Sei sulla pagina 1di 295

MEDICAL RADIOLOGY

Diagnostic Imaging
Editors:
A. L. Baert, Leuven
K. Sartor, Heidelberg
Springer
Berlin
Heidelberg
New York
Hong Kong
London
Milan
Paris
Tokyo
c. Procacci . A. J. Megibow (Eds.)

Imaging
of the Pancreas
Cystic and Rare Tumors
With Contributions by
C. Bassi· R. Bettini· C. Biasiutti . G. Bogina . A. Brighenti . P. Capelli· G. Carbognin
L. Casetti . E Cirillo· D. Coser . E. Della Chiara· M. Falconi· M. Ferdeghini . M. Ferrari
E Fornasa . I. R. Francis· I. Frigerio . A. Fuini . A. Guarise . W. Mantovani . G. Martignoni
A. J. Megibow· E. Molinari· T. Oweity· N. Pagnotta· P. Pederzoli· A. Pesci . E. Petrella
C. Procacci· R. Salvia· A. Scarpa· G. Schenal· M. Tapparelli· R.E Thoeni· S. Vasori
S. Venturini· A. B. West· G. Zamboni

Foreword by
A.L. Baert

With 238 Figures in 598 Separate Illustrations, 130 in Color and 17 Tables

' " Springer


CARLO PROCACCI, MD
Professor, Istituto di Radiologia
Dipartimento di Scienze Morfologico-Biomediche
Universita degli Studi di Verona
Policlinico "Giambattista Rossi"
P.zza L.A. Scuro 10
37134 Verona
Italy

ALEC J. MEG !BOW, MD, MPH, FACR


Professor of Radiology
NYU School of Medicine
Department of Radiology
560 First Avenue
New York, NY 10016
USA

MEDICAL RADIOLOGY' Diagnostic Imaging and Radiation Oncology


Series Editors: A. L. Baert . L. W. Brady· H.-P. Heilmann· M. Molls· K. Sartor
Continuation of
Handbuch der medizinischen Radiologie
Encyclopedia of Medical Radiology

ISBN-13978-3-642-63942-5 Springer-Verlag Berlin Heidelberg New York

Library of Congress Cataloging-in-Publication Data

Imaging of the pancreas: cystic and rare tumors / C. Procacci, A. J. Megibow (eds.) ;
with contributions by C. Bassi ... [et al.] ;foreword by A. L. Baert.
p. ; cm. -- (Medical radiology)
Includes bibliographical references and index.
ISBN- 13 978-3-642-63942-5 (alk.paper)
1. Pancreas--Tumors--Imaging. 2. Pancreas--Cysts--Imaging. I. Procacci, C. (Carlo),
1950- II. Megibow, Alec J. III. Bassi, C (Claudio) IV. Series.
[DNLM: 1. Pancreatic Neoplasms--diagnosis. 2. Carcinoma. 3. Diagnostic
Imaging--methods. 4. Pancreatic Neoplasms--pathology. WI 810 131 2003]
RC280.P25 1434 2003
616.99'2370757--dc21 2002026896

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, specifi-
cally the rights of translation, reprinting, reuse of illustrations, recitations, broadcasting, reproduction on microfilm or
in any other way, and storage in data banks. Duplication of this publication or parts thereof is permitted only under the
provisions of the German Copyright Law of September 9, 1965, in its current version, and permission for use must always
be obtained from Springer-Verlag. Violations are liable for prosecution under the German Copyright Law.
Springer- Verlag Berlin Heidelberg New York
a member of BertelsmannSpringer Science+Business Media GmbH

httpllwww. springer.de
[SBN- \3 978-3-642-63942-5 e-ISBN-13 978-3-642-59344-4
DOl : 10.1007/978-3-642-59344-4

© Springer-Verlag Berlin Heidelberg 2003


Softcover reprint of the hardcover I st edition 2003

The use of general descriptive names, trademarks, etc. in this publication does not imply, even in the absence of a specific
statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general
use.
Product liability: The publishers cannot guarantee the accuracy of any information about dosage and application contained
in this book. In every case the user must check such information by consulting the relevant literature.
Cover- Design and Typesetting: Verlagsservice Teichmann, 69256 Mauer
2113150 - 543 2 1 0 - Printed on acid-free paper
Foreword

The pathology of the pancreas, which was a "hidden" retroperitoneal organ for many years,
can now be marvelously depicted by the modern sectional imaging techniques. As a result
great progress has been achieved during the past decade in the diagnosis and differential
diagnosis of pancreatic tumors.
Among the tumors of the pancreas the so-called rare tumors need to be well known by
radiologists because they pose challenging problems for differential diagnosis and for clini-
cal management. This is particularly true because these tumors are frequently discovered
incidentally.
This volume covers comprehensively all aspects of cystic tumors , intraductal cystic
tumors, endocrine tumors, rare tumors and secondary tumors of the pancreas and features
numerous superb illustrations. It really provides a wealth of information on a relatively less
well known area of the digestive system.
The book has been prepared and edited by two outstanding abdominal radiologists, Alec
Megibow and Carlo Procacci, both widely known internationally for their many brilliant
contributions to the scientific literature on pancreatic imaging. The other contributors come
also from both sides of the Atlantic and were invited to provide chapters because of their
exceptional expertise and experience.
I would like to congratulate the editors and the authors most sincerely for their efforts,
which have resulted in a volume exceptional both in its contents and in its presentation. I am
confident that this outstanding book will meet with great interest not only from general and
abdominal radiologists but also from gastroenterologists and abdominal surgeons. I believe
that it will encounter the same success with readers as previous volumes published in this
series.

Leuven ALBERT 1. BAERT


Preface

The availability and the widespread diffusion of ever more sophisticated radiological instru-
ments has significantly improved radiologists' ability to image pancreatic diseases. Imaging
studies now playa decisive role not only in the diagnosis, but also in the therapy of pancreatic
disorders. However, our improved ability to image diseases has brought new challenges in
understanding the nature and significance of our findings. The technological improvements
require radiologists to update and revise their current imaging techniques and integrate new
information from novel imaging procedures. To attempt to aid this process, we undertook a
project dedicated to pancreatic diseases. At the conclusion of this writing, we expect a four-
volume set encompassing imaging findings in pancreatic diseases.
For the first volume, we purposefully chose cystic and rare tumors of the pancreas. These
tumors are becoming more frequently observed and less well understood. Not surprisingly,
these lesions have been the focus of a burst ofliterature from many sources. Researchers and
physicians are beginning to accept a more uniform classification scheme by which the lesions
can be more accurately compared in terms of their biologic and clinical behavior. This infor-
mation will lead to a more uniform approach to therapy.
Subsequent volumes will focus on pancreatitis, acute and chronic (volume 2); ductal
carcinoma (volume 3); and imaging techniques (volume 4). We chose to place the volume
on techniques at the end of the series so that, at completion, the series will reflect the most
current imaging protocols and procedures.
With this book, we hope to attract the attention not only of radiologists, but also of pathol-
ogists, gastroenterologists and surgeons. In order to meet this goal, a complete description
of the clinical aspects, the therapeutic options and the pathological characteristics of these
tumors precedes all of the imaging chapters. This model integration between the clinical,
pathologic and imaging analyses reflects the collaborations which characterize the New York
University and Verona teams, both of which have been researching pancreatic imaging and
pathology for many years. Furthermore, this book boldly attempts to demonstrate how dif-
ferent authors, working on different continents, achieve consensus and thereby add to the
knowledge of those interested in this pathology.
Presently the second volume, dedicated to inflammatory diseases of the pancreas, is in
preparation.
We would like to thank the Editor-in -Chief, Prof. Albert Baert, for his valuable suggestions,
and Ursula N. Davis of Springer-Verlag for her constant and patient collaboration. We would
also like to sincerely thank all the authors for their extremely hard work in preparing their
contributions.
Last, but certainly not least, special thanks to our families and collaborators for all their
encouragement and support.

Verona CARLO PROCACCI


New York ALEC J. MEGIBOW
Contents

Cystic Tumors ............................................................. .

Clinical Manifestations and Therapeutic Management


CLAUDIO BASSI, ENRICO MOLINARI, MASSIMO FALCONI, and PAOLO PEDERZOLI 3

2 Pathology of Cystic Tumors


GIUSEPPE ZAMBONI, PAOLA CAPELLI, ANNA PESCI, and ANTONIETTA BRIGHENTI .. 9

3 Serous Cystic Tumors


GIOVANNI CARBOGNIN, MARGHERITA TAPPARELLI, ENRICO PETRELLA,
ARNALDO FUINI, and CARLO PROCACCI ..................................... 31

4 Mucinous Cystic Tumors


CARLO BIASIUTTI, FRANCESCA FORNASA, SILVIA VENTURINI,
NICOLETTA PAGNOTTA, GIACOMO SCHENAL, and CARLO PROCACCI 57

Intraductal Cystic Tumors .................................................... 75

5 Clinical Manifestations and Therapeutic Management


ROBERTO SALVIA, MASSIMO FALCONI, LUCA CASETTI, WILLIAM MANTOVANI,
ISABELLA FRIGERIO, and PAOLO PEDERZOLI ................................. 77

6 Pathology of Intraductal Cystic Tumors


GIUSEPPE ZAMBONI, PAOLA CAPELLI, GIACOMO BOGINA, ANNA PESCI,
and ANTONIETTA BRIGHENTI .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

7 Intraductal Papillary Mucinous Tumors: Imaging


CARLO PROCACCI, GIACOMO SCHENAL, EMILIANO DELLA CHIARA,
ARNALDO FUINI, and ALESSANDRO GUARISE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97

Endocrine Thmors .......................................................... 139

8 Clinical Manifestations and Therapeutic Management


of Hyperfunctioning Endocrine Tumors
FERNANDO CIRILLO, MASSIMO FALCONI, and ROSSELLA BETTINI ............... 141

9 Clinical Manifestations and Therapeutic Management


of Hyperfunctioning Endocrine Tumors
MASSIMO FALCONI, ROSELLA BETTINI, CLAUDIO BASSI, ROBERTO SALVIA,
and PAOLO PEDERZOLI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. 153
10 Pathology
PAOLA CAPELLI, GIUSEPPE ZAMBONI,ANNA PESCI, GUIDO MARTIGNONI,
and ALDO SCARPA. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. 161

11 Endocrine Hyperfunctioning Tumors


RUEDI F. THOENI ......................................................... 177

12 Nonhyperfunctioning Endocrine Tumors


CARLO PROCACCI, MARCO FERDEGHINI, ALESSANDRO GUARISE, SIMONE VASORI,
DANIELA COSER, and MAURO FERRARI ...................................... 197

Rare Tumors ................................................................ 217

13 Rare Solid Tumors: Clinical Manifestations and Pathology


THAIRA OWElTY and A. BRIAN WEST ....................................... 219

14 Unusual Pancreatic Neoplasms: Imaging


ALEC J. MEGIBOW and ISAAC R. FRANCIS .................................... 249

Secondary Tumors .......................................................... 267

15 Secondary Tumors: Clinical Manifestations and Pathology


THAIRA OWElTY and A. BRIAN WEST ....................................... 269

16 Secondary Pancreatic Tumors: Imaging


ALEC J. MEGIBOW ........................................................ 277

Subject Index ............................................................... 289

List of Contributors .......................................................... 297


Cystic Tumors
1 Clinical Manifestations and Therapeutic Management
C. BASSI, E. MOLINARI, M. FALCONI, and P. PEDERZOLI

CONTENTS prevalence in imaging, led by ultrasound, may be the


main reason for the increased detection of pancreatic
1.1 Introduction 3 cystic tumors (BASSI et al. 2001).
1.2 Clinical Data 3 In this chapter, we shall analyze the clinical and
1.2.1 Clinical History and Lifestyle 3
1.2.2 Symptoms 4
laboratory data presented by patients suffering
1.3 Laboratory 5 from cystic tumors of the pancreas and then go on
1.3.1 Hemo-Chemistry Routine 5 to discuss their prognostic significance and further
1.3.2 Markers 5 management.
1.4 Index of Suspicion from Clinical Data 5
1.5 From Assessment to Management 5
1.5.1 Diagnostic Suspicion 5
1.5.2 Definite Diagnosis 6
1.5.2.1 Operable Patients 6 1.2
1.5.2.2 Inoperable Patients 7 Clinical Data
1.5.2.3 Follow-Up 7
References 7
1.2.1
Clinical History and Lifestyle

The most impressive epidemiological data regarding


1.1 peripheral cystic tumors of the pancreas is their nota-
Introduction bly more frequent occurrence in women (ratio of 4:
1). In fact, in our series of serous cystic tumors (SCT),
Cystic tumors of the pancreas were first observed in 87% are women (BASSI et al. 2002), while almost all
1830 (BECOURT and BECOURT 1830). Even so, there peripheral mucinous cystic tumors (MCT), benign
still remain many unanswered questions as to their or malignant, are found in women (ZAMBONI et al.
identification, classification, staging, history, and 1999; PEDERZOLI et al. 2000). Frequently, peripheral
treatment (TALAMINI et al. 1992). Modern under- mucinous tumors display ovarian-like stroma in the
standing of these tumors stems from the work of rim of the lesion at histology. The presence of ovar-
CAMPAGNO and OERTEL (1978). These pathologists ian-like stroma is thought to represent a common
first grouped these lesions into mucinous-macro- pathway between the pancreatic peripheral muci-
cystic and serous-micro cystic type. More recent nous tumor and its counterpart in the hepatobiliary
classifications are based on the appearance of the system, ovary, and retroperitoneum (ZAMBONI et al.
epithelium (KUJPPEL et al. 1996) or as microcystic 1999). However, there are no epidemiological data
and oligocystic variants (LEWANDROWSKI et al. 1992; available that might suggest any correlation in this
EGAWA et al. 1994). sense between the tumor's appearance, menarche,
Nowadays, in virtually all but the most remote irregular menstruation, pregnancy, estro-progesto-
countries worldwide, every patient presenting with gen treatment, etc. The female predilection is not
abdominal symptoms will undergo ultrasound true for intraductal papillary mucinous tumor.
imaging due to its wide availability and low cost. The The patient age range is very large, with an aver-
age age which seems to vary depending on the type
and malignancy of the neoplasm. The average age for
C. BASSI, MD; E. MOLINARI, MD; M. FALCONI, MD;
P. PEDERZOLI, MD serous forms is around 50 years, while the mucinous
Surgical-Gastroenterological Department, Hospital 'G .B. Rossi', variant mainly affects patients in their 60s. Patients
University of Verona, 37134 Verona, Italy presenting with malignant mucinous tumors tend to
4 C. Bassi et al.

be older, suggesting the possible progression of the tumors are more frequently symptomatic (dyspep-
tumor over time from initial benignity to variable sia, pain, weight loss, and jaundice) compared with
degrees of malignancy (BASSI et al. 2001; TALAMINI their benign counterparts. Tables 1.1 and 1.2 show
et al. 1992; ZAMBONI et al. 1999; PEDERZOLI et al. the symptoms reported in a series of 85 patients
2000; LE BORGNE 1998; YANG et al. 1994; WARSHAW undergoing surgical resection of serous (n=44) or
et al. 1990). mucinous (n=41) cystic pancreatic tumors (BASSI et
Serous cysts are reported to be associated al. 2001,2002).
with other neoplasms and chronic diseases; how- Considering all the tumors surgically or medically
ever, the only established association is with von treated, 46% of patients were asymptomatic: 56%
Hippel-Lindau disease (PYKE et al. 1992). A review serous tumor and 28% mucinous tumor. In the latter,
of 55 patients with von Hippel-Lindau disease symptoms referable to the malignant behavior of the
shows the presence of serous pancreatic cysts in tumor (anorexia and weight loss) were more fre-
15% (NEUMANN et al. 1991). The diagnosis of von quent, especially in mucinous cystadenocarcinoma
Hippel-Lindau disease must always be considered (Table 1.3).
when multiple pancreatic cysts are found in an The precise pathophysiological basis for an individ-
asymptomatic patient without clinical suspicion of ual patient's complaint is almost impossible because
pancreatitis. Imaging and clinical evaluation of the the symptoms are so nonspecific. Even when present,
central nervous system, eyeball, kidneys, adrenal the clinical symptoms often do not warrant immediate
gland, and epididymis should be incorporated into concern; therefore, it is not uncommon that, in retro-
the work-up (GIRELLI et al. 1997). The presence of spect, the onset of symptoms occurred years (about a
symptoms related to the larger pancreatic lesions will year and a half in the case of serous tumors) before
require a careful evaluation of the type of treatment, the diagnosis (TALAMINI et al. 1992; YANG et al. 1994;
with particular attention given to the most conserva- WARSHAW et al. 1990; BASSI et al. 2002).
tive attitude, which will be dealt with in more detail The most common symptom is abdominal pain,
later (BASSI et al. 2002). Lastly, the mainly autosomal but in reality, only about 35% of patients suffer-
hereditary nature of the syndrome will require an ing from symptomatic serous and mucinous cystic
accurate report of the family case history data and
genetic screening of the patient's relatives, on the
basis of which method and time for follow-up can be Table 1.1. Symptoms of patients affected with serous cystic
planned so as to make an early diagnosis, especially tumors of the pancreas, surgically treated (n=44)
of renal neoplasm which is, at the moment, the main Paina 33 (75%)
risk and mortality factor in these patients. Anorexia and dyspepsia 7 (15.9%)
Weight loss and dyspepsia 6 (13.6%)
Obstruction of upper GI tractb 3 (6.8%)

1.2.2 a Five patients (11.3%) suffered from pain associated with


Symptoms dyspepsia
b One case with associated jaundice
The most common clinical complaint is some degree
of abdominal pain or discomfort. Other frequent
symptoms are weight loss, palpable mass, post- Table 1.2. Symptoms in patients with mucinous cystic tumors
prandial fullness, nausea, and vomiting. Onset with of the pancreas, surgically treated (n=41)
jaundice, acute pancreatitis, or bleeding is rare. Lastly, Pain 41 (lOO%)
the percentage of patients discovered incidentally is Weight loss and dyspepsia 14 (34%)
constantly increasing. Anorexia and dyspepsia 3 (7.3%)
In truth, both symptomatic cases and the percent- Jaundice 1 (2.5%)
age of incidental findings appear to vary depending
on the benign or malignant nature of the tumor.
In two American series (WARSHAW et al. 1990; Table 1.3. Symptoms of patients with mucinous cystadenocar-
cinoma, surgically treated (n=40)
PYKE et al. 1992),40% and one-third of all patients,
respectively, were asymptomatic, but when study- Pain 22 (55%)
ing the mucinous and the serous tumors separately, Weight loss and dyspepsia 25 (62.5%)
Anorexia and dyspepsia 21 (52.5%)
the percentage increases to 50% in the former and
Jaundice 5 (12.5%)
decreases to 20% in the latter. Malignant mucinous
Clinical Manifestations and Therapeutic Management 5

tumors experience a pain complex (such as radia- predictive value in discriminating between benign
tion to the flanks) which would suggest a possible or malignant cystic masses. When the cyst content
pancreatic etiology (BASSI et al. 2002; EGAWA et al. displays elevated Ca 19-9 and/or CEA levels and an
1994). Moreover, the clinical picture does not always elevated CA 72-4, the lesion should be assumed to
correlate to the location and dimension of the neo- be a malignant neoplasm and resected if possible
plasia. Therefore, the presence of 'pancreatic-type (HAMMEL et al. 1998; SPERTI et al. 1996). However, at
pain' is associated with a negative prognosis. Many least at the time of this writing, the presence and value
patients presenting with radiating pain will prove to of intracystic tumor markers should be considered in
have invasive cystadenocarcinoma infiltrating the the context of the patient's clinical presentation and
retroperitoneum. When such pain is accompanied by results of radiological studies (SAND et al. 1996).
symptoms like weight loss, anorexia, and jaundice, it It is hoped that molecular findings will further
is obvious why the suspicion of malignancy becomes improve specificity, but this is still in the experimen-
necessarily high. tal stages.

1.3 1.4
Laboratory Index of Suspicion from Clinical Data

1.3.1 By what has been previously stated, it is evident how


Hemo-Chemistry Routine the index of suspicion for pancreatic cystic tumor is
severely limited based on available clinical and labo-
Routine laboratory tests are only helpful in the work- ratory data. Only with imaging correlation can the
up and staging of pancreatic cystic neoplasia in terms pancreas be implicated as the source of the patient's
of further confirmation of the advanced stages of complaint.
malignant pathologies which have generally already Only in very particular cases, such as an already
been clinically and radiologically documented. Dia- diagnosed von Hippel-Lindau syndrome associated
betes is not thought to have a prognostic significance with specific pancreatic symptoms, can a suspicion
in peripheral tumors. of pancreas-related disease be suggested from the
clinical findings alone.
The appearance of specific pancreatic symptoms
1.3.2 associated with neoplastic marker positiveness, like
Markers Ca 19-9, as can be determined in the case of invasive
and/or metastatic cystadenocarcinoma, more easily
Unfortunately, serous tumor markers are neither leads to the suspicion of the more common advanced
diagnostic nor indicative of the biological behav- ductal adenocarcinoma. Thus, without the help of
ior of the lesion. However, aspiration of the fluid imaging, every suspicion of a cystic tumor cannot
content of the cyst may provide significant informa- reasonably be assumed.
tion (LEWANDROWSKI et al. 1993, 1995; GUPTA and
ALANSARI 1994). To gather these data and plan the
surgical treatment, it is necessary to perform a fine
needle aspiration biopsy (FNAB) to evaluate the 1.5
chemical nature of the cyst content and to elucidate From Assessment to Management
the presence of dysplastic or neoplastic cells. The
question is whether this approach is justifiable in all 1.5.1
cases of pancreatic cystic lesions or whether it should Diagnostic Suspicion
only be performed on patients selected on the basis
of the lesion's clinical and radiological characteristics Although there are rare exceptions, SCTs almost always
(see Section 1.5.1 Diagnostic Suspicion). demonstrate a benign biological and clinical course
The absolute concentrations of serum markers (GEORGE et al. 1989; KAMEl et aI. 1991; YOSHIMI et al.
found within the fluid content of cystic masses is 1992;ABE et aI. 1998; WIDMAIER et al.1996).Mucinous
variable (Ca 19-9, CEA, Ca 125). The presence of tumors are either already malignant at diagnosis or
newer markers, such as CA 72-4, may offer greater will degenerate in time (BASSI et al. 2001).
6 C. Bassi et al.

Diagnosis of tumor type is predominantly based 1.5.2


on the radiological findings since, as discussed Definite Diagnosis
above, the lack of specificity of the clinical picture is
not helpful. The discovery by imaging, usually ultra- Generally speaking, the biological behavior of cystic
sound, of a finding that could suggest a large cystic tumors of the pancreas leaves little room for the
lesion in the pancreatic area is frequently the starting conservative management, making three progres-
point. The patient should therefore be investigated in sive steps in their work-up and staging mandatory:
greater detail as regards the case history, clinical and confirmation of the intraglandular origin of the
morphological data to indicate which of the different neoplasia; exclusion of an inflammatory cause, i.e.,
radiological examinations should be carried out (CT, pseudocyst; identification of a surgical indication for
MR, MRCP, and ERCP). actual or potential malignancy (LE BORGNE 1998).
Another possible step is an image-guided FNAB This last concept could be better expressed as the
either with ultrasound or CT guidance. In our opin- correct identification of clearly benign serous tumors
ion, this procedure should not be carried out as a and, therefore, the only clear indication for conserva-
matter of routine for all cases of cystic pancreatic tive management (BASSI et al. 2001). If it cannot be
lesions, but only in selected patients. Information identified with absolute certainty in the diagnostic
gained from cytologic analysis of the cyst's content work-up, laparotomy is the next inevitable step
can undoubtedly be invaluable when positive. The even for serous tumors discovered casually during
inability to confirm malignancy cytologically does the course of a definitive and complete histological
not rule out its presence, and further investigations diagnosis. Resection, moreover, represents the only
are necessary. Another aspect arguing against the therapy possible in symptomatic cases except when
generalized use of FNAB is the aforementioned the risk of resection is unacceptable, and a digestive
lack of diagnostic reliability of serum markers. The bypass is performed to decompress the cyst (TALA-
procedure is best used for the detection of elevated MINI et al. 1992; YANG et al. 1994; GRIESHOP et al.
isoamylase levels in distinguishing a pancreatitis- 1994).
related fluid collection from a cystic neoplasm.
Neither the intracystic Ca 19-9 nor CEA is of any 1.5.2.1
help in distinguishing between serous and mucinous Operable Patients
cystadenomas (LEWANDROWSKI et al. 1993, 1995;
GUPTA and ALANSARI 1994). The appearance of the Suspected or established malignancy, persistent
aspirate - fluid and transparent on the one hand, symptomatology, and diagnostic confirmation rep-
and dense, streaky, and/or hemorrhagic on the other resent the principal surgical indications. The serial
- clearly distinguishes between the serous and muci- demonstration by imaging of volumetric growth of
nous forms. In the latter, a high concentration of Ca asymptomatic serous forms could be added (BASSI
19-9 and CEA is indicative of possible malignancy, et al. 2001; GRIESHOP et al. 1994; LE BORGNE 1998;
making it appears that these findings, associated PYKE et al. 1992; TALAMINI et al. 1992; WARSHAW et
with a high viscosity and a reliable cytologic result, al. 1990).
represent a definite final diagnosis of the disease. Once an indication has been established, the sur-
This procedure seems to be applicable in those gical approach will be based on the site and nature
patients whose radiological data suggest the suspi- of the lesion. In patients with suspect, potential, and
cion of a mucinous form when they are not suitable clear malignancy, the standard rules of oncology
for surgical resection due to their clinical condition. with resections from the left or widened pancreato-
It is better to proceed with the operation on the other duodenectomies together with a lymphadenectomy
patients, bearing in mind the absolute necessity of are followed (LE BORGNE 1998; PYKE et al. 1992;
giving the pathologist the entire lesion so that a PEDERZOLI et al. 2000).
complete analysis of the epithelium of the internal In the possibly benign forms, particularly in
covering of the cyst can be carried out. On the one young patients, more conservative approaches to the
hand, the intracavity pressure can strip whole areas, pancreatic parenchyma are also possible, for example
making them unsuitable for a reliable diagnosis, and intermediate resection and cephalic resection pre-
on the other, there is the possibility that the malig- serving the duodenum; laparoscopic resection is also
nant degeneration has a focal character, thus sparing possible for small (diameter less than 4 cm) lesions of
extensive areas of epithelium. the tail (PEDERZOLI et al. 2000).
Clinical Manifestations and Therapeutic Management 7

1.5.2.2 Bassi C, Falconi M, Pederzoli P (2001) Non adenocarcinoma


Inoperable Patients of the pancreas: cystic tumors. In: Schein M, Wise L (eds)
Controversies in surgery, vol 4. Springer, Berlin Heidelberg
New York, pp 130-139
Concerning the symptomatic tumors, the patient Bassi C, Salvia R, Molinari E, Biasiutti C, Falconi M, Pederzoli P
who cannot be operated on due to the local diffu- (2003) Management of 100 consecutive cases of pancreatic
sion of the disease or co-morbidity could undergo cystoadenoma: wait for symptoms and see at imaging, or
digestive or biliary bypass. When tumors of any vice versa. World J Surg, in press
Becourt PJ, Becourt G (1830) Recherches sur Ie pancreas: ses
nature appear clinically with obstructive symptoms,
fonctions et ses alterations organiques. Levrautl, Stras-
whether digestive or biliary, and are not suitable for bourg
traditional surgical correction, the endoscopic or Compagno J, Oertel JE (1978) Microcystic adenomas of the
radiological alternatives commonly carried out in pancreas (glycogen-rich cystadenomas): a clinicopatho-
nonresectable cases of ductal adenocarcinoma will logic study of 34 cases. Am J Clin Pathol 69:289-298
Egawa N, Maillet B, Schroder S, Mukai K, Kloppel G (1994)
be opted for.
Serous oligo cystic and ill-demarcated adenoma of the
It is also compulsory with these patients to per- pancreas: a variant of serous cystic adenoma. Virchows
form a cytology by aspiration of the primitive lesion Arch 424:13-17
or, if in stage IV, of the hepatic metastases, especially George DH, Murphy F, Michalski R, Ulmer BG (1989) Serous
when a neo-adjuvant therapy is possible. cystadenoma of the pancreas: a new entity? Am J Surg
Pathol 13:61-66
Girelli R, Bassi C, Falconi M, De San tis L, Bonora A, Caldiron E,
1.5.2.3 Sartori N, Salvia R, Briani GF, Pederzoli P (1997) Pancreatic
Follow-Up cystic manifestations in von Hippel - Lindau disease. Int J
PancreatoI22:101-109
The vast majority of patients undergoing radical Grieshop NA, Wiebke EA, Kratzer SS, Madura JA (1994) Cystic
neoplasms of the pancreas. Am Surg 60:509-514
resection for cystic tumors report relief of symptoms
Gupta RK, Alansari AG (1994) Needle aspiration cytology in
in the postoperative period. the diagnosis of mucinous cystadenocarcinoma of the pan-
As far as the mucinous forms are concerned, to creas. A study of five cases with an emphasis on utility and
obtain long-term survival, a distinction must be differential diagnosis. Int J PancreatoI15:149-153
made between the invasive and noninvasive types. Hammel P, Voitot H, Vilgrain V, Levy P, Ruszniewski P, Ber-
nades P (1998) Diagnostic value of Ca 72-4 carcinoembry-
The former, with peri tumoral infiltration outside
onic antigen determination in the fluid of pancreatic cystic
the cystic wall, has a survival rate of 5 years like that lesions. Eur J Gastroenterol Hepatol 10:345-348
of ductal pancreatic carcinoma, for less than 20% of Kamei K, Funabiki T, Ochiai M, Amano H, Kasahara M, Saka-
patients. Some 40% of patients with only cystic wall moto T (1991) Multifocal pancreatic serous cystoadenoma
invasion are still alive after 5 years, while all those with atypical cells and focal perineural invasion. Int J Pan-
creatoll0:161-172
who underwent radical surgical resection, with or
Kloppel G, Solcia E, Longnecker DS, Capella C, Sobin LH (1996)
without focal malignancy but only at the intratu- Histological typing of tumors of the exocrine pancreas. In:
moral level, are alive and well (ZAMBONI et al. 1999). Kloppel et al (eds) International histological classification
These considerations once again suggest how the of tumors, 2nd edn. Springer, Berlin Heidelberg New York
management of cystic lesions of the pancreas must Lawandrowski KB, Southern JF, Pins MR, Compton CC,
Warshaw AL (1993) Cyst fluid analysis in the differential
take on a totally aggressive attitude as the only guar-
diagnosis of pancreas cystic. A comparison of pseudo cysts,
antee of radical curative treatment in a large percent- serous cystadenomas, mucinous cystic neoplasms, and
age of patients. It is obvious that given the difficult mucinous cystadenocarcinoma. Ann Surg 217:41-47
staging and the risky treatment in terms of surgical Lewandrowski K, Warshaw A, Compton C (1992) Macrocystic
morbidity and mortality, it is better that such an serous cystadenoma of the pancreas: a morphologic vari-
approach be performed only in specialized centers ant differing from micro cystic adenoma. Hum Pathol 23:
871-875
with a vast amount of reference volumes of activity. Lewandrowski K, Lee J, Southern J, Centeno B, Warshaw A
(1995) Cyst fluid analysis in the differential diagnosis of
pancreatic cysts a new approach to the preoperative assess-
ment of pancreatic cysts lesions. AJR Am J Roentgenoll64:
815-819
References Le Borgne J (1998) Cystic tumors of the pancreas. Br J Surg
85:577-579
Abe H, Kubota K, Mori M, Miki K, Minagawa M, Noie T, Neumann HP, Dinkel E, Brambs H, Wimmer B, Friedburg H,
Kimura W, Makuuchi M (1998) Serous cystadenoma of the Yolk B, Sigmund G,Riegler P, Haag K, Schollmeyer P (1991)
pancreas with invasive growth: benign or malignant? Am J Pancreatic lesions in the von Hippel-Lindau Syndrome.
Gastroenterol 93: 1963-1966 Gastoenterology 101:465-471
8 C. Bassi et al.

Pederzoli P, Salvia R, Falconi M, Casetti L, Marcucci S, Bassi C Warshaw AL, Compton CC, Lewandrowski K, Cardenosa G,
(2000) Cistoadenomi e cistoadenocarcinomi del pancreas: Mueller PR (1990) Cystic tumors of the pancreas. New
inquadramento generale e strategia chirurgica. Osp Ital clinical, radiologic, and pathologic observation in 67
Chir 6:435-440 patients. Ann Surg 212:432-443
Pyke CM, van Heerden JA, Colby TV, Sarr MG, Weaver AL (1992) Widmaier U, Mattfeld T, Siech M, Beger HG (1996) Serous
The spectrum of serous cystoadenoma of the pancreas. Clini- cystadenocarcinoma of the pancreas. It J Pancreatol 20:
cal, pathologic, and surgical aspects. Ann Surg 215:l32-l39 l35-l39
Sand JA, Hyoty MK, Mattila J, Dagorn JC, Nordback IH (1996) Yang EYT, Joehl RJ, Talamonti MS (1994) Cystic neoplasms of
Clinical assessment compared with cyst fluid analysis in the pancreas. J Am Coll Surg 179:747-757
the differential diagnosis of cystic lesions in the pancreas. Yoshimi N, Sugie S, Tanaka T, Aijin W, Bunai Y, Tatematsu A
Surgery 119:275-280 Okada T,Mori H (1992) A rare case of cystadenocarcinoma
Sperti C, Pasquali C, Guolo P, Polverosi R, Liessi G, Pedrazzoli of the pancreas. Cancer 69:2449-2453
S (1996) Serum tumors markers and cystic fluid analysis Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini
are useful for the diagnosis of pancreatic cystic tumors. G, Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM,
Cancer 78:237-243 Klappel G (1999) Mucinous cystic tumors of the pancreas:
Talamini MA, Pitt HA, Hruban RH, Boitnott JK, Coleman J, clinicopathological features, prognosis, and relationship
Cameron JL (1992) Spectum of cystic tumors of the pan- to other mucinous cystic tumors. Am J Surg Pathol 23:
creas. Am J Surg 163:117-123 410-422
2 Pathology of Cystic Tumors
G. ZAMBONI, P. CAPELLI, A. PEsel, and A. BRIGHENTI

CONTENTS techniques, such as ultrasonography (US) and com-


puted tom6graphy (CT). They have been encountered
2.1 Introduction 9
in 1.4% of all abdominal CT scans (PARIENTY et al.
2.2 Serous Cystadenomas 10
2.2.1 Macroscopy 10 1980), in 10% of all pancreatic cysts (HODGKINSON
2.2.2 Microscopy 11 et al. 1978; WARSHAW et al. 1990) and in 1%-7.1% of
2.2.3 Differential Diagnosis 12 all pancreatic neoplasms (CUBILLA and FITZGERALD
2.2.4 Treatment and Outcome 14 1984; MOROHOSHI et al. 1983).
2.3 Mucinous Cystic Neoplasms 14
Although all pancreatic neoplasia, including
2.3.1 Macroscopy 15
2.3.2 Microscopy 16 ductal adenocarcinoma, may clinically appear as a
2.3.3 Pathogenesis --17 cystic lesion due to degenerative changes in a solid
2.3.4 Differential Diagnosis 18 tumor (ADSAY et al. 2000b), the most typical cystic
2.3.5 Treatment and Outcome 19 neoplasms of the pancreas are cysts lined by differ-
2.4 Solid Pseudopapillary Tumor 20
ent epithelium. Two decades ago, the classification of
2.5 Acinar Cell Cystic Tumors 23
2.5.1 Acinar Cell Cystadenoma 23 cystic tumors of the pancreas seemed straightforward,
2.5.2 Acinar Cell Cystadenocarcinoma 24 and two lesions were distinguishable: micro cystic or
2.6 Other Cystic Tumors 25 glycogen-rich adenomas (also called serous cystic),
References 25 with an excellent prognosis and only minimal risk
of malignant transformation, and mucinous cystic
neoplasms (MCNs), which encompass a broad spec-
trum of morphological changes and showed 'overt' or
2.1 'latent' malignancy (COMPAGNO and OERTEL 1978b;
I ntrod uction COMPAGNO et al. 1979; GIBSON and SOBIN 1978).
It must be stressed that the main diagnostic cri-
The category of cystic lesions of the pancreas includes terion is the morphology of the epithelium, whereas
cystic formations of differing morphology, pathogen- the shape and volume of cysts are of no importance
esis, and biology. Although these lesions are uncom- (KU'lPPEL et aL 1996). The spectrum of pancreatic
mon compared with solid tumors of the pancreas, tumors with cystic features is broad and includes
they constitute an important group of lesions with serous micro cystic or oligocystic (macro cystic ) tumor
distinctive clinical and pathological features. The (COMPAGNO and OERTEL 1978a; EGAWA et al. 1994;
therapeutic approach and the prognosis differ sub- LEWANDROWSKI et al. 1992), MCNs (SOLCIA et al.
stantially, depending on the specific diagnosis (ADSAY 1997; ZAMBONI et al. 2000), acinar cell cystadenoma
et al. 2000b). An incorrect diagnosis may therefore (ZAMBONI et al. 2002), acinar cell cystadenocarci-
result in inappropriate therapy, leading for instance noma (CANTRELL et al. 1981; STAMM et al. 1987},solid
to incomplete removal of a tumor that requires total pseudopapillary tumor with cystic degeneration (i.e.,
resection to prevent malignant degeneration (SACHS et solid cystic tumor) (KU'lPPEL et al. 1991; MATSUNOU
al. 1989; WARSHAW and RUTLEDGE 1987). The current et al. 1990), and endocrine tumor with cystic degen-
interest in identifying cystic pancreatic lesions arises eration (IACONO et al. 1992). Nonneoplastic cysts,
from their easier recognition with modern imaging such as congenital cysts (KLOPPEL 2000), single true
cysts (HOWARD 1989), lymphoepithelial cysts (ADSAY
et al. 2000), enterogenous cysts (PILCHER et al. 1982),
G. ZAMBONI, MD; P. CAPELLI, MD; A. PEsel, MD;
A. BRIGHENTI, MD and endometrial cysts (MARCHEVSKY et al. 1984), are
Department of Pathology, University of Verona, Strada Le extremely rare, but an exact estimation of their true
Grazie 8,37134 Verona, Italy prevalence is so far not available.
10 G. Zamboni et al.

The World Health Organization (WHO) clas- tions were found in either K-ras or p53. No patient
sification of exocrine pancreatic tumors (KU)PPEL showed micro satellite instability of the type seen in
et al. 1996) is used in this chapter to classify cystic mismatch repair-deficient tumors.
neoplasms. It divides neoplasms on the basis of their SCAs may coexist either with other pancreatic
histological type and biological behavior into benign tumors or with extrapancreatic neoplasms. In the
tumors, borderline tumors, and malignant tumors. pancreas, SCAs were found to be associated with
both ductal adenocarcinoma (MONTAG et al. 1990)
and endocrine tumor (KEEL et al. 1996).
A precise histogenesis has not been established.
2.2 Ultrastructural analogies with normal pancreatic
Serous Cystadenomas centro acinar cells have been demonstrated (LAITIO
et al. 1974), as well as the presence of abundant intra-
Serous cystadenomas are cystic tumors formed by cytoplasmic glycogen in fetal centro acinar cells, which
glycogen-rich, periodic acid-Schiff (PAS)-positive is almost absent in normal adult centroacinar cells
epithelial cells which produce serous fluid. They have (LAITIO et al. 1974).
frequently been reported as micro cystic or glycogen- SCAs in our series represent 2% of all pancreatic
rich cystadenomas. They differ from the mucinous tumors and 44% of cystic tumors. We observed a
neoplasms of the pancreas, both MCNs and intra- marked female predominance and a mean age (57
ductal papillary mucinous neoplasms (IPMNs), years) which was higher than that of benign MCNs
which are potentially or overtly malignant. The (47 years) and almost identical to that of malignant
absence of atypia and the indolent course are the two MCNs (55 years). Topographically, the body-tail
most important characteristics of this tumor. Serous location was similar in frequency to the pancreatic
tumors are classified in the WHO classification into head. Two patients had diffuse pancreatic multi cystic
serous cystadenoma (SCA), the vast majority of cases lesions associated with VHL syndrome. None of the
being clearly benign, and serous cystadenocarcinoma lesions in our 61 patients exhibited malignant behav-
(SCAC), with only a few reported cases. SCAs are sub- ior; nevertheless, a diffuse involvement of the entire
classified according to the macroscopic variation of pancreas was observed later in 3 patients. One patient
the cysts into serous micro cystic adenoma and serous died of concomitant pancreatic ductal carcinoma.
oligocystic or macro cystic adenoma.
SCAs, which account for 1%-2% of all exocrine
neoplasms, occur more often in women at an aver- 2.2.1
age age of 66 years (KU:>PPEL et al. 1996). About one- Macroscopy
third of patients are asymptomatic, and the tumors
are discovered incidentally. Two-thirds of patients SCAs frequently appear as a well-circumscribed,
complain of symptoms related to local mass effects, round, cystic mass filled with clear watery fluid and
such as abdominal pain, nausea, and vomiting; in with bulging edges (Fig. 2.1). The lesions range in
some cases, the mass can be palpable.
Clinically, they can be sporadic or associated
with von Hippel-Lindau syndrome (VHL). Pancre-
atic involvement is frequent in VHL, occurring in
60%-80% of patients. In these patients, the pancreatic
lesions are diffuse or multifocal (HORTON et al. 1976)
and frequently associated with cysts in other organs
(liver, lung, kidney, spleen, epididymis) (GIRELLI et al.
1997). There is genetic evidence supporting the role of
the VHL gene, located at chromosome 3p25.5, in SCA
tumorigenesis, in both VHL-associated and sporadic
pancreatic SCAs (VORTMEYER et al. 1997). In a study
by our group (MOORE et al. 2001), we demonstrated
the involvement of chromosomal arms 3p in 40% of
our SCAs and VHL gene involvement in a subset of
sporadic cases. Loss of chromosome lOq was the most Fig. 2.1. Serous cystadenoma. Cut section showing a well-cir-
frequent event in SCAs (50% of patients). No muta- cumscribed, round mass with bulging edges
Pathology of Cystic Tumors 11

diameter from 1 to 25 cm (average 6-11 cm). This


broad size range encompasses large lesions which
are often symptomatic and small lesions incidentally
found in pancreas resected for other reasons or dis-
covered during radiological study for unrelated prob-
lems. They are usually single, but in some cases they
can be multifocal or characterized by contiguous and
confluent tumors, which can rarely extend to involve
the entire parenchyma. seAs show no communication
with the pancreatic ducts.
Macroscopically, they are subdivided according to
the number and dimensions of the individuallocules
into two subtypes: micro cystic and macro cystic.
a

1. Microcystic or classic type (KU)PPEL et al. 1996):


The tumors appear as relatively well-circumscribed
lesions with multilobular margins. On cut section,
they are sponge-like, formed by innumerable cysts
which range from 1 to 5 mm in diameter, larger ones
(up to 1-2 cm) being rarer and frequentlyperipher-
ally located. The cysts are filled with a clear, watery
fluid. Typically, they have a central stellate scar made
up of white, fibrous nodules with peripheral fibrous
bands (Fig. 2.2). The scar frequently shows calcium
deposits, the pathological basis of the typical 'sun-
burst' pattern of calcification seen on radiological
studies. In our series of 61 patients, the microcystic
b
variant was seen in the large majority of cases (38
patients, 64%); 31 patients were women and 7 men, Fig. 2.2a,b. Serous cystadenoma, micro cystic type. a Gross
with an average age of 61 years. appearance showing a well-circumscribed tumor made up
of innumerable small cysts, with central scar and peripheral
fibrous bands. b Whole-mount macrosections showing the
2. Macrocystic or oligo cystic type (EGAWA et al. 1994; small cysts and the central fibrous scar
LEWANDROWSKI et al. 1992): These tumors, charac-
terized by a small number oflocules, are less demar-
cated than the micro cystic variant due to the pres- The macro cystic subtype, which frequently contains
ence of cysts extending into the adjacent pancreatic hemorrhagic fluid and calcifications, is seldom dif-
parenchyma. The cyst fluid may vary from classic ferentiated preoperatively from either pseudocyst or
clear and watery to bloody and brown. The cut sur- mucinous cystic tumors and may occasionally be con-
face shows the presence of a countable number of fused with these entities microscopically (LEWAND-
cysts (oligocystic: Fig. 2.3a) or sometimes a single ROWSKI et al. 1992). The final pathological diagnosis
cyst (unilocular: Fig. 2.3b) with a variable diameter may necessitate extensive sampling of the cyst wall,
of 2-15 cm. The oligo cystic variant characteristically because epithelial denudation occurs frequently. The
lacks a central scar and usually has ill-defined tumor correct diagnosis of macro cystic serous adenoma is of
growth, whereas the multilocular variant may show the utmost importance, since surgical resection may
a central scar (Fig. 2.3c). In our series, 21 patients not be necessary in patients at high surgical risk.
(36%) had unilocular or multilocular macro cysts,
ranging in dimension from 2 to 7 cm. This subtype
was far more common in women (20 women and 1 2.2.2
man); the average age was 50 years. Fourteen lesions Microscopy
could be considered 'pure', whereas 7 had a mixture
of micro- and macrolocules. The patients with 'pure' Microscopically, the micro cystic and macro cystic
macro cystic types were younger than those with the types are indistinguishable and show the typical
'mixed' types: 43 vs 51 years. cuboidal to almost flat serous epithelium charac-
12 G. Zamboni et al.

terized by clear cytoplasm and round nuclei with


inconspicuous nucleoli. Neither cytologic atypia
nor mitotic activity is found in either type. Intra-
cellular glycogen is characteristic of this neoplasm
(diastase-sensitive PAS-positive deposits) (Fig. 2.4).
Intracellular mucins are not present. Occasional
cases may show the presence of intracystic tufting
of the epithelium with the formation of true papil-
lae (Fig. 2.5). In some cases and in focal areas, the
cytoplasm may be eosinophilic and granular. The
neoplastic stroma is highly vascular, creating a fine
supporting network. The septa separating the larger
cysts are hyalinized and contain hemosiderin-laden
macrophages and sometimes entrapped islets of
Langerhans and exocrine acini. Central fibrous scar
formed by hyalinized tissue is present in the major-
ity of micro cystic types, frequently associated with
calcification. Although the micro cystic variant is
usually more sharply demarcated, both types lack a
well-formed fibrous pseudocapsule.
Serous adenomas with identical cytological fea-
tures and immunohistochemical profile without cystic
locules and characterized by an exclusively solid pat-
tern of growth are reported as solid serous adenomas
(PEREZ-ORDONEZ et al. 1996; COMPTON 2000) (Fig.2.6).
They are usually small lesions, measuring 2-4 cm at
the most and are made up of small acini lined by typi-
cal serous cells.
b

c
Fig. 2.3a-c. Serous cystadenoma, oligo cystic or macrocystic
type. Whole-mount macrosections showing an ill-defined
lesion made up of a few large cysts extending into the pan-
creatic parenchyma (a), unilocular lesion with a thick fibrous
pseudo capsule (b), and multilocular tumor with central fibrous
scar adjacent to the duct of Wirsung (c)

Fig. 2.4a,b. Serous cystadenoma. Typical histological appear-


ance of lining epithelium, composed of cuboidal cells with
clear cytoplasm (a), filled with PAS-positive glycogen (b) b
Pathology of Cystic Tumors 13

honeycomb appearance, is almost always correctly


identified at preoperative imaging. In these cases,
the pathologist is asked to confirm a radiologic
diagnosis. Serous micro cystic adenoma has to be
differentiated from solid-pseudopapillary tumor of
the pancreas (ZAMBONI et al. 1993), from endocrine
tumor with clear cells, and from metastatic clear-cell
carcinoma of the kidney. An interesting differential
diagnosis is with a clear-cell 'sugar' tumor of the
pancreas (ZAMBONI et al. 1996), an extremely rare
tumor belonging to the family of lesions character-
ized by the presence of perivascular epithelioid cells
(PEC) and described in mUltiple organs and sites
Fig. 2.5. Serous cystadenoma. Small intracystic papillary
(PEA et al. 1991, 1996; BONETTI et al. 1992, 1994).
structures
The latter is characteristically HMB45-positive and
cytokeratin -negative.
The preoperative radiological differential diagno-
sis of the macro cystic or oligo cystic variant of serous
adenoma with pseudo cysts and mucinous cystic
neoplasms is at the moment extremely difficult. In
fact, these two lesions frequently appear as either
oligo cystic or unilocular cysts.
Fine needle aspiration biopsy under ultrasound
guidance for cytological and biochemical analysis
may help in the differential diagnosis (YOUNG et al.
1991). The cytological smears are characterized by
sparse cellularity in a background devoid of debris
and, most importantly, of mucus. The neoplastic
cells, individually dispersed or rarely aggregated in
a flat sheet with a honeycomb pattern, have a mono-
Fig. 2.6. Serous cystadenoma, solid type. Whole-mount macro- morphic appearance with poorly defined cytoplasm
section showing a small lesion, measuring 2 cm at its largest, and small, round, oval nuclei. PAS staining performed
made up of small, packed acini with and without diastase shows abundant cytoplas-
mic glycogen and the absence of mucins. Frequently,
histiocytes and hemosiderin-laden macrophages are
Immunohistochemically, the epithelium shows present. Immunohistochemical staining for cyto-
diffuse cytoplasmic staining for low-molecular- keratin and antimacrophage markers can help in
weight cytokeratins (CK 7,8, 18 and 19) and diffuse the diagnosis (Fig. 2.7). The sensitivity of cytologi-
membrane staining using antibodies against epithe- cal examination is only about 50%-60% because of
lial membrane antigen (EMA) (SOLCIA et al. 1997; inadequate sampling, with an absence of diagnostic
ALPERT et al. 1988; ALBORES-SAAVEDRA et al. 1990). epithelial components or poor cellular preserva-
The neoplastic cells may focally express CA 19-9 and tion (CARLSON et al. 1998; CENTENO et al. 1994;
B72.3 (ISHIKAWA et al. 1998) but are CEA-negative LAUCIRICA et al. 1992).
(SOLCIA et al. 1997). Endocrine and acinar cell dif- The biochemical and immunological analysis of
ferentiation is not found (SOLCIA et al. 1997). the cystic fluid may assist in the differential diag-
nosis with mucinous tumors and pseudo cysts. The
cystic fluid of serous adenoma does not have the
2.2.3 high levels of either enzymes (amylase and lipase)
Differential Diagnosis or tumor markers (CEA, CA 19.9, CA 125) observed
in pseudo cysts and mucinous tumors, respectively
The differential diagnosis of SCAs differs according (LEWANDROWSKI et al. 1993; COMPTON 2000).
to the two morphological subtypes. The microcys- The pathological differential diagnosis with
tic variant, which has a typical macroscopic solid lymphangiomas is quite easy morphologically due to
14 G. Zamboni et al.

a b

Fig.2.7a,b. Serous cystadenoma. a Fine needle aspiration biopsy specimen showing sparse cellularity with epithelial-like cells mixed
with macrophages with foamy cytoplasm; b immunohistochemical stain for cytokeratins 8, 18, 19 in the neoplastic cells

the presence of flat cells lacking glycogen and immu- 2.3


noreactivity for factor VIII and cytokeratins (SOLCIA Mucinous Cystic Neoplasms
et al. 1997).
These cystic tumors, occurring almost exclusively
in women, are formed of epithelial cells producing
2.2.4 mucin, supported by ovarian-type stroma, showing
Treatment and Outcome no communication with the pancreatic ductal system
(Fig. 2.8). According to the grade of epithelial dys-
Surgical resection is only necessary when the plasia, they may be classified into adenoma, border-
tumors are symptomatic and the benefits of the line tumor, and carcinoma, noninvasive or invasive
operation outweigh the risk (SOLCIA et al. 1997) or (KU:'>PPEL et al. 1996; ZAMBONI et al. 2000).
to prevent the development of complications such In 1978, Compagno and Oertel (COMPAGNO and
as erosion into adjacent organs, hemorrhage, and OERTEL 1978b) reported their experience with 41
biliary and gastrointestinal obstruction. Resection patients with MCN. They defined these tumors as
may be contraindicated in elderly or asymptomatic cystic masses containing mucin-producing colum-
patients, and follow-up examination can be accept- nar epithelium supported by 'ovarian-like stroma'
able (COMPAGNO and OERTEL 1978a; WARSHAW et al. and emphasized that all MCNs of the pancreas must
1990; HODGKINSON et al. 1978). be considered and treated as potentially malignant,
The prognosis of both microcystic and macrocys- regardless of their epithelial differentiation. They
tic types of serous cystadenoma is excellent. How- therefore discouraged the use of the terms 'cyst-
ever, there are rare exceptions in which there is local adenoma' and 'cystadenocarcinoma' and instead
destructive growth with progressive involvement of proposed the term 'mucinous cystic neoplasm with
the entire pancreas and the slight risk of malignant overt or latent malignancy'. Although their work
transformation. In a recent review of SCAC of the pan- resulted in the appropriate therapy for MCNs, i.e.,
creas' Compton found 11 reported cases (COMPTON complete resection instead of a drainage procedure,
2000). However, the interpretation of these neoplasms it prevented a more individualized approach to the
as malignant is not so clear. In fact, they show the tumors, such as that developed for MCN s of the ovary
features of aggressive growth with perineural and (RUTGERS and SCULLY 1988; BOSTWICK et al. 1986).
perivascular invasion, involvement of regional lymph However, a cystic structure and the presence of
nodes and adjacent organs, such as the stomach and mucin-secreting epithelium are not sufficient to clas-
liver. Surgical resection in all cases proved to be cura- sify a neoplasm as an MCN (COMPAGNO and OERTEL
tive, however; not one patient died of disease (GEORGE 1978b; FURUKAWA et al. 1992; YAMADA et al. 1991),
et al. 1989; ABE et al. 1998; KAMEl et al. 1992; OHTA because IPMNs with duct ectasia and mucin hyper-
et al. 1993; YOSHIMI et al. 1992; ZIRINSKY et al. 1984; secretion (YAMADA et al. 1991; RICKAERT et al. 1991)
FRIEDMAN 1990; WIDMAIER et al. 1996; ALPERT et al. also show these features. While most cystic tumors
1988; FUJII et al. 1998; COMPTON 2000). are easily differentiated from each other, distinguish-
Pathology of Cystic Tumors 15

a b

Fig.2.8a,b. Mucinous cystic tumor. A round cystic lesion in the tail of the pancreas. The pancreatic duct, which does not com-
municate with the cyst lumen, has been opened (a); pancreatography of the resected specimen shows no connection between
the pancreatic ducts and the cystic tumor (b)

ing MCNs from the duct ectatic variant of IPMN We reviewed the anatomic and clinical features of
may be difficult. This histological similarity has led our panel of 56 mucinous cystic tumors (ZAMBONI
some pathologists to interpret MCN and IPMN as et al. 1999). All patients were women, ranging in age
a single entity (ITAI et al. 1986). A clear distinction from 18 to 78 years (mean 48.2 years). The 56 MCNs
between these two tumors therefore requires precise included 22 mucinous cystic adenomas (MCAs), 12
definitions: (1) MCN has unilocular or multilocular mucinous cystic borderline neoplasms (MCBs), and
cysts with no or minimal connection with the main 22 mucinous cystic carcinomas (MCCs). Sixteen of
pancreatic duct, lined by mucin -secreting epithelium the MCCs were invasive. The difference between the
supported by ovarian type stroma (YAMADA et al. mean ages of MCC patients (54.2 years) and patients
1991); (2) IPMN is characterized by dilatation of the with MCA and MCB (44.7 years) was statistically sig-
main and/or branch ducts, mucinous epithelium with nificant (p<0.04).
mucus hyperproduction, and intraductal growth
(RICKAERT et al. 1991; MOROHOSHI et al. 1989).
MCN is still considered a rare lesion, represent- 2.3.1
ing approximately 2%-5.7% of all exocrine pancre- Macroscopy
atic tumors (SOLCIA et al. 1997; THOMPSON et al.
1999; WILENTZ et al. 1999,2000). These figures may The vast majority of lesions occur in the body-tail
be biased because of the change in the diagnostic of the pancreas (COMPAGNO and OERTEL 1978b;
criteria over the years and the probable over-rep- THOMPSON et al. 1999; WILENTZ et al. 1999; YAMADA
resentation of MCNs in pathology files due to their et al. 1991; ZAMBONI et al. 1999). The head is only
high resectability rate compared with that of ductal rarely involved, with a predilection for mucinous
adenocarcinoma (ZAMBONI et al. 2000). cystadenocarcinomas (THOMPSON et al. 1999; ZAM-
It is evident from all reports on MCNs that the BONI et al. 1999).
neoplasms are much more common in women than MCNs appear as a round mass with a smooth sur-
in men (SOLCIA et al. 1997; THOMPSON et al. 1999; face and a fibrous pseudo capsule of variable thick-
WILENTZ et al. 1999) or occur almost exclusively in ness and with frequent calcifications. The diameter
women (ZAMBONI et al. 1999). It is likely that many of of the tumor ranges from 2 to 35 cm, with an average
the cases reported in men in the early literature were between 6 and 10 cm. The cut section shows either
IPMNs (COMPAGNO and OERTEL 1978b). The peak unilocular or multilocular tumor with cystic spaces
age occurs in the fifth decade (range 20-82 years, ranging from a few millimeters to several centime-
mean 49 years) (SOLCIA et al. 1997). The clinical ters in diameter, containing either thick mucin or a
presentation depends on the size of the tumor. Small mixture of mucin and hemorrhagic-necrotic mate-
tumors are generally found incidentally, whereas rial. The internal surface of unilocular tumors is
larger tumors produce symptoms which are usually usually smooth and glistening (Fig. 2.9), whereas the
derived from compression of adjacent structures and multilocular tumors often show papillary projections
are often accompanied by a palpable abdominal mass and mural nodules (Fig. 2.10). There is no significant
(SOLCIA et al. 1997). difference in size among the different categories of
16 G. Zamboni et al.

Fig. 2.9. Mucinous cystic tumor. Benign unilocular tumor with Fig. 2.10. Mucinous cystic tumor. Malignant tumor with mul-
smooth and glistening internal surface tiple, solid, nodular areas

MCN, whereas the malignancy of the tumors cor- differentiation. About half of the tumors contain
relates significantly with the presence of papillary scattered argyrophil and argentaffin endocrine
projections and/or mural nodules and multilocu- cells at the base of the columnar cells (ALB ORES-
larity (ZAMBONI et al. 1999). As a rule, there is no SAAVEDRA et al. 1987; COMPAGNO and OERTEL
communication between the tumor and the duct of 1978b; YAMAGUCHI and ENJOJI 1987). The spectrum
Wirsung or the secondary ducts, but exceptions have of differentiation ranges from histologically benign
been reported (YAMADA et al. 1991). columnar epithelium to severely atypical epithelium.
MCNs are classified according to the degree of epi-
thelial dysplasia into the following different types, as
2.3.2 suggested by the WHO.
Microscopy Mucinous cystadenomas (MCA) show only mild
epithelial dysplasia characterized by a slight increase
MCNs show two distinct components: an inner in the size of the basally located nuclei and the
epithelial layer and an outer densely cellular layer absence of mitoses.
of 'ovarian-like' stroma (Fig. 2.11). The epithelium Mucinous cystic neoplasms of borderline malignant
frequently displays areas with pseudopyloric, gastric potential (MCB) exhibit moderate dysplasia, charac-
foveolar, small-intestinal, colonic, and squamous terized by papillary projections or crypt -like invagina-
tions, and cellular pseudostratification with crowding
of atypical nuclei with rare mitoses (Fig. 2.12).
Mucinous cystadenocarcinomas (MCC) have
severe dysplasia/carcinoma-in-situ changes which
usually occur focally and may only be detected after
careful search of multiple sections from different
regions of the tumor. The epithelial cells, which often
form papillae with irregular branching and budding,
show nuclear stratification, severe nuclear atypia, and
frequent mitoses. The presence of carcinomatous
stromal invasion characterizes the invasive mucinous
cystadenocarcinoma. The invasive component usu-
ally resembles the common ductal adenocarcinoma
(Fig. 2.13). However, MCCs with invasive adeno-
Fig. 2.11. Mucinous cystic adenoma. Cyst wall lined with squamous carcinoma, osteoclast-like giant cells, or
mildly dysplastic columnar epithelium supported by 'ovar- choriocarcinoma have been reported (COMPAGNO
ian-like' stroma and OERTEL 1978b; POSEN 1981; REGO et al. 1991;
Pathology of Cystic Tumors 17

ZAMBONI et al. 1999; MOLBERG et al. 1998; WESTRA


et al. 1998). Occasionally, MCNs have mural nodules
with a sarcomatous stroma or an associated sarcoma
(WENIG et al. 1997; THOMPSON et al. 1999; ZAMBONI
etal.1999).
In the series we reported in 1999, the invasive
carcinomatous foci of 16 MCCs were limited to the
intratumoral septa in 3 cases (19%), to the tumor wall
in 5 cases (31 %), of which 3 were at the site of a previ-
ous cystojejunostomy, and in 8 cases (50%) there was
peri tumoral extension. The 7 grossly visible mural
nodules were undifferentiated carcinoma (1), adeno-
squamous carcinomas (2), osteoclast-like giant-cell
tumors (2), malignant sarcoma (1), and reactive
inflammatory process (1) (ZAMBONI et al. 1999).
Fig. 2.13. Mucinous cystadenocarcinoma. Cyst wall with tubu-
The epithelial cells show immunoreactivity to lar-type invasive carcinoma within the intratumoral stroma
epithelial markers including EMA, CEA, cytokeratins (bottom)
7,8,18 and 19, to gastric-type mucin markers (PGII
and M1), and to the pancreatic-type mucin marker
DUPAN-2. Noninvasive MCNs are positive for (PR) (Fig. 2.14b), and estrogen receptors (ER). The
MUC5AC and DPC4 and negative for MUCl, whereas luteinized cells stain for alpha-inhibin (Fig. 2.14c)
the carcinomatous invasive component is DPC4-neg- (ZAMBONI et al. 1999).
ative and MUCl-positive (LUTTGES et al. 2002).
The ovarian-like stroma, composed of densely
packed, spindle-shaped cells with round or elongated 2.3.3
nuclei and sparse cytoplasm, frequently exhibits a Pathogenesis
variable degree ofluteinization (Fig. 2.14a). The stro-
mal cells are positive for vim entin, smooth-muscle Pancreatic MCNs are exclusively, or almost exclusively,
actin, and desmin (focally), progesterone receptors limited to women (WARSHAW et al. 1990; COMPAGNO
and OERTEL 1978b; YAMAGUCHI and ENJOJI 1987;
ALBORES-SAAVEDRA et al. 1987), a feature that they
share with their counterparts in the liver (AKWARI et
al. 1990; WHEELER and EDMONDSON 1985) and retro-
peritoneum (TENTI et al. 1994). All these mucinous
cystic tumors can be considered a biphasic tumor
with the same pattern of cell lineage differentiation
in both the epithelium and the stroma. The stromal
component stains for PR, ER (CZERNOBILSKY et al.
1989; ZAMBONI et al. 1994), and inhibin, indicating
stromal luteinization (ZAMBONI et al. 1999). Stro-
mal luteinization was found in a similar percentage
in ovarian MCNs (SCULLY 1979; ROME et al. 1981;
FLEMMING et al. 1996). The demonstration of inhibin
in ovarian (FLEMMING et al. 1996; ZHENG et al. 1997)
and in pancreatic MCNs is of special interest, because
inhibin has proved to be a sensitive serum marker for
ovarian tumors, including MCNs (HEALY et al. 1993).
Inhibin may therefore also serve as a tumor marker
for pancreatic MCNs. These findings indicate a strik-
ing resemblance between pancreatic, hepatobiliary,
Fig. 2.12. Mucinous cystic neoplasm of borderline malignant
retroperitoneal, and ovarian MCNs, which suggests
potential. Papillary projections lined with moderately dysplas- a common pathway of tumor development. The pos-
tic epithelium sible derivation of the stromal component of MCNs
18 G. Zamboni et al.

--
,-
• •
;.--

a " b

Fig. 2.14a-c. Mucinous cystic neoplasm. Ovarian-type stroma


with luteinized cells, characterized by clear cytoplasm, large
nuclei with evident nucleoli (a), showing immunostaining for
c progesterone receptors (b) and inhibin (c)

from the ovarian primordium is supported by its of the pancreas. This could explain the fact that the
morphology, tendency to undergo luteinization, and pancreatic head is only rarely involved by MeN. The
immunophenotypic features. One possibility is that occurrence of retroperitoneal MeN can easily be
there may be ectopic ovarian stroma in the pancreas related to the localization of the genital ridges.
(particularly in the tail), along the biliary tree, and in
the retroperitoneum, which may release hormones
and growth factors, causing endodermally derived 2.3.4
epithelium in its vicinity to proliferate and form cystic Differential Diagnosis
tumors. It is important to note that the left primordial
gonad and the dorsal pancreatic anlage, which gives The differential diagnosis of MeN is usually easy
rise to the pancreatic body and tail and a small part from serous cystadenoma, mucinous noncystic carci-
of the pancreatic head, lie side by side at an early noma, acinar cystadenocarcinoma, solid -pseudopap-
stage (4th to 5th week) of development (NISHIMURA illary tumor, and cystic endocrine tumor because of
1983; TUCHMANN-DuPLESSIS 1968). Theoretically, the lack of mucin-producing columnar epithelium.
therefore, during that period, primordial ovarian cells It may be more difficult to distinguish MeN from a
could easily become incorporated into the pancreas, pancreatic pseudocyst (SACHS et al. 1989; WARSHAW
explaining the predilection of MeN for the body-tail and RUTLEDGE 1987; REMINE et al. 1987; TALAMINI
region of the pancreas. The development ofhepatobili- et al. 1992). In our series, approximately 10% of the
ary MeN may be due to a similar process because the patients were initially treated by cystojejunostomy,
hepatobiliary anlage is also found in close proximity suggesting that they were mistaken for a pseudocyst.
to the right genital ridge. The hepatobiliary anlage, Differentiating a pseudocyst from MeN may not only
on the other hand, separates the ventral pancreatic be a clinical problem but also a morphological one,
anlage from the genital ridge, since it buds off from since in MeN, due to degenerative processes, the
the duodenum between the ventral and dorsal anlage tumor epithelium may be denuded and the contents
Pathology of Cystic Tumors 19

hemorrhagic. The necessity of appropriate sampling 2.3.5


for making the correct diagnosis must be emphasized Treatment and Outcome
(COMPAGNO and OERTEL 1978b).
The best approach to obtain an exact preoperative The treatment of choice is complete surgical resec-
diagnosis is the combined evaluation of all available tion, with the lesion being examined histologically
clinical, serological, radiological, and cytological find- in its entirety to avoid designating an invasive MCN
ings. An increase in the peripheral blood serum tumor incorrectly as an adenoma, borderline tumor, or in-
markers CEA or CA 19-9 or high cyst fluid levels of situ carcinoma (WILENTZ et al. 2000). Internal (cyst-
CEA, CA-19-9, TAG-72, CA-15-3,or the mucin-like car- enteric anastomosis) or external drainage of the cystic
cinoma-associated antigen, together with a low amylase tumors has to be avoided, since there are many well-
level, is suggestive of MCN. The highest levels of these documented cases of apparent histologically benign
markers are seen in cystadenocarcinoma (YAMAGUCHI mucinous cystic tumors which have recurred after
and ENJOJI 1987; ALLES et al. 1994; LEWANDROWSKI et drainage procedures as invasive cystadenocarcinomas
al. 1993; RUBIN et al. 1994; SPERTI et al. 1997). (CAMPELL and CRUICKSHANK 1962; COMPAGNO and
Fine needle aspiration biopsy can be performed OERTEL 1978b; HODGKINSON et al. 1978; ZAMBONI
percutaneously with CT or US guidance or intraop- et al. 1994) (Fig. 2.16). The prognosis of a mucinous
eratively. The latter appears to be a valid alternative cystadenoma, borderline tumor, and mucinous cyst-
to the classic surgical biopsy that can cause com- adenocarcinoma without invasion, regardless of the
plications such as fistulas and protracted bleeding degree of cellular atypia, is excellent if the tumor can
(BLANDAMURA et al. 1995). Aspirates are charac- be completely removed (Fig. 2.17) (ZAMBONI et al.
terized by the presence of background mucin and 1999; COMPAGNO and OERTEL 1978b; CORBALLY et
columnar cells, with a clear and vacuolated cyto- al. 1989; DIDOLKAR et al. 1975; HODGKINSON et al.
plasm, arranged in sheets and in papillae (Fig. 2.15). 1978; WARSHAW et al. 1990). However, in a minor-
Significant cytological atypia is absent in many cases. ity of cases, recurrence and metastasis have been
The disordered arrangement of cells, with nuclear reported after a curative resection of a noninvasive
crowding and overlapping, the presence of single MCN (ALB ORES-SAAVEDRA et al. 1987; COMPAGNO
cells, large nuclei with distinct nucleoli, irregular and OERTEL 1978b; WARREN and HARDY 1968;
chromatin distribution, and marked variations in THOMPSON et al. 1999; WILENTZ et al. 1999). In con-
size and shape are the most important cytologic fea- nection with this problem, we would like to emphasize
tures of mucinous cystadenocarcinoma (LAUCIRICA the necessity of appropriate sampling for making the
et al. 1992; YOUNG et al. 1991; SCHWERK 1981). Fine correct diagnosis (ZAMBONI et al. 1999). The chance
needle aspiration biopsy is used to confirm the pres- of finding carcinomatous foci is greater in the grossly
ence of malignancy and prevent unnecessary surgery conspicuous areas than in flat and thin portions of the
in unresectable cases. cystic wall. It is worth noting that all cases of invasive
MCC usually show foci with MCB and MCA changes.

Fig. 2.16. Mucinous cystadenocarcinoma. A large cystic lesion,


Fig. 2.15. Mucinous cystic tumor. Fine needle aspiration treated 11 years previously with cystojejunostomy due to an
biopsy specimen showing moderately atypical aggregates of incorrect diagnosis of inflammatory pseudocyst, with malig-
cells in a mucinous background nant progression into invasive cystadenocarcinoma
20 G. Zamboni et a1.

a b

Fig. 2.17a,b. Mucinous cystadenocarcinoma. a Cut section of the tumor characterized by multiloculated cyst and'irregular solid
areas, with a thick, fibrous pseudo capsule. b Whole-mount section showing multilocular cystic spaces with multiple papillary
projections, but no invasion into the thick fibrous pseudocapsule

It is conceivable that limited tumor sampling might as site, tumor size, macroscopic appearance, grade of
be the reason why in these rare cases in which metas- differentiation, presence/absence/luteinization of the
tases were observed, the original resection specimen stroma, and p53 positivity have no prognostic signifi-
was thought to be noninvasive. cance (ZAMBONI et al. 1999).
The prognosis of invasive mucinous cystadeno-
carcinoma depends on the extent of tumor invasion.
Tumor recurrence and tumor-related death correlate
with tumor wall and peritumoral invasion (ZAMBONI 2.4
et al. 1999) (Fig. 2.18). Patients over than 50 years old Solid Pseudopapillary Tumor
have been shown to have a poorer survival rate.
Tumor ploidy and S-phase fraction may predict Solid pseudopapillary tumor (SPT) of the pancreas
survival in patients with MeN. Aneuploid neoplasms is a distinctive low-grade malignancy that primarily
with a high S-phase had a worse prognosis than occurs in girls and young women (KLOPPEL et al.1981;
patients with diploid tumors with a low S-phase ZAMBONI et al. 1993) and is composed of monomor-
(SOUTHERN etal. 1996; THOMPSON et al.1999; FLEJOU phous, small cells. It has been referred to as solid-cystic
et al. 1996; BRENIN et al. 1995). Other variables such tumor or papillary-cystic tumor. It has been reported
rarely in older women (LIEBER et al. 1987) and men
(KLOPPEL et al. 1991; LIEBER et al. 1987) and at extra-
pancreatic sites (KLOPPEL et al. 1991; ISHIKAWA et al.
1990; KIM et al. 1990). SPTs represent less than 1% of
all pancreatic tumors. In our series, they represented
11.5% of all cystic tumors; 15 occurred in women
(average age 31 years, range 14-69 years) and 1 in a
male adolescent (14 years old), with a predilection for
the head of the pancreas (12 cases).
Macroscopically, SPTs are frequently large, round,
well-circumscribed masses and show variable pro-
portions of solid and cystic areas filled with hem-
orrhagic fluid and necrotic debris (Fig. 2.19a,b). At
Fig. 2.18. Mucinous cystadenocarcinoma. Cut section of malig-
the extreme ends of the spectrum of macroscopic
nant cystadenocarcinoma showing gelatinous carcinomatous appearances, some cases can be exclusively solid
infiltration of the pseudocapsule (top, right) (usually the smaller lesions), whereas others (usu-
Pathology of Cystic Tumors 21

Fig. 2.20. Solid pseudopapillary tumor. Whole-mount macro-


section of a large, predominantly cystic lesion. The tumor shows
extensive hemorrhagic changes; the viable neoplastic compo-
nent is limited to a small semicircular area (middle, left)

posItive globules, stromal myxoid degeneration,


necrotic changes with foam cells, and hemorrhage
are characteristically present (Fig. 2.21b). Mitotic
figures are virtually absent, with a low proliferative
fraction (Ki-67 index <2%).
Clinically and radiologically, the differential diag-
nosis of endocrine tumors vs exclusively solid SPTs
that lack degenerative changes cannot be resolved.
Sometimes this may also be difficult during patho-
logical examination. Immunohistochemical studies
help to distinguish the two lesions. SPTs are positive
for neuron-specific enolase, CD56, CDI0, vimentin,
a-I-antitrypsin, and a-l-antichymotrypsin, whereas
the immunoreactivity for a broad spectrum of keratin
markers is usually negative (MOROHOSHI et al. 1989;
KU)PPEL et al. 1991; ZAMBONI et al. 1993; NOTOHARA
et al. 2000; KOSMAHL et al. 2000) (Fig. 2.22). Recently,
immunohistochemical positivity for p-catenin has
C L-_ _
been reported (Fig. 2.23) (TANAKA et al. 2001). SPTs
have not shown the molecular changes reported in
Fig. 2.19a-c. Solid pseudopapillary tumor. Cut sections of
predominantly solid (a), mixed solid and cystic (b), and pre- ductal adenocarcinomas, such as p53, Ki-ras, DPC-4,
dominantly cystic tumors (c) and p16 (KLIMSTRA et al. 2000).
A preoperative diagnosis may be obtained using
fine needle aspiration biopsy under ultrasound guid-
ally the larger tumors) may be entirely cystic, and the ance (ZAMBONI et al. 1993; PELOSI et al. 1995). The
lesion easily mistaken for a pseudocyst (Fig. 2.19c). cytological smears are highly cellular and character-
Some tumors may present a fibrous pseudo capsule ized by the presence of mildly atypical, monomor-
with focal calcifications (KU)PPEL et al. 2000). phous cells, with scanty eosinophilic cytoplasm and
Microscopically, the tumors are composed of round, oval nuclei with grooves and finely granular
a mixture of solid and cystic areas, usually sur- chromatin. Branching papillae, formed by delicate
rounded by a fibrous capsule (Fig. 2.20). The tumor central fibrovascular stalks covered by one or more
cells, forming solid areas or lining pseudopapillae, layers of tumor cells, are frequently observed together
are monomorphous, with round to oval nuclei and with irregular clusters, acinar structures, and individ-
eosinophilic, granular cytoplasm (Fig. 2.21a). PAS- ual dissociated cells (Fig. 2.24). Eosinophilic, hyaline
22 G. Zamboni et al.

a ......~........1000 b

Fig. 2.21a,b. Solid pseudopapillary tumor. a Pseudopapillary structures with fibrovascular cores lined by small monomorphic
cells. b Solid area showing foam cells and hemorrhage

Fig. 2.22. Solid pseudopapillary tumor. Exclusively solid tumor, without degenerative changes (H&E: second section from the left),
showing negative immunoreactivity for cytokeratins 8, 18, 19 (first section) and intense positivity for vimentin (third section)
and neuron-specific enolase (NSE) (fourth section)

globules and numerous foam cells may also be pres-


ent. The most important differential diagnosis is from
nonfunctioning islet cell tumors (IACONO et al. 1992),
pancreatoblastoma (CUBILLA and FITZGERALD 1984),
and acinar cell tumor (KUMSTRA et al. 1992).
Although the histogenesis remains obscure, the
immunohistochemical and ultrastructural findings,
demonstrating a prevalence of ductular cells with
minimal acinar and endocrine differentiation, favor
the hypothesis of a ductular cell origin with divergent
differentiation (MATSUNOU et al. 1990). The pathoge-
netic role of sex hormones is suggested by the predi-
lection of SPT for young, fertile women. Controversial
data have been provided for the presence of progester-
one receptors (PR) and/or estrogen receptors (ER) in
Fig. 2.23. Solid pseudopapillary tumor. Cytoplasmic and
nuclear ~-catenin immunostaining in the neoplastic cells
SPT (CARBONE et al. 1989; WRBA et al. 1988; KOSMAHL
(left); slight membranous positivity is present in the normal et al. 2000). We reported the immunohistochemical
pancreatic tissue (right) detection of PR in the large majority of neoplastic
Pathology of Cystic Tumors 23

2.5.1
Acinar Cell Cystadenoma

We recently described the detailed clinicopathologi-


cal features of ten patients with acinar cell cystad-
enoma (ACA), a previously unrecognized type of
cystic pancreatic lesion, which is characterized by
acinar cell differentiation of the lining cells, absence
of distinct cellular atypia, focal or diffuse involvement
of the pancreas, and a good prognosis (ZAMBONI et
al. 2002). In our ten patients, ACAs showed no clear
age predilection; the patients' age ranged from 16 to
66 years. There was, however, a female predilection:
seven women and three men, with the largest lesions
Fig. 2.24. Solid pseudopapillary tumor. Fine needle aspiration occurring in women. There was no clear preferential
biopsy showing branching papillae, formed by delicate, cen- localization within the pancreas. Though the largest
tral, fibrovascular stalks covered by monomorphic cells with
cysts had developed in the head of the pancreas, the
eosinophilic cytoplasm and round nuclei
remaining cysts also involved the body and tail of
the gland, and in one case, the entire pancreas was
cells in ten patients with SPT, whereas no ER-posi- affected. All patients remained alive and well during
tivity was found (ZAMBONI et al. 1993). This finding a follow-up period of up to 84 months.
supports the hypothesis of a possible pathogenetic Macroscopically, the lesions show a well-circum-
role of progesterone in SPT, regardless of the patient's scribed cystic pattern with a thin, fibrous pseudo capsule
age and sex. However, it is not possible to exclude that (Fig. 2.25), which in some cases can be thick and pres-
it simply represents an epiphenomenon, if one consid- ent with calcification. On cut section, they were either
ers that PR is also expressed in pancreatic endocrine unilocular (with a diameter between 0.5 and 10 em)
tumors (VIALE et al. 1992) as well as in normal islets or multilocular, with the largest diameter being 7.5 em.
(DOGLIONI et al. 1990). In all cases, the internal surface was smooth and shiny,
SPT has to be regarded as a carcinoma with with no solid areas or papillary projections. The cysts
low malignant potential and a favorable clinical contained serous fluid.
course, although invasion of vital structures and Microscopically, the cysts were lined by a single
metastases have been reported (CAPPELLARI et al. layer of cuboidal or columnar cells with little tendency
1990; MATsuNou et al. 1990; SCLAFANI et al. 1991; to pseudostratification or crowding (Fig. 2.26a). Their
NISHIHARA et al. 1993; KLIMSTRA et al. 2000). More cytoplasm was typical for acinar cells, with deeply
than 95% of patients with SPT limited to the pancreas eosinophilic granules in the apical part and basophilic
are cured by complete surgical resection (KLIMSTRA
et al. 2000). Only a few patients have died of metas-
tasizing tumor (COMPAGNO et al. 1979; MATSUNOU et
al. 1990; Ky et al. 1998).

2.5
Acinar Cell Cystic Tumors

Acinar cell carcinomas are tumors with evidence


of pancreatic enzyme production by the neoplastic
cells (SOLCIA et al. 1997). The vast majority are solid
tumors (KLIMSTRA et al. 1992). The cystic variant of
these tumors has only rarely been reported. We can
now consider two categories of lesions: acinar cell Fig. 2.25. Acinar cell cystadenoma. Intraoperative picture
cystadenocarcinoma and the newly reported acinar showing a large, multilocular cystic lesion in the body of the
cell cystadenoma. pancreas
24 G. Zamboni et al.

staining at the base of the cells. In addition, the apical all cases. All seven stained cases were positive for the
cytoplasm showed characteristic PAS-positivity which cytokeratin markers CAM 5.2 (cytokeratins 8, 18, 19),
was diastase-resistant. The nuclei were basally located, KLl (cytokeratins 1,2,5,6,7,8,11,14,16,17,18), and
uniform, and normochromic with small but evident cytokeratin 7. The proliferative fraction was very small
nucleoli; atypia was very mild or absent. Mitoses were in all cases, with a Ki-67 index <1%. No case showed
extremely rare. The cysts were usually connected with nuclear positivity for the p53 tumor suppressor gene.
small clusters of acinar cells forming acini that opened Although at present a nonneoplastic nature
into the cystic lumen. These acini were often dilated cannot be completely excluded, we refer to this tumor
and had the appearance of tubular complexes. Intra- as acinar cell cystadenoma (and suggest that it might
cystic club-like pseudopapillae with a fibrous stalk represent the benign counterpart of the well-recog-
were a distinctive feature (Fig. 2.26b). In some cases, nized acinar cell cystadenocarcinoma (CANTRELL
the cysts contained eosinophilic secretions. et al. 1981; ISHIZAKI et al. 1995; JOUBERT et al. 1998;
Immunohistochemically, the epithelial cells stained STAMM et al. 1987).
positively with at least one of the acinar markers:
trypsin (Fig. 2.27), chymotrypsin, and lipase. All but
one of the cases stained positively for pancreatic stone 2.5.2
protein (PSP). The eosinophilic secretions also stained Acinar Cell Cystadenocarcinoma
for trypsin and PSP. The neuroendocrine markers
chromogranin A and synaptophysin were negative in Rare examples of cystic acinar cell carcinoma have
been reported as 'acinar cell cystadenocarcinomas'
(CANTRELL et al. 1981; STAMM et al. 1987; JOUBERT
et al. 1998; ISHIZAKI et al. 1995). The patients are
frequently men, with a mean age of 50-60 years.
Macroscopically, these neoplasms are large masses
with diameters up to 35 cm containing multiple cysts,
with a diameter ranging from a few millimeters to
many centimeters. Hemorrhage and necrosis have
been reported (KLIMSTRA et al. 2000). Microscopi-
cally, the tumors are composed of multiple cysts of
varying sizes, mixed with tubular and solid areas. The
lining cells show the typical acinar differentiation, the
cytoplasm filled with deeply eosinophilic granules in
the apex and with basophilic staining at the base. The

.,

Fig. 2.26a,b. Acinar cell cystadenoma. Portion of cyst lined Fig. 2.27. Acinar cell cystadenoma. Immunohistochemical
by columnar cells with acinar differentiation. Note the small expression of the pancreatic enzyme trypsin in the lining
acini connected to the lining cells (a); cyst showing a collar of epithelium
dilated acinar structures opening into the lumen, with club-
like pseudopapillae (b)
Pathology of Cystic Tumors 25

cells composing cystic acinar cell carcinomas show columnar epithelium. Suppurative inflammation is
clear signs of atypia and many mitoses; necrosis is frequently found. They have no malignant potential.
frequently found.
The prognosis of reported cases was poor and
similar to that of the solid counterpart; metastases
were evident either at the time of diagnosis or within
a few months of surgery (KLIMSTRA et al. 2000). References

Abe H, Kubota K, Mori M, Miki K, Minagawa M, Noie T,


Kimura W, Makuuchi M (1998) Serous cystadenoma of the
pancreas with invasive growth: benign or malignant? Am J
2.6 GastroenteroI93:1963-1966
Other Cystic Tumors Adsay N, Klimstra D (2000) Cystic forms of typically solid
pancreatic tumors. Semin Diagn PathoI17:81-88
This category includes very rare lesions, which Adsay NV, Hasteh F, Cheng JD, Klimstra DS (2000a) Squa-
account for less than 5% of our cystic tumors. mous-lined cysts of the pancreas: lymphoepithelial cysts,
dermoid cysts (teratomas), and accessory-splenic epider-
Cystic endocrine tumor is an endocrine tumor char- moid cysts. Semin Diagn PathoI17:56-65
acterized by solid growth with massive degenerative Adsay NV, Klimstra DS, Compton CC (2000b) Cystic lesions of
changes, which may be confused with other cystic neo- the pancreas. Introduction. Semin Diagn PathoI17:1-6
plasms. The preoperative diagnosis may be solved with Akwari OE, Turker A, Seigler HF, Itani KMF (1990) Hepato-
fine needle aspiration biopsy showing the characteris- biliary cystadenoma with mesenchymal stroma. Ann Surg
211:18-27
tic cytological features (AOSAY and KLIMSTRA 2000). Albores-Saavedra J, Angeles-Angeles A, Nadji M, Henson DE,
The diagnosis may be confirmed by the immunohisto- Alvarez L (1987) Mucinous cystadenocarcinoma of the
chemical demonstration of endocrine markers such as pancreas. Morphologic and immunohistochemical obser-
chromogranin A and synaptophysin and the detection vations. Am J Surg Pathol 11: 11-20
of hormone production (IACONO et al. 1992). Albores-Saavedra J, Gould E, Angeles-Angeles A, Henson DE
(1990) Cystic tumors of the pancreas. Pathol Annu:19-50
Lymphoepithelial cyst is a benign, uncommon Alles AJ, Warshaw AL, Southern JF, Compton CC,Lewandrowski
lesion characterized by mature squamous epithe- KB (1994) Expression of CA 72-4 (TAG-72) in the fluid
lium associated with lymphoid tissue (AOSAY et al. contents of pancreatic cysts. A new marker to distinguish
2000a; IACONO et al. 1996). They are more common malignant pancreatic cystic tumors from benign neo-
in men, the mean age of the patients being 56 years plasms and pseudocysts. Ann Surg 219:131-134
Alpert LC, Truong LD, Bossart MI, Spjut HJ (1988) Microcystic
(AOSAY et al. 2000a). Macroscopically, they may
adenoma (serous cystadenoma) of the pancreas. A study of
occur predominantly as extrapancreatic lesions and 14 cases with immunohistochemical and electron-micro-
in any case are well demarcated from the surround- scopic correlation. Am J Surg PathoI12:251-263
ing pancreatic parenchyma. They can be either mul- Blandamura S, Costantin G, Nitti D, Boccato P (1995) Intra-
tilocular (60% of cases) or unilocular (40% of cases); operative cytology of pancreatic masses. A 1O-year experi-
ence. Acta Cytol 39:23-27
their mean diameter is 4.7 cm. The cyst wall is usually Bonetti F, Pea M, Martignoni G, Zamboni G (1992) PEC and
thin, and the inner surface of the cysts is smooth or sugar. Am J Surg PathoI16:307-308
finely granular. Microscopically, the cysts are lined Bonetti F, Pea M, Martignoni G, Doglioni C, Zamboni G,
by stratified squamous epithelium mixed with flat or Capelli P, et al (1994) Clear cell ('sugar') tumor of the lung
cuboidal epithelium. The cyst wall and the septa are is a lesion strictly related to angiomyolipoma-the concept
of a family of lesions characterised by the presence of the
filled with dense lymphoid tissue composed of CD3- perivascular epithelioid cells (PEC). Pathology 26:230-236
positive T-cells, frequently associated with germinal Bostwick DG, Tazelaar HD, Bailon SC, Endrickson MR, Kemp-
centers formed by B-cells. The differential diagnosis son RL (1986) Ovarian epithelial tumors of borderline
is essentially from serous cystadenoma and mature malignancy: a clinical and pathologic study of 109 cases.
Cancer 58:2052-2065
teratoma. They have no malignant potential.
Brenin D, Talamonti M, Yang E, Sener S, Haines G, Joehl R,
Mature teratoma is a benign, extragonadal germ- Nahrwold D (1995) Cystic neoplasms of the pancreas. A
cell tumor with mature tissues derived from all three clinicopathologic study, including DNA flow cytometry.
germ layers (AOSAY et al. 2000a; IACONO et al. 1993). Arch Surg 130: 1048-1054
They have no gender predominance; the mean age of Campell J, Cruickshank A (1962) Cystoadenoma and cystoad-
enocarcinoma of the pancreas. J Clin PathoI15:432-437
the patients is 29 years. Macroscopically, they have
Cantrell BB, Cubilla AL, Erlandson RA, Fortner J, Fitzgerald PJ
both solid and cystic areas, with frequent calcifica- (1981) Acinar cell cystadenocarcinoma of human pancreas.
tion. Microscopically, the cystic spaces are lined by Cancer 47:410-416
squamous epithelium, mixed with respiratory and Cappellari JO, Geisinger KR, Albertson DA, Wolfman NT,
26 G. Zamboni et al.

Kute TE (1990) Malignant papillary cystic tumor of the ment and extension visualized by three-dimensional com-
pancreas. Cancer 66:193-198 puterized mapping. Cancer 70:1505-1513
Carbone A, Ranelletti F, Rinelli A, Vecchio F, Lauriola L, George DH, Murphy F, Michalski R, Ulmer BG (1989) Serous
Piantelli M, Capelli A (1989) Type II estrogen receptors cystadenocarcinoma of the pancreas: a new entity? Am J
in the papillary cystic tumor of the pancreas. Am J Clin Surg Pathol13:61-66
Pathol 92:572-576 Gibson JB, Sobin LH (1978) Histological typing of tumor of
Carlson SK, Johnson CD, Brandt KR, Batts KP, Salomao DR the liver, biliary tract and pancreas, 1st edn. WHO Interna-
(1998) Pancreatic cystic neoplasms: the role and sensitiv- tional Histological Classification of Tumours, Geneva
ity of needle aspiration and biopsy. Abdom Imaging 23: Girelli R, Bassi C, Falconi M De Santis L, Bonora A, Caldiron E,
387-393 Sartori N, Salvia R, Briani G, Pederzoli P (1997) Pancreatic
Centeno B, Lewandrowski K, Warshaw A, Compton C, South- cystic manifestations in von Hippel-Lindau disease. Int J
ern J (1994) Cyst fluid cytologic analysis in the differential Pancreatol 22: 10 1-109
diagnosis of pancreatic cystic lesions. Am J Clin Pathol Healy DL, Burger HG, Mamers P, Jobling T, Bangh M, Quinn
101:483-487 M, Grant P, Day AJ, Rome R, Campbell JJ (1993) Elevated
Compagno J, Oertel J (1978a) Microcystic adenomas of the serum inhibin concentrations in postmenopausal women
pancreas (glycogen-rich cystadenomas): a clinicopatho- with ovarian tumors. N Engl J Med 329:1539-1542
logical study of 34 cases. Am J Clin Pathol 69:289-298 Hodgkinson DJ, ReMine WH, Weiland LH (1978) A clinico-
Compagno J, Oertel J (1978b) Mucinous cystic neoplasms of pathologic study of 45 cases of pancreatic cystadenocarci-
the pancreas with overt and latent malignancy (cystadeno- noma. Ann Surg 188:679-684
carcinoma and cystadenoma): a clinicopathologic study of Horton WA, Wong V, Eldridge R (1976) Von Hippel-Lindau
41 cases. Am J Clin Pathol 69:573-580 disease: clinical and pathological manifestations in nine
Compagno J, Oertel JE, Kremzar M (1979) Solid and papillary families with 50 affected members. Arch Intern Med l36:
epithelial neoplasm of the pancreas, probably of small duct 769-777
origin: a clinicopathologic study of 52 cases. Lab Invest 40: Howard JM (1989) Cystic neoplasms and true cysts of the
248-249 pancreas. Surg Clin North Am 69:651-665
Compton CC (2000) Serous cystic tumors of the pancreas. Iacono C, Serio G, Fugazzola C, Zamboni G, Bergamo Andreis
Semin Diagn PathoI17:43-55 lA, Iannucci A, Zicari M, Dagradi A (1992) Cystic islet cell
Corbally MT, McAnena OJ, Urmacher C, Herman B, Shiu MH tumor of the pancreas. A clinico-pathological report of
(1989) Pancreatic cystadenoma. A clinicopathologic study. two nonfunctionig cases and review of the literature. Int J
Arch Surg 124:1271-1274 Pancreatol 11: 199-208
Cubilla A, Fitzgerald P (1984) Tumors of the exocrine pan- Iacono C, Zamboni G, Di Marcello R, Zicari M, Maran M, Mon-
creas. AFIP Atlas of tumor pathology, 2nd ser, vol 19. tresor E, Nicoli N, Serio G (1993) Dermoid cyst of the head
Armed Forces Institute of Pathology, Washington DC of the pancreas area. Int J PancreatoI14:269-273
Czernobilsky B, She zen E, Lifshitz-Mercer B (1989) Alpha Iacono C, Cracco N, Zamboni G, Bernardello F, Zicari M,
smooth muscle actin (a-SM actin) in normal human Marino F, Montresor E, Serio G (1996) Lymphoepithelial
ovaries, in ovarian stromal hyperplasia and ovarian neo- cyst of the pancreas. Report of two cases and review of the
plasms. Virch Arch B Cell Pathol 57:55-61 literature. Int J PancreatoI19:71-76
Didolkar M, Malhotra Y, Holyoke E, Elias E (1975) Cyst- Ishikawa 0, Ishiguro S, Ohhigashi H, Sasaki Y, Yasuda T,Imaoka
adenoma of the pancreas. Surg Gynecol Obstet 140: S, Iwanaga T, Nakaizumi A, Fujita M, Wada A (1990) Solid
925-928 and papillary neoplasm arising from an ectopic pancreas
Doglioni C, Gambacorta M, Zamboni G, Coggi G, Viale G in the mesocolon. Am J Gastroenterol 85:597-601
(1990) Immunocytochemical localization of progesterone Ishikawa T, Nakao A, Nomoto S, Hosono J, Harada A, Nonami
receptors in endocrine cells of the human pancreas. Am J T, Takagi H (1998) Immunohistochemical and molecular
Pathol137:999-1005 biological studies of serous cystadenoma of the pancreas.
Egawa N, Maillet B, Schroder S, Mukai K, Kloppel G (1994) Pancreas 16:40-44
Serous oligocystic and ill-demarcated adenoma of the Ishizaki A, Koito K, Namieno T, Nagakawa T, Murashima Y,
pancreas: a variant of serous cystic adenoma. Virchows Suga T (1995) Acinar cell carcinoma of the pancreas: a rare
Arch 424: 13-17 case of an alpha-fetoprotein-producing cystic tumor. Eur J
Flejou JF, Boulange B, Bernades P, Belghiti J, Henin D (1996) RadioI21:58-60
p53 protein expression and DNA ploidy in cystic tumors of Itai Y, Ohhashi K, Nagai H, Murakami Y, Kokubo T, Makita
the pancreas. Pancreas 13:247-252 K, Ohtomo K (1986) Ductectatic mucinous cystadenoma
Flemming P, Grothe W, Maschek H, Petry KU, Wellmann A, and cystadenocarcinoma of the pancreas. Radiology 161:
Georgii A (1996) The site of inhibin production in ovarian 697-700
neoplasms. Histopathology 29:465-468 Joubert M, Fiche M, Hamy A, Heymann M, Sagan C, Valette P,
Friedman HD (1990) Nonmucinous, glycogen-poor cystad- Le BM (1998) Extension of an acinar cell pancreatic car-
enocarcinoma of the pancreas Arch Pathol Lab Med 114: cinoma with cystic changes invading the Wirsung canal.
888-891 Gastroenterol Clin Bioi 22:465-468
Fujii H, Kubo S, Hirohashi K, Kinoshita H, Yamamoto T, Kamei K, Funabiki T, Ochiai M, Amano H, Marugami Y, Kasahara
Wakasa K (1998) Serous cystadenoma of the pancreas M, Sakamoto T (1992) Some considerations on the biology of
with atypical cells. Case report. Int J Pancreatol 23: pancreatic serous cystadenoma. Int J Pancreatol 11 :97-104
165-169 Keel SB, Zukerberg L, Graeme-Cook F, Compton CC (1996) A
Furukawa T, Takahashi T, Kobari M, Matsuno S (1992) The pancreatic endocrine tumor arising within a serous cystad-
mucus-hypersecreting tumor of the pancreas. Develop- enoma of the pancreas. Am J Surg PathoI20:471-475
Pathology of Cystic Tumors 27

Kim YI, Kim ST, Lee GK, Choi BI (1990) Papillary cystic tumor (1984) Endometrial cyst of the pancreas. Gastroenterology
of the liver. A case report with ultrastructural observation. 86:1589-1591
Cancer 65:2740-2746 Matsunou H, Konishi F, Yamamichi N, Takayanagi N, Mukai
Klimstra DS, Heffes CS, Oertl J, Rosai J (1992) Acinar cell car- M (1990) Solid, infiltrating variety of papillary cystic neo-
cinoma of the pancreas. A clinicopathologic study of 28 plasm of the pancreas. Cancer 65:2747-2757
cases. Am J Surg PathoI16:815-837 Molberg K, Heffess C, Delgado R, Albores-Saavedra J (1998)
Klimstra DS, Wenig BM, Heffess CS (2000) Solid-pseudopapil- Undifferentiated carcinoma with osteoclast-like giant cells
lary tumor of the pancreas: a typically cystic carcinoma of of the pancreas and periampullary region. Cancer 82:
low malignant potential. Semin Diagn PathoI17:66-80 1279-1287
KlOppei G (2000) Pseudo cysts and other non-neoplastic cysts Montag A, Fossati N, Michelassi F (1990) Pancreatic microcys-
of the pancreas. Semin Diagn PathoI17:7-15 tic adenoma coexistent with pancreatic ductal carcinoma.
Kloppel G, Morohoshi T, John HD, Oehmichen W, Opitz K, Am J Surg PathoI14:352-355
Angelkort A, Lietz H, Riickert K (1981) Solid and cystic Moore PS, Zamboni G, Brighenti A, Lissandrini D, Antonello D,
acinar cell tumour of the pancreas. A tumour in young Capelli P, Rigaud G, Falconi M, Scarpa A (2001) Molecular
women with favourable prognosis. Virchows Arch 392: characterization of pancreatic serous micro cystic adeno-
171-183 mas: evidence for a tumor suppressor gene on chromo-
KlOppei G, Maurer R, Hofmann E, Luthold K, Oscarson J, some lOq. Am J PathoI158:317-321
Forsby N, Ihse I, Ljungberg 0, Heitz PU (1991) Solid- Morohoshi T, Held G, Kloppel G (1983) Exocrine pancreatic
cystic (papillary-cystic) tumours within and outside the tumours and their histological classification. A study based
pancreas in men: report of two cases. Virchows Arch (A) on 167 autopsy and 97 surgical cases. Histopathology 7:
418:179-183 645-661
Kloppel G, Solcia E, Longnecker DS, Capella C, Sobin LH (1996) Morohoshi T, Kanda M, Asanuma K, Kloppel G (1989) Intra-
Histological typing of tumours of the exocrine pancreas, ductal papillary neoplasms of the pancreas. A clinicopatho-
2nd edn. WHO International Histological Classification of logic study of six patients. Cancer 64: 1329-1335
Tumours. Springer, Berlin Heidelberg New York Nishihara K, Nagoshi M, Tsuneyoshi M, Yamaguchi K, Hayashi
KlOppel G, Hruban RH, Luttges J, Kern S, Klimstra D, Adler I (1993) Papillary cystic tumors of the pancreas. Assess-
G (2000) Solid-pseudopapillary neoplasm. In: Hamilton ment of their malignant potential. Cancer 71:82-92
SR, Aaltonen LA (eds) Pathology and genetics of tumours Nishimura H (1983) Atlas of human prenatal histology, 1st
of the digestive system. WHO Classification of Tumours. edn. Igaku-Shoin, Tokyo
IARC Press, Lyon, pp 246-248 Notohara K, Hamazaki S, Tsukayama C, Nakamoto S, Kawabata
Kosmahl M, Seada LS, Janig U, Harms D, Kloppel G (2000) K, Mizobuchi K, Sakamoto K, Okada S (2000) Solid-pseu-
Solid-pseudopapillary tumor of the pancreas: its origin dopapillary tumor of the pancreas: immunohistochemical
revisited. Virchows Arch 436:473-480 localization of neuroendocrine markers and CDlO. Am J
Ky A, Shilyansky J, Gerstle J, Taylor G, Filler RM, Grace N, Surg Pathol 24:1361-137l
Superina R (1998) Experience with papillary and solid Ohta T, Nagakawa T, !toh H, Fonseca L, Miyazaki I, Terada T
epithelial neoplasms of the pancreas in children. J Pediatr (1993) A case of serous cystadenoma of the pancreas with
Surg 33:42-44 focal malignant changes. Int J PancreatoI14:283-289
Laitio M, Lev R, Oriic D (1974) The developing human fetal Parienty RA, Ducellier R, Lubrano JM, Piccard JD, Pradel J,
pancreas: an ultrastructural and histochemical study Solarski N (1980) Cystadenomas of the pancreas: diagno-
with special reference to exocrine cells. J Anat 117: sis by computed tomography. J Comput Assist Tomogr 4:
619-634 364-367
Laucirica R, Schwartz M, Ramzy I (1992) Fine needle Pea M, Bonetti F, Zamboni G, Martignoni G, Fiore-Donati L,
aspiration of cystic epithelial neoplasms. Acta Cytol 36: Doglioni C (1991) Clear cell tumor and angiomyolipoma.
881-886 Am J Surg PathoI15:199-202
Lewandrowski KB, Warshaw AL, Compton CC (1992) Macro- Pea M, Martignoni G, Zamboni G, Bonetti F (1996) Perivascu-
cystic serous cystadenoma of the pancreas: a morphologic lar epithelioid cell. Am J Surg PathoI20:1149-1153
variant differing from micro cystic adenoma. Hum Pathol Pelosi G, Iannucci A, Zamboni G, Bresaola E, Iacono C, Serio
23:871-875 G (1995) Solid and cystic papillary neoplasm of the pan-
Lewandrowski KB, Southern JF, Pins MR, Compton CC, War- creas: a clinico-cytopathologic and immunocytochemical
shaw AL (1993) Cystic fluid analysis in the differential study of 5 new cases diagnosed by fine-needle aspiration
diagnosis of pancreatic cysts. A comparison of pseudo- cytology and a review of the literature. Diagn Cytopathol
cysts, serous cystadenomas, mucinous cystic neoplasms, 13:233--246
and mucinous cystadenoma. Ann Surg 217:41-47 Perez-Ordonez B, Naseem A, Lieberman PH, Klimstra DS
Lieber MR, Lack EE, Robert JR, Patterson K, Restrepo C, Solo- (1996) Solid serous adenoma of the pancreas. The solid
mon D, Chandra R, Triche TJ (1987) Solid and papillary variant of serous cystadenoma? Am J Surg Pathol 20:
epithelial neoplasm of the pancreas. An ultrastructural 1401-1405
and immunohistochemical study of six cases. Am J Surg Pilcher CS, Bradley EL III, Majmudar B (1982) Enterogenous
Patholll:185-193 cyst of the pancreas. Am J Gastroenterol 77:576-577
Luttges J, Feyerabend B, Buchelt T, Pacena M, Kloppel G (2002) Posen J (1981) Giant cell tumor of the pancreas of the osteo-
The mucin profile of noninvasive and invasive mucinous clastic type associated with a mucous secreting cystadeno-
cystic neoplasms of the pancreas. Am J Surg Pathol 26: carcinoma. Hum PathoI12:944-947
466-47l Rego J, Ruvira L, Garcia A, Freijanes P, Penaranda J, Soto J
Marchevsky AM, Zimmerman MJ, Aufses AH Jr, Weiss H (1991) Pancreatic mucinous cystadenocarcinoma with
28 G. Zamboni et al.

pseudosarcomatous mural nodules. A report of a case with Viale G, Doglioni C, Gambacorta M, Zamboni G, Coggi G,
immunohistochemical study. Cancer 67:494-498 Bordi C (1992) Progesterone recector immunoreactivity
ReMine SG, Frey D, Rossi RL, Munson JL, Braasch JW (1987) in pancreatic endocrine tumors. An immunohistochemi-
Cystic neoplasms of the pancreas. Arch Surg 122:443-446 cal study of 156 neuroendocrine tumors of the pancreas,
Rickaert F, Cremer M, Deviere 1, Tavares L, Lambilliotte of the gastrointestinal and respiratory tracts, and the skin.
JP, Schroder S, Wurbs D, Kloppel G (1991) Intraductal Cancer 70:2268-2277
mucin-hypersecreting neoplasms of the pancreas. A clini- Vortmeyer AO, Lubensky lA, Fogt F, Linehan WM, Khettry U,
copathologic study of eight patients. Gastroenterology 101: Zhuang Z (1997) Allelic deletion and mutation of the von
512-519 Hippel-Lindau (VHL) tumor suppressor gene in pancreatic
Rome RM, Fortune DW, Quinn MA, Brown JB (1981) Func- microcystic adenomas. Am J PathoI151:951-956
tioning ovarian tumors in postmenopausal women. Obstet Warren K, Hardy K (1968) Cystadenocarcinoma of the pan-
GynecoI57:705-7l0 creas. Surg Gynecol Obstet 127:734-736
Rubin D, Warshaw AL, Southern JF, Pins M, Compton CC, Warshaw A, Rutledge P (1987) Cystic tumors mistaken for
Lewandrowski KB (1994) Expression of CA 15.3 protein pancreatic pseudo cysts. Ann Surg 205:393-398
in the cyst contents distinguishes benign from malignant Warshaw AL, Compton CC, Lewandrowsk KB, Cardenosa G,
pancreatic mucinous cystic neoplasms. Surgery 115:52-55 Mueller PR (1990) Cystic tumors of the pancreas. New
Rutgers JL, Scully RE (1988) Ovarian mullerian mucinous clinical, radiologic, and pathologic observations in 67
papillary cystadenomas of borderline malignancy. A clini- patients. Ann Surg 4:432-445
copathologic analysis. Cancer 61 :340-348 Wenig BM, Albores-Saavedra J, Buetow PC, Heffes CS (1997)
Sachs JR, Deren JJ, Sohn M, Nusbaum M (1989) Mucinous Pancreatic mucinous cystic neoplasm with sarcomatous
cystadenoma: pitfalls of differential diagnosis. Am J Gas- stroma. Report of three cases. Am J Surg Pathol 21:
troenterol 84:811-816 70-80
Schwerk W (1981) Ultrasonically guided percutaneous punc- Westra WH, Sturm P, Drillenburg P, Choti MA, Klimstra DS,
ture and analysis of aspirated material of cystic pancreatic Albores-Saavedra J, Montag A, Offerhaus GJ, Hruban RH
lesions. Digestion 21:184-192 (1998) K-ras oncogene mutations in osteoclast-like giant
Sclafani LM, Reiter VE, Coit DG, Brennan MF (1991) The cell tumors of the pancreas and liver: genetic evidence to
malignant nature of papillary and cystic neoplasm of the support origin from the duct epithelium. Am J Surg Pathol
pancreas. Cancer 68:153-158 22:1247-1254
Scully RE (1979) Tumors of the ovary and maldevelopment Wheeler DA, Edmondson HA (1985) Cystadenoma with mes-
gonads. Atlas of tumour pathology, vol 16. Armed Forces enchymal stroma (CMS) in the liver and bile ducts A clini-
Institute of Pathology, Washington DC copathologic study of 17 cases, 4 with malignant changes.
Solcia E, Capella C, Kloppel G (1997) Tumors of the pancreas. Cancer 56:1434-1445
AFIP Atlas of tumor pathology, vol 20. Armed Forces Insti- Widmaier U, Mattfeldt T, Siech M, Beger HG (1996) Serous
tute of Pathology, Washington DC cystadenocarcinoma of the pancreas. Int J Pancreatol 20:
Southern 1, Warshaw A, Lewandrowski K (1996) DNA ploidy 135-139
analysis of mucinous cystic tumors of the pancreas. Corre- Wilentz RE, Albores-Saavedra J, Zahurak M, Talamini MA, Yeo
lation of aneuploidy with malignancy and poor prognosis. q, Cameron JL, Hruban RH (1999) Pathologic examina-
Cancer 77:58-62 tion accurately predicts prognosis in mucinous cystic neo-
Sperti C, Pasquali C, Pedrazzoli S, Guolo P, Liessi G (1997) plasms of the pancreas. Am J Surg Pathol 23: 1320-1327
Expression of mucin-like carcinoma-associated antigen in Wilentz RE, Albores-Saavedra J, Hruban RH (2000) Mucinous
the cyst fluid differentiates mucinous from nonmucinous cystic neoplasms of the pancreas. Semin Diagn Pathol 17:
pancreatic cysts. Am J Gastroenterol 92:672-675 31-42
Stamm B, Burger H, Hollinger A (1987) Acinar cell cystadeno- Wrba F, Chott A, Ludvik B, Schratter M, Sponas J, Reiner A,
carcinoma of the pancreas. Cancer 60:2542-2547 Schernthaner G, Krisch K (1988) Solid and cystic tumor of
Talamini MA, Pitt HA, Hruban RH, Boitnott JK, Coleman JA, the pancreas; a hormonal-dependent neoplasm? Histopa-
Cameron JL (1992) Spectrum of cystic tumors of the pan- thology 12:338-340
creas. Am J Surg 163:117-124 Yamada M, Kozuka S, Yamano K, Nakazawa S, Naitoh Y, Tsu-
Tanaka Y, Kato K, Notohara K, Hojo H, Ijiri R, Miyake T, kamoto Y (1991) Mucin-producing tumor of the pancreas.
Nagahara N, Sasaki F, Kitagawa N, Nakatani Y, Kobayashi Cancer 68:159-168
Y (2001) Frequent beta-catenin mutation and cytoplasmic/ Yamaguchi K, Enjoji M (1987) Cystic neoplasms of the pan-
nuclear accumulation in pancreatic solid -pseudopapillary creas. Gastroenterology 92:1934-1943
neoplasm. Cancer Res 61:8401-8404 Yang JM, Southern JF, Warshaw AL, Lewandrowski KB (1996)
Tenti P, Romagnoli S, Pellegata NS, Zappatore R, Giunta P, Proliferation tissue polypeptide antigen distinguishes
Ranzani GN, Carnevali L (1994) Primary retroperitoneal malignant mucinous cystadenocarcinomas from benign
mucinous cystadenocarcinomas: an immunohistochemical cystic tumors and pseudocysts. Am J Surg 17l:126-129;
and molecular study. Virchows Arch 424:53-57 discussion 129-130
Thompson LD, Becker RC, Przygodzki RM, Adair CF, Heffess Yoshimi N, Sugie S, Tanaka T, Aijin W, Bunai Y, Tatematsu A,
CS (1999) Mucinous cystic neoplasm (mucinous cystad- Tsuneyoshi 0, Mori H (1992) A rare case of serous cystad-
enocarcinoma of low-grade malignant potential) of the enocarcinoma of the pancreas. Cancer 69:2449-2453
pancreas: a clinicopathologic study of 130 cases. Am J Young NA, Villani MA, Khoury P,Naryshkin S (1991) Differen-
Surg PathoI23:1-16 tial diagnosis of cystic neoplasms of the pancreas by fine-
Tuchmann-Duplessis H (1968) Embryologie. Travaux pra- needle aspiration. Arch Pathol Lab Med 115:571-577
tiques et enseignement dirige. Masson, Paris Zamboni G, Bonetti F, Scarpa A, Pelosi G, Doglioni C, Iannucci
Pathology of Cystic Tumors 29

A, Castelli P, Balercia G, Aldovini D, Bellomi A, Iacono C, G, Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM,
Serio G, Mariuzzi GM (1993) Expression of progesterone Kleppe! G (1999) Mucinous cystic tumors of the pancreas:
receptors in solid-cystic tumour of the pancreas: a clinico- clinicopathological features, prognosis and relationship to
pathological and immunohistochemical study of ten cases. other mucinous tumors. Am J Surg Pathol 23:410-422
Virchows Arch A 423:425-431 Zamboni G, Kleppel G, Hruban RH, Longnecker DS, Adler
Zamboni G, Scarpa A, Franzin G, Capelli P, Bogina G, Iannucci G (2000) Mucinous cystic neoplasms of the pancreas. In:
A, Bassi C, Pederzoli P, Iacono C, Serio G, Procacci C, Mari- Hamilton SR, Aaltonen LA (eds) Pathology and genetics
uzzi GM (1994) Pathology of cystic tumors of the pancreas. of tumours of the digestive system. WHO Classification of
In: Pederzoli P, Bassi C, Cavallini G, Falconi M (eds) Facing Tumours. IARC Press, Lyon, pp 234-236
the pancreatic dilemma. Springer, Berlin Heidelberg New Zamboni G, Terris B, Scarpa A, Kosmahl M, Capelli P, Klimstra
York, pp 368-397 DS, Lam PW, Kleppe! G (2002) Acinar cell cystadenoma of
Zamboni G, Pea M, Martignoni G, Zancanaro C, Faccioli G, the pancreas: a new entity? Am J Surg Pathol 26:698-704
Gilioli E, Pederzoli P, Bonetti F (1996) Clear cell 'sugar' Zheng W, Sung q, Hanna I, DePetris G, Lambert-Messrlian
tumor of the pancreas A novel member of the family of G, Steinhoff M, Lauchlan SC (1997) Alpha and beta sub-
lesions characterised by the presence of perivascular epi- units of inhibin/activin as sex cord-stromal differentiation
thelioid cells. Am J Surg PathoI20:722-730 markers. Int J Gynecol PathoI16:263-271
Zamboni G, Bonetti F, Iannucci A (1998) Needle aspiration Zirinsky K, Abiri M, Baer JW (1984) Computed tomography
cytology of the pancreas. Pathologica 90:318-320 demonstration of pancreatic micro cystic adenoma. Am J
Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini Gastroenterol 79: 139-142
3 Serous Cystic Tumors
G. CARBOGNIN, M. TAPPARELLI, E. PETRELLA, A. FUINI, and C. PROCACCI

CONTENTS (GEORGE et al. 1989; SOYER et al. 1994; KLOPPEL et


al. 1996; WIDMAIER et al. 1996; ABE et al. 1998; ERI-
3.1 Introduction 31 GUCHI et al. 1998; Wu et al. 1999). Authors are still
3.2 Imaging 32 not in complete agreement about the therapeutic
3.2.1 Transabdominal US 32
3.2.2 Computed Tomography 33 management of SCT. Reports are divided in the sur-
3.2.2.1 General Principles of Technique 33 gical literature between those who advocate resec-
3.2.2.2 CT Findings 34 tion in all cases (PADOVANI et al. 1991; TALAMINI
3.2.3 Magnetic Resonance Imaging 41 et al. 1992; NAGEL et al. 2000; Box and DOUGLAS
3.2.3.1 Technique 41 2000; SHEENAN et al. 2000) and those who accept
3.2.3.2 MR Findings 42
3.2.4 Other Imaging Modalities 44 the imaging diagnosis of SCT, allowing the lesions
3.2.4.1 Plain Radiographs 44 to be followed (ISELIN et al. 1993; SARR et al. 2001).
3.2.4.2 Contrast Examination Conservative management is favored in patients with
of the Upper Gastrointestinal Tract 44 large masses who are asymptomatic; elderly patients
3.2.4.3 Angiography 44 or those who may be poor surgical candidates; lesions
3.2.4.4 Endoscopic Retrograde
Cholangiopancreatography 46 in the pancreatic head where the morbidity and mor-
3.2.4.5 Endoscopic US 49 tality associated with pancreaticoduodenectomy may
3.3 Differential Diagnosis 50 be higher than the presence of the untreated lesion;
References 53 and above all, those small tumors which occasionally
show up in otherwise completely healthy or young
patients (KIMURA and MAKUUCHI 1999). COMPAGNO
and OERTEL (l978a, b) originally observed the ana-
3.1 tomic-pathological appearance of SCT. Their studies
Introduction characterized the morphology of all cystic lesions
containing serous fluid as 'micro cystic'. This unique
The serous cystic tumor (SCT) of the pancreas is morphology narrows down the differential diagno-
most often incidentally detected when an imaging sis of pancreatic masses with this appearance. This
study is performed for clinical symptoms which may classification has been further expanded by the
or may not be directly related to the pancreas. Once identification of an oligo cystic type (KLOPPEL et al.
detected, accurate characterization of a pancreatic 1996; SOLCIA et al. 1997), resulting in a considerable
mass as an SCT is of primary importance since this increase in differential diagnosis problems.
tumor is benign, unlike all the other cystic tumors As stated above, these lesions are detected acciden-
of the pancreas. There are no more than 10 reports tally during ultrasound (US), computed tomography
of serous cystadenocarcinoma in the world literature (CT) or magnetic resonance (MR) imaging. The other
conventional radiological investigations (plain film
G. CARBOGNIN, MD; M. TAPPARELLI, MD; E. PETRELLA, MD; of the abdomen, barium study of the upper gastroin-
C. PROCACCI, MD testinal tract, angiography, etc.) have no application
Istituto di Radiologia, Dipartimento di Scienze Morfologico- in the study of this lesion. Endoscopic ultrasound
Biomediche, Universita degli Studi di Verona, Policlinico
(EUS) or endoscopic retrograde cholangiopancrea-
'Giambattista Rossi', P.zza L.A. Scuro 10,37134 Verona, Italy
A. FUINI,MD tography (ERCP) may be utilized in certain cases
Servizio di Gastroenterologia, Ospedale Civile Maggiore, P.le when the previously mentioned imaging methods do
Stefani, 1,37124 Verona, Italy not suggest a conclusive diagnosis.
32 G. Carbognin et al.

3.2 ticular features (obesity, meteorism). The successful


Imaging identification of seT with us is solely based on the
unique morphological characteristics of the tumor.
3.2.1 The microcystic type SeT can be easily classified
Transabdominal US when the us investigations show a mass which usu-
ally has a multilobulated border, no posterior acoustic
US investigations have significantly contributed to enhancement, and an internal 'honeycomb' architec-
the increase in the number of seT observations ture (Fig. 3.1a) due to the presence of multiple septa
due to its widespread use in clinical practice. Only which delimit small «2 cm diameter) cystic spaces
in a minority of cases do us investigations identify (WOLFMAN et al. 1982; VILGRAIN et al. 1987; JOHNSON
the lesion following a request to clarify the cause of etal.1988;BuCK and HAYES 1990;PADOVANI etal.1991;
symptoms, even if they are nonspecific (epigastric YANG et al. 1994; FUGAZZOLA et al. 1991; PROCACCI et
pain, vomiting, weight loss, jaundice), supported by al. 1997, 2001a,b). More frequently, the multiplicity of
a mass of conspicuous size. Identification usually the interfaces results in a predominantly echogenic
occurs completely by chance, especially where small mass in which cystic spaces (Fig. 3.1b) can be recog-
tumors are concerned (PROCACCI et al.I997,2001a,b; nized (BUCK and HAYES 1990; PADOVANI et al. 1991).
MATHIEU et al. 1989). The false-negative rate of us Often, together with the micro cysts, larger cystic
in identifying seT is low; recognition of a small seT spaces (>2 cm diameter) can be found at the periphery
can be missed either because its site is more difficult of the lesion, resulting in a mixed pattern (Fig. 3.1c),
to explore (tail) and/or because of the patient's par- with a micro cystic or mainly solid, central portion and

a b

c d

Fig. 3.1a-d. Microcystic serous cystic tumor (SCT) (ultrasound, US evaluation). a Multilobulated mass with multiple internal
cystic spaces, delimitated by septations, giving the tumor a 'honeycomb' appearance. b Mass with solid appearance (calipers)
with a mainly echogenic texture; minute transonic cystic spaces are recognizable within. c Mixed appearance tumor (micro-
cystic and macrocystic): large cysts (>2 em in diameter: asterisk) are distinguishable at the periphery of the tumor. d Large
echogenic mass, with solid appearance. Calcifications appear like hyperechoic spots with posterior shadowing (arrows). L, liver;
ICV, inferior caval vein; A, aorta; P, pancreas; K, kidney
Serous Cystic Tumors 33

peripheral macro cysts (PROCACCI et al. 1997,2001b). Second, in the case of a mixed tumor (a micro cystic
The macrocysts can grow as large as 8-10 cm in dia- center and peripheral macrocysts), the macro cystic
meter, making it difficult to recognize the true nature component can conceal the micro cysts with the con-
of the tumor. Sometimes, in more 10%-30% of cases sequent misdiagnosis of a unilocular or multilocular
(PADOVANI et al. 1991; TORRESAN et al. 1997; COMP- macro cystic mass.
TON 2000; PROCACCI et al. 2001b), there can be hyper- The macrocystic type SCT, whether unilocular or
echogenic spots with posterior shadowing (Fig. 3.1d), multilocular (Fig. 3.3), is easily detectable even when
due to the presence of calcifications within the septa. its dimensions are small. It appears as a sharply
Less frequently, when there is a calcified scar, large marginated, hypoechoic mass with the transmission
hyperechogenic spots can be found in the center. SCT characteristics of a cyst. There may be sparse, thin,
is more frequently located in the pancreatic head, and central septa (MORI et al. 1995; GOUHIRI et al. 1999;
the gland is usually normal upstream. In extreme PROCACCI et al. 1997, 2001a,b; KANETO et al. 2000).
cases, dilation of either the duct ofWirsung or the bili- In this case, the possibility of a differential diagnosis
ary tree can be found due to obstruction by the mass. from the other cystic masses of the pancreas is lower
The micro cystic appearance is also seen in the (GOUHIRI et al. 1999; PROCACCI et al. 1997,2001a,b).
serous cystadenoma which accompanies von Hippel-
Lindau disease. In this case the tumor is multicentric
or diffusely involves and enlarges the pancreas. Typical 3.2.2
of SCT, these masses also display a multilobulated out- Computed Tomography
line and an inhomogeneous echotexture secondary to
the coexistence of cystic and solid areas, and frequent The most reliable method of detecting and char-
hyperechoic spots due to the calcifications (CHOYKE acterizing SCT is intravenous contrast-enhanced
et al. 1995; HES and FELDBERG 1999). In patients with computed tomography (CT). Characterization
von Hippel-Lindau disease, it is necessary to perform will probably be further improved as experience
renal US to exclude clear-cell carcinoma. with multidetector-row CT (MDCT) accumulates
There are two reasons why US may fail to charac- (FISHMAN et al. 2000; NINO-MuRCIA et al. 2001).
terize a pancreatic mass accurately as an SCT. First,
in the presence of a 'sponge-like' mass, the tumor can 3.2.2.1
manifest a homogenously echogenic aspect (Fig. 3.2) General Principles of Technique
with sharp borders, just like a solid tumor (JOHNSON
et al. 1988; BUCK and HAYES 1990; PADOVANI et al. As stated above, most SCT are detected accidentally
1991; PROCACCI et al. 1997, 2001a,b), because the during CT examinations for other or nonspecific
multiplicity of the small cysts and thick fibrous diagnoses. In most cases, proper characterization of
stroma produce innumerable acoustic interfaces the mass will require an additional dedicated pan-
yielding the false impression that the lesion is solid. creatic study. Regardless of the type of scanner one

a b

Fig. 3.2a,b. Microcystic SCT (US evaluation). Two cases. In the oblique subcostal (a) and axial (b) scans, a sharply demarcated,
uniformly hyperechoic mass, mimicking a solid tumor, can be seen. In the first case (a) the mass is compressing the biliary duct
( CBD) which appears dilated upstream. L, liver; K, kidney
34 G. Carbognin et al.

a b

Fig. 3.3a,b. Oligo cystic SCT (US evaluation). Two cases. a Ovoid, transonic, unilocular mass occupying the body of the pancreas.
Note relation to the aorta (A) and the inferior vena cava (ICV). b Large, transonic, multilocular mass in the pancreatic head
with lobulated contours and thick septum (arrow)

uses, the imaging protocol must ensure the following: 3.2.2.1.2


distension of the stomach and the duodenum with Multidetector-row CT
oral contrast or, preferably, with water; appropri-
ately delivered intravenous (IV) contrast medium, Multidetector-row CT substantially increases the
administered by power injector (automatic), so that diagnostic information available from the CT study.
scanning can occur during a phase in which there MDCT offers significantly increased diagnostic
is maximal pancreatic parenchymal enhancement; information from a single CT examination. This
and finally, acquisition with collimation of less than includes the ability to create an arterial and venous
3mm. map of the pancreatic area (FISHMAN et al. 2000) and
the ability to view the data in high quality 3D, improv-
3.2.2.1.1 ing our understanding of the relation of the mass to
Single Detector CT surrounding structures (NINO-MuRCIA et al. 2001).
Pancreatic imaging with MDCT is performed in a
Single detector CT examination starts with a pre- similar fashion to SCT; the major difference is that one
contrast (unenhanced) acquisition, indispensable must choose both an acquisition collimation and a dis-
for detecting calcifications or hemorrhage within play collimation. Two acquisitions are utilized, the first
the tumor (ITAI et al. 1988; SaYER et al. 1994; (pancreatic phase) is acquired 34-40 s following the IV
COMPTON 2000; PROCACCI et al. 1997, 2001a,b). Fol- injection oflow osmolality contrast medium at a rate of
lowing this, 120-150 ml of iodinated contrast agent not less than 3 mlls. The scan range is prescribed in an
is administered at a minimal rate of 3 mlls. A second identical fashion to that in Section 3.2.2.1.1. We utilize
acquisition is then initially performed after 30-35 s a 4x 1 mm detector row configuration and create 3-mm
delay (the pancreatic phase), and a third one after slices for image analysis. Additionally, we retain the
70-80 s delay (venous phase). In the pancreatic phase volume of slices and use them on the workstation for
acquisition, a slice thickness of 3 mm coupled with 3D evaluation. The second acquisition (venous phase)
a reconstruction interval of 2 mm is used, and the uses a 4x2.5 mm detector configuration. This scan is
images are acquired from the porta hepatis through prescribed from the top of the diaphragm through the
the transverse duodenum. In the venous phase, a iliac crests. This scan begins approximately 70-80 s fol-
slice thickness of 5-7 mm is used coupled with a lowing the initiation of the injection. From this acquisi-
reconstruction interval of 4-6 mm at a pitch of 1.5 tion, 5- to 7-mm slices can be created for viewing.
from the diaphragm to the lower pole of the kidneys.
In approximately 10%-15% of cases, after 120 s, a 3.2.2.2
further acquisition, limited to the pancreatic area, is CTFindings
performed with the aim of better demonstrating the
enhancement of the internal septations (PROCACCI The CT appearance of SCT depends upon two factors:
et al. 1999, 2001b). the macroscopic features (microcystic or macro cystic
Serous Cystic Tumors 35

structure) of the mass and the time of data acquisition et al. 1994; LE BORGNE et al. 1999; PROCACCl et al. 1999;
as it relates to the contrast injection (unenhanced, pan- CURRY et al. 2000). These calcifications are punctate or
creatic or venous) during which the mass is examined. globular (Figs. 3.7a, 3.8a), as opposed to the lamellar
calcifications seen in the mucinous cystic tumors. It is
3.2.2.2.1 sometimes possible to find a large calcification with
Microcystic SeT a rounded or 'star-shape' morphology, situated in the
central scar(s) (Fig. 3.9a). Usually, the scar is visible in
On unenhanced images, the tumor displays mass effect the larger masses since it forms later on (HEALY et al.
or a simple deformation of the contour of the gland. 1994). The presence of central calcifications in corre-
The lesion has multilobulated borders and a homoge- spondence with septa or scars is an important finding
neous density, slightly superior to that of water, and as it definitively characterizes the mass as SCT (WAR-
appears isodense or slightly hypodense compared SHAW et al. 1990; SOYER et al. 1994; CURRY et al. 2000).
with the pancreatic parenchyma (Figs. 3.4a, 3.Sa, 3.6a). Unfortunately, this pattern of calcification is seen in, at
Sometimes, by reducing the width of the image most, 30% of cases (WARSHAW et al. 1990; COMPTON
window, it is possible to demonstrate the septum and/ 2000). The presence of macro cysts at the periphery
or the central fibrous scar (Fig. 3.5c). When calcification are usually not recognized on the precontrast images
is present, it is almost always central (Figs. 3.7a, 3.8a) (Fig.3.6a).
(ITAl et al. 1982,1988; WOLFMAN et al. 1982; VlLGRAlN Maximal visualization of the septa and therefore the
et al. 1987; JOHNSON et al. 1988; SOYER et al. 1994; YANG greatest opportunity to correctly characterize the mass

a b

c d

Fig. 3.4a-d. Microcystic SCT (CT evaluation). a Non-contrast: bulky, lobulated mass with homogeneous density, slightly superior to
that of water. b Pancreatic phase: 'honeycomb' appearance of the mass due to the presence of multiple enhancing septa. c, d Portal
venous and late phases: the septa appear isodense compared to the gland; the 'honeycomb' aspect is, however, clearly recognizable
36 G. Carbognin et al.

a b

c d

Fig. 3.5a-d. Microcystic SCT (CT evaluation). a, b First case. Non-contrast phase (a), the sharply bordered lesion has homo-
geneous density. In the pancreatic phase (b), the radial orientation of hyperdense septa is recognizable, originating from the
central fibrous scar to the margins of the neoplasm. c, d Second case. On the un enhanced images viewed with a narrow window
(c), it is possible to demonstrate the septa and the central fibrous scar. In the contrast-enhanced pancreatic phase (d), the septa
coming from the central scar can be seen more clearly

a b

Fig. 3.6a,b. Microcystic SCT (CT evaluation). a On non-contrast enhanced images, the tumor has a homogeneous density, and
the peripheral macro cysts are not distinguishable from the micro cystic nucleus. b In the pancreatic phase, the peripheral mac-
rocysts and the micro cystic nucleus are readily demonstrated
Serous Cystic Tumors 37

c d

Fig. 3.7a-d. Microcystic SCT (CT evaluation). a On non-contrast enhanced images, the mass appears homogeneously attenuat-
ing. Small calcifications are scattered around the peripheral portions (but not the wall) of the tumor. b In the pancreatic phase,
hyperdense septa create a sponge-like pattern where an inhomogeneous solid center and multiple peripheral cysts can be
identified. c In the portal venous phase, the mass maintains its central solid appearance with peripheral cysts. d In the delayed
phase, the marked enhancement of the large central fibrous scar is highlighted

occurs during the pancreatic phase (see 3.2.2.1.112). to the precontrast or unenhanced images. When thin
The 'honeycomb' appearance so characteristic of this «5 mm) slices are obtained in the pancreatic phase,
mass is best seen during this phase (Fig. 3.4b). In larger it is possible to evaluate the tumor's relationships to
tumors it is possible to demonstrate the radial orien- adjacent structures more accurately. The SCT, depend-
tation of the septa (Figs. 3.5b, 3.9b) emanating from ing on its size, can compress surrounding viscera. It is
the central scar and extending towards the margins also possible to document the mass effect on adjacent
of the lesion (JOHNSON et al. 1988; ITAI et al. 1988, vessels. It is quite unusual to find occlusion or throm-
PADOVANI et al. 1991; SOYER et al. 1994; PROCACCI et bosis which, in association with the collateral circula-
al. 1997; LUNDSTEDT and DAWISKIBA 2000; ZIRINSKY tion, is more common in pseudo cysts or malignant
et al. 1984). The mass may appear as inhomogeneously cystic tumors. In extreme cases, it is possible to find
hyperdense due to the combination of the vascular- biliary obstruction in the presence of a large mass in
ized septa and the interspersed micro cysts (Fig. 3.7b). the pancreatic head. However, even large SCT rarely
Peripheral macrocysts related to the central micro- involve the duct ofWirsung. When this happens, how-
cystic nucleus may have diameters that vary from 2 ever rarely, the differential diagnosis may be difficult
to 10 em (Figs. 3.6b, 3.7b, 3.8b) and are even more as far as intraductal tumors of the collateral ducts are
easily recognizable (PROCACCI et al. 1997). In this concerned (Fig. 3.1O).
pancreatic phase, the intense enhancement of the In the venous phase, the tumor appears isodense
septa can obscure calcifications, thereby adding value compared to the gland; the 'honeycomb' aspect is
38 G. Carbognin et al.

a b

c d

Fig. 3.8a-d. Microcystic SCT (CT evaluation). a Non-contrast enhanced images reveal a multilobulated mass with large calcifica-
tions. b In the pancreatic phase, the mixed aspect of the tumor is recognizable with a large micro cystic area and a peripheral
macrocyst. c, d In the venous (c) and late (d) phases, no significant variations are evident

a b

Fig. 3.9a,b. Microcystic SCT (CT evaluation). a On noncontrast enhanced image, the mass appears homogeneous with a slightly
higher density to that of water and with a large central calcification. b In the pancreatic phase, septa radiating from the central
scar are recognizable
Serous Cystic Tumors 39

..

t~~
, ". . )~.,
c d

Fig. 3.lOa-d. Microcystic SCT (CT evaluation). a-c Axial scans, obtained in the craniocaudal direction during the pancreatic
phase, show dilation of the duct ofWirsung, stopping at the uncinate process. A small lesion (c) is seen, which has a micro cystic
architecture and a small stellate central calcification. d MRCP confirms the dilation of the duct of Wirsung and the micro cystic
lesion in the pancreatic head

easily visualized (Figs. 3.4c, 3.5d, 3.7c, 3.8c), and any presence of multiple cystic formations of varying
calcifications are identifiable due to the lower density sizes. Some of these contain calcification and infre-
of the septa. In this phase and in later acquisitions, quently may be solid (CHOYKE et al. 1995; PROCACCI
when the tumor is sponge-like, the cystic component et al. 1997, 2001a; HES and FELDBERG 1999). Other
may be difficult to recognize (Fig. 3.11) (ITAI et al. organs, especially the kidneys, are more commonly
1988; BUCK and HAYES 1990; PROCACCI et al. 1997; involved. The diagnosis is made easier by the diffuse
COMPTON 2000). The diagnosis is, however, easier if involvement of the gland, the coexistence of lesions
larger cysts are present at the periphery (Fig. 3.7c). in other organs and, above all, by familial history
The study of the tumor during a delayed phase (Fig. 3.12).
related to the contrast injection (Figs. 3.4d, 3.7d, SCT has been reported to coexist with other
3.8d) could simulate a unilocular macro cystic tumor tumors. This rare occurrence has been described
because of the nonrecognition of septa, indistin- between the micro cystic form of SCT and the endo-
guishable from the liquid content of the cyst (BUCK crine tumor (HERESBACH et al. 1993; KEEL et al. 1996;
and HAYES 1990; PROCACCI et al. 1997). TUNG et al. 2001). For this reason, the structure of the
Serous cystadenomas seen in patients with von lesion should be accurately evaluated, so as to exclude
Hippel-Lindau disease have a unique appearance. the presence of solid components, which would indi-
The pancreas appears enlarged due to the variable cate an associated tumor.
40 G. Carbognin et al.

a b

Fig. 3.11a,b. Microcystic SCT (CT evaluation). a, b Axial scans, obtained in the craniocaudal direction in the portal venous phase,
show a neoplasm with homogeneously solid density, within the limits of the pancreatic parenchyma. The lack of pancreatic
phase images disguises the presence of septa and leads to misdiagnosis

a b

C L-_""";'--_ d

Fig. 3.12a-d. Von Hippel-Lindau disease (CT evaluation). a, b First case. In the venous phase there is a diffuse enlargement of
the entire pancreas. The parenchyma has been replaced by multiple, water-density masses of variable dimensions. The larg-
est (asterisk in b) is in the head. c, d Second case (brother of the previous patient). In the portal venous phase, the gland has
increased in size and contains multiple cystic formations of uniform dimensions. There are also liquid and solid lesions in the
kidneys, the largest being in the right kidney
Serous Cystic Tumors 41

3.2.2.2.2 3.2.3
Oligo cystic SeT Magnetic Resonance Imaging

Oligo cystic SCT, as already highlighted by many MR is assuming an increasingly important role in
authors (LEWANDROWSKl et al. 1992; MORl et al. 1995; characterizing cystic tumors, particularly SCT, since
PROCACCl et al. 1997, 2001a,b; GOUHlRl et al. 1999; the multiplicity of the sequences gives greater and
KANETO et al. 2000; COMPTON 2000),is indistinguish- more accurate information about the structure of
able from other macro cystic masses of the pancreas, the lesion, most particularly the presence of septa.
primarily the mucinous cystic tumor. On CT, one Furthermore, the use of magnetic resonance chol-
detects the presence of a unilocular or multilocu- angiopancreatography (MRCP) gives a better evalu-
lar cystic mass, usually with thin walls and deeply ation of the relationship of the cystic mass to the
embedded 1n the pancreatic gland (PROCACCl et al. main pancreatic duct, which can be decisive in the
1997,1999, 2001a; GOUHIRl et al. 1999). Calcifications differential diagnosis between SCT and intraductal
are not frequent as in the micro cystic variety. In the papillary mucinous tumor (IPMT) of the collateral
pancreatic or venous phases of contrast enhance- ducts.
ment, both the walls and the scant, thin central
septations, when present, are highlighted (Fig. 3.13). 3.2.3.1
When immediate surgical treatment is not con- Technique
sidered appropriate, it is advisable to proceed
with needle aspiration under US or CT guidance Multiple MR sequences can provide a comprehensive
(LEWANDROWSKl et al. 1993; CARLSON et al. 1998; evaluation of the pancreatic mass, its internal struc-
HAMMEL 2000; SPERTl et al. 2000). ture and enhancement characteristics. Most studies

a b

c d

Fig. 3.13a-d. Oligocystic SCT (CT evaluation). a, b First case. The axial scans obtained in the craniocaudal direction during the
pancreatic phase show a unilocular cystic mass with multilobulated borders. c, d Second case. At CT (c) in the portal venous phase,
there is a large cystic lesion of the pancreatic head with a central scar and radially oriented thin septa. Contrast injection into the
duct ofWirsung of the resected specimen (splenopancreatectomy (d) shows that the large cyst of the pancreatic head does not com-
municate with the main duct, which appears merely displaced, while the collateral ducts are displaced in a basket-like fashion
42 G. Carbognin et al.

require several sequences from which this informa- 3.2.3.2


tion can be extracted. MRFindings
Initially, T2-weighted fast or turbo-spin-echo
(TSE) images are obtained, which in suspected 3.2.3.2.1
micro cystic SCT are immediately followed by Microcystic SeT
MRCP. The information obtainable from these ini-
tial sequences is often enough to make a conclusive With GRE Tl-weighted images the lesion is easily
diagnosis. If necessary, contrast-enhanced imaging demonstrated and appears typically hypointense
utilizing intravenous gadolinium-DTPA (Gd-DTPA) compared with the adjacent parenchyma (Figs. 3.14a,
is carried out. In this case, whenever possible, it is 3.15a). The mass displays a homogeneous signal
better to use spoiled gradient refocused echo (GRE) since the septa and calcifications are not identifi-
3D sequences with fat suppression. First, precontrast able (PROCACCI et al. 1997, 2001a). Extremely rarely,
images are acquired both in-phase and opposed- hyperintense hemorrhagic foci within the mass may
phase. Then acquisitions in the pancreatic, venous be detected (MERGO et al. 1997).
and late phases are obtained after the rapid bolus IV The T2-weighted TSE images are almost always
injection of Gd-DTPA (MERGO et al. 1997; PROCACCI decisive in diagnosing SCT since the contrast reso-
et al. 1997; KANEMATSU et al. 2000). The advantage of lution afforded by MR can easily demonstrate the
3D sequences is to offer, for each acquisition, the pos- small amounts of fluid interspersed within the
sibility of analyzing the enhancement of the paren- dense septa of the 'sponge-like' mass or, conversely,
chyma and, at the same time, of obtaining the lesion's will show the thin septa within the fluid of more
vascular map with postprocessing, which aids in typical adenomas (Figs. 3.14b, 3.15b, 3.16a,b, 3.17a,b)
surgical planning for the larger masses. (NISHIHARA et al. 1996; MERGO et al. 1997; PROCACCI

c d

Fig. 3.14a-d. Microcystic SCT (MR evaluation). a Tl W SE sequence: the lesion appears homogeneously hypointense towards the
surrounding parenchyma. Septa and calcifications are not recognizable. b T2W SE sequence: the mass is hyperintense because
of the liquid content while the septa appear hypointense. c, d In the pancreatic (c) and venous (d) phases, there is enhancement
of the walls and the septa, giving the mass its 'honeycomb' aspect
a b

Fig. 3.1Sa-d. Microcystic SCT (MR evaluation; same patient as in Fig. 3.8). a Opposed phase, unenhanced TI W GRE sequence:
a homogeneously hypointense mass is located in the body of the pancreas. b T2W TSE sequence: the mass has a microcystic
architecture with septa radiating from the large central scar. A peripheral macrocyst, characterized by a higher signal than the
micro cysts, is evident. c, d Gd-DTPA-enhanced fat-suppressed TIW GRE sequence: in the pancreatic (c) and venous (d) phases
there is a progressive enhancement of the walls and septa, resulting in the typically 'honeycomb' appearance of the mass. The
peripheral macrocyst is easily identified. The large central calcification is not seen (see Fig. 3.8)

et al. 1997, 2001a,b). MR has a disadvantage in that it tion duct between the cystic mass and the main duct
is insensitive to calcification. In the case of SCT with a (Fig. 3.17c,d). The normality of the duct of Wirsung
mixed micro cystic and macro cystic architecture, the and the lack of a communication duct can exclude the
homogeneously hyperintense peripheral macro cysts presence of IPMT of the collateral ducts and allow for
and the central nucleus of microcysts are perfectly a certain diagnosis of SCT (YAMAGUCHI et al. 1998;
recognizable (Figs. 3.15b, 3.17a,b). In this event, the DUPAS and LE BORGNE 2000).
signal intensity of the peripheral macrocysts will be In the contrast-enhanced phase, the MR findings
higher than that of the central micro cystic nucleus are essentially equivalent to CT. The mass will be
(PROCACCI et al. 1997). In the T2-weighted image hypo intense in the pre contrast phase; enhancement
sequence, significant replacement of the pancreatic of both the septa and the walls is seen in the postcon-
gland is immediately demonstrated in von Hippel- trast pancreatic phase, at which time the 'honeycomb'
Lindau disease, particularly the multiple cystic areas or 'sponge-like' patterns become evident (Figs. 3.14c,
that replace most of the parenchyma (Fig. 3.18). 3.15c) (MERGO et al. 1997; PROCACCI et al. 2001a,b).
MRCP must be carried out routinely in the study In the venous phase the 'honeycomb' appearance is
of these tumors (Figs. 3.lOd, 3.16c, 3.17c,d) since it even more easily recognizable (Figs. 3.14d, 3.15d,
can contribute to distinguishing micro cystic SCT 3.19), while the mass with the 'sponge-like' aspect can
from intraductal tumors of the collateral ducts, sometimes appear solid which, theoretically, could
which often have a septate appearance as well. MRCP lead to the misdiagnosis of a solid tumor. However,
images should be obtained in the coronal plane, with the T2-weighted image sequences this mistake
according to multiple rotation angles, as well as the need not be made. Unfortunately, it is not possible to
axial plane to best detect dilatation of the duct of find calcifications in any of these phases. The identifi-
Wirsung or the presence or absence of a communica- cation of possible peripheral macro cysts is, however,
44 G. Carbognin et al.

Fig. 3.16a-d. Microcystic SCT (MR evaluation). T2W half-Fourier single-shot turbo-spin-echo (HASTE) sequence obtained in the axial
(a) and coronal (b) planes: a small, multilobulated lesion can be seen with a micro cystic aspect and a small, hypointense, central scar.
e MRCP highlights the normal duct of Wirsung overlaid by the small mass, making it impossible to establish the presence or absence
of communication between the two structures. d Only ERCP is able to exclude the communication between the main duct and the
cystic mass, allowing for a definite diagnosis of microcystic SCT. Only a small imprint (arrow) is recognizable on the ductal wall

easy (Fig. 3.1Sd). If a GRE 3D sequence is performed ment of gas-filled hollow viscera. Punctate, globular,
in the same pancreatic and venous phases, postpro- or rarely lamellar calcifications can be revealed in
cessing can provide an accurate vascular map of the the region of the pancreas (MATHIEU et al. 1989;
pancreatic area, identifying phenomena of compres- VILGRAIN et al. 1999; FRIEDMAN et al. 1983).
sion, more rarely occlusive, of the peripancreatic
vascular structures. 3.2.4.2
Contrast Examination
3.2.3.2.2 of the Upper Gastrointestinal Tract
Oligocystic SeT
SCT, more commonly situated in the head, can pro-
The oligocystic form has a nonspecific MR aspect with duce a widening of the duodenal loop, and the rare
characteristics analogous to those demonstrated by CT positioning in the body-tail can impress the stomach
(Fig. 3.20). Once again, calcifications eventually present or the colon (MATHIEU et al. 1989; VILGRAIN et al.
are not distinguishable (PROCACCI et al. 1997). 1999). These findings are incidentally noted during
barium studies of the digestive tracts, in which pan-
creatic disease is not suspected as the cause of the
3.2.4 patient's symptoms.
Other Imaging Modalities
3.2.4.3
3.2.4.1 Angiography
Plain Radiographs
Angiography has often been used in the study of
When the masses are sufficiently large, plain film cystic masses of the pancreas since it is able to distin-
examination of the abdomen will demonstrate a soft- guish hypervascular lesions, almost always benign,
tissue mass in the epigastric region, with a displace- from hypovascular ones, which are malignant or
Serous Cystic Tumors 4S

a b

c d

Fig. 3.17a-d. Microcystic SCT (MR evaluation). a, b T2W HASTE sequences obtained in the craniocaudal direction reveal a lesion
of the pancreatic body. The mass gently displaces the duct ofWirsung. The lesion has a mixed micro cystic and macro cystic appear-
ance. The macro cystic component has a higher signal intensity than the micro cystic one. c, d At MRCP the mass overlies the main
duct in the coronal plane (c), while in the axial (d) plane, the absence of a communicating duct between the lesion and the main
duct leads to a diagnosis of serous cystadenoma

Fig. 3.18a,b. Von Hippel-Lindau disease (MR evaluation; same patient as in Fig. 3.12c,d). In the T2W TSE images obtained in
a craniocaudal direction, the pancreatic gland is notably increased in volume and replaced by multiple cystic formations; the
duct of Wirsung is not dilated. There are also lesions with analogous signal behavior in the right kidney (b)
46 G. Carbognin et ai.

a b

Fig. 3.19a,b. Microcystic SCT (MR evaluation). After the administration of Gd-DTPA on both the axial (a) and coronal (b) II W
GRE scans, a large mass of the body-tail with the typical 'honeycomb' aspect is evident

a b

Fig. 3.20a,b. Oligo cystic SCT (MR evaluation). In the IIW (a) and T2W (b) images obtained in the axial plane, the mass shows
a unilocular macrocystic aspect. Typical internal septations are not evident in this variety of path-proven SCT

potentially so (WARTER et al. 1981). In fact, in the solid tumor (a nonfunctioning endocrine tumor, for
case of a micro cystic tumor, selective arteriography example). In the presence of a tumor with a micro-
of the celiac trunk and the superior mesenteric artery cystic nucleus surrounded by macrocysts, arteriog-
demonstrates the presence of a hypervascular mass raphy can document the intense enhancement of the
with dilated afferent arteries, neo-vascularization micro cystic hypervascularization and the 'basket-
and 'blush' capillaries (Fig. 3.21). The hypervascu- like' imprint by the non-vascularized macro cysts on
larization is due to the extended capillary network the vessels at the periphery (Fig. 3.22).
present in the septa (WARTER et al. 1981; WARSHAW Oligo cystic SCT inevitably has a nonspecific
and RUTLEDGE 1987; BUCK and HAYES 1990). Signs of angiographic appearance, showing up as an avascu-
vascular encasement are, however, not recognizable lar mass with possible smooth displacement of arte-
(FRIEDMAN et al. 1983; ITAI et al. 1988). In the arterial rial and venous peripancreatic vessels (Fig. 3.23), an
phase, radiolucent areas, equal to or less than 2 em, appearance identical to mucinous cystic tumor.
are recognizable around the mass (Fig. 3.21a). In the
parenchymal phase, the intense enhancement of the 3.2.4.4
septa, especially in the sponge-like form, renders the Endoscopic Retrograde Choiangiopancreatography
mass uniformly dense (Fig. 3.21b). The intense and
homogeneous enhancement of the lesion can some- ERCP has occasionally been used in the study of
times lead to the misdiagnosis of a hypervascular cystic tumors with the aim of characterizing the
Serous Cystic Tumors 47

a b

Fig. 3.2Ia,b. Microcystic SCT (angiographic evaluation). a Selective injection into the superior mesenteric artery shows the
presence of a large hypervascular mass in the pancreatic head. There is a capillary 'blush', around which the afferent arteries
appear dilated. The enhancement is inhomogeneous due to the presence of radiolucent areas inside the mass. In the portal
phase (b) the intense enhancement persists with imprinting on the veins

a b

Fig. 3.22a,b. Microcystic SCT. a CT evaluation: in the portal venous phase, a mass with a micro cystic nucleus outlined by mac-
rocysts is recognizable. b Angiographic evaluation: in the arterial phase, selective arteriography of the celiac trunk shows the
intense enhancement of the micro cystic nucleus and the basket-like dislocation of the branches of the pancreatic arches by the
non -vascularized macro cysts

lesion. In reality, in the presence of a SCT, the ERCP clarified the morphological characteristics of the
examination is invariably normal (WARSHAW and collateral duct IPMT, whose appearance is analo-
RUTLEDGE 1987). Only in the larger lesions is it pos- gous to SCT, particularly in small masses, ERCP is
sible to find a non-specific mass effect on the duct of justified for the differential diagnosis (Fig. 3.16d).
Wirsung which otherwise retains its regular caliber In fact, this is the only method able to confirm the
and morphology (Fig. 3.24). An obstruction in the absence of communication between the cystic mass
main duct, relatively common in malignant cystic and the main pancreatic duct, allowing for a definite
tumors, is extremely rare in SCT (FRIEDMAN et al. diagnosis of microcystic SCT (PROCACCI et al. 1996,
1983; GAZELLE et al. 1993). More recently, having 2001b). There is only one reported case of a micro-
48 G. Carbognin et al.

a b

c d

Fig. 3.23a-d. Oligo cystic SCT. a CT evaluation: in the venous phase, a bi-lobed, macrocystic lesion is recognizable in the pancre-
atic tail. b-d Angiographic evaluation: in the arterial (b), venous (c) and delayed (d) phases, as shown by CT, a non enhancing
area is visible in the pancreatic tail (asterisk) at the site of the lesion

Fig. 3.24a,b. Oligo cystic SCT. a CT evaluation: in the late phase, a large, unilocular cystic mass is recognizable in the pancreatic
body. b ERCP evaluation: it is possible to distinguish the mass effect (arrows) on the duct of Wirsung that has an otherwise
regular caliber and morphology
Serous Cystic Tumors 49

cystic adenoma communicating with the collateral tion duct. Definitive evaluation of the communication
duct at ERCP (DELCENSERIE et al. 1988). with the pancreatic duct is not possible in every case.
Recently, it was suggested that EUS should be used
3.2.4.5 in the study of unilocular or multilocular macrocys-
Endoscopic US tic masses as the more accurate structural analysis,
even where large masses are concerned, would allow
EUS can be used in the study of cystic masses of the visualization of the parietal microcysts which are
pancreas with the aim of characterizing the lesion otherwise not distinguishable with other imaging
when this has not been done by cross-sectional imag- techniques. SCTs that manifest with an apparent
ing. In fact, this technique's contrast and spatial reso- oligo cystic aspect in other investigations can also be
lution are particularly high (ARIYAMA et al. 1998). characterized if small micro cystic foci are present
It is certainly not only superior to transabdominal (LEVY et al. 1995; PALAZZO et al. 1999; KANETO et
US, but to other cross-sectional imaging methods as al. 2000). Nevertheless, there remains a percentage of
well (ARIYAMA et al. 1998). In the classic micro cystic macro cystic lesions with unilocular appearance and
forms, which may falsely appear solid, EUS can visu- totally anechoic texture where EUS fails to demon-
alize the micro cysts (smaller than 2 mm) within the strate the presence of peripheral micro cysts within or
mass (NAPOLEON et al. 1993; PALAZZO et al. 1999) just close to the wall. In this case, a diagnosis of nature
(Fig. 3.25). In our personal experience and that of based only on morphological criteria is feasible in
other authors, EUS has proved particularly useful in no more than 50% (LEVY et al. 1995; MALLERY et al.
studying small lesions since it can demonstrate the 1998), while the differential diagnosis from a branch
micro cystic architecture and help clarify the nature duct IPMT appearing as an unilocular cyst is hardly
of a cystic pancreatic mass (Fig. 3.26). In the presence feasible (PALAZZO et al. 1999; VALETTE et al. 2001). In
of cystic lesions smaller than 2 cm, EUS becomes an the specific case of unilocular lesions, needle aspira-
essential examination because the morphological tion under endosonographic guidance (FNA) may be
definition and classification have a high accuracy of help. The sample is taken in a more precise way
(KoITO et al. 1997,2001). Even the small serous cys- than that obtained under transabdominal guidance
toadenomas demonstrate an internal structure similar and with a smaller risk of dissemination (PALAZZO et
to that typical honeycomb (BRUGGE 2000) (Fig. 3.27). al. 1999; BHUTANI et al. 1997; SUITS et al. 1999).
The relationship between the small tumor and the The cytologic examination, the biochemical
pancreatic duct can also be evaluated with greater analysis of the fluid and the dosage of the tumoral
accuracy so as to exclude (SCT) or demonstrate markers (CEA, CA 19-9, CA 72-4, Ki-ras gene muta-
(branch duct IPMT) the presence of a communica- tions, etc.) can contribute to improving the diagno-

a b

Fig. 3.25a,b. Microcystic SCT (EUS evaluation). a Large lesion, adjacent to the duodenal wall. Within the apparently solid struc-
ture, multiple cystic lesions are appreciable. b Different case: multiple micro cysts are visible within the smalliesion(arrow).
PV, portal vein
50 G. Carbognin et al.

a b

Fig. 3.26a-c. Microcystic SCT. a Transabdominal US evalu-


ation: small cystic and apparently unilocular lesion in the
head of the pancreas. b CT evaluation: the unilocular aspect
of the lesion is confirmed. c EUS evaluation: the lesion has
lobulated contours and central septa which allows recognition
c of a micro cystic SCT

sis. In particular, CEA is useful for discriminating like' aspect of the lesion. Finding a mass with these
between mucinous and nonmucinous lesions (LEVY characteristics in the pancreatic head in a woman,
et al. 1995; MALLERY et al. 1998; BRUGGE et al. 2001). especially when the pancreatic gland upstream of the
A lower titer of CEA, the absence of mucin, the pres- lesion appears normal with no dilation of the duct
ence of typical glycogen-rich, PAS-positive cells and of Wirsung, and central calcification when present
the absence of malignant cells lead to the diagnosis of (in 10%-30% of cases according to COMPTON 2000)
serous cystadenoma. results in a definite diagnosis of SCT. In this case, there
is no need for further diagnostic investigation since
the therapeutic choice, especially in advocating radi-
cal surgery, will be made on the basis of the clinical
3.3 picture and the patient'S general condition. The diag-
Differential Diagnosis nosis of SCT can also be considered definite when the
lesion shows a mixed aspect with macro cysts (more
In light of what has been reported in the previous sec- than 3 cm in diameter) situated on the periphery
tion, whichever diagnostic method is used, the differ- of the micro cystic nucleus. If surgical intervention
ential diagnosis problems that SCT poses for imaging is decided against, the lesion must be monitored.
depend on whether the micro cystic or the oligocystic The development of a serous cystadenoma is vari-
form is being considered. US and/or CT, which are able since the mass in some patients can remain
the more commonly used imaging techniques in the unchanged for many years (Fig. 3.28), while in others
study of SCT, give three possible diagnoses. a progressive growth is recorded (Box and DOUGLAS
In the majority of cases, these methods are able to 2000) and, in relation to the appearance of signs of
document the micro cystic, 'honeycomb' or 'sponge- compression on the adjacent structures, may justify
Serous Cystic Tumors 51

a b

c d

Fig. 3.27a-d. Microcystic SCT (EUS evaluation). a Small lesion of the pancreatic body (arrow), with honeycomb architecture.
b Different case: middle-sized lesion with multiple septa and small peripheral cystic areas (white arrows). c Another case: small
lesion, composed of micro cysts separated by thin septa. Changing the scan angle (d), the same lesion shows microscopic septa
and a peripheral cystic area. PV, portal vein; SV, splenic vein; SMV, superior mesenteric vein

a b

Fig. 3.28a,b. Follow-up of micro cystic SCT (CT evaluation). The mass with a typical microcystic appearance (a) does not show
significant morphological or dimensional variations in the follow-up performed 2 years after the initial scan (b)
52 G. Carbognin et al.

late, but inevitable, radical surgery (Figs. 3.29, 3.30). a communicating duct between the mass and the duct
In some cases the volume may spontaneously of Wirsung. MRCP could be used for this purpose, but
decrease, and eventual macro cysts, situated on the the diagnosis can only be considered definite when
periphery of the micro cystic nucleus, may even dis- the absence of a communication duct between the
appear (Fig. 3.31). mass and the main duct is shown. This can be done by
Despite demonstrating a micro cystic aspect, the acquiring multiple plane images (Fig. 3.17). However,
correct diagnosis of the lesion is less certain when- when the lesion is superimposed upon or adjacent to
ever the mass is found in a male patient, whether the duct of Wirsung (Figs. 3.lOd, 3.16a-c), it is neces-
located in the uncinate process and/or associated sary, even today, to carry out an ERCP (Fig. 3.16d).
with dilation of the duct of Wirsung (Fig. 3.10). In For various reasons, the tumor can appear as a
this event, in fact, the differential diagnosis of branch solid mass with US or CT (Figs. 3.2, 3.11), resulting
ducts IPMT may be more appropriate. in misdiagnosis. In particular, and above all with CT,
To make a differential diagnosis, the primary aim of the bright enhancement of the tumor can lead to the
imaging is to demonstrate the presence or absence of misdiagnosis of a non-functioning endocrine tumor.

a b

Fig. 3.29a,b. Follow-up of micro cystic SCT (US evaluation). The mass located in the tail of the pancreas has a typical micro-
cystic aspect and shows significant growth after 5 years. At the second follow-up (b) the compressing effect on the splenic vein
(arrows) is more evident. At surgery, benign micro cystic SCT was diagnosed

a b

Fig.3.30a,b. Follow-up of microcystic SCT (CT evaluation). The lesion with a micro cystic structure located in the tail of the
pancreas (a) shows conspicuous growth at the second follow-up (b) carried out after 3 years. At surgery, benign microcystic
SCT was diagnosed
Serous Cystic Tumors 53

a b

Fig. 3.31a,b. Follow-up of micro cystic SCT (CT evaluation). In the second follow-up done after 28 months, the mass has changed
aspect in that the macrocyst, recognizable on the previous study (asterisk in a), has been reabsorbed (b)

In some cases the use of both techniques can correct References


the reciprocal errors; in others, in order to resolve the
diagnostic doubt, it is necessary to turn to MR which, Abe H, Kubota K, Mori M, Mild K, Minagawa M, Noie T,
mostly thanks to T2-weighted image sequences, is Kimura W, Makuuchi M (1998) Serous cystadenoma of the
pancreas with invasive growth: benign or malignant? Am J
always able to recognize the micro cystic aspect of Gastroenterol 93: 1963-1966
the lesion. Ariyama J, Suyama M, Satoh K, Wakabayashi K (1998) Endo-
The diagnosis is necessarily non-specific when scopic ultrasound and intraductal ultrasound in the
the SCT has a unilocular or multilocular macro cystic diagnosis of small pancreatic tumors. Abdom Imaging
aspect (Figs. 3.3, 3.13, 3.20, 3.24), analogous to that 23:380-386
Bhutani MS, Hawes RH, Baron PL, Sanders-Cliette A, van Velse
of many other cystic lesions of the pancreas. In the A, Osborne JF, Hoffman BJ (1997) Endoscopic ultrasound
past, in the cases examined with conventional CT, guided fine needle aspiration of malignant pancreatic
such misclassification was due to not recognizing lesions. Endoscopy 29:854-858
central septations which were easily demonstrated Box JC, Douglas HO (2000) Management of cystic neoplasms
by US or MR. At present, the macro cystic lesion seen of the pancreas. Am Surg 66:495-501
Brugge WR (2000) The role of EUS in the diagnosis of cystic
with all imaging modalities is related to an oligocys- lesions of the pancreas. Gastrointest Endosc 52 [Suppl):
tic SCT. Proper characterization of this mass may be S18-S22
impossible: the only technique able to supply supple- Brugge WR, Saltzman JR, Scheiman JM, Wallace MB, Jowell
mentary information seems to be EUS, which can PS, Pochapin MB, Stevens PD, Gress FG, Lewandrowski K
demonstrate the micro cystic aspect of rather small (2001) Diagnosis of cystic neoplasms of the pancreas by
EUS: the report of the cooperative pancreatic cyst study.
lesions (Figs. 3.25, 3.26) or the presence of parietal Gastrointest Endosc 53:AB71
microcysts around a macrocystic mass. Thanks to Buck JL, Hayes WS (1990) From the archives of the AFIP:
such findings, not obtainable with the other methods, micro cystic adenoma of the pancreas. Radiographics 10:
it is possible to correctly classify the lesion. In the 313-322
majority of these cases, unfortunately the diagnosis Carlson SK, Johnson CD, Brandt KR, Batts KP, Salomao DR
(1998) Pancreatic cystic neoplasms: the role and sensitiv-
is still non-specific; needle aspiration of the lesion ity of needle aspiration and biopsy. Abdom Imaging 23:
is also not always able to supply decisive elements 387-393
for characterizing the lesion. Therefore, the smaller Choyke PL, Glenn GM, Walther MM, Patronas NJ, Linehan
lesions, particularly in elderly patients, are followed WM, Zbar B (1995) Von Hippel-Lindau disease: genetic,
up, while the inevitable final step for the others is clinical, and imaging features. Radiology 194:629-642
Compagno J, Oertel JE (1978a) Mycrocystic adenomas of the
surgical intervention. pancreas (glycogen-rich cystadenomas). A clinicopatho-
logic study of 34 cases. Am J Clin Pathol 69:289-298
Compagno J, Oertel JE (1978b) Mucinous cystic neoplasms
of the pancreas with overt and latent malignancy (cyst-
adenocarcinoma and cystadenoma). Am J Clin Pathol 69:
573-580
54 G. Carbognin et al.

Compton CC (2000) Serous cystic tumors of the pancreas. graphic assessment. AJR Am J RoentgenoI151:1133-1138
Semin Diagn Pathol 17:43-55 Jung HK, Son HY, Lee HC, Young SY (2001) Microcystic ade-
Curry CA, Eng J, Horton KM, Urban B, Siegelman S, Kuszyk BS, noma coexistent with low-grade malignant islet cell tumor
Fishman EK (2000) CT of primary cystic pancreatic neo- of the pancreas. J Clin Gastroenterol 32:441-443
plasms: can CT be used for patient triage and treatment? Kanematsu M, Shiratori Y, Hoshi H, Kondo H, Matsuo M,
AJR Am J RoentgenoI175:99-103 Moriwaki H (2000) Pancreas and peripancreatic vessels:
Delcenserie R, Dupass JL, Joly JP, Descombes P, Mortier F, effect of imaging delay on gadolinium enhancement at
Capron JP (1988) Microcystic adenoma of the pancreas dynamic gradient-recalled-echo MR imaging. Radiology
demonstrated by endoscopic retrograde pancreatography. 215:95-102
Gastrointest Endosc 34:52-54 Kaneto H, Endo T, Ozeki I, Itoh H, Sasaki S, Mukaiya M, Ikeda
Dupas B, Le Borgne J (2000) L'apport de la cholangiopan- K, Koito K, Imai K (2000) Macrocystic serous cystade-
creatographie-IRM dans les tumeurs kystiques du pan- noma of the pancreas: importance of coexistent tiny cysts
cnias. Ann Chir 125:571-577 depicted by EUS. J Gastroenterol 35 :472-475
Eriguchi N, Aoyagi S, Nakayama T et al (1998) Serous cyst- Keel SB, Zukerberg L, Graeme-Cook F, Compton CC (1996) A
adenocarcinoma of the pancreas with liver metastases. J pancreatic endocrine tumor arising within a serous cystad-
Hepatob Pancreat Surg 5:467-470 enoma of the pancreas. Am J Surg PathoI20:471-475
Fishman EK, Horton KM, Urban BA (2000) Multidetector CT Kimura W, Makuuchi M (1999) Operative indications for cystic
angiography in the evaluation of pancreatic carcinoma: pre- lesions of the pancreas with malignant potential - our expe-
liminary observations. J Comput Assist Tomogr 24:849-853 rience. Hepatogastroenterology 46:483-491
Friedman AC, Lichtenstein JE, Dachman AH (1983) Cystic Kloppel G, Solcia E, Longnecker DS, Capella C, Sobin LH
neoplasms of the pancreas. Radiological-pathological cor- (1996) Histological typing of tumors of the exocrine pan-
relation. Radiology 149:45-50 creas. International histological classification of tumors,
Fugazzola C, Procacci C, Bergamo Andreis lA, Iacono C, 2nd ed. Berlin: Springer-Verlag
Portuese A, Dompieri P, Laveneziana S, Zampieri PG, Ian- Koito K, Namieno T, Nagakawa T, Shyonai T, Hirokawa N, Morita
nucci A, Serio G, Pistolesi GF. (1991) Cystic tumors of the K (1997) Solitary cystic tumor of the pancreas: EUS-patho-
pancreas: evaluation by ultrasonography and computed logic correlation. Gastrointest Endosc 45:268-276
tomography. Gastrointest Radiol 16:53-61 Koito K, Namieno T, Nagakawa T, Hirokawa N, Ichimura
Gazelle GS, Mueller PR, Raafat N, Halpern EF, Cardenosa G, T, Syonai T, Yama N, Someya M, Nakata K, Sakata K,
Warshaw AL (1993) Cystic neoplasms of the pancreas: Hareyama M (2001) Pancreas: imaging diagnosis with
evaluation with endoscopic retrograde pancreatography. color/power doppler, ultrasonography, endoscopic ultra-
Radiology 188:633-636 sonography, and intraductal ultrasonography. Eur J Radiol
George DH, Murphy F, Michaski R, Ulmer BG (1989) Serous 38:94-104
cystadenocarcinoma of the pancreas: a new entity? Am J Le Borgne J, de Calan L, Partensky C (1999) Cystadenomas and
Surg Pathol13:61-66 cystadenocarcinomas of the pancreas: a multiinstitutional
Gouhiri M, Soyer P, Barbagelatta M, Rymer R (1999) Macro- retrospective study of 398 cases. Ann Surg 230:152-161
cystic serous cystadenoma of the pancreas; CT and endo- Levy M, Levy P, Hammel P, Zins M, Vilgrain V, Amouyal G,
sonographic features. Abdom Imaging 24:72-74 Amouyal P, Molas G, FJejou JF, Voitot H, Sauvanet A, Bara
Hammel P (2000) Interet diagnostique de l'analyse du liquide J, Menu Y, Belghiti J, Bernades P (1995) Diagnostic des
de ponction des lesions kystiques du pancreas: donnees cystadenomes et cystadenocarcinomes du pancreas. Gas-
actuelles, limites et perspectives. J Radiol 81 :487-490 troenterol Clin Bioi 19: 189-196
Healy JC, Davies SE, Reznek RH (1994) CT of Microcystic Lewandrowski KB, Warshaw A, Compton C (1992) Macrocystic
(serous) Pancreatic Adenoma. J Comput Assist Tomogr serous cystadenoma of the pancreas: a morphologic vari-
18:146-148 ant differing from microcystic adenoma. Hum Pathol 23:
Heresbach D, Robert I, Le Berre N, Raoul JL, Siproudhis L, 871-875
Bretagne JF, Ramee MP, Gosselin M (1993) Tumeurs kys- Lewandrowski KB, Southern JF, Pins MR, Compton CC, War-
tiques et tumeur endocrine du pancreas. Gastroenterol shaw AL (1993) Cyst fluid analysis in the differential diag-
Clin Bioi 17:968-971 nosis of pancreatic cysts. A comparison of pseudo cysts,
Hes FJ, Feldberg MAM (1999) Von Hippel-Lindau disease: serous cystadenomas, mucinous cystic neoplasms, and
strategies in early detection (renal-, adrenal-, pancreatic mucinous cystadenocarcinoma. Ann Surg 217:41-47
masses). Eur RadioI9:598-610 Lundstedt C, Dawiskiba S (2000) Serous and mucinous
Iselin CE, Meyer P, Hauser H, Kurt A-M, Vermeulen BJM, cystadenoma/cystadenocarcinoma of the pancreas. Abdom
Rohner A. (1993) Computed tomography and fine-needle Imaging 25:201-206
aspiration cytology for preoperative evaluation of cystic Mallery S, Quirk D, Lewandrowski K, Centeno B, Warshaw A,
tumors of the pancreas. Br J Surg 80: 1166-1169 Brugge W (1998) EUS-guided FNA with cyst fluid analysis
Itai Y, Moss AA, Ohtomo K (1982) Computed tomography of in pancreatic cystic lesions. Endoscopy 30 [SuppllJ:AI80
cystadenoma and cystadenocarcinoma of the pancreas. Mathieu D, Guigui B, Valette pJ, Dao TH, Bruneton IN, Bruel
Radiology 145:419-425 JM, Pringot J, Vasile N (1989) Pancreatic cystic neoplasms.
Itai Y, Ohhashi K, Furui S, Araki T, Murakami Y, Ohtomo K, Radiol Clin North Am 27:163-176
Atomi Y (1988) Microcystic adenoma of the pancreas: Mergo PJ, Heimberger TK, Buetow PC, Heimberger RC, Ros PR
spectrum of computed tomographic findings. J Comput (1997) Pancreatic neoplasms: MR imaging and pathologic
Assist Tomogr 12:797-803 correlation. Radiographies 17:281-301
Johnson CD, Stephens DH, Charboneau JW, Capenter HA, Mori K, Takeyama S, Hirosawa H, Watanabe T, Taniya T,
Welch TJ (1988) Cystic pancreatic tumors: CT and sono- Arakawa H, Nagakawa T, Ohta T, Miyazaki I, Nakanuma
Serous Cystic Tumors 55

Y (1995) A case of macrocystic serous cystadenoma of the Laissy JP, Archour E, Levesque M (1994) Cystic tumors of
pancreas. Int J PancreatoI17:91-93 the pancreas: dynamic CT studies. J Com put Assist Tomogr
Nagel M, Dobrowolski F, Bunk A, Saeger HD (2000) Das Serose 18:420-426
Cystadenom des Pankreas. Chirurg 71:424-428 Sperti C, Pasquali C, Perasole A, Liessi G, Pedrazzoli S (2000)
Napoleon B, Pujol B, Ponchon T, Valette PJ, Souquet JC (1993) Macrocystic serous cystadenoma of the pancreas: clinico-
Tumeurs kystiques du pancreas: l'echoendoscopie per met- pathologic features in seven cases. Int J PancreatoI28:1-7
elle de choisir entre chirurgie et surveillance. Gastroenterol Suits J, Frazee R, Erickson RA (1999) Endoscopic ultrasound
Clin Bioi 17 [SuppI2J:A89 and fine needle aspiration for the evaluation of pancreatic
Nino-Murcia M, Jeffrey RB, Beaulieu CF, Li KCP, Rubin GD masses. Arch Surg l34:639-642
(2001) Multidetector CT of the pancreas and bile duct Talamini MA, Pitt HA, Hruban RH, Boitnott JK, Coleman JA,
system: value of curved planar reformations. AJR Am J Cameron JL (1992) Spectrum of cystic tumors of the pan-
Roentgenol 176:689-693 creas. Am J Surg 163:117-124
Nishihara K, Kawabata A, Ueno T, Miyahara M, Hamanaka Y, Torresan F, Casadei R, Solmi L, Marrano D, Gandolfi L (1997)
Suzuki T (1996) The differential diagnosis of pancreatic The role of ultrasound in the differential diagnosis of
cysts by MR imaging. Hepatogastroenterology 43:714-720 serous and mucinous cystic tumours of the pancreas. Eur
Padovani B. Neuveut P, Chanalet S, Benchimol D, Saint-Paul J Gastroenterol HepatoI9:169-172
MC, Michiels JF, Arpurt JP, Serres JJ (1991) Microcystic Valette 0, Cuilleron M, Debelle L, Antunes L, Mosnier JF,
adenoma of the pancreas: report on four cases and review Regent D, Veyret C (2001) Imagerie des tumeurs intra-
of the literature. Gastrointest RadioI16:62-66 canalaires papillaires mucineuses du pancreas: revue de la
Palazzo L, Hammel L, Cellier C, Ruszniewski P (1999) Le litterature. J Radiol 82:633-645
tumeurs kystiques du pancreas. Acta Endoscop 29:418-422 Vilgrain V, Menu Y, Lorphelin JM, Nahum H (1987) Cysta-
Procacci C, Graziani R, Bicego E, Bergamo-Andreis lA, Main- denomes pancreatiques. Pieges et limites du diagnostic
ardi P, Zamboni G, Perderzoli P, Cavallini G, Valdo M, Pisto- radiologique. J Radiol 68:455-463
lesi GF (1996) Intraductal mucin-producing tumors of the Vilgrain V, Mathieu D, Bruel J-M, Flejou J-F, Rahmouni A,
pancreas: imaging findings. Radiology 198:249-257 Taourel P (1999) Cystic tumors of the pancreas, 2nd rev
Procacci C, Graziani R, Bicego E, Bergamo-Andreis lA, Guar- edn. Springer, Berlin Heidelberg New York
ise A, Valdo M, Bogina G, Solarino U, Pistolesi GF (1997) Warshaw AL, Rutledge PL (1987) Cystic tumors mistaken for
Serous cystadenoma of the pancreas: report of 30 cases pancreatic pseudocysts. Ann Surg 205:393-398
with emphasis on the imaging findings. J Comput Assist Warshaw AL, Compton CC, Lewandrowski K, Cardenosa G,
Tomogr 21:373-382 Mueller PR (1990) Cystic tumors of the pancreas. New
Procacci C, Biasiutti C, Carbognin G, Accordini S, Bicego E, clinical, radiologic and pathologic observations in 67
Guarise A, Spoto E, Bergamo Andreis lA, De Marco R, patients. Ann Surg 212:432-443
Megibow AJ (1999) Characterisation of cystic tumors of Warter J, Walter P, Warter A, Bareiss P, Sibilly A (1981) Cysta-
the pancreas: CT accuracy. J Comput Assist Tomogr 23: denomes du pancreas. Considerations sur leur vascularisa-
906-912 tion et les maladies qui leur sont associees. Sem Hop Paris
Procacci C, Biasiutti C, Carbognin G, Capelli P, EI-Dalati G, 57:529-537
Falconi M, Misiani G, Ghirardi C, Zamboni G (2001a) Pan- Widmaier U, Mattfeldt T, Siech M et al (1996) Serous cystade-
creatic neoplasms and tumor-like conditions. Eur Radiol nocarcinoma of the pancreas. Int J PancreatoI20:l35-l39
11 [SuppI2J:167-192 Wolfman NT, Ramquist NA, Karstaedt N, Hopkins MB (1982)
Procacci C, Carbognin G, Biasiutti C, Ghirardi C, Misiani G, Cystic neoplasms of the pancreas: CT and sonography AJR
Schenal G, Tapparelli M (2001b) Cistoadenoma Sieroso del Am J Roentgenoll38:37-41
Pancreas. I. Rilievi dell'Imaging. Radiol Med lO2:23-31 Wu CM, Fishman EK, Hruban RK, Schlott WD, Cameron JL
Sarr MG, Kendrick ML, Nagorney DM, Thompson GB, Farley (1999) Serous cystic neoplasm involving the pancreas and
DR, Farnell MB (2001) Cystic neoplasms of the pancreas: liver: an unusual clinical entity. Abdom Imaging 24:75--77
benign to malignant epithelial neoplasms. Surg Clin North Yamaguchi K, Chijiwa K, Shimizu S, Yokohata K, Morisaki T,
Am 81:497-509 Tanaka M (1998) Comparison of endoscopic retrograde and
Sheehan M, Latona C, Aranha G, Pickleman J (2000) The magnetic resonance cholangiopancreatography in the surgi-
increasing problem of the unusual pancreatic tumors. cal diagnosis of pancreatic diseases. Am J Surg 175:203-208
Arch Surg 135:644-650 Yang EYT, Joehl RJ, Talamonti MS (1994) Cystic neoplasms of
Solcia E, Capella C, Kloppel G (1997) Tumors of the pancreas. the pancreas. J Am Coli Surg 179:747-757
Atlas of tumor pathology, 3rd ser, fasc 20. Armed Forces Zirinsky K, Abiri M, Baer JW (1984) Computed tomography
Institute of Pathology, Washington DC demonstration of pancreatic micro cystic adenoma. Am J
Soyer P, Rabenandrasana A, Van Beers B, Barge J, Sibert A, GastroenteroI79:139-142
4 Mucinous Cystic Tumors
C. BIASIUTTI, F. FORNASA, S. VENTURINI, N. PAGNOTTA, G. SCHENAL, and C. PROCACCI

CONTENTS of epithelial abnormality coexisting in the same


tumor (ZAMBONI et al. 1999; SARR et al. 2000), MCT
4.1 Introduction 57 will display a more uniform epithelial profile within
4.2 Imaging 57 the same lesion. However, most authorities believe
4.2.1 Transabdominal US 57
4.2.2 Computed Tomography 60
that MCT is, at best, a premalignant lesion, and there-
4.2.3 Magnetic Resonance Imaging 64 fore it is important to distinguish it from other cystic
4.2.4 Other Imaging Modalities 64 tumors of the pancreas. A complicating factor is that
4.2.4.1 Plain Films 64 because many are benign adenomas when resected,
4.2.4.2 Contrast Examination it is also desirable to be able to define features char-
of the Upper Gastrointestinal Tract 69
4.2.4.3 Angiography 69 acterizing either a benign or malignant tumor to aid
4.2.4.4 Endoscopic Retrograde in establishing the prognosis and to help in choosing
Cholangiopancreatography 69 the appropriate therapy (PROCACCI et al. 2001b).
4.2.4.5 Endoscopic US 70 In the majority of cases, an MCT is identified acci-
4.3 Prognostic Evaluation 71 dentally during radiological examinations carried
4.4 Differential Diagnosis 71
References 72
out for other clinical indications not directly refer-
able to the pancreas. Therefore, the roles of imaging
include identifying, characterizing, and classifying
a given lesion as MCT and, when appropriate, sug-
gesting a differential diagnosis. Today, ultrasound
4.1 (US), computed tomography (CT), and magnetic
Introduction resonance imaging (MR) are the three imaging pro-
cedures called upon to answer these questions. There
The mucinous cystic tumor (MCT) was first clas- is no role for radiological examinations, such as plain
sified by OERTEL and COMPAGNO as 'macro cystic film of the abdomen, upper gastrointestinal tract
cystadenoma/cystadenocarcinoma' to differentiate examination, or angiography. ERCP, endoscopic US
it from the microcystic serous cystic tumor (SCT) (EUS), and fine needle aspiration biopsy (FNAB) may
(COMPAGNO and OERTEL 1978). Subsequently, a provide definitive ancillary information (FUGAZZOLA
further subdivision between peripheral MCT and etal.1991).
intraductal papillary mucinous tumor (IPMT) was
made. Although both of these lesions are lined with
a mucin-secreting epithelium, the clinical and mac-
roscopic pathological characteristics are completely 4.2
different (SHYR et al. 1996). The WHO classification Imaging
of 1996 distinguishes MCT from IPMT. While IPMT
may display four different grades, from adenoma 4.2.1
through low-grade/high-grade dysplasia to invasive Transabdominal US
carcinoma in accordance with the degree of epithelial
dysplasia (WILENTZ et al. 2000), with different grades The increasing use of US in recent years has certainly
contributed to the rise in the number of diagnoses
of MCT. Moreover, the technological refinements in
C. BIASIUTTI, MD; F. FORNASA, MD; S. VENTURINI, MD; N.
PAGNOTTA, MD; G. SCHENAL, MD; C. PROCACCI, MD US in the last decade have improved pancreatic US
Istituto di Radiologia, Policlinico Universitario 'GB Rossi', P.le evaluation even in obese patients or those with large
L.A. Scuro, 10,37134 Verona, Italy amounts of intestinal gas.
58 C. Biasiutti et al.

MCT displays two characteristic imaging patterns locules (Fig. 4.1 b) (FRIEDMAN et al. 1983). Scattered
on all cross-sectional modalities (ALGARD et al. 1986; echoes can be recognized in the dependent portion
JOHNSON et al. 1988; FUGAZZOLA et al. 1991). The of the cyst (Fig. 4.1c), indicative of prior hemorrhage
macro cystic multilocular pattern is the most common (BUETOW et al. 1998). In calcified lesions, hyperechoic
variety of MCT. Although it is not pathognomonic lamellar foci will be present in the peripheral por-
(SPERTI et al. 1993), this variety is frequently located tions of the tumor (Fig. 4.1d) (WOLFMAN et al. 1982;
in the body-tail, appearing as a rounded, sharply FUGAZZOLA et al. 1991; FERNANDEZ DEL CASTILLO
defined mass with a variably thickened wall (BASTID and WARSHAW 1995; SCOTT et al. 2000). Larger cal-
et al. 1989; BUETOW et ai. 1998). Centrally located thin cifications produce the expected shadowing. With
septa delimit wide cystic spaces (FRIEDMAN et al. US examinations using tissue harmonics, exquisitely
1983; FUGAZZOLA et al. 1991). The lesion has a multi- thin peripheral septa will frequently be recognized.
locular, hypoechoic appearance displaying increased The importance of recognizing septa as a diagnostic
through transmission (Fig. 4.1a) (ALGARD et al. 1986; feature of MCT cannot be overemphasized. Careful
BUETOW et al. 1998). The cyst content can have vari- attention to technique and examining the pancreas
able acoustic characteristics within the same mass, from multiple orientations aid in the localization of
related to the amount of mucin within any of the these septa (FRIEDMAN et al. 1983). The thickness of

a b

c d

Fig. 4.1a-d. Macrocystic multilocular mucinous cystic tumor (MeT) (US evaluation). a A fluid-containing mass with sharp bor-
ders; there are thin septa (arrows) within the cystic space. The increased through transmission results in acoustic enhancement
of the posterior wall (asterisk). b Multilocular cystic formation with thin wall. The cystic space on the left is more echo genic
due to the greater amount of mucin (arrow). c Multilocular cystic formation with hemorrhagic debris; fine echoes are seen in
the dependent portion of the mass (arrows), a sign of previous hemorrhage. d Multilocular cystic formation with a calcification
(arrow) along one of the septa results in posterior shadowing (asterisk)
Mucinous Cystic Tumors 59

the septa, like that of the marginal capsule, is vari- including pseudocyst, cannot be made easily
able, from 2 mm to over 1 cm (Fig. 4.2) (ALGARD et (SPERTI et al. 1993; BUETOW et al. 1998). Smaller
al. 1986). When the wall is thicker, it is more common lesions often have this appearance. It should be
to find papillary proliferations originating from the emphasized that if one encounters a unilocular
wall or its septa. The thick septa and/or papillary pro- mass with a thick irregular wall, MeT should be
liferations are often proof of the malignant degenera- the primary diagnostic consideration (Fig. 4.3b)
tion of the MeT (WOLFMAN et al. 1982; BUETOW et (SCOTT et al. 2000). There can sometimes be intra-
al. 1998). mural hyperechoic foci along the thick wall which
The macrocystic unilocular pattern is less are due to calcifications. A thick wall and periph-
common and rather less specific than the multiloc- eral calcifications are ominous signs of malignant
ular one, in that it simulates any pancreatic cystic degeneration. The content of the unilocular MeT
mass (BUETOW et al. 1998; SCOTT et al. 2000). The may be inhomogeneous due to the presence of
tumor shows up as a hypoechoic, sharply circum- low-level echoes, producing a fluid-fluid level in the
scribed mass with enhanced through transmission. dependent portion of the lesion. These are presum-
If the walls are smooth and thin (Fig. 4.3a), differen- ably the result of episodes of recent hemorrhage
tiation between this lesion and other cystic masses, into the mass (BUETOW et al. 1998).

a b

Fig. 4.2a,b. Macrocystic multilocular MCT (US evaluation). Two large lesions with clear content and peripheral septa. The septa
thickness is variable, being either thin (long arrow in a) or thick (short arrow in b). Thick septa raise the suspicion of malignant
degeneration of the lesion

Fig.4.3a,b. Macrocystic unilocular MCT (US evaluation). Unilocular, thin-walled, fluid-containing intrapancreatic mass. In (a),
because of no other findings, the appearance is nonspecific, and a diagnosis cannot be made. In (b), however, the suspicion
of MCT can be more confidently raised due to the presence of mural vegetation (arrow). These mural elements also suggest
malignant degeneration
60 C. Biasiutti et al.

4.2.2
Computed Tomography

Thin-section helical/spiral CT is the primary modal-


ity for detecting and characterizing MCT. Multislice
CT offers significant improvements over single-slice
techniques. This is due to the ability to acquire thin-
section data and evaluate the images in any viewing
plane. The use of thin sections allows better delin-
eation of structural findings (septa, calcifications)
which, located in the superior or inferior poles of
the mass, can sometimes go unrecognized in axial
a
scans. The technique for studying these masses
either by single or multislice CT is identical to that
described in Chapter 3.2.2.1. The reader is referred
to this discussion.
The macrocystic multilocular pattern is best
demonstrated following IV contrast enhancement,
although careful attention should be paid to unen-
hanced images if they are obtained (BUETOW et al.
1998; PROCACCI et al. 2001a). On unenhanced images,
the tumor is easily identifiable (Figs. 4.4a, 4.5a, 4.6a,b,
4.7a). In this phase, the wall and the septa are identifi-
able only when they are thick or when there are lamel-
lar calcifications (BUETOW et al. 1998; CURRY et al.
2000; PROCACCI et al. 2001a; SARR et al. 2001). Precon- b
trast unenhanced images are particularly helpful for
documenting calcifications (Fig. 4.5a). In this phase,
variability in the density of the cyst contents is easily
recognizable (Figs. 4.5a, 4.6a,b). The variability is the
result of either greater or lesser quantities of mucin
or fluid-fluid levels resulting from prior hemorrhage
(ITAI and OHTOMO 1996; BUETOW et al. 1998).
Analysis of enhanced images during the pancre-
atic phase, the time during which the gland is maxi-
mally enhanced, reveals that the walls and the septa
appear to display a slightly lower attenuation value
compared with the densely enhanced surrounding
pancreatic parenchyma because of the fibrous tissue
composition and minimal vascularization (Figs. 4.4b,
c
4.5b, 4.6c,d, 4.7b) (SOYER et al. 1994). The outer wall
and the septa have a similar thickness (Figs. 4.4, 4.5). Fig. 4.4a-c. Macrocystic multilocular MCT (CT evaluation).
The septa are gently curved and can extend across a Unenhanced image: a large, thin-walled mass containing septa
the diameter of the tumor or bridge adjacent regions (arrows) expands the pancreatic tail and displaces the stomach
of the same wall. It is sometimes possible to recog- anteriorly. b Pancreatic phase image: notable enhancement of
the pancreatic parenchyma with respect to the mass. c Venous
nize small peripheral cysts located on, or sometimes
phase image: the septa become even more apparent
within, the wall (Fig. 4.8). When present in malignant
lesions, papillary projections or nodules can be
detected in the pancreatic phase of enhancement of the wall (FuGAZZOLA et al. 1991; PROCACCI et al.
(Figs. 4.6c, 4.7b, 4.8a) (ITAI et al. 1982; BUETOW et al. 2001a; SARR et al. 2001). Differential densities within
1998; CURRY et al. 2000; Ros et al. 2000). In mucinous the mass are less evident following contrast enhance-
cystadenocarcinoma, there can also be extracapsular ment (Fig. 4.5a,b). The combined use of thin sections
extension of the lesion as well as nodular thickening for optimal enhancement of peripancreatic vessels
a
a

Fig. 4.5a-c. Macrocystic multilocular MCT (CT evaluation). c


a Unenhanced image: large mass in the body-tail of the pancreas
with a mural (peripheral) calcification along the anterior wall
(arrow). Thin septa delimit different cystic compartments. The
variable attenuation within the loculations results from prior
hemorrhage into the largest one (asterisk). b Pancreatic phase
image: although the enhancement of the surrounding pancreas
makes localization apparent, the mural calcification is more dif-
ficult to detect. The septa are well seen. c Venous phase image:
progressive enhancement of the septa which have become hyper-
dense (short arrows) compared with the pancreatic parenchyma

Fig. 4.6a-d. Macrocystic multilocular MCT (CT evaluation). a,


b Unenhanced images: a large mass almost totally replaces the
pancreatic gland. The lesion contains multiple septa delimiting
cystic spaces with various attenuation resulting from prior hem-
orrhage. c, d Pancreatic phase image: enhancing mural nodules
d
(arrow) can be distinguished from hemorrhagic debris
62 C. Biasiutti et al.

Fig.4.7a-c. Macrocystic multilocular MCT (CT evaluation).


a Unenhanced image: a large, partially cystic mass in the
body-tail of the pancreas with prominent solid component
and thick septa. b Pancreatic phase image: early enhancement
of the solid components. The mass displaces the splenic vein
(arrows). c Venous phase image: progressive enhancement of
the capsule delimits the mass from the pancreatic gland. The

..
splenic vein is displaced, but not infiltrated, by the mass

Fig. 4.8a,b. Macrocystic multilocular MCT (CT evaluation).


Pancreatic phase image. A bi-lobate mass in the pancreatic
body-tail with thick septa and thick walls. Nodules (arrow in a)
are present along the wall and the septal margins. Small cystic
formations (arrowheads in a and b) are visible. The mass com-
presses and displaces the stomach anteriorly (asterisk) and the
c descending colon (C) posterolaterally
Mucinous Cystic Tumors 63

provides data sets which can accurately depict the result of the presence of fibrous tissue within the wall
tumor's relationships with the peripancreatic vessels. and stroma. This phenomenon results in increased
MeT will displace and compress surrounding arter- conspicuity of the tumor margin against the pancreatic
ies; frank encasement is unusual. Venous thrombosis parenchyma (Figs. 4.4c, 4.5c, 4.7c). The other findings
may occur resulting from local mass effect, but this are identical to those obtained in the pancreatic phase.
phenomenon is also unusual (Figs. 4.6c, 4. 7b) (SOYER Venous phase images are primarily used to survey the
et al. 1994). The relationships of the tumor with the entire abdomen and pelvis for extrapancreatic foci of
pancreatic duct and/or the common bile duct can disease. The most frequent locations of metastases are
also be depicted in both axial scans and reconstruc- within the peritoneum (JOHNSON et al. 1988) and liver
tions. Similar to SeT, the MeT will compress or dis- (SHAMSI et al. 1993; BUETOW et al. 1998).
place the ducts without frank invasion (WARSHAW The macrocystic unilocular pattern is easily con-
et al. 1990; STEPHENS 1997; PROCACCI et al. 2001a). fused with other cystic pancreatic masses (Fig. 4.9).
The compression rarely results in duct obstruction The demonstration of a thick irregular wall, calcifica-
despite the large size of the lesions (MARTIN et al. tions (Fig. 4.9c), and/or parietal nodules (Fig. 4.9d)
1998). Reconstructed thin section images can effec- allows a diagnosis of MeT (MARTIN et al. 1998; ITOH
tively show the relationship of the mass to the sur- et al. 1992; ITAI et al. 1982). When these findings are
rounding GI tract (Figs. 4.4-4.8). absent, and when the tumor appears as an unilocular
Delayed enhancement of the tumor wall and septa cystic mass with a thin wall (Fig. 4.9a,b), MeT is no
may be observed in the venous phase. This is a direct longer distinguishable from other cystic masses of

c d

Fig. 4.9a-d. Macrocystic unilocular MCT. a, b Pancreatic phase images: two different patients display thin-walled unilocular
cystic lesions within the pancreatic parenchyma: the characteristics of the mass do not allow for classification. c, d Venous
phase images: two different cases. The presence of calcifications (arrow in c) within the wall of an unilocular cystic mass of
the pancreas, even though it is a nonspecific finding, is suspicious of MCT. Similarly, the demonstration of the thick wall with
nodules (arrowhead in d) leads to the diagnosis of MCT
64 C. Biasiutti et al.

the pancreas, even pseudocyst (JOHNSON et al. 1988; the papillary projections (Fig. 4.10c-f). T2-weighted
FUGAZZOLA et al. 1991; SOYER et al. 1994; BUETOW images (highly sensitive to fluid) are optimal for
et al. 1998; CURRY et al. 2000). In this case, it may be viewing the duct of Wirsung (Fig. 4.10c,d). When
necessary to perform a fine needle aspiration biopsy the mass clearly appears isolated from the duct of
of the lesion. Increasing experience with MDCT Wirsung, thereby excluding the possibility of an
promises that architectural features may be better intraductal tumor, further examination with MRCP
recognized (Fig. 4.10). is superfluous (KoITO et al. 1998; MERA et al. 1999;
DUPAs and LE BORGNE 2000).
Following intravenous contrast administration
4.2.3 (Gd-DTPA), the pancreas can be examined in an
Magnetic Resonance Imaging identical fashion as with CT, i.e., a pancreatic and
venous phase (Figs. 4.11c,d,4.12c,d, 4.13b). Enhance-
Current MR technology provides acquisition times ment produced by contrast medium administration
of sufficiently short duration to allow breathhold makes it much easier to recognize the lesion's cap-
dynamic contrast-enhanced study of the pancreas sule and the septa, as well as any mural excrescence
and the ability to acquire data in 3D (MINAMI et al. or vegetation when compared with the unenhanced
1989; THOENI and BLANKENBERG 1993; KEHAGIAS scan (MERGO et al. 1997; BUETOW et al. 1998;
et al. 2000). The addition of MRCP provides further PROCACCI et al. 2001a). Maximal enhancement of
imaging with the ability to visualize the pancreatic septa is observed in the venous phase (Figs. 4.11d,
duct (KoITO et al. 1998; MERA et al. 1999; ALBERT et 4.12d, 4.13b). Data acquisition in the coronal plane
al. 2000; MEGIBOW et al. 2001a). aids in the recognition of these anatomic elements
The overall MR evaluation consists of a series of (Fig. 4.13b). Obviously, if a 3D sequence is used, the
acquisitions performed prior to and following IV best reconstruction planes can be determined at the
contrast administration. On precontrast Tl-weighted workstation during postprocessing. Unfortunately,
GRE images (optimally performed with fat suppres- the recognition of calcification is not improved with
sion), lesions displaying the macrocystic multilocular IV contrast (MERGO et al. 1997; BUETOW et al. 1998;
pattern (Figs. 4.11, 4.12) appear as rounded, sharply PROCACCI et al. 2001a). 3D postprocessing provides
circumscribed masses mainly located in the body- vascular maps of the peripancreatic vessels, which
tail (MINAMI et al. 1989; ITo et al. 2001). The con- aids in surgical planning.
tent is homogeneous and hypointense (Fig. 4.11b). The macrocystic unilocular pattern as studied by
Thick mucin or foci of hemorrhage may increase MR is also problematic in that septa may go unrec-
the signal (Fig. 4.12b) (MINAMI et al. 1989; Ros et ognized. The lack of septa documentation will result
al. 2000; PROCACCI et al. 2001a; ITO et al. 2001). On in a broad differential diagnosis of the cystic mass
this sequence, the central septa and the calcifications (Fig. 4.14), unless the presence of a thick wall and,
are almost always unrecognizable. Identification of a above all, parietal nodules leads to the suspicion of
thick wall and of peripheral mural nodularity is dif- MCT (Fig. 4.15).
ficult (Fig. 4.13a).
T2 characteristics may be rapidly visualized
by so-called 'turbo' or 'fast spin-echo' techniques, 4.2.4
such as rapid acquisition relaxation enhancement Other Imaging Modalities
(RARE) or half-Fourier single-shot turbo spin-echo
(HASTE). The predominantly fluid content of these 4.2.4.1
masses renders them bright on all T2-weighted Plain Films
images. The presence, characteristics, and distribu-
tion of internal septa are best appreciated with these There is no primary use for plain films in the study
images (Figs. 4.10c-f, 4.11a, 4.12a). Calcifications are of MCT. As with large SCT, a soft-tissue mass can be
not appreciated unless they are large. The intracystic demonstrated, which may displace gas-distended
fluid is slightly hypointense compared with the pure hollow organs (DE LIMA et al. 1999). Calcifications
water signal which might arise from the gallbladder. appear at the periphery of the lesion (Fig. 4.16)
The decrease in signal is directly related to the mucin (FRIEDMAN et al. 1983; BUETOW et al. 1998; SARR et
composition or the presence of hemorrhagic foci al. 2001). It is important to remember that other cystic
(MEGIBOW et al. 2001a) (Fig. 4.12a). The sequence masses, including pseudocyst and cystic endocrine
allows for accurate examination of the septa and tumor, can demonstrate the same calcifications.
Mucinous Cystic Tumors 65

Fig. 4. lOa-f. Macrocystic multilocular MCT. a, b Venous phase images. CT evaluation of a unilocular mass of the pancreatic tail.
In the scan (a) there is a thin septum, which is not visible inferiorly (b). c, d Axial HASTE T2W images: a hyperintense mass is
recognizable, containing a single, thin, hypointense septum (black arrow in c) and thick irregular walls. The duct of Wirsung
appears normal downstream; dilation of some collateral branches is appreciable in the tail. e, f Coronal HASTE T2W images:
the ability to acquire images in a variety of planes leads to the demonstration of multiple thin septa (black arrowheads), lead-
ing the diagnosis towards MCT
66 C. Biasiutti et al.

a
__ ~~ __ a

d ~U~ ."'C'''Ol J. al~;':) a .lCl.l51l;;, '-l\)u.\... JBQ\)\) 11J l.U~ UVUY-lau

pancreas with a thick, sharply marginated wall. Multiple septa


are visible within the lesion. b-d Axial GRE Tl W images. On
the unenhanced image (b), the content of the mass is homoge-
neously hypo intense, and the septa are faintly visible. Follow-
ing intravenous gadolinium, the septa are better seen during
the pancreatic phase (c) . In the portal venous phase (d), wall
enhancement is demonstrated; also, the enhanced internal
septa stand out from the hypointensity of the mucin
Mucinous Cystic Tumors 67

Fig. 4.12a-d. Macrocystic multilocular MCT. a Axial SE T2W


image demonstrates a large, multiloculated lesion in the
pancreatic body-tail. The signal from the mass is variable,
related to the presence of different mucin composition and
hemorrhagic components: on T2W images, mucin (asterisk) is
hyperintense, and hemorrhagic content is responsible for the
decreased signal in the larger portion of the cyst. b Axial SE
II W image at the same level as (a). Demonstration of the dif-
ferent components of the lesion: one (asterisk) is hypointense
compared with the other. A thick, slightly hypointense wall is
appreciable at the periphery of the mass. c, d Axial GRE II W
images in the pancreatic (c) and venous (d) phases following
IV gadolinium. Moderate enhancement of the wall and the
septa are better seen during the venous phase
OIl

Fig. 4.14a-c. Macrocystic unilocular MCT. a Axial GRE T2W


image reveals a unilocular, sharply marginated, cystic mass in
Fig.4.13a,b. Macrocystic multilocular MCT. Coronal GRE II W the pancreatic tail. b, c Axial GRE 3D II W FS images. Unen-
images. a In the precontrast phase, a large, rounded lesion is hanced image (b), the lesion appears homogeneously hypoin-
demonstrated in the pancreatic body. The lesion appears het- tense. In the pancreatic phase following IV gadolinium (c),
erogeneous due to the presence of filling defects within. b In the lesion persists as hypointense. No filling defects or septa
the portal venous phase following IV gadolinium, enhance- are visible within. A specific diagnosis of MCT is impossible
ment of the filling defects and the wall is demonstrated in this case by imaging alone
68 C. Biasiutti et al.

a
a

b b
Fig. 4.15a,b. Macrocystic unilocular MCT. Axial GRE TlW FS
images. a Unenhanced image: a unilocular, slightly hyperintense
mass is present in the pancreatic tail. A papillary proliferation
originates from the nondependent portion of the lesion. b In the
venous phase, the thick wall is enhanced, as well as the papillary
projection: these findings lead to the diagnosis of MCT

Fig. 4.16a-c. Macrocystic multilocular MCT. a Plain film:


incidental finding of thin, curvilinear calcifications delimiting
a right upper quadrant ovoid mass; a second linear calcifica-
tion is oriented transversely (arrow). b, c CT evaluation: portal
venous phase. CT confirms the presence of a cystic lesion of the
pancreatic head with mural calcifications (arrow in b) and the
presence of multiple, thin, calcified septa (arrow in c)
Mucinous Cystic Tumors 69

4.2.4.2 one. In the more advanced malignant forms, how-


Contrast Examination of the Upper Gastrointestinal ever, it is possible to demonstrate encasement of the
Tract peripancreatic vessels (BUETOW et al. 1998). There is
no role for diagnostic angiography today. Improving
The findings obtained in the presence of MCT 3D capabilities from cross-sectional imaging may
(displacement/imprinting of the digestive tract) are obviate the need to obtain angiograms for 'surgical
all nonspecific and analogous to those found with planning'.
other pancreatic tumors. Again, these findings are
incidental during the search for other pathologies. 4.2.4.4
Endoscopic Retrograde Cholangiopancreatography
4.2.4.3
Angiography ERCP is used when the location of the lesion (unci-
nate process) and/or its relationships with the duct
Angiography was used to distinguish MCT (nonvas- ofWirsung raises the suspicion of a collateral branch
cular) from SCT (richly vascularized) (FRIEDMAN IPMT (SHYR et al. 1996). ERCP is able to document
et al. 1983; PINSON et al. 1990; BUETOW et al. 1998). whether there is a communication between the cystic
Obviously, it is not possible with angiography to dis- formation and the duct (Fig. 4.17) (PINSON et al. 1990;
tinguish the multilocular form from the unilocular OBARA et al. 1991; SHIMA et al. 2000).

a b

c d

Fig. 4. 17a-d. Macrocystic unilocular MCT. a A rounded, sharply marginated, hypodense lesion embedded in the pancreatic tail
without septa or calcifications is detected. In the more caudal scan (b), the lesion appears to be adjacent to the duct ofWirsung.
Determination of possible communication is difficult. cHASTE T2W image obtained after the intravenous administration of
secretin in the parasagittal plane confirms the mass near the duct of Wirsung, which has a normal caliber. The presence of
communication is still questionable. d ERCP reveals the normal duct ofWirsung and fails to demonstrate communication with
the mass, excluding the possibility of an intraductal tumor of the collateral ducts
70 C. Biasiutti et al.

4.2.4.5 municating duct with the duct of Wirsung) (KoITO


Endoscopic US et al. 1997; GRESS et al. 2000). Furthermore,EUS aids
in the recognition of slight thickening or parietal
EUS, with superior spatial resolution compared vegetation (Fig. 4.18a-c) aiding in accurate classifi-
with transabdominal US (KoITO et al. 1997), is cation (INUI et al. 1998; BRUGGE 2000).
helpful in classifying those masses when it has not When confronted with a large, unilocular cystic
been possible to make a diagnosis with the exami- mass, EUS can demonstrate thin septa or nodulation,
nations carried out up to that point (KoITO et al. leading the diagnosis in the direction of a possible
1997; ARIYAMA et al. 1998). The most important MCT (INUI et al. 1998; BRUGGE 2000).
contribution of EUS is particularly in the study Last but not least, in nonspecific cases, the pos-
of small cystic lesions of the pancreas (Fig. 4.18), sibility of classification by means of fine-needle
(ARIYAMA et al. 1998). When faced with a long list aspiration (FNA) under endosonographic guidance
of differential diagnostic possibilities of a small, should be pointed out (Fig. 4.18d) (BRUGGE 2000;
cystic, pancreatic mass, EUS can distinguish MCT BRUGGE et al. 2001; GRESS et al. 2000). The malignant
(macro cystic multilocular or unilocular pattern) cells at cytology, the presence of mucin, or a high
from SCT (microcystic appearance) and from the titer of CEA, all contribute to the diagnosis of MCT
collateral branch IPMT (demonstration of the com- (LEWANDROWSKI et al. 1993).

a c

b d

Fig. 4.1Sa-d. Macrocystic unilocular MCT (EUS evaluation). a A unilocular cyst with a thick wall and hyperechoic mural nodules. b,
c Second case, different planes: a cyst with a thick wall and one single, thick, hyperechoic septum (arrow). d Fine-needle aspiration
(FNA) of a unilocular pancreatic cyst. The needle is visible inside the cyst (courtesy of Prof. G. Caletti, University of Bologna)
Mucinous Cystic Tumors 71

4.3 3. The unilocular mass with thick wall demands sur-


Prognostic Evaluation gical intervention when calcifications and/or pari-
etal nodulations are present because these have a
The potential or overtly malignant nature of MCT high likelihood of malignancy. When only a thick
makes surgical resection appropriate when a mass wall is present, however, there could be justifica-
with a macrocystic multilocular appearance is identi- tion enough to avoid intervention and proceed to
fied. Although this approach may lead to the resec- follow-up, as in point 2 (SACHS et al. 1989).
tion of a benign mass such as an oligo cystic SCT, 4. The unilocular mass with thin wall is absolutely
the overwhelming majority will be MCT (JOHNSON nonspecific, and if it is an MCT, it has a very high
et al. 1988; SPERTI et al. 1993; SOYER et al. 1994; possibility of being benign. In this case, follow-up
BUETOW et al. 1998; LUNDSTEDT and DAWISKIBA or further diagnostic investigations, including an
2000; PROCACCI et al. 2001b). Many patients with FNAB, should be carried out (SACHS et al. 1989;
MCT will be older and carry a high surgical risk. CARLSON et al. 1998).
Therefore, it is important to try and glean prognostic
information from the imaging procedure (ISELIN et
al. 1993; LE BORGNE et al. 1999; BAGDASARIAN and
FUHRMAN 2000; PROCACCI et al. 2001b). 4.4
MCT, unilocular or multilocular, appears with Differential Diagnosis
rather variable degrees of wall and septal thickness
and the presence or absence of parietal nodulations The macrocystic multilocular pattern is considered
and/or calcifications. A thick wall, thick septa, parietal typical but not pathognomonic. A mass with these
nodules, and, above all, calcifications are considered to characteristics in an adult woman, especially when
be the best predictors of the malignant nature of MCT it is located in the body-tail, is almost certainly
(ZAMBONI et al. 1999; SHIMA et al. 2000; PROCACCI et MCT. As already said, mural calcifications and thick
al. 2001b). It has been shown that when the three signs septa lead to the prediction of the malignant nature
are present (calcifications, thick wall, and thick septa), of the lesion (PROCACCI et al. 2001 b). Oligo cystic
the probability of a malignant MCT is 95%. When SCT, solid pseudopapillary tumor (cystic variant),
fewer than three signs are present, the probability of cystic endocrine tumor, and necrotic metastases can
malignancy decreases until it almost cancels itself out have an identical appearance (JOHNSON et al. 1988;
when there are no calcifications and septa, and the wall FUGAZZOLA et al. 1991; SOYER et al. 1994; HAMMEL
is thin. Therefore, CT is the primary imaging modal- and BERNADES 1996; BUETOW et al. 1998; CURRY et
ity for these patients because calcifications cannot be al. 2000). In this case, the clinical history and the
visualized with MR. On CT, four classes of lesion can laboratory data are critical for an accurate diagnosis
be distinguished, each requiring a different therapeu- (PINSON et al. 1990).
tic approach (PROCACCI et al. 2001b): Oligocystic SCT, accounting for less than 3% of
1. The macrocystic multilocular mass with thick SCT, has an appearance identical to benign MCT
wall, thick septa, and calcifications has the high- (Fig. 4.19a) (LUNDSTEDT and DAWISKIBA 2000;
est probability of being a malignant MCT. Imme- PROCACCI et al. 2001a). These lesions will never have
diate surgical resection is indicated if possible a thick wall, thick septa, or calcifications. This lesion
(BUETOW et al. 1998). is almost never correctly diagnosed preoperatively.
2. The macro cystic multilocular mass with thin wall Cystic solid pseudopapillary tumor results from pre-
and thin septa, without calcifications is strongly vious necrosis and hemorrhage within the tumor. On
indicative of MCT, but the probability of malig- US, these lesions show a finely hyperechoic appearance.
nancy is low. In this case, surgical resection without On unenhanced CT, the cyst contents may be hyper-
performing an FNAB is advised for young and/or dense. On MR, depending on the degree of necrosis
healthy patients because even though the lesion and the extent of hemorrhage, the signal will appear
at the time of resection may appear benign, most high on II-weighted and low or high on T2-weighted
authorities feel these are premalignant lesions. In sequences (PROCACCI et al. 2001a). Most lesions dis-
older and/or clinically compromised patients, a play a complex alternating pattern combining solid
high -risk operation could be avoided by careful (neoplastic) and fluid components (Fig. 4.19b).
follow-up. If the presence of the mass produces The endocrine cystic tumor also owes its appear-
symptoms from local compression of adjacent ance to previous necrosis and hemorrhage. Similarly
viscera, surgical resection is appropriate. to the solid pseudopapillary tumor, calcification and
72 C. Biasiutti et al.

solid components may be identified (PROCACCI et


al. 2001a). Clinical syndromes related to endocrine
hyperfunction are rare. Cystic endocrine tumors may
be found in association with multi endocrine type
MEN 1 syndrome. A cystic tumor as described above
occurring in a man should raise the clinical suspicion
of a cystic neuroendocrine tumor (Fig. 4.19c).
The pseudocyst, even though rare, can have a
macrocystic multilocular appearance, with a thick
wall and calcifications. Nevertheless, in this case, the
clinical history of the patient (previous acute pancre-
atitis) and/or the presence at the pancreatic level of
the typical signs of chronic pancreatitis (calcifica-
a
tions, dilation of the duct of Wirsung, etc.) help make
the correct diagnosis (PINSON et al. 1990; SPERTI et
al. 1993; PROCACCI et al. 1999; CURRY et al. 2000; Ros
et al. 2000; PROCACCI et al. 2001a; MEGIBOW et al.
2001b).
The macro cystic unilocular pattern, as already
said, is completely nonspecific (HAMMEL and BER-
NADES 1996). In this case, the first problem to arise
is the differential diagnosis between it and a pseudo-
cyst. The clinical history is imperative for the diagno-
sis (MARTIN et al. 1998; KUBA et al. 1998; PROCACCI et
al. 1999). However, when the clinical picture is silent
or nonspecific, the diagnosis of pseudocyst becomes
less likely, and the cystic tumor should be suspected.
If the unilocular lesion has a thick wall, nodulations,
b and calcifications, not only the mucinous nature can
be assumed, but also its malignancy (PROCACCI et al.
2001 b). Aspiration of the contents of the cyst should
help discriminate between pseudocyst (high amy-
lase) and cystic neoplasm (high mucin level).
It should be pointed out that if an unilocular
cystic mass has been diagnosed with only one of the
previously described imaging modalities, it is always
better to perform more than one single diagnostic
technique in order to look more carefully for signs
(small septa, calcifications) that can contribute to
making a more specific diagnosis.

c References

Fig. 4.19a-c. Macrocystic unilocular MCT-like lesions. Differ- Albert J, Schilling D, Breer H, Jungius KP, Riemann JF, Adamek
ent cases. a Oligocystic SCT, un enhanced CT evaluation. The HE (2000) Mucinous cystadenomas and intraductal papil-
lesion displays a thin wall and small calcification (arrow). lary mucinous tumors of the pancreas in magnetic reso-
b Solid pseudo-papillary tumor, portal venous phase CT nance cholangiopancreatography. Endoscopy 32:472-476
evaluation. In the axial image, both the hypodense (fluid) and Algard M, Ponsot P, Hautefeuille M, Menu Y, Fekete F, Paolaggi
hyperdense (solid) components are appreciable. c Endocrine JA (1986) Pancreatic cystadenoma: diagnostic value of
tumor, portal venous phase CT evaluation. The central necro- ultrasonics and x-ray computed tomography. Gastroen-
sis is responsible for the cystic appearance, mimicking a MCT. terol Clin Bioi 10:23-28
A thin septum along with a small calcification (black arrow) Ariyama J, Suyama M, Satoh K, Wakabayashi K (1998) Endo-
are visible within the lesion scopic ultrasound and intraductal ultrasound in the
Mucinous Cystic Tumors 73

diagnosis of small pancreatic tumors. Abdom Imaging Ito K, Koike S, Matsunaga N (2001) MR imaging of pancreatic
23:380-386 diseases. Eur J Radiol 38:78-93
Bagdasarian RW, Fuhrman GM (2000) Cystic tumors of the Itoh S, Ishiguchi T, Ishigaki T, Sakuma S, Maruyama K, Senda
pancreas: an update. Curr Surg 57:522-528 K (1992) Mucin-producing pancreatic tumor: CT findings
Bastid C, Sahel J, Sastre B, Schurgers P, Sarles H (1989) Muci- and histopathologic correlation. Radiology 183:81-86
nous cystadenocarcinoma of the pancreas. Ultrasono- Johnson CD, Stephens DH, Charboneau JW, Carpenter HA,
graphic findings in 5 cases. Acta Radiol 30:45-47 Welch TJ (1988) Cystic pancreatic tumors: CT and sono-
Brugge WR (2000) The role of EUS in the diagnosis of cystic graphic assessment. AJR 151:1133-1338
lesions of the pancreas. Gastrointest Endosc 52:S18-S22 Kehagias D, Smyrniotis V, Gouliamos A, Vlahos L (2000) Cystic
Brugge WR, Saltzman JR, Scheiman JM, Wallace MB, Jowell pancreatic neoplasms: computed tomography and magnetic
PS, Pochapin MB, Stevens PD, Gress FG, Lewandrowski K resonance imaging findings. Int J Pancreatol 28:223-230
(2001) Diagnosis of cystic neoplasms of the pancreas by Koito K, Namieno T, Nagakawa T, Shyonai T, Hirokawa N, Morita
EUS: the report of the cooperative pancreatic cyst study. K (1997) Solitary cystic tumor of the pancreas: EUS-patho-
Gastrointest Endosc 53:AB71 logic correlation. Gastrointest Endosc 45:268-276
Buetow PC, Rao P, Thompson LD (1998) From the archives of Koito K, Namieno T, Ichimura T, Yama N, Hareyama M, Morita
the AFIP. Mucinous cystic neoplasms of the pancreas: radio- K, Nishi M (1998) Mucin-producing pancreatic tumors:
logic-pathologic correlation. Radiographies 18:433-449 comparison of MR cholangiopancreatography with endo-
Carlson SK, Johnson CD, Brandt KR, Batts KP, Salomao DR scopic retrograde cholangiopancreatography. Radiology
(1998) Pancreatic cystic neoplasms: the role and sensitivity of 208:231-237
needle aspiration and biopsy. Abdom Imaging 23:387-393 Kuba H, Yamaguchi K, Shimizu S, Yokohata K, Sugitani A,
Compagno J, Oertel JE (1978) Mucinous cystic neoplasms of Chijiiwa K, Tanaka M (1998) Chronic asymptomatic pseu-
the pancreas with overt and latent malignancy (cystadeno- docyst with sludge aggregates masquerading as mucinous
carcinoma and cystadenoma). A clinicopathologic study of cystic neoplasm of the pancreas. J GastroenteroI33:766-769
41 cases. Am J Clin Pathol 69:573-580 Le Borgne J,de Calan L,Partensky C (1999) Cystadenomas and
Curry CA, Eng J, Horton KM, Urban B, Siegelman S, Kuszyk cystadenocarcinomas of the pancreas: a multiinstitutional
BS, Fishman EK (2000) CT of primary cystic pancreatic retrospective study of 398 cases. French surgical associa-
neoplasms: can CT be used for patient triage and treat- tion. Ann Surg 230:152-161
ment? AJR 175:99-103 Lewandrowski KB, Southern JF, Pins MR, Compton CC,
De Lima JE Jr, Javitt MC, Mathur SC (1999) Mucinous cystic Warshaw AL (1993) Cyst fluid analysis in the differential
neoplasm of the pancreas. Radiographies 19:807-811 diagnosis of pancreatic cysts. A comparison of pseudo-
Dupas B, Le Borgne J (2000) MRI-Cholangiopancreatography cysts, serous cystadenomas, mucinous cystic neoplasms,
characteristics of cystic pancreatic tumors. Ann Chir 125: and mucinous cystadenocarcinoma. Ann Surg 217:41-47
571-577 Lundstedt C, Dawiskiba S (2000) Serous and mucinous
Fernandez-del Castillo C, Warshaw AL (1995) Cystic tumors of cystadenoma/cystadenocarcinoma of the pancreas. Abdom
the pancreas. Surg Clin North Am 75:1001-1016 Imaging 25:201-206
Friedman AC, Lichtenstein JE, Dachman AH (1983) Cystic Martin I, Hammond P, Scott J, Redhead D, Carter DC, Garden
neoplasms of the pancreas. Radiological-pathological cor- OJ (1998) Cystic tumours of the pancreas. Br J Surg 85:
relation. Radiology 149:45-50 1484-1486
Fugazzola C, Procacci C, Bergamo Andreis lA, Iacono C, Por- Megibow AJ, Lavelle MT, Rofsky NM (2001a) MR imaging of
tuese A, Dompieri P, Laveneziana S, Zampieri PG, Jannucci the pancreas. Surg Clin North Am 81:307-320
A, Serio Get al (1991) Cystic tumors of the pancreas: evalu- Megibow AJ, Lavelle MT, Rofsky NM (2001b) Cystic tumors
ation by ultrasonography and computed tomography. Gas- of the pancreas: the radiologist. Surg Clin North Am 81:
trointest Radiol 16:53-61 489-495
Gress F, Gottlieb K, Cummings 0, Sherman S, Lehman G Mera K, Tajiri H, Muto M, Ohtsu A, Furuse J, Maru Y, Kinoshita
(2000) Endoscopic ultrasound characteristics of mucinous T, Ryu M, Nawano S, Murakami K, Moriyama N, Yoshida S
cystic neoplasms of the pancreas. Am J Gastroenterol 95: (1999) Clinical significance of magnetic resonance chol-
961-965 angiopancreatography for the diagnosis of cystic tumor
Hammel P, Bernades P (1996) Les lesions kystiques du pan- of the pancreas compared with endoscopic retrograde
creas de diagnostic difficile. Presse Med 25:1794-1800 cholangiopancreatography and computed tomography.
Inui K, Nakazawa S, Yoshino J, Yamachika H, Kanemaki Jpn J Clin Oncol 29:294-298
M, Wakabayashi T, Okushima K, Taki N, Nakamura Y, Mergo PJ, Heimberger TK, Buetow PC, Heimberger RC, Ros PR
Takashima T, Hattori T, Miyoshi H (1998) Mucin-produc- (1997) Pancreatic neoplasms: MR imaging and pathologic
ing tumor of the pancreas-intraluminal ultrasonography. correlation. Radiographics 17:281-301
Hepatogastroenterology 45:1996-2000 Minami M, Itai Y, Ohtomo K, Yoshida H, Yoshikawa K, Iio M
Iselin CE, Meyer P, Hauser H, Kurt AM, Vermeulen JM, Rohner (1989) Cystic neoplasms of the pancreas: comparison of
A. (1993) Computed tomography and fine-needle aspira- MR imaging with CT. Radiology 171:53-56
tion cytology for preoperative evaluation of cystic tumours Obara T, Maguchi H, Saitoh Y, Ura H, Koike Y, Kitazawa S,
of the pancreas. Br J Surg 80:1166-1169 Namiki M (1991) Mucin-producing tumor of the pancreas:
Itai Y, Ohtomo K (1996) Cystic tumours of the pancreas. Eur a unique clinical entity. Am J Gastroenterol 86:1619-1625
Radiol 6:844-850 Pinson CW, Munson JL, Deveney CW (1990) Endoscopic
Itai Y, Moss AA, Ohtomo K (1982) Computed tomography of retrograde cholangiopancreatography in the preoperative
cystadenoma and cystadenocarcinoma of the pancreas. diagnosis of pancreatic neoplasms associated with cysts.
Radiology 145:419-425 Am J Surg 159:510-513
74 C. Biasiutti et al.

Procacci C, Biasiutti C, Carbognin G, Accordini S, Bicego E, Shima Y, Mori M, Takakura N, Kimura T, Yagi T, Tanaka N
Guarise A, Spoto E, Bergamo Andreis lA, De Marco R, (2000) Diagnosis and management of cystic pancreatic
Megibow AJ (1999) Characterization of cystic tumors of tumours with mucin production. Br J Surg 87:1041-1047
the pancreas: CT accuracy. J Comput Assist Tomogr 23: Shyr YM, Su CR, Tsay SH, Lui WY (1996) Mucin-producing
906-912 neoplasms of the pancreas. Intraductal papillary and muci-
Procacci C, Biasiutti C, Carbognin G, Capelli P, El-Dalati G, nous cystic neoplasms. Ann Surg 223:141-146
Falconi M, Misiani G, Ghirardi C, Zamboni G (2001a) Pan- Soyer P, Rabenandrasana A, Van Beers B, Barge 1, Sibert A,
creatic neoplasms and tumor-like conditions. Eur Radiol Laissy JP, Achour E, Levesque M (1994) Cystic tumors of
11:S167-S192 the pancreas: dynamic CT studies. J Comput Assist Tomogr
Procacci C, Carbognin G, Accordini S, Biasiutti C, Guarise A, 18:420-426
Lombardo F, Ghirardi C, Graziani R, Pagnotta N, De Marco Sperti C, Cappellazzo F, Pasquali C, Militello C, Catalini S,
R (2001b) CT features of malignant mucinous cystic Bonadimani B, Pedrazzoli S (1993) Cystic neoplasms of
tumors of the pancreas. Eur Radiol 11: 1626-1630 the pancreas: problems in differential diagnosis. Am Surg
Ros LH, Helmberger T, Ros PR (2000) Cystic pancreatic 59:740-745
lesions. Eur Radiol 1O:S209-S217 Stephens DH (1997) CT of pancreatic neoplasms, part II. The
Sachs JR, Deren JJ, Sohn M, Nusbaum M (1989) Mucinous unusual tumors. Curr Prob Diagn RadioI26:81-106
cystadenoma: pitfalls of differential diagnosis. Am J Gas- Thoeni RF, Blankenberg F (1993) Pancreatic imaging. Com-
troenteroI84:811-816 puted tomography and magnetic resonance imaging.
Sarr MG, Carpenter HA, Prabhakar LP, Orchard TF, Hughes S, Radiol Clin North Am 31:1085-1113
van Heerden JA, DiMagno EP (2000) Clinical and patho- Warshaw AL, Compton CC, Lewandrowski K, Cardenosa G,
logic correlation of 84 mucinous cystic neoplasms of the Mueller PR. (1990) Cystic tumors of the pancreas. New
pancreas: can one reliably differentiate benign from malig- clinical, radiologic, and pathologic observations in 67
nant (or premalignant) neoplasms? Ann Surg 231:205-212 patients. Ann Surg 212:432-443
Sarr MG, Kendrick ML, Nagorney DM, Thompson GB, Farley Wilentz RE, Albores-Saavedra 1, Hruban RH (2000) Mucinous
DR, Farnell MB (2001) Cystic neoplasms of the pancreas: cystic neoplasms of the pancreas. Semin Diagn Pathol 17:
benign to malignant epithelial neoplasms. Surg Clin North 31-42
Am 81:497-509 Wolfman NT, Ramquist NA, Karstaedt N, Hopkins MB (1982)
Scott J, Martin I, Redhead D, Hammond P, Garden OJ (2000) Cystic neoplasms of the pancreas: CT and sonography. AJR
Mucinous cystic neoplasms of the pancreas: imaging fea- 138:37-41
tures and diagnostic difficulties. Clin Radiol 55:187-192 Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini G,
Shamsi K, Deckers F, De Shepper A, Hauben E, Van Marek E Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM, Kloppel
(1993) Mucinous cystadenocarcinoma of the pancreas with G (1999) Mucinous cystic tumors of the pancreas: clinico-
liver metastases. An unusual presentation of a rare tumor. pathological features, prognosis, and relationship to other
Ann RadioI36:328-331 mucinous cystic tumors. Am J Surg PathoI23:410-422
Intraductal Cystic Tumors
5 Clinical Manifestations and Therapeutic Management
R. SALVIA, M. FALCONI, L. CASETTI, W. MANTOVANI, I. FRIGERIO, and P. PEDERZOLI

CONTENTS The tumor originates from the main duct or


peripheral ducts and can involve the whole pancre-
5.1 Introduction 77 atic gland, having different histological grades within
5.2 Clinical Data 78 the same tumor.
5.2.1 Sex and Age 78
5.2.2 Habits 78 The disease originates from the epithelium of the
5.2.3 Pain and Previous Diagnosis pancreatic ducts and can evolve through all stages
of Acute Pancreatitis 78 from slight dysplasia to carcinoma. Currently, most
5.2.4 Weight Loss 79 researchers agree that their evolution towards the
5.2.5 Asthenia 79 carcinoma stage is slow, but probably inexorable
5.2.6 Diabetes 79
5.2.7 Jaundice 79 (BRAT et al. 1998; FURUKAWA et al. 1994; KLOPPEL
5.2.8 Clinical Evaluation Summary 80 1996; LONGNECKER 1998; OBARA et al. 1993; RIVERA
5.3 Laboratory Findings 80 et al. 1997; SOLCIA 1997; YAMAGUCHI et al. 1996;
5.4 Clinical Aspects in Differential Diagnosis 80 Z'GRAGGEN et al. 1997).
5.4.1 Chronic Pancreatitis 80 At the beginning, the major clinical problem
5.4.2 Serous Cystadenoma 80
5.4.3 Mucinous Cystic Tumor 81 was to recognize IPMT and to differentiate it from
5.5 From Assessment to Management 81 chronic pancreatitis (CP). The majority of undiag-
5.5.1 Surgical Therapy 82 nosed IPMTs are, in fact, wrongly interpreted as CPo
5.5.2 Follow-Up 82 Increased awareness of these tumors has decreased
References 83 the number of incorrect diagnoses. Nowadays, pre-
operative recognition of the histological grading of
these tumors is desirable (YAMAGUCHI et al. 1996).
The need for this is related to a series of consid-
5.1 erations concerning the patients and the disease:
Introduction the only therapeutic option considered in the lit-
erature for the treatment of these tumors is partial
Intraductal papillary mucinous tumors (IPMTs) of or total surgical resection of the pancreatic gland
the pancreas are a relatively new entity among cystic (CUILLERIER et al. 1998; PARTENSKY et al. 1996;
tumors. Described for the first time in 1982 as neo- RIVERA et al. 1997; SIECH et al. 1999; SUGIYAMA
plasms with mucin hyperproduction, dilatation of the and ATOM! 1998; TRAVERSO 1998), but this option
duct ofWirsung, and protruding papilla (the Ohhashi applies to patients who are generally elderly (65-70
triad) (OHHASHI 1982), they have undergone a true years old) with multiple medical problems. However,
epidemiological explosion in recent years (AZAR et patients with a malignant tumor certainly benefit
al. 1996; FALCONI et al. 2001; NAVARRO et al. 1999; from partial or total pancreatic resection. There is
OHHASHI 1982; SHIMA et al. 2000; SUGIYAMA and little justification for the risks and costs of surgery in
ATOMI 1999; TAOUL! et al. 2000; TRAVERSO 1998). high-risk patients harboring a benign tumor, which
could simply be followed up.
Collaboration between the clinician and radiolo-
gist, therefore, focuses on two aspects: diagnosis of
R. SALVIA, MD; M. FALCONI, MD; 1. CASE TTl, MD;
W. MANTOVANI, MD; 1. FRIGERIO, MD; P. PEDERZOLI, MD the disease and evaluation of the clinical and radio-
Surgical-Gastroenterological Department, Hospital 'G .B. Rossi', logical data that might be helpful in the assessment of
University of Verona, 37124 Verona, Italy the stage of the disease before surgery.
78 R. Salvia et al.

S.2 of patients suffering from IPMT reported alcohol


Clinical Data abuse which, according to the author, would also be
a predictive factor of malignancy (TRAVERSO 1998).
The parameters considered in the literature for the On the contrary, alcohol abuse is almost constant
diagnosis of IPMT that can discriminate between the in patients with CPo Smoking does not seem to be
benign or malignant neoplasia are (Table 5.1): demo- a discriminating factor. In personal experience,
graphic data, daily habits, clinical history, symptoms, 42% of patients suffering from IPMT were smokers
laboratory findings, and, of course, radiological find- (FALCONI et al. 2001).
ings (CUILLERIER et al. 1998; SUGIYAMA and ATOMI
1998; TRAVERSO 1998; YAMAGUCHI et al. 1996,2000).
5.2.3
Pain and Previous Diagnosis
5.2.1 of Acute Pancreatitis
Sex and Age
Recurrent pain is certainly the most common of
The patients suffering from this tumor are in their symptoms and is described by many authors as
60s and 70s; there is an equal distribution of men 'pancreatitis-like' (AZAR et al. 1996; NAVARRO et al.
and women (YAMAGUCHI et al. 2000; YAMAO et al. 1999; WARSHAW 1991). The painful symptomatology
2000). In personal experience (Table 5.1), the aver- is generally referred to as continuous, related to meals,
age age of IPMT patients was 64 years, with an equal localized in the upper area of the abdomen, radiating
distribution between the sexes (M:F=l:1) (FALCONI to the back. Well-investigated symptoms like those
et al. 2001). This is in distinction to patients with CP, described above suffice to focus clinical suspicion on
where there is a marked male predominance, and the pancreas. The percentage of patients complaining
the average age at diagnosis is 42 years (CAVALLINI of pain is higher when those with a previous history
et al. 1998). of acute pancreatitis are also taken into consideration.
The pain from IPMT and from acute pancreatitis have
the same origin: the hyperproduction of mucin is
5.2.2 responsible for hindering the outflow of pancreatic
Habits juices (WARSHAW 1991). An episode of acute pancre-
atitis in the case history can make it more difficult to
Evaluation of the alcohol and tobacco intake is distinguish an IPMT of the peripheral ducts from a
important in terms of the differential diagnosis in pseudocyst. However, in personal experience, an epi-
patients with CPo Traverso points out how only 12% sode of acute pancreatitis which was severe enough

Table 5.1. Main demographic and clinical characteristics of 88 patients suffering from surgically resected intraductal papillary
mucinous tumors (IPMTs; personal series) (IPMA/B, intraductal papillary mucinous adenoma and borderline; IPMC/IC, intra-
ductal papillary mucinous carcinoma and invasive carcinoma; AP, acute pancreatitis; CP, chronic pancreatitis)
Characteristic Total IPMA/B (n=47) IPMC/IC (n=41) p
Sex (M/F) 47/41 28119 19122 NS
Average age (years) 62.7 62.4 63.0 NS
Average alcohol intake (g/day) 38 35.4 40.6 NS
Smokers (no. of patients) 41 (46.6%) 23 18 NS
Symptoms:"
Recurrence of painful attacks 58 (66%) 34 (72.3) 24 (58.5) NS
Weight loss 37 (42%) 18 (38.3) 19 (47.5) NS
Asthenia 14 (16%) 3 (6.4) 11 (26.8) 0.02
Fever 5 (5.6%) o (0) 5 (12.2) 0.02
Onset or worsening of diabetes 10 (11.4%) 1 (2.1) 9 (22) 0.003
Jaundice 11 (12.5%) 1 (2.1) 10 (24.4) 0.002
Incidental diagnosis 14 (16%) 9 (19) 5 (12.2) NS
Previous diagnosis of AP 17 (19%) 11 (23.4) 6 (14.6) NS
Previous diagnosis of CP 8 (9%) 6 (12.8) 2 (5) NS
Previous diagnosis of pseudocyst 6 (6.8%) 4 (8.5) 2 (5) NS

"Patient with more symptoms


Clinical Manifestations and Therapeutic Management 79

to potentially develop a pseudocyst occurred in less 5.2.6


than 2% of all patients with IPMTs observed. Diabetes

The sudden onset of diabetes almost always leads


5.2.4 to the suspicion of ductal adenocarcinoma. Some
Weight Loss 11 % of patients suffering from IPMT have diabetes,
almost all in the advanced stages of the disease (AZAR
Another frequent symptom is weight loss (AZAR et al. et al. 1996; FALCONI et al. 2001; KIMURA et al. 1996;
1996; RICKAERT et al. 1991), which is found in 42% of KOBARI et al. 1999; LOFTUS et al. 1996; NAVARRO et
patients in a personal series. Weight loss might have al. 1999; PARTENSKY et al. 1996; PAYE et al. 1999;
two different physiopathological mechanisms related RICKAERT et al. 1991; SUGIYAMA and ATOMI 1998;
to the stage of the disease. In the early phases, the TAOULI et al. 2000). In personal experience, too, the
hyperproduction of mucin obstructs normal pancre- recent onset of diabetes or its worsening within a year
atic secretion, causing the pain related to meals. Thus, from diagnosis of an IPMT more frequently occurred
just as with CP, the patients stop eating in order to with advanced tumors (p<0.005). Other authors do
avoid pain, with subsequent weight loss (CAVALLINI et not confirm this correlation between diabetes and the
al. 1996; WARSHAW 1991). In the advanced stage of the advanced stage of the disease (TRAVERSO 1998). The
disease, the weight loss is more likely due to the produc- symptom, when present, therefore has a double value:
tion of neoplastic factors responsible for cachexia. suspicion of the neoplasm and its malignancy.
Weight loss is common to all neoplastic patholo-
gies and should therefore be considered nonspecific.
Persistent weight loss arouses the suspicion of the 5.2.7
malignancy of the tumor. Jaundice

Jaundice, like diabetes, plays an important role in


5.2.5 being a typical symptom of pancreatic head dis-
Asthenia eases. The frequency of this symptom varies from
5% to 50% of IPMTs (Table 5.2) in the literature
Asthenia is usually associated with weight loss, but (CUILLERIER et al. 1998; ITAI et al. 1987; ITOH et al.
it is difficult to interpret, as it is subject to the char- 1992; KIMURA et al. 1996; OBARA et al. 1991; OHTA
acter and mood of the patient. In fact, the majority 1992; PAYE et al. 1999; RAIJMAN et al. 1994; SUGIYAMA
of authors do not describe the symptom. In personal and ATOMI 1998; UEHARA et al. 1994; YAMAGUCHI
experience, it was present in 16% of patients. and TANAKA 1991). Jaundice is a sign of the tumor
It is interesting to point out that this symptom in its advanced stages (Table 5.1), as confirmed by
was more frequent in those patients with advanced many authors (ITAI et al. 1987; MILCHGRUB et al.
disease (p<0.05). 1992; TRAVERSO 1998).

Table 5.2. Symptoms reported by the principal authors, review of the literature

Publication No. of Pancreatitis Abdominal Weight Diabetes Jaundice Other


cases (% ) pain (%) loss (%) (%) (% ) (% )

TRAVERSO (1998) 33 75 82 36 27 9
RICKAERT et al. (1991) 8 60 87 50 25
LOFTUS et al. (1996) 14 15 73 27
KIMURA et al. (1996) 222 41 52 11 19 18
CUILLERIER et al. (1998) 41 24 7 7 14
PARTENSKY et al. (1996) 54 12 36 24 18 9 2
SUGIYAMA and ATOMI (1998) 41 51 44 7 22 10
PAYE et al. (1999) 41 19 34 37 20 5 7
FALCONI et al. (2001) 63 35 66 42 11 12 16
NAVARRO et al. (1999) 111 29 11 2
SIECH et al. (1999) 8 33 100
TAOUL! et al. (2000) 36 56 41 19
AZAR et al. (1996) 32 42 44 50 37 6
KOBARI et al. (1999) 30 100 54
80 R. Salvia et al.

5.2.8 et al. 1996). In other series, the M:F ratio is, however,
Clinical Evaluation Summary only slightly higher than one (YAMAGUCHI et al. 2000;
YAMAO et al. 2000). Patients with CP (mean age 42
In conclusion, an incidental diagnosis of IPMT occurs years) are about 20 years younger on average than
in 30%-35% of patients, the majority of whom are those with IPMT (mean age 64 years) (CAVALLINI et
symptomatic (PARTENSKY et al. 1996; CUILLERIER et al. 1996; FALCONI et al. 2001). A protracted clinical
al. 1998; PAYE et al. 1999; SHIMA et al. 2000). These history could indicate an inflammatory pathology,
symptoms, characterized by recurrent abdominal pain but this is only a hypothesis, given that the natural
correlated to meals, weight loss, diabetes, jaundice, history of IPMT is not yet completely known (BARBE
previous diagnosis of acute pancreatitis (Table 5.2), et al. 1997; OBARA et al. 1993).
lead to the suspicion of pancreatic pathology. Demo- Lastly, there are symptoms that do not lead to any
graphic data (sex, age) and lifestyle habits may con- kind of discrimination, like nicotine abuse, abdomi-
tribute to the differential diagnosis between IPMT nal pain correlated to meals, the onset of diabetes,
and chronic pancreatitis, while the presence of jaun- and obstructive jaundice, as they are present in both
dice and diabetes, with the diagnosis already made, diseases.
are indications of the malignancy of the lesion.

5.4.2
Serous Cystadenoma
5.3
Laboratory Findings The differential diagnosis between serous cyst-
adenoma and IPMT is difficult and particularly
The laboratory does not contribute in any way to
the diagnosis of IPMT. No blood tests, even tumoral
markers, are specific for these tumors (ALONSO
Table 5.3. Principal laboratory data reported in the two
CASADO et al. 2000; AZAR et al. 1996; OBARA et al. groups of benign and malignant IPMTs (mean values), per-
1993; UEHARA et al. 1997). sonal series
Once the diagnosis of IPMT has been obtained,
Parameter Benign Malignant p
elevated blood glucose, alkaline phosphatase (ALP), (n=47) (n=41)
alanine aminotransferase (ALT), and gamma-glu-
Glycemia (mg/dl) 101.1 121 0.003
tamyl transferase (GGT) are more common with
Albuminemia (gil) 42 40.6 NS
malignant tumors (Tables 5.3,5.4). Total bilirubin (mg/dl) 0.7 1.2 NS
Direct bilirubin (mg/dl) 0.3 0.7 NS
AST (U/I) aspartate aminotrans- 23.6 30.2 NS
ferase
ALT (U/I) alanine aminotrans- 25.7 61.3 0.004
5.4 ferase
Clinical Aspects in Differential Diagnosis LDH (UIl) lactate dehydrogenase 141 l31.3 NS
ALP (U/I) alkaline phosphatase 76.6 171.5 0.006
The problems in the differential diagnosis of IPMT GGT (U/I) gamma-glutamyl 27.2 88.9 0.007
vary according to the tumor's site of growth. IPMTs transferase
Serous amylase (UIl) 242.8 122.3 NS
of the main duct can, in fact, simulate CP, while neo-
Urinary amylase (U/I) 994 473 NS
plasms involving the secondary ducts, 'branch side C-reactive protein (mg/I) 9.8 9.3 NS
IPMTs', should be differentiated from cystic tumors
(FUKUKURA et al. 2000).
Table 5.4. Principal neoplastic markers reported in the two
groups of benign and malignant IPMTs (mean values), per-
sonal series
5.4.1
Chronic Pancreatitis Marker Benign Malignant p
(n=47) (n=41)

In the differential diagnosis between IPMT and CP, CEA (ng/ml) carcinoembryonic antigen 4.8 3.4 NS
some demographic and clinical data can be helpful. Ca 19-9 (U/ml) 23.9 145.1 NS
In personal experience, the vast majority of patients Ca 15.3 (U/ml) 19 16.3 NS
Ca 125 (U/ml) 9.7 12.8 NS
with CP are male, with a M:F ratio of 6:1 (CAVALLINI
Clinical Manifestations and Therapeutic Management 81

important since the former is almost always benign. 5.5


In fact, malignancy has been described in only a few From Assessment to Management
isolated cases (ABE et al. 1998; KAMEl et al. 1991;
WIDMAIER et al. 1996; YOSHIMI et al. 1992). Patients Once the diagnosis of IPMT has been made with the
with a histological diagnosis of serous cystadenoma, contribution of imaging, the clinician will need fur-
when asymptomatic, can be followed up without ther information before going on to therapy.
surgical intervention. In relation to the site, several authors have sug-
Serous cystadenoma is definitely more common in gested classifying these lesions into two groups:
women (F:M=6.7: 1) with an average age of 51.8 years, IPMTs of the main duct and IPMTs of the peripheral
10 years younger than those with IPMT. The tumor ducts (FURUKAWA et al. 1994; KOBARI et al. 1999;
is mainly located in the head of the pancreas. In NAKAGOHRI et al. 1999; TERRIS et al. 2000).
personal experience, about 45% of serous cystad- All the authors agree on the appropriateness of sur-
enomas were located within the pancreatic head gery( assumingaresectablesituation,see Section5.5.1)
and neck, 27% in the body, and 28% in the tail. The for main duct tumors, due to the high malignancy
demographic characteristics, the case history, and the rate of this group.
habits do not lead to differentiation between the two On the other hand, the therapeutic choice for
types of tumor. The presence of symptoms, mainly branch duct IPMTs is more debatable. The literature,
jaundice, diabetes, and 'pancreatitis-like' pain, may like personal experience, confirms that tumors aris-
indicate IPMT since almost all serous cystadenomas ing from the peripheral ducts are less aggressive. It
(75%) were discovered incidentally, due to their lack is assumed that branch duct IPMTs are a different
of symptoms. This hypothesis is confirmed in the entity, or that diagnosis was made earlier in the
literature (GRIESHOP et al. 1994), while the behavior tumor group originating in the periphery. This latter
of IPMT, with only 16% incidental diagnosis, can be hypothesis, however, would contrast with the finding
considered the opposite. that IPMTs of the peripheral ducts are less symptom-
atic and that their diagnosis is more often incidental
and therefore late.
5.4.3 The parameters recently suggested in the evalua-
Mucinous Cystic Tumor tion of these neoplasms are the dimensions, the pres-
ence or absence of papillary growth, and the thick-
The differences in the clinical presentation between ness of the walls of the cystic lesion (LIM et al. 2001;
IPMT and mucinous cystic tumor are less important SILAS et aI. 2001; TAOUL! et al. 2000; WAKABAYASHI
because, due to the potential malignancy of these et al. 2001).
neoplasms, surgical treatment is indicated in both As can be seen in Table 5.5, in our experience, the
cases. aggressive behavior of these neoplasms was justified
Mucinous cystic tumor occurs almost exclusively by 31% (9/29) of peripheral IPMTs being identified
in women of around 45 years of age. The average age as carcinomas at the final histological examination,
is higher when the neoplasm takes on malignant 17.2% (5/29) of which showed signs of invasion. The
characteristics. The topography of the neoplasm can finding of intraductal carcinomas among the branch
be useful for the differential diagnosis since IPMT is side IPMTs has also been reported by some authors
frequently located in the uncinate process, while the (KOBARI et al. 1999, NAKAGOHRI et al. 1999), while
mucinous cystic tumor more commonly involves the being excluded by others (TERRIS et al. 2000).
body-tail. In some series, there is a 93% incidence of It is, in any case, right to point out that invasive
tumors in the tail (ZAMBONI et al. 1999). carcinomas are always characterized by thick walls
Moreover, it is necessary to point out that and endoluminal papillary growth, while only carci-
IPMT is almost always symptomatic, and in those
cases involving the main duct, it can simulate CP,
Table 5.5. Anatomopathological findings in 88 resected IPMTs,
while the mucinous cystic tumor is almost always distinguished according to site of origin, personal series
asymptomatic. It is obvious that all of this clinical
information is helpful in the diagnosis, which is Tumor Central Peripheral
based essentially on imaging findings. It is there- Adenoma (20) 5 (25%) 15 (75%)
fore essential that, at the time of investigation, the Borderline (27) 22 (81.5%) 5 (18.5%)
radiologist is aware of all appropriate history and Noninvasive carcinoma (11) 7 (63.6%) 4 (36.4%)
Invasive carcinoma (30) 25 (83.3%) 5 (16.7%)
clinical information.
82 R. Salvia et al.

noma in situ can have small dimensions, thin walls, noma, unlike patients with muconodular infiltration,
and no endoluminal vegetation. However, despite the who have a considerably better prognosis.
histological diagnosis, the behavior of the tumor may Once the resectability has been assessed, the clini-
be quite different. For this reason, despite the differ- cian needs to know the site of the neoplasm in order
ent opinions, in clinical practice, a peripheral lesion to decide on the type of surgical resection. Again, from
with a diameter of less than 3 cm, thin walls, and no personal data (Table 5.6) and the literature, we see
papillary growth in an elderly, asymptomatic patient that 70% of IPMTs are located in the head of the pan-
can be followed up (KOBARI et al. 1999; NAKAGOHRI creas (SHIMA et al. 2000; SUGIYAMA and ATOMI 1998;
et al. 1999; TAOULI et al. 2000; TERRIS et al. 2000). TAOULI et al. 2000). In personal experience, no IPMT
was found in the body-tail, while this site is indicated
in 0% to 14.3% of peripheral IPMTs in the literature
5.5.1 (NAKAGOHRI et al. 1999; TERRIS et al. 2000).
Surgical Therapy
Table 5.6. Anatomical location and site of origin of 88 resected
IPMTs, personal series
Before performing surgery, it is necessary to consider
operability, which is different to resectability. In fact, Part of pancreas Central Peripheral
operability indicates whether or not it is possible to Head (60) (68.2%) 33 (55%) 27 (45%)
carry out surgery in terms of the patient's general Neck (11) (12.5%) 9 (82%) 2 (18%)
clinical condition. When these conditions are partic- Body-tail (14) (15.9%) 14 (100%) o (0)
Diffuse (3) (3.4%) 3 (100%) 0(0)
ularly poor, surgical resection may not be advisable.
Operability criteria are made strictly on the basis of
a clinical evaluation. Indications for surgery can also arise from the
The clinician will obviously ask the radiologist for occurrence of complications, as in those patients
information on resectability. For IPMTs, as for ductal considered nonresectable but symptomatic of
adenocarcinomas, at the more common cephalic obstructive jaundice or obstruction of the alimentary
location, encasement of the vascular structures adja- tract. In these cases, the surgeon is forced to oper-
cent to the gland, in particular the splenomesenteric- ate and perform a biliodigestive and/or alimentary
portal confluence, but also the superior mesenteric bypass. This is why it is necessary to know radiologi-
artery and the celiac trunk, is a contraindication of cally if there is any obstruction of the main biliary
resectability for tumors in the head of the pancreas. duct and/or alimentary tract.
Tumors located within the body-tail sites can be Faced with a resectable lesion, the best interven-
resected even when infiltration of the splenic artery tion is pancreoduodenectomy for cephalic IPMTs
and vein has occurred, since the spleno-pancreatec- and splenopancreatectomy for those with a body-tail
tomy involves the tying of both vessels. Possible location (ALONSO CASADO et al. 2000; FALCONI et al.
evaluation can be made of how much the macro- 2001; KOBARI et al. 1999; SUGIYAMA and ATOMI 1998;
scopic infiltration influences the prognosis of these TERRIS et al. 2000). Limited or atypical resections,
neoplasms. The differential diagnosis between IPMT as some authors suggest (SCIAUDONE et al. 2000;
and ductal adenocarcinoma of the body-tail involv- TAKEYOSHI et al. 1999; TERRIS et al. 2000), involve a
ing the splenic vessels is important since the latter greater risk oflocal recurrence (FALCONI et al. 2001).
has a low indication of resectability and, in any case, a Intraoperatively, it is crucial to ensure that a nega-
poor prognosis. On the contrary, IPMTs, even if infil- tive margin for dysplasia is found, even if it leads to
trating, should be resected; they are not sensitive to total pancreatectomy. In this case, considering the
chemotherapy or radiotherapy, leaving no alternative patient's age and general condition, the surgeon may
treatment to surgical intervention (KLOPPEL 1998). decide not to perform a total pancreatectomy if low
The presence of hepatic metastases precludes or moderate dysplasia is present on the edge of the
resectability. In our experience, the latter event only section (FALCONI et al. 2001).
appeared in 2% of patients. This is clearly different
from patients with ductal adenocarcinomas, 50% of
whom are affected by hepatic metastases at diagno- 5.5.2
sis. The prognosis of infiltrating IPMTs depends on FollOW-Up
the histological type of infiltration (FUKUSHIMA et
al. 1997). Patients with the tubular type of infiltration At the moment, the data in literature and from
have a poor prognosis similar to ductal adenocarci- personal experience are not sufficient to consider
Clinical Manifestations and Therapeutic Management 83

follow-up as a standard choice for these patients Fukushima N, Mukai K, Kanai Y, Hasebe T, Shimada K, Ozaki
because of the potentially malignant nature of these H, Kinoshita T, Kosuge T (1997) Intraductal papillary
tumors. Nevertheless, small lesions of the pancreatic tumors and mucinous cystic tumors of the pancreas: clini-
copathologic study of 38 cases. Hum PathoI28:1010-1017
head, originating from the branch ducts, with no Furukawa T, Tsukamoto Y, Naitoh Y, Mitake M, Hirooka Y,
past history of diabetes or jaundice, without risk Hayakawa T (1994) Differential diagnosis of pancreatic
factors, and in the absence of radiological signs of diseases with an intraductal ultrasound system. Gastroin-
malignancy can be strictly followed up, as some test Endosc 40:2l3-219
authors advise (KIMURA and MAKUUCHI 1999; Grieshop NA, Wiebke EA, Kratzer SS, Madura JA (1994) Cystic
neoplasms of the pancreas. Am Surg 60:509-514; discus-
YAMAO et al. 2000).
sion 514-515
Itai Y, Itai Y, Ohtomo K, Itoh K, Kawauchi N, Minami M (1987)
Mucin-hypersecreting carcinoma of the pancreas. Radiol-
ogy 165:51-55
References Itoh S, Ishiguchi T, Ishigaki T, Sakuma S, Maruyama K, Senda
K (1992) Mucin-producing pancreatic tumor: CT findings
and histopathologic correlation. Radiology 183:81-86
Abe H, Kubota K, Mori M, Mild K, Minagawa M, Noie T, Kamei K, Funabiki T, Ochiai M, Amano H, Kasahara M, Saka-
Kimura W, Makuuchi M (1998) Serous cystadenoma of the moto T (1991) Multifocal pancreatic serous cystadenoma
pancreas with invasive growth: benign or malignant? Am J with atypical cells and focal perineural invasion. Int J Pan-
GastroenteroI93:1963-1966 creatollO:161-172
Alonso Casado 0, Hernandez Gallardo D, Moreno Gonzalez Kimura W, Makuuchi M (1999) Operative indications for
E, Manzanera Diaz M, Gimeno Calvo A, Perez Saborido B, cystic lesions of the pancreas with malignant potential:
Marques Medina E, Gutierrez Martin A (2000) Intraductal our experience. Hepatogastroenterology 46:483-491
papillary-mucinous tumors: an entity which is infrequent and Kimura W, Sasahira N, Yoshikawa T, Muto T, Makuuchi M
difficult to diagnose. Hepatogastroenterology 47:275-284 (1996) Duct-ectatic type of mucin producing tumor of the
Azar C, Van de Stadt J, Rickaert F, Deviere M, Baize M, Kloppel pancreas: new concept of pancreatic neoplasia. Hepatogas-
G, Gelin M, Cremer M (1996) Intraductal papillary muci- troenterology 43:692-709
nous tumours of the pancreas. Clinical and therapeutic Kloppel G (1998) Clinicopathologic view of intraductal papil-
issues in 32 patients. Gut 39:457-464 lary-mucinous tumor of the pancreas. Hepatogastroenter-
Barbe L, Ponsot P, Vilgrain V, Terris B, Fiejou JF, Sauvanet A, ology 45:1981-1985
Belghiti J, Hammel P, Ruszniewski P, Bernades P (1997) Kloppel G (1996) Histogical typing of tumors of the eso-
Intraductal papillary mucinous tumors of the pancreas. crine pancreas. World Health Organization International
Clinical and morphological aspects in 30 patients. Gastro- Histological Classification of Tumours. Springer, Berlin
enterol Clin Bioi 21:278-286 Heidelberg New York
Brat DJ, Lillemoe KD, Yeo CJ, Warfield PB, Hruban RH (1998) Kobari M, Egawa S, Shibuya K, Shimamura H, Sunamura M,
Progression of pancreatic intraductal neoplasias to infil- Takeda K, Matsuno S, Furukawa T (1999) Intraductal papil-
trating adenocarcinoma of the pancreas. Am J Surg Pathol lary mucinous tumors of the pancreas comprise 2 clinical
22:163-169 subtypes: differences in clinical characteristics and surgi-
Cavallini G, Bovo P, Vaona B, DiFrancesco V, Frulloni L, Rigo cal management. Arch Surg l34: 1l31-1136
L, Brunori MP, Andreaus MC, Tebaldi M, Sgarbi D, Angelini Lim JH, Yi CA, Lim HK, Lee WJ, Lee SJ, Kim SH (2001) Radio-
G, Talamini G, Procacci C, Pederzoli P, Filippini M (1996) logic spectrum of intraductal papillary mucinous tumor
Chronic obstructive pancreatitis in humans is a lithiasic of the pancreas. Radiographies 21:323-337; discussion
disease. Pancreas l3:66-70 337-340
Cavallini G, Frulloni L, Pederzoli P, Talamini G, Bovo P, Bassi Loftus EV Jr., Olivares-Pakzad BA, Batts KP, Adkins MC,
C, Di Francesco V, Vaona B, Falconi M, Sartori N, Angelini Stephens DH, Sarr MG, DiMagno EP (1996) Intraductal
G, Brunori MP, Filippini M (1998) Long-term follow-up of papillary-mucinous tumors of the pancreas: clinicopatho-
patients with chronic pancreatitis in Italy. Scand J Gastro- logic features, outcome, and nomenclature. Members of the
enterol 33:880-889 Pancreas Clinic, and Pancreatic Surgeons of Mayo Clinic.
Cuillerier E, Cellier C, Palazzo L, Deviere J, Rickaert F, Flejou Gastroenterology 110: 1909-1918
JF, Van de Stadt J, Landi B, Pare R, Cremer M, Cugnenc PH, Longnecker DS (1998) Observations on the etiology and
Barbier JP (1998) Intraductal papillary mucinous tumors pathogenesis of intraductal papillary-mucinous neoplasms
of the pancreas: are there any preoperative clinical and of the pancreas. Hepatogastroenterology 45:1973-1980
laboratory factors predictive of degeneration. Results of a Milchgrub S, Campuzano M, Casillas J, Albores-Saavedra J
French-Belgium collective series. Ann Chir 52:215-222 (1992) Intraductal carcinoma of the pancreas. Cancer 69:
Falconi M, Salvia R, Bassi C, Zamboni G, Talamini G, Peder- 651-656
zoli P (2001) Clinicopathological features and treatment of Nakagohri T, Kenmochi T, Kainuma 0, Tokoro Y, Asano T
intraductal papillary mucinous tumor of the pancreas. Br (1999) Intraductal papillary mucinous tumors of the pan-
J Surg 88:376-381 creas. Am J Surg 178:344-347
Fukukura Y, Fujiyoshi F, Sasaki M, Inoue H, Yonezawa S, Navarro F, Michel J, Bauret P, Ramos J, Blanc P, Fabre JM,
Nakajo M. (2000) Intraductal papillary mucinous tumors Millat B, Desrousseaux B, Domergue J (1999) Management
of the pancreas: thin-section helical CT findings. AJR Am J of intraductal papillary mucinous tumors of the pancreas.
RoentgenoI174:441-447 Eur J Surg 165:43-48
84 R. Salvia et al.

Obara T, Maguchi H, Saitoh Y, Itoh A, Arisato S, Ashida T, Y (2000) Intraductal papillary mucinous tumors of the
Nishino N, Ura H, Namiki M (1991) Mucin-producing pancreas: helical CT with histopathologic correlation.
tumor of the pancreas: a unique clinical entity. Am J Gas- Radiology 217:757-764
troenterol 86: 1619-1625 Terris B, Vilgrain V, Vullierme MP, Terris B, Denys A, Sauvanet
Obara T, Maguchi H, Saitoh Y, Itoh A, Arisato S, Ashida T, A, Hammel P, Menu Y (2000) Intraductal papillary muci-
Nishino N, Ura H, Namiki M (1993) Mucin-producing nous tumors of the pancreas confined to secondary ducts
tumor of the pancreas: natural history and serial pancrea- show less aggressive pathologic features as compared with
togram changes. Am J Gastroenterol 88:564-569 those involving the main pancreatic duct. Am J Surg Pathol
Ohhashi K (1982) Four cases of mucus secreting pancreatic 24:1372-1377
cancer. Prog Dig Endosc 20:48-51 Traverso LW (1998) Intraductal neoplasms of the pancreas.
Ohta T (1992) The ,duct-ectatic' variant of mucinous cystic Am J Surg 175:426-432
neoplasm of the pancreas: clinical and radiologic studies Uehara H, Nakaizumi A, Iishi H, Tatsuta M, Kitamra T, Okuda
of seven cases. Am J Gastroenterol 87:300-304 S, Ohigashi H, Ishikawa 0, Takenaka A, Ishiguro S (1994)
Partensky C, Berger F, Ponchon T, Valette PJ (1996) Pancre- Cytologic examination of pancreatic juice for differential
atectomy for intraductal papillary mucinous tumor of the diagnosis of benign and malignant mucin-producing
pancreas. Gastroenterol Clin Bioi 20:938-945 tumors of the pancreas. Cancer 74:826-833
Paye F, Ponsot P, Paye F, Hammel P, Sauvanet A, Molas G, Uehara H, Nakaizumi A, Tatsuta M, Iishi H, Kitamura T,Ohi-
Bernades P, Belghiti J, Ruszniewski P, Flejou JF (1999) gashi H, Ishikawa 0, Takenaka A (1997) Diagnosis of car-
Intraductal papillary mucinous tumors of the pancreas: cinoma in situ of the pancreas by peroral pancreatoscopy
diagnosis, treatment and prognosis. Ann Chir 53:598-604 and pancreatoscopic cytology. Cancer 79:454-461
Raijman I, Kortan P, Walden D, Kandel G, Marcon NE, Haber Wakabayashi T, Kawaura Y, Morimoto H, Watanabe K, Toya D,
GB (1994) Mucinous ductal ectasia: cholangiopancreato- Asada Y, Satomura Y, Watanabe H, Okai T, Sawabu N (2001)
graphic and endoscopic findings. Endoscopy 26:303-307 Clinical management of intraductal papillary mucinous
Rickaert F, Cremer M, Deviere J, Tavares L, Lambilliotte tumors of the pancreas based on imaging findings. Pan-
JP, Schroder S, Wurbs D, Kloppel G (1991) Intraductal creas 22:370-377
mucin-hypersecreting neoplasms of the pancreas. A clini- Warshaw AL (1991) Mucinous cystic tumors and mucinous
copathologic study of eight patients. Gastroenterology 101: ductal ectasia of the pancreas. Gastrointest Endosc 37:
512-519 199-201
Rivera JA, Fernandez-del Castillo C, Pins M, Compton CC, Widmaier U, Mattfeldt T, Siech M, Beger HG (1996) Serous
Lewandrowski KB, Rattner DW, Warshaw AL (1997) Pan- cystadenocarcinoma of the pancreas. Int J Pancreatol 20:
creatic mucinous ductal ectasia and intraductal papillary 135-139
neoplasms. A single malignant clinicopathologic entity. Yamaguchi K, Tanaka M (1991) Mucin-hypersecreting tumor
Ann Surg 225:637-644; discussion 644-646 of the pancreas with mucin extrusion through an enlarged
Sciaudone G, Perniceni T, Levy P, Bougaran J, Gayet B (2000) papilla. Am J Gastroenterol 86:835-839
Enucleation of intraductal papillary-mucinous tumor of Yamaguchi K, Ogawa Y, Chijiiwa K, Tanaka M (1996) Mucin-
the head of the pancreas. Report of 2 cases. Gastroenterol hypersecreting tumors of the pancreas: assessing the grade
Clin Bioi 24:121-124 of malignancy preoperatively. Am J Surg 17l:427-431
Shima Y, Mori M, Takakura N, Kimura T, Yagi T, Tanaka N Yamaguchi K, Yokohata K, Noshiro H, Chijiiwa K, Tanaka M.
(2000) Diagnosis and management of cystic pancreatic (2000) Mucinous cystic neoplasm of the pancreas or intra-
tumors with mucin production. Br J Surg 87:1041-1047 ductal papillary-mucinous tumor of the pancreas. Eur J
Siech M, Tripp K, Schmidt-Rohlfing B, Mattfeldt T, Gorich J, Surg 166:141-148
Beger HG (1999) Intraductal papillary mucinous tumor of Yamao K, Ohashi K, Nakamura T, Suzuki T, Shimizu Y, Naka-
the pancreas. Am J Surg 177: 117-120 mura Y, Horibe Y, Yanagisawa A, Nakao A, Nimuara Y, Naito
Silas AM, Morrin MM, Raptopoulos V, Keogan MT (2001) Y, Hayakawa T (2000) The prognosis of intraductal papil-
Intraductal papillary mucinous tumors of the pancreas. lary mucinous tumors of the pancreas. Hepatogastroenter-
AJR Am J RoentgenoI176:179-185 ology 47:1129-1134
Solcia E (1997) Tumors of the pancreas. Atlas of Tumor Pathol- Yoshimi N, Sugie S, Tanaka T, Aijin W, Bunai Y, Tatematsu A,
ogy, 3rd ser, fasc 20. Armed Forces Institute of Pathology, Okada T, Mori H (1992) A rare case of serous cystadeno-
Washington DC carcinoma of the pancreas. Cancer 69:2449-2453
Sugiyama M, Atomi Y (1998) Intraductal papillary mucinous Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini
tumors of the pancreas: imaging studies and treatment G, Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM,
strategies. Ann Surg 228:685-691 Kloppel G (1999) Mucinous cystic tumors of the pancreas:
Sugiyama M,Atomi Y (1999) Extrapancreatic neoplasms occur clinicopathological features, prognosis, and relationship
with unusual frequency in patients with intraductal papil- to other mucinous cystic tumors. Am J Surg Pathol 23:
lary mucinous tumors of the pancreas. Am J Gastroenterol 410-422
94:470-473 Z'Graggen K, Rivera JA, Compton CC, Pins M, Werner J,
Takeyoshi I, Ohwada S, Nakamura S, Ogawa T, Kawashima Y, Fernandez-del Castillo C, Rattner DW, Lewandrowski KB,
Ikeya T, Morishita Y (1999) Segmental pancreatectomy for Rustgi AK, Warshaw AL (1997) Prevalence of activating
mucin-producing pancreatic tumors. Hepatogastroenterol- K-ras mutations in the evolutionary stages of neoplasia
ogy 46:2585-2588 in intraductal papillary mucinous tumors of the pancreas.
Taouli B, Vilgrain V, O'Toole D, Vullierme MP, Terris B, Menu Ann Surg 226:491-498; discussion 498-500
6 Pathology of Intraductal Cystic Tumors
G. ZAMBONI, P. CAPELLI, G. BOGINA, A. PESCI, and A. BRIGHENTI

CONTENTS papillary neoplasms (MOROHOSHI et al. 1989), dif-


fuse intraductal papillary adenocarcinoma (CONLEY
6.1 Introduction 85 et al. 1987), villous adenoma of the main pancreatic
6.2 Macroscopic Examination 85 duct (PAYAN et al. 1990), villous adenomatosis of duct
6.3 Microscopic Examination 88
6.4 Immunohistochemical and Molecular of Wirsung (BRESLER et al. 1992) are among these
Characteristics 90 names.
6.5 Preoperative and Intraoperative Apart from their macroscopically heterogeneous
Pathological Diagnosis 91 appearance, all these tumors can be regarded as a
6.6 Differential Diagnosis 93 single clinicopathological entity. The revised WHO
6.7 Prognosis 93
References 93 classification of exocrine pancreatic neoplasms
(KU)PPEL et al. 1996), reported in the Armed Forces
Institute of Pathology (AFIP) fascicle (SOLCIA et al.
1997), suggests the term intraductal papillary muci-
nous tumor (IPMT), calling attention to a possible
6.1 adenoma-carcinoma sequence; the transition from
Introduction benign to malignant epithelium, which can be seen
in a single IPMT, with identifiable adenomatous, bor-
In recent years, increasing attention has been paid derline, and malignant components is morphological
to intraductal papillary mucinous tumors (IPMTs) evidence of tumor progression.
because of their clinical picture, favorable prognosis, The pathologic hallmarks of IPMT are: (1) diffuse
unclear nature, and potential relationship with ductal or segmental dilatation of the main and/or branch
adenocarcinoma. While the diagnosis of IPMTs has pancreatic ducts, with no mechanical strictures, (2)
improved considerably over the past few years (AZAR intraductal growth of the mucin-producing epithelial
et al. 1996; LOFTUS et al. 1996; SESSA et al. 1994), the lining cells, with a 'flat' or 'papillary-villous' appear-
nature of these neoplasms, their pathogenesis, and their ance, (3) protrusion and dilatation of the major and/
relationship to ductal adenocarcinoma and its variants, or minor papilla of Vater, with mucous excretion.
specifically mucinous noncystic carcinoma (MNCC), To be classified as an IPMT, a neoplasm must
are still largely unknown (LONGNECKER 1998). involve the main pancreatic duct and/or major
In the past, this entity has been reported under branches and should be grossly or radiologically
different names, reflecting either the various authors' visible (TERRIS et al. 2000). If it is not, it would be
specialty (radiology, gastroenterology, endoscopy, sur- better classified as a pancreatic intraepithelial neo-
gery, pathology) or the different features of the tumors plasia (PanIN), a proliferative epithelial lesion that
(KLOPPEL 1998). Mucous-secreting cancer (OHASHI et typically involves the smaller pancreatic ducts and
al. 1982), mucin-producing tumor (OBARA et al. 1991; ductules (HRUBAN et al. 2001; Fig. 6.1).
YAMADA et al. 1991), mucus-hypersecreting tumor
(FURUKAWA et al. 1992), intraductal mucin-hyperse-
creting neoplasm (RICKAERT et al. 1991), ductectatic
mucinous cystadenoma (ITAI et al. 1986), intraductal 6.2
Macroscopic Examination
G. ZAMBONI, MD; P. CAPELLI, MD; A. BRIGHENTI, MD
Department of Pathology University of Verona, Verona, Italy The macroscopic examination of the surgical speci-
G. BOGINA, MD; A. PESCI, MD men should document the intraductal origin and
Ospedale S.Cuore-Don Calabria, Verona, Italy growth of the lesion, the distribution and type of
86 G. Zamboni et al.

The large majority of cases arises in the main pancre-


atic duct of the head of the pancreas and progresses
in a retrograde direction over a variable length with
frequent extension into the branch ducts (Fig. 6.4).
Extension of the IPMT to the major and/or minor
papilla creates the typical protrusion into the duode-
num, which can be seen either at endoscopy or at US
and CT. The patulous orifice is observed in 50%-60%
of cases (Fig. 6.5). Unfortunately, it is present only in
the advanced stages of disease, being present in 73%
of patients with either in situ carcinoma or invasive
carcinoma as opposed to 39% of patients with hyper-
plasia and adenoma (HARA et al. 2002). In advanced
Fig. 6.1. Pancreatic intraepithelial neoplasia of the pancreas cases, IPMT may form fistulas with adjacent organs.
(PanIN). Branch duct and small branches with microscopic According to the study of 36 patients with IPMTs with
alteration, previously unidentified at both imaging and macro- fistulae (KOBAYASHI et al. 1993), the organs involved
scopic examination, characterized by papillary formation were the stomach (17%), duodenum (64%), and bile
duct (56%). When the tumor creates a biliopancre-
tumor, and search for an invasive carcinoma com- atic fistula, it is important to differentiate a primary
ponent. fistula from neoplastic invasion of the bile duct
Macroscopically, two main types can be distin- (KURIHARA et al. 2000). In fact, half of the patients
guished: the ductectatic mucin-hypersecreting vari- with fistulae retain the basic characteristics of IPMT
ant in which the dilated main duct or branch ducts with no carcinomatous invasion around the fistula.
are filled with tenacious mucin, and the cyst lining Mechanical pressure due to the tumor's expansion
is flat and smooth (Fig. 6.2), and the papillary-villous and/or thick mucus 'bursting' into the bile duct are
variant, characterized by soft and friable, nodular or the proposed pathophysiologic mechanisms in this
papillary proliferation associated with mucous mate- subset of patients. The other half appeared with foci
rial within the dilated pancreatic duct (Fig. 6.3). The of invasive colloid carcinoma in the stroma around
polypoid proliferations, together with mucin plugs, are the fistula. Local recurrence and diffuse peritoneal
responsible for the typical filling defects seen at endo- carcinomatosis have been reported (KURIHARA et al.
scopic retrograde cholangiopancreatography (ERCP). 2000). The pancreatic acinar parenchyma surround-
IPMT may also be divided into main duct and ing the central type IPMT is frequently replaced by
branch duct types. The main duct type involves the fibrous tissue, related to local chronic pancreatitis
main duct, either in a diffuse or segmental manner. resulting from long-lasting duct obstruction.

Fig. 6.3. IPMT papillary-villous type. Duodenopancreatectomy


specimen showing a nodular, papillary proliferation within a
Fig. 6.2. IPMT ductectatic mucin-hypersecreting type. A cystically dilated duct of Wirsung with extension to the duct of Santorini,
dilated branch duct of the uncinate process is filled with sticky associated with a scirrhous carcinomatous area involving the
mucus. The lesion communicates with the main pancreatic duct head of the pancreas and infiltrating the terminal bile duct
Pathology of Intraductal Cystic Tumors 87

IPMTs of the branch ducts arise anywhere within


the pancreas, although the head and neck of the pan-
creas and the uncinate process are the more common
locations. They often have a grape-like appearance
and may protrude from the outer part of the pancre-
atic parenchyma (Fig. 6.6). Cystic dilatation ranges
from 3 mm to 50-60 mm. The cystic changes in this
subtype can sometimes be so extensive that they may
be confused with mucinous cystic tumor. The main
criterion for distinguishing the two lesions is that in
IPMT the cysts communicate with the duct system
(Fig. 6.7), whereas mucinous cystic tumors do not.
The adjacent pancreatic parenchyma in the branch-
Fig. 6.4. IPMT main duct type. Total pancreatectomy showing type IPMT is usually normal.
an advanced stage of the disease with diffuse dilatation of the Searching for invasive carcinomatous components
main duct and the duct of Santorini with multiple gelatinous in IPMTs is extremely important for many reasons.
areas in the head and tail of the pancreas
The presence of carcinomatous invasion is considered
the most important predictive factor for survival and
disease recurrence along with the presence of lymph
node metastases (CUILLERIER et al. 2000). Although
carcinomatous invasion is reported in 30%-50% of
patients (KLOPPEL 1998; FUKUSHIMA et al. 2001;
GIGOT et al. 2001; ADSAY et al. 2002; SUGIURA et al.
2002; SOHN et al. 2001; CUILLERIER et al. 2000), the
sensitivity of imaging in preoperatively identifying
invasive cases is less than 80% (CUILLERIER et al.
2000). IPMTs with carcinomatous transformation are
frequently found in the main duct type, characterized
by marked longitudinal extension and involvement of
secondary ducts (Fig. 6.4), but can also be observed
in variable proportion in the peripheral branch duct
type. The macroscopic findings, which correlate with
the carcinomatous nature of the lesions, are thick
ductal walls and endoluminal papillary or nodular
Fig. 6.5. IPMT main duct and branch duct type. Whole-mount
vegetations. In the muconodular or colloid type
macro section showing papillary proliferations involving the
main and branch ducts of the head of the pancreas with exten- carcinoma, the invasive component is constituted
sion to the intraduodenal portion of the main duct and to the by multiple, well-circumscribed, gelatinous nodules,
ampulla of Vater which can be confined to the pancreatic parenchyma

a h----------------------------------- b
Fig.6.6a,b. IPMT branch duct type. A grape-like aspect with protrusion from the outer part of the uncinate process of the
pancreas (a); on section, the main cyst measured 2 cm in diameter at its largest point (b)
88 G. Zamboni et al.

a b

Fig.6.7a,b. IPMT branch duct type of the uncinate process. a A mucus-filled cystic lesion of the uncinate process; the probe
indicates the bile duct. b Whole-mount macro section of the lesion showing a connection with the main duct, the presence of
sticky mucus, and a smooth internal surface of the cyst

or extend into the peripancreatic adipose tissue, with findings (FURUKAWA et al. 1997; KOITO et al. 2001).
or without lymph node involvement. Identification of The combination of these two new diagnostic pre-
the tubular type carcinoma within an IPMT depends operative techniques is highly sensitive in differenti-
on the extension of the carcinomatous foci. Grossly, ating between benign and malignant IPMTs (HARA
they appear as a firm mass with ill-defined margins, et al. 2002). The presence or absence of protruding
merging imperceptibly with the surrounding pan- lesions, their morphology, height, and the presence or
creatic parenchyma, like common ductal carcinomas absence of tumor vessels in the protrusions are useful
of the pancreas. This macroscopic appearance may features for the differential diagnosis (Fig. 6.9). The
be difficult to differentiate from the fibrous scars of visualization by POPS of villous or vegetative types
chronic pancreatitis. of growth and the presence of tumor vessels correlate
with malignancy, as well as the presence of lesions
protruding more than 4 mm on mus.

6.3
Microscopic Examination

Microscopically, the epithelial component lining the


ectatic main duct or the cystically dilated branch
ducts is represented by tall mucin-producing colum-
nar cells, but lacks the 'ovarian-type' stroma seen in
mucinous cystic tumors (MCTs) (Fig. 6.8). This
epithelium frequently forms papillary projections
and shows dysplastic modifications. The papillary
proliferations may consist of microscopic folds, usu-
ally lined by slightly atypical columnar cells, or florid
branching papillae measuring up to several centi-
meters. The latter prevalently show intestinal-type
epithelial lining, similar to villous adenoma of the
large bowel, or may rarely appear with a pancreato-
biliary-type epithelium, which is less mucinous than
the former and is more likely to be highly dysplastic
(ADSAY et al. 2002). The pathological features of the
papillary proliferations constitute the anatomical
basis for the interpretation of peroral pancreatoscopy
(POPS) and intraductal ultrasonography (mUS) Fig. 6.8. IPMT with tall columnar epithelium and mild atypia
Pathology of Intraductal Cystic Tumors 89

Fig. 6.9. IPMT with micropapillary proliferations of the epithe-


liallining, showing fibrous stalks with small vessels

Individual IPMTs, or sometimes within the single


lesion, show a spectrum of changes with the preva-
lence in some areas of ductal ectasia and the presence
Fig. 6.10. IPMT with diffuse involvement of central and
of papillary proliferations in contiguous areas. The peripheral ducts, with ductal ectasia and papillary prolifera-
lesions, either the central or the peripheral types, are tion, with no tumoral pseudocapsule
usually poorly circumscribed and without a fibrous
capsule delimitating them from the parenchyma.
However, the large, dilated ducts may have a fibrous
wall (Fig. 6.10).
Recently, an oncocytic variant of IPMT was iden-
tified (ADSAY et al. 1996). The clinical-pathological
features seem to be similar to those of the classical
type, with a slight prevalence in elderly, male patients
and the pancreatic head. The tumors have mucin-
filled cysts containing nodular papillary projections
lined by cells with abundant granular eosinophilic
cytoplasm (Fig. 6.11) (oncocytic cells rich in mito-
chondria), mixed with both goblet and mucin-con-
taining cells. Characteristically, these oncocytic cells
show intracytoplasmic lumina (Fig. 6.12). Most of
them were noninvasive carcinomas; one case showed
focal microinvasion, and another displayed wide-
spread invasive carcinoma.
IPMTs are classified on the basis of the greatest
degree of dysplasia, according to the WHO classifica-
tion, into adenoma, borderline tumor, and carcinoma Fig. 6.11. IPMT oncocytic type. Presence of papillary prolifera-
(KLOPPEL et al. 1996). tions, lined by cells with finely granular cytoplasm and nuclei
Intraductal papillary-mucinous adenoma is char- with prominent nucleoli
acterized by tall columnar mucin-producing cells with
mild dysplasia and the absence of mitosis (Fig. 6.8); changes with the presence of irregular branching
borderline intraductal papillary-mucinous tumor is papillae or cribriform growth. The epithelial cells
characterized by papillary projections, cellular pseu- show nuclear stratification, severe nuclear atypia, and
do stratification with crowding of moderately atypical frequent mitosis. Because of the great variability within
and hyperchromatic nuclei (Fig. 6.13); intraductal a tumor, it is important to emphasize the necessity of
papillary-mucinous carcinoma has severe dysplastic extensive tumor sampling in order to make the correct
90 G. Zamboni et al.

diagnosis and assess the presence of tumor invasion,


giving special emphasis to papillary or nodular prolif-
erations and sclerotic areas.
Carcinomatous stromal invasion by invasive IPMT
is characterized by the presence of mucin-filled cystic
spaces, partially lined by atypical cells and containing
floating mucus-secreting cell nuclei (Fig. 6.14). This
pattern of invasion, which has been called'muconodu-
lar' (YAMADA et al. 1991), assumes the appearance of
MNCC (SOLCIA et al.1997; KLOPPEL et al. 1996) or col-
loid carcinoma (ADSAY et al. 200l). The overwhelming
majority of colloid carcinomas in the pancreas arises
from pre-existing IPMTs. When colloid tumors of the
pancreas have been extensively sampled, pre-existing
IPMTs were found in all cases (SEIDEL et al. 2002),
Fig. 6.12. IPMT oncocytic type. Oncocytic cells showing intra-
cytoplasmic lumina whereas with less extensive pathological sampling,
ADSAY et al. reported 59% of association with IPMTs
(ADSAY et al. 2001). The finding of a colloid carcinoma
in the pancreas should prompt the pathologist to
search for an IPMT. On the contrary, the presence of
a glandular component, which characterizes the'tubu-
lar' pattern (similar to the usual ductal carcinoma), is
rarely encountered (YAMADA et al. 1991). The 'tubular'
type carcinoma within an IPMT is characterized by
the presence of malignant glands, similar to those of
common ductal carcinoma (Fig. 6.15).

6.4
Immunohistochemical and Molecular
Characteristics
a Although IPMTs represent an extremely interest-
ing system for studying tumor progression in the
pancreas, very little is known about their molecular
abnormalities. HER-2neu overexpression has been
found in about 75% of cases (SESSA et al. 1994). K-ras
mutation has been reported in 30%-80% of IPMTs
(SESSA et al. 1994; TADA et al. 1991; Z'GRAGGEN et
al. 1997; YANAGISAWA et al. 1991). Independent and
different K-ras mutations have been identified in
IPMTs, suggesting a field cancerization and giving
a molecular basis for the risk of synchronous and!
or metachronous tumors (IzAwA et al. 200l). The
retention of tumor suppressor functions in IPMTs
may explain their more favorable prognosis com-
b pared with ductal carcinoma. Mutations in the p53
tumor-suppressor gene were detected in only 8%
Fig. 6.13a,b. IPMT borderline. a Intestinal-type papillae, of cases (SESSA et al. 1994). Recently, a 25% rate of
morphologically similar to those in colonic villous adenoma.
b Papillary projections characterized by cellular pseudostrati- inactivation of the STKll!LKBl tumor-suppressor
fication with crowding of moderately atypical and hyperchro- gene responsible for Peutz-Jeghers syndrome has
matic nuclei; mitosis is observed been reported (SATO et al. 200l). This gene, which is
Pathology of Intraductal Cystic Tumors 91

a _____...-.. b

Fig. 6.14a,b. IPMT with 'muconodular' carcinomatous transformation. a Whole-mount macro section of duodenopancreatec-
tomy showing a diffuse ductal ectasia of the head of the pancreas and multiple nodular gelatinous areas. b Histologically, the
mucin-filled cystic spaces are partially lined by atypical cells and contain floating mucus-secreting cells

involved in growth suppression activity, also associ-


ates with p53 in apoptosis control.
Depending on their mucin expression profiles, one
common (MUC2-positive) and two rarer (MUCl/
MUC2 and MUC1) types of IPMTs (LUTTGES et al.
2001) can be identified. The MUC2-positive IPMTs
typically form papillary structures. When invasive,
all MUC2-positive IPMTs assume the appearance
of a MNCC. The IPMTs showing focal MUC1/MUC2
expression are characterized by oncocytic histology.
The third group seems to be related to ductal carci-
noma because of its MUC1 positivity in the absence
of, or very slight, MUC2 staining and due to the prev-
alence of the 'tubular' pattern of their invasive carci-
nomatous component. This assumption is supported
Fig. 6.15. IPMT with 'tubular' carcinomatous transforma-
by the DPC4 findings. DPC4 is a tumor-suppressor tion. Tubular-type carcinoma is characterized by infiltrating,
gene on chromosome 18q, which mediates the effect irregular tubular structures, similar to those of ordinary
of TGF-beta signaling, resulting in growth inhibi- ductal carcinoma
tion (DAr et al. 1998). While DPC4 is consistently
expressed in the MUC2 and MUCl/MUC2 types of
IMPTs, both in the intraductal and in the muconodu- 6.S
lar carcinomatous component, 50% of the MUC1- Preoperative and Intraoperative
positive IPMTs and the 'tubular' invasive carcinomas Pathological Diagnosis
show the loss of DPC4/SMAD4 expression (LUTTGES
et al. 2001; IACOBUZIO-DoNAHUE et al. 2000).A simi- A preoperative pathological diagnosis of IPMT
lar proportion of deletion (55%) has been reported in is possible with both cytology and histology. The
conventional ductal carcinomas (HAHN et al. 1996). cytological examination of the intraductal material
As DPC4 expression in conventional carcinoma cor- collected during the endoscopic examination (ERCP)
relates with longer survival (WrLENTz et al. 2000), reveals the presence of abundant extracellular mucin
it is possible to hypothesize that the prolonged and with a floating epithelial component, either in singly
indolent clinical course of IPMT compared with dispersed cells or in a sheet-like arrangement. While
conventional ductal carcinomas may be related in it is possible to subdivide low-grade and high-grade
part to the persistent expression of DPC4 in IPMTs tumors, it is not possible to predict the invasiveness
(IACOBUZIO-DoNAHUE et al. 2000). of the tumor. The examination of K-ras mutation
92 G. Zamboni et a1.

from pancreatic juice, in association with cytologi-


cal examination, proved to be useful, although the
accuracy rate is low (HARA et al. 2002; TADA et al.
1991; YANAGISAWA et al. 1991). The histological
diagnosis can be performed on biopsy material
obtained from the major/minor papilla or directly
from the main duct during a pancreatoscopy. The
disadvantage of histology is that a single localized
biopsy may not accurately reflect the region of the
severest dysplasia.
As positive resection margins and postoperative a
recurrence have been reported in up to 50% of patients
(SHO et al. 1998; FALCONI et al. 2001; AZAR et al. 1996;
RIVERA et al. 1997; TRAVERSO et al. 1998; SHO et al.
1998), the determination of tumor-free resection lines
is extremely important. It is crucial for the surgeon to
assess the intraductal spread of the disease in order to
achieve a total resection. Preoperative imaging, com-
puted tomography (CT), magnetic resonance imaging
(MRI), endoscopic ultrasonography (EUS), and ERCP
do not provide accurate information, failing to pre-
dict neoplasm extension in more than 20% of cases
(CUILLERIER et al. 2000). Partial pancreatic resection
b
should be guided by intra-operative frozen section
examination until disease-free margins are obtained.
Pathologists face many difficulties. One of these is the
differential diagnosis between papillary hyperplasia
and low-grade dysplasia; the pathologist should report
only the moderate to high-grade dysplasia. A second
problem is represented by the extensive inflammatory
reaction with epithelial denudation, which is frequently
found in association with dilatation of the main duct.
The mere absence of dysplastic epithelium in these
cases is not sufficient to define a negative margin. We
experienced two stump recurrences in such cases (Fig.
6.16). Another important point to stress is that careful
attention must be paid to epithelial changes of the small,
peripheral ducts, since the tumor may develop over a
c
wide area in the branches (NAGASAKA and NAKASHIMA
2001). In fact, a tumor component with low-grade dys- Fig. 6.16a-c. IPMT with stump recurrence. a The resected jeju-
plasia remaining in the resected margins may explain nopancreatostomy showing a gelatinous enlargement of the
the high percentage oflocal tumor recurrence in IPMTs. anastomotic area. b A whole-mount macro section showing
the pancreatic stump with multiple areas of colloid carcinoma
For this reason, a negative margin in IPMT patients cor- involving the anastomotic area. c The resection margin of the
responds only to a normal ductal epithelium. Recently, previous duodenopancreatectomy showing a central duct of
the importance of using intraoperative annular array Wirsung with extensive inflammatory reaction and epithelial
US (KANAZUMI et al. 2001), intraductal ultrasonogra- denudation; some branch ducts were dilated and filled with
phy (GIGOT et al. 2001), and pancreatoscopy (HARA et mucus, but lacked evident cellular atypia
al. 2002) has been emphasized to highlight the small
intraluminal proliferations of the duct ofWirsung, lead- This approach seems particularly effective for detecting
ing to biopsy and frozen section examination. In the longitudinal spread of the disease and the multicentric
series reported by HARA et al., a postoperative recur- micropapillary lesions which are not macroscopically
rence was observed in only one patient, and the 3-year visible, and for improving the postoperative outcome
cumulative survival rate was 93% (HARA et al. 2002). (GIGOT et al. 2001; HARA et al. 2002).
Pathology of Intraductal Cystic Tumors 93

The use of pancreatogastric anastomosis in patients become invasive and metastasize (KLOPPEL 1998;
with IPMT treated with pancreatoduodenectomy FUKUSHIMA et al. 2001; GIGOT et al. 2001; ADSAY
improves the long-term oncological follow-up. This et al. 2002; SUGIURA et al. 2002; SOHN et al. 2001;
reconstructive technique gives direct access to the CUILLERIER et al. 2000), and the 5-year survival rate
pancreatic stump by endoscopy during the follow-up, for all patients with IPMT ranges from 50% to 80%
allowing direct opacification of the pancreatic duct, (SOHN et al. 2001; ADSAY et al. 2002; KANAZUMI et
sampling of the pancreatic juice for cytological exami- al. 2001; CUILLERIER et al. 2000). The prognosis of
nation, and ductal mapping with multiple biopsies for IPMTs depends on the presence of carcinomatous
histological examination (NAVARRO et al. 1999; GIGOT invasion at the time of resection (KANAZUMI et al.
et al. 2001). 2001; KIMURA et al. 1998). Therefore, a careful dis-
section and a complete submission of the resected
specimen are mandatory to search for the presence
of microscopically invasive carcinoma. The type
6.6 of carcinomatous invasion seems to be significant
Differential Diagnosis since 'colloid' -type carcinoma (Fig. 6.14) has a more
favorable prognosis than 'tubular' -type carcinoma.
The differential diagnosis of IPMT is MCT, chronic However, patients with 'tubular'-type invasive carci-
pancreatitis with ductal papillary hyperplasia, and noma (Fig. 6.15) follow a better course than patients
ductal adenocarcinoma. It is quite easy to differ- with conventional ductal carcinoma (ADSAY et al.
entiate IPMT from most cystic lesions, but dis- 2002). The prognosis is also influenced by the type
tinguishing the 'ductectatic mucin-hypersecreting of surgery (total pancreatectomy vs partial pancre-
variant' of IPMT from MCT may be difficult. This atectomy; FUKUSHIMA et al. 2001; SEIDEL et al. 2002;
histological similarity has led some pathologists to CUILLERIER et al. 2000). Extrapancreatic invasion,
interpret MCT and IPMT as a single entity (ITAI lymph node metastases, venous invasion, and peri-
et al. 1986). A clear distinction between these two neural invasion, although less frequent than in ductal
tumors therefore requires precise definitions: the carcinoma (SOHN et al. 2001), are all indicators of a
growth within the pancreatic duct system (dem- poor prognosis (FUKUSHIMA et al. 2001; CUILLERIER
onstrated on ERCP, MRCP with macroscopic and et al. 2000; SUGIYAMA and ATOMI 1998).
microscopic confirmation) and the absence of a Among the IPMTs with a good prognosis are those
subepithelial ovarian-type stroma typically pres- originating in the branch ducts instead of the main
ent in MCT (ZAMBONI et al. 1999). The differential duct (TERRIS et al. 2000). However, there is no com-
diagnosis with chronic pancreatitis may be difficult plete agreement: in situ and invasive carcinoma have
in the early stages of IPMT, in which the only histo- been reported in branch duct type IPMTs (YAMADA
logical modifications are the papillary hyperplasia et al. 1991; FALCONI et al. 2001; KANAZUMI et al.
and a mild dilatation of the pancreatic duct system 2001; FUKUSHIMA et al. 2001; KOBARI et al. 1999;
(BASTID et al. 1991). Such a misdiagnosis causes NAKAGOHRI et al. 1999). The dimension of the lesion
delay in administering the appropriate therapy (greater than 3 cm), luminal vegetations, the thick-
and may favor carcinomatous transformation. The ness of the walls, and a dilated main pancreatic duct
diagnosis of ductal adenocarcinoma is usually easy are currently considered as negative prognostic fac-
because only a solid mass is present. There may tors and suggest surgical resection (WAKABAYASHI et
be some problems with ductal carcinoma associ- al. 2001; LIM et al. 2001).
ated with marked ductal ectasia (secondary to the
obstruction) or with a relevant intraductal compo-
nent, which is, however, detected only at a micro-
scopic level (ADSAY and KLIMSTRA 2000). References
Adsay N, Klimstra D (2000) Cystic forms of typically solid
pancreatic tumors. Semin Diagn PathoI17:81-88
Adsay N,Adair C, Heffes C, Klimstra D (1996) Oncocytic pap-
6.7 illary mucinous cystic tumor of the pancreas. Am J Surg
Pathol 20:980-994
Prognosis
Adsay NV, Pierson C, Sarkar F, Abrams J, Weaver D, Conlon
KC, Brennan MF, Klimstra DS (2001) Colloid (mucinous
Although IPMTs are generally considered slow-grow- noncystic) carcinoma of the pancreas. Am J Surg Pathol
ing tumors, approximately 30%-50% of cases may 25:26-42
94 G. Zamboni et al.

Adsay NV, Conlon KC, Zee SY, Brennan MF, Klimstra DS Iacobuzio-Donahue CA, Klimstra DS, Adsay NV, Wilentz RE,
(2002) Intraductal papillary-mucinous neoplasms of the Argani P, Sohn TA, Yeo q, Cameron JL, Kern SE, Hruban
pancreas: an analysis of in situ and invasive carcinomas in RH (2000) Dpc-4 protein is expressed in virtually all
28 patients. Cancer 94:62-77 human intraductal papillary mucinous neoplasms of the
Azar C, Van de Stadt J, Rickaert F, Deviere M, Baize M, Kloppel pancreas: comparison with conventional ductal adenocar-
G, Gelin M, Cremer M (1996) Intraductal papillary muci- cinomas. Am J PathoI157:755-761
nous tumors of the pancreas. Clinical and therapeutic Itai Y, Ohhashi K, Nagai H, Murakami Y, Kokubo T, Makita
issues in 32 patients. Gut 39:457-464 K, Ohtomo K (1986) Ductectatic mucinous cystadenoma
Bastid C, Bernard J, Sarles H, Payan M, Sahel J (1991) Muci- and cystadenocarcinoma of the pancreas. Radiology 161:
nous ductal ectasia of the pancreas: a premalignant disease 697-700
and a cause of obstructive pancreatitis. Pancreas 6:15-22 Izawa T, Obara T, Tanno S, Mizukami Y, Yanagawa N, Kohgo
Bresler L, Gay G, Boissel P, Grosdidier J (1992) Villous Y (2001) Clonality and field cancerization in intraductal
adenomatosis of duct of Wirsung revealed by Wirsungor- papillary-mucinous tumors of the pancreas. Cancer 92:
ragie: evolution and surgical management. Dig Dis Sci 37: 1807-1817
470-473 Kanazumi N, Nakao A, Kaneko T, Takeda S, Harada A, Inoue
Conley C, Scheithauer B, Weiland L, van Heerden J (1987) Dif- S, Nagasaka T, Nakashima N (2001) Surgical treatment of
fuse intraductal papillary adenocarcinoma of the pancreas. intraductal papillary-mucinous tumors of the pancreas.
Ann Surg 205:246-249 Hepatogastroenterology 48: 967-971
Cuillerier E, Cellier C, Palazzo L, Deviere 1, Wind P, Rickaert Kimura W, Makuuchi M, Kuroda A (1998) Characteristcs and
F, Cugnenc PH, Cremer M, Barbier JP (2000) Outcome treatment of mucin-producing tumor of the pancreas.
after surgical resection of intraductal papillary mucinous Hepatogastroenterology 45:2001-2008
tumors of the pancreas. Am J Gastroenterol 95:441-445 Kloppel G (1998) Clinicopathologic view of intraductal papil-
Dai J, Turnacioglu K, Schuttle M, Sugar A, Kern S (1998) Dpc4 lary-mucinous tumor of the pancreas. Hepatogastroenter-
transcriptional activation and dysfunction in cancer cells. ology 45:1981-1985
Cancer Res 58:4592-4597 Kliippel G, Solcia E, Longnecker D, Capella C, Sobin L (1996)
Falconi M, Salvia R, Bassi C, Zamboni G, Talamini G, Peder- Histological typing of tumors of the exocrine pancreas.
zoli P. (2001) Clinicopathological features and treatment of International histological classification of tumors. Springer,
intraductal papillary mucinous tumor of the pancreas. Br Berlin Heidelberg New York
J Surg 88:376-381 Kobari M, Egawa S, Shibuya K (1999) Intraductal papillary
Fukushima N, Mukai K, Sakamoto M, Hasebe T, Shimada K, mucinous tumors of the pancreas comprise 2 clinical sub-
Kosuge T, Kinoshita T, Hirohashi S (2001) Invasive carci- types: differences in clinical characteristics and surgical
noma derived from intraductal papillary-mucinous carci- management. Arch Surg l34: 1131-1l36
noma of the pancreas: clinicopathologic and immunohis- Kobayashi G, Fujita N, Mochizuki F, Yamazaki T (1993) Study
tochemical study of eight cases. Virchows Arch 439:6-l3 of cases of mucin producing tumors of the pancreas show-
Furukawa T, Takahashi T, Kobari M, Matsuno S (1992) The ing penetration of other organs (in Japanese). Nippon Sho-
mucus-hypersecreting tumor of the pancreas. Develop- kakibyo Gakkai Zasshi 90:3081-3089
ment and extension visualized by three-dimensional com- Koito K, Namieno T, Nagakawa T, Hirokawa N, Ichimura T,
puterized mapping. Cancer 70:1505-15l3 Syonai T, Yam a N, Someya M, Nakata K, Sakata K, Hareyama
Furukawa T, Ohhashi K, Yamao K, Naitoh Y, Hirooka Y, Taki M (2001) Pancreas: imaging diagnosis with color/power
T, Itoh A, Hayakawa S, Watanabe Y, Goto H, Hayakawa T Doppler ultrasonography, endoscopic ultrasonography,
(1997) Intraductal ultrasonography of the pancreas: devel- and intraductal ultrasonography. Eur Radiol 38:94-104
opment and clinical potential. Endoscopy 29:561-569 Kurihara K, Nagai H, Kasahara K, Kanazawa K, Kanai N (2000)
Gigot JF, Deprez P, Sempoux C, Descamps C, Metairie S, Glineur Biliopancreatic fistula associated with intraductal papil-
D, Gianello P (2001) Surgical management of intraductal lary-mucinous pancreatic cancer: institutional experience
papillary mucinous tumors of the pancreas: the role of rou- and review of the literature. Hepatogastroenterology 47:
tine frozen section of the surgical margin, intraoperative 1164-1167
endoscopic staged biopsies of the Wirsung duct, and pan- Lim JH, Lee G, Oh YL (2001) Radiologic spectrum of intra-
creaticogastric anastomosis. Arch Surg l36:1256-1262 ductal papillary mucinous tumor of the pancreas. Radio-
Hahn SA, Schutte M, Hoque A, Moskaluk CA, da Costa LT, graphies 21: 323-337; discussion 337-340
Rozenblum E, Weinstein CL, Fischer A, Yeo C1, Hruban RH, Loftus EV Jr, Olivares-Pakzad BA, Batts KP, Adkins MC,
Kern SE (1996) DPC4, a candidate tumor suppressor gene Stephens DH, Sarr MG, DiMagno EP (1996) Intraductal
at human chromosome 18q21.1. Science 271:350-353 papillary-mucinous tumors of the pancreas: clinicopatho-
Hara T, Yamaguchi T, Ishihara T, Tsuyuguchi T, Kondo F, Kato logic features, outcome, and nomenclature. Members of the
K, Asano T, Saisho H (2002) Diagnosis and patient man- Pancreas Clinic, and Pancreatic Surgeons of Mayo Clinic.
agement of intraductal papillary-mucinous tumor of the Gastroenterology 11 0: 1909-1918
pancreas by using peroral pancreatoscopy and intraductal Longnecker DS (1998) Observations on the etiology and patho-
ultrasonography. Gastroenterology 122:34-43 genesis of intraductal papillary- mucinous neoplasms of
Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Gar- the pancreas. Hepatogastroenterology 45: 1973-1980
rett ES, Goodman SN, Kern SE, Klimstra DS, Kloppel G, Luttges J, Zamboni G, Longnecker D, Kloppel G (2001) The
Longnecker DS, Luttges 1, Offerhaus GJ (2001) Pancreatic immunohistochemical mucin expression pattern distin-
intraepithelial neoplasia: a new nomenclature and clas- guishes different types of intraductal papillary mucinous
sification system for pancreatic duct lesions. Am J Surg neoplasms of the pancreas and determines their relation-
Pathol 25:579-586 ship to mucinous noncystic carcinoma and ductal adeno-
Pathology of Intraductal Cystic Tumors 95

carcinoma. Am J Surg Pathol 25:942-948 Sohn TA, Yeo CJ, Cameron JL, Iacobuzio-Donahue CA, Hruban
Morohoshi T, Kanda M, Asanuma K, Kleppel G (1989) Intra- RH, Lillemoe KD (2001) Intraductal papillary mucinous
ductal papillary neoplasms of the pancreas. A clinicopatho- neoplasms of the pancreas: an increasingly recognized
logic study of six patients. Cancer 64: 1329-1335 clinicopathologic entity. Ann Surg 234:313-321; discussion
Nagasaka T, Nakashima N (2001) Problems in histologi- 321-312
cal diagnosis of intraductal papillary-mucinous tumor Solcia E, Capella C, Kleppe! G (1997) Tumors of the pancreas.
(IPMT). Hepatogastroenterology 48:972-976 AFIP Atlas of tumor pathology, vol 20. Armed Forces Insti-
Nakagohri T, Kenmochi T, Kainuma 0, Tokoro Y, Asano T tute of Pathology, Washington DC
(1999) Intraductal papillary mucinous tumors of the pan- Sugiura H, Kondo S, Islam HK, Ito K, Ono K, Morikawa T,
creas. Am J Surg 178:344-347 Okushiba S, Katoh H (2002) Clinicopathologic features and
Navarro F, Michel J, Bauret P, Ramos J, Blanc P, Fabre JM, outcomes of intraductal papillary-mucinous tumors of the
Millat B, Desrousseaux B, Domergue J (1999) Management pancreas. Hepatogastroenterology 49:263-267
of intraductal papillary mucinous tumours of the pancreas. Sugiyama M, Atomi Y (1998) Intraductal papillary mucinous
Eur J Surg 165: 43-48 tumors of the pancreas: imaging studies and treatment
Obara T, Maguchi H, Saitoh Y, Ura H, Koike Y, Kitazawa S, strategies. Ann Surg 228:685-691
Namiki M (1991) Mucin-producing tumor of the pancreas: Tada M, Ornata M, Ohto M (1991) Ras gene mutations in intra-
a unique clinical entity. Am J Gastroentrol 86: 1619-1625 ductal papillary neoplasms of the pancreas. Analysis in five
Ohhashi K, Murakami Y, Maruyama M (1982) Four cases of cases. Cancer 67:634-637
mucous secreting pancreatic cancer (in Japanese). Prog Dig Terris B, Ponsot P, Paye F, Hammel P, Sauvanet A, Molas G,
Endosc 20: 348-351 Bernades P, Belghiti J, Ruszniewski P, Fiejou JF (2000)
Payan M, Xerri L, Moncada K, Bastid C, Agostini S, Sastre Intraductal papillary mucinous tumors of the pancreas
B, Sahel J, Choux R (1990) Villous adenoma of the main confined to secondary ducts show less aggressive patho-
pancreatic duct: a potentially malignant tumor? Am J Gas- logic features as compared with those involving the main
troenterol 85: 459-463 pancreatic duct. Am J Surg PathoI24:1372-1377
Rickaert F, Cremer M, Deviere J, Tavares L, Lambilliotte JP, Traverso LW, Peralta EA, Ryan JA Jr, Kozarek RA (1998) Intra-
Schroder S, Wurbs D, Kloppel G (1991) Intraductal mucin- ductal neoplasms of the pancreas. Am J Surg 175:426-432
hypersecreting neoplasms of the pancreas. A clinicopatho- Wakabayashi T, Kawaura Y, Morimoto H, Watanabe K, Toya D,
logic study of eight patients. Gastroenterology 101:512-519 Asada Y, Satomura Y, Watanabe H, Okai T, Sawabu N (2001)
Rivera JA, Fernandez-del Castillo C, Pins M, Compton CC, Clinical management of intraductal papillary mucinous
Lewandrowski KB, Rattner DW, Warshaw AL (1997) Pan- tumors of the pancreas based on imaging findings. Pan-
creatic mucinous ductal ectasia and intraductal papillary creas 22:370-377
neoplasms. A single malignant clinicopathologic entity. Wilentz RE, Su GH, Dai JL, Sparks AB, Argani P, Sohn TA, Yeo
Ann Surg 225:637-644; discussion 644-636 CJ, Kern SE, Hruban RH (2000) Immunohistochemistry
Sato N, Rosty C, Jansen M, Fukushima N, Ueki T, Yeo CJ, Cam- labeling for Dpc4 mirrors genetic status in pancreatic and
eron JL, Iacobuzio-Donahue CA, Hruban RH, Goggins M peripancreatic adenocarcinomas: a new marker of DPC4
(2001) STKll/LKB1 Peutz-Jeghers gene inactivation in inactivation. Am J PathoI156:37-43
intraductal papillary-mucinous neoplasms of the pancreas. Yamada M, Kozuka S, Yamano K, Nakazawa S, Naitoh Y, Tsu-
Am J PathoI159:2017-2022 kamoto Y (1991) Mucin-producing tumor of the pancreas.
Seidel G, Zahurak M, Iacobuzio-Donahue C, Sohn TA, Adsay Cancer 68:159-168
NV, Yeo CJ, Lillemoe KD, Cameron JL, Hruban RH, Wilentz Yanagisawa A, Kato Y, Ohtake K, Kitagawa T, Ohashi K, Hori M,
RE (2002) Almost all infiltrating colloid carcinomas of Takagi K, Sugano H (1991) c-Ki-ras point mutations in duc-
the pancreas and periampullary region arise from in situ tectatic-type mucinous cystic neoplasms of the pancreas.
papillary neoplasms: a study of 39 cases. Am J Surg Pathol Jpn J Cancer Res 82:1057-1060
26:56--63 Zamboni G, Scarpa A, Bogina G, Iacono C, Bassi C, Talamini
Sessa F, Solcia E, Capella C, Bonato M, Scarpa A, Zamboni G, G, Sessa F, Capella C, Solcia E, Rickaert F, Mariuzzi GM,
Pellegata NS, Ranzani GN, Rickaert F, Kloppel G (1994) Kloppel G (1999) Mucinous cystic tumors of the pancreas:
Intraductal papillary-mucinous tumors represent a dis- clinicopathological features, prognosis, and relationship
tinct group of pancreatic neoplasms: an investigation of to other mucinous cystic tumors. Am J Surg Pathol 23:
tumor cell differentiation and K-ras, p53 and c-erbB-2 410-422
abnormalities in 26 patients. Virchows Arch 425:357-367 Z'Graggen K, Rivera JA, Compton CC, Pins M, Werner J,
Sho M, Nakajima Y, Kanehiro H, Hisanaga M, Nishio K, Nagao Fernandez-del Castillo C, Rattner DW, Lewandrowski KB,
M, Ikeda N, Kanokogi H, Yamada T, Nakano H (1998) Pat- Rustgi AK, Warshaw AL (1997) Prevalence of activating
tern of recurrence after resection for intraductal papil- K-ras mutations in the evolutionary stages of neoplasia
lary mucinous tumors of the pancreas. World J Surg 22: in intraductal papillary mucinous tumors of the pancreas.
874-878 Ann Surg 226:491-498; discussion 498-500
7 Intraductal Papillary Mucinous Tumors: Imaging
C. PROCACCI, G. SCHENAL, E. DELLA CHIARA, A. FUINI, and A. GUARISE

CONTENTS al. 1986,1987; BASTID et al. 1991; YAMADA et al. 1991;


OBARA et al. 1993a,b; YANAGISAWA et al. 1993; RAT-
7.1 Introduction 97 TNER and PINS 1994; RAIJMAN et al. 1994). In 1997,
7.2 IPMT of the Main Duct 98 the tumor was classified with other cystic tumors of
7.2.1 Transabdominal US 98
7.2.2 Computed Tomography 101 the pancreas, and the name of 'intraductal papillary
7.2.3 Magnetic Resonance Imaging 106 mucinous tumor' was proposed as the proper term
7.2.4 Endoscopic Retrograde referring to these lesions. The name emphasizes the
Cholangiopancreatography 11 0 neoplasm's intraductal location, the macroscopic
7.2.5 Endoscopic US 111
papillary architecture, and the abnormal production
7.3 IPMT of the Collateral Ducts 113
7.3.1 Transabdominal US 113 of mucin (SOLCIA et al. 1997; WADE et al. 1997).
7.3.2 Computed Tomography 114 Reports of this neoplasm have been few for many
7.3.3 Magnetic Resonance Imaging 116 years because the lesion has often been misdiagnosed
7.3.4 Endoscopic Retrograde as chronic pancreatitis when its origin is in the main
Cholangiopancreatography 120 duct (AGOSTINI et al. 1989; AZAR et al. 1996; TENNER
7.3.5 Endoscopic US 122
7.4 IPMT of the Mixed Type 123 et al. 1996; PROCACCI et al. 1999; CASADO et al. 2000)
7.5 Differential Diagnosis 124 or has often been included in the mucinous cystic
7.6 Biological Behavior and Recurrences 129 tumor group when its origin is in the collateral ducts
References 135 (arising from the parenchyma as opposed to the
duct epithelium) with which it shares its histologi-
cal characteristics (OHTA et al. 1992; TENNER et al.
1996; SOLCIA et al. 1997; SUGIYAMA et al. 1997). In
7.1 the past 10 years, however, recognition of this neo-
Introduction plasm has increased such that it can be considered
the most frequently occurring cystic tumor of the
The first description of the radiological appearance pancreas. Furthermore, the literature underestimates
of intraductal papillary mucinous tumors (IPMTs) the prevalence of the tumor because not all cases
dates back to 1982 when OHHASHI and co-authors are reported (PROCACCI et al. 1999,2001; LIM et al.
(OHHASHI et al. 1982) described the lesion on endo- 2001). The widespread use of imaging techniques,
scopic retrograde cholangiopancreatography (ERCP). particularly US and CT, and the greater familiarity
Since then, this tumor has been called many different with the typical imaging findings is the most sig-
names: mucinous ductal ectasia, ductectatic muci- nificant reason that the lesion is being recognized
nous cystic tumor, mucinous villous adenomatosis, increasingly often (OHTA et al. 1992; YANAGISAWA et
intraductal mucin-hypersecreting neoplasm (ITAI et al. 1993; KOBAYASHI et al. 1995).
IPMT develops exclusively from the epithelial lining
of pancreatic ducts. The macroscopic characteristics
C. PROCACCI, MD; G. SCHENAL, MD; E. DELLA CHIARA, MD
Istituto di Radiologia, Dipartimento di Scienze Morfologico- and the corresponding appearance at imaging vary sig-
Biomediche, Universita degli Studi di Verona, Policlinico nificantly depending on whether the tumor is located
'Giambattista Rossi', P.zza L.A. Scuro lO, 37134 Verona, Italy in the main duct, the collateral ducts, or both (ITAI et
A. FUINI,MD al. 1986, 1987; MOROHOSHI et al. 1989; FURUKAWA et
Servizio di Gastroenterologia, Ospedale Civile Maggiore, P.le
al. 1992; PROCACCI et al. 1996, 1999). The radiologist's
Stefani, 1,37124 Verona, Italy
A. GUARISE, MD job is to recognize the lesion as soon as possible, to
Servizio di Radiologia, Ospedale S. Cuore-Don Calabria, Via D. give an evaluation of its extent, and whenever possible,
Sempreboni, 37024 Negrar, Italy to express a judgment on its biological behavior. The
98 C. Procacci et al.

tumors can vary histologically from hyperplasia to involved (Fig. 7.1b), it is difficult to distinguish the
invasive carcinoma and are classified by the Armed dilatation from chronic pancreatitis (AGOSTINI et al.
Forces Institute of Pathology (AFIP) in the 'benign', 1989; AZAR et al. 1996; TENNER et al. 1996; PROCACCI
'borderline', and 'malignant' categories. Frequently, et al. 1999). The prompt diagnosis of IPMT of the
multiple forms of epithelial change are present in the main duct is of paramount importance, because
same lesion (FURUKAWA et al. 1992). The prognosis when these tumors involve the main duct to any
of this tumor is significantly more favorable than that degree, they must be considered malignant. Patients
of a ductal adenocarcinoma (MOROHOSHI et al. 1989; should be immediately evaluated for possible surgical
TENNER et al. 1996; LOFTUS et al. 1996; WADE et al. resection, which unfortunately, in the more advanced
1997; CELLIER et al. 1998; PROCACCI et al. 1999; YAMAO stages, can advocate a total pancreatectomy (TENNER
et al. 2000; KANEKO et al. 2001). Even patients with the et al. 1996; KOBARI et al. 1999; LIM et al. 2001).
malignant form who are adequately treated by surgery
can have a significant long-term survival (RAIJMAN
et al. 1994; MADURA et al. 1997; CASADO et al. 2000). 7.2.1
When, however, there is the least suspicion of a benign Transabdominal US
lesion, the management of this tumor need not be sur-
gical, and the choice of follow-up can be opted for. The primary objective of transabdominal US is to iden-
The imaging findings of each of the three tumor tify the lesion; once an abnormally dilated pancreatic
forms (tumor of the main duct, of the collateral duct is identified, the differential diagnostic consider-
branches, and of both) will be discussed individu- ations are limited (PAVONE et al. 1997; LIM et al. 2001;
ally. Following this, the differential diagnoses will be KANEKO et al. 2001; SILAS et al. 2001). The major limi-
presented. In the conclusion, the potential of imaging tation with the US examination is that excessive bowel
in defining the biological behavior of these tumors gas can limit a complete visualization of the pancreas
and the problems encountered at follow-up will be in about 25% of patients (PAVONE et al. 1997).
analyzed. Dilation of the main duct is usually demonstrated
(ITAI et al. 1987; MADURA et al. 1997; WADE et al. 1997;
KANEKO et al. 2001). In the segmental form, the dila-
tion can be in the pancreatic head (Fig. 7.2a) or, less
7.2 frequently, in the tail (Fig. 7.2b) (PROCACCI et al. 1999).
IPMT of the Main Duct Suspicion of segmental IPMT is mostly based on the
absence of an obstructing mass and the absence of
Main duct IPMT presents as segmental (early) or a history of episodes of acute pancreatitis that could
diffuse (late) dilatation of the main pancreatic duct. lead to the suspicion of a postinflammatory stenosis
Segmental IPMT is the most difficult to diagnose (DABEZIES et al. 1990). The pancreatic parenchyma usu-
(Fig. 7.1a). Once the entire duct becomes diffusely ally has normal thickness, as long as there is no cystic

a b

Fig. 7.1a,b. Intraductal papillary mucinous tumor (IPMT) of the main duct. a Segmental lesion: ectasia of the main duct, involv-
ing a portion of the main duct. b Diffuse form: cystic ectasia of the collateral ducts is associated with dilatation of the entire
main duct. The dilated papilla protrudes into the duodenal lumen
Intraductal Papillary Mucinous Tumors: Imaging 99

a b

c d

e f

Fig.7.2a-f. IPMT of the main duct, segmental type (a, b) and diffuse type (c-f) (US evaluation). a Segmental dilation of the
main duct in the pancreatic head is shown in the axial scan. The content is inhomogeneously hyperechoic due to the presence
of mucin. b Another patient. Cystic ectasia of the distal tract of the duct of Wirsung is shown in the axial scan; within, there are
some hyperechoic formations attributed to mucin deposits and papillary proliferations. c, d Axial image shows marked dilation
of the main duct (calipers in c); both the Santorini and Wirsung ducts (arrows in d) are dilated. e, fDifferent patient. Dilation of
the duct of Wirsung in the body-tail is shown in the axial scans and cystic ectasia of the portion of the duct in the pancreatic
head within which echoic formations (arrows) can be seen, presumably due to papillary growth
100 C. Procacci et al.

dilatation of the duct, with resulting adjacent parenchy- nant degeneration of the tumor can be presumed. In
mal atrophy (PROCACCI et al. 1999). The duct's cystic this case, usually the neoplastic layer is indistinguish-
transformation (Fig. 7.2b) has sometimes been reported able from the residual, infiltrated parenchyma.
in the tail (KANEKO et al. 2001; LIM et al. 2001). With transabdominal US, it is not possible to docu-
In the diffuse form, the whole duct is dilated ment the dilated, protruding duodenal papilla seen
(Fig. 7.2c,d), to a greater or lesser degree, depend- frequently at endoscopy (PROCACCI et al. 1999). It is
ing on the time of onset of the neoplasm (PROCACCI easy to recognize cystic ectasia of the collateral ducts
et al. 1999). In this event, cystic ectasia of the duct which appears in the form of small, multiple, anechoic
is likely, more commonly in the head of the gland, structures located near the main duct or, more com-
associated with mild dilation of the tract upstream monly, grouped in bunches in the uncinate process
(Fig. 7.2e,f). Obviously, in this case, it is difficult to (Fig. 7.3a,b) (KANEKO et al. 2001).
establish whether the whole duct is affected by the The content of the dilated main or collateral ducts
neoplasm or if the cephalic tract neoplasm is associ- is usually transonic but can be inhomogeneous
ated with the dilation of the tract upstream simply because of the frequent presence of echogenic areas
due to obstruction. located in the sloping portion of the duct or near
Parenchymal atrophy is usually proportional to the the walls (Fig. 7.2) (TAKI et al. 1997). It is not always
ductal dilation (OBARA et al. 1993a). When the mark- possible to establish with US if these echo genic
edly dilated duct is bordered by a thick, echogenic formations are produced by mucin globules or by
wall (Fig. 7.2a) displaying luminal irregularity, malig- papillary proliferations (KANEKO et al. 2001). The

a b

c d

Fig. 7.3a-d. IPMT of the main duct (US evaluation) a, b In the axial scans, dilation of the main duct (calipers in a) is demon-
strated with transonic content and cystic ectasia of the collateral ducts of the uncinate process (calipers in b). c, d Another
patient. Significant dilatation of the entire main duct in which intensely echoic formations are seen in the dependent position
(asterisk in c), attributed to mucin and other less echoic formations (arrows in c, d) attributed to papillary growths
Intraductal Papillary Mucinous Tumors: Imaging 101

papillary growths, however, tend to be less echogenic Patients with known or suspected IPMT should be
than mucin (Fig. 7.3c,d); these growths have a height studied utilizing a dedicated pancreatic technique.
of between 1.5 and 21 mm, with an average value of The details of this technique are elaborated in Sec-
6.7 mm (FUKUSHIMA et al. 1997; KANEKO et a1. 2001). tion 3.2.2.1 and the following.
Rarely, one can differentiate luminal projections On non-IV contrast images, it is possible to evalu-
from mucin by varying the patient's position: mucin ate the gland's morphology, identifying the localized
can be seen to vary in position within the duct, while increase in gland size due to the cystic ectasia of
the papillary projections remain fixed (KANEKO et a1. the main duct or of the collateral ducts (FUKUKURA
2001). In some cases, the large volumes of mucinous et a1. 2000). In this phase, if the duodenal lumen
concretions within the ductal content can render has been distended using an oral radiopaque con-
the appearance of the dilated duct echogenic, and trast medium, it may be possible to recognize the
because the dilated ductal lumen is not distinguish- hypo dense protruding papilla (Fig. 7.4a) (PROCACCI
able from the adjacent parenchyma, the entire lesion et a1. 1999; FUKuKuRA et al. 2000). If water is used
may go undiagnosed (PROCACCI et a1. 1996). Careful to identify the duodenal lumen, this feature may
attention to 'gain' settings is necessary so that this not be recognized at this stage of the examination.
error may be avoided. Sometimes the echogenicity of Calcifications may be most apparent on non-IV
these intraductal mucinous formations can be par- contrast-enhanced images. Calcification located at
ticularly high in relation to the deposits of calcium the margin of the duct with the parenchyma may be
within them (ITAI et a1. 1987; PROCACCI et a1. 1996); due to superimposed chronic pancreatitis (Fig. 7.5)
nonetheless, the posterior shadow is not appreciable. (PROCACCI et al. 1996), whereas those located centrally
result from deposits of calcium within the mucin glob-
ules (Fig. 7.6) (TAOUL! et al. 2000).
7.2.2 On IV contrast-enhanced images, the lesion is
Computed Tomography better outlined against the bright background of
contrast-enhanced parenchyma (ITOH et a1. 1992).
CT has contributed significantly to the improved In this phase, it is possible to define the parenchymal
recognition of IPMT, even surpassing ERCP as the thickness and the extent of the parenchymal atrophy
primary imaging test for diagnosis. CT recognition (ITAI et a1. 1987; RICKAERT et a1. 1991; ITOH et a1.
has improved with the advent of helical/spiral data 1992; PROCACCI et a1. 1996). The main duct, vari-
acquisition (FUKUKURA et a1. 2000; TAOUL! et a1. ably dilated, appears distinctly hypodense compared
2000). It is presumed that a further significant diag- with the glandular tissue (Figs. 7.5, 7.7-7.9). Careful
nostic improvement will be gained from using the inspection reveals that the lumen is seldom homoge-
multislice technique. neous (Fig. 7.7), since mucin deposits and/or papil-

a b

Fig.7.4a,b. IPMT of the main duct (CT evaluation) a On non-IV contrast images, modest enlargement of the pancreatic head
and a large defect (arrow) in the duodenal lumen, opacified by diluted oral contrast (Gastrografin), representing a dilated major
papilla are demonstrated. b On IV contrast-enhanced images, significant ectasia of the main duct is shown in direct continuity
with the dilated papilla protruding into the duodenal lumen that, in this phase, is distended by the hypodense contents (water)
102 C. Procacci et a1.

a b

c d

Fig. 7.5a-d. IPMT of the main duct (CT evaluation). Axial scans, carried out in a craniocaudal direction with IV contrast, reveal
mild ectasia of the duct of Wirsung in the body-tail (a) and cystic ectasia of the duct within the pancreatic head (b-d). The
major papilla (arrowhead) and minor papilla (arrow) bulge into the duodenal lumen. Multiple parenchymal calcifications are
present, presumably secondary to chronic obstructive pancreatitis

lary proliferations can be revealed, characterized


by higher density {Fig. 7.8} {FUKUKURA et al. 2000}.
Nonenhancing mucin deposits can therefore be dis-
tinguished from contrast-enhancing papillary prolif-
erations. Alternatively, CT can differentiate between
mucin and papillary proliferations by changing the
patient's position (KANEKO et al. 2001; PROCACCI et
al. 2001).
Pancreatic ductal dilation is accompanied by a
proportional thinning and atrophy of the parenchyma
{Figs. 7.7, 7.8}. Parenchymal calcifications may appear
in long-standing cases (ITAI et al. 1987; RICKAERT et al.
1991; ITOH et al. 1992; PROCACCI et al. 1996; SUGIYAMA
and ATOMI 1999). Whenever significant ductal dilation
with normal or even increased parenchymal thickness
Fig. 7.6. IPMT of the main duct (CT evaluation). The axial is present, one must assume malignant degeneration
scan, carried out with delayed IV contrast-enhanced images, of the tumor {Fig. 7.9}. The suspicion of malignancy
demonstrates marked cystic-type dilatation of the main duct is also supported by the presence of multiple papillary
in the body-tail, with significant atrophy of the parenchyma
surrounding the lesion. Within the duct, there are multiple proliferations {Fig. 7.8}. Even if a tumor in its advanced
amorphous calcifications, presumably located in the mucin stages invades the pancreatic parenchyma, the cystic
deposits component remains recognizable {Fig. 7.9}, which
Intraductal Papillary Mucinous Tumors: Imaging 103

_ _ ___ d
c

e f

Fig.7.7a-f. IPMT of the main duct (CT evaluation). a, b First patient. Significant dilation of the distal duct of Wirsung with
protrusion of the dilated papilla (arrow in b) into the duodenum (d). The duct ofWirsung is not recognizable in the body-tail.
c-f Different patient. Significant dilatation of the main duct in the portion between the pancreatic neck (arrow in c) and the
papilla which, dilated, protrudes into the duodenal lumen (arrow in e); there is coexisting cystic ectasia of the collateral ducts
of the uncinate process (arrows in f)
a b

c d
Fig. 7.8a-d. IPMT of the main duct (CT evaluation). In the IV contrast-enhanced images obtained in the craniocaudal direc-
tion, significant dilatation of the duct of Wirsung and dilation of the papilla (long arrow in d) is present. Marked parenchymal
atrophy is also present. Multiple small formations are recognizable originating on the ductal walls (small arrows) , corresponding
to papillary proliferations

a b

Fig.7.9a,b. IPMT of the main duct (CT evaluation). Modest dilatation of the duct of Wirsung in the body-tail. Thick-walled,
cystic dilation of the duct in the head of the gland is also appreciable
Intraductal Papillary Mucinous Tumors: Imaging 105

makes it possible to distinguish IPMT of the main above-mentioned findings (ductal dilation with
duct from the classic ductal adenocarcinoma (ITOH mucin deposits or papillary proliferations: Fig. 7.8),
et al. 1992). The component which invades the paren- there are another two helpful findings. First, cystic
chyma will demonstrate greater enhancement than ectasia of the collateral ducts is more or less con-
the atrophic and fibrotic parenchyma that delimits its stant, even if modest, above all in the uncinate
exterior (ITOH et al. 1992). process (Fig. 7.70. Second, dilation of the major
In the segmental form, the CT picture is more often and/or minor papillae (Figs. 7.7e, 7.8d, 7.12c,d),
nonspecific (TAOULI et al. 2000; PROCACCI et al. 2001), which protrude into the duodenal lumen (FuKu-
since only the segmental ductal dilation can be docu- KURA et al. 2000), is seen more frequently than in
mented, even when there is no significant parenchymal the segmental form (Figs. 7.5, 7.7b). On IV con-
atrophy (Fig. 7.10). Segmental IPMT seldom appears trast-enhanced images, when the duodenal lumen
with cystic dilation of the main duct; one will see a has been distended with water, the papilla, with
cystic mass most commonly in the tail (Fig. 7.11). In this a hypo dense content, is perfectly distinguishable
case, in order to avoid the risk of misdiagnosing a muci- from the intraluminal content thanks to the intense
nous cystic tumor, one must demonstrate that the cyst enhancement of the walls (Fig. 7.4b). If, however,
communicates with the duct ofWirsung (Fig. 7.11a). the duodenal lumen has been distended with radi-
With the diffuse form, the diagnosis of IPMT is opaque contrast medium, the papilla will appear as
certainly much easier because, in addition to the a hypodense intraluminal mass (Fig. 7.4a).

a _ """-_ _ b

Fig. 7.10a,b. IPMT of the main duct (CT evaluation). a Initial examination shows modest dilatation of the main duct in the tail.
b At a second examination 10 years later, there is more pronounced dilatation of the duct of Wirsung

a b

Fig. 7. 11 a,b. IPMT of the main duct (CT evaluation). Cystic dilatation of the proximal portion of the duct ofWirsung; the cyst
appears to be in continuity with the main duct in the body (a). There is a small mural nodule in the posterior wall of the cystic
formation (arrow in b)
106 C. Procacci et a1.

a b

c _ ..._ __
d

Fig. 7.12a-d. IPMT of the main duct (CT evaluation). IV contrast-enhanced scans carried out in the craniocaudal direction
reveal marked dilation of the main duct with adjacent parenchymal atrophy, most evident in the pancreatic neck (a). There is
coexisting dilation of both minor (arrowhead in c) and major (arrow in d) papillae, both of which protrude into the duodenal
lumen (d)

7.2.3 T2-weighted half-Fourier single-shot turbo spin-echo


Magnetic Resonance Imaging (HASTE) acquisition. The latter is performed in mul-
tiple planes including axial and coronal (ONAYA et al.
Current magnetic resonance (MR) technology, with 1998; ARAKAWA et al. 2000). The oral administration of
the use of fast, breathhold sequences and the possibil- superparamagnetic contrast medium greatly improves
ity of obtaining visualization of the biliary and pan- MRCP quality. The intravenous administration of
creatic ducts, has significantly increased its clinical gadolinium during the GRE Tl-weighted sequences,
utility (SUGIYAMA et al. 1997; SUGIYAMA and ATOMI with a timing analogous to that of the CT IV contrast
1998; SUGIYAMA et al. 1998a; ARAKAWA et al. 2000). phases, improves contrast resolution in the pancreas
MR study of pancreatic diseases requires the utiliza- and allows for preoperative planning by the ability to
tion of un enhanced and IV gadolinium -enhanced gra- create MR angiographic renderings.
dient recalled echo (GRE) Tl-weighted fat-suppressed The unenhanced GRE Tl-weighted sequences give
(FS) sequences, and fast T2-weighted sequences. Added the first indication of the extent (segmental or diffuse)
to these are magnetic resonance cholangiopancreatog- of duct involvement. With segmental IPMT, it is pos-
raphy (MRCP) renderings obtained either with a long sible to demonstrate the typical hypo intense image of
echo train, single-shot thick slab technique or a heavily the dilated duct (Fig. 7.13a). Without IV gadolinium,
Intraductal Papillary Mucinous Tumors: Imaging 107

a b

c d

Fig. 7. 13a-d. IPMT of the main duct (MR evaluation). a On this noncontrast opposed-phase GRE Tl-weighted image, there is a
marked hypointense signal within the pancreatic tail. b, c On images acquired in the craniocaudal direction utilizing a HASTE
T2-weighted sequence, segmental dilatation of the duct of Wirsung with adjacent parenchymal atrophy is demonstrated. Cystic
ectasia of a collateral duct is also present (arrow in b). d MRCP confirms the segmental dilation of the caudal portion of the
duct of Wirsung and the cystic ectasia of a collateral duct (arrow)

the signal of the lumen is not significantly different and/or minor papilla, protruding into the duodenal
from that of the adjacent pancreatic tissue, unless fat lumen, can also be seen (LIM et al. 2001). Moreover,
suppression is employed (Fig. 7.14a,b). In the diffuse with these sequences it is possible to recognize
form, the gland atrophies so that it is often impossible papillary proliferations within the dilated ducts
to distinguish the thin and hypo intense parenchyma (Figs. 7.15b, 7.17), which appear hypointense com-
from the dilated duct lumen with the same signal, pared with the intraductal mucin (IRIE et al. 2000).
especially if there are mucin and/or papillary pro- The nondependent location of these intraluminal
liferations inside the main duct (Fig. 7.15a). Unlike defects confirms their neoplastic nature. The maxi-
with CT, it is not possible to document the presence mum resolution is obtained with HASTE sequences,
of calcifications. where identification of the tiniest intraluminal
The HASTE T2-weighted sequences are acquired defects can be made, which are sometimes underes-
with a 4-mm slice thickness in both the axial and timated with MRCP (IRIE et al. 2000). MRCP, carried
coronal planes. These images are vital in diagnosing out with breath-holding and thick slabs, supplies an
IPMT, since they provide an accurate evaluation of accurate demonstration of the pancreaticobiliary
the duct and its contents (IRIE et al. 2000; ARAKAWA ducts, revealing the location and the extent of the
et al. 2000). The segmental or diffuse dilation main duct dilation (Figs. 7.13d, 7.14e, 7.16b) and the
(Figs. 7.13b,c, 7.14c,d, 7.16a) of the main duct and associated collateral ducts dilation (ARAKAWA et al.
the possible coexistence of cystic ectasia of the 2000). It also allows for the recognition of dilatation
collateral ducts are visible (SUGIYAMA et al. 1998; of the duodenal papilla. Documentation of possible
ARAKAWA et al. 2000). The dilation of the major intraluminal defects is less accurate. Both small pap-
108 C. Procacci et al.

c d

Fig. 7.14a-e. rPMT of the main duct (MR evaluation). a, b Axial


non contrast GRE FS II-weighted image reveals segmental
dilatation of the duct of Wirsung in the head and neck of the
pancreas. Decreased signal is apparent within the dilated duct
compared with the parenchyma. There is also modest dilation
of the common bile duct (arrow in b). c, d Axial HASTE T2-
weighted images (same region as a, b): the above-mentioned
findings are confirmed. Within the common bile duct, a
fluid-fluid level image is recognizable due to the presence of
layering sludge (arrow in d). e MRCP confirms the segmental
dilatation of the duct ofWirsung with associated cystic ectasia
of the collateral branches in the uncinate process (arrow). The
e common bile duct is slightly and regularly dilated

illary proliferations and mucin deposits located within The GRE Tl-weighted sequences, carried out in
a dilated duct can, in fact, be hidden by volume aver- the IV contrast phase, allow for the analysis of the
aging within the hyperintense signal within the thick enhancement and preservation of the pancreatic
slab image. The use of diffusion-weighted echo-planar parenchyma (Fig. 7.16c,d). Just like with spiral CT in
imaging has been proposed to differentiate the mucin the IV contrast phase, it is even possible to recognize
from the pancreatic juice (YAMASHITA et al. 1998), but the presence of hard, neoplastic rind and/or papillary
not all the authors agree on that (IRIE et al. 2002). proliferations, which show intense enhancement.
Intraductal Papillary Mucinous Tumors: Imaging 109

a b

Fig. 7.15a,b. IPMT of the main duct (MR evaluation). a Axial noncontrast opposed-phase GRE Tl-weighted image. The pan-
creas appears homogeneously hypointense. b Axial SE T2-weighted image. The duct of Wirsung appears markedly dilated with
significant parenchymal atrophy. Within it, large hypointense filling defects attributed to papillary growths are seen

a b

c d

Fig. 7.16a-d. IPMT of the main duct (MR evaluation). a The axial HASTE T2-weighted image shows dilatation of the duct of
Wirsung in the body-tail, with cystic ectasia oflocal collateral ducts (arrows). The dilatation of the duct ofWirsung is segmental:
the duct in the head of the gland is normal. b MRCP confirms the segmental dilatation of the proximal segment of the duct of
Wirsung with cystic ectasia of the collateral ducts. c, d In the gadolinium-enhanced GRE Tl-weighted sequence, carried out in
the arterial (nonfat-suppressed: c) and venous (fat-suppressed: d) phases, the pancreatic duct is clearly hypointense compared
with the parenchyma, which shows normal enhancement
110 C. Procacci et al.

into the duodenum, because of the patulous papillary


orifice (CROSS 1980; ITAI et al. 1987). In some cases,
an adequate pancreatogram can be obtained using
a balloon occlusion technique within the papillary
orifice (ITAI et al. 1987; OBARA et al. 1993a,b). Finally,
incomplete opacification of the ductal tree (Fig. 7.18)
can be caused by the obstructing mucin deposits
(TENNER et al. 1996; PAVONE et al. 1997; SUGIYAMA et
al. 1998a; YAMAGUCHI et al. 1999). These limitations
notwithstanding, ERCP provides optimal demonstra-
tion of the main pancreatic duct and precise docu-
mentation of segmental or diffuse dilatation (Figs.
7.19-7.21), and reveals the presence of ectasia within
the collateral branches (Fig. 7.19b) (FUKUKURA
Fig. 7.17. IPMT of the main duct (MR evaluation). The coro- et al. 2000). Further specific findings of IPMT are
nal heavily T2-weighted image highlights cystic dilatation of intraluminal filling defects resulting from mucinous
the main duct (asterisk), which appears to be surrounded by
ectatic collateral ducts. On the main duct walls, there are small, deposits or neoplastic papillary proliferations. Mucin
multiple, hypointense nodules attributed to papillary growths. deposits appear as elongated and amorphous intra-
A stone is present in the gallbladder luminal filling defects (Figs. 7.18, 7.19), isolated from
the ductal walls because of the interposition of the
contrast medium which, due to the injection pres-
sure, can mobilize them. On the contrary, the papil-
7.2.4 lary growths arise from the ductal walls, appearing
Endoscopic Retrograde as multilobulated, more constant defects within the
Cholangiopancreatography contrast-enhanced lumen (Fig. 7.21).
In case of doubt, it is possible not only to carry out
In 1982, OHHASHI and co-workers reported the first aspiration of the pancreatic juice and the subsequent
description of IPMT based on findings at ERCP cytological tests, but also to biopsy the ductal walls
(OHHASHI et al. 1982). ERCP remains today the 'gold or the papillary proliferations directly (BLOCK et al.
standard' imaging modality in studying IPMT. For
many years, its use was considered indispensable
in diagnosing this pathology (RAIJMAN et al. 1994;
RATTNER and PINS 1994; TENNER et al. 1996; LIM et
al. 2001). Today, its role is diminished because of the
increasing recognition of this entity on noninvasive
cross-sectional imaging, in particular MRCP. The
information that can be gained is twofold: endo-
scopic and radiographic (PAVONE et al. 1997).
The definitive diagnosis is established endoscopi-
cally by first recognizing the dilated papillae protrud-
ing into the lumen (PAVONE et al. 1997; FUKUKURA et
al. 2000) and, second, the abundant mucin pouring
from the patulous papillary orifice into the duodenal
lumen (DABEZIES et al. 1990; TENNER et al. 1996;
PAVONE et al. 1997; FUKUKURA et al. 2000). The secre-
tion of large quantities of mucin is typical of malig-
nant tumors (OBARA et al. 1993a; NAKAGOHRI et al.
1999). In more advanced stages, it is even possible
to find the opening of fistulous tracts which reach
the duodenal lumen from the common bile duct or
Fig. 7.18. IPMT of the main duct (ERCP evaluation). Opacifica-
from the main pancreatic duct. Opacification of the tion of the distal portion of the markedly dilated duct of Wir-
pancreatic duct is not always easy to accomplish, sung, with a voluminous amorphous filling defect attributed
especially when there is reflux of contrast medium to mucin. Normal-sized common bile duct (arrows)
Intraductal Papillary Mucinous Tumors: Imaging III

a ~----~------------~.. b

Fig. 7.19a,b. IPMT of the main duct (ERCP evaluation). a Segmental IPMT within the distal duct of Wirsung associated with
cystic dilatation and a voluminous filling defect (arrow). The main duct appears regularly dilated upstream. b Segmental IPMT
of the proximal portion of the main duct, which appears markedly dilated, with cystic ectasia of the collateral ducts, within which
small filling defects, attributed to mucin deposits (arrows), are highlighted. The remaining ductal segments appear normal

1996; SHIMA et al. 2000). However, biopsy can lead to


a misdiagnosis of benignity (HURWITZ et al. 2001).

7.2.5
Endoscopic US

This technique is mostly used in the study of IPMT


of the collateral ducts when cross-sectional imaging
has not resolved the problem of differential diagnosis
with other cystic masses of the pancreas. When seg-
mental or diffuse ectasia of the main duct has been
identified by other imaging modalities, but the ques-
tion of pancreatitis versus neoplasm remains uncer-
Fig. 7.20. IPMT of the main duct (intraoperative pancreato- tain, EUS may be valuable. However, the number of
gram). Opacification by means of direct injection into the duct nonspecific diagnoses is declining due to the increas-
of Wirsung shows its cystic dilatation in the head and body ing awareness of the imaging features of this tumor.
with mild dilatation in the tail The high resolution of EUS, related to the contiguity
of the probe with the structure under examination,
can lead to the finding of the smallest useful elements
for diagnosis (FUKUSHIMA et al. 1997; NAKAGOHRI
et al. 1999). Dilation of the main duct in these cases
has usually already been demonstrated; but it is pos-
sible to find the small cystic ectasia of the collateral
branches and the presence of intensely echogenic
mucin deposits and/or small hyperechoic papil-
lary proliferations within the main duct (Fig. 7.22)
(PAVONE et al. 1997; YAMAGUCHI et al. 1999). Thick-
ening and irregularities of the walls and hyperechoic
parietal septations or nodules correspond to areas
Fig. 7.21. IPMT of the main duct (pancreatogram on surgical of malignant transformation. The presence of mucin,
specimen: total pancreatectomy). The same patient as in Fig. 7.15.
Opacification by means of direct injection into the duct of Wir- in the form of hyperechoic aggregates or filaments,
sung confirms its dilation along with the irregular contour of the within the lumen of the cystically dilated branches is
walls due to the presence of multiple papillary growths also well shown (Fig. 7.23) (SUGIYAMA et al. 1998b;
112 C. Procacci et al.

a b

Fig. 7.22a,b. IPMT of the main duct (EUS evaluation). a Significant dilatation of the duct of Wirsung (W), starting from the
papilla. The ectatic hypoechoic collateral branches (black arrows) are visible. b Marked dilatation of the pancreatic duct (W)
in the body-tail, associated with atrophy of the adjacent parenchyma. The content of the duct is hyperechoic due to intraductal
mucin. PV, portal vein; SV, splenic vein; SMV, superior mesenteric vein

a b

Fig. 7.23a-c. IPMT of the main duct (EUS evaluation). a The


cephalic portion of the pancreatic duct at the ampullary orifice
into the duodenum (black arrow) is distended and contains
mural irregularities with small nodules (white arrow). b
Same image as a, magnified. Both the papillary projections
(white arrow) and the main duct opening into the duodenal
lumen (black arrow) are better visualized. c Different patient.
Distended main pancreatic duct within which a hyperechoic
nodule is appreciable (white arrow). PV, portal vein; SMV,
c superior mesenteric vein
Intraductal Papillary Mucinous Tumors: Imaging 113

ARIYAMA et al. 1998). EUS has been used to evalu- 7.3


ate the longitudinal extent of the neoplastic portion IPMT of the Collateral Ducts
of the main duct, useful for defining the therapeutic
approach of these forms (CELLIER et al. 1998). Find- IPMTs of the collateral ducts are more easily identifi-
ings which closely correlate to the probability of able than those of the main duct since they always
malignancy include a duct of Wirsung more than 10 appear with a mass effect. The considerable increase in
mm in diameter in main duct lesions, or alternatively, reports of this tumor, recorded over the past few years,
the presence of cystic lesions larger than 40 mm, con- is due to the ever more frequent casual finding of the
taining septa or nodules greater than 10 mm in size lesion with noninvasive imaging techniques like US
(KuBo et al. 2001). and CT (YANAGISAWA et al. 1993; CELLIER et al. 1998;
The introduction of high frequency miniprobes LIM et al. 2001). The lesion, when limited to the col-
(15-20 MHz) has made it possible to extend EUS lateral ducts, is almost always asymptomatic; clinical
by actually performing intraductal US examination symptoms related to this tumor often occur following
(intraductal ultrasonography, IDUS). This technique the secondary involvement of the main duct and are
is particularly useful in studying some IPMTs of the identical to those reported for main duct IPMTs.
main duct since the introduction of a miniprobe into
the papilla of Vater during the ERCP examination
is made easier. With IDUS, the identification of all 7.3.1
the aforementioned alterations is enhanced. Conse- Transabdominal US
quently, papillary projections, mural nodules, and
extraductal proliferations with parenchymal inva- The lesion has multilobulated outlines and a tran-
sion, suggestive of a carcinomatous transformation, sonic texture at US investigation (LIM et al. 2001).
are better visualized (FURUKAWA et al. 1997; MUKAI Identification is facilitated by the fact that the tumor
et al. 1998; YAMAO et al. 2001; KOITO et al. 2001). The is most commonly found in the head or the uncinate
association of EUS and IDUS seems to increase the process (Figs. 7.24a, 7.25a, 7.26a, 7.27a) and less com-
accuracy in the diagnosis and staging of the invasive monly in the body and tail (DABEZIES et al. 1990; LIM
forms (KoITO et al. 2001). et al. 2001). With current sonographic technology, a
Performing an endoscopically directed fine- needle skilled operator should be able to identify lesions
aspiration biopsy (FNAB) can refine the differential with a diameter of 1-2 cm (LIM et al. 2001). The lesion
diagnostic possibilities either by collecting muci- can have a 'honeycomb' micro cystic or an unilocular
nous fluid, if this is not practicable by the retrograde or multilocular macro cystic architecture (Figs. 7.24a,
approach, or by carrying out multiple biopsies of all 7.25a, 7.26a, 7.27a) (KANEKO et al. 2001; LIM et al.
the focal lesions protruding into the ductal lumen 2001). Despite the multiplicity of the septa, the micro-
or infiltrating the wall towards the parenchyma cystic form never appears as a solid echoic mass, as
(BRUGGE 2000). happens with a 'sponge-like' SCT. The diagnosis of

a b

Fig.7.24a,b. Microcystic IPMT of the collateral ducts (US and CT evaluations). The US (a) and CT (b) axial scans highlight
confluent cystic structures occupying the pancreatic head
114 C. Procacci et al.

Fig. 7.25a-c. Microcystic rPMT of the collateral ducts (US and


CT evaluations). In the longitudinal US (a) and axial CT (b, c)
scans, a small, ovoid mass with cystic architecture is visualized
in the uncinate process

b c

IPMT is further insured by finding intraluminal cystic tumors of the pancreas should be considered
defects (Fig. 7.27a) that can appear with an intensely in the differential diagnosis. Therefore, the primary
echoic texture in the presence of mucin deposits or role of transabdominal US is the identification of the
with a hypoechoic texture, originating on the ductal lesions, rather than their classification.
wall in the case of papillary proliferations (KANEKO
et al. 2001; LIM et al. 2001).
With US, it is difficult to demonstrate the com- 7.3.2
municating duct between the tumor and the main Computed Tomography
pancreatic duct; for this reason, the differential
diagnosis between collateral branch IPMT and The recognition of IPMT by computed tomography
either a serous cystadenoma ('honeycomb' micro- (CT) has been responsible for reducing the ERCP
cystic aspect: Figs. 7.24a, 7.25a, 7.26a) or mucinous evaluation of this lesion. Spiral and now multi slice CT
cystic tumor (unilocular or multilocular macrocys- techniques have allowed the pancreas to be evaluated
tic aspect: Fig. 7.27a) may be impossible. US is, how- with thinner sections (improving resolution), with
ever, particularly sensitive in documenting possible optimal contrast enhancement (facilitating recogni-
associated dilation, both segmental and diffuse, of tion of smaller lesions), and in 3D (refining the ability
the main duct. of the procedure to determine whether or not a cyst
Finding a lesion with the above-mentioned charac- communicates with the main duct) (FUKUKURA et
teristics near the uncinate process in a male patient, al. 2000). For this reason, it is particularly useful to
especially without symptoms, must lead to the suspi- acquire scans at a slice thickness of 3 mm on single-
cion of IPMT (DABEZIES et al. 1990). However, if the slice systems or 1-1.25 mm with multislice systems.
lesion is found in a woman, is not located in the unci- In the non-IV contrast phase, IPMT of the col-
nate process, does not have a communicating duct, and lateral ducts is recognizable when, depending on
is not associated with dilation of the main duct, other its size, it is able to produce a localized mass effect
Intraductal Papillary Mucinous Tumors: Imaging 115

c
Fig. 7.26a-d. Microcystic IPMT of the collateral ducts (US and CT evaluations). In the US (a) and axial CT (b-d) images obtained
following IV contrast enhancement, a fluid-containing lesion is identified in the pancreatic head, made up of microcysts and
macrocysts

within the gland. The lesion will appear isodense and following) (Figs. 7.28 c, d, 7.29b). It is important
or slightly hypodense compared with the adjacent to assess the thickness of the wall and septa: the rec-
un enhanced parenchyma (Figs. 7.28a, b, 7.29a). Cal- ognition of increased thickness is a useful indicator
cifications are rare but could be located in the septa of malignancy. On the other hand, thin wall and septa
or within mucin deposits. They are more often found do not exclude malignancy. With CT studies, it is still
in larger tumors. Ductal calcifications may occur as difficult to distinguish mucin deposits from papil-
a result of chronic pancreatitis resulting from long- lary proliferations unless the latter are not located
standing obstruction of the duct of Wirsung by the in a nondependent position. Varying the patient's
downstream tumor. position can be useful for differentiating mobile
In the pancreatic IV contrast phase (PROCACCI et mucin from fixed neoplastic elements. With IV con-
al. 1996), the difference between a low attenuation trast-enhanced spiral or multislice techniques, it is
tumor and the normal parenchyma is highlighted possible to demonstrate the considerable enhance-
(Figs. 7.24b, 7.25b,c, 7.26b-d, 7.27b-d). Optimal ment of the papillary proliferations which, unlike
enhancement of the pancreatic parenchyma is the mucin deposits, will demonstrate an increase in
obtained in the pancreatic phase (see Section 3.2.2.1 Hounsfield value. When the communicating duct is
116 C. Procacci et al.

a b

c d

Fig. 7.27a-d. Macrocystic IPMT of the collateral ducts (US and CT evaluations). a In the axial transabdominal US, a large,
sharply marginated mass can be seen in the pancreatic head. The complex echo pattern results from the presence of papillary
proliferations and mucin deposits. b-d Axial IV contrast-enhanced CT images reveal a large, unilocular mass in the uncinate
process. Multiple nodules are seen along the wall, the central portion containing inhomogeneous fluid

not visible, the differential diagnosis between IPMT chyma is less obvious because the enhancement of
and other cystic masses of the pancreas is difficult the pancreatic parenchyma approaches the baseline.
(Fig. 7.27b-d). IPMT of the collateral ducts more However, the greater density of the septa, due to the
often, but not necessarily, has a unifocal character. slower enhancement, makes it easier to demonstrate
In fact, there are situations in which multiple lesions, the internal architecture of the tumor (Figs. 7.24b,
even of small dimensions, involve the whole pancre- 7.25b, c, 7.26b-d, 7.28e, f, 7.29c).
atic gland (Fig. 7.30). At other times, there can be a
dual location at both the uncinate process and the tail
(NAKAGOHRI et al. 1999). 7.3.3
Following IV contrast administration, the main Magnetic Resonance Imaging
duct, which can either be quite normal or have some
degree of segmental or diffuse dilation (Fig. 7.28c), is The study protocol is the same as described above for
recognizable (SUGIYAMA and ATOMI 1998). This last IPMT of the main duct. However, in this case, par-
finding certainly contributes to the diagnosis of IPMT. ticular attention should be paid to the obliteration of
In the delayed IV contrast phases, the difference in signal coming from the liquid content of the stomach
density between the lesion and the pancreatic paren- and duodenum by the use of an oral contrast agent.
Intraductal Papillary Mucinous Tumors: Imaging 117

a b

c d

e f

Fig. 7.28a-f. IPMT of the collateral ducts (CT evaluation). a, b In the noncontrast images, there is a mild increase in the size of
the pancreatic head. c, d On the pancreatic phase IV contrast-enhanced images, there is intense enhancement of the head (c),
revealing an enlarged duct of Wirsung. Downstream (d), a multilobulated cystic lesion can be recognized. e, f In the venous
phase, the multilocular architecture of the cystic mass is more evident due to the greater enhancement of the septa
118 C. Procacci et a1.

a b

Fig. 7.29a-c. IPMT of the collateral ducts (CT evaluation). a


On the non contrast image, there is a multilobulated lesion,
slightly hypodense compared with the surrounding pancreatic
tissue. b Following IV contrast enhancement, in the pancreatic
phase the lesion is contrasted against the intensely enhancing
parenchyma. c In the venous phase, the central and thin septa
c are seen

a b

Fig. 7.30a,b. Multicentric IPMT of the collateral ducts (CT evaluation). Multiple hypodense lesions with clear outlines can be
seen throughout the gland (asterisks in a); the largest is located in the uncinate process (asterisk in b)
Intraductal Papillary Mucinous Tumors: Imaging 119

a b

______ .~~ .. ~~ ___________ ~ d

Fig. 7.31a-d. IPMT of the collateral ducts (MR evaluation). a In the axial unenhanced GRE FS Tl-weighted image, a tubular
lesion, hypointense compared with the pancreatic parenchyma, can be seen located in the uncinate process. b HASTE T2-
weighted sequence: the lesion is homogeneously hyperintense; the multilobulated contour and the communication with the
main duct (arrow) are more easily recognizable. c, d Gd-DTPA-enhanced, Tl-weighted GRE images obtained in the pancreatic
and venous phases show the intense enhancement of the parenchyma delimiting the tumor. The duct of Wirsung is identifiable
in the head of the pancreas (arrow)

During the MRCP study, it is also necessary to use The signal intensity is proportional to the size of the
multiple acquisition planes to attempt to document cystic space. The septa are hypointense, as are the
the presence or absence of a communication between mural papillary proliferations; mucin is indistin-
the lesion and the main duct. Unfortunately, at the guishable from the pancreatic juice. Further, with this
time of this writing, there is no 3D MRCP acquisi- sequence, it is possible to evaluate the relationships
tion technique. between the lesion and the duct ofWirsung (Figs. 7.31 b,
In the GRE FS Tl-weighted sequence, the lesion, 7.32a-c), particularly in the axial section (ARAKAWA et
despite its size and structural characteristics, appears al. 2000). The demonstration of the status of the main
hypo intense compared with the normal pancreatic duct is facilitated (ARAKAWA et al. 2000). This particu-
parenchyma (Fig. 7.31a). It is impossible to define the lar sequence is felt to have the highest sensitivity for
microcystic or macro cystic architecture of the tumor. detecting these lesions (Fig. 7.33a-d). Therefore, T2-
The pancreatic signal is brightest when the images weighted HASTE sequences are indispensable in the
are obtained with an 'opposed-phase' protocol. MR evaluation of these lesions.
On the T2-weighted HASTE sequence, the lesion Long echo-train, thick slab MRCP acquisitions
always appears hyperintense with a macro cystic or are not to be omitted from the study of these tumors
micro cystic architecture (Figs. 7.31b, 7.32a-c, 7.33a-d). (SUGIYAMA et al. 1998a; IRIE et al. 2000). This tech-
120 C. Procacci et al.

......._ _ ~_ ;.J_ ..._ _ _.......... b

Fig.7.32a-d. rPMT of the collateral ducts (MR evaluation). a-c HASTE T2-weighted axial scans show a tubular hyperintense
lesion with central septa communicating with (arrow) the main duct (arrowhead in a); the latter appears moderately dilated
upstream. The tumor is located in the posterior portion of the head. d MRCP confirms the presence of a multilobulated cystic
lesion in the pancreatic head, partially superimposed over the enlarged duct of Wirsung

nique gives a complete map of the pancreatic duct to predict the biological behavior of the lesion (IRIE
and the lesions, whether single or multiple (Figs. et al. 2000). In fact, as with CT, the presence of thick
7.32d, 7.33e,f), that surround it (ARAKAWA et al. walls and septa (Fig. 7.31c,d) testifies to the malig-
2000). Carrying out multiple acquisitions in dif- nant nature of the tumor, while thin septa do not
ferent spatial planes contributes to the recognition exclude malignant transformation of the lesion.
of the communicating duct (ONAYA et al. 1998). In
the larger masses, the papillary proliferations are
sometimes obscured due to volume averaging effects 7.3.4
related to the thick slab acquisition. Endoscopic Retrograde
MR investigations, especially when performed for Cholangiopancreatography
monitoring a previously found lesion, are made up
exclusively of the previously mentioned sequences. ERCP still represents the 'gold standard' of IPMT
The use of IV contrast medium (gadolinium-DTPA) diagnosis even today, although MRCP has recently
is useful once a diagnosis of IPMT has been obtained replaced a good deal of its diagnostic role. Having
Intraductal Papillary Mucinous Tumors: Imaging 121

c _~_--'-=-' " d

e f

Fig. 7.33a-f. Multicentric IPMT of the collateral ducts (MR evaluation). a-c On axial HASTE T2-weighted scans, small cystic
formation in the pancreatic tail (black arrow in a) and a dominant mass with micro cystic architecture in the pancreatic head
can be seen; the duct of Wirsung in the pancreatic head is moderately dilated. d In the coronal plane, the caudal extension and
the micro cystic aspect of the larger lesion are confirmed as well as the modest dilatation of both the common bile duct and
the duct of Wirsung. e, f MRCP examination, carried out in the coronal (e) and axial (f) planes, confirms the aforementioned
cystic lesions and the mild dilation of both the duct of Wirsung and the common bile duct
122 C. Procacci et al.

said that, in doubtful cases (SUGIYAMA and ATOMI luminal defects easier, which significantly modify
1998) and, above all, just before surgical resection, the therapeutic strategy of these lesions (LIM et al.
this technique is still utilized. The opacification of 2001). Finally, ERCP is able to document a multicen-
the lesion, especially when the pancreatic duct is tric IPMT of the collateral ducts even if its sensitivity
normal, is almost always possible (Figs. 7.34, 7.35a) seems to be inferior to that of MRCP in the dem-
unless a mucin deposit obstructs the orifice of the onstration of single lesions (SUGIYAMA and ATOMI
communicating duct and the lesion does not fill (LIM 1998; NAKAGOHRI et al. 1999; IRIE et al. 2000).
et al. 2001; SUGIYAMA and ATOMI 1998; OBARA et al.
1993a). ERCP shows, like the other imaging modali-
ties, the micro cystic or macro cystic architecture of 7.3.5
the mass, along with the aforementioned mucin Endoscopic US
deposits or intraluminal growths (OBARA et al.
1993a,b; SUGIYAMA and ATOMI 1998; LIM et al. 2001). EUS is only occasionally used in the study of IPMT
The distension of the duct of Wirsung, even when of the collateral ducts since other imaging modalities,
the size is normal, can make recognition of intra- especially MRCP, are able to characterize the lesion
more often than not. The advantage of this technique
is its accurate definition of the architecture of the
lesion (Fig. 7.36), where even the thinnest septa,
mucin deposits, and small papillary proliferations
can be seen (OBARA et al.1993a; SUGIYAMA et al.1997,
1998b; SUGIYAMA and ATOMI 1998). Furthermore, it
is sometimes possible to recognize the communicat-
ing duct. The cystic IPMTs can have a wide range of
morphological variations, particularly if located in
the uncinate process. They cause problems of differ-
ential diagnosis at EUS when they are confused with
other cystic tumors, especially when they present as a
unilocular cyst. Cysts with this appearance are found
when the IPMT is in an advanced stage and there is
a higher probability of malignancy.
When the results of the imaging studies are
Fig. 7.34. IPMT of the collateral ducts (ERCP evaluation). The
duct of Wirsung is dilated in the head of the pancreas; on its inconclusive, lesions located in the head or uncinate
right side an ovoid, cystic lesion (asterisk) opacities, oriented process can be further analyzed by transduodenal
parallel to the duct of Wirsung FNA with analysis of the aspirated liquid, cytologic

a b

Fig. 7.35a,b. IPMT of the collateral ducts (ERCP and CT evaluations). a Cystic ectasia of two collateral branches in the pancreatic
head is shown (arrows). b CT axial scan shows the two cystic lesions (arrows) located near the enlarged duct ofWirsung
Intraductal Papillary Mucinous Tumors: Imaging 123

a b

Fig.7.36a,b. IPMT of the collateral ducts (EUS evaluation). Cystic ectasia of a collateral branch in the uncinate process with
hyperechoic mucin globs on the inside (arrows). A magnified view (b) shows the mucin aggregate better (arrows). Ao, aorta;
SMV, superior mesenteric vein

analysis, and assay for tumor markers (BRUGGE 2000; power Doppler, it is possible to demonstrate the flow
GRESS et al. 2000; BOUNDS and BRUGGE 2001;AITHAL within the papillary proliferations (Fig. 7.39b,c). In
et al. 2001). the more advanced stages of the disease, it is pos-
sible to find peripancreatic adenopathy or malignant
ascites.
CT examination documents the multiplicity of
7.4 collateral ductal ectasia associated with the dilation
IPMT of the Mixed Type of the main duct (Fig. 7.37c,d). The gland can increase
in volume, particularly in the head. There is compres-
The mixed form deserves a separate classification. sion of the duodenal lumen (Figs. 7.38c-f, 7.40, 7.41)
When tumors involve both the collateral as well as and/or papillae protrusion into it (Figs. 7.38f, 7.41b).
the main duct, the clinical picture is severer, and Mucin deposits can always be recognized in these
the patients will present with a symptom complex advanced tumors. The neoplastic rind around it
which is not seen in the previously mentioned forms. (Fig. 7.41a) or the large papillary proliferations (Fig.
Because both duct systems are involved, the site of 7.42a,b) are commonly demonstrated as signs of
origin (e.g., main duct into collateral duct or collat- malignant degeneration of the tumor. It can happen,
eral duct into main duct) cannot be discerned. however, that the multiple lesions, despite their large
At transabdominal US, it is possible to recognize size, have thin walls. They can protrude towards the
the dilation of the duct of Wirsung associated with peritoneal cavity or the retroperitoneal space with
the cystic ectasia of the collateral ducts (Fig. 7.3 7a,b). the possible appearance of ascites, due to peritoneal
In some cases, it is possible to distinguish the main carcinomatosis. In this case, mucin deposits are
duct from the collateral ducts. In others, there might sometimes visible as they are well delimited com-
be one large mass that occupies the pancreatic head pared with the ascitic fluid (Fig. 7.41), surrounded by
inside of which cystic spaces can be seen, whose a thin, dense rim. It is possible to recognize adenopa-
origin is difficult to establish (Fig. 7.38a,b). Dilation thy while hepatic metastases are uncommon.
of the common bile duct is often found secondary to MR, especially with the use of intravenous contrast
compression or direct involvement by the mass. It is medium (gadolinium), gives the same information as
more difficult to establish the lesion's relationship CT with the added benefit of MRCP renderings of the
with the duodenum. Within the dilated ducts, it is ductal tree and its contents (Figs. 7.42c,d, 7.43, 7.44).
common to find deposits of hyperechoic mucin or The utilization of ERCP or EUS in studying this
hypoechoic papillary proliferations (Fig. 7.39a). With advanced form of IPMT is not often necessary
124 C. Procacci et a!.

a b

c d

Fig. 7.37a-d. IPMT of the mixed type (US and CT evaluations). In the oblique subcostal (a) and axial (b) scans, a small, fluid-
containing lesion with micro cystic architecture is visible in the pancreatic neck (arrow) with dilation of the duct of Wirsung
upstream (caliper). c, d In the axial, IV contrast-enhanced CT images, the corresponding cystic lesion with microcystic archi-
tecture can be found in the pancreatic neck with dilation of the duct of Wirsung upstream

because useful information, not only for diagnosis lateral duct form is being examined. Mixed type
but also for the therapeutic decision-making, has lesions, on the other hand, are easily characterized
already been gained through noninvasive techniques. by imaging modality.
Obviously, whenever there is the tiniest suspicion of With main duct IPMTs, the greatest difficulty in
the nature of the lesion being different (chronic classification arises in the presence of the segmen-
obstructive pancreatitis), it is advisable to perform tal form. When the involvement is minimal, US or
an ERCP. This technique gives an optimal endoscopic CT will reveal only a simple ectasia of the affected
evaluation of the papillae and the possible fistulous portion of the main duct (Fig. 7.10). Supporting
tracts with mucin leakage. Complete opacification of findings (ectasia of the collateral branches, papil-
the ducts is difficult because of the patulous orifice lary proliferations, mucin deposits) will not easily
of the papilla and the dilution and obstruction of the be recognized. MRCP is more helpful and shows the
contrast by the thick mucin. continuity between the normal duct and the dilated
segment. In this case, the most important aim of
imaging is to show that there is no postinflammatory
or neoplastic stenosis responsible for the segmental
7.5 dilation of the main duct. MRCP is able to demon-
Differential Diagnosis strate the absence of a stenotic segment of the duct
of Wirsung, but ERCP is, undoubtedly, the modality
The problems of differential diagnosis of IPMT which gives the most specific results and the only one
depend on whether the main duct form or the col- able to demonstrate, with certainty, the patency of the
Intraductal Papillary Mucinous Tumors: Imaging 125

a b

c d

e f

Fig. 7.38a-f. IPMT of the mixed type (US and CT evaluations). a, b In the axial US scans, there is marked ectasia of the main
duct (caliper) and a large lesion of mixed content (solid and fluid) in the head (calipers). c-f In the IV contrast-enhanced CT
scans, marked ectasia of the duct of Wirsung and a predominantly cystic mass in the head of the pancreas are visible. Some of
the cystic spaces show papillary growths within (arrows in d, e). f Conspicuous dilation of the major papilla is present (asterisk)
which bulges into the duodenal lumen
126 C. Procacci et al.

a b

Fig. 7.39a-c. IPMT of the mixed type (US evaluation). High-


lighted in the US axial scan (a), cystic ectasia of the main
duct within which numerous papillary growths can be seen,
especially along the posterior wall (arrows). With power Dop-
pler (b, c), arterial flow within the stalk of the largest papillary
c growth is seen

a b

Fig.7.40a,b. IPMT of the mixed type (CT evaluation). IV contrast-enhanced CT images in the craniocaudal direction reveal
diffuse enlargement of the entire pancreatic gland, especially in the head, which deforms the inferior vena cava. The significant
dilation of the main duct and the cystic ectasia of the collateral ducts are clearly recognizable
Intraductal Papillary Mucinous Tumors: Imaging 127

a b

Fig. 7.41a,b. IPMT of the mixed type (CT evaluation). In the CT carried out following IV contrast administration, marked ectasia
of the main duct with multiple solid parietal growths is shown. There is also dilation of the major papilla which protrudes into
the duodenal lumen and cystic ectasia of the collateral ducts of the uncinate process. Moreover, ascites is recognizable, within
which there are omental masses (asterisks) with soft-tissue attenuation

a b

c d

Fig. 7.42a-d. IPMT of the mixed type (CT and MR evaluations). a, b CT scans carried out in the venous phase demonstrate ectasia
of the duct of Wirsung, with cystic appearance in the cephalic portion. Multiple hypodense parietal nodules are recognizable.
c, d In the MR scans, acquired with HASTE T2-weighted sequence, the significant ectasia of the main duct and the collateral
ducts are confirmed. Furthermore, the multiple hypointense parietal nodules are also demonstrated
128 C. Procacci et al.

a b

Fig. 7.43a-c. IPMT of the mixed type (MR evaluation). Same


patient as in Fig. 4.39. Conspicuous dilation of the duct of
Wirsung with parenchymal atrophy is present. In the body,
two papillary projections originating from the posterior wall
are highlighted (arrows), which are slightly hyperintense in
non-IV contrast images (GRE FS II WI in a), hypointense in
c T2 (b), and hyperintense after contrast medium (c)

a b

_ _ _..... d
c

Fig. 7.44a-d. IPMT of the mixed type (MR evaluation). The axial GRE FS II-weighted venous phase-enhanced images docu-
ment dilation of the main duct and the collateral ducts, within which multiple hypointense parietal nodules are recognizable.
Dilation of the papilla (arrow in d) which protrudes into the duodenal lumen is also appreciable
Intraductal Papillary Mucinous Tumors: Imaging 129

main duct over its entire extent. Further, ERCP may known. More recently, the opposite error has been
occasionally demonstrate intraluminal filling defects made, that is chronic obstructive or cystic fibrosis
(mucin or papillary proliferations). Therefore, US, CT correlated pancreatitis has mistakenly been labeled as
or MR demonstration of the segmental dilation of the IPMT and submitted to surgical resection (Fig. 7.48).
duct ofWirsung (Fig. 7.10), especially when noted in Because of all these considerations, whenever a diag-
a patient with no previous pancreatic clinical history, nosis of IPMT of the main duct or chronic obstructive
must lead to the suspicion of the possible presence of pancreatitis leads to the decision to proceed to surgi-
segmental main duct IPMT. ERCP is recommended in cal resection or derivative surgery, it is imperative to
these cases for the optimal depiction of ductal anat- establish the diagnosis by means of an endoscopic
omy (PAVONE et al. 1997; YAMAGUCHI et al. 1999). biopsy, analysis of pancreatic juice (for mucin or
The segmental involvement of the main duct can amylase), or during surgery with immediate frozen
more rarely appear as a cystic dilation (Figs. 7.3a, section reporting.
b, 7.11b). In these cases, the lesion will be misdiag- Collateral branches IPMTs, easily recognizable as
nosed as an extraductal cystic mass, particularly a a cystic mass of the pancreas, may be mistaken for
mucinous cystic tumor (FUKUKURA et al. 2000). In cystic tumors which do not arise from the walls of
the past, some reports of mucinous cystic tumor, the pancreatic ducts The diagnosis, despite the size
in male patients, might really have represented seg- of the tumor, is definitively established when the
mental IPMTs with cystic ectasia of the main duct communication with the main duct is documented
(PAVONE et al. 1997). The communication of the (FUKUKURA et al. 2000). This goal is difficult to
cystic ally dilated segment with the normal duct can reach with US but easier with CT (FUKUKURA et
be suspected with CT (Fig. 7.11b) and, above all, with al. 2000). Demonstrating such continuity is easier
MRCP, but without doubt, it can only be confirmed at when the main duct is dilated near the lesion. If the
ERCP (PAVONE et al. 1997). communication is not visible, IPMT cannot be dif-
In the diffuse form, the diagnosis of IPMT of the ferentiated with certainty from serous cystadenoma
main duct is easier. In this case, other signs, such as (when of micro cystic architecture), mucinous cystic
cystic ectasia of the collateral branches and the dila- tumor, retention cysts, or lymphangioma (when of
tion of the papillae protruding into the duodenal macro cystic unilocular or multilocular architecture)
lumen, are present in association with the previously (PAVONE et al. 1997; GRIFFITH et al. 1998). In this
mentioned findings of mucin deposits and papillary case, MRCP can significantly aid in establishing the
proliferations (PAVONE et al. 1997). These signs are correct diagnosis providing that acquisitions are
not always visible, however (Fig. 7.45). Furthermore, taken along multiple spatial planes (FUKUKURA et
there may also be findings typically associated with al. 2000). While the failure to recognize the com-
chronic obstructive pancreatitis, in particular calci- munication does not exclude this tumor, ERCP may
fications. For this reason, it is sometimes difficult to be necessary to find it. If even this investigation fails
differentiate the diffuse form of main duct IPMT and to provide a conclusive diagnosis, the use of biopsy
chronic pancreatitis with US, CT, and conventional of the lesion utilizing EUS guidance may be defini-
MR (CASADO et al. 2000). The diagnostic difficulties tive. It has been suggested to limit needle aspirations
are also related to the clinical picture of main duct by means of an endoscope so as to lower the risk of
IPMTs which may be identical to those of relapsing mucin and neoplastic dissemination along the tract
acute or chronic pancreatitis (TRAVERSO et al. 1998; (DABEZIES et al. 1990; PAVONE et al. 1997).
NAVARRO et al. 1999; TIBAYAN et al. 2000).
In our personal experience, there have been cases
of main duct IPMT mistakenly interpreted as chronic
obstructive pancreatitis and submitted to surgi- 7.6
cal intervention (pancreatojejunostomy: Fig. 7.46), Biological Behavior and Recurrences
which were correctly diagnosed several years later
because of the significant progression of the disease. The difficulty in the therapeutic management of
There have also been cases where the radiological intraductal tumors derives from the wide range of
signs useful for the diagnosis were present in radio- epithelial changes manifested by this neoplasm, rang-
logical examinations, especially CT, carried out up to ing from simple hyperplasia to aggressive carcinoma.
10 years previously (Fig. 7.47). The misdiagnoses of It is not uncommon to find all grades of epithelial
chronic pancreatitis were done mainly in the 1990s change in the same lesion. Therefore, imaging is
when IPMT of the main duct was not very well required not only to identify and characterize the
130 C. Procacci et al.

c .....'--_ _ _
d

e f

Fig. 7.4Sa-f. IPMT of the mixed type (CT evaluation). The craniocaudal scans, taken in the craniocaudal direction, show a cystic
lesion (asterisk in a) of the pancreatic tail and uniform dilation of the duct of Wirsung up to the papilla

lesion, but also to make an assessment of its biologi- 1999; NAKAGOHRI et al. 1999; TERRIS et al. 2000; IRIE
cal behavior. Pathological correlation has shown that et al. 2000; WAKABAYASHI et al. 2001). Any sized pap-
there are considerable differences between the main illary proliferations (Figs. 7.42, 7.43) and/or a thick
duct and collateral duct forms (TERRIS et al. 2000). rind with irregular contour delimiting the dilated
IPMT of the main duct, whether segmental of duct (Figs. 7.41, 7.44) are always signs of malignant
diffuse, must be considered a malignant tumor, and degeneration of the tumor (SUGIYAMA et al. 1997;
surgical resection is recommended (SUGIYAMA and SUGIYAMA and ATOMI 1998; YAMAGUCHI et al. 1999;
ATOMI 1998; YAMAGUCHI et al. 1999; KOBARI et al. IRIE et al. 2000; TAOULI et al. 2000; LIM et al. 2001;
Intraductal Papillary Mucinous Tumors: Imaging 131

a b

c d

Fig.7.46a-d. IPMT of the mixed type (CT and ERCP evaluations). a-c CT scans, carried out in the IV contrast-enhanced phase,
reveal cystic dilation (asterisk) of the duct of Wirsung in the head and neck of the pancreas. d ERCP confirms the cystic dila-
tion of the duct within the head (asterisk). This patient had undergone previous pancreatojejunostomy (arrow), and contrast
material opacifies the jejunal loop

WAKABAYASHI et al. 2001). Moreover, marked dila- ing the biological behavior of the tumor, considering
tion of the main duct and protrusion of the dilated lesions greater than 3 cm as suspicious for malignant
papillae into the duodenum (Figs. 7.4, 7.5, 7.8, 7.12) degeneration (SUGIYAMA and ATOMI 1998; KIMURA
are almost always indicative of malignant behavior and MAKUUCHI 1999; IRIE et al. 2000; WAKABAYASHI
(LIM et al. 2001). In the absence of these findings, it et al. 2001). In reality, it is possible to find very large
is still impossible to exclude the presence of foci of cystic masses with thin walls where only the presence
malignant degeneration (carcinoma in situ) (SUGI- of hyperplasia is later documented. Finding thick
YAMA and ATOMI 1998; IRIE et al. 2000). walls and/or septa and, above all, the presence of
The definition of the biological behavior of IPMT papillary growths (Fig. 7.27) is certainly much more
of the collateral ducts is of notable clinical importance reliable for assessing malignancy.
since the occurrence of benign tumors is rather high, Unfortunately, even with small lesions, the absence
and it is theoretically possible to avoid surgical inter- of the above-mentioned signs cannot justify the defi-
vention and follow these lesions with serial imaging nite diagnosis of a benign tumor since microscopic
studies. It should be remembered that IPMT of the foci of malignant epithelium may be present in any
collateral ducts is often multicentric (NAKAGOHRI given lesion (AGOSTINI et al. 1989). However, it has
et al. 1999), involving the entire gland so that only been shown that these lesions grow very slowly, with
a total pancreatectomy would guarantee its surgical no significant variation in the majority of cases, even
resection. after a number of years (KAWARADA et al. 1992;
Some authors think that the size of the lesion in OBARA et al. 1993a; BARBE et al. 1998; MEGIBOW et
the collateral ducts is a useful parameter for defin- al. 2001; WAKABAYASHI et al. 2001). Therefore, in the
132 C. Procacci et al.

3 b

c d

e f

Fig.7.47a-f. rPMT of the mixed type (CT evaluation). 3-d Contrast-enhanced CT scans in the venous phase demonstrate mild
dilation of the duct of Wirsung in the pancreatic body-tail, and significant, cystic-like dilation of the duct in the neck and head
of the pancreas. Additionally, protrusion of the dilated papilla into the duodenum (arrow in d) can be seen. Papillary growths
and mucin deposits are present within the dilated ducts. e, f The same investigation, carried out 10 years previously in the
same patient, documented the initial ectasia only in the cephalic portion of main duct and collateral branches within the head
(arrows in f)
Intraductal Papillary Mucinous Tumors: Imaging l33

a b

c d

e f

Fig.7.48a-f. Chronic pancreatitis secondary to cystic fibrosis, erroneously diagnosed as main duct IPMT (CT and MR evalua-
tions). a, b CT scans carried out on non-IV contrast images (a) and in IV contrast-enhanced images in the pancreatic phase (b)
demonstrate dilation of the entire main duct. Multiple, nonenhancing, solid deposits (arrows in a, b) located in the dependent
portion are present. c-d Axial HASTE T2-weighted MR images reveal dilatation of the duct of Wirsung with parenchymal
atrophy. Numerous, markedly hypointense deposits are also present. e MRCP confirms the diffuse dilation of the main duct
and the multiple defects. f On the specimen (total pancreatectomy), the dilated main duct is occupied by multiple amorphous
calcifications
134 C. Procacci et al.

a _ _ ........ b

c d

e f

Fig.7.49a-f. rPMT of the mixed type (CT evaluation). a-c CT scans performed during the contrast-enhanced venous phase
show cystic-like segmental dilation of the duct of Wirsung in the pancreatic body (asterisk in a) with modest upstream dilata-
tion; the pancreatic head is normal. d-f The follow-up examination carried out after 5 years, following intermediate resection,
demonstrates significant atrophy of the pancreatic tail and neoplastic transformation of the duct of Wirsung with the dilated
papilla protruding into the duodenum (arrow)
Intraductal Papillary Mucinous Tumors: Imaging 135

presence of a cystic lesion presumably originating in Arakawa A, Yamashita Y, Namimoto T, Tang Y, Tsuruta J,
the collateral ducts with benign characteristics and Kanemitsu K (2000) Intraductal papillary tumors of the
pancreas. Histopathologic correlation of MR colangiopan-
in an elderly patient, it is possible to avoid surgical
creatography findings. Acta Radiol 41:343-347
intervention and monitor the lesion with US or CT Azar C, Van de Stadt J, Rickaert F, Deviere J, Baize M, Kloppel G,
(KIMURA and MAKUUCHI 1999; YAMAO et al. 2000). Gelin M, Cremer M (1996) Intraductal papillary mucinous
But when the lesion appears in a young patient and tumours of the pancreas. Clinical and therapeutic issues in
there is diagnostic doubt as to its nature, it is better 32 patients. Gut 39:457-464
Bastid C, Bernard JP, Sarles H, Payan MJ, Sahel J (1991) Muci-
to carry out an immediate ERCP. Once the IPMT has
nous ductal ectasia of the pancreas: a premalignant disease
been confirmed, thanks to the demonstration of its and a cause of obstructive pancreatitis. Pancreas 6: 15-22
communication with the main duct, according to Barbe L, Levy P, Mal F, Gayet B (1998) Benign intraductal
some authors it is better to proceed immediately to papillary mucinous tumors of the pancreas with a 30-year
surgical resection (SUG IYAMA and ATOMI 1998; SIECH follow-up. Gastroenterol Clin Bioi 22:91-93
Block KP, Mahvi D, Voytovich M, Watkins JL, Mosley R,
et al. 1999; VAN DE STADT et al. 1999; YAMAGUCHI et
Reichelderfer M (1996) Mucinous ductal ectasia in an
al. 1999; TIBAYAN et al. 2000; FALCONI et al. 2001). octogenarian: successful treatment with the Whipple pro-
Others favor a varied approach and accept follow-up cedure. Am J Gastroenterol 91:388-390
in selected cases (SHO et al. 1998; TAKEYOSHI et al. Bounds BC, Brugge WR (2001) EUS guided fine needle aspi-
1999; NAKAGOHRI et al. 1999; SCIAUDONE et al. 2000; ration cytology and tumor marker analysis of pancreatic
ductal fluid differentiates between IPMT and Chronic
YAMAO et al. 2000; CASADO et al. 2000; WAKABAYASHI
Pancreatitis. Gastrointest Endosc 53:AB163
et al. 2001). Watchful monitoring is particularly Brugge WR (2000) The role of EUS in the diagnosis of cystic
advisable in the case of cystic lesions smaller than lesions of the pancreas. Gastrointest Endosc 52 [Suppl]:
2.5 cm in diameter with a normal main duct, since it S18-S22
has been documented that these lesions are normally Casado OA, Gallardo DH, Gonzalez EM, Diaz MM, Calvo AG,
Saborido BP, Medina EM, Martin AG (2000) Intraductal
benign and grow very slowly. Resection is, however,
papillary mucinous tumors: an entity which is infrequent
preferable when the main duct is dilated, partial and difficult to diagnose. Hepatogastroenterology 47:
resection being an option when there is segmental 275-284
ductal involvement. Unfortunately, in the presence of Cellier C, Cullerier E, Palazzo L, Rickaert F, Flejou JF, Napoleon
diffuse dilation of the main duct, whether primary B, Van Gansbeke D, Bely N, Ponsot P, Partensky C, Cugnenc
PH, Barbier JP, Deviere J, Cremer M (1998) Intraductal
or associated with a collateral duct tumor, it is not
papillary and mucinous tumors of the pancreas: accuracy
possible to define, by imaging, the true intraductal of preoperative computed tomography, endoscopic retro-
extension of the neoplasm. For this reason, a total grade pancreatography and endoscopic ultrasonography,
pancreatectomy should be performed. As an alterna- and long-term outcome in a large surgical series. Gastro-
tive, it is possible to initially resect the more highly intest Endosc 47:42-49
Cross MR (1980) Mucinous cystadenoma of the pancreas:
involved part of the gland, extending the resection
Endoscopy as an aid to diagnosis. Gastroenterol 79:
distally or proximally until the margins at frozen sec- 944-947
tion are free of tumor (SUGIYAMA and ATOMI 1998; Cuillerier E, Cellier C, Palazzo L, Deviere J, Wind P, Rickaert
CUILLERIER et al. 2000). Unfortunately, this method F (2000) Outcome after surgical resection of intraductal
does not exclude 'skip lesions' from which tumor may papillary and mucinous tumors of the pancreas. Am J
GastroenteroI95:441-445
recur several years following resection (Fig. 7.49)
Dabezies MA, Campana T, Friedman AC (1990) ERCP in the
(CUILLERIER et al. 2000). diagnosis of ductectatic mucinous cystadenocarcinoma of
the pancreas. Gastrointest Endosc 36:410-411
Falconi M, Salvia R, Bassi C, Pederzoli P (2001) Clinicopatho-
logical features and treatment of intraductal papillary
References mucinous tumours of the pancreas. Br J Surg 88:376-381
Fukukura Y, Fujiyoshi F, Sasaki M, Inoue H, Yonezawa S,
Agostini S, Choux R, Payan MJ, Sastre B, Sahel J, Clement JP Nakajo M (2000) Intraductal papillary mucinous tumors
(1989) Mucinous pancreatic duct ectasia in the body of the of the pancreas: thin-section helical CT findings. AJR Am J
pancreas. Radiology 170:815-816 RoentgenoI174:441-447
Aithal GP, Cunningham JT, Kim EY, Wallace MB, Hoffman BJ Fukushima N, Mukai K, Kanai Y, Hasebe T, Shimada K, Ozaki
(2001) Accuracy of endoscopic ultrasound in suspecting H, Kinoshita T, Kosuge T (1997) Intraductal papillary
the diagnosis of intraductal papillary mucinous tumor of tumors and mucinous cystic tumors of the pancreas: clini-
the pancreas. Gastrointest Endosc 53:AB166 copathologic study of 38 cases. Hum PathoI28:1010-1017
Ariyama J, Sugiyama M, Satoh K, Wakabayashi K (1998) Furukawa T, Takahashi T, Kobari M, Matsuno S (1992) The
Endoscopic ultrasound and intraductal ultrasound in the mucus-hypersecreting tumor of the pancreas. Develop-
diagnosis of small pancreatic tumors. Abdom Imaging 23: ment and extension visualized by three-dimensional com-
380-386 puterized mapping. Cancer 70:1505-1513
136 C. Procacci et al.

Furukawa T, Ohhashi K, Yamao K, Naitoh Y, Taki T, Itoh A, between benign and malignant tumors by endoscopic
Hayakawa S, Watanabe Y, Goto H,Hayakawa T (1997) Intra- ultrasonography. Am J Gastroenterol 96:1323-1325
ductal ultrasonography of the pancreas: development and Lim JH, Lee G, Oh YL (2001) Radiologic spectrum of intra-
clinical potential. Endoscopy 29:561-569 ductal papillary mucinous tumor of the pancreas. Radio-
Gress F, Gottlieb K, Cummings 0, Sherman S, Lehman G (2000) Graphics 21:323-340
Endoscopic ultrasound characteristics of mucinous cystic Loftus EV, Olivares-Pakzad B, Batts KP, Adkins MC, Stephens DH,
neoplasm of the pancreas. Am J Gastroenterol 95:961-965 Sarr MG,Dimagno EP (1996) Intraductal papillary-mucinous
Griffith J, Ahuja A, Yeung D, Metreweli C (1998) Case report: tumors of the pancreas: clinicopathologic features, outcome
mucinous ductal ectasia of the pancreas. Clin Radiol 53: and nomenclature. Gastroenterology 110:1909-1918
536-538 Madura JA, Wiebke EA, Howard TJ, Cummings OW, Hull
Hurwitz LK, Daniels A, Barkin JS, Petruff C, Gaertner E, Moses MT, Sherman S, Lehman GA (1997) Mucin-hypersecret-
F (2001) Preoperative diagnosis of intraductal papillary ing intraductal neoplasms of the pancreas: a precursor to
mucinous tumors of the pancreas by endoscopic pancre- cystic pancreatic malignancies. Surgery 122:786-793
atic biopsy. Gastrointest Endosc 53:510-513 Megibow AJ, Lombardo FP, Guarise A, Carbognin G, Scholes
Irie H, Honda H,Aibe H, Kuroiwa T, Yoshimitsu K, Shinozaki K, J, Rofsky NM, Macari M, Balthazar EJ, Procacci C (2001)
Yamaguchi K, Shimada M, Masuda K (2000) MR cholangio- Cystic pancreatic masses: cross-sectional imaging observa-
pancreatographic differentiation of benign and malignant tions and serial follow-up. Abdom Imaging 26:640-647
intraductal mucin-producing tumors of the pancreas. AJR Morohoshi T, Kanda M, Asanuma K, Kloppel G (1989) Intra-
Am J RoentgenoI174:1403-1408 ductal papillary neoplasms of the pancreas. A clinicopatho-
Irie H, Honda H, Kuroiwa T, Yoshimitsu K, Aibe H, Shinozaki logic study of six patients. Cancer 64: 1329-1335
K, Masuda K (2002) Measurement of the apparent diffusion Mukai H, Yasuda K, Nakajima M (1998) Differential diagnosis
coefficient in intraductal mucin-producing tumors of the of mucin-producing tumors of the pancreas by intraductal
pancreas by diffusion-weighted echo-planar MR imaging. ultrasonography and peroral pancreatoscopy. Endoscopy
Abdom Imaging 27:82-87 30 [Supplll:A99-A102
Itai Y, Ohhashi K, Nagai H, Murakami Y, Kokubo T, Makita Nakagohri T, Kenmochi T, Kainuma 0, Tokoro Y, Asano T
K, Ohtomo K (1986) 'Ductectatic' mucinous cystadenoma (1999) Intraductal papillary mucinous tumors of the pan-
and cystadenocarcinoma of the pancreas. Radiology 161: creas. Am J Surg 178:344-347
697-700 Navarro F, Michel J, Bauret P, Ramos J, Blanc P, Fabre JM, Millat
Itai Y, Kokubo T, Atomi Y, Kuroda A, Haraguchi Y, Terano A B (1999) Management of intraductal papillary mucinous
(1987) Mucin-hypersecreting carcinoma of the pancreas. tumours of the pancreas. Eur J Surg 165:43-48
Radiology 165:51-55 Obara T, Maguchi H, Saitoh Y, Itoh A, Arisato S, Ashida T,
Itoh S, Ishiguchi T, Ishigaki T, Sakuma S, Maruyama K, Senda Nishino N, Ura H, Namiki M (1993a) Mucin-producing
K (1992) Mucin-producing pancreatic tumor: CT findings tumor of the pancreas: natural history and serial pancrea-
and histopathologic correlation. Radiology 183:81-86 togram changes. Am J Gastroenterol 88:564-569
Kaneko T, Nakao A, Inoue S, Sugimoto H, Hatsuno T, Ito A, Obara T, Maguchi H, Saitoh Y, Sohma M, Tsuji K, Koike Y,
Hirooka Y, Nagasaka T, Nakashima N (2001) Intraoperative Takemura K, Ura H, Namiki M (1993b) Intraductal papil-
ultrasonography by high-resolution annular array trans- lary neoplasms of the pancreas: diagnosis by endoscopic
ducer for intraductal papillary mucinous tumors of the pancreatic biopsy. Endoscopy 25:290-293
pancreas. Surgery 129:55-65 Ohhashi K, Murakami Y, Maruyama M, Takekoshi T, Ohta H,
Kawarada Y, Yano T, Yamamoto T, Yokoi H, Imai T, Ogura Y, Ohhashi I, Takagi K, Kato Y (1982) Four cases of mucus
Mizumoto R (1992) Intraductal mucin-producing tumors secreting pancreatic cancer. Pro Dig Endosc 20:348-351
of the pancreas. Am J Gastroenterol 87:634-638 Ohta T, Nagakawa T, Akiyama T, Fukushima W, Ueno K,
Kimura W, Makuuchi M, (1999) Operative indications for Miyazaki I, Suzuki M, Matsui 0, Terada T, Nakanuma Y,
cystic lesions of the pancreas with malignant potential-our Kanno M, Uogishi M, Sodani H (1992) The 'duct-ectatic'
experience. Hepatogastroenterology 46:483-491 variant of mucinous cystic neoplasm of the pancreas:
Kobari M, Egawa S, Shibuja K, Shimamura H, Sunamura M, clinical and radiologic studies of seven cases. Am J Gas-
Takeda K, Matsuno S, Fukukawa T (1999) Intraductal papil- troenterol 87:300-304
lary mucinous tumors of the pancreas comprise 2 clinical Onaya H, Itai Y,Niitsu M, Chiba T,Michishita N, Saida Y (1998)
subtypes: differences in clinical characteristics and surgi- Ductectatic mucinous cystic neoplasms of the pancreas:
cal management. Arch Surg 134: 1131-1136 evaluation with MR cholangiopancreatography. AJR Am J
Kobayashi H, Itoh T, Konishi J (1995) Duct ectasia due to RoentgenoI171:171-177
mucus-producing cancers with intraductal extension: his- Pavone E, Mehta SN, Hilzenrat N, Bret P, Lough J, Goresky CA,
topathologic correlation with radiologic imagings. Abdom BarkunAN,Jabbari M (1997) Role ofERCP in the diagnosis
Imaging 20:341-347 of intraductal papillary mucinous neoplasms. Am J Gastro-
Koito K, Namieno T, Nagakawa T, Hirokawa N, Ichimura enterol 92:887-890
T, Syonai T, Yam a N, Someya M, Nakata K, Sakata K, Procacci C, Graziani R, Bicego E, Bergamo-Andreis lA, Main-
Hareyama M (2001) Pancreas: imaging diagnosis with ardi P, Zamboni G, Perderzoli P, Cavallini G, Valdo M, Pisto-
color/power Doppler ultrasonography, endoscopic ultra- lesi GF (1996) Intraductal mucin-producing tumors of the
sonography, and intraductal ultrasonography. Eur Radiol pancreas: imaging findings. Radiology 198:249-257
38:94-104 Procacci C, Megibow A, Carbognin G, Guarise A, Spoto E,
Kubo H, Chijiiwa Y, Akahoshi K, Hamada S, Harada N, Sumii Biasiutti C, Pistolesi GF (1999) Intraductal papillary muci-
T, Takashima M, Nawata H (2001) Intraductal papillary nous tumor of the pancreas: a pictorial essay. Radiograph-
mucinous tumors of the pancreas: differential diagnosis ics 19:1447-1463
Intraductal Papillary Mucinous Tumors: Imaging 137

Procacci C, Carbognin G, Biasiutti C, Guarise A, Ghirardi C, Taki T, Goto H, Naitoh Y, Hirooka Y, Furukawa T, Hayakawa
Schenal G (2001) Intraductal papillary mucinous tumors T (1997) Diagnosis of mucin-producing tumor of the
of the pancreas: spectrum of CT and MR findings with pancreas with intraductal ultrasonographic system. I
pathologic correlation. Eur Radiol11:1939-1951 Ultrasound Med 16:1-6
Raijman I, Kortan P, Walden D, Kandel G, Marcon NE, Haber Taouli B, Vilgrain V, Vullierme MP, Terris B, Denys A, Sauvanet
GB (1994) Mucinous ductal ectasia: cholangiopancreato- A, Hammel P, Menu Y (2000) Intraductal papillary muci-
graphic and endoscopic findings. Endoscopy 26:303-307 nous tumors of the pancreas: helical CT with histopatho-
Rattner DW, Pins MR (1994) Case records of the Massachusetts logic correlation. Radiology 217:757-764
General Hospital. Weekly clinicopathologic exercises. Case Tenner S, Carr-Locke DL, Banks PA, Brooks DC, Van Dam I,
23-1994. A 37-year-old man with several bouts of acute Farraye FA, Turner IR, Lichtenstein DR (1996) Intraductal
pancreatitis and pancreatic-duct filling defects. N Engl I mucin hypersecreting neoplasm 'mucinous ductal ectasia':
Med 330:1671-1676 endoscopic recognition and management. Am I Gastroen-
Rickaert F, Cremer M, Deviere I, Tavares L, Lambilliotte teroI91:2548-2554
IP, Schroder S, Wurbs D, Kloppel G (1991) Intraductal Terris B, Ponsot P, Paye F, Hammel P, Sauvanet A, Molas G,
mucin-hypersecreting neoplasms of the pancreas. A clini- Bernades P, Belghiti I, Ruszniewski P, Flejou IF (2000)
copathologic study of eight patients. Gastroenterology 101: Intraductal papillary mucinous tumors of the pancreas
512-519 confined to secondary ducts show less aggressive patho-
Sciaudone G, Perniceni T, Levy P, Bougaran I, Gayet B (2000) logic features as compared with those involving the main
Enucleation of intraductal papillary-mucinous tumor of pancreatic duct. Am I Surg Pathol 24:1372-1377
the head of the pancreas. Report of 2 cases. Gastroenterol Tibayan F, Vierra M, Mindelzun B, Tsang D, Young H, True-
Clin Bioi 24:121-124 blood W (2000) Clinical presentation of mucin-secreting
Shima Y, Mori M, Takakura N, Kimura T, Yagi T, Tanaka N tumors of the pancreas. Am J Surg 179:349-351
(2000) Diagnosis and management of cystic pancreatic Traverso LW, Peralta EA, Ryan A, Kozerek RA (1998) Intra-
tumours with mucin production. Br I Surg 87:1041-1047 ductal neoplasms of the pancreas. Am I Surg 175:426-432
Sho M, Nakajima Y, Kanehiro H, Hisanaga M, Nishio K, Nagao Van de Stadt J, Closset J, Gelin M (1999) Intraductal papillary
M, Ikeda N, Kanokogi H, Yamada T, Nakano H (1998) Pat- mucinous tumors (IPMT). Ann Surg 230:828-829
tern of recurrence after resection for intraductal papilla Wade TP, Feldman MS, Andrus CH (1997) Spectrum of
mucinous tumors of the pancreas. World I Surg 22: mucus-secreting pancreatic neoplasia. Am J Gastroenterol
874-878 92:154-155
Siech M, Tripp K, Schmidt-Rohlfing B, Mattfeldt T, Gorich I, Wakabayashi T, Kawaura Y, Morimoto H, Watanabe K, Toya D,
Beg HG (1999) Intraductal papillary mucinous tumor of Asada Y, Satomura Y, Watanabe H, Okai T, Sawabu N (2001)
the pancreas. Am I Surg 177:117-120 Clinical management of intraductal papillary mucinous
Silas AM, Morrin MM, Raptopoulos V, Keogan MT (2001) tumors of the pancreas based on imaging findings. Pan-
Intraductal papillary mucinous tumors of the pancreas. creas 22:370-377
AIR Am I RoentgenoI176:179-185 Yamada M, Kozuka S, Yamao K, Nakazawa S, Naitoh Y, Tsu-
Solcia E, Capella C, Kloppel G (1997) Tumors of the pancreas. kamoto Y (1991) Mucin-producing tumor of the pancreas.
Atlas of tumor pathology, 3rd ser, fasc 20. Armed Forces Cancer 68: 159-168
Institute of Pathology, Washington DC Yamaguchi K, Chijiiwa K, Shimizu S, Yokohata K, Morisaki T,
Sugiyama M, Atomi Y (1998) Intraductal papillary mucinous Yonemasu H, Tanaka M (1999) Intraductal papillary neo-
tumors of the pancreas: imaging studies and treatment plasm of the pancreas: a clinical review of 13 benign and
strategies. Ann Surg 228:685-691 four malignant tumours. Eur J Surg 165:223-229
Sugiyama M,Atomi Y (1999) Extrapancreatic neoplasms occur Yamao K, Ohhashi K, Nakamura T, Suzuki T, Shimizu Y,
with unusual frequency in patients with intraductal papil- Nakamura Y, Horibe Y, Yanagisawa A, Nakao A, Nimuara Y,
lary mucinous tumors of the pancreas. Am I Gastroenterol Naito Y, Hayakawa T (2000) The prognosis of intraductal
94:470-473 papillary mucinous tumors of the pancreas. Hepatogastro-
Sugiyama M, Atomi Y, Kuroda A (1997) Two types of mucin- enterology 47:1129-1134
producing cystic tumors of the pancreas: diagnosis and Yamao K, Ohhashi K, Nakamura T, Suzuki T, Watanabe Y, Shi-
treatment. Surgery 122:617-625 mizu Y, Nakamura Y, Ozden I (2001) Evaluation of various
Sugiyama M, Atomi Y, Hachiya I (1998a) Intraductal papil- imaging methods in the differential diagnosis of Intra-
lary tumors of the pancreas: evaluation with magnetic ductal Papillary Mucinous Tumor (IPMT) of the pancreas.
resonance cholangiopancreatography. Am I Gastroenterol Hepatogastroenterology 48:962-966
93:156-159 Yamashita Y, Namimoto T, Mitsuzaki K, Urata J, Tsuchigame T,
Sugiyama M, Atomi Y, Saito M (1998b) Intraductal papillary Takahashi M, Ogawa M (1998) Mucin-producing tumor of
tumors of the pancreas: evaluation with endoscopic ultra- the pancreas: diagnostic value of diffusion - weighted echo
sonography. Gastrointest Endosc 48:164-171 - planar MR imaging. Radiology 208:605-609
Takeyoshi I, Ohwada S, Nakamura S, Ogawa T, Kawashima Y, Yanagisawa A, Ohashi K, Hori M, Takagi K, Kitagawa T, Sugano
Ike T, Morishita Y (1999) Segmental pancreatectomy for H, Kato Y (1993) Ductectatic-type mucinous cystadenoma
mucin-producing pancreatic tumors. Hepatogastroenter- and cystadenocarcinoma of the human pancreas: a novel
ology 46:2585-2588 clinicopathologic entity. Ipn I Cancer Res 84:474-479
Endocrine Tumors
8 Clinical Manifestations and
Therapeutic Management
of Hyperfunctioning Endocrine Tumors
F. CIRILLO, M. FALCONI, and R. BETTINI

CONTENTS 8.1
Introduction
8.1 Introduction 141
8.2 Clinical Data 142
8.2.1 Epidemiological Data A significant percentage of neuroendocrine malig-
and Benign/Malignant Ratio 142 nancies of the gastrointestinal tract arise within
8.2.1.1 Insulinoma 142 the pancreas. From a clinical point of view, pan-
8.2.1.2 Gastrinoma 142 creatic endocrine tumors can be divided into two
8.2.1.3 VIPoma 142 types: nonfunctional (nonsecreting, 'inactive') and
8.2.1.4 Glucagonoma 142
8.2.1.5 Somatostatinoma 143 functional (secreting, 'active'). Functional secreting
8.2.1.6 Other Tumors 143 neuroendocrine tumors are capable of producing
8.2.2 Hormonal Pathophysiology and secreting hormone substances that can evoke
and Clinical Symptoms 144 physiological effects producing well-known symp-
8.2.2.1 Insulinoma 144 toms and signs. Typically, these include flushing and
8.2.2.2 Gastrinoma 144
8.2.2.3 VIPoma 145
chronic diarrhea seen in carcinoid syndrome; hypo-
8.2.2.4 Glucagonoma 145 glycemic crisis in insulin om a; diabetes mellitus and
8.2.2.5 Somatostatinoma 146 necrolytic erythema associated with glucagonoma;
8.2.3 Laboratory Data and Evocative Tests 146 erosive gastritis sometimes associated with diarrhea
8.2.3.1 Insulinoma 146 in the case of gastrinoma.
8.2.3.2 Gastrinoma 147
8.2.3.3 VIPoma 147
The occurrence of pancreatic tumors can be spo-
8.2.3.4 Glucagonoma 147 radic or hereditary and prevalent in individuals with
8.2.3.5 Somatostatinoma 147 a positive family history of hormonal disorders.
8.3 Therapeutic Strategy 148 The increase in the number of diagnosed cases of
8.3.1 Insulinoma 148 digestive endocrine tumors over the past few years is
8.3.2 Gastrinoma 148
8.3.3 VIPoma 149 a result of increased awareness by clinicians, imagers
8.3.4 Glucagonoma 149 and pathologists (CHIT! et al. 2000; ERIKSSON et al.
8.3.5 Somatostatinoma 149 2000a), the routine availability of serum hormone
References 150 assays (ERIKSSON et al. 2000b; SEREGNI et al. 2000),
and improved treatment options (ARNOLD et al.
1996; OBERG 2000).
A recent complete pathological overview of all
these digestive tumors has enabled pathologists
F. CIRILLO, MD to distinguish between well-differentiated tumors
Department of General Surgery II, 'Istituti Ospitalieri' Hos- and endocrine carcinoma with different grades of
pital, Cremona and Post-Graduate School of Endocrinology malignancy (SOLCIA et al. 2000). The combination of
and Metabolic Diseases, Viale Concordia, 1, University of
the old prognostic criteria (tissue type, grading, and
Parma, 26100 Cremona, Italy
M. FALCONI, MD size) with new parameters, e.g., the tumor's biological
Department of Surgical, Gastroenterological Endocrine and activity, angio-invasion, cell mitotic and proliferative
Pancreatic Unit, Hospital 'GB Rossi', University of Verona, index, is crucial for a correct diagnostic and prog-
37134 Verona, Italy nostic assessment.
R. BETTINI, MD
The aim of this chapter is to focus on all these
Department of Surgical, Gastroenterological Endocrine and
Pancreatic Unit, Hospital 'GB Rossi', University of Verona, aspects of functional endocrine tumors arising in
37134 Verona, Italy the pancreas.
142 F. Cirillo et al.

8.2 in the literature, 72% of the primary tumors were


Clinical Data located in the pancreas. Localization in the gastroin-
testinal tract (duodenum, liver biliary ducts, jejunum,
8.2.1 omentum) is less common, and other extragastroin-
Epidemiological Data testinallocations (such as the parathyroid, kidneys,
and Benign/Malignant Ratio ovaries, bone, lung, spleen, and skin) are even more
uncommon (DE BELLIS et al. 1988,1990; PERCOPO et
The most frequent pancreatic functioning endocrine al. 1990a; SANTINI et al. 1991). On average, patients
tumors are insulinomas (representing 17% of cases), are in their 40s at the time of diagnosis, while 2%-
gastrinomas (9% occurrence rate), while VIPomas, 5% of tumors are diagnosed in childhood. Men are
glucagonomas, and somatostatinomas account for more frequently affected than women (63%-7l %)
less than 2%, respectively (CIRILLO 2001). Other, (SANTINI et al. 1991; SOLCIA et al. 1993; CIRILLO
rarer forms of tumor are those involving an ectopic 2001).
production of hormones such as adrenocortical tro- Malignant pancreatic gastrinoma is present in
phic hormone (ACTH), calcitonin, (growth-releasing 70% of cases (SOLCIA et al. 1993); symptomatic dis-
factor (GRF), and parathormone. Table 8.1 summa- ease is associated with a high incidence of metastases.
rizes all the main features of these tumors, which are Pancreatic gastrinoma more frequently presents with
better specified in the individual sections below. nodal metastases than does duodenal gastrinoma
(39% vs 28%), while the difference is slight for hepatic
8.2.1.1 metastases (27% vs 3%) (SOLCIA et al. 1993). In 50%
Insulinoma of patients, gastrinomas represent a hereditary dis-
ease (JENSEN et al. 1983; PIPELEERS-MARICHAL et al.
At this writing, approximately 4000 cases of insu- 1993). In such cases, the parathyroid glands are more
linoma have been reported. Insulinoma represents frequently involved (80.2%), followed by hypophysis
the most common islet-cell tumor with an incidence (19.5%) (CIRILLO 2001).
of 11100,000 cases yearly. Men are affected half as
often as women, and 20%-30% of patients report a 8.2.1.3
positive family history of diabetes mellitus. The peak VIPoma
incidence is between the 4th and 5th decade of life.
Most insulinomas (85%) are single lesions. Child- Over 250 cases of VIPoma have been reported in
hood insulinomas, however, are multiple lesions the literature. The average age at onset is 50 years. It
predominantly located in the head of the pancreas affects women more often than men. Some 80% of
and are known as nesidioblastosis. This entity is VIPomas are single lesions with a diameter ranging
recognized in the presence of focal or diffused b-cell from 1 to 7 cm (a 20 cm nodule was reported in one
hyperplasia. The symptoms related to this entity can case) (STINNER and ROTHMUND 1992).
be managed with pharmaceutical treatment. Extra- Pancreatic VIPomas are localized in 80% of
pancreatic insulinomas are observed rarely (only patients, and 50% of them are malignant; 10%
0.5% of cases). of tumors are localized elsewhere in the adrenal
The rate of malignancy among insulinomas cortex, retroperitoneum, and neck. In childhood, the
reaches 10% and should be suspected in cases observation of elevated plasma levels of vasoactive
where the symptoms are recent and are accompa- intestinal polypeptide (VIP) is usually indicative of
nied by high plasma levels of insulin or pro-insulin. neuroblastoma.
Malignant tumors usually have larger dimensions
(diameter in excess of 2.5 cm), and metastases may 8.2.1.4
already be present at the time of diagnosis (KLOPPEL Glucagonoma
and HEITZ 1990). In 5%-10% of cases, there is a posi-
tive family history (MERREL 1988). The number of documented cases of glucagonoma is
247 (CASADEI et al. 1999). However, the nonspecific
8.2.1.2 nature of many of its symptoms suggests that this
Gastrinoma disease may have a higher incidence (estimated at
around 0.2/100,000/year) (WHIPPLE and FRANTZ
The incidence of gastrinomas is approximately II 1935). The average age at onset is approximately
500,000. Over 2500 cases have been described so far 65 years (range 19-84 years), and 75% of cases
Clinical Manifestations and Therapeutic Management of Hyperfunctioning Endocrine Tumors 143

Table 8.1. Main features of pancreatic hyperfunctioning endocrine tumors


Name a Incidence M/F Age Tumor location Size Malignant Associated Symptoms
(cases/ (years) (em) (%) with
million pop. MEN I
per year) (%)

Insulinoma 10 112 40-60 Pancreas 99.5% 0.12-S 10 5-10 Whipple syndrome (fasting
(beta cells, (head 50%, hypoglycemia attacks,
insulin) tail 50%); glycemia <40 mg/dl during
extrapancreatic attacks, remission of symp-
0.5% toms following food intake);
neuropsychiatric symptoms;
tachycardia; obesity
Gastrinoma 2 5/3 40 Pancreas SS%; O.I-IS 70 20-25 Zollinger-Ellison syndrome
(G cells, duodenum 13%; (pancreas); (peptic ulcer 93%, diarrhea
gastrin) other sites 2% 38 40%, steatorrhea 30%)
(duodenum)

VIPoma (VIP) 0.05-0.2 3/5 45-55 Pancreas 90%; 0.6-20 SO Verner Morrison syndrome
extrapancreatic (diarrhea 90%, electrolyte
(adrenal cortex, imbalance 50%-70%, hyper-
retroperitoneum, glycemia 50%, flushing
neck) 10% 20%)
Glucagonoma 0.2 111 45-70 Pancreas 3-35 65 Diabetes 90%; necrolytic
(alpha cells, (head 12%, erythema 70%; diarrhea
glucagon) body 38%, 20%
tail 50%)
Somato- Rare 111 40-50 Pancreas 35%; 2-10 70
statinoma extrapancreatic: (pancreas) Diabetes; gallstones;
(delta cells, duodenum 52%, SO diarrhea/steatorrhea
somatostatin) other 13% (duodenum)

aIn parentheses: type of cells and peptide secreted

have been diagnosed in individuals between 45 and in the duodenum than in the pancreas (51.6% and
70 years of age (GUILLAUSSEAU et al. 1995). No gen- 34.7%, respectively). The rate of malignancy varies
der differences have been noted (BODEN et al. 1977; depending on the primary location of the tumor: 70%
PRINZ et al. 1981). Only rarely has it been possible in the pancreas, 50% in the duodenum, and practi-
to observe hereditary forms of glucagonoma with cally all rare somatostatinomas located outside of the
contemporary elevation of different peptides such gastrointestinal tract (NEGRO and PERCOPO 1988;
as insulin, gastrin, somatostatin, VIP, and pancreatic CIRILLO 2001).
polypeptide (PP) (OHNEDA et al. 1979).
Glucagonomas are indeed malignant in 60%-70% of 8.2.1.6
cases. Most metastases involve the liver, other possible Other Tumors
locations being the vertebrae, adrenal cortex, and nodes.
In 50%-77% of cases, the primary tumor is located Several other types of functional tumors have been
in the pancreatic tail and may reach a large diam- described in the literature even though they occur
eter. Lastly, most glucagonomas are functional tumors infrequently. The only reliable epidemiological data
(BODEN 1987; MOZELL et al. 1990; NORTON 1994). concern carcinoids located in the pancreas, of which
43 cases between 1838 (one year before Merling's
8.2.1.5 description of intestinal carcinoid) and 1998 have
Somatostatinoma been reported. They account for 0.5% of all gastro-
enteropancreatic neuroendocrine tumors. This rare
This tumor most frequently affects individuals in variety mainly involves the pancreatic head and
their 40s and 50s with no particular gender pre- tail, while 29.2% of cases presented with multiple
dominance. Somatostatinomas more frequently arise localizations.
144 F. Cirillo et al.

The patients affected were all men in their 50s. The As a result, these patients may be erroneously
carcinoid syndrome was present in 64.3% of cases, considered as psychiatric, cardiopathic, or epileptic
and in 88.4% the tumor proved to be malignant, with individuals and be treated as such, even for long
metastases present at the moment of diagnosis in the periods of time.
great majority of cases.
8.2.2.2
Gastrinoma
8.2.2
Hormonal Pathophysiology The term gastrinoma refers to a type of pancreatic
and Clinical Symptoms tumor originating from a cellular population called
G-cells. This tumor is responsible for the pathological
8.2.2.1 production of gastrin, the hormone capable of stimu-
Insulinoma lating acid secretion in the stomach.
This tumor and the associated clinical symptoms
The term insulinoma defines a tumor originating were first reported by ZOLLINGER and ELLISON in
from the B-cells of the pancreas. This tumor was 1955 (GREGORY et al. 1960; ZOLLINGER et al. 1980).
first described by NICHOLLS in 1902 (NICHOLLS Several years later, Gregory succeeded in isolating
1902; WHIPPLE et al. 1935). However, the detailed gastrin, the peptide responsible for the clinical symp-
description of this pathology is credited to WHIPPLE toms, from the tumor cells (GREGORY et al. 1960). The
(WHIPPLE and FRANTZ 1935). He described a triad of presence of hypergastrinemia, resulting in a second-
clinical symptoms characteristic of the insulinoma ary hypersecretion of chloridric acid, constitutes the
syndrome: hypoglycemic attacks during fasting, Zollinger-Ellison syndrome.
glycemia levels of less than 40 mg/dl during attacks, The specific symptoms of gastrinomas are ab-
and a rapid remission of symptoms following food dominal pain (>75%) and diarrhea (50%).
consumption. Hypoglycemia is a frequent symptom In 93% of cases, the abdominal pain can be related
of insulinoma with the typical neuropsychiatric, car- to the presence of ulcer disease (JENSEN 1996), to an
diovascular, and digestive implications. increase in bowel movements in 14%, and to neo-
Neuropsychiatric symptoms are present in 80% plastic infiltration of the nerves in 2.4% (JENSEN et al.
of cases and may be triggered by fasting or physical 1983). In 25% of patients, abdominal pain is the only
exercise. Harrington defined them as 'funny turns' symptom (JENSEN and GARDNER 1993). Peptic ulcers
in order to underline the extreme variability of are located in the proximal portion of the duodenum
behavior that these symptoms cause (HARRINGTON in 75% of cases and less frequently in the distal part
1983). Manifestations may include neurovegetative of the stomach. Location in the distal part of the
disorder (such as nausea, vomiting, dizziness, duodenum or the proximal portion of the jejunum is,
asthenia, diplopia), automatism, psychomotor however, quite rare. The esophagus may be involved
agitation, epileptic-type attacks, paresis, paraly- in 35% of cases, with possible complications such as
sis, irritability, disorientation, and torpor and, in perforation, neoplastic transformation, or Barrett
some cases, may result in coma secondary to a esophagus.
sharp decline in glycemic levels. These symptoms The diarrhea, initially of an intermittent nature,
may be preceded by a perceptive aura (FILIPI and is associated with steatorrhea in a third of patients,
HIGGINS 1973; STEFANINI et al. 1974; PERCOPO et and in 7% it is the only clinical manifestation in the
al. 1990b). In 7% of cases, there may be permanent absence of ulcer disease. Four different pathophysi-
brain damage. ological theories have been put forward to explain
Cardiovascular manifestations may include the presence of diarrhea and steatorrhea in patients
tachycardia, extrasystoles, atrial fibrillation, angina, affected by gastrinoma:
sweating, and pallor. These symptoms, present in - the increased gastric secretion reduces the
approximately 10%-15% of patients, are secondary intestinal absorption; the presence of hypergas-
to the liberation of adrenaline during marked hypo- trinemia induces a reduction in the absorption
glycemic episodes. of water and electrolytes in the gut. Indeed, the
Hunger pains mainly represent the gastrointes- increased secretion of chloridric acid induces the
tinal symptoms (4%-8% of cases). Patients with conversion of pepsinogen into pepsin, the action
insulinoma thus become polyphagic and, in the long of which alters the permeability of the intestinal
term, obese. mucosa;
Clinical Manifestations and Therapeutic Management of Hyperfunctioning Endocrine Tumors 145

- the lowering of the duodenal pH results in the hypokalemia, hypochlorhydria, and consequently
inactivation of the pancreatic lipases and the metabolic acidosis (VERNER and MORRISON 1958,
precipitation of biliary acids, thus making fat 1974). MARKS applied the acronym WDHA to this
absorption difficult; syndrome (watery diarrhea hypokalemia hypochlor-
- the nonabsorbed fatty acids stimulate the secretive hydria) (STINNER and ROTHMUND 1992).
activity of the colon; This syndrome has also been defined as 'pan-
- an additional cause of diarrhea could be the creatic cholera'. This name is inappropriate when the
hypersecretion of other enteric hormones that disease arises in locations other than the pancreas.
have a stimulating effect on the motility and The observation of elevated plasma levels of vasoac-
secretive activity of the gastrointestinal tract. tive intestinal peptide (VIP) gave rise to the currently
used term - VIPoma.
The alteration of the gastrointestinal motility The VIPoma syndrome is mainly characterized by
is another factor which plays a significant role in the presence of profuse and progressively worsening
the pathogenesis of diarrhea in patients with gas- watery diarrhea (l-61!day). Electrolyte imbalances
trinoma. It has, in fact, been demonstrated that the are secondary to the diarrhea and may result in
gastric emptying in these individuals is possibly lethargy, muscular asthenia, cardiac conduction dis-
accelerated because the receptors are not able to orders, weight loss, abdominal pain, and dyspepsia.
regulate the gastroduodenal peristalsis. Flushing may be present in 20% of cases and, more
Furthermore, 7% of all gastrin om as are clinically rarely, acute tubular necrosis.
silent. In these cases, the onset of disease may take During its initial phases, the Verner-Morrison
a different course, appearing as a result of digestive syndrome may be confused with other forms of
disturbances or compressive disease. Gastrointes- diarrhea of bacterial, viral, or parasitic origin or with
tinal hemorrhages have been described in 46% of ulcerative diseases (such as ulcerative colitis and
patients, intestinal occlusion in 20%, and perforation Crohn disease). The differential diagnosis should
in 6% (DE BELLIS et al. 1990). also be made in the face of other endocrine diar-
Gastrinomas may be present in 0.1 %-1 % of all rheogenic tumors.
patients suffering from peptic ulcer disease. How-
ever, the diagnosis may not be immediate (the period 8.2.2.4
of latency may extend for up to 6 years) (JENSEN and Glucagonoma
GARDNER 1993). This is partly due to the fact that the
abdominal pain might not be recognized as a symp- Glucagon, secreted by the alpha-cells of the pan-
tom of gastrinoma and also to the indiscriminate use creatic islets, is a hormone which plays a fundamental
of antisecretive drugs able to mask the clinical man- role in the metabolism of sugars. An inappropriate
ifestations of these tumors, particularly in the initial secretion of glucagon gives rise to a syndrome first
phases (ZOLLINGER et al. 1980; DE BELLIS et al. 1988). described by BECKER in 1942 (BECKER et al. 1942),
Such a late diagnosis may account for the frequent with manifestations involving the skin, such as
observation of metastases at the time of diagnosis Wilkinson migrant necrolytic erythema (WILKINSON
(50%-60% of cases). 1973), angular stomatitis, chelitis, atrophic glossitis,
Therefore, the diagnosis of a gastrinoma should and, more rarely, purpura.
always be suspected in patients with the following: (1) The pathogenesis of the cutaneous lesions is prob-
multiple ulcerations of the upper digestive tract resis- ably multifactorial. The reduced concentration of
tant to pharmacological treatment or recurring quickly alanine and glutamine amino acids in the blood may
after targeted surgical treatment; (2) secretive diarrhea be one of its causes, thus excluding a direct relation
of unknown origin; (3) highly positive family history of between the occurrence of skin disease and elevated
peptic ulcer disease; (4) a positive individual or family plasma concentrations of glucagon (NORTON et al.
history of parathyroid and/or hypophysial tumors; (5) 1979).
acid hypersecretion and/or hypergastrinemia. Apart from the skin lesions, this syndrome is char-
acterized by a marked loss of weight, normochromic
8.2.2.3 normocytic anemia, reduced plasma concentrations of
VIPoma alanine, glutamine, essential fatty acids, and zinc, all due
to the catabolic effect of glucagon. Some 20% of patients
In 1958, VERNER and MORRISON described two have diarrhea, which may be caused by the hypersecre-
patients with a syndrome characterized by diarrhea, tion of other enteric hormones besides glucagon.
146 F. Cirillo et al.

In other cases, constipation may be present, the D-cells have been reported to be involved in
gravity of which may reach the stage of intestinal other digestive endocrine tumors, too (such as
subocclusion; glucagon is indeed a powerful inhibitor gastrinomas and insulinomas), even if no clear
of intestinal motility. somatostatinoma-related syndrome is present
Other symptoms reported include neurological (WHEELER et al. 1986).
alterations probably due to the action of a circu- In the majority of cases, the diagnosis of
lating factor capable of inducing encephalomyelitis. somatostatinoma is retrospective, mostly due to
Thrombophlebitis and pulmonary embolism, whose the nonspecific clinical presentation. In other cases,
elevated frequency justifies the use of prophylactic the tumor may be recognized incidentally during
treatment, may be among the manifestations with an intervention to remove gallstones, or follow-
unclear pathogenesis. ing other investigations for disorders that are part
of the complications of the primary or metastatic
8.2.2.S lesion (such as hemorrhage, abdominal pain, and
Somatostatinoma jaundice). In such cases, the tumors may reveal sig-
nificant growth (over 5 cm in diameter) and distant
Somatostatin is a hormone primarily isolated from metastases in almost 74% of patients (MOZELL et al.
the pituitary as a metabolic regulator of the growth 1990; DELCORE and FRIESEN 1994; NORTON 1994).
hormone. Somatostatin is synthesized in the delta- In 75% of patients, the tumor may induce the secre-
cells present in the pancreas and throughout the tion of other hormones (such as calcitonin, PP, glu-
entire gastrointestinal tract (SOLCIA et al. 1993). cagon ACTH), which may account for the variable
The first observation of a tumor with a pre- clinical presentation.
dominant secretion of somatostatin dates back to
1977 and was reported by LARSSON et al. (1977). This
tumor can be either functional or asymptomatic. In
the latter case, the diagnosis can be secondary to the 8.2.3
occurrence of biliary obstruction due to the frequent Laboratory Data and Evocative Tests
localization of the tumor in the pancreatic ampulla.
When biologically active, this tumor is characterized 8.2.3.1
by the presence of a syndrome of extremely variable Insulinoma
clinical symptoms, which reflect the entire range of
the pharmacological actions of somatostatin. The laboratory diagnosis of insulinom a is based on
Acquired diabetes is present, even if modest, due to the determination of alterations in serum glucose
the inhibitory effect of somatostatin on both insulin and insulin levels measured at baseline or during
and glucagon. Biliary stasis may occur secondary to attacks. It is especially important to carry out a con-
the reduced function of the gallbladder, which subse- temporary measurement of both serum glucose and
quently results in the formation of stones (8l.5%). insulin under fasting conditions. The diagnostic test
The altered biliary and intestinal motility, in asso- of choice is the fasting glucose tolerance test, with
ciation with the reduced pancreatic secretion, causes insulin and C-peptide levels taken every 6 h from
diarrhea and steatorrhea (77.8%). In several cases, the onset of symptoms. Patients in whom the serum
hypochromic anemia (55.5%), weight loss (43%), glucose/insulin ratio is less than or equal to 1 should
and hypo- or achlorhydria (29.6%) may be present be suspected of having insulinoma.
(NEGRO and PERCOPO 1988). Most insulinomas can produce an insulin pre-
The classic triad (diabetes/gallstones/diarrhea- cursor known as pro-insulin. The determination
steatorrhea) can, in some cases, be accompanied by of the levels of this metabolite and of C-peptide, a
clinical symptoms secondary to the production of by-product of the conversion process of pro-insu-
other enteric hormones. Such variability of the clin- lin to insulin, may further facilitate the diagnosis
ical presentation may also be due to the secretion of of this type of tumor (DELCORE and FRIESEN 1994;
atypical derivatives of somatostatin characterized by NORTON 1994). It should be stressed, however, that
different biological actions. 10%-25% of patients with insulinoma may have
The simultaneous occurrence of somatostatinoma, a normal level of pro-insulin and also that a high
pheochromocytoma, and neurofibromatosis of von concentration of this metabolite may be observed
Recklinghausen is the cause of a rare polyendo- in otherwise healthy obese individuals (GRANT
crinopathy. 1993).
Clinical Manifestations and Therapeutic Management of Hyperfunctioning Endocrine Tumors 147

The measurement of C-peptide plasma levels may 8.2.3.3


be extremely useful in those patients who do not have VIPoma
overt hypoglycemia under fasting conditions.
In addition to the above alterations, patients with In 75% of patients, hypercalcemia combined with
insulinoma may also have an elevated level of chori- hypophosphatemia is also present, probably due to
onic gonadotropin (HCG) (KAHN et al. 1977; OBERG the presence of associated hyperparathyroidism. The
and WIDE 1981). hormones secreted by the tumor may themselves
The use of pharmacological loading tests (using induce hypercalcemia.
tolbutamide calcium, glucagon, or leucine) has been Approximately half of the patients with VIPoma
discontinued due to the high percentage of false pos- have hyperglycemia due to reduced glucose tol-
itives and their potential risks. On the contrary, those erance. Hypo-achlorhydria is present in 40% of
dynamic tests still practiced (long-term fasting and patients, flushing in 20%.
administration of exogenous insulin) tend to high- Patients with VIPoma have extremely elevated
light the increase in the glucose/insulin ratio under plasma concentrations of VIP (in different molecular
specific conditions. forms). Other diarrheogenic substances, such
as secretin, pancreatic polypeptide (PP), gastric
8.2.3.2 inhibitory polypeptide (GIP), prostaglandin E2,
Gastrinoma and peptide histidine isoleucine (PHI), may also be
increased (BLOOM et al. 1983).
All patients with gastrinoma have markedly elevated
serum gastrin levels (exceeding 1000 pg/ml), estab- 8.2.3.4
lishing the diagnosis. It must be remembered that Glucagonoma
serum gastrin may be elevated in other diseases, and
therefore, more refined laboratory tests may be applied The existence of glucagonoma should always be
to make a specific diagnosis. suspected when diabetic patients display the typical
Elevated basal acid output (BAO) indicates a rash. The diagnosis is confirmed by the observa-
pathologic hypersecretion (>15 mEq/h) in 85% of tion of elevated plasma levels of glucagon. Other
patients. This observation is significant in the diag- conditions characterized by high glucagon blood
nosis of gastrinoma only when the serum gastrin concentration are chronic renal failure, chronic liver
level exceeds 1000 pg/ml. However, patients with disease, Cushing syndrome, prolonged fasting, and
gastrinoma with BAO values of less than 5 mEq/h acute pancreatitis. Loading tests (using arginine
have been described. In the absence of serum gas- tolbutamide calcium or calcitonin) may be useful
trin levels> 1000 pg/ml, a BAO/MAO (maximal acid in diagnosing doubtful cases. The continuous oral
output) relationship >0.6 is not pathognomonic of or parenteral administration of somatostatin and
gastrinoma. glucose further facilitates the diagnosis.
Loading tests may be of help in the differential
diagnosis of patients with gastrin levels ranging from 8.2.3.5
100 to 1000 pg/ml, gastric pH <2.5, and clinical symp- Somatostatinoma
toms suggestive, but not specific, of gastrinoma. The
loading test using secretin is currently considered the Low, reduced plasma concentrations of growth
most reliable as regards sensitivity and specificity. hormone and glucagon, together with the reduced
In 90% of patients with gastrin om a, the adminis- sensitivity of blood insulin levels to exogenous glu-
tration of secretin induces a rapid (within 2 min) cose treatment, may steer the clinician towards the
and massive (>200 pg/ml) increase of gastrin blood diagnosis of somatostatinoma. Other dynamic tests
concentration, which returns to the pretest level after (tolbutamide calcium-pentagastrin secretin) may be
30 min (MCGUIGAN and WOLFE 1980). This test will useful in dubious cases. However, a definitive diag-
be positive in 87% of patients with serum gastrin lev- nosis can be formulated when the plasma concen-
els varying between 100 and 1000 pg/ml. In patients trations of somatostatin exceed 100 pmolll.
with achlorhydria with pH levels> 2.5, false positives In 75% of patients, the tumor may induce the
may be encountered (FELMAN et al. 1987). secretion of other hormones (such as calcitonin, PP,
Other commonly employed loading tests are those glucagon, ACTH), which may account for a variable
using bombesin, glucagon, calcium, and standard clinical presentation.
proteic meal.
148 F. Cirillo et aI.

8.3 cannot control the clinical symptoms satisfactorily


Therapeutic Strategy (DOHERTY et al. 1991; VINIK et al. 1991; MILLER
1993).
The object of the therapy is, whenever possible, pri- Surgical treatment of benign insulinom as may be
marily cure. If this aim cannot be achieved, then the limited to the simple excision of the mass (if single)
clinician must at least try to manage the symptoms or to the resection of a pancreatic segment (usually
of endocrine hyperfunction, since these are some- caudal). Malignant insulinomas on the contrary
times so incapacitating that they seriously affect the require a more extensive and radical treatment. Such
patient's quality of life (NORTON 1994). When sur- aggressive resection involves all the metastatic local-
gery is radical, it is able to achieve both objectives. As izations and ensures an improvement in the quality
regards the therapeutic strategy of these neoplasms, of life. In fact, a partial or incomplete removal of
this is still the first option to consider. This is also true the tumor does not allow adequate control of the
for those cases where, when the clinical and labora- hypoglycemia (possible in approximately 20% of
tory diagnosis is certain, the radiologist is not able patients).
to identify the disease. Where radical resection is not In metastatic disease, diazoxide and somatostatin
possible, the surgeon has more options. Debulking analogues are considered. Octreotide controls symp-
increases the possibility of reducing the symptoms toms well in 25%-30% of patients but only at a high
and may be an option because of the slow growth dosage (VERSCHOOR et al. 1986).
of many of these lesions. Nevertheless, the surgeon
should not forget, especially when controlling the
symptoms, that different therapeutic choices also 8.3.2
exist (analogues, diazoxide, interferon, proton pump Gastrinoma
inhibitors, etc.). The therapeutic strategy should be
dealt with according to the particular aspects of each Gastrinomas, too, can also be clinically diagnosed
type of tumor. without definite identification of the site of the dis-
ease. A correct intraoperative examination leads to
the identification of 95% of those gastrinomas not
8.3.1 seen at imaging. Aside from careful exploration and
Insulinoma palpation of the pancreatic parenchyma, a thorough
study of the entire peritoneal cavity should be done,
The presence of characteristic clinical symptoms since there is also the possibility of disease in the
and the positive results of valid laboratory tests extrapancreatic area. Small gastrin om as located in
make the diagnosis of insulinoma definite even the submucosa of the duodenal wall can be found by
when the tumor is still hidden in 10%-20% of trans-illumination or by longitudinal duodenectomy
patients despite significant improvement in imag- of about 3 cm for a correct palpation of the entire
ing. In this situation, a rigorous surgical procedure mucosal surface, even of the medial wall. Norton
leads to the identification of a tumor (GIANELLO et recently pointed out that a duodenectomy for gastri-
al. 1988; ZEIGER et al. 1993; ROTHMUND 1994). In nomas in this site is the best solution (NORTON 1999)
any case, the surgeon must perform a wide exposure as it is able to identify small gastrinomas that were
to make the entire gland accessible to palpation. 'missed' at palpation, intraoperative ultrasound, and
The whole parenchyma should be studied with the trans-illumination. Intraoperative ultrasound shows
help of an intraoperative ultrasound using a 10 or 91 % of gastrinomas located in the pancreas and
7.5 MHz probe (KLOTTER et al. 1987; GRANT et al. about 30% of the duodenal ones (NORTON 1995). In
1988; NORTON et al. 1988; BOTTGER et al. 1990). In all, this procedure is able to identify from 64% to 92%
this way, small lesions can be seen, which gives an of lesions (NORTON et al. 1992).
important advantage over mere palpation, especially If at the end of the staging procedures no lesion
when the neoplasm is in the head of the pancreas has been identified, the surgeon should not turn to a
(NORTON 1999). 'blind resection' or a total gastrectomy. It should not
Whenever localization is impossible, even during be forgotten that, with proton pump inhibitors, med-
surgery, reports in the literature state that 'blind ical therapy today is able to control symptoms in the
resection' can be carried out only when the 'venous majority of patients.
sampling' is definitely positive in 'regionalizing' the The therapeutic approach to gastrinoma has
lesion (NORTON 1994) and when medical therapy changed radically since the advent of synthetic
Clinical Manifestations and Therapeutic Management of Hyperfunctioning Endocrine Tumors 149

antisecretive molecules. The increasingly accurate 8.3.3


topographic diagnosis has led to the definitive VIPoma
suspension of the once abused gastrectomy. These
changes created the opportunity for better-targeted Given the presence of invalidating symptoms, sur-
interventions, which vary from a simple excision in gery is the first treatment and is often indicated
cases of a single and well-delineated benign pancre- when there are also metastases (AKERSTROM 1992;
atic lesion to the more articulated intervention of STINNER and ROTHMUND 1992; GRANT 1993; OBERG
pancreatic surgery. The best intervention is debatable 1996; WIEDENMANN et al. 1998). Treatment with
since the authors who propose surgical enucleation somatostatin analogues is able to reduce diarrhea
as a first choice report that only 51 % of patients are by regularizing the electrolyte imbalance. Shrinkage
clear of disease immediately following surgery. Their of metastases was observed in a few cases (CLEMENTS
choice is due to the fact that the survival rate is still and ELIAS 1986).
94% after 10 years (NORTON et al. 1999). According to
other authors (IMAMURA et al. 1989; DELCORE and
FRIESEN 1992; UDELSMAN et al. 1993; STADIL 1995; 8.3.4
PHAN et al. 1998; THODIYIL et al. 2000), this opinion Glucagonoma
is not altogether correct since, in experienced cen-
ters, the mortality rate following a pancreaticodu- Due to the nonspecific clinical presentation, this
odenectomy is less than 2% and guarantees greater tumor is normally diagnosed late, when metastases
radicality. are already present. To date, surgical treatment is
In the past, the 5-year survival rate of patients considered the only effective approach, giving satis-
with gastrinoma was approximately 69% in patients factory results in approximately one-third of patients
undergoing radical surgery and 38% in patients with (HIGGINS et al. 1979). Surgical intervention will con-
advanced disease. The respective survival rates at sist of the most radical pancreatic resection possible
10 years were 62% and 27% (ELLISON et al. 1987). depending on the location of the lesion within the
Norton's more recent results indicate how an even pancreas. Moreover, the surgeon is perfectly justi-
earlier diagnosis followed by an adequate surgical fied in performing debulking interventions, even
and pharmacological treatment could improve the repeatable ones, in the attempt to control particularly
survival of these patients and reduce the number of incapacitating clinical symptoms. For these patients,
neoplastic complications (NORTON et al. 1999).A sig- the preoperative use of somatostatin analogues can
nificant number of patients (25%) tend, in any case, be helpful in controlling clinical symptoms (AKER-
to develop distant and mainly skeletal metastases STROM 1992; STINNER and ROTHMUND 1992; GRANT
(30% with liver metastases), which further impair 1993; OBERG 1996; WIEDENMANN et al. 1998).
the patient's prognosis (BARTON et al. 1986; NORTON Chemotherapy may produce a response in a num-
and JENSEN 1991; ZAYENE et al. 1997). ber of patients without prolonging survival (KESS-
Long-term treatment of gastric acid hyperse- INGER et al. 1983; MOERTEL et al. 1992; DELCORE and
cretion involves antisecretory molecules like Hz- FRIESEN 1994). There is evidence of partial remission
antagonists (ranitidine) or proton pump inhibitors after treatment with somatostatin analogues and with
(in particular omeprazole) and somatostatin ana- interferon (SILLER et al. 1994; STROHM 1996). In any
logues (octreotide, lanreotide). Omeprazole provides case, metastasis recurrence is extremely frequent.
for a good control of symptoms with a dosage vary-
ing from 40 to 160 mg daily (METz and JENSEN 1995).
The other pump inhibitors, such as pantoprazole, 8.3.S
lansoprazole, and rabeprazole, show different char- Somatostatinoma
acteristics of efficacy and potency. Octreotide, even
in low doses, seems able to control gastrin secretion As already described for VIPomas and
in order to reduce the daily antisecretory therapy. somatostatinomas, the surgical approach must be
Proglumide prostaglandin analogues and anticho- as aggressive as possible, justifying the association
linergic molecules show a low antisecretory activity of pancreatic demolition, even with vascular recon-
in association with many side effects. struction, to extensive debulking of eventual hepatic
metastases (AKERSTROM 1992; STINNER and ROTH-
MUND 1992; GRANT 1993; OBERG 1996; WIEDENMANN
et al. 1998). Nevertheless, given the typically virulent
150 F. Cirillo et al.

development of this tumor, the survival rate 1 year Doherty GM, Doppman JL, Shawker TH, Miller DL, Eastman
after diagnosis is approximately 48%, whatever the RC, Gorden P, Norton JA (1991) Results of a prospective
treatment (HARRIS et al. 1987; KONOMI et al. 1990). strategy to diagnose, localize, and resect insulinomas. Sur-
gery 110:989-996; discussion 996-997
Ellison EC, Carey LC, Sparks J, 0' Dorisio TM, Mekhjian HS,
Fromkes JJ, Caldwell JH, Thomas FB (1987) Early surgical
treatment of gastrinoma. Am J Med 82: 17 - 24
References Eriksson B, Bergstrom M, Orlefors H, Sundin A, Oberg K,
Uingstrom B (2000a) Use of PET in neuroendocrine
Akerstrom G (1992) Surgical treatment of endocrine pan- tumors. In vivo applications and in vitro studies. Q J Nucl
creatic tumors. Diagn Oncol 2:332-337 Med 44:68-76
Arnold R, Trautmann ME, Creutzfeldt W, Benning R, Benning Eriksson B, Oberg K, Stridsberg M (2000b) Tumor markers in
M, Neuhaus C, Jurgensen R, Stein K, Schafer H, Bruns C, neuroendocrine tumors. Digestion 62 [SuppllJ:33-38
Dennler HJ (1996) Somatostatin analogue octreotide and Felman M, Schiller LR, Walsh JH, Fordtran JS, Richardson CT
inhibition of tumour growth in metastatic endocrine gas- (1987) Positive intravenous secretin test in patients with
troenteropancreatic tumours. Gut 38:430-438 achlorhydria-related hypergastrinemia. Gastroenterology
Barton JC, Hirschowitz BI, Maton PN, Jensen RT (1986) Bone 93:59-62
metastases in malignant gastrinoma. Gastroenterology 91: Filipi CJ, Higgins GA (1973) Diagnosis and management of
1179-1185 insulinoma. Am J Surg 125:231-239
Becker SW, Kahn D, Rothman S (1942) Cutaneus manifest- Gianello P, Gigot JF, Berthet F, Dardenne AN, Lambotte L,
ations of intestinal malignant tumors. Arch Dermatol Rahier J, Otte JB, Kestens PJ (1988) Pre- and intraopera-
Syph 45:1069-1080 tive localization of insulinomas: report of 22 observations.
Bloom SR, Christofides ND, Yiangou T, Blank MA, Tatemoto World J Surg 12:389-397
K, Polak JM (1983) Peptide histidine isoleucine (PHI) and Grant CS (1993) Surgical management of malignant islet cell
Verner-Morrison syndrome. Gut 24:A473 tumors. World J Surg 17:498-503
Boden G (1987) Insulinoma and glucagonoma. Semin Oncol Grant CS, van Heerden J, Charboneau JW, James EM, Reading
14:253-262 CC (1988) Insulinoma. The value of intraoperative ultraso-
Boden G, Owen OE, Rezvani I, Elfenbein BI, Quickel KE (1977) nography. Arch Surg 123:843-848
An islet cell carcinoma containing glucagon and insulin. Gregory R, Tracy HJ, French JM, Sircus W (1960) Extraction of
Chronic glucagon excess and glucose homeostasis. Diabe- gastrin-like substance from a pancreatic tumor in case of
tes 26:128-137 Zollinger-Ellison syndrome. Lancet 1:1045-1048
Bottger TC, Weber W, Beyer J, Junginger T (1990) Value of Guillausseau P, Guillausseau-Scholer C (1995) Glucagonomas:
tumor localization in patients with insulinoma. World J clinical presentation, diagnosis and advances in manage-
Surg 14: 107-112; discussion 112-114 ment. Endocrine tumors of the pancreas. M Mignon and R
Casadei R, Tomassetti P, Rossi C, La Donna M, Migliori M, Mar- Jensen, Basel. Front Gastrointest Res 23:183-193
rano D (1999) Treatment of metastatic glucagonoma to the Harrington MG (1983) A prospective survey for insulinomas
liver: case report and literature review. Ital J Gastroenterol in a neurology department. Lancet 1:1094-1095
Hepatol 31 :308-312 Harris GJ, Tio F, Cruz AB Jr (1987) Somatostatinoma: a case
Chiti A, BrigantiV, Fanti S, Monetti N, Masi R, Bombardieri report and review of the literature. J Surg Oncol 36:8-16
E (2000) Results and potential of somatostatin receptor Higgins GA, Recant C, Fischmann AB (1979) The glucagonoma
imaging in gastroenteropancreatic tract tumours. Q J Nucl syndrome: surgically curable diabetes. Am J Surg 137:
Med 44:42-49 142-148
Cirillo F (2001) Clinica e fisiopatologia dei tumori neuro- Imamura M, Takahashi K, Isobe Y, Hattori Y, Satomura K, Tobe
endocrini gastroentero-pancreatici. In: Cirillo F (ed) I T (1989) Curative resection of multiple gastrinomas aided
tumori neuroendocrini gastroenteropancreatici. Manuale by selective arterial secretin injection and intraoperative
di diagnosi e trattamento, 3rd edn. Casa Editrice Ambro- secretin test. Ann Surg 210:71 0-715
siana, Milano Jensen RT (1996) Gastrointestinal endocrine tumours. Gastri-
Clements D, Elias E (1986) Regression of metastatic VIPoma noma. Baillieres Clin Gastroenterol 10:603-643
with somatostatin analogue SMS (letter). Lancet 1:874 Jensen RT, Gardner JD (1993) Gastrinoma. In: Go VLD, Di
De Bellis E,Percopo V, Lorenzo M, Odierna G, Tesauro B (1988) Magno EP, Gardner JD et al (eds) The pancreas: biology,
Gastrinomas. First world post graduate surgical week of pathobiology and disease, 2nd edn. Raven, New York,
the University of Milan, Lecture book III pp 931-978
De Bellis E, Percopo V, Lorenzo M, Odierna G, Miceli G, De Jensen RT, Gardner JD, Raufman JP, Pandol SJ, Doppman JL,
Bellis M (1990) Biologically active gastrinomas. Current Collen MJ (1983) Zollinger-Ellison syndrome: current con-
therapeutic approach. Second world week of professional cepts and management. Ann Intern Med 98:59-75
updating in surgery and in surgical and oncological disci- Kahn CR, Rosen SW, Weintraub BD, Fajans SS, Gorden P
plines of the University of Milan (1977) Ectopic production of chorionic gonadotropin
Delcore R, Friesen SR (1992) Role of pancreatoduodenectomy and its subunits by islet cell tumors. N Engl J Med 297:
in the management of primary duodenal wall gastrinomas 565-569
in patients with Zollinger-Ellison syndrome. Surgery 112: Kessinger A, Foley JF, Lemon HM (1983) Therapy of malignant
10 16-1022; discussion 1022-1023 APUD cell tumors. Effectiveness of DTIC. Cancer 51:
Delcore R, Friesen SR (1994) Gastrointestinal neuroendocrine 790-794
tumors. J Am Coli Surg 178:187-211 Kloppel G, Heitz PU (1990) Morphology and functional activity
Clinical Manifestations and Therapeutic Management of Hyperfunctioning Endocrine Tumors 151

of gastroenteropancreatic neuroendocrine tumours. Rec Res Oberg K (2000) Interferon in the management of neuroen-
Cancer Res 118:27-36 docrine GEP-tumors: a review. Digestion 62 [Suppl1J:
Klotter HJ, Ruckert K, Kummerle F, Rothmund M (1987) The 92-97
use of intraoperative sonography in endocrine tumors of Oberg K, Wide L (1981) hCG and hCG subunits as tumour
the pancreas. World J Surg 11:635-641 markers in patients with endocrine pancreatic tumours
Konomi K, Chijiiwa K, Katsuta T, Yamaguchi K (1990) Pan- and carcinoids. Acta Endocrinol (Copenh) 98:256-260
creatic somatostatinoma: a case report and review of the Ohneda A, Otsuki M, Fujiya H,Yaginuma C, Kokubo T,Ohtani
literature. J Surg Oncol 43:259-265 H (1979) A malignant insulinoma transformed into a glu-
Larsson U, Hirsch MA, Holst JJ, Ingemansson S, Kuhl C, Jensen cagonoma syndrome. Diabetes 28:962-969
SL, Lundqvist G, Rehfeld JF, Schwartz TW (1977) Pancre- Percopo V, Angrisani L, Lorenzo M, Angrisani G, Di Prisco B,
atic somatostatinoma. Clinical features and physiological Tesauro B (1990a) Jejunal gastrinoma, an extremely rare
implications. Lancet 26: 666-668 location. Second world week of professional updating in
McGuigan JE, Wolfe MM (1980) Secretin injection test in the surgery and in surgical and in oncological disciplines of
diagnosis of gastrinoma. Gastroenterology 79:l324-l331 the University of Milan, Abstract Book. M Montorsi and
Merrel RC (1988) Insulinoma. First world post graduate sur- P Granelli, p 119
gical week of the University of Milan, Lecture Book III. M Percopo V, Negro G, Taddeo F, Cobellis L, Maglio MND, Sor-
Montorsi and P Granelli, pp 1282-1283 rentino L (1990b) Biologically active insulinoma: current
Metz DC, Jensen RT (1995) Advances in gastric antisecretory therapeutic approach. Second world week of professional
therapy in Zollinger Ellison syndrome. Front Gastrointest updating in surgery and in surgical and in oncological
Res 23: 240-257 disciplines of the University of Milan, Lecture Book III. M
Miller DL (1993) Endocrine angiography and venous sam- Montorsi and F Zennaro, pp l335-l337
pling. Radiol Clin North Am 31:1051-1067 Phan GQ, Yeo CJ, Hruban RH, Lillemoe KD, Pitt HA, Cameron
Moertel CG, Lefkopoulo M, Lipsitz S, Hahn RG, Klaassen D JL (1998) Surgical experience with pancreatic and peripan-
(1992) Streptozocin-doxorubicin, streptozocin-fluorou- creatic neuroendocrine tumors: review of 125 patients. J
racil or chlorozotocin in the treatment of advanced islet- Gastrointest Surg 2:472-482
cell carcinoma. N Engl J Med 326:519-523 Pipeleers-Marichal M, Donow C, Heitz PU Kloppel G (1993)
Mozell E, Stenzel P, Woltering E (1990) Functional endocrine Pathologic aspects of gastrinomas in patients with
tumors of the pancreas: clinical presentation, diagnosis, Zollinger-Ellison syndrome with and without multiple
and treatment. Curr Prob Surg 27:301-386 endocrine neoplasia type I. World J Surg 17:481-488
Negro G, Percopo V (1988) Somatostatinomas. Fist world post Prinz RA, Dorsch TR, Lawrence AM (1981) Clinical aspects of
graduate surgical week of the University of Milan, Lecture glucagon-producing islet cell tumors. Am J Gastroenterol
book III. M Montorsi and P Granelli, pp 1285-88 76:125-l31
Nicholls A (1902) Simple adenoma of the pancreas arising Rothmund M (1994) Localization of endocrine pancreatic
from an island of Langherans. J Med Res 3:985 tumours. Br J Surg 81:164-166
Norton JA (1994) Neuroendocrine tumors of the pancreas and Santini D, Pasquinelli G, D' Alessandro L, Mazzoleni G,
duodenum. Curr Prob Surg 31:77-156 Taffurelli M, Campione 0, Marrano D, Martinelli GN (1991)
Norton JA (1995) Surgical treatment of islet cell tumors with spe- Parathyroid gastrin and parathormone-producing tumour
cial emphasis on Operative Ultrasound. Endocrine tumors of in the Zollinger-Ellison syndrome of MEN 1 origin. Vir-
the pancreas: recent advances in research and management. chows Arch A Pathol Anat HistopathoI419:433-437
M Mignon and RT Jensen. Karger, Basel, 23:333-41 Seregni E, Ferrari L, Stivanello M, Dogliotti L (2000) Labo-
Norton JA (1999) Intraoperative methods to stage and localize ratory tests for neuroendocrine tumours. Q J Nucl Med
pancreatic and duodenal tumors. Ann Oncol10 [SuppI4J: 44:22-41
182-184 Siller GM, Strutton GM, Moore GA, Kanowski DM, Nedwich
Norton JA, Jensen RT (1991) Unresolved surgical issues in the JA (1994) Glucagonoma syndrome with increased lactate
management of patients with Zollinger-Ellison syndrome. dehydrogenase isoenzymes: octreotide treatment. Aust J
World J Surg 15:151-159 DermatoI35:11-14
Norton JA, Kahn CR, Schiebinger R, Gorschboth C, Brennan Solcia E, Fiocca R, Rindi G, Villani L, Cornaggia M, Capella
MF (1979) Amino acid deficiency and the skin rash asso- C (1993) The pathology of the gastrointestinal endocrine
ciated with glucagonoma. Ann Intern Med 91:2l3-215 system. Endocrinol Metab Clin North Am 22:795-821
Norton JA, Cromack DT, Shawker TH, Doppman JL, Comi Solcia E, Kloppel G, Sobin LH (2000) Histological typing of
R, Gorden P, Maton PN, Gardner JD, Jensen RT (1988) the endocrine tumours. Springer, Berlin Heidelberg New
Intraoperative ultrasonographic localization of islet cell York
tumors. A prospective comparison to palpation. Ann Surg Stadil F (1995) Treatment of gastrinomas with pancreatico-
207:160-168 duodenectomy. In: Mignon M, Jensen RT (eds) Endocrine
Norton JA, Doppman JL, Jensen RT (1992) Curative resection tumors of the pancreas: recent advances in research and
in Zollinger-Ellison syndrome: results of a 10-year pro- management. Karger, Basel 23:333-341
spective study. Ann Surg 215:8-18 Stefanini P, Carboni M, Patrassi N, Basoli A (1974) Beta-islet
Norton JA, Fraker DL,Alexander HR, Venzon DJ, Doppman JL, cell tumors of the pancreas: results of a study on 1,067
Serrano J, Goebel SU, Peghini PL, Roy PK, Gibril F, Jensen cases. Surgery 75:597-609
RT (1999) Surgery to cure the Zollinger-Ellison syndrome Stinner B, Rothmund M (1992) Actual surgical strategies for
(see comments). N Engl J Med 341:635-644 endocrine tumors. Onkologie 15:346-354
Oberg K (1996) Neuroendocrine gastrointestinal tumours. Strohm WD (1996) Paraneoplastic spastic tetraparesis in glu-
Ann Oncol 7:453-463 cagonoma syndrome. Successful therapy with octreotide,
152 F. Cirillo et al.

dacarbazine and interferon-alpha. Z Gastroenterol 34: Wheeler M, Curley IR, Williams ED (1986) The association
438-445 of neurofibromatosis, pheocromocytoma and soma-
Thodiyil PA, El-Masry NS, Williamson RC (2000) Achieving tostatin-rich duodenal carcinoid tumor. Surgery 12:
eugastrinemia in Zollinger-Ellison Syndrome: resection or 1163-1168
enucleation? Dig Surg 18:118-123 Whipple A, Frantz VK (1935) Adenoma of the islet cell with
Udelsman R, Yeo CJ, Hruban RH, Pitt HA, Niederhuber JE, hyperinsulinemia. Ann Surg 101:1299-1305
Coleman J, Cameron JL (1993) Pancreaticoduodenectomy Wiedenmann B, Jensen RT, Mignon M, Modlin CI, Skogseid
for selected pancreatic endocrine tumors. Surg Gynecol B, Doherty G, Oberg K (1998) Preoperative diagnosis and
Obstet 177:269-278 surgical management of neuroendocrine gastroenteropan-
Verner JV, Morrison AB (1958) Islet-cell tumor and a syn- creatic tumors: general recommendations by a consensus
drome of refractory watery diarrhea,hypokalemia and workshop. World J Surg 22:309-318
hypochlorhydria. Clin Gastroenterol 3:395-408 Wilkinson DS (1973) Necrolytic migratory erythema with
Verner JV, Morrison AB (1974) Endocrine pancreatic islet dis- carcinoma of the pancreas. Trans St Johns Hosp Dermatol
ease with diarrhea. Report of a case due to diffuse hyper- Soc 59:244-250
plasia of nonbeta islet tissue with a review of 54 additional Zayene A, Cadiot G, Vaugnat A, Bonnaud G, Mignon M (1997)
cases. Arch Intern Med 133:492-499 Survival factors in patients with liver metastases. Gastro-
Verschoor L, Uitterlinden P, Lamberts SWJ, Del Pozo E (1986) enterology 112:A496
On the use of a new somatostatin analogue in the treat- Zeiger MA, Shawker TH, Norton JA (1993) Use of intra-
ment of hypoglycaemia in patients with insulinoma. Clin operative ultrasonography to localize islet cell tumors.
Endocrinol (Oxf) 25:555-560 World J Surg 17:448-454
Vinik AI, Delbridge L, Moattari R, Cho K, Thompson N (1991) Zollinger RM, Ellison EC, Fabri PJ, Johnson J, Sparks J, Carey
Transhepatic portal vein catheterization for localization of LC. (1980) Primary peptic ulcerations of the jejunum asso-
insulinomas: a ten-year experience. Surgery 109:1-11; ciated with islet cell tumors. Twenty-five-year appraisal.
discussion III Ann Surg 192:422-430
9 Clinical Manifestations and Therapeutic Management
of Nonfunctioning Endocrine Tumors
M. FALCONI, R. BETTINI, C. BASSI, R. SALVIA, and P. PEDERZOLI

CONTENTS use of diagnostic testing, and improved characteriza-


tion of pancreatic neoplasms by pathologists rather
9.1 Introduction 153 than to a real epidemiological increase (YEO et al.
9.2 Clinical Data 153 1993; SALEH et al. 1996; DI STASI et al. 1998). The
9.2.1 Clinical History, Habits and Heredity 153
9.2.2 Symptoms 154 cyto-/histological definition of the endocrine origin
9.3 Laboratory 154 of these tumors is indispensable for establishing the
9.4 Clinical Data 155 correct therapy and the prognosis, which are very
9.5 From Assessment to Management 155 different from those of the more common ductal
9.5.1 Diagnostic Suspicion 155 adenocarcinoma.
9.5.2 Definite Diagnosis 156
9.5.2.1 Surgically Treatable Patients 156
9.5.2.2 Patients Suitable for Debulking
and Multidisciplinary Approach 157
9.5.2.3 Nonresectable Patients 9.2
and Nonsurgical Therapies 157 Clinical Data
9.6 Follow-up 158
References 158
9.2.1
Clinical History, Habits and Heredity

These neoplasms are usually diagnosed in patients


9.1 in their 50s and 60s with a ratio of 1:1 for men
Introduction and women (THOMPSON 1988; EVANS et al. 1993;
MADURA et al. 1997; PHAN et al. 1998; MATTHEWS et
Nonfunctioning endocrine tumors arise from pan- al. 2000). The neoplasm can be idiopathic or detected
creatic insular cells. Sharing the same histological as part of the multiple endocrine neoplasia (MEN 1)
and immunohistochemical features as functioning syndrome or, more rarely, of the von Hippel-Lindau
endocrine tumors, they are characterized by the syndrome. MEN 1 syndrome is an autosomal domi-
lack of clinical symptoms due to hormonal hyper- nant disease with a variable degree of penetration in
production (AKERSTROM 1992; STINNER et al. 1992; which tumors can develop in different organs. Almost
OBERG 1996; WIEDENMANN et al. 1998; AZIMUDDIN all patients have parathyroid hyperplasia. The diag-
and CHAMBERLAIN 2001; RIGAUD et al. 2001). nosis of primary hyperparathyroidism often leads to,
In the different reported series, their percentage or precedes, the diagnosis of other neoplasms. Fewer
varies from 15% to 52% of pancreatic endocrine than 50% of patients also develop pituitary tumors
lesions (PHAN et al. 1998). These tumors have been
increasingly observed (Fig. 9.1) over the last few
years (THOMPSON 1988; YEO et al. 1993; FALCONI et
al. 1999; FALCONI 2000). This finding seems to be
related more to greater clinical awareness, increasing

M. FALCONI, MDj R. BETTINI, MDj C. BASSI,MDj R. SALVIA,MDj 84--'85 86-'87 88-'89 90-'91 92-'93 94-'95 96-'97 98-'99 00-'01

P. PEDERZOLI, MD
Surgical-Gastroenterological Department, Endocrine and Fig. 9.1. Yearly distribution of functioning and nonfunctioning
Pancreatic Unit, Hospital 'G.B. Rossi', University of Verona, pancreatic endocrine tumors observed between 1984 and 2001
37134 Verona, Italy in personal experience
154 M. Falconi et al.

(usually prolactinomas), while about 75% develop 60% of patients, followed by nausea, vomiting, and
concurrent islet-cell tumors of different types. Non- jaundice. A greater frequency of cephalic tumors
functioning tumors represent only 20% of these (Table 9.1) is reported in the literature (DIAL et
(NORTON 1994). In patients with MEN 1 syndrome, al. 1985; ECKHAUSER et al. 1986). Pain, weight loss,
the pancreatic tumors are recognized at an average symptoms of exocrine and/or endocrine pancreatic
age of 25 years (WIEDENMANN et al. 1998). insufficiency (steatorrhea and diabetes mellitus), and
As far as the influence of nonessential factors such asthenia are often expressions of advanced disease.
as alcohol or coffee abuse and cigarette smoking is Other rarely reported symptoms are those due to
concerned, there is no evidence of any correlation recurrent pancreatitis or gastrointestinal bleeding.
between them and the occurrence of these neo- In endocrine tumors, there is a discrepancy
plasms. between the extent of the disease and the usually
limited symptoms. This is probably due to their low
biological aggressiveness and slow growth. It should
9.2.2 be noted that the symptom complex is virtually iden-
Symptoms tical to that of ductal adenocarcinoma.
Table 9.2 summarizes the symptoms and their fre-
By definition, there are no clinical symptoms related quency in the main series reported in the literature.
to hormonal hyperproduction in nonfunctioning
endocrine lesions. In 10%-15% of patients, the
diagnosis is made following characterization of an
incidentally detected pancreatic mass (KENT et al. 9.3
1981). In these cases, imaging is usually carried out Laboratory
for nonspecific dyspeptic symptoms or the presence
of a palpable mass. Alternatively, the patient may be Traditional routine laboratory analysis is useful when
referred to the radiologist for symptoms related to the tumor has caused an obstruction at the biliary
infiltration or local compression of adjacent struc- duct level or when there is a massive replacement
tures by the tumor or its metastases. Pain is, in fact, of hepatic parenchyma with subsequent cholestasis
the most common symptom reported, occurring in and/or tissue necrosis.

Table 9.1. Location of nonfunctioning endocrine tumors in the different series (NA, not available)

Location Personal BARTSCH PHAN Lo EVANS THOMPSON KENT


experience (2000) et al. (2000) et al. (1998) et al. (1996) et al. (1993) (1988) et al. (1981)
(n=115) (n=18) (n=58) (n=34) (n=73) (n=27) (n=25)

Head/neck/uncus 56% 61 % 50% 47% 59% Predominantly 56%


Body/tail 44% 39% 17% 47% 41 % NA 12%
Multicentric NA 19% 6% 0 NA 12%

Table 9.2. Clinical presentation in the different series (NA, not available)

Complaints/Symptoms Personal MATTHEWS BARTSCH KENT MADURA PHAN


experience (2000) (2000) (2000) (1981) (1997) (1988)
(n=115) (n=28) (n=18) (n=25) (n=14) (n=58)

Incidental diagnosis 22.4% NA 11% 16% NA NA


Abdominal pain 60.3% 67.9% 72% 36% 57% 56%
Nausea/vomiting 10.3% 35.7% 11% NA 43% 21%
Weight loss 31% 32.1 % 39% NA 50% 46%
Anorexia 16.3% 21.4% 33% NA NA 14%
Jaundice 11.2% 21.4% 22% 28% 50% 35%
Malaise NA 14.3% NA NA 21% 7%
Pancreatitis NA 7.1 % 11% NA NA NA
Abdominal mass 6.9% 7.1% 17% 2% NA NA
Gastrointestinal 4.3% 3.6% 5.5% NA NA NA
bleeding/acute abdomen
New onset diabetes NA NA NA NA 14% NA
Clinical Manifestations and Therapeutic Management of Nonfunctioning Endocrine Tumors 155

Nonfunctioning endocrine tumors are frequently 9.4


associated with elevated chromogranin A values. This Clinical Data
is a glycoprotein contained in intracellular granules of
the insular cells. Chromogranin A is, in fact, the most Generally speaking, once a pancreatic mass has been
sensitive of the markers for nonfunctioning endocrine identified, the suspicion of a nonfunctioning endo-
tumors (68%) and simultaneously shows good speci- crine neoplasm arises when the biological behavior
ficity (86%) (FERRARI et al. 1999). However, in 19% of the tumor is 'non aggressive'. Despite the fact that
of nonselected patients, the chromogranin A value is a nonfunctioning endocrine tumor is diagnosed in
high even with no endocrine tumor. The correlation a high percentage of patients in the advanced stages
between the marker value and the size of the tumor of the disease (Table 9.3), the patient rarely shows
is not certain (SCHURMANN et al. 1992; BAUDIN et al. significant symptoms. The absence of a hormonal
1998; FERRARI et al. 1999; GOEBEL et al. 1999). Lastly, clinical syndrome allows these tumors to grow unde-
the marker can be useful in identifying recurrence and tected, and therefore they are larger and present with
as a marker of successful therapy, although the value a greater frequency of hepatic metastases than the
is limited in patients being treated with somatostatin functioning types. In personal experience, the per-
analogues (STIVANELLO et al. 2001). centage of hepatic metastases at diagnosis was 33%
The neuronspecific enolase (NSE) has no sig- compared with 16% for functioning types. Moreover,
nificant role at present in defining endocrine tumors. on average, the dimension of the primary neoplasm
Although it shows an analogous specificity to chro- at diagnosis is 32 mm (Table 9.3) compared with 15
mogranin A, the sensitivity is inferior (33%) (FER- mm for functioning ones. Despite the larger tumor
RARI et al. 1999). Nevertheless, it would seem that the size and aggressive appearance, the patients rarely
high levels of this marker are associated with a poor appear to be suffering from metastatic cancer.
cellular differentiation (BAUDIN et al. 1998).
Pancreatic polypeptide (PP) is elevated in about
60% of pancreatic endocrine tumors. Fractions of
beta- and alpha-subunits of human chorionic gonad- 9.5
otropin (HCG) may also be useful, as they seem to From Assessment to Management
be correlated to a worse prognosis, even though only
20%-30% of patients show an increase of the serum 9.S.1
levels (ERIKSSON et al. 1990a, 2000). Diagnostic Suspicion
Some hormones, characteristic of the functional
types, can also have higher values in the blood and Nowadays, cyto/histological confirmation of the clin-
be used as markers, even if they do not produce a ical suspicion of nonfunctioning endocrine tumor is
clinical syndrome. possible with fine needle aspiration biopsy of both
Furthermore, in patients suffering from MEN 1 the primary pancreatic mass and hepatic metastases
syndrome, some authors have suggested the use of (SALEH et al. 1996; DI STASI et al. 1998). This proce-
the 'meal stimulatory test' for PP as a method of early dure does not predict tumor resectability. The cyto-
diagnosis (CIRILLO 2001; LANGER et al. 2001). logical and/or histological confirmation, however, is

Table 9.3. Biological behavior and main surgical and pathological features in the different series (NA, not available)
Personal BARTSCH PHAN Lo EVANS THOMPSON KENT
experience (2000) et al. (2000) et al. (1998) et al. (1996) et al. (1993) (1988) et al. (1981)
(n=1l5) (n=18) (n=58) (n=34) (n=73) (n=27) (n=25)

Diameter of primary tumor:


Median (cm) 3.2 5.0 4.0 NA NA NA NA
Range (cm) 0.5-14 3-10 0.5-5
Malignant at diagnosis 69% 83% 60% 84%" 60% NA NA
Resectable 62%b 83% 79.3% 53% 43.8% 51% 60%
Curative resection 50.4%b 62% NA 35% 26% 37% 48%
Operative mortality 0.8% 16.6% NA 2% 0 3% 8%
" Distant metastases: 63%; locoregional metastases: 34%
b All patients included
156 M. Falconi et al.

mandatory for the future therapeutic strategy when is less common compared with ductal carcinoma
the disease is either locally advanced or metastatic. (EELKEMA et al. 1984; MADURA et al. 1997) and, in
In fact, the diagnosis of pancreatic endocrine tumor any case, does not exclude the possibility of radical
gives the clinician a choice of therapeutic options. resection. The choice of procedure will depend on
Surgical resection is still justifiable for advanced each individual case.
forms, and if this cannot be performed, palliative The possible surgical options are: limited resec-
surgery can be carried out. Furthermore, in cases of tion (enucleation or middle pancreatectomy) and
nonresectability, evaluation of the histological differ- conventional resection (pancreoduodenectomy or
entiation and the Ki-67 cellular mitotic index leads distal pancreatectomy with or without spleen preser-
to the choice of therapy. vation). In general, limited resection has the advan-
tage of preserving the largest possible part of the
pancreatic parenchyma, but it is less 'sure' in onco-
9.5.2 logical terms. This can usually be performed in those
Definite Diagnosis cases where the lesions are single, well-localized, of
limited size «4 em in diameter), and do not involve
The therapeutic approach for a patient with a non- or are at a 'sufficient' distance from the pancreatic
functioning pancreatic endocrine tumor is often duct (PARK et al. 1998). Nevertheless, conventional
multidisciplinary. Nevertheless, surgery, when pos- resection, depending on the site, appears to be the
sible, is the only possibility for cure and is, even today, most standard procedure (DELCORE and FRIESEN
the first therapeutic option to consider (OBERG 1996). 1992; UDELSMAN et al. 1993; NORTON 1994; PHAN et
From a management point of view, then, it would al. 1998; JORDAN 1999).
seem useful to distinguish those patients eligible for The long-term results vary in accordance with
surgical resection from those who are not. the histological grade of the lesion as determined
by microscopic pathology. Table 9.4 summarizes
9.5.2.1 the survival percentage of the main surgical series
Surgically Treatable Patients published in the literature. Generally, for endocrine
tumors presently defined as benign by the World
Compared with ductal carcinomas, these neoplasms Health Organization (WHO) 2000 classification
have a very high rate of resectability. This varies, in (SOLCIA 2000), the survival rate after radical surgery
the different series, from 26% to 62% (KENT et al. is 100%. This rate decreases in patients resected for
1981; DIAL et al. 1985; THOMPSON 1988; BARTSCH et endocrine carcinoma (PHAN et al. 1998). During
al. 2000). Such a high resectability rate does not mean, follow-up, 66% of patients show recurrence of the
however, that surgical resection always corresponds disease, usually within the liver (CHAMBERLAIN et al.
to radical surgery, whose incidence is, in fact, lower 2000; PEDERZOLI et al. 1999).
(Table 9.3). As for the synchronous or metachronous metasta-
In the presence of nonfunctioning endocrine ses, whenever a resection must leave no residual dis-
tumor, the surgeon must try to completely resect all ease, hepatic resection should be performed (DAN-
of the visible tumor mass along with total debulking FORTH et al. 1984; NORTON et al. 1986; McENTEE et
of all visible intra- and extrapancreatic disease. al. 1990; CARTY et al. 1992). Contemporary resection
Surgical resection is indicated in all cases where of the primary tumor and all the hepatic metastases
the disease appears to be 'limited' to the pancreas. or the removal of subsequent ones does not, in fact,
However, some have shown that a survival benefit seem to be an unfavorable prognostic factor (CHEN et
can be gained by radical resection of the tumor and al. 1998; CHAMBERLAIN et al. 2000).
surrounding structures (stomach, colon, kidney, Unfortunately, in 90% of patients, the liver metas-
adrenal gland) or by further vascular resection (DIAL tases are multifocal or bilateral (MATTHEWS et al.
et al. 1985; DELCORE and FRIESEN 1994; MADURA 2000), making radical surgery impossible. In fact,
et al. 1997; BARTSCH et al. 2000; MATTHEWS et al. fewer than 18% of patients have a localized disease
2000). In 1985, DIAL did not exclude resection of the (unilobar or less than 75% involvement of the hepatic
involved portal vein (DIAL et al. 1985). More recently, parenchyma) which can be considered for resection
EVANS suggested the possible segmental resection with intent of cure (CHAMBERLAIN et al. 2000).
of the portal-mesenteric vein confluence (EVANS et This approach leads to good results, with the pres-
al. 1993). Involvement of the superior mesenteric ent survival rate of 76% at 5 years (CHEN et al. 1998).
artery and the celiac trunk by endocrine tumors The survival rate seems to be directly related to the
Clinical Manifestations and Therapeutic Management of Nonfunctioning Endocrine Tumors 157

Table 9.4. The 2-, 5-, and 10-year survival rates for different surgical series (NA, not available)
Survival Personal BARTSCH PHAN EVANS THOMPSON KENT
experience (2000) et al. (2000) et al. (1998) et al (1993) (1988) et al. (1981)
(n=1l5) (n=18) (n=58) (n=73) (n=27) (n=25)

2-year 85% NA 79% NA 58% at 3 years 60% at 3 years


5-year 70% 65% 52% 50% NA 44%
lO-year 45% 49% 42% NA NA NA

level of hepatic involvement (SOREIDE et al. 1992; pression of adjacent viscera. The disease can be
QUE et al. 1995; CHEN et al. 1998; FUJII et al. 1999). 'restricted' to the liver, thus facilitating subsequent
appropriate therapies.
9.5.2.2 The palliative resection of the residual hepatic
Patients Suitable for Debulking metastases after radical resection of the primary
and Multidisciplinary Approach tumor has limited applications, as it does not lead to
a significant increase in the survival rate (FALCONI et
In some clinical situations, the information gained al. 1999; CHAMBERLAIN et al. 2000).
from preoperative examinations or during a lapa- In nonfunctioning lesions, where less than 90% of
rotomy does not give a reasonably sure prediction of the tumor is resectable and there are no symptoms of
achieving total surgical removal of all sites of disease. locoregional compression, catheter embolization is
In these cases, resection would leave a macroscopic the best treatment (CHAMBERLAIN et al. 2000).
residual disease (R2) due to the presence of'signifi- As an alternative to traditional surgical tech-
cant and massive' local infiltration (vessels, organs, niques for the treatment of residual hepatic dis-
retroperitoneum) and/or not completely resectable ease, embolization and chemo-embolization may
hepatic metastases. be proposed (MARLINK et al. 1990; NESOVIC et al.
In this case, many surgeons have suggested deb- 1992; MAVLIGIT et al. 1993; RUSZNIEWSKI et al. 1993;
ulking with follow-up treatment of residual disease CLOUSE et al. 1994; ERIKSSON et al. 1998; FALCONI
with locoregional therapies. It should be remem- et al. 1999). Ligation of the hepatic artery alone is
bered that, to be therapeutically successful, the deb- not enough because a collateral circulation quickly
ulking must reduce the mass by at least 90%, which appears (WIEDENMANN et al. 1998). At present, the
is seldom possible. Debulking has further been sug- first results with dia-thermo-ablation are beginning
gested as a preventive method against complications to emerge (OBERG 1999,2000). The experience with
related to local growth of the tumor, such as recur- this technique and its practice is currently limited to
rent pancreatitis, biliary or intestinal occlusion, and a few centers.
gastrointestinal bleeding. Indications for hepatic transplant are also experi-
In truth, surgical resection does not appear justi- mental. The criteria for patient selection include:
fiable in all cases of locally incompletely resectable patients under 60 years old, nonresectable hepatic
disease. In fact, existing data do not justify a local metastases, no extrahepatic abdominal tumor, and
partial resection of the mass, fearing fragmentation failure of conventional therapies. Rapid growth of
with subsequent dissemination of the tumor in the the tumor as an indication for transplant is con-
peritoneal cavity. Moreover, the vascularity of these troversial (LANG et al. 1997; FRILLING et al. 1998).
neoplasms introduces a high risk of bleeding, and Data on patients selected according to the above
palliation of the symptoms secondary to the mass indications show a global survival rate at 5 years of
is only temporary, since local relapse is the rule. 36%, with 17% of survivors completely disease-free.
Furthermore, the survival rate of unresected patients Unfortunately, the postoperative mortality is 30% (LE
with locally advanced disease remains at 44% at 5 TREUT et al. 1997).
years (EVANS et al. 1993).
Debulking can, however, play a role if intended 9.5.2.3
as a total resection of the primary tumor, even in Nonresectable Patients and Nonsurgical Therapies
the presence of residual hepatic metastatic disease
(PEDERZOLI et al. 1999). Even when the metastatic Those patients unsuitable for surgical resection and
residue is more than 10%, resection of the primary with symptoms of compression of ducts and/or vis-
tumor will help avoid symptoms related to com- cera related to the mass are candidates for palliative
158 M. Falconi et al.

surgical therapy. Because these patients live longer slow, and the therapeutic programs adopted are quite
than those with adenocarcinoma of the pancreas, different. In general, the follow- up intervals should be
surgical bypass is preferred. Biliary enteric bypass closer during the initial phase following diagnosis,
can be combined with gastric drainage procedures when therapeutic variations occur, or whenever the
to prevent duodenal obstruction (SPEER et al. 1987; disease shows progression.
ANDERSEN et al. 1989; WELVAART 1992). Although The following outline seems to be rational and
endoscopic palliative treatment is available, the common, mainly based on clinical experience.
longer life expectancy results in multiple procedures - Patients treated with radical surgery but with
to replace the endoprostheses (DELCORE and FRI- unfavorable histological and prognostic factors
ESEN 1994; PEDERZOLI et al. 1999). cannot be considered cured of disease and should
Medical therapy is indicated for those patients be followed up every 4-6 months. On the basis
suffering from nonresectable, nondebulkable dis- of the WHO 2000 classification (SOLCIA 2000),
ease. Somatostatin analogues (octreotide, lanreotide) negative prognostic factors include: presence of
associated or unassociated with interferon are first a mass larger than 2 cm, a mitotic index greater
line agents. The presence of somatostatin receptors, than 2%, and/or the presence of vascular or
imaged with indium-lll-pentrotide (octreoscan) microscopic neural invasion.
scintigraphy, correlates with response to treatment. - Patients with favorable prognostic factors should
Treatment with somatostatin analogues leads to con- be followed up every 12 months.
trol of the tumor in at least 20% of patients suffering - Patients with metastatic endocrine tumor should
from metastatic gastro-entero-pancreatic neuroen- be followed up every 4 months in order to detect
docrine tumor (ARNOLD et al. 1996). The antitumor any eventual progression of the disease.
activity can be increased by association with alpha- - Patients with residual disease after surgery should
interferon, which inhibits the transitory growth of be followed up at the same regular intervals.
the tumor by 67% (FRANK et al. 1999).
Chemotherapy is reserved for cases of progressive With further experience accrued over the years,
disease, when the tumor is anaplastic. The optimal these outlines will be re-evaluated. In light of the
regimen employs streptozocin (STZ) associated with previously recently revised histopathological classi-
either 5-fluorouracil (5-FU) or doxorubicin (MOER- fication, results from many series will become easier
TEL et al. 1980,1982). The rate of objective response is to interpret and compare.
around 50% for both combinations, and the response
duration is about 2 years (ERIKSSON et al. 1990b;
ERIKSSON and OBERG 1993). For those patients with
rapid growth anaplastic forms, treatment with cispla- References
tin (CDDP) + etoposide (VPI6) seems to give better
results at the moment (67% of objective response; Akerstrom G (1992) Surgical treatment of endocrine pancre-
atic tumors. Diagn Oncol 2:332-337
MOERTEL et al. 1991). Andersen JR, Sorensen SM, Kruse A, Rokkjaer M, Matzen
Although gastrointestinal endocrine tumors P (1989) Randomised trial of endoscopic endoprothesis
have classically been considered as radioresistant, versus operative bypass in malignant obstructive jaundice.
radiometabolic therapy-based trials with iodine- Gut 30:1132-1135
131-metaiodobenzylguanidine (l3l I-MIBG) or with Arnold R, Trautmann ME, Creutzfeldt W, Benning R, Benning
M, Neuhaus C, Jurgensen R, Stein K, Schafer H, Bruns C,
yttrium-90-1,4,7,10-tetra-azacyclododecane tetra- Dennler HJ (1996) Somatostatin analogue octreotide and
acetic acid eOY-DOTA) are underway at present. The inhibition of tumor growth in metastatic endocrine gastro-
results, however, are preliminary. enteropancreatic tumours. Gut 38:430-438
Azimuddin K, Chamberlain RS (2001) The surgical manage-
ment of pancreatic neuroendocrine tumors. Surg Clin North
Am 81:511-525
Bartsch DK, Schilling T, Ramaswamy A, Gerdes B, Celik I,
9.6 Wagner HI, Simon B, Rothmund M (2000) Management
Follow-up of nonfunctioning islet cell carcinomas. World J Surg 24:
1418-1424
Follow-up monitoring of the success of surgical or Baudin E, Gigliotti A, Ducreux M, Ropers J, Comoy E, Sab-
ourin JC, Bidart JM, Cailleux AF, Bonacci R, Ruffie P,
medical therapy is based on clinical, laboratory, and Schlumberger M (1998) Neuron-specific enolase and chro-
imaging examinations. Appropriate follow-up inter- mogranin A as markers of neuroendocrine tumours. Br J
vals are not standardized; the tumor growth is usually Cancer 78:1102-1107
Clinical Manifestations and Therapeutic Management of Nonfunctioning Endocrine Tumors 159

Carty SE, Jensen RT, Norton JA (1992) Prospective study of Bassi C. (eds) Pancreatic Tumors. Achievement and pro-
aggressive resection of metastatic pancreatic endocrine spective. Thieme, Stuttgart, pp 368_397
tumors. Surgery 112:1024-1031; discussion 1031-1032 Falconi M, Bassi C, Bonora A, Sartori N, Procacci C, Talamini
Chamberlain RS, Canes D, Brown KT, Saltz L, Jarnagin W, G, Mansueto GC, Pederzoli P (l999) Role of chemoembo-
Fong Y, Blumgart LH (2000) Hepatic neuroendocrine lization in synchronous liver metastases from pancreatic
metastases: does intervention alter outcomes? J Am Coli endocrine tumors. Dig Surg 16:32-38
Surg 190:432-445 Ferrari L, Seregni E, Bajetta E, Martinetti A, Bombardieri E
Chen H, Hardacre JM, Uzar A, Cameron JL, Choti MA (1998) (l999) The biological characteristics of chromogranin A
Isolated liver metastases from neuroendocrine tumors: and its role as a circulating marker in neuroendocrine
does resection prolong survival? J Am Coli Surg 187:88-92; tumors. Anticancer Res 19:3415-3427
discussion 92-93 Frank M, Klose KJ, Wied M, Ishaque N, Schade-Brittinger C,
Cirillo F (2001) Clinica e fisiopatologia dei tumori neuroendo- Arnold R (1999) Combination therapy with octreotide and
crini gastroentero-pancreatici. Casa Editrice Ambrosiana, alpha-interferon: effect on tumor growth in metastatic
Milano endocrine gastroenteropancreatic tumors. Am J Gastro-
Clouse ME, Perry LJ, Stuart K, Stokes KR (1994) Hepatic arte- enteroI94:1381-1387
rial chemoembolization for metastatic neuroendocrine Frilling A, Rogiers X, Malago M, Liedke 0, Kaun M, Broelsch
tumors. Digestion 55 [Suppl 3]:92-97 CE (l998) Liver transplantation in patients with liver
Danforth DN Jr, Gorden P, Brennan MF (1984) Metastatic insu- metastases of neuroendocrine tumors. Transplant Proc
lin-secreting carcinoma of the pancreas: clinical course 30:3298-3300
and the role of surgery. Surgery 96:1027-1037 Fujii K, Yamamoto J, Shimada K, Kosuge T, Yamasaki S, Kanai
Delcore R, Friesen SR (1992) Role of pancreatoduodenectomy Y (1999) Resection of liver metastases after pancreatoduo-
in the management of primary duodenal wall gastrinomas denectomy: report of seven cases. Hepatogastroenterology
in patients with Zollinger-Ellison syndrome. Surgery 112: 46:2429-2433
1016-1022; discussion 1022-1023 Goebel SU, Serrano J, Yu F, Gibril F, Venzon DJ, Jensen RT (1999)
Delcore R, Friesen SR (1994) Gastrointestinal neuroendocrine Prospective study of the value of serum chromogranin A or
tumors. J Am Coli Surg 178:187-211 serum gastrin levels in the assessment of the presence,
Di Stasi M, Lencioni R, Solmi L, Magnolfi F, Caturelli E, De Sio I, extent, or growth of gastrinomas. Cancer 85:1470-1483
Salmi A, Buscarini L (l998) Ultrasound-guided fine needle Jordan PH Jr (1999) A personal experience with pancreatic
biopsy of pancreatic masses: results of a multi-center study. and duodenal neuroendocrine tumors. J Am Coli Surg
Am J GastroenteroI93:1329-1333 189:470-482
Dial PF, Braasch JW, Rossi RL, Lee AK, Jin GL (1985) Manage- Kent RB III, van Heerden JA, Weiland LH (1981) Nonfunction-
ment of nonfunctioning islet cell tumors of the pancreas. ing islet cell tumors. Ann Surg 193:185-190
Surg Clin North Am 65:291-299 Lang H, von Woellwarth J, Oldhafer KJ, Behrend M, Schlitt HJ,
Eckhauser FE, Cheung PS, Vinik AI, Strodel WE, Lloyd RV, Nashan B, Pichlmayr R (l997) Liver transplantation for
Thompson NW (1986) Nonfunctioning malignant neuroen- metastatic neuroendocrine tumors. Ann Surg 225:347-354
docrine tumors of the pancreas. Surgery 100:978-988 Langer P, Wild A, Celik I, Kopp I, Bergenfelz A, Bartsch DK
Eelkema EA, Stephens DH, Ward EM, Sheedy PF Jr (l984) CT (2001) Prospective controlled trial of a standardized meal
features of nonfunctioning islet cell carcinoma. AJR Am J stimulation test in the detection of pancreaticoduodenal
RoentgenoI143:943-948 endocrine tumors in patients with multiple endocrine
Eriksson B, Oberg K (l993) An update of the medical treat- neoplasia type 1. Br J Surg 88:1403-1407
ment of malignant endocrine pancreatic tumors. Acta Le Treut YP, Delpero JR, Dousset B, Cherqui D, Segol P, Man-
Oncol 32:203-208 tion G, Hannoun L, Benhamou G, Launois B, Boillot 0,
Eriksson B, Arnberg H, Lindgren PG, Lorelius LE, Magnusson Domergue J, Bismuth H (1997) Results of liver trans-
A, Lundqvist G, Skogseid B, Wide L, Wilander E, Oberg K plantation in the treatment of metastatic neuroendocrine
(1990a) Neuroendocrine pancreatic tumors: clinical pre- tumors. A 31-case French multicentric report. Ann Surg
sentation, biochemical and histopathological findings in 84 225:355-364
patients. J Intern Med 228:103-113 Lo CY, Heerden JA van, Thompson GB, Grant CS, Soreide JA,
Eriksson B, Skogseid B, Lundqvist G, Wide L, Wilander E, Harmsen WS (1996) Islet cell carcinoma of the pancreas.
Oberg K (1990b) Medical treatment and long-term sur- World J Surg 20: 878-883
vival in a prospective study of 84 patients with endocrine Madura JA, Cummings OW, Wiebke EA, Broadie TA, Goulet RL
pancreatic tumors. Cancer 65:1883-1890 Jr, Howard TJ (1997) Nonfunctioning islet cell tumors of
Eriksson BK, Larsson EG, Skogseid BM, Lofberg AM, Lorelius the pancreas: a difficult diagnosis but one worth the effort.
LE, Oberg KE (1998) Liver embolizations of patients with Am Surg 63:573-577; discussion 577-578
malignant neuroendocrine gastrointestinal tumors. Cancer Marlink RG, Lokich JJ, Robins JR, Clouse ME (1990) Hepatic
83:2293-2301 arterial embolization for metastatic hormone-secret-
Eriksson B, Oberg K, Stridsberg M (2000) Tumor markers in ing tumors. Technique, effectiveness, and complications.
neuroendocrine tumors. Digestion 62 [SuppI1]:33-38 Cancer 65:2227-2232
Evans DB, Skibber JM, Lee JE, Cleary KR, Ajani JA, Gagel RF, Matthews BD, Heniford BT, Reardon PR, Brunicardi FC,
Sellin RV, Fenoglio q, Merrell RC, Hickey RC (l993) Non- Greene FL (2000) Surgical experience with nonfunction-
functioning islet cell carcinoma of the pancreas. Surgery ing neuroendocrine tumors of the pancreas. Am Surg 66:
114:1175-1181; discussion 1181-1182 1116-1122; discussion 1122-1123
Falconi M, Bonora A, Bassi C, Contro C, Marcucci S, Marchiori Mavligit GM, Pollock RE, Evans HL, Wallace S (l993) Durable
L, Talamini G, Scarpa A, Pederzoli P (2000) In: Pederzoli P, hepatic tumor regression after arterial chemoemboliza-
160 M. Falconi et al.

tion-infusion in patients with islet cell carcinoma of the P, Pederzoli P, Scarpa A (2001) High resolution allelotype
pancreas metastatic to the liver. Cancer 72:375-380 of nonfunctional pancreatic endocrine tumors: identifica-
McEntee GP, Nagorney DM, Kvols LK, Moertel CG, Grant CS tion of two molecular subgroups with clinical implications.
(1990) Cytoreductive hepatic surgery for neuroendocrine Cancer Res 61:285-292
tumors. Surgery 108:1091-1096 Ruszniewski P, Rougier P, Roche A, Legmann P, Sibert A,
Moertel CG, Hanley JA, Johnson LA (1980) Streptozocin alone Hochlaf S, Ychou M, Mignon M (1993) Hepatic arterial
compared with streptozocin plus fluorouracil in the treat- chemoembolization in patients with liver metastases of
ment of advanced islet-cell carcinoma. N Engl J Med 303: endocrine tumors. A prospective phase II study in 24
1189-1194 patients. Cancer 71 :2624-2630
Moertel CG, Rubin J, van Hazel GA, Schutt AJ, O'Connell MJ Saleh HA, Masood S, Khatib G (1996) Percutaneous and intra-
(1982) Phase II trial of doxorubicin therapy for advanced operative aspiration biopsy cytology of pancreatic neuro-
islet cell carcinoma. Cancer Treat Rep 66: 1567-1569 endocrine tumors: cytomorphologic study with an immu-
Moertel CG, Kvols LK, O'Connell MJ, Rubin J (1991) Treatment nocytochemical contribution. Acta CytoI40:182-190
of neuroendocrine carcinomas with combined etoposide Schurmann G, Raeth U, Wiedenmann B, Buhr H, Herfarth C
and cisplatin. Evidence of major therapeutic activity in the (1992) Serum chromogranin A in the diagnosis and follow-
anaplastic variants of these neoplasms. Cancer 68:227-232 up of neuroendocrine tumors of the gastroenteropancre-
Nesovic M, Ciric J, Radojkovic S, Zarkovic M, Durovic M atic tract. World J Surg 16:697-701; discussion 701-702
(1992) Improvement of metastatic endocrine tumors of Solcia E (2000) Histological typing of the endocrine tumors.
the pancreas by hepatic artery chemoembolization. J Springer, Berlin Heidelberg New York
Endocrinol Invest 15:543-547 Soreide 0, Berstad T, Bakka A, Schrumpf E, Hanssen LE, Engh
Norton JA (1994) Neuroendocrine tumors of the pancreas and V, Bergan A, Flatmark A (1992) Surgical treatment as a
duodenum. Curr Prob Surg 31:77-156 principle in patients with advanced abdominal carcinoid
Norton JA, Sugarbaker PH, Doppman JL, Wesley RA, Maton tumors. Surgery 111:48-54
PN, Gardner JD, Jensen RT (1986) Aggressive resection Speer AG, Cotton PB, Russell RC, Mason RR, Hatfield AR, Leung
of metastatic disease in selected patients with malignant JW, MacRae KD, Houghton J, Lennon CA (1987) Randomised
gastrinoma. Ann Surg 203:352-359 trial of endoscopic versus percutaneous stent insertion in
Oberg K (1996) Neuroendocrine gastrointestinal tumors. Ann malignant obstructive jaundice. Lancet 2:57-62
Oncol 7:453-463 Stinner B, Beyermann K, Hasselmann U, Rothmund M (1992)
Oberg K (1999) Neuroendocrine gastrointestinal tumors Actual surgical strategies for endocrine tumors. Onkologie
- a condensed overview of diagnosis and treatment. Ann 15:346-354
Oncoll0 [SuppI2]:S3-S8 Stivanello M, Berruti A, Torta M, Termine A, Tampellini M,
Oberg K (2000) State of the art and future prospects in the man- Gorzegno G, Angeli A, Dogliotti L (2001) Circulating chro-
agement of neuroendocrine tumors. Q J Nucl Med 44:3-12 mogranin A in the assessment of patients with neuroendo-
Park BJ, Alexander HR, Libutti SK, Huang J, Royalty D, Skarulis crine tumors. A single institution experience. Ann Oncol12
MC, Jensen RT, Gorden P, Doppman JL, Shawker TH, Fraker [SuppI2]:S73-S77
DL, Norton JA, Bartlett DL (1998) Operative management Thompson GB (1988) Islet cell carcinomas of the pancreas: a
of islet-cell tumors arising in the head of the pancreas. twenty-year experience. Surgery 104:1011-1017
Surgery 124:1056-1061; discussion 1061-1062 Udelsman R, Yeo CJ, Hruban RH, Pitt HA, Niederhuber JE,
Pederzoli P, Falconi M, Bonora A, Salvia R, Sartori N, Contro Coleman J, Cameron JL (1993) Pancreaticoduodenectomy
C, Marcucci S, Bassi C (1999) Cytoreductive surgery in for selected pancreatic endocrine tumors. Surg Gynecol
advanced endocrine tumors of the pancreas. Ital J Gastro- Obstet 177:269-278
enterol Hepatol31 [SuppI2]:S207-S212 Welvaart K (1992) Operative bypass for incurable cancer of the
Phan GQ, Yeo CJ, Hruban RH, Lillemoe KD, Pitt HA, Cameron head of the pancreas. Eur J Surg OncoI18:353-356
JL (1998) Surgical experience with pancreatic and peripan- Wiedenmann B, Jensen RT, Mignon M, Modlin CI, Skogseid
creatic neuroendocrine tumors: review of 125 patients. J B, Doherty G, Oberg K (1998) Preoperative diagnosis and
Gastrointest Surg 2:472-482 surgical management of neuroendocrine gastroenteropan-
Que FG, Nagorney DM, Batts KP, Linz LJ, Kvols LK (1995) creatic tumors: general recommendations by a consensus
Hepatic resection for metastatic neuroendocrine carcino- workshop. World J Surg 22:309-318
mas. Am J Surg 169:36-42; discussion 42-43 Yeo CJ, Wang BH, Anthone GJ, Cameron JL (1993) Surgical
Rigaud G, Missiaglia E, Moore PS, Zamboni G, Falconi M, Tala- experience with pancreatic islet-cell tumors. Arch Surg
mini G, Pesci A, Baron A, Lissandrini D, Rindi G, Grigolato 128: 1143-1148
10 Pathology
P. CAPELLI, G. ZAMBONI, A. PESCI, G. MARTIGNONI, and A. SCARPA

CONTENTS types have preferred sites (SOLCIA et al. 1997). They


originate from multipotent cells which have retained
10.1 Introduction 161 the ability to proliferate and differentiate into various
10.2 General Features 161 endocrine cell types and to produce several types of
10.2.1 Cytology 162
10.2.2 Macroscopic Features 162 hormones, both 'eutopic' and 'ectopic' in respect of
10.2.3 Histologic Features 163 those normally produced by this organ (CARNEY
10.3 Ancillary Methods for 1976; HEITZ et al. 1982; KLOPPEL and HEITZ 1988;
Histopathological Evaluation 166 PHILIPPE et al. 1988; SOLCIA et al. 1989,2000).
10.3.1 Immunohistochemistry 166 From the clinical point of view, PET are subdi-
10.3.2 Histochemistry 168
10.3.3 Electron Microscopy 168 vided into functioning and nonfunctioning endo-
10.4 Functioning Endocrine Tumors 168 crine tumors depending on whether or not they are
10.4.1 Insulinomas 168 accompanied by a clinical syndrome caused by the
10.4.2 Gastrinomas 168 hormone produced. The majority of well-differenti-
10.4.3 Glucagonomas 169 ated endocrine tumors of the pancreas are function-
10.4.4 VIPomas 169
10.4.5 Somatostatinomas 169
ing (SOLCIA et a1.1997).
10.4.6 Functioning Endocrine Tumors Sporadic PET are mostly solitary, while the heredi-
Producing Ectopic Hormones 169 tary ones are multifocal, such as those associated with
10.5 Nonfunctioning Endocrine Tumors 169 multiple endocrine neoplasia type I (MEN I) (BRANDI
10.6 Endocrine Tumors in MEN I Syndrome 170 et al. 1987; CHANDRASEKHARAPPA et al. 1997; DUH
10.7 Criteria of Malignancy
and Tumor Classification 170
et al. 1987; HESSMAN et al. 1998; LARSSON et al. 1988;
10.8 Differential Diagnosis 171 MOORE et al. 2001a; PADBERG et al. 1995; ROSAI et al.
References 172 1972; SAMAAN et al. 1989). In accordance with the
recent World Health Organization (WHO) classifica-
tion, PET are classified into well differentiated and
poorly differentiated tumors (SOLCIA et al. 2000).
10.1 While the poorly differentiated tumors are invariably
Introduction malignant and are therefore known as poorly differ-
entiated endocrine carcinomas, the behavior of the
Pancreatic endocrine tumors (PET) are either benign well differentiated tumors, which are more common,
or malignant epithelial tumors with endocrine differ- is unpredictable. A well differentiated tumor is defined
entiation characterized, in the majority of cases, by a as a malignant, well differentiated endocrine carci-
low grade of histological aggressiveness. noma only when invasion of the neighboring tissue
These tumors are rare, representing about 1%-2% and/or metastases are also present.
of all pancreatic tumors. They occur with similar fre-
quency in male and female adults and can be located
in any part of the pancreas, although the different
10.2
General Features
P. CAPELLI, MD; G. MARTIGNONI, MD; A. SCARPA, MD
Istituto di Anatomia Patologica, Policlinico Universitario 'G.B.
In the majority of patients, the cytological-histologi-
Rossi', P.le L.A. Scuro, 10,37134 Verona, Italy
G. ZAMBONI, MD; A. PESCI, MD cal characteristics are enough to suggest an endocrine
Servizio di Anatomia Patologica, Ospedale S. Cuore-Don Cal- origin. There is a well-defined spatial ratio between
abria, Via D. Sempreboni, 37024 Negrar, Italy the tumor cells and the rich vascular network and
162 P. Capelli et al.

the rather uniform appearance of the majority of the The cellular elements can show particular fea-
cells. The endocrine functionality is demonstrated by tures: oncocytic-like appearance, clear cytoplasm,
immunohistochemical methods. a prominent pleomorphic fused appearance, lead-
ing to a wide range of differential diagnoses such
as common ductal adenocarcinoma, rare anaplastic
10.2.1 carcinoma, mesenchymal or metastatic tumors. The
Cytology presence of papillary-like structures characterized
by a thin stromal core with a vessel in the center or
Examination of cytological material obtained by by acinar-like structures simulates solid pseudopap-
image-guided fine needle aspiration biopsy (FNAB) illary tumor (KLIMSTRA et al. 2000) and acinar cell
is a technique increasingly utilized for the diagnosis carcinoma (KLIMSTRA et al. 1992).
of a pancreatic mass. The material obtained is usually The amount of neoplastic cells obtained with
extremely cellular. At low magnification, the morpho- FNAB can also be low because of the presence of a
logical picture is characterized by the presence of large fibrous component of the tumor. Sometimes,
small to medium-sized, monomorphic cells, which the cytological smear contains a large quantity of
are homogeneously distributed and poorly cohesive. amorphous material, eosinophilic to hematoxylin-
The cells usually have a round or oval, central or eosin and polychromatic with Papanicolaou staining,
outlying nucleus (superficially resembling plasma similar to amyloid. In this case, the rare neoplastic
cells). The nucleus has finely distributed chromatin cells must be looked for very carefully.
with a 'salt and pepper' pattern (with tiny clumps of Cytological evaluation of the malignant potential
dense heterochromatin scattered through the nuclei) of endocrine tumors, with the exception of the most
and displays an inconspicuous nucleolus. Cytoplasm poorly differentiated carcinomas, is not reliable. In
is often slight, homogeneously eosinophilic and only fact, there are no definite criteria able to identify
rarely finely granular (Fig. 10.1). potentially aggressive tumors. A high nucleus-cyto-
Although most cases have these features, atypical plasmic ratio, nucleus characterized by thickened
cytological aspects can be found, which can make chromatin and sometimes with clearer areas against
the diagnosis difficult. The tumor cells can assume the nuclear membrane, the presence of frequent
particular features or arrange themselves in a pattern signs of apoptosis with a background of cellular
similar to other pancreatic cell types. The presence debris have negative prognostic significance. The
of a conspicuous nucleolus associated with a finely presence of single or rare cells with notable pleomor-
granular cytoplasm simulates acinar cell carcinoma phism, gigantic nucleus, and intranuclear cytoplas-
(KLIMSTRA et al. 1992). matic pseudoinclusions is not a sign of malignancy
(SOLCIA et al. 1997).

10.2.2
Macroscopic Features

The majority of PET is a single lesion with a rounded


or multilobulated border and sharp demarcation
from the surrounding parenchyma, despite the lack
of a pseudo capsule. Most masses are between 1 and 5
cm in diameter, but larger masses are not uncommon.
These characteristics indicate an expansive pattern
of growth which differs from the infiltrative pattern
of a ductal adenocarcinoma (Figs. 10.2, 10.3).
PETs usually have a solid appearance; cystic degen-
erative areas are rare; however, cystic PETs are well
described (DAVTYAN et al. 1990; LIGNEAU et al. 2001;
Fig. 10.1. Endocrine tumor: fine needle aspiration biopsy.
WARSHAW et al. 1990). When present, the preoperative
Extremely cellular aspirate composed of uniform, small to
medium-sized, poorly cohesive cells with eosinophilic cyto- diagnosis (SOLCIA et al. 1997) compared with a solid
plasm, rounded nucleus, 'salt and pepper' chromatin, and pseudopapillary tumor and other cystic masses may
inconspicuous nucleolus. Some cells resemble plasma cells be impossible (Fig. lOA).
Pathology 163

Fig. 10.4. Endocrine cystic tumor. Whole-mount macrosection


of an endocrine tumor with pseudo capsule and cystic change
Fig. 10.2. Endocrine tumor: Whipple resection specimen. Endo-
crine tumor of the head of the pancreas. Cut surface shows a
solid, well-circumscribed, whitish tumor compressing the
common bile duct (arrow)

Fig. 10.3. Endocrine tumor: left-side pancreatectomy specimen. Fig. 10.5. Insulinoma. Enucleation of an intrapancreatic endo-
Endocrine tumor of the body of the pancreas. Cut surface shows crine tumor. The tumor nodule appears well-circumscribed,
a solid, intrapancreatic tumor, well circumscribed compared has a homogeneous aspect and an intense red, hemorrhagic
with the surrounding parenchyma, brownish in color color

The color and the consistency depend on the splenic vessels with vascular thrombosis can cause
amount of stroma and the degree of vascularization. splenic infarcts (Fig. 1O.6).
The color varies from brown to reddish, and the con- Sometimes the tumor reaches a remarkable size,
sistency in most cases is higher than the surrounding even tens of centimeters, and infiltrates the walls of
pancreatic parenchyma. In rare cases, these masses the neighboring organs, forming ulcers responsible
appear considerably hemorrhagic, with a purple to for significant digestive bleeding and anemia.
bluish color and soft consistency. Sometimes, however, The presence of multiple lesions should lead to
the fibrosis is considerable, giving a firm consistency the suspicion of pancreatic involvement with MEN
and a whitish color. There may be necrotic and hem- I (BRANDI et al. 1987; SAMAAN et al. 1989). Multiple
orrhagic foci, especially in malignant masses, which insulinomas may be present in 7.5%-13% of patients,
appear as yellowish or brownish areas with a soft and gastrinomas in up to 30% (SOLCIA et al. 1997).
consistency. Nonfunctioning PETs are usually large;
insulinomas are among the smallest (Fig. 1O.5).
Sometimes, a PET may demonstrate the features of 10.2.3
a malignant tumor: ill-defined margins, infiltration Histologic Features
of the perivisceral adipose tissue, mainly as satellite
nodules, infiltration of the duodenal wall, common In the majority of cases, the histological appearance
bile duct, spleen, or large vessels. Involvement of the of the tumor is enough to suggest an endocrine
164 P. Capelli et al.

(3) solid-nodular ('insular') pattern, typical of malig-


nant tumors (Fig. 10.8c), where the fibrous stroma of
the mass delineates cohesive groups of cells, resulting
in the characteristic nodular or insular appearance
(MUKAI et al. 1982; SOLCIA et al. 1997). Although one
pattern is generally prevalent, there may be different
growth patterns within the same tumor. Reports of
a papillary growth pattern (Fig. 10.8d) or rare cases
with mucus (TOMITA et al. 1981), hyaline periodic
acid-Schiff (PAS)-positive globules, cells with clear
(Fig. 10.9a; GUARDA et al. 1983; HOANG et al. 2001),
vacuolated (ORDONEZ and SILVA 1997), or oxyphilic
cytoplasm (Fig. 10.9b; GOTCHALL et al. 1987; RADI
et al. 1985) can be observed, but these forms are
extremely unusual.
Fig. 10.6. Endocrine carcinoma. The tumor completely occu- The hyperdense radiological appearance is related
pies the tail of the pancreas, infiltrates the peripancreatic adi-
to the rich vascularization, which characterizes the
pose tissue and the splenic capsule. Cut surface of the spleen
shows signs of embolization of the splenic vessels large majority of these lesions (Fig. 10. lOa). Only
rare cases show prevalent wide bands of sclerohya-
line stroma, sometimes with calcified foci. When the
ongm because the majority are well-differenti- sclerosis is widespread, the cells can appear to float in
ated tumors (Fig. 10.7). These are characterized by this fibrous tissue (Fig. 10.1 Ob). Calcifications, either
a homogeneous cellularity, composed of small to as large deposits or as psammoma bodies, are rare.
medium-sized elements, similar in shape and size, The presence of amyloid, as deposits of extracellu-
which can occasionally have an irregular shape with a lar, amorphous, eosinophilic material, is frequently
voluminous nucleus and frequent cytoplasmic inclu- observed in insulinomas.
sions. Three main growth patterns can be observed: Since these tumors grow slowly, normal pancreatic
(1) trabecular pattern (Fig. 10.8a) with cords and structures such as ducts and islets can be entrapped
ribbons, frequent in benign tumors where the cells within the tumor.
are lengthened, the main axis perpendicular to that Infiltration of local structures characterizes malig-
of the trabecula, and the nucleus polarized; (2) glan- nant forms and should be identified by histological
dular-like or acinar pattern (Fig. 10.8b) comprising examination. Infiltration is not always evident from
acini and tubules, often containing secreted material; macroscopic examination and therefore should be
looked for with care. Generally, there are foci of
tumoral infiltration of the muscular tunic of the duo-
denum, the choledochal wall, or lymph node metasta-
ses; evaluation of perivisceral adipose tissue infiltra-
tion can be more difficult especially when the tumor
has expansive growth margins and a pseudocapsule.
In very rare cases, solid and wide areas with
poorly differentiated, small cells with hyperchro-
mic nucleus and little cytoplasm characterize the
tumor. The mitotic activity is generally high because
of widespread apoptosis; necrotic areas can some-
times be very large, with an appearance known as a
'geographic map' (Fig. 10.11). In this case, the tumor
is poorly differentiated, similar to a small-cell pul-
monary carcinoma, and requires the use of immu-
nohistochemical methods to identify its endocrine
differentiation.
Fig. 10.7. Endocrine tumor. Endocrine tumors are typically
composed of small to medium-sized cells with uniform, It should be remembered from the latter that the
round, or oval, central or outlying nuclei, and eosinophilic tumor's histological aspect alone does not lead to any
and granular cytoplasm conclusions about its functional state or the type of
Pathology 165

a b

Fig. 1O.8a-d. Endocrine tumors growth pattern: trabecular pattern (a), microglandular pattern (b), solid-nodular pattern (c),
unusual papillary pattern (d)

a b

Fig. 10.9a,b. Endocrine tumors, unusual cell types: cells with clear cytoplasm (a), oncocytic cells with deeply eosinophilic
cytoplasm (b)
166 P. Capelli et al.

Fig.lO.lOa,b. Endocrine tumors, the stroma: richly vascularized stroma (a), fibrous stroma (b)

a
Fig. 10.11a,b. Poorly differentiated endocrine carcinoma. a Tumor with solid pattern made up of atypical, small cells with high
nucleus-cytoplasm ratio, poor, scanty cytoplasm, and frequent mitosis. b Abundant necrosis is present

hormone produced. There are two exceptions: the Endocrine differentiation of the tumor can be
presence of stroma with amyloid deposits indica- highlighted by immunophenotype markers common
tive of insulin om a, and a glandular-like growth pat- to the cells of the endocrine system (BORDI et al.
tern with psammoma bodies seen in periampullary 1979): neurospecific enolase (NSE) (SCHMECHEL et
somatostatinoma. al. 1978), protein gene product (PGP) 9.5 (RODE et
al. 1985), synaptophysin (GOULD et al. 1987), and
chromogranins A, B, and C (HAGN et al. 1986; LLOYD
et al. 1984). It is important to subdivide these into
10.3 cytoplasmic markers, cytosolic and microvesicular,
Ancillary Methods for Histopathological and granular.
Evaluation NSE and PGP 9.5 are cytoplasmic proteins, while
synaptophysin belongs to the complex family of
10.3.1 proteins of small vesicle membrane. The staining
Immunohistochemistry intensity does not depend on the granular secre-
tion or the type of hormone produced (BaRD! et al.
The immunohistochemical method is extremely 1979). The staining due to antibodies that recognize
useful both for the diagnosis of endocrine tumors neurosecretory granules depends on the granule
and for their classification. content of the cells. Positivity for chromogranin
Pathology 167

A is the same as the identification of granules by 1981). The hormone produced and responsible for
silver impregnation methods (coloring according the clinical syndrome can be highlighted (HEITZ et
to Grimelius). al. 1982; SOLCIA et al. 1997). The staining intensity
Generic endocrine markers are most important or the number of positive cells does not correlate
when used for confirming the diagnosis, especially with the severity of the symptoms. Moreover, the
with poorly differentiated tumors. Nevertheless, cells may not demonstrate positivity for any hor-
highly specific granular markers are only faintly mones because of their hormonal overproduction.
positive or completely negative in poorly differenti- This classification is used in the differentiated forms,
ated tumors. Therefore, with these tumors, it would whereas hormone production is never seen in poorly
be better to use cytoplasmic markers, which persist differentiated endocrine carcinomas. The type of
in the neoplastic cells. Synaptophysin has a higher hormