
radius and most of its shaft. A and B were taken 2 years apart. (Courtesy Dr. T. Bauer, Cleveland.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1091

A B

C D
FIGURE 15-32  ■  Intraosseous ganglion: microscopic features. A, Low power photomicrograph of fibrous cyst. Note the absence
of specific lining. B, Fibrous membrane lining a ganglion cyst with prominent myxoid change. C and D, Low and higher
power photomicrographs of fibrous membrane lining a ganglion cyst with extensive myxoid change. (A-C, ×100; D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1092 15  Cystic Lesions

A B

C D
FIGURE 15-33  ■  Intraosseous ganglion: microscopic features. A–C, Fibrous membrane lining the cyst. Note prominent stromal myxoid
change and the presence of proteinaceous myxoid material in the lumen. D, More cellular component of the fibrous cyst with myxoid
change (A-D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1093

preoperatively on the bases of radiographic features and lesions lined by squamous epithelium. The displacement
the location of the lesion. A subchondral cyst is microscopi- theory (developmental or injury related) is frequently
cally indistinguishable from an intraosseous ganglion, but postulated as a possible pathogenetic mechanism for the
the former is associated both microscopically and radio- development of epidermal cysts in bone and other organs.
graphically with changes consistent with osteoarthri- Displacement is certainly a mechanism for lesions that
tis.121,126 Intraosseous ganglion also can be confused with develop in association with a penetrating injury (inclusion
other myxoid lesions of bone, such as fibromyxoma or cysts). In many instances, such lesions arise by means of
chondromyxoid fibroma, but this is rare. Fibromyxoma and aberrant squamous differentiation of mesenchymal cells
chondromyxoid fibroma may contain free mucinous (developmental cysts) rather than by displacement. Inclu-
material, but the periphery of the lesion usually contains sion and developmental epidermal cysts of bone have
diagnostic cellular tissue. distinct age and skeletal distribution patterns.146 The age-
related distribution and the most frequent sites of skeletal
involvement are presented in Figure 15-34.
Treatment and Behavior
Epidermal inclusion inclusion cysts typically occur in
Curettage and bone grafting are sufficient treatment for the acral skeleton and primarily involve the phalan-
the vast majority of cases. A small percentage of patients ges.132,140,143,147 They are related to penetrating injuries
may have recurrences, but these cases are successfully and frequently occur in the hands of skeletally mature
treated by second curettage. individuals. Involvement of a long bone is extraordinarily
rare.136 Radiographically, they represent well-demarcated
lytic defects with sclerotic margins (Fig. 15-35). The
EPIDERMAL INCLUSION CYST bone contour can be expanded, but cortical disruption is
AND DERMOID CYST rarely seen. Pathologic fracture may be a presenting sign
(Fig. 15-35). Histologically, these lesions are cysts lined
Cysts of bone lined by squamous epithelium are relatively by squamous epithelium and filled with keratin debris
rare. In the course of embryonal development, cells of (Fig. 15-36). Disruption of the cyst may result in a leak
the epidermis may be displaced into bone and form cystic of keratin material into the adjacent tissue, which causes
Incidence

Phalanges
Skull

1 2 3 4 5 6 7 8
Age in decades

FIGURE 15-34  ■  Epidermal cyst. Peak age incidence and most frequent sites of skeletal involvement. Most frequent site is indicated
by solid black arrow.

ERRNVPHGLFRVRUJ
1094 15  Cystic Lesions

A B C

D
FIGURE 15-35  ■  Epidermal inclusion cyst: radiographic features. A, Cystic lucency in distal phalanx of man who sustained crush injury
of this finger 3 years previously. Slowly enlarging cyst expands bone contour and shows associated bone sclerosis proximally.
B, Pathologic fracture of distal phalanx through area of radiolucency. C, Expansile lytic lesion in distal phalanx of middle finger 1
year after a penetrating injury. D, Expansile lytic lesion of distal phalanx erodes cortex and is associated with soft tissue edema.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1095

A B

C D
FIGURE 15-36  ■  Epidermal inclusion cyst: microscopic features. A, Cystic space filled with keratin and lined by stratified squamous
epithelium. B, Disrupted cyst with lumen partially lined by squamous epithelium exhibiting keratin debris in soft tissue. C and
D, Disrupted cysts showing keratin debris within soft tissue associated with inflammatory response. (A, C, and D, ×100; B, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1096 15  Cystic Lesions

an inflammatory response with foreign-body multinucle- 10. Clarke SJ, MacKay JS, Young GK: Recurrence of aneurysmal
ated giant cells. The associated soft tissue edema may bone cyst of the fourth metatarsal. J Foot Ankle Surg 33:467–471,
1994.
clinically or radiographically raise suspicion of an aggres- 11. Clough JR, Price CH: Aneurysmal bone cyst: pathogenesis and
sive lesion. long term results of treatment. Clin Orthop 97:52–63, 1973.
Developmental epidermal cysts occur predominantly 12. Cohen RS: Aneurysmal bone cyst of the upper maxilla. Rev Lar-
in the skull and are most frequently diagnosed in yngol Otol Rhinol (Bord) 114:29–32, 1993.
13. Dabska M, Buraczewski J: Aneurysmal bone cyst: pathology, clini-
children during the first decade of life.139,145,149-151 Most of cal course and radiographic appearances. Cancer 23:371–389,
these lesions are present at birth, but some may be 1969.
asymptomatic for many years and are eventually diag- 14. Dahlin DC, Unni KK: Bone tumors: general aspects and data on 8,542
nosed during adulthood. Radiographically, they appear as cases, ed 4, Springfield, Ill, 1986, Charles C Thomas.
lytic defects with scalloped sclerotic margins. Epidermal 15. Dailey R, Gilliland G, McCoy GB: Orbital aneurysmal bone cyst
in a patient with renal cell carcinoma. Am J Ophthalmol 117:643–
cysts may be encountered in any calvarial bone involving 646, 1994.
outer and inner tables or diploe. Outward expansion can 16. Dehner LP, Risdall RJ, L’Heureux P: Giant-cell containing
produce a palpable mass. Inward growth may be asymp- “fibrous” lesion of the sacrum: a roentgenographic, pathologic,
tomatic for a long period, but eventually it becomes and ultrastructural study of three cases. Am J Surg Pathol 2:55–70,
1978.
symptomatic.130,131,133,134,137,139,145 17. Dicaprio MR, Murphy MJ, Camp RL: Aneurysmal bone cysts of
Dermoid cysts, or dermoids, are distinguished from the spine with familial incidence. Spine 25(12):1589–1592, 2000.
epidermal cysts by the presence of additional dermal 18. Ellison DA, Sawyer JR, Parham DM, et al: Soft-tissue aneurysmal
structures—skin adnexae (sebaceous glands, hair folli- bone cyst: report of a case with t(5;17)(q33;p13). Pediatr Dev
cles). They represent a form of mature cystic teratoma Pathol 10:46–49, 2007.
19. Falappa P, Fassari FM, Fanelli A, et al: Aneurysmal bone cysts:
and most frequently occur in the midline.135,141,142,144,148 treatment with direct percutaneous Ethibloc injection: long term
These cysts usually occur in the regions of the anterior results. Cardiovasc Intervent Radiol 25:282–290, 2002.
fontanel and sacrum. Pathogenetically, they belong to the 20. Geiersbach K, Rector LS, Sederberg M, et al: Unknown partner
family of germ-cell tumors, and their extensive descrip- for USP6 and unusual SS18 rearrangement detected by fluores-
cence in situ hybridization in a solid aneurysmal bone cyst. Cancer
tion is beyond the scope of this text. Cholesteatoma is an Genet 204:195–202, 2011.
old descriptive term used to designate a cystic lesion filled 21. Gupta VK, Gupta SK, Khosla VK, et al: Aneurysmal bone cysts
with grossly cheesy keratinous debris.104 of the spine. Surg Neurol 42:428–432, 1994.
Developmental epidermoid cysts, or dermoids, of the 22. Haber HP, Drews K, Scheel-Walter H, et al: Aneurysmal bone
skull may be associated with a malignant component. cyst in early childhood: ultrasound findings. Pediatr Radiol 23:405–
406, 1993.
Considering the relative rarity of these lesions, there are 23. Herens C, Thiry A, Dresse MF, et al: Translocation (16;17)
quite a few reports of malignant transformation, which is (q22;p13) is a recurrent abnomaly of aneurysmal bone cysts.
usually in the form of squamous carcinoma.138,152 To the Cancer Genet Cytogenet 127:83–84, 2001.
best of our knowledge, there is no report of malignant 24. Hunter JV, Yokoyama C, Moseley IF, et al: Aneurysmal bone cyst
of the sphenoid with orbital involvement. Br J Ophthalmol 74:505–
transformation of epidermal inclusion cysts of the acral 508, 1990.
skeleton. Squamous cell carcinomas of bone are typically 25. Hsu CC, Wang JW, Huang CH, et al: Osteosarcoma at the site
associated with epidermalization of sinus tracts of chronic of a previously treated aneurysmal bone cyst. A case report. J Bone
osteomyelitis and are discussed in Chapter 24. Joint Surg Am 87(2):395–398, 2005.
26. Jaffe HL: Aneurysmal bone cyst. Bull Hosp Joint Dis 11:3–13, 1950.
27. Jaffe HL, Lichtenstein L: Solitary unicameral bone cyst, with
REFERENCES emphasis on the roentgen picture, the pathologic picture, and the
pathogenesis. Arch Surg 46:1004–1025, 1942.
Aneurysmal Bone Cyst 28. Karabela-Bouropoulou V, Liapi-Avgeri G, Paxinos O, et al: Solid
1. Abdelwahab IF, Kenan S, Hermann G, et al: Case report 845: variant of aneurysmal bone cyst: a case report with bilateral
fluid-filling giant cell tumor with an aneurysmal bone cyst com- involvement of the distal femoral metaphyses. Virchows Arch
ponent. Skeletal Radiol 23:317–319, 1994. 425:531–535, 1994.
2. Aho HJ, Aho AJ, Einola S: Aneurysmal bone cyst: a study of 29. Kenney B, Richkind KE, Zambrano E: Solid variant of aneurys-
ultrastructure and malignant transformation. Virchows Arch A mal bone cyst with a novel (X;9) translocation. Cancer Genet Cyto-
Pathol Anat Histol 395:169–179, 1982. genet 178:155–159, 2007.
3. Aho HJ, Aho AJ, Pelliniemi LJ, et al: Endothelium in aneurysmal 30. Kransdorf MJ, Sweet DE: Aneurysmal bone cyst: concept, con-
bone cyst. Histopathology 9:381–387, 1985. troversy, clinical presentation, and imaging. AJR Am J Roentgenol
4. Alles JU, Schulz A: Immunohistochemical markers (endothelial 164:573–580, 1995.
and histiocytic) and ultrastructure of primary aneurysmal bone 31. Kyriakos M, Hardy D: Malignant transformation of aneurysmal
cysts. Hum Pathol 17:39–45, 1986. bone cyst, with an analysis of the literature. Cancer 68:1770–1780,
5. Althof PA, Ohmori K, Zhou M, et al: Cytogenetic and molecular 1991.
cytogenetic findings in 43 aneurysmal bone cysts: aberrations of 32. Leither A, Windhager R, Kainberger F, et al: A case of aneurysmal
17p mapped to 17p13.2 by fluorescence in situ hybridization. Mod bone cyst in father and son. Eur J Radiol 29:28–30, 1998.
Pathol 17:518–525, 2004. 33. Levy WM, Miller AS, Bonakdarpour A, et al: Aneurysmal bone
6. Amir G, Mogle P, Sucher E: Case report #729: myositis ossificans cyst secondary to other osseous lesions: report of 57 cases. Am J
and aneurysmal bone cyst. Skeletal Radiol 21:257–259, 1992. Clin Pathol 63:1–8, 1975.
7. Biesecker JL, Marcove RC, Huvos AG, et al: Aneurysmal bone 34. Libera DD, Redlich G, Bittesini L, et al: Aneurysmal bone cyst
cysts: a clinicopathologic study of 66 cases. Cancer 26:615–625, of the larynx presenting with hypoglottic obstruction. A case
1970. report and review of the literature. Arch Pathol Lab Med 125:673–
8. Carlson DH, Wilkinson RH, Bhakkaviziam A: Aneurysmal bone 676, 2001.
cysts in children. Am J Roentgen Rad Ther Nucl Med 116:644–650, 35. Lichtenstein L: Aneurysmal bone cyst: observations on 50 cases.
1972. J Bone Joint Surg 39A:873–882, 1957.
9. Chakravarty K, Brett F, Merry P: Aneurysmal bone cyst: an 36. Martinez V, Sissons HA: Aneurysmal bone cyst: a review of 123
unusual presentation of neck pain in a young adult. Br J Rheumatol cases including primary lesions and those secondary to other bone
33:597–598, 1994. pathology. Cancer 61:2291–2304, 1988.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1097

37. Masuda-Robens J, Kutney SN, Qi H, et al: The TRE17 onco- 63. Svensson B, Isacsson G: Benign osteoblastoma associated with an
gene encodes a component of a novel effector pathway for Rho aneurysmal bone cyst of the mandibular ramus and condyle. Oral
GTPases Cdc42 and Rac1 and stimulates actin remodeling. Mol Surg Oral Med Oral Pathol 76:433–436, 1993.
Cell Biol 23:2151–2161, 2003. 64. Tillman BP, Dahlin DC, Lipscomb PR, et al: Aneurysmal
38. McCarthy EF, Dorfman HD: Vascular and cartilaginous hamar- bone cyst: an analysis of 95 cases. Mayo Clin Proc 43:478–495,
toma of the ribs in infancy with secondary aneurysmal bone cyst 1968.
formation. Am J Surg Pathol 4:247–253, 1980. 65. Trent C, Byl FM: Aneurysmal bone cyst of the mandible. Ann Otol
39. McQueen MM, Chalmers J, Smith GD: Spontaneous healing of Rhinol Laryngol 102:917–924, 1993.
aneurysmal bone cysts: a report of two cases. J Bone Joint Surg Br 66. Tresserra F, Rami R, Salas A, et al: Polyostotic coexisting fibrous
67:310–312, 1985. dysplasia and aneurysmal bone cyst of the chest wall. Thorac Car-
40. Mirra JM: Bone tumors: clinical, radiologic, and pathologic correlations, diovasc Surg 41:321–324, 1993.
Philadelphia, 1989, Lea & Febiger. 67. Vergel De Dios AM, Bond JR, Shives TC, et al: Aneurysmal bone
41. Motamedi MH, Yazdi E: Aneurysmal bone cyst of the jaws: analy- cyst: a clinicopathologic study of 238 cases. Cancer 69:2921–2931,
sis of 11 cases. J Oral Maxillofac Surg 52:471–475, 1994. 1992.
42. Murphy WA, Strecker EB, Schoenecker P: Transcatheter emboli- 68. Wang XL, Gielen JL, Salgado R, et al: Soft tissue aneurysmal
sation therapy of an ischial aneurysmal bone cyst. J Bone Joint bone cyst. Skeletal Radiol 33:477–480, 2004.
Surg Br 64:166–168, 1992. 69. Winnepenninckx V, Debiec-Rychter M, Jorissen M, et al: Aneu-
43. Nakamura T, Hillova J, Mariage-Samson R, et al: Molecular rysmal bone cyst of the nose with 17p13 involvement. Virchows
cloning of a novel oncogene generated by DNA recombination Arch 439:636–639, 2001.
during transfection. Oncogene Res 2:357–370, 1988. 70. Wojno KJ, McCarthy EF: Fibro-osseous lesions of the face and
44. Oliveira AM, Perez-Atayde AR, Inwards CY, et al: USP6 and skull with aneurysmal bone cyst formation. Skeletal Radiol 23:15–
CDH11 oncogenes identify the neoplastic cell primary aneurysmal 18, 1994.
bone cysts and are absent in so called secondary aneurysmal bone 71. Wyatt-Ashmead J, Bao L, Eilert RE, et al: Primary aneurysmal
cysts. Am J Pathol 165:1773–1780, 2004. bone cysts: 16q22 and /or 17p13 chromosomes abnormalities.
45. Oliveira AM, Perez-Atayde AR, Dal Cin P, et al: Aneurysmal bone Pediatr Dev Pathol 4:418–419, 2001.
cyst variant translocations upregulate USP6 transcription by pro-
moter swapping with ZNF9, COL1A1, TRAP150, and OMD Solitary Bone Cyst
genes. Oncogene 24:3419–3426, 2005. 72. Abdelwahab IF, Hermann G, Norton KI, et al: “Simple” bone
46. Pan Z, Sanger WG, Bridge JA, et al: A novel t(6;13)(q15;q34) cysts of the pelvis in adolescents: a report of four cases. J Bone
translocation in a giant cell reparative granuloma (solid aneurys- Joint Surg 73A:1090–1094, 1991.
mal bone cyst). Hum Pathol 43:952–957, 2012. 73. Adler CP: Tumor-like lesions in the femur with cementum-like
47. Panagopoulos I, Mertens F, Lofvenberg R, et al: Fusion of the material: does the “cementoma” of long bone exist? Skeletal Radiol
COL1A1 and USP6 genes in a benign bone tumor. Cancer Genet 14:26–37, 1985.
Cytogenet 180:70–73, 2008. 74. Baschang A, von Laer L: Indication and procedure of the opera-
48. Panoutsakopoulos G, Pandis N, Kyriazoglou I, et al: Recurrent tive treatment of benign bone cysts in children and adolescents.
t(16;17)(q22;p13) in aneurysmal bone cyst. Genes Chromosomes Eur J Pediatr Surg 1:207–209, 1991.
Cancer 26:265–266, 1999. 75. Blumberg ML: CT of lliac unicameral bone cysts. AJR Am J
49. Paulding CA, Ruvolo M, Haber DA: The Tre2 (USP6) oncogene Roentgenol 136:1231–1232, 1981.
is a hominoid-specific gene. Proc Natl Acad Sci USA 100:2507– 76. Boseker EH, Bickel WH, Dahlin DC: A clinicopathologic study
2511, 2003. of simple unicameral bone cysts. Surg Gynecol Obstet 127:550–560,
50. Petrik PK, Findlay JM, Sherlock RA: Aneurysmal cyst, bone type, 1968.
primary in an artery. Am J Surg Pathol 17:1062–1066, 1993. 77. Campanacci M, Capanna R, Picci P: Unicameral and aneurysmal
51. Raftopoulos C, Hurrel A, Ticket L, et al: Total recuperation in a bone cysts. Clin Orthop 204:25–36, 1986.
case of sudden total paraplegia due to an aneurysmal bone cyst of 78. Capanna R, Albisinni U, Caroli GC, et al: Contrast examination
the thoracic spine. Childs Nerv Syst 10:464–467, 1994. as a prognostic factor in the treatment of solitary bone cyst by
52. Rai AT, Collins JJ: Percutaneous treatment of pediatric aneurys- cortisone injection. Skeletal Radiol 12:97–102, 1984.
mal bone cysts at C1: a minimally invasive alternative: a case 79. Chang CH, Staton RP, Glutting J: Unicameral bone cyst treated
report. AJNR Am J Neuroradiol 26:30–33, 2005. by injection of bone marrow or methylprednisolone. J Bone Joint
53. Rao GS, Keogh P, Webster H, et al: Aneurysmal bone cysts in Surg Br 84:407–412, 2002.
the hand treated by free non-vascular transfer of metatarsal or 80. Chigira M, Maehara S, Arita S, et al: The aetiology and treat-
proximal phalanx from the foot. J Hand Surg [Br] 18:736–741, ment of simple bone cysts. J Bone Joint Surg Br 65:633–637,
1993. 1983.
54. Ruiter DJ, van Rijssel TG, van der Velde EA: Aneurysmal bone 81. Cohen J: Etiology of simple bone cyst. J Bone Joint Surg 52A:1493–
cysts: a clinicopathological study of 105 cases. Cancer 39:2231– 1497, 1970.
2239, 1977. 82. Conway WF, Hayes CW: Miscellaneous lesions of bone. Radiol
55. Sanerkin NG, Mott MG, Roylance J: An unusual intraosseous Clin North Am 31:339–358, 1993.
lesion with fibroblastic, osteoclastic, osteoblastic, aneurysmal and 83. Ehara S, Rosenberg AE, el-Khoury GY: Sacral cysts with exo-
fibromyxoid elements: “solid” variant of aneurysmal bone cyst. phytic components: a report of two cases. Skeletal Radiol 19:117–
Cancer 51:2278–2286, 1983. 119, 1990.
56. Scheil-Bertman S, Hartwig E, Bruderlein S, et al: Metachronous 84. Fernbach SK, Blumenthal DH, Poznanski AK, et al: Radiographic
and multiple aneurysmal bone cysts: a rare variant of primary changes in unicameral bone cysts following direct injection of
aneurysmal bone cysts. Virchows Arch 444:293–299, 2004. steroids: a report on 14 cases. Radiology 140:689–695, 1981.
57. Sciot R, Dorfman H, Brys P, et al: Cytogenetic-morphologic cor- 85. Garcia Filho RJ, Dos Santos JB, Korukian M, et al: Conservative
relations in aneurysmal bone csyt, giant cell tumor of bone and treatment of “solitary” bone cysts: a study of 55 patients. Rev Paul
combined lesions. A report from the CHAMP study group. Mod Med 110:131–137, 1992.
Pathol 13:1206–1210, 2000. 86. Grabias S, Mankin HJ: Chondrosarcoma arising in histologically
58. Scully SP, Temple HT, O’Keefe RJ, et al: Case report 830: aneu- proved unicameral bone cyst: a case report. J Bone Joint Surg
rysmal bone cyst. Skeletal Radiol 23:157–160, 1994. 56A:1501–1509, 1974.
59. Sharma P, Elangovan S, Ratnakar C: Case report: calcification 87. Graham JJ: Solitary unicameral bone cyst: follow-up study of 31
within aneurysmal bone cyst. Br J Radiol 67:306–308, 1994. cases with proven pathological diagnoses. Bull Hosp Joint Dis
60. Sherman RS, Soong KY: Aneurysmal bone cyst: its roentgen diag- 13:106–130, 1952.
nosis. Radiology 68:54–64, 1957. 88. Hoeffel JC: “Simple” bone cysts of the pelvis in adolescents: a
61. Steiner GC, Kantor EB: Ultrastructure of aneurysmal bone cyst. report of four cases. J Bone Joint Surg 74A:951–952, 1992.
Cancer 40:2967–2978, 1977. 89. Jaffe HL, Lichtenstein L: Solitary unicameral bone cyst: with
62. Sundaram M, McDonald DJ, Steigman CK, et al: Metachronous emphasis on the roentgen picture, the pathologic appearance and
multiple aneurysmal bone cysts. Skeletal Radiol 26:561–567, 1997. the pathogenesis. Arch Surg 48:1004–1025, 1944.

ERRNVPHGLFRVRUJ
1098 15  Cystic Lesions

90. Johnson LC, Vetter H, Putschar WGJ: Sarcomas arising in bone Intraosseous Ganglion
cysts. Virchows Arch Pathol Anat Physiol Klin Med 335:428–451, 115. Bauer TW, Dorfman HD: Intraosseous ganglion: a clinicopatho-
1962. logic study of 11 cases. Am J Surg Pathol 3:207–213, 1982.
91. Kragel PJ, Williams J, Garvin DF, et al: Solitary bone cyst of the 116. Crabbe WA: Intra-osseous ganglia of bone. Br J Surg 53:15–17,
radius containing Liesegang’s rings. Am J Clin Pathol 92:831–833, 1966.
1989. 117. Feldman F, Johnston A: Intraosseous ganglion. AJR Am J Roent-
92. Kuttenberger JJ, Farmand M, Stoss H: Recurrence of a “solitary” genol 118:328–343, 1971.
bone cyst of the mandibular condyle in a bone graft: a case report. 118. Glass TA, Dyer R, Fisher L, et al: Expansile subchondral bone
Oral Surg Oral Med Oral Pathol 74:550–556, 1992. cyst. AJR Am J Roentgenol 139:1210–1211, 1982.
93. Matsumoto K, Fujii S, Mochizuki T, et al: “Solitary” bone cyst of 119. Grange WJ: Subperiosteal ganglion: a case report. J Bone Joint
a lumbar vertebra: a case report and review of literature. Spine Surg Br 60:124–125, 1978.
15:605–607, 1990. 120. Kambolis C, Bullough PG, Jaffe HI: Ganglionic cystic defects of
94. McKay DW, Nason SS: Treatment of unicameral bone cysts by bone. J Bone Joint Surg 55A:496–505, 1973.
subtotal resection without grafts. J Bone Joint Surg 59A:515–519, 121. Landells JW: The bone cysts of osteoarthritis. J Bone Joint
1977. Surg Br 35:643–649, 1953.
95. Mirra JM, Bernard GW, Bullough PG, et al: Cementum-like 122. Mainzer F, Minagi H: Intraosseous ganglion: a solitary subchon-
bone production in solitary bone cysts (so-called “cementoma” of dral lesion of bone. Radiology 94:387–389, 1970.
long bones): report of three cases—electron microscopic observa- 123. McBeath AA, Neidhart DA: Acetabular cyst with communicating
tion supporting a synovial origin to the simple bone cyst. Clin ganglion: a case report. J Bone Joint Surg 58A:267–269, 1976.
Orthop 135:295–307, 1978. 124. Ostlere SJ, Seeger LL, Eckardt JJ: Subchondral cysts of the tibia
96. Mylle J, Burssens A, Fabry G: “Simple” bone cysts: a review of 59 secondary to osteoarthritis of the knee. Skeletal Radiol 19:287–289,
cases with special reference to their treatment. Arch Orthop 1990.
Trauma Surg 111:297–300, 1992. 125. Pope TL, Jr, Fechner RE, Keats TE: Intra-osseous ganglion:
97. Nakagawa T, Kawano H, Kubota T: “Solitary” bone cyst of the report of four cases and review of the literature. Skeletal Radiol
cervical spine: case report. Neurol Med Chir (Tokyo) 34:558–560, 18:185–187, 1989.
1994. 126. Rahney K, Lamb DW: The cysts of osteoarthritis of the hip: a
98. Neer CS, Francis KC, Marcove RC, et al: A follow-up study of radiographic and pathological study. J Bone Joint Surg Br 37:663–
175 cases: treatment of unicameral bone cyst. J Bone Joint Surg 675, 1955.
48A:731–745, 1966. 127. Schajowicz F, Clavel Sainz M, Slullitel JA: Juxta-articular bone
99. Ovadia D, Ezra E, Segev E, et al: Epiphyseal involvement of cysts (intra-osseous ganglia): a clinicopathological study of eighty-
simple bone cyst. J Pediatr Orthop 23:222–229, 2003. eight cases. J Bone Joint Surg Br 61:107–116, 1979.
100. Pogoda P, Priemel M, Linhart W, et al: Clinical relevance of 128. Sim FH, Dahlin DC: Ganglion cysts of bone. Mayo Clin Proc
calcaneal bone cyst: a study of 50 cyst in 47 patients. Clin Orthop 46:484–488, 1971.
Relat Res 424:202–210, 2004. 129. Yale JF, Gaeta M: Intraosseous ganglion of the navicular. J Am
101. Rougraff BT, Kling TJ: Treatment of active unicameral bone Podiatr Med Assoc 84:124–126, 1994.
cyst with percutaneous injection of demineralized bone matrix
and autogenous bone marrow. J Bone Joint Surg 84A:921–929,
2002. Epidermal and Dermoid Cysts
102. Rud B, Pedersen NW, Thomsen PB: “Simple” bone cysts in 130. Alpers BJ, Harrow R: Cholesteatoma originating in the skull
children treated with methylprednisolone acetate. Orthopedics bones causing symptoms of intracranial pressure. Am J Surg 18:
14:185–187, 1991. 51–57, 1932.
103. Saito Y, Hoshina Y, Nagamine T, et al: “Simple” bone cyst: a 131. Canale DJ, Clarke HA, Pak YH, et al: Giant primary intradiploic
clinical and histopathologic study of 15 cases. Oral Surg Oral Med epidermoid tumor of the skull: case report. Surg Neurol 2:51–54,
Oral Pathol 74:487–491, 1992. 1974.
104. Sanerkin NG: Old fibrin coagula and their ossification in simple 132. Carroll RE: Epidermoid (epithelial) cyst of the hand skeleton. Am
bone cysts. J Bone Joint Surg Br 61:194–199, 1979. J Surg 85:327–332, 1953.
105. Scaglietti O, Marchetti PG, Bartolozzi P: The effects of 133. Constans JP, Meder JF, De Devitiis E, et al: Giant intradiploic
methylprednisolone acetate in the treatment of bone cysts: epidermoid cysts of the skull: report of two cases. J Neurosurg
results of 3 years follow-up. J Bone Joint Surg Br 61:200–204, 62:445–448, 1985.
1979. 134. Cronqvist S, Wholey MH: Intracranial involvement in epider-
106. Sethi A, Agarwal K, Sethi S, et al: Allograft in the treatment of moid and dermoid tumors of the skull. Acta Radiol Diagn (Stockh)
benign cystic lesions of bone. Arch Orthop Trauma Surg 112:167– 2:194–200, 1964.
170, 1993. 135. Demaerel P, Wilms G, Lammens M, et al: Extradural epidermoid
107. Smith RW, Smith CF: Solitary unicameral bone cyst of the calca- tumor of the frontal bone. Neuroradiology 33:349–351, 1991.
neus. J Bone Joint Surg 56A:49–56, 1974. 136. Exner G, Hort W, Boger A: Epidermoidzyster der tibia. Z Orthop
108. Struhl S, Edelson C, Pritzker H, et al: Solitary (unicameral) bone Ihre Grenzgeb 116:362–368, 1978.
cyst: the fallen fragment sign revisited. Skeletal Radiol 18:261–265, 137. Garcia J, Lagier R, Hoessly M: Computed tomography–pathology
1989. correlation in skull epidermoid cyst. J Comput Assist Tomogr 6:818–
109. Taneda H, Azuma H: Avascular necrosis of the femoral epiphysis 820, 1982.
complicating a minimally displaced fracture of “solitary” bone cyst 138. Haig PV: Primary epidermoids of the skull: including a case
of the neck of the femur in a child: a case report. Clin Orthop with malignant changes. Am J Roentgenol Radium Ther Nucl Med
304:172–175, 1994. 76:1076–1080, 1956.
110. Telfer MR, Jones GM, Pell GM, et al: Primary bone cyst of 139. Harrison MJ, Morgello S, Post KD: Epithelial cystic lesions of
the mandibular condyle. Br J Oral Maxillofac Surg 28:340–343, the sellar and parasellar region: a continuum of ectodermal deriva-
1990. tives? J Neurosurg 80:1018–1025, 1994.
111. Tsai JC, Dalinka MK, Fallon MD, et al: Fluid-fluid level: a non- 140. Lerner MR, Southwick WO: Keratin cysts in phalangeal bones:
specific finding in tumors of bone and soft tissue. Radiology report of an unusual case. J Bone Joint Surg 50A:365–372, 1968.
175:779–782, 1990. 141. Lunardi P, Missori P, Gagliardi Fortuna A: Dermoid and epider-
112. Violas P, Salmeron F, Chappuis M, et al: Simple bone cyst of the moid cysts of the midline in the posterior cranial fossa. Neurosurg
proximal humerus complicated with growth arrest. Acta Orthop Rev 15:171–175, 1992.
Belg 70:166–170, 2004. 142. Neblett CR, Caram PC, Morris R: Lateral congenital dermal
113. Watanabe H, Arita S, Chigira M: Aetiology of a “simple” bone sinus tract associated with an intradiploic dermoid tumor. Case
cyst: a case report. Int Orthop 18:16–19, 1994. report. J Neurosurg 33:103–105, 1970.
114. Wu KK: A surgically treated unicameral (“solitary”) bone cyst of 143. Oda Y, Hashimoto H, Tsuneyoshi M, et al: Case report #742:
the talus with a 15-year follow-up. J Foot Ankle Surg 32:242–244, intraosseous epidermoid cysts arising in the fifth metacarpal bone.
1993. Skeletal Radiol 21:343–345, 1992.

ERRNVPHGLFRVRUJ
15  Cystic Lesions 1099

144. Reddick EJ, Ettinger D, Madauss WC: Dermoid cysts of 148. Siddiqui MA, Francis DA, Zaman MN: Giant epidermoid cyst
the anterior fontanelle: case reports. Mil Med 148:548–550, near the anterior fontanelle. Br J Hosp Med 50:484–485,1993.
1983. 149. Swisher RC, Jr, Tesluk H: Epidermoid cyst of the skull causing
145. Rengachary S, Kishore PRS, Watanabe I: Intradiploic epidermoid displacement of brain. JAMA 210:1280–1281, 1969.
cyst of the occipital bone with torcular obstruction. Case report. 150. Vogel HJ, Goddard RE, van den Bouwhuysen PW, et al: Epider-
J Neurosurg 48:475–478, 1978. mal cyst of the skull. J Belge Radiol 76:418, 1993.
146. Roth SI: Squamous cysts involving the skull and distal phalanges. 151. Wax MK, Briant TD: Epidermoid cysts of the cranial bones. Head
J Bone Joint Surg 46A:1442–1450, 1964. Neck 14:293–296, 1992.
147. Schajowicz F, Aiello CL, Slullitel I: Cystic and pseudocystic 152. Yanai Y, Tsuji R, Ohmori S, et al: Malignant change in an intra-
lesions of the terminal phalanx with special reference to epider- diploic epidermoid: report of a case and review of the literature.
moid cyst. Clin Orthop 68:84–92, 1970. Neurosurgery 16:252–256, 1985.

ERRNVPHGLFRVRUJ
C H A P T E R 1 6 

Miscellaneous Mesenchymal
Tumors

CHAPTER OUTLINE

LIPOMA PERIVASCULAR EPITHELIOID CELL TUMOR


Intramedullary Lipoma LEIOMYOSARCOMA
Intracortical and Subperiosteal Lipoma
Parosteal Lipoma RHABDOMYOSARCOMA

LIPOSARCOMA MULTIPOTENTIAL PRIMITIVE SARCOMA,


POLYHISTIOMA, AND MALIGNANT
VASCULAR-CARTILAGINOUS HAMARTOMA OF MESENCHYMOMA
THE CHEST WALL
MELANOTIC NEUROECTODERMAL TUMOR OF
INFANCY

LIPOMA (Figs. 16-2 and 16-3). Magnetic resonance imaging may


Definition show no discernible signal alteration because lipoma is
composed of mature adipose tissue with signal character-
Lipoma is a rare primary tumor of bone composed istics similar to normal fatty marrow (Fig. 16-1). Lipomas
of mature adipose tissue. Lipomas involving bone with extensive fat necrosis may appear as low signal
are divided into three types: intramedullary (intraosse- density lesions with signal void in areas of calcifications
ous), intracortical/subperiosteal, and parosteal. Lipomas (Fig. 16-4).
involving synovial tissue are described in Chapter 20.
Gross Findings
Incidence and Location
The gross appearance of lipoma is that of a well-
Lipomas of bone are extremely rare; fewer than 50 cases circumscribed, lobular mass of adipose tissue (see
are described in the literature. Fig. 16-2).

Intramedullary Lipoma Microscopic Findings


Intramedullary lipomas usually occur in the metaphyseal Histologically, lipomas consist of mature adipose tissue
parts of major long bones of the lower extremity—the and do not differ from ordinary lipomas of soft tissue
femur, tibia, and fibula.3-6,8-14,16,17,20,21,23,25,26,28,29,34 The cal- (Fig. 16-4). The presence of fat necrosis with dystrophic
caneus is also a common site.1,12,19,21,22,27,31,33 Lipomas also calcifications, which correspond to the central, niduslike
have been reported in the sacrum, coccyx, mandible, opacity seen radiographically, is frequently seen (Figs.
maxilla, skull, ribs, and metatarsals.36,37 Pain and swelling 16-3 and 16-4).
are the most frequent presenting symptoms, but most of
these lesions are asymptomatic and may only be inciden-
tally noted on radiographs.
Intracortical and Subperiosteal Lipoma
Intracortical and subperiosteal lipomas are extremely
rare.2,24,28,38 They radiographically present as intracortical
Radiographic Imaging
or subperiosteal lucent lesions with intralesional trabecu-
Radiographically, intramedullary lipoma is seen as a lytic lations and foci of dotlike calcifications. Histologically,
lesion with trabeculation (Figs. 16-1 and 16-2). Bone these lesions consist of mature adipose tissue and, similar
expansion with a thinned but intact cortex may be to intramedullary lipomas, they do not differ from ordi-
present (Fig. 16-2). Central, niduslike calcification is a nary lipomas of soft tissue.
frequent radiographic feature of intramedullary lipoma Text continued on p. 1105

1100
ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1101

C
FIGURE 16-1  ■  Intramedullary lipoma: radiographic features. A and B, Anteroposterior and lateral radiographs show lytic lobulated
lesion involving proximal tibial metaphysis in skeletally mature patient. Biopsy revealed mature adipose tissue. C, Coronal magnetic
resonance image of intramedullary lipoma of tibia shows no discernible marrow signal alteration because lipoma is composed
of mature adipose tissue. There is some expansion of contour of medial cortex overlying lesion. (Courtesy of Dr. German Steiner,
New York.)

ERRNVPHGLFRVRUJ
1102 16  Miscellaneous Mesenchymal Tumors

B
FIGURE 16-2  ■  Intramedullary lipoma: radiographic and gross features. A, Anteroposterior radiograph shows lytic lesion involving
distal end of fibula. Note bone contour expansion and central niduslike opacities corresponding to areas of fat necrosis. B, Fibular
resection specimen shows lesion composed of adipose tissue. (Courtesy Dr. Leonard Kahn, New York.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1103

A B

C D
FIGURE 16-3  ■  Lipoma of bone: cystic tumor of calcaneus with fat necrosis. A, Lateral radiograph of foot of a 40-year-old woman
who had pain for 1 year. Anterior portion of calcaneus shows lytic lesion with ringlike calcific density in its center. B, Coronal com-
puted tomogram shows lucent lesion in calcaneus that has central cyst; walls of cyst are calcified. C, On curettage, lesion was partly
cystic and contained calcified necrotic fat; fragment of cyst wall. D, Calcified necrotic fat in center of lipoma. (C, ×40; D, ×100) (C and
D, hematoxylin-eosin.) (Courtesy Dr. M. Nuovo, New York.)

ERRNVPHGLFRVRUJ
1104 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-4  ■  Intramedullary lipoma: radiographic and microscopic features. A, Anteroposterior radiograph of proximal femur shows
lytic intertrochanteric lesion with well demarcated sclerotic margins. Note multifocal coalescent opacities corresponding to calcified
areas of fat necrosis. B, Magnetic resonance image (MRI) shows a well demarcated intramedullary lesion with signal void in areas
corresponding to calcifications. Inset, T2-weighted MRI shows signal enhancement with signal void in areas corresponding to cal-
cifications. C, Lesion is composed of mature adipose tissue. Note the absence of bone trabeculae, a feature helpful in distinguishing
this lesion from normal fatty bone marrow. D, Central portion of lesion exhibits features of fat necrosis and dystrophic calcification.
(C and D, ×50) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1105

Parosteal Lipoma Clinical Behavior


Parosteal lipomas are not true bone tumors and belong Approximately 50% of patients who were reported to
to the category of soft tissue lesions. Because of their have primary liposarcoma of bone had metastases after
association with the bone surface, parosteal lipomas may amputation.
have some distinctive radiographic and microscopic
features. Similar to intramedullary lipomas, parosteal
lipomas of bone are extremely rare.7,15,17,18,20,30,32,35 They
usually involve the metaphyseal parts of long tubular
VASCULAR-CARTILAGINOUS
bones, but they may involve other bones, such as the flat HAMARTOMA OF THE CHEST WALL
bones of the trunk and the bones of the upper extremities Definition
(Fig. 16-5). A characteristic feature of parosteal lipoma
(not always seen) is a sclerotic, osseous pedicle that grows Vascular-cartilaginous hamartoma is a rare and distinct
from the surface of the bone into the adipose tissue. This clinical, radiologic, and pathologic entity that presents at
bony excrescence can dominate the lesion and can result birth or early infancy as an expansile intraosseous lesion
in radiographic misdiagnosis of the lesion as an osteo- involving the central portion of one or more ribs.
chondroma. Foci of cartilaginous metaplasia with forma-
tion of a cartilaginous cap may be present. Incidence and Location
McLeod and Dahlin56 reported nine cases from the Mayo
Clinic’s consultation files. The lesion typically involves the
LIPOSARCOMA chest wall and most likely originates within the ribs.49-59
Definition
Gross Findings
Liposarcoma is an extremely rare primary sarcoma of
bone; it is composed of immature adipose tissue. The gross findings of these tumors are those of smooth,
well-circumscribed, red-brown masses that arise from the
surface of the rib. The cut surface is marked by the pres-
Incidence and Location
ence of many dilated channels filled with blood. These
Liposarcoma of bone is one of the rarest of primary bone spaces are separated by a friable, red-brown stroma that
tumors.39-48 In 1982, Addison and Payne accepted only six contains variable amounts of gritty, shiny, white, carti-
examples of this entity in bone from previously published laginous tissue.
reports.24 The patients ranged in age from 15 to 53 years
but usually had symptoms during the third and fourth Radiographic Imaging
decades of life. Reported cases of intraosseous liposar-
coma involved the major long tubular bones, such as the Radiographically vascular-cartilaginous hamartoma pres-
femur, tibia, and humerus. ents as an expansile lesion of the rib. Magnetic reso-
nance imaging typically shows signal enhancement in
T2-weighted images corresponding to the high water
Radiographic Imaging
content in both cartilaginous and vascular components
Radiographic features are nonspecific and show a bone- of the lesion (Figs. 16-6 and 16-8).
destructive process indicative of a malignant tumor.
Microscopic Findings
Microscopic Findings
Microscopically, these lesions show a mixture of cellular
Microscopically, liposarcomas in bone represent high- fibrous tissue that contains many blood-filled, endothelial-
grade pleomorphic or round-cell (signet-ring) lesions. lined vascular spaces; numerous islands of randomly dis-
Some of these tumors may have features of myxoid tributed mature hyaline cartilage; and reactive new bone
liposarcoma. (Figs. 16-6 and 16-7). The cartilage islands represent
well-developed nodules of cartilaginous tissue that are
well demarcated from the surrounding vascular compo-
Differential Diagnosis
nent and occasionally form irregular clusters. The vascu-
The high-grade pleomorphic liposarcoma should be lar component is similar to conventional hemangioma
differentiated from malignant fibrous histiocytoma. In the and may contain large, highly cellular areas composed of
evaluation of adipose tissue and differentiation of intraos- spindle cells forming narrow vascular channels. The
seous lesions, the presence of residual nonneoplastic spindle-cell areas may show prominent mitotic activity,
tissue of fatty marrow infiltrated by a nonlipomatous but atypical mitoses are absent. Larger-caliber vessels are
tumor should be ruled out before the lesion is classified also present and may form clusters mimicking common
as a liposarcoma. It is our impression that at least some (cavernous) hemangioma. Vascular-cartilaginous hamar-
of the cases previously reported as primary liposarcomas toma frequently shows features of superimposed second-
of bone would be currently classified as malignant fibrous ary aneurysmal bone cyst.49,54-56 In fact, aneurysmal bone
histiocytomas. Text continued on p. 1110

ERRNVPHGLFRVRUJ
1106 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-5  ■  Parosteal lipoma: radiographic, gross, and microscopic features. A, Lateral radiograph of femur shows multilobulated,
partially lucent parosteal tumor containing areas of mineralization. B, Corresponding magnetic resonance image demonstrates
parosteal location of tumor, which for the most part shows same signal intensity as marrow fat. C, Gross specimen of excised
tumor mass shown in A and B. It consists of lobulated adipose tissue covered by thin fibrous capsule. Bony spur was
present at point of attachment to cortex. D, Photomicrograph shows mature adipose tissue traversed by fibrous bands. (D, ×100)
(D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1107

A B

C D
FIGURE 16-6  ■  Vascular-cartilaginous hamartoma of rib in infancy. A, Chest radiograph shows large, ill-defined intrathoracic expansile
mass arising from the ninth and tenth ribs of a 7-month-old boy. B, Bulging encapsulated tumor mass compresses lung. Most of
this mass was composed of aneurysmal bone cyst component of cartilaginous hamartoma. C, Radiograph of resection specimen
of cartilaginous hamartoma that arose in rib with secondary aneurysmal bone cyst formation. Expansile lytic lesion with focal cal-
cification is seen in central rib. D, Whole-mount section of rib shows intramedullary cartilage lobules and dilated vascular channels
(hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1108 16  Miscellaneous Mesenchymal Tumors

B
FIGURE 16-7  ■  Vascular-cartilaginous hamartoma of rib in infancy: microscopic features. A, Islands of mature cartilage within fibro-
vascular stroma containing numerous capillary-type vessels. B, Higher magnification of A shows islands of cartilage within spindle-
cell stroma with numerous capillary vessels. Inset, Higher magnification of spindle-cell area. Note focal brisk mitotic activity.
(A, ×100; B, ×200; Inset, ×400) (A, B, and Inset, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1109

C
FIGURE 16-8  ■  Vascular-cartilaginous hamartoma of rib in infancy: radiographic features. A and B, T1-weighted magnetic resonance
images (MRI) showing a well demarcated low signal lesion of the chest wall. C, T2-weighted MRI showing signal enhancement.

ERRNVPHGLFRVRUJ
1110 16  Miscellaneous Mesenchymal Tumors

cyst features can dominate the lesion. Some of the lesions epithelial alveolar structures are loosely distributed. This
may contain coalescent nodules of mature hyaline carti- may create a more disorganized pattern of neuroblastic
lage that obscure the spindle and vascular component of cells and solid ephithelial proliferations. The presence
the lesion (Fig. 16-9). of heterologous elements (i.e., mesenchymal spindle
cells and woven bone) has also been described but is
Treatment and Behavior extremely rare.83
The tumor cells, particularly the epithelial pigment–
These rare lesions are benign and may be cured by en containing cells, are positive for keratin and occasionally
bloc excision of the mass in continuity with the involved positive for epithelial membrane antigen and the HMB-45
rib. Rapid enlargement of the lesion can be clinically marker. In addition, both components (i.e., small neuro-
observed if vascular-cartilaginous hamartoma is compli- blastic cells and epithelial cells) are positive for neuron-
cated by a superimposed aneurysmal bone cyst.49,54-56 specific enolase and Leu-7 and occasionally are positive
for synaptophysin. Glial fibrillary acidic and S-100
proteins may be focally positive in a small-cell neuroblas-
tic component. This immunohistochemical pattern is
MELANOTIC NEUROECTODERMAL consistent with the postulated neuroectodermal deriva-
TUMOR OF INFANCY tion of these tumors.73,84,86,92,81
Definition
Treatment and Behavior
Melanotic neuroectodermal tumor of infancy is a rare,
distinctive neoplasm that frequently affects children Melanotic neuroectodermal tumors of infancy generally
younger than age 1 year. The tumor is composed of nests behave as benign lesions, but local recurrence is possible.
of cells that have melanin pigment and is also referred to In addition, approximately 6% of patients develop distant
as retinal anlage tumor, melanotic progonoma, or melanotic metastases. The development of distant metastases cannot
ameloblastoma. be correlated with any specific microscopic features,
the distribution of DNA ploidy, or tumor proliferation
parameters.
Incidence and Location
Melanotic neuroectodermal tumors usually involve the
maxilla or mandible but also occur in the soft tissue,
the epididymis, and the mediastinum or may present as
PERIVASCULAR EPITHELIOID
intracranial (dural or brain) masses.60,62-64,67-69,71,72,79,80,82,85 CELL TUMOR
They may present as lesions in the long bones. The Definition
lesion can be clinically associated with increased levels
of catecholamines and urinary excretion of vanillylman- Perivascular epithelioid cell tumor (PEComa) is a rare
delic acid.61,70 distinct neoplasm of bone made up of epithelioid clear
cells rich in glycogen and characterized by coexpres-
sion of muscle and melanocytic markers. This term has
Radiographic Imaging
been applied to an expanding family of tumors that are
Radiographically, the tumor presents as a well- presumed to originate from microscopically and pheno-
demarcated, lytic bone lesion.63,72,80 The preoperative typically unique perivascular epithelioid cells, although
diagnosis can be suspected radiographically if the lesion there is no known normal cellular counterpart to these
presents in typical locations—the maxilla or mandible— cells, and precursor lesions for PEComas have not been
in a child during the first year of life. identified.88-91,99,101 It includes angiomyolipoma (both renal
and extrarenal), lymphangiomyomatosis, clear cell sugar
tumor of the lung, and a variety of spindled and epithe-
Gross Findings
lioid neoplasms involving various organs, soft tissue, and
The lesion is fibrous and tan, brown, or black, depending bone with a similar phenotype.88,92,93,96,98,100,105,108-110,114,116
on the proportion of the pigmented component. The unifying concept proposed for this family of tumors
evolved over the past century and was developed by iden-
tifying unique clear cell features as well as the smooth
Microscopic Findings
muscle and melanocytic characteristics of the tumor cells.
Microscopic features are characteristic and consist of two A distinct type of benign clear cell tumor referred to as
basic cell populations: small neuroblastic cells and larger, clear cell sugar tumor was described in the lung in 1963 by
melanin-containing epithelial cells arranged in a distinct Liebow and Castleman.107 The name refers to the clear
architectural pattern.65,66,73-78,81 The tumor cells form cytoplasm of the cells, which is rich in glycogen. Despite
nests within a predominantly fibrous stroma. The nests their epithelioid appearance, the clear cells are negative
are delineated by epithelial, pigment-containing cells for epithelial markers and exhibit distinctive coexpression
(Figs. 16-10 to 16-14). The central portion of these struc- of muscle and melanoma markers.88,89,97,100,105,106,112,115 The
tures with alveolar or tubular arrangements is filled by same immunophenotypic features are also present in a
small, loosely arranged neuroblastic cells in a neurofibril- subset of cells in lymphangiomatosis and angiomyolipoma.
lary matrix (Fig. 16-13, B to D). The neuroblastic cells On the basis of the common and distinctive phenotype,
occasionally form larger, more diffuse areas in which Text continued on p. 1117

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1111

A B

C D
FIGURE 16-9  ■  Vascular-cartilaginous hamartoma of rib in infancy: microscopic features. A and B, Low power view of predominantly
cartilaginous component of the lesion with small islands of fibrovascular cores showing enlarged vascular channels. C and D, Higher
magnification showing mature hyaline cartilage and fibrous component with prominent vascular channels. (A and B, ×50; C and
D, ×100.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1112 16  Miscellaneous Mesenchymal Tumors

A B

C D

FIGURE 16-10  ■  Melanotic neuroectodemal tumor of infancy: radiographic features. A and B, Axial and coronal magnetic resonance
images show variable signal intensity tumor involving left lateral orbit in the greater wing of sphenoid bone in a 3-month-old girl.
C and D, Axial computed tomograms of the lesion in A and B showing mixed sclerotic and lytic tumor involving left lateral orbit.

ERRNVPHGLFRVRUJ
A

FIGURE 16-11  ■  Melanotic neuroectodermal tumor of infancy: microscopic features. A, Epithelial cells form tubular or alveolar struc-
tures in fibrous stroma. B, Central portions of epithelial-lined structures are filled with smaller, loosely arranged neuroblastic cells.
Inset (top), Higher magnification of A documents epithelial appearance of cells lining tubular structures that contain melanotic
granules. Inset (bottom), Higher magnification of B shows small neuroblastic cells filling larger alveolar spaces. Note loose fibrillar
intercellular matrix. (A and B, ×100; Insets, ×200) (A, B, and Insets, hematoxylin-eosin.) (Courtesy Dr. M.A. Luna, Houston.)

ERRNVPHGLFRVRUJ
1114 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-12  ■  Melanotic neuroectodermal tumor of infancy: microscopic features. A, Low power magnification of complex tubular
structures in fibrous stroma. B, Higher power view of tubular structures lined by epithelial cells containing melanin pigment. C, Low
power magnification of large alveolar structures. D, Higher magnification of C shows small neuroblastic cells filling alveolar space.
(A and C, ×100; B, ×400; D, ×200) (A-D, hematoxylin-eosin). (Courtesy Dr. M.A. Luna, Houston.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1115

A B

C D
FIGURE 16-13  ■  Melanotic neuroectodemal tumor of infancy: microscopic features. A, Intermediate power view of a tumor showing
proliferation of small loosely arranged neuroblastic cells. B, Higher magnification of A showing proliferation of primitive neuroblastic
cells. C, Intermediate power view showing proliferation of epithelioid cells and small loosely arranged cluster of small neuroblastic
cells. D, Higher magnification of C showing clusters of small neuroblastic cells adjacent to areas of solid proliferations of epithelioid-
appearing cells. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1116 16  Miscellaneous Mesenchymal Tumors

B C
FIGURE 16-14  ■  Melanotic neuroectodemal tumor of infancy: microscopic features. A, Low power view showing strong positive
staining for cytokeratin in epithelioid cells. Small neuroblastic cells are negative. B, Higher magnification of A showing strong posi-
tive staining for keratin in the epithelioid component of the tumor. C, Coexpression of HMB-45 in the epithelioid component of the
tumor. (A, ×100; B and C, ×200)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1117

Bonetti et al, Pea et al, and Zamboni et al defined a family negative for epithelial markers such as epithelial mem-
of lesions characterized by the presence of this peculiar brane antigen and keratins (CAM5.2 and AE1:AE3) but
cell type, the perivascular epithelioid cell (PEC), and coexpress muscle markers (smooth muscle actin [SMA],
designated the group of tumors with similar features as muscle-specific actin [MSA], and desmin) and melano-
PEComas.88,89,90,118 These tumors have also been referred cytic antigens (HMB45 and melanin A) (Fig. 16-18, D).
to as PEST (primary extrapulmonary sugar tumors).114 These lesions are also positive for neuron-specific enolase,
Patients with one or more of the PEC family of lesions CD68, and sometimes for S-100. Ultrastructurally the
may have the tuberous sclerosis complex. The tuber- tumor cells form well-developed nests surrounded by
ous sclerosis complex is typically associated with angio- rudimentary basement membrane with prominent capil-
myolipomas and lymphangiomyomatosis, but PEComas lary network encircling individual tumor cell nests. The
in other anatomic sites can develop in this syndrome.91 cytoplasm is rich in glycogen and contains multiple mito-
Two-step inactivation of the TSC1 and TSC2 genes acti- chondria. Premelanosomes and dense granules are often
vating the Rheb/mTOR/p70S6K pathway underlie the observed but are not always present. Poorly developed
development of PEComas in tuberous sclerosis.101,117 intercellular junctions are present but true desmosomes
Comparative genomic hybridization studies of PEComas are not found.
disclose frequent losses and gains of multiple chro-
mosomes, implicating major chromosomal rearrange-
Treatment and Behavior
ments.111 Some PEComas show rearrangements of the
transcription factor E3 (TFE3) gene and overexpression Long-term follow-up on the four cases indicates that
of the TFE3 protein immunohistochemically.87,95,104,113 clear cell sugar tumors of bone are of uncertain malignant
potential. Three of our cases behaved as benign tumors
Incidence and Location but one metastasized to the lung after many years of
follow-up. Reported cases of PEComas in bone indicate
Our experience is restricted to four cases of primary clear that both primary and metastatic lesions can be found in
cell sugar tumor of bone seen in consultation. These pre- the skeleton. Clinical follow-up with primary cases is very
sented in the calcaneus of a 9-year-old boy, the humerus limited, but indicates a metastatic potential for those
of a 10-year-old girl, the patella of a 29-year-old man, and tumors that originate as primary bone lesions.94,102,106,117
the proximal phalanx of the middle finger of the left hand
in a 33-year-old woman. One of the cases showed a clonal
cytogenetic abnormality consisting of reciprocal chromo- LEIOMYOSARCOMA
somal translocation t(X;2) (q11-12; q34-35). Definition

Radiographic Imaging Leiomyosarcoma is a malignant mesenchymal tumor


composed predominantly of spindle cells that exhibit
Radiographic features are nonspecific and consist of smooth muscle differentiation.
a well-demarcated lucent lesion with a trabeculated
appearance and sclerotic margins (Figs. 16-15 and 16-
Incidence and Location
16). In general, the tumors arise in skeletal sites that are
relatively rare for primary bone tumors. Primary leiomyosarcoma in bone is extremely rare, and
fewer than 100 well-documented cases have been
described. The peak age incidence and most frequent
Microscopic Findings
sites of skeletal involvement are shown in Figure 16-19.
Microscopically clear cell sugar tumors consist of clear Many bona fide primary leiomyosarcomas of bone may
cells arranged in nests with prominent arborizing capil- prove to be metastatic, and the primary lesions are most
lary network demarcating the tumor cell nests (Figs. frequently located in the uterus and gastrointestinal
16-17 and 16-18). The nuclei are uniform, oval or round, tract.123,129,142,154 Some of these lesions present many years
with prominent nucleoli and fine granular chromatin. after excision of allegedly benign smooth muscle tumors
Mitoses are rare and atypical mitoses are absent. Some in the uterus or gastrointestinal tract. The ages of affected
tumor cells contain pink granular cytoplasm but most patients are widely distributed, but primary leiomyosar-
are vacuolated, which gives the tumor a clear cell appear- coma of bone is rare during the first two decades of
ance. Reported cases of PEComas in bone display a spec- life.120-122,126,132,145,150,155,159
trum of microscopic features ranging from clear cell The most common site is the distal femur followed
lesions, somewhat similar to those described for pulmo- by the proximal tibia. These bones are involved in
nary clear cell sugar tumors, to highly pleomorphic almost 50% of reported cases, but any bone can be
lesions with necrosis and brisk mitotic activities reminis- affected.119,121,128,130-132,136,141,144,153,157 Other major long
cent of those observed in a subset of epithelioid renal tubular bones of the extremities are also frequently
angiomyolipomas.94,102,103,106,117 involved. The pelvis is the most frequently involved trunk
bone. Leiomyosarcoma of bone also has been described
as one of the rare complications of radiation therapy.140
Special Techniques
Leiomyosarcomas occur in immunocompromised
The cytoplasm of clear cells is rich in glycogen and stains patients such as those who have undergone organ trans-
positively with periodic acid–Schiff (PAS). Despite their plants and patients with acquired immunodeficiency
epithelioid appearance the clear cells are consistently Text continued on p. 1122

ERRNVPHGLFRVRUJ
1118 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-15  ■  Clear cell sugar tumor (PEComa): radiographic features. A, Well demarcated lucent lesion with a trabeculated appear-
ance and sclerotic margins involving the calcaneus of a 9-year-old boy. B, Intramedullary lucency with focal sclerosis and trabecula-
tion involving the proximal phalanx of the middle finger in a 33-year-old woman. C, Lucent lesion with trabeculation and focal
penetration of the cortex in the patella of a 29-year-old man. D, Sagittal magnetic resonance image of the lesion shown in C depict-
ing an intramedullary low signal intensity mass in the patella.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1119

A B

C D
FIGURE 16-16  ■  Clear cell sugar tumor (PEComa): radiographic features. A and B, Anteroposterior and lateral views of the lesion
involving distal humerus of a 10-year-old girl. Note an expansile lytic lesion with trabeculations. The cortex shows thinning and
endosteal scalloping. C and D, T1- and T2-weighted axial magnetic resonance images showing an expansile intramedullary lesion
with thin but intact overlying cortex.

ERRNVPHGLFRVRUJ
1120 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-17  ■  Clear cell sugar tumor (PEComa): microscopic features. A and B, Low and intermediate power view of PEComa
composed of clear cells arranged in nests with prominent capillary network. C and D, Higher magnification of PEComa showing
nests of tumor cells with clear cytoplasm and nuclei containing prominent typically singular nucleoli. (A, ×25; B, ×50; C, ×100;
D, ×400.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1121

C D
FIGURE 16-18  ■  Clear cell sugar tumor (PEComa): microscopic features. A, Nests of primarily granular cells surrounded by prominent
capillary network. Inset, Higher magnification of a nest of tumor cells with predominantly granular cytoplasm and focal clear cell
features. B, Nests of clear cells demarcated by prominent capillary network. C, Higher magnification of B showing nests of clear
cells. D, Strong diffuse cytoplasmic stain of PEComa cells for HMB-45 protein. (A and B, ×100; Inset, C and D, x 200) (A-C and
inset, hematoxylin-eosin; D, DAB.)

ERRNVPHGLFRVRUJ
1122 16  Miscellaneous Mesenchymal Tumors

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 16-19  ■  Leiomyosarcoma of bone. Peak age incidence and most frequent sites of skeletal involvement. Most frequent sites
are indicated by solid black arrows.

Gross Findings
syndrome (AIDS) with increased frequency as compared
to the general population.124,125,151,158,160 Leiomyosarcomas When the lesion is examined in situ, the cut surface is
associated with AIDS predominantly occur in children gray to white with areas of necrosis (Fig. 16-25). The
and young adults. These tumors are often multifocal tumor is usually well demarcated from the surrounding
and primarily involve the gastrointestinal tract, liver, and bone or soft tissue, and this feature can be misleading. In
lung.127,139,146,147,163 The development of leiomyosarco- fact, microscopic margins show an infiltrative irregular
mas in immunocompromised patients is associated with pattern of involvement of adjacent structures.
Epstein-Barr virus (EBV) infection; EBV surface antigen
(CD21) has been documented in these lesions.134,135,138 Microscopic Findings
Although some rare examples of primary leiomyosarcoma
of bone occur in children, no association with immuno- The microscopic appearance of primary leiomyosarcoma
suppression and EBV infection has been reported for of bone does not differ from that of more common
these tumors. Leiomyosarcomas of the soft tissue and uterine, gastrointestinal, or soft tissue leiomyosarco-
bone may also develop in familial retinoblastoma syn- mas.150,159,161 The tumor is composed of interweaving fas-
drome caused by germline mutations of RB1 mapping to cicles of spindle cells that have prominent eosinophilic
13q14 locus (Figs. 16-24 and 16-29). cytoplasm and elongated, blunt-ended nuclei (Figs. 16-27
and 16-28). The stroma can have variable amounts of
collagen, and the lesion may be focally or diffusely heavily
Radiographic Imaging
hyalinized. Large areas of necrosis can be present. Focally
Radiographic features are nonspecific but are consis- the tumor cells may be more rounded with prominent
tent with those of a malignant lesion. On plain radio- eosinophilic cytoplasm; that is, they may have a round-
graphs, leiomyosarcoma presents as a destructive lytic cell or epithelioid appearance. Leiomyosarcomas in bone
lesion with cortical disruption and extension into soft frequently exhibit an associated giant-cell reaction. Occa-
tissue.122,126,149,152,162 The margins of the lesion are ill sionally, aggregates of multinucleated, osteoclast-like
defined with frequent moth-eaten patterns of bone giant cells can be so prominent as to obscure the spindle
destruction (Figs. 16-20 to 16-24). Text continued on p. 1129

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1123

A B
FIGURE 16-20  ■  Leiomyosarcoma of bone: radiographic features. A and B, Anteroposterior and lateral radiographs of distal femoral
shaft of a 33-year-old man show lytic lesion with permeative destruction at margins and focal cortical erosion. Small amount of
periosteal new bone formation is seen along one border. Patient had experienced increasing pain in this area for 2 years.

ERRNVPHGLFRVRUJ
1124 16  Miscellaneous Mesenchymal Tumors

A B

D
FIGURE 16-21  ■  Leiomyosarcoma of bone: radiographic features. A, Lateral radiograph shows destructive lesion of proximal end of
fibula in a 32-year-old man. This well-differentiated leiomyosarcoma is sharply demarcated and expansile, without periosteal reac-
tion. B, Coronal magnetic resonance image (MRI) shows marrow replacement and cortical breakthrough laterally. C, Axial MRI shows
cortical thinning and disruption, with extension into soft tissue. D, Specimen radiograph demonstrates sharp circumscription of
proximal fibular leiomyosarcoma. Remnants of preexisting bone are seen in tumor, but reactive sclerosis is not apparent.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1125

B
FIGURE 16-22  ■  Leiomyosarcoma of bone: radiographic features. A and B, Anteroposterior and lateral radiographs of knee of a
51-year-old man show ill-defined lesion in proximal tibial metaphysis. Tumor has produced permeative pattern of bone destruction
with no discernible periosteal reaction.

ERRNVPHGLFRVRUJ
1126 16  Miscellaneous Mesenchymal Tumors

C D
FIGURE 16-23  ■  Leiomyosarcoma of bone: radiographic features. A and B, Anteroposterior and lateral radiographs of ankle region of
a 68-year-old woman show ill-defined destructive lesion of distal tibial shaft with large mass in posterior soft tissue. C, Computed
tomogram shows intraosseous tibial tumor and large mass in posterior soft tissue. D, Coronal cut through distal tibia and ankle
(amputation specimen) shows gray-white, firm intraosseous tumor filling marrow cavity and eroding tibial cortex.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1127

A B C
FIGURE 16-24  ■  Leiomyosarcoma of bone in familial retinoblastoma: radiographic features. A, Plain radiographic image showing
intramedullary diaphyseal lesion of the humerus with lytic destructive growth pattern and pathologic fracture in a 40-year-old woman
who had a son with bilateral retinoblastoma. B and C, T1- and T2-weighted magnetic resonance images of the lesion shown in
A showing a destructive intramedullary lesion with associated pathologic fracture. (For microscopic features, see Figure 16-26.)

ERRNVPHGLFRVRUJ
1128 16  Miscellaneous Mesenchymal Tumors

A C
FIGURE 16-25  ■  Leiomyosarcoma of bone: gross features. A, Sagittally sectioned tibial resection specimen with destructive gray to
white mass involving distal area. Note massive amount of cortical destruction, with circumferential extension into soft tissue.
B, Proximal tibial resection specimen shows destructive gray to white intramedullary mass. C, Coronal section of proximal tibial
resection shows intramedullary mass with central necrosis within fleshy site.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1129

cells and smooth muscle features of the tumor cells. Epi- that involve bone in children are metastatic or represent
thelioid change can be focally present but is rarely a secondary involvement by contiguous growth of a soft
dominant feature in primary leiomyosarcoma of bone.137 tissue lesion.169,170,171 The latter is typically seen in the
Therefore if such change is present, it suggests that the head and neck region (orbit). In patients older than age
lesion in question may be metastatic rather than primary 50 years, the presence of rhabdomyoblastic differentia-
(Fig. 16-26). Pleomorphic or dedifferentiated leiomyo- tion within a bone tumor is more likely to represent a
sarcoma is also extremely unusual in bone (Fig. 16-29), phenotypic switch (divergent differentiation) within a
and tumors with such features often are metastatic. high-grade sarcomatous component of dedifferentiated
chondrosarcoma rather than a pure primary rhabdomyo-
sarcoma of bone.164,166,167 However, there are extremely
Special Techniques
rare, well-documented cases of pure primary rhabdomyo-
Immunohistochemically, the tumor cells are strongly and sarcomas of bone.165,168,172,173 Radiographic features are
diffusely positive for SMA and MSA (Figs. 16-27, D and nonspecific and show bone destruction by a lytic process
16-29, C-D).121,133,143 Similar to other leiomyosarcomas, (Fig. 16-31). We have seen one case of a pure primary
especially those of the gastrointestinal tract, leiomyosar- rhabdomyosarcoma of bone.
coma of bone can exhibit focal positivity for keratin. The cases described in bone do not differ from rhab-
Electron microscopic examination can provide supportive domyosarcomas of soft tissue, and both embryonal and
evidence for the diagnosis of leiomyosarcoma (Fig. 16-30). adult (pleomorphic) types of rhabdomyosarcoma can
Ultrastructurally these tumors are composed of spindle occur in bone.
cells containing a centrally placed elongated nucleus with Typical microscopic features include round, ribbon-
blunt ends. Although the nuclei of smooth muscle cells shaped, and strap rhabdomyoblasts (Fig. 16-32). Large,
are oval under the light microscope, ultrastructurally they round, pleomorphic cells with dense eosinophilic cyto-
may show multiple infoldings of the nuclear envelope. plasm, which are typical of the pleomorphic variant
The cytoplasm contains fine (6- to 80-nm thick) filaments of rhabdomyosarcoma, also may be present. In typical
consistent with actin. Many electron-dense patches are cases, the tumor cells are positive for MyoD1, myo-
present within the filaments. These structures appear to genin, myoglobin, and desmin, but the intensity of
represent focal condensation of filaments. Smooth muscle staining is proportional to the degree of skeletal muscle
cells also contain numerous pinocytotic vesicles and are differentiation.
surrounded by basal lamina. In well-differentiated tumors, Cells with microscopically recognizable skeletal
the described features are present in every tumor cell and muscle differentiation are typically strongly positive for
are easy to identify. On the other hand, in more undif- these markers. The staining is usually minimal or even
ferentiated (high-grade) lesions, ultrastructural features absent in less-differentiated round- or spindle-cell areas.
of smooth muscle differentiation can be focal. Leiomyo- It is important to realize that MSA and desmin are also
sarcomas show complex chromosomal aberrations with positive in smooth muscle. Therefore the use of these
frequent loss of 3p21-23, 8p21-pter, 13q12-13, and markers does not help in the determination between skel-
13q32-qter and gain of the 1q21-31 region.148,156 etal and smooth muscle differentiation.
The presence of other component,s such as cartilage,
bone, and undifferentiated sarcomatous elements, in
Differential Diagnosis
bone raise the suspicion that the lesion in question may
In any case of leiomyosarcoma involving bone, a meta- represent a dedifferentiation phenomenon rather than a
static lesion must be ruled out before the lesion in ques- primary rhabdomyosarcoma of bone.
tion can be designated as a primary tumor. Microscopically
leiomyosarcoma must be differentiated from malignant
fibrous histiocytoma or fibrosarcoma and spindle-cell carcinoma. MULTIPOTENTIAL PRIMITIVE
These lesions have prominent herringbone or storiform
patterns that are typically not present in leiomyosarcoma. SARCOMA, POLYHISTIOMA, AND
Immunohistochemical staining for smooth muscle MALIGNANT MESENCHYMOMA
markers can be somewhat confusing because some of the
malignant fibrous histiocytoma cells can be positive for The term malignant mesenchymoma was proposed by
SMA and MSA. On the other hand, strong uniform posi- Stout184 to designate a group of malignant neoplasms that
tivity of tumor cells for these markers is rarely seen in had two or more distinct mesenchymal components
malignant fibrous histiocytomas or fibrosarcomas. exhibiting divergent differentiation. Subsequently the
Differentiation from spindle-cell sarcomatoid carci- term was applied to a variety of neoplasms that involved
noma can be accomplished in a majority of cases if clinical various organs. Rare cases of this questionable entity have
data are available. In addition, most sarcomatoid carcino- been reported in bone.174,178,181-184 In our opinion, most
mas, at least focally, show strong positivity for epithelial lesions diagnosed as malignant mesenchymomas in bone
markers. are better understood if they are classified as dedifferenti-
ated chondrosarcomas with divergent differentiation in
their high-grade sarcomatous components.179 Several
RHABDOMYOSARCOMA well documented cases of osteosarcoma combined with
rhabdomyosarcomatous differentiation have recently
Rhabdomyosarcoma is one of the rarest primary neo- been reported as primary bone lesions (Fig. 16-33).181,183,185
plasms of bone. The majority of rhabdomyosarcomas Text continued on p. 1138

ERRNVPHGLFRVRUJ
A

B
FIGURE 16-26  ■  Metastatic leiomyosarcoma of bone with epithelioid features. A and B, Low and intermediate power views of leio-
myosarcoma involving thoracic vertebrae. Note uniform epithelioid change of tumor cells and prominent hemangiopericytoma-like
pattern of branching vascular channels. This extent of epithelial change is rarely seen in primary leiomyosarcoma of bone and
suggests metastatic lesion. In this case, primary tumor was found in stomach. Inset (top) shows epithelioid features of tumor cells.
(A, ×100; B, ×200; Inset, ×400.) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1131

A B

C D
FIGURE 16-27  ■  Leiomyosarcoma of bone: microscopic features. A, Interlacing bundles of spindle cells with inconspicuous nuclear
atypia. B and C, Features of smooth muscle are evident under higher magnification. Note bundles of cells with elongated endonuclei
and clearly darker dense, eosinophilic cytoplasm. D, Strong positivity of tumor cells for smooth muscle actin (A, ×100; B-D, ×200).

ERRNVPHGLFRVRUJ
1132 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-28  ■  Leiomyosarcoma of bone: radiographic and microscopic features. A, Lytic lesion of distal tibia with cortical penetra-
tion in a 14-year-old girl. B, T1-weighted magnetic resonance image showing intramedullary lesion with low signal intensity. C, Low
power magnification of metastatic leiomyosarcoma in bone forming a subpleural nodule. D, Microscopic image of primary leiomyo-
sarcoma of the distal tibia showing interlacing bundles of spindle cells with prominent atypia. (C, ×4; D, ×100) (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1133

A B

C D
FIGURE 16-29  ■  Leiomyosarcoma of bone in familial retinoblastoma: microscopic features. A, Leiomyosarcoma composed of interlac-
ing spindle cells juxtaposed on areas with epithelioid appearance (×50). B, Highly pleomorphic dedifferentiated component of the
tumor shown in A (×200). C, Immunohistochemical stain for smooth muscle actin showing strong positive staining in spindle and
epithelioid areas and no staining in pleomorphic dedifferentiated component (×25). D, Immunohistochemical stain for desmin
showing strong positive staining of spindle cells and negative staining in pleomorphic areas (×50). Inset, Higher magnification of
pleomorphic component of the tumor with negative staining for desmin (×200). (A and B, hematoxylin-eosin; C, D, and Inset, DAB
with hematoxylin.) (Radiographic features of this case are shown in Figure 16-20.)

ERRNVPHGLFRVRUJ
1134 16  Miscellaneous Mesenchymal Tumors

B
FIGURE 16-30  ■  Leiomyosarcoma of bone: ultrastructural features. A, Low power magnification shows spindle-shaped tumor cells
with centrally placed oval nuclei. Note prominent bundles of actin filaments in peripheral areas of cytoplasm. B, Higher magnifica-
tion of A shows actin filaments with focal condensation forming patchy, electron-dense areas. Inset (top), Numerous pinocytotic
vesicles and presence of basal lamina are frequent features of cells exhibiting smooth muscle differentiation. Inset (bottom), Oval
nucleus of leiomyosarcoma with prominent nucleolus and irregular nuclear membrane forming deep infoldings. (A and bottom
Inset, ×3,500; B, ×12,000; top Inset, ×35,000)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1135

B
FIGURE 16-31  ■  Rhabdomyosarcoma in bone: radiographic and gross features. A, Computed tomogram of pelvis of a 31-year-old
man with intraosseous radiolucent mass that has caused cortical destruction and has extended into soft tissue. B, Gross photograph
of wing of left ilium sectioned to show sarcoma that apparently arose in medullary cavity and penetrated cortex at several points.
Extension into soft tissue was noted during microscopic examination. (Courtesy Dr. M. Tsuneyoshi, Fukuoka, Japan.)

ERRNVPHGLFRVRUJ
1136 16  Miscellaneous Mesenchymal Tumors

A B

C D
FIGURE 16-32  ■  Rhabdomyosarcoma of bone: microscopic features. A-D, Spindle-cell malignant neoplasm with multiple strap cells
and occasional multinucleated large cells. (A-D, ×200) (A-D, hematoxylin-eosin.) (Courtesy Dr. M. Tsuneyoshi, Fukuoka, Japan.)

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1137

A B

C D

E F G
FIGURE 16-33  ■  Polyphenotypic sarcoma: radiographic and microscopic features. A, Computed tomogram scan of a destructive
osteoblastic lesion at the angle of the left mandible, with evidence of soft tissue extension. B, Areas of the osteosarcoma with
osteoblastic histology. There are pleomorphic tumor cells with extensive bone matrix production and osteoclasts. C, Areas of the
osteosarcoma with chondroblastic differentiation. There is extensive cartilaginous matrix production intermingled with osteoblastic
elements. D, Higher magnification of B showing osteoblastic differentiation and prominent osteoid deposition. E, Foci of tumor with
rhabdomyoblastic differentiation. The rhabdomyoblastic tumor cells have characteristic cross striations (arrow). F and G, Immuno-
phenotypic features support rhabdomyoblastic differentiation with positive staining for desmin and myogenin. (B and C, ×50;
D, F, and G, ×100; E, ×600) (B-E, hematoxylin-eosin.) (Reprinted with permission from Seethala RR, et al: Arch Pathol Lab Med 130:385–
388, 2006.)

ERRNVPHGLFRVRUJ
1138 16  Miscellaneous Mesenchymal Tumors

Some of these polyphenotypic sarcomas develop as com- 20. Kubo T, Matsui Y, Goto T, et al: MRI characteristics of parosteal
plications of radiotherapy.183 lipomas associated with the HMGA2-LPP fusion gene. Anticancer
Res 26:2253–2257, 2006.
The terms primitive multipotential sarcoma and polyhis- 21. Kwak HS, Lee KB, Lee SY, et al: On the AJR viewbox. MR find-
tioma were proposed by Hutter et al in 1966176 and ings of calcaneal intraosseous lipoma with hemorrhage. AJR Am
Jacobson in 1977,177 respectively. These terms were J Roentgenol 185:1378–1379, 2005.
used to designate a group of extremely rare primary 22. Lagier R: Calcaneus lipoma with bone infarct. Rofo 142:472–474,
1985.
sarcomas of bone in which a population of primitive 23. Lagier R: Fibular lipoma with areas of bone infarct calcification.
undifferentiated cells, similar to those seen in Ewing’s Eur J Radiol 5:226–227, 1985.
sarcoma, was accompanied by elements that exhibited 24. Lee SJ, Yoon JH, Bae JI, et al: Intracortical lipoma of the femur,
microscopically recognizable polyphenotypic differentia- Skeletal Radiol online publication, 2006.
tion along several mesenchymal or even epithelial path- 25. Leeson MC, Kay D, Smith BS: Intraosseous lipoma. Clin Orthop
181:186–190, 1983.
ways (also see Chapter 11).175 26. Milgram JW: Intraosseous lipomas with reactive ossification in
Although a link between this neoplasm and a true the proximal femur: report of eight cases. Skeletal Radiol 7:1–13,
Ewing’s sarcoma was originally postulated and seems to 1981.
be logical, no consistent molecular data are available to 27. Mohan V, Gupta SK, Cherian J, et al: Intra-osseous lipoma of the
calcaneum. J Postgrad Med 27:127–128, 1981.
support this concept. On the other hand, some authors 28. Moorefield WGJ, Urbaniak JR, Gonzalvo AA: Intramedullary
say that review of a large series of small-cell tumors dis- lipoma of the distal femur. South Med J 69:1210–1211, 1976.
closes the presence of a new base of primitive sarcomas 29. Murphey MD, Carroll JF, Flemming DJ, et al: From the archives
with at least focal Ewing’s sarcoma–like features.180 Poly- of the AFIP: benign musculoskeletal lipomatous lesions. Radio-
phenotypic differentiation, usually epithelial, muscular, graphics 24:1433–1466, 2004.
30. Murphy NB: Ossifying lipoma. Br J Radiol 47:97–98, 1974.
and neural, has been documented in some of these lesions 31. Nahles G, Schaeper F, Bier J, et al: An intraosseous lipoma in the
as well as the expression of MIC2 gene product.180 frontal bone—a case report. Int J Oral Maxillofac Surg 33:408–410,
2004.
REFERENCES 32. Plaut GS, Salm R, Truscott DE: Three cases of ossifying lipoma.
J Pathol Bacteriol 78:292–295, 1959.
Lipoma 33. Poussa M, Holmstrom T: Intraosseous lipoma of the calcaneus:
1. Appenzeller J, Weitzner S: Intraosseous lipoma of os calcis: case report of a case and a short review of the literature. Acta Orthop
report and review of literature of intraosseous lipoma of extremi- Scand 47:570–574, 1976.
ties. Clin Orthop 101:171–175, 1974. 34. Radl R, Leithner A, Machacek F, et al: Intraosseous lipoma: ret-
2. Blair TR, Resnick D: Subperiosteal lipoma: a case report. Clin rospective analysis of 29 patients. Int Orthop 28:374–378, 2004.
Imaging 24:381–383, 2000. 35. Rau T, Soeder S, Olk A, et al: Parosteal lipoma of the thigh with
3. Campbell RS, Grainger AJ, Mangham DC, et al: Intraosseous cartilaginous and osseous differentiation: an osteochondrolipoma.
lipoma: report of 35 new cases and a review of the literature. Ann Diagn Pathol 10:279–282, 2006.
Skeletal Radiol 32:209–222, 2003. 36. Uguz B, Aydin AT: Intraosseous lipoma of the fifth metatarsal: a
4. Cebesoy O, Altinel L: Rare case of intraosseous lipoma with case report. J Foot Ankle Surg 43:119–122, 2004.
proximal femoral localization causing cortical expansion. J Natl 37. Zorn DT, Cordray DR, Randels PH: Intraosseous lipoma of
Med Assoc 98:1702–1704, 2006. bone involving the sacrum. J Bone Joint Surg 53A:1201–1204,
5. Delee JC: Intra-osseous lipoma of the proximal part of the femur: 1971.
case report. J Bone Joint Surg 61A:601–603, 1979. 38. Yamamoto T, Marui T, Akisue T, et al: Intracortical lipoma of the
6. Eroglu A, Gundogdu C, Turkyilmaz A, et al: Intraosseous lipoma femur. Am J Surg Pathol 26:804–808, 2002.
of the rib. J Thorac Cardiovasc Surg 130:1468–1469, 2005.
7. Fleming RJ, Alpert M, Garcia N: Parosteal lipoma. AJR Am J Liposarcoma
Roentgenol 87:1075, 1962. 39. Addison AK, Payne SR: Primary liposarcoma of bone: case report.
8. Gero MJ, Kahn LB: Case report 498: intraosseus lipoma of the J Bone Joint Surg 64A:301–304, 1982.
distal end of the fibula with focal infarction. Skeletal Radiol 17:443– 40. Catto M, Stevens J: Liposarcoma of bone. J Pathol Bacteriol
446, 1988. 86:248–253, 1963.
9. Goto T, Jojima T, Iijima T, et al: Intraosseous lipoma: a clinical 41. Dawson EK: Liposarcoma of bone. J Pathol Bacteriol 70:513–520,
study of 12 patients. J Orthop Sci 7:274–280, 2002. 1955.
10. Greenfield GB: Radiology of bone diseases, Philadelphia, 1969, 42. Kenan S, Klein M, Lewis MM: Juxtacortical liposarcoma: a case
JB Lippincott. report and review of the literature. Clin Orthop 243:225–229,
11. Gunterberg B, Kindblom LG: Intraosseous lipoma: a report of 1989.
two cases. Acta Orthop Scand 49:95–97, 1978. 43. Kenan S, Lewis MM, Abdelwahab IF, et al: Case report 652:
12. Hanelin LG, Schlamberg EL, Bardsley JL: Intraosseous lipoma primary intraosseous low grade myxoid sarcoma of the scapula
of the coccyx: report of a case. Radiology 114:343–344, 1975. (myxoid liposarcoma). Skeletal Radiol 20:73–75, 1991.
13. Hart JA: Intraosseous lipoma. J Bone Joint Surg Br 55:624, 1973. 44. Larsson SE, Lorentzon R, Boquist L: Primary liposarcoma of
14. Hermann G, Rosen Y: Radiologic notes: case no. 395—ossifying bone. Acta Orthop Scand 46:869–876, 1975.
lipoma. Mt Sinai J Med 44:292–295, 1977. 45. Pardo-Mindán FJ, Ayala H, Joly M, et al: Primary liposarcoma of
15. Jacobs P: Parosteal lipoma with hyperostosis. Clin Radiol 23:196– bone: light and electron microscopic study. Cancer 48:274–280,
198, 1972. 1981.
16. Jebson PJ, Schock EJ, Biermann JS: Intraosseous lipoma of the 46. Schneider HM, Wunderlich T, Puls P: The primary liposarcoma
proximal radius with extraosseous extension and a secondary pos- of the bone. Arch Orthop Trauma Surg 96:235–239, 1980.
terior interosseous nerve palsy. Am J Orthop 31:413–416, 2002. 47. Srivastava KP, Chandra SH, Sharma RD, et al: Primary liposar-
17. Jones JG, Habermann ET, Dorfman HD: Case report 553: coma of the skull. Int Surg 61:234, 1976.
parosteal ossifying lipoma of femur. Skeletal Radiol 18:537–540, 48. Torok G, Meller Y, Maor E: Primary liposarcoma of bone: case
1989. report and review of the literature. Bull Hosp Joint Dis Orthop Inst
18. Kenin A, Levine J, Spinner M: Parosteal lipoma: a report of two 43:28–37, 1983.
cases with associated bone changes. J Bone Joint Surg 41A:1122–
1126, 1959. Vascular-Cartilaginous Hamartoma of the Chest Wall
19. Ketyer S, Brownstein S, Cholankeril J: CT diagnosis of intraos- 49. Ayala AG, Ro JY, Bolio-Solis A, et al: Mesenchymal hamartoma
seous lipoma of the calcaneus. J Comput Assist Tomogr 7:546–547, of the chest wall in infants and children: a clinicopathological
1983. study of five patients. Skeletal Radiol 22:469–576, 1993.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1139

50. Blumenthal BI, Capitanio MA, Queloz JA, et al: Intrathoracic 74. Kaya S, Unal OF, Sarac S, et al: Melanotic neuroectodermal
mesenchymoma: observation in two infants. Radiology 104:107– tumor of infancy: report of two cases and review of literature. Int
109, 1972. J Pediatr Otorhinolaryngol 52:169–172, 2000.
51. Calleja MD, Castillo CB, Blanco-Rodriguez G: Vascular and car- 75. Khoddami M, Squire J, Zielenska M, et al: Melanotic neuroecto-
tilaginous hamartoma of the chest wall in an infant (in Spanish). dermal tumor of infancy: a molecular genetic study. Pediatr Dev
Bol Med Hosp Infant Mex 41:677–681, 1984. Pathol 1:295–299, 1998.
52. Davis RI, Macloon J, Sloan JM: Vascular and cartilaginous ham- 76. Mello RJ, Vidal AK, Fittipaldi HM, Jr, et al: Melanotic neuroec-
artoma: a case report. Histopathology 20:269–270, 1992. todermal tumor of infancy: clinicopathologic study of a case, with
53. Hall DP, Ellison RG: Osteochondrosarcoma of the chest wall in emphasis on the chemotherapeutic effects. Int J Surg Pathol
a newborn infant: a case report 6 years after surgery. Am Surg 8:247–251, 2000.
30:745–747, 1964. 77. Mirich DR, Blaser SI, Harwood-Nash DC, et al: Melanotic
54. Levy WM, Miller AS, Bonakdarpour A, et al: Aneurysmal bone neuroectodermal tumor of infancy: clinical, radiologic, and
cysts secondary to other lesions: report of 57 cases. Am J Clin pathologic findings in five cases. Am J Neuroradiol 12:689–697,
Pathol 63:1–8, 1975. 1991.
55. McCarthy EF, Dorfman HD: Vascular and cartilaginous hamar- 78. Navas Palacios JJ: Malignant melanotic neuroectodermal tumor:
toma of the ribs in infancy with secondary aneurysmal bone cyst light and electron microscopic study. Cancer 46:529–536, 1980.
formation. Am J Surg Pathol 4:247–253, 1980. 79. Nitta T, Endo T, Tsunoda A, et al: Melanotic neuroectodermal
56. McLeod RA, Dahlin DC: Hamartoma (mesenchymoma) of the tumor of infancy: a molecular approach to diagnosis—case report.
chest wall in infancy. Radiology 131:657–661, 1979. J Neurosurg 83:145–148, 1995.
57. Moghal N: Vascular and cartilaginous hamartoma (mesenchy- 80. Parizek J, Nemecek S, Cernoch Z, et al: Melanotic neuroectoder-
moma) of the ribs in infancy. J Pak Med Assoc 40:114–115, 1990. mal tumor of infancy of extra-, intra-, and subdural right temporal
58. Seibert JJ, Rossi NP, McCarthy EF: A primary rib tumor in a location: CT examination, surgical treatment, literature review.
newborn. J Pediatr Surg 11:1031–1032, 1976. Neuropediatrics 17:115–123, 1986.
59. Ulloa-Patino P, Baeza-Flores E, Montalvo-Marin A, et al: Vascu- 81. Pettinato G, Manivel JC, d’Amore ES, et al: Melanotic neuroec-
lar and cartilaginous hamartoma of the thoracic wall. Bol Med Hosp todermal tumor of infancy: a reexamination of a histogenetic
Infant Mex 45:467–471, 1988. problem based on immunohistochemical, flow cytometric, and
ultrastructural study of 10 cases. Am J Surg Pathol 15:233–245,
Melanotic Neuroectodermal Tumor of Infancy 1991.
60. Allen MS, Jr, Harrison W, Jahrsdoerfer RA: “Retinal anlage” 82. Shokry A, Briner J, Makek M: Malignant melanotic neuroecto-
tumors: melanotic progonoma, melanotic adamantinoma, pig- dermal tumor of infancy: a case report. Pediatr Pathol 5:217–223,
mented epulis, melanotic neuroectodermal tumor of infancy and 1986.
benign melanotic neuroectodermal tumor of infancy. Am J Clin 83. Slootweg PJ: Heterologous tissue elements in melanotic neu-
Pathol 51:309–314, 1969. roectodermal tumor of infancy. J Oral Pathol Med 21:90–92,
61. Borello ED, Gorlin RJ: Melanotic neuroectodermal tumor of 1992.
infancy: a neoplasm of neural crest origin: report of a case associ- 84. Stirling RW, Powell G, Fletcher CD: Pigmented neuroectoder-
ated with high urinary excretion of vanilmandelic acid. Cancer mal tumor of infancy: an immunohistochemical study. Histopathol-
19:196–206, 1966. ogy 12:425–435, 1988.
62. Carpenter BF, Jimenez C, Robb IA: Melanotic neuroectodermal 85. Toda T, Sadi AM, Kiyuna M, et al: Pigmented neuroectodermal
tumor of infancy. Pediatr Pathol 3:227–244, 1985. tumor of infancy in the epididymis. A case report. Acta Cytol 42:
63. Cheung LK, Piette EM, Tideman H: Melanotic neuroectodermal 775–780, 1998.
tumour of infancy: a case report emphasizing the importance 86. Zarbo RJ, Raju U, Regezi J, et al: Melanotic neuroectodermal
of computed tomography. Dentomaxillofac Radiol 21:172–174, tumors (MNT) of infancy: intermediate filament, neuroendocrine
1991. and melanoma associated antigen profile. Lab Invest 60:109A,
64. Cohen BH, Handler MS, deVivo DC, et al: Central nervous 1989.
system melanotic neuroectodermal tumor of infancy: value of
chemotherapy in management. Neurology 38:163–164, 1988. Perivascular Epithelioid Cell Tumor (PEComa)
65. Cutler LS, Chaudry AP, Topazian R: Melanotic neuroectodermal 87. Argani P, Aulmann S, Illei PB, et al: A distinctive subset of
tumor of infancy: an ultrastructural study, literature review, and PEComas harbors TFE3 gene fusions. Am J Surg Pathol 34:1395–
re-evaluation. Cancer 48:257–270, 1981. 1406, 2010.
66. Dehner LP, Sibley RK, Sauk JJ, Jr, et al: Malignant melanotic 88. Bonetti F, Pea M, Martignoni G, et al: PEC and sugar. Am J Surg
neuroectodermal tumor of infancy, a clinical, pathologic, ultra- Pathol 16:307–308, 1992.
structural and tissue culture study. Cancer 43:1389–1410, 1979. 89. Bonetti F, Pea M, Martignoni G, et al: Clear cell (“sugar”) tumor
67. Eckardt A, Swennen G, Teltzrow T: Melanotic neuroectodermal of the lung is a lesion strictly related to angiomyolipoma—the
tumor of infancy involving the mandible: 7-year follow-up after concept of a family of lesions characterized by the presence of the
heminmandibulectomy and costochondral graft reconstruction. perivascular epithelioid cells (PEC). Pathology 26(3):230–236,
J Craniofac Surg 12:349–354, 2001. 1994.
68. Franchi G, Sleilati F, Soupre V, et al: Melanotic neuroectodermal 90. Bonetti F, Pea M, Martignoni G, et al: The perivascular epitheli-
tumour of infancy involving the orbit and maxilla: surgical man- oid cell and related lesions. Adv Anat Pathol 4:343–358, 1997.
agement and follow-up strategy. Br J Plast Surg 55:526–529, 2002. 91. Bonetti F, Martignoni G, Colato C, et al: Abdominopelvic
69. Haider N, Mc Dermott M, Fitzgerald RJ: Melanotic neuroecto- sarcoma of perivascular epithelioid cells. Report of four cases in
dermal tumour of infancy arising in skull. Med Pediatr Oncol young women, one with tuberous sclerosis. Mod Pathol 14:563–
41:495–496, 2003. 568, 2001.
70. Hoshino S, Takahashi H, Shimura T, et al: Melanotic neuroecto- 92. Bosincu L, Rocca PC, Martignoni G, et al: Perivascular epitheli-
dermal tumor of infancy in the skull associated with high serum oid cell (PEC) tumors of the uterus: a clinicopathologic study of
levels of catecholamine: case report. J Neurosurg 80:919–924, two cases with aggressive features. Mod Pathol 18:1336–1342,
1994. 2005.
71. Johnson RE, Scheithauer BW, Dahlin DC: Melanotic neuroecto- 93. de Saint Aubain Somerhausen N, Gomez Galdon M, Bouffioux
dermal tumor of infancy: a review of seven cases. Cancer 52:661– B, et al: Clear cell ‘sugar’ tumor (PEComa) of the skin: a case
666, 1983. report. J Cutan Pathol 32:441–444, 2005.
72. Jones HH, Parker BR, Ballerio CG, et al: Case report 617. Mela- 94. Desy NM, Berstein M, Nahal A, et al: Primary perivascular
notic neuroectodermal tumor of infancy (melanotic progonoma) epithelioid cell neoplasm (PEComa) of bone: report of two
involving the calvaria. Skeletal Radiol 19:527–530, 1990. cases and review of the literature. Skeletal Radiol 41:1469–1474,
73. Kapadia SB, Frisman DM, Hitchcock CL, et al: Melanotic neu- 2012.
roectodermal tumor of infancy: clinicopathological, immunohis- 95. Dickson BC, Brooks JS, Pasha TL, et al: TFE3 expression in
tochemical, and flow cytometric study. Am J Surg Pathol 17: tumors of the microphthalmia-associated transcription factor
566–573, 1993. (Mi TF) family. Int J Surg Pathol 19:26–30, 2011.

ERRNVPHGLFRVRUJ
1140 16  Miscellaneous Mesenchymal Tumors

96. Eble JN: Angiomyolipoma of kidney. Semin Diagn Pathol 15:21–


40, 1998. Leiomyosarcoma
97. Fetsch P, Fetsch J, Marincol F, et al: Comparison of melanoma 119. Abdelwahab IF, Hermann G, Kenan S, et al: Case report 794:
antigen recognized by T cells (MART-1) to HMB-45: additional primary leiomyosarcoma of the right femur. Skeletal Radiol 22:
evidence to support a common lineage for angiomyolipoma, 379–381, 1993.
lymphangiomyomatosis, and clear cell sugar tumor. Mod Pathol 120. Angervall L, Berlin O, Kindblom LG, et al: Primary leiomyosar-
11(8):699–703, 1998. coma of bone: a study of five cases. Cancer 46:1270–1279, 1980.
98. Folpe AL, Goodman ZD, Ishak KG, et al: Clear cell myomelano- 121. Antonescu CR, Erlandson RA, Huvos AG: Primary leiomyosar-
cytic tumor of the falciform ligament/ligamentum teres: a novel coma of bone: a clinicopathologic, immunohistochemical, and
member of the perivascular epithelioid clear cell family of tumors ultrastructural study of 33 patients and a literature review. Am J
with a predilection for children and young adults. Am J Surg Surg Pathol 21:1281–1294, 1997.
Pathol 24:1239–1246, 2000. 122. Berlin Ö, Angervall L, Kindblom LG, et al: Primary leiomyosar-
99. Folpe AL, Mentzel T, Lehr HA, et al: Perivascular epithelioid cell coma of bone: a clinical, radiographic, pathologic-anatomic, and
neoplasms of soft tissue and gynecologic origin: a clinicopatho- prognostic study of 16 cases. Skeletal Radiol 16:364–376, 1987.
logic study of 26 cases and review of the literature. Am J Surg 123. Birla RK, Bowden L: Solitary bony metastasis as the first sign of
Pathol 29:1558–1575, 2005. malignant gastric tumor or of its recurrence. Ann Surg 182:45–49,
100. Gaffey M, Mills S, Askin F, et al: Clear cell tumor of the lung: a 1975.
clinicopathologic, immunohistochemical, and ultrastructural 124. Bluhm JM, Yi ES, Diaz G, et al: Multicentric endobronchial
study of eight cases. Am J Surg Pathol 14(3):248–259, 1990. smooth muscle tumors associated with the Epstein-Barr virus in
101. Goldblum JR, Folpe AL, Weiss SW: Soft tissue tumors, ed 6, Phila- an adult patient with the acquired immunodeficiency syndrome.
delphia, 2014, Elsevier Saunders. Cancer 80:1910–1913, 1997.
102. Insabato L, De Rosa G, Terracciano LM, et al: Primary mono- 125. Boman F, Gultekin H, Dickman PS: Latent Epstein-Barr virus
typic epithelioid angiomyolipoma of bone. Histopathology 40:286– infection demonstrated in low grade leiomyosarcomas of adults
290, 2002. with acquired immunodeficiency syndrome, but not in adjacent
103. Kazzaz D, Khalifa M, Alorjan M, et al: Malignant PEComa of the Kaposi’s lesion or smooth muscle tumors in immunocompetent
lumbar vertebra: a rare bone tumour. Skeletal Radiol 41:1465– patients. Arch Pathol Lab Med 121:834–838, 1997.
1568, 2012. 126. Bouaziz MC, Chaabane S, Mrad K, et al: Primary leiomyosar-
104. Kuroda N, Goda M, Kazakov DV, et al: Perivascular epithelioid coma of bone: report of four cases. J Comput Assist Tomogr 29:254–
cell tumor of the nasal cavity with TFE expression. Pathol Int 259, 2005.
59:769–770, 2009. 127. Chadwick EG, Connor EJ, Guerra Hanson C, et al: Tumors of
105. Lantuejoul S, Isaac S, Pinel N, et al: Clear cell tumor of the lung: smooth-muscle origin in HIV infected children. JAMA 263:3182–
an immunohistochemical and ultrastructural study supporting a 3184, 1990.
pericytic differentiation. Mod Pathol 10(10):1001–1008, 1997. 128. Eady JL, McKinney JD, McDonald EC: Primary leiomyosarcoma
106. Lehman NL: Malignant PEComa of the skull base. Am J Surg of bone: a case report and review of the literature. J Bone Joint
Pathol 28:1230–1232, 2004. Surg 69A:287–289, 1987.
107. Liebow AA, Castleman B: Benign clear cell tumor of the lung. Am 129. Fornasier VL, Paley D: Leiomyosarcoma in bone: primary or
J Pathol 43:13a–14a, 1963. secondary? A case report and review of the literature. Skeletal
108. Mentzel T, Reisshauer S, Rutten A, et al: Cutaneous clear cell Radiol 10:147–153, 1983.
myomelanocytic tumour: a new member of the growing family of 130. Goto T, Ishida T, Motoi N, et al: Primary leiomyosarcoma of the
perivascular epithelioid cell tumours (PEComas). Clinicopatho- femur. J Orthop Sci 7:267–273, 2002.
logical and immunohistochemical analysis of seven cases. Histopa- 131. Inoue S, Tanaka K, Sakamoto A, et al: Primary leiomyosarcoma
thology 46:498–504, 2005. of the patella. Skeletal Radiol 30:530–533, 2001.
109. Pan CC, Yang AH, Chiang H: Malignant perivascular epithelioid 132. Jundt G, Moll C, Nidecker A, et al: Primary leiomyosarcoma of
cell tumor involving the prostate. Arch Pathol Lab Med 127:96–98, bone: report of eight cases. Hum Pathol 25:1205–1212, 1994.
2003. 133. Kawai T, Suzuki M, Mukai M, et al: Primary leiomyosarcoma of
110. Pan CC, Yu IT, Yang AH, et al: Clear cell myomelanocytic bone: an immunohistochemical and ultrastructural study. Arch
tumor of the urinary bladder. Am J Surg Pathol 27:689–692, Pathol Lab Med 107:433–437, 1983.
2003. 134. Kingma DW, Shad A, Tsokos M, et al: Epstein-Barr virus (EBV)-
111. Pan CC, Jong YJ, Chai CY, et al: Comparative genomic hybrid- associated smooth muscle tumor arising in a post-transplant
ization study of perivascular epithelioid cell tumor: molecular patient treated successfully for two PT-EBV associated large-cell
genetic evidence of perivascular epithelioid cell tumor as a distinc- lymphomas: case report. Am J Surg Pathol 20:1511–1519, 1996.
tive neoplasm. Hum Pathol 37:606–612, 2006. 135. Lee E, Locker J, Nalesnik M, et al: The association of Epstein-
112. Panizo-Santos A, Sola I, de Alava E, et al: Angiomyolipoma and Barr virus with smooth muscle tumors occurring after organ
PEComa are immunoreactive for MyoD1 in cell cytoplasmic transplantation. N Engl J Med 332:19–25, 1995.
staining pattern. Appl Immunohistochem Mol Morphol 11:156–160, 136. Lo TH, van Rooij WJ, Teepen JL, et al: Primary leiomyosarcoma
2003. of the spine. Neuroradiology 37:465–467, 1995.
113. Tanaka M, Kato K, Gomi K, et al: Perivascular epithelioid cell 137. Lopez-Barea F, Rodriguez-Peralto JL, Sanchez-Herrera S, et al:
tumor with SFPQ/PSF-TFE3 gene fusion in a patient with Primary epithelioid leiomyosarcoma of bone. Case report and
advanced neuroblastoma. Am J Surg Pathol 33:1416–1420, 2009. literature review. Virchows Arch 43:367–371, 1999.
114. Tazelaar HD, Batts KP, Srigley JR: Primary extrapulmonary sugar 138. McClain KL, Leach CT, Jenson HB, et al: Association of Epstein-
tumor (PEST): a report of four cases. Mod Pathol 14(6):615–622, Barr virus with leiomyosarcomas in young people with AIDS.
2001. N Engl J Med 332:12–18, 1995.
115. Vang R, Kempson RL: Perivascular epithelioid cell tumor 139. McLoughlin LC, Nord KS, Joshi VV, et al: Disseminated leio-
(‘PEComa’) of the uterus: a subset of HMB-45–positive epitheli- myosarcoma in a child with acquired immune deficiency syn-
oid mesenchymal neoplasms with an uncertain relationshiop to drome. Cancer 67:2618–2621, 1991.
pure smooth muscle tumors. Am J Surg Pathol 26:1–13, 2002. 140. Martin-Hirsch DP, Habashi S, Benbow EW, et al: Post-irradiation
116. Yamamoto H, Oda Y, Yao T, et al: Malignant perivascular epithe- leiomyosarcoma of the maxilla. J Laryngol Otol 105:1068–1071,
lioid cell tumor of the colon: report of a case with molecular 1991.
analysis. Pathol Int 56:46–50, 2006. 141. Marymont JV, Clanton TO: Leiomyosarcoma of the os calcis. Foot
117. Yamashita K, Fletcher CD: PEComa presenting in bone: clinico- Ankle 10:239–242, 1990.
pathologic analysis of 6 cases and literature review. Am J Surg 142. Meltzer CC, Fishman EK, Scott WW, Jr: Computed tomography
Pathol 34:1622–1629, 2010. appearance of bone metastases of leiomyosarcoma. Skeletal Radiol
118. Zamboni G, Pea M, Martignoni G, et al: Clear cell “sugar” tumor 21:445–447, 1992.
of the páncreas. A novel member of the family of lesions charac- 143. Myers JL, Arocho J, Bernreuter W, et al: Leiomyosarcoma of
terized by the presence of perivascular epithelioid cells. Am J Surg bone: a clinicopathologic, immunohistochemical, and ultrastruc-
Pathol 20(6):722–730, 1996. tural study of five cases. Cancer 67:1051–1056, 1991.

ERRNVPHGLFRVRUJ
16  Miscellaneous Mesenchymal Tumors 1141

144. Narvaez JA, De Lama E, Portabella F, et al: Subperiosteal leio- 167. Niezabitowski A, Edel G, Roessner A, et al: Rhabdomyosarcoma-
myosarcoma of the tibia. Skeletal Radiol 34:42–46, 2005. tous component in dedifferentiated chondrosarcoma. Pathol Res
145. Overgaard J, Frederiksen P, Helmig O, et al: Primary leiomyosar- Pract 182:275–282, 1987.
coma of bone. Cancer 39:1664–1671, 1977. 168. Oda Y, Tsuneyoshi M, Hashimoto H, et al: Primary rhabdomyo-
146. Pritzker KPH, Huang SN, Marshall KG: Malignant tumors fol- sarcoma of the iliac bone in an adult: a case mimicking fibrosar-
lowing immunosuppressive therapy. Can Med Assoc J 103:1362– coma. Virchows Arch [A] 423:65–69, 1993.
1365, 1970. 169. Ruymann FB, Newton WA, Jr, Ragab AH, et al: Bone marrow
147. Ross JS, Del Rosario A, Bui HX, et al: Primary hepatic leiomyo- metastases at diagnosis in children and adolescents with rhabdo-
sarcoma in a child with the acquired immunodeficiency syndrome. myosarcoma: a report from the intergroup rhabdomyosarcoma
Hum Pathol 23:69–72, 1993. study. Cancer 53:368–373, 1984.
148. Sandberg AA: Updates on the cytogenetics and molecular genetics 170. Shapeero LG, Couanet D, Vanel D, et al: Bone metastases as the
of bone and soft tissue tumors: leiomyosarcoma. Cancer Genet presenting manifestation of rhabdomyosarcoma in childhood.
Cytogenet 161:1–19, 2005. Skeletal Radiol 22:433–438, 1993.
149. Sanerkin NG: Primary leiomyosarcoma of the bone and its com- 171. Simmons M, Tucker AK: The radiology of bone changes in rhab-
parison with fibrosarcoma. Cancer 44:1375–1387, 1979. domyosarcoma. Clin Radiol 29:47–52, 1978.
150. Shamsuddin AK, Reyes F, Harvey JW, et al: Primary leiomyosar- 172. Thomas F, Lipton JF, Barbera C, et al: Primary rhabdomyosar-
coma of bone. Hum Pathol 11:581–583, 1980. coma of the humerus: a case report and review of the literature.
151. Shen SC, Yunis EJ: Leiomyosarcoma developing in a child during J Bone Joint Surg 84A:813–817, 2002.
remission of leukemia. J Pediatr 89:780–782, 1976. 173. Wang JW, Eng HL, Huang CH: Primary rhabdomyosarcoma
152. Sundaram M, Akduman I, White LM, et al: Primary leiomyosar- of long bone. A case report. Clin Orthop Relat Res 377:205–209,
coma of bone. Am J Roentgenol 172:771–776, 1999. 2000.
153. Takami KM, Ishida T, Ieguchi M, et al: Primary leiomyosarcoma
or leiomyoma of the pubic bone? A case report. Int Orthop 18:248– Multipotential Primitive Sarcoma, Polyhistioma,
251, 1994. and Malignant Mesenchymoma
154. Takemori M, Nishimura R, Sugimura K, et al: Thoracic vertebral 174. Chow LT, Kumta SM: Primary osteochondrorhabdomyosarcoma
bone metastasis from uterine leiomyosarcoma. Gynecol Oncol 51: (malignant mesenchymoma) of the fibula: a rare tumour in an
244–247, 1993. unusual site—case report and review of the literature. APMIS
155. Templeton K: Leiomyosarcoma of bone. Am J Orthop 34:249–251, 112:617–623, 2004.
2005. 175. Frydman CP, Klein MJ, Abdelwahab IF, et al: Primitive multipo-
156. Verelst SJ, Hans J, Hanselmann RG, et al: Genetic instability in tential primary sarcoma of bone: a case report and immunohisto-
primary leiomyosarcoma of bone. Hum Pathol 35:1404–1412, chemical study. Mod Pathol 4:768–772, 1991.
2004. 176. Hutter RV, Foote JW, Jr, Francis KC, et al: Primitive multipoten-
157. von Hochstetter AR, Eberle A, Rüttner JR: Primary leiomyosar- tial primary sarcoma of bone. Cancer 19:1–25, 1966.
coma of extragnathic bones: case report and review of literature. 177. Jacobson SA: Polyhistioma: a malignant tumor of bone and
Cancer 53:2194–2200, 1984. extraskeletal tissues. Cancer 40:2116–2130, 1977.
158. Walker D, Gill TJ, III, Corson JM: Leiomyosarcoma in a renal 178. Kessler S, Mirra JM, Ishii T, et al: Primary malignant mesenchy-
allograft recipient treated with immunosuppressive drugs. JAMA moma of bone: case report, literature review, and distinction of
215:2084–2086, 1971. this entity from mesenchymal and dedifferentiated chondrosar-
159. Wang TY, Erlandson RA, Marcove RC, et al: Primary leiomyo- coma. Skeletal Radiol 24:291–295, 1995.
sarcoma of bone. Arch Pathol Lab Med 104:100–104, 1980. 179. Ling LL, Steiner GC: Primary multipotential malignant neo-
160. Wirbel RJ, Verelst S, Hanselmann R, et al: Primary leiomyo- plasm of bone: chondrosarcoma associated with squamous cell
sarcoma of bone: clinicopathologic, immunohistochemical, and carcinoma. Hum Pathol 17:317–320, 1986.
molecular biologic aspects. Ann Surg Oncol 5:635–641, 1998. 180. Llombart-Bosch A, Contesso G, Peydro-Olaya A: Histology,
161. Young MP, Freemont AJ: Primary leiomyosarcoma of bone. His- immunohistochemistry, and electron microscopy of small round
topathology 19:257–262, 1991. cell tumors of bone. Semin Diagn Pathol 13:153–170, 1996.
162. Young CL, Wold LE, McLeod RA, et al: Primary leiomyosar- 181. Mrad K, Sassi S, Smida M, et al: Osteosarcoma with rhabdomyo-
coma of bone. Orthopedics 11:615–618, 1988. sarcomatous component or so-called malignant mesenchymoma
163. Zhao X, Sun NC, Witt MD, et al: Changing pattern of AIDS: a of bone. Pathologica 96:475–478, 2004.
bone marrow study. Am J Clin Pathol 121:393–401, 2004. 182. Scheele PM, Jr, Von Kuster LC, Krivchenia G, II: Primary malig-
nant mesenchymoma of bone. Arch Pathol Lab Med 114:614–617,
Rhabdomyosarcoma 1990.
164. Astorino RN, Tesluk H: Dedifferentiated chondrosarcoma with 183. Seethala RR, Sturgis EM, Raymond AK, et al: Postirradiation
a rhabdomyosarcomatous component. Hum Pathol 16:318–320, osteosarcoma of the mandible with heterologous differentiation.
1985. Arch Pathol Lab Med 130:385–388, 2006.
165. Hsueh S, Hsih SN, Kuo TT: Primary rhabdomyosarcoma of long 184. Stout AP: Mesenchymoma, the mixed tumor of mesenchymal
bone: a case report. Orthopedics 9:705–707, 1986. derivatives. Ann Surg 127:278, 1948.
166. Lamovec J, Zidar A, Bracko M, et al: Primary bone sarcoma with 185. Van Dorpe J, Sciot R, Samson I, et al: Primary osteorhabdomyo-
rhabdomyosarcomatous component. Pathol Res Pract 190:51–60, sarcoma (malignant mesenchymoma) of bone: a case report and
1994. review of the literature. Mod Pathol 10:1047–1053, 1997.

ERRNVPHGLFRVRUJ
C H A P T E R 1 7 

Adamantinoma of Long Bone


CHAPTER OUTLINE

CLASSIC ADAMANTINOMA OSTEOFIBROUS DYSPLASIA


DIFFERENTIATED (OSTEOFIBROUS
DYSPLASIA–LIKE) ADAMANTINOMA

An epithelial neoplasm originating in the long bones of studying the relationship between adamantinoma and
the skeleton, referred to as adamantinoma, which almost fibroosseous lesions of the long bones. Further studies
exclusively affects the tibia and fibula, and its relationship of such composite lesions showed that the histologic
to a coexisting peculiar fibroosseus lesion, has been and radiologic features of the fibroosseous components
intriguing both pathologists and clinicians for more than were more consistent with those of osteofibrous dyspla-
a century. In 1913, Fischer17 described a peculiar tumor sia.1,32,47,51 These studies also showed that although osteo-
occurring predominantly in the tibia and occasionally in fibrous dysplasia–like changes are frequently present
the fibula that was somewhat similar to a more common at the periphery of adamantinomas, in some instances
odontogenic adamantinoma of the jaw bones. Despite they may dominate the entire lesion.62,64-66 Most current
histologic similarity, there has been no proof that these studies have shown that long bone adamantinoma, despite
tumors—of the jaw bone and of the long bone—have a the considerable range of its histologic patterns, appears
similar histogenetic origin. Adamantinoma of long bones to be of an epithelial nature.11,52-58
is extremely rare; there are approximately 300 well- The correlation of clinical, radiologic, and his-
documented cases published in the world literature.48-50 tologic data of our 25 consultation cases showed that
The largest individual series of long bone adamantinoma long bone adamantinomas could be divided into two
was reported by a group from the Mayo Clinic.37,51,67,68 main groups (Fig. 17-1).11 The first, defined as classic
Long bone adamantinomas present with a wide range of adamantinoma, is characterized by an abundance of
morphologic patterns that can mimic various primary or tumor cells and destructive growth pattern radiographi-
even metastatic bone lesions. The morphologic diversity cally. The second group is defined as differentiated
of long bone adamantinomas also accounts for the con- adamantinoma and is characterized histologically by a
tinuing controversy regarding their histogenesis. Another predominance of an osteofibrous dysplasia–like pattern
interesting phenomenon is the association of these tumors with a small, inconspicuous component of epithelial
with areas resembling osteofibrous dysplasia (ossifying tumor elements. On radiographs, differentiated adaman-
fibroma of long bones). tinomas are intracortical and often multicentric lesions.
The first description of areas of rarefaction associ- The histogenesis of these peculiar neoplasms is still
ated with long bone adamantinoma interpreted to be unclear, and various concepts of their origin as derived
osteitis fibrosa was provided in 1942 by Dockerty and from eccrine gland, synovial, vascular, or pluripotential
Myerding.15 In a subsequent article by Baker et al.,2 these mesenchyma have been postulated.9,12,16,28,29,38,43 The
lesions were interpreted to be fibrous dysplasia. The first current literature continues to subdivide adamantinomas
comprehensive description of long bone adamantinoma of long bones into the classic and differentiated osteofi-
associated with areas resembling fibrous dysplasia was brous dysplasia-like forms.13,19,26,41 A summary of clinical,
provided in 1962 by Cohen et al.10 These authors stated radiographic, and histologic features of the two subsets
that the radiologic pattern of fibrous dysplasia associated of long bone adamantinoma is provided in Table 17-1. In
with adamantinoma was different from that of classic the description in this chapter, classic and differentiated
fibrous dysplasia. Moreover, café au lait spots, which adamantinoma are discussed separately.
are frequent in polyostotic fibrous dysplasia, were not
seen in association with adamantinomas. Therefore these
authors were the first to postulate that, despite the histo-
logic similarity to fibrous dysplasia, fibroosseous lesions CLASSIC ADAMANTINOMA
associated with adamantinoma may represent a distinc- Definition
tive feature related to these tumors. The description of
ossifying fibroma of long bone by Kempson38 in 1966 and Classic adamantinoma is a distinct, rare biphasic neo-
the subsequent redesignation of this lesion as osteofibrous plasm of long bones that predominantly involves the tibia
dysplasia by Campanacci et al.7,8 provided a new basis for and occasionally the fibula and is characterized by a
1142
ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1143

Differentiated
(osteofibrous dysplasia–like)
adamantinoma Classic adamantinoma

Beyond cortex

Intracortical

Age
of 0 10 20 30 40 50 60 70 80
patient
Predominance of Predominance of
Metastases
osteofibrous dysplasia tumor cells
FIGURE 17-1  ■  Separation of long bone adamantinomas into two main groups (differentiated and classic). Separation is based on
combination of clinical, radiologic, and microscopic data.

TABLE 17-1 Summary of Features Characteristic of Classic and Differentiated Types of Adamantinoma
of Long Bone
Feature Classic Differentiated
Age Usually after first 20 years First 20 years
Location Tibia or fibula Tibia or fibula
Radiograph Expand beyond periosteum or intracortical Intracortical
Histology Predominance of tumor cells forming basaloid, Predominance of osteofibrous dysplasia
spindled, tubular, or squamous pattern pattern
Immunohistochemistry Predominance of cytokeratin-positive cells Scattered elements positive for cytokeratin

variety of histologic patterns. It has an overall histologic Radiographic Imaging


similarity to the more common odontogenic adamanti-
noma of the jaw bones. Features of classic adamantinoma on radiographs are dis-
tinct and often diagnostic.8,11,48,51 According to the radio-
graphic appearance, the lesion can be divided into two
Incidence and Location
major groups: intracortical and with radiologic evidence
Classic adamantinoma is extremely rare; while its true of complete cortical disruption and involvement of the
incidence is unknown, it clearly accounts for less than 1% medullary cavity, adjacent soft tissue, or both. Approxi-
of all bone neoplasms.48,49 This tumor typically affects mately 70% of lesions are located in the midshaft of the
patients older than age 20 years (Fig. 17-2), but occasion- tibia. The remaining 30% involve the proximal or distal
ally it may occur during the first two decades of life. The tibial regions. In the tibia, involvement of the anterolat-
youngest patient with classic adamantinoma in our series eral cortex and the presence of a moderate to severe
was a 13-year-old boy who subsequently developed lung bowing deformity are typical. The common appearance
metastases.11 More recently, a case of classic adamanti- is of mixed lytic and sclerotic lesions or multiple lucencies
noma has been observed in the tibia of a 3-year-old boy.40 with sclerosis of the intervening bone (Figs. 17-3 and
The involved bones are usually the tibia, fibula, or both. 17-4). The lucent lesions have well-defined margins. Fea-
Involvement of other skeletal sites is rare but has been tures of a destructive growth pattern with cortical disrup-
reported.49 According to our experience, the involvement tion are often seen. On radiographs, some lesions appear
of other skeletal sites should make the clinician question as destructive lytic defects involving two adjacent bones,
the diagnosis of adamantinoma and rule out other meta- the tibia and fibula. This is typically seen when the lesions
static neoplasms. Synchronous involvement of the tibia are located at the proximal or distal end of the tibia (Fig.
and fibula by two independent foci or by contiguous foci 17-3). The synchronous involvement by separate foci in
is typical of adamantinoma. The tibia and fibula are syn- the tibia and fibula is less frequent in this form of ada-
chronously involved in approximately 50% of cases. mantinoma (i.e., classic versus differentiated). The eccen-
tricity of the lesions is less evident in the fibula, in which
Clinical Symptoms the lesions are more centrally located and represent
sharply demarcated lytic foci with sclerotic margins.
Pain, bowing deformity of the tibia, or both of these Expansion of the bone contour without bowing defor-
symptoms are the most frequent clinical findings, and mity is typical of fibular lesions. A recent study of the role
these may be present for decades before diagnosis. A of magnetic resonance imaging (MRI) characteristics
palpable mass may be present. found that the method did not add to the specificity of

ERRNVPHGLFRVRUJ
1144 17  Adamantinoma of Long Bone

(osteofibrous dysplasia–like)
adamantinoma
Differentiated
Incidence

Classic adamantinoma

1 2 3 4 5 6 7 8
Age in decades

FIGURE 17-2  ■  Adamantinoma of long bones. Age distribution of classic and differentiated adamantinoma and most frequent sites
of skeletal involvement are indicated by a solid black arrow.

the distinction between histologic subtypes, but was of various sizes within the masses of cells usually separate
helpful in evaluating soft tissue and intramedullary the outer layer of cells from the inner masses. The solid
extension.69 areas are composed of cells forming sheets with a roughly
parallel arrangement of their oval nuclei.
The spindle pattern is characterized by the presence
Gross Findings
of spindle-shaped cells with elongated nuclei (Fig. 17-9).
Classic adamantinoma evaluated in a resection specimen This pattern also lacks the peripheral palisading orienta-
presents as a fleshy, soft mass. The lesion can be multifo- tion of the cells that is characteristic of the basaloid
cal or can involve a large segment of the medullary cavity pattern. The areas of pure spindle-cell pattern resemble
(Figs. 17-5 and 17-6). Gradual transition from soft, fleshy mesenchymal spindle-cell tumors such as fibrosarcoma,
areas to gritty fibrous tissue can be seen. Complete corti- even exhibiting a herring-bone pattern typical of that
cal disruption and contiguous involvement of adjacent tumor.
soft tissue or paired bone may be present. The tubular pattern consists of small, flattened or
cuboid cells that line spaces of varying sizes (Fig. 17-10).
In some areas the tubular pattern is produced by small
Microscopic Findings
glandular spaces separating spindle cells. Sometimes,
The histologic patterns of adamantinoma are extremely tubular structures lined by one or several layers of cuboi-
variable within each case and among cases. However, dal cells or flattened cells are embedded in fibrous stroma.
five basic patterns can be recognized: basaloid, spindle, Tubular structures give the impression of vascular chan-
tubular, squamous, and osteofibrous dysplasia–like.70 nels of various sizes or resemble glandular structures.
The basaloid pattern consists of nests or large masses Small tubular spaces occasionally resemble primitive
of epithelioid tumor cells that have a peripheral layer of capillary channels. Careful examination, however, some-
cells oriented roughly at right angles to the inner masses times reveals the presence within these structures of
of cells (Figs. 17-7 and 17-8). Peripheral cuboid or cells with features of epithelial differentiation or even
columnar cells with elongated nuclei occasionally produce keratinization.
sharply demarcated palisading structures. Cystlike spaces Text continued on p. 1153

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1145

C
FIGURE 17-3  ■  Classic adamantinoma: radiographic features. A, Multifocal lytic lesions involving distal tibial shaft. Separate lesion
of distal fibula is present. B, Adamantinoma contiguously involves fibula and tibia. C, Lytic lesion of distal tibia involves fibula
contiguously.

ERRNVPHGLFRVRUJ
1146 17  Adamantinoma of Long Bone

A C
FIGURE 17-4  ■  Plain radiograph and corresponding computed tomograms of adamantinoma of tibia. A, Multifocal, predominantly
osteosclerotic lesions of tibial shaft. Note focal, lytic, intramedullary lesion. B, Axial computed tomogram (CT) shows dense sclerotic
lesion of tibial shaft. C, Axial CT from lytic focus shows expanded intramedullary lesion.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1147

B
FIGURE 17-5  ■  Classic adamantinoma: gross features. A, Coronal section of amputation specimen shows destructive fleshy lesion
that expanded contour of distal fibula and eroded tibia contiguously. Note separate intramedullary lesion in tibial shaft. Inset, Whole-
mount specimen showing destructive lesion in the tibial shaft. B, Coronal section of amputation specimen shows destructive lesion
of distal tibia that involves fibula contiguously. Same specimen as shown in A. Inset, Intramedullary extension of the lesion in fibula.

ERRNVPHGLFRVRUJ
1148 17  Adamantinoma of Long Bone

A B C

D E
FIGURE 17-6  ■  Classic adamantinoma: gross features. A, Bisected tibial resection specimen shows a destructive fleshy intramedullary
lesion that expanded contour of bone. B, Sagittal section of tibia showing multifocal intramedullary and intracortical lesions.
C, Expanded view of B showing multifocal cortical and medullary lesions consisting of fleshy tissue. Note sclerosis of the overlying
cortex and irregular corticomedullary border. D, Coronal section of tibial amputation specimen shows destructive fleshy lesions that
erodes overlying cortex. E, Whole-mount specimen showing an intramedullary extension of adamantinoma.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1149

A B

C D
FIGURE 17-7  ■  Classic adamantinoma: microscopic features. A, Basaloid pattern with peripheral layers of cuboidal cells oriented at
right angles to inner spindled cell mass. B, Solid proliferations of tumor cells outlined focally by basaloid patterns. C, Spindled
pattern of loosely arranged tumor cells. D, Nests of tumor cells in fibrous stroma (A-D, ×100) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1150 17  Adamantinoma of Long Bone

A B

C D
FIGURE 17-8  ■  Adamantinoma: microscopic features. A, Basaloid pattern with peripheral layers of cuboidal cells. B, Higher magnifica-
tion of A showing basaloid arrangement of cuboidal cells outlining the nests of tumor cells. C, Irregular nests of tumor cells in
fibrous stroma. D, Loose cluster of epithelial cells in fibrous stroma. (A, ×100; B-D, ×200) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1151

A B

C D
FIGURE 17-9  ■  Classic adamantinoma: microscopic features. A, Solid proliferation of spindled tumor cells. B, Higher magnification
of A showing spindled tumor cells. C, Solid proliferation of spindled tumor cells. D, Higher magnification of C showing spindled
tumor cells. (A and C, ×100; B and D, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1152 17  Adamantinoma of Long Bone

B C
FIGURE 17-10  ■  Classic adamantinoma: microscopic variations of tubular pattern. A, Small open spaces (tubular) lined by flattened
or cuboidal cells. B, Irregular nests of tumor cells with small cystic spaces. C, Myriad small tubular spaces lined by flattened cells.
(A and C, ×200; B, ×100) (A-C, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1153

The squamous pattern is characterized by areas that however, it is clearly positive in endothelial cells lining
show evidence of squamous differentiation, including the blood vessels. All tumor cells and various components
eosinophilic cytoplasm and the presence of round, oval, of the osteofibrous dysplasia–like pattern are uniformly
or polygonal cytoplasm with occasional pyknosis of the negative for S-100 protein. Staining for actin is negative
nucleus (Fig. 17-11). Cells with these features form areas in all types of tumor cells. In contrast, fibroblast-like
of various sizes embedded within solid masses of non- stromal cells are focally weakly positive for this marker.
squamous cells. Features of squamous differentiation also Classic adamantinomas express E-, P-, and N-cadherins,
are commonly seen in the peripheral parts of the solid which are typically not found in osteofibrous dysplasia–
tumor nests, with individual squamous cells embedded in like adamantinomas.44
surrounding stromal tissue. Cytogenetic studies revealed numerical chromosomal
The osteofibrous dysplasia–like pattern is seen at least aberrations with extra copies of chromosomes 7, 8, 12,
focally in all cases of adamantinoma with adequate sam- 19, and 21 that could be detected in both classic and
pling of the tumor and its periphery. It represents areas osteofibrous dysplasia–like adamantinomas.4,35 The simi-
of loose fascicles of elongated fibroblast-like cells that larities of those aberrations in two subtypes of adaman-
often have a storiform pattern. Trabeculae of bone sur- tinomas further support the relationship between classic
rounded by rims of osteoblasts are present within this and osteofibrous dysplasia–like adamantinomas.4 Struc-
tissue. The zonal architecture of the fibrous dysplasia– tural chromosomal aberrations with translocations and
like changes can be seen in adequately sampled lesions, marker chromosomes resulting in complex karyotypes
which show progressive maturation and increasing are typically seen in classic adamantinoma and are not
numbers of bone trabeculae at the periphery of the lesion. observed in osteofibrous dysplasia–like adamantino-
When examined with the aid of polarized light, the mas.6,27,46,50 Taken together these observations support
increasing maturation from woven bone trabeculae in the progressive nature of chromosomal aberrations when
the central parts to predominantly lamellar bone at the disease evolves from osteofibrous dysplasia–like adaman-
periphery of the lesion is seen. The main feature differ- tinoma to classic adamantinoma. Occasional transloca-
entiating this pattern from typical osteofibrous dysplasia tions involving the 13q14 region of the tumor and
is the presence of small nests of epithelial cells in germline in the same patient have also been reported.60
the fibroblastic stroma that show occasional squamous In general, on total DNA measurements, the epithelial
differentiation. In addition to the solid epithelial nests, component of adamantinoma is highly aneuploid and
small tubular structures lined by cuboidal or flattened osteofibrous dysplasia is euploid.22
cells scattered in the fibrous stroma are commonly
found. Differential Diagnosis
The classic adamantinomas are characterized micro-
scopically by the abundance of tumor cells and a frequent Adamantinoma of long bones can be confused with a
mixture of several histologic patterns. The osteofibrous variety of primary and metastatic epithelial and soft tissue
dysplasia–like pattern is seen focally only and never dom- neoplasms.
inates the histologic picture. In limited biopsy specimens, epithelial elements, espe-
cially basaloid/tubular or squamous patterns, can be mis-
diagnosed as metastatic carcinoma. Identification of a
Special Techniques
peculiar mixture of various patterns present in the major-
Ultrastructurally, adamantinoma is epithelial in nature ity of adamantinomas, as well as the location of the lesion
with prominent desmosomes, tubular structures, and fea- in the tibia, the fibula, or both areas, should help avoid
tures of keratinization seen focally.16,39,52,57,58 Tumor cells this error.
of basaloid, spindled, tubular, and squamous patterns are Dominant spindle-cell or small tubular patterns can
consistently strongly positive for keratin (Figs. 17-12 and be misdiagnosed as fibrosarcoma or a vascular neoplasm.
17-13).3,11,30,58 The fibrous stroma and bone trabeculae of Identification of the epithelial nature of cells by appropri-
the osteofibrous dysplasia–like pattern are negative for ate immunohistochemical stains and radiographic data is
keratin; however, small nests of epithelial cells and tubular helpful in identifying the lesion as adamantinoma.
structures scattered in the stromal tissue are strongly More difficult problems exist in biopsy specimens con-
positive for keratin. More detailed studies reveal that taining predominantly fibroosseous areas that can be
adamantinomas of long bone express classes of keratins confused with fibrous dysplasia or ossifying fibroma (osteo-
similar to those found in the basal layer of the epider- fibrous dysplasia). The distinction from fibrous dysplasia
mis.3,33,34,44,45 Typically the epithelial component shows can be easily made if radiographic features of the lesion
expression of keratins 5, 14, 9, 1, 13, and 17.23 Keratins are considered. Fibrous dysplasia typically presents as an
8 and 18 are not expressed. Interestingly, epithelial com- intramedullary lesion. Scrupulous search and immuno-
ponents additionally coexpress epithelial membrane histochemical stains can disclose inconspicuous epithelial
antigen, p63, vimentin, and podoplanin indicative of a elements in a dominant osteofibrous dysplasia–like
mixed epithelial/mesenchymal phenotype.14,33,36,59 Stain- pattern. In such instances, the distinction between
ing for vimentin is uniformly strongly positive in tumor classic and differentiated adamantinoma should be made
cells of all patterns and in fibroblast-like stroma, includ- (Table 17-1).
ing blood vessels and osteoblasts. Staining for factor VIII, In rare instances, morphologic overlap has been
Ulex europaeus, CD31, and CD34 is negative in all pat- observed between adamantinoma and Ewing’s sarcoma.5,18,21
terns of tumor cells, fibrous stroma, and osteoblasts; These perplexing cases have been referred to as both

ERRNVPHGLFRVRUJ
1154 17  Adamantinoma of Long Bone

A B

C
FIGURE 17-11  ■  Spindled and squamous patterns of adamantinoma. A, Spindled pattern showing palisading of nuclei. B, Nests of
squamous differentiation in adamantinoma. C, Large areas of squamous differentiation in adamantinoma. (A and B, ×200; C, ×150)
(A-C, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1155

A B

C D
FIGURE 17-12  ■  Classic adamantinoma: immunohistochemical features. A, Solid areas of keratin positive tumor cells in fibrous
stroma. B, Higher magnification of A showing strong positivity of tumor cells for keratin. C, Solid nests of tumor cells positive for
vimentin. D, Scattered keratin positive cells in fibrous stroma. (A, ×100; B and C, ×200; D, ×150)

ERRNVPHGLFRVRUJ
1156 17  Adamantinoma of Long Bone

A B

C D
FIGURE 17-13  ■  Classic adamantinoma: immunohistochemical features. A, Solid nests of keratin-positive cells in fibrous stroma.
B, Small tubular structures with strong positivity for keratin in fibrous stroma. C, Keratin-positive tubular structures in fibrous stroma.
D, Scattered keratin-positive tumor cells in fibrous stroma. (A, C, and D, ×200; B, ×150)

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1157

Ewing’s-like adamantinoma and as adamantinoma-like disruption with involvement of the medullary cavity or
Ewing’s because they may not only share phenotypic epi- extension into adjacent soft tissue disqualifies the lesion
thelial features (cytokeratin immunoreactivity and well- as a differentiated adamantinoma.
formed desmosomes ultrastructurally), but also O-13
antigen, S-100, and NSE immunoreactivity, dense core
Gross Findings
granule presence ultrastructurally, as well as 11;22 trans-
location [t(11;22) (q24;q12)], resulting in the fusion of Resection specimens show multiple areas of fibrous-
EWSR1 and FLI1 genes. (See also discussion in Chapter appearing tissue involving the anterolateral cortex of the
11.) Although such exceptional cases definitely enter into tibia. The lesions expand the cortex toward the medullary
the differential diagnosis of adamantinoma, they are cavity and anteriorly and are always delineated by a rim
properly regarded as variants of Ewing’s sarcoma and not of sclerotic cortical bone. True disruption of the cortex
the other way around, in our opinion. with extension into the medullary cavity, soft tissue, or
both is not present. The bowing deformity of the resected
segment of bone is typically present.
DIFFERENTIATED (OSTEOFIBROUS
DYSPLASIA–LIKE) ADAMANTINOMA Microscopic Findings

Definition The microscopic hallmark of this form of adamantinoma


is its overall similarity to osteofibrous dysplasia. The
Differentiated (osteofibrous dysplasia–like) adamanti- predominance of osteofibrous dysplasia–like changes is
noma is exclusively intracortical in the tibia with fre- characterized by the presence of fascicles of fibroblastic
quent synchronous involvement of the ipsilateral fibula. cells with a storiform pattern (Figs. 17-17 and 17-18).
Microscopically, it is characterized by small nests of Trabeculae of bone with prominent rims of osteoblasts
epithelial neoplastic cells, scattered individual epithelial are present within this tissue. The zonal architecture with
cells, or both features, best documented by immuno- progressive trabeculation from woven to lamellar bone
histochemical staining for keratin in a dominant back- and increasing numbers of bone trabeculae at the periph-
ground of osteofibrous dysplasia–like pattern (Figs. 17-14 ery are seen in an appropriately sampled lesion or in the
through 17-17). Sometimes these lesions are designated resection specimens and are similar to the findings in de
as intracortical, juvenile, or osteofibrous dysplasia–like novo osteofibrous dysplasia (Figs. 17-17 and 17-18). The
adamantinomas.65,66 small nests of epithelial tumor cells along with individual
keratin-positive epithelial cells scattered in the fibrous
Incidence and Location stroma are characteristic features (Figs. 17-16 through
17-18). A recent report cited a case of differentiated ada-
This form of adamantinoma is rare. In a series of 25 mantinoma with rhabdoid-like epithelial cell morphol-
consultation cases, 8 could be classified as differentiated ogy.38 Epithelial elements may be present only focally
adamantinoma. Typically it affects patients during the and usually in the central portion of individual lucencies.
first two decades of life. It seems that it is more frequent We postulated that the distinct histologic, radiologic, and
during the second decade of life than in the first. The immunohistochemical features of this form of adaman-
lesion exclusively involves the tibia and the fibula. In 3 of tinoma are the result of indolent growth of tumor cells
our cases, synchronous lesions in the tibia and the fibula and the predominance of a secondary reparative process
were present. The involvement of the anterolateral cortex that overgrows the entire lesion. The reparative origin of
of the tibial midshaft is common. the osteofibrous dysplasia–like pattern in adamantinoma
is supported by its frequent location at the periphery of
the classic adamantinoma and by its zonal architecture
Clinical Symptoms
when it dominates the lesion in the differentiated type
The main symptom is a long history of dull pain in the of adamantinoma.
lower extremity. Clinically evident anterior bowing of the
tibia is frequently present. Treatment and Behavior (Classic and
Differentiated Adamantinoma)
Radiographic Imaging
Adamantinomas of long bones are slow-growing, locally
On radiographs the lesions are indistinguishable from destructive tumors with a high recurrence rate after local
conventional osteofibrous dysplasia. They present as excision.13,24,51,63 It is estimated that metastases, most fre-
multiple lucencies, sclerotic foci, or both features, involv- quently in the lungs, develop in approximately 25% of
ing the anterolateral cortex of the tibia with associated patients with classic adamantinoma. Typically the metas-
cortical expansion and sclerosis of the intervening cortex tases occur late during the clinical course, often several
(Figs. 17-14 and 17-15).11 Anterior bowing of the tibia is (2 to 5) years after diagnosis. The metastases develop
frequently present. Although the cortex can be markedly typically in regional lymph nodes and the lungs but any
expanded, each focus is always delineated by a ring of site may be involved, including the skeleton, liver, and
intact cortical bone. The intracortical location of this brain, especially in the terminal phase. The available
form of adamantinoma is best evaluated by computed incomplete follow-up data of our consultation cases
tomography and MRI. The presence of complete cortical suggest that metastases occur exclusively in classic

ERRNVPHGLFRVRUJ
1158 17  Adamantinoma of Long Bone

A B C D
FIGURE 17-14  ■  Radiographic features of adamantinoma: comparison of classic and differentiated (osteofibrous dysplasia–like) ada-
mantinoma. A, Differentiated adamantinoma involving anterolateral cortex of tibia. Note marked anterior bowing of tibial surface.
B, Differentiated adamantinoma showing multiple intracortical lesions in tibia and fibula of same extremity. C, Classic adamantinoma
represented by large expanding lesion in distal fibula. D, Classic adamantinoma represented by eccentric lytic lesion that has
expanded beyond cortex of tibia. Note sclerotic margins at its periphery. (From Czerniak B, Rojas-Corona RR, Dorfman HD: Cancer
64:2319–2333, 1989.)

adamantinoma. At the time of this writing, we have no Personal Comments


example of differentiated (osteofibrous dysplasia–like)
adamantinoma that metastasized. Therefore in addition Describing the relationship between adamantinoma and
to microscopic and radiographic features, differentiated osteofibrous dysplasia of long bone, we have proposed
adamantinoma may have clinical behavior that overlaps two distinct pathways that the tumor may follow (Fig.
with osteofibrous dysplasia.34 Several cases of differenti- 17-19). The first is that exemplified by the classic ada-
ated osteofibrous dysplasia–like adamantinoma that pro- mantinoma, in which the tumor grows progressively and
gressed to classic adamantinoma have been reported.20,61 may, at least in some instances, invade the surrounding
In addition, in rare instances, classic adamantinoma tissue and metastasize. In the second, characterized by the
may evolve into the so-called dedifferentiated adamanti- differentiated adamantinoma, an intracortically situated
noma progressing to a highly pleomorphic sarcoma-like adamantinoma becomes dominated by a secondary repar-
malignancy.25,31 ative process with the histologic features of osteofibrous
Because differentiated adamantinoma may initially dysplasia. It is possible that the continuation of this repar-
present as clinically indistinguishable from osteofibrous ative process may lead to the total elimination of recog-
dysplasia, it is essential to establish the diagnosis by nizable tumor cells from the lesion. At this stage, it may
biopsy. Osteofibrous dysplasia lesions that have been no longer be possible to identify the underlying primary
adequately sampled may be followed until puberty and condition. The notion that the differentiated type of ada-
then treated by extraperiosteal excision or segmental mantinoma and osteofibrous dysplasia may represent dif-
resection for larger lesions.42 Most adamantinomas, either ferent stages of the same process is supported by the
classic or differentiated, are now treated surgically, with finding, in some cases, of inconspicuous focal remnants
limb salvage as the standard. Wide operative margins can of epithelial elements in a lesion otherwise indistinguish-
be obtained in most cases, with a limb preservation rate able from osteofibrous dysplasia. The close relationship
of more than 80% and a survival rate of close to 90% at between these two lesions is further supported by their
10 years.56 similar anatomic location, age distribution, and tendency

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1159

A B
FIGURE 17-15  ■  Similarity of radiographic features of differentiated adamantinoma and osteofibrous dysplasia. A and B, Differenti-
ated adamantinoma (A) and osteofibrous dysplasia (B) were located in anterolateral cortex of proximal shaft of tibia. Note that both
lesions have associated anterior bowing of tibia.

to grow intracortically. Indeed, the radiologic appearance osteofibrous dysplasia–like pattern is insufficient for a
of these two entities is often identical. diagnosis of differentiated adamantinoma. The lesion
The concept of differentiated adamantinoma implies may appear radiologically to be intracortical for the most
the existence of a continuum of lesions with classic ada- part; however, its focal expansion into the surrounding
mantinoma at one end and osteofibrous dysplasia at tissue should be a warning signal of at least local aggres-
the other. Therefore one can expect intermediate cases sion and indicates the primary site for biopsy. Such lesions
exhibiting features of both subsets of adamantinoma. can contain large components of intracortically located
The diagnosis of differentiated adamantinoma depends differentiated adamantinoma, with focal extension beyond
on factors such as the intracortical location of the the cortex and histologic features of classic adamanti-
entire lesion, uniform predominance of an osteofibrous noma. In such instances, the diagnosis should be based
dysplasia–like pattern, and scattered positivity of epithe- on the most aggressive portion of the tumor. It also
lial elements for cytokeratin. Focal predominance of an must be mentioned that our concept of differentiated

ERRNVPHGLFRVRUJ
1160 17  Adamantinoma of Long Bone

A B

C D

FIGURE 17-16  ■  Differentiated adamantinoma: microscopic features. A, Nests of epithelial cells in central portion of differentiated
adamantinoma. B, Higher magnification of A showing irregular nests of tumor cells in fibrous stroma. C, Residual nests of epithelial
cells in central portion of adamantinoma. D, Higher magnification of A showing inconspicuous nests of tumor cells in fibrous
stroma. Inset, Strong positivity of tumor cells for keratin in differentiated adamantinoma. (A and C, ×100; B and D, ×200)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1161

A B C D
FIGURE 17-17  ■  Differentiated adamantinoma of tibia and fibula: zonal architecture. A, Radiograph shows multilocular lesions in
anterolateral cortex of tibial shaft and in fibula. B-D, Osteofibrous dysplasia–like pattern in peripheral, intermediate, and central
zones of tibial lesion. Note progressive maturation of bone trabeculae from central to peripheral zone and few inconspicuous epi-
thelial nests present in central part of lesion (arrows). (B-D, ×50) (B-D, hematoxylin-eosin.)

adamantinoma and its proposed relationship to osteofi- as osteofibrous dysplasia to be reclassified later as ada-
brous dysplasia does not rule out the possibility of mantinoma61 (Fig. 17-20). In our opinion, such cases
the existence of de novo osteofibrous dysplasia not related represent a sampling problem or misdiagnosis rather
to adamantinoma. It is also suggested that because of than the true evolution of the fibroosseous lesion into an
its intracortical location and similarity to osteofibrous epithelial neoplasm.
dysplasia, the differentiated type of adamantinoma
may represent a prognostically more favorable variant
compared with the classic type of adamantinoma. OSTEOFIBROUS DYSPLASIA
However, this statement remains unproved until suffi-
cient follow-up data on a representative number of cases Osteofibrous dysplasia is a rare lesion that occurs exclu-
are available. sively in the tibia and fibula. Originally the lesion was
Since the publication of our concept of differentiated considered a variant of fibrous dysplasia or a process
adamantinoma and its relationship to osteofibrous analogous to ossifying fibroma of jaw bones.80,87 We
dysplasia–like change, several other groups have con- regard osteofibrous dysplasia of long bones to be patho-
firmed the existence of scattered epithelial elements in genetically distinct from ossifying fibroma of the jaws and
some cases of osteofibrous dysplasia.30,41,66 Our original from fibrous dysplasia. Initially this lesion was designated
concept was that the presence of osteofibrous dysplasia– as ossifying fibroma of long bones or intracortical fibrous dys-
like areas in adamantinoma might indicate a healing or plasia.81,87 These terms had the disadvantage of uninten-
regressive process. The relationship between these two tionally linking this lesion with other more common
entities has become less controversial since we initially fibroosseous disorders. In 1976, Campanacci78,79 intro-
proposed it in 1989, with a number of other authors duced the term osteofibrous dysplasia of long bones for
postulating the opposite pathway, suggesting that osteo- these lesions and proposed the concept of a self-healing
fibrous dysplasia can be a precursor lesion for adamanti- process that evolves until puberty and then stabilizes.
noma.20,22,62 It is not unusual for cases originally diagnosed The relationship between osteofibrous dysplasia and

ERRNVPHGLFRVRUJ
1162 17  Adamantinoma of Long Bone

A B C D

FIGURE 17-18  ■  Osteofibrous dysplasia of tibia: zonal architecture. A, Radiograph shows multilocular lesion in anterolateral cortex
of proximal shaft of tibia. B-D, Osteofibrous dysplasia pattern in peripheral, intermediate, and central zones of lesion. Note progres-
sive maturation of bone trabeculae from central to peripheral zones. (B-D, ×50) (B-D, hematoxylin-eosin.)

adamantinoma of long bones, which is discussed in the fibula, or both locations. Synchronous involvement of the
section on adamantinoma of long bone in this chapter, is tibia and fibula on one side is typical. The peak rate of
well documented in literature.72,75,76,83,85,91,93,95-98,100-102,104 incidence and skeletal sites of involvement are shown in
However, there are unquestionable cases of de novo Figure 17-21.
osteofibrous dysplasia unrelated to adamantinoma.71,82,86
Rare forms of inherited familial osteofibrous dysplasia Radiographic Imaging
have also been described.89 The following description
is restricted to this de novo variant of osteofibrous The lesion typically exhibits multiloculated radiolucent
dysplasia. expansion of the anterolateral cortex of the tibia (Figs.
17-22 and 17-23).74,88 Anterior bowing associated with
cortical thickening and sclerosis in the midshaft is very
Definition
characteristic. Additional synchronous, predominantly
Osteofibrous dysplasia is a disorder characterized by the lytic foci may be present near the distal or proximal end
presence of fibrous tissue and bone trabeculae rimmed by or, less commonly in the fibula. The anterior bowing
osteoblasts. The lesion occurs exclusively in the tibia, deformity is usually present at the time of diagnosis and
fibula, or both locations and predominantly affects infants may become more pronounced with progression of the
and children. disease.
In young children, especially newborns, the lesion may
present as a purely lytic, sharply demarcated, expansile
Incidence and Location
defect in the tibial diaphysis (Figs. 17-24 and 17-25).
Osteofibrous dysplasia is a rare lesion, accounting for less In this age group, the origin of the lesion in the antero-
than 1% of all bone tumors. Although it typically occurs lateral cortex may not be obvious on radiographs. Com-
during the first two decades of life, some cases have been puted tomography and MRI can be used to demonstrate
present at birth.71,81,94,99,103 There is no clear sex predilec- multiloculated anterolateral intracortical lesions with
tion. All reported cases have been located in the tibia, cortical expansion and sclerosis of the intervening bone

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1163

they anastomose and finally merge with the thickened


cortical bone. There is centrifugal maturation of woven
bone to lamellar bone. The central portion of the lesion
is predominantly fibrous and may exhibit myxomatous or
cystic changes. Pathologic fracture, hemorrhage, and
Progression
focal collections of multinucleated giant cells can alter
this classic pattern. Single, scattered, keratin-positive
cells that are verified ultrastructurally as epithelial ele-
ments are frequently present and support the concept
that osteofibrous dysplasia may represent an indolent
form of adamantinoma.73 In fact, in several resected spec-
imens, we found small epithelial nests in the fibrous
central zone in otherwise typical osteofibrous dysplasia.
This is not to deny the existence of cases in which a
scrupulous search is negative for any epithelial compo-
Fibroosseous nent. The identification of clonal chromosomal abnor-
overgrowth malities in osteofibrous dysplasia supports the concept
that an underlying indolent neoplastic process may be
involved in the pathogenesis of osteofibrous dysplasia.77
In the original description of this entity, Kempson90
used the term ossifying fibroma to distinguish it from
fibrous dysplasia. He postulated that this was an aggres-
Further sive lesion with a tendency for local recurrence and
fibroosseous progression. Campanacci and Laus79 designated the
De novo
overgrowth same lesion as osteofibrous dysplasia and observed that it
osteofibrous
dysplasia
initially grows in a locally aggressive way but that the
process has a tendency to stabilize at puberty. During
the second decade of life, at least some of these lesions
become stationary and develop gradual sclerosis, but the
deformities may persist. This observation necessitated a
more conservative approach to the treatment of osteofi-
brous dysplasia, which should be limited to corrective
FIGURE 17-19  ■  Adamantinoma: pathways of development. procedures rather than radical excisions.
Graphic presentation of concept of differentiated adamanti-
noma of long bones and its relationship to osteofibrous
dysplasia–like changes. Differential Diagnosis
Clinically, radiographically, and microscopically, osteofi-
(Figs. 17-22 and 17-23). In extremely rare cases, osteofi- brous dysplasia must be differentiated from conventional
brous dysplasia may involve both extremities and present fibrous dysplasia and from both forms of adamantinoma:
as bilateral tibial lesions (Fig. 17-25). classic and differentiated. Although the microscopic fea-
tures of fibrous dysplasia and osteofibrous dysplasia can
Gross Findings be somewhat overlapping, the distinct radiographic pre-
sentation, almost exclusive involvement of the tibia and
Sections of resected segments of the tibial shaft show a the absence of GNS mutations in this disorder distin-
multilocular intracortical lesion in the anterolateral guishes it pathogenetically from fibrous dysplasia.92 The
cortex that is composed of gritty fibrous tissue. Expansion detailed differential diagnosis of osteofibrous dysplasia
of the cortex with sclerosis is usually clearly seen. Occa- and fibrous dysplasia is described in Chapter 8 in the
sionally the lesion may consist of a single larger focus of section on fibrous dysplasia. The differential diagnosis
fibrous tissue surrounded by an expanded shell of scle- between osteofibrous dysplasia and adamantinoma of
rotic cortex. Cortical disruption and invasion into soft long bones is discussed in the section on osteofibrous
tissue are typically not present. dysplasia–like adamantinoma in this chapter.
Our delineation of two subsets of adamantinoma,
namely the classic and differentiated (osteofibrous
Microscopic Findings
dysplasia–like) types, suggests that the latter lesion,
The lesion is composed of loose bundles of fibroblast-like despite focal presence of epithelial elements, pursues a
cells that often show a storiform pattern. Within this clinical course similar to that of conventional osteofi-
tissue, trabeculae of bone are present and are surrounded brous dysplasia.84 Other authors indicate that some cases
by a row of rimming osteoblasts (Fig. 17-26). In appro- of osteofibrous dysplasia may progress to classic adaman-
priately sampled cases, zonal architecture can be observed tinoma.93 Hence, ruling out the possibility of adamanti-
with progressive widening, maturation, and increased noma continues to be a major problem in planning the
numbers of mineralized bone trabeculae at the periphery management of osteofibrous dysplasia–like lesions.
of the lesion. As the trabeculae become more numerous, Text continued on p. 1171

ERRNVPHGLFRVRUJ
1164 17  Adamantinoma of Long Bone

A B C
FIGURE 17-20  ■  Slow local growth of differentiated adamantinoma of tibia in an 18-year-old woman with diagnosis of osteofibrous
dysplasia 11 years previously. A, Multifocal lytic changes of tibia diagnosed as osteofibrous dysplasia when patient was 8 years
old. B, Persistent multifocal lesion 3 years later. Note slow progression of lesion, with sclerosis and mild anterior bowing of tibia.
C, Same lesion 11 years after original diagnosis. Note almost no progression compared with B remodeling of tibial cortex and partial
correction of bowing. At this time, biopsy with immunohistochemical studies revealed scattered epithelial elements, and lesion was
reclassified as differentiated adamantinoma.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1165

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 17-21  ■  Osteofibrous dysplasia. Peak age incidence and typical sites of skeletal involvement are indicated by a solid black
arrow.

ERRNVPHGLFRVRUJ
1166 17  Adamantinoma of Long Bone

A C
FIGURE 17-22  ■  Osteofibrous dysplasia: radiographic features. A, Lateral radiograph of tibia of an 8-year-old boy with anterior bowing
deformity associated with intracortical lesion of tibia. B, T1-weighted magnetic resonance imaging shows cortical thickening.
C, Computed tomogram shows intracortical lesion narrowing medullary cavity.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1167

A B

C
FIGURE 17-23  ■  Osteofibrous dysplasia: radiographic features. A, Lateral view of tibia of young patient with anterior bowing deformity
and fusiform cortical expansion of anterior cortex. Fibula is not involved. B, Pathologic fracture ensured through tibial lesion shown
in A. C, Computed tomogram shows multilocular, low-signal defects in thickened anteromedial cortex of tibia.

ERRNVPHGLFRVRUJ
1168 17  Adamantinoma of Long Bone

B
FIGURE 17-24  ■  Osteofibrous dysplasia: radiographic features. A, Bowing deformity of left tibia in a 4-month-old boy with large lytic,
expansile lesion in anterolateral cortex. B, Lateral view of lesion shown in A. Biopsy showed features of osteofibrous dysplasia.
Lesion was first discovered when boy was 1 month old.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1169

A B
FIGURE 17-25  ■  Osteofibrous dysplasia: radiographic features. A, Radiographs of left and right tibiae show bilateral involvement in
a young child. B, Lateral view of right tibia shown in A demonstrates marked anterior bowing deformity.

ERRNVPHGLFRVRUJ
1170 17  Adamantinoma of Long Bone

A B

C D
FIGURE 17-26  ■  Osteofibrous dysplasia (ossifying fibroma): microscopic features. A-C, Osteofibrous dysplasia with irregular, imma-
ture bone trabeculae surrounded by prominent osteoblastic rims. Note fibrous stroma. D, Example of typical fibrous dysplasia. Note
absence of osteoblastic rims and bone trabeculae. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1171

REFERENCES 25. Hazelbag HM, Laforga JB, Roels JH, et al: Dedifferentiated ada-
mantinoma with revertant mesenchymal phenotype. Am J Surg
Classic Adamantinoma Pathol 27:1530–1537, 2003.
1. Alguacil-Garcia A, Alonso A, Pettigrew NM: Osteofibrous dys- 26. Hazelbag HM, Taminiau AH, Fleuren GJ, et al: Adaman-
plasia (ossifying fibroma) of the tibia and fibula and adamanti- tinoma of the long bones: a clinicopathological study of 32
noma. Am J Clin Pathol 82:470–474, 1984. patients with emphasis on histological subtype, precursor lesion,
2. Baker PL, Dockerty MB, Coventry MB: Adamantinomas (so and biological behavior. J Bone Joint Surg 76A:1482–1499,
called) of long bones: review of literature and report of three new 1994.
cases. J Bone Joint Surg 36A:704–720, 1954. 27. Hazelbag HM, Wessels JW, Mollevangers P, et al: Cytogenetic
3. Benassi MS, Campanacci L, Gamberi G, et al: Cytokeratin analysis of adamantinoma of long bones: further indications for a
expression and distribution in adamantinoma of the long bones common histogenesis with osteofibrous dysplasia. Cancer Genet
and osteofibrous dysplasia of tibia and fibula: an immunohisto- Cytogenet 97(1):5–11, 1997.
chemical study correlated to histogenesis. Histopathology 25:71–76, 28. Hicks JD: Synovial sarcoma of the tibia. J Pathol Bacteriol 67:115–
1994. 161, 1956.
4. Bridge JA, Dembinski A, DeBoer J, et al: Clonal chromosomal 29. Huvos AG, Marcove RC: Adamantinoma of long bones: a clini-
abnormalities in osteofibrous dysplasia. Implications for histo- copathological study of 14 cases with vascular origin suggested.
pathogenesis and its relationship with adamantinoma. Cancer J Bone Joint Surg 57A:148–154, 1975.
73(6):1746–1752, 1994. 30. Ishida T, Iijima T, Kikuchi F, et al: A clinicopathological and
5. Bridge JA, Fidler ME, Neff JR, et al: Adamantinoma-like Ewing’s immunohistochemical study of osteofibrous dysplasia, differenti-
sarcoma: genomic confirmation, phenotypic drift. Am J Surg ated adamantinoma, and adamantinoma of long bones. Skeletal
Pathol 23(2):159–165, 1999. Radiol 21:493–502, 1992.
6. Camp MD, Tompkins RK, Spanier SS, et al: Best cases from the 31. Izquierdo FM, Ramos LR, Sanchez-Herraez S, et al: Dedifferen-
AFIP: adamantinoma of the tibia and fibula with cytogenetic tiated classic adamantinoma of the tibia: a report of a case with
analysis. Radiographics 28:1215–1220, 2008. eventual complete revertant mesenchymal phenotype. Am J Surg
7. Campanacci M, Giunti A, Bertoni F, et al: Adamantinoma of the Pathol 34:1388–1392, 2010.
long bones: the experience at the Istituto Ortopedico Rizzoli. Am 32. Johnson LC: Congenital pseudoarthrosis, adamantinoma of long
J Surg Pathol 5:533–542, 1981. bone, and intracortical fibrous dysplasia of the tibia. J Bone Joint
8. Campanacci M, Laus M: Osteofibrous dysplasia of the tibia and Surg 54:1355, 1972. (abstract).
fibula. J Bone Joint Surg 63A:367–375, 1981. 33. Jundt G, Remberger K, Roessner A, et al: Adamantinoma of long
9. Changus GW, Speed JS, Stewart FW: Malignant angioblastoma bones. A histopathological and immunohistochemical study of 23
of bone: a reappraisal of adamantinoma of long bone. Cancer cases. Pathol Res Pract 191:112–120, 1995.
10:540–559, 1957. 34. Kahn LB: Adamantinoma, osteofibrous dysplasia and differenti-
10. Cohen DM, Dahlin DC, Pugh DG: Fibrous dysplasia associated ated adamantinoma. Skeletal Radiol 32(5):245–258, 2003.
with adamantinoma of the long bones. Cancer 15:515–521, 35. Kanamori M, Antonescu CR, Scott M, et al: Extra copies of chro-
1962. mosomes 7, 8, 12, 19 and 21 are recurrent in adamantinoma.
11. Czerniak B, Rojas-Corona RR, Dorfman HD: Morphologic J Mol Diagn 3(1):16–21, 2001.
diversity of long bone adamantinoma: the concept of differenti- 36. Kashima TG, Dongre A, Flanagan AM, et al: Podoplanin expres-
ated (regressing) adamantinoma and its relationship to osteofi- sion in adamantinoma of long bones and osteofibrous dysplasia.
brous dysplasia. Cancer 64:2319–2334, 1989. Virchows Arch 459:41–46, 2011.
12. Dameron TB, Jr: Adamantinoma of the appendicular skeleton. 37. Keeney GL, Unni KK, Beabout JW, et al: Adamantinoma of long
Johns Hopkins Med J 145:107–111, 1979. bones. A clinicopathologic study of 85 cases. Cancer 64:730–737,
13. Desai SS, Jambhekar N, Agarwal M, et al: Adamantinoma of tibia: 1989.
a study of 12 cases. J Surg Oncol 93:429–433, 2006. 38. Kempson RL: Ossifying fibroma of the long bones. Arch Pathol
14. Dickson BC, Gortzak Y, Bell RS, et al: p63 expression in adaman- 82:218–233, 1966.
tinoma. Virchows Arch 459:109–113, 2011. 39. Kohler G, Rossner JA, Waldherr R: Zur Structur und Differen-
15. Dockerty MB, Myerding HW: Adamantinoma of the tibia: report tialdiagnose des sog. Tibia—Adamantinomes. Eine Licht-und
of two new cases. JAMA 119:932–937, 1942. electronenoptische Untersuchung. Verh Dtsch Ges Pathol 58:454–
16. Eisenstein W, Pitcock JA: Adamantinoma of the tibia: an eccrine 458, 1974.
carcinoma. Arch Pathol Lab Med 108:246–250, 1984. 40. Kumar D, Mulligan ME, Levine AM, et al: Classic adamantinoma
17. Fischer B: Über ein primares Adamantinom der Tibia. Frankfurt in a 3-year-old. Skeletal Radiol 27(7):406–409, 1998.
Z Pathol 12:422–441, 1913. 41. Kuruvilla G, Steiner GC: Osteofibrous dysplasia-like adamanti-
18. Fujii H, Honoki K, Enomoto Y, et al: Adamantinoma-like Ewing’s noma of bone: a report of five cases with immunohistochemical
sarcoma with EWS-FLI1 fusion gene: a case report. Virchows Arch and ultrastructural studies. Hum Pathol 29(8):809–814, 1998.
449(5):579–584, 2006. 42. Lee RS, Weitzel S, Eastwood DM, et al: Osteofibrous dysplasia
19. Gleason BC, Liegl-Atzwanger B, Kozakewich HP, et al: Osteofi- of the tibia. Is there a need for radical surgical approach? J Bone
brous dysplasia and adamantinoma in children and adolescents: Joint Surg Br 88(5):658–664, 2006.
a clinicopathologic reappraisal. Am J Surg Pathol 32:363–376, 43. Llombart-Bosch A, Ortuno-Pacheco G: Ultrastructural findings
2008. supporting the angioblastic nature of the so-called adamantinoma
20. Hatori M, Watanabe M, Hosaka M, et al: A classical adamanti- of the tibia. Histopathology 2:189–200, 1978.
noma arising from osteofibrous dyplasia-like adamantinoma in the 44. Maki M, Athanasou N: Osteofibrous dysplasia and adamantinoma:
lower leg: a case report and review of the literature. Tohoku J Exp correlation of proto-oncogene product and matrix protein expres-
Med 209(1):53–59, 2006. sion. Hum Pathol 35:69–74, 2004.
21. Hauben E, van den Broek LC, Van Marck E, et al: Adamantinoma- 45. Maki M, Saitoh K, Kaneko Y, et al: Expression of cytokeratin 1,
like Ewing’s sarcoma and Ewing’s-like adamantinoma. The 5, 14, 19 and transforming growth factors-β1, β3 in osteofibrous
t(11;22), t(21;22) status. J Pathol 195(2):218–221, 2001. dysplasia and adamantinoma: a possible association of transform-
22. Hazelbag HM, Fleuren GJ, Cornelisse CJ, et al: DNA aberrations ing growth factor-β with basal cell phenotype promotion. Pathol
in the epithelial cell component of adamantinoma of long bones. Int 50(10):801–807, 2000.
Am J Pathol 147:1770–1779, 1995. 46. Mandahl N, Heim S, Rydholm A, et al: Structural chromosome
23. Hazelbag HM, Fleuren GJ, van den Broek LJ, et al: Adamanti- aberrations in an adamantinoma. Cancer Genet Cytogenet 42:187–
noma of the long bones: keratin subclass immunoreactivity pattern 190, 1989.
with reference to its histogenesis. Am J Surg Pathol 17:1225–1233, 47. Markel SF: Ossifying fibroma of long bone: its distinction from
1993. fibrous dysplasia and its association with adamantinoma of long
24. Hazelbag HM, Hogendoorn PC: Adamantinoma of the long bone. Am J Clin Pathol 69:91–97, 1978.
bones: anatomo-clinical review and its relationship with osteofi- 48. Moon NF: Adamantinoma of the appendicular skeleton in chil-
brous dysplasia. Ann Pathol 21:499–511, 2001. dren. Int Orthop 18:379–388, 1994.

ERRNVPHGLFRVRUJ
1172 17  Adamantinoma of Long Bone

49. Moon NF, Mori H: Adamantinoma of the appendicular skeleton: chemical study correlated to histogenesis. Histopathology 25:71–76,
updated. Clin Orthop Rel Res 204:215–237, 1986. 1994.
50. Parham DM, Bridge JA, Lukacs JL, et al: Cytogenetic distinction 74. Bethapudi S, Ritchie DA, Macduff E, et al: Imaging in osteofi-
among benign fibro-osseous lesions of bone in children and ado- brous dysplasia, osteofibrous dysplasia-like adamantinoma, and
lescents: value of karyotypic findings in differential diagnosis. classic adamantinoma. Clin Radiol 69:200–208, 2014.
Pediatr Dev Pathol 7:148–158, 2004. 75. Blasius S, Edel G, Wuisman P, et al: Osteofibrous dysplasia of
51. Park YK, Unni KK, McLeod RA, et al: Osteofibrous dysplasia: long bones: a reactive process to adamantinomatous tissue.
clinicopathologic study of 80 cases. Hum Pathol 24:1339–1347, J Cancer Res Clin Oncol 118:152–156, 1992.
1993. 76. Bloem JL, van der Heul RO, Schuttevaer HM, et al: Fibrous
52. Pieterse AS, Smith PS, McClure J: Adamantinoma of long bones: dysplasia vs adamantinoma of tibia: differentiation based on dis-
clinical, pathological and ultrastructural features. J Clin Pathol criminant analysis of clinical and plain film findings. Am J Roent-
35:780–786, 1982. genol 156:1017–1023, 1991.
53. Povysil C, Kohout A, Urban K, et al: Differentiated adamanti- 77. Bridge JA, Dembinski A, DeBoer J, et al: Clonal chromosomal
noma of the fibula: a rhabdoid variant. Skeletal Radiol 33(8):488– abnormalities in osteofibrous dysplasia: implications for histo-
492, 2004. pathogenesis and its relationship with adamantinoma. Cancer
54. Povysil C, Matejovsky Z: Ultrastructure of adamantinoma of long 73:1746–1752, 1994.
bones. Virchows Arch [A] 393:233–244, 1981. 78. Campanacci M: Osteofibrous dysplasia of long bones: a new clini-
55. Povysil C, Matejovsky Z, Horak M: Classic adamantinoma, dif- cal entity. Ital J Orthop Traumatol 2:221–237, 1976.
ferentiated adamantinoma and osteofibrous dysplasia of long 79. Campanacci M, Laus M: Osteofibrous dysplasia of tibia and fibula.
bones. Cas Lek Cesk 143(5):329–335, 2004. J Bone Joint Surg 63A:367–375, 1981.
56. Qureshi AA, Shott S, Mallin BA, et al: Current trends in the 80. Campbell CJ, Hawk T: A variant of fibrous dysplasia (osteofibrous
management of adamantinoma of long bones. An international dysplasia). J Bone Joint Surg 64A:231–236, 1982.
study. J Bone Joint Surg 82A(8):1122–1131, 2000. 81. Castellote A, Garcia-Pena P, Lucaya J, et al: Osteofibrous
57. Rosai J: Adamantinoma of the tibia: electron microscopic evidence dysplasia: a report of two cases. Skeletal Radiol 17:483–486,
of its epithelial origin. Am J Clin Pathol 51:786–792, 1969. 1988.
58. Rosai J, Pinkus GS: Immunohistochemical demonstration of epi- 82. Cetinkaya M, Ozkan H, Koksal N, et al: Neonatal osteofi-
thelial differentiation in adamantinoma of the tibia. Am J Surg brous dysplasia associated with pathological tibia fracture: a case
Pathol 6:427–434, 1982. report and review of the literature. J Pediatr Orthop B 21:183–186,
59. Sarita-Reyes CD, Greco MA, Steiner GC: Mesenchymal- 2012.
epithelial differentiation of adamantinoma of long bones: an 83. Cohen DM, Dahlin DC, Pugh DG: Fibrous dysplasia associated
immunohistochemical and ultrastructural study. Ultrastruct Pathol with adamantinoma of long bones. Cancer 15:515–521, 1962.
36:23–30, 2012. 84. Czerniak B, Rojas-Corona RR, Dorfman HD: Morphologic
60. Sozzi G, Miozzo M, Di Palma S, et al: Involvement of the region diversity of long bone adamantinoma: the concept of differenti-
13q14 in a patient with adamantinoma of the long bones. Hum ated (regressing) adamantinoma and its relationship to osteofi-
Genet 85:513–515, 1990. brous dysplasia. Cancer 64:2319–2334, 1989.
61. Springfield DS, Rosenberg AE, Mankin HJ, et al: Relationships 85. Hazelbag HM, Taminiau AH, Fleuren GJ, et al: Adamantinoma
between osteofibrous dysplasia and adamantinoma. Clin Orthop of long bones: a clinicopathological study of 32 patients with
Relat Res 309:234–244, 1994. emphasis on histological subtype, precursor lesion, and biological
62. Sweet DE, Vinh TN, Devaney K: Cortical osteofibrous dysplasia behavior. J Bone Joint Surg 76A:1482–1499, 1994.
of long bone and its relationship to adamantinoma: a clinicopatho- 86. Jobke B, Bohndorf K, Vieth V, et al: Congenital osteofibrous
logic study of 30 cases. Am J Surg Pathol 16:282–290, 1992. dysplasia Campanacci: spontaneous postbioptic regression.
63. Szendroi M, Antal I, Arato G: Adamantinoma of long bones: a J Pediatr Hematol Oncol 36:249–252, 2014.
long-term follow-up study of 11 cases. Pathol Oncol Res 15:209– 87. Johnson LC: Congenital pseudoarthrosis, adamantinoma of long
216, 2009. bone, and intracortical fibrous dysplasia of tibia. J Bone Joint Surg
64. Taylor RM, Kashima TG, Ferguson DJ, et al: Analysis of stromal 54A:1355, 1972. (abstract).
cells in osteofibrous dysplasia and adamantinoma of long bones. 88. Jung JY, Jee WH, Hong SH, et al: MR findings of the osteofi-
Mod Pathol 25:56–64, 2012. brous dysplasia. Korean J Radiol 15:114–122, 2014.
65. Ueda Y, Blasius S, Edel G, et al: Osteofibrous dysplasia of long 89. Karol LA, Brown DS, Wise CA, et al: Familial osteofibrous dys-
bones: a reactive process to adamantinomatous tissue. J Cancer Res plasia. A case series. J Bone Joint Surg 87A:2297–2307, 2005.
Clin Oncol 118:152–156, 1992. 90. Kempson RL: Ossifying fibroma of long bones: a light and elec-
66. Ueda Y, Roessner A, Bosse A, et al: Juvenile intracortical adaman- tron microscopic study. Arch Pathol 82:218–233, 1966.
tinoma of the tibia with predominant osteofibrous dysplasia–like 91. Klein MJ: Osteofibrous dysplasia: unique lesion, fibrous dysplasia
features. Pathol Res Pract 187:1039–1043, 1991. variant, or reactive process associated with other lesions? Adv Anat
67. Unni KK: Dahlin’s bone tumors: general aspects and data on 11,087 Pathol 2:28–32, 1995.
cases, ed 5, Philadelphia, 1996, Lippincott-Raven. 92. Liang Q, Wei M, Hodge L, et al: Quantitative analysis of activat-
68. Unni KK, Inwards CY, Bridge JA, et al: Tumor of the bones and ing alpha subunit of the G protein (Gsα) mutation by pyrose-
joint, AFIP Atlas of Tumor Pathology 4th Series: Fascicle 2, 2005, quencing in fibrous dysplasia and other bone lesions. J Mol Diagn
Washington, DC. 13:137–142, 2011.
69. Van der Woude HJ, Hazelberg HM, Bloem JL, et al: MRI of 93. Mankin HJ, Mindell ER: Relationship between osteofibrous dys-
adamantinoma of long bones in correlation with histopathology. plasia and adamantinoma. Clin Orthop 309:234–244, 1994.
AJR Am J Roentgenol 183(6):1737–1744, 2004. 94. Nakashima Y, Yamamuro T, Fujiwara Y, et al: Osteofibrous dys-
70. Weiss SW, Dorfman HD: Adamantinoma of long bone: an analy- plasia (ossifying fibroma of long bones), a study of 12 cases. Cancer
sis of nine new cases with emphasis on metastasizing lesions and 52:909–914, 1983.
fibrous dysplasia–like changes. Hum Pathol 8:141–153, 1977. 95. Park YK, Unni KK, McLeod RA, et al: Osteofibrous dysplasia:
clinicopathologic study of 80 cases. Hum Pathol 24:1339–1347,
Osteofibrous Dysplasia 1993.
71. Anderson MJ, Townsend DR, Johnston JO, et al: Osteofibrous 96. Resnik CS, Young JW, Levine AM, et al: Case report 604: osteo-
dysplasia in newborn: report of case. J Bone Joint Surg 75A:265– fibrous dysplasia (ossifying fibroma) of tibia. Skeletal Radiol
267, 1993. 19:217–219, 1990.
72. Baker PL, Dockerty MB, Coventry MD: Adamantinoma 97. Roessner A, Bosse A, Edel G, et al: Juvenile intracortical adaman-
(so-called) of long bones: review of literature and report of three tinoma of tibia with predominant osteofibrous dysplasia like fea-
new cases. J Bone Joint Surg Am 36:704–720, 1954. tures. Pathol Res Pract 187:1039–1043, 1991.
73. Benassi MS, Campanacci L, Gamberi G, et al: Cytokeratin 98. Schoenecker PL, Swanson K, Sheridan JJ: Ossifying fibroma of
expression and distribution in adamantinoma of long bones tibia: report of a new case and review of the literature. J Bone Joint
and osteofibrous dysplasia of tibia and fibula: an immunohisto- Surg 63A:483–488, 1981.

ERRNVPHGLFRVRUJ
17  Adamantinoma of Long Bone 1173

99. Sissons H, Kancheria P, Wallace B: Ossifying fibroma of bone: 102. Unni KK: Adamantinoma of long bones: the mystery endures.
report of two cases. Bull Hosp J Dis 43:1–4, 1983. Adv Anat Pathol 3:16–21, 1996.
100. Smith NM, Byard RW, Foster B, et al: Congenital ossifying 103. Wang JW, Shih CH, Chen WJ: Osteofibrous dysplasia (ossifying
fibroma (osteofibrous dysplasia) of tibia: a case report. Pediatr fibroma of long bones): a report of four cases and review of litera-
Radiol 21:449–451, 1991. ture. Clin Orthop 278:235–243, 1992.
101. Sweet DE, Vinh TN, Devaney K: Cortical osteofibrous dysplasia 104. Weiss SW, Dorfman HD: Adamantinoma of long bone. An analy-
of long bone and its relationship to adamantinoma: a clinicopatho- sis of nine new cases with emphasis on metastasizing lesions and
logic study of 30 cases. Am J Surg Pathol 16:282–290, 1992. fibrous dysplasia-like changes. Hum Pathol 8:141–153, 1977.

ERRNVPHGLFRVRUJ
C H A P T E R 1 8 

Chordoma and Related Lesions


CHAPTER OUTLINE

NOTOCHORDAL RESTS AND BENIGN CHONDROID CHORDOMA


NOTOCHORDAL TUMORS
DEDIFFERENTIATED CHORDOMA
CHORDOMA

In evolutionary biology, the notochord or chorda dorsalis indicate that Rho-GTPase activity protein (XrGAP),
is typically discussed as a prototypic internal skeleton axial protocadherin, and antagonists of β-catenin are
of prevertebrate animals. In vertebrates, the notochord critical mediators for notochord formation.1,2,4,6 It should
exists transiently and in addition to providing structural be viewed as a tissue that is evolutionary and functionally
integrity to the embryo, it secretes factors that are essen- related to cartilage and shares many features that are
tial for body patterning with respect to the dorsal-ventral common for notochord and cartilage cells. It expresses
and left-right axes.10 These signals are required for posi- such characteristic cartilage components as type II and
tional orientation and fate information concerning the type IX collagen, aggrecan, Sox9, and chondromodulin.
development of several abdominal and thoracic organs as In contrast to cartilage cells, notochord cells produce a
well as the development of the central nervous system. In thick basement membrane and retain hydrated materials
human embryonal development, the notochord forms an in large vacuoles. The expression profiles characteristic
axial structure that attains its complete muturation in of chordoid and chondroid lesions that may have diag-
the 10- to 11-mm embryo. Subsequently, it undergoes nostic significance are summarized in Table 18-1.129
gradual involution and fragmentation by the ossification
centers of the axial skeleton (Fig. 18-1).4,7,8 During the
second month of embryonal development, the notochord NOTOCHORDAL RESTS AND BENIGN
is restricted to the intervertebral residues (Fig. 18-2). It NOTOCHORDAL TUMORS
is generally received that in adults, it forms the nucleus
pulposus of the intervertebral disks, but recent data show In 1857, Luschka, Hasse, Zenker, and Virchow described
that the cells of the nucleus pulposus do not express small gelatinous nodules projecting into the cranial cavity
brachyury or epithelial markers, bringing into question from the clivus.2,5,19,31 Virchow considered these lesions
the notochord origin of the central portion of human as cartilaginous and, because of the prominent vesicular
intervertebral disks.129 Occasionally, its remnants can be cytoplasmic change of their cells, designated them ecchor-
found in the peripheral parts of the vertebral bodies. dosis physaliphora.27 In 1858, H. Muller performed a devel-
They are also present in the sphenooccipital area and in opmental study of human notochord and concluded that
the sacrococcygeal region. They occasionally form larger the “ecchordoses” were derived from the notochordal
masses that are probably derived from the persistent tissue.22 In 1894, Ribbert confirmed Muller’s opinion;
intravertebral notochord canal and are located more since then, it has been generally accepted that the gelati-
centrally within the vertebral bodies.2,3,11,12,100 The rem- nous nodules occasionally found in the clivus or dorsum
nants of notochord tissue are referred to as ecchordosis sellae are “ecchordoses” composed of chordal tissue and
physaliphora. that chordomas arise from the remains of chordal tissue.24
Microscopically, the notochord tissue is somewhat Klebs in 1864 and Grahl in 1903 described the first large
similar to immature cartilage and is composed of oval “ecchordoses” causing symptoms.14,17,28
cells with central nuclei and vacuolated cytoplasm embed- In the past notochord rests were identified as inciden-
ded in an eosinophilic myxomatous stroma2,3,4,7,9 (Fig. tal autopsy findings or were found in vertebral biopsy
18-2). The characteristic physaliphorous cells are focally samples taken for other reasons. With the development
present. Immunohistochemically, the notochord cells of modern computerized imaging techniques, they are
have some features of cartilage and epithelial cells and are more often identified before surgery during the diag-
strongly positive for S-100 protein and keratin and epi- nostic workup of the axial skeleton.23,29 Several cases
thelial membrane antigen (EMA) and brachyury (Fig. recently presented, discussed at meetings of the Interna-
18-3).8 The coexpression of S-100 protein, epithelial tional Skeletal Society, and published in reports provide
markers, and brachyury is a hallmark of notochord tissue evidence that notochordal lesions represent a spectrum
and is retained in chordoma.9 The molecular mechanisms of conditions ranging from occult microscopic remnants
governing the development and involution of the noto- to large symptomatic masses.13,15,16,18,20,21,25,29 Large noto-
chord are still poorly understood. The prelimary data chordal lesions with bland histologic appearance typically
1174
ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1175

Residual
notochord
Somite

Noto-
chord
Intervertebral Residual
disk Vertebral notochord
body tissue

Residual
Neural Sclerotome notochord
tube
Notochord
A B C D
FIGURE 18-1  ■  Embryonal development of notochord and relationship to other structures of central axis. A, Fully developed noto-
chord forming axial structure located anteriorly to neural tube. B, Notochord surrounded by sclerotomes forming ossification centers
of vertebral column. C, Notochord obliterated by ossification centers of vertebral bodies. Note intravertebral notochord canal, which
may persist focally. D, Final stage of involution with residual notochordal tissue forming nucleus pulposus of intervertebral disks.

A B
FIGURE 18-2  ■  Development of notochord: microscopic features. A, Whole-mount photomicrograph of vertebral column from 3-month
human embryo showing vertebral ossification centers with residual notochord forming disklike structures within the intervertebral
cartilages. B, Higher magnification of residual notochordal tissue within an intervertebral cartilage. C and D, Residual notochordal
tissue located in place of future nucleus pulposus showing cordlike arrangement of cells within myxoid stroma. (A, ×4; B, ×20;
C, ×60; D, ×180.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1176 18  Chordoma and Related Lesions

8 14%

7 Number of lesions
Frequency depending upon anatomical segments 12%

6
10%
Number of lesions

Frequency
8%
4
6%
3

4%
2

1 2%

0 0%
T5
C1
C2
C3
C4
C5
C6
C7
T1
T2
T3
T4

T6
T7

T9
T10
T11
T12

S1
S2
S3
S4
S5
T8

L5
L1
L2
L3
L4

Coccyx
Clivus

Cervical Thoracic Lumbar Sacro-coccygeal


FIGURE 18-3  ■  Anatomic distribution of intraosseous notochord rests. The lesions are most frequently found in the clivus and sacro-
coccygeal vertebra followed by the cervical and lumbar spine. The distribution, depending on anatomic segments, is identical to
that of classic chordomas. (Contributed by Yamaguchi T, et al, unpublished data.)

TABLE 18-1 Expression Profiles of Chordoid (Fig. 18-3). Microscopically such rests are composed of
and Chondroid Tumors solid areas of notochordal cells without nuclear atypia or
mitotic activity (Fig. 18-4). Small cystic spaces filled with
Common serous fluid can be present. The oval cytoplasm of the
SOX-9 notochordal cells shows various degree of vacuolization.
Collagen type II fibromodulin Typical physaliphorous cells are present focally. Some
Cartilage oligomeric matrix protein of such rests can be composed of cells with extensively
Aggrecan
Cartilage link protein 1
vacuolated cytoplasm that resemble fat or lipoma. The
S100 calcium-binding protein A1 trabecular bone of the medulla is intact and the nests of
Chondroitin sulfate proteoglycan 4 notochordal cells grow between the trabecular spaces.
Proline/arginine-rich end leucine-rich repeat protein The overall architecture is that of a stationary lesion
Chondroitin 6 focally surrounded by the embracing trabeculae of the
Chordoid medullary bone (Figs 18-5 and 18-6).
Brachyury The published reports and our own experience indi-
Keratin 8 cate that symtomatic notochordal rests typically measure
Keratin 15 at least 10 mm and may be multifocal involving several
Keratin 18 vertebral bodies. Microscopically such lesions are com-
Keratin 19
Discoidin domain receptor 1 posed of a uniform, sheetlike proliferation of notochordal
CD24 cells, which permeate and fill the intratrabecular spaces
(Fig. 18-7). The trabecular bone is preserved but may
Chondroid
show features of remodeling and sclorosis focally. Thick-
Collagen type X
Platelet-derived growth factor α
ened trabecular bone may have central cores of hyaline
Reticulocalbin-3 cartilage with peripheral seams of partially calcified or
ossified cartilage. The lesion has a nonagressive growth
pattern with pushing borders and is sharply delineated
measuring greater than 10 mm are designated as giant from the adjacent tissue. The notochordal cells are closely
notochordal rests or giant notochordal hamartomas. packed with no intercellular stroma. Most cells have clear
Detailed mapping studies of the axial skeleton indicates cytoplasm and resemble fat at low magnification.8,21,30 In
that the distribution of small notochordal rests measuring contrast, to adipocytes in the nuclei of notochordal cells
less than 5 mm is similar to that of conventional chordoma are frequently centrally located and are surrounded by

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1177

A B

FIGURE 18-4  ■  Notochord rest: microscopic features. A and B, Notochord rest of the clivus beneath sella turcica composed of uniform
cells filling intertrabecular spaces with preservation of the trabecular bone. C, Higher magnification of A and B showing sheetlike
proliferation of cells resembling fat with uniform mostly centrally located nuclei. Inset, Higher magnification of notochordal cells
with vacuolated cytoplasm. (A, ×4; B, ×16; C, ×200; Inset, ×400) (A-C and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1178 18  Chordoma and Related Lesions

B C
FIGURE 18-5  ■  Notochord rest: gross and microscopic features. A, Whole-mount photomicrograph of vertebral column with a small
notochordal rest. B, Higher magnification of A showing overall architectural arrangement of the rest with preservation of the tra-
becular bone. Note pushing borders with trabecular bone partially outlining the rest. C, Higher magnification of A and
B showing sheetlike proliferation of notochordal cells with vacuolated cytoplasms resembling fat. (A, ×4; B, ×16; C, ×200)
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1179

A B
FIGURE 18-6  ■  Giant notochord rest: radiographic features. A, T1-weighted sagittal magnetic resonance image (MRI) () shows low-
signal lesion in the body of L3 (arrows). B, T2-weighted MRI of lesion shown in A exhibits signal enhancement (arrows).

the cytoplasm, which forms eosinophilic strands delineat- addition, cellular atypia and heterogeneity is lacking in
ing vacuolar structures. benign notochordal lesions. The bone both trabecular
Immunohistochemically both small and giant noto- and cortical is typically destroyed in chordomas and is
chord rests retain phenotypic features of normal noto- intact or focally thickened in benign notochordal lesions.
chord and stain positive for keratins AE1/AE3, CAM5.2,
EMA, S-100 protein, and brachyury (Fig. 18-8).13 They
are negative for p53 protein, Ki67 (MIB-1), and high
molecular weight keratin 34βE12 (keratin 903).18 CHORDOMA
Notochordal rests may progress to a conventional Definition
chordoma. Chordomas associated with notochordal
rests range from incidental microscopic findings to Chordoma is a slow-growing neoplasm that exhibits
large destructive symptomatic lesions (Figs 18-9 and notochordal differentiation and most frequently arises in
18-10).26,30 the sacrococcygeal and sphenooccipital regions. Some
chordomas originate in the notochord rests, but it is
unclear whether all tumors of this type arise from the
Differential Diagnosis
persistent remnant of notochord tissue.
It is important that notochordal rests not be confused
with chordoma. The correlation of microscopic features Incidence and Location
with radiologic images helps avoid this error. Extensive
bone destruction with associated paravertebral soft tissue Chordoma is a relatively common tumor, accounting for
mass is the hallmark of chordoma. Microscopically noto- 3% to 4% of primary malignant bone tumors in major
chordal rests lack the lobulation, mucinous stromal pools, series. The peak age incidence and the frequent sites of
and syncytial cell chords characteristic of chordoma. In Text continued on p. 1184

ERRNVPHGLFRVRUJ
1180 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-7  ■  Giant notochord rest: microscopic features. A, A uniform sheetlike proliferation of notochordal cells filling up the
intratrabecular spaces. Note preservation of the trabecular bone. B, Focus on thicken bone trabeculae with scallop borders forming
interconnecting network, signifying bone remodeling frequently seen in giant notochord rest. C, Solid sheetlike proliferation of
notochordal cells with clear cytoplasm resembling fat. D, Pushing border sharply delimiting the notochord cells from the adjacent,
uninvolved fatty marrow. Inset, Higher magnification of a physaliphorous cell typically present in notochord rests. (A and D, ×100;
B and C, ×60; Inset, ×600) (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1181

A B

C D
FIGURE 18-8  ■  Notochord rest: microscopic and immunohistochemical features. A, Notochord tissue with vacuolization of cytoplasm.
B, Physaliphorous cells in notochord rest. C, Strong positivity of notochord tissue for keratin. D, Positive staining for S-100 protein.
(A and C, ×300; B and D, ×600) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1182 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-9  ■  Chordoma developing in association with giant notochord rest: microscopic features. A and B, Whole-mount photo-
micrographs of coccyx showing a giant notochord rest and a focus of chordoma (arrows). Note that focus of chordoma shows
distinctive lobular architecture with myxoid stromal change and exhibits destructive growth pattern, breaking through the cortex
into the periosteal fibroadipose tissue. B, Higher magnifications of A and B showing a focus of chordoma with lobular architecture
and myxoid stroma. C, Higher magnification of A showing chordoma cells infiltrating periosteal stromal tissue. D, Higher magnifica-
tion of B showing chordoma cells infiltrating periosteal stromal tissue. Inset, Microscopic features of giant notochordal cells with
solid sheetlike proliferation of bland looking notochordal cells with clear cytoplasm resembling fat. (A and B, ×10; C and D, ×100;
Inset, ×100)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1183

A B

C D
FIGURE 18-10  ■  Chordoma developing in association with giant notochord rest: radiographic, gross, and microscopic features.
A, Gadolinium-diethylenetriamine pentaacetic acid enhanced T1-weighted magnetic resonance image. Tumor protrudes anteriorly
from the coccyx and consists of intermediate density lobules separated by thin contrasted septations. Two intraosseous lesions of
low density corresponding to notochord rests are identified (arrows). B, Gross photograph of the resected specimen discloses a
lobulated tumor composed of gelatinous tissue with foci of hemorrhage. Two intraosseous lesions corresponding to multifocal giant
notochord rests are also present (arrows). C, Microscopic features of a giant notochord rest showing sheetlike solid proliferation
of cells with vacuolated cytoplasm. D, Microscopic features of notochordal rest (upper part) and chordoma (lower part).
Note myxoid appearance of stroma in chordoma and bland looking solid proliferation of benign notochord cells. (C and D, ×80)
(C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1184 18  Chordoma and Related Lesions

Sphenooccipital area

Incidence

50

1 2 3 4 5 6 7 8
Age in decades

FIGURE 18-11  ■  Chordoma. Peak age incidence and frequent sites of skeletal involvement. Most frequent sites are indicated by solid
black arrows.

skeletal involvement are shown in Figure 18-11. Chor- spine is rare. Extremely rare examples of multicentric
doma appears to be the fourth most frequent primary chordoma involving several anatomic sites have been
malignant tumor of bone after osteosarcoma, chondro- reported.34
sarcoma, and Ewing’s sarcoma. Approximately 8% of all
primary malignant bone tumors were reported as chor- Clinical Symptoms
domas to the National Cancer Institute’s Surveillance,
Epidemiology, and End Results (SEER) Program. The Pain is a common symptom in chordoma and can last for
age-specific incidence and frequency distribution show months to several years.58 Compression of the nerves in
gradual increases and peak during the fifth and sixth the sacrococcygeal region and obstruction of the rectum
decades of life (Fig. 18-12). It must be mentioned that a resulting in constipation are frequent presentations.
small number of chordomas occur during the first and Sphenooccipital chordomas produce symptoms related to
second decades of life.42,52 In this age group, the involve- compression or destruction of various adjacent struc-
ment of the second cervical vertebra is typical. Very rare tures, such as the optic nerves or pituitary gland.47,60,83
cases of congenital chordoma have been described.89 They also can present with symptoms characteristic of a
Some of the major series report a male sex predominance. tumor of the pontocerebellar region. Spinal chordomas
However, SEER data show an almost equal male-to- frequently produce compression of the cord and
female sex distribution.57 It appears that, similar to nerves.43,67 Cervical chordoma frequently presents as a
Ewing’s sarcoma, chordomas are less frequent in black pharyngeal mass.53,81,90
patients. In the SEER data, only 18 (2.1%) of 650 chor-
domas were reported in black patients. Radiographic Imaging
Chordoma almost exclusively involves the axial
skeleton, which is in keeping with its notochordal On plain radiographs, chordoma typically presents as a
origin.55,7,12,20-22,59,95 Nearly 90% of cases occur in the lytic lesion (Fig. 18-13).40,84,102,104,105 Scattered, discrete
sacrococcygeal region and in the base of the skull. opacities representing intralesional calcifications can be
The remaining cases are reported predominantly in the present. Sphenooccipital lesions destroy the clivus, the
cervical and lumbar spine. Involvement of the thoracic sella turcica, or both areas (Figs 18-14 and 18-15).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1185

0.25 12

10
Incidence Rate (per 1000,000)

0.2

Frequency %
0.15

0.1
4

0.05
2

0 0
4

+
0-

5-

-1

-1

-2

-2

-3

-3

-4

-4

-5

-5

-6

-6

-7

-7

-8

85
10

15

20

25

30

35

40

45

50

55

60

65

70

75

80
Age Group

Source: SEER Incidence Rate Frequency


FIGURE 18-12  ■  Epidemiology of chordoma. (Data from National Cancer Institute’s Surveillance, Epidemiology, and End Results Program,
1975-2010. Age-adjusted incidence rates and age-specific frequency, all races, both sexes, 650 cases.)

A B
FIGURE 18-13  ■  Chordoma: radiographic features. A, Anteroposterior radiograph of sacrum shows expansive lytic lesion of body of
sacrum. B, Lateral radiograph of lumbar spine shows unusual sclerotic appearance of body of L4 produced by vertebral
chordoma.

ERRNVPHGLFRVRUJ
1186 18  Chordoma and Related Lesions

FIGURE 18-14  ■  Intracranial chordoma: radiographic features. Lateral view of skull shows destruction of sella turcica region of base
produced by sphenooccipital chordoma (arrow).

FIGURE 18-15  ■  Chordoma: radiographic features. T2-weighted magnetic resonance image of chordoma at base of skull shows high
signal intensity mass in region of sphenoid bone (arrows).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1187

Sacrococcygeal lesions expand the bone contour and textbooks, are relatively sparse or may not be present
obliterate normal markings (Fig. 18-16). Vertebral chor- at all in otherwise typical chordoma. The classic large
domas produce lytic lesions that may involve two or more physaliphorous cell has a centrally located nucleus sur-
adjacent vertebral bodies. Chordomas of the vertebral rounded by a narrow rim of cytoplasm that in turn, is
column and the sacrum originate in the vertebral bodies encircled by a ring of more peripherally located cytoplas-
and produce masses that grow anteriorly to the dural sac mic vacuoles. Extensive vacuolization of the cytoplasm
(Figs 18-17 to 18-19). Lesions of the coccygeal region, may cause a clear-cell appearance of the tumor (Fig.
however, may grow both anteriorly and posteriorly to the 18-28). Such lesions are descriptively referred to as
sacrum (Fig. 18-16). Chordoma may rarely present as a lipoma-like chordomas and may be confused with benign
sclerotic lesion (Fig. 18-13, B). Computed tomography or malignant lipomatous tumors. Some chordomas may
and magnetic resonance imaging are indispensable in have a sarcoma-like appearance caused by elongation of
evaluating both the extent of the lesion and the involve- their nuclei and the presence of nuclear atypia (Fig.
ment of adjacent structures.72,74,75,91,97,103 Magnetic reso- 18-23). On the other hand, highly cellular lesions with
nance imaging, in particular, can reveal the lobulated minimal vacuolization and dense eosinophilic cytoplasm
nature of the lesion, and T2-weighted images typically may mimic epithelial neoplasms.
show signal enhancement (Fig. 18-15). Chordoma may also exhibit cartilaginous differentia-
tion. Areas of cartilage can range from small microscopic
Gross Findings foci to large prominent areas. Therefore chordomas can
be occasionally difficult to distinguish from chondrosar-
Chordomas are soft, gray-tan, multilobulated masses coma. The term chondroid chordoma has been proposed to
(Fig. 18-20 to 18-22). They are frequently myxoid or designate hybrid lesions that exhibit features of both
gelatinous in appearance and can mimic chondrosarcoma lesions—chordoma and chondrosarcoma. The contro-
or mucinous adenocarcinoma. The expanded bone versy over this entity and its potential for clinical signifi-
contour is frequently delineated by a thin rim of perios- cance are discussed separately.
teum. The intraosseous borders are typically indistinct.
Despite good demarcation, the tumor usually extends
Cytology
beyond its grossly recognizable borders. The recurrent
lesions are typically in the form of multiple nodules. Fine-needle aspirates from chordoma usually are cellular
Chordomas often involve the medullary canal, compress- and contain abundant myxoid material admixed with
ing the spinal cord and its nerves. clear, vacuolated neoplastic cells33,38,54,68 (Fig. 18-31). The
presence of abnormal multivacuolated cytoplasm with
Histology centrally placed, scalloped nucleus (that is, physalipho-
rous cells) is the hallmark of chordoma. Such classical
Chordomas are composed of cords or nests of solid cells are, however, rare; in the vast majority, the cells
areas of cells that have vacuolated cytoplasm embed- show dense eosinophilic cytoplasm or are lipoblast-like
ded in a myxoid intercellular matrix (Fig. 18-23). The with single or several larger cytoplasmic vacuoles displac-
nuclei are round or oval and have a prominent nucleolus. ing the nucleus peripherally. Cytologic features com-
Mitoses are rare, and atypical mitotic figures usually are bined with radiologic and clinical presentation allow
not present. The level of cellularity can vary consid- correct cytologic diagnosis in most cases of chordoma.
erably among cases and in different areas of the same The immunohistochemical features such as coexpression
tumor. Some tumors are highly cellular with minimal or of epithelial markers and S-100 protein can be also tested
no intercellular matrix. More often, there is a moder- in cytologic preparations.
ate amount of cellular tissue and clearly recognizable
intercellular myxoid stroma. In a typical case, the tumor Special Techniques
cells form an anastomosing pattern of cords with focal
solid areas separated by a prominent intercellular matrix Ultrastructurally, the physaliphorous cells show numer-
(Fig. 18-24). In general, most chordomas exhibit clearly ous vacuolated spaces that may occupy large portions
recognizable nuclear pleomorphism with occasional large of the cytoplasm (Fig. 18-30).48,59,61,62,69,80,85,87,90,96,101,126 A
atypical cells. prominent Golgi center, rough endoplasmic reticulum,
Vacuolization of the cytoplasm is almost invariably and patchy areas of glycogen are present. Chordoma
present (Figs. 18-24 to 18-28). Frequently, it is in the cells contain intermediate filaments and are connected by
form of large (single or several) vacuoles displacing the desmosome-like junctions. The extracellular matrix con-
nucleus peripherally and causing the so-called signet- sists of a fine, granular substance of low electron density.
ring-like appearance of the tumor cells. It can also Some chordomas may show prominent aggregates of
be in the form of smaller (multivesicular) structures. microtubules in their cytoplasm.
Occasionally, the vacuoles encircle the nucleus, which Immunohistochemically, chordomas coexpress S-100
remains centered in the cytoplasm and produces the protein, epithelial markers (i.e., cytokeratins, EMA) and
so-called physaliphorous appearance. The physaliphorous brachyury (Figs 18-32 and 18-33).32,73,76,92,98,115 They may
cell is a hallmark of chordoma (Figs 18-29 and 18-30). It also be positive for neurofilaments and may express the
must be mentioned, however, that classic physaliphorous carcinoembryonic antigen (CEA). Chordomas also show
cells, as they are frequently illustrated in many pathology overexpression of cathepsin K and reduced expression of

ERRNVPHGLFRVRUJ
1188 18  Chordoma and Related Lesions

B
FIGURE 18-16  ■  Sacrococcygeal chordoma: radiographic features. A, Computed tomography shows lobulated mass protruding pos-
teriorly and anteriorly from origin in sacrococcygeal junction. B, T1-weighted sagittal magnetic resonance image of chordoma shown
in A with larger posterior and anterior low signal mass (arrows).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1189

FIGURE 18-17  ■  Chordoma of cervical spine: radiographic features. Lateral radiograph of cervical spine shows destructive lesion of
body of C2 (arrows).

E-cadherin.64,79 The cytoplasm of tumor cells contains to 5q, 7q, 12q, and 20q may play a role in the develop-
material positive for periodic acid-Schiff, which is dia- ment of these tumors. A rare syndrome of familial chor-
stase sensitive. In general chordoma recapitulate a gene doma was linked to chromosome 7q33.56,70
expression signature of notochordal cells.63,65,78,95 DNA
ploidy measurements show that chordoma may be aneu- Differential Diagnosis
ploid or diploid, but DNA ploidy patterns do not cor-
relate with clinical outcome. Among all of the markers, Chordoma must be distinguished from chondrosarcoma.
brachyury appears to be the most specific for chordoma The presence of physaliphorous cells and a trabecular or
and notochordal tissue and is useful in the differential cordlike arrangement together with a myxoid matrix are
diagnosis.41,93 Brachyury is an important transcriptional typical for chordoma. However, conventional chondro-
regulator in the FGFR/MEK/ERK pathway, playing an sarcoma occasionally may show a cordlike arrangement of
essential role in chordoma cell growth and sur- cells and myxoid stroma. In such instances, positivity for
vival.60,66,82,88,94,107 A more detailed description of the bio- S-100 protein and negative staining for epithelial markers
logic role of brachyury in notochord development and favor chondrosarcoma. Other myxoid bone tumors, such
chordoma is provided in Chapter 3. as chondromyxoid fibroma, which rarely occurs in the axial
Cytogenetic studies have been performed on a limited skeleton, are very unlikely to be confused with chordoma.
number of cases.45,71,77,99 It seems that abnormalities of Highly cellular areas with cohesive solid cellular sheets
chromosome 21, including loss or structural rearrange- and prominent nuclear atypia can be confused, especially
ment of 21(q22), are frequent in these tumors. The most in a limited biopsy specimen, with an epithelial neoplasm.
common cytogenetic abnormality is hypodiploidy with Extensive clear-cell change and the signet-ring appear-
loss of the short arm of chromosome 3, and loss of proxi- ance of chordoma cells can lead to a misdiagnosis of meta-
mal 1p as well as monosomy of chromosome 10.99 Com- static adenocarcinoma or less frequently lipomatous tumor.
parative genomic hybridization studies disclosed that On the other hand metastatic carcinomas can be misdiag-
losses of 1p and 3p could be detected in 50% of chordo- nosed as chordoma. Among various epithelial tumors
mas. Chordomas are also characterized by frequent gains metastatic to the skeleton a chromophobe variant of renal cell
of genetic material on 5q, 7q, 12q, and 20q. The gains carcinoma may superficially resemble a chordoma. Strict
on 17q appear to be a dominant event present in 70% of adherence to clinical and radiographic data usually helps
tumors. The cytogenetic data suggests that tumor sup- to avoid this error. It must be mentioned that the
pressor genes on 1p and 3q as well as oncogenes mapping Text continued on p. 1206

ERRNVPHGLFRVRUJ
1190 18  Chordoma and Related Lesions

A B
FIGURE 18-18  ■  Chordoma of cervical spine: radiographic features. A, T1-weighted magnetic resonance image (MRI) of cervical spine
shows expansive, low signal lesion of body of C2, with narrowing of spinal canal and compression of spinal cord. B, T2-weighted
MRI shows high signal intensity in lesion shown in A.

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1191

C
A
FIGURE 18-19  ■  Vertebral chordoma: radiographic features. A, Sagittal magnetic resonance image (MRI) of vertebral column shows
lesion of high signal intensity at thoracolumbar junction (T12-L1) (arrow). B and C, T1- and T2-weighted axial MRIs of lesion shown
in A that encircles vertebral body (arrows).

ERRNVPHGLFRVRUJ
1192 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-20  ■  Chordoma: radiographic, gross, and microscopic features. A, Axial computed tomography shows destructive mass
protruding posteriorly and anteriorly from the sacrum (arrows). B, T1-weighted sagittal magnetic resonance image of chordoma
shown in A with predominantly anterior low signal mass (arrows). C, Sagitally cut resection specimen of the tumor shown in A and
B demonstrates a lobulated, fleshy tumor mass destroying the sacrum and extending to the soft tissue anteriorly and posteriorly.
D, Histologic section of same tumor shows cords of chordoma cells with multivesicular cytoplasm growing in a myxoid stroma.
(D, ×200) (D, hematoxylin-eosin.) (Reprinted with permission from Czerniak B, et al: Aspiration cytology of bone tumors. In: Koss LG, ed:
Diagnostic cytology and its histopathologic bases, ed 5. Lippincott Williams & Wilkins, New York, 2005.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1193

A B

C D
FIGURE 18-21  ■  Chordoma: gross features. A, Resection specimen of sacral chordoma showing a gelatinous tumor growing anteriorly
from the coccyx. Note extensive interstitial hemorrhage and areas of necrosis. B, Higher magnification of A showing a tumor growing
anteriorly out of coccyx. C, Gross photograph of a bisected lumbar (L5) vertebral resection specimen showing a gelatinous tumor
mass replacing almost the entire vertebral body. D, Specimen radiograph of tumor shown in C showing destructive lesion replacing
the vertebral body.

ERRNVPHGLFRVRUJ
1194 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-22  ■  Chordoma: gross features. A, Bisected tumor from sacral region shows tan-gray soft tissue with myxoid appearance.
B, Bisected tumor mass from sacral region shows multilobulated myxoid tumor tissue with hemorrhagic areas. C, Autopsy specimen
shows recurrent chordoma of cervical spine invading medullary canal and compressing spinal cord (arrows). D, Sagittally cut resec-
tion specimen of recurrent sacrococcygeal chordoma shows multiple gray, fleshy nodules within coccyx and adjacent soft tissue
(arrows).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1195

A B

C D
FIGURE 18-23  ■  Chordoma: microscopic features. A, Solid sheets of chordoma cells with vacuolated cytoplasm and areas of tumor
with more pronounced atypia. B, Higher magnification of A showing tumor cells with pronounced atypia. C, Higher magnification
of another area in A showing tumor cells with extensive vacuolated cytoplasms. D, Higher magnification of chordoma cells showing
pronounced atypia. Inset, Higher magnification of tumor cells showing vacuolated cytoplasm and physaliphorous features. (A, ×60;
B and C, ×100; D, ×200; Inset, ×300) (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1196 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-24  ■  Chordoma: microscopic features. A, Low power view of lobulated chordoma with mucinous stromal change. B, Higher
magnification of A showing nests and cords of highly atypical tumor cells loosely arranged in a myxoid stroma. C, View of chordoma
showing tumor cells loosely arranged in a myxoid stroma. D, Higher magnification of C showing predominantly cellular makeup of
the tumor. Note extensive vacuolization of the cytoplasm. Insets, Individual chordoma cells with pronounced nuclear atypia. (A and
C, ×60; B and D, ×120; Insets, ×200) (A-D and Insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1197

C
FIGURE 18-25  ■  Chordoma: microscopic features. A, Solid area of highly vacuolated cells with occasional atypical nuclei. B, Intercon-
necting cords of small cells with prominent vacuolization and abundant myxoid stroma. C, Cords and trabeculae of large eosinophilic
cells with focal vacuolar changes. (A-C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1198 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-26  ■  Chordoma: microscopic features. A, Solid proliferation of chordoma cells with predominantly granular cytoplasms.
B, Solid nests of chordoma cells dispersed in a myxoid stroma. C, Cords of tumor cells growing in myxoid stroma. D, Unusual
microscopic appearance of chordoma with loosely connected predominantly spindled tumor cells separated by interstitial fluid.
(A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1199

A B

C D
FIGURE 18-27  ■  Chordoma: microscopic features. A, Low magnification of chordoma showing lobular arrangement. Note increased
cellularity at the periphery of lobule. B, Cords of chordoma cells with epithelioid features. C, Loosely arranged tumor cells with
extensively vacuolated cytoplasm growing in a myxoid stroma. D, Solid area of tumor cells with granular cytoplasm and epithelioid
features. Tumors with such features can be confused with an epithelial neoplasm. Inset, Higher magnification of chordoma cells
with granular cytoplasms. (A, ×60; B-D, ×100; Inset, ×300) (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1200 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-28  ■  Chordoma: microscopic features. A and B, Unusual appearance of chordoma with tumor cells showing extensively
vacuolated cytoplasms resembling fat. Tumors with such features are referred to as lipoma-like chordomas and may be confused
with liposarcoma, especially when they involve the retroperitoneal tissue. C and D, Higher magnification of chordoma with exten-
sively vacuolated cytoplasm with pronounced nuclear atypia. (A and B, ×100; C and D, ×200) (A-D, hematoxlyin-eosin).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1201

A B

C D
FIGURE 18-29  ■  Chordoma: physaliphorous cells. A-D, Microscopic appearances of physaliphorous cells. (A-D, ×600)
(A-D, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
1202 18  Chordoma and Related Lesions

B
FIGURE 18-30  ■  Chordoma: ultrastructural features. A, Ultrastructure of physaliphorous cell. Multiple cytoplasmic vacuoles of low
electron density are present. B, Low power magnification of cord of chordoma cells connected by desmosome-like junctions. Light
microscopic appearance of physaliphorous cells. Inset left, light miscroscoptic features of physaliphorous cells. Inset right, higher
magnification of desmosome-like junction. (A, ×3000; B, ×2000; Inset, right, ×16,000; Inset, left, ×600).

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1203

A B

C D

FIGURE 18-31  ■  Chordoma: histologic and cytologic features. A and B, Histologic features of chordoma demonstrating tumor cells
with vacuolated cytoplasm growing in cords and trabeculae within loose myxoid stroma. B, A larger tumor cell with vacuolated
cytoplasm surrounding the nucleus (physaliphorous cell) (arrow). C, Fine-needle aspirate containing loosely arranged chordoma
cells with myxoid extracellular matrix. D, Higher magnification of chordoma cells with ill-defined cytoplasm and larger cell with a
prominent vacuolated cytoplasm corresponding to a physaliphorous cell (arrows). Inset, Higher magnification of chordoma cells
with vesicular cytoplasm and prominent nucleoli. (A, ×200; B, ×300; C, ×100; D, ×400; Inset, ×600) (A-D and Inset, hematoxylin-eosin.)
(Reprinted with permission from Czerniak B et al: Aspiration cytology of bone tumors. In: Koss LG, ed: Diagnostic cytology and its histo-
pathologic bases, ed 5. Lippincott Williams & Wilkins, New York, 2005.)

ERRNVPHGLFRVRUJ
1204 18  Chordoma and Related Lesions

A B

C D
FIGURE 18-32  ■  Chordoma: histologic and immunohistochemical features. A, Histologic features of chordoma demonstrating solid
tumor growth composed of cells with prominent eosinophilic and vacuolated cytoplasm. B, Strong positivity of chordoma cells for
pancytokeratin. C and D, Strong nuclear positivity of chordoma cells for brachyury. (A-D, ×100) (A, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1205

A B

C D
FIGURE 18-33  ■  Chordoma: immunohistochemical features. A, Cords of chordoma cells in mucoid stroma. B, Positive staining for
S-100 protein. C, Positive staining for keratin (AE1/AE3) in chordoma cells. D, Positivity for periodic acid-Schiff reflects high content
of glycogen in chordoma cell cytoplasm. (A-D, ×400) (A, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1206 18  Chordoma and Related Lesions

coexpression of S-100 protein and epithelial markers is distinguishing it from cartilaginous tumors that show
not specific for chordoma and is sometimes seen in various negative immunoreactivity to the epithelial markers. In
neoplasms, including adenocarcinomas. addition, 58-nucleotidase can be histochemically docu-
mented in chordoma but not in chondroid chordoma or
chondrosarcoma.106
Treatment and Behavior
In 1986, Abenoza and Sibley32 reported that three
Chordoma is typically a locally aggressive tumor with chondroid chordomas were positive for the epithelial
a high propensity for local recurrence. Because of its markers in both chondroid and chordoid areas, whereas
unique location in the axial skeleton, it frequently affects the chondrosarcomas they studied were negative for
vital structures and causes death. Chordoma is associated cytokeratin and EMA. They concluded that immunohis-
with a high delayed mortality rate and a mean survival tochemical studies were useful in the differential diag-
of approximately 4 years.44 Survival longer than 10 years nosis between these tumors. In the late 1980s, Brooks
is extremely rare.44,90 A significant improvement in the et al.107,108 reported totally discordant results because
overall survival rate was noted in SEER data for 1973 cases previously classified as chondroid chordomas
to 1987. Over this period, the 5-year survival rates of were negative for cytokeratins and EMA, even in their
patients with chordoma improved from 40% to 65%. chordoma-like areas. As a result of their immunohisto-
Complete surgical excision, if possible, is the treatment of chemical study and review of the literature on chondroid
choice. Unresectable tumors are treated by debulking and chordomas, Brooks et al. concluded that chondroid chor-
adjuvant radiation therapy. The rate of metastases varies doma did not exist and that the tumors at the skull base
widely among the series, with an average of 40%.46,49,50,51 with cartilaginous appearance were better classified as
The unusually high rates of metastases reported in some low-grade chondrosarcoma. However, their “chondroid
series are probably the result of consideration in the anal- chordomas” represented a peculiarly nonchordomatous
ysis of the local or regional spread of recurrent tumors. group of tumors. None of these tumors contained clear-
True distant metastases are relatively rare and most likely cut chordoid areas, and all were basically cartilaginous
occur in less than 10% of patients. Tumor recurrences or myxocartilaginous. In 1992, Wojno et al.130 repeated
are typically in the form of multiple bone and soft tissue their immunohistochemical study on 17 cases of car-
masses or as subcutaneous nodules.35,36,39 Distant metas- tilaginous tumors of the craniospinal axis. The chon-
tases usually occur late during the course of disease and droid differentiation in these tumors was confirmed not
typically involve the lungs (Fig. 18-34). Metastases in the only by light microscopic findings, but also by immu-
regional lymph nodes can also be seen. Chordoma may nohistochemical expression for type II collagen. A total
occasionally exhibit unusually aggressive clinical behavior of 4 of 17 tumors were virtually indistinguishable from
with early distant metastases and massive vascular tumor chondrosarcomas. The remaining 13 tumors had areas
emboli involving multiple sites.37,86 of recognizable conventional chordoma in addition to
the cartilaginous areas. The cells within the chondroid
components of most of these 13 tumors stained for the
epithelial markers. One of 4 tumors composed entirely
CHONDROID CHORDOMA of cartilage was positive for the epithelial markers. The
remaining 3 tumors were negative for these markers and
Definition
were classified as low-grade chondrosarcomas. These
Chondroid chordoma is a tumor that contains a mixture authors concluded that chordomas can contain cartilage
of chondroid and cartilaginous elements. It predomi- and that chordomas with cartilage (“chondroid chordo-
nantly occurs in the central axis and involves the skull mas”) can be distinguished from chondrosarcomas. This
base, spine, or sacrococcygeal region (Figs 18-35 and article appeared to reverse the trend of doubt about the
18-36). existence of chondroid chordoma. In modern literature,
chondroid chordomas continue to be listed mostly as
single case reports. A detailed molecular profiling dis-
Controversy over Chondroid Chordoma
closes, in spite of significant molecular overlap, distinct
Chondroid chordoma is relatively rare. The first report profiles for chondroid and chordoid lesions, with brachy-
published on this entity, by Heffelfinger et al.,112 identi- ury being a major discriminator.129 On the other hand,
fied 22 chondroid chordomas in a series of 155 conven- it appears that cartilage-appearing components of chon-
tional chordomas. Since the original article in 1973, this droid chordomas express both keratins and brachyury
entity has become accepted, and the term chondroid chor- and are likely to be of notochordal origin.125,129
doma has become a widely used designation for chondroid
tumors of the skull base.109,111,116,117,120,127,123,128,132 However, Personal Comments
advances in immunohistochemistry have caused a debate
over the existence of this entity.118,119,122,124 Both cartilage Review of our consultation files of cartilaginous and
cells and tumor cells of chordomas are positive for chordoid tumors of the skull base supported by the
S-100 protein. In addition to immunoreactivity for S-100 immunohistochemical studies led us to the following
protein and vimentin, chordoma cells show positive conclusions114:
immunoreaction to epithelial markers such as cytokeratin 1. Histologically, chondrosarcomas of the skull base
and EMA and occasionally to CEA.32,73,76,92,98,106,112,113,121 may show myxoid change of the matrix and a
This immunophenotype of chordoma cells is useful in linear arrangement of tumor cells.107,108,110,112 These

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1207

B
FIGURE 18-34  ■  Metastatic chordoma in lung. A, Multiple tumor emboli in pulmonary vessels. B, High magnification showing
tumor embolus in peribronchial vessel. Inset, Microscopic details of tumor cells with clear cytoplasm. (A, ×40; B, ×100; Inset, ×200)
(A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1208 18  Chordoma and Related Lesions

A B

C E
FIGURE 18-35  ■  Chondroid chordoma: microscopic features. A, Area of conventional chordoma with prominent vacuolization of tumor
cells. B, Cartilaginous differentiation in chondroid chordoma. C, Higher magnification of chondroid differentiation. D, Positive reac-
tivity of chondroid area for keratin. E, Positive reactivity of chondroid area for epithelial membrane antigen. (A and B, ×100;
C-E, ×200) (A-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1209

A B

C D
FIGURE 18-36  ■  Comparison of microscopic features of conventional chordoma and chondroid chordoma. A, Conventional chordoma
with interconnecting cords of tumor cells in prominent mucoid stroma. B, Area of chondroid matrix in chondroid chordoma. C and
D, Physaliphorous cells showing prominent bubbly cytoplasmic change in conventional-appearing areas of chondroid chordoma.
(A and B, ×100; C, ×400; D, ×600). (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1210 18  Chordoma and Related Lesions

findings are somewhat reminiscent of the cohesive


arrangement of chordoma cells. It is on the basis of DEDIFFERENTIATED CHORDOMA
this histologic pattern that chondrosarcomas of the Definition
skull base have been misinterpreted as chondroid
chordoma. However, the typical cords or nest for- Dedifferentiated chordoma is a tumor composed of two
mation (two or more cells in thickness) usually distinct elements: conventional chordoma and high-
found in chordoma is lacking in chondrosarcomas. grade spindle-cell or pleomorphic sarcoma. The phe-
It is important to recognize that a linear arrange- nomenon of dedifferentiation in chordoma is similar to
ment of tumor cells can occur in chondrosarcomas that originally postulated for dedifferentiated chondro-
and that a strandlike appearance is insufficient evi- sarcoma. Chordomas with a high-grade sarcomatous
dence for a diagnosis of chordoma. component were described as early as 1915 and 1923 by
2. Chondroid differentiation in chordomas appears Albert and Burrow, respectively.133,131 The term dediffer-
not to be such a rare phenomenon if tiny foci of entiated chordoma was postulated by Meis et al. in 1987.141
cartilage are included. Previously reported chon-
droid chordomas that were positive for cytokeratin Incidence and Location
and EMA may represent tumors of this category.
The proportion of chondroid and chordoid areas Dedifferentiation in chordoma is rare, and only individ-
in such tumors may range from a predominance of ual cases have been described in the literature. The age
chordoma to an equal volume of each component. and anatomic sites involved are similar to those seen in
This immunophenotype indicates that chordoma conventional chordoma. All cases described are located
cells have an ability to differentiate into cartilage in the axial skeleton, predominantly in the sphenooccipi-
cells. Although chondroid chordomas were initially tal and sacrococcygeal regions.
considered to be confined to the sphenooccipital
region, 8 of 14 cytokeratin- and EMA-positive Radiographic Imaging
chondroid chordomas reported by Wojno et al.130
arose from the spine or sacrococcygeal region. This Dedifferentiated chordoma presents as a destructive lytic
seems to indicate that chondroid differentiation in lesion. Identification of its components—conventional
chordomas is not limited to those that originate in chordoma and high-grade sarcoma—is not resolved by
the sphenooccipital region. conventional radiography. However, the possibility of
3. It is also important to mention that some of the dedifferentiation can be suspected in a case of a rapidly
reports concerning chondroid chordomas dealt enlarging mass with progressive symptoms in a patient
with tumors that were virtually cartilaginous or with a history of conventional chordoma.
myxocartilaginous in nature and were classified as
chondroid chordomas on the basis of microscopic Gross Findings
impressions. These tumors were consistently nega-
tive for cytokeratins and EMA.76,106,123 The two components of the tumor can be distinguished
4. Although chondroid chordomas continue to be on gross examination. The conventional chordomatous
reported in the literature as distinct entities, component has the typical gross appearance of a gray-tan
the clinical significance of precise differentiation myxomatous mass and is present within or at the periph-
between chordoma and chondroid chordoma ery of a fleshy sarcomatous component. The two compo-
appears to be less significant than originally pro- nents are typically well demarcated, but a small island of
posed. Moreover, recent molecular profiling reports chordoma can be embedded in the sarcomatous compo-
suggest that cartilage-appearing components of nent. Hemorrhage and necrosis are frequently present
chondroid chordoma are notochordal in nature. within the dedifferentiated component.
In summary, after more than four decades of debate the
identification of this specific subtype of chordoma con- Microscopic Findings
tinues to be controversial.
The hallmark of dedifferentiated chordoma is the coex-
Clinical Significance istence of both conventional chordoma and high-grade
sarcoma (Figs 18-37 to 18-39).132,136-140,142,143-145 Typically,
In the original report,112 mean survival of patients with there is a sharp separation between conventional chor-
chondroid chordomas was about 6 to 10 years (3 times) doma and the dedifferentiated component. Sometimes,
longer than the mean survival for conventional chordo- small distinct nodules of chordoma are embedded in
mas. However, there has been no report in which a high-grade sarcoma. The dedifferentiated compo-
systematic follow-up study was performed on immuno- nent typically exhibits features of high-grade spindle-
histochemically classified cases. Therefore it is necessary cell or pleomorphic sarcoma with malignant fibrous
to investigate the long-range treatment results in the histiocytoma-like features. Similar to other dedifferenti-
major three categories of chondroid and cartilaginous ated tumors, the sarcomatous component of dediffer-
tumors of the central axis to evaluate the clinical signifi- entiated chordoma may show a phenotypic switch to a
cance of chordoma with chondroid differentiation (so- rhabdomyoblastic lineage and may exhibit features of
called chondroid chordoma). rhabdomyosarcoma.134

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1211

B
FIGURE 18-37  ■  Dedifferentiated chordoma: microscopic features. A, Interface of two distinct tumor patterns in dedifferentiated
chordoma. Conventional chordoma (left) directly abuts highly cellular, pleomorphic spindle-cell sarcoma (right). B, Higher magnifi-
cation of high-grade sarcomatous component shown in A. Inset (left), Classic microscopic appearance of conventional chordoma
component. Inset (right), Whole-mount photomicrograph shows nodules of chordoma (arrows) within cellular sarcomatoid compo-
nents. (A and right Inset, ×50; B, ×80; left Inset, ×200) (A, B, and Insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1212 18  Chordoma and Related Lesions

B
FIGURE 18-38  ■  Dedifferentiated chordoma: microscopic features. A, Sharp demarcation of two distinct tumor components: conven-
tional chordoma (left) and high-grade spindle-cell sarcoma (right). B, High magnification of dedifferentiated component. Inset, High
magnification of conventional chordoma component. (A, ×50; B and Inset, ×150) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1213

A B

C D
FIGURE 18-39  ■  Dedifferentiated chordoma: microscopic features. A, Island of conventional chordoma within high-grade sarcoma.
B, High magnification showing area of conventional chordoma in A. C, High-grade sarcomatous component. D, High magnification
showing nuclear features of high-grade sarcoma in C. Inset, Higher magnification of physaliphorous cell in B. (A and C, ×100; B and
D, ×400; Inset, ×600) (A-D and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1214 18  Chordoma and Related Lesions

11. Ulich TR, Mirra JM: Ecchordosis physaliphora vertebralis. Clin


Differential Diagnosis Orthop 163:282–289, 1982.
12. Wyatt RB, Schochet SS, Jr, McCormick WF: Ecchordosis phys-
Conventional chordoma may show prominent nuclear aliphora: an electron microscopic study. J Neurosurg 34:672–677,
atypia focally or diffusely or may mimic an epithelial 1971.
neoplasm. These features should not be confused with
Notochordal Rests and Benign Notochordal Tumors
dedifferentiation.
13. Amer HZ, Hameed M: Intraosseous benign notochordal cell
Dedifferentiated chondrosarcoma has very similar archi- tumor. Arch Pathol Lab Med 134:283–288, 2010.
tectural features to conventional chordomas (i.e., the 14. Darby AJ, Cassar-Pullicino VN, McCall IW, et al: Vertebral intra-
clearly separated components). The difference is in a osseous chordoma or giant notochordal rest? Skeletal Radiol 28:
low-grade precursor lesion that has features of conven- 342–346, 1999.
tional chordoma and that coexpresses S-100 protein and 15. Ho KL: Ecchorodosis physaliphora and chordoma: a comparative
ultrastructural study. Clin Neuropathol 4:77–86, 1985.
epithelial markers. 16. Iorgulescu JB, Laufer I, Hameed M, et al: Benign notochordal cell
Because the dedifferentiation in chordoma has fea- tumors of the spine: natural history of 8 patients with histologi-
tures that mimic malignant fibrous histiocytoma, small cally confirmed lesions. Neurosurgery 73:411–416, 2013.
biopsy samples may be misinterpreted as malignant 17. Klebs E: Ein Fall von Ecchondrosis spheno-occipitalis amylacea.
Virchows Arch 31:396, 1864.
fibrous histiocytoma when the chordomatous elements 18. Kyriakos M, Totty WG, Lenke LG: Giant vertebral notochordal
are not found. Attention to the radiographic findings and rest. A lesion distinct from chordoma: discussion of an evolving
location of the lesion in the sites typical for chordoma concept. Am J Surg Pathol 27(3):396–406, 2003.
are essential in guiding the selection of a biopsy site that 19. Lushka H: Ueber die Gallertgeschwülste des Clivus Blumen-
would include both possible tumor components. bachii. Virchows Arch 11:8–11, 1857.
20. Macdonald RL, Cusimano MD, Deck JHN, et al: Cerebrospinal
fluid fistula secondary to ecchordosis physaliphora. Neurosurgery
Treatment and Behavior 26:515–519, 1990.
21. Mirra JM: Giant notochordal hamartoma of intraosseous origin:
Dedifferentiation can occur in primary or recurrent chor- a newly reported benign entity to be distinguished from chor-
doma. Report of two cases. Skeletal Radiol 30:698–709, 2001.
domas.140,142 The behavior of dedifferentiated chordoma 22. Muller H: Ueber das Vorkommen von Resten der Chorda
is similar to that of dedifferentiated chondrosarcoma— dorsalis bei Menschen nach der Geburt und uber-ihr Verhaltnis
the tumor is highly lethal with almost no survivors 1 zu den Gallert-geschwulsten am Clivus. Ztschr f rat Med 2:202,
to 2 years after diagnosis. After diagnosis, widespread 1858.
metastases are the rule, and patients do not seem to 23. Ng SH, Ko SF, Wan YL, et al: Cervical ecchordosis physaliphora:
CT and MR features. Brit J Radiol 71:329–331, 1998.
benefit significantly from chemotherapy or radiotherapy. 24. Ribbert H: Ubert die Eccordrosis physaliphora spheno-occipitalis.
However, in some cases, a transient response to aggres- Zentralb Allg Path Path Anat 5:457–461, 1894.
sive chemotherapy protocols has been reported.135,139 The 25. Salisbury JR: The pathology of the human notochord. J Pathol
relationship between irradiation and the transformation 171:253–255, 1993.
26. Shen J, Li CD, Yang HL, et al: Classic chordoma coexisting with
of conventional chordoma into a high-grade sarcoma has benign notochordal cell rest demonstrating different immunohis-
been postulated. togical expression patterns of brachyury and galectin-3. J Clin
Neurosci 18:96–99, 2011.
REFERENCES 27. Virchow R: Untersuchungen ueber die Entwicklung des Schaedel-
grundes, Berlin, 1857, G Rimer.
Introduction 28. Wills RA: Pathology of tumours, ed 4, New York, 1967, Appleton-
1. Cheyette BNR, Waxman JS, Miller JR, et al: Dapper, a disheveled- Century-Crofts, pp 937–943.
associated antagonist of β-Catenin and JNK signaling, is required 29. Wolfe JT, Scheithauer BW: “Intradural chordoma” or “giant
for notochord formation. Dev Cell 2:449–461, 2002. ecchordosis physaliphora”? Report of two cases. Clin Neuropathol
2. Horten BC, Montague SR: Human ecchordosis physaliphora and 6:98–103, 1987.
chick embryonic notochord: a comparative electron microscopic 30. Yamaguchi T, Yamato M, Saotome K: First histologically con-
study. Virchows Arch [Pathol Anat] 371:295–303, 1976. firmed case of a classic chordoma arising in a precursor benign
3. Horwitz T: Chordal ectopia and its possible relationship to chor- notochordal lesion: differential diagnosis of benign and malignant
doma. Arch Pathol 31:354–362, 1941. notochordal lesions. Skeletal Radiol 31:413–418, 2002.
4. Horwitz T: The human notochord: a study of its development and 31. Zenker FA: Ueber die Gallertgeschwülste des Clivus Blumen-
regression—variations and pathologic derivative chordoma, Indianapo- bachii. Virchows Arch 12:108–110, 1857.
lis, 1977, limited private printing.
5. Kim J, Shim S, Choi SC, et al: A putative xenopus Rho-GTPase Chordoma
activating protein (XrGAP) gene is expressed in the notochord 32. Abenoza P, Sibley RK: Chordoma: an immunohistological study.
and brain during the early embryogenesis. Gene Expr Patterns Hum Pathol 17:744–747, 1986.
3:219–223, 2003. 33. Agarwal S, Agarwal T, Agarwal R, et al: Fine needle aspiration of
6. Kuroda H, Inui M, Sugimoto K, et al: Axial protocadherin is bone tumors. Cancer Detect Prev 24:602–609, 2000.
a mediator of prenotochord cell sorting in xenopus. Develop Biol 34. Anderson WB, Meyers HI: Multicentric chordoma: report of a
244:267–277, 2002. case. Cancer 21:126–128, 1968.
7. Muller F, O’Rahilly R: The prechordal plate, the rostral end of 35. Ariel IM, Verdu C: Chordoma: an analysis of 20 cases treated over
the notochord and nearby median features in staged human a 20 year span. J Surg Oncol 7:27–44, 1975.
embryos. Cells Tissues Organs 173:1–20, 2003. 36. Ashwood N, Hoskin PJ, Saunders MI: Metastatic chordoma:
8. Salisbury JR, Deverell MH, Cookson MJ, et al: Three-dimensional pattern of spread and response to chemotherapy. Clin Oncol (R Coll
reconstruction of human embryonic notochords: clue to the Radiol) 6:341–342, 1994.
pathogenesis of chordoma. J Pathol 171:59–62, 1993. 37. Auger M, Raney B, Callender D, et al: Metastatic intracranial
9. Salisbury JR: The pathology of the human notochord. J Pathol chordoma in a child with massive pulmonary tumor emboli.
171:253–255, 1993. Pediatr Pathol 14:763–770, 1994.
10. Stemple DL: Structure and function of the notochord: an essential 38. Ayala AG, Ro JY, Fanning CV, et al: Core needle biopsy and fine-
organ for chordate development. Development 132:2503–2512, needle aspiration in the diagnosis of bone and soft-tissue lesions.
2005. Hematol Oncol Clin North Am 9:633–651, 1995.

ERRNVPHGLFRVRUJ
18  Chordoma and Related Lesions 1215

39. Azzarelli A, Quagliuolo V, Cerasoli S, et al: Chordoma: natural 68. Kay PA, Nascimento AG, Unni KK, et al: Chordoma. Cytomor-
history and treatment results in 33 cases. J Surg Oncol 37:185–191, phologic findings in 14 cases diagnosed by fine needle aspiration.
1988. Acta Cytol 47:202–208, 2003.
40. Baker HW, Coley BL: Chordoma of lumbar vertebra. J Bone Joint 69. Kay S, Schatzki PF: Ultrastructural observations of a chordoma
Surg 35A:403–408, 1953. arising in the clivus. Hum Pathol 3:403–413, 1972.
41. Barresi V, Ieni A, Branca G, et al: Brachyury: a diagnostic marker 70. Kelley MJ, Korczak JF, Sheridan E, et al: Familial chordoma, a
for the differential diagnosis of chordoma and hemangioblastoma tumor of notochordal remnants, is linked to chromosome 7q33.
versus neoplastic histological mimickers. Dis Markers 514753, Am J Hum Genet 69:454–460, 2001.
2014. 71. Klingler L, Shooks J, Fiedler PN, et al: Microsatellite instability
42. Benk V, Liebsch NJ, Munzenrider JE, et al: Base of skull and in sacral chordoma. J Surg Oncol 73:100–103, 2000.
cervical spine chordomas in children treated by high-dose irradia- 72. Krol G, Sundaresan N, Deck M: Computed tomography of axial
tion. Int J Radiat Oncol Biol Phys 31:577–581, 1995. chordomas. J Comput Assist Tomogr 7:286–289, 1983.
43. Bergh P, Kindblom LG, Gunterberg B, et al: Prognostic factors 73. Kubota T, Sato K, Kabuto M, et al: Immunohistochemical and
in chordoma of the sacrum and mobile spine. Cancer 88:2122– ultrastructural study of skull base chordomas. Noshuyo Byori
2134, 2000. 11:23–28, 1994.
44. Bervin S, Zurakowski D, Mankin HJ, et al: Clinical outcome in 74. Larson TC, III, Houser OW, Laws ER, Jr: Imaging of cranial
chordoma. Utility of flow cytometry in DNA determination. Spine chordomas. Mayo Clin Proc 62:886–893, 1987.
27:374–379, 2002. 75. Leproux F, de Toffol B, Aesch B, et al: MRI of cranial chordomas:
45. Bridge JA, Pickering D, Neff JR: Cytogenetic and molecular cyto- the value of gadolinium. Neuroradiol 35:543–545, 1993.
genetic analysis of sacral chordoma. Cancer Genet Cytogenet 75:23– 76. Meis JM, Giraldo AA: Chordoma: an immunohistochemical study
25, 1994. of 20 cases. Arch Pathol Lab Med 112:553–556, 1988.
46. Brondi LA, Podrecca S, Catoldo I: Chordoma: study of 245 cases 77. Mertens F, Kreicbergs A, Rydholm A, et al: Clonal chromosome
verified at the Istituto Nazionale dei Tumori de Milano. Rev Paul aberrations in three sacral chordomas. Cancer Genet Cytogenet
Med 95:71–73, 1980. 73:147–151, 1994.
47. Campbell WM, McDonald TJ, Unni KK, et al: Nasal and para- 78. Miller J: Relationship of the notochord to the cartilage of the skull
nasal presentations of chordomas. Laryngoscope 90:612–618, 1980. and its correlation with the location and frequencies of chordo-
48. Cancilla P, Morecki R, Hurwitt ES: Fine structure of a recurrent mata. Anat Rec 112:362–365, 1952.
chordoma. Arch Neurol 11:289–295, 1964. 79. Mori K, Chano T, Kushima R, et al: Expression of E-cadherin in
49. Chalmers J, Coulson WF: A metastasing chordoma. J Bone Joint chordomas: diagnostic marker and possible role of tumor cell
Surg Br 42:556–559, 1960. affinity. Virchows Arch 440:123–127, 2002.
50. Chalmers J, Heard BE: A metastasing chordoma: a further note. 80. Murad TM, Murthy MS: Ultrastructure of a chordoma. Cancer
J Bone Joint Surg Br 54:526–529, 1972. 25:1204–1215, 1970.
51. Chambers PW, Schwinn CP: Chordoma: a clinicopathologic 81. Murali R, Rovit RL, Benjamin MV: Chordoma of the cervical
study of metastasis. Am J Clin Pathol 72:765–776, 1979. spine. Neurosurgery 9:253–256, 1981.
52. Coffin CM, Swanson PE, Wick MR, et al: Chordoma in child- 82. Nibu Y, Jose-Edwards DS, Di Gregorio A: From notochord for-
hood and adolescence: a clinicopathologic analysis of 12 cases. mation to hereditary chordoma: the many roles of brachyury.
Arch Pathol Lab Med 117:927–933, 1993. Biomed Res Int 826435, 2013.
53. Coraddu M, Floris F, Nurchi GC, et al: Chordoma of the cervical 83. O’Connell JX, Renard LG, Liebsch NJ, et al: Base of skull chor-
spine: case report. J Neurol Sci 38:51–53, 1994. doma: a correlative study of histologic and clinical features of 62
54. Crapanzano JP, Ali SZ, Ginsberg MS, et al: Chordoma: a cyto- cases. Cancer 74:2261–2267, 1994.
logic study with histologic and radiologic correlation. Cancer 84. Pascaud JL, Dudognon P, Vigneu P, et al: Chordoma of the base
(Cancer Cytopathology) 93:40–51, 2001. of the skull and tomodensitometry. Ann Radiol (Paris) 26:513–514,
55. Dahlin DC, MacCarty CS: Chordoma: a study of 59 cases. Cancer 1983.
5:1170–1178, 1952. 85. Pena CE, Horvat BL, Fisher ER: The ultrastructure of chordoma.
56. Dalpra L, Malgara R, Miozzo M, et al: First cytogenetic study of Am J Clin Pathol 53:544–551, 1970.
a recurrent familial chordoma of the clivus. Int J Cancer 81:24–30, 86. Peramezza C, Cellini A, Berardi P, et al: Chordoma with multiple
1999. skin metastases. Dermatology 186:266–268, 1993.
57. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210, 87. Pluot M, Bernard MH, Rousseaux P, et al: Two cases of sellar
1995. chordomas: ultrastructural and histochemical study. Semaine des
58. Eriksson B, Gunterberg B, Kindblom LG: Chordoma: a clinico- Hospitaux 57:579–585, 1981.
pathologic and prognostic study of a Swedish national series. Acta 88. Presneau N, Shalaby A, Ye H, et al: Role of the transcription
Orthop Scand 52:49–58, 1981. factor T (brachyury) in the pathogenesis of sporadic chordoma:
59. Erlandson RA, Tandler B, Lieberman PH, et al: Ultrastructure of a genetic and functional-based study. J Pathol 2223:327–335,
human chordoma. Cancer Res 28:2115–2125, 1968. 2011.
60. Forsyth PA, Cascino TL, Shaw EG, et al: Intracranial chordomas: 89. Probst En, Zanella FE, Vortmeyer AO: Congenital clivus chor-
a clinicopathological and prognostic study of 51 cases. J Neurosurg doma. Am J Neuroradiol 14:537–539, 1993.
78:741–747, 1993. 90. Richter HJ, Batsakis JG, Boles R: Chordomas: nasopharyngeal
61. Friedmann I, Harrison DFN, Bird ES: The fine structure of presentation and atypical long survival. Ann Otol Rhinol Laryngol
chordoma with particular reference to the physaliphorous cell. 84:327–332, 1975.
J Clin Pathol 15:116–125, 1962. 91. Rosenthal DI, Scott JA, Mankin HJ, et al: Sacrococcygeal chor-
62. Gessaga EC, Mair WG, Grant DN: Ultrastructure of a sacrococ- doma: magnetic resonance imaging and computed tomography.
cygeal chordoma. Acta Neuropathol 25:27–35, 1973. AJR Am J Roentgenol 145:143–147, 1985.
63. Gottschalk D, Fehn M, Patt S, et al: Matrix gene expression 92. Rutherfoord GS, Davies AG: Chordomas: ultrastructure and
analysis and cellular phenotyping in chordoma reveals focal dif- immunohistochemistry: a report on the examination of six cases.
ferentiation pattern of neoplastic cells mimicking nucleus pulpo- Histopathology 11:775–787, 1987.
sus development. Am J Pathol 158:1571–1578, 2001. 93. Sangoi AR, Karamchandani J, Lane B, et al: Specificity of brachy-
64. Haeckel C, Krueger S, Kuester D, et al: Expression of cathepsin ury in the distinction of chordoma from clear cell renal cell car-
K in chordoma. Hum Pathol 31:834–840, 2000. cinoma and germ cell tumors: a study of 305 cases. Mod Pathol
65. Heaton JM, Turner DR: Reflections on notochordal differentia- 24:425–429, 2011.
tion arising from a study of chordomas. Histopathology 9:543–550, 94. Scheil-Bertram S, Kappler R, von Baer A, et al: Molecular profil-
1985. ing of chordoma. Int J Oncol 44:1041–1055, 2014.
66. Hu Y, Mintz A, Shah SR, et al: The FGFR/MEK/ERK/brachyury 95. Shen J, Shi Q, Lu J, et al: Histological study of chordoma origin
pathway is critical for chordoma cell growth and survival. Carci- from fetal notochordal cell rests. Spine 38:2165–2170, 2013.
nogenesis 35:1491–1499, 2014. 96. Smith J, Ludwig RL, Marcove RC: Sacrococcygeal chordoma: a
67. Kaiser TE, Pritchard DJ, Unni KK: Clinicopathologic study of clinicoradiological study of 60 patients. Skeletal Radiol 16:37–44,
sacrococcygeal chordoma. Cancer 54:2574–2578, 1984. 1987.

ERRNVPHGLFRVRUJ
1216 18  Chordoma and Related Lesions

97. Spjut HJ, Luse SA: Chordoma: an electron microscopic study. 123. Moriki T, Takahashi T, Wada M, et al: Chondroid chordoma:
Cancer 17:643–656, 1964. fine-needle aspiration cytology with histopathological, immuno-
98. Stephens GC, Schwartz HS: Lumbosacral chordoma resection: histochemical, and ultrastructural study of two cases. Diagn Cyto-
image integration and surgical planning. J Surg Oncol 54:226–232, pathol 21:335–339, 1999.
1993. 124. Persson S, Kindblom LG, Angervall L: Classical and chondroid
99. Szadowska A, Giryn I: Electron-microscopic studies of the case of chordoma: a light-microscopic, histochemical, ultrastructural and
chordoma. Acta Med Pol 20:461–462, 1979. immunohistochemical analysis of the various cell types. Pathol Res
100. Tallini G, Dorfman H, Brys P, et al: Correlation between clinico- Pract 187:828–838, 1991.
pathological features and karyotype in 100 cartilaginous and chor- 125. Romeo S, Hogendoorn PC: Brachyury and chordoma: the
doid tumours. A report from the CHromosomes And MorPhology chondroid-chordoid dilemma resolved? J Pathol 209(143):146,
(CHAMP) collaborative study group. J Pathol 196:194–203, 2002. 2006.
101. Taylor JR: Persistence of the notochordal canal in vertebrae. 126. Rutherford GS, Davies AG: Chordomas: ultrastructure and
J Anat 111:211–217, 1972. immunohistochemistry: a report based on the examination of six
102. Thiery JP, Mazabraud A, Mignot J, et al: Electron microscopic cases. Histopathology 11:775–787, 1987.
study of a sacral chordoma: characterization of various develop- 127. Salisbury JR: Demonstration of cytokeratins and an epithelial
ment stages of the tumoral cells. Ann Anat Pathol 22:193–204, membrane antigen in chondroid chordoma. J Pathol 153:37–40,
1977. 1987.
103. Utne JR, Pugh DG: The roentgenologic aspects of chordoma. Am 128. Solares CA, Umana E, Lopez EM, et al: Chondroid chordoma of
J Roentgenol Radium Ther Nucl Med 74:593–608, 1955. the cervical spine. Acta Neurochir 144:1059–1060, 2002.
104. Vanel D, Rebibo G, Hales H, et al: Contribution of computed 129. Vujovic S, Henderson S, Presneau N, et al: Brachyury, a crucial
tomography in six sacrococcygeal chordomas. Eur J Radiol 5:87– regulator of notochordal development, is a novel biomarker for
90, 1985. chordomas. J Pathol 209:157–165, 2006.
105. Watkins L, Khudados ES, Kaleoglu M, et al: Skull base chordo- 130. Valderrama E, Kahn LB, Lipper S, et al: Chondroid chordoma:
mas: a review of 38 patients, 1958-88. Br J Neurosurg 7:241–248, electron-microscopic study of two cases. Am J Surg Pathol 7:625–
1993. 632, 1983.
106. Weber AL, Liebsch NJ, Sanchez R: Sweriduk ST Jr: Chordomas 131. Walker WP, Landas SK, Bromley CM, et al: Immunohistochemi-
of the skull base: radiologic and clinical evaluation. Neuroimaging cal distinction of classic and chondroid chordomas. Mod Pathol
Clin North Am 4:515–527, 1994. 4:661–666, 1991.
107. Yakkioui Y, van Overbeeke JJ, Santegoeds R, et al: The origin of 132. Wojno KJ, Hruban RH, Garin-Chesa P, et al: Chondroid chor-
chordoma. Biochim Biophys Acta [epub ahead of print]. 2014. domas and low-grade chondrosarcomas of the craniospinal axis:
an immunohistochemical analysis of 17 cases. Am J Surg Pathol
Chondroid Chordoma 16:1144–1152, 1992.
108. Bottles K, Beckstead JH: Enzyme histochemical characterization
of chordomas. Am J Surg Pathol 8:443–447, 1984. Dedifferentiated Chordoma
109. Brooks JJ, LiVolsi VA, Trojanowski JQ: Does chondroid chor- 133. Belza MG, Urich H: Chordoma and malignant fibrous histiocy-
doma exist? Acta Neuropathol 72:229–235, 1987. toma: evidence for transformation. Cancer 58:1082–1087, 1986.
110. Brooks JJ, Trojanowski JQ, LiVolsi VA: Chondroid chordoma: a 134. Bisceglia M, D’Angelo VA, Guglielmi G, et al: Dedifferentiated
low-grade chondrosarcoma and its differential diagnosis. Curr Top chordoma of the thoracic spine with rhabdomyosarcomatous dif-
Pathol 80:165–181, 1989. ferentiation. Report of a case and review of the literature. Ann
111. Chu TA: Chondroid chordoma of the sacrococcygeal region. Arch Diagn Pathol 11:262–273, 2007.
Pathol Lab Med 111:861–864, 1987. 135. Burrow J, Le F, Stewart MJ: Malignant spheno-occipital chor-
112. Coltrera MD, Goorge PB, Harrist TJ, et al: Chondrosarcoma of doma. J Neurol Psychopath 4:205–217, 1923.
the temporal bone: diagnosis and treatment of 13 cases and review 136. Choi YJ, Kim TS: Malignant fibrous histiocytoma in chordoma:
of the literature. Cancer 58:2689–2696, 1986. immunohistochemical evidence of transformation from chordoma
113. Hasegawa M, Nishijima W, Watanabe I, et al: Primary chondroid to malignant fibrous histiocytoma. Yonsei Med J 35:239–243, 1994.
chordoma arising from the base of the temporal bone: a 10-year 137. Fleming GF, Heimann PS, Stephens JK, et al: Dedifferentiated
post-operative follow-up. J Laryngol Otol 99:485–489, 1985. chordoma: response to aggressive chemotherapy in two cases.
114. Heffelfinger MJ, Dahlin DC, MacCarty CS, et al: Chordomas and Cancer 72:714–718, 1993.
cartilaginous tumors at the skull base. Cancer 32:410–420, 1973. 138. Halpern J, Kopolovic J, Catane R: Malignant fibrous histiocytoma
115. Horn KD, Fowler JC, Carrau R, et al: Cytokeratin immunophe- developing in irradiated sacral chordoma. Cancer 53:2661–2662,
notyping of an unusual cervical vertebral chordoma with extensive 1984.
chondroid foci and perilaryngeal recurrence: a case report with 139. Hanna SA, Tirabosco R, Amin A, et al: Dedifferentiated chor-
review of the literature. Am J Otolaryngol 22:428–434, 2001. doma: a report of four cases arising ‘de novo. J Bone Joint Surg Br
116. Ishida T, Dorfman HD: Chondroid chordoma versus low-grade 90:652–656, 2008.
chondrosarcoma of the base of the skull: can immunohistochem- 140. Hruban RH, Traganos F, Reuter VE, et al: Chordomas with
istry resolve the controversy? J Neurooncol 18:199–206, 1994. malignant spindle cell components: a DNA flow cytometric and
117. Kay PA, Nascimento AG, Unni KK, et al: Chordoma. Cytomor- immunohistochemical study with histogenetic implications. Am J
phologic findings in 14 cases diagnosed by fine needle aspiration. Pathol 137:435–447, 1990.
Acta Cytol 47:202–208, 2003. 141. Knechtges TC: Sacrococcygeal chordoma with sarcomatous fea-
118. Mierau GW, Weeks DA: Chondroid chordoma. Ultrastruct Pathol tures (spindle cell metaplasia). Am J Clin Pathol 53:612–616,
11:731–737, 1987. 1970.
119. Miettinen M, Lehto VP, Dahl D, et al: Differential diagnosis of 142. Makek M, Leu HJ: Malignant fibrous histiocytoma arising in a
chordoma, chondroid, and ependymal tumors as aided by anti- recurrent chordoma: case report and electron microscopic find-
intermediate filament antibodies. Am J Pathol 112:160–169, 1983. ings. Virchows Arch [Pathol Anat] 397:241–250, 1982.
120. Miettinen M: Chordoma: antibodies to epithelial membrane 143. Meis JM, Raymond AK, Evans HL, et al: Dedifferentiated” chor-
antigen and carcinoembryonic antigen in differential diagnosis. doma: a clinicopathologic and immunohistochemical study of
Arch Pathol Lab Med 108:891–892, 1984. three cases. Am J Surg Pathol 11:516–525, 1987.
121. Mitchell A, Scheithauer BW, Unni KK, et al: Chordoma and 144. Miettinen M, Lehto VP, Virtanen I: Malignant fibrous histocy-
chondroid neoplasms of the spheno-occiput: an immunohisto- toma within a recurrent chordoma: a light microscopic and immu-
chemical study of 41 cases with prognostic and nosologic implica- nohistochemical study. Am J Clin Pathol 82:738–743, 1984.
tions. Cancer 72:2943–2949, 1993. 145. Ng WK, Tang V: Crush preparation findings of “sarcomatoid”
122. Mori K, Chano T, Kushima R, et al: Expression of E-cadherin in chordoma of the sacrum: report of a case with histologic, immu-
chordomas: diagnostic marker and possible role of tumor cell nohistochemical, and ultrastructural correlation. Diagn Cytopathol
affinity. Virchows Arch 440:123–127, 2002. 25:406–410, 2001.

ERRNVPHGLFRVRUJ
C H A P T E R 1 9 

Metastatic Tumors in Bone


John D. Reith, MD

CHAPTER OUTLINE

BIOLOGY OF METASTASES Carcinoma of the Thyroid


GENERAL FEATURES OF SKELETAL METASTASES Carcinoma of the Pancreas

SKELETAL METASTASES IN SELECTED COMMON METASTATIC MELANOMA


NEOPLASMS SKELETAL METASTASES IN SELECTED PEDIATRIC
Carcinoma of the Lung TUMORS
Gastrointestinal Carcinoma Rhabdomyosarcoma
Carcinoma of the Breast Neuroblastoma
TUMORS WITH A UNIQUE PROPENSITY FOR Clear Cell Sarcoma of the Kidney
SKELETAL METASTASES
Carcinoma of the Prostate
Carcinoma of the Kidney

Bone is one of the most common anatomic sites for two biologic hallmarks of malignancy. To successfully
metastases, along with the lungs and liver, and metastases metastasize, tumor cells must (1) proliferate within the
are the most commonly diagnosed neoplasms encoun- host organ and gain the ability to invade the surrounding
tered in the skeleton. Although most metastases encoun- tissue and matrix; (2) invade blood vessels, whether
tered in the adult skeleton are either carcinoma or induced by angiogenic properties of the tumor cells or
melanoma, sarcomas may also metastasize to bone, par- normal vessels at the site; (3) enter the circulatory system
ticularly in the pediatric population. The general surgical and survive both intracellular and extracellular influences
pathologist is far more likely to encounter a skeletal focus attempting to destroy them; (4) adhere to and migrate
of metastatic cancer than any of the primary malignant across sinusoidal walls at distant sites; and (5) form depos-
bone tumors. Patients frequently present with signs and its capable of surviving in the new environment10 (Fig.
symptoms related to metastatic bone lesions before a 19-1, A). The basic biology, molecular mechanisms, and
primary tumor has come to clinical attention, and severe genetic and epigenetic changes that drive these steps are
pain or a pathologic fracture is commonly the presenting extremely complex and probably differ depending on the
complaint. Despite the often extensive immunohisto- source of the metastasis and specific type of tumor.
chemical evaluation of metastatic deposits, a primary Before metastasizing, tumor cells prepare distant sites
source can be elusive and sometimes never located. The to receive metastatic deposits by creating a premetastatic
incidence and localization, radiologic characteristics, pat- niche.6,9 By modulating hematopoietic cells to increase
terns of spread, and histologic and immunohistochemical fibronectin production and by directly producing factors
features of various metastatic lesions in bone are consid- that render the bone microenvironment receptive, tumors
ered in some detail in this chapter. Because fine-needle are able to prepare the bone for metastases. Such factors
aspiration cytology plays a major role in diagnosing met- include osteopontin,2 matrix metalloproteinases, and
astatic lesions affecting the skeleton, we include in this parathyroid hormone-related protein (PTHrP).5 Once
chapter a brief description of cytologic features of meta- tumor cells have become disseminated, the local environ-
static tumors in bone. First, however, it is important to ment within the metastatic site, or metastatic niche, is
discuss some of the newer information on the biology of important in determining whether tumor cells are able to
tumor metastasis that has been derived from the methods survive.12,16
of molecular pathology. This information is important After induction neovascularization by tumor cells in
for many reasons, including the understanding of the the primary organ, the tumor cells invade into the vessels
pharmacologic strategies for treating bone metastases. and are embolized to distant sites. Those tumor cells that
are able to survive in transit become lodged in capillaries
within a secondary site such as bone, adhere to the endo-
BIOLOGY OF METASTASES thelium, and eventually extravasate through the capillary
wall and into the stroma, where they proliferate and ulti-
The ability of tumor cells to invade within their host mately form clinically significant masses. Tumor cells
organ and their ability to metastasize to distant sites are home in to bone via the same protein interactions that
1217
ERRNVPHGLFRVRUJ
1218 19  Metastatic Tumors in Bone

A B
FIGURE 19-1  ■  Major biologic phenomena associated with tumor progression and metastasis. A, To metastasize, tumor cells must
gain several unique biologic properties such as invasive growth, induction of vascular growth, vascular invasion, adherence to
endothelial cells or thrombosis of peripheral sinusoids, continuation of invasive growth with extravasation, and formation of primary
and secondary metastatic foci. Although biologic phenomena associated with tumor progression and metastasis are usually present
as sequence of orderly events, in reality tumor cells may develop these properties in a virtually haphazard pattern. Moreover, not
all tumor cells may develop all properties listed; some cell clones may subspecialize (e.g., have high invasive properties), whereas
other cells may take advantage of stromal and vascular destruction and metastasize. B, Summary of molecular changes associated
with tumor cell interaction with various stromal components essential for invasive growth and metastasis. See text for more specific
description.

hematopoietic stem cells use, relying on integrins, che- The tumor cells must invade and destroy the extracel-
mokines, bone morphogenetic proteins, and osteopontin, lular matrix at several stages of the process to metastasize.
among others, to settle in bone.18 Thus the interaction of tumor cells with extracellular
An important element of metastasis is the ability of matrix seems to be one of the major mechanisms of the
tumor cells to both adhere to and degrade the extracel- metastatic cascade.3,17 The extracellular matrix can be
lular matrix, invade blood vessels, and evade cell death, divided into two main components: a basement mem-
both within their primary organ and within bone.3,17 This brane and interstitial connective tissue. The basement
is achieved through a complex interaction between inte- membrane is a sheetlike structure that separates epithelial
grins; extracellular matrix components such as type I col- and endothelial cells from the interstitial matrix. It con-
lagen and fibronectin; and proteolytic enzymes such as tains type IV collagen and several distinct noncollagenous
matrix metalloproteinases (Fig. 19-2). Integrins, a super- proteins, such as laminin, heparan, and sulfated proteo-
family of transmembrane receptors that modulate cell-to- glycan. Laminin, together with collagen IV, is a major
cell and cell-to-matrix interactions by binding with a component of the basement membrane and serves as an
variety of ligands, play a key role in skeletal metastases integration unit of the structure. Laminin also plays a
on many levels, matrix and blood vessel invasion, osteo- major role as a cellular adhesion molecule. This large
clast signaling, neovascularization, and colonization of complex molecule has three short arms containing col-
bone.4,8,15,20 CD44, a hyaluronic acid receptor that has lagen binding sites and a single long arm with a heparan
been studied in a wide variety of tumors, appears to binding site. The central region of the molecule, where
be extremely important in the early development of all four arms are connected in a cross-shaped structure,
metastases.19 contains the laminin receptor binding site for cells.

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1219

A B
FIGURE 19-2  ■  Regulation of bone resorption and bone formation. A, Both systemic factors and locally acting factors induce the
formation and activity of osteoclasts. Systemic hormones such as parathyroid hormone, 1,25-dihydroxyvitamin D3, and thyroxine
(T4), stimulate the formulation of osteoclasts by inducing the expression of receptor activator of nuclear factor-κB ligand (RANKL)
on marrow stromal cells and osteoblasts. In addition, osteoblasts produce interleukin-6, interleukin-1, prostaglandins, and colony-
stimulating factors (CSFs), which induce the formation of osteoclasts. Accessory cells such as T cells can produce cytokines that
can inhibit the formation of osteoclasts, such as interleukin-4, interleukin-18 and interferon-γ. TGF-β denotes transforming growth
factor-β. Plus signs indicate stimulation, and minus signs inhibition. B, Both systemic factors and locally acting factors can enhance
the proliferation and differentiation of osteoblasts. These include parathyroid hormone, prostaglandins, and cytokines as well as
growth factors such as platelet-derived growth factor (PDGF) produced by lymphocytes. In addition, bone matrix is a major source
of growth factors, which can enhance the proliferation and differentiation of osteoblasts. These include the bone morphogenetic
proteins (BMPs), TGF-β, insulin-like growth factors (IGFs), and fibroblast growth factors (FGFs). Corticosteroids can induce apoptosis
of osteoblasts and block bone formation. (Modified and reprinted with permission from Roodman GD: Mechanisms of bone metastasis.
N Engl J Med 350:1655–1664, 2004.)

Recent advancements have elucidated some additional To invade the intercellular matrix and to penetrate the
components of the so-called cell-to-cell and cell-to- vascular channels, the tumor cells have to follow three
matrix adhesion system and its potential role in invasive general steps:
growth.8,13,15,17,20 A new class of transmembrane cell adhe- 1. They must maintain some degree of their attach-
sion receptors (integrins) that has a unique structure and ment to the matrix components (mainly laminin
ability to bind fibronectin and laminin has been identi- and fibronectin).
fied.13,15,17,20 These receptors integrate the extracellular 2. Degradation of the matrix components must
matrix anchorage with an intracytoplasmic cytoskeleton proceed through the action of proteolytic enzymes.
and provide a pathway for signal transduction. 3. Tumor cells must continue to migrate into the
Collagen I and III are the most prominent compo- degraded area of the extracellular matrix.
nents of the interstitial extracellular matrix.3,17 Proteogly- Major molecular phenomena associated with the
cans fill the interstitial space among the collagen fibers, development of the metastatic phenotype are summa-
and their major role is to retain water and to maintain rized in Figure 19-1, B.
the shape and volume of the interstitial space. Fibronec- Paradoxically, it has been shown that tumor cells
tin is one of the major noncollagenous components of the exhibiting increased levels of laminin and fibronectin
interstitial matrix. It is biochemically distinct from surface receptors have a higher metastatic potential com-
laminin but has a similar biologic function. It serves as a pared with those that have minimal levels of these recep-
major adhesion molecule in the intracellular matrix. tors. Thus to develop metastatic foci, the tumor cells

ERRNVPHGLFRVRUJ
1220 19  Metastatic Tumors in Bone

must maintain some degree of adherence to the intercel- important of which is PTHrP, an osteoclast activator.1,10
lular matrix elements. In general, some insufficiency of As osteoclasts resorb bone, TGFβ is released from bone
cell adhesion and junction systems can be documented in matrix, resulting in increased expression of PTHrP by
most cancers.3,13,15,17 tumor cells13; PTHrP may also further increase tumor
For tumor cells to occupy a territory of the stromal cell proliferation and act as an angiogenic factor.
matrix, the existing elements of the stromal matrix have PTHrP and other factors stimulate osteoblasts to
to be at least partly destroyed or degraded. Tumor cells produce the cytokine RANK-ligand (receptor activator of
that invade vessels have a higher capacity to degrade the nuclear factor-κβ ligand), a member of the tumor necrosis
stromal matrix than other tumor cells. The enzymatic factors, which binds to and activates the RANK receptor
activity of tumor cells plays an important role in the on osteoclast precursors, inducing both differentiation
development of metastases. The involvement of several and activation of these bone resorbing cells. PTHrP
different proteases, including urokinase, plasminogen also down regulates osteoprotegerin, a decoy receptor
activator, cathepsins B and D, and various metalloprote- for RANKL.7 Blockade of the RANK-RANKL system
ases produced directly by tumor cells, play an important is the mechanism by which the monoclonal antibody
role in the invasive growth and development of metasta- denosumab serves as an effective therapeutic agent in the
sis.12 Virtually all proteases appear to be controlled by a treatment of metastatic carcinoma.11,14 Bisphosphonates—
cascade of complex-activating and complex-inhibiting inhibitors of osteoclastic bone resorption—are also stan-
factors. Therefore their roles in invasive growth and dard pharmacologic agents used for the care of patients
metastasis depend not only on the level of the enzyme, with bone metastases, among other conditions.
but also on the presence of adequate amounts of their Not all types of metastatic carcinoma induce osteoly-
activators and inhibitors. The major component of the sis. Carcinomas of the prostate and breast, as well as
basement membrane (type IV collagen) is degraded by a neuroendocrine tumors, can induce predominately blastic
specific metalloprotease known as type IV collagenase. or sclerotic metastases. The mechanisms that cause
The activity of this enzyme can be correlated with the blastic metastases are not as well understood as those for
metastatic potential of several experimental and human osteolytic metastases. Factors such as endothelin-1,
tumors. Overall, an upregulation of multiple proteolytic platelet-derived growth factor (PDGF), TGFβ2, insulin-
enzymes of the so-called plasminogen cascade have been like growth factor (IGF), and bone morphogenetic
documented in malignant tumor cells and have been proteins (BMPs) all stimulate bone formation and
linked to their invasive and metastatic potential. are produced by various types of carcinomas. These
The ability to induce vascular growth is another factor factors induce bone formation by inducing osteoclast
that secures the survival of an enlarging tumor mass.13 It apoptosis while stimulating osteoblast differentiation and
appears that the tumors that induce a rich vascular proliferation.
network have a higher metastatic potential. The ability
of tumor cells to induce proliferation of vascular cells via
a number of growth factors such as endothelial growth GENERAL FEATURES OF
factor and fibroblastic growth factor has recently been SKELETAL METASTASES
extensively studied.3 It seems that tumors with higher
levels of these factors are more clinically aggressive com- The skeleton is one of the most common sites for the
pared with those that have low levels of these factors. The metastasis of virtually all common human malignant neo-
tumor cells that invade the vessels and circulate in the plasms, and almost every malignant neoplasm has been
lymph or blood interact with cellular and humoral com- described as being capable of metastasizing to bone.22,46
ponents of the environment. The interaction of tumor In general, metastatic neoplastic cells reach the bones
cells with platelets and other blood clotting factors, both through the complex arterial and venous systems. The
circulating and cell fixed, is an important element in the blood supply to the skeleton represents a significant pro-
promotion of tumor cell thrombosis of peripheral sinu- portion of the body’s vasculature. In addition, the verte-
soids and growth of metastatic foci.13 In addition, the bral plexus of veins is valveless, and the retrograde venous
retention on tumor cells of a unique class of carbohydrate pressure is often increased in the abdominal and chest
antigens that interact with endothelial surface antigens is regions. This enables a retrograde flow of blood to bypass
a critical factor for settlement in metastatic foci. More- the caval system and to reach the bones of the vertebral
over, the tumor cells of a metastatic focus must retain column.24 These two basic anatomic and physiologic fea-
their stromal destructive activities and interact with other tures explain why metastases preferentially involve the
cells of the new environment to survive and form clini- bones of the axial skeleton.25 From a biologic point of
cally detectable nodules. view, it is very unlikely that the abundance of the vascular
Following the successful invasion and colonization of network within bone is the only factor that predisposes
the bone, metastatic deposits most commonly induce to metastasis because metastases rarely develop in other
osteolysis and less commonly new bone formation by tissues that have an equally rich vascular network, such
interacting with complex bone resorption and formation as the spleen. Thus the biologic conditions of bone tissue
regulatory pathways (Fig. 19-2). Osteolysis is driven by must also be important factors in promoting the growth
osteoclastic resorption of bone, the control of which of tumor cells that reach the marrow via the venous and
comes under a variety of influences. For some metastatic arterial blood network.
carcinomas, particularly breast cancer, osteoclastogenesis Autopsy reports of large series of patients have shown
is stimulated by a number of cytokines, one of the most that, with gross examination and limited sampling,

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1221

skeletal metastases can be documented in 30% of patients thyroid, and colon.50 Rarely, bilateral symmetric metas-
who died of carcinoma, with particular carcinomas such tases involving the left and right sides can be present.43,44
as breast or prostate cancer present in nearly 85% of Bilateral, symmetric, osteoblastic metastases can simulate
patients autopsied.21 These findings are supported by osteopoikilosis. The small tubular bones of the hands and
bone scintigraphy studies, which suggest that approxi- feet are rarely affected by metastases34,38,41 (Fig. 19-6).
mately 85% of patients with common carcinomas have This rare phenomenon more often involves the bones of
skeletal involvement. This is in keeping with observations the feet. Although it can occur in many common cancers,
by Jaffe, who stated that if extensive skeletal sampling carcinoma of the lung is the most frequent malignancy
were to be performed, metastases could be documented in which so-called acral metastases occur. In general,
in 70% of patients who died of carcinoma.36 acral metastases are most often seen in the small bones
Conventional radiographs are not particularly sensi- of the feet in highly aggressive malignant neoplasms of
tive in identifying early metastatic lesions in the skele- visceral and thoracic organs. In extremely rare instances,
ton.53 Radioisotopic scanning can demonstrate abnormal metastatic lesions can have a misleading radiographic
uptake of bone-seeking isotope approximately 4 months appearance; that is, their radiographic appearance may
on average before a lesion can be identified on plain support the diagnosis of a benign condition or a primary
radiographs.37 Radioisotopic scans, however, are not bone lesion.
specific, and they identify an increased turnover state Tenderness, swelling, and pain are typical presenting
associated with osteoblastic activity rather than the pro- symptoms of skeletal metastases. The symptoms are
liferation of tumor cells. Therefore they may not detect insidious in onset and gradually increase in intensity over
metastatic tumors that are primarily associated with bone weeks to months, often preceding changes that are rec-
destruction and minimal or no osteoblastic activity. ognizable on conventional radiographs. An abrupt onset
Diffusion-weighted magnetic resonance imaging (MRI) of symptoms (typically severe pain) is usually associated
and whole-body fluorodeoxyglucose positron emission with pathologic fracture (Figs. 19-7 and 19-8). In general,
tomography/computed tomography (FDG-PET/CT) the presence of skeletal metastases is a sign of dissemi-
scans have been shown to be very sensitive and accurate nated multisystem disease, and other organs are likely to
in identifying metastatic deposits in bone from a variety be involved. However, in a minority of patients, a solitary
of sources.33,51,52 metastatic focus in the skeleton can be the only identifi-
Metastatic tumor in bone typically presents as a able site of the disease. In such cases, a metastasectomy
destructive focus that has an aggressive pattern of bone is advocated because it can significantly prolong a patient’s
destruction (Fig. 19-3). Cortical disruption, extension life.23,49 The management of skeletal metastases typically
into soft tissue, and periosteal new bone formation can includes internal fixation or joint replacement for frac-
be present.26,27,39,42,45 Some metastatic tumors can provoke tures or impending fractures, radiation and chemother-
an osteoblastic reaction with new bone formation apy, and pain control. Pharmacologic therapy with
and appear to be a densely sclerotic lesion32 (Fig. 19-4). bisphosphonates inhibits bone resorption by blocking
Often, focal sclerosis is seen within lytic lesions, and osteoclastic activity and has become very important in the
therefore many skeletal metastases produce a mixture management of patients with metastases. These drugs
of lytic and blastic appearances. In general, a lytic improve bone stock in cases of metastatic carcinoma but
versus blastic radiographic appearance of a metastatic do not improve overall patient survival.35
tumor in bone results from a prevalent bone resorptive Because a metastasis to the skeleton is often a patient’s
(destructive) or stimulating (osteoblastic) activity of the initial clinical manifestation of a carcinoma, the surgical
tumor.28,30,31,40 pathologist may be asked to assist in identifying the
Multiple lesions are a radiographic hallmark of meta- source of the tumor. Although it is not possible to char-
static skeletal disease (Fig. 19-4). Although any skeletal acterize the site of origin in all cases of metastatic carci-
site can be affected by metastasis, the majority of lesions noma of unknown primary site (Fig. 19-9), some tumors,
involve the axial skeleton and proximal portions of the such as metastatic renal cell carcinoma and metastatic
appendicular skeleton (Fig. 19-5). Consequently, such thyroid carcinomas, have characteristic light microscopic
bones as the vertebrae, pelvis, ribs, skull, sternum, proxi- features. In cases of metastatic carcinoma lacking charac-
mal femur, and humerus are most frequently involved. teristic histologic findings, the use of a battery of com-
These sites correspond to areas that contain hematopoi- monly employed immunostains can often direct the
etic marrow, which has a rich sinusoidal vascular network. clinical team to investigate a limited number of potential
This feature and the presence of venous plexus connected primary sites, if not identify the specific source for the
with abdominal and thoracic organs may promote metas- metastasis. Some tumors, such as squamous cell carci-
tasis in these regions. Metastases that predominantly noma, lack a specific immunohistochemical profile that
involve fatty marrow distal to elbow and knee joints and allows the distinction of one primary site from another
the mandible are unusual in adults. with certainty. A focused history and physical examina-
Occasionally a single metastatic focus of carcinoma tion coupled with a thorough radiographic evaluation of
can be present. In some patients, it can be a presenting the chest, abdomen, and pelvis are also extremely valu-
sign of a clinically silent primary tumor that, most often, able in identifying the source of metastases to the skele-
is located within the thoracic or abdominal organs.29 ton.47,48 Figure 19-10 represents an algorithm for the
Although a solitary skeletal metastasis can be a presenting immunohistochemical evaluation of the commonly
sign in any type of common carcinoma, it is most fre- encountered metastases in bone.
quent in carcinomas of the kidney, lung, breast, pancreas, Text continued on p. 1230

ERRNVPHGLFRVRUJ
1222 19  Metastatic Tumors in Bone

A B

C D
FIGURE 19-3  ■  Solitary metastasis: radiographic features. A, Anteroposterior view of a predominately lytic, destructive lesion in the
distal femur from a solitary metastatic deposit from a primary breast carcinoma. B, Coronal computed tomography (CT) scan allows
for easier visualization of the metastatic deposit. C, Radioisotope scan shows significant uptake in a solitary bone metastasis.
D, The gross appearance of the lesion correlates well with the radiographic findings, particularly those seen on CT scan.

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1223

A B

C D
FIGURE 19-4  ■  Widespread osteoblastic skeletal metastases in prostate adenocarcinoma: radiographic features. A, Anteroposterior
view of several densely osteoblastic metastases in the proximal humerus and scapula. B, Coronal computed tomography (CT) scan
highlights osteoblastic lesions in the humerus and scapula. C, Radioisotope scan show multiple foci of increased uptake throughout
the axial and appendicular skeleton, including a large lesion in the proximal left femur. D, Coronal CT scan shows the destructive
lesion in the proximal femur, which has both lytic and blastic foci.

ERRNVPHGLFRVRUJ
1224 19  Metastatic Tumors in Bone

FIGURE 19-5  ■  Metastases in vertebral column: radiographic and gross features. A, Radiograph of sagittally bisected vertebral column
with multiple lytic destructive lesions in vertebral bodies taken during autopsy. B, Gross appearance of sagittally bisected vertebral
column with massive blastic metastases seen in vertebral bodies. C, Gross appearance of sagittally bisected lumbar spine and
sacrum with multifocal diffuse involvement by sclerotic metastatic foci of L3 and L5 and sacrum.

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1225

A B C D

FIGURE 19-6  ■  Acral metastases: gross and radiographic features. A, Destructive lytic lesion of proximal phalanx of third toe repre-
sents metastasis from adenocarcinoma of lung. B, Closer oblique view of case shown in A. C and D, Destructive lytic lesion of middle
phalanx of thumb in non–small-cell carcinoma of the lung. E, Gross photograph shows metastatic renal cell carcinoma in talus.

ERRNVPHGLFRVRUJ
1226 19  Metastatic Tumors in Bone

B
FIGURE 19-7  ■  Metastatic carcinoma with pathologic fracture: radiographic and histologic features. A and B, Low and high power
photomicrographs of the lesion shown in the inset demonstrating characteristic histologic features of metastatic hepatocellular
carcinoma. Inset, Anteroposterior view of a pathologic fracture through a subtle radiolucent lesion involving the proximal diaphysis
of the humerus (A, ×100; B, ×400) (A and B, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1227

A B

C
FIGURE 19-8  ■  Metastatic carcinoma: gross features. A, Metastatic renal cell carcinoma in proximal humerus. Note pathologic fracture
and extension into soft tissue. B, Metastatic renal cell carcinoma in proximal humerus. Tumor was embolized before surgery.
C, Metastatic breast carcinoma in femoral head.

ERRNVPHGLFRVRUJ
1228 19  Metastatic Tumors in Bone

A B

FIGURE 19-9  ■  Metastatic carcinoma to rib, unknown primary. A and B, Photomicrographs show a poorly differentiated carcinoma
composed of sheets and nests of atypical epithelioid cells, many of which have clear cytoplasm. The tumor elicits reactive
bone formation. C, Gross photograph of the metastasis to the rib, which has a heavily blastic appearance. Inset, An immunohisto-
chemical stain for CAM5.2 is strongly positive in tumor cells; immunohistochemical and radiographic evaluation failed to identify
a primary site, and the patient died with disseminated disease before a source could be located, (A, ×100; B and Inset, ×200; A and
B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1229

Destructive skeletal lesion

Primary bone neoplasm Hematolymphoid process,


or myeloma

Keratin- Keratin+
Metastasis

S-100+, markers of Markers of


melanocytic differentiation neuroendocrine differentiation
Keratin- Screening Keratin+

Melanoma Sarcoma Carcinoma Small cell carcinoma TTF1+, variable CD56,


chromogranin+, synaptophysin+

Pheochromocytoma/ chromogranin+,
paraganglioma synaptophysin+

Neuroblastoma NFP+, chromogranin+,


(first r/o Ewing/PNET) synaptophysin+, CD56+

Prostatic PSA+, PSAP+


adenocarcinoma

CK7-/CK20- Renal cell CD10+, RCC+, PAX8+


Alveolar soft TFE3+, desmin+/-, muscle
carcinoma
part sarcoma specific actin+/-
Hepatocellular pCEA+, HepPar1+, AFP+
Angiosarcoma ERG+, CD31+
carcinoma
Leiomyosarcoma Smooth muscle actin+,
muscle specific actin+, Mammary ER+, GCDFP+,
desmin+, caldesmon+, adenocarcinoma mammaglobin+, GATA3+
EMA+/-
Pulmonary TTF1+, napsin A+
Liposarcoma S-100 protein+ (adipocytic adenocarcinoma
component/lipoblasts in all
liposarcoma subtypes), Well-differentiated TTF1+, thyroglobulin+
MDM2/CDK4+ thyroid carcinomas
(dedifferentiated liposarcoma)
CK7+/CK20- Gynecologic WT1+/ER-, PAX8+
Malignant S-100 protein+/- (50%) serous carcinomas
peripheral nerve
sheath tumor Gynecologic ER+/WT1-, PAX8+
endometrioid
Rhabdomyosarcoma Desmin+, myogenin+, carcinomas
CD99+/-
Pancreatic DPC4-
Synovial sarcoma Cytokeratin+ (positive in carcinoma
(SS) glandular component of
biphasic SS, patchy in Upper GI CDX2+, villin+
monophasic and poorly adenocarcinoma
differentiated SS), EMA+
(similar pattern to
cytokeratin), CD99+, BCL2+, Colorectal and CDX2+, villin+
CK7-/CK20+
TLE1+ some appendiceal
carcinomas
Undifferentiated Vimentin+, smooth muscle
pleomorphic actin+/-, CD68+/-
sarcoma Urothelial Uroplakin+, GATA3+
carcinoma

CK7+/CK20+ Some pancreatic MUC5+, CK19+


adenocarcinomas

Upper GI CDX2+, villin+


adenocarcinoma

FIGURE 19-10  ■  Algorithm for the immunohistochemical evaluation of a metastasis of unknown primary. (Modified and reprinted with
permission from Bhargava R, Dabbs DJ: Immunohistology of metastatic carcinomas of unknown primary. In Dabbs DJ, editor: Diagnostic
immunohistochemistry. Theranostic and genomic applications, Philadelphia, 2010, Saunders Elsevier, pp 206–255.)

ERRNVPHGLFRVRUJ
1230 19  Metastatic Tumors in Bone

Radioisotopic scans reveal abnormal uptake suggestive of


SKELETAL METASTASES IN SELECTED metastasis in approximately 45% of patients who have
COMMON NEOPLASMS stomach cancer.56 The sites most frequently involved are
similar to those affected by other common neoplasms:
In this section, some basic clinical, radiographic, and the spine, ribs, pelvis, femur, and skull. Most metastases
pathologic features of bone involvement in the most occur approximately 1 year after diagnosis in patients
common malignant neoplasms are briefly discussed. The with stage III disease.73 Occasionally, skeletal metastasis,
entities discussed in this section represent only the either as a solitary lesion or as multiple lesions, can be a
malignancies for which skeletal metastases are most fre- presenting sign.67 This is more typical of poorly differ-
quently a presenting sign or occur during the course of entiated gastric carcinoma such as a diffuse type of signet-
the disease. ringcell carcinoma or a small cell variant (Figs. 19-13
and 19-14).
Colorectal carcinoma is very common in developed
Carcinoma of the Lung countries, but bone metastases from these tumors occur
Lung cancer is a leading cause of death worldwide and less commonly than from gastric carcinomas.55 The inci-
the most frequently occurring human malignancy that dence of bone metastasis from colorectal cancer is
has a high propensity to metastasize to a variety of distant reported to be approximately 23% in autopsy cases.62
sites.76 All major histologic subtypes of lung cancer— Cancers involving the rectum and cecum are more likely
including small cell and non–small cell carcinomas—have to develop bone metastasis than tumors involving other
a high tendency to metastasize to the skeleton, particu- portions of the colon.69 Poorly differentiated carcinoma
larly the small cell types.70,77 Carcinoma of the lung typi- and signet-ring cell carcinomas have a higher propensity
cally produces lytic metastases (Fig. 19-11) and is the for metastasizing to bone than other histologic types of
most frequent human tumor to present as a solitary meta- colon cancer (i.e., conventional, well to moderately dif-
static lesion in bone; lung cancers are the most frequent ferentiated adenocarcinoma of the colon). Skeletal metas-
tumor that metastasizes to the acral skeleton. Bilateral tases typically coexist with lung and liver metastases.
symmetric metastases that involve unusual sites, such as Solitary skeletal metastasis can be a presenting sign in
the patella, can also occur.71 Finally, lung cancer is often colorectal cancer. In fact, colon carcinoma is the most
centered on the cortex of the appendicular skeleton and frequent gastrointestinal cancer in which solitary skeletal
may cause unusual radiographic presentations such as metastasis can appear as the initial clinical event. Some
entirely intracortical or subperiosteal lesions. of these lesions may involve unusual sites such as the acral
A great deal is known about the molecular alterations skeleton, skull, or patella or even the temporomandibular
in lung cancer, which can impact the use of targeted thera- joint.59,65,72,74,78 Microscopically, these lesions have typical
pies in the treatment of these patients. Epidermal growth features of gastrointestinal adenocarcinoma with variable
factor receptor (EGFR), the anaplastic lymphoma kinase degrees of differentiation, mucin production, and signet-
gene (ALK), and the K-RAS gene should all be evaluated ring cell features.
in lung cancers, including metastatic deposits when the Aspirates from metastatic colon carcinoma are usually
primary tumor has not been sampled. ALK gene rear- very cellular and show cells organized in three-dimensional
rangements occur in young, nonsmoking patients with clusters, large sheets, small groups, and dispersed singly.
adenocarcinoma. Metastatic pulmonary adenocarcinoma Nuclei of cancer cells, arranged in palisade at borders of
may harbor mutations of the EGFR gene, and in such the large sheet composed of columnar cancer cells, is a
cases patients frequently respond favorably to tyrosine characteristic feature of colon carcinoma (Fig. 19-15, A,
kinase inhibitors.63 Patients with K-RAS mutated adeno- B). Necrosis is very frequent in cytologic preparations
carcinoma may be resistant to tyrosine kinase inhibitors. from metastatic colon carcinoma. In many of the cases,
Aspirates from non–small cell metastatic lung cancer cytologic features of metastasis allow recognition of the
are highly cellular and contain large epithelial cells with primary site even without clinical data.
pronounced atypia (Fig. 19-12, A). Keratinization and
extensive necrosis are frequently seen. Dispersed cells Carcinoma of the Breast
with scanty cytoplasm, occasionally forming small three-
dimensional clusters, characterize metastatic small cell Breast carcinomas have a high propensity for metastasiz-
carcinoma (Fig. 19-12, B). Nuclei of these cells have ing to bone, and bone is the most common site of distant
coarsely granular chromatin with a few larger chromo- (nonnodal) metastases for these tumors.54,57,58,61,64,66 They
centers and no visible nucleoli. Positive immunohisto- occur in typical skeletal sites, such as the vertebral bodies,
chemical staining for thyroid transcription factor-1 pelvis, proximal femur, and humerus. Involvement of the
(TTF-1) can be helpful to establish the origin of metas- proximal femur (intertrochanteric area and femoral neck
tasis from lung. Small cell carcinomas typically show neu- with pathologic fracture) and the presence of cranial and
roendocrine differentiation and are positive for a wide dural metastases are typical signs of advanced-stage breast
range of neuroendocrine markers. carcinoma. Carcinoma of the breast usually produces
osteolytic metastases, but some can be of mixed type; less
frequently, they are purely sclerotic60,68,79 (Fig. 19-16).
Gastrointestinal Carcinoma Cancers that produce lytic skeletal lesions destroy bone
Most carcinomas of the gastrointestinal tract are highly with the assistance of bone-resorbing osteoclasts. The in
aggressive lesions with a high propensity for metastasis.75 Text continued on p. 1237

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1231

C D
FIGURE 19-11  ■  Lytic pulmonary adenocarcinoma metastasis in humerus: radiographic, microscopic, and immunohistochemical
features. A, Anteroposterior radiograph showing a metastatic pulmonary adenocarcinoma resulting in a large radiolucent defect in
the humerus with cortical destruction and soft tissue extension. B, Positron emission tomography (PET) scan showing innumerable
PET-avid metastases throughout the skeleton. C and D, Microscopic features of the metastasis seen in A, showing a relatively poorly
differentiated adenocarcinoma with a small amount of adjacent reactive bone. Insets, The tumor was immunoreactive for cytokeratin
7 (left) and TTF-1 (right), supporting the lung as the source of the metastasis. (C and Insets, ×200; D, ×400.) (C, D, and Insets,
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
A B

C D

FIGURE 19-12  ■  Metastatic carcinoma of the lung and follicular carcinoma of the thyroid: cytologic features. A, A cluster of the lung
adenocarcinoma cells showing obvious cytologic atypia. Inset, Higher magnification of individual cancer cells with hyperchromatic
dark nuclei. B, A cluster of poorly differentiated cancer cells with somewhat elongated nuclei, pronounced atypia, and brisk mitotic
activity in a case of small cell carcinoma metastatic to bone. C and D, Groups of loosely arranged cells forming small follicular
structures characteristic of thyroid cancer. Note traces of colloid substance in the background of the smears. Inset, Higher magni-
fication of thyroid follicular cancer cells with visible nucleoli. (A, C, and D, ×400; B and Insets, ×600.) (A-D and insets, hematoxylin-
eosin.) (Reprinted with permission from Czerniak B, et al: Bone tumors. In Koss LG, Melamed MR, editors: Koss’ diagnostic cytology and
its histopathologic bases, ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1233

A B

C D

FIGURE 19-13  ■  Widespread blastic gastric adenocarcinoma metastases: radiographic, microscopic, and immunohistochemical fea-
tures. A, An axial computed tomography scan of the spine shows several osteoblastic foci of metastatic carcinoma within the
vertebral body. B, Radioisotope scan shows increased uptake in widespread metastases throughout the skeleton. C, Microscopic
features of metastatic poorly differentiated adenocarcinoma filling the intertrabecular spaces associated with reactive bone
formation. D, An immunohistochemical stain for cytokeratin AE1/3 is strongly reactive in tumor cells. (C and D, ×400;
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1234 19  Metastatic Tumors in Bone

A B

C D
FIGURE 19-14  ■  Metastatic esophageal adenocarcinoma in femur: radiographic and microscopic features. A, Anteroposterior
radiograph shows a blastic metastatic focus from an esophageal adenocarcinoma. B and C, Coronal and axial computed tomog-
raphy scans highlight the blastic reaction to the metastatic deposits within the medullary cavity as well as periosteal reaction.
D, Microscopic features of metastatic, moderately differentiated adenocarcinoma that originated in the esophagus. (D, ×200)
(D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1235

A B

C D

FIGURE 19-15  ■  Colonic and breast carcinomas metastatic to bone: cytologic features. A and B, Large sheets of colonic carcinoma
cells showing overlapping nuclei and focal peripheral palisading. Inset, Higher magnification of tumor cells. Note that individual
nuclei of tumor cells vary in size and contain coarse chromatin with prominent irregular nucleoli. C and D, Metastatic ductal breast
carcinoma of the breast with cells arranged in loose clusters. Inset, Details of nuclear morphology with coarse chromatin and
prominent irregular nucleoli (A-D, ×400; Insets, ×600) (A-D and insets, hematoxylin-eosin). (Reprinted with permission from Czerniak
B, et al: Bone tumors. In Koss LG, Melamed MR, editors: Koss’ diagnostic cytology and its histopathologic bases, ed 5, Philadelphia, 2006,
Lippincott Williams & Wilkins.)

ERRNVPHGLFRVRUJ
1236 19  Metastatic Tumors in Bone

A B

C D
FIGURE 19-16  ■  Metastatic breast carcinoma: radiographic and microscopic features. A and B, Axial computed tomography scans
show foci of metastatic carcinoma adjacent to the sacroiliac joint and within the iliac wing. C and D, Microscopically, this poorly
differentiated adenocarcinoma had foci with signet-ring cell features similar to the primary tumor within the breast (C, ×200; D, ×400)
(C and D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1237

vivo model of metastatic breast carcinoma supports the 10) or in small cell variants of prostate carcinoma. In
concept that the ability of tumor cells to stimulate osteo- typical cases, the unique morphologic characteristics of
clasts plays a role in bone destruction that is associated well-developed small glands of prostate carcinoma are
with tumor deposits.1 easy to recognize in metastases. In addition, the expres-
Aspirates from metastatic breast carcinoma show sion in metastatic sites of two organ-specific makers,
three-dimensional aggregates, small clusters and single, prostate-specific antigen (PSA) and prostate-specific acid
dispersed cancer cells of various sizes. Differentiation phosphatase (PSAP), which can be identified immuno-
between ductal and lobular carcinoma in bone aspirates histochemically, is a helpful feature for diagnosis. Some
is rarely possible.153 Ductal carcinomas are characterized poorly differentiated prostate carcinomas can be negative
by the presence of large cancer cells with abundant cyto- for both PSA and PSAP. Some of these neoplasms can
plasm containing highly atypical nuclei with a clumped have neuroendocrine features. The small cell carcinomas
chromatin pattern and prominent nucleoli (Fig. 19-15, of the prostate are microscopically indistinguishable from
C, D). These cells form irregular clusters with crowding other small cell carcinomas, such as those of lung origin.
and overlapping nuclei. Aspirates from metastatic lobular A significant proportion of small cell carcinomas of the
carcinoma are characterized by the presence of relatively prostate express TTF-1, and care must be taken to distin-
small, oval cells with eccentric, fairly monotonous hyper- guish such lesions from metastatic small cell carcinoma
chromatic nuclei. Some of these cells contain a large of pulmonary origin.136
vacuole with central inclusion of condensed mucous The osteoblastic reaction associated with metastatic
material. Nearly all patients have a past history of treated neoplastic cells within the bone can be so pronounced
mammary carcinoma. In the rare cases without this that it can overshadow cancer cells. In such cases, the
history, the disease should be suspected in women of the metastatic foci predominantly consist of sclerotic bone
appropriate age group. with scattered small clusters of tumor cells. It is postu-
lated that prostate carcinoma stimulates new bone pro-
duction by secretion of several ubiquitous kinins and
TUMORS WITH A UNIQUE PROPENSITY pro-osteoblastic factors that drive osteoblast differentia-
FOR SKELETAL METASTASES tion and bone formation.81 Occasionally, painless scle-
rotic metastases of prostatic carcinoma that are negative
Some malignancies have unique biologic properties that on biopsy samples (i.e., contain only sclerotic bone) can
make them prone to metastasize to the skeleton. In fact, be confused with nonneoplastic disorders such as osteo-
for such malignancies, solitary or multiple skeletal metas- poikilosis and bone islands. Treated prostate carcinoma
tases are frequently an integral component of the clinical can be very inconspicuous; that is, the tumor cells can
picture at presentation, especially when the primary mimic nonneoplastic cells such as histiocytes. In such
tumor remains occult. Even if they are not evident at cases, documentation of the epithelial nature of these
presentation, skeletal metastases eventually develop later cells with epithelial markers such as cytokeratin can aid
during the course of the disease in this group of tumors. the diagnosis. In addition to radiotherapy and bisphos-
phonates, a variety of radiopharmaceuticals have recently
been identified to specifically treat prostate carcinoma
Carcinoma of the Prostate bone metastases by homing in on bone lesions while
Carcinoma of the prostate is one of the most common limiting local toxicity, thus providing both palliative and
human cancers, accounting for nearly 25% of all malig- survival benefits.84,96
nancies in males, and is a model for epithelial malignancy Aspirates from metastatic prostatic adenocarcinoma
that is likely to metastasize to bone.80,85,88,95,115 Not all are cellular; cells may be dispersed singly or create sheets
prostate cancers have this unique propensity, and some and glandlike structures (Fig. 19-17, A, B). Cancer cells
prostate carcinomas will behave in indolent fashion. The have no distinguishing features and are usually large with
risk factors for dissemination of the disease and develop- cytoplasmic borders and nuclei with clearly visible large
ment of metastases in distant sites such as the skeleton nucleoli. Cytologic preparations from well or moderately
include tumors with a high histologic grade (Gleason’s differentiated prostate cancer may contain recognizable
combined score ≥7), high stage, large diameter, and microglandular structures that resemble small rosette-
metastasis to pelvic lymph nodes.119,124,141,142 More precise like formations with nuclei at the periphery and a center
volumetric measurements of whole organ-embedded formed by amorphous cytoplasmic material. Immunohis-
specimens indicate that the volume of cancer is an impor- tochemical stains for PSA and PSAP may be used to
tant prognostic factor for dissemination.142 Studies have confirm the prostatic origin of the lesion, if there is no
shown that nearly 90% of men who develop metastases past history of the disease or if there is the possibility of
from prostate cancer will have bone involvement.136 another primary tumor.
Skeletal metastases of prostate carcinoma are typi-
cally osteoblastic (Fig. 19-4). Sclerotic lesions in the
pelvis and vertebral bodies are seen most often. In rare
Carcinoma of the Kidney
instances, massive sclerotic metastases can develop within Renal cell carcinoma is a prototype of a tumor that fre-
major portions of the skeleton and are associated with quently presents as a skeletal (often solitary) metastasis
pronounced periosteal new bone formation. Osteo- with a clinically occult primary tumor.131,135 Nearly 30%
lytic metastases typically occur in poorly differentiated of patients with renal cell carcinomas have metastatic
prostate carcinomas (Gleason’s combined score of 9 to disease at presentation.129 Dissemination in the form of

ERRNVPHGLFRVRUJ
1238 19  Metastatic Tumors in Bone

C D

FIGURE 19-17  ■  Prostatic and renal carcinomas metastatic to bone: cytologic features. A and B, Large cohesive sheets of prostatic
carcinoma cells. Note that nuclei of tumor cells are uniform in size and display only minimal atypia. Inset, Higher magnification of
tumor cells showing details of nuclear morphology, which contain prominent nucleoli and evenly distributed chromatin. C and
D, Metastatic clear cell carcinoma of the kidney shows loosely arranged clusters of tumor cells with ill-defined clear and finely
granular cytoplasm. Inset, Tumor cells with clear cytoplasm. Note mild atypia of tumor cells characterized by the presence of round
to oval nuclei with clearly visible nucleoli (A-D, ×400; Inset A, ×600; Inset D, ×400) (A-D and insets, hematoxylin-eosin.) (Reprinted
with permission from Czerniak B et al: Bone tumors. In Koss LG, Melamed MR, editors: Koss’ diagnostic cytology and its histopathologic
bases, ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1239

multiple or solitary skeletal metastases will eventually with a propensity for local recurrence and lymph node
develop in up to 50% of patients originally treated for metastases to highly malignant, undifferentiated (ana-
localized disease (Figs. 19-18 and 19-19).98,122 Occasion- plastic) carcinoma.93,120,134 All variants of thyroid car-
ally, partial or complete regression of metastatic renal cell cinoma can metastasize to bones (Fig. 19-20), but the
carcinoma is observed,91,100,106,114 which can be spontane- well-differentiated carcinomas (papillary, follicular, and
ous or induced by the reduction of tumor burden. Hürthle cell types) have a unique propensity to metastasize
Renal cell carcinomas are known to metastasize to to the skeleton.90,94,97,108,109,111,112,116,118,123,126,132,133,139,140,143
unusual distant sites such as the skin, tongue, eye, heart Among the well-differentiated variants, follicular carci-
muscle, acral skeleton, and breast.99,103,107,125,137 Autopsy noma is particularly prone to metastasize to bone. Well-
data reveal that approximately 8% of patients who died differentiated thyroid carcinoma can present clinically
of renal cell carcinoma have metastasis confined to one as skeletal metastases (either solitary or multiple lesions)
organ. Renal cell carcinomas preferentially metastasize to with an occult primary tumor.101 It appears that in thyroid
the femur, humerus, pelvis, and spine; although osteo- cancer, after metastases to the neck lymph nodes, the
blastic lesions have been described,128 the vast majority skeleton and lungs are the most frequent site of metas-
are lytic. tases.113 Skeletal metastases can occur even more than 20
Typically, metastatic renal cell carcinomas have clear years after treatment of the primary lesion. Thyroid car-
cell features, and the renal origin of skeletal metastasis is cinomas that affect younger individuals and children are
suspected after microscopic examination of biopsy more aggressive and have a higher propensity to metas-
samples (Fig. 19-19). Characterization of a metastatic tasize to the skeleton and lungs, although age greater
clear cell carcinoma as originating from the kidney is than 55 years is a strong predictor of death following
easily achieved with commonly utilized immunohisto- disseminated disease.102
chemical markers such as RCCma, CD10, or PAX-8. Radiographically, metastatic thyroid carcinoma typi-
Approximately 10% of renal cell carcinomas dedifferenti- cally presents as a destructive lytic lesion of bone.
ate into high-grade spindle-cell or pleomorphic sarcoma- Metastatic thyroid carcinoma in bone has easily recog­
toid carcinoma. The sarcomatoid elements may be the nizable microscopic features and can be clinically associ-
only components present in bone metastases. Some of ated with thyrotoxicosis105 (Figs. 19-20 and 19-21). The
these lesions can have features suggesting an undifferen- microscopic diagnosis can be aided with appropriate
tiated spindle-cell or pleomorphic sarcoma, and can be immunohistochemical stains for thyroglobulin or TTF-1
misdiagnosed as primary bone lesions. Review of clinical in follicular and papillary carcinomas, and calcitonin in
data and identification of the epithelial nature of cells medullary carcinomas.
with appropriate immunohistochemical markers are Aspirates from metastatic thyroid follicular carcinoma
helpful for classifying the lesion as a metastasis. show a uniform population of small cells lying singly in
Resection of solitary skeletal metastasis significantly three-dimensional clusters, sometimes forming follicles
prolongs life.89,110,127,130 Nephrectomy or metastasectomy with central colloid. Characteristically, the nuclei show
is sometimes performed in patients with disseminated granular chromatin and clearly visible nucleoli (Fig.
disease to reduce the tumor burden, with the hope that 19-12, C, D). Intranuclear cytoplasmic inclusions and
remaining foci may stabilize or regress.132 Patients with a nuclear “creases” are commonly seen in the spherical
solitary metastasis, metastatic deposits with low Fuhrman nuclei. Colloid can be seen as an amorphous substance
histologic grade, and late onset of metastatic disease have dispersed in a background of the smear or can be present
longer survival.92,129,138 inside the core follicles. Cytologic features of metastatic
Aspirates from metastatic renal cell carcinoma tend to thyroid follicular cell carcinoma are very distinctive and
be bloody, with cancer cells lying singly or arranged in permit the identification of thyroid origin in the majority
small groups, and even occasionally larger three- of cases.49 Immunohistochemical stains for thyroglobulin
dimensional structures. Cancer cells have abundant clear can be used as a marker in questionable cases.
cytoplasm with multiple delicate vacuoles and ill-defined
borders (Fig. 19-17, C, D). Nuclei have delicate chroma- Carcinoma of the Pancreas
tin and conspicuous nucleoli. Cytologic preparations
from the metastatic sarcomatoid variant of renal cell car- Carcinoma of the pancreas is known for its poor progno-
cinoma show malignant spindle and pleomorphic cells sis. It is associated with the lowest 5-year survival rate
arranged in large sheets, small clusters, or singly that may (10%) of all types of cancer surveyed in the National
be very misleading. Nuclei show pronounced atypia char- Cancer Institute’s Surveillance, Epidemiology, and End
acterized by irregular chromatin clumping and promi- Results (SEER) Program.86 It seems that cancers that
nent nucleoli. In general, the cytologic features of arise in the body and the tail of the pancreas have a higher
metastatic sarcomatoid carcinoma are indistinguishable tendency for distant metastasis compared with those that
from those of primary malignant fibrous tumors of bone. arise in the head of the pancreas.104 In addition to regional
Immunohistochemical stains for keratin and vimentin, lymph nodes and liver, lung, and bone are the most favor-
combined with a clinical history of primary renal carci- able distant sites involved.82,87 Pancreatic carcinomas,
noma, are helpful to establish the correct diagnosis. especially those that arise in the body or tail, can present
clinically as a solitary bone metastasis, and may produce
either lytic or osteoblastic lesions.83,117,121 The axial skel-
Carcinoma of the Thyroid eton is more frequently affected by metastatic pancreatic
The behavior of carcinomas of the thyroid varies widely carcinoma than the bones of the extremities.
from indolent, slow-growing, well-differentiated tumors Text continued on p. 1244

ERRNVPHGLFRVRUJ
1240 19  Metastatic Tumors in Bone

A B

C
FIGURE 19-18  ■  Multifocal metastatic renal cell carcinoma. A and B, T1- and T2-weighted coronal magnetic resonance images (MRIs)
show a large deposit of metastatic renal cell carcinoma that has destroyed the iliac wing, forming a large extraosseous mass. The
lesion is low signal intensity on T1-weighted images (A) and high-signal intensity on T2-weighted images (B). C, An axial computed
tomography scan shows the large lesion in the right ilium as well as a second lesion near the left sacroiliac joint.

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1241

A B

C D
FIGURE 19-19  ■  Solitary renal cell carcinoma metastasis in humerus: radiographic, gross, and microscopic features. A, Anteroposte-
rior radiograph shows a solitary lytic lesion in the proximal humeral diaphysis. B, Radioisotope scan shows a solitary focus of uptake
corresponding to the lesion seen in A. Inset, Gross appearance of metastatic renal cell carcinoma. C and D, Microscopically, the
lesion is composed of nests of clear cells with intervening fine capillaries, typical of renal cell carcinoma (C, ×100; D, ×400) (C and
D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1242 19  Metastatic Tumors in Bone

A B

C D
FIGURE 19-20  ■  Lytic metastatic carcinoma of thyroid: radiographic features. A, Lytic destructive lesion of rib. B, Specimen radiograph
shows destructive lytic lesion with moth-eaten pattern. Note absence of sclerotic reaction. C, Gross photograph of bisected rib shows
intramedullary gray-tan lesion with cortical disruption. D, Photomicrograph shows characteristic follicular growth pattern with colloid
of metastatic follicular variant of papillary carcinoma of thyroid. (D, ×100) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1243

A B

C D
FIGURE 19-21  ■  Metastatic anaplastic thyroid carcinoma to bone mimicking sarcoma. A and B, Low power photomicrographs show
a predominantly spindle-cell neoplasm with abundant collagenous stroma, interpreted on limited needle biopsy as undifferentiated
carcinoma. C, The sarcomatoid component adjacent to a focus with overt epithelial differentiation. D, The epithelial component
resembles the follicular variant of papillary carcinoma. (A, ×100; B-D, ×200.) (A-D, hematoxyin-eosin.)

ERRNVPHGLFRVRUJ
1244 19  Metastatic Tumors in Bone

Aspirates from metastatic pancreatic carcinoma show metastases, owing in part to a shorter interval to metas-
clusters of obviously atypical adenocarcinoma cells with tasis for the axial lesions.145
prominent eosinophilic cytoplasm. The crowded sheets Aspirates of melanoma show the presence of cells with
of epithelial cells with a high ratio of nuclear variability oval densely eosinophilic cytoplasm and eccentrically
are typically present (Fig. 19-25, C, D). located nuclei showing obvious atypia and prominent
nucleoli. Melanin deposits are frequently present. Mela-
noma cells show a high degree of morphologic variability,
METASTATIC MELANOMA and cytologic presentation may vary from large pleomor-
phic to small inconspicuous cells as well spindle-cell vari-
Melanoma is a relatively uncommon cutaneous malig- ants (see Figs. 19-24, A and 19-25, A, B).
nancy, accounting for fewer than 5% of all cancers. Mela-
noma most commonly metastasizes to regional lymph
nodes, but skeletal metastases also occur, most often
involving axial sites such as the ribs, jaw, pelvis, and SKELETAL METASTASES IN SELECTED
spine; appendicular metastases are encountered less PEDIATRIC TUMORS
frequently.153,154
Radiographically, metastatic melanoma appears as a Many extraskeletal pediatric tumors can metastasize to
lytic lesion with an aggressive pattern of bone destruction bone. In general, most skeletal metastases that occur in
(Fig. 19-22); however, a small percentage of metastatic children are osteolytic. In addition, they more often
melanomas can have either a deceptively nonaggressive involve the appendicular skeleton compared with the
pattern of bone destruction or can cause an osteoblas- axial skeletal involvement seen in adults. However, metas-
tic lesion.148 Metastatic melanoma may appear hyper- tases from neoplasms such as medulloblastoma and other
intense on T1-weighted MRI due to the presence of glial tumors, osteosarcoma, carcinoid tumor, and some-
both melanin pigment and hemorrhage within the times retinoblastoma can induce prominent osteoblastic
lesion.150,152 MRI appears to be a more sensitive modal- reactions and present as sclerotic lesions on radio-
ity than CT for identifying metastatic melanoma in graphs165,171,198 (Fig. 19-26).
bone.151 Rhabdomyosarcoma, neuroblastoma, and clear cell
Metastatic melanoma can present the same wide array sarcoma of the kidney are the three most notable exam-
of histologic appearances that is seen in primary melano- ples of pediatric malignancies that have a particularly
mas (Fig. 19-23). Tumor cells can vary from epithelioid high propensity for metastasizing to the skeleton, a
to spindled in appearance, and may or may not contain feature that represents an integral part of the clinical
melanin pigment. While establishing a diagnosis of meta- behavior of these tumors. The first two tumors, together
static melanoma is not difficult in the setting of a known with Ewing’s sarcoma, belong to the category of nonhe-
primary cutaneous tumor, the diagnosis can be extremely matologic small round-cell malignancies.
challenging when the primary lesion has either not been
identified or has undergone regression. In such cases, a
battery of immunostains including S-100 protein, Melan-
Rhabdomyosarcoma
A, HMB-45, microphthalmia-associated transcription Rhabdomyosarcoma can be classified into several differ-
factor (MITF), or tyrosinase, is necessary to establish the ent subtypes. Embryonal and alveolar rhabdomyosar-
melanocytic nature of the tumor and distinguish the coma are seen mainly in the pediatric population, whereas
lesion from other types of primary and secondary malig- pleomorphic rhabdomyosarcoma, a relatively rare tumor,
nant neoplasms. occurs mainly in the adult population. Embryonal rhab-
In the setting of an unknown primary tumor, meta- domyosarcoma has a peak age incidence between ages 3
static melanoma must be distinguished from poorly dif- and 5 years. Occasionally, it may present in newborns as
ferentiated carcinoma; amelanotic tumors may also be an orbital or perineal mass. One subtype of embryonal
mistaken for poorly differentiated carcinoma or even sar- rhabdomyosarcoma that affects adolescents and is more
comas when predominately spindled in appearance.147 frequently located in the spermatic cord is the spindle-
This separation is usually possible with the use of a cell variant. Overall, the head and neck region, genito-
battery of immunostains, including cytokeratins and urinary organs, and soft tissue of the extremities are the
various melanocytic markers. Rare examples of HMB-45– most frequently affected sites. The alveolar variant of
immunoreactive primary perivascular epithelioid cell rhabdomyosarcoma, which more commonly arises in the
tumors (“PEComas”) of bone should be distinguished extremities, has a poorer prognosis than either embryo-
from metastatic melanoma primarily by their lack of nal rhabdomyosarcoma or fusion-negative alveolar rhab-
cytologic atypia.146,155 domyosarcomas193 (see below). Rhabdomyosarcoma is a
After a diagnosis of metastatic melanoma, the surgical highly aggressive neoplasm; however, modern multimo-
pathologist may be asked to assess the tumor for either dality treatment regimens have significantly improved
BRAF, NRAS, or KIT mutations,144,149 particularly if such 5-year survival to greater than 70%, and 5-year survival
testing has not been obtained on a primary tumor. The for embryonal rhabdomyosarcoma approaches 90%.
status of these mutations can have implications for tar- Many rhabdomyosarcomas have clearly recognizable
geted therapies. Survival following metastatic melanoma rhabdomyoblastic microscopic features and can be diag-
to bone is poor. Patients with appendicular metastases nosed with the aid of appropriate immunostains and
seem to have slightly better survival than those with axial Text continued on p. 1250

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1245

A B

C D
FIGURE 19-22  ■  Metastatic melanoma in femur: radiographic features. A, Lateral radiograph of the knee shows subtle disruption of
the posterior femoral cortex in the region of the lateral femoral condyle. B, Sagittal computed tomography scan more clearly shows
a radiolucent lesion in the distal femur with destruction of the posterior metaphyseal cortex. C, Sagittal, proton density magnetic
resonance image (MRI) and D, T2-weighted coronal MRI show the metastatic melanoma in the posterior portion of the lateral femoral
condyle.

ERRNVPHGLFRVRUJ
1246 19  Metastatic Tumors in Bone

B
FIGURE 19-23  ■  Metastatic melanoma: microscopic features. A and B, Photomicrographs of the lesion in Figure 19-18 showing nests
of pleomorphic, epithelioid cells typical of melanoma. Inset, Tumor cells are strongly immunoreactive for S-100 protein. (A and
Inset, ×200; B, ×400.) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1247

A B

C D
FIGURE 19-24  ■  Metastatic melanoma, neuroblastoma and rhabdomyosarcoma: cytologic features. A, Aspirates from metastatic
melanoma showing large epithelioid cells with eccentric nuclei. B, Metastatic neuroblastoma showing poorly differentiated small
cells in a background of delicate fibrillar matrix. C and D, Aspirate of metastatic alveolar rhabdomyosarcoma showing dispersed
rhabdoid cells with oval cytoplasm. (A, B, and D, ×600; C, ×400.) (A-D, hematoxylin-eosin.) (Courtesy of Dr. John Stewart, MD Anderson
Cancer Center, Houston, Texas.)

ERRNVPHGLFRVRUJ
1248 19  Metastatic Tumors in Bone

A B

C D
FIGURE 19-25  ■  Metastatic melanoma and pancreatic carcinoma: cytologic features. A, Aspirates of melanoma showing epithelioid
cells with dense eosinophilic cytoplasm and eccentric nuclei. Inset, Melanoma cells with melanin deposits. B, Spindle-cell melanoma
with extensive melanin deposits. C and D, Crowded sheets of epithelial cells with high nuclear size variability among tumor cells.
(A-D, ×400; Inset, ×600.) (A-D and Inset, hematoxylin-eosin.) (Courtesy of Dr. Gregory Starkel, MD Anderson Cancer Center, Houston,
Texas.)

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1249

A B

C
FIGURE 19-26  ■  Lytic and osteoblastic metastases: radiographic features. A, Blowout destructive lytic lesion of second left rib (arrows)
confirmed by microscopic analysis to be metastatic carcinoma of thyroid. B, Computed tomogram of lesion in A documents destruc-
tive low-signal lesion in proximal portion of second left rib (arrows). Note residual patchy mineralization and thin line of expanded
periosteum. These features suggest that this metastatic carcinoma most likely contains a secondary aneurysmal bone cyst compo-
nent, which was confirmed microscopically. C, Anteroposterior radiograph of pelvis of a 9-year-old child shows multiple sclerotic
foci that represent widespread skeletal metastases of medulloblastoma.

ERRNVPHGLFRVRUJ
1250 19  Metastatic Tumors in Bone

molecular or molecular cytogenetic studies. Rhabdomyo- biopsy samples is frequently seen and can be documented
sarcoma must be differentiated from a variety of other in nearly 50% of neuroblastomas. On the other hand,
hematologic and nonhematologic round-cell malignan- radiographically demonstrable destructive bone lesions
cies. The skeleton is a relatively common site for meta- (Fig. 19-28) are seen less frequently in approximately
static rhabdomyosarcoma, and skeletal metastases in 25% of children with neuroblastoma. In typical cases, the
rhabdomyosarcoma are typically lytic167 (Fig. 19-27). presence of neuropil and ganglionic differentiation are
Diffuse involvement of bone with a permeative growth microscopic features diagnostic of neuroblastoma (Fig.
pattern similar to that seen in other small cell malignan- 19-29). Neuroblastoma serves as an example of a tumor
cies may be seen with alveolar rhabdomyosarcoma,182 and for which molecular studies can not only aid the diagno-
pediatric patients typically undergo bone marrow biopsy sis, but are also of prognostic value. Clinically aggressive
as a component of their staging studies. tumors with a high propensity for dissemination and less
Rhabdomyosarcoma is another example of a neoplasm favorable response to treatment, as well as presenting in
for which molecular techniques can be used as an adjunct an advanced stage, are characterized by MYCN amplifica-
in diagnosis. Similar to neuroblastoma, these data can tion, and loss of heterozygosity of 1p, 3p, and 11q, and
provide some prognostic information. Alveolar rhabdo- gains of 1q and 17q.160,162,163,175,178-180,188,190,195 MYCN also
myosarcoma shows consistent chromosomal transloca- regulates the expression of several micro-RNAs, which
tion t(2;13)(q35;q14), and less commonly t(1;13)(p36;q1 play a role in the pathogenesis of neuroblastoma.166 The
4).158,173,176,191,197,199,200 These cytogenetic abnormalities are PHOX2B and ALK genes have also been linked to familial
associated with the formation of PAX3-FOXO1 or PAX7- neuroblastomas. Low-risk neuroblastomas are character-
FOXO1 gene fusions.159,161,170,192 The PAX3-FOXO1 gene ized by hyperdiploidy or near triploidy, particularly in
fusion occurs nearly four times as frequently in alveolar patients younger than age 1.5 years.169
rhabdomyosarcoma and is associated with a worse prog- Aspirates from metastatic neuroblastoma are usually
nosis than those tumors that harbor a PAX7-FOXO1 very cellular and consist of small round cells occasionally
fusion.181 These abnormalities can be identified using arranged in clusters and lying singly. Formation of
either reverse transcriptase polymerase chain reaction or rosettes composed of spherically arranged nuclei with a
fluorescence in situ hybridization, valuable ancillary tests fibrillar center may also be present, but this is a rather
that can be used to differentiate rhabdomyosarcoma from rare finding in cytologic preparations. The presentation
other round-cell tumors.174 The amplification of the may be very similar to Ewing’s tumor (Fig. 19-25, B).
MYCN gene is not entirely specific for neuroblastoma
and can occur in rhabdomyosarcoma, but it is typically
seen only in the alveolar variant.172,196
Clear Cell Sarcoma of the Kidney
The smears from rhabdomyosarcoma are usually very Clear cell sarcoma of the kidney is a rare neoplasm
cellular and show a population of malignant small round that represents the third most common pediatric renal
cells with scanty cytoplasm and round or oval nuclei (Fig. neoplasm after Wilms’ tumor and renal cell carci-
19-24, C, D). The presence of larger cells with abundant noma.157,177,185,201 This tumor primarily affects young chil-
eosinophilic cytoplasm containing fibrils or showing dren and has a peak age incidence between age 2 and 3
cross-striations indicates rhabdomyoblastic differentia- years. Clear cell sarcoma of the kidney has a strong pro-
tion. However, rhabdomyoblasts displaying unequivocal pensity to metastasize to the skeleton.157,189,194 Multiple
cross-striation are difficult to find and, in most cases, lytic osseous metastases during the clinical course of this
immunohistochemical stains are necessary to classify the tumor represent a clinical hallmark of this rare tumor. In
lesion. fact, this unique feature was originally used to separate
this lesion from the Wilms’ tumor group. It frequently
Neuroblastoma presents with skeletal metastases and an occult primary
tumor. Microscopically, most tumors are composed of
Solitary or multiple, typically lytic, bone lesions are fre- nests of spindled or ovoid cells surrounded by fibrovas-
quent presentations of neuroblastoma. This tumor has a cular septa (Fig. 19-30); however, a small number of
unique predilection to metastasize to the orbit, jaws, and tumors may have variant patterns described as myxoid,
metaphyseal parts of long tubular bones.168 In fact, neu- sclerosing, cellular, epithelioid, palisading, spindle-cell,
roblastoma is a prototypic pediatric malignancy that can storiform, or anaplastic.157 Clear cell sarcomas do not
present as a solitary bone metastasis and a clinically occult have a characteristic immunohistochemical profile. A
primary lesion. This tumor has a predilection to metas- unique translocation—t(10;17)(q22;p13)—has been iden-
tasize to the orbit, jaws, and metaphyseal parts of long tified in clear cell sarcoma164,183,184,186,187; however, the
bones. The latter presentation, due to similarity in clini- molecular characteristics of the breakpoints have not
cal symptoms, can be confused with osteomyelitis. In been defined.
typical cases, the presence of intercellular fibrillar matrix Aspirates from metastatic clear cell sarcoma of the
separating the loosely arranged tumor cell nuclei is a kidney are usually cellular and show a large population
helpful feature in differentiating neuroblastomas from of pleomorphic sarcomatous cells. Deep nuclear indenta-
other round-cell malignancies. The changes in the long tions and nuclear grooves, as well as a granular cytoplasm,
tubular bones can be similar to those seen with acute are characteristic features. In some cases, aspirates may
leukemia, or they may be associated with periosteal reac- show biphasic pattern with pleomorphic sarcomatous
tions and can be radiographically confused with osteomy- cells admixed with spindle-shaped cells.
elitis.156 Discrete diffuse involvement of bone marrow in Text continued on p. 1255

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1251

A
B

C D

FIGURE 19-27  ■  Metastatic rhabdomyosarcoma in bone: radiographic, gross, and histologic features. A, Anteroposterior radiograph
showing a destructive lytic lesion with pathologic fracture. B, Coronally bisected gross photograph of the resected femur shown in
A with a destructive ill-defined fleshy tumor of the distal femur. Note cystic change within the center of the lesion filled with muci-
nous fluid related to treatment effect. C and D, Low power photomicrograph showing microscopic features of metastatic rhabdo-
myosarcoma infiltrating fatty marrow. Insets show microscopic details of metastatic rhabdomyosarcoma with maturation effect
induced by chemotherapy in C. (C and D, ×100; Insets, ×200.) (C, D and inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1252 19  Metastatic Tumors in Bone

C D
FIGURE 19-28  ■  Metastatic neuroblastoma: radiographic features. A, Lateral radiograph of the spine showing an osteoblastic lesion
in the S1 portion of the sacrum. B, Axial computed tomography scan of the spine through the S1 lesion seen in A. C, T1-weighted
sagittal magnetic resonance image (MRI) shows only the metastasis in S1, which appears dark (very low signal intensity).
D, However, T1-weighted sagittal postcontrast MRI reveals multiple lesions in the lower thoracic and lumbar vertebral bodies. Inset,
Radioisotope scan shows multiple areas of increased uptake in the lower spine.

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1253

B
FIGURE 19-29  ■  Metstatic neuroblastoma: histologic and radiographic features. A and B, Photomicrographs show nests of round
cells deposited in an eosinophilic, fibrillary matrix. Ancillary tests are also useful in differentiating metastatic neuroblastoma from
other round cell malignancies. Inset, Axial computed tomography scan showing a metastatic neuroblastoma deposit within the
calvarium (A, ×200; B, ×400) (A and B, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1254 19  Metastatic Tumors in Bone

B
FIGURE 19-30  ■  Metastatic clear cell sarcoma of kidney: microscopic features. A and B, Solid proliferation of round primitive cells
with clear cytoplasm is characteristic of this tumor (A, ×200; B, ×400) (A and B, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1255

REFERENCES 28. Cramer SF, Fried L, Carter KJ: The cellular basis of metastatic
bone disease in patients with lung cancer. Cancer 48:2649–2660,
Biology of Metastases 1981.
1. Akhtari M, Mansuri J, Newman KA, et al: Biology of breast cancer 29. Didolkar MS, Fanous N, Elias EG, et al: Metastatic carcinomas
bone metastasis. Cancer Biol Ther 7:3–9, 2008. from occult primary tumors. Ann Surg 186:625–630, 1977.
2. Anborgh PH, Mutrie JC, Tuck AB, et al: Role of the metastasis- 30. Eilon G, Mundy GR: Direct resorption of bone by human breast
promoting protein osteopontin in the tumour microenvironment. cancer cells in vitro. Nature 276:726–728, 1978.
J Cell Mol Med 14:2037–2044, 2010. 31. Galasko CS: Mechanisms of lytic and blastic metastatic disease of
3. Bernstein LR, Liotta LA: Molecular mediators of interactions bone. Clin Orthop 169:20–27, 1982.
with extracellular matrix components in metastasis and angiogen- 32. Ghandur-Mnaymneh L, Broder LE, Mnaymneh WA: Lobular
esis. Curr Opin Oncol 6:106–113, 1994. carcinoma of the breast metastatic to bone with unusual clinical,
4. Giancotti FG, Mainiero F: Integrin-mediated adhesion and sig- radiologic, and pathologic features mimicking osteopoikilosis.
naling in tumorigenesis. Biochim Biophys Acta 1198:47–64, 1994. Cancer 53:1801–1803, 1984.
5. Guise TA, Yin JJ, Taylor SD, et al: Evidence for a causal role of 33. Goudarzi B, Kishimoto R, Komatsu S, et al: Detection of bone
parathyroid hormone-related protein in the pathogenesis of metastases using diffusion weighted magnetic resonance imaging:
human breast cancer-mediated osteolysis. J Clin Invest 98:1544– comparison with 11C-methionine PET and bone scintigraphy.
1549, 1996. Magn Reson Imaging 28:372–379, 2010.
6. Hiratsuka S, Watanabe A, Aburatani H, et al: Tumour-mediated 34. Healey JH, Turnbull ADM, Miedema B, et al: Acrometastases: a
upregulation of chemoattractants and recruitment of myeloid cells study of twenty-nine patients with osseous involvement of the
predetermines lung metastasis. Nat Cell Biol 10:1349–1355, 2008. hands and feet. J Bone Joint Surg 68A:743–746, 1986.
7. Hofbauer LC, Neubauer A, Heufelder AE: Receptor activator of 35. Hortobagyi GN, Theriault RL, Porter L, et al: Efficacy of pami-
nuclear factor-κB ligand and osteoprotegerin: potential implica- dronate in reducing skeletal complications in patients with breast
tions for the pathogenesis and treatment of malignant bone dis- cancer and lytic bone metastases. Protocol 19 Aredia Breast
eases. Cancer 92:460–470, 2001. Cancer Study Group. N Engl J Med 335:91–1996, 1785.
8. Hynes RO: Integrins: bidirectional, allosteric signaling machines. 36. Jaffe HL: Tumors and tumorous conditions of the bones and joints,
Cell 110:673–687, 2002. Philadelphia, 1958, Lea & Febiger.
9. Kaplan RN, Riba RD, Zacharoulis S, et al: VEGFR1-positive 37. Joo KG, Parthasarathy KL, Bakshi SP, et al: Bone scintigrams:
haematopoietic bone marrow progenitors initiate the pre- their clinical usefulness in patients with breast carcinoma. Oncology
metastatic niche. Nature 438:820–827, 2005. 36:94–98, 1979.
10. Mundy GR: Metastasis to bone: causes, consequences and thera- 38. Knapp D, Abdul-Karim FW: Fine needle aspiration cytology
peutic opportunities. Nat Rev Cancer 2:584–593, 2002. of acrometastasis: a report of two cases. Acta Cytol 38:589–591,
11. Peters S, Meylan E: Targeting receptor activator of nuclear 1994.
factor-κ B as a new therapy for bone metastasis in non-small cell 39. Koskinen EV, Nieminen RA: Surgical treatment of metastatic
lung cancer. Curr Opin Oncol 25:137–144, 2013. pathological fracture of major long bones. Acta Orthop Scand
12. Psaila B, Lyden B: The metastatic niche: adapting the foreign soil. 44:539–549, 1973.
Nat Rev Cancer 9:285–293, 2009. 40. Koutsilieris M: Osteoblastic metastasis in advanced prostate
13. Roodman GD: Mechanisms of bone metastasis. N Engl J Med cancer. Anticancer Res 13:443–449, 1993.
350:1655–1664, 2004. 41. Libson E, Bloom RA, Husband JE, et al: Metastatic tumors of the
14. Scagliotti GV, Hirsh V, Siena S, et al: Overall survival improve- bones of the hand and foot: a comparative review and report of
ment in patients with lung cancer and bone metastases treated 43 additional cases. Skeletal Radiol 16:387–392, 1987.
with denosumab versus zoledronic acid: subgroup analysis from a 42. Norman A, Ulin R: A comparative study of periosteal new-bone
randomized phase 3 study. J Thorac Oncol 7:1823–1829, 2012. response in metastatic bone tumors (solitary) and primary sarco-
15. Schneider JG, Amend SR, Weilbaecher KN: Integrins and bone mas. Radiology 92:705–708, 1969.
metastasis: integrating tumor cell and stromal cell interactions. 43. Ozarda AT: Bilateral symmetrical metastatic osseous lesions. Am
Bone 48:54–65, 2011. Surg 31:56–57, 1965.
16. Sleeman JP: The metastatic niche and stromal progression. Cancer 44. Panebianco AC, Kaupp HA: Bilateral thumb metastasis from
Metastasis Rev 31:429–440, 2012. breast carcinoma. Arch Surg 96:216–218, 1968.
17. Stetler-Stevenson WG, Aznavoorian S, Liotta LA: Tumor cell 45. Pollack HM: Magnetic resonance imaging of bone metastases.
interactions with the extracellular matrix during invasion and Adv Exp Med Biol 324:205–208, 1992.
metastasis. Annu Rev Cell Biol 9:541–573, 1993. 46. Powell JM: Metastatic carcinoid of bone: report of two cases and
18. Weilbaecher KN, Guise TA, McCauley LK: Cancer to bone: a review of the literature. Clin Orthop 230:266–272, 1988.
fatal attraction. Nat Rev Cancer 11:411–425, 2011. 47. Rougraff BT: Evaluation of a patient with carcinoma of unknown
19. Yu Q, Toole BP, Stamenkovic I: Induction of apoptosis of meta- origin metastatic to bone. Clin Orthop Rel Res 415S:S105–S109,
static mammary carcinoma cells in vivo by disruption of tumor 2003.
cell surface CD44. J Exp Med 186:1985–1996, 1997. 48. Rougraff BT, Kneisl JS, Simon MA: Skeletal metastases of
20. Ziober BL, Lin CS, Kramer RH: Laminin-binding integrins in unknown origin: a prospective study of a diagnostic strategy.
tumor progression and metastasis. Semin Cancer Biol 7:119–128, J Bone Joint Surg 75A:1276–1281, 1993.
1996. 49. Scher HI, Chung LW: Bone metastases: improving the therapeu-
tic index. Semin Oncol 21:630–656, 1994.
General Features of Skeletal Metastases 50. Simon MA, Bartucci EJ: The search for the primary tumor
21. Abrams HL, Spiro R, Goldstein N: Metastases in carcinoma: in patients with skeletal metastases of unknown origin. Cancer
analysis of 1000 autopsied cases. Cancer 3:74–85, 1950. 58:1088–1095, 1986.
22. Assor D: Basal cell carcinoma with metastasis to bone: report of 51. Takenaka D, Ohno Y, Matsumoto K, et al: Detection of bone
two cases. Cancer 20:2125–2132, 1967. metastases in non-small cell lung cancer patients: comparison
23. Barr LC, Skene AI, Thomas JM: Metastasectomy. Br J Surg of whole-body diffusion-weighted imaging (DWI), whole-body
79:1268–1274, 1992. MR imaging without and with DWI, whole-body FDG-PET/
24. Batson OV: The function of the vertebral veins and their role in CT, and bone scintigraphy. J Magn Reson Imaging 30:298–308,
the spread of metastases. Ann Surg 112:138–149, 1940. 2009.
25. Berrettoni BA, Carter JR: Mechanisms of cancer metastasis to 52. Wu Y, Li P, Zhang H, et al: Diagnostic value of fluorine 18 fluo-
bone. J Bone Joint Surg 68A:308–312, 1986. rodeoxyglucose positron emission tomography/computed tomog-
26. Bloom RA, Libson E, Husband JE, et al: The periosteal sunburst raphy for the detection of metastases in non-small-cell lung cancer
reaction to bone metastases: a literature review and report of 20 patients. Int J Cancer 132:E37–E47, 2012.
additional cases. Skeletal Radiol 16:629–634, 1987. 53. Young JM, Funk FJ, Jr: Incidence of tumor metastasis to the
27. Borak J: Relationship between the clinical and roentgenological lumbar spine: a comparative study of roentgenographic changes
findings in bone metastases. Surg Gynecol Obstet 75:599–604, 1942. and gross lesions. J Bone Joint Surg 35A:55–64, 1953.

ERRNVPHGLFRVRUJ
1256 19  Metastatic Tumors in Bone

Skeletal Metastases in Selected Common Neoplasms 82. Blastic M, Plavecz E, Zalatnai A: Pancreatic carcinomas in a
54. Berg JW, Hutter RVP: Breast cancer. Cancer 75S:257–269, 1995. 60-year, institute-based autopsy material with special emphasis of
55. Bosman FT: Prognostic value of pathological characteristics of metastatic pattern. Pancreas 40:478–480, 2011.
colorectal cancer. Eur J Cancer 31:1216–1221, 1995. 83. Borad MJ, Saadti H, Lakshmipathy A, et al: Skeletal metastases
56. Choi CW, Lee DS, Chung JK, et al: Evaluation of bone metasta- in pancreatic cancer: a retrospective study and review of the litera-
ses by Tc-99m MDP imaging in patients with stomach cancer. Clin ture. Yale J Biol Med 82:1–6, 2009.
Nucl Med 20:310–314, 1995. 84. Brady D, Parker CC, O’Sullivan JM: Bone-targeting radiophar-
57. Costelloe CM, Rohren EM, Madewell JE, et al: Imaging bone maceuticals including Radium-223. Cancer J 19:71–78, 2013.
metastases in breast cancer: techniques and recommendations for 85. Bubendorf L, Schopfer A, Wagner U, et al: Metastatic patterns of
diagnosis. Lancet Oncol 10:606–614, 2009. prostate cancer: an autopsy study of 1589 patients. Hum Pathol
58. Diel IJ, Kaufmann M, Costa SD, et al: Monoclonal antibodies to 31:578–583, 2000.
detect breast cancer cells in bone marrow. Important Adv Oncol 86. Carriaga MT, Henson DE: Liver, gallbladder, extrahepatic bile
143–164, 1994. ducts, and pancreas. Cancer 75S:171–190, 1995.
59. Ezra E, Vardy S, Rose G: Metastatic colon adenocarcinoma of the 87. Cubilla AL, Fortner J, Fitzgerald PJ: Lymph node involvement in
orbit with intraneural extension from the brow to the brainstem. carcinoma of the head of the pancreas areas. Cancer 41:880–887,
Eye 9:371–372, 1995. 1978.
60. Hamaoka T, Madewell JE, Podoloff DA, et al: Bone imaging in 88. DeAntoni EP, Crawford ED: Pretreatment of metastatic disease:
metastatic breast cancer. J Clin Oncol 22:2942–2953, 2004. prostate cancer in the older male. Cancer 74S:2182–2187, 1994.
61. Jardines L, Callans LS, Torosian MH: Recurrent breast cancer: 89. Durr HR, Maier M, Pfahler M, et al: Surgical treatment of
presentation, diagnosis, and treatment. Semin Oncol 20:538–547, osseous metastases in patients with renal cell carcinoma. Clin
1993. Orthop Rel Res 367:283–290, 1999.
62. Katoh M, Unakami M, Hara M, et al: Bone metastasis from 90. Farooki A, Leung V, Tala H, et al: Skeletal-related events due to
colorectal cancer in autopsy cases. J Gastroenterol 30:615–618, bone metastases from differentiated thyroid cancer. J Clin Endo-
1995. crinol Metab 97:2433–2439, 2012.
63. Krawczyk P, Nicoś M, Ramlau R, et al: The incidence of EGFR- 91. Fidler IJ: The biology of renal cancer metastasis. Semin Urol
activating mutations in bone metastases of lung adenocarcinoma. 10:3–11, 1992.
Pathol Oncol Res 2013. [epub ahead of print]. 92. Fottner A, Szalantzy M, Wirthmann L, et al: Bone metastases
64. Leonard RC, Rodger A, Dixon JM: ABC of breast diseases: meta- from renal cell carcinoma: patient survival after surgical treat-
static breast cancer. Br Med J 309:1501–1504, 1994. ment. BMC Musculoskelet Disord 11:145, 2010.
65. MacAfee KA, II, Quinn PD, Abaza NA: Adenocarcinoma of the 93. Franceschi S, La Vecchia C: Thryoid cancer. Cancer Surv 19-
colon metastatic to the temporomandibular joint: a case report. 20:393–422, 1994.
J Oral Maxillofac Surg 51:793–797, 1993. 94. Freeman JL, Gershon A, Liavaag PG, et al: Papillary thyroid
66. Miller F, Whitehill R: Carcinoma of the breast metastatic to the carcinoma metastasizing to the sphenoid-ethmoid sinuses and
skeleton. Clin Orthop 184:121–127, 1984. skull base. Thyroid 6:59–61, 1996.
67. Mohandas KM, Swaroop VS, Krishnamurthy S, et al: Unusual 95. Gilliland FD, Key CR: Male genital cancers. Cancer 75S:295–315,
bone metastasis as the initial symptom of gastric cancer: a report 1995.
of four cases. Indian J Cancer 30:146–150, 1993. 96. Goyal J, Antonarakis ES: Bone-targeting radiopharmaceuticals for
68. Mootoosamy IM, Anchor SC, Dacie JE: Expanding osteolytic the treatment of prostate cancer with bone metastases. Cancer Lett
bone metastases from carcinoma of the breast: an unusual appear- 323:135–146, 2012.
ance. Skeletal Radiol 14:188–190, 1985. 97. Hefer T, Joachims HZ, Hashmonai M, et al: Highly aggressive
69. Nozue M, Oshiro Y, Kurata M, et al: Treatment and prognosis in behaviour of occult papillary thyroid carcinoma. J Laryngol Otol
colorectal cancer patients with bone metastasis. Oncol Rep 9:109– 109:1109–1112, 1995.
112, 2002. 98. Henriksson C, Haraldsson G, Aldenborg F, et al: Skeletal metas-
70. Patel AM, Peters SG: Clinical manifestations of lung cancer. Mayo tases in 102 patients evaluated before surgery for renal cell carci-
Clin Proc 68:273–277, 1993. noma. Scand J Urol Nephrol 26:363–366, 1992.
71. Pauzner R, Istomin V, Segal-Lieberman G, et al: Bilateral patellar 99. Holt BA, Holmes SA, Kirby RS: Renal cell carcinoma presenting
metastases as the clinical presentation of bronchogenic adenocar- with orbital metastases. Br J Urol 75:246–247, 1995.
cinoma. J Rheumatol 23:939–941, 1996. 100. Ibayashi K, Ando M, Gotoh E: Regression of pulmonary and
72. Rodriguez-Garcia JL, Martinez-San-Millan J, Cuesta C, et al: multiple skeletal metastases from renal cell carcinoma by nephrec-
Colorectal cancer presenting as isolated skull metastasis. Ann tomy and alpha-interferon therapy: a case report. Jpn J Clin Oncol
Oncol 3:321–322, 1992. 23:378–383, 1993.
73. Santini D, Tampellini M, Vincenzi B, et al: Natural history of 101. Inci S, Akbay A, Bertan V, et al: Solitary skull metastasis from
bone metastasis in colorectal cancer: final results of a large Italian occult thyroid carcinoma. J Neurosurg Sci 38:63–66, 1994.
bone metastases study. Ann Oncol 23:2072–2077, 2012. 102. Ito Y, Kudo T, Kobayashi K, et al: Prognostic factors for recur-
74. Schmidt RG, Cecchini AJ, Mayer DP, et al: Magnetic resonance rence of papillary thyroid carcinoma in the lymph nodes, lung,
diagnosis of an occult metastatic colon carcinoma of the calcaneus. and bone: analysis of 5768 patients with an average 10-year
Foot Ankle Int 15:334–339, 1994. follow-up. World J Surg 36:1274–1278, 2012.
75. Thomas RM, Sobin LH: Gastrointestinal cancer. Cancer 75S:154– 103. Jacobsen KD, Folleras G, Fossa SD: Metastases from renal cell
170, 1995. carcinoma to the humerus or the shoulder girdle. Br J Urol
76. Travis WD, Travis LB, Devesa SS: Lung cancer. Cancer 75S:191– 73:124–128, 1994.
202, 1995. 104. Joffe N, Antonioli DA: Osteoblastic bone metastases secondary to
77. Trillet-Lenoir VN, Arpin D, Brune J: Bone marrow metastases adenocarcinoma of the pancreas. Clin Radiol 29:41–46, 1978.
detection in small cell lung cancer: a review. Anticancer Res 105. Kasagi K, Takeuchi R, Miyamoto S, et al: Metastatic thyroid
14:2795–2797, 1994. cancer presenting as thyrotoxicosis: report of three cases. Clin
78. Urvoy P, Mestdagh H, Butin E, et al: Patellar metastasis from a Endocrinol (Oxf) 40:429–434, 1994.
large bowel adenocarcinoma. Acta Orthop Belg 59:409–411, 1993. 106. Kerbl K, Pauer W: Spontaneous regression of osseous metastasis
79. Yoneda T, Sasaki A, Mundy GR: Osteolytic bone metastasis in in renal cell carcinoma. Aust NZ J Surg 63:901–903, 1993.
breast cancer. Breast Cancer Res Treat 32:73–84, 1994. 107. Kobus RJ, Leinberry C, Kirkpatrick WH: Metastatic renal carci-
noma in the hand: treatment with preoperative irradiation and ray
resection. Orthop Rev 21:983–984, 990–995, 1992.
Tumors with a Unique Propensity for Skeletal Metastases 108. Koranda P, Ryznar V, Dockal M, et al: Papillary adenocarcinoma
80. Barry MJ: Natural history of clinically localized prostate cancer. of the thyroid gland metastasizing into the epidural space of the
Semin Surg Oncol 11:3–8, 1995. upper thoracic spine, without vertebral involvement: case report.
81. Bichler KH, Petri E, Hub A, et al: Investigations on bone metabo- Acta Univ Palacki Olomuc Fac Med 135:31–32, 1993.
lism of urological tumors forming metastases. Urol Int 56:61–68, 109. Kosuda S, Yokoyama H, Katayama M, et al: Technetium-99m
1996. tetrofosmin and technetium-99m sestamibi imaging of multiple

ERRNVPHGLFRVRUJ
19  Metastatic Tumors in Bone 1257

metastases from differentiated thyroid carcinoma. Eur J Nucl Med 136. Wang W, Epstein JI: Small cell carcinoma of the prostate: a mor-
22:1218–1220, 1995. phologic and immunohistochemical study of 95 cases. Am J Surg
110. Kozlowski JM: Management of distant solitary recurrence in the Pathol 32:65–71, 2008.
patient with renal cancer: contralateral kidney and other sites. Urol 137. Wolin MJ: Metastatic renal cell carcinoma manifesting as an
Clin North Am 21:601–624, 1994. orbital mass. Am J Ophthalmol 115:542–543, 1993 (letter).
111. Loree TR: Therapeutic implications of prognostic factors in dif- 138. Woodward E, Jagdev S, McParland L, et al: Skeletal complica-
ferentiated carcinoma of the thyroid gland. Semin Surg Oncol tions and survival in renal cell carcinoma patients with bone
11:246–255, 1995. metastases. Bone 48:160–166, 2011.
112. Margo CE, Levy MH: Orbital metastasis from medullary carci- 139. Yamasoba T, Kikuchi S, Sugasawa M, et al: Occult follicular car-
noma of the thyroid. Am J Ophthalmol 115:394–395, 1993. cinoma metastasizing to the sinonasal tract. ORL J Otorhinolaryn-
113. Muresan MM, Olivier P, Leclere J, et al: Bone metastases from gol Relat Spec 56:239–243, 1994.
differentiated thyroid carcinoma. Endocr Relat Cancer 15:37–49, 140. Yen TC, King KL, Yang AH, et al: Comparative radionuclide
2008. imaging of metastatic insular carcinoma of the thyroid: value of
114. Nakajima T, Suzuki M, Ando S, et al: Spontaneous regression of technetium-99m-(V)DMSA. J Nucl Med 37:78–80, 1996.
bone metastasis from renal cell carcinoma: a case report. BMC 141. Zetter BR, Chackal-Roy M, Smith R: The cellular basis for pros-
Cancer 13:6, 2006. tate cancer metastasis. Adv Exp Med Biol 324:39–43, 1992.
115. Osborn JL, Getzenberg RH, Trump DL: Spinal cord compression 142. Zhau HE, Pisters LL, Hall MC, et al: Biomarkers associated with
in prostate cancer. J Neurooncol 23:135–147, 1995. prostate cancer progression. J Cell Biochem 19:208–216, 1994.
116. Ozaki O, Kitagawa W, Koshiishi H, et al: Thyroid carcinoma 143. Zohar Y, Strauss M: Occult distant metastases of well-differentiated
metastasized to the sternum: resection of the sternum and recon- thyroid carcinoma. Head Neck 16:438–442, 1994.
struction with acrylic resin. J Surg Oncol 60:282–285, 1995.
117. Pneumaticos SG, Savidou C, Korres DS, et al: Pancreatic cancer’s Metastatic Melanoma
initial presentation: back pain due to osteoblastic bone metastasis. 144. Beadling C, Jacobson-Dunlop E, Hodi FS, et al: KIT gene muta-
Eur J Cancer Care (Engl) 19:137–140, 2010. tions and copy number in melanoma subtypes. Clin Cancer Res
118. Redleaf MI, Bauman NM, Robinson RA, et al: Medullary carci- 14:6821–6828, 2008.
noma of the thyroid metastatic to the temporal bone. J Laryngol 145. DeBoer DK, Schwartz HS, Thelman S, et al: Heterogeneous sur-
Otol 109:1200–1203, 1995. vival rates for isolated skeletal metastases from melanoma. Clin
119. Rinker-Schaeffer CW, Partin AW, Isaacs WB, et al: Molecular Orthop Rel Res 323:277–283, 1996.
and cellular changes associated with the acquisition of metastatic 146. Desy NM, Bernstein M, Nahal A, et al: Primary perivascular
ability by prostatic cancer cells. Prostate 25:249–265, 1994. epithelioid cell neoplasm (PEComa) of bone: report of two
120. Robbins J: Prognostic factors in the management of thyroid cases and review of the literature. Skeletal Radiol 41:1469–1474,
cancer. J Endocrinol Invest 18:159–160, 1995. 2012.
121. Saif MW, Galanina N, Ravage-Mass L, et al: Bone metastasis as 147. Deyrup AT, Montag AG: Epithelioid and epithelial neoplasms of
the only metastatic site in a patient with pancreatic cancer follow- bone. Arch Pathol Lab Med 131:205–216, 2007.
ing distal pancreatectomy. Case Rep Med 2010. Article ID 634975. 148. Fon GT, Wong WS, Gold RH, et al: Skeletal metastases of mela-
122. Saitoh H: Distant metastasis of renal adenocarcinoma. Cancer noma: radiologic, scintigraphic, and clinical review. AJR 137:103–
48:1487–1491, 1981. 108, 1981.
123. Schlumberger M, Challeton C, De Vathaire F, et al: Radioactive 149. Long GV, Menzies AM, Nagrial AM, et al: Prognostic and clini-
iodine treatment and external radiotherapy for lung and bone copathologic associations of oncogenic BRAF in metastatic mela-
metastases from thyroid carcinoma. J Nucl Med 37:598–605, 1996. noma. J Clin Oncol 29:1239–1246, 2011.
124. Schroder F, Tribukait B, Bocking A, et al: Clinical utility of cel- 150. McMenamin DS, Stuckey SL, Potgieter GJ: T1 hyperintense
lular DNA measurements in prostate carcinoma: consensus con- vertebral column melanoma metastases. Am J Neuroradiol 28:
ference on diagnosis and prognostic parameters in localized 1817–1818, 2007.
prostate cancer, Stockholm, Sweden, May 12-13, 1993. Scand J 151. Muller-Horvat C, Radny P, Eigentler TK, et al: Prospective com-
Urol Nephrol 162:51–63, 1994. parison of the impact on treatment decisions of whole-body mag-
125. Sesenna E, Tullio A, Piazza P: Treatment of craniofacial metastasis netic resonance imaging and computed tomography in patients
of a renal adenocarcinoma: report of case and review of literature. with metastatic malignant melanoma. Eur J Cancer 42:342–350,
J Oral Maxillofac Surg 53:187–193, 1995. 2006.
126. Simpson H, Orr DJ, John PJ, et al: Follicular carcinoma of 152. Premkumar A, Marincola F, Taubenberger J, et al: Metastatic
thyroid presenting as back pain leading to a delay in diagnosis. Br melanoma: correlation of MRI characteristics and histopathology.
J Clin Pract 48:334–336, 1994. J Magn Reson Imaging 1:190–194, 1996.
127. Smith EM, Kursh ED, Makley J, et al: Treatment of osseous 153. Rivera RD, Diamante M, Kasten SJ, et al: Metastatic melanoma
metastases secondary to renal cell carcinoma. J Urol 148:784–787, to the mandible: case report and review of the literature. J Oral
1992. Maxillofac Surg 68:2903–2906, 2010.
128. Sneag DB, Krajewski KM, Howard S, et al: Sclerotic osseous 154. Stewart WR, Gelberman RH, Harrelson JM, et al: Skeletal
metastases from renal cell carcinoma. Skeletal Radiol 41:1169– metastases of melanoma. J Bone Joint Surg 60A:645–649, 1978.
1175, 2012. 155. Yamashita K, Fletcher CDM: PEComa presenting in bone: clini-
129. Szendroi A, Dinya E, Kardos M, et al: Prognostic factors and copathologic analysis of 6 cases and literature review. Am J Surg
survival of renal clear cell carcinoma patients with bone metasta- Pathol 34:1622–1629, 2010.
ses. Pathol Oncol Res 16:29–38, 2010.
130. Takashi M, Takagi Y, Sakata T, et al: Surgical treatment of renal Skeletal Metastases in Selected Pediatric Tumors
cell carcinoma metastases: prognostic significance. Int Urol 156. Applegate K, Connolly LP, Treves ST: Neuroblastoma presenting
Nephrol 27:1–8, 1995. clinically as hip osteomyelitis: a “signature” diagnosis on skeletal
131. Tongaonkar HB, Kulkarni JN, Kamat MR: Solitary metastases scintigraphy. Pediatr Radiol 25(Suppl 1):S93–S96, 1995.
from renal cell carcinoma: a review. J Surg Oncol 49:45–48, 1992. 157. Argani P, Perlman EJ, Breslow NE, et al: Clear cell sarcoma of
132. Van Poppel H, Baert L: Nephrectomy for metastatic renal cell the kidney. A review of 351 cases from the National Wilms Tumor
carcinoma and surgery for distant metastases. Acta Urol Belg Study Group Pathology Center. Am J Surg Pathol 24:4–18, 2000.
64:11–17, 1996. 158. Barr FG, Biegel JA, Sellinger B, et al: Molecular and cytogenetic
133. Vanderpump MP, Tunbridge WM: Hurthle cell carcinoma pre- analysis of chromosomal arms 2q and 13q in alveolar rhabdomyo-
senting with retroorbital metastasis. J R Soc Med 85:493–494, sarcoma. Gene Chromosomes Cancer 3:153–161, 1991.
1992. 159. Barr FG, Galili N, Holick J, et al: Rearrangement of the PAX3
134. Vassilopoulou-Sellin R: Management of papillary thyroid cancer. paired box gene in the paediatric solid tumour alveolar rhabdo-
Oncology 9:145–151, 1995. myosarcoma. Nat Genet 3:113–117, 1993.
135. Wahner-Roedler DL, Sebo TJ: Renal cell carcinoma: diagnosis 160. Benard J: Genetic alterations associated with metastatic dissemi-
based on metastatic manifestations. Mayo Clin Proc 72:935–941, nation and chemoresistance in neuroblastoma. Eur J Cancer
1997. 31A:560–564, 1995.

ERRNVPHGLFRVRUJ
1258 19  Metastatic Tumors in Bone

161. Biegel JA, Nycum LM, Valentine V, et al: Detection of the t(2;13) 182. Nishida Y, Tsukushi S, Urakawa H, et al: High incidence of
(q35;q14) and PAX3-FKHR fusion in alveolar rhabdomyosarcoma regional and in-transit lymph node metastasis in patients with
by fluorescence in situ hybridization. Gene Chromosomes Cancer alveolar rhabdomyosarcoma. Int J Clin Oncol 2013. [epub ahead
12:186–192, 1995. of print].
162. Brodeur GM, Seeger RC, Schwab M, et al: Amplification of 183. Punnett HH, Halligan GE, Zaeri N, et al: Translocation 10;17 in
N-myc in untreated human neuroblastomas correlates with clear cell sarcoma of the kidney: a first report. Cancer Genet Cyto-
advanced disease stage. Science 224:1121–1124, 1984. genet 41:123–128, 1989.
163. Brodeur GM, Sekhon GS, Goldstein MN: Chromosomal aberra- 184. Rakheja D, Weinberg AG, Tomlinson GE, et al: Translocation
tions in human neuroblastomas. Cancer 40:2256–2263, 1977. (10;17)(q22;p13): a recurring translocation in clear cell sarcoma
164. Brownlee NA, Perkins LA, Stewart W, et al: Recurring transloca- of the kidney. Cancer Genet Cytogenet 154:175–179, 2004.
tion (10;17) and deletion (14q) in clear cell sarcoma of the kidney. 185. Ries LAG, Smith MA, Gurney JG, et al, editors: Cancer incidence
Arch Pathol Lab Med 131:446–451, 2007. and survival among children and adolescents: United States SEER
165. Brutschin P, Culver GJ: Extracranial metastases from medullo- program 1975-1995, National Cancer Institute, SEER Program. NIH
blastoma. Radiology 107:359–362, 1973. Pub. No. 99-4649, Bethesda, MD, 1999.
166. Buechner J, Einvik C: N-myc and noncoding RNAs in neuroblas- 186. Royer-Pokora B: Genetics of pediatric renal tumors. Pediatr
toma. Mol Cancer Res 10:1243–1253, 2012. Nephrol 28:13–23, 2013.
167. Caffey J, Andersen DH: Metastatic embryonal rhabdomyosar- 187. Schuster AE, Schneider DT, Fritsch MK, et al: Genetic and
coma in the growing skeleton. Am J Dis Child 95:581, 1958. genetic expression analyses of clear cell sarcoma of the kidney. Lab
168. D’Ambrosio N, Lyo J, Young R, et al: Common and unusual Invest 83:1293–1299, 2003.
craniofacial manifestations of metastatic neuroblastoma. Neurora- 188. Schwab M, Varmus HE, Bishop JM, et al: Chromosome localiza-
diology 52:549–553, 2010. tion in normal human cells and neuroblastomas of a gene related
169. Davenport KP, Blanco FC, Sandler AD: Pediatric malignancies. to c-myc. Nature 308:288–291, 1984.
Neuroblastoma, Wilm’s tumor, hepatoblastoma, rhabdomyosar- 189. Sebire NJ, Vujanic GM: Paediatric renal tumours: recent develop-
coma, and sacrococcygeal teratoma. Surg Clin North Am 92:745– ments, new entities and pathological features. Histopathology 54:
767, 2012. 516–528, 2009.
170. Davis RJ, D’Cruz CM, Lovell MA, et al: Fusion of PX7 to FKHR 190. Seeger RC, Brodeur GM, Sather H, et al: Association of multiple
by the variant t(1;13)(p36;q14) translocation in alveolar rhabdo- copies of the N-myc oncogene with rapid progression of neuro-
myosarcoma. Cancer Res 54:2869–2872, 1994. blastomas. N Engl J Med 313:1111–1116, 1985.
171. Debnam JW, Staple TW: Osseous metastases from cerebellar 191. Seidal T, Mark J, Hagmar B, et al: Alveolar rhabdomyosarcoma:
medulloblastoma. Radiology 107:363–365, 1973. a cytogenetic and correlated cytological and histological study.
172. Dias P, Kumar P, Marsden HB, et al: N-myc gene is amplified in Acta Pathol Microbiol Immunol Scand 90:345–354, 1982.
alveolar rhabdomyosarcoma (RMS) but not in embryonal RMS. 192. Shapiro DN, Sublett JE, Li B, et al: Fusion of PAX3 to a member
Int J Cancer 45:593–596, 1990. of the fork head family of transcription factors in human alveolar
173. Douglass EC, Valentine M, Etcubanas E, et al: A specific chro- rhabdomyosarcoma. Cancer Res 53:5108–5112, 1993.
mosomal abnormality in rhabdomyosarcoma. Cytogenet Cell Genet 193. Skapek SX, Anderson J, Barr FG, et al: PAX-FOXO1 fusion status
45:148–155, 1987. drives unfavorable outcome for children with rhabdomyosarcoma:
174. Downing JR, Khandekar A, Shurtleff SA, et al: Multiplex RT-PCR a Children’s Oncology Group report. Pediatr Blood Cancer 60:
assay for the differential diagnosis of alveolar rhabdomyosarcoma 1411–1417, 2013.
and Ewing’s sarcoma. Am J Pathol 146:626–634, 1995. 194. Sotelo-Avila C, Gonzalez-Crussi F, Sadowinski S, et al: Clear cell
175. Fong CT, Dracopoli NC, White PS, et al: Loss of heterozygosity sarcoma of the kidney: a clinicopathologic study of 21 patients
for the short arm of chromosome 1 in human neuroblastomas: with long-term follow-up evaluation. Hum Pathol 16:1219–1230,
correlation with N-myc amplification. Proc Natl Acad Sci U S A 1985.
86:3753–3757, 1989. 195. Sridhar S, Al-Moallem B, Kamal H, et al: New insights into the
176. Galili N, Davis RJ, Fredericks WJ, et al: Fusion of a fork head genetics of neuroblastoma. Implications for diagnosis and therapy.
domain gene to PAX3 in the solid tumour alveolar rhabdomyo- Mol Diagn Ther 17:63–69, 2013.
sarcoma. Nat Genet 5:230–235, 1993. 196. Tonelli R, McIntyre A, Camerin C, et al: Antitumor activity of
177. Gooskens SLM, Furtwangler R, Vujanic GM, et al: Clear cell sustained N-myc reduction in rhabdomyosarcomas and transcrip-
sarcoma of the kidney: a review. Eur J Cancer 28:2219–2226, tional block by antigene therapy. Clin Cancer Res 18:796–807,
2012. 2012.
178. Hayashi Y, Kanda N, Inaba T, et al: Cytogenetic findings and 197. Turc-Carel C, Lizard-Nacol S, Justrabo E, et al: Consistent chro-
prognosis in neuroblastoma with emphasis on marker chromo- mosomal translocation in alveolar rhabdomyosarcoma. Cancer
some 1. Cancer 63:126–132, 1989. Genet Cytogenet 19:361–362, 1986.
179. Kaneko Y, Kanda N, Maseki N, et al: Different karyotypic pat- 198. Vieco PT, Azouz EM, Hoeffel JC: Metastases to bone in medul-
terns in early and advanced stage neuroblastomas. Cancer Res loblastoma: a report of five cases. Skeletal Radiol 18:445–449, 1989.
47:311–318, 1987. 199. Wang-Wuu S, Soukup S, Ballard E, et al: Chromosomal analysis
180. Kohl NE, Kanda N, Schreck RR, et al: Transposition and ampli- of 16 human rhabdomyosarcomas. Cancer Res 48:983–987, 1988.
fication of oncogene-related sequences in human neuroblastomas. 200. Whang-Peng J, Knutsen T, Theil K, et al: Cytogenetic studies in
Cell 35:359–367, 1983. subgroups of rhabdomyosarcoma. Gene Chromosomes Cancer
181. Missiaglia E, Williamson D, Chisholm J, et al: PAX3/FOXO1 5:299–310, 1992.
fusion gene status is the key prognostic molecular marker in rhab- 201. Zhuge Y, Cheung MC, Yang R, et al: Pediatric non-Wilms renal
domyosarcoma and significantly improves current risk stratifica- tumors: subtypes survival, and prognostic indicators. J Surg Res
tion. J Clin Oncol 30:1670–1677, 2012. 163:257–263, 2010.

ERRNVPHGLFRVRUJ
C H A P T E R 2 0 

Synovial Lesions

CHAPTER OUTLINE

SYNOVIAL CHONDROMATOSIS SYNOVIAL LIPOMA


SYNOVIAL CHONDROSARCOMA SYNOVIAL HEMANGIOMA
PIGMENTED VILLONODULAR SYNOVITIS

are bilateral. The peak age incidence and the most fre-
SYNOVIAL CHONDROMATOSIS quent joints involved are shown in Figure 20-1.
Definition
Clinical Symptoms
Synovial chondromatosis is a metaplastic condition
involving articular or tendon sheath synovial membranes The clinical presentation consists of pain, swelling, and
in which multiple nodules of cartilage are produced. limitation of motion. The patient may have a history of
Many of the nodules subsequently become detached from symptoms lasting several months to several decades, but
the synovial membrane and float in the joint. The process usually symptoms have persisted for several years, with
typically involves the synovium diffusely, has a high pro- the average being 5 years.
pensity for recurrences, and may severely compromise
the function of a joint. Radiographic Imaging
The exact pathogenesis of synovial chondromatosis is
unknown, but the involvement of chondroprogenitor If the process is well developed (i.e., sufficient mineraliza-
cells with dysregulation of the hedgehog signaling tion of the cartilaginous matrix has occurred), the changes
pathway and overexpression of bone morphogenetic pro- shown on plain radiographs are characteristic, and the
teins 2 and 4 (BMP2 and 4) has been postulated.6,11,23,24 diagnosis can be established preoperatively.8,22,25,38,39
Overexpression of fibroblast growth factor receptor 3 However, in approximately 10% of synovial chondroma-
(FGFR3), an important regulator of chondroprogenitor toses, no calcifications can be documented on plain films.
cells, and its specific ligand fibroblast growth factor 9 In typical cases involving a major joint, synovial chondro-
(FGF9) was identified in proliferating cells at the periph- matosis presents as multifocal, articular, or periarticular
ery of cartilage nodules.28 nodules with stippled or ringlike calcifications (Figs. 20-2
to 20-5). The level of mineralization gradually increases
Incidence and Location if the lesions are untreated. Older lesions are heavily
calcified and easy to recognize on radiographs. Superim-
Synovial chondromatosis is thought to occur rarely, but position of enchondral ossification is associated with the
data on its true incidence are not available. The peak development of linear calcifications. The nodules may
incidence is in the fifth decade of life, but the age range eventually develop ringlike or eggshell-like calcifications
varies widely from the first to seventh decades. Male with central lucencies (Fig. 20-2). These structures cor-
patients are predominantly affected, and the male-to- respond to well-developed ossified nodules that are com-
female ratio is approximately 2 : 1. posed of a shell of bone with a central area mimicking a
Synovial chondromatosis preferentially involves the medullary cavity. Occasionally the involvement may be
major weight-bearing joints, but any joint may be quite extensive, and the disease may pursue an aggressive
involved.4,5,7,10,13,15,18 Joints such as the knee, hip, and clinical course (Fig. 20-2). In such cases, there is extensive
elbow are typically involved. Approximately 70% of involvement of tendons, multiple joints, or both tendons
cases involve the area around the knee. A few cases and joints in the same anatomic region. Bone erosion can
involve joints of the shoulder and acral skeleton, the be present and is not an indication of aggressive behav-
intervertebral facet joint, and the temporomandibular ior.17,25,26,28 Computed imaging techniques, especially
joint.1,2,14,16,17,29,31,40,41 Approximately 10% of cases, espe- magnetic resonance imaging, are helpful in identifying
cially those involving the knee, hip, elbow, or shoulder, Text continued on p. 1264

1259
ERRNVPHGLFRVRUJ
1260 20  Synovial Lesions

Incidence

1 2 3 4 5 6 7 8
Age in decades
FIGURE 20-1  ■  Synovial chondromatosis. Peak age incidence and most frequent joints involved. Most frequent sites are indicated by
solid black arrows.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1261

B
FIGURE 20-2  ■  Synovial chondromatosis: radiographic features. A and B, Lateral (A) and anteroposterior (B) radiographs of foot and
ankle show long-standing synovial chondromatosis involving tendon sheaths. Tarsal joints and ankle joint appear to be preserved.
Most nodules are of bony consistency. Note extensive involvement of extraarticular synovium of tendons and eggshell pattern of
mineralization with central lucency in some of the nodules.

ERRNVPHGLFRVRUJ
1262 20  Synovial Lesions

C
FIGURE 20-3  ■  Synovial chondromatosis: radiographic features. A, Lateral radiograph of knee shows stippled calcification in posterior
aspect. B, Computed tomogram documents multifocal calcification in periarticular soft tissue. C, Specimen radiograph of resected
synovial pad with multifocal calcified nodules representing areas of cartilaginous metaplasia.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1263

B
FIGURE 20-4  ■  Synovial osteochondromatosis: radiographic features. A, Sagittal magnetic resonance image shows high signal in
loose bodies with areas of signal void representing mineralization of cartilage and bone matrix. B, Lateral radiograph of knee shows
cartilaginous and bony loose bodies in knee joint below patella; joint space is preserved.

ERRNVPHGLFRVRUJ
1264 20  Synovial Lesions

(Fig. 20-10). Each cartilage nodule ranges in size from a


small focus (<1 mm) that requires a magnifying glass to
be seen to a larger lesion measuring more than 1 cm in
diameter. In well-established, diffuse lesions, there are
multiple free cartilaginous nodules, and the joint space
occasionally is packed with loose bodies that range in size
from 1 mm to 1 to 2 cm9,10 (Fig. 20-10). In addition to
individual nodules focally, larger cartilaginous areas rep-
resent coalescent smaller nodules of cartilage. These
larger cartilaginous masses have multinodular (granular)
surfaces, and their multifocal origin can still be seen
on cut surface. Occasionally in long-standing cases, the
adjacent bursae or tendons may be involved. On the
basis of gross features, synovial chondromatosis can be
divided into two forms: diffuse and localized. The diffuse
form is characterized by multiple nodules that involve
almost the entire synovium of the joint. In the localized
form a discrete mass is present and represents a coales-
cence of individual nodules of metaplastic cartilage
(Fig. 20-11). In rare cases a nodular form of synovial
chondromatosis can have a configuration of a polypoid
pedunculated mass protruding into the joint space. The
process typically involves synovial membranes of the joint
capsule. Exclusively extraarticular cases are rare.33,34
Extraarticular involvement is associated more often with
an articular form.

Microscopic Findings
Fully developed synovial chondromatosis consists of mul-
tiple, well-demarcated nodules of cartilage with hyaline
or myxoid features19,20,21,25,35,38-40 (Fig. 20-12). Early lesions
are composed of ill defined hypercellular nodules com-
posed of chondroblastic cells with minimal matrix forma-
FIGURE 20-5  ■  Synovial chondromatosis of ankle joint: radio-
graphic features. Lateral radiograph shows popcornlike calcifi-
tion (Figs. 20-13 and 20-14). In typical cases the cartilage
cation in cartilaginous loose bodies and in synovium above cells form loose clusters, but a uniform distribution of
posterior aspect of calcaneus and overlying the distal end of cartilage cells can also be seen. The cellularity of cartilage
tibia, fibula, and talus. Note enlargement of tarsal sinus and nodules is often increased. The nuclei of cartilage cells
thinning of neck of talus secondary to pressure of intraarticular have open chromatin and small nucleoli and may exhibit
bodies.
significant nuclear atypia (Figs. 20-15 to 20-17). Binucle-
ated or larger cartilage cells are frequently present. The
microscopic features of cartilage atypia may be compat-
the lesion in its early stages when there is insufficient ible with those seen in grade 1 and 2 chondrosarcomas if
mineralization to be documented in conventional radio- taken out of context of a metaplastic synovial condition.
graphs.4,12,36 In later stages, these techniques provide the These features should not be considered as indicative of
means to evaluate the extent of involvement of the joint malignant transformation and cannot be correlated with
and its adjacent structures (Figs. 20-4, 20-6, and 20-7). a more aggressive clinical behavior of the disease.
T1-weighted images typically show a punctate signal void The development of cartilage nodules in this meta-
in the lesion involving the joint capsule. However, plastic process is somewhat similar to normal phases of
T2-weighted images show signal enhancement and may cartilage development. However, the majority of patients
show the lesion to have a multinodular architecture.4,8,29 have a long history of symptoms when they are first seen.
Lesions involving smaller joints become symptomatic in Therefore the early phases of this process are rarely seen.
earlier stages and may show discrete, ill-defined calcifica- The earliest phases of synovial cartilage metaplasia are
tions or no calcification on plain radiographs3 (Figs. 20-8 most frequently seen in smaller joints, such as the tem-
and 20-9). At the other end of the spectrum are long- poromandibular joint or the small joints of the acral skel-
lasting lesions that present as a consolidated, heavily cal- eton, in patients with a short history of symptoms. In the
cified mass.30 earliest stages of synovial chondromatosis, small islands
of cartilage cells with chondroblastic features form. Indi-
Gross Findings vidual chondroblastic cells dispersed within the stromal
tissue of the joint capsule also can be seen (Fig. 20-18).
In advanced cases, the synovium is diffusely involved Loosely arranged clusters of immature cartilage cells may
and may be studded with multiple cartilage nodules Text continued on p. 1278

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1265

FIGURE 20-6  ■  Localized synovial chondromatosis of hip joint: radiographic features. Computed tomogram of hip shows femoral
head within acetabulum. Note enlargement of acetabular fossa and calcified intrasynovial cartilaginous bodies characteristic of
localized synovial chondrometaplasia.

ERRNVPHGLFRVRUJ
1266 20  Synovial Lesions

B
FIGURE 20-7  ■  Synovial chondromatosis: radiographic features. A and B, Computed tomogram of skull shows involvement of tem-
poromandibular joint by synovial chondromatosis. Calcifications in loose bodies can be seen adjacent to mandibular condyle.
C, Magnetic resonance image of temporomandibular joint shown in A and B. Note expansion of joint and areas of signal void within
joint and in thickened synovium. These represent calcification in metaplastic cartilage.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1267

B
FIGURE 20-8  ■  Synovial chondromatosis: radiographic features. A, Plain radiograph of wrist and hand shows soft tissue calcifications
in wrist joint overlying triquetrum, hamate, and distal end of ulna (arrows). B, Lateral view of wrist (same case as A) shows soft
tissue calcified bodies that are both dorsal and volar in location (arrows).

ERRNVPHGLFRVRUJ
1268 20  Synovial Lesions

B
FIGURE 20-9  ■  Synovial chondromatosis of tendon sheath: radiographic features. A, Anteroposterior radiograph of foot shows soft
tissue calcification between first and second metatarsals with ringlike and punctate appearance. B, Oblique radiograph shows soft
tissue calcifications representing loose cartilaginous bodies in tendon sheath. Note pressure erosions along inferolateral cortex of
first metatarsal.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1269

A B

C D
FIGURE 20-10  ■  Synovial chondromatosis (diffuse form in major joints): gross features. A and B, Synovectomy specimen in case of
diffuse synovial chondromatosis of knee joint. Synovial membrane is studded with multiple cartilage nodules that vary in size.
C and D, Loose cartilaginous bodies and synovial membrane from two cases of synovial chondromatosis that diffusely involved
knee joint. Cartilage nodules are present in synovium and free in joint. Bodies vary in size, and some are conglomerated to form
larger masses.

ERRNVPHGLFRVRUJ
1270 20  Synovial Lesions

A B

C D
FIGURE 20-11  ■  Synovial chondromatosis (localized form): gross and radiographic features. A, Localized synovial chondromatosis
presents as conglomerate of individual cartilage nodules. B, Pedunculated polypoid lesion of knee joint is composed of individual
cartilage nodules and interlacing synovial membrane. C, Radiograph of shoulder showing a conglomerate of individual mineralized
nodules with punctate and ringlike calcification patterns. D, Central calcification in metaplastic chondroid nodule of synovium in
knee joint. (D, ×25) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1271

A B

C D
FIGURE 20-12  ■  Synovial chondromatosis: microscopic features. A, Low power photomicrograph of cross section through synovium
of knee joint in synovial chondromatosis. Most nodules are composed of hyaline cartilage. Some are calcified, and early endochon-
dral ossification is present in others. B, Medium power photomicrograph of typical chondroid metaplastic nodules in synovium
of knee joint. C, Low power photomicrograph of synovium with conversion of cartilage nodules to bone. This is late stage
of the phenomenon. D, Almost complete ossification of hyaline cartilage nodule in synovium to bone. (B, ×25; D, ×50)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1272 20  Synovial Lesions

B
FIGURE 20-13  ■  Synovial chondromatosis: microscopic features. A, Ill defined nodules composed of chondroblastic cells in the syno-
vial membrane representing early phases of synovial chondromatosis. B, Higher magnification of A showing a ill defined subsynovial
nodule composed of chondroblastic cells with early evidence of matrix deposition in the center. Inset, Higher magnification showing
nuclear morphology of chondroblastic cell. (A, ×15; B, ×25; Inset, ×200) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1273

B C
FIGURE 20-14  ■  Synovial chondromatosis: microscopic features. A, Low power view showing hypercellular ill defined nodules com-
posed of chondroblastic cells. Some of the nodules show cartilage matrix deposition. B, Higher magnification of A showing an ill
defined nodule composed of chondroblastic cells. C, Higher magnification of A showing a cartilage nodule. (A, ×15; B and C, ×25)
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1274 20  Synovial Lesions

B C
FIGURE 20-15  ■  Synovial chondromatosis: microscopic features. A, Medium power photomicrograph of chondroid metaplastic
nodules in synovium with clear cell features. B, Higher magnification of A showing cartilage nodules in the synovial membrane
with clear cell features and nuclear hyperchromasia. C, Higher magnification of metaplastic synovial cartilage nodules with clear
cell features and nuclear hyperchromasia. (A, ×15; B and C, ×50) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1275

B
FIGURE 20-16  ■  Synovial chondromatosis: microscopic features. A, Low power photomicrograph of synovium containing nodules
of metaplastic cartilage with hyaline and myxoid features. B, Medium power photomicrograph shows hyaline cartilage arising
through metaplasia of synovial membrane. Inset on left shows early stage of development with myxoid differentiation of connective
tissue cells and intercellular matrix. Inset on right shows chondrocytes with open chromatin in hyaline matrix. (A, ×100; B and
left Inset, ×200; right Inset, ×400) (A, B, and Insets, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1276 20  Synovial Lesions

A B

C D

FIGURE 20-17  ■  Synovial chondromatosis: microscopic features. A and B, Low power photomicrographs showing extensive cartilage
metaplasia of the synovial membrane with confluent cartilage nodules. C and D, Intermediate and high power photomicrographs
showing myxoid and hyaline chondroid matrix formation. Note pronounced cellular variability and nuclear hyperchromasia (A and
B, ×25; C, ×100; D, ×200) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1277

A B

C D

FIGURE 20-18  ■  Synovial chondromatosis: microscopic features. A and B, Low and medium power photomicrographs of chondroid
metaplastic nodules in the synovium. C and D, Medium and high power photomicrographs showing loosely arranged cells with
chondroblastic features within myxoid areas adjacent to discrete cartilage nodules. Note variation in nuclear size and hyperchro-
matism, which should not be interpreted as indicative of malignancy (A, ×25; B, ×50; C, ×100; D, ×200) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1278 20  Synovial Lesions

exhibit myxoid or clear-cell features. Occasionally, larger, Treatment and Behavior


loose areas of cells with microscopically recognizable
chondroblastic features can be seen within the soft tissue Synovial chondromatosis is treated by removing the loose
in the vicinity of more mature cartilage islands (Fig. bodies and the synovial membrane. In some cases, it
20-19). These foci are present beneath the synovial lining follows a self-limited course. After many years, these
and do not directly involve the synovium. Smaller, more lesions reach a quiescent stage in which only loose
densely packed foci of chondroblastic cells with dense intraarticular bodies are present and there is no active
eosinophilic cytoplasm and large hyperchromatic nuclei process in the joint capsule. Extraarticular extension with
may also be present within well-established cartilage tendon involvement does not indicate aggressive behav-
nodules (Fig. 20-20). It is our impression that immaturity ior. Synovial chondromatosis is a local, nonneoplastic,
of cartilage cells is responsible for prominent atypia in a self-limiting process. Removal of the loose bodies and of
majority of cases. Rarely is the formation of cartilage the involved synovium is sufficient to control the disease
nodules associated with prominent giant cell osteoclastic in the majority of cases. Multiple and often short-interval
reaction (Fig. 20-21). recurrences raise suspicion of synovial chondrosarcoma.
Enchondral ossification of cartilage nodules is a fre-
quent feature of well-developed synovial chondromato-
sis. There are well-developed peripheral rings or eggshells SYNOVIAL CHONDROSARCOMA
of lamellar bone (Fig. 20-12). Occasionally the nodules
consist of well-formed rings of lamellar bone with a Chondrosarcoma of the synovium is extremely rare.42-53
central, fatty bone marrow–like space. Lesions with well- The largest series of 10 cases was published by research-
developed enchondral ossification are also referred to as ers at the Mayo Clinic.43 Chondrosarcoma can develop
osteochondromatosis. within the joint capsule as a de novo (primary) lesion or
Lesions that present as a single cartilaginous nodule may be secondary to preexisting synovial chondromato-
within the joint capsule are sometimes referred to as sis. Primary synovial chondrosarcoma is the rarest form.
synovial chondromas.8,10,27,30,32,34,37 Single, isolated cartilage In a series of 10 reported cases, only 2 were primary
nodules are typically seen in smaller joints of the acral lesions, and the remaining 8 developed in a setting of
skeleton (Fig. 20-22) or in the temporomandibular joint. synovial chondromatosis. The majority of reported cases
These lesions typically show prominent mineralization of involve the major joints of the lower extremities. Knee
cartilage matrix. It is still controversial whether a single involvement is the most frequent, followed by involve-
discrete nodule within synovium or periarticular soft ment of the ankle and hip. Synovial chondrosarcoma may
tissue is a clonal neoplastic proliferation of cartilage cells develop in other major joints of the upper extremities and
(i.e., chondroma) or represents a localized unifocal variant in the smaller joints of the acral skeleton. Our experience
of a metaplastic process (i.e., synovial chondromatosis). is restricted to three cases that involved the knee, elbow,
and foot. In all three cases, the tumor developed in a
setting of synovial chondromatosis.
Differential Diagnosis
Radiographically, chondrosarcomas of the synovium
The most important consideration in differential diag- should be suspected if a consolidated calcified mass is
nosis is chondrosarcoma. The cytologic atypia of carti- present in the joint area. In most cases, the presence
lage cells is often worrisome and raises the suspicion of of a calcified consolidated mass is associated with bone
chondrosarcoma. The approach used to evaluate synovial erosion. Grossly, the lesion represents a consolidated
chondromatosis is the same as that used to evaluate car- cartilage mass and typically involves the adjacent bone
tilage lesions of bone. Thus as with cartilage lesions, all (Fig. 20-23).
clinical and radiographic findings should be considered. Histologically, the lesions are typically grade 1 and 2
First, rare but well-known secondary involvement of chondrosarcomas (Figs. 20-24 and 20-25). Sometimes,
the joint by a chondrosarcoma of the adjacent bone, such the tumors are of myxoid type. Features such as loss of
as the femur or tibia, must be clinically and radiographi- clustered growth pattern of cartilage cells, prominent and
cally ruled out. This dilemma arises most frequently in uniform myxoid appearance, necrosis, and presence of
evaluating hip lesions, in which the joint capsule may be spindling at the periphery of nodules should raise the
secondarily involved by a chondrosarcoma of the adjacent suspicion of malignancy.42,52,53 The diagnosis of chondro-
pelvic and femoral bones. When the clinical, radio- sarcoma of the synovium should be based on the analysis
graphic, and gross findings are consistent with the of clinical, radiographic, and microscopic evidence.
process confined to the joint capsule and space, the atypia The microscopic features described are valid if they
of cartilage cells can be discounted as a feature of malig- coincide with the radiographic and gross documentation
nant transformation, and the lesion can be classified as of a consolidated mass that is not a coalescence of indi-
synovial chondromatosis. However, these cases must be vidual metaplastic nodules. It is common for the final
differentiated from extremely rare primary synovial diagnosis of synovial chondrosarcoma to be made after
chondrosarcoma or chondrosarcoma developing in syno- several local recurrences of lesions initially considered
vial chondromatosis (see later section). In rare instances to be synovial chondromatosis. Pulmonary metastases
synovial chondromatosis may develop in a bursa overly- develop in approximately 50% of patients with reported
ing an osteochondroma. Such cases may, radiographi- cases of synovial chondrosarcomas, usually within 3 years
cally, mimic a secondary chondrosarcoma arising in the after initial diagnosis. Synovial chondrosarcomas are
cartilaginous cap of the osteochondroma.9 Text continued on p. 1286

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1279

A B

C D
FIGURE 20-19  ■  Synovial chondromatosis: microscopic features. A and B, Low and medium power photomicrographs showing con-
fluent cartilage nodules in the synovial membrane with hypercellular areas and hyperchromatism. C and D, Medium and high power
photomicrographs showing ill defined hypercellular cartilage nodules with clear cell features and nuclear hyperchromasia (A, ×25;
B, ×50; C and D, ×100) (A-D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1280 20  Synovial Lesions

B
FIGURE 20-20  ■  Synovial chondromatosis: microscopic features. A, Metaplastic chondroid nodule showing hypercellular metaplastic
cartilage nodule with nuclear hyperchromasia. B, Loosely arranged cells with chondroblastic features within myxoid area. Inset
shows variation in nuclear size and hyperchromasia, which should not be interpreted as indicative of malignancy. (A and B, ×200;
Inset, ×400) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1281

B C
FIGURE 20-21  ■  Synovial chondromatosis: microscopic features. A, Low power photomicrograph showing an unusual association
of synovial chondroid cartilage metaplasia with prominent giant cell reaction. B and C, Interface between cartilage nodule
and hypercellular areas with prominent giant cell reaction with multinucleated osteoclastic cells (A, ×25; B and C, ×100)
(A-C, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1282 20  Synovial Lesions

A B

C D

FIGURE 20-22  ■  Tenosynovial chondrometaplasia (localized nodular form): radiographic and microscopic features. A and B, Lateral
radiographs of synovial chondrometaplasia in dorsal and volar tendon sheath of fingers. Note pressure erosion of phalanx in A and
focal soft tissue calcification in B. C and D, Low power photomicrographs of tenosynovial chondrometaplasia (synovial chondroma-
tosis) in digits. There is nodular hyaline cartilage differentiation and patchy calcification. (C, × whole mount; D, ×25.) (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1283

FIGURE 20-23  ■  Synovial chondrosarcoma developing secondary to synovial chondromatosis: gross features. A, Gross photograph
of synovial chondrosarcoma developing in left elbow of a man with a 10-year history of synovial chondromatosis. Grade 2 chon-
drosarcoma arose in synovium of elbow and spread beyond joint into soft tissue and bone. Multiple recurrences were followed by
amputation. B, Gross photograph of sagittally cut ankle and foot of patient with 15-year history of recurrent tenosynovial chondro-
matosis who had a grade 1 chondrosarcoma that invaded tarsal and metatarsal bones.

ERRNVPHGLFRVRUJ
1284 20  Synovial Lesions

B
FIGURE 20-24  ■  Synovial chondrosarcoma arising in synovial chondromatosis: microscopic features. A and B, Medium power
photomicrographs show ill defined nodularity of malignant cartilage cells with nuclear irregularity and hyperchromatism.
Inset shows hyperchromatic malignant cartilage cells in hyaline cartilage matrix (A and B, ×100; Inset, ×200) (A, B, and
Inset, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1285

B
FIGURE 20-25  ■  Synovial chondrosarcoma: microscopic features. A, Grade 2 chondrosarcoma that developed in synovium of elbow
(same case as Fig. 20-20, A). B, Higher magnification of A shows so-called open chromatin of cartilage cells and myxoid stroma.
Inset shows pronounced nuclear atypia of cartilage cells. (A, ×100; B, ×200; Inset, ×400) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1286 20  Synovial Lesions

appears that the polyclonal nonneoplastic histiocytic cells


TABLE 20-1 Forms of Pigmented
are attracted to the lesion by overexpression of CSF1.
Villonodular Synovitis
Cytogenetic studies also indicate that the most common
Intraarticular fusion partner for CSF1 should be located on 2q35-37,
Diffuse Diffuse involvement of joint synovium resulting in 1p11-2q37 balanced translocation.123 In fact,
Localized Localized (polypoid or nodular) joint synovial this translocation produces a CSF1-COL6A3 chimeric
lesion gene65,123 reminiscent of the translocation that defines
Extraarticular dermatofibrosarcoma protuberans, in which t(17;22)
Diffuse Diffuse multifocal involvement of brings PDGF-β under control of the strong COL1A1
extraarticular synovial membrane (tendons) promoter.88 This data suggests that only a minority of
Nodular Localized extraarticular nodular lesion, also cells in pigmented villonodular synovitis are clonal and
referred to as giant cell tumor of the neoplastic and that the majority of histiocytic cells in the
tendon sheath
lesion are nonneoplastic. These observations are consis-
tent with the biology of CSF1, which mediates the pro-
liferation, differentiation, and function of macrophages
treated by aggressive surgery (i.e., amputation or extraar- and their precursors. The clonal neoplastic cells, with the
ticular resection with amputation). CSF1 translocation and its overexpression, most likely
recruit CSF1R-expressing macrophages and induce the
formation of multinucleated giant cells (Fig. 20-26).
PIGMENTED VILLONODULAR SYNOVITIS These findings explain the lack of evidence for clonality
in previous chromosome X inactivation studies.104
The unified clinicopathologic concept of pigmented vil- DNA microarray studies show that the genes differ-
lonodular synovitis or tenosynovitis proposed by Jaffe et al. entially expressed in pigmented villonodular synovitis
in 194180 encompasses a diverse group of fibrohistiocytic are involved in cell proliferation, apoptosis, matrix
proliferations that can occur in several, sometimes over- degradation, and inflammation, in part explaining the
lapping, clinical settings (Table 20-1). The process may aggressive, occasionally bone destructive nature of the
involve joints, bursae, tendon sheaths, the adjacent fascial disease.70,75,116,119,122 Occasional aneuploidy seen primarily
and ligamentous tissue, or a combination of these areas. in the diffuse forms of the disease confirms the neo-
Pigmented villonodular synovitis can be divided into plastic nature of the disorder.55 The increased prolifera-
several forms on the basis of site and extent of involve- tive rate of monocyte-macrophage lineage (histiocytic)
ment: intraarticular versus extraarticular and diffuse cells with overexpression of fibroblast growth factor and
versus localized or nodular. It is common for a nodular the tyrosine kinase receptors Tie-1/Tie-2 have been
or more diffuse lesion to occur in the same case. Simi- documented in some cases of pigmented villonodular
larly, intraarticular and extraarticular involvement can synovitis.32,92,108,110,117
coexist. At the time of initial presentation, most lesions
can be classified into the basic forms listed in Table 20-1.
Definition
In the past it was generally accepted that the process
represents a reactive inflammatory condition. However, Pigmented villonodular synovitis is a destructive, fibro-
in some cases, the clinical aggressiveness—bone destruc- histiocytic proliferation with production of villous and
tion, recurrences, or both—raised the suspicion of low- nodular protrusions of articular and extraarticular syno-
grade, locally aggressive neoplasia. In fact, in some vial membranes. Microscopically the lesion is composed
lesions, especially in the nodular extra-articular form of fibroblasts, histiocytic cells, and inflammatory cell
(giant cell tumor of tendon sheaths), clonal chromosomal infiltrates and contains prominent hemosiderin deposi-
aberrations and aneuploidy have been noted.54,67,72,91,102 tion, which is responsible for its brown color.
Trisomies 5 and 7 appear to be dominant, nonrandom
alterations in this disorder.66,72 Translocations involving
Incidence and Location
chromosomes 1 and 2 and chromosomes 1 and 14 have
also been reported.77 In general, the cells involved in The true incidence of this condition is unknown, but it
pigmented villonodular synovitis express a wide range of is relatively rare and accounts for less than 5% of all
histiocyte/macrophage and osteoclast markers.55 primary soft tissue tumors. The peak age incidence and
Several reports indicate that clonal cytogenetic abnor- most frequent sites of involvement are shown in Figure
malities involving 1p11-13 are most frequent in pig- 20-27. The age of patients varies widely from the first to
mented villonodular synovitis and may contribute to the seventh decades of life. The peak age incidence is in the
growth advantage in providing gene activation through a third and fourth decades of life. There are significant
balanced translocation.67,91,95,98,103,106 It was recently shown differences in the skeletal locations and age distribution
that mononuclear and osteoclast-like giant cells in pig- patterns among the various forms of pigmented villon-
mented villonodular synovitis express high levels of colony odular synovitis.78,99,100,101,117 The intraarticular forms,
stimulating factor 1 receptor (CSF1R). In addition, the both diffuse and localized, predominantly involve the
colony stimulating factor 1 (CSF1) gene encoding ligand major weight-bearing joints of the lower extremities.
for CSF1R is translocated in the majority of intraarticular In the intraarticular form, the knee is involved in approxi-
and extraarticular pigmented villonodular synovitis71,123 mately 80% of cases, followed by the hip (15%). Rare
(Fig. 20-26). Surprisingly, only a minority of tumor cells examples of intraarticular forms can be found in
(<20%) carry the translocation and express CSF1.65,123 It the ankle, foot, elbow, shoulder, sacral, vertebral facet,

ERRNVPHGLFRVRUJ
Chr 1 CSF1

Exon 1

Exon 2

Exon 3

Exon 4

Exon 5

Exon 6

Exon 7
Exon 8

Exon 9
36.3
Chr 2 36.2

cds
36.1
25.3
25.1 25.2 35
24 34.3 34.2
34.1
23 33
32.3
22 32.2
21
32.1
31.3 CSF1
31.2
1 554 aa
16 31.1
15 22.3
14 22.2 Four-helical cytokine-like, core domain
13 22.1
12 21 B
11.2 13.3
11.1 11.1 CSF1
11.2 13.1 13.2
11
12
13
12
11
COL6A3
14.1 12
14.2

Exon 10
Exon 11
Exon 12
Exon 13
Exon 14
Exon 15
Exon 16

Exon 17
Exon 18
Exon 19
Exon 20
Exon 21
Exon 22
Exon 23
Exon 24
Exon 25
Exon 26
Exon 27
Exon 28
Exon 29
Exon 30
Exon 31
Exon 32
Exon 33
Exon 34
Exon 35
Exon 36
Exon 37
Exon 38
Exon 39
Exon 40
Exon 41
Exon 42
Exon 43
Exon 44
14.3

Exon 1
Exon 2
Exon 3

Exon 4

Exon 5
Exon 6
Exon 7
Exon 8
Exon 9
21.1 21.2 21.1
21.2 21.3 21.3

cds
22 22
23
23 24
24.1 24.2 25
24.3
31 31
32.1
32.2
COL6A3
32.3 32.1
33 32.2 1 3177 aa
32.3
34 41
35 42.1 von Willebrand factor, type A domain Immunoglobulin-like fold domain
36 42.2 42.3
43 Collagen triple helix repeat domains Proteinase inhibitor I2, Kunitz metazoa domain
37.1 44
37.2 COL6A6
37.3
C
A

D E F

G H I J

CSF1R
R

CSF1R
F1
1R
CS 1R
CSF1R

COL6A3:CSF1
C CSF1

CSF1R
1R

FIGURE 20-26  ■  Pigmented villonodular synovitis: activation of CSF1 by a translocation and its landscape effect. A, Positions of CSF1
and COL6A3 on chromosomes 1 and 2 involved in 1P13-2q37 balanced translocation are shown. The nature of the break-point and
the specific structure of the fusion gene are not known at this time. B, Exon-intron structure of the CSF1 gene and of its encoded
protein are shown. C, Exon-intron structure of the COL6A3 gene and of its encoded protein are shown. D, Fluorescence in situ
hybridization (FISH) with the CSF1-spanning BAC probe RP11–19F3B in extraarticular villonodular synovitis confirms that the CSF1
gene is split by the trans-location in TGCT. E, The fusion of CSF1 and COL6A3 confirmed by FISH. The COL6A3 locus is represented
by CTD–2344F21 (labeled in white), the region telomeric to CSF1 is represented by RP11–96F24 (labeled in green), and the region
centromeric to CSF1 is represented by RP11–354C7 (labeled in orange). F, FISH with BAC probe RP11–25803 that covers the region
telomeric to COL6A3 fails to split. G and H, Two nuclei with the fusion of CSF1 and COL6A3 demonstrated by FISH in extraarticular
villonodular synovitis. The COL6A3 locus is represented by CTD–2344F21 (labeled in white), the region telomeric to CSF1 is repre-
sented by RP11–96F24 (labeled in green), and the region centromeric to CSF1 is represented by RP11–354C7 (labeled in orange).
I, Lack of translocation in a nucleus from the same sample as D and E shows an intact CSF1 gene. J, Combined interphase FISH
with the region telomeric to CSF1 represented by RP11–96F24 (labeled in green) and the region centromeric to CSF1 represented
by RP11–354C7 (labeled in orange), and CSF1 immunohistochemistry (labeled in red) demonstrated that only the cells with the
translocation expressed CSF1. K, Autocrine and paracrine scheme for CSF1 landscaping effect. CSF1 is produced by neoplastic cells,
with translocation resulting in increased numbers of neoplastic cells through an autocrine loop with CSF1R. CSF1 also recruits non-
neoplastic CD163- and CSF1R-expressing cells of monocyte/macrophage lineage. (Modified and reprinted with permission from West
RB, et al: A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor
cells. PNAS 103(3):690–695, 2006.)
ERRNVPHGLFRVRUJ
1288 20  Synovial Lesions

Extraarticular
Incidence

Intraarticular
1 2 3 4 5 6 7 8
Age in decades

FIGURE 20-27  ■  Pigmented villonodular synovitis. Peak age incidence and most frequent sites of involvement. Most frequent sites
are indicated by solid black arrows.

and temporomandibular joints.62,73,74,84,87,120 The nodular mass57,59,61,63,64,68,69,76,79,83,86,107,111 (Figs. 20-28 to 20-30). It


extraarticular variant of pigmented villonodular tenosy- is frequently associated with degenerative joint disease
novitis typically involves the sacral soft tissue and is pre- and multiple subchondral cysts in the bones around the
dominantly found in the fingers and metacarpal and joint.57,107 The cysts are usually present on both sides
carpal areas.100 Tendons of the forearm are rarely involved. of the affected joint. Computed tomography and mag-
When all forms of the disease are analyzed, the fingers netic resonance imaging reveal the extent of involve-
are involved in nearly 60% of cases, whereas the knee is ment and are particularly useful in documenting the
involved in approximately 30% of cases.117 The toes are presence of localized pedunculated lesions within major
involved in about 10% of cases. Involvement of several joints.58,82,85,90,112,113 T1-weighted magnetic resonance
joints or severe polyarticular forms are seen more often images show low-signal inhomogeneous masses that
in younger patients.56,64,118,121 In rare instances pigmented may reveal a punctate signal void related to hemosiderin
villonodular tenosynovitis can coexist with other synovial deposits (Figs. 20-31 and 20-32). Erosion of bone com-
conditions such as synovial chondromatosis.81 pressing the affected joint is frequently present85,90,112,113
(Figs. 20-28, 20-30, and 20-31).
Clinical Symptoms
Gross Findings
Symptoms include pain, swelling of the joint, and limita-
tion of motion. The presence of hemorrhagic joint The various forms of pigmented villonodular synovitis
effusion is frequent. The aspirated fluid is typically hem- are defined by a combination of clinical, gross, and
orrhagic. The patient usually has a relatively long history microscopic findings. Therefore gross examination of the
of symptoms lasting from 2 to 3 years. The symptoms removed tissue is an important part of determining the
can be intermittent or steadily progressive. correct diagnosis. In diffuse intraarticular forms, most or
all of the synovium is involved. It is covered with tan to
brown, irregular papillary projections and larger nodular
Radiographic Imaging
protrusions (Fig. 20-33). The irregularity or heterogene-
On plain radiographs, pigmented villonodular tenosy- ity of synovial projections is very prominent. They range
novitis presents as an ill-defined, periarticular soft tissue from slender villous structures to thick papillae to large

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1289

B
FIGURE 20-28  ■  Pigmented villonodular synovitis: radiologic features. A, T1-weighted sagittal magnetic resonance image of knee
joint with diffuse pigmented vil-lonodular synovitis. Note nodular thickening of synovium with areas of signal void at sites of hemo-
siderin deposition and erosion in anterior part of tibial plateau. B, Lateral radiograph of knee. Note preservation of joint space and
anterior erosion of tibial plateau.

ERRNVPHGLFRVRUJ
1290 20  Synovial Lesions

A
FIGURE 20-29  ■  Localized nodular tenosynovitis: radiographic features. A, Radiograph of finger shows soft tissue mass devoid of
calcification alongside proximal phalanx. B, Corresponding T1-weighted magnetic resonance image of phalanx adjacent to soft
tissue mass shows nodular tenosynovitis (giant cell tumor of tendon sheath).

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1291

attachment to the synovial membrane or can present as


a pedunculated polypoid lesion. The nodule is frequently
necrotic because of mechanical injury or torsion of its
pedunculated stalk. Extraarticular pigmented villonodu-
lar synovitis can present as a diffuse or nodular disease
similar to the intraarticular form (Fig. 20-35). A nodular
growth pattern is typical for extra articular forms.

Microscopic Findings
The lesion is composed of several cell types that are
commonly found in a group of lesions identified as
fibrohistiocytic proliferations or reactions. The lesion is
composed of a variable number of mononuclear histio-
cytic cells and irregularly interspersed multinucleated
giant cells forming ill defined nodules or villous struc-
tures in the synovial membrane (Figs. 20-36 through
20-41).69,80,89,94,96,101,105 Hemosiderin deposits are promi-
nent. In addition, a variable number of foamy histiocytic
cells are individually interspersed or form clusters (Fig.
20-39). The villous structures are irregular and are
covered by a thickened synovial layer that merges with
an underlying cellular infiltrate (Fig. 20-37 to Fig. 20-40).
The overlying synovium can be denuded from the large
areas, and the stromal cellular infiltrate can be in direct
contact with synovial fluid. Cellularity varies in different
areas of the lesion. Ill-defined nodular aggregates of his-
tiocytic cells with enlarged nuclei and prominent, well-
defined eosinophilic cytoplasm are occasionally present
(Fig. 20-36). Mitotic figures are frequently seen, but the
atypical mitotic figures are absent. The mononuclear his-
tiocytic cells, as well as the multinucleated giant cells, are
positive for kinesin-like protein 1 (KP1 [CD68]), HAM56,
α1-antichymotrypsin, and MAC387 markers.55,89,93,96,105,108
The immunophenotype is consistent with the monocyte-
macrophage lineage origin. These findings are in keeping
with earlier electron microscopic observations that dis-
closed ultrastructural features consistent with the histio-
cytic origin of mononuclear and multinucleated giant
cells in this disorder.93,105 Results of staining for epithelial
membrane antigen and keratin are negative on cellular
infiltrates. More recent findings indicate that the vast
majority of cells in pigmented villonodular synovitis
express CSF1R.71,123 In contrast, only a small proportion
of cells (>20%) express CSF1. In some of the cases, these
FIGURE 20-30  ■  Localized nodular tenosynovitis: radiographic CSF1 overexpressing cells show the clonal 1p11-2q37
features. Lateral radiograph of thumb shows pressure erosion translocation that produces CSF1-COL6A3 chimeric
and reactive sclerosis of proximal phalanx produced by long- gene.123 Preliminary data indicate that overexpression of
standing, overlying tendon sheath nodule.
CSF1 is common in a wide range of reactive synovial
conditions, including reheumatoid arthritis, but the
irregular nodules. The involved joint capsule is thick- translocations of CSF1 appeared to be specific for pig-
ened, and the lesion is poorly demarcated from the adja- mented villonodular synovitis.65
cent periarticular soft tissue. Cross sections reveal soft
tissue that varies in color. The areas with hemosiderin Differential Diagnosis
deposition are tan to brown. The lighter or yellowish
regions correspond to lipid deposition in foamy histio- Pigmented villonodular synovitis must be differentiated
cytes. The lesion has ill-defined borders that impercep- from other reactive inflammatory conditions involving
tibly merge with the periarticular soft tissue and involve synovial membranes that may be associated with villous
the skeletal muscles. The articular cartilage and the adja- or nodular changes of the synovium, such as rheumatoid
cent bone may be eroded. The localized intraarticular arthritis, osteoarthritis, posttraumatic reactive synovitis, and
form presents as a discrete nodule protruding into the hemarthrosis associated with hemophilia.
joint space (Fig. 20-34). It can have a broad base of Text continued on p. 1303

ERRNVPHGLFRVRUJ
1292 20  Synovial Lesions

B
FIGURE 20-31  ■  Pigmented villonodular synovitis: radiographic features. A, T2-weighted magnetic resonance image shows intraspinal
mass arising from synovium of cervical facet joint, which is affected by pigmented villonodular synovitis. Note low-signal areas
corresponding to hemosiderin deposits. B, Computed tomogram of cervical spine of patient shown in A. Lamina is eroded and
expanded by synovium of facet joint, which is involved by pigmented villonodular synovitis.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1293

B
FIGURE 20-32  ■  Diffuse pigmented villonodular synovitis of tendon sheaths of wrist and hand: radiographic and gross features.
A and B, Coronal and axial magnetic resonance images of hand show nodular masses in thenar eminence and wrist region. Irregular
areas of low signal represent hemosiderin deposition. C, Gross photograph of resected synovium of tendon sheath studded with
multiple nodules that range from rubbery gray-white and fibrous to soft yellow streaked with brown on cut section.

ERRNVPHGLFRVRUJ
1294 20  Synovial Lesions

A B

C D
FIGURE 20-33  ■  Pigmented villonodular synovitis of diffuse involvement with bone erosion: gross features. A, Lateral radiograph of
knee shows swelling of soft tissue and erosion of femur, tibia, and patella. B and C, Gross specimens of synovectomies of knee
performed for diffuse pigmented villonodular synovitis. Note chocolate-brown pigmentation produced by hemosiderin deposition.
Villous and nodular proliferation of synovium is evident. D, Low power photomicrograph shows villous architecture of the synovial
membrane with histiocytic infiltration. (D, ×15) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1295

A B

C D
FIGURE 20-34  ■  Pigmented villonodular synovitis of localized form in knee joint: gross features. A, Pedunculated mass attached to
inner surface of synovial fat pad of knee; meniscus is attached. Polypoid mass has been sectioned longitudinally. B, Pedunculated
nodular mass on inner surface of synovial fat pad of knee. C and D, Medium power photomicrographs show histiocytic infiltrate
with hemosiderin deposition and scattered osteoclast-like giant cells characteristic of pigmented villonodular synovitis. (C and
D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1296 20  Synovial Lesions

A B

C D
FIGURE 20-35  ■  Localized nodular tenosynovitis: radiographic and gross features. A, Plain radiograph of finger shows noncalcified
mass in soft tissue adjacent to proximal phalanx with pressure erosion of cortex. B, Bivalved tendon sheath nodule shows firm,
lipid-rich (yellow) mass with brown streaks. C, Whole-mount histologic section shows nodular infiltrates of histiocytes. Surface layer
of synovium is seen in D. D, Photomicrograph at low magnification of nodule shown in C. Note histiocytic infiltrate with hemosiderin
and multinucleated giant cells. (C and D, ×50) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1297

C D

FIGURE 20-36  ■  Pigmented villonodular synovitis: microscopic features. A and B, Surface of synovial membrane with irregular villous
structures. C and D, Higher magnification of B shows villous structures infiltrated by histiocytic cells.(A and B, ×4; C, ×10; D, ×15.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1298 20  Synovial Lesions

B C
FIGURE 20-37  ■  Pigmented villonodular synovitis: microscopic features. A, Villous structures of the synovial membrane with extensive
histiocytic infiltrate. B and C, Medium-power photomicrographs showing extensive stromal histiocytic infiltrate and scattered
osteoclast-like giant cells. (A, ×50; B and C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1299

B C
FIGURE 20-38  ■  Pigmented villonodular synovitis: microscopic features. A, Irregular villous structures of the synovial membrane
with stromal histiocytic infiltrate. B, Higher magnification of A showing irregular villous structures with stromal histiocytic
infiltrate. C, Medium power photomicrograph showing mixed lymphocytic and histiocytic infiltrate. (A, ×25; B, ×50; C, ×100)
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1300 20  Synovial Lesions

B C

FIGURE 20-39  ■  Pigmented villonodular synovitis: microscopic features. A, Synovial membrane with irregular villous structures.
B, Histiocytic infiltrate with focal aggregate of xanthomatous cells and scattered osteoclast-like giant cells. C, Histiocytic infiltrate
with occasional osteoclast-like giant cells. (A, ×25; B, ×50; C, ×100) (A-C, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1301

B C
FIGURE 20-40  ■  Pigmented villonodular synovitis: microscopic features. A, Surface of synovial membrane with irregular villous stru-
cures. B, Higher magnification of A showing extensive histiocytic infiltrate within the stroma of villous protrusions. C, Sheets of
histiocytes and osteoclast-like giant cells. (A, ×25; B, ×50; C, ×100) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1302 20  Synovial Lesions

A B

C D

E F
FIGURE 20-41  ■  Pigmented villonodular synovitis: microscopic features. A-F, Extensive stromal histiocytic infiltrate with hemosiderin
deposition and scattered osteoclast-like giant cells. (A-F, ×100) (A-F, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1303

Rheumatoid arthritis associated with villous synovial fusely involves the synovial membrane and forms multi-
changes usually shows prominent plasma cell infiltrates ple papillary excrescences composed of mature adipose
and formation of lymphoid follicles, which are not fea- tissue lined by synovial cells (Fig. 20-46). The adipose
tures of pigmented villonodular synovitis. tissue is highly vascular, and often there is a noticeable
Posttraumatic synovitis can have villous change, but a inflammatory cell infiltrate composed of lymphocytes
peculiar cellular infiltrate seen in pigmented villonodular and plasma cells. In fact, some of the villous structures
synovitis is not present. may represent reactive change with inflammatory infil-
Hemarthrosis in hemophilia usually does not show trates. Fibrosis within papillary projections and promi-
hemosiderin deposits in the joint capsule. Detritic syno- nant vascular channels are frequent findings in synovial
vitis associated with the dispersion of foreign material lipomas (Figs. 20-47 and 20-48). In extremely rare cases,
related to prosthetic replacement joints is easily identified these may involve several joints.125,126,133,139,140,141 Most
under polarized light. lesions occur in the knee, but other large joints of the
The extraarticular nodular form, especially in highly extremities may also be involved.
cellular lesions with “epithelioid” histiocytes, must be Lipomas involving tendons occur less frequently than
differentiated from epithelioid sarcoma (Fig. 20-42). As those involving the articular capsule.142,145 The tendon
mentioned, pigmented villonodular synovitis is negative sheaths of the hands and wrists are common sites of involve-
for epithelial markers. Geographic necrosis mimicking a ment. Multifocal tumors or bilateral involvement of both
rheumatoid nodule is not a feature in this process, and hands may occur. Similar to lipomas of the joint capsule,
atypical mitoses are absent. In rare instances, pigmented lipomas of the tendons can present as discrete circum-
villonodular synovitis may show cords of epithelioid his- scribed masses or may represent a diffuse, ill-defined over-
tiocytic cells in hyalinized stroma. Such areas can be growth of adipose tissue. Lipomas of the tendons and joint
misinterpreted as low-grade epithelioid sclerosing fibro- capsule may erode the adjacent bone, but complete excision
sarcoma (Fig. 20-43). is curative with virtually no recurrences.
Plain radiographs are nonspecific and may disclose a
soft tissue or joint capsule swelling. Magnetic resonance
Treatment and Behavior
imaging is diagnostic and shows a typical pattern of
Locally aggressive behavior with multiple recurrences is villous lipomatous proliferations of the synovium. T1–
typical for this disorder.59,78,97 Erosion of adjacent bone weighted images show villous synovial proliferations of
and severe compromise of joint function can occur. The similar density as subcutaneous fat. T2–weighted images
treatment of choice is excision of the affected synovium. with fat-suppressed spin show the low signal intensity of
The recurrence rate after surgical treatment ranges within the lesion, similar to subcutaneous fat.126,130,133,143
30%. Some cases with severe involvement of synovium
and adjacent bone require prostatic replacement.114,115 In
clinically aggressive cases, low-dose radiotherapy is some- SYNOVIAL HEMANGIOMA
times used to control the process and has been shown to
be beneficial in preventing further destruction of the joint Similar to synovial lipomas, hemangiomas involving the
and in reducing the risk of recurrences.97 synovium of a joint capsule, periarticular soft tissue, or
In extremely rare cases, transformation to a spindle- tendon sheath belong to the category of soft tissue
cell malignancy with metastases to the contralateral thigh tumors.146-166 The peak age incidence and the most fre-
has been documented 40 years or more after initial pre- quently involved sites are shown in Figure 20-49. Extraar-
sentation; it was preceded by multiple local recurrences.60 ticular synovial hemangiomas involving the tendon
Unusually aggressive behavior with involvement of the sheath are rare and almost exclusively occur in the hand
pelvic bone and subsequent extension into the peritoneal or wrist.148,162,159 They are soft, nodular masses treated by
cavity has also been reported.109 These cases are rare; in simple excision, which is curative.
most cases, locally destructive growth with recurrences is Although hemangiomas involving articular synovium
typical. Cases with usually aggressive behavior are some- occur more frequently than extraarticular hemangiomas
times referred to as malignant giant cell tumor of synovium.93 of tendons, they are still considered rare. Fewer than 200
cases of articular synovial hemangiomas have been
described in the world literature. These lesions almost
SYNOVIAL LIPOMA exclusively involve the knee and rarely involve the elbow
or the ankle. Some patients may have associated angioma-
Lipomas involving the articular capsule and tendons tosis in another location. Typically the lesion manifests
belong to the broad category of soft tissue lipomas. The during the second or third decade of life. Most patients
peak age incidence and the most frequent sites of involve- have a long history of dull pain, limitation of motion, and
ment are shown in Figure 20-44. A circumscribed lipoma a soft palpable mass. The mass decreases in size when the
of the joint capsule is extremely rare. The tumor is micro- extremity is elevated. Plain radiographs show an ill-
scopically identical to an ordinary lipoma in soft tissue defined, periarticular mass, which may contain phlebo-
and consists of mature adipose tissue. It may be pedun- liths. An associated degenerative change in the affected
culated or polypoid and may protrude into the joint joint and cystic lesions in the adjacent bone may be present.
space. Local excision is curative. Microscopically, synovial hemangiomas do not differ
A more common variant of lipoma involving the joint from ordinary hemangiomas of the soft tissue (Fig.
capsule is a diffuse adipose tissue overgrowth referred to 20-50). Typically, they are of capillary or mixed (capillary
as lipoma arborescens124-129,130-132,134-138,144 (Fig. 20-45). It dif- Text continued on p. 1313

ERRNVPHGLFRVRUJ
1304 20  Synovial Lesions

B
FIGURE 20-42  ■  Diffuse tenosynovial pigmented villonodular synovitis, extraarticular form: microscopic features. A, Low power
photomicrograph shows nodular infiltration of histiocytic cells with scattered multinucleated giant cells in synovium of tendon
sheath. B, High power photomicrograph shows sheets of histiocytes forming coalescent nodules. Cells containing vesicular nuclei
with prominent nucleoli but no cytologic atypia are noted. (A, ×10; B, ×200) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1305

B C
FIGURE 20-43  ■  Pigmented villonodular synovitis: microscopic features. A, Cords of histiocytic cells in hylanized stroma. B and
C, Higher magnification of A showing cords of epithelioid histiocytic cells in hyalinized fibrous stroma. This unique feature of
pigmented villonodular synovitis can be misinterpreted as diagnostic of low-grade epithelioid sclerosing fibrosarcoma. (A, ×100;
B and C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1306 20  Synovial Lesions

Incidence

Tendons

Articular
synovium

1 2 3 4 5 6 7 8
Age in decades

FIGURE 20-44  ■  Synovial lipoma. Peak age incidence and most frequently involved sites. Most frequent site is indicated by solid black
arrow.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1307

A B

C D

FIGURE 20-45  ■  Synovial lipoma. A, Gross specimen of excised pedunculated fatty tumor of synovium of knee joint. Tumor is covered
by thin layer of synovial lining. B, Lipoma arborescens representing broad-based polypoid fatty mass attached to synovium of knee
joint. Fine, villous fingerlike projections and clubbed fatty polyps are seen studding surface of tumor, which is composed of mature
fat. C, Diffuse lipomatosis of synovium of knee joint. Discrete polypoid masses of adipose tissue, each covered by a delicate synovial
lining, are seen with intervening zone of flat, uninvolved synovium. D, Higher magnification of specimen shown in C reveals discrete
polypoid masses of yellow adipose tissue.

ERRNVPHGLFRVRUJ
1308 20  Synovial Lesions

B
FIGURE 20-46  ■  Synovial lipoma: microscopic features. A, Lipoma arborescens of synovial membrane. Note multiple polypoid projec-
tions composed of adipose tissue. B, Some polypoid structures represent hyperplastic synovium with chronic inflammation.
Inset shows fingerlike projection, which represents chronically inflamed synovium. (A and B, ×25; Inset, ×50) (A, B, and
Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1309

B
FIGURE 20-47  ■  Synovial lipoma arborescens: microscopic features. A, Low power photomicrograph of polypoid projections of syno-
vial membrane. B, Higher magnification of A shows adipose tissue within one of polypoid projections. Inset shows pronounced
fibrosis, which is frequent in this type of lipoma. (A, ×10; B, ×25; Inset, ×50) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1310 20  Synovial Lesions

FIGURE 20-48  ■  Synovial lipoma: microscopic features. A and B, Prominent vasculature is frequent feature of synovial lipomas
(A, ×50; B, ×100) (A and B, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1311

Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 20-49  ■  Synovial hemangioma. Peak age incidence and most frequent involvement sites. Most frequent sites are indicated
by solid black arrows.

ERRNVPHGLFRVRUJ
1312 20  Synovial Lesions

A B

C D
FIGURE 20-50  ■  Synovial hemangioma: gross and microscopic features. A, Gross photograph of bivalved synovial nodule from knee
of patient who experienced recurrent hemorrhagic effusions without trauma. Note blood-filled dilated vascular channels embedded
in fat. B, Low power photomicrographs of cavernous hemangioma. C, Medium power photomicrograph of synovial hemangioma
showing cavernous and capillary vessels. D, Synovial hemangioma composed of small capillary-type vessels. (B, ×10; C and D, ×50)
(B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1313

and cavernous) type. Similar to hemangiomas in other 26. Norman A, Steiner GC: Bone erosion in synovial chondromatosis.
sites, synovial lesions may occasionally develop a papil- Radiology 161:749–752, 1986.
27. O’Connell JX: Pathology of the synovium. Am J Clin Pathol
lary endothelial hyperplasia of Masson’s type.152 114:773–784, 2000.
28. Pedowitz DI, Reddy SC, Parekh SG, et al: Recurrent para-
REFERENCES articular chondroma of the ankle: a case report. Foot Ankle Int
27:216–219, 2006.
Synovial Chondromatosis 29. Robinson D, Hasharoni A, Evron Z, et al: Synovial chondroma-
1. Abdelwahab IF, Contractor D, Bianchi S, et al: Synovial chondro- tosis: the possible role of FGF 9 and FGF receptor 3 in its pathol-
matosis of the lumbar spine with compressive myelopathy: a case ogy. Int J Exp Pathol 81:183–189, 2000.
report with review of the literature. Skeletal Radiol 37:863–867, 30. Rosati LA, Stevens C: Synovial chondromatosis of the temporo-
2008. mandibular joint presenting as an intracranial mass. Arch Otolar-
2. Adachi PL, Kaba SP, Martins MT, et al: Arthrocentesis in the yngol Head Neck Surg 116:1334–1337, 1990.
treatment of loose bodies of the temporomandibular joint associ- 31. Sarmiento A, Elkins RW: Giant intra-articular osteochondroma
ated with synovial chondromatosis. Br J Oral Maxillofac Surg of the knee. J Bone Joint Surg 57A:560–561, 1975.
46:320–321, 2008. 32. Shearer H, Stern P, Brubacher A, et al: A case report of bilateral
3. Ballard R, Weiland LH: Synovial chondromatosis of the temporo- synovial chondromatosis of the ankle. Chiropr Osteopat 15:18,
mandibular joint. Cancer 30:791–795, 1972. 2007.
4. Blandino A, Salvi L, Chirico G, et al: Synovial osteochondroma- 33. Silverman JS, Knapip M: Fibroma and giant cell tumor of tendon
tosis of the ankle: MR findings. Clin Imaging 16:34–36, 1992. sheath: a comparative histological and immunohistological study.
5. Bloom R, Pattinson JN: Osteochondromatosis of the hip joint. Mod Pathol 9:82–84, 1996.
J Bone Joint Surg 33A:80–84, 1951. 34. Sim FH, Dahlin DC, Ivins JC: Extra-articular synovial chondro-
6. Crawford A, Frazer A, Lippitt JM, et al: A case of chondromatosis matosis. J Bone Joint Surg 59A:492–495, 1977.
indicates a synovial stem cell aetiology. Rheumatology (Oxford) 45: 35. Steiner GC, Meushar N, Norman A, et al: Intracapsular and para-
1529–1533, 2006. articular chondromas. Clin Orthop 303:231–236, 1994.
7. Dunn AW, Whisler JH: Synovial chondromatosis of the knee with 36. Sviland L, Malcolm AJ: Synovial chondromatosis presenting as
associated extracapsular chondromas. J Bone Joint Surg 55A:1747– painless soft tissue mass: a report of 19 cases. Histopathology
1748, 1973. 27:275–279, 1995.
8. Edeiken J, Edeiken BS, Ayala AG, et al: Giant solitary synovial 37. Tan CH, Rai SB, Chandy J: MRI appearances of multiple rice
chondromatosis. Skeletal Radiol 23:23–29, 1994. body formation in chronic subacromial and subdeltoid bursitis, in
9. Errani C, Jutte PC, De Paolis M, et al: Secondary synovial chon- association with synovial chondromatosis. Clin Radiol 59:753–757,
dromatosis in a bursa overlying an osteochondroma mimicking a 2004.
peripheral chondrosarcoma—a case report. Acta Orthop 78:701– 38. Tibrewal SB, Iossifidis A: Extra-articular synovial chondromatosis
704, 2007. of the ankle. J Bone Joint Surg Br 77:659–660, 1995.
10. Fechner RE: Neoplasms and neoplasm-like lesions of the 39. Trias A, Quintana O: Synovial chondrometaplasia: review of world
synovium. In Ackerman LV, Spjut HJ, Abell MA, editors: Bones literature and study of 18 Canadian cases. Can J Surg 19:151–158,
and joints, IAP monograph, no. 17. Baltimore, 1976, Williams & 1976.
Wilkins. 40. Villacin AB, Brigham LN, Bullough PG: Primary and secondary
11. Hopyan S, Nadesan P, Yu C, et al: Dysregulation of hedgehog synovial chondrometaplasia: histopathologic and clinicoradiologic
signalling predisposes to synovial chondromatosis. J Pathol 206: differences. Hum Pathol 10:439–451, 1979.
143–150, 2005. 41. Wilmoth CL: Osteochondromatosis. J Bone Joint Surg Br 23:367–
12. Ida M, Yoshitake H, Okoch K, et al: An investigation of magnetic 374, 1971.
resonance imaging features in 14 patients with synovial chondro- 42. Xu WH, Ma XC, Guo CB, et al: Synovial chondromatosis of the
matosis of the temporomandibular joint. Dentomaxillofac Radiol temporomandibular joint with middle cranial fossa extension. Int
37:213–219, 2008. J Oral Maxillofac Surg 36:652–665, 2007.
13. Jeffreys TE: Synovial chondromatosis. J Bone Joint Surg Br 49:
530–534, 1967. Synovial Chondrosarcoma
14. Keogh CF, Torreggiani WC, Munk PL: Bilateral synovial chon- 43. Bhadra AK, Pollock R, Tirabosco RP, et al: Primary tumours of
dromatosis of the temporomandibular joint. Clin Radiol 57:862, the synovium. A report of four cases of malignant tumour. J Bone
2002. Joint Surg Br 89:1504–1508, 2007.
15. Kobayashi T, Fujikawa K, Sasazaki Y, et al: Osteochondromatosis 44. Bertoni F, Unni KK, Beabout JW, et al: Chondrosarcomas of the
with high concentration of procollagen II C peptide in joint fluid. synovium. Cancer 67:155–162, 1991.
Knee 9:165–167, 2002. 45. Campanacci DA, Matera D, Franchi A, et al: Synovial chondro-
16. Lee SK, Choy WS, Lee KW, et al: Synovial chondromatosis of sarcoma of the hip: report of two cases and literature review. Chir
the radiocarpal joint. Orthopedics 31:811, 2008. Organi Mov 92:139–144, 2008.
17. Lieger O, Zix J, Stauffer-Brauch EJ, et al: Synovial chondroma- 46. Dunn EJ, McGavran MH, Nelson P, et al: Synovial chondrosar-
tosis of the temporomandibular joint with cranial extension: a case coma: report of a case. J Bone Joint Surg 56A:811–813, 1974.
report and literature review. J Oral Maxillofac Surg 65:2073–2080, 47. Hamilton A, Davis RI, Hayes D, et al: Chondrosarcoma develop-
2007. ing in synovial chondromatosis: a case report. J Bone Joint Surg
18. McIvor RR, King D: Osteochondromatosis of the hip joint. J Bone Br 69:137–140, 1987.
Joint Surg 49A:530–534, 1967. 48. Kenan S, Abdelwahab IF, Klein MJ, et al: Case report 817: syno-
19. Milgram JW: The classification of loose bodies in human joints. vial chondrosarcoma secondary to synovial chondromatosis. Skel-
Clin Orthop 124:282–291, 1977. etal Radiol 22:623–626, 1993.
20. Milgram JW: The development of loose bodies in human joints. 49. Kindblom LG, Angervall L: Myxoid chondrosarcoma of the syno-
Clin Orthop 124:292–303, 1977. vial tissue: a clinicopathologic, histochemical and ultrastructural
21. Milgram JW: Synovial osteochondromatosis: a histopathological analysis. Cancer 52:1886–1895, 1983.
study of thirty cases. J Bone Joint Surg 59A:792–801, 1977. 50. King JW, Spjut HJ, Fechner RE, et al: Synovial chondrosarcoma
22. Mohr W: Is synovial osteo-chondromatosis a proliferative disease? of the knee joint. J Bone Joint Surg 49A:1389–1396, 1967.
Pathol Res Pract 198:585–588, 2002. 51. Manivel JC, Dehner LP, Thompson R: Case report 460: synovial
23. Murphy FP, Dahlin DC, Sullivan CR: Articular synovial chondro- chondrosarcoma of left knee. Skeletal Radiol 17:66–71, 1988.
matosis. J Bone Joint Surg 44A:77–86, 1962. 52. Perry BE, McQueen DA, Lin JJ: Synovial chondromatosis with
24. Nakanishi S, Sskamoto K, Yoshitake H, et al: Bone morphoge- malignant degeneration to chondrosarcoma: report of a case.
netic proteins are involved in the pathobiology of synovial chon- J Bone Joint Surg 70A:1259–1261, 1988.
dromatosis. Biochem Biophys Res Commun 379:914–919, 2009. 53. Sperling BL, Angel S, Stoneham G, et al: Synovial chondromato-
25. Nishimura K, Solchaga LA, Caplan AI, et al: Chondroprogenitor sis and chondrosarcoma: a diagnostic dilemma. Sarcoma 7:69–73,
cells of synovial tissue. Arthritis Rheum 42:2631–2637, 1999. 2003.

ERRNVPHGLFRVRUJ
1314 20  Synovial Lesions

54. Wittkop B, Davies AM, Mangham DC: Primary synovial chon- 76. Geldyyev A, Koleganova N, Piecha G, et al: High expression level
dromatosis and synovial chondrosarcoma: a pictorial review. Eur of bone degrading proteins as a possible inducer of osteolytic
Radiol 12:2112–2119, 2002. features in pigmented villonodular synovitis. Cancer Lett 8(255):
275–283, 2007.
Pigmented Villonodular Synovitis 77. Goldman AB, DiCarlo EF: Pigmented villonodular synovitis:
55. Abdul-Karim FW, El-Naggar AK, Joyce MJ, et al: Diffuse and diagnosis and differential diagnosis. Radiol Clin North Am 26:1327–
localized tenosynovial giant cell tumor and pigmented villonodu- 1347, 1988.
lar synovitis: a clinicopathologic and flow cytometric DNA analy- 78. Gonzalez-Campora R, Salas Herrero E, Otal-Salaverri C, et al:
sis. Hum Pathol 23:729–735, 1992. Diffuse tenosynovial giant cell tumor of soft tissues: report of a
56. Berger I, Weckauf H, Helmchen B, et al: Rheumatoid arthritis case with cytologic and cytogenetic findings. Acta Cytol 39:770–
and pigmented villonodular synovitis: comparative analysis of cell 776, 1995.
polyploidy, cell cycle phases and expression of macrophage and 79. Granowitz SP, D’Antonio J, Mankin HL: The pathogenesis and
fibroblast markers in proliferating synovial cells. Histopathology long-term end results of pigmented villonodular synovitis. Clin
46:490–497, 2005. Orthop 114:335–351, 1976.
57. Bobechko WP, Kostiuk JP: Childhood villonodular synovitis. Can 80. Greenfield MM, Wallace KM: Pigmented villonodular synovitis.
J Surg 11:480–486, 1968. Radiology 54:350–356, 1950.
58. Breimer CW, Freiberger RH: Bone lesions associated with villo- 81. Jaffe HL, Lichtenstein L, Sutro CJ: Pigmented villonodular syno-
nodular synovitis. Am J Roentgenol Radium Ther Nucl Med 79:618– vitis, bursitis and tenosynovitis. Arch Pathol 31:731–765, 1941.
629, 1958. 82. Janssens X, Veys EM, Cuvelier C: Pigmented villonodular syno-
59. Butt WP, Hardy G, Ostlere SJ: Pigmented villonodular synovitis vitis of the hip: association with osteochondromatosis. Clin Exp
of the knee: computed tomographic appearances. Skeletal Radiol Rheumatol 5:329–334, 1987.
19:191–196, 1990. 83. Jelinek JS, Kransdorf MJ, Shmookler BM, et al: Giant cell tumor
60. Byer PD, Cotton RE, Deacon OW, et al: The diagnosis and treat- of the tendon sheath: MR findings in nine cases. AJR Am J Roent-
ment of pigmented villonodular synovitis. J Bone Joint Surg Br 50: genol 162:919–922, 1994.
290–305, 1968. 84. Jergesen HE, Mankin HJ, Schiller AL: Diffuse pigmented villon-
61. Choong PF, Willen H, Nilbert M, et al: Pigmented villonodular odular synovitis of the knee mimicking primary bone neoplasms:
synovitis: monoclonality and metastasis—a case for neoplastic a report of two cases. J Bone Joint Surg 60A:825–829, 1978.
origin? Acta Orthop Scand 66:64–68, 1995. 85. Kang GH, Chi JG, Choi IH: Pigmented villonodular synovitis in
62. Chung SMK, Janes JM: Diffuse pigmented villonodular synovitis the sacral joint with extensive bone destruction in a child. Pediatr
of the hip joint: review of the literature and report of four cases. Pathol 12:725–730, 1992.
J Bone Joint Surg 47A:293–303, 1965. 86. Khoury GM, Shimkin PM, Kleinman GM, et al: Computed
63. Clark LJ, McCormick PW, Domenico DR, et al: Pigmented vil- tomography and magnetic resonance imaging findings of pig-
lonodular synovitis of the spine. Case report. J Neurosurg 79:456– mented villonodular synovitis of the spine. Spine 16:1236–1237,
459, 1993. 1991.
64. Cotten A, Flipo RM, Chastanet P, et al: Pigmented villonodular 87. Kindblom LG, Gunterberg B: Pigmented villonodular synovitis
synovitis of the hip: review of radiographic features in 58 patients. involving bone: case report. J Bone Joint Surg 60A:830–832,
Skeletal Radiol 24:1–6, 1995. 1978.
65. Crosby EB, Inglis A, Bullough PG: Multiple joint involvement 88. Levin EJ, Gannon W: Diffuse villonodular synovitis of the
with pigmented villonodular synovitis. Radiology 122:671–672, shoulder. Am J Roentgenol Radium Ther Nucl Med 89:1302–1304,
1977. 1963.
66. Cupp JS, Miller MA, Montgomery KD, et al: Translocation and 89. Linn SC, West RB, Pollack JR, et al: Gene expression patterns
expression of CSF1 in pigmented villonodular synovitis, tenosy- and gene copy number changes in dermatofibrosarcoma protuber-
novial giant cell tumor, rheumatoid arthritis and other reactive ans. Am J Pathol 163:2383–2395, 2003.
synovitides. Am J Surg Pathol 31:970–976, 2007. 90. Maluf HM, DeYoung BR, Swanson PE, et al: Fibroma and giant
67. Dahlén A, Broberg K, Domanski HA, et al: Analysis of the distri- cell tumor of tendon sheath: a comparative histological and immu-
bution and frequency of trisomy 7 in vivo in synovia from patients nohistological study. Mod Pathol 8:155–159, 1995.
with osteoarthritis and pigmented villonodular synovitis. Cancer 91. Mandelbaum BR, Grant TT, Hartzman S, et al: The use of MRI
Genet Cytogenet 131:19–24, 2001. to assist in diagnosis of pigmented villonodular synovitis of the
68. Dal Cin P, Sciot R, Samson I, et al: Cytogenetic characterization knee joint. Clin Orthop 231:135–139, 1988.
of tenosynovial giant cell tumors (nodular tenosynovitis). Cancer 92. Mertens F, Orndal C, Mandahl N, et al: Chromosome aberrations
Res 1(54):3986–3987, 1994. in tenosynovial giant cell tumors and nontumorous synovial tissue.
69. Docken WP: Pigmented villonodular synovitis: a review with Genes Chromosomes Cancer 6:212–217, 1993.
illustrative case reports. Semin Arthritis Rheum 9:1–22, 1979. 93. Nakashima M, Uchida T, Tsukazaki T, et al: Expression of tyro-
70. Dorwart RH, Genant HK, Johnston WH, et al: Pigmented sine kinase receptors Tie-1 and Tie-2 in giant cell tumor of the
villonodular synovitis of synovial joints: clinical, pathologic, tendon sheath: a possible role in synovial proliferation. Pathol Res
and radiologic features. AJR Am J Roentgenol 143:877–885, Pract 197:101–107, 2001.
1984. 94. Nielsen AL, Kiaer T: Malignant giant cell tumor of synovium and
71. Finis K, Sültmann H, Ruschhaupt M, et al: Analysis of pigmented locally destructive pigmented villonodular synovitis: ultrastruc-
villonodular synovitis with genome-wide complementary DNA tural and immunohistochemical study and review of the literature.
microarray and tissue array technology reveals insight into poten- Hum Pathol 20:765–771, 1989.
tial novel therapeutic approaches. Arthritis Rheum 54:1009–1019, 95. Nilsonne U, Moberger G: Pigmented villonodular synovitis of
2006. joints: histological and clinical problems in diagnosis. Acta Orthop
72. Fiocco U, Sfriso P, Lunardi F, et al: Molecular pathways involved Scand 40:448–460, 1969.
in synovial cell inflammation and tumoral proliferation in diffuse 96. Nilsson M, Höglund M, Panagopoulos I, et al: Molecular cytoge-
pigmented villonodular synovitis. Autoimmun Rev 9:780–784, netic mapping of recurrent chromosomal breakpoints in tenosy-
2010. novial giant cell tumors. Virchows Arch 441:475–480, 2002.
73. Fletcher JA, Henkle C, Atkins L, et al: Trisomy 5 and trisomy 7 97. O’Connell JX, Fanburg JC, Rosenberg AE: Giant cell tumor of
are non-random aberrations in pigmented villonodular synovitis: tendon sheath and pigmented villonodular synovitis: immunophe-
confirmation of trisomy 7 in uncultured cells. Genes Chromosomes notype suggests a synovial cell origin. Hum Pathol 26:771–775,
Cancer 4:264–266, 1992. 1995.
74. Fujita S, Iizuka T, Tuboi Y, et al: Synovial chondromatosis of the 98. O’Sullivan B, Cummings B, Catton C, et al: Outcome following
temporomandibular joint with immunohistochemical findings: radiation treatment for high-risk pigmented villonodular synovi-
report of a case. J Oral Maxillofac Surg 49:880–883, 1991. tis. Int J Radiat Oncol Biol Phys 32:777–786, 1995.
75. Gehweiler JA, Wilson JW: Diffuse villonodular synovitis of the 99. Ohjimi Y, Iwasaki H, Ishiguro M, et al: Short arm of chromosome
shoulder. Am J Roentgenol Radium Ther Nucl Med 93:845–851, 1 aberration recurrently found in pigmented villonodular synovi-
1969. tis. Cancer Genet Cytogenet 90:80–85, 1996.

ERRNVPHGLFRVRUJ
20  Synovial Lesions 1315

100. Peterson LFA, Johnson EW, Jr, Woolner LB: Extra-articular pig- a translocation in a minority of tumor cells. Proc Natl Acad Sci
mented villonodular synovitis of the knee. Am J Clin Pathol U S A 103:690–695, 2006.
30:158–162, 1958.
101. Phalen GS, McCormick LJ, Gazale WJ: Giant-cell tumor of the Synovial Lipoma
tendon sheath (benign synovioma) in the hand: evaluation of 56 125. Adelani MA, Wupperman RM, Holt GE: Benign synovial disor-
cases. Clin Orthop 15:140–151, 1959. ders. J Am Acad Orthop Surg 16:268–275, 2008.
102. Rao AS, Vigorita VJ: Pigmented villonodular synovitis (giant-cell 126. Arzimanoglu A: Bilateral arborescent lipoma of the knee: a case
tumor of the tendon sheath and synovial membrane): a review of report. J Bone Joint Surg 39A:976–979, 1957.
81 cases. J Bone Joint Surg 66A:76–94, 1984. 127. Bejia I, Younes M, Moussa A, et al: Lipoma arborescens affecting
103. Ray RA, Morton CC, Lipinski KK, et al: Cytogenetic evidence of multiple joints. Skeletal Radiol 34:536–538, 2005.
clonality in a case of pigmented villonodular synovitis. Cancer 128. Blais RE, LaPrade RF, Chaljub G, et al: The arthroscopic appear-
67:121–125, 1991. ance of lipoma arborescens of the knee. Arthroscopy 11:623–627,
104. Rowlands CG, Roland B, Hwang WS, et al: Diffuse-variant teno- 1995.
synovial giant cell tumor: a rare and aggressive lesion. Hum Pathol 129. Burgan DW: Lipoma arborescens of the knee: another cause
25:423–425, 1994. of filling defects on a knee arthrogram. Radiology 101:583–584,
105. Sakkers RJ, de Jong D, van der Heul RO: X chromosome inactiva- 1971.
tion in patients who have pigmented villonodular synovitis. J Bone 130. Chaljub G, Johnson PR: In vivo MRI characteristics of lipoma
Joint Surg 73A:1532–1536, 1991. arborescens utilizing fat suppression and contrast administration.
106. Schumacher HR, Lotke P, Athreya B, et al: Pigmented villonodu- J Comput Assist Tomogr 20:85–87, 1996.
lar synovitis: light and electron microscopic studies. Semin Arthri- 131. Cil A, Atay OA, Aydingöz U, et al: Bilateral lipoma arborescens
tis Rheum 12:32–43, 1982. of the knee in a child: a case report. Knee Surg Sports Traumatol
107. Sciot R, Rosai J, Dal Cin P, et al: Analysis of 35 cases of localized Arthrosc 13:463–467, 2005.
and diffuse tenosynovial giant cell tumor: a report from the Chro- 132. Davies AP, Blewitt N: Lipoma arborescens of the knee. Knee
mosomes and Morphology (CHAMP) study group. Mod Pathol 12:394–396, 2005.
12:576–579, 1999. 133. Dawson JS, Dowling F, Preston BJ, et al: Case report: lipoma
108. Scott PM: Bone lesions in pigmented villonodular synovitis. arborescens of the sub-deltoid bursa. Br J Radiol 68:197–199,
J Bone Joint Surg Br 50:306–311, 1968. 1995.
109. Seki K, Hirose T, Hasegawa T, et al: Giant cell tumor of tendon 134. Dinauer P, Bojescul JA, Kaplan KJ, et al: Bilateral lipoma arbo-
sheath: an immunohistochemical observation on the characteris- rescens of the bicipitoradial bursa. Skeletal Radiol 31:661–665,
tics and the capacity of proliferation of tumor cells. Zentralbl 2002.
Pathol 139:287–294, 1993. 135. Grieten M, Buckwalter KA, Cardinal E, et al: Case report 873:
110. Shinjo K, Miyake N, Takahashi Y: Malignant giant cell tumor of lipoma arborescens (villous lipomatous proliferation of the syno-
the tendon sheath: an autopsy report and review of the literature. vial membrane). Skeletal Radiol 23:652–655, 1994.
Jpn J Clin Oncol 23:317–324, 1993. 136. Hallel T, Lew S, Bansal M: Villous lipomatous proliferation of
111. Silverman JS, Knapik MI: Fibroma of tendon sheath and tenosy- the synovial membrane (lipoma arborescens). J Bone Joint Surg
novial giant cell tumors are rich in factor XIIIA+ dendrophages. 70A:264–270, 1988.
J Histotech 19:45–53, 1996. 137. Laorr A, Peterfy CG, Tirman PF, et al: Lipoma arborescens of
112. Smith JH, Pugh DG: Roentgenographic aspects of articular pig- the shoulder: magnetic resonance imaging findings. Can Assoc
mented villonodular synovitis. Am J Roentgenol Radium Ther Nucl Radiol J 46:311–313, 1995.
Med 87:1146–1156, 1962. 138. Martinez D, Millner PA, Coral A, et al: Case report 745: synovial
113. Steinbach LS, Neumann CH, Stoller DW, et al: MRI of the knee lipoma arborescens. Skeletal Radiol 21:393–395, 1992.
in diffuse pigmented villonodular synovitis. Clin Imaging 13:305– 139. Noel ER, Tebib JG, Dumontet C, et al: Synovial lipoma arbore-
316, 1989. scens of the hip. Clin Exp Rheumatol 6:92–96, 1987.
114. Sundaram M, McGuire MH, Fletcher J, et al: Magnetic resonance 140. Pandey T, Alkhulaifi Y: Bilateral lipoma arborescens of the sub-
imaging of lesions of synovial origin. Skeletal Radiol 15:110–116, deltoid bursa. Australas Radiol 50:487–489, 2006.
1986. 141. Sağlik Y, Akmeşe R, Yildiz Y, et al: Lipoma arborescens occurring
115. Tartaglia L, Chiroff RT: Diffuse pigmented villonodular synovitis: in both knees at different times: a case report. Acta Orthop Trau-
an indication for total hip replacement in the young patient. Clin matol Turc 40:176–180, 2006.
Orthop 115:172–176, 1976. 142. Siva C, Brasington R, Totty W, et al: Synovial lipomatosis
116. Tashiro H, Iwasaki H, Kikuchi M, et al: Giant cell tumors of (lipoma arborescens) affecting multiple joints in a patient with
tendon sheath: a single and multiple immunostaining analysis. congenital short bowel syndrome. J Rheumatol 29:1088–1092,
Pathol Int 45:147–155, 1995. 2002.
117. Uchibori M, Nishida Y, Tabata I, et al: Expression of matrix 143. Sullivan CR, Dahlin DC, Bryan RS: Lipoma of the tendon sheath.
metalloproteinases and tissue inhibitors of metalloproteinases in J Bone Joint Surg 38A:1275–1280, 1956.
pigmented villonodular synovitis suggests their potential role for 144. Vilanova JC, Barceló J, Villalón M, et al: MR imaging of lipoma
joint destruction. J Rheumatol 31:110–119, 2004. arborescens and the associated lesions. Skeletal Radiol 32:504–509,
118. Ushijima M, Hashimoto H, Tsuneyoshi M, et al: Giant cell tumor 2003.
of the tendon sheath (nodular tenosynovitis): a study of 207 cases 145. Weitzman G: Lipoma arborescens of the knee: report of a case.
to compare the large joint group with the common digit group. J Bone Joint Surg 47A:1030–1033, 1965.
Cancer 57:875–884, 1986. 146. White JR: Arborescent lipomata of tendon sheaths; a report of
119. Wagner ML, Spjut HJ, Dutton RV, et al: Polyarticular pigmented- two cases. Surg Obstet Gynecol 38:489–490, 1924.
villonodular synovitis. AJR Am J Roentgenol 136:821–823, 1981.
120. Weckauf H, Helmchen B, Hinz U, et al: Expression of cell cycle- Synovial Hemangioma
related gene products in different forms of primary versus recur- 147. Abe T, Tomatsu T, Tazaki K: Synovial hemangioma of the knee
rent PVNS. Cancer Lett 210:111–118, 2004. in young children. J Pediatr Orthop B 11:293–297, 2002.
121. Weidner N, Challa VR, Bonsib SM, et al: Giant cell tumors of 148. Atkinson TJ, Wolf S, Anavi Y, et al: Synovial hemangioma of the
synovium (pigmented villonodular synovitis) involving the verte- temporomandibular joint: report of a case and review of the lit-
bral column. Cancer 57:2030–2036, 1986. erature. J Oral Maxillofac Surg 46:804–808, 1988.
122. Wendt RG, Wolfe F, McQueen D, et al: Polyarticular pigmented 149. Bate TH: Hemangiomata of the tendon sheath. J Bone Joint Surg
villonodular synovitis in children: evidence for a genetic contribu- 36A:104–109, 1954.
tion. J Rheumatol 13:921–926, 1986. 150. Bruns J, Eggers-Stroeder G, von Torklus D: Synovial hemangi-
123. Yoshida W, Uzuki M, Kurose A, et al: Cell characterization of oma: a rare benign synovial tumor—report of four cases. Knee
mononuclear and giant cells constituting pigmented villonodular Surg Sports Traumatol Arthrosc 2:186–189, 1994.
synovitis. Hum Pathol 34:65–73, 2003. 151. Busa R, Adani R, Marcuzzi A, et al: Acute posterior interosseous
124. West RB, Rubin BP, Miller MA, et al: A landscape effect in teno- nerve palsy caused by a synovial haemangioma of the elbow joint.
synovial giant-cell tumor from activation of CSF1 expression by J Hand Surg [Br] 20:652–654, 1995.

ERRNVPHGLFRVRUJ
1316 20  Synovial Lesions

152. Cotten A, Flipo RM, Herbaux B, et al: Synovial haemangioma of 159. Lenchik L, Poznanski AK, Donaldson JS, et al: Case report 681:
the knee: a frequently misdiagnosed lesion. Skeletal Radiol 24:257– synovial hemangioma of the knee. Skeletal Radiol 20:387–389,
261, 1995. 1991.
153. Devaney K, Vinh TN, Sweet DE: Synovial hemangioma: a report 160. Lewis RC, Jr, Coventry MB, Soule EH: Hemangioma of the
of 20 cases with differential diagnostic considerations. Hum Pathol synovial membrane. J Bone Joint Surg 41A:264–271, 1959.
24:737–745, 1993. 161. Llauger J, Monill JM, Palmer J, et al: Synovial hemangioma of
154. Eichhorn JH, Rosenberg AE: Intravascular papillary endothelial the knee: MRI findings in two cases. Skeletal Radiol 24:579–581,
hyperplasia involving the synovium. Arch Pathol Lab Med 112:647– 1995.
650, 1988. 162. Moon NF: Synovial hemangioma of the knee joint: a review of
155. Forrest J, Staple TW: Synovial hemangioma of the knee: demon- previously reported cases and inclusion of two new cases. Clin
stration by arthrography and arteriography. Am J Roentgenol Orthop 90:183–190, 1973.
Radium Ther Nucl Med 112:512–516, 1971. 163. Rico AA, Holguin PH, Gonzalez IG, et al: Flexor tendon synovial
156. Greenspan A, Azouz EM, Matthews J, II, et al: Synovial heman- sheath haemangioma mimicking subacute tenosynovitis. J Hand
gioma: imaging features in eight histologically proven cases, Surg [Br] 19:704–705, 1994.
review of the literature, and differential diagnosis. Skeletal Radiol 164. Sarma UC, Dave PK, Verma K, et al: Synovial haemangioma of
24:583–590, 1995. knee joint. Indian J Pediatr 57:122–124, 1990.
157. Halborg A, Hansen H, Sneppen HO: Haemangioma of the knee 165. Seimon LP, Hekmat F: Synovial hemangioma of the knee.
joint. Acta Orthop Scand 39:209–216, 1968. J Pediatr Orthop 6:356–359, 1986.
158. Karlholm S, Stjernsward J: Hemangioma of the knee joint. Acta 166. Wirth T, Rauch G, Ruschoff J, et al: Synovial haemangioma of
Orthop Scand 33:306–311, 1963. the knee joint. Int Orthop 16:130–132, 1992.

ERRNVPHGLFRVRUJ
C H A P T E R 2 1 

Sclerosing Bone Lesions

CHAPTER OUTLINE

OSTEOMA AND BONE ISLAND OSTEOPOIKILOSIS (SPOTTED BONE DISEASE)


Ivory Exostosis OSTEOPETROSIS
Sinoorbital Osteoma
Bone Island (Enostosis) MELORHEOSTOSIS
Surface Osteoma of Long Bones

Sclerosing bone lesions represent a unique group of Some authors consider these lesions to be hamartoma-
dysplastic anomalies with a wide range of clinical presen- tous or dysplastic in nature, but there is a rationale for
tations, radiographic characteristics, and genetic back- regarding those that affect the orbit, facial bones, and
grounds.7 They can be separated into two major groups paranasal sinuses (sinoorbital osteoma) as benign bone-
with hereditary and nonhereditary clinical presentations.3 forming tumors.
A comprehensive description of all conditions included
in the group is beyond the scope of this book. In this
chapter, we describe some of these conditions that may
Ivory Exostosis
occasionally simulate a neoplasm and may require micro- Ivory exostoses represent a type of osteoma that affects
scopic examination to confirm the diagnosis or to rule bones formed by membranous ossification, chiefly affect-
out a neoplastic process. ing calvarial and mandibular surfaces2,4,6,10–12,20,22,28 (Fig.
In the radiologic literature, these conditions are 21-2). Multiple osteomas of this type are frequently asso-
grouped into two major categories: those associated with ciated with colonic polyposis and fibrous soft tissue
abnormal enchondral ossification and those primarily tumors, including mesenteric fibromatosis and epidermal
affecting the sites of membranous bone formation. It is inclusion cysts in the familial disorder known as Gard-
generally accepted that the common feature of the condi- ner’s syndrome.13,14,17,24,29 Some patients also had distinct
tions described in this chapter is that they all arise from hyperplastic changes of the retinal pigmented epithe-
defects in bone resorption or formation during the lium.22,32,33 The development of ocular lesions may
process of skeletal development, maturation, and remod- precede the development of colonic and osseous mani-
eling.1,5,8,10 Sometimes several such conditions can coexist, festations. The polyps of the gastrointestinal tract are
resulting in the so-called overlapping dysplastic syndromes, adenomatous and typically involve the colon but can also
which further supports the concept of their unified be present in the duodenum and stomach. The adenoma-
pathogenesis.5,6,9 An abbreviated list of sclerosing bone tous polyps of the gastrointestinal tract have a high pro-
dysplasias classified on the basis of the so-called target- pensity for malignant transformation. In fact, colon
site approach originally proposed by Norman and cancer develops in all affected individuals unless prophy-
Greenspan5,10 (i.e., specific phase, site of skeletal develop- lactic colectomy is performed.23 The disorder is transmit-
ment affected, or both) is provided in Table 21-1. A ted by an autosomal dominant pattern of inheritance.
summary of clinical presentations, radiographic charac- Similar to other major familial colonic polyposis syn-
teristics, and genetic alterations implicated in the devel- dromes, such as flat adenoma and Turcot’s syndrome
opment of hereditary bone dysplasia is provided in (colorectal polyposis with brain tumor), Gardner’s syn-
Table 21-2.7 drome is linked to a malfunctioning adenomatous pol-
yposis coli (APC) gene mapped to chromosome 5q21.26
Mutations involving different regions of APC are associ-
OSTEOMA AND BONE ISLAND ated with the severity of the clinical syndrome and the
extent of extracolonic manifestations. It has been shown
Several benign, slow-growing bony lesions are identified that mutations involving the 38 end around codon 1444
under the general heading of osteoma (Fig. 21-1). They of the APC gene are associated with particularly severe
can be divided into (1) calvarial and mandibular ivory osseous manifestations.16 Patients with this type of APC
exostoses; (2) osteomas of the paranasal sinuses, facial alteration also have a higher incidence of soft tissue fibro-
bones, and orbit (sino-orbital osteomas); (3) enostoses or matosis. The soft tissue fibrous lesions range from aggres-
bone islands; and (4) surface (juxtacortical) osteomas of sive fibromatosis at one end of the spectrum to a lesion
long bones. which is histologically similar to nuchal-type fibroma.15,19,27
1317
ERRNVPHGLFRVRUJ
1318 21  Sclerosing Bone Lesions

Bone island
(enostosis)

Sinoorbital
Ivory Surface
osteoma
exostosis osteoma
of long bones

Associated with
Gardner’s syndrome

FIGURE 21-1  ■  Clinicopathologic variants of osteoma. Lesions classified microscopically as osteomas can be separated into several
distinct clinicopathologic variants; ivory exostosis, sinoorbital osteoma, bone island, and surface osteoma of long bones.

stain positively for CD34 and CD99. Many of these


TABLE 21-1 Classification of Sclerosing
lesions show positive nuclear immune-reactivity for
Dysplasias of Bone
β-catenin.15,19 Approximately 50% of patients with
Dysplasias of Enchondral Bone Formation Gardner-associated fibromas eventually develop a
Affecting primary spongiosa (immature bone) desmoid-type fibromatosis at the same site. Overall, 10%
Osteopetrosis* to 15% of patients with Gardner’s syndrome develop
Affecting secondary spongiosa (mature bone) aggressive desmoid-type fibromatosis. They typically
Osteoma*
Bone islands*
develop intraabdominally spontaneously or more often
Osteopoikilosis* after surgery. Similar to Gardner-associated fibromas,
desmoid fibromatoses associated with this syndrome
Dysplasias of Membranous Bone Formation harbor the inactivating mutations of the APC gene.
Progressive diaphyseal dysplasia However, similar to sporadic fibromatoses, they are posi-
Hereditary multiple diaphyseal sclerosis
Endosteal hyperostosis
tive immunohistochemically for nuclear β-catenin.15,19 In
addition to osteomas, which are often multiple, dental
Mixed Sclerosing Dysplasias (Affecting Both Enchondral malformation or more diffuse sclerotic changes in the
and Membranous Ossification) craniofacial region may develop in patients with Gard-
Melorheostosis (predominantly membranous ossification)* ner’s syndrome.25 Osteomas in Gardner’s syndrome may
*Described in this chapter. increase in size over time; that is, progressively more
Modified and used with permission from Greenspan A: Skelet severe malformation of the craniofacial bones may
Radiol 20:561–583, 1991. develop.31 Osseous lesions in Gardner’s syndrome do not
behave as true neoplasms, and there is no evidence of
their evolution into malignant lesions. Moreover, the
These fibromas of low cellularity have been designated presence of osteoblastoma-like areas, predominantly seen
as Gardner fibromas, and their significance as early indi- in sporadic sinoorbital osteomas, is not a feature of
cators of the development of other manifestations of osseous lesions in Gardner’s syndrome.
Gardner’s syndrome such as desmoid-type fibromatosis Microscopically, ivory exostoses are button-like excres-
has been proposed.15,34 Unlike typical nuchal-type fibro- cences of mature lamellar bone limited to the cortical
mas, which most often affect patients in the third through surfaces and usually continuous with the outer table of
fifth decades of life, Gardner-associated fibromas arise in the skull or the mandibular cortex. The dense cortical
younger patients and the majority of them present in the lamellar bone making up these exostoses contains osteons
first decade of life. The most common location is in the with haversian canals. Rare cases of compact surface
back and paraspinal region followed by the head and neck osteoma have been reported in other flat bones as well as
and extremities. Immunohistochemically, these lesions on long bones.44

ERRNVPHGLFRVRUJ

TABLE 21-2 Summary of the Hereditary Sclerosing Bone Dysplasias


Dysplasia Inheritance Pattern Genetics Type of Dysplasia Age at Onset Osseous Findings Other Findings
Osteopetrosis AR, AD TCIRG1 (ATP6i), Endochondral (primary Stillbirth or infancy Diffusely increased Anemia, cranial nerve
CLCN7, GL spongiosa) to adulthood bone density; type deficits (rare with AD
(OSTM1), RANKL, 1: uniform sclerosis inheritance pattern)
CA2, PLEKHM1 of the skull, spine,
and long bones;
type 2: endobone
appearance
Pyknodysostosis AR Cathepsin K Endochondral (primary Infancy or early Hyperostosis of long Dwarfiism, acro-osteolysis,
spongiosa) childhood bones with wormian bones
preserved
medullary cavities
Osteopoikilosis AD LEMD3 Endochondral Childhood or Multiple enostoses Dermatofibrosis
(secondary spongiosa) adulthood lenticularis disseminate
Osteopathia striata X linked Unknown Endochondral Childhood or Dense striations in No other associated
(secondary spongiosa) adulthood metadiaphyses of abnormalities
(incidental finding) long bones
Progressive AD LAP of TGFB1 Intramembranous Childhood Bilateral/symmetric Gait disturbances, pain,
diaphyseal ossification periosteal and weakness
dysplasia endosteal cortical
thickening involving
long bones or
calvaria

ERRNVPHGLFRVRUJ
Hereditary multiple AR Unknown Intramembranous After puberty Unilateral/asymmetric Milder neuromuscular
diaphyseal ossification cortical thickening symptoms than with
sclerosis involving long progressive diaphyseal
bones only dysplasia
Hyperostosis AR (Van Buchem SOST (Wnt signaling Intramembranous Childhood Endosteal cortical Van Buchem disease:
corticalis disease, pathway), LRP5 ossification thickening involving facial nerve palsy;
generalisata sclerosteosis), AD (Wnt signaling the long bones, sclerosteosis: facial
(Worth disease) pathway) skull, and facial nerve palsy, syndactyly,
bones; mandible bone overgrowth; Worth
enlargement disease: no cranial nerve
palsy, torus palatinus

Note: AD, Autosomal dominant; AR, Autosomal recessive; ATP, Adenosine triphosphate; CA2, Carbonic anhydrase II; CLCN7, Chloride 7 channel; GL, Gray-lethal; LAP, Latency-associated
protein; LEMD3, LEM domain-containing protein 3; LRP5, Lipoprotein receptor-related protein 5; OSTM1, Osteopetrosis-associated transmembrane protein 1; PLEKHM1, Pleckstrin
homology domain-containing family M member 1; RANKL, Receptor activator of nuclear factor κ-B ligand; SOST, Sclerostin; TCIRG1, T-cell immune regulator 1; TGFB1, Transforming
growth factor β1.
Reprinted with permission from Ihde LL et al: RadioGraphics 31:1865–1882, 2011.
21  Sclerosing Bone Lesions
1319
1320 21  Sclerosing Bone Lesions

A B

C
FIGURE 21-2  ■  Ivory exostosis (parosteal osteoma) of skull: radiographic and histologic features. A, Lateral radiograph of skull shows
frontal region with buttonlike surface osteoma. B, Specimen radiograph of excised, ivory-like excrescence removed from frontal
regions of skull. C, Medium power photomicrograph of specimen shown in B. Compact bone with fibrous stroma shows focal
osteoclastic resorptive activity (C, ×25) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1321

Sinoorbital Osteoma Biopsy specimens from small bone islands are rarely
obtained, but larger lesions may raise the suspicion of a
Osteomas involving paranasal sinuses, orbit, and facial neoplasm, such as a sclerotic variant of osteosarcoma. We
bones tend to project into the sinus cavity from a broad also have seen a giant bone island that was misinterpreted
attachment to the affected bone (Figs. 21-3 and 21- microscopically as a low-grade intraosseous fibroblastic
4).21,33,34,36,39,41,42,45,46 Some osteomas involving this region, osteosarcoma. Although some osteoblastic rimming may
especially those that are multifocal, are associated with be present within dilated haversian channels of giant bone
Gardner’s syndrome. Microscopically, they are indistin- islands, the spindle-cell stroma and infiltrative pattern of
guishable from ivory exostosis. In contrast, the so-called growth characteristic of low-grade intraosseous fibroblas-
sporadic sinoorbital osteomas (not associated with Gardner’s tic osteosarcoma are not present in these lesions.
syndrome) are usually composed of dense, immature
bone and may have osteoblastic rimming as well as signs
of bone resorption (Fig. 21-5). These lesions may contain
Surface Osteoma of Long Bones
central or peripheral foci of spongy or cancellous appear- Surface osteoma that involves the clavicle, pelvic, or long
ance (Figs. 21-6 and 21-7). Sometimes the latter areas bones of the extremities is the rarest form of osteoma.61-69
show active remodeling, with cellular fibrous tissue filling On radiographs, it appears to resemble ivory exostosis
the interstices, osteoblastic activity, and osteoclastic (surface osteoma) of the craniofacial bones (Fig. 21-14).
resorption.33 In ethmoid and frontal sinus osteomas, Similar to other osteomas, this lesion is composed of
these lesions may occasionally contain radiolucent areas mature lamellar bone organized into osteons and haver-
that are histologically indistinguishable from conven- sian canals. The outer surface is covered by a thin layer
tional osteoblastoma and even aggressive osteoblastoma of fibrous tissue. There is no cartilaginous cap covering
(Fig. 21-8).38,40,47 The latter cases sometimes resemble the lesion. The underlying cortex may be thickened but
multifocal osteoblastomas arising within osteomas. The is otherwise intact. No continuity between the lesion and
clinical behavior of these lesions is similar to that seen in the underlying medullary cavity can be seen grossly or on
osteoblastomas and aggressive osteoblastoma groups; radiographs. Osteoma that involves the surface of a long
that is, they exhibit a local destructive growth pattern and bone must be radiographically and microscopically dif-
recurrences. For this reason, we prefer to regard the ferentiated from parosteal osteosarcoma, melorheostosis, and
sporadic osteomas that arise in the orbit, paranasal osteochondroma. The spindle-cell fibroblastic stroma that
sinuses, and facial bones, especially those that contain is characteristic of parosteal osteosarcoma is not present
osteoblastoma-like foci or features of active bone produc- in osteoma. Moreover, the bone in osteoma is mature and
tion and remodeling, as benign bone-forming neoplasms forms solid areas with recognizable haversian canals,
rather than as hamartomas or dysplasias. Osteomas in this which are typically not present in parosteal osteosarcoma.
region typically do not exceed 2 cm in diameter. Occa- The distinction from melorheostosis is based mainly on
sionally, they attain a larger size (giant osteoma) that the radiographic pattern of the bone surface lesion. Mul-
compresses adjacent structures and cause a disfiguring tifocality and the so-called wax-dripping patterns are not
deformity.28,35,37,43 present in surface osteoma. In contrast to osteochon-
droma, parosteal osteoma is not covered by a cartilagi-
Bone Island (Enostosis) nous cap, and there is no continuation between the lesion
and the underlying medullary cavity.
Bone islands are foci of dense, compact bone within
the medullary cavity (i.e., within cancellous bone) (Fig.
21-9).48,50,51,54 Occasionally, they may be attached to the OSTEOPOIKILOSIS (SPOTTED
inner surface of the cortex. They are ovoid, with the long BONE DISEASE)
axis parallel to the cortex of the affected bone. Most bone
islands are small (less than 1 cm in diameter), and the Osteopoikilosis is closely related to osteomas and bone
majority of lesions measure from 0.1 to 2.0 cm. Extremely islands. The difference is most likely related to the degree
rare examples of giant bone islands that measure more of phenotypic penetration of the genetic defect. Some
than 2.0 cm in diameter have been described (Figs. 21-10 investigators have suggested that a bone island represents
and 21-11).18,30,49,52,57,60 The characteristic feature is the a unifocal minor form of osteopoikilosis.6,10 Osteopoiki-
presence of so-called thorny spicules or pseudopodia at losis is inherited in an autosomal dominant pattern by
the periphery of the bone island.53,54,56 inactivating mutations involving the LEM domain-
Microscopically, bone islands are composed of compact containing protein 3 gene. LEMD3 interacts with trans-
(cortical) lamellar bone. The thornlike projections repre- forming growth factor TGF-β and bone morphogenetic
sent thickened trabeculae radiating from the main mass protein signaling, causing focal deposit of compact lamel-
and blending with the surrounding bone (Fig. 21-12). lar bone.7 On radiographs, osteopoikilosis presents as
The lesions show prominent haversian canals that may multiple bone islands clustered at the ends of the
contain osteoblasts and osteoclasts within Howship’s metaphyses.70–72 It is rare, with only about 400 cases
lacunae. Features of active bone deposition and remodel- described in the literature. Osteopoikilosis can be associ-
ing are more often seen in larger lesions (Fig. 21-13). ated with the hereditary dermatologic condition known
This explains the increased amount of bone turnover in as dermatofibrosis lenticularis disseminata (Buschke-Ollendorff
bone islands, which can be demonstrated by skeletal scin- syndrome), which is characterized by multiple papular
tigraphy.48,50,58 In fact, some bone islands may slowly fibromas on the back, arms, and thighs.5,71
increase in size (growing bone islands).48,50,55,59 Text continued on p. 1333

ERRNVPHGLFRVRUJ
1322 21  Sclerosing Bone Lesions

A
FIGURE 21-3  ■  Sinoorbital osteoma: radiographic and microscopic features. A, Large, sessile, radiodense mass within frontal sinus.
B, Computed tomogram of frontal sinus osteoma.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1323

B
FIGURE 21-4  ■  Sinoorbital osteoma: radiographic features. A, Plain radiograph of large, solid orbital mass composed of compact
bone. B, Computed tomogram of mass in A.

ERRNVPHGLFRVRUJ
1324 21  Sclerosing Bone Lesions

A B

C D
FIGURE 21-5  ■  Sinoorbital osteoma: mircroscopic features. A, Low power photomicrograph shows dense compact bone of osteoma.
B, Higher magnification of A shows compact bone with prominent haversion systems and focal resorption activity. C, Low power
photomicrograph shows compact bone of osteoma. D, Higher magnification of C showing compact sclerotic bone with prominent
haversian systems and accentuated ossification lines. Note irregularity of haversian systems due to resorptive activity. (A and C,
×20; B and D, ×50) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1325

B
FIGURE 21-6  ■  Sinoorbital osteomas with osteoblastoma-like areas: radiographic and microscopic features. A, Low power photomi-
crograph shows compact bone with less dense areas showing bone remodeling that resembles osteoblastoma. B, Higher magnifica-
tion of A corresponding to less dense area shows loose bone trabeculae with osteoblastic lining and osteoclastic activity resembling
those seen in osteoblasoma. Inset, Computed tomogram shows radiodense oral mass composed of compact bone. (A, ×10; B, ×50)
(A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1326 21  Sclerosing Bone Lesions

A B

C D
FIGURE 21-7  ■  Sinoorbital osteoma: microscopic features. A, Low power photomicrograph shows dense compact bone. B, Less
mature pattern of woven bone and osteoblastic rimming that is seen in some osteomas. C and D, Periphery of osteoma shows loose
pattern of woven bone with osteoblastic rimming and cellular fibroblastic stromal tissue representing growing component of the
tumor. (A and B, ×25; C and D, ×50) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1327

FIGURE 21-8  ■  Sinoorbital osteoma with areas of osteoblastoma: radiographic and microscopic features. A, Low power photomicro-
graph of a lesion corresponding to solid component shows compact, mature bone with resorptive activity. B, Osteoblastoma-like
area corresponding to low-signal area of A. Inset (top), Computed tomogram of lesion in maxillary sinus that is composed of solid
compact mass and has low-signal area with patchy mineralization corresponding to conventional osteoma and osteoblastoma com-
ponents, respectively. Inset (bottom), Higher magnification of B showing interconnected bone trabeculae with prominent osteoblasts
and engorged vasculature characteristic of osteoblastoma. The lesion recurred after the initial surgery, and the recurrent lesion
showed features of aggressive osteoblastoma. (A, ×50; B, ×200; Inset [bottom], ×100) (A, B, and Inset [bottom], hematoxylin-eosin.)
ERRNVPHGLFRVRUJ
1328 21  Sclerosing Bone Lesions

FIGURE 21-9  ■  Bone island (enostosis): radiographic features. A, Anteroposterior radiograph of right hip shows radiodense ovoid
intramedullary density in femoral neck. B, Computed tomogram of lesion in A reveals radiodense island of compact bone within
cancellous bone of femoral neck.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1329

A B
FIGURE 21-10  ■  Giant bone island: radiographic features. A, Anteroposterior radiograph shows large radiodense intramedullary mass
with irregular borders. B, Magnetic resonance image of lesion in A demonstrates absence of signal and irregular contour of large
intramedullary bone island.

ERRNVPHGLFRVRUJ
1330 21  Sclerosing Bone Lesions

C
FIGURE 21-11  ■  Giant bone island: radiographic features. A, Bone scintigram shows increased uptake of technetium 99 in large bone
island within medullary cavity of distal femur. B, Anteroposterior radiograph of intramedullary giant bone island of distal femoral
metaphysis; lesion was asymptomatic, incidentally discovered opacity. C, Axial computed tomogram shows ivory-like density and
sharp circumscription.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1331

C
FIGURE 21-12  ■  Bone island: microscopic features. A, Low power photomicrograph showing irregular contour with multiple pseudo-
podia and dense compact bone. B, Higher magnification of the edge of bone island showing long, projecting surface process con-
tinuous with dense bony focus. C, Detail of lamellar bone architecture with haversian canals within bone island. (A, ×5; B, ×20;
C, ×50) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1332 21  Sclerosing Bone Lesions

B
FIGURE 21-13  ■  Giant bone island: microscopic features. A, Low power photomicrograph shows thickened coarse trabeculae of lamel-
lar bone with interstices containing loose fibrous vascular tissue. B, Higher magnification shows lamellar architecture and loose
fibrous tissue stroma in narrow spaces. (A, ×25; B, ×50) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1333

genetic defects, including inactivating mutations of T-cell


immune regulator 1 (TCIRG1); homozygous mutations
in the chloride 7 channel (CLCN7); a defect in the gray-
lethal or osteopetrosis-associated transmembrane protein
gene (GL); and mutations of the receptor activator of
nuclear factor κ-B ligand (RANKL).7,86,88,89 The autoso-
mal dominant subtype, referred to as osteopetrosis type
2, was linked to mutations inactivating one allele of the
chloride 7 channel gene (CLCN7), and mutations involv-
ing carbonic anhydrase II (CA2), T-cell immune regula-
tor 1 (TCIRG1), osteopetrosis-associated transmembrane
protein 1 (OSTM1), and pleckstrin homology domain-
containing family M member 1 (PLEKHM1).7,76,86,88,89 At
least eight subtypes of osteopetrosis, each with unique
clinical presentation and mode of transmission, have been
described in humans.73,74,77 Deficiency of the carbonic
anhydrase II isoenzyme is associated with autosomal
recessive osteopetrosis. The majority of malignant auto-
somal recessive osteopetrosis is caused by mutations in
the TCIRG1 gene, whereas mutations in the gene encod-
ing the CLCN7 chlorine channel pump are responsible
for the majority of the most common autosomal domi-
nant osteopetrosis.75,78,80,83,84,85,86,88,89
The adult form occurs more frequently than the con-
genital form. Under normal conditions, the primary
spongiosa is resorbed by osteoclasts. Focal ineffective
resorption of primary spongiosa results in accumulated
masses of calcified cartilage in the medullary cavity. Con-
genital osteopetrosis is typically fatal and in many
instances results in stillbirth. When a child is born alive,
FIGURE 21-14  ■  Surface osteoma of long bone: radiographic fea- the disease rapidly progresses, and most patients die in
tures. Anteroposterior radiograph of midshaft of fibula of young early infancy after manifesting profound anemia and
adult with juxtacortical osteoma. This rare, benign tumor was other sequelae of bone marrow compromise. Adults with
composed of mature compact bone without cartilage cap and osteopetrosis usually live a long time. Many patients are
was treated by segmented resection.
asymptomatic, and the diagnosis is based on incidental
findings on radiographs made for other reasons or as a
result of pathologic fractures.
The radiologic hallmark of all variants of osteopetrosis
The symmetric distribution of multiple bone islands is generalized opacification of bone (Figs. 21-17 and
in the metaphyseal ends of the long tubular bones and in 21-18). The skeletal sites that are formed by membranous
the carpal or tarsal bones is a characteristic radiographic ossification are usually unaffected. Thus in the skull, scle-
feature (Figs. 21-15 and 21-16). Less frequently the rosis is more prominent at the base. In the long bones,
lesions may also be found in other parts of the skeleton the metaphyseal parts typically show prominent sclerosis.
in the vicinity of joints (e.g., around the acetabulum or In the spine, the sclerosis of the end plates gives the
glenoid fossa). The skull, spine, and ribs are extremely vertebrae a “sandwiched” appearance. The metaphyseal
rare sites of involvement. regions of long bones appear clublike without the normal
The lesions in osteopoikilosis are intramedullary but flare (Erlenmeyer flask deformity). The metaphyses may
may also be attached to the inner surface of the cortex. also show horizontal striations of lucent and sclerotic
Microscopically, they are composed of lamellar bone areas.
identical to that seen in bone islands (Fig. 21-16). Microscopically, the sclerotic areas of bone show an
accumulation of cartilage with an interconnecting
network of bone trabeculae87 (Figs. 21-18 and 21-19).
OSTEOPETROSIS Osteoclasts may be present on bone surfaces, but they do
not show Howship’s lacunae and lack ruffled borders.
Osteopetrosis (Albers-Schönberg disease) occurs in two These features indicate that the basic defect in this dis-
basic clinical settings: as an autosomal recessive condition order involves osteoclastic cells that cannot resorb bone.
with early manifestation and as an autosomal dominant This is consistent with the observation that the metaphy-
condition with delayed manifestation.77,81,82 These two seal region closest to the growth plate is packed with
forms of osteopetrosis are referred to as congenital or thickened trabeculae of woven bone that has a central
malignant osteopetrosis and adult osteopetrosis or osteopetro- core of calcified cartilage (i.e., lack of resorption of
sis tarda. The autosomal recessive subtype, also referred primary spongiosa). This concept is further supported by
to as osteopetrosis type 1, has been linked to several Text continued on p. 1339

ERRNVPHGLFRVRUJ
1334 21  Sclerosing Bone Lesions

B
FIGURE 21-15  ■  Osteopoikilosis: radiographic features. A, Incidentally discovered speckled, pea-sized densities within cancellous bone
of proximal femur, pelvis, and sacrum. B, Distal tibiae and fibulae showing multiple small islands of compact bone.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1335

A B

FIGURE 21-16  ■  Osteopoikilosis: radiographic and histologic features. A, Plain radiograph showing punctate radiodensities in pha-
langes. B, Anteroposterior radiograph of left hip of a 21-year-old man in whom speckled radiodensities were incidentally discovered
in pelvis, sacrum, and proximal femur. C, Whole-mount photomicrograph of femoral head showing multiple small islands of compact
bone within cancellous bone (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1336 21  Sclerosing Bone Lesions

B C D

FIGURE 21-17  ■  Osteopetrosis (Albers-Schönberg disease): radiographic features. A, Anteroposterior radiograph of pelvis and hips
of child with marble bone disease. Note diffuse ivory-like density of bones, including epiphyses. B, Erlenmeyer flask deformities
seen in distal femoral shaft. Note ill-defined delineation of cortical and cancellous bone borders. C, Distal ends of tibia and fibula
showing increased density adjacent to growth plates and horizontal striations in distal tibial shaft. D, Lateral radiograph of lumbar
spine showing ivory density in upper and lower borders of each vertebral body.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1337

B C
FIGURE 21-18  ■  Osteopetrosis: radiographic and microscopic features. A, Lateral radiograph of skull shows thickening of base with
ivory density of bone preformed in cartilage. B, “Bone-within-a-bone” appearance of first metatarsal with diffuse osteosclerosis.
C, Low-power photomicrograph of osteopetrotic bone shows persistence of calcified cartilage cores in cancellous bone trabeculae
(primary spongiosa) as well as absence of normal marrow spaces. (C, ×100) (C, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
1338 21  Sclerosing Bone Lesions

A B

C D
FIGURE 21-19  ■  Osteopetrosis: microscopic features. A-D, Lower and corresponding intermediate power microphotographs show
persistence of prominent calcified cartilage cores in bone trabeculae of primary spongiosa and the fibrosis of marrow spaces with
the absence of hematopoetic element. (A and C, ×100; B and D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1339

A B
FIGURE 21-20  ■  Melorheostosis: radiographic features. A and B, Anteroposterior and lateral radiographs of leg show cortical thicken-
ing of fibular diaphysis resembling candle dripping, characteristic of melorheostosis.

the significant normalization of skeletal density and the sclerotomes.101 It can also be associated with other lesions
reversal of bone opacification that can be induced by such as arteriovenous aneurysms and scleroderma.99,100,103
bone marrow transplantation in patients with congenital Microscopically, these lesions represent hyperostosis of
osteopetrosis.79,89 compact cortical bone similar to that seen in osteomas,
bone islands, and osteopoikilosis (Fig. 21-24). Features
of immaturity (absence of well-organized osteons and a
MELORHEOSTOSIS woven bone appearance) can be present. Melorheostosis
typically involves the surface of bone but in rare cases can
Melorheostosis is a rare condition characterized by a also involve the medullary cavity. It is believed that
localized, diffuse thickening of the cortical bone (Figs. hyperostosis tends to progress through childhood into
21-20 through 21-23). The lesions frequently are adult life. It is characterized by periods of exacerbation
described radiographically as resembling wax dripping and arrest.95,98 Overlying soft tissues and nearby joints
down the side of a candle (guttering).90,92,93,96,102 The may be involved with fibrous thickening and contrac-
condition may involve one bone (monostotic form) or tures. Radioisotopic bone scans are usually positive in
may affect multiple bones in one extremity (monomelic melorheostosis and show increased uptake at the sites of
form). Rarely, it presents as a disseminated skeletal dis- lesions. Mineralized soft tissue masses may occur in as
order that involves more than one limb (polyostotic many as 27% of patients with melorheostosis.101 These
polymelic form) (Fig. 21-23).91 The most prominent may be found adjacent to areas of cortical hyperostosis
hyperostosis is usually at the site developmentally con- or at a distance, usually in paraarticular location.97 His-
sidered to be formed by membranous ossification (shafts tologically, these bony and cartilaginous soft tissue masses
in the long tubular bones). In the polyostotic involvement often contain a core of mature osseous tissue with a
of one limb, the distribution of lesions in melorheostosis peripheral cap of hyaline cartilage.94
seems to correlate with the anatomic distribution of Text continued on p. 1344

ERRNVPHGLFRVRUJ
1340 21  Sclerosing Bone Lesions

FIGURE 21-21  ■  Melorheostosis: radiographic features. Anteroposterior radiograph of tibia shows thickening of medial cortex in upper
diaphysis. Lucent defect represents biopsy site.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1341

B
FIGURE 21-22  ■  Melorheostosis: radiographic features. A and B, Lateral radiographs of calcaneus of a 46-year-old woman show
sclerotic changes within bone and on dorsal surface.

ERRNVPHGLFRVRUJ
1342 21  Sclerosing Bone Lesions

A B

C D
FIGURE 21-23  ■  Melorheostosis, polyostotic (polymelic form): radiographic features. A and B, Anteroposterior radiographs of shoul-
der and elbow of a 35-year-old woman show surface thickening of cortex of humerus and soft tissue bony densities. C, Technetium
99 radioisotope scan shows markedly increased uptake in shoulder and humeral shaft. D, Radiograph of wrist and hand of patient
in A and C. Note sclerotic lesions of second metacarpal and carpal bones and phalanx.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1343

B C
FIGURE 21-24  ■  Melorheostosis: microscopic features. A, Low power photomicrograph of cortex and intramedullary dense bone
in calcaneus shown in Figure 21-22. Note that cortex (right) is thickened, and intramedullary sclerotic bone (left) is immature.
B and C, Intermediate and high power photomicrographs of intramedullary dense immature bone. (A, ×50; B and C, ×100.)
(A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1344 21  Sclerosing Bone Lesions

REFERENCES 28. Noren GD, Roche WC: Huge osteoma of the mandible: report
of case. J Oral Surg 36:375–379, 1978.
Osteoma and Bone Islands 29. Perniciaro C: Gardner’s syndrome. Dermatol Clin 13:51–56,
1. Beighton P, Cremin BJ: Sclerosing bone dysplasias, Berlin, 1980, 1995.
Springer. 30. Smith J: Giant bone islands. Radiology 107:35–36, 1973.
2. Boffano P, Roccia F, Campisi P, et al: Review of 43 osteomas of 31. Takeuchi T, Takenoshita Y, Kubo K, et al: Natural course of jaw
the craniomaxillofacial region. J Oral Maxillofac Surg 70:1093– lesions in patients with familial adenomatosis coli (Gardner’s syn-
1095, 2012. drome). Int J Oral Maxillofac Surg 22:226–230, 1993.
3. De Vernejoul MC, Kornak U: Heritable sclerosing bone disor- 32. Traboulsi EI, Maumenee IH, Krush AJ, et al: Congenital hyper-
ders: presentation and new molecular mechanisms. Ann N Y Acad trophy of the retinal pigment epithelium predicts colorectal
Sci 1192:269–277, 2010. polyposis in Gardner’s syndrome. Arch Ophthalmol 108:525–526,
4. Gondak RO, Mariano FV, Vargas PA, et al: Bilateral osteoma of 1990.
the maxillary sinus or anatomic variation? J Craniofac Surg 33. Traboulsi EI, Murphy SF, de la Cruz ZC, et al: A clinicopatho-
25:1133–1134, 2014. logic study of the eyes in familial adenomatous polyposis with
5. Greenspan A: Sclerosing bone dysplasias: a target-site approach. extracolonic manifestations (Gardner’s syndrome). Am J Ophthal-
Skeletal Radiol 20:561–583, 1991. mol 110:550–561, 1990.
6. Greenspan A, Steiner G, Sotelo D, et al: Mixed sclerosing bone 34. Wehrli BM, Weiss SW, Yandow S, et al: Gardner-Associated
dysplasia coexisting with dysplasia epiphysealis hemimelica fibromas (GAF) in young patients. Am J Surg Pathol 25:645–651,
(Trevor-Fairbank disease). Skeletal Radiol 15:452–454, 1986. 2001.
7. Ihde LL, Forrester DM, Gottsegen CJ, et al: Sclerosing bone
dysplasia: review and differentiation from other causes of osteo- Sino-orbital Osteoma
sclerosis. Radiographics 31:1865–1882, 2011. 33. Bullough PG: Ivory exostosis of the skull. Postgrad Med 41:277–
8. Kaufmann HJ: Classification of the skeletal dysplasias and the 281, 1965.
radiologic approach to their differentiation. Clin Orthop 114:12– 34. Childrey JH: Osteoma of sinuses, the frontal and the sphenoid
17, 1976. bone: report of 15 cases. Arch Otolaryngol 30:63–72, 1939.
9. Muderris T, Sevil E, Bercin S, et al: Giant paranasal sinus osteo- 35. Gawlik G, Turowski K, Trojanowski T: Giant osteoma of the
mas: surgical treatment options. J Craniofac Surg 25:1287–1291, skull. Neurol Neurochir Pol 14:453–455, 1980.
2014. 36. Hallberg OE, Begley JW, Jr: Origin and treatment of osteomas
10. Norman A, Greenspan A: Sclerosing dysplasias of bone. In of the paranasal sinuses. Arch Otolaryngol 51:750–760, 1950.
Taveras JM, Ferrucci JT, editors: Radiology: diagnosis, imaging, 37. Hudolin V, Riessner D, Kadrnka S, et al: A huge osteoma in the
intervention, vol 5, Philadelphia, 1986, JB Lippincott. anterior cranial fossa. J Neurol Neurosurg Psychiatry 24:80–83,
11. Tippu SR, Rahman F, Sharma N, et al: Collision tumor of the 1961.
palate: a rare case report. Contemp Clin Dent 5:102–105, 2014. 38. Lehmer LM, Kissel P, Ragsdale BD: Frontal sinus osteoma with
12. Wei LA, Ramey NA, Durairaj VD, et al: Orbital osteoma: clinical osteoblastoma-like histology and associated intracranial pneuma-
features and management options. Ophthal Plast Reconstr Surg tocele. Head Neck Pathol 6:384–388, 2012.
30:168–174, 2014. 39. Liston SL, Barker BF, Cocayne DR: Multiple osteomas of the
maxillary sinus. J Oral Surg 37:113–114, 1979.
Ivory Exostosis 40. McHugh JB, Mukherji SK, Lucas DR: Sino-orbital osteoma: a
13. Bulow S, Sondergaard JO, Witt I, et al: Mandibular osteomas in clinicopathologic study of 45 surgically treated cases with empha-
familial polyposis coli. Dis Colon Rectum 27:105–108, 1984. sis on tumors with osteoblastoma-like features. Arch Pathol Lab
14. Chang CH, Piatt ED, Thomas KE, et al: Bone abnormalities in Med 133:1587–1593, 2009.
Gardner’s syndrome. Am J Roentgenol Radium Ther Nucl Med 41. Mehta BS, Grewal GS: Osteoma of the paranasal sinuses along
103:645–652, 1968. with a case report of an orbitoethmoidal osteoma. J Laryngol
15. Coffin CM: Gardner fibroma. In Fletcher CDM, Bridge JA, 77:601–610, 1963.
Hogendoorn PCW, Mertens F, editors: WHO classification of 42. Mikaelian DO, Lewis WJ, Behringer WH: Primary osteoma of
tumours of soft tissue and bone, Lyon, 2013, p 68. the sphenoid sinus. Laryngoscope 86:728–733, 1976.
16. Davies DR, Armstrong JG, Thakker N, et al: Severe Gardner 43. Miller NR, Gray J, Snip R: Giant, mushroom-shaped osteoma of
syndrome in families with mutations restricted to a specific region the orbit originating from the maxillary sinus. Am J Ophthalmol
of the APC gene. Am J Hum Genet 57:1151–1158, 1995. 83:587–591, 1977.
17. Gardner EJ, Richards RE: Multiple cutaneous and subcutaneous 44. Mirra JM, Gold RH, Pignatti G, et al: Case report 497: compact
lesions occurring simultaneously with hereditary polyposis and osteoma of iliac bone. Skeletal Radiol 17:437–442, 1988.
osteomatosis. Am J Hum Genet 5:139–147, 1953. 45. Montgomery WW: Osteoma of the frontal sinus. Ann Otol Rhinol
18. Gold RH, Mirra JM, Remotti F, et al: Case report 527: giant bone Laryngol 69:245–255, 1960.
island of tibia. Skeletal Radiol 18:129–132, 1989. 46. Samy LL, Mostafa H: Osteomata of the nose and paranasal sinuses
19. Goldblum JR, Folpe AL, Weiss SW: Soft tissue tumors, ed 5, Phila- with a report 21 cases. J Laryngol Otol 85:449–469, 1971.
delphia, 2014, Elsevier. 47. Yazici Z, Yazici B, Yalcinkaya U, et al: Sino-orbital osteoma with
20. Graham MD: Osteomas and exostoses of the external auditory o steoblastoma-like features: case reports. Neuroradiology 54:765–
canal: a clinical, histopathological and scanning electron micro- 769, 2012.
scopic study. Ann Otol Rhinol Laryngol 88:566–572, 1979.
21. Green AE, Bowerman JE: An osteoma of the mandible. Br J Oral Bone Island (Enostosis)
Surg 12:225–228, 1974. 48. Blank N, Lieber A: The significance of growing bone islands.
22. Iwama T, Mishima Y, Okamoto N, et al: Association of congenital Radiology 85:508–511, 1965.
hypertrophy of the retinal pigment epithelium with familial ade- 49. Brien EW, Mirra JM, Latanza L, et al: Giant bone island of femur.
nomatous polyposis. Br J Surg 77:273–276, 1990. Case report, literature review, and its distinction from low grade
23. Jones K, Korzcak P: The diagnostic significance and management osteosarcoma. Skeletal Radiol 24:546–550, 1995.
of Gardner’s syndrome. Br J Oral Maxillofac Surg 28:80–84, 1990. 50. Davies JA, Hall FM, Goldberg RP, et al: Positive bone scan in a
24. Katou F, Motegi K, Baba S: Mandibular lesions in patients with bone island. J Bone Joint Surg 61A:943–945, 1979.
adenomatosis coli. J Craniomaxillofacial Surg 17:354–358, 1989. 51. Gerges M, Glard Y, Luanay F, et al: Small bone islands: unusual
25. Kubo K, Miyatani H, Takenoshita Y, et al: Widespread radiopac- clinical symptomatology. Orthopedics 37:79–82, 2014.
ity of jaw bones in familial adenomatosis coli. J Craniomaxillofacial 52. Greenspan A, Klein MJ: Giant bone island. Skeletal Radiol 35:67–
Surg 17:350–353, 1989. 69, 1996.
26. Lasser DM, DeVivo DC, Garvin J, et al: Turcot’s syndrome: evi- 53. Greenspan A, Steiner G, Knutzon R: Bone island (enostosis):
dence for linkage to the adenomatous polyposis coli (APC) locus. clinical significance and radiologic and pathologic correlations.
Neurology 44:1083–1086, 1994. Skeletal Radiol 20:85–90, 1991.
27. Michal M, Fetsch JF, Hess O, et al: Nuchal type fibroma: a clin- 54. Kim SK, Barry WF, Jr: Bone islands. Radiology 90:77–78, 1968.
copathologic study of 52 cases. Cancer 85:1563, 1999. 55. Ngan H: Growing bone islands. Clin Radiol 23:199–201, 1972.

ERRNVPHGLFRVRUJ
21  Sclerosing Bone Lesions 1345

56. Onitsuka H: Roentgenologic aspects of bone islands. Radiology of human autosomal recessive osteopetrosis. Nat Genet 25:343–
123:607–612, 1977. 346, 2000.
57. Park HS, Kim JR, Lee SY, et al: Symptomatic giant (10-cm) bone 81. Horton WA, Schimke RN, Iyama T: Osteopetrosis: further het-
island of the tibia. Skeletal Radiol 34:347–350, 2005. erogeneity. J Pediatr 97:580–585, 1980.
58. Sickles EA, Genant HK, Hoffer PB: Increased localization of 82. Kahler SG, Burns JA, Aylsworth AS: A mild auto-recessive form
99mTc-pyrophosphate in a bone island: case report. J Nucl Med of osteopetrosis. Am J Med Genet 17:451–464, 1984.
17:113–115, 1976. 83. Kornak U, Kasper D, Bosl MR, et al: Loss of the CIC7 chloride
59. Simon K, Mulligan ME: Growing bone islands revisited: a case channel leads to osteopetrosis in mice and man. Cell 104:205–215,
report. J Bone Joint Surg 67A:809–811, 1985. 2001.
60. Trombetti A, Noel E: Giant bone islands: a case with 31 years of 84. Letizia C, Taranta A, Migliaccio S, et al: Type II benign osteope-
follow-up. Joint Bone Spine 69:81–84, 2002. trosis (Albers-Schönberg disease) caused by a novel mutation in
CLCN7 presenting with unusual clinical manifestations. Calcif
Surface Osteoma of Long Bones Tissue Int 74:42–46, 2004.
61. Bertoni F, Unni KK, Beabout JW, et al: Parosteal osteoma of 85. Michigami T, Kageyama T, Satomura K, et al: Novel mutations
bones other than of the skull and face. Cancer 75:2466–2473, 1995. in the α3 subunit of vacuolar H+-adenosine triphosphatase in
62. Geschickter CF, Copeland MM: Parosteal osteoma of bone: a new a Japanese patient with infantile malignant osteopetrosis. Bone
entity. Ann Surg 133:790–807, 1951. 30:436–439, 2002.
63. Lambiase RE, Levine SM, Terek RM, et al: Long bone surface 86. Michou L, Brown JP: Genetics of bone diseases: Paget’s disease,
osteomas: imaging features that may help avoid unnecessary biop- fibrous dysplasia, osteopetrosis, and osteogenesis imperfecta. Joint
sies. AJR Am J Roentgenol 171:775–778, 1998. Bone Spine Rev Rhum 78:252–258, 2011.
64. Meltzer CC, Scott WW, Jr, McCarthy EF: Case report 698: 87. Shapiro F, Glimcher MJ, Holtrop ME, et al: Human osteopetro-
osteoma of the clavicle. Skeletal Radiol 20:555–557, 1991. sis: a histological, ultrastructural, and biochemical study. J Bone
65. O’Connell JX, Rosenthal DI, Mankin JH, et al: Solitary osteoma Joint Surg 62A:384–399, 1980.
of a long bone: a case report. J Bone Joint Surg 75A:1830–1834, 88. Sobacchi C, Schulz A, Coxon FP, et al: Osteopetrosis: genetics,
1993. treatment and new insights into osteoclast function. Nature Rev
66. Soler RR, Martinez S, de Marcos JA, et al: Parosteal osteoma of Endocrinol 9:522–536, 2013.
the iliac bone. Skeletal Radiol 27:161–163, 1993. 89. Stark Z, Savarirayan R: Osteopetrosis. Orphanet J Rare Dis 4:5,
67. Sundaram M, Falbo S, McDonald D, et al: Surface osteomas of 2009.
the appendicular skeleton. AJR Am J Roentgenol 167:1529–1533,
1996. Melorheostosis
68. Tamarit LV, Pardo J: Parosteal osteoma: a clinicopathologic 90. Campbell CJ, Papademetriou T, Bonfiglio M: Melorheostosis:
approach. Pathol Annu 12:373–387, 1977. a report of the clinical, roentgenographic and pathological find-
69. Yun SJ, Jin W, Park YK, et al: Simultaneously detected parosteal ings in 14 cases. J Bone Joint Surg 50A:1281–1304, 1968.
osteoma and osteochondroma in the distal femur of a single 91. Elkeles A: Melorheostosis: report of a case affecting all extremi-
patients. Clin Imaging 37:950–953, 2013. ties. Indian J Radiol Souvenir 1–7:1956.
92. Gagliardi GG, Mahan KT: Melorheostosis: a literature review and
Osteopoikolosis (Spotted Bone Disease) case report with surgical considerations. J Foot Ankle Surg 49:80–
70. Di Primio G: Benign spotted bones: a diagnostic dilemma. CMAJ 85, 2010.
Can Med Assoc J 183:456–459, 2011. 93. Gold RH, Mirra JM: Case report 35: melorheostose. Skeletal
71. Green AE, Ellswood WH, Collins JR: Melorheostosis and osteo- Radiol 2:57–58, 1977.
poikilosis with a review of the literature. AJR Am J Roentgenol 94. Goldman AB, Schneider R, Huvos AS, et al: Case Report 778:
87:1096–1111, 1962. Melorheostosis presenting as two soft-tissue masses with osseous
72. Lagier R, Mbakop A, Bigler A: Osteopoikilosis: a radiological and changes limited to the axial skeleton. Skeletal Radiol 22:206–210,
pathological study. Skeletal Radiol 11:161–168, 1984. 1993.
95. Hove E, Sury B: Melorheostosis: report on five cases with
Osteopetrosis follow-up. Acta Orthop Scand 42:315–319, 1971.
73. Andersen PE, Jr, Bollerslev J: Heterogeneity of autosomal domi- 96. Jain VK, Arya RK, Bharadwaj M, et al: Melorheostosis: clinico-
nant osteopetrosis. Radiology 164:223–225, 1987. pathological features, diagnosis, and management. Orthopedics 32:
74. Beighton P, Hamersma H, Cremin BJ: Osteopetrosis in South 512, 2009.
Africa: the benign, lethal and intermediate forms. S Afr Med J 97. Judkiewicz AM, Murphey M, Resnik CS, et al: Advanced imaging
55:659–665, 1979. of melorheostosis with emphasis on MRI. Skeletal Radiol 30:447–
75. Benichou O, Cleiren E, Gram J, et al: Mapping of autosomal 453, 2001.
dominant osteopetrosis type II (Albers-Schönberg disease) to 98. Morris JM, Samilson RL, Corey CL: Melorheostosis: review of
chromosome 16p13.3. Am J Hum Genetics 63:647–654, 2001. the literature and report of an interesting case with 19-year
76. Bollerslev J, Henriksen K, Nielsen MF, et al: Autosomal dominant follow-up. J Bone Joint Surg 45A:1191–1206, 1963.
osteopetrosis revisited: lessons from recent studies. Eur J Endocri- 99. Muller S, Henderson ED: Melorheostosis with linear sclero-
nol 169:39–57, 2013. derma. Arch Dermatol 88:142, 1963.
77. Brown DM, Dent PB: Pathogenesis and osteopetrosis: a compari- 100. Murray RO: Melorheostosis associated with congenital arteriove-
son of human and animal spectra. Pediatr Res 5:181, 1971. nous aneurysms. Proc R Soc Med 44:473, 1950.
78. Cleiren E, Benichou O, Van Hul E, et al: Albers-Schönberg 101. Murray RO, McCredie J: Melorheostosis and the sclerotomes: a
disease (autosomal dominant osteopetrosis, type II) results from radiological correlation. Skeletal Radiol 4:57–71, 1979.
mutations in the CLCN7 chloride channel gene. Hum Mol Genet 102. Suresh S, Muthukumar T, Saifuddin A: Classical and unusual
10:2861–2867, 2001. imaging appearances of melorheostosis. Clin Radiol 65:593–600,
79. Coccia PF, Krivit W, Cervenka J, et al: Successful bone-marrow 2010.
transplantation for infantile malignant osteopetrosis. N Engl J 103. Thompson NM, Allen CEL, Andrews GS, et al: Scleroderma and
Med 302:701–708, 1980. melorheostosis: report of a case. J Bone Joint Surg Br 33B:430,
80. Frattini A, Orchard PJ, Sobacchi C, et al: Defects in TCIRG1 1951.
subunit of the vacuolar proton pump are responsible for a subset

ERRNVPHGLFRVRUJ
C H A P T E R 2 2 

Tumor-Induced Osteomalacia
and Rickets

Modern understanding of rachitic syndromes, originally The typical presentation is a gradual onset of bone pain
referred to as vitamin D-resistant rickets, is based on the in weight bearing areas such as the legs, ankles, hips, and
identification of a novel phosphate-regulating homeo- back. Some patients have pathologic fractures. The pain
static system and its underlying diverse genetic back- is frequently accompanied by generalized muscular weak-
ground.7,10 Essential to this understanding were discoveries ness, leading to a severely debilitated state in which the
of two mutated genes involved in the development of patient is bedridden. Hence, in some patients an initial
somewhat overlapping yet distinct phosphaturic disor- misdiagnosis of a primary muscular disorder is made.
ders. The first one involved the identification of phos- Symptoms are usually present anywhere from 3 months
phate regulating endopeptidase homolog on chromosome to 17 years before diagnosis. In some cases, a mass was
X (PHEX) as the mutated gene in X-linked phosphate- noted up to 20 years before the onset of generalized
mia.56 The second was the discovery that a mutant fibro- symptoms. Occasionally an asymptomatic bone tumor is
blast growth factor 23 (FGF23) plays a role in the identified in radiographs in a patient with a diagnosis of
development of tumor induced osteomalacia and rickets vitamin-D resistant rickets.40 Children may initially have
as well as an autosomal dominant X-linked variant of malaise, gait disturbance, swollen joints, and genu valgum
hypophosphatemic rickets. A more complete list of phos- or varum.
phatemic disorders with their genetic background and Characteristic metabolic abnormalities include a low
biochemistry is provided in Table 22-1.7 Their detailed serum phosphorus level (2.2 to 0.33 mg/dL), normal or
description is beyond the scope of this book, and this slightly low serum calcium level, and variably elevated
chapter focuses primarily on tumor-induced osteomalacia alkaline phosphatase activity (occasionally up to 1700 to
and rickets. 2300 IU). The urinary phosphate level is usually ele-
The cause-and-effect relationship between certain vated, and most cases exhibit decreased tubular reabsorp-
mesenchymal tumors and severe osteomalacia or rickets tion of phosphate (23% to 70%). Urinary calcium levels
was first recognized in 1959 by Prader et al.54 The origi- tend to be low. Serum levels of 1,25-dihydroxyvitamin
nal description of this peculiar paraneoplastic syndrome D3 are low or detectable, whereas serum levels of
was provided in 1947 by McCance,41 who reported a case 25-hydroxyvitamin D3 are generally normal. In addition,
of resolution of vitamin D-resistant rickets after removal aminoaciduria and glycosuria with normal serum glucose
of “degenerated osteoid tissue.” levels may be present.

Radiographic Imaging
TUMOR-INDUCED OSTEOMALACIA
In skeletally immature patients, radiographs show wid­
Tumor-induced osteomalacia (oncogenic osteomalacia) is ening of growth plates and radiolucent zones in the
a clinicopathologic entity in which vitamin D-resistant metaphyses consistent with rachitic changes (Fig. 22-1).
osteomalacia or rickets occurs in association with a bone Lower-extremity long bones may show bowing deformi-
or soft tissue tumor.1,64,72 These tumors have exhibited a ties. In skeletally mature patients, radiographic features
wide spectrum of histologic features that have only consist of generalized osteopenia with Milkman’s pseu-
recently been gathered under a unifying concept of phos- dofractures or Looser’s lines, characteristic of osteomala-
phaturic mesenchymal tumors.20,82,83 Overlapping clinical cia. In addition, skeletal radiographs may reveal the
presentation and biochemical abnormalities can be presence of an asymptomatic or symptomatic tumor.11
present in X-linked hypophosphatemia and autosomal Pathologic fractures in adults, particularly of the femoral
dominant hyphosphatemia.7,37 A similar syndrome has neck, are relatively common.
also been reported in association with the linear seba-
ceous nevus syndrome and fibrous dysplasia.2,15,25,29 Pathologic Findings
Histomorphometry on undecalcified bone sections shows
Clinical Data
hyperosteoidosis with increased osteoid volume and
The majority of patients with tumor-induced osteo­ increased width of osteoid seams. In approximately 50%
malacia are adults; two thirds are age 30 years or of cases, the tumor is primary in bone.49 Half of the cases
older.36,45,55,58,63,74,89 The ages of reported patients range involving bone occurred in the long bones of the lower
from 7 to 73 years, and the male-to-female ratio is 1.2 : 1. extremities, particularly in the femur and tibia. The
1346
ERRNVPHGLFRVRUJ
22  Tumor-Induced Osteomalacia and Rickets 1347

TABLE 22-1 Biochemistry and Genetics of Hypophosphatemic Disorders


Phosphate Vitamin D
Calcium Metabolism Metabolism Metabolism Mutation
SERUM URINE SERUM GI CALCIUM SERUM SERUM
CALCIUM CALCIUM PTH ABSORPTION PHOSPHATE TMP/GFR 1,25(OH)2D GENE

FGF23-mediated
  XLH N ↓ N, ↑ ↓ ↓ ↓ (↓) PHEX
  ADHR N ↓ N ↓ ↓ ↓ (↓) FGF23
  ARHR1 N N, ↓ N, ↑ ? ↓ ↓ (↓) DMP1
  TIO N N, ↓ N, ↓, ↑ ↓ ↓ ↓ ↓ FGF23
  ARHR2 N N, ↓ N ? ↓ ↓ (↓) ENPP1
  McCune-Albright N, ↑ N N, ↑ ? N, ↓ N, ↓ N, ↓ GNAS1
  ENS N, ↑ N, ↓ N, ↑ ? N, ↓ N, ↓ (↓) ? FGFR3*
  NF N, ↓ N, ↓ N ? N, ↓ N, ↓ N, (↓) NF1
  OGD N N N ? ↓ ↓ (↓) FGFR1
  HRHPT N, ↑ N, ↑ ↑ ? ↓ ↓ (↓) KL (klotho)
Non-FGF23-mediated
  XLRH N ↑ N, ↓, ↑ N, ↑ ↓ ↓ ↑ CLCN5
  HHRH N, ↑ ↑ N, ↓ ↑ ↓ ↓ ↑ SLC34A3
  Fanconi N, ↓, ↑ N, ↑ N ? N, ↓ N, ↓ N, ↓ Various (SLC34A1)
  NHERF1 N ? (stones) N ? ↓ ↓ N, ↑ NHERF1

Reprinted with permission from Carpenter TO, et al: J Bone Miner Metab 30:1–9, 2012.
ADHR, autosomal dominant hypophosphatemic rickets; ARHR, autosomal recessive hypophosphatemic rickets; ENS, epidermal
nevus syndrome; HHRH, hereditary hypophosphatemic rickets with hypercalciuria; HRHPT, hypophosphatemic rickets with
hyperparathyroidism; NF neurofibromatosis; N, normal; NHERF, Na+/H+ exchanger regulatory factor; OGD, osteoglophonic dysplasia;,
TIO, tumor-induced osteomalacia; XLH, X-linked hypophosphatemia; XLRH, X-linked recessive hypophosphatemia (Dent’s disease);
↓, decreased; (↓), decreased relative to the serum phosphorus concentration; ↑, increased; ?, not well documented.
*Mosaic FGF23 mutations account for some cases of ENS; however, in reported cases due to this mutation hypophosphatemia is not
reported.

second most common location of osseous tumors is in the histologically from similar lesions not associated with this
craniofacial bones.31,47 Soft tissue tumors comprise the syndrome (see Fig. 22-2).
other half of all cases and follow the same pattern of It was recognized by several authors that many of these
anatomic distribution as skeletal tumors—they occur in tumors show a distinctive morphology and have micro-
the soft tissues of the lower extremities and craniofacial scopic features in common.8,18,50,63 In 1987, Weidner and
region. Santa Cruz first introduced the term phosphaturic mesen-
Histologically, 40% of reported cases are vascular chymal tumor, mixed connective tissue variant, proposing
tumors (Table 22-2). Of these, hemangiopericytoma that these tumors of wide-ranging histologic patterns had
is the most common, representing about half of the vas- common features of spindle cells, prominent blood
cular tumors and approximately 20% of the total (Fig. vessels, osteoclast-like giant cells, cystic spaces, metaplas-
22-2).6,24,26,42,60,65,77,83,84,89 Primary hemangiopericytoma of tic bone, and cartilage-like matrix, which indicated that
bone is extremely rare and comprises 0.08% of all primary they represent a distinct clinicopathologic entity.83
tumors of bone.79 Other vascular tumors include heman- A collaborative multiinstitutional study of 32 examples
gioma, angiofibroma, low-grade hemangioendothelioma, of phosphaturic mesenchymal tumors in which reverse
and very rarely, high-grade angiosarcoma.1,13,39,43,63,81,89 transcription polymerase chain reaction (RT-PCR) dem-
The second group of tumors associated with rickets onstration of FGF23 gene expression in two cases and
or osteomalacia can be histologically classified as having immunohistochemical demonstration of FGF23 protein
features of fibrohistiocytic and giant cell lesions exhibit- expression in 17 cases has strengthened the concept of a
ing prominent vasculature (Fig. 22-3). The specific recognizable clinicopathologic entity with a demonstra-
lesions of this group are nonossifying fibromas and giant ble phosphaturic factor in the tumor cells. These authors
cell reparative granuloma, giant cell tumor, and pig- pointed out that wider recognition of the histologic fea-
mented villonodular synovitis.3 A small proportion of tures of phosphaturic mesenchymal tumors could occa-
reported tumors represent malignant lesions, which sionally identify patients with previously undiagnosed
include osteosarcoma and mesenchymal chondrosar- osteomalacia.22
coma.28,46,48,75,76,87 Some of the tumors defy clear-cut clas-
sifications and are descriptively designated as fibrovascular Treatment and Behavior
or mesenchymal. The majority of tumors are relatively
small—within the range of 1 to 4 cm—but occasionally Conventional treatment for rickets or osteomalacia con-
measure 7 cm or larger. Rarely malignant tumors such as sisting of increasing doses of vitamin D or phosphate
osteosarcoma are associated with rickets or osteomalacia provides only transient improvement in approximately
(Fig. 22-4). 15% of patients with oncogenic osteomalacia.3,4,12,51,52
Ultrastructurally or immunohistochemically, the Removal of the tumor is required and results in complete
tumors inducing rickets or osteomalacia do not differ cure of osteomalacia or rickets in approximately 80% of

ERRNVPHGLFRVRUJ
1348 22  Tumor-Induced Osteomalacia and Rickets

B
FIGURE 22-1  ■  Oncogenic rickets: radiographic features. A, Radiograph of knee of same case in A showing radiolucent zones on
metaphyseal sides of growth plates and circumscribed eccentric lesion of distal femur diagnosed as hemangiopericytoma. B, Radio-
graph of wrist with radiolucent zones on metaphyseal sides of the growth plates and flaring of the metaphyses characteristic of
rachitic changes.

ERRNVPHGLFRVRUJ
22  Tumor-Induced Osteomalacia and Rickets 1349

sarcomatosis.38,59,83 Another death occurred in the case


TABLE 22-2 Types of Tumors Producing
of nasal hemangiopericytoma and was attributed to per-
Osteomalacia and Rickets
sistent bleeding.9 A reported complication of medical
Histologic Type No. of Cases therapy with calcitriol and phosphates is the development
of parathyroid autonomy (tertiary hyperparathyroidism).
Vascular tumors 31
  Hemangiopericytomas 14
  Hemangiomas 7 Pathogenesis
  “Fibrovascular” lesions 6
  Angiosarcomas 2 A wide variety of causes may give rise to osteomalacia or
  Hemangioendothelioma 1
  Angiolipoma 1
rickets, and their description is beyond the scope of this
Giant cell lesions 9 book. In developed countries, however, given that inad-
  GCT of bone 2 equate nutrition is infrequent, some of the rare causes are
  Soft tissue GCT 2 X-linked hypophosphatemia and autosomal dominant
  Other 5 hypophosphatemia.7,10,17 Oncogenic osteomalacia pres-
  Malignant GCT
  PVNS ents with analogous symptoms and chemical abnormali-
  GCRG ties requiring large doses of vitamin D and phosphate for
  Giant cell variant of soft-part chondroma mild, transitory improvement.21 However, serum calcium
  Giant cell MFH levels usually are normal in contrast to other forms of
Nonossifying fibromas 8
osteomalacia.
“Mesenchymal” tumors (malignant) 9
Osteoblastomas 4 After the tumor is excised, a dramatic reversion to
Cartilaginous 2 normal bone metabolism implies that the tumor is directly
  Chondroma 1 responsible for skeletal changes. Immunoassays per-
  Mesenchymal chondrosarcoma 1 formed on saline tumor extracts show no evidence of
Odontogenic 2
Undifferentiated sarcomas 2
parathyroid hormone or calcitonin.61,62 Elaboration by
Other 5 the tumor of a “phosphaturic substance” that would act
  “Degenerate osteoid tissue” on the proximal renal tubule and cause a renal phosphate
  Neuroma leak has been proposed.35,43,50,61,62 Some investigators were
  Osteosarcoma able to induce phosphaturia in experimental animals by
  Fibrous xanthoma
  “Vascular” fibrous histiocytoma injection of saline tumor extract that did not contain
parathyroid hormone or calcitonin.2,34,53 A substance
Modified from Nuovo MA, et al: Am J Surg Pathol 13:588–599, affecting phosphaturia alone would not explain the low
1989. or undetectable levels of 1,25-dihydroxyvitamin D3.16 It
GCT, Giant-cell tumor; GCRG, giant-cell reparative granuloma;
PVNS, pigmented villonodular tenosynovitis; MFH, malignant has been postulated that the decreased levels of 1,25
fibrous histiocytoma. dihydroxyvitamin D3 are due to elaboration and secretion
by the tumor of a factor that would inhibit the hydroxy-
lase, which acts on the mitochondria of the proximal
patients.14,84 If the tumor cannot be completely removed renal tubule. Phosphaturia would be a direct effect
because of its location, partial removal is necessary to of 1,25-dihydroxyvitamin D3 deficiency, because it has
support the medical treatment to reverse symptoms been proved that the latter increases renal phosphate
and skeletal changes. Recurrence of the original tumor resorption.32 Transplantation of soft tissue hemangioperi-
(benign or malignant) results in the reappearance of the cytoma causing osteomalacia into athymic mice resulted
chemical findings with or without accompanying clinical in hyperphosphatemia, high serum alkaline phosphate
symptoms or skeletal changes. In fact, chemical monitor- levels, and increased urinary phosphorus excretion.45 On
ing can be used for identification of local recurrence or the other hand, decreased levels of the metabolically
metastasis. active form of vitamin D3 alone do not fully account for
Correction of the laboratory abnormalities usually the phosphate depletion because increasing the dosage
occurs from 1 day to 20 months after surgery. Symptoms does not correct the defect.23,62,63,73,80,86
may disappear from the first day to 1 year after resec- The circulating phosphaturic factor secreted by the
tion.43,55,67,71 Continuation of medical treatment (vitamin tumor cells was originally termed phosphatonin but is now
D, phosphate) is required in patients with advanced skel- recognized to represent fibroblast growth factor-23
etal changes and persistent symptoms.13,44 In some cases (FGF23).17,22,32,33,70 Missense mutations in the gene encod-
the persistence of symptoms, chemical abnormalities, and ing FGF23 are associated with the development of
skeletal changes can be attributed to incomplete removal tumor-induced osteomalacia and rickets in the majority
of the tumor.18,57 In malignant tumors, local recurrence of cases. They also play a role in autosomal dominant
and metastases are associated with reappearance of chem- hypophosphatemic rickets, which has clinical manifesta-
ical changes, skeletal abnormalities, and clinical symp- tions virtually identical to those of X-linked hypophos-
toms of rickets or osteomalacia.38,61,62 phatemic rickets and tumor-induced osteomalacia.
Follow-up of reported cases varies from 3 days to Mutations of the PHEX gene are believed to play a role
30 years. In eight cases, the asymptomatic patients in the development of X-linked hypophosphatemia. The
were followed for more than 5 years. Among the 10 FGF23 gene, located on human chromosome 12p1,3 is
histologically malignant tumors, only 1 recurred and 2 one of 23 known members of the FGF family.68,70 The
metastasized, leading to death as a result of generalized full-length FGF23 is an 26-kDa (251 amino acids) protein

ERRNVPHGLFRVRUJ
1350 22  Tumor-Induced Osteomalacia and Rickets

A B

C D
FIGURE 22-2  ■  Hemangiopericytoma associated with osteomalacia: radiographic and microscopic features. A, Radiograph of chest
showing an expansile lesion involving the rib. Inset, T2- weighted magnetic resonance image of the same lesion. Note high signal
intensity of the tumor. B, Photomicrograph of tumor composed of oval and spindle cells with branching stag-horn pattern of vascular
spaces. C, Different area of tumor shown in B with tightly packed parieytic cells and prominent cystic vascular spaces. D, Higher
magnification of C showing solid area of pericytic cells. (B and C, ×100; D, ×400) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
22  Tumor-Induced Osteomalacia and Rickets 1351

A B C

D E

FIGURE 22-3  ■  Nonossifying fibroma associated with rickets: radiographic and microscopic features. A and B, Radiograph of the
knee showing radiolucent zones on the metaphyseal sides of growth plates. Note an inconspicuous eccentric lesion with sclerotic
margins involving the femoral metastasis diagnosed as nonossifying fibroma (arrows). C-E, Photomicrographs of nonossifying
fibroma showing proliferation of spindle cells arranged in storiform pattern with interstitial hemorrhage and scattered osteoclast-like
multinucleated giant cells. (C-E, ×200) (C-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1352 22  Tumor-Induced Osteomalacia and Rickets

B C

D E

FIGURE 22-4  ■  Intracortical osteosarcoma associated with rickets: radiographic and microscopic features. A, Radiograph of tibia
showing radiolucent intracortical lesion involving the proximal metaphysis. Note radiolucent zones on metaphyseal sides of growth
plates characteristic of rickets. B, Magnetic resonance image of same case as A showing eccentric intracortical lesions of low signal
intensity. C, Gross photograph of case shown in A and B, showing expansile intracortical lesion with heavily mineralized matrix.
D, Low power photomicrograph showing intracortical lesion with prominent production of tumor osteoid. E, Higher magnification
of D showing tumor osteoid production and its deposition on the preexisting host bone. Inset, Atypia of tumor cells producing
osteoid. (D, ×40; E and Inset, ×400) (D, E, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
22  Tumor-Induced Osteomalacia and Rickets 1353

Chr12 FGF23
13.3 FGF23

Exon 1

Exon 2

Exon 3
13.1 13.2
12.3

cds
12.2 12.1
11.2
11 11.1
12
13.1
tel cen
13.3 13.2
14
15 FGF23

4
21.1
21.2
21.3 1 251 aa
22 176 RXXR 179
23
Missense substitution
24.1
Substitution - coding silent
24.2
24.31 24.32
24.33
A

Osteoblast Fi
PHEX PHEX
Calcium in diet and Phosphate in diet and
1,25-(OH)2D Fa
serum increases serum increases
Osteocyte

FGF23 FGF23a FGF23i

PT DCT
3Na
Klotho
FGFR1

2Pi
Tubular reabsorption
PTH FGF23 of phosphate 1a–hydroxylase 1,25–(OH)2D

B C
FIGURE 22-5  ■  The FGF23 gene and its regulatory pathway. A, Chromosomal location of the FGF23 gene on the short arm of chro-
mosome 12 (left diagram). The exon-intron structure of the FGF23 gene is shown (upper right diagram). The linear structure of the
encoded FGF23 protein with the position of the mutations in the RXXR motif are shown (lower right diagram). B, Regulatory pathway
of FGF23. Solid lines represent promotion and dotted lines represent inhibition. Calcium, phosphorus, and 1,25-(OH)2D in food and
serum can stimulate the synthesis of FGF23. FGF23 directly inhibits the secretion of 1,25-(OH)2D, and inhibits parathyroid hormone
and blocks the synthesis of 1,25(OH)2D. In contrast, FGF23 inhibits the reabsorption of phosphorus in the renal tubules to decrease
serum phosphate levels, whereas hypophosphatemia stimulates secretion of 1,25(OH)2D. As a result, the serum level of 1,25(OH)2D
depends on both FGF23 and the phosphate level. C, Interaction of FGF23 and PHEX, and their influence on serum 1,25(OH)2D and
phosphate levels. FGF23 secreted from osteocytes binds to FGFR1, and inhibits the NaPi symporter and the activity of 1α-hydroxylase
(CPT27B1). PHEX from osteoblasts activates inactive factor (Fi) to produce active factor (Fa), which exists upstream of FGF23. Fa
inactivates FGF23. Elevation of 1,25(OH)2D inhibits PHEX activity through a negative feedback pathway. (B and C, Adapted and reprinted
with permission from Liao E: Front Med 27:65–80, 2013.)

with an NH2-terminal region containing the FGF (FGFR1c and FGFR3c) interacting with the klotho
homology domain and a novel 71-amino acid COOH protein.27 In physiologic states, FGF23 plays a central
terminus.70 Cleavage of FGF23 at the 176-RXXR-179 role in phosphate homeostasis, interacting with 1,25-
motif generates biologically inactive NH2- and COOH- (OH)2D, parathyroid hormone, and tubular reabsorption
terminal fragments.76 The basic physiologic defects in of phosphate (Fig. 22-5). It is also a PHEX substrate that
tumor-induced osteomalacia and rickets depends on provides a functional link between FGF23-mediated
increased activity of circulating mutant FGF23 produced phosphaturic syndrome and PHEX mutations in X-linked
by tumor cells that cannot be cleaved into inactive frag- phosphatemia.5 Its primary hyperactivation by mutations
ments.69 The increased FGF23 activity reduces the that prevent degradation of the active protein or by over-
expression of NaPi-2α and β (type 2 sodium phosphate expression by the upstream regulatory factors decreases
cotransporters) on the apical surface of renal proximal the mineralization of bone and inhibits renal tubular
tubules.37 The signal transduction of FGF23 is accom- reabsorption of phosphate, causing the phosphate waste
plished by forming a complex with FGF receptors related syndromes (Fig. 22-6).

ERRNVPHGLFRVRUJ
1354 22  Tumor-Induced Osteomalacia and Rickets

PHEX
Mutations
inhibit
proteolysis Oligopeptide Substrates
Subtilisin-like
proprotein convertase
RXXR
FGF23 MEPE
N FGF-like domain C N FGF-like domain C
1 176 179 251 1 176 179 251

Inactive FGF23N- and C-


Terminal Fragments

Proliferative
FGFR3C
Hypertrophic
Chondrocytes
PHEX

Proximal
tubule
25(OH)D
Osteoblasts
O s
1a hydroxylase 1,25(OH)2D
Abnormal
1,25–(OH)2D3
Na+ PTHr
NaPi2 Phex Rickets / Osteomalacia
R
PTH
PO43– FGFR2C
Phosphaturia PTH
FGFR3C

Kidney Skeleton
FIGURE 22-6  ■  Biologic effects of FGF23 and PHEX, explaining the pathogenesis of autosomal dominant phosphatemic rickets,
X-linked hypophosphatemia, and sporadic oncogenic rickets and osteomalacia. In this model, mutations of PHEX and FGF23 are
linked to autosomal dominant phosphatemic rickets, X-linked hypophosphatemia, and the tumor-induced rickets and osteomalacia.
Increased levels of mutant uncleaved FGF23, by interaction with FGF receptors (FGFRs) 2C and 3C in kidney and skeleton, and MEPE
or its fragments acting to modulate mineralization of extracellular matrix, are responsible for the phosphaturia and rickets/
osteomalacia. (Modified and reprinted with permission from Quarles DL: Am J Physiol Endocrinol Metab 285:E1–E9, 2003.)

The RXXR motif of FGF23 is similar to the consensus 4. Boriani S, Campanacci M: Osteoblastoma associated with osteo-
recognition site for precursor convertases, which belong malacia (presentation of a case and review of the literature). Ital J
to the family of serine proteases of the subtilisin/kexin Orthop Traumatol 4:379–382, 1978.
5. Bowe AE, Finnegan R, Jan de Beur SM, et al: FGF-23 inhibits
type. The increased level of uncleaved FGF23 in periph- renal tubular phosphate transport and is a PHEX substrate. Biochem
eral blood can be used to confirm preoperative diagnosis Biophys Res Commun 284:977–981, 2001.
of tumor-induced osteomalacia and to monitor the treat- 6. Camus JP, Crouzet J, Prier A, et al: Hypophosphoremic osteo-
ment.78,85,88 The demonstration that some of the tumors malacia secondary to benign connective tissue tumors: a report
on three unusual cases with vitamin D metabolites measure-
involved in the syndrome may express somatostatin ments (author’s transl). Ann Med Interne (Paris) 131:422–426,
receptors has led to the use of octreotide imaging using 1980.
labeling with indium-111 to detect occult phosphaturic 7. Carpenter TO: The expanding family of hypophosphatemic syn-
mesenchymal tumors and in some cases to treat overpro- dromes. J Bone Miner Metab 30:1–9, 2012.
duction of FGF23 secretion by these tumors.19,22,30,66 8. Castleman B, McNeely BU: Case records of the Massachusetts
General Hospital. N Engl J Med 273:494–504, 1965.
9. Chacko V, Joesph B: Osteomalacia associated with haemangioperi-
cytoma. J Indian Med Assoc 76:173–175, 1981.
REFERENCES 10. Chong WH, Molinolo AA, Chen CC, et al: Tumor-induced osteo-
1. Agus ZS: Oncogenic hypophosphatemic osteomalacia. Kidney Int malacia. Endocr Relat Cancer 18:R53–R77, 2011.
24:113–123, 1983. 11. Chong WH, Yavuz S, Patel SM, et al: The importance of whole
2. Aschinberg LC, Solomon LM, Zeis PM, et al: Vitamin D-resistant body imaging in tumor-induced osteomalacia. J Clin Endocrinol
rickets associated with epidermal nevus syndrome: demonstration Metab 96:3599–3600, 2011.
of a phosphaturic substance in the dermal lesions. J Pediatr 91:56– 12. Cotton GE, Van Puffelen P: Hypophosphatemic osteomalacia sec-
60, 1977. ondary to neoplasia. J Bone Joint Surg 68A:129–133, 1986.
3. Asnes RS, Berdon WE, Bassett CA: Hypophosphatemic rickets in 13. Daniels RA, Weisenfeld I: Tumorous phosphaturic osteomalacia.
an adolescent cured by excision of a nonossifying fibroma. Clin Report of a case associated with multiple hemangiomas of bone.
Pediatr (Phila) 20:646–648, 1981. Am J Med 67:155–159, 1979.

ERRNVPHGLFRVRUJ
22  Tumor-Induced Osteomalacia and Rickets 1355

14. Dent CE, Gertner JM: Hypophasphatemic osteomalacia in fibrous 39. Linsey M, Smith W, Yamauchi H, et al: Nasopharyngeal angiofi-
dysplasia. Q J Med 45:411–420, 1976. broma presenting as adult osteomalacia: case report and review of
15. Dent CE, Friedman M: Hypophosphataemic osteomalacia with the literature. Laryngoscope 93:1328–1331, 1983.
complete recovery. Br Med J 5399:1676–1679, 1964. 40. Martini A, Notarangelo LD, Barberis L, et al: Acquired vitamin
16. Drezner MK, Feinglos MN: Osteomalacia due to 1α,25- D-resistant rickets caused by prolonged latency in appearance of
dihydroxycholecalciferol deficiency. Association with a giant cell bone tumor. Am J Dis Child 137:1205–1206, 1983.
tumor of bone. J Clin Invest 60:1046–1053, 1977. 41. McCance RA: Osteomalacia with Looser’s nodes (Milkman’s syn-
17. Econs MJ, Drezner MK: Tumor-induced osteomalacia: unveiling a drome) due to raised resistance to vitamin D acquired about the
new hormone (editorial comment). N Engl J Med 330:1679–1681, age of 15 years. QJM 16:33–47, 1947.
1994. 42. McClure J, Smith PS: Oncogenic osteomalacia. J Clin Pathol
18. Evans DJ, Azzopardi JG: Distinctive tumours of bone and soft 40:446–453, 1987.
tissue causing acquired vitamin-D-resistant osteomalacia. Lancet 43. Miyauchi A, Fukase M, Tsutsumi M, et al: Hemangiopericytoma-
1:353–354, 1972. induced osteomalacia: tumor transplantation in nude mice
19. Ferraz MP, Watanabe T, Sado HN, et al: Concordance between causes hypophosphatemia and tumor extracts inhibit renal 25-
whole-body scintigraphy 111In-octreotide and 99mTc-sestamibi hydroxyvitamin D 1-hydroxylase activity. J Clin Endocrinol Metab
uptake in the detection of four tumor-induced osteomalacia cases. 67:46–53, 1988.
J Clin Endocrinol Metab 99:699–700, 2014. 44. Moncrieff MW, Brenton DP, Arthur LJ: Case of tumour rickets.
20. Folpe AL, Fanburg-Smith JC, Billings SD, et al: Most osteomalacia- Arch Dis Child 53:740–745, 1978.
associated mesenchymal tumors are a single histopathologic entity. 45. Moser CR, Fessel WJ: Rheumatic manifestations of hypophospha-
an analysis of 32 cases and a comprehensive review of the literature. temia. Arch Intern Med 134:674–678, 1974.
Am J Surg Pathol 28:1–30, 2004. 46. Nakashima A, Unni KK, Shives TC, et al: Mesenchymal chondro-
21. Fukumoto Y, Tarui S, Tsukiyama K, et al: Tumor-induced vitamin sarcoma of bone and soft tissue: a review of cases. Cancer 57:2444–
D-resistant hypophosphatemic osteomalacia associated with proxi- 2453, 1986.
mal renal tubular dysfunction and 1,25-dihydroxyvitamin D defi- 47. Nitzan DW, Marmary Y, Azaz B: Mandibular tumor-induced mus-
ciency. J Clin Endocrinol Metab 49:873–878, 1979. cular weakness and osteomalacia. Oral Surg Oral Med Oral Pathol
22. Fukumoto S, Yamashita T: Fibroblast growth factor-23 is the phos- 52:253–256, 1981.
phaturic factor in tumor-induced osteomalacia and may be phos- 48. Nomura G, Koshino Y, Morimoto H, et al: Vitamin D resistant
phatonin. Curr Opin Nephrol Hypertens 11(4):385–389, 2002. hypophosphatemic osteomalacia associated with osteosarcoma of
23. Fukumoto S: Diagnostic modalities for FGF23-producing tumors the mandible: report of a case. Jpn J Med 21:35–39, 1982.
in patients with tumor-induced osteomalacia. Endocrinol Metab 49. Nuovo MA, Dorfman HD, Sun CC, et al: Tumor-induced osteo-
29:136–143, 2014. malacia and rickets. Am J Surg Pathol 13:588–599, 1989.
24. Furuya K, Isobe Y, Morita M: Hypertrophic osteoarthropathy and 50. Olefsky J, Kempson R, Jones H, et al: “Tertiary” hyperparathyroid-
hypophosphatemic osteomalacia associated with tumor]. Gan to ism and apparent “cure” of vitamin-D-resistant rickets after
Kagaku Ryoho 13:2056–2064, 1986. removal of an ossifying mesenchymal tumor of the pharynx. N Engl
25. Gardner KH, Shon W, Folpe AL, et al: Tumor-induced osteoma- J Med 286:740–745, 1972.
lacia resulting from primary cutaneous phosphaturic mesenchymal 51. Parker MS, Klein I, Haussler MR, et al: Tumor-induced osteoma-
tumor: a case and review of the medical literature. J Cutan Pathol lacia. Evidence of a surgically correctable alteration in vitamin D
40:780–784, 2013. metabolism. JAMA 245:492–493, 1981.
26. Gitelis S, Ryan WG, Rosenberg AG, et al: Adult-onset hypophos- 52. Pollack JA, Schiller AL, Crawford JD: Rickets and myopathy cured
phatemic osteomalacia secondary to neoplasm. A case report and by removal of a nonossifying fibroma of bone. Pediatrics 52:364–
review of the pathophysiology. J Bone Joint Surg 68A:134–138, 371, 1973.
1986. 53. Popovtzer MM: Tumor-induced hypophosphatemic osteomalacia
27. Goetz R, Nakada Y, Hu MC, et al: Isolated C-terminal tail of (TUO): evidence for a phosphaturic cyclic AMP-independent
FGF23 alleviates hypophosphatemia by inhibiting FGF23-FGFR- action of tumor extract. Clin Res 29:418A, 1981.
Klotho complex formation. PNAS 107:407–412, 2010. 54. Prader VA, Illig R, Vehlinger E, et al: Rachitis infolge Knochentu-
28. Hasegawa T, Shimoda T, Yokoyama R, et al: Intracortical osteo- mors. Helv Paediatr Acta 14:554–565, 1959.
blastic osteosarcoma with oncogenic rickets. Skeletal Radiol 18:41– 55. Prowse M, Brooks PM: Oncogenic hypophosphatemic osteomala-
45, 1999. cia associated with a giant cell tumour of a tendon sheath. Aust NZ
29. Hosalkar HS, Jones DH, Offiah A, et al: Lincar sebaceous naevus J Med 17:330–332, 1987.
syndrome and resistant rickets. J Bone Joint Surg Br 85:578–583, 56. Quarles LD: FGF23, PHEX, and MEPE regulation of phosphate
2003. homeostasis and skeletal mineralization. Am J Physiol Endocrinol
30. Houang M, Clarkson A, Sioson L, et al: Phosphaturic mesenchy- Metab 285(1):E1–E9, 2003.
mal tumors show positive staining for somatostatin receptor 2A 57. Renton P, Shaw DG: Hypophosphatemic osteomalacia secondary
(SSTR2A). Hum Pathol 44:2711–2718, 2013. to vascular tumors of bone and soft tissue. Skeletal Radiol 1:21–24,
31. Jefferis AF, Taylor PC, Walsh-Waring GP: Tumour-associated 1976.
hypophosphataemic osteomalacia occurring in a patient with an 58. Riberio Leite MO, Borelli A, Barros de Ulhoa Cintra A: Osteoma-
odontogenic tumour of the maxilla. J Laryngol Otol 99:1011–1017, lacia hipofosfatemica associada a hemangio-fibroma: consideracoes
1985. etiopatogenicas. Rev Hosp Clin Fac Med Sao Paulo 33:65–67, 1978.
32. Kumar R: Tumor-induced osteomalacia and the regulation of phos- 59. Rico H, Fernandez-Miranda E, Sanz J, et al: Oncogenous osteo-
phate homeostasis. Bone 27(3):333–338, 2000. malacia: a new case secondary to a malignant tumor. Bone 7:325–
33. Kumar R: New insights into phosphate homeostasis: fibroblast 329, 1986.
growth factor 23 and frizzled-related protein-4 are phosphaturic 60. Robertson A: Case of the winter season. Hypophosphatemic osteo-
factors derived from tumors associated with osteomalacia. Curr malacia secondary to hemangiopericytoma of the right femur.
Opin Nephrol Hypertens 11(5):547–553, 2002. Semin Roentgenol 18:5–6, 1983.
34. Lau K, Stom MC, Goldberg M, et al: Evidence for a humoral 61. Ryan EA, Reiss E: Oncogenous osteomalacia: review of the world
phosphaturic factor in oncogenic hypophosphatemic osteomalacia. literature of 42 cases and report of two new cases. Am J Med 77:
Clin Res 27:421A, 1979. 501–512, 1984.
35. Leehey DJ, Ing TS, Daugirdas JT: Fanconi syndrome associated 62. Ryan WG, Gitelis S, Charters JR: Studies in a patient with tumor-
with a non-ossifying fibroma of bone. Am J Med 78:708–710, 1985. induced hypophosphatemic osteomalacia. Calcif Tissue Int 38:358–
36. Lejeune E, Bouvier M, Meunier P, et al: Osteomalacia of mesen- 362, 1986.
chymatous tumors. Apropos of a new case]. Rev Rhum Mal Osteo- 63. Salassa RM, Jowsey J, Arnaud CD: Hypophosphatemic osteomala-
artic 46:187–193, 1979. cia associated with “nonendocrine” tumors. N Engl J Med 283:65–
37. Liao E: FGF23 associated bone diseases. Front Med 7:65–80, 2013. 70, 1970.
38. Linovitz RJ, Resnick D, Keissling P, et al: Tumor-induced osteo- 64. Santamaria Sandi J, Elorza Olabegoya R, Beitia Martin JJ, et al:
malacia and rickets: a surgically curable syndrome. Report of two Tumoral hypophosphatemic osteomalacia]. Med Clin (Barc) 85:191–
cases. J Bone Joint Surg 58A:419–423, 1976. 193, 1985.

ERRNVPHGLFRVRUJ
1356 22  Tumor-Induced Osteomalacia and Rickets

65. Seshadri MS, Cornish CJ, Mason RS, et al: Parathyroid hormone- 78. Takeuchi Y, Suzuki H, Ogura S, et al: Venous sampling for fibro-
like bioactivity in tumours from patients with oncogenic osteoma- blast growth factor-23 confirms preoperative diagnosis of tumor-
lacia. Clin Endocrinol (Oxf) 23:689–697, 1985. induced osteomalacia. J Clin Endocrinol Metab 89:3979–3982,
66. Seufert J, Ebert K, Muller J, et al: Octreotide therapy for tumor- 2004.
induced osteomalacia. N Engl J Med 345:1883–1888, 2001. 79. Tang JS, Gold RH, Mirra JM, et al: Hemangiopericytoma of bone.
67. Shenker Y, Grekin RJ: Oncogenic osteomalacia. Isr J Med Sci Cancer 62:848–859, 1988.
20:739–741, 1984. 80. Taylor HC, Santa-Cruz D, Teitelbaum SL, et al: Assessment
68. Shimada T, Mizutani S, Muto T, et al: Cloning and characteriza- of calcitriol and inorganic phosphate therapy before cure of oncog-
tion of FGF23 as a causative factor of tumor-induced osteomalacia. enous osteomalacia by resection of a mixed mesenchymal tumor.
Proc Natl. Acad Sci USA 98:6500–6505, 2001. Bone 9:37–43, 1988.
69. Shimada T, Muto T, Urakawa I, et al: Mutant FGF-23 responsible 81. Turner ML, Dalinka MK: Osteomalacia: uncommon causes. AJR
for autosomal dominant hypophosphatemic rickets is resistant to Am J Roentgenol 133:539–540, 1979.
proteolytic cleavage and causes hypophosphatemia in vivo. Endo- 82. Weidner N, Bar RS, Weiss D, et al: Neoplastic pathology of onco-
crinology 143(8):3179–3182, 2002. genic osteomalacia/rickets. Cancer 55:1691–1705, 1985.
70. Shimada T, Urakawa I, Yamazaki Y, et al: FGF-23 transgenic mice 83. Weidner N, Santa Cruz D: Phosphaturic mesenchymal tumors.
demonstrate hypophosphatemic rickets with reduced expression of A polymorphous group causing osteomalacia or rickets. Cancer
sodium phosphate cotransporter type IIa. Biochem Biophys Res 59:1442–1454, 1987.
Commun 314(2):409–414, 2004. 84. Wener M, Cohen L, Bar RS, et al: Regulation of phosphate and
71. Siris ES, Clemens TL, Dempster DW, et al: Tumor-induced osteo- calcium metabolism by vitamin D metabolites: studies in a patient
malacia. Kinetics of calcium, phosphorus, and vitamin D metabo- with oncogenic osteomalacia. Arthritis Rheum 22:672–673, 1979.
lism and characteristics of bone histomorphometry. Am J Med 85. Westerberg PA, Olauson H, Toss G, et al: Preoperative tumor
82:307–312, 1987. localization by means of venous sampling for fibroblast growth
72. Sissons HA: Osteoporosis and osteomalacia, Baltimore, 1976, Williams factor-23 in a patient with tumor-induced osteomalacia. Endocr
& Wilkins. Pract 14:362–367, 2008.
73. Sparagana M: Tumor-induced osteomalacia: long-term follow-up 86. Willhoite DR: Acquired rickets and solitary bone tumor: the ques-
of two patients cured by removal of their tumors. J Surg Oncol tion of a causal relationship. Clin Orthop 109:210–211, 1975.
36:198–205, 1987. 87. Wyman AL, Paradinas FJ, Daly JR: Hypophosphataemic osteoma-
74. Stanbury SW: Tumor-associated hypophosphatemic osteomalacia lacia associated with a malignant tumour of the tibia: report of a
and rickets. Clin Endocrinol (Oxf) 1:256–266, 1972. case. J Clin Pathol 30:328–335, 1977.
75. Stone E, Bernier V, Rabinovich S, et al: Oncogenic osteomalacia 88. Yamazaki Y, Okazaki R, Shibata M, et al: Increased circulatory
associated with a mesenchymal chondrosarcoma. Clin Invest Med level of biologically active full-length FGF-23 in patients with
7:179–185, 1984. hypophosphatemic rickets/osteomalacia. J Clin Endocrinol Metab
76. Sundaram M, McCarthy EF: Oncogenic osteomalacia. Skeletal 87(11):4957–4960, 2002.
Radiol 29:117–124, 2000. 89. Yoshikawa S, Kawabata M, Hatsuyama Y, et al: Atypical vitamin-D
77. Sweet RA, Males JL, Hamstra AJ, et al: Vitamin D metabolite levels resistant osteomalacia. report of a case. J Bone Joint Surg 46A:998–
in oncogenic osteomalacia. Ann Intern Med 93:279–280, 1980. 1007, 1964.

ERRNVPHGLFRVRUJ
C H A P T E R 2 3 

Reactive and Metabolic Conditions


Simulating Neoplasms of Bone

CHAPTER OUTLINE

MYOSITIS OSSIFICANS Cortical Irregularity Syndrome


(Periosteal Desmoid)
MYOSITIS OSSIFICANS PROGRESSIVA
Stress Fractures
REACTIVE LESIONS OF THE BONE SURFACE
PUBIC OSTEOLYSIS
Florid Reactive Periostitis
Bizarre Parosteal Osteochondromatous TUMORAL CALCINOSIS
Proliferation (Nora’s Lesion) TOPHACEOUS PSEUDOGOUT
Acquired Osteochondroma (Turret Exostosis)
Subungual (Dupuytren’s) Exostosis TOPHACEOUS GOUT

EXUBERANT FRACTURE CALLUS CHRONIC RECURRENT MULTIFOCAL


OSTEOMYELITIS
AVULSION AND STRESS INJURIES
AMYLOIDOSIS OF BONE
Avulsion Injuries

subcutaneous tissue.12,13,15 Rare examples of intraabdomi-


MYOSITIS OSSIFICANS nal myositis ossificans have been also reported.35 A lesion
Definition located deep in soft tissue may present radiographically
and clinically as a lesion of the bone surface. Some lesions
Myositis ossificans is a reactive, self-limiting condition that originate in the parosteal soft tissue and do not
characterized by prominent heterotopic ossification. It involve the bone surface de novo may eventually, in the
predominantly occurs deep in soft tissue, and it undergoes process of their evolution, adhere to the adjacent bone.
zonal maturation. If the lesion is located in the vicinity This typically occurs in the later stages of the lesion’s
of bone (i.e., involves the structures of the bone surface), evolution.
a periosteal reaction contributes to its formation. This Most intramuscular lesions that involve deep soft
may alter the classic clinicopathologic presentation of the tissue are located in the lower extremities, whereas more
lesion. For this reason, we distinguish the classic (soft superficial lesions typically affect the upper extremities
tissue) and bone surface forms of myositis ossificans (par- and occur more frequently in women than in men. A
osteal myositis ossificans) (Fig. 23-1). The pathogenesis similar process seen predominantly in the acral parts has
of myositis ossificans is unknown, but it is generally con- been designated fibroosseous pseudotumor of the digits. It is
sidered a reactive process. Chromosome X inactivation described in this chapter under the term florid reactive
analysis indicates that it is a polyclonal disorder, but some periostitis in the section on reactive lesions of the bone
recent studies show rearrangement of the USP6 gene in surface.
a subset of myositis ossificans, which affects only a minor A peculiar form of heterotopic bone formation that
population of the stromal cells within the lesion.17,30 resembles myositis ossificans occurs in comatose patients
who have sustained head injuries.8 It is typically located
in the soft tissue around the hip joints, but other major
Incidence and Location
joints such as the shoulder area also can be affected.
Myositis ossificans predominantly affects adolescents and Heterotopic ossification of the soft tissue has also been
young, athletically active adults. Typically, it develops at described as a complication of many neurologic disor-
a site of clearly identifiable trauma, but in some cases, no ders, including tetanus, myelodysplasia, poliomyelitis,
history of injury can be obtained.1,4,5,16,22,24,26,29 The peak spinal cord injury, anoxic brain damage, and stroke.19,27,32,33
age incidence is during the second and third decades of These lesions resemble myositis ossificans in the sense
life, and nearly 50% of patients with this diagnosis are in that the fundamental soft tissue reaction is that of het-
this age group. It is usually found deep in soft tissue erotopic bone formation. However, they show no radio-
within the muscles of the lower or upper extremities (Fig. graphic or microscopic evidence of a zonal architecture
23-2). However, it may also involve fasciae, tendons, or nor do they ultimately form a peripheral shell of bone.
1357
ERRNVPHGLFRVRUJ
1358 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

Periosteum

Cortex
With periosteal With focal attachment
Medullary cavity reaction and no bone to bone surface
attachment

Soft tissue Parosteal


FIGURE 23-1  ■  Myositis ossificans in relation to bone surface. Parosteal lesions may induce periosteal reaction without true bone
attachment. Lesions located closer to bone may show focal or broad-based attachment to bone surface. Note that lesions attached
to bone surface do not show continuous fusion with underlying cortex. Prominent cartilage metaplasia (blue) within lesion and in
periosteal new bone formation can be present in the region corresponding to bone attachment.

Soft
Incidence

tissue

1 2 3 4 5 6 7 8
Age in decades

FIGURE 23-2  ■  Myositis ossificans. Peak age incidence and most frequently involved sites are indicated by arrows.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1359

Radiographic Imaging and a peripheral shell of reactive bone (Figs. 23-12 to


23-14). The outer surface of the reactive shell of the bone
Myositis ossificans has several ill-defined phases that may, is more sclerotic and better delineated from the adjacent
to some extent, be identified on radiographs.7,9,14,21,31 soft tissue than its inner surface. The inner border of the
These evolutionary phases follow an orderly time sclerotic rim merges imperceptibly with a softer, often
sequence and reflect maturation and then regression. Ini- hemorrhagic, center. The hemorrhagic center is seen in
tially, radiographs show an ill-defined mass in the soft younger lesions. More mature, older lesions show tan,
tissue. A well-developed zonal calcification pattern, char- fibrous, sometimes prominently vascularized tissue.
acteristic of the mature lesion, is typically seen 4 to 6 Myxoid or cystic changes are frequently seen in fully
weeks after an injury (Figs. 23-3 and 23-4). The mature matured or regressing older lesions. Initially the shell of
phase of myositis ossificans presents radiographically as the bone is ill-defined and sparsely mineralized with a
a well-demarcated, calcified mass. The calcifications are gritty consistency. At the other end of the spectrum are
accentuated at the periphery of the lesion and may form well-developed lesions that have a well-mineralized and
a shell at its periphery (Figs. 23-3 to 23-5). Less evident clearly demarcated peripheral shell of bone, which
faint, flocculent, irregular opacities, referred to as a requires decalcification for specimen processing. The
dotted veil, can be seen much earlier. A regressive phase lesions are typically less than 10 cm in size, and most are
is associated with some reduction in size and increased relatively small, measuring 3 to 6 cm.
mineralization at the periphery. Zonal architecture and
mineralization pattern are particularly well documented Microscopic Findings
by computed tomography and magnetic resonance
imaging.7,14 T1-weighted images show shell-like signal The distinct zonal architecture with predominantly
void of the periphery of the lesion (Figs. 23-3 and 23-5). peripheral heterotopic ossification is a hallmark of myo-
Central portions of the lesion show varying degrees of sitis ossificans (Figs. 23-12 to 23-14).1,2,3,6,11,16,18,31,34 This
signal enhancement on T2-weighted images (Figs. 23-3 feature is most evident in mature, fully developed lesions
and 23-5). at least 4 weeks old. The inner central portion of the
Myositis ossificans developing in the vicinity of bone lesion is composed of loose fibrovascular tissue. It resem-
(parosteal myositis ossificans) can be accompanied by a bles, to some extent, granulation tissue or a type of
periosteal new bone formation that typically has a multi- spindle-cell proliferation seen in nodular fasciitis (Fig.
layered onion-skin pattern (Figs. 23-4 and 23-6). The 23-15). Ultrastructural analyses show that the proliferat-
presence of a radiolucent cleft (space) that separates the ing spindle cells in this process are myofibroblasts.25
lesion from the periosteum helps distinguish the lesions Areas with ganglion-like giant mesenchymal cells that
that affect the soft tissue from those that primarily affect have prominent nucleoli, similar to those seen in prolif-
a bone surface (Figs. 23-4 and 23-8).20,21 The lesion may erative myositis, also may be present. These usually are
show a focal attachment to the bone surface (see Fig. seen in younger, evolving lesions. Lesions with a pre-
23-6). In areas of attachment, especially in a lesion of dominance of these types of cells are sometimes referred
long duration, it may be difficult to demonstrate a fibrous to as proliferative fasciitis. Evidence of fresh and old hem-
zone that separates the lesion from the adjacent cortex. orrhage with deposition of hemosiderin may be present.
However, even if there is radiographic evidence of attach- Prominent histiocytic and giant-cell reactions may also
ment, microscopically there is usually a narrow fibrous be seen and are usually concentrated in the areas of hem-
zone that separates the lesion from the cortex. Occasion- orrhage. Degenerated, entrapped skeletal muscle cells are
ally, myositis ossificans presents as a convex lesion seen focally. The peripheral parts of the lesion contain a
attached to the surface of the bone by a broad base network of reactive bone that shows prominent osteo-
(Figs. 23-7 and 23-8). Long-standing lesions show a blastic rimming (Fig. 23-15). The amount of osteoid and
linear pattern of mineralization at the periphery and may its maturation increases toward the periphery. Early
show a broad, bony stalk attached to the bone surface, lesions are dominated by florid stromal cell proliferations
which is fused with its cortex (Figs. 23-5, 23-9, and with nuclear atypia showing various stages of osteoid
23-10). This phase of myositis ossificans somewhat over- matrix deposition and mineralization (Figs. 23-16 and
laps radiographically and microscopically with acquired 23-17). Fully developed lesions are outlined by a clearly
osteochondroma or the so-called turret exostosis that is defined shell of mature bone. In well-developed lesions,
seen predominantly in the acral skeleton. progressive maturation from woven to lamellar bone may
Myositis ossificans occasionally involves the soft tissues be identified under polarized light. The outer edges of
of the trunk or the head and neck region (Fig. 23-11). If the lesion are separated from the surrounding skeletal
these sites are involved in patients younger than age 20 muscle by a loose fibrous capsule. Foci of metaplastic
years and especially if multifocal ossifications are present cartilage with enchondral ossification evolving into bone
in the soft tissue, myositis ossificans progressiva (fibro- trabeculae may occasionally be seen (Figs. 23-18 and 23-
dysplasia ossificans progressiva) must be ruled out. 19). Metaplastic cartilage is more likely to be present in
deeply sited lesions and often corresponds to the
Gross Findings areas of bone surface attachment (parosteal myositis ossi-
ficans). In this area a clear zonal architecture may be
The gross appearance of myositis ossificans corresponds disturbed by a periosteal reaction that contains foci
to the radiographic presentation of the lesion: a zonal of cartilage. In such cases, metaplastic hypercellular
architecture with a hemorrhagic, more cellular, center cartilage can dominate the lesion focally. In areas of

ERRNVPHGLFRVRUJ
1360 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-3  ■  Myositis ossificans: radiographic features. A, Plain radiograph shows ringlike calcification delineating mass in soft
tissue of medial aspect of proximal thigh (arrows). B, Computed tomogram of lesion in A shows well-circumscribed mass in soft
tissue with incomplete ring of peripheral calcification. C and D, Magnetic resonance images of same lesion show well-circumscribed
mass. Note that peripheral ring-shaped signal void corresponds to mineralization of bone within lesion.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1361

A B

C D
FIGURE 23-4  ■  Myositis ossificans: radiographic features. A, Ill-defined soft tissue mass with parallel periosteal reaction on surface
of adjacent bone. B, Same lesion as shown in A, x-rayed approximately 1 month later, shows distinct mineralization without typical
shell-like structure at the periphery. C, Juxtacortical myositis ossificans shows prominent peripheral maturation 6 months after direct
trauma. Note continuous radiolucent zone between cortex and bony parosteal mass. D, Specimen radiograph (slab x-ray film) of
lesion shown in A. Note mature bony shell and central lucency.

ERRNVPHGLFRVRUJ
1362 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A C

D E
FIGURE 23-5  ■  Myositis ossificans: radiographic features. A and B, Ill-defined soft tissue mass of the proximal thigh with mineraliza-
tion pattern accentuated at the periphery of the lesion. Note the attachment of the lesion to the underlying bone surface. C and
D, T1-weighted axial and coronal magnetic resonance images (MRI) show shell-like signal void at the periphery of the lesion.
E. Strong signal enhancement of the lesion on T2-weighted MRI.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1363

FIGURE 23-6  ■  Parosteal myositis ossificans of scapula: specimen radiograph. Note focal attachment of lesion to surface of adjacent
bone (left) and parallel periosteal new bone formation (arrows) on surface of adjacent bone (right).

attachment to a bone’s surface, focal proliferation of bone It is occasionally difficult to distinguish parosteal myo-
and metaplastic cartilage can mimic the areas seen in sitis ossificans from osteosarcoma (parosteal and periosteal
florid reactive periostitis or bizarre osteochondromatous osteosarcoma) of a bone surface, especially when the lesion
proliferation (Fig. 23-18) (also see the section on reactive is located near the surface of a major long tubular bone
lesions of the bone surface in this chapter). Early lesions of the lower extremities. Lesions located on the poste-
may show less evident zonal architecture, and the level of rior aspect of the distal femoral shaft and superior to
cellularity can be very high. In such lesions, florid prolif- the popliteal fossa should be carefully evaluated because
eration of spindle cells with giant cells may be the domi- parosteal osteosarcoma frequently is found at this site.
nant feature. Parosteal myositis ossificans usually shows a clear zone
of separation from the underlying cortical bone that can
be seen on radiographs. Lesions that show wide, broad
Differential Diagnosis
fusion with the underlying bone in this location should be
Distinguishing myositis ossificans from extraskeletal osteo- considered suspicious. Microscopically, parosteal osteo-
sarcoma and osteosarcoma of a bone surface is of para- sarcoma has unique features of hypercellular, fibroblas-
mount importance.5,11,16,21,31 The zonal architecture in tic stroma and a network of relatively well-developed
myositis ossificans is the single most important feature tumor bone. Paradoxically, the level of bone matura-
helpful in distinguishing this lesion from malignant tion in parosteal osteosarcoma often exceeds the level
bone-forming tumors. The zonal architecture of the of bone maturation in myositis ossificans. The overall
lesion can be seen on radiographs and in gross and micro- radiographic pattern of mineralization in parosteal osteo-
scopic examinations and provides important clues to the sarcoma is different from that seen in myositis ossificans
correct diagnosis. The microscopic features of immature because parosteal osteosarcoma has a heavily mineralized
bone trabeculae with osteoblastic rimming and progres- central or basal portion that is fused with the underlying
sion to more mature bone are not seen in osteosarcoma. cortex.
Moreover, extraskeletal osteosarcomas involving soft The occasional presence of cartilage with hypercellu-
tissue are usually high-grade lesions with obvious nuclear larity and nuclear atypia may require differentiation
atypia. Atypical mitoses are not present in myositis between myositis ossificans and periosteal osteosarcoma.
ossificans. Text continued on p. 1377

ERRNVPHGLFRVRUJ
1364 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B
FIGURE 23-7  ■  Parosteal myositis ossificans: radiographic features. A, Lateral radiograph of ankle of young adult shows partly calci-
fied mass in soft tissue adjacent to distal end of tibia. Peripheral density and central radiolucency indicate zonal maturation.
B, Juxtacortical mineralized mass adjacent to posterior cortex of femoral shaft. The lesion is in early stage of evolution and does
not yet show zonal maturation.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1365

A C
FIGURE 23-8  ■  Parosteal myositis ossificans: radiographic features. A, Lateral plain radiograph shows convex bony lesion on bone
surface of distal femur. Note lucent zone of separation from underlying cortex. B and C, Computed tomograms show lesion that
involves bone surface of anterolateral aspect of femoral shaft. Note line of separation in B. Underlying cortex is intact. Focal attach-
ment in central portion of base is seen in C.

ERRNVPHGLFRVRUJ
1366 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B
FIGURE 23-9  ■  Parosteal myositis ossificans, late phase: radiographic features. A and B, Lateral and oblique plain radiographs of
distal femur with exostosis-like lesion of bone surface.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1367

A
FIGURE 23-10  ■  Parosteal myositis ossificans, late phase: radiographic features. A, Exostosis-like bony outcropping on cortical surface
of humeral shaft in young adult with history of trauma. B, Computed tomogram shows cortically based, mature, bony excrescence,
which represents late stage of development (posttraumatic exostosis).

ERRNVPHGLFRVRUJ
1368 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-11  ■  Myositis ossificans: radiographic features A and B, Anteroposterior and lateral views of cervical spine show mineral-
ized mass in soft tissue anterolateral to C4 to C5 within paraspinal musculature (arrows). There is suggestion of peripheral matura-
tion and central lucency.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1369

A B

C D

FIGURE 23-12  ■  Myositis ossificans: microscopic features. A and B, Whole-mount photomicrographs of myositis ossificans demon-
strate zonal maturation of bone peripherally. C and D, Low power photomicrographs demonstrate zonal maturation of osteoid and
bone formation peripherally. (A and B, ×3; C and D, ×10.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1370 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D E
FIGURE 23-13  ■  Myositis ossificans, mature phase: microscopic features. A and B, Whole-mount photomicrographs of myositis ossi-
ficans demonstrate zonal maturation of bone peripherally. C, Well-developed shallow bone at periphery of lesion. D, Intermediate
zone showing well-organized trabeculae of bone rimmed by osteblasts. E, Central less ossified part of lesion is composed of thin
bone trabeculae and loose fibroblastic tissue with prominent vasculature. (A and B, ×3; C-E, ×25.) (A-E, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1371

B C

FIGURE 23-14  ■  Myositis ossificans, mature phase: microscopic features. A, Fragment of whole-mount section of entire lesion with
mature, well-developed shell of bone delineating its periphery. B, Central nonossified part of lesion is composed of loose fibroblastic
tissue that resembles nodular fasciitis. C, Well-developed shell of bone is seen at periphery of lesion. Note sharp delineation of
outer surface from surrounding soft tissue. (A, ×3; B, ×50; C, ×25) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1372 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

D
FIGURE 23-15  ■  Myositis ossificans: microscopic features. A-D, Zonal maturation of bone formation. A, Central nonossified part of
lesion is composed of loose fibroblastic proliferation and fresh hemorrhage that resembles nodular fasciitis or proliferative myositis.
B, Early phases of osteoid formation in the intermediate zone. C, Well-organized irregular trabeculae of osteoid rimmed by osteo-
blasts. D, Well-developed shell of bone at periphery of the lesion. Note sharp delineation of outer surface from surrounding soft
tissue. (A-D, ×5) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1373

D
FIGURE 23-16  ■  Myositis ossificans, early phase: miscroscopic features. A-D, Early phases of osteoid formation in myositis ossificans
(A-D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1374 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-17  ■  Myositis ossificans, early phase: microscopic features. A, Early osteoid formation. B, Florid proliferation of
fibroblastic cells in central zone of myositis ossificans. C, Early osteoid formation. D, Florid proliferation of plump spindle cells
in central portion of the lesion. Note cellular variability and some degree of hyperchromatism. (A and B, ×100; C and D, ×200)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1375

B C
FIGURE 23-18  ■  Parosteal myositis ossificans: microscopic features. A-C, Bizarre osteochondromatous proliferation dominates area
of bone surface attachment in parosteal myositis ossificans (A-C, ×25) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1376 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-19  ■  Parosteal myositis ossificans: microscopic features. A and B, Bizarre osteochondromatous proliferation dominates
area of bone surface attachment in parosteal myositis ossificans. C and D, Anastomosing pattern of osteoid deposition with promi-
nent osteoblastic rimming in areas of bone surface attachment in periosteal myositis ossificans. (A and C, ×25; B and D, ×50.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1377

The overall radiographic appearance of periosteal osteo- ossificans progressiva. Upregulation of the RUNX2/
sarcoma is different from that of myositis ossificans. Peri- CBFA1 gene in stromal cells of myositis ossificans pro-
osteal osteosarcoma is a true lesion of the bone surface gressiva increases the recruitment of chondroprogenitor
and has an associated periosteal reaction that is usually in and osteoprogenitor cells from the pool of uncom-
the form of perpendicular striations on the bone surface. mitted primitive mesenchymal cells.48 The resulting
In addition, true sarcomatous elements with direct tumor downstream deregulation of the bone morphogenetic
bone formation are a classic feature of periosteal osteo- protein network through the SMAD and MAPK kinase
sarcoma. Moreover, periosteal osteosarcoma shows a pathways causes ligand-independent activation of bone
seamless fusion with the underlying cortex, which is rela- morphogenetic protein signaling and ligand-dependent
tively uniform over the entire length of the lesion. hyper-responsiveness to bone morphogenetic protein
stimulation.36,44,64 Animal model studies indicate that
hematopoietic stem cells contribute to early phases of
Treatment and Behavior
heterotopic ossification induced by BMP-4.51 Although
Myositis ossificans is a benign, self-limiting condition. the genetic defects causing fibrodysplasia ossificans pro-
Conservative excision of the lesion is curative. This is gressiva are not completely understood, the clinical and
best performed during the mature phase of the disease, animal models implicate mutations in bone morphoge-
when the lesion is well delineated and a mass is clearly netic proteins, their receptors, and activin receptor type
identifiable. Incompletely excised lesions, especially those IA (ACVR1) as well as mutations of the noggin (NOG)
in the early phase of development, continue to grow for gene in the development of this disorder.56
a limited period (several weeks to months). Eventually,
these lesions stabilize and may undergo partial or com- Clinical Symptoms
plete regression.
Myositis ossificans is one of the rare extraskeletal con- Multifocal involvement of soft tissues, predominantly of
ditions that may be complicated by a superimposed sec- the trunk, a progressive chronic clinical course, and
ondary aneurysmal bone cyst.2 Several reports on a familial incidence and occurrence in young children
secondary malignant transformation in myositis ossifi- distinguish this disease from the more common, local-
cans are not convincing.10,23,28 Most likely, they represent ized, and solitary (posttraumatic) form of myositis
lesions that were originally misdiagnosed and were de ossificans.37,38,40,41,43,52,57
novo malignant lesion of the soft tissue. The first clinical manifestation is a localized, painful
A more recently published case by E. Konishi et al swelling in the soft tissue. The onset is often precipitated
may represent an extremely rare true example of an by a minor injury or an infectious disease. The lesion
osteosarcoma that has developed in association with a typically appears first within the muscles of the trunk (the
long-standing myositis ossificans.13 back, shoulder, and paravertebral region). The head and
neck region is also a frequent site of initial presentation.
Lesions of the head and neck are most often located in
MYOSITIS OSSIFICANS PROGRESSIVA the scalp and within the sternocleidomastoid muscle. As
it progresses, the disease moves into the proximal parts
Definition
of the extremities until it eventually involves their distal
Myositis ossificans progressiva (fibrodysplasia ossificans parts, which is typical of a more advanced, long-term
progressiva) represents an extremely rare, multifocal, process.40,41
generalized chronic progressive disease of heterotopic The disorder is often associated with abnormalities of
ossification within the body’s musculature and frequently bone and joint formation that principally affect the acral
shows a familial pattern of occurrence.38,41,42,57,63 It pre- skeleton.60 The changes predominantly consist of bilat-
dominantly affects children during the first decade of life, eral microdactyly and the absence of both thumbs and
primarily between birth and age 6 years. great toes. If great toes are present, they frequently show
Myositis ossificans progressiva is an autosomal domi- deviation (bilateral hallux valgus). Deafness and sexual
nant disorder caused by germline mutations of the activin infantilism can also be present.
A type 1 receptor (bone morphogenetic protein type 1
receptor) gene mapping to the 2q23-34 chromosomal Radiographic Imaging
region in both inherited and sporatic cases.58,59 The
majority of patients carry a germline (617G→A; R206H) On radiographs, the disorder presents as multifocal ossi-
mutation in the glycine-serine (Gs) activation domain of fications of soft tissue that progress to form a network of
the ACVR1 gene. Other less frequent mutations such as interconnecting bridges between adjacent bones and
(1067G→A; G356D) also involving the ACVR1 gene joints, causing ankylosis, muscle stiffness, and ultimately,
have been identified.46,47 Another predisposing locus heterotopic bone formation at the affected sites (Fig.
mapping to the 17q21-32 region with mutations of the 23-20).39,54,61,62
noggin (NOG) gene may be involved in a subset of
patients with myositis ossificans progressiva.45,55 Microscopic Findings
It is postulated that the mutations of ACVR1 cause
destabilization of the GS domain and permanently acti- The early lesions consist of multifocal proliferations
vate ACVR1, which in turn trigger exuberant ectopic of plump fibroblasts within the skeletal muscle, similar
chondrogenesis and osteogenesis, a hallmark of myositis to those seen in fibromatosis. In fact, early lesions of

ERRNVPHGLFRVRUJ
1378 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-20  ■  Myositis ossificans progressiva (fibrodysplasia ossificans progressiva): radiographic features. A, Multiple foci of
heterotopic ossification of soft tissue in forearm. Some foci are attached to adjacent bone surfaces. B, Lesion’s progression is shown
by formation of multiple bridges between ulna and radius.

myositis ossificans progressiva, especially in the sterno- tion of loose fibroblastic cells that are somewhat similar
cleidomastoid muscle, are often confused with juvenile to those seen in fasciitis or fibromatosis. In the second
fibromatosis. The lesions undergo mineralization and phase, thick collagen fibers appear among the fibroblasts
develop extensive bone metaplasia with formation of before osteoid deposition, mineralization, and the meta-
fusing bridges between adjacent bones and joints. The plastic change into bone and cartilage. In the third
development of bone can be associated with cartilage (regressing) phase, there is some reduction in size, and
metaplasia and can have features of enchondral ossifica- maturation is identified by the network of lamellar bone
tion.53 The zonal architecture typical of common solitary and the formation of firm bridges among individual
myositis ossificans is not present (Fig. 23-21). lesions and adjacent structures (articular ankylosis). Con-
Pathogenetically, the disease has three main phases trary to localized solitary myositis ossificans, this process
that somewhat parallel those of conventional solitary is multifocal, and the ossifications develop in the centers
myositis ossificans. The first phase consists of a prolifera- of individual lesions rather than at their periphery.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1379

A B

C D
FIGURE 23-21  ■  Myositis ossificans progressiva (fibrodysplasia ossificans progressiva): microscopic features. A, Multiple foci of het-
erotopic ossification within soft tissue. B, Focus of ossification within fibrous area. C and D, Higher power magnification shows
maturation of heterotopic bone with formation of interconnecting network of lamellar bone trabeculae. Note absence of zonal
architecture and tendency toward uniform ossification pattern that preferentially involves central portion of lesion. (A and B, ×5;
C and D, ×25.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1380 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

Differential Diagnosis maturation and organization rather than on a distinct


pathogenesis.107 The basic architectural features of these
Myositis ossificans progressiva should be differentiated conditions are summarized in Figure 23-23. Although
microscopically from fibromatosis and the localized the radiographic and microscopic data of these condi-
common form of myositis ossificans. The analysis of the tions clearly overlap, recent cytogenetic studies have
entire clinical and radiographic picture is usually very identified distinctive chromosomal rearrangements in
helpful: occurrence during the first decade of life, initial subungual (Dupuytren’s) exostosis and bizarre parosteal
involvement of trunk musculature, and presence of mul- ostechondromatous prolifereration (Nora’s lesion), which
tifocal proliferations. Radiographic and microscopic evi- suggests that these lesions are in fact neoplastic and may
dence of bone formation helps differentiate the lesions evolve via different molecular mechanisms.79,86,94,101,108
from fibromatosis. In addition, the clinical differential Several reports indicate that Nora’s lesion is charac-
diagnosis includes battered child syndrome, ectopic bone terized by a clonal balanced translocation t(1;17)(q32;
formation with multiple congenital anomalies, and q21) or variant translocations involving 1q32 or t(1:17)
pseudohypoparathyroidism. (q42:q23) (Fig 23-24).79,94 Conversely, the cells in subun-
gual exostosis carry a balanced clonal translocation t(x;6)
(q13-14;q22).86,91,101,108 The chromosomal breakpoints of
Treatment and Behavior
subungual exostosis cluster in 6q13-14 and Xq22 harbor-
Typically the disease is fatal and causes death after 10 to ing the collagen genes CoL12A1 and COL4A5, respec-
15 years of prolonged progressive illness with transient tively.101 The exact structure of putative chimeric gene(s)
spontaneous remissions.37,40,41,49,50,57,61 The fatal outcome and their fusion transcripts in subungual exostosis are
frequently results from the progressive immobilization of unknown at this time.
the thorax, which causes severe respiratory insufficiency.
Biopsy, trauma, and infections can exacerbate the process
and lead to the development of new lesions.
Florid Reactive Periostitis
The term florid reactive periostitis was proposed by Spjut
and Dorfman100 in 1981 to designate a rare lesion on the
bone surface that most frequently affects the short tubular
REACTIVE LESIONS OF bones of the hands and, less commonly, the feet.80,82,96,103
THE BONE SURFACE The peak incidence is during the third and fourth decades
of life. Nearly 70% of cases are diagnosed in patients
Periosteal (bone surface) reactive lesions that mimic between ages 20 and 40 years, at a mean age of 34 years.
primary neoplasms of bone occur in several distinct clini- There is no sex predilection. The lesions usually involve
copathologic settings and are designated as florid reactive the proximal and middle phalanges and less frequently,
periostitis, bizarre parosteal osteochondromatous proliferation, the metatarsals and the metacarpals but have also been
acquired osteochondroma (turret exostosis), and subungual reported in the long bones.68
(Dupuytren’s) exostosis.77,95,100 In the pathology literature Radiographically, there is a critical difference between
concerning soft tissue, these lesions are designated these proliferations and ordinary cartilage-capped osteo-
descriptively as fibroosseous pseudotumors.76,78,97 Common chondromas. There is no flaring out of the cortex of
to all these lesions is an initial hemorrhagic subperiosteal the adjacent bone or continuity of the central part of
collection that often can be related to trauma. the lesion with the underlying osseous medulla, as is seen
The lesion undergoes organization referred to as mat- in typical osteochondroma. The lesion appears to arise
uration. Because of its peculiar localization, a periosteal from the surface of the bone without disturbing the
reaction (i.e., new bone and cartilage of periosteal origin) architecture of the adjacent bony structure. The overall
participates in the process. Initially, spindle and giant shape of the lesion is that of a pedunculated or, more
cells, as well as hemorrhage with minimal bone and car- often, flat cap attached to the bone surface (Fig. 23-25).
tilage proliferation, dominate the lesion (florid reactive Early lesions present as an ill-defined density of soft
periostitis). Later, periosteal new bone formation and tissue that is attached to the bone surface and contains
metaplastic cartilage proliferation are dominant elements varying amounts of calcific material. Sometimes, rapid
(bizarre parosteal osteochondromatous proliferation, or growth is noted over several days or weeks. Most symp-
Nora’s lesion). Finally, the focus of ossification matures toms are related to the mechanics of the lesion, such as
with the formation of a bony base that is incorporated discomfort from wearing shoes or pain when flexing
into the underlying cortex and that more peripherally is digits.
covered with a cartilaginous cap (acquired osteochon- The size of the lesions varies from 0.5 to 3.0 cm. Most
droma, turret exostosis, subungual exostosis). lesions are grossly described as resembling osteochon-
All these lesions predominantly affect the phalanges of dromas because they have a bony matrix covered by car-
the hands and feet and have their peak incidence in the tilage. Microscopically, an irregular cartilage cap covers
third and fourth decades of life (Fig. 23-22). They rarely the outer surface of the lesion. The cartilage exhibits
involve the long tubular bones of the extremities. The marked proliferative activity. The osteocartilaginous
bones of the forearm are more often affected than other interface is irregular and strongly resembles a callus. Car-
long bones. In extremely rare instances, the lesions can tilage cells are numerous and have enlarged nuclei.
involve other bones. Spindle-cell fibroblastic proliferation with giant-cell
It is postulated that the radiologic and histologic reaction and fresh hemorrhage can dominate the lesion
features of these lesions depend on their stages of focally.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1381

Predominantly florid periostitis


Incidence

and bizarre
osteochondromatous
proliferations

1 2 3 4 5 6 7 8
Age in decades

Predominantly subungual
exostosis

FIGURE 23-22  ■  Reactive lesions of bone surface. Peak age incidence and most frequent sites of skeletal involvement are indicated
by solid black arrow.

Cartilage
Metaplastic cap
Periosteum
cartilage (blue)
Cortex
Medullary cavity

Florid reactive Bizarre Reactive


periostitis osteocartilaginous osteoochondroma
proliferation (Nora's lesion) (turret exostosis)
FIGURE 23-23  ■  Unified concept of reactive lesions of bone surface. It is postulated that several separately described entities (florid
reactive periostitis, bizarre osteochondromatous proliferation, acquired osteochondroma, and subungual exostosis) represent dif-
ferent phases of reactive processes initiated by trauma and subperiosteal hemorrhage with subsequent organization and maturation.
End stage of these processes and final common pathway is acquired exostosis (turret exostosis).

ERRNVPHGLFRVRUJ
1382 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

19 20 21 22 X Y
A

der(1)
I

1 der(1)

17 der(17) der(17)
B C 1

der(1)
1
17

D der(17) E F G H J
FIGURE 23-24  ■  Bizarre parosteal osteocartilaginous proliferation (Nora’s lesion): cytogenetic abnormalities. A, G-banding karyogram
showing reciprocal 1;17 translocation. (A, Case courtesy of Dr. Nina Tatevian, Texas Children’s Hospital, Houston, TX) B, Partial karyo-
gram showing reciprocal 1;17 translocation. Breakpoints are indicated by arrows. C, Three-color metaphase fluorescent in situ
hybridization (FISH) using BAC probes 99A19 (green), 145I20 (red), and 351B8 (yellow). Signals from all three probes are present in
the der(17)t(1;17), whereas the reciprocal der(1) has a signal from only the proximal probe 99A19. The insert presents only green
signals to demonstrate the uneven split of 99A19. D, Split of BAC probe 219F9 (yellow). Signals are seen on der(1), der(17), and
chromosome 17. Signals from the more proximal probe 243D13 (red) are present on der(17) and chromosome 17. The whole chro-
mosome 1 paint probe (green) demonstrates the presence of chromosome 1 material. E, Interphase nucleus (case 1) showing a split
in the 1q probes. Chromosome 1 is indicated by joined red and green signals (arrow), der(1) is indicated by a red signal (black
arrowhead), and der(17) is indicated by a green signal (white arrowhead). F, Interphase nucleus showing a split in the 17q probes.
Chromosome 17 is indicated by joined red and green signals (arrow), der(17) is indicated by a red signal (black arrowhead), and
der(1) is indicated by a green signal (white arrowhead). G, Interphase nucleus (case 2a) showing fusion of the proximal 1q and distal
17q probes. The fusion and der(1) are indicated by joined red and green signals (arrow), chromosome 1 is indicated by a red signal
(black arrowhead), and chromosome 17 is indicated by a green signal (white arrowhead). H, Normal tissue hybridized with the 1q
probes, where red and green signals are seen together on chromosome 1 (arrows). I and J, FISH probes used for detecting break-
points in chromosomes 1 and 17. BAC clones are represented by vertical lines. Probes that were split by the 1;17translocation are
marked with stars, and those used in interphase FISH analyses are shown in bold type. Probes situated on the proximal side and
on the distal side of the breakpoints are labeled red and green, respectively. The localization and size of BAC clones are given
according to the UCSC Genome Browser and NCBI Map Viewer. (B-J, Modified and reprinted with permission from Nilsson M, et al:
Human Pathol 35:1063–1069, 2004.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1383

A B

C D
FIGURE 23-25  ■  Florid reactive periostitis: radiographic and microscopic features. A, Swelling of soft tissue adjacent to proximal
phalanx of fifth finger shortly after injury. B, One month later, there is ill-defined calcification of soft tissue and periosteal new bone
formation. C and D, Low and intermediate power photomicrographs show proliferation of spindle cells and trabeculae of woven
bone. (C, ×25; D, ×50.) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1384 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

Bizarre Parosteal Osteochondromatous proliferation of cartilage is less evident, and the cartilage
Proliferation (Nora’s Lesion) cap is thinner and more regular and exhibits less cytologic
atypia. In contradiction to ordinary osteochondroma or
Bizarre parosteal osteochondromatous proliferation pre- osteocartilaginous exostosis, the underlying cortex is
dominantly involving the bones of the hands and feet has intact, and there is no continuity between the center of
been described by Nora et al.95 The peak age incidence the lesion and the medullary cavity of the affected bone.
is during the third and fourth decades of life, and the In summary, the microscopic features overlap with those
lesion predominantly affects the short tubular bones of of so-called subungual exostosis.
the hands and feet.65,71,74,75,85,87,90,105 The peak age inci-
dence and the anatomic sites involved closely overlap
with those of florid reactive periostitis. In fact, the lesion
Subungual (Dupuytren’s) Exostosis
is closely related to other reactive lesions of the bone Subungual exostosis is an osteocartilaginous reactive
surface and most likely represents an intermediate step lesion similar to acquired osteochondroma but arising
between florid reactive periostitis and acquired osteo- from a distal phalanx.66,67,69,70,72,73,81,84,88,89,93,98,99,102,104 It was
chondroma that is either subungual or the so-called first described by Dupuytren in 1847 as a bony growth
turret exostosis found on the shafts of phalanges and of the distal phalanx of the great toe.77 In fact, this is the
metacarpals. most frequent location of the lesion. The peak incidence
Radiographically, Nora’s lesion presents as a well- is in the second and third decades of life, at a mean age
delineated, cap-shaped lesion attached to the bone of 24 years. Nearly 80% of reported cases involve the
surface. Plain radiographs typically disclose a well- great toe, and the remaining cases involve other toes.
developed patchy or linear mineralization pattern Subungual exostosis rarely involves the fingers.
(Figs. 23-26 and 23-27). The radiographic features are typical. Frequently, the
Prominent periosteal new bone and cartilage forma- exostosis arises from the dorsal aspect of the tip of the
tion is a dominant feature of the lesion (Figs. 23-27 and distal phalanx (Figs. 23-31 and 23-32). In mature, well-
23-28). The cartilage shows hypercellularity, an open developed lesions, there is a clear trabecular pattern at
chromatin structure, and binucleated cartilage cells. In the base that connects the lesion to the underlying pha-
summary, the lesion exhibits striking architectural and langeal bone tuft. The peripheral parts of the exostosis
cytologic atypia that may lead to the diagnosis of malig- have ill-defined margins and a hazy density in the soft
nancy if microscopic features are evaluated without tissue, which is the typical presentation of an early lesion.
knowledge of the entire clinicoradiologic presentation. As these lesions mature, they develop calcification and
Despite pronounced cytologic atypia of both osseous and trabecular patterns (compare photographs in Fig. 23-32).
cartilaginous elements, a low power examination dis- In the early stages of development, the lesion consists
closes a peculiar zonal architecture—the bony elements of proliferating fibroblasts in direct continuity with the
are located within the central/basal region, and the car- nail bed, where cartilaginous metaplasia can be seen
tilage forms irregular, caplike structures toward the (Figs. 23-33 and 23-34). The cartilage gradually under-
periphery. In some cases, a more organized, somewhat goes calcification, increases in volume, and develops
parallel arrangement of reactive bone trabeculae can be enchondral ossification. The areas of enchondral ossifica-
seen at low power magnification. tion gradually progress to woven bone and then lamellar
bone. Eventually the bone forms the core, or base, of the
Acquired Osteochondroma lesion and is covered by a thick cartilaginous cap (Figs.
23-33 and 23-34). During evolution of the lesion, the
(Turret Exostosis) continuity between the bony stalk and the underlying
The term turret exostosis was proposed by Wissinger cortex is established. The cartilage cap may show striking
et al.106 in 1966 in reference to reactive lesions on the cellularity with plump nuclei and binucleated and multi-
bone surface of the phalanges that they descriptively des- nucleated chondrocytes. The surrounding proliferating
ignated as ossifying hematoma of the phalanges.83,92 The fibrous tissue can be hypercellular, and it is composed of
clinicoradiologic features of this condition overlap with myofibroblasts. In the initial phase, the proliferation of
the two other reactive lesions of the bone surface cartilage contributes to the exophytic growth of the
described in this section: florid reactive periostitis and lesion. In later phases, an interconnecting network of
bizarre parosteal osteochondromatous proliferation. mature lamellar bone dominates the lesion (Fig. 23-35).
Radiographically, acquired osteochondroma is typi- Limited biopsy samples taken from the periphery of the
cally well delineated and has a well-developed linear min- lesion may create the impression of a malignant cartilage
eralization pattern at the base, which is fused with the tumor. The microscopic features of this condition should
cortex (Fig. 23-29). Microscopically, the lesion consists be evaluated with the clinicopathologic presentation: the
of an ossified core or base attached to the underlying anatomic site and radiographic features of a lesion at the
cortex. The periphery of the lesion is covered by a carti- bone surface. An associated infection can cause a consid-
laginous cap. erably acute and chronic inflammatory infiltrate.
Depending on the level of maturation of osseous and Subungual exostosis is a benign condition with self-
cartilaginous elements, the lesion may exhibit more or limited growth potential. These lesions usually develop
less architectural and cytologic atypia. Typically the zonal during the course of several months, but we have seen
architecture is well developed; that is, it has cartilage at cases in which the lesion had been present for 10 years
the periphery and bone at the base (Fig. 23-30). The before the patient sought help. The growth causes only

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1385

A B

C D
FIGURE 23-26  ■  Bizarre parosteal osteocartilaginous proliferation (Nora’s lesion): radiographic and microscopic features. A and
B, Two examples of bony and cartilaginous exostotic lesions of phalanx and metacarpal bone in a 38-year-old man and a 61-year-
old woman, respectively. C, Photomicrograph of lesion of phalanx shown in A demonstrates island of hyaline cartilage that blends
with woven bone in fibrous background. D, Basophilic bone elsewhere in C. This type of immature bone is found in both florid
reactive periostitis and Nora’s lesion. (C and D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1386 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-27  ■  Bizarre parosteal osteochondromatous proliferation (Nora’s lesion): radiographic and microscopic features. A, Cir-
cumscribed bony and cartilaginous excrescence attached to surface of distal ulnar shaft is shown in this lateral radiograph.
B, Computed tomogram shows exostotic mass composed of bone and cartilage attached to cortical surface of ulna. C, Photomicro-
graph of exostosis shows disorganized pattern of bone and cartilage in fibrous matrix. D, Basophilic bone in fibrous stroma.
(C, ×50; D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1387

B C

FIGURE 23-28  ■  Bizarre parosteal osteochondromatous proliferation (Nora’s lesion): microscopic features. A, Low power photomi-
crograph shows proliferation of disorganized metaplastic cartilage with enchondral ossification. Note that formation of irregular
cartilage cap is evident at periphery (top). B, Higher magnification of A shows atypical features of cartilage cells. C, Higher magni-
fication of A showing hypercellular spindle-cell proliferation and osteoid deposition adjacent to cartilaginous areas. (A, ×100; B and
C, ×200) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1388 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A
FIGURE 23-29  ■  Acquired, posttraumatic exostosis (turret exostosis): radiographic features. A and B, Anteroposterior and lateral
radiographs of third finger of adult who sustained injury to middle phalanx 1 year previously. Note broad-based, well-circumscribed
exostosis and intact underlying cortex. In some cases, this lesion represents end stage of florid reactive periostitis.

local discomfort. Consequently, it is not surprising that a EXUBERANT FRACTURE CALLUS


patient may wait several years before seeking medical
attention. Treatment is usually prompted by superim- The intensity and extent of repair are a complex process
posed infection. The recurrence rate is not well docu- regulated by multiple growth factors and modified by the
mented, but a partial biopsy and incomplete excision severity of injury and underlying pathologic condi-
stimulate further growth in approximately 50% of tions.109,112,114,115 Submitted tissue from fracture callus
patients. Definitive excision is curative. A permanent cure rarely presents a diagnostic problem for a pathologist.
is usually accomplished if complete excision is performed However, occasionally it can be so exuberant that, radio-
on mature, well-established exostoses. The gradual graphically and clinically, it raises the suspicion of a neo-
merging of the fibrocartilage cap with the nail bed, most plastic process. The development of an exuberant callus
evident in early lesions, makes it impossible to develop a typically follows major trauma with or without fracture
cleavage plane. Therefore if excision is attempted before (Fig. 23-36). Some rare conditions, such as osteogenesis
the lesion is mature, the overlying nail bed must be imperfecta and osteopetrosis, predispose to the develop-
removed. Although the deformity of the nail after this ment of exuberant fracture callus even after minor
procedure rarely presents a problem in the toe, the trauma.110,111,113
patient whose lesion is in a finger may require a nail bed Typical fracture callus shows a gradual transition from
graft. The best cosmetic results are accomplished if the hypercellular areas containing spindle cells with early
lesion becomes a purely osseous structure and cleavage osteoid formation of an interconnecting network of
planes are more easily identified. reactive bone with prominent osteoblastic rimming

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1389

B
FIGURE 23-30  ■  Acquired exostosis: microscopic features. A, Note well-organized ossification pattern, which is linear and forms
interconnecting trabeculae (lower aspect of photograph). Peripheral parts are covered by cartilaginous cap. B, Higher magnification
of A shows hypercellularity of cartilaginous cap. (A, ×50; B, ×100) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1390 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

C
FIGURE 23-31  ■  Subungual exostosis: radiographic features. A and B, Anteroposterior and lateral radiographs of great toe show very
large mature bony excrescence attached to dorsomedial surface of distal phalanx by narrow base. C, Clinical photograph of fungat-
ing, ulcerated mass arising in nail bed of 11-year-old boy 5 months after treatment for paronychia. Excision revealed subungual
exostosis (same case as in A and B).

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1391

A B

C D
FIGURE 23-32  ■  Subungual exostosis: radiographic features. A, Anteroposterior radiograph of foot shows ill-defined exostotic bony
mass on medial aspect of distal phalanx of great toe with swelling of surrounding soft tissue. B, More mature subungual exostosis
of distal phalanx of great toe. Mass is attached to phalangeal cortex, and its periphery is sharply defined. C, Lateral view of mature
subungual exostosis shown in B. D, Lateral view of great toe with maturing subungual exostosis.

(Figs. 23-37 and 23-38). In more chronic proliferation, a bone-forming neoplastic process. They typically occur
such as that initiated by prolonged repeated trauma (i.e., in the parosteal soft tissue without an evident frac-
unrecognized malunited fracture), disorganized fibrocar- ture, presenting on radiographs as a mass on the bone
tilaginous proliferation can be a dominant feature of surface or in the soft tissue (Fig. 23-40). A fracture
the process (Fig. 23-39). More mature lesions show pro- superimposed on osteopetrosis can result in the forma-
gressive maturation to lamellar bone. The peculiar dis- tion of an exuberant callus with abundant cartilage that
tinctive architecture of a fracture callus is best evaluated exhibits highly atypical features. Microscopically, these
under low power magnification. The continuity among reactions show a fracture callus that lacks maturation
various microscopic elements of callus tissue is the most patterns into lamellar bone and shows the disorga-
helpful microscopic feature in distinguishing this reactive nized florid formation of metaplastic cartilage and early
process from a neoplasm. osteoid (Figs. 23-40 to 23-42). These lesions may contain
Osteogenesis imperfecta is a rare condition that predis- areas of young granulation or fibroblastic tissue with
poses a bone to fracture. Moreover, patients with this hemorrhage and an occasional giant-cell reaction. Meta-
disorder are particularly prone to the development of plastic cartilage with irregular zones of enchondral ossi-
florid exuberant callus tissue in response to even mild fication is present. These zones show continuity with
trauma; this evolution may not be evident clinically areas of reactive bone formation rimmed by palisading
or may not be documented radiographically. Radio­ osteoblasts.
graphically and clinically, these reactions can simulate Text continued on p. 1402

ERRNVPHGLFRVRUJ
1392 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-33  ■  Subungual exostosis: microscopic features. A, Low power photomicrograph shows cellular chondroid tissue merging
with peripheral zone of proliferating fibrous tissue in nail bed. Cartilage matrix calcification and early stages of enchondral ossifica-
tion are seen below. Inset, Higher magnification of cartilaginous cap showing hypercellularity and atypia of cartilage cells. B, Higher
magnification of an area of A below the cartilaginous cap showing early phases of enchondral ossification and hypercellular fibro-
blastic stroma. (A, ×25; Inset, ×50; B, ×100) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1393

B
FIGURE 23-34  ■  Subungual exostosis: microscopic features. A, Low power photomicrograph shows metaplastic cartilage cap overly-
ing zone of enchondral ossification. Inset, Higher magnification of cartilaginous cap with hypercellularity and atypia. B, Somewhat
disorderly zone of enchondral ossification is evolving into subungual exostosis. (A and B, ×100; Inset, ×200) (A, B, and Inset,
hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1394 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-35  ■  Subungual exostosis: microscopic features. A, Low power photomicrograph of late stage in development of subun-
gual exostosis shows it projecting into nail bed (below). The cartilage cap has been almost completely replaced, through enchondral
ossification, to form bony mass with lamellar maturation. B, Higher power magnification of A shows trabecular bone with lamellar
architecture and almost complete replacement of cartilaginous cap. The bone blends with fibrous tissue of nail bed. Inset, Higher
magnification shows irregular trabecular bone with osteoblastic rimming blending with fibrous tissue of nail bed. (A, ×15; B, ×25;
Inset, ×100) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1395

B
FIGURE 23-36  ■  Exuberant fracture callus: radiographic features. A and B, Anteroposterior and lateral radiographs of foot in young
man with metatarsal shaft fracture exhibiting excessive callus formation that can be mistaken for bone-forming tumor.

ERRNVPHGLFRVRUJ
1396 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-37  ■  Hyperplastic fracture callus: microscopic features. A and B, Low power views of abandoned callus growth in unrec-
ognized fracture composed of well-organized woven and lamellar bone trabeculae. C, Higher magnification of abandoned callus
growth showing well-developed trabecular bone formation arranged in plexiform pattern with zoning. D, Higher magnification of
fracture callus composed of mixture of hyaline cartilage and both woven and lamellar bone trabeculae. (A and B, ×15; C and D, ×20.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1397

B
FIGURE 23-38  ■  Fracture callus: microscopic features. A and B, Anastomosing weblike pattern of woven bone trabeculae with promi-
nent osteoblastic rimming in fracture callus. Inset, Anastomosing woven bone trabeculae rimmed by prominent osteoblastic cells.
(A and B, ×100; Inset, ×200.) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1398 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-39  ■  Hyperplastic fracture callus: microscopic features. A, Low power view of abundant callus growth in unrecognized
fracture of rib; growth is composed of mixture of hyaline cartilage and both woven and lamellar bone trabeculae. Note that trabeculae
are arranged in plexiform pattern with zoning. B, Chondroid and chondromyxoid areas in long-term fracture callus.
C, Juxtaposed hyaline cartilage and trabecular bone in long-term persistent fracture callus. D, Granulation tissue and relatively
mature bone trabeculae with osteoclastic remodeling. (A, ×10; B and D, ×50; C, ×200) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1399

A B C

D
FIGURE 23-40  ■  Exuberant hyperplastic callus in osteogenesis imperfecta: radiographic features. A, Fluffy mineralization is noted
juxtacortically at midshaft of femur (arrows) in a 13-year-old boy known to have osteogenesis imperfecta. He had history of multiple
long bone fractures since age 2. No definite history of trauma could be obtained, and fracture line is very indistinct. B, Circumfer-
ential new bone formation involving midshaft (arrows) of a 4-year-old child who has osteogenesis imperfecta and history of several
fractures sustained after minor injuries. No recent history of trauma could be obtained. Note bony deformation and thickening of
posterior cortex. C, Mass on medial aspect of surface of right femoral shaft in child with osteogenesis imperfecta. There is no
radiographic evidence of fracture, and no recent history of significant trauma could be obtained. D, Microscopic features of florid
fracture callus with early osteoid and metaplastic cartilage matrix deposition of the lesion shown in C (200; hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1400 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-41  ■  Exuberant fracture callus in osteogenesis imperfecta: microscopic features. A, Low power view of periosteal mass
in soft tissue shows florid fracture callus with extensive cartilage metaplasia and early osteoid formation. B, Area of metaplastic
cartilage with nuclear atypia. Inset, Early deposition of osteoid. (A, ×25; B and Inset, ×100) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1401

B
FIGURE 23-42  ■  Exuberant fracture callus in osteogenesis imperfecta: microscopic features. A, Low power view showing florid frac-
ture callus with extensive areas of cartilage metaplasia. B, Area of metaplastic cartilage with hypercelluarity and nuclear atypia.
Inset, Early osteoid deposition in florid callus. (A, ×25; B and Inset, ×100) (A, B, and Inset, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1402 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

AVULSION AND STRESS INJURIES of previously unrecognized acute episodes. Chronic avul-
sions are characterized by the formation of callus and
Avulsion and stress injuries are caused by distinct trau- organized bone in relation to a separated osseous frag-
matic episodes that may synergistically contribute to the ment. The bone prominences in the lower extremities,
development of a single lesion. Microscopically, these particularly around the pelvis and hip, are particularly
conditions are characterized by the presence of a fracture susceptible to avulsion injuries.120,122,125,136,137,147,156 The
callus in different phases of formation. They can be sepa- most frequent sites of involvement include the ischial
rated into specific clinicoradiologic entities if clinical apophysis, the anterior/inferior iliac spine, and the lesser
history (type of injury or physical activity involved) and and greater trochanters of the femur. In the upper
radiologic features are considered. Occasionally, these extremities the insertion of the deltoid muscle tendon
conditions clinically and radiographically simulate a neo- (lateral aspect of proximal humeral shaft) is the most
plasm and may require a biopsy to rule out a malignant typical site of an avulsion injury.125,152 The chronic
process. The most typical sites of avulsion and stress variant of this injury may clinically and radiographically
injuries, as well as their peak age incidence, are shown in raise the suspicion of a neoplasm, prompting biopsy and
Figure 23-43. microscopic evaluation (Fig. 23-44). Lesions involving
areas of ligament insertion can predispose to avulsion
injuries.127,152
Avulsion Injuries Microscopically, the lesion shows fracture callus in
Avulsion injuries are caused by a traumatic episode of different phases of formation. Long-standing lesions may
muscle spasm or traction causing the detachment of the show well-developed woven or lamellar bone, but more
muscle’s tendinous attachment.120,122,125,126,136,139,140,150,155 typically, repeated stress stimulates florid proliferations
Acute avulsion is recognized by the displacement of a of fibroblastic and granulation tissue with prominent for-
bone that frequently corresponds to the site of insertion. mation of reactive woven bone and cartilage metaplasia;
Acute injuries are typically not subjected to microscopic that is, features overlap with those seen in the tissue
evaluation. More insidious or chronic lesions are associ- of bizarre parosteal osteochondromatous proliferation
ated with repeated stress or represent the healing phase (Figs. 23-45 and 23-46).

Avulsion
injury
Incidence

Avulsion
injuries

1 2 3 4 5 6 7 8
Stress
Age in decades injuries

FIGURE 23-43  ■  Avulsive and stress injuries. Peak age incidence and typical sites of skeletal involvement are indicated by arrows.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1403

FIGURE 23-44  ■  Avulsive injury to ischial apophysis: radiographic features. Note irregularity of ischial apophysis in teenage boy with
groin pain and stiffness (arrows).

Cortical Irregularity Syndrome the second or third metatarsal neck region, which com-
(Periosteal Desmoid) monly affects soldiers in training, is popularly referred to
as march fracture.116,127,131,143,145 Long-distance running
In the past the term periosteal desmoid has been inappro- (jogging) typically causes stress fractures involving the
priately applied to this avulsive lesion, which involves distal fibula and the tibial shaft.122,143 Underlying skeletal
the insertion of the adductor muscle tendon on the disorders, such as osteoporosis, can predispose to fatigue
posteromedial aspect of the distal end of the femur (stress) fractures even in relation to normal activities.142,145
(Fig. 23-47).117,124,132,134,135,148,149,153,157 Cortical irregularity On radiographs, the lesion may show a transverse frac-
syndrome especially affects young, athletic individuals, ture or sclerosis with associated periosteal reaction (Fig.
predominantly boys, before adolescence. Bony irregulari- 23-49). The actual fracture may not be revealed by
ties and excavations in relation to the medial side of the computed tomography or even magnetic resonance
distal end of the femur are common and characteristic of imaging.151,154 The so-called gray cortex represents focal
this lesion.76,81,101-103,110,111,146 In rare circumstances, this density and can be an early radiographic sign of stress
lesion, as it appears on radiographs, can raise suspicion fracture.141 In such cases, the lesion presents only as peri-
of a neoplastic disorder.107 In such cases, biopsy samples osteal new bone formation on radiographs. Some of these
shows young, fibroblastic tissue without cytologic atypia lesions may raise the suspicion of a neoplasm on the bone
(Fig. 23-48). The presence of thick collagen fibers surface, and biopsy should be performed. Microscopic
with parallel arrangement consistent with tendinous or examination reveals fracture callus in various phases of
ligamentous tissue can be seen. If the microscopic fea- development.
tures are evaluated without clinicoradiologic correlation,
the lesion may be confused with fibromatosis or even
fibrosarcoma. PUBIC OSTEOLYSIS
Pubic osteolysis is a rare lesion characterized by a
Stress Fractures rapidly progressive osteolysis in and around the pubis.
Stress fractures are traumatic lesions that are the result The condition usually affects middle-aged to elderly
of prolonged minor, repeated traumas usually associated osteopenic women. The major diagnostic significance
with athletic or occupational activities.116,119,127-130,133,144 of pubic osteolysis is that it may have radiographic and
These lesions usually occur in young, athletically active microscopic features that overlap with those of chon-
adults. There also is some association between this type of drosarcoma.158,161,164,165,167,170 Our consultation practice
injury and osteoporosis in elderly patients. The repeated indicates that most cases of pubic osteolysis are submit-
stress finally culminates in an acute fracture of the bone. ted for second opinion with the preliminary diagnosis of
Stress fractures usually involve the bones of the lower chondrosarcoma.118
extremities.121,123,129,130,133,138 The frequent involvement of Text continued on p. 1409

ERRNVPHGLFRVRUJ
1404 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-45  ■  Chronic avulsive injury: microscopic features. A and B, Low power magnification of atypical pattern of callus seen
in longstanding or persistent avulsive injury consisting of alternating zones of metaplastic hyaline cartilage and woven bone tra-
beculae. C, Prominent hyaline cartilage differentiation in fracture callus with focal conversion to bone. D, Anastomosing pattern of
woven bone trabeculae in fracture callus (A-D, ×25) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1405

A B

C D
FIGURE 23-46  ■  Chronic avulsive injury: microscopic features. A, Anastomosing weblike pattern of woven bone trabeculae in fracture
callus. B, Chondroid and chondromyxoid areas juxtaposed on woven bone in long-term fracture callus. C, Chondroid areas with
disorganized enchondral ossification pattern in persistent fracture callus. D, Juxtaposed hyaline cartilage and trabecular bone in
long-term persistent fracture callus. (A, C, and D, ×25; B, ×50) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1406 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

C D
FIGURE 23-47  ■  Cortical irregularity syndrome: radiographic features. A, Lateral radiograph of knee shows roughening of supracon-
dylar ridge and overlying shadow of soft tissue, which represents chronic avulsive lesion of insertion of adductor magnus
muscle (arrows). B, Computed tomogram shows concave erosion of linea aspera, into which adductor magnus tendon inserts
(arrow). C, Photomicrograph showing proliferation of myofibroblastic cells without cytologic atypia. D, Proliferation of
fibroblast-like spindle cells in fibrous stroma resembling fibromitosis frequently seen in this reactive process. (C and D, ×25) (C and
D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1407

B C
FIGURE 23-48  ■  Cortical irregularity syndrome: radiographic features. A-C, Proliferation of myofibroblastic cells without cytologic
atypia in dense fibrous stroma resembling fibromatosis (A-C, ×100) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1408 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B
FIGURE 23-49  ■  Stress fracture: radiographic features. A and B, Anteroposterior and lateral radiographs of tibia in child show posterior
cortical disruption in upper third of shaft, with periosteal new bone formation and surrounding bone sclerosis. Such injuries can
mimic osteoid osteoma or even osteosarcoma.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1409

The exact pathogenesis of this condition is not known. of this condition involving the temporomandibular joint
It is generally postulated that trauma and osteopenia are also have been described.209 Bilateral symmetric involve-
the two major predisposing factors.159,160,162,163,168,169,171 ment of corresponding anatomic regions is frequent. The
Similar lesions have been described in athletes and in peak age incidence and the most frequently affected sites
association with urologic surgery, renal failure, and are shown in Figure 23-52. Familial occurrences have also
obesity.162,166,169,170 The presence of radiographically doc- been reported. Lesions have been described in siblings
umented pathologic fracture was evident in only one of and in several generations of one family. An autosomal
our cases. dominant pattern of inheritance with variable clinical
On radiographs the lesion represents a lysis of the expressivity is postulated in familial cases.199 Germline
bone in the pubic area (Fig. 23-50). Development of mutations of several genes, such as fibroblast growth factor
fracture, fragmentation, or both frequently occur later in 23 (FGF23), UDP-N-acetyl-alpha-d-galactosamine: poly-
the course of the disease. An area of lysis is usually sur- peptide N-acetylgalactosaminyl transferase 3 (GALNT3),
rounded by swelling of the adjacent soft tissue. Virtually sterile alpha motif domain containing 9 (SAMD9), and
all lesions eventually develop calcification (sclerosis) klotho (KL), have been identified in familial forms of
within lytic areas of the affected bone and in the adjacent tumoral calcinosis.175-182,186-189,196,197,213,216,218 Some of these
soft tissue. familial syndromes (e.g., associated with mutations of
Microscopically, the lesion shows ineffective and exu- GALNT3) are clinically presenting with widespread
berant bony callus. Prominent areas of metaplastic carti- ectopic calcifications and severe phosphatemia.175,179
lage are always present and may dominate the lesion
(Fig. 23-51). Occasional binucleate chondrocytes, a high
Clinical Symptoms
degree of cellularity, and myxoid changes are seen within
cartilaginous areas. Bone metaplasia is also frequently The mass is typically asymptomatic. It may cause local
present with the production of abundant immature woven discomfort such as tenderness or pain. Laboratory data
bone. The bone metaplasia is identical to that seen in show no evidence of abnormal calcium levels. However,
lesions of exuberant callus formation secondary to frac- many patients may have mild to moderate hyperphospha-
ture healing or in active myositis ossificans. The risk of temia.169,198,199,216 Calcitriol (1, 25-dihydroxyvitamin D3)
misdiagnosis is compounded if this fracture callus extends levels may also be elevated.200,207,211 Serum uric acid and
into soft tissue. The osteocartilaginous material may also alkaline phosphatase levels are normal.
show focal bone necrosis, fibrosis of the marrow, and
proliferation of capillary vessels with hemosiderin depos- Radiographic Imaging
its. The angiomatoid pattern of granulation tissue might
suggest angioma, but this is typically seen in fracture Plain radiographs reveal conglomerates of well-
healing. Neuropathic change is suggested by the occa- demarcated opacities in soft tissue near a major joint
sional presence of fragments of necrotic bone and carti- (Fig. 23-53, A and B).170,173,194,195,210,217 Occasionally, fluid
lage debris that result from fragmentation of the fracture levels can be documented on plain radiographs.194 They
site. There is loosely textured edematous chondroid and can usually be seen in larger, long-lasting lesions. The
fibrous intervening tissue with vascular proliferation and foci of calcification range in size from several millimeters
a granulation-like tissue appearance. Inflammatory cells to 1 to 2 cm. Computed tomography or magnetic reso-
are typically not present; thus confusion with osteitis nance imaging shows multifocal lesions. In some of the
pubis does not arise. larger calcified nodules, cystic changes with fluid levels
can be documented using these techniques.
TUMORAL CALCINOSIS
Gross Findings
Definition
Tumoral calcinosis consists of an encapsulated firm mass
Tumoral calcinosis is a distinct clinicopathologic entity of multiple calcified nodules that focally coalesce into
characterized by a tumorlike deposition of calcium larger, lobulated masses. Cystic changes are frequently
without an obvious associated metabolic calcium disorder. present in some of the nodules. The nodules are filled
with yellow-gray, pasty masses or a milky fluid that can
Incidence and Location be washed out from individual lesions.

Tumoral calcinosis is very rare and seems to affect Afri-


Microscopic Findings
cans and African-Americans predominantly, but it can
also affect other ethnic groups.129,171,183,185,190,193,201,205,206,214 Microscopically, the lesion consists of large areas of amor-
It typically occurs in otherwise healthy children, adoles- phous or granular calcium masses with occasional inter-
cents, and young adults.202 The lesions are often multiple, spersed, concentrically laminated calcospherites172,185,215
and they may affect two or more siblings in the same (Fig. 23-53, C and D). Chemical and ultrastructural
family. Calcifications are usually located in the periarticu- analysis of this substance reveals calcium crystals con-
lar soft tissue in the vicinity of major joints, such as the sistent with hydroxyapatite.192,208 The active phase of
hips, shoulders, and posterior elbows. Occasionally, they the disorder is associated with a prominent infiltration
may affect the knee and spine regions.108 Tumoral calci- by macrophages, osteoclast-like giant cells, and other
nosis is extremely rare in the hands and feet. Examples inflammatory cells that border and encircle calcium

ERRNVPHGLFRVRUJ
1410 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

B
FIGURE 23-50  ■  Pubic osteolysis: radiographic features. A, Lysis of bones in pubic area and widening of pubic symphysis. B, Lysis
of pubic bone with fracture and fragmentation.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1411

A B

C D
FIGURE 23-51  ■  Pubic osteolysis: microscopic features. A and B, Proliferation of spindle cells with prominent vasculature. C, Reactive
bone with osteoblastic rimming. D, Metaplastic cartilage with hypercellularity and nuclear atypia. (A and C, ×50; B, ×100; D, ×200.)
(A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1412 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

Periarticular
soft tissue
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 23-52  ■  Tumoral calcinosis. Peak age incidence and most frequent sites of skeletal involvement are indicated by solid black
arrows.

deposits. The inactive or mature phase merely shows antacids have been used and offer an alternative mode of
calcified deposits in a dense fibrous stroma. conservative therapy for lesions that cannot be surgically
removed for technical reasons.204 Other modes of non-
surgical therapy (radiotherapy, cortisone, low-phosphorus
Differential Diagnosis
diet, aluminum hydroxide gels) have been shown to be
Similar tumoral calcifications can be found in patients ineffective.204,212
with several underlying metabolic calcium disorders,
such as chronic renal disease or secondary hyperparathy-
roidism.174,184,203 The patients with such conditions are TOPHACEOUS PSEUDOGOUT
typically older, have a history of an underlying disorder,
or exhibit features of generalized metabolic calcium dis- Definition
order. In addition, they show disseminated calcification
in visceral organs. Lesions similar to tumoral calcinosis Tophaceous pseudogout (tumoral calcium pyrophosphate
can occur in patients with primary hyperparathyroidism, dihydrate crystal deposition disease) was first described in
hypervitaminosis D, milk-alkali syndrome, and sclero- 1962 by McCarty et al.226 The crystals were identified in
derma.191 However, these lesions are typically associated the synovial fluid of patients who had goutlike symptoms
with abnormal serum levels of calcium. without sodium urate crystals. Consequently the entity
was designated as pseudogout. The terms chondrocalcinosis
and calcium pyrophosphate crystal deposition arthropathy were
Treatment and Behavior
independently applied to the same condition, which is
Surgical removal of the lesion is the treatment of choice. characterized on radiographs by prominent multifocal
Incompletely excised lesions recur. Long-standing lesions calcification in the cartilages of joints and intervertebral
may develop secondary infection with abscess formation. disks.225,228 The condition is now designated as calcium
It is recommended that lesions be surgically removed as pyrophosphate dihydrate crystal deposition disease. The term
early as possible, when they are small and amenable to encompasses pseudogout, chondrocalcinosis, and pyro-
complete excision. Calcium deprivation and PO4-binding phosphate arthropathy.219,220,222-225,227

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1413

C D
FIGURE 23-53  ■  Tumoral calcinosis: radiographic and microscopic features. A, Lateral radiograph of elbow shows coarse deposits of
calcific material in region of olecranon bursa. B, Massive calcinosis in soft tissue around parascapular area and axilla in teenage
boy. C, Low power photomicrograph shows circumscribed foci of calcific material with central cystification and peripheral foreign-
body giant-cell reaction in tumoral calcinosis. D, Higher power magnification shows calcified bodies and more finely dispersed,
noncrystalline calcifications. (C, ×50; D, ×25) (C-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1414 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

Calcium pyrophosphate dihydrate deposition com- developed in another patient. One patient was asymp-
monly occurs in the articular and periarticular tissue. tomatic, and the lesion was discovered incidentally on
However, tophaceous calcium pyrophosphate dihydrate imaging studies done for other reasons. No patient’s
deposition (i.e., tumoral or massive calcium pyrophos- serum tests showed an abnormal level of calcium or phos-
phate dihydrate crystal deposition) is very rare.221,229,230 phate. None of the patients in our series had radiographic
Because of the unusual location and prominent cartilage evidence of chondrocalcinosis or clinical symptoms in
formation, the tumorlike masses in calcium pyrophos- any other joints. The peak age incidence and sites of
phate dihydrate crystal deposition disease may be misin- skeletal involvement of our consultation cases are shown
terpreted as a benign or even a malignant cartilage in Figure 23-54.
neoplasm, especially chondrosarcoma.
Radiographic Imaging
Clinical Data
Radiographically, the lesion appears to be a slightly or
We have seen seven cases of tophaceous pseudogout in densely calcified mass; the pattern of calcification is gran-
consultation. The ages of the patients ranged from 31 to ular or fluffy (Figs. 23-55 and 23-56). Unlike myositis
86 years (average, 60.7 years). One patient was a man, ossificans, peripheral density is not observed. The bone
and six were women. The temporomandibular joint was adjacent to the calcified mass shows pressure erosion in
involved in three patients, and the metatarsophalangeal some cases. The calcification itself and its pattern are
joint of the great toe was involved in two patients. The demonstrated well on computed tomograms and mag-
hip joint and cervical spine were involved in one patient netic resonance images (Fig. 23-57).
each. Long-standing swelling of the affected area was
noted in two patients with temporomandibular joint
Gross Findings
involvement. Two patients had a painful mass of the great
toe, which was clinically considered to be gouty tophus. The lesion is a circumscribed, white-gray mass with a
One patient noticed increasing hip pain over 18 months. more or less chalky appearance. The sizes of the lesions
Rapid, progressive myelopathy at an upper cervical level in our series ranged from 2 to 4 cm.
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 23-54  ■  Tophaceous pseudogout (tumoral calcium pyrophosphate dihydrate crystal deposition disease). Peak age incidence
and most frequent sites of skeletal invovlement.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1415

pyrophosphate dihydrate crystals (Fig. 23-58). The cel-


lular tissues contain large numbers of histiocytes and
foreign body–type giant cells surrounding the calcified
areas and representing a foreign body granulomatous
reaction, whereas the less cellular tissues consist of meta-
plastic chondroid tissues. The chondroid cells can be
atypical, with variable nuclear size and shape, and can
mimic a malignant cartilage tumor. The chondroid
cells can be seen in and around the calcified areas (see
Fig. 23-58).
The calcium pyrophosphate dihydrate crystals are
removable by decalcification, and their basophilic fea-
tures are lost in demineralized sections. Moreover, in
decalcified sections, the amorphous intercellular matrix,
which is somewhat similar to the chondroid matrix, is left
behind in the areas of calcium pyrophosphate dihydrate
deposition (Fig. 23-59).

Differential Diagnosis
The metaplastic chondrocytes found in our series of
patients sometimes showed cytologic atypia (three
patients) analogous to that seen in some patients with
synovial chondromatosis and superficially resembled
chondrosarcoma. Particularly in decalcified sections from
A which calcium pyrophosphate dihydrate crystals are lost,
atypical features in metaplastic chondrocytes and the
presence of myxoid stroma may lead to the histologic
misdiagnosis of chondrosarcoma. Grossly, tophaceous
pseudogout is a calcified, chalky mass unlike the calcifica-
tion seen in chondrosarcoma. A foreign body–type gran-
ulomatous reaction containing histiocytes and giant cells
is a helpful finding to distinguish it from chondrosar-
coma. More important, however, is the identification by
polarized light microscopy of birefringent crystals char-
acteristic of calcium pyrophosphate dihydrate. The exact
nature of these crystals can be determined by radio-
graphic diffraction or electron probe analysis. Even in the
decalcified sections, empty outlines of crystals in the
chondroid matrix can be seen, although birefringent
crystals are not identified directly.
B Synovial chondromatosis and chondromas in soft
tissue are the other lesions that can be confused with
FIGURE 23-55  ■  Calcium pyrophosphate dihydrate deposition tophaceous pseudogout, especially in the conditions in
disease (tophaceous pseudogout): radiographic features. which a heavily calcified cartilage matrix may obscure the
A, Radiograph of right hip shows calcified mass in soft tissue
adjacent to acetabular margin; this mass represents deposits of
cartilaginous nature. However, the calcified areas of
calcium pyrophosphate dihydrate in capsule of hip joint synovial chondromatosis do not contain the rhomboid
(arrows). B, Computed tomogram of case shown in A demon- crystals characteristic of calcium pyrophosphate dihy-
strates circumscribed calcification in soft tissue that is adjacent drate deposition. Indeed, radiographic diffraction studies
to acetabular labium (arrows). (From Ishida T, Dorfman HD, in both synovial chondromatosis and chondroma of
Bullough PG: Hum Pathol 26:587–593, 1995.)
soft tissue show that the mineral deposits represent
hydroxyapatite.

Microscopic Findings Treatment and Behavior


Histologically, the lesion consists of cellular or less Tophaceous pseudogout may recur after complete or
cellular tissues containing small to large islands of incomplete surgical excision. Six cases that recurred 11
intensely basophilic calcified crystalline material (Fig. months to 6 years after surgery have been reported.
23-58). The crystals are short rhomboid or, rarely, Three recurrences were noted in one patient. One of the
needle shaped, similar to urate crystals. Under compen- patients in our series had two recurrences during a
sated polarized light microscopy, the crystals exhibit the 20-year span.
weak positive birefringence characteristic of calcium Text continued on p. 1420

ERRNVPHGLFRVRUJ
1416 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

FIGURE 23-56  ■  Calcium pyrophosphate dihydrate deposition disease (tophaceous pseudogout): radiographic features. A and
B, Anteroposterior and lateral radiographs of foot show massive calcification of soft tissue around metatarsophalangeal joint of
great toe resulting from localized deposit of calcium pyrophosphate dihydrate crystals. (From Ishida T, Dorfman HD, Bullough PG:
Hum Pathol 26:587–593, 1995.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1417

B
FIGURE 23-57  ■  Calcium pyrophosphate deposition disease (tophaceous pseudogout): radiographic features. A, Sagittal magnetic
resonance image (MRI) of skull and cervical spine of elderly woman who had calcified extradural mass at junction of C1 and C2
with posterior displacement and kinking of dural sac and cord (arrow). B, Computed tomogram of cervical spine of patient shown
in A reveals extradural calcific mass compressing dural sac (arrows). Inset, T2-weighted axial MRI of skull of 55-year-old woman
shows tumorous mass of infratemporal fossa adjacent to temporomandibular joint on left side of photograph. (From Ishida T, Dorfman
HD, Bullough PG: Hum Pathol 26:587–593, 1995.)

ERRNVPHGLFRVRUJ
1418 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-58  ■  Calcium pyrophosphate dihydrate deposition disease (tophaceous pseudogout): gross and microscopic features.
A, Calcified tophaceous deposit from plantar aspect of foot shows whitish chalky appearance similar to that seen in monosodium
urate tophus. B, Metaplastic chondroid change surrounds islands of basophilic calcified crystalline calcium pyrophosphate dihydrate.
C, Some basophilic calcified deposits provoke foreign body–type, giant-cell reaction without chondroid metaplasia. D, Compensated
polarization microscopy demonstrates positive birefringence of rhomboid calcium pyrophosphate dihydrate crystals. (B and C, ×100;
D, ×200) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1419

A B

C D
FIGURE 23-59  ■  Tumoral calcium pyrophosphate dihydrate crystal deposition disease (tophaceous pseudogout): radiographic and
microscopic features. A, Computed tomogram (CT) of the skull showing a calcified soft tissue mass in the area of the right tem-
poromandibular joint. B, 3-D CT reconstruction image showing a mass in the right temporomandibular joint. C, Photomicrograph
of decalcified specimen showing lobules of myxoid material corresponding to areas of metaplastic cartilage and crystalline material
deposits. D, Higher magnification of C showing an interface between hypercellular stroma and myxoid areas with prominent plump
histiocytic cells exhibiting nuclear atypia. (C, ×100; D, ×200) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1420 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

TOPHACEOUS GOUT of acute attacks increase in duration and become more


frequent. Approximately 50% of the patients with gouty
A comprehensive description of the various forms of gout arthritis have nodular deposition of monosodium urate
and their pathogenesis is beyond the scope of this book. crystals that are referred to as tophi. Patients who have
This description is restricted to the tophaceous form higher levels of uric acid have a high propensity for the
of gout and the basic microscopic features that are development of tophi. The peak age incidence and the
helpful in distinguishing this metabolic disorder from a most frequent sites of skeletal involvement are shown in
neoplasm. Figure 23-60.

Clinical Data Radiographic Imaging


In general, gout is characterized by hyperuricemia and The formation of tophi occurs 3 to 40 years (average, 12
the deposition of monosodium urate crystals in synovial years) after the first attack of gout. Radiographically,
tissues.232,233,235,236 tophaceous gout typically presents as a periarticular non-
Nearly 90% of cases of gout occur in men, and the calcified mass with frequent erosion of the adjacent bones
peak incidence is in patients older than age 50 years. It (Figs. 23-61 and 23-62). Early lesions present as ill-
is rare in women, but when it does affect a female patient, defined swelling in the soft tissue. Approximately 20% to
the onset of symptoms typically occurs in the postmeno- 30% of tophi show calcifications. Occasionally, topha-
pausal period. Many patients with hyperuricemia are ceous gout can produce large calcified periarticular
asymptomatic. About 20% have renal lithiasis or episodes masses (Fig. 23-63).231,234,237,238
of acute gouty arthritis. Acute gout can present as a
monoarticular or oligoarticular arthritis. The disease
Gross Findings
usually involves the joints of the foot and ankle. The knee
joint is also frequently involved. The involvement of the Tophaceous deposits in gout appear as white to yellow
small joints of the hands is next in frequency. The periods chalky nodular areas surrounded by an intervening
Incidence

1 2 3 4 5 6 7 8
Age in decades

FIGURE 23-60  ■  Tophaceous gout. Peak age incidence and typical sites of skeletal involvement. Solid black arrows indicate most
frequent sites of involvement.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1421

tion (Fig. 23-65). The intervening stromal tissue shows


histiocytic and multinucleated giant-cell reaction.

CHRONIC RECURRENT
MULTIFOCAL OSTEOMYELITIS
Chronic recurrent multifocal osteomyelitis is a more
recently described rare variant of osteomyelitis that pri-
marily affects children and young adults. Clinically, it is
characterized by an insidious onset of fever, local swell-
ing, and pain in the affected bones.239,243,252-261 The disease
mainly affects the metaphyses of the long tubular bones
and the clavicle. Sometimes, it has a symmetric distribu-
tion; that is, similar sites in both extremities are involved
(Fig. 23-66).245,246 The most commonly involved skeletal
sites are the tibia, femur, clavicle, and fibula. On radio-
graphs the lesion presents as lytic foci with associated
sclerosis (Figs. 23-66 to 23-68).245,249,253,256-258
The presence of associated autoimmune disorders
and familial clustering with linkage to a predisposing
locus at 18q21-22 suggests an autoimmune etiology
and genetic predisposition.248 In fact, mutations of the
LPIN2 gene cause a familial form of chronic recurrent
multifocal osteomyelitis known as Majeed syndrome
while mutations in Pstpip2 cause a murine form of the
disorder.244,251 The roles of LPIN2 and of the human
homolog of Pstpip2 (PSTPIP2) in the etiology of chronic
recurrent multifocal osteomyelitis are uncertain at the
time of this writing.
Microscopically, early lesions contain polymorpho-
nuclear (neutrophilic) leukocytic infiltrate in the
marrow.241 Collections of neutrophils that mimic bone
marrow abscesses can be present and are sometimes sur-
rounded by a collar of lymphocytic infiltrates. Bone
resorption with prominent osteoclasts is a common
feature. Long-standing lesions show fibrosis with pre-
dominantly lymphocytic infiltrates (Fig. 23-67, D, and
Fig. 23-68, D). Prominent formation of new reactive
bone can be a dominant feature in later phases of the
disease. Peculiar granulomas made up of collections
FIGURE 23-61  ■  Tophaceous gout: radiographic features. Mar-
ginal erosions of tarsal bones and metatarsals are sharply out- of neutrophils surrounded by a rim of epithelioid histio-
lined and completely radiolucent (arrows). cytes can be present, but there is no caseous necrosis.
Results of bacterial, viral, and fungal cultures, as well as
special stains for infectious organisms, are consistently
negative.
fibrous tissue. Smaller lesions predominantly show Some patients may have associated recurrent skin
deposits of urate crystals. Larger, older lesions may lesions (pustolosis palmoplantaris).239,240 Typically, the
exhibit multilobular architecture with prominent fibrosis emergence of skin changes parallels exacerbations of the
and calcifications (Fig. 23-64). skeletal lesions. In fact, it is postulated that chronic recur-
rent multifocal osteomyelitis is a manifestation of the
spectrum of disorders characterized by sterile neutro-
Microscopic Findings
philic infiltrate primarily in the skin referred to as neu-
Microscopically, monosodium urate crystals are needle trophilic dermatoses. They include palmoplantar pustulosis,
shaped and demonstrate strongly negative birefringence psoriasis, acne fulminans, neutrophilic eccrine hidradeni-
under compensated polarization microscopy. They are tis, acute febrile netrophilic dermatosis, and pyoderma
found floating freely and intracellularly in leukocytes. gangrenosum. The frequent association of chronic recur-
Monosodium urate crystals are also found in tophi, where rent multifocal osteomyelitis with synovitis, acne, pustu-
they form multicentric, radially oriented deposits sur- losis, hyperostosis, and osteitis has been defined as a new
rounded by a matrix of amorphous material, lipids, and (SAPHO) syndrome.262
proteinaceous debris. They show a multilobular or In most cases the erythrocyte sedimentation rate is
nodular pattern of deposition that may undergo calcifica- elevated. Some authors have reported a high incidence

ERRNVPHGLFRVRUJ
1422 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B
FIGURE 23-62  ■  Tophaceous gout: radiographic features. A and B, Anteroposterior and lateral views of knee of patient with long-
standing gout show bone erosion by tophaceous deposits of monosodium urate (arrows). Note radiolucent defects with sclerotic
borders in femoral condyle, tibial plateau, patella, and fibular head. Subchondral erosions show typical overhanging edge.

of prior throat infections and elevation of antistreptoly- radiographic presentation of the lesion. Lymphoma of
sin O titers. Clinical outcome of patients with chronic the bone almost never occurs in young patients whose
recurrent multifocal osteomyelitis is generally good, disease presents with multifocal involvement of metaphy-
but some patients may have persistent disease after a seal parts. On the other hand, a destructive (permeative)
long follow-up.242,247,250 A minority of patients can have a lesion that occurs in a patient older than age 40 years and
severe and prolonged disease course.242 that is shown under a microscope to contain lymphoid
Because of the involvement of the metaphyseal parts cells is very unlikely to represent chronic osteomyelitis.
of the long bones in young patients and the sclerotic
radiographic appearance, chronic recurrent multifocal
osteomyelitis must be differentiated from more common AMYLOIDOSIS OF BONE
bone-forming lesions that affect the same sites in young
patients (i.e., osteosarcoma). Lesions of the clavicle Two basic forms of amyloidosis—secondary and
may simulate Ewing’s sarcoma. Biopsy of the lesions is primary—can affect the skeleton. Primary amyloidosis,
performed to confirm the inflammatory nature of the with no associated generalized disorder, is very rare in the
process, and the lesions are rarely confused with a non- skeleton.
hematologic neoplasm. However, the late phases of the The most frequent form of amyloidosis involving the
process, characterized by the predominant lymphoid skeleton is associated with multiple myeloma. It can
infiltrate, can be confused with lymphoma of the bone. We affect the skeleton diffusely as a part of disseminated
have also seen lymphoma of the bone misclassified as multiorgan disorder. Disseminated and tumoral forms
chronic osteomyelitis. It is helpful to consider in the can coexist. Tumoral amyloidosis is defined by focal
differential diagnosis the entire clinical picture and deposition of amyloid in a sufficient quantity to produce

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1423

FIGURE 23-63  ■  Tophaceous gout: radiographic features. Olecranon deposit of monosodium urate crystals (tophus) shows sufficient
calcium deposits to render it radiopaque. This phenomenon occurs in only one third of gouty tophi, which are usually
radiolucent.

a lytic tumorlike lesion that can be detected on radio- Tumoral deposition of amyloid in bone predisposes a
graphs (Fig. 23-69). Less frequently diffuse or tumoral person to pathologic fracture, especially in weight-
amyloidosis can be associated with chronic autoimmune bearing bones, and may compress adjacent vital struc-
disorder. In older textbooks, secondary amyloidosis, such tures such as the spinal cord.264-268,276
as that associated with tuberculosis or chronic infectious Microscopically, amyloid represents amorphous eosin-
osteomyelitis, has been emphasized. These types of amy- ophilic deposits within stromal tissue and around blood
loidosis are now exceedingly rare. A new form of skeletal vessels. Tumoral amyloidosis is typically associated with
amyloidosis has been described in patients receiving an abundance of amyloid deposits in well-demarcated
long-term dialysis.263,265,267,269,273,274,281,284,287 In this instance, areas associated with interspersed multinucleated giant
the deposits represent β2-microglobulin and may form cells of the foreign body type, mononuclear histiocytes,
lytic lesions several centimeters in diameter.279,282 This and plasma cells (Fig. 23-70). Tumoral deposition of
form of skeletal amyloidosis appears to occur second amyloid in bone is frequently associated with cystic
most frequently, following skeletal amyloidosis associated degeneration.278,286 Amyloid deposits are typically meta-
with multiple myeloma. Several cases of tumorlike amy- chromatic and stain positively with Congo red and
loidosis associated with Bence Jones proteinuria and crystal violet. Congo red preparations show distinct
other laboratory parameters of plasma cell dyscrasias green birefringence under polarized light. Minimal
have been described. Extremely rare cases of tumoral amounts of amyloid deposit may not be recognizable on
amyloid deposition in bone without any other associated conventional histologic preparations. The presence of
disorders have been also described in the skele- amyloidosis in such cases can be disclosed by all or some
ton.264,266,268,270,272,285 However, the type of amyloid has not of the mentioned special stains.265,266,274,281,283 Congo red
been characterized in these cases.265 These lesions seem preparations examined under polarized light appear to be
to be similar to solitary amyloids that occasionally are the most sensitive stains.
identified in other organs and soft tissue.271,275,277,280,288 Text continued on p. 1431

ERRNVPHGLFRVRUJ
1424 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D

FIGURE 23-64  ■  Tophaceous gout: gross and microscopic features. A, Tophaceous gout forming large mass in olecranon area.
B, Bisected tophus shows areas of chalky material surrounded by fibrous tissue. C, Whole-mount specimen of tophaceous gout
involving interphalangeal joint. Note that tophaceous deposit of chalky material erodes bone and disrupts joint surfaces. D, Higher
power magnification of C depicts tophaceous deposit and disruption of interphalangeal joint surface and deposition of material in
adjacent medullary cavity. (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1425

A B

C D
FIGURE 23-65  ■  Tophaceous gout: radiographic and microscopic features. A, Plain radiograph of right hand shows multiple lytic
periarticular defects with overhanging edges typical of tophaceous gout. B, Nodular (tophaceous) deposition of monosodium urate
crystals surrounded by fibrosis with reactive changes such as mononuclear cells and multinucleated giant cells. C, Polarized micros-
copy of tissue processed and fixed in nonaqueous solution shows strong birefringence of needle-shaped monosodium urate crystals.
D, Higher power photomicrograph under compensated polarization microscopy shows negatively birefringent needle-shaped mono-
sodium urate crystals. (B and C, ×50; D, ×100) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1426 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-66  ■  Chronic recurrent multifocal osteomyelitis: radiographic features. A, Lytic and sclerotic changes in proximal metaphy-
sis of tibia in patient with episodes of fever and bone pain. B, Radioisotopic bone scan shows increased uptake in both femoral and
tibial metaphyses. C, Lytic sclerotic changes in distal fibular and tibial metaphysis (same patient as in A and B). D, Radioisotopic
bone scan shows increased uptake in both distal tibial metaphyses.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1427

A B

C D
FIGURE 23-67  ■  Chronic recurrent multifocal osteomyelitis: radiographic and microscopic features. A, Mixed sclerotic and lytic lesion
involving middle phalanx of second finger. B, Radioisotopic scan of same patient shows increased uptake in middle phalanx of
second finger as well as radius. C, Sclerotic lesion involving distal radial metaphysis (same patient as in A and B). D, Chronic inflam-
matory cell infiltrate with fibrosis corresponding to late phase of disease. (D, ×200) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1428 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

D
FIGURE 23-68  ■  Chronic recurrent multifocal osteomyelitis: radiographic and microscopic features. A, Multifocal lytic and sclerotic
lesion involving both clavicles and third right rib. B, Radioisotopic bone scan of same patient as shown in A shows multifocal
increased uptake in right third rib and both clavicles. C, Diffuse lytic and sclerotic process with bone expansion involving left
clavicle (same patient as in A and B). D, Chronic inflammatory infiltrate with predominance of lymphocytes and associated fibrosis.
(D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1429

C
FIGURE 23-69  ■  Tumoral amyloidosis: radiographic features. A, Lytic lesion involving proximal humerus. Note well-demarcated scle-
rotic margins and incomplete trabeculation. No other underlying condition was identified. B, T2-weighted magnetic resonance image
shows high signal intensity in central (cystic) portion of lesion. Note peripheral rim of signal void (same lesion as in A). Lesion
represents cyst with extensive deposits of amyloid in its wall. C, Pathologic fracture through tumor amyloidosis involving distal
femoral end in patient with plasma cell dyscrasia and associated disseminated amyloidosis.

ERRNVPHGLFRVRUJ
1430 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

A B

C D
FIGURE 23-70  ■  Tumoral amyloidosis of bone: microscopic features. A, Extensive deposit of eosinophilic masses. B, Crystal violet
stain of amyloid deposit. C, Congo red stain of amyloid deposit. D, Apple-green birefringence of amyloid stained with Congo red
and examined under polarized light (A-D, ×50) (A, hematoxylin-eosin; B, crystal violet; C and D, Congo red)

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1431

REFERENCES 28. Shanoff LB, Spira M, Hardy SB: Myositis ossificans: evolution to
osteogenic sarcoma: report of a histologically verified case. Am J
Myositis Ossificans Surg 113:537–541, 1967.
1. Ackerman LV: Extra-osseous localized non-neoplastic bone and 29. Skajaa T: Myositis ossificans traumatica. Acta Chir Scand 116:68–
cartilage formation (so-called myositis ossificans): clinical and 74, 1958.
pathological confusion with malignant neoplasms. J Bone Joint 30. Sukov WR, Franco MF, Erickson-Johnson M, et al: Frequency
Surg 40A:279–298, 1958. of USP6 rearrangements in myositis ossificans, brown tumor,
2. Amir G, Mogle P, Sucher E: Case report 729: myositis ossificans and cherubism: molecular cytogenetic evidence that a subset of
and aneurysmal bone cyst. Skeletal Radiol 21:257–259, 1992. “myositis ossificans-like lesions” are the early phases in the forma-
3. Chaplin DM, Harrison MH: Pseudomalignant osseous tumour of tion of soft-tissue aneurysmal bone cyst. Skeletal Radiol 37:321–
soft tissue: report of two cases. J Bone Joint Surg Br 54:334–340, 327, 2008.
1972. 31. Sumiyoshi K, Tsuneyoshi M, Enjoji M: Myositis ossificans: a clini-
4. Cushner FD, Morwessel RM: Myositis ossificans in children. copathologic study of 21 cases. Acta Pathol Jpn 35:1109–1122,
Orthopedics 18:287–291, 1995. 1985.
5. Dahl I, Angervall L: Pseudosarcomatous lesions of the soft tissues 32. Tibone J, Sakimura I, Nickel VL, et al: Heterotopic ossification
reported as sarcoma during a 6-year period (1958-1963). Acta around the hip in spinal cord–injured patients: a long term
Pathol Microbiol Scand 85:917–930, 1977. follow-up study. J Bone Joint Surg 60A:769–775, 1978.
6. Dahl I, Angervall L: Pseudosarcomatous proliferative lesions of 33. Wittenberg RH, Peschke U, Botel U: Heterotopic ossification
soft tissue with or without bone formations. Acta Pathol Microbiol after spinal cord injury: epidemiology and risk factors. J Bone Joint
Scand 85:577–589, 1977. Surg Br 74:215–218, 1992.
7. De Smet AA, Norris MA, Fisher DR: Magnetic resonance imaging 34. Zadek I: Ossifying hematoma in the thigh: a case report. J Bone
of myositis ossificans: analysis of seven cases. Skeletal Radiol Joint Surg 51A:386–390, 1969.
21:503–507, 1992. 35. Zamolyi RQ, Souza P, Nascimento AG, et al: Intraabdominal
8. Garland DE, Blum CE, Waters RL: Periarticular heterotopic myositis ossificans: a report of 9 new cases. Int J Surg Pathol
ossification in head-injured adults: incidence and location. J Bone 14:37–41, 2006.
Joint Surg 62A:1143–1146, 1980.
9. Hutcheson J, Klatte EC, Kremp R: The angiographic appearance Myositis Ossificans Progressiva
of myositis ossificans circumscripta: a case report. Radiology 102: 36. Billings PC, Fiori JL, Bentwood JL, et al: Dysregulated BMP
57–58, 1972. signaling and enhanced osteogenic differentiation of connective
10. Järvi OH, Kvist HT, Vainio PV: Extraskeletal retroperitoneal tissue progenitor cells from patients with fibrodysplasia ossificans
osteosarcoma probably arising from myositis ossificans. Acta progressiva (FOP). J Bone Miner Res 23:305–313, 2008.
Pathol Microbiol Scand 74:11–25, 1968. 37. Bridges AJ, Hsu KC, Singh A, et al: Fibrodysplasia (myositis)
11. Jeffreys TE, Stiles PJ: Pseudomalignant osseous tumor of soft ossificans progressiva. Semin Arthritis Rheum 24:155–164, 1994.
tissue. J Bone Joint Surg Br 48:488–492, 1966. 38. Buyse G, Silberstein J, Goemans N, et al: Fibrodysplasia ossificans
12. Kaleli T, Temiz A, Oztürk H: Pseudomalignant myositis ossificans progressiva: still turning into wood after 300 years? Eur J Pediatr
of the wrist causing compression of the ulnar nerve and artery. A 154:694–699, 1995.
case report. Acta Orthop Belg 69:289–291, 2003. 39. Caron KH, DiPietro MA, Aisen AM, et al: MR imaging of
13. Konishi E, Kusuzaki K, Murata H, et al: Extraskeletal osteosar- early fibrodysplasia ossificans progressiva. J Comput Assist Tomogr
coma arising in myositis ossificans. Skeletal Radiol 30:39–43, 2001. 14:318–321, 1990.
14. Kransdorf MJ, Meis JM, Jelinek JS: Myositis ossificans: MR 40. Cohen RB, Hahn GV, Tabas JA, et al: The natural history of
appearance with radiologic-pathologic correlation. AJR Am J heterotopic ossification in patients who have fibrodysplasia ossifi-
Roentgenol 157:1243–1248, 1991. cans progressiva: a study of 44 patients. J Bone Joint Surg 75A:215–
15. Kwittken J, Branche M: Fasciitis ossificans. Am J Clin Pathol 219, 1993.
51:251–255, 1969. 41. Connor JM, Evans DA: Fibrodysplasia ossificans progressiva: the
16. Lagier R, Cox JN: Pseudomalignant myositis ossificans: patho- clinical features and natural history of 34 patients. J Bone Joint
logical study of 8 cases. Hum Pathol 6:653–665, 1975. Surg Br 64:76–83, 1982.
17. Leithner A, Weinhaeusel A, Zeitlhofer P, et al: Evidence of a 42. Connor JM, Skirton H, Lunt PW: A three generation family
polyclonal nature of myositis ossificans. Virchows Arch 446:438– with fibrodysplasia ossifican progressiva. J Med Genet 30:687–689,
441, 2005. 1993.
18. Mallory TB: A group of metaplastic and neoplastic bone- and 43. Cramer SF, Ruehl A, Mandel MA: Fibrodysplasia ossificans pro-
cartilage-containing tumors of soft parts. Am J Pathol 9:765–776, gressiva: a distinctive bone-forming lesion of the soft tissue.
1933. Cancer 48:1016–1021, 1981.
19. Miller LF, O’Neill C: Myositis ossificans in paraplegics. J Bone 44. Fiori JL, Billings PC, de la Peña LS, et al: Dysregulation of the
Joint Surg 31:283–288, 1949. BMP-p38 MAPK signaling pathway in cells from patients with
20. Norman A, Dorfman HD: Juxtacortical circumscribed myositis fibrodysplasia ossificans progressiva (FOP). J Bone Miner Res
ossificans: evolution and radiographic features. Radiology 96:301– 21:902–909, 2006.
306, 1970. 45. Fontaine K, Sémonin O, Legarde JP, et al: A new mutation of the
21. Nuovo MA, Norman A, Chumas J, et al: Myositis ossificans with noggin gene in a French fibrodysplasia ossificans progressiva
atypical clinical, radiographic, or pathologic findings: a review of (FOP) family. Genet Couns 16:149–154, 2005.
23 cases. Skeletal Radiol 21:87–101, 1992. 46. Fukuda T, Kanomata K, Nojima J, et al: A unique mutation of
22. Ogilvie-Harris DJ, Fornasier VL: Pseudomalignant myositis ossi- ALK2, G356D, found in a patient with fibrodysplasia ossificans
ficans: heterotopic new-bone formation without a history of progressiva is a moderately activated BMP type I receptor. Biochem
trauma. J Bone Joint Surg 62A:1274–1283, 1980. Biophys Res Commun 19(377):905–909, 2008.
23. Pack GT, Braund RR: Development of sarcoma in myositis ossi- 47. Furuya H, Ikezoe K, Wang L, et al: A unique case of fibrodyspla-
ficans. JAMA 119:776–779, 1942. sia ossificans progressiva with an ACVR1 mutation, G356D,
24. Paterson DC: Myositis ossificans circumscripta: report of four other than the common mutation (R206H). Am J Med Genet A
cases without history of injury. J Bone Joint Surg Br 52:296–301, 15(146A):459–463, 2008.
1970. 48. Hegyi L, Gannon FH, Glaser DL, et al: Stromal cells of fibro-
25. Povysil C, Matejovsky Z: Ultrastructural evidence of myofibro- dysplasia ossificans progressiva lesions express smooth muscle
blasts in pseudomalignant myositis ossificans. Virchows Arch [A] lineage markers and the osteogenic transcription factor Runx2/
381:189–203, 1979. Cbfa-1: clues to a vascular origin of heterotopic ossification?
26. Samuelson KM, Coleman SS: Nontraumatic myositis ossificans in J Pathol 201:141–148, 2003.
healthy individuals. JAMA 235:1132–1133, 1976. 49. Illingworth RS: Myositis ossificans progressiva (Münchmeyer’s
27. Sazbon L, Najenson T, Tartakovsky M, et al: Widespread periar- disease): Brief review with report of two cases treated with corti-
ticular new-bone formation in long-term comatose patients. costeroids and observed for 16 years. Arch Dis Child 46:264–268,
J Bone Joint Surg Br 63:120–125, 1981. 1971.

ERRNVPHGLFRVRUJ
1432 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

50. Kaplan FS, Glaser DL, Hebela N, et al: Heterotopic ossification. 78. Ehara S, Nishida J, Abe M, et al: Magnetic resonance imaging
J Am Acad Orthop Surg 12:116–125, 2004. of pseudomalignant osseous tumor of the hand. Skeletal Radiol
51. Kaplan FS, Glaser DL, Shore EM, et al: Hematopoietic stem-cell 23:513–516, 1994.
contribution to ectopic skeletogenesis. J Bone Joint Surg 89A:347– 79. Endo M, Hasegawa T, Tashiro T, et al: Bizarre parosteal osteo-
357, 2007. chondromatous proliferation with a t(1;17) translocation. Virchows
52. Kaplan FS, McCluskey W, Hahn G, et al: Genetic transmission Arch 447:99–102, 2005.
of fibrodysplasia ossificans progressiva: report of a family. J Bone 80. Hirao K, Sugita T, Yasunaga Y, et al: Florid reactive periostitis of
Joint Surg 75A:1214–1220, 1993. the metatarsal bone: a case report. Foot Ankle Int 26:771–774,
53. Kaplan FS, Tabas JA, Gannon FH, et al: The histopathology 2005.
of fibrodysplasia ossificans progressiva: an enchondral process. 81. Ilyas W, Geskin L, Joseph AK, et al: Subungual exostosis of the
J Bone Joint Surg 75A:220–230, 1993. third toe. J Am Acad Dermatol 45:S200–S201, 2001.
54. Kaplan FS, Xu M, Glaser DL, et al: Early diagnosis of 82. Jambhekar NA, Desai SS, Puri A, et al: Florid reactive periostitis
fibrodysplasia ossificans progressiva. Pediatrics 121:e1295–e1300, of the hands. Skeletal Radiol 33:663–665, 2004.
2008. 83. Kontogeorgakos VA, Lykissas MG, Mavrodontidis AN, et al:
55. Lucotte G, Lagarde JP: FOP European Research Group: Muta- Turret exostosis of the hallux. J Foot Ankle Surg 46:130–132,
tions of the noggin and of the activin A type I receptor genes in 2007.
fibrodysplasia ossificans progressiva (FOP). Genet Couns 18:349– 84. Landon GC, Johnson KA, Dahlin DC: Subungual exostosis.
352, 2007. J Bone Joint Surg 61A:256–259, 1979.
56. Miao J, Zhang C, Wu S, et al: Genetic abnormalities in fibrodys- 85. Le Bellec Y, Asfazadourian H: [Bizarre parosteal osteochon-
plasia ossificans progressiva. Genes Genet Syst 87:213–219, 2012. dromatous proliferation (Nora’s lesion). Two case reports]. Chir
57. Rogers JG, Geho WB: Fibrodysplasia ossificans progressiva: Main 24:265–269, 2005.
a survey of 42 cases. J Bone Joint Surg 61A:909–914, 1979. 86. Lee SK, Jung MS, Lee YH, et al: Two distinctive subungual
58. Shore EM, Kaplan FS: Insights from a rare genetic disorder of pathologies: subungual exostosis and subungual osteochondroma.
extra-skeletal bone formation, fibrodysplasia ossificans progressiva Foot Ankle Int 28:595–601, 2007.
(FOP). Bone 43:427–433, 2008. 87. Lindeque BG, Simson IW, Fourie PA: Bizarre parosteal osteo-
59. Shore EM, Xu M, Feldman GJ, et al: A recurrent mutation in the chondromatous proliferation of a phalanx. Arch Orthop Trauma
BMP type I receptor ACVR1 causes inherited and sporadic fibro- Surg 110:58–60, 1990.
dysplasia ossificans progressiva. Nat Genet 38:525–527, 2006. 88. Loewenthal K: Subungual exostosis on a forefinger. N Y State J
60. Schroeder HW, Jr, Zasloff M: The hand and foot malformations Med 64:2691–2695, 1964.
in fibrodysplasia ossificans progressiva. Johns Hopkins Med J 147: 89. Matthewson MH: Subungual exostoses of the fingers: are they
73–78, 1980. really common? Br J Dermatol 98:187–189, 1978.
61. Smith R, Russell GG, Woods RG: Myositis ossificans progressiva: 90. Meneses MF, Unni KK, Swee RG: Bizarre parosteal osteochon-
clinical features of eight patients and their response to treatment. dromatous proliferation of bone (Nora’s lesion). Am J Surg Pathol
J Bone Joint Surg Br 58:48–57, 1976. 17:691–697, 1993.
62. Thickman D, Bonakdar-pour A, Clancy M, et al: Fibrodysplasia 91. Mertens F, Moller E, Mandahl N, et al: The t(X;6) in subungual
ossificans progressiva. Am J Roentgenol 139:935–941, 1982. exostotis results in transciptional deregulation of the gene for
63. Waller SL, Porter DE, Huntley JS: Myositis ossificans progres- insulin receptor substrate 4. Int J Cancer 128:487–491, 2011.
siva. Br J Hosp Med (Lond) 67:606–607, 2006. 92. Mohanna PN, Moiemen NS, Frame JD: Turret exostosis of the
64. Yu PB, Deng DY, Lai CS, et al: BMP type I receptor inhibition thumb. Br J Plast Surg 53:629–631, 2000.
reduces heterotopic [corrected] ossification. Nat Med 14:1363– 93. Muse G, Rayan G: Subungual exostosis. Orthopedics 9:997–998,
1369, 2008. 1986.
94. Nilsson M, Domanski HA, Mertens F, et al: Molecular cytoge-
Reactive Lesions of the Bone Surface netic characterization of recurrent translocation breakpoints in
65. Abramovici L, Steiner GC: Bizarre parosteal osteochondromatous bizarre parosteal osteochondromatous proliferation (Nora’s
proliferation (Nora’s lesion): a retrospective study of 12 cases, lesion). Hum Pathol 35:1063–1069, 2004.
2 arising in long bones. Hum Pathol 33:1205–1210, 2002. 95. Nora FE, Dahlin DC, Beabout JW: Bizarre parosteal osteochon-
66. Apfelberg DB, Drucker D, Maser MR, et al: Subungual osteo- dromatous proliferations of the hands and feet. Am J Surg Pathol
chondroma: differential diagnosis and treatment. Arch Dermatol 7:245–250, 1983.
115:472–473, 1979. 96. Rogers GF, Brzezienski MA: Florid reactive periostitis of the
67. Bendl BJ: Subungual exostosis. Cutis 26:260–262, 1980. middle phalanx: a case report and review of the literature. J Hand
68. Brien EW, Zahiri CA, Mirra JM: Florid reactive periostitis ossi- Surg [Am] 24:1014–1018, 1999.
ficans of the proximal aspect of the tibia: a lesion that must be 97. Rosenberg AE: Pseudosarcomas of soft tissue. Arch Pathol Lab Med
distinguished from osteosarcoma. A case report. J Bone Joint Surg 132:579–586, 2008.
81A:1002–1007, 1999. 98. Sánchez-Castellanos ME, Sandoval-Tress C, Ramírez-Bárcena P:
69. Cavalo DJ, D’Amelio JP, Hirsch AL, et al: Juvenile subungual Subungual exostosis. Arch Dermatol 143:1234, 2007.
osteochondroma: case presentation. J Am Podiatry Assoc 71:81–83, 99. Sherman BD, Sherman RE: Subungual osteochondroma: a case
1981. report. J Am Podiatry Assoc 61:434–436, 1971.
70. Chessler SM, Basler RS: Subungual exostosis. J Am Podiatry Assoc 100. Spjut HJ, Dorfman HD: Florid reactive periostitis of the tubular
68:732–734, 1978. bones of the hands and feet: a benign lesion which may simulate
71. Claude V, Couture C, Battin-Bertho R, et al: [Unusual parosteal osteosarcoma. Am J Surg Pathol 5:423–433, 1981.
osteochondromatous proliferation or Nora’s tumor. A clinico- 101. Storlazzi CT, Wozniak A, Panagopoulos I, et al: Rearrangement
pathological analysis of 4 cases]. Ann Pathol 23:258–260, 2003. of the COL12A1 and COL4A5 genes in subungual exostosis:
72. Cohen HJ, Frank SB, Minkin W, et al: Subungual exostoses. Arch molecular cytogenetic delineation of the tumor-specific transloca-
Dermatol 107:431–432, 1973. tion t(X;6)(q13-14;q22). Int J Cancer 118:1972–1976, 2006.
73. Dave S, Carounanidy U, Thappa DM, et al: Subungual exostosis 102. Suga H, Mukouda M: Subungual exostosis: a review of 16 cases
of the thumb. Dermatol Online J 10:15, 2004. focusing on postoperative deformity of the nail. Ann Plast Surg
74. de Lange EE, Pope TL, Jr, Fechner RE, et al: Case report 428: 55:272–275, 2005.
bizarre parosteal osteochondromatous proliferation (BPOP). Skel- 103. Sundaram M, Wang L, Rotman M, et al: Florid reactive periostitis
etal Radiol 16:481–483, 1987. and bizarre parosteal osteochondromatous proliferation: pre-
75. Dhondt E, Oudenhoven L, Khan S, et al: Nora’s lesion, a distinct biopsy imaging evolution, treatment and outcome. Skeletal Radiol
radiological entity? Skeletal Radiol 35:497–502, 2006. 30:192–198, 2001.
76. Dupree WB, Enzinger FM: Fibro-osseous pseudotumor of the 104. Suresh SS, Zaki H: Subungual exostosis of the big toe: Case
digits. Cancer 58:2103–2109, 1986. reports. J Plast Reconstr Aesthet Surg 2009.
77. Dupuytren G: On the injuries and diseases of bones, London, 1847, 105. Twiston Davies CW: Bizarre parosteal osteochondromatous pro-
Publications of the Sysdenham Society (translated by F LeGros liferation in the hand: a case report. J Bone Joint Surg 67A:648–
Clard). 650, 1985.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1433

106. Wissinger HA, McClain EJ, Boyes JH: Turret exostosis: ossifying 135. Marek FM: Fibrous cortical defect (periosteal desmoid). Bull Hosp
hematoma of the phalanges. J Bone Joint Surg 48A:105–110, 1966. J Dis 16:77–87, 1955.
107. Yuen M, Friedman L, Orr W, et al: Proliferative periosteal pro- 136. Metzmaker JN, Pappas AM: Avulsion fractures of the pelvis. Am
cesses of phalanges: a unitary hypothesis. Skeletal Radiol 21:301– J Sports Med 13:349–358, 1985.
303, 1992. 137. Mirbey J, Besancenot J, Chambers RT, et al: Avulsion fractures of
108. Zambrano E, Nosé V, Perez-Atayde AR, et al: Distinct chromo- the tibial tuberosity in the adolescent athlete: risk factors, mecha-
somal rearrangements in subungual (Dupuytren) exostosis and nism of injury, and treatment. Am J Sports Med 16:336–340, 1988.
bizarre parosteal osteochondromatous proliferation (Nora lesion). 138. Mulligan ME: The “gray cortex”: an early sign of stress fracture.
Am J Surg Pathol 28:1033–1039, 2004. Skeletal Radiol 24:201–203, 1995.
139. Nimityongskul P, Montague WL, Anderson LD: Avulsion frac-
Exuberant Fracture Callus ture of the tibial tuberosity in late adolescence. J Trauma 28:505–
109. Bolander ME: Regulation of fracture repair by growth factors. 509, 1988.
Proc Soc Exp Biol Med 200:165–170, 1992. 140. Orava S, Ala-Ketola L: Avulsion fractures in athletes. Br J Sports
110. Burchardt AJ, Wagner AA, Basse P: Hyperplastic callus formation Med 11:65–71, 1977.
in osteogenesis imperfecta: a case report. Acta Radiol 35:426–428, 141. Orava S, Karpakka J, Hulkko A, et al: Diagnosis and treatment of
1994. stress fractures located at the mid-tibial shaft in athletes. Int J
111. de Palma L, Tulli A, Maccauro G, et al: Fracture callus in osteo- Sports Med 12:419–422, 1991.
petrosis. Clin Orthop 308:85–89, 1994. 142. Orcel P, de Vernejoul MC, Prier A, et al: Stress fractures of the
112. Joerring S, Krogsgaard M, Wilbek H, et al: Collagen turnover lower limbs in osteoporotic patients treated with fluoride. J Bone
after tibial fractures. Arch Orthop Trauma Surg 113:334–336, 1994. Miner Res 5:191–194, 1990.
113. Massey T, Garst J: Compartment syndrome of the thigh with 143. Pester S, Smith PC: Stress fractures in the lower extremities of
osteogenesis imperfecta: a case report. Clin Orthop 267:202–205, soldiers in basic training. Orthop Rev 21:297–303, 1992.
1991. 144. Paluska SA: An overview of hip injuries in running. Sports Med
114. Oni OO, Dunning J, Mobbs RJ, et al: Clinical factors and the size 35:991–1014, 2005.
of the external callus in tibial shaft fractures. Clin Orthop 273:278– 145. Rammelt S, Heineck J, Zwipp H: Metatarsal fractures. Injury
283, 1991. 35:SB77–SB86, 2004.
115. Postacchini F, Gumina S, Perugia D, et al: Early fracture callus in 146. Resnick D, Greenway G: Distal femoral cortical defects, irregu-
the diaphysis of human long bones: histologic and ultrastructural larities, and excavations. Radiology 143:345–354, 1982.
study. Clin Orthop 310:218–228, 1995. 147. Sferopoulos NK, Rafailidis D, Traios S, et al: Avulsion fractures
of the lateral tibial condyle in children. Injury 37:57–60, 2006.
Avulsion and Stress Injuries 148. Simon H: Medial distal metaphyseal femoral irregularity in chil-
116. Anderson EG: Fatigue fractures of the foot. Injury 21:275–279, dren. Radiology 90:258–260, 1968.
1990. 149. Sklar DH, Phillips JJ, Lachman RS: Case report 683: distal
117. Anderson SE, Johnston JO, O’Donnell R, et al: MR imaging of metaphyseal femoral defect (cortical desmoid; distal femoral corti-
sports-related pseudotumor in children: mid femoral diaphyseal cal irregularity). Skeletal Radiol 20:394–396, 1991.
periostitis at insertion site of adductor musculature. AJR Am J 150. Sundar M, Carty H: Avulsion fractures of the pelvis in children:
Roentgenol 176:1227–1231, 2001. a report of 32 fractures and their outcome. Skeletal Radiol 23:85–
118. Barnes GR, Jr, Gwinn JL: Distal irregularities of the femur simu- 90, 1994.
lating malignancy. Am J Roentgenol 122:180–185, 1974. 151. Tyrrell PN, Davies AM: Magnetic resonance imaging appearance
119. Beals RK, Cook RD: Stress fractures of the anterior tibial diaphy- of fatigue fractures of the long bones of the lower limb. Br J Radiol
sis. Orthopedics 14:869–875, 1991. 67:332–338, 1994.
120. Blumberg N, Arbel R, Dabby D: Avulsion of the triceps tendon 152. van Laarhoven HA, te Slaa RL, van Laarhoven EW: Isolated
during acceleration stress. Aviat Space Environ Med 73:931–933, avulsion fracture of the lesser tuberosity of the humerus. J Trauma
2002. 39:997–999, 1995.
121. Buckley SL, Robertson WW, Jr, Shalaby-Rana E: Stress fractures 153. Velchik MG, Heyman S, Makler PT, Jr, et al: Bone scintigraphy:
of the femoral diaphysis in young children: a report of 2 cases. differentiating benign cortical irregularity of the distal femur from
Clin Orthop 310:165–169, 1995. malignancy. J Nucl Med 25:72–74, 1984.
122. Chadwick CJ: Tendinitis of the pectoralis major insertion with 154. Volpin G, Hoerer D, Groisman G, et al: Stress fractures of the
humeral lesions: a report of two cases. J Bone Joint Surg Br 71:816– femoral neck following strenuous activity. J Orthop Trauma 4:394–
818, 1989. 398, 1990.
123. Clement DB, Ammann W, Taunton JE, et al: Exercise-induced 155. Wootton JR, Cross MJ, Holt KW: Avulsion of the ischial apophy-
stress injuries to the femur. Int J Sports Med 14:347–352, 1993. sis: the case for open reduction and internal fixation. J Bone Joint
124. Dunham WK, Marcus NW, Enneking WF, et al: Developmental Surg Br 72:625–627, 1990.
defects of the distal femoral metaphysis. J Bone Joint Surg 156. Yamazaki T, Maruoka S, Takahashi S, et al: MR findings of avul-
62A:801–806, 1980. sive cortical irregularity of the distal femur. Skeletal Radiol 24:43–
125. Earwaker J: Isolated avulsion fracture of the lesser tuberosity of 46, 1995.
the humerus. Skeletal Radiol 19:121–125, 1990. 157. Young DW, Nogrady MB, Dunbar JS, et al: Benign cortical irreg-
126. Fernbach SK, Wilkinson RH: Avulsion injuries of the pelvis and ularities in the distal femur of children. J Can Assoc Radiol 23:107–
proximal femur. Am J Roentgenol 137:581–584, 1981. 115, 1972.
127. Giladi M, Milgrom C, Simkin A, et al: Stress fractures: identifi-
able risk factors. Am J Sports Med 19:647–652, 1991. Pubic Osteolysis
128. Ha KI, Hahn SH, Chung MY, et al: A clinical study of stress 158. An HS, Jackson WT, Hawthorne KB: Osteolysis of the pubic bone
fractures in sports activities. Orthopedics 14:1089–1095, 1991. simulating malignancy. Orthopedics 11:951–954, 1988.
129. Hershman EB, Lombardo J, Bergfeld JA: Femoral shaft stress 159. Burns JR, Gregory JG: Osteomyelitis of the pubic symphysis after
fractures in athletes. Clin Sports Med 9:111–119, 1990. urologic surgery. J Urol 118:803–805, 1977.
130. Hershman EB, Mailly T: Stress fractures. Clin Sports Med 9:183– 160. Casey D, Mirra J, Staple TW: Parasymphyseal insufficiency
214, 1990. fractures of the os pubis. AJR Am J Roentgenol 142:581–586,
131. Kadel NJ, Teitz CC, Kronmal RA: Stress fractures in ballet 1984.
dancers. Am J Sports Med 20:445–449, 1992. 161. Goergen TG, Resnick D, Riley RR: Post traumatic abnormalities
132. Kimmelstiel P, Rapp IH: Cortical defect due to periosteal des- of the pubic bone simulating malignancy. Radiology 126:85–87,
moids. Bull Hosp J Dis 12:286–297, 1951. 1978.
133. Kozlowski K, Azouz M, Hoff D: Stress fracture of the fibula in 162. Hall FM, Goldberg RP, Kasdon EJ, et al: Post traumatic osteolysis
the first decade of life: report of eight cases. Pediatr Radiol 21:381– of the pubic bone simulating a malignant lesion. J Bone Joint Surg
383, 1991. 66A:121–126, 1984.
134. Kumar R, Swischuk LE, Madewell JE: Benign cortical defect: site 163. Harris NH, Murray RO: Lesions of the symphysis in athletes. Br
for an avulsion fracture. Skeletal Radiol 15:553–555, 1986. Med J 4:211–214, 1974.

ERRNVPHGLFRVRUJ
1434 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

164. McCarthy B, Dorfman HD: Pubic osteolysis: a benign lesion of tions in the glycosyl transferase domain of the GALNT3 gene.
the pelvis closely mimicking a malignant neoplasm. Clin Orthop J Clin Endocrinol Metab 91:4472–4475, 2006.
251:300–307, 1990. 189. Ichikawa S, Lyles KW, Econs MJ: A novel GALNT3 mutation in
165. McGuigan LE, Edmonds JP, Painter DM: Pubic osteolysis. J Bone a pseudoautosomal dominant form of tumoral calcinosis: evidence
Joint Surg 66A:127–129, 1984. that the disorder is autosomal recessive. J Clin Endocrinol Metab
166. Rask MR: Fatigue fracture ossa pubes due to obesity: report of a 90:2420–2423, 2005.
case. Clin Orthop 142:182–184, 1979. 190. Jain SP: Tumoral calcinosis in Somalia and Ethiopia: a report of
167. Schabel SI, Burgener FA: Osteitis pubis in renal failure simulating 21 cases and brief review of literature. East Afr Med J 66:476–480,
chondrosarcoma. Br J Radiol 48:1027–1028, 1975. 1989.
168. Takada J, Yamashita T, Takebayashi T, et al: Osteoporosis in 5 191. Katayama I, Higashi K, Mukai H, et al: Tumoral calcinosis in
elderly women with pubic osteolysis. Acta Orthop Scand 74:59–61, scleroderma. J Dermatol 16:82–85, 1989.
2003. 192. Kindblom LG, Gunterberg B: Tumoral calcinosis: an ultrastruc-
tural analysis and consideration of pathogenesis. APMIS 96:368–
Tumoral Calcinosis 376, 1988.
169. Baldursson H, Evans EB, Dodge WF, et al: Tumoral calcinosis 193. Knowles SA, Declerck G, Anthony PP: Tumoral calcinosis. Br J
with hyperphosphatemia: a report of a family with incidence in Surg 70:105–107, 1983.
four siblings. J Bone Joint Surg 51A:913–925, 1969. 194. Kolawole TM, Bohrer SP: Tumoral calcinosis with “fluid levels”
170. Barton DL, Reeves RJ: Tumoral calcinosis: report of three in the tumoral masses. Am J Roentgenol 120:461–465, 1974.
cases and review of the literature. Am J Roentgenol 86:351, 195. Krueger-Franke M, Siebert CH, Weiss M, et al: Tumoral calci-
1961. nosis of the ischium. Arch Orthop Trauma Surg 111:284–286, 1992.
171. Berg D: Tumoral calcinosis. Br J Surg 59:570–571, 1972. 196. Laleye A, Alao MJ, Gbessi G, et al: Tumoral calcinosis due to
172. Boskey AL, Vigorita VJ, Sencer O, et al: Chemical, microscopic, GALNT3 C.516-2A >T mutation in a black African family. Genet
and ultrastructural characterization of the mineral deposits in Couns 19:183–192, 2008.
tumoral calcinosis. Clin Orthop 178:258–269, 1983. 197. Larsson T, Davis SI, Garringer HJ, et al: Fibroblast growth
173. Brown ML, Thrall JH, Cooper RA, et al: Radiography and scin- factor-23 mutants causing familial tumoral calcinosis are differen-
tigraphy in tumoral calcinosis. Radiology 124:757–758, 1977. tially processed. Endocrinology 146:3883–3891, 2005.
174. Buchkremer F, Farese S: Uremic tumoral calcinosis improved by 198. Lufkin EG, Wilson DM, Smith LH, et al: Phosphorus excretion
kidney transplantation. Kidney Int 74:1498, 2008. in tumoral calcinosis: response to parathyroid hormone and acet-
175. Chefetz I, Kohno K, Izumi H, et al: GALNT3, a gene associated azolamide. J Clin Endocrinol Metab 50:648–653, 1980.
with hyperphosphatemic familial tumoral calcinosis, is transcrip- 199. Lyles KW, Burkes EJ, Ellis GJ, et al: Genetic transmission of
tionally regulated by extracellular phosphate and modulates tumoral calcinosis: autosomal dominant with variable clinical
matrix metalloproteinase activity. Biochim Biophys Acta 61-67:2009, expressivity. J Clin Endocrinol Metab 60:1093–1096, 1985.
1792. 200. Lyles KW, Halsey DL, Friedman NE, et al: Correlation of serum
176. Chefetz I, Ben Amitai D, Browning S, et al: Normophosphatemic concentrations of 1,25-dihydroxy vitamin D, phosphorus, and
familial tumoral calcinosis is caused by deleterious mutations in parathyroid hormone in tumoral calcinosis. J Clin Endocrinol
SAMD9, encoding a TNF-alpha responsive protein. J Invest Der- Metab 67:88–92, 1988.
matol 128:1423–1429, 2008. 201. Maathuis JB, Koten JW: Kikuyu-bursa and tumoral calcinosis.
177. Chefetz I, Heller R, Galli-Tsinopoulou A, et al: A novel homozy- Trop Geogr Med 21:389–396, 1969.
gous missense mutation in FGF23 causes Familial Tumoral Cal- 202. McClatchie S, Bremner AD: Tumoral calcinosis: an unrecognized
cinosis associated with disseminated visceral calcification. Hum disease. Br Med J 1:153–155, 1969.
Genet 118:261–266, 2005. 203. Meltzer CC, Fishman EK, Scott WW, Jr: Tumoral calcinosis
178. Dereure O: [SAMD9 mutation in normophosphatemic familial causing bone erosion in a renal dialysis patient. Clin Imaging
tumoral calcinosis]. Ann Dermatol Venereol 134:505, 2007. 16:49–51, 1992.
179. Dumitrescu CE, Kelly MH, Khosravi A, et al: A case of familial 204. Mozaffarian G, Lafferty FW, Pearson OH: Treatment of tumoral
tumoral calcinosis/hyperostosis-hyperphosphatemia syndrome calcinosis with phosphorus deprivation. Ann Intern Med 77:741–
due to a compound heterozygous mutation in GALNT3 demon- 745, 1972.
strating new phenotypic features. Osteoporos Int 20:1273–1278, 205. Murthy DP: Tumoral calcinosis: a study of cases from Papua, New
2008. Guinea. J Trop Med Hyg 93:403–407, 1990.
180. Fathi I, Sakr M: Review of tumoral calcinosis: a rare clinical– 206. Noyez JF, Murphree SM, Chen K: Tumoral calcinosis, a clinical
pathological entity. World J Clin Cases 2:409–414, 2014. report of 11 cases. Acta Orthop Belg 59:249–254, 1993.
181. Garringer HJ, Malekpour M, Esteghamat F, et al: Molecular 207. Prince MJ, Schaeffer PC, Goldsmith RS, et al: Hyperphospha-
genetic and biochemical analyses of FGF23 mutations in familial temic tumoral calcinosis: association with elevation of serum
tumoral calcinosis. Am J Physiol Endocrinol Metab 295:E929–E937, 1,25-dihydroxycholecalciferol concentrations. Ann Intern Med
2008. 96:586–591, 1982.
182. Garringer HJ, Fisher C, Larsson TE, et al: The role of 208. Slavin RE, Wen J, Kumar D, et al: Familial tumoral calcinosis: a
mutant UDP-N-acetyl-alpha-D-galactosamine-polypeptide N- clinical, histopathological, and ultrastructural study with an analy-
acetylgalactosaminyltransferase 3 in regulating serum intact fibro- sis of its calcifying process and pathogenesis. Am J Surg Pathol
blast growth factor 23 and matrix extracellular phosphoglycoprotein 17:788–802, 1993.
in heritable tumoral calcinosis. J Clin Endocrinol Metab 91:4037– 209. Sledz K, Ortiz O, Wax M, et al: Tumoral calcinosis of the tem-
4042, 2006. poromandibular joint: CT and MR findings. Am J Neuroradiol
183. Geirnaerdt MJ, Kroon HM, van der Heul RO, et al: Tumoral 16:782–785, 1995.
calcinosis. Skeletal Radiol 24:148–151, 1995. 210. Steinbach LS, Johnston JO, Tepper EF, et al: Tumoral calcinosis:
184. Gupta A, Sun Y, Konstantinov KN, et al: Tumoral calcinosis radiologic-pathologic correlation. Skeletal Radiol 24:573–578,
without hyperparathyroidism in a patient on continuous ambula- 1995.
tory peritoneal dialysis. Adv Perit Dial 24:132–136, 2008. 211. Steinherz R, Chesney RW, Eisenstein B, et al: Elevated serum
185. Hacihanefioglu U: Tumoral calcinosis: a clinical and pathological calcitriol concentrations do not fall in response to hyperphospha-
study of eleven unreported cases in Turkey. J Bone Joint Surg temia in familial tumoral calcinosis. Am J Dis Child 139:816–819,
60A:1131–1135, 1978. 1985.
186. Ichikawa S, Imel EA, Kreiter ML, et al: A homozygous missense 212. Tezelman S, Siperstein AE, Duh QY, et al: Tumoral calcinosis:
mutation in human KLOTHO causes severe tumoral calcinosis. controversies in the etiology and alternatives in the treatment.
J Musculoskelet Neuronal Interact 7:318–319, 2007. Arch Surg 128:737–744, 1993.
187. Ichikawa S, Imel EA, Kreiter ML, et al: A homozygous missense 213. Topaz O, Indelman M, Chefetz I, et al: A deleterious mutation in
mutation in human KLOTHO causes severe tumoral calcinosis. SAMD9 causes normophosphatemic familial tumoral calcinosis.
J Clin Invest 117:2684–2691, 2007. Am J Hum Genet 79:759–764, 2006.
188. Ichikawa S, Imel EA, Sorenson AH, et al: Tumoral calcinosis 214. Vasudev KS, Tapp L, Harris M, et al: Tumoral calcinosis in
presenting with eyelid calcifications due to novel missense muta- Britain. Br Med J 1:767, 1973.

ERRNVPHGLFRVRUJ
23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone 1435

215. Veress B, Malik MO, El Hassan AM: Tumoural lipocalcinosis: 242. Catalano-Pons C, Comte A, Wipff J, et al: Clinical outcome in
a clinicopathological study of 20 cases. J Pathol 119:113–118, children with chronic recurrent multifocal osteomyelitis. Rheuma-
1976. tology (Oxford) 47:1397–1399, 2008.
216. Wilber JF, Slatopolsky E: Hyperphosphatemia and tumoral calci- 243. Cyrlak D, Pais MJ: Chronic recurrent multifocal osteomyelitis.
nosis. Ann Intern Med 68:1043–1049, 1968. Skeletal Radiol 15:32–39, 1986.
217. Yaghmai I, Mirbod P: Tumoral calcinosis. Am J Roentgenol Radium 244. El-Shanti HI, Ferguson PJ: Chronic recurrent multifocal osteo-
Ther Nucl Med 111:573–578, 1971. myelitis: a concise review and genetic update. Clin Orthop Relat
218. Yu X, White KE: FGF23 and disorders of phosphate homeostasis. Res 462:11–19, 2007.
Cytokine Growth Factor Rev 16:221–232, 2005. 245. Giedion A, Holthusen W, Masel LF, et al: Subacute and chronic
“symmetrical” osteomyelitis. Ann Radiol (Paris) 15:329–342, 1972.
Tophaceous Pseudogout 246. Girardet JP, Sambucy F, Douillet P, et al: Osteomyelite symetrique
219. Fam AG: Calcium pyrophosphate crystal deposition disease and subaigue. Arch Fr Pediatr 36:418–422, 1979.
other crystal deposition diseases. Curr Opin Rheumatol 4:574–582, 247. Girschick HJ, Zimmer C, Klaus G, et al: Chronic recurrent mul-
1992. tifocal osteomyelitis: what is it and how should it be treated? Nat
220. Hamilton EB: Diseases associated with CPPD deposition disease. Clin Pract Rheumatol 3:733–738, 2007.
Arthritis Rheum 19:353–357, 1976. 248. Golla A, Jansson A, Ramser J, et al: Chronic recurrent multifocal
221. Ishida T, Dorfman HD, Bullough PG: Tophaceous pseudogout osteomyelitis (CRMO): evidence for a susceptibility gene located
(tumoral calcium pyrophosphate dihydrate crystal deposition on chromosome 18q21.3-18q22. Eur J Hum Genet 10:217–221,
disease). Hum Pathol 26:587–593, 1995. 2002.
222. Jensen PS, Putman CE: Current concepts with respect to chon- 249. Gustavson KH, Wilbrand HF: Chronic symmetric osteomyelitis.
drocalcinosis and the pseudogout syndrome. AJR Am J Roentgenol Acta Radiol 15:551–557, 1974.
Radium Ther Nucl Med 123:531–539, 1975. 250. Hoffmann T, Finger D: Chronic recurrent multifocal osteomyeli-
223. Markel SF, Hart WR: Arthropathy in calcium pyrophosphate tis. J Rheumatol 34:2499–2500, 2007.
dihydrate crystal deposition disease. Arch Pathol Lab Med 106:529– 251. Jansson A, Renner ED, Ramser J, et al: Classification of non-
533, 1982. bacterial osteitis: retrospective study of clinical, immunological
224. Martel W, McCarter D, Solsky MA, et al: Further observations and genetic aspects in 89 patients. Rheumatology (Oxford) 46:154–
on the arthropathy of calcium pyrophosphate crystal deposition 160, 2007.
disease. Radiology 141:1–15, 1981. 252. Kawakami T, Toyoshima R, Furuse K, et al: So-called sterno-
225. McAllister CM, Dorfman HD: Crystal deposition disease in bone: costo-clavicular hyperostosis: its etiology and manifestations.
a review. Einstein Q J Biol Med 7:122–130, 1989. Rinsho Seikei Geka 15:650, 1980.
226. McCarty DJ, Kohn NN, Faires JS: The significance of calcium 253. Kozlowski K, Masel J, Harbison S, et al: Multifocal chronic osteo-
phosphate crystals in the synovial fluid of arthritic patients: the myelitis of unknown etiology. Pediatr Radiol 13:130–136, 1983.
“pseudogout syndrome.” I. Clinical aspects. Ann Intern Med 254. Machiels F, Seynaeve P, Lagey C, et al: Chronic recurrent multi-
56:711–737, 1962. focal osteomyelitis with MR correlation: a case report. Pediatr
227. Resnick CS, Resnick D: Crystal deposition disease. Semin Arthritis Radiol 22:535–536, 1992.
Rheum 12:390–403, 1983. 255. Murray SD, Kehl DK: Chronic recurrent multifocal osteomyeli-
228. Richards AJ, Hamilton BB: Destructive arthropathy in chondro- tis: a case report. J Bone Joint Surg 66A:1110–1112, 1984.
calcinosis articularis. Ann Rheum Dis 33:196–203, 1974. 256. Probst FP: Chronic multifocal cleido-metaphyseal osteomy-
229. Shaffrey CI, Munoz EL, Sutton CL, et al: Tumoral calcium pyro- elitis of childhood. Acta Radiol Diagn (Stockholm) 17:531–537,
phosphate dihydrate deposition disease mimicking a cervical spine 1976.
neoplasm: case report. Neurosurgery 37:335–339, 1995. 257. Probst FP, Bjorksten B, Gustavson KH: Radiological aspects of
230. Yu SL, Li RZ, Bian ZH: Tumoral calcium pyrophosphate dihy- chronic recurrent multifocal osteomyelitis. Ann Radiol (Paris)
drate deposition disease: report of a case with review of the litera- 21:115–125, 1978.
ture. Chin Med J 105:780–784, 1992. 258. Ravelli A, Marseglia GL, Viola S, et al: Chronic recurrent multi-
focal osteomyelitis with unusual features. Acta Paediatr 84:222–
Tophaceous Gout 225, 1995.
231. Bloch C, Hermann G, Yu TF: A radiologic reevaluation of 259. Segev E, Hayek S, Lokiec F, et al: Primary chronic sclerosing
gout: a study of 2,000 patients. AJR Am J Roentgenol 134:781–787, (Garré’s) osteomyelitis in children. J Pediatr Orthop B 10:360–364,
1980. 2001.
232. Keith MP, Gilliland WR: Updates in the management of gout. 260. Solheim LF, Paus B, Liverud K, et al: Chronic recurrent multifo-
Am J Med 120:221–224, 2007. cal osteomyelitis: a new clinical-radiological syndrome. Acta
233. Kelley WN, Fox IH: Gout and related disorders of purine metab- Orthop Scand 51:37–41, 1980.
olism. In Kelley WN, Harris ED, Jr, Ruddy S, et al, editors: 261. Stanton RP, Lopez-Sosa FH, Doidge R: Chronic recurrent mul-
Textbook of rheumatology, ed 4, 2 vols, Philadelphia, 1993, WB tifocal osteomyelitis. Orthop Rev 22:229–233, 1993.
Saunders. 262. Tlougan BE, Podjasek JO, O’Haver J, et al: Chronic recurrent
234. Monu JU, Pope TL, Jr: Gout: a clinical and radiologic review. multifocal osteomyelitis (CRMO) and synovitis, acne, pustulosis,
Radiol Clin North Am 42:169–184, 2004. hyperostosis, and osteitis (SAPHO) syndrome with associated
235. Peh WC: Tophaceous gout. Am J Orthop 30:665, 2001. neutrophilic dermatoses: a report of seven cases and review of the
236. Pennes DR, Martel W: Hyperuricemia and gout. Semin Roentgenol literature. Pediatr Dermatol 26:497–505, 2009.
21:245–255, 1986.
237. Resnick D, Broderick TW: Intraosseous calcifications in topha- Amyloidosis of Bone
ceous gout. AJR Am J Roentgenol 137:1157–1161, 1981. 263. Casey TT, Stone WJ, DiRaimondo CR, et al: Tumoral amyloido-
238. Resnick D, Niwayama G: Gouty arthritis. In Resnick D, editor: sis of bone of beta 2-microglobulin origin in association with
Diagnosis of bone and joint disorders, ed 3, Philadelphia, 1994, WB long-term hemodialysis: a new type of amyloid disease. Hum
Saunders. Pathol 17:731–738, 1986.
264. Cloft HJ, Quint DJ, Markert JM, et al: Primary osseous amy-
Chronic Recurrent Multifocal Osteomyelitis loidoma causing spinal cord compression. Am J Neuroradiol
239. Bergdahl K, Bjorksten B, Gustavson K-H, et al: Pustulosis palmo- 16:1152–1154, 1995.
plantaris and its relation to chronic recurrent multifocal osteomy- 265. Cohen AS: Proteins of the systemic amyloidoses. Curr Opin
elitis. Dermatologica 159:37–45, 1979. Rheum 6:55–67, 1994.
240. Bjorksten B, Gustavson K-H, Eriksson B, et al: Chronic recurrent 266. Cornwell GG, III, Johnson KH, Westermark P: The age related
multifocal osteomyelitis and pustulosis palmoplantaris. J Pediatr amyloids: a growing family of unique biochemical substances.
93:227–231, 1978. J Clin Pathol 48:984–989, 1995.
241. Bjorksten B, Boquist L: Histopathological aspects of chronic 267. Davidson GS, Montanera WJ, Fleming JF, et al: Amyloid destruc-
recurrent multifocal osteomyelitis. J Bone Joint Surg Br 62:376– tive spondyloarthropathy causing cord compression: related to
380, 1980. chronic renal failure and dialysis. Neurosurgery 33:519–522, 1993.

ERRNVPHGLFRVRUJ
1436 23  Reactive and Metabolic Conditions Simulating Neoplasms of Bone

268. Dickman CA, Sonntag VK, Johnson P, et al: Amyloidoma of the 278. Naito M, Ogata K, Shiota E, et al: Amyloid bone cysts of the
cervical spine: a case report. Neurosurgery 22:419–422, 1988. femoral neck: impending fractures treated by curettage and bone
269. Drueke T, Touam M, Zingraff J: Dialysis-associated amyloidosis. grafting. J Bone Joint Surg Br 76:922–925, 1994.
Adv Renal Replace Ther 2:24–39, 1995. 279. Onishi S, Andress DL, Maloney NA, et al: Beta 2-microglobulin
270. Ferreiro JA, Bhuta S, Nieberg RK, et al: Amyloidoma of the skull deposition in bone in chronic renal failure. Kidney Int 39:990–995,
base. Arch Pathol Lab Med 114:974–976, 1990. 1991.
271. Fierens J, Mees U, Vanbockrijck M, et al: Amyloidoma of the 280. Pasternak S, Wright BA, Walsh N: Soft tissue amyloidoma of the
chest wall: a rare entity. Interact Cardiovasc Thorac Surg 7:1194– extremities: report of a case and review of the literature. Am J
1195, 2008. Dermatopathol 29:152–155, 2007.
272. Griffin M, Parai M, Fernandez D, et al: Amyloid tumor of the 281. Sipe JD: Amyloidosis. Crit Rev Clin Lab Sci 31:325–354, 1994.
sacrum: a case report. Acta Cytol 39:503–506, 1995. 282. Sprague SM, Popovtzer MM: Is beta-2-microglobulin a mediator
273. Isaacs M, Bansal M, Flombaum CD, et al: Case report 772: stress of bone disease? Kidney Int 47:1–6, 1995.
fracture of the hip secondary to renal osteodystrophy and erosion 283. Tan SY, Pepys MB: Amyloidosis. Histopathology 25:403–414,
of ischium due to amyloid deposition. Skeletal Radiol 22:129–133, 1994.
1993. 284. Tateishi H, Maeda M, Yoh K, et al: Pathologic fracture associated
274. Kisilevsky R: Amyloid and amyloidosis: differences, common with amyloid deposition in the bone of a chronic hemodialysis
themes, and practical considerations. Mod Pathol 4:514–518, 1991. patient: a case report. Clin Orthop 274:300–304, 1992.
275. Krishnan J, Chu WS, Elrod JP, et al: Tumoral presentation of 285. Unal F, Hepgul K, Bayindir C, et al: Skull base amyloidoma: case
amyloidosis (amyloidomas) in soft tissue: a report of 14 cases. Am report. J Neurosurg 76:303–306, 1992.
J Clin Pathol 100:135–144, 1993. 286. Urban BA, Fishman EK, Humphrey RL: Primary amyloidosis of
276. Lai KN, Chan KW, Siu DL, et al: Pathologic hip fractures sec- the sternum: imaging findings. Clin Imaging 18:75–78, 1994.
ondary to amyloidoma: case report and review of the literature. 287. Villarejo F, Perez Diaz C, Perla C, et al: Spinal cord compression
Am J Med 77:937–943, 1984. by amyloid deposits. Spine 19:1178–1181, 1994.
277. Lipper S, Kahn LB: Amyloid tumor: a clinicopathologic study of 288. Weiss SW: Tumoral amyloidosis of soft tissue (amyloidoma): new
four cases. Am J Surg Pathol 2:141–145, 1978. approaches to an old problem. Am J Clin Pathol 100:91, 1993.

ERRNVPHGLFRVRUJ
C H A P T E R 2 4 

Precancerous Conditions

CHAPTER OUTLINE

PAGET’S DISEASE OF BONE OSTEOGENESIS IMPERFECTA


OSTEOMYELITIS SYNDROMES PREDISPOSING TO MALIGNANCY
IN BONE
BONE INFARCTS
Rothmund-Thomson Syndrome
METALLIC IMPLANTS Li-Fraumeni Syndrome
RADIATION INJURY

The presence of a neoplasm in bone is manifested in a are listed in Table 24-1. In this chapter the discussion
variety of ways. Pain and localized swelling or pathologic of precancerous lesions is limited to those conditions
fracture are the most frequent presenting complaints, but that play a major role in increasing the risk for bone
rarely is a bone tumor discovered as an incidental finding malignancy.
on radiographic images made for other reasons (e.g., after
trauma). Occasionally, the appearance of new symptoms
or a changing clinical picture in the presence of a known
skeletal disorder may signal the onset of malignant trans- PAGET’S DISEASE OF BONE
formation in a benign precursor lesion. Definition
Early diagnosis of bone neoplasms is complicated
by the fact that except for osteoid osteoma and the Osteitis deformans, as described by Sir James Paget, rep-
extremely uncommon intraosseous glomus tumor, small resents a prototype skeletal disorder that predisposes to
bone tumors are usually asymptomatic. Primary malig- the development of sarcoma in bone.12,13 A comprehen-
nant tumors of bone generally reach substantial dimen- sive description of this unique disease is beyond the scope
sions before they produce symptoms (predominantly pain of this book; thus the discussion of Paget’s disease is
or pathologic fracture). restricted to the basic clinicopathologic features of oste-
Although the majority of primary bone malignancies itis deformans that are relevant for its recognition when
arise de novo, it is increasingly apparent that some it is associated with sarcoma of bone. From the pathoge-
develop in association with recognizable precursors. The netic point of view, the disease can be explained by
likelihood of discovering these associated lesions can be increased transient but progressive and multifocal osteo-
facilitated by attention to clinicopathologic correlation of clastic activity, with bone resorption followed by new
all available data before arriving at a diagnosis. In bone, bone formation, and ultimately bone sclerosis.
the inclusion of radiographic imaging data in the diag- Ultrastructural and immunohistochemical analyses
nostic process offers a unique opportunity to discover have demonstrated cytoplasmic and nuclear inclusions in
clues to causal relationships that may not be reflected in the osteoclasts of pagetic bone that are similar to those
histologic patterns or in other laboratory data. This is seen in paramyxovirus infection.4,8,10 As a result of these
especially true when serial radiographs are available for observations, some authors support the hypothesis that
review. Paget’s disease may be induced by a slow viral infec-
Paget’s disease, radiation injury, and some of the more tion.8,17 When there is a high rate of bone turnover with
common benign cartilaginous dysplasias are the most rapid bone growth, cell proliferation, and osteoclastic
clearly established precancerous conditions. The relative activity, it is conceivable that somatic mutations and
rarity of malignant transformation in fibrous dysplasia, genomic deletions will occur with a high frequency and
osteomyelitis, bone cysts, osteogenesis imperfecta, and thus provide a molecular basis for malignant transforma-
bone infarction places these conditions in a separate cat- tion. Other authors have alluded to the possibility that
egory. The possible relationship of secondary malignancy an infectious etiology is a cofactor in the development of
to metallic implants and joint prostheses is a subject malignancy in Paget’s disease.10 However, more recent
of increasing concern, although its statistical validity is attempts to identify paramyxovirus coding sequences by
still in doubt. Additional neoplastic and nonneoplastic the use of polymerase chain reactions have failed to dem-
lesions that may be precursors of malignancy in bone onstrate the presence of a paramyxoviral genome in
1437
ERRNVPHGLFRVRUJ
1438 24  Precancerous Conditions

A [GOLGA6A]).4,15 The environmental factors that are


TABLE 24-1 Neoplastic and Nonneoplastic
suggested as possible contributors for Paget’s disease
Precursors of Malignancy in Bone
include dietary deficiencies (calcium and vitamin D),
Nonneoplastic chronic infection with measles virus, canine distemper
Conditions Benign Tumors virus, and respiratory syncytial virus.4,18 The potential
Paget’s disease* Osteochondroma contribution of chronic infections with measles virus has
(see Chapter 6) been the most extensively studied. In animal models, the
Chronic osteomyelitis* Enchondroma (see Chapter 6) expression of measles virus nucleocapsid gene (MVNP)
Bone infarct* Ollier’s disease and Maffucci’s in osteoclasts induces pagetic lesions. The overexpression
syndrome (see Chapter 6)
Metallic implants* Chondromyxoid fibroma
of MVNP in osteoclastic cells activates NFκB and results
(see Chapter 6) in increased levels of IL-6 and ephrinB2. This, in turn,
Radiation, osteitis* Giant cell tumor elevates ephrinB4 in osteoblastic cells, leading to the
(see Chapter 10) increased bone formation implicated in the development
Osteogenesis Metaphyseal fibrous defect of pagetoid sclerosis. These new molecular developments
imperfecta* (see Chapter 8)
Fibrous dysplasia Osteoblastoma (see Chapter 4) provide interesting clues to the pathobiology of Paget’s
(see Chapter 8) disease, but full understanding of this complex and still
Bone cysts Osteofibrous dysplasia and mysterious disorder is far from complete.
(see Chapter 15) adamantinoma of long bones
(see Chapters 8 and 17)
Synoval chondromatosis Incidence and Location
(see Chapter 20)
Paget’s disease is widespread in many countries. Autopsy
*These conditions are described in this chapter. data indicate that it can be found in 3% to 4% of
For a description of the other lesions and their roles as precursors unselected patients older than age 45 years who died of
of malignancy in bone, refer to the appropriate chapters.
various causes.2,17 The rate of radiographically diagnosed
Paget’s disease varies from 1 in 500 to 1 in 12,000 hospital
admissions.1 It appears that male and female patients are
Paget’s disease. Modern concepts postulate that the etiol- almost equally affected by the disease, but in many series,
ogy of Paget’s disease is heterogeneous and involves both the incidence is slightly higher in men.
genetic and environmental components (Fig. 24-1).1,4,15 Paget’s disease is common in Europe. The incidence
The gene with mutations in the coding region associated is highest in the United Kingdom, France, and Germany.
with Paget’s disease is sequestosome 1 (SQSTM1) encod- It is less frequent in Scandinavia, Spain, Italy, Central
ing the p62 protein.5 All mutations involving p62 identi- Europe, and Russia. The incidence in Australia and New
fied in Paget’s patients cause the loss of function of the Zealand is similar to that in Great Britain. Families with
C-terminal ubiquitin-associated (UBA) domain. These several generations affected by Paget’s disease have been
mutations result in elevation of NFκB in pagetic osteo- described. The rate of familial variants is unclear, and
clastic cells but are insufficient to induce full pagetic most authors report that less than 10% of cases have a
phenotype, and additional genetic as well as environmen- familial pattern. A mendelian-dominant pattern of trans-
tal factors are needed for full expression of the pagetic mission has been documented in some families.
phenotype. The scaffold p62 protein has specific interac- The onset of Paget’s disease is insidious, and the
tions that mediate activations of RANKL, a key regulator disease can be asymptomatic for many years. The full
of osteoclast function. The most common mutation, clinical picture with characteristic bone deformities
SQSTM1C1215T, causes amino acid substitution and is appears after 20 to 30 years. Cases with widespread
found in approximately 10% of sporadic and 30% of involvement of the skeleton and characteristic deformi-
familial Paget’s disease. Recently, SQSTM1 mutations ties are rare. It is estimated that there is 1 such case per
have been identified in patients with amyotrophic lateral every 100 patients with indolent asymptomatic disease.
sclerosis and frontotemporal lobar degeneration, neuro- The disease is rarely diagnosed in people younger than
degenerative disorders in which there is previously unrec- age 40 years. The disease in its fully developed clinical
ognized coexistence with Paget’s disease of bone.16 The picture is typically seen in patients in the sixth through
mutations in these disorders can overlap with those iden- eighth decades of life.
tified in Paget’s disease, but some of them appear to be
specific for the neurodegenerative disease. These find-
Clinical Symptoms
ings provide an unexpected link between amyotrophic
lateral sclerosis/frontotemporal lobar degeneration and The typical skeletal deformities of fully developed Paget’s
Paget’s disease of bone. disease consist of an enlarged head, outward bowed
The genetic linkage studies have identified seven femora, forward bowed tibiae, and bowed back. The long
predisposing loci that involve 1p12.3 (macrophage bones appear to be thickened on external examination. A
colony stimulating factor [M-CSF; CSF1]), 18q21.33 single bowed and enlarged long bone, such as the tibia
(RANK [TNFRSF11A]), 8q22.3 (dendritic-cell-specific or femur, may be the only clinical sign. In most such
transmembrane protein [DCSTAMP]), 10p13(optineurin cases, a radiographic examination documents the involve-
[OPTN]), 7q33 (nucleoporin 205κDa [NUP205]), 14q32 ment of additional bones. Clinically silent Paget’s disease
(Ras and Rab interactor 3 [RIN3]), 15q24 (promyelo- is most frequently identified on radiographs of the pelvis,
cytic leukemia [PML] and golgin A6 family, member sacrum, and lumbar spine. In general, the spine, pelvis,

ERRNVPHGLFRVRUJ
SQSTM1
Chr 5

Exon 1

Exon 2

Exon 3
Exon 4
Exon 5
Exon 6

Exon 7
Exon 8

Exon 9
15.3

cds
15.2
15.1
14
13.3
13.1 13.2
12
11 11.1
11.2

P392L
12
13.1
13.3 13.2 SQSTM1
14
15
21 1 440 aa
22
23.1
3 PB1 102 122 ZZ 167 LIR KIR 387 UBA 436
23.2
23.3 B
31.1
31.2
31.3
33.1 32
33.3 33.2
34
35.1 35.2
35.3 SQSTM1
A

PB1 Phox/Bem1p domain


ZZ Zinc finger type domain
LIR LC3-interacting region
KIR Keap1-interacting region
UBA Ubiquitin associated domain

Synonymous substitution
Missense substitution
Substitution - coding silent
Deletion frameshift

p65 NF-κB
Activation & IL-6 gene expression

NF-κB activation
1,25-(OH)2D3 signaling
D E
FIGURE 24-1  ■  Molecular mechanisms of Paget’s disease. A, Chromosomal location and exon-intron structure of the sequestosome
1 (SQSTM1) gene. B, Linear diagram of the p62 protein showing specific domains and motifs with the position of mutations. The
mutations involved in Paget’s disease of bone cluster in the ubiquitin-associated domain (UBA). C, The structural diagrams of the
p62 protein showing domains and motifs with selected mutations identified in amyotrophic lateral sclerosis (ALS), frontotemporal
lobar degeneration (FTLD), and Paget’s disease of bone. The diagram shows domain specific peptides: Phox and Bem1p domain
(PB1), LC3-interacting region (LIR), Keap1-interaction region (KIR), and UBA bound to their respective recognition proteins with the
highlighted mutations. Mutation sites common to both ALS/FTLD and PDB with (A390X, P392L, G411S, G425R) or very close (P387L)
to the UBA domain are highlighted. D, Molecular mechanisms activated by overexpression of MVNP in pagetic osteoclastic cells,
which increases TBK1, resulting in activation of NFκB, leads to nuclear translocation. MVNP also decreases Sirt1 deacetylation of
NFκB, leading to increased NFκB activity. In addition, TBK1 cooperates with MVNP to increase TAF12 and phosphoATF7, causing
hypersensitivity of VDR to 1,25(OH)2D3. E, Activation of TNFα and RANKL in Paget’s disease of bone is mediated by mutant p62.
Binding of TNFα activates RIP1 and TRAF6 by inducing K63-linked ubiquitination. The scaffold protein p62 interacts with RIP1 and
TRAF6, leading to downstream activation of NFκB with complementary biologic effects to MVNP. Mutant p62 has faulty interactions
with intermediary Cyld protein and does not associate with RIP1 and TRAF6, leading to decreased signal attenuation resulting in
hyperactive NFκB. Both MVNP and mutant p62 hyperactivation of NFκB in pagetic osteoclastic cells increase IL-6 and ephrinB2 in
osteoclasts and cause ephrinB4 dependent hyperactivation of osteoblasts resulting in increased bone formation. (C, Modified and
reprinted with permission from Rea SL, Majcher V, Searle MS, et al: Exp Cell Res 325:27–37, 2014. D and E, modified and reprinted with
permission from Galson DL, Roodman GD: J Bone Metab 21:85–98, 2014.)
ERRNVPHGLFRVRUJ
1440 24  Precancerous Conditions

28

Peak age 31
incidence
of Paget's
sarcoma
11
1

76
Incidence

14
Peak age 31
incidence
of Paget's
disease

1 2 3 4 5 6 7 8
Age in decades 14

FIGURE 24-2  ■  Paget’s disease and sarcomatoid transformation. Skeletal sites most frequently involved by Paget’s disease are indi-
cated by shaded areas. Numbers show skeletal distribution of 208 published cases of Paget’s sarcoma. Note that sarcomatous
transformation predominantly occurs in sites frequently affected by Paget’s disease.

skull, and femur are considered the most frequently Bowing deformities and pathologic fractures are typically
affected parts of the skeleton (Fig. 24-2). The tibia and seen in weight-bearing sites.
the humerus are less frequently affected.
Microscopic Findings
Radiographic Imaging
Microscopically, the initial early phase of the disease
Radiographically, the lesions present as areas of irregular shows increased osteoclastic activity with fibrosis and
increased density with a cotton wool–like appearance prominent vascularization of the intertrabecular spaces.
interspersed by radiolucent areas1,11 (Fig. 24-3). More The fully developed, active phase of Paget’s disease is a
consolidated and larger areas of sclerosis can also be mixture of osteoclastic activity that may lead to the for-
present. The bones are usually enlarged, and their normal mation of large clusters adjacent to Howship’s lacunae
shape or contour is altered by the development of promi- (Fig. 24-9). In addition, prominent osteoblastic activity
nent bony deformities (Fig. 24-4). In the long bones, the results in the production of new osteoid. Bone in Paget’s
disease typically starts within the end portion and pro- disease represents irregular trabeculae with scalloped
gresses toward the shaft (Fig. 24-5). When the disease is contours and irregular lines of mineralization that mimic
confined to the epiphyses of long bones, it can be con- a mosaic pattern. The end of the last phase is dominated
fused on radiographs with other conditions such as giant by large areas of bone sclerosis in which multiple irregu-
cell tumor. Radiolucent areas represent younger, active lar lines of mineralization are present. Typically, different
foci. Patchy sclerosis is the intermediate phase. More phases of the disease are present in different areas of the
consolidated larger areas of sclerosis represent the final affected bone (Figs. 24-9 to 24-11).
stages of the process. Early lesions of the skull represent
well-demarcated circular lytic areas, referred to as osteo-
Malignant Transformation
porosis circumscripta (Figs. 24-6 and 24-8). Characteristi-
cally, in areas with paired bones such as the forearm or The development of bone sarcoma in this condition is
leg, only one bone is altered (Fig. 24-7). The other bone the most serious complication and, although uncommon,
either is intact or shows only minimal involvement. Text continued on p. 1450

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1441

B
FIGURE 24-3  ■  Paget’s disease of skull: radiographic features. A and B, Advanced Paget’s disease of skull presenting as large, ill-
defined lytic area with cotton wool–like opacities.

ERRNVPHGLFRVRUJ
1442 24  Precancerous Conditions

A
FIGURE 24-4  ■  Paget’s disease of bone: radiographic features. A and B, Lateral radiographs of two examples of Paget’s disease
involving tibia. Note anterior bowing deformity and coarse trabecular pattern with obscured corticomedullary demarcation in both
radiographs. Incomplete transverse fracture is seen in proximal tibial shaft in B.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1443

B
FIGURE 24-5  ■  Paget’s disease of humerus: radiographic features. A and B, Lytic lesion involving proximal end of humerus. Note sharp
demarcation of advancing edge. Lesion is associated with pathologic fracture and was considered to represent giant cell tumor.

ERRNVPHGLFRVRUJ
1444 24  Precancerous Conditions

B
FIGURE 24-6  ■  Paget’s disease of skull: radiographic features. A, Early phase of Paget’s disease in skull presents as well-delineated
lytic area referred to as osteitis circumscripta (arrows). B, Progression of lesion. Note large advancing lytic edge at periphery (arrows)
and development of bony opacities in central, older areas of lesion (asterisk).

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1445

A B

C D
FIGURE 24-7  ■  Paget’s disease of long bones: radiographic features. A, Early localized phase of Paget’s disease of radius with patho-
logic fracture (arrows). B, Early predominantly lytic phase of Paget’s disease of radius. Note involvement of only one bone of pair
and bony deformity of affected bone. C and D, Fully developed phase of Paget’s disease with prominent osteoclastic activity and
irregular scalloped trabeculae of bone. Osteoblastic activity is prominent focally. Note that osteoclasts are markedly enlarged and
occur in clusters. (C and D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1446 24  Precancerous Conditions

A B

C D
FIGURE 24-8  ■  Paget’s disease: gross, radiographic, and microscopic features. A, Autopsy specimen showing advanced Paget’s
disease of skull. B, Early phase of Paget’s disease in skull presents as lytic area refered to as osteitis circumscripta. C, Sclerotic
phase of Paget’s disease shows cement (reversal) lines in mosaic pattern. D, Specimen radiograph of slice of parietal bone with
marked thickening and multiple cotton wool opacities that form larger sclerotic areas. Note the obliteration of corticomedullary
borders. (C, ×50) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1447

A B

C D
FIGURE 24-9  ■  Paget’s disease: microscopic features. A, Active “hot” phase with extensive osteoclastic bone resorption and fibrous
replacement of marrow. B, At higher magnification, osteoclast can be seen resorbing bone; this is early phase of Paget’s disease.
Note fibrovascular stroma. C, Mosaic pattern in thickened bone trabeculae of late-stage Paget’s disease. D, Higher magnification
shows complex pattern of cement (reversal) lines in mosaic pattern. (A and C, ×50; B, ×200; D, ×100) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1448 24  Precancerous Conditions

A B

C D
FIGURE 24-10  ■  Paget’s disease of bone: microscopic features. A and B, Microscopic features of Paget’s disease in region of active
“hot” phase include scalloped bone trabeculae, increased osteoclastic activity, and intertrabecular fibrosis. C, Higher magnification
reveals osteoclasts in Howship’s lacunae. D, Sclerotic phase of Paget’s disease is characterized by thickened bone trabeculae and
irregular mosaic lines of mineralization. (A, B, and D, ×50; C, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1449

A B

C D
FIGURE 24-11  ■  Paget’s disease of bone: radiographic and microscopic features. A, Specimen radiograph of slice of parietal bone
shows marked thickening with multiple cotton-wool opacities that fuse and form larger sclerotic areas. Note indistinct corticomedul-
lary border. B, Microscopic features of Paget’s disease in region of active “hot” phase include scalloped bone trabeculae, increased
osteoclastic activity, and intertrabecular fibrosis. C, Higher magnification reveals osteoclasts in Howship’s lacunae. D, Sclerotic phase
of Paget’s disease is characterized by thickened bone trabeculae and irregular mosaic lines of mineralization. (B and D, ×50; C, ×200)
(B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1450 24  Precancerous Conditions

accounts for 20% of the bone sarcomas occuring in and facial bones, and although benign, such tumors can
patients older than age 40 years.21 The incidence of be locally destructive and extend into soft tissues. The
Paget’s sarcoma increases steadily with age until the tumors may have the histologic appearance of giant cell
seventh decade of life,14 and the rate of sarcomatous tumors of bone, but some authors have emphasized the
transformation ranges between 1% and 10% depending resemblance to giant cell reparative granuloma.11 Many
on the extent and severity of the disease. Data from the patients with these types of tumors have a common
Surveillance, Epidemiology, and End Results program ancestry from Avellino, Italy.20 It is still not clear whether
confirm this epidemiologic pattern of incidence and show genetic factors, environmental factors, or both play a role
that the overall incidence rate of Paget’s sarcoma is in the link between patients and the development of this
approximately 0.1 to 0.2 per 100,000.3 In patients older unusual neoplasm.
than age 50 years, it is significantly higher and varies from
0.4 to 0.7 per 100,000. Because so many cases of Paget’s
disease are asymptomatic, the interval between onset and OSTEOMYELITIS
the development of a sarcoma is uncertain. Sarcoma com-
plicating Paget’s disease is typically diagnosed in patients Long-standing inflammation with intermittent sinus
older than age 50 years.3 Clinically, the most common tract formation in chronic osteomyelitis and reactive
presenting symptoms are a progressive increase of local- hyperplasia of the squamous epithelium provides the
ized pain and a palpable mass; less often, a pathologic setting for malignant transformation in approximately
fracture is the first symptom. Serum levels of alkaline 0.5% of patients affected by this condition. Therefore the
phosphatase can be increased compared with previous risk of malignant transformation in chronic osteomyelitis
levels. The sites most frequently involved by sarcomatous is relatively small. The tibia is most frequently affected
changes are the pelvis, humerus, and femur. Overall the by chronic osteomyelitis and consequently is the typical
distribution of sarcomatous changes parallels the distri- site of secondary malignancy in this setting (Fig. 24-18).
bution of skeletal sites involved by the disease (i.e., the A tumor mass or an ulceration with indurated borders
bones frequently altered by Paget’s disease are likely to typically develops within the sinus tract and may extend
develop sarcomatous transformation) (Fig. 24-2). Typi- down into the bone (Figs. 24-19 and 24-20). Less fre-
cally the bone involved by sarcoma also exhibits both the quently, squamous cell carcinoma develops in the epithe-
radiographic and gross features that are characteristic of lialized lining of the bone defect.24
Paget’s disease (Figs. 24-8 and 24-12). Atypical pseudoepitheliomatous hyperplasia heralds
Radiographically, the predominant tumor pattern is the transformation into squamous cell carcinoma. Some-
osteolytic.9,19 Mixed and osteoblastic patterns occur less times it is difficult to separate the two conditions in the
frequently. Cortical breakthrough without periosteal limited, small, and superficial biopsy material. Differen-
reaction and bulky extension into soft tissue are other tial diagnosis between florid pseudoepitheliomatous pro-
signs of malignancy (Figs. 24-13 and 24-14). Computed liferation and well-differentiated squamous cell carcinoma
tomography and magnetic resonance imaging may be is the common problem in evaluating sinus tract biopsy
more sensitive for the early detection of sarcomatous specimens from patients with chronic osteomyelitis (Figs.
degeneration. 24-21 and 24-22). Some authors advocate amputation as
The most common histologic type of Paget’s sarcoma an appropriate treatment for both conditions; it is fol-
is osteosarcoma (Figs. 24-15 and 24-16), although fibro- lowed by evaluation of large tissue sections from postop-
sarcoma, chondrosarcoma (Fig. 24-17), malignant giant erative material to distinguish between the two conditions
cell tumor, and malignant fibrous histiocytoma can also with certainty. Regional lymph nodes are often enlarged
occur.6,7,9 Because of the variability in histologic patterns, as a reaction to the inflammatory process, but metastasis
some authors designate these tumors only as Paget’s occurs in only 10% to 20% of cases. However, this rate
sarcomas. is high enough to require that the surgical approach must
The prognosis is poor for the majority of patients. The include lymph node biopsies.23 Tumor histology and
5-year survival rate for osteosarcoma is 8% compared regional lymph node involvement are related to survival.
with the current 50% to 65% 5-year survival rate for Pulmonary and visceral metastases are exceedingly rare
de novo osteosarcoma. In addition to the higher grade in this form of secondary malignancy.
of the tumors, the fact that these patients are in an Squamous cell carcinoma is the most common type of
older age group that is typified by decreased immunity, malignant tumor that evolves at the site of chronic osteo-
poor general health, and low tolerance for conventional myelitis, and the latency period is between 20 and 50
chemotherapy and radiotherapy may account for this dis- years. The lower extremities, in particular the tibial
crepancy.21 In addition, the unusual anatomic location of region, are most commonly affected.24
the tumors, such as the axial skeleton and craniofacial Clinical signs that should arouse suspicion about
bones, accounts for the difficulty in treatment by com- malignant transformation include increased pain with
plete resection. The most frequent cause of death is foul or bloody discharge from the sinus, a progressively
distant metastasis. Multicentric Paget’s sarcoma has been enlarging mass in and around the sinus tract opening, and
described and interpreted by some as metastases; others progressive bone destruction.27
believe that each site is a separate primary tumor. Multiple biopsies are advisable for adequate assess-
Benign giant cell tumors have also been described in ment of the affected tissues. The histologic features of
association with Paget’s disease (see Chapter 10). The malignancy include cellular atypia, abnormal mitosis,
most common locations for these tumors are the skull Text continued on p. 1462

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1451

A B

C D
FIGURE 24-12  ■  Paget’s sarcoma: gross features. A, Destructive tumor mass of proximal humerus with pathologic fracture. B, Tan-
white tumor with matrix mineralization (histologically osteosarcoma) of distal femoral end with circumferential extension into soft
tissue. Note thickened sclerotic bone proximally. C, Resected femoral head with pathologic fracture. Note intramedullary tan-gray
tumor mass. D, Destructive tumor mass with pathologic fracture of femoral shaft. Note diffuse sclerotic change in femoral shaft,
feature consistent with Paget’s disease.

ERRNVPHGLFRVRUJ
1452 24  Precancerous Conditions

B
FIGURE 24-13  ■  Paget’s sarcoma of skull: radiographic features. A, Magnetic resonance image of skull shows destructive mass in
right frontal area. Note thickened sclerotic bone of skull. B, Computed tomogram shows bone destruction with extension into soft
tissue of right frontal area. Note thick sclerotic bones of skull and multiple coalesced opacities.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1453

B C

D
FIGURE 24-14  ■  Paget’s sarcoma: radiographic, gross, and microscopic features. A, Advanced Paget’s disease of tibia with bowing
deformity. Note absence of involvement of fibula. Destructive process of proximal tibial end extends into soft tissue, and cloudy
opacities within tumor mass extend into popliteal soft tissue (arrows). Histologically, this tumor was high-grade osteosarcoma.
B, Extensive involvement of tibia by Paget’s disease. Note large mass that extends into soft tissue at proximal end; this is consistent
with sarcomatous transformation. C, Sagittal section of amputated specimen shows abnormal sclerotic tibia with destructive min-
eralized mass at its proximal end. D, Microscopically the tumor is high-grade osteosarcoma (D, ×200) (D, hematoxylin-eosin).

ERRNVPHGLFRVRUJ
1454 24  Precancerous Conditions

B C

FIGURE 24-15  ■  Paget’s disease with secondary osteosarcoma. A, Low power photomicrograph of osteosarcoma associated with
Paget’s disease. B, Low power photomicrograph of sclerotic tumor with a solid area of osteoid showing different level of mineraliza-
tion juxtaposed on the preexisting host bone. C, Higher magnification showing atypical osteoblastic cells and tumor osteoid deposi-
tion. (A, ×50; B, ×100; C, ×400) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1455

A B

C D
FIGURE 24-16  ■  Paget’s disease with secondary osteosarcoma. A, Lateral radiograph of an 85-year-old man with long-standing
Paget’s disease of humerus and osteosarcoma destroying distal end of bone; soft tissue involvement is apparent. B, Sclerotic bone
with irregular mosaic lines of mineralization consistent with sclerotic phase of Paget’s disease adjacent to tumor. C and D, Photo-
micrographs of high-grade osteoblastic osteosarcoma shown in A. Note lacelike osteoid pattern and nuclear anaplasia. (B, ×50;
C and D, ×100.) (B-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1456 24  Precancerous Conditions

A B

C D
FIGURE 24-17  ■  Chondrosarcoma, grade ii secondary to Paget’s disease of femur. A, Lateral radiograph of knee of a 66-year-old man
with Paget’s disease of distal femur and tibia who had secondary chondrosarcoma in femur. B, Amputation was performed after
biopsy revealed presence of malignant cartilage tumor that broke out of bone and invaded soft tissue and synovium of knee joint.
C, Close-up view of B. Note that tumor extends across joint to invade tibia, which is also affected by Paget’s disease. D, Photomi-
crograph of tumor shown in C reveals grade 2 myxoid chondrosarcoma. (D, ×400) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1457

A
FIGURE 24-18  ■  Chronic osteomyelitis: radiographic features. A, Anteroposterior radiograph shows sclerotic changes of tibial shaft
and lateral bowing deformity. B, Lateral radiograph shows sclerosis of tibial shaft and anterior cortical deformity corresponding to
draining fistula.

ERRNVPHGLFRVRUJ
1458 24  Precancerous Conditions

A B

C D
FIGURE 24-19  ■  Squamous cell carcinoma associated with chronic osteomyelitis. A, Clinical photograph of draining fistulas of tibia.
B, Bisected tibia with several bone defects and draining tracts. C, Higher magnification of B shows two fistulas connected to tibial
bone defects. D, Histologic section of bone in vicinity of larger bone defect shows infiltrating well-differentiated squamous cell
carcinoma. (D ×100, hematoxylin-eosin)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1459

B
FIGURE 24-20  ■  Squamous cell carcinoma in association with chronic osteomyelitis. A, Coronally bisected specimen from below-knee
amputation. B, Higher magnification of A shows area of fistulous tract with destructive cheesy mass consistent with keratinized
squamous cell carcinoma.

ERRNVPHGLFRVRUJ
1460 24  Precancerous Conditions

B
FIGURE 24-21  ■  Chronic osteomyelitis: microscopic features of pseudoepitheliomatous hyperplasia. A, Low power magnification of
hyperplastic squamous epithelium lining fistulous tract. B, Higher magnification of A. Note preserved maturation pattern of squa-
mous epithelium and absence of nuclear atypia. Tongues of hyperplastic epithelium have smooth regular borders and gradually
merge with overlapping epithelium. (A ×50; B ×100) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1461

FIGURE 24-22  ■  Pseudoepitheliomatous hyperplasia versus well-differentiated squamous cell carcinoma: microscopic features.
A, Pseudoinfiltration pattern of stroma in hyperplastic epithelium. Note smooth outlines of epithelial nests and their orderly inter-
connecting arrangement. B, Well-differentiated squamous cell carcinoma. Note irregular outlines of disorderly arranged tumor cell
nests and presence of keratinized pearls within tumor cell nests. (A and B, ×100) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1462 24  Precancerous Conditions

lymphatic permeation, and invasion into blood vessels. in the fourth and fifth decades of life (5 cases each), fol-
Abnormal maturation patterns that show formation of lowed by the sixth and seventh decades (4 cases each).
squamous pearls deep within the infiltration tongues There were 13 men and 9 women. Except for 1 patient
of squamous epithelium help distinguish squamous car- whose race was unknown, 11 were white patients and 10
cinoma from pseudoepitheliomatous hyperplasia (Fig. were black patients.
24-23). The diagnostic difficulties are amplified by the The duration of symptoms of our 22 cases ranged
irregular shapes of the fistulous tracts, which cause the from 1 month to 2 years (average, 6 months). The most
squamous lining to be cut tangentially on histologic sec- frequent presenting symptom was local pain or tender-
tions. Moreover, squamous cell carcinomas that develop ness, in 15 of 19 patients in whom symptoms were
in sites of chronic osteomyelitis frequently tend to be recorded; pain was unassociated with other symptoms in
moderately to well-differentiated lesions that on small 7 patients, whereas 5 reported associated swelling. Seven
biopsy samples can easily be confused with benign squa- patients had pathologic fracture, in 4 of whom it was the
mous epithelium proliferation. presenting symptom.
In addition to squamous cell carcinoma, other tumors Fifteen (68%) of the patients had no medical condition
have been described in association with osteomyelitis. or work history associated with bone infarction; 7 had
These include basal cell carcinoma, adenocarcinoma, such a condition or history, with 3 having sickle cell
myeloma, fibroblastic osteosarcoma, angiosarcoma, rhab- disease or trait, 2 with decompression bone disease
domyosarcoma, and lymphoma.22,24-26 Because the sarco- (caisson disease), 1 with alcoholism, and 1 with Gaucher’s
mas metastasize in 56% of cases, they have a worse disease. In the 2 cases of caisson disease, the interval
prognosis than that for secondary carcinoma.23 between the last exposure to decompression and the clini-
cal manifestation of the sarcoma was 17 and 25 years,
respectively.
Skeletal distribution of bone sarcomas associated with
BONE INFARCTS infarcts in our 22 cases is shown in Figure 24-24. The
Definition area around the knee was the site of tumor in 13 (59%)
of the patients; the distal femur was involved in 6 patients
Bone infarction is a relatively common lesion that typi- and the proximal tibia in 7. Most lesions were located in
cally occurs in the metaphyseal region of the long bones, the metaphysis; one each occurred in the diaphysis of the
often around the knee. The terms aseptic and avascular humerus and the femur.
necrosis are generally applied to areas of epiphyseal or
subarticular involvement, as commonly seen in the
Radiographic Imaging
femoral head.
A variety of conditions predispose to bone infarction, On radiographs, chronic bone infarcts typically appear as
including alcoholism; corticosteroid therapy; renal dialy- irregular but sharply demarcated intramedullary densities
sis; exposure to compressed air, such as occurs in caisson in the metaphyseal or metadiaphyseal region (Figs. 24-25
workers and divers; Legg-Calvé-Perthes disease; hemo- to 24-28). Serpiginous or wavy rims and coils of calcific
globinopathies, especially sickle cell disease and trait; density are characteristic of bone infarcts. Oval shadows
Gaucher’s disease; chronic pancreatitis; gout; pregnancy; outlined by a thin radiopaque zone of margination can
exposure to radiation; and collagen or vascular disorders. be present in some cases.
Although the majority of the reported cases of infarct- Thirteen (59%) patients in our series had radiographic
related sarcomas have been associated with idiopathic evidence of infarcts that affected multiple bones; 9 (41%)
bone infarcts, those found in 37% of our patients stemmed had a single infarct only. Of patients with multiple
from some prior medical or occupational cause, such as infarcts, 7 had symmetric infarcts in the opposite bone
exposure to compressed air or rapid decompression,33,42-44 without associated tumor.
sickle cell disease or trait,34,45 alcohol abuse,34,40 or previ- Secondary sarcoma appears as irregular areas of
ous steroid therapy.46 One patient had development of destruction of the medullary and cortical bones and has
an angiosarcoma in a bone infarct; this patient had a a permeative margin within or at the edge of the infarct
history of Hodgkin’s disease and had undergone chemo- that is usually accompanied by a soft tissue mass, with or
therapy.29 Arnold30 described a family with hereditary without pathologic fracture (Figs. 24-25 to 24-28). An
bone infarcts, and two family members had associated associated periosteal reaction can be present. Poorly
fibrosarcomas. It is difficult to assess the real risk of defined mineralization can be seen in matrix-producing
malignant transformation associated with bone necrosis lesions such as osteosarcoma.
because many bone infarcts are asymptomatic. Based on The computed tomographic scans available in two of
the relatively low number of published cases, overall risk our cases showed both a well-defined endosteal calcific
of development of sarcoma in these settings appears to shell and focal disruption of the cortex with a soft tissue
be low.32,38 mass. Bone scans reveal increased uptake of isotope in the
regions corresponding prominently to the tumor and, to
a lesser degree, the bone infarct. Specimen radiographs
Clinical Findings
disclose ill-defined, irregular, lytic lesions of the metaph-
We have seen 22 cases of bone sarcomas associated with ysis with focal areas of calcification at the periphery,
infarcts. The ages of our patients ranged from 28 to 82 which correspond to bone infarct.
years (average age, 51.5 years). The highest incidence was Text continued on p. 1469

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1463

C
FIGURE 24-23  ■  Squamous cell carcinoma developing within fistulous tract of chronic osteomyelitis. A, Invasive squamous cell
carcinoma in fistulous tract. B, Higher magnification of A shows well-differentiated squamous cell carcinoma mimicking pattern
seen in pseudoepitheliomatous hyperplasia. C, Higher magnification of B shows abnormal maturation pattern with keratinized pearls
in deeper invasive portion of the lesion. (A, ×5; B, ×15; C, ×100) (A-C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1464 24  Precancerous Conditions

6 1
Males 13

Females 9
5
TOTAL 22

4
Incidence

2
3
1

2 6

7
1

0
3
1 2 3 4 5 6 7 8 9
Age in decades
FIGURE 24-24  ■  Sarcomas associated with bone infarcts. Age-specific and skeletal distribution patterns of 22 sarcomas associated
with bone infarcts.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1465

A B

D
C

FIGURE 24-25  ■  Sarcoma associated with bone infarcts: radiographic features. A and B, Anteroposterior and oblique views on plain
radiographs of knee show extensive bone infarct. Note lytic, ill-defined area involving lateral condyle. Biopsy material from this
region shows high-grade malignant fibrous histiocytoma. C, Bone scintigram shows increased uptake, predominantly in distal
femoral metaphysis. D, Microscopically tumor is malignant fibrous histiocytoma (D, ×100) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1466 24  Precancerous Conditions

C
FIGURE 24-26  ■  Sarcoma associated with bone infarcts: radiographic and microscopic features. A and B, Plain radiographs show
extensive multifocal infarcts of femur and tibia. Note destructive lesion with cortical disruption in proximal tibial metaphysis (better
seen in B [arrows]). C, Biopsy material shows high-grade malignant fibrous histiocytoma (C, ×100) (C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1467

A B

C D

FIGURE 24-27  ■  Sarcoma associated with bone infarcts: radiographic features. A, Specimen radiograph shows fracture through distal
femoral metaphysis with geographic mineralization pattern consistent with infarct. B, Coronally bisected amputation specimen
shows pathologic fracture through destructive tumor mass. Fibrous and mineralized pattern in distal fragment corresponds to infarct.
C, Multiple symmetric bone infarcts of femur and tibia were present in contralateral lower extremity of same patient. D, Microscopi-
cally, tumor represents malignant fibrous histiocytoma (D, ×100) (D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1468 24  Precancerous Conditions

A C
FIGURE 24-28  ■  Sarcoma associated with bone infarct: radiographic and microscopic features. A, Specimen radiograph of distal
femur with geographic mineralization pattern consistent with infarct and a large destructive intramedullary mass penetrating the
cortex and extending to the soft tissue. Inset, Specimen radiograph of the tibia from the same amputation specimen with geographic
mineralization pattern consistent with infarct. B, Low power photomicrograph showing interface between bone infarct and associ-
ated with high-grade sarcomatoid neoplasm. C, Microsocopically, tumor represents malignant fibrous histiocytoma. (B, ×25; C, ×100)
(B and C, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1469

Gross Findings infarcts seems to have a poorer prognosis and to affect


older patients.
Bone infarcts appear as sharply demarcated, irregular,
yellow-white zones in medullary bone and are sometimes
Personal Comments
accompanied by fibrous-walled, cystlike structures and
areas of cystic fat necrosis. Infarct-associated tumors Men seem to be more affected by bone infarcts associated
range in size from 2 to 13 cm (mean, 6.7 cm). The tumors with sarcomas than women. This difference can be attrib-
generally have ill-defined, gray-tan, firm, and rather uted to the fact that many conditions that predispose to
gritty appearances that blend into the surrounding med- bone infarction are predominantly related to male-
ullary tissue and often erode or destroy the cortex while oriented occupations or propensities, such as tunnel
infiltrating into the adjoining muscle and fascia. Hemor- workers, divers, and heavy alcohol abusers. Black patients
rhage and necrosis are usually obvious. account for a large proportion (48%) of cases, partly
because of the association between infarcts and sickle cell
Microscopic Findings disease and trait. The ages of previously reported patients
ranged from 18 to 82 years (mean, 53.7 years). Malignant
Histologic examination of the bone infarcts shows irregu- fibrous histiocytoma is the most frequent infarct-related
lar fragments of necrotic bone trabeculae and intervening sarcoma in our series, as well as in previously reported
hyalinized fibrous tissue and fat necrosis with a variable cases.28,31,34-37,40-45 As in individuals who have malignant
amount of dystrophic calcification (Fig. 24-29). Cysts fibrous histiocytomas that develop in bones affected
lined by loose fibrous tissue, old focal hemorrhage, and by preexisting conditions (low-grade chondrosarcoma,
calcified debris are also observed. Abnormal cells provide Paget’s disease, or previous irradiation), patients with
a clue to the predisposing factor, such as foamy histio- sarcoma secondary to bone infarct are generally older
cytes in Gaucher’s disease (Fig. 24-30) or sickled eryth- than those in whom sarcomas develop de novo with no
rocytes in patients with sickle cell disease. underlying condition.
The peripheral areas of bone infarction in the transi- The increased radiographic density of chronic bone
tion zone adjacent to frank sarcoma contain focal resorp- infarcts is probably due to a combination of heavy calcium
tion of dead bone trabeculae; layering of new viable deposition in necrotic marrow and an increased amount
bone on the dead trabecular surfaces; and formation of reactive bone surrounding the necrotic component.
of granulation tissue composed of scattered infiltrates Such a radiographic appearance may sometimes mimic
of chronic inflammatory cells, proliferation of small that of calcified enchondroma. However, the foci of calci-
vessels, and cellular fibrous tissue with occasional atypical fied matrix in enchondroma are discrete and scattered
spindle cells. Atypical spindle cells, whose nuclei are diffusely throughout the lesion and have punctate to pop-
somewhat larger and hyperchromatic, are present in cornlike densities; the margin of the lesion is not so
these areas. The cellularity of reparative tissue ranges clearly outlined as that of an infarct. Approximately 75%
from isolated cells to relatively hypercellular with inter- of the patients with infarct-associated sarcomas have
lacing spindle-cell fascicles. The fascicles of these cells multiple infarcts, usually in the bone symmetrically
are sometimes arranged in a storiform pattern with des- opposite the tumor-bearing bone.
moplastic collagen. Farther from the infarct, atypical The proposition that infarct-associated sarcoma arises
spindle cells merge into a high-grade, spindle-cell sarco- as a result of prolonged excessive activity or a high degree
matous component. of chronic proliferative activity of reparative tissue adja-
Infarct-associated sarcomas show the typical micro- cent to the infarct has been questioned by some authors.48
scopic features of malignant fibrous histiocytoma, It has been suggested that large infarcts are not totally
low-grade osteosarcoma, and conventional high-grade replaced because of the eventual cessation of the repara-
osteosarcoma.18,36,39,40,44 Low-grade osteosarcomas associ- tive process. No studies have documented excessive or
ated with bone infarcts are mostly composed of interlac- persistent repair adjacent to metaphyseal or diaphyseal
ing bundles of spindle cells interspersed with strands of infarcts. On the other hand, scintigraphy shows intense
osteoid or mineralized woven bone and a small number isotope uptake not only in the sarcomatous areas, but also
of associated osteoclast-like giant cells. The tumor to a lesser degree in the infarcted lesions. We have
cells have uniform nuclei with only slight atypia and observed cases of multiple bone infarcts that were not
rare mitotic figures. Malignant fibrous histiocytoma associated with secondary sarcomas in which “hot” lesions
and osteosarcoma associated with bone infarcts do not (those with increased uptake) corresponded to chronic
differ microscopically from their conventional de novo bone infarcts. The reparative process surrounding bone
counterparts. infarction has been described in association with revas-
cularization and increased accumulation of the radioiso-
tope.47 On the basis of the microscopic evidence from our
Treatment and Behavior
studies with proliferative markers (i.e., proliferating cell
Among 12 of the 22 patients whose follow-up data were nuclear antigen) and scintigraphic evidence, the active
available, 9 (75%) died of metastases from 2 months to 5 repair process adjacent to bone infarcts appears likely to
years after diagnosis (mean, 19 months), and 3 (25%) continue in some instances, although the cause of its
were alive and well from 9 to 19 years (mean, 15.5 years) persistence is unclear.
after treatment. As is the case with tumors secondary to The exact reason for the preponderance of malig-
other underlying conditions, sarcoma arising in bone nant fibrous histiocytoma as the tumor most commonly

ERRNVPHGLFRVRUJ
1470 24  Precancerous Conditions

A B

C D
FIGURE 24-29  ■  Sarcoma associated with bone infarct: microscopic features. A and B, Periphery of infarct shows regenerative granu-
lation tissue adjacent to hyalinized tissue that has dystrophic calcification. High-grade sarcomatoid neoplasm is seen in the upper
half of the photomicrograph. C, Example of malignant fibrous low-grade fibroblastic osteosarcoma associated with bone infarct.
D, High-grade pleomorphic variant of malignant fibrous histiocytoma associated with bone infarct infiltrating fatty bone marrow.
(A and B, ×50; C, ×100; D, ×200.) (A-D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1471

B
FIGURE 24-30  ■  Malignant fibrous histiocytoma associated with Gaucher’s disease: radiographic and microscopic features. A, Low-
power magnification of spindle-cell neoplasm is consistent with malignant fibrous histiocytoma. B, Higher-power magnification of
A shows spindle tumor cells with prominent atypia. Insets, Anteroposterior and oblique views on plain radiographs show lytic mass
of proximal tibial shaft. Note increased mineralization pattern of bone adjacent to lytic area corresponding to bone infarcts. Gaucher’s
disease was diagnosed in early childhood. (A, ×100; B, ×200) (A and B, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1472 24  Precancerous Conditions

associated with bone infarcts and the relative infrequency metastatic sarcomatous malignancy.62,65 Radiographically
of osteosarcoma and angiosarcoma is unknown. The and microscopically, these tumors do not differ from
tumors expected to develop, however, would reflect the their conventional de novo counterparts (Figs. 24-33
cell types involved in such a reparative process (i.e., fibro- and 24-34).
blasts, osteoblasts, and endothelial cells). Thus it seems A recent report of 20 examples of malignant fibrous
clear that a close pathogenetic relationship exists between histiocytomas developing as induced sarcomas in bone
bone infarcts and the associated sarcomas. The repara- noted that a median dose of 5700 rads was given for
tive, predominantly fibroblastic tissue, which has a rela- either nonosseous conditions or preexisting skeletal
tively high degree of proliferative activity in the border lesions.64
zone, could be the presumptive origin of this malignancy. A latency period of 2.75 to 13 years separated the
irradiation from the appearance of a malignant tumor in
one series,68 but some authors have recorded delays of up
METALLIC IMPLANTS to 30 years. In a series of 91 bone sarcomas that devel-
oped in patients treated with radiotherapy with other
Malignant tumors reported to arise at the site of metallic modalities for previous malignancies and in patients who
implants are exceedingly rare, with only 12 cases recorded had genetic predispositions, the latency period was short-
up to 1990.49,51,53-55,60 Some authors believe that the asso- ened by the use of adjuvant chemotherapy.68 Typically
ciation is coincidental and that the risk of inducing postradiation sarcoma develops 5 to 10 years after expo-
neoplasia is minimal.52 Osteosarcoma, Ewing’s sarcoma, sure (Figs. 24-33 and 24-34).
hemangioendothelioma, malignant fibrous histiocytoma, Internal deposits of radioactive elements of radium
lymphoma, and synovial sarcoma have been associated and mesothorium from industrial exposure have been
with metallic implants.53-56,60 Experimental investigations found in painters of watch dials, chemists, and techni-
support the carcinogenic effect of heavy metals such as cians. Internal deposits of radium salts and thorium have
cobalt, cadmium, and nickel in a possible cause-and- been found in some individuals who use them in medical
effect relationship for neoplastic induction by orthopedic work. Once in the body, these substances behave like
devices composed of metals.56-58 These observations are calcium, depositing in areas of active bone turnover.
of concern when considering the long-term effect of Their deposition predisposes the bone to the develop-
metallic implants. ment of osteosarcoma and other malignancies. Bone
The interaction between the body’s saline environ- damage, including osteopenia, necrosis, spontaneous
ment and the implanted metal sets the background for fractures, and osteosarcoma, results from many decades
potential production of corrosive products. The rate of of constant irradiation of bone cells and matrix and
corrosion varies according to particle size; metal solubil- mineral formation affected by the α-rays emitted from
ity; environmental pH; and concentrations of the metal, these deposits.61 The anatomic sites of the induced tumors
negative ions, and oxygen. The continuous exposure of are evenly distributed and frequently are in the skull,
tissue to metallic corrosion products is considered to be pelvis, and short tubular bones. Multiple neoplasms may
an initiating factor.50,59 Radiographically, these tumors occur.
present as destructive masses in the vicinity of metallic
implants (Figs. 24-31 and 24-32).
OSTEOGENESIS IMPERFECTA
RADIATION INJURY Osteogenesis imperfecta is an inherited disorder of
connective tissue in which deficient osteoid formation
The bone damage produced by ionizing radiation from leads to multiple fractures. It has a complex genetic
either internal or external sources is often referred to as background with 17 genetic causes transmitted in auto-
radiation osteitis. The necrotic bone in this condition leads somal dominant or recessive patterns.78 Osteogenesis
to reparative changes characterized by a highly cellular imperfecta patients are classified in four distinctive
proliferation of fibroblastic tissue and reactive new bone syndromes with different clinical characteristics and
formation. Unlike the reparative tissue adjacent to simple inheritance patterns. Mutations of collagen 1A (COL1A2)
bone infarcts, atypical mesenchymal cells with pleomor- genes encoding collagen alpha-1 chains play a major
phic and hyperchromatic nuclei are present. It is from role in the development of this disorder. In radiographic
this substrate that sarcomas may develop after exposure and microscopic diagnosis of bone tumors, the paradoxi-
to radiation.61,63,66 The likelihood of sarcomatous change cal predisposition to form exuberant fracture callus
is directly related to the number of irradiation courses that may be mistaken for osteosarcoma is of major
and the absorbed dose of radiation in bone. With the importance.69,71,72,76,79,80
dose range used in modern radiotherapy (4000 to 7000 Few case reports have dealt with the association
rads), the risk for sarcomatous transformation is low.67 of osteogenesis imperfecta and osteosarcoma.70,73-75,77
The histologic types of induced sarcoma include osteo- Most authors stress the importance of distinguishing
sarcoma, chondrosarcoma, and malignant fibrous histio- between exuberant fracture callus and osteosarcoma (see
cytoma. The latter tumor is emerging as an important Chapter 23).
sequel to therapeutic irradiation. It also appears that In addition to the clinical evaluation, laboratory and
radiation therapy can induce the transformation of a radiographic studies and adequate biopsy material are
low-grade, barely aggressive lesion into a high-grade needed to make a correct diagnosis. The characteristic

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1473

A B

C D
FIGURE 24-31  ■  Sarcoma of bone associated with metallic implant. A, Radiograph of resection specimen shows femoral head pros-
thesis and lytic area of proximal femur. B, Gross photograph of same specimen shows hemorrhagic destructive mass surrounding
femoral head of metallic implant; hemorrhagic mass extends into soft tissue (arrows). C and D, Malignant fibrous histiocytoma
associated with metallic implant (same case as shown in A and B). (C and D, ×100) (C and D, hematoxylin-eosin.)

ERRNVPHGLFRVRUJ
1474 24  Precancerous Conditions

unique genetic predisposition. The risk of developing


sarcoma in bone because of genetic predisposition has
not yet been estimated. In general, familial syndromes
that may predispose to cancer should be clinically sus-
pected when (1) clusters of cancer occur in one family,
regardless of whether the tumors are the same or differ-
ent types that originate in the same or different organs;
(2) cancer occurs in an unusual age, typically in younger
patients; and (3) multiple independent primary tumors
affect a single individual.

Rothmund-Thomson Syndrome
This extremely rare syndrome is an autosomal recessive
disorder that consists of skin lesions (atrophy, hyper-
pigmentation, subepidermal fibrosis, and telangiecta-
sias referred to as poikiloderma), cataracts, and skeletal
abnormalities such as dysostoses and dysplasias that
result in abnormally shaped bones (Figs. 24-35 and 24-
36).91,92,107,112,116,119,120 Abnormal developmental anoma-
lies of the skeleton are frequently associated with short
stature and facial deformities. The skeletal anomalies can
be settled and focal or may be widespread, resulting in
major disfiguring deformities. The clinical features of the
syndrome usually manifest in childhood. Abnormal skin
pigmentation is typically the presenting sign. Benign skin
FIGURE 24-32  ■  Sarcoma of bone associated with metallic
implant. Large destructive mass with patchy matrix mineralized adnexal lesions are the most frequent neoplasms seen in
in area of metallic implant (hip prosthesis). this condition. An increased incidence of osteosarcoma
is the most serious complication of Rothmund-Thomson
syndrome. The analysis of large clinical data indicates
that 30% of patients with clinical manifestations of the
malignant changes of osteosarcoma—pleomorphism, syndrome develop osteosarcoma.84,89,122 Osteosarcomas
hyperchromatism, and bizarre mitoses—are absent in the in this syndrome have a tendency to develop in unusual
hyperplastic callus. sites, and some are multifocal. These osteosarcomas
develop in patients who are younger than those who have
conventional osteosarcomas. Mutations of the RECQL4
gene encoding a RCO DNA helicase are present in a
SYNDROMES PREDISPOSING TO large proportion of patients affected by the Rothmund-
MALIGNANCY IN BONE Thomson syndrome (Fig. 24-37).85,93,105,109,121 In contrast,
mutations of RECQL4 are infrequent in sporadic osteo-
There is increased awareness that hereditary etiologies sarcoma.93,100 A more detailed description of the role of
with mendelian inheritance in addition to congenital syn- helicases in the biology of tumors and their associated
dromes without clear inheritance patterns are responsible clinical syndrome is provided in Chapter 3. The RECQ
for the development of bone and soft tissue tumors. family of DNA helicases plays a role in DNA repair,
During the past decade, there has been an exponential replication, and recombination pathways.98 In humans,
increase in the identification of germline mutations that mutations of these enzymes encoded by the BLM, WRN,
predispose individuals to the development of bone tumors and RECQL4 genes are associated with Bloom’s, Werner’s
in these syndromes. Therefore, a detailed family history and Rothmund-Thomson syndromes, respectively.98,99 In
is an important component of prevention and surveil- general, these disorders are characterized by premature
lance, facilitating early detections in affected families. aging and predisposition to cancer. Increased numbers
The list of hereditary disorders that are associated with of chromosomal breaks in somatic cells and sensitivity to
the development of various skeletal and soft tissue tumors ultraviolet light, consistent with a defect in DNA repair,
in which bone and cartilage forming tumors may develop are downstream effects of malfunctioning DNA helicases
is provided in Table 24-2. Their detailed description is in these syndromes.103,104,110,123
beyond the scope of this book. Two familial syndromes,
Rothmund-Thomson and Li-Fraumeni, that predispose
individuals to the development of sarcoma in bone are
Li-Fraumeni Syndrome
described in this section. As in the case of retinoblastoma- In 1988, Li and Fraumeni described 24 kindreds affected
associated osteosarcoma (see Chapters 3 and 5), both may by a wide range of cancer types. The most frequent
have a unique inherited molecular mechanism. They are cancer types were breast carcinoma and soft tissue sarco-
extremely rare, but it is estimated that up to 10% of mas. The members of these families were less frequently
common cancers in humans may develop because of a Text continued on p. 1480

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1475

D
FIGURE 24-33  ■  Postradiation sarcoma: radiographic features. A, Plain radiograph shows destruction of C2 bone in a 30-year-old
woman in whom this lesion developed 5 years after exposure to radiation from nuclear plant accident. Microscopically, tumor was
malignant fibrous histiocytoma. B, Computed tomogram shows destructive lesion involving C2 body and posterior elements, pro-
truding into spinal cord and extending into soft tissue. C and D, Coronal and sagittal magnetic resonance images show destructive
low-signal mass encircling spinal cord. Mass has destroyed C2 bone elements and has extended into soft tissue.

ERRNVPHGLFRVRUJ
1476 24  Precancerous Conditions

B
FIGURE 24-34  ■  Postradiation sarcoma: radiographic features. A and B, Anteroposterior and oblique views of postradiation sarcoma
of scapula 8 years after radiation therapy for breast carcinoma. Note lytic destructive mass of scapula (arrow) and patchy lytic areas
of radionecrosis in proximal humerus.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1477

TABLE 24-2 Hereditary Disorders Associated with Tumors of Soft Tissue and Bone, Ordered
by Genes Known to be Involved
Gene Locus Inherited Syndrome Inheritance Associated Tumors
ACP5 19p13 Spondyloenchondrodysplasia AR Enchondromas
ACVR1 2q24 Fibrodysplasia ossificans Sporadic/AD Progressive heterotopic ossification
progressiva (myositis ossificans progressiva),
proximal tibial osteochondromas
ANTXR2 4q21 Fibromatosis, juvenile hyaline AR Fibromatosis
APC 5q21 Desmoid disease, hereditary AD Desmoid tumors
Familial adenomatous polyposis 1 AD Craniofacial osteomas, desmoid tumors,
(Gardner’s syndrome included) Gardner fibromas
BRAF 7q34 Cardiofaciocutaneous syndrome AD Giant cell lesions of small bones (central)
BUB1B 15q15 Mosaic variegated aneuploidy AR Embryonal rhabdomyosarcomas
syndrome 1
TP53 17p13 Li-Fraumeni syndrome 1 AD Osteosarcomas, rhabdomyosarcomas,
and other soft tissue sarcomas
CHEK2 22q12 Li-Fraumeni syndrome 2 AD Osteosarcomas, rhabdomyosarcomas,
and other soft tissue sarcomas
EXT1 8q24 Multiple osteochondromas AD Osteochondromas, secondary peripheral
chondrosarcomas
Trichorhinophalangeal syndrome Sporadic Osteochondromas, secondary peripheral
type 2 chondrosarcomas
EXT2 11p11 Multiple osteochondromas AD Osteochondromas, secondary peripheral
chondrosarcomas
Potocki-Shaffer syndrome Sporadic Osteochondromas, secondary peripheral
chondrosarcomas
GLMN 1p22 Glomovenous malformations AD Glomus tumors
GNAS 20q13 Pseudohypoparathyroidism, type 1A AD Cutaneous osteomas
McCune-Albright syndrome, incl. Sporadic Polyostotic fibrous dysplasia,
Mazabraud syndrome osteosarcomas (Mazabraud syndrome:
intramuscular myxomas)
Osseous heteroplasia, progressive AD Cutaneous osteomas
Pseudopseudohypoparathyroidism AD Cutaneous osteomas
IDH1 2q34 Enchondromatosis Sporadic Enchondromas, chondrosarcomas
IDH2 15q26 (Ollier’s disease and Maffucci’s (Maffucci’s syndrome: hemangiomas,
PTH1R 3p21 syndrome) angiosarcomas)
LEMD3 12q14 Buschke-Ollendorff (osteopoikilosis AD Enostoses
isolated incl.)
MID1 Xp22 Opitz GBBB syndrome, X-linked XR Cranial osteomas
PORCN Xp11 Focal dermal hypoplasia XD Giant cell tumors of bone
PRKAR1A 17q24 Carney complex, type 1 AD Osteochondromyxomas, cardiac and
other myxomas, melanocytic
schwannomas
PTPN11 12q24 LEOPARD syndrome 1 AD Granular cell tumors
Metachondromatosis AD Enchondromas, osteochondroma-like
lesions
Noonan syndrome 1 AD Granular cell tumors, giant cell lesions of
small bones (central)
RB1 13q14 Retinoblastoma AD Osteosarcomas, soft tissue sarcomas
RECQL4 8q24 Baller-Gerold syndrome AR Osteosarcomas
RAPADILINO syndrome AR Osteosarcomas
Rothmund-Thomson syndrome AR Osteosarcomas
WRN 8p12 Werner syndrome AR Bone and soft tissue sarcomas
BLM 15q26 Bloom syndrome AR Osteosarcomas
SH3BP2 4p16 Cherubism AD Giant cell lesions
T 6q27 Chordoma, familial AD Chordomas
TNFRSF11A 18q22 Paget’s disease of bone AD Osteosarcomas
Polyostotic osteolytic dysplasia, AD Osteosarcomas
hereditary expansile

AD, Autosomal dominant; AR, Autosomal recessive; GIST, Gastrointestinal stromal tumor; XD, X-linked dominant; XR, X-linked recessive.
Modified from Bridge JA, Mertens F: Tumour syndromes: introduction. In Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F,
editors: WHO classification of tumours of soft tissue and bone, ed 4. Lyon, 2013, IARC.

ERRNVPHGLFRVRUJ
1478 24  Precancerous Conditions

A D

B C

FIGURE 24-35  ■  Rothmund-Thomson syndrome: clinical and radiographic features. A, Anteroposterior view of pelvis. Note abnormally
shaped iliac bones and multiple lytic defects with scalloped sclerotic margins. B and C, Clinical photographs showing deformities
of both lower extremities. Note abnormal skin pigmentation and bulging mass corresponding to left proximal fibular region.
D, Radiograph of right lower extremity with deformity and sclerosis of tibia.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1479

A B C
FIGURE 24-36  ■  Osteosarcoma in Rothmund-Thomson syndrome: radiographic and gross features. A and B, Anteroposterior and
lateral views of osteoblastic mass of left proximal fibula. Note deformity and sclerosis of tibia. C, Gross photograph of amputation
specimen shows calcified mass of proximal fibular end and severe bowing deformity of tibia (same case as Fig. 24-35).

ERRNVPHGLFRVRUJ
1480 24  Precancerous Conditions

Chr 8 RECQL4
23.3 23.2
23.1

Exon 10
Exon 11

Exon 12

Exon 13

Exon 14

Exon 15

Exon 16
Exon 17
Exon 18

Exon 19

Exon 20

Exon 21
Exon 1
Exon 2
Exon 3

Exon 4

Exon 5

Exon 6

Exon 7
Exon 8
Exon 9
22
21.3 21.2

cds
21.1
12
11.2 11.1
11.1
11.22 11.21
12 11.23 Helicase region Splicing mutation
13 Intronic deletion
21.1
21.2
21.3 RECQL4

5
22.1
22.2
22.3
23 1 1208 aa
24.1
24.2 24.3 Zinc finger CCHC-type domain Missense substitution
RECQL4 P-loop containing nucleoside triphosphate hydrolase domains Frameshift mutation
Helicase superfamily 1/2 ATP-binding domain Nonsense mutation
A Helicase C-terminal domain
B

N-End Rule Base Excision


Pathway Repair

UBR1/2 PARP1

RECQL4

Rad51 Cut5

RECQL4-interacting Partners

Repair of DNA
DSBs Replication
Proposed Cellular Pathway
C
FIGURE 24-37  ■  Molecular alterations in Rothmund-Thomson syndrome. A, Location of the RECQL4 gene on the long arm of chromo-
some 8 is shown. B, Mutations of the RECQL4 gene frequently present in patients with Rothmund-Thomson syndrome. The positions
of mutations involving the non-coding sequence are shown on the upper diagram depicting the exon-intron structure of the RECQL4
gene. The positions of mutations involving the coding sequences are shown on the lower diagram depicting the structure of the
RECQL4 protein. C, Interacting partners of the RCO DNA helicase and putative pathways controlled by this enzyme responsible for
clinical manifestations in the Rothmund-Thomson syndrome. Modified from Wang LL, Gannavarapu A, Kozinetz CA, et al: JNCI 95:669–
674, 2003 and Dietschy T, Shevelev I, Stagljar I: Cell Mol Life Sci 64:796–802, 2007.

affected by other cancers, including osteosarcoma of mal control of the cell cycle.81,85,87,92,101 Alterations of
bone.81,82,96,97 The syndrome is unique in the sense that it other genes such as MDM2 and CHEK2, as well as
predisposes the affected family to a wide range of malig- shorter telomere length, have been postulated in the
nant neoplasms in different organs. In contrast, other development of Li-Fraumeni syndrome, but linkage
well-known familial cancer syndromes, such as hereditary between the alterations in these genes and the develop-
colon and breast cancers as well as rare examples of ment of sarcoma in the absence of TP53 mutations is
familial osteosarcomas, predispose to a limited range of uncertain at the time of this writing.83,86,108,111,115
cancers (see Chapter 5). It has been shown that families
REFERENCES
with Li-Fraumeni syndrome carry germline mutations
of the TP53 gene.87,88,94,95,106,113,114,117,118,124 Therefore, Paget’s Disease of Bone
Li-Fraumeni syndrome is the prototype clinical condi- 1. Bertoldi I, Cantarini L, Filippou G, et al: Paget’s disease. Reuma-
tismo 66:171–183, 2014.
tion that links genetic predisposition to a wide range of 2. Collins DH: Paget’s disease of bone: incidence and subclinical
cancers with the malfunction of a single tumor-suppressor forms. Lancet 2:51, 1956.
gene.90,101,102 However, the syndrome may not have a 3. Dorfman HD, Czerniak B: Bone cancers. Cancer 75:203–210,
uniform TP53-related pathogenesis because mutations of 1994.
the TP53 gene were not identified in some affected fami- 4. Galson DL, Roodman GD: Pathobiology of Paget’s disease of
bone. J Bone Metab 21:85–98, 2014.
lies. In addition to TP53 mutations, fibroblastic cells of 5. Guay-Belanger S, Picard S, Gagnon E, et al: Detection of
patients with Li-Fraumeni syndrome show an increased SQSTM1/P392L post-zygotic mutations in Paget’s disease of
rate of replicative errors (genetic instability) and abnor- bone. Hum Genet 134:53–65, 2015.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1481

6. Hadjipavlou A, Lander P, Srolovitz H, et al: Malignant trans- 33. Dorfman HD, Norman A, Wolff H: Fibrosarcoma complicating
formation in Paget’s disease of bone. Cancer 70:2802–2808, bone infarction in a caisson worker: a case report. J Bone Joint
1992. Surg 48A:528–532, 1966.
7. Haibach H, Farrell C, Dittrich FJ: Neoplasms arising in Paget’s 34. Frierson HF, Fechner RE, Stallings RG, et al: Malignant fibrous
disease of bone: a study of 82 cases. J Clin Pathol 83:594–600, histiocytoma in bone infarct: association with sickle cell trait and
1985. alcohol abuse. Cancer 59:496–500, 1987.
8. Harvey L: Viral aetiology of Paget’s disease of bone: a review. J R 35. Furey JG, Ferrer-Torells M, Reagan JW: Fibrosarcoma arising at
Soc Med 77:943–948, 1984. the site of bone infarcts: a report of two cases. J Bone Joint Surg
9. Huvos AG, Butler A, Bretsky SS: Osteogenic sarcoma associated 42A:802–810, 1960.
with Paget’s disease of bone: a clinicopathologic study of 65 36. Galli SJ, Weintraub HP, Proppe KH: Malignant fibrous histiocy-
patients. Cancer 52:1489–1495, 1983. toma and pleomorphic sarcoma in association with medullary
10. Mills BG, Singer FR: Nuclear inclusions in Paget’s disease of bone infarcts. Cancer 41:607–619, 1978.
bone. Science 194:201–202, 1976. 37. Grootboom MJ, Charles RW: Sarcoma associated with bone
11. Mirra JM, Brien EW, Tehranzadeh J: Paget’s disease of bone: infarction: a report of 2 cases. S Afr Med J 75:293–294, 1989.
review with emphasis on radiologic features. Part II. Skeletal Radiol 38. Houdek MT, Wyles CC, Sierra RJ: Osteonecrosis of the femoral
24:173–184, 1995. head: treatment with ancillary growth factors. Curr Rev Musculo-
12. Paget J: On a form of chronic inflammation of bones (osteitis skelet Med 2015. [Epub ahead of print].
deformans). Med Chir Trans 60:37, 1877. 39. Heater K, Collins PA: Osteosarcoma in association with infarction
13. Paget J: Additional cases of osteitis deformans. Med Chir Trans of bone. J Bone Joint Surg 69A:300–302, 1987.
65:225, 1882. 40. Heselson NG, Price SK, Mills EE, et al: Two malignant fibrous
14. Price CH, Goldie W: Paget’s sarcoma of bone: a study of eighty histiocytomas in bone infarcts. J Bone Joint Surg 65A:1166–1171,
cases from the Briston and the Leeds bone tumour registries. 1983.
J Bone Joint Surg Br 51:205–224, 1969. 41. Heselson NG, Webber BL, Goldberg S, et al: Bone infarction
15. Ralston SH, Albagha OM: Genetics of Paget’s disease of bone. complicated by sarcomatous change: a case report. S Afr Med J
Curr Osteoporos Rep 12:263–271, 2014. 50:1942–1944, 1976.
16. Rea SL, Majcher V, Searle MS, et al: SQSTM1 mutations— 42. Kitano M, Iwasaki H, Yoh SS, et al: Malignant fibrous histiocy-
bridging Paget disease of bone and ALS/FTLD. Exp Cell Res toma at the site of bone infarction in association with DCS.
325:27–37, 2014. Undersea Biomed Res 11:305–314, 1984.
17. Rebel A, Basle M, Pouplard A, et al: Viral antigens in osteoclasts 43. Michael RH, Dorfman HD: Malignant fibrous histiocytoma asso-
from Paget’s disease of bone contained viral antigenic material: ciated with bone infarcts: report of a case. Clin Orthop 118:180–
ultrastructural and immunological studies suggested that measles 183, 1976.
or measles-related virus was the agent involved. Lancet 16:344– 44. Mirra JM, Bullough PG, Marcove RC, et al: Malignant fibrous
346, 1980. histiocytoma and osteosarcoma in association with bone infarcts:
18. Singer FR: Paget’s disease of bone: possible viral basis. Trends report of four cases, two in caisson workers. J Bone Joint Surg
Endocrinol Metab 7:258–261, 1996. 56A:932–940, 1974.
19. Smith J, Botet JF, Yeh SD: Bone sarcomas in Paget disease: a study 45. Mirra JM, Gold RH, Marafiote R: Malignant (fibrous) histiocy-
of 85 patients. Radiology 152:583–590, 1984. toma arising in association with a bone infarct in sickle-cell
20. Upchurch KS, Simon LS, Schiller AL, et al: Giant cell reparative disease: coincidence or cause-and-effect? Cancer 39:186–194,
granuloma of Paget’s disease of bone: a unique clinical entity. Ann 1977.
Intern Med 98:35–40, 1983. 46. Péré P, Adolphe J, Delgoffe C, et al: Ostéonécroses épiphysaires,
21. Wick MR, Siegal GP, Unni KK, et al: Sarcomas of bone compli- infarctus osseux multiples et histiocytome fibreux malin. Rev
cating osteitis deformans (Paget’s disease). Am J Surg Pathol Rhum Mal Osteoartic 51:427–430, 1984.
5:47–59, 1981. 47. Sweet DE, Madewell JE: Pathogenesis of osteonecrosis. In
Resnick D, Niwayama G, editors: Diagnosis of bone and joint disor-
Osteomyelitis ders, ed 2, Philadelphia, 1988, WB Saunders.
22. Akbarnia BA, Wirth CR, Colman N: Fibrosarcoma arising from 48. Torres FX, Kyriakos M: Bone infarct-associated osteosarcoma.
chronic osteomyelitis. J Bone Joint Surg 58A:123–125, 1976. Cancer 70:2418–2430, 1992.
23. Giunti A, Laus M: Malignant tumors in chronic osteomyelitis. Ital
J Orthop Traumatol 4:171–182, 1978. Metallic Implants
24. Johnson LL, Kempson RL: Epidermoid carcinoma in chronic 49. Bago-Granell J, Aguirre-Canyadell M, Nardi J, et al: Malignant
osteomyelitis: diagnostic problems and management. J Bone Joint fibrous histiocytoma of bone at the site of a total hip arthroplasty:
Surg 47A:133–144, 1965. a case report. J Bone Joint Surg Br 66:38–40, 1984.
25. Johnston RM, Miles JS: Sarcomas arising from chronic osteomy- 50. Black J: Does corrosion matter? J Bone Joint Surg Br 70:517–520,
elitis sinuses: a report of two cases. J Bone Joint Surg 55A:162–168, 1988.
1973. 51. Brien WW, Salvati EA, Healey JH, et al: Osteogenic sarcoma
26. Morris JM, Lucas DB: Fibrosarcoma within a sinus tract of arising in the area of a total hip replacement: a case report. J Bone
chronic draining osteomyelitis: case report and review of litera- Joint Surg 72A:1097–1099, 1990.
ture. J Bone Joint Surg 46A:853–857, 1964. 52. Gillespie WJ, Frampton CMA, Henderson RJ, et al: The inci-
27. Wagner RF, Grande DJ: Pseudoepitheliomatous hyperplasia vs. dence of cancer following total hip replacement. J Bone Joint
squamous cell carcinoma arising from chronic osteomyelitis of the Surg Br 70:539–542, 1988.
humerus. J Dermatol Surg Oncol 12:632–635, 1986. 53. Haas M, Adler CP: Malignant fibrous histiocytoma in association
with hip replacement. J Bone Joint Surg Br 71:701, 1989.
Bone Infarcts 54. Hughes AW, Sherlock DA, Lamblen DL, et al: Sarcoma at the
28. Abdelwahab IF, Hermann G, Lewis MM, et al: Transformation site of a single hip screw: a case report. J Bone Joint Surg Br
of an idiopathic bone infarct into malignant fibrous histiocy- 69:470–472, 1987.
toma in a female: a case report. Bull Hosp Joint Dis 48:197–203, 55. Lamovec J, Zidar A, Cucek-Planicar M: Synovial sarcoma associ-
1988. ated with total hip replacement. J Bone Joint Surg 70A:1558–1560,
29. Abdelwahab IF, Kenan S, Klein MJ, et al: Case report: angiosar- 1988.
coma occurring in a bone infarct. Clin Radiol 45:412–414, 1992. 56. Martin A, Bauer TW, Manley MT, et al: Osteosarcoma at the site
30. Arnold WH: Hereditary bone dysplasia with sarcomatous degen- of total hip replacement: a case report. J Bone Joint Surg 70A:1561–
eration: study of a family. Ann Intern Med 78:902–906, 1973. 1567, 1988.
31. Breen TF, Healy WL: Malignant fibrous histiocytoma arising in 57. Memoli VA, Urban RM, Alroy J, et al: Malignant neoplasms asso-
medullary long bone infarcts: a case report. Orthopedics 10:1169– ciated with orthopedic implant materials in rats. J Orthop Res
1173, 1987. 4:346–355, 1986.
32. Dorfman HD, Habermann ET: The risk of bone sarcoma in 58. Penman HG, Ring PA: Osteosarcoma in association with total hip
patients with bone infarction. Proc Am Acad Orthop Surg 1988. replacement. J Bone Joint Surg Br 66:632–634, 1984.

ERRNVPHGLFRVRUJ
1482 24  Precancerous Conditions

59. Sinibaldi KR, Pugh J, Rosen H, et al: Osteomyelitis and neoplasia 85. Dietschy T, Shevelev I, Stagljar I: The molecular role of the
associated with use of the Jonas intramedullary splint in small Rothmund-Thomson-, RAPADILINO- and Baller-Gerold-gene
animals. J Am Vet Med Assoc 181:885–890, 1982. product, RECQL4: recent progress. Cell Mol Life Sci 64:796–802,
60. Swann M: Malignant soft-tissue tumour at the site of a total hip 2007.
replacement. J Bone Joint Surg Br 66:629–631, 1984. 86. Evans DG, Birch JM, Narod SA: Is CHEK2 a cause of the
Li-Fraumeni syndrome? J Med Genet 45:63–64, 2008.
Radiation Injury 87. Frebourg T, Barbier N, Yan YX, et al: Germ-line p53 mutations
61. Evans R: The effect of skeletal deposited alpha-ray emitters in in 15 families with Li-Fraumeni syndrome. Am J Hum Genet
man. Br J Radiol 39:881–895, 1966. 56:608–615, 1995.
62. Halpern J, Kopolovic J, Catane R: Malignant fibrous histiocytoma 88. Iwakuma T, Lozano G, Flores ER: Li-Fraumeni syndrome: a p53
developing in irradiated sacral chordoma. Cancer 53:2661–2662, family affair. Cell Cycle 4:865–867, 2005.
1984. 89. Judge MR, Kilby A, Harper JI: Rothmund-Thomson syndrome
63. Hutchison GB: Late neoplastic changes following medical irradia- and osteosarcoma. Br J Dermatol 129:723–725, 1993.
tion. Radiology 105:645–652, 1972. 90. Lang GA, Iwakuma T, Suh YA, et al: Gain of function of a p53
64. Huvos AG, Woodard II, Heilweil M: Postradiation malignant hot spot mutation in a mouse model of Li-Fraumeni syndrome.
fibrous histiocytoma of bone: a clinicopathologic study of twenty Cell 119:861–872, 2004.
patients. Am J Surg Pathol 10:9–18, 1986. 91. Larizza L, Roversi G, Volpi L: Rothmund-Thomson syndrome.
65. Rock MG, Sim FH, Unni KK, et al: Secondary malignant giant- Orphanet J Rare Dis 5:2, 2010.
cell tumor of bone: clinicopathological assessment of nineteen 92. Larralde M, Morales MS, Gonzalez JA, et al: What syndrome is
patients. J Bone Joint Surg 68A:1073–1079, 1986. this? Rothmund-Thomson syndrome. Pediatr Dermatol 12:277–
66. Sim FH, Cupps RE, Dahlin DC, et al: Post-radiation sarcoma of 280, 1995.
bone. J Bone Joint Surg 54A:1479–1489, 1972. 93. Lu L, Jin W, Liu H, et al: RECQ DNA helicases and osteosar-
67. Tountas AA, Fornasier VL, Harwood AR, et al: Post-radiation coma. Adv Exp Med Biol 804:129–145, 2014.
sarcoma of bone. Cancer 43:182–187, 1979. 94. MacGeoch C, Turner G, Bobrow LG, et al: Heterogeneity in
68. Weatherby RP, Dahlin DC, Ivins JC: Post-radiation sarcoma of Li-Fraumeni families: p53 mutation analysis and immunohisto-
bone. Mayo Clin Proc 56:294–306, 1981. chemical staining. J Med Genet 32:186–190, 1995.
95. Malkin D: p53 and the Li-Fraumeni syndrome. Cancer Genet Cyto-
Osteogenesis Imperfecta genet 66:83–92, 1993.
69. Banta JV, Schreiber RR, Kulik WJ: Hyperplastic callus formation 96. Malkin D: Li-Fraumeni syndrome. Genes Cancer 2:475–484,
in osteogenesis imperfecta simulating osteosarcoma. J Bone Joint 2011.
Surg 53A:115–122, 1971. 97. McBride KA, Ballinger ML, Killick E, et al: Li-Fraumeni syn-
70. Bedi HS, Kaufman DV, Choong PF, et al: Osteosarcoma of the drome: cancer risk assessment and clinical management. Nat Rev
scapula arising in osteogenesis imperfecta. Pathology 31:52–54, Clin Oncol 11:260–271, 2014.
1999. 98. Mohaghegh P, Hickson ID: DNA helicase deficiencies associated
71. Dobrocky I, Seidl G, Grill F: MRI and CT features of hyperplastic with cancer predisposition and premature ageing disorders. Hum
callus in osteogenesis imperfecta tarda. Eur Radiol 9:665–668, Mol Genet 10:741–746, 2001.
1999. 99. Mohaghegh P, Hickson ID: Premature aging in RecQ helicase-
72. Kahn LB, Wood F, Ackerman LV: Fracture callus associated with deficient human syndromes. Int J Biochem Cell Biol 34:1496–1501,
benign and malignant bone lesions and mimicking osteosarcoma. 2002.
Am J Clin Pathol 52:14–24, 1969. 100. Nishijo K, Nakayama T, Aoyama T, et al: Mutation analysis of the
73. Klenerman L, Ockenden BG, Townsend AC: Osteosarcoma RECQL4 gene in sporadic osteosarcomas. Int J Cancer 111:367–
occurring in osteogenesis imperfecta: report of two cases. J Bone 372, 2004.
Joint Surg 49B:314–323, 1967. 101. Olive KP, Tuveson DA, Ruhe ZC, et al: Mutant p53 gain of func-
74. Lasson U, Harms D, Wiedemann H-R: Osteogenic sarcoma com- tion in two mouse models of Li-Fraumeni syndrome. Cell 119:
plicating osteogenesis imperfecta tarda. Eur J Pediatr 129:215– 847–860, 2004.
218, 1978. 102. Olivier M, Goldgar DE, Sodha N, et al: Li-Fraumeni and related
75. Maiya S, Grimer RJ, Ramaswamy R, et al: Osteosarcoma occur- syndromes: correlation between tumor type, family structure, and
ring in osteogenesis imperfecta tarda. Int Orthop 26:126–128, TP53 genotype. Cancer Res 63:6643–6650, 2003.
2002. 103. Orstavik KH, McFadden N, Hagelsteen J, et al: Instability of
76. Rieker O, Kreitner KF, Karbowski A: Hyperplastic callus forma- lymphocyte chromosomes in a girl with Rothmund-Thomson
tion in osteogenesis imperfecta: CT and MRI findings. Eur Radiol syndrome. J Med Genet 31:570–572, 1994.
8:1137–1139, 1998. 104. Paules RS, Levedakou EN, Wilson SJ, et al: Defective G2 check-
77. Rutkowski R, Resnick P, McMaster JH: Osteosarcoma occurring point function in cells from individuals with familial cancer syn-
in osteogenesis imperfecta. J Bone Joint Surg 61A:606–608, 1979. dromes. Cancer Res 55:1763–1773, 1995.
78. Van Dijk FS, Sillence DO: Osteogenesis imperfecta: clinical diag- 105. Petkovic M, Dietschy T, Freire R, et al: The human Rothmund-
nosis, nomenclature and severity assessment. Am J Med Genet Thomson syndrome gene product, RECQL4, localizes to distinct
164:1470–1481, 2014. nuclear foci that coincide with proteins involved in the mainte-
79. Vieira RL, Amaral DT, Jesus-Garcia FR, et al: Hyperplastic callus nance of genome stability. J Cell Sci 118:4261–4269, 2005.
formation in osteogenesis imperfecta type V mimicking osteosar- 106. Ponz de Leon M: Li-Fraumeni syndrome. Recent Results Cancer
coma: 4-year follow-up with resolution. Skeletal Radiol 35:402– Res 136:275, 1994.
405, 2006. 107. Rizzari C, Conter V: Rothmund-Thomson syndrome, malignant
80. Yekeler E, Kumbasar B, Dursun M, et al: Pseudomass of the diseases, and treatment opportunities. Pediatr Hematol Oncol 13:
sternal manubrium in osteogenesis imperfecta. Skeletal Radiol 195–196, 1996.
32:371–373, 2003. 108. Ruijs MW, Schmidt MK, Nevanlinna H, et al: The single-
nucleotide polymorphism 309 in the MDM2 gene contributes to
Syndromes Predisposing to Malignancy in Bone the Li-Fraumeni syndrome and related phenotypes. Eur J Hum
81. Birch JM: Li-Fraumeni syndrome. Eur J Cancer 30A:1935–1941, Genet 15:110–114, 2007.
1994. 109. Sangrithi MN, Bernal JA, Madine M, et al: Initiation of DNA
82. Birch JM, Hartley AL, Tricker KJ, et al: Prevalence and diversity replication requires the RECQL4 protein mutated in Rothmund-
of constitutional mutations in the p53 gene among 21 Li-Fraumeni Thomson syndrome. Cell 121:887–898, 2005.
families. Cancer Res 54:1298–1304, 1994. 110. Shinya A, Nishigori C, Moriwaki S, et al: A case of Rothmund-
83. Birch JM, Heighway J, Teare MD, et al: Linkage studies in a Thomson syndrome with reduced DNA repair capacity. Arch
Li-Fraumeni family with increased expression of p53 protein but Dermatol 129:332–336, 1993.
no germline mutation in p53. Br J Cancer 70:1176–1181, 1994. 111. Siddiqui R, Onel K, Facio F, et al: The TP53 mutational spectrum
84. Cumin I, Cohen JY, David A, et al: Rothmund-Thomson syn- and frequency of CHEK2*1100delC in Li-Fraumeni-like kin-
drome and osteosarcoma. Med Pediatr Oncol 26:414–416, 1996. dreds. Fam Cancer 4:177–181, 2005.

ERRNVPHGLFRVRUJ
24  Precancerous Conditions 1483

112. Sim FH, DeVries EM, Miser JS, et al: Case report 760: osteoblas- 119. Vennos EM, Collins M, James WD: Rothmund-Thomson syn-
tic osteosarcoma (grade 4) with Rothmund-Thomson syndrome. drome: review of the world literature. J Am Acad Dermatol 27:750–
Skeletal Radiol 21:543–545, 1992. 762, 1992.
113. Strauss EA, Hosler MR, Herzog P, et al: Complex replication 120. Vennos EM, James WD: Rothmund-Thomson syndrome. Der-
error causes p53 mutation in a Li-Fraumeni family. Cancer Res matol Clin 13:143–150, 1995.
5:3237–3241, 1995. 121. Wang LL, Gannavarapu A, Kozinetz CA, et al: Association
114. Strong LC, Williams WR, Tainsky MA: The Li-Fraumeni syn- between osteosarcoma and deleterious mutations in the RECQL4
drome: from clinical epidemiology to molecular genetics. Am J gene in Rothmund-Thomson syndrome. J Natl Cancer Inst 95:
Epidemiol 135:190–199, 1992. 669–674, 2003.
115. Tabori U, Nanda S, Druker H, et al: Younger age of cancer initia- 122. Wang LL, Levy ML, Lewis RA, et al: Clinical manifestations in
tion is associated with shorter telomere length in Li-Fraumeni a cohort of 41 Rothmund-Thomson syndrome patients. Am J Med
syndrome. Cancer Res 67:1415–1418, 2007. Genet 102:11–17, 2001.
116. Tong M: Rothmund-Thomson syndrome in fraternal twins. 123. Werner SR, Prahalad AK, Yang J, et al: RECQL4-deficient cells
Pediatr Dermatol 12:134–137, 1995. are hypersensitive to oxidative stress/damage: insights for osteo-
117. Varley JM, McGown G, Thorncroft M, et al: A previously unde- sarcoma prevalence and heterogeneity in Rothmund-Thomson
scribed mutation within the tetramerisation domain of TP53 in a syndrome. Biochem Biophys Res Commun 345:403–409, 2006.
family with Li-Fraumeni syndrome. Oncogene 12:2437–2442, 1996. 124. Wu CC, Shete S, Amos CI, et al: Joint effects of germ-line p53
118. Varley JM, Thorncroft M, McGown G, et al: A novel deletion mutation and sex on cancer risk in Li-Fraumeni syndrome. Cancer
within exon 6 of TP53 in a family with Li-Fraumeni–like syn- Res 66:8287–8292, 2006.
drome and LOH in a benign lesion from a mutation carrier.
Cancer Genet Cytogenet 90:14–16, 1996.

ERRNVPHGLFRVRUJ

You might also like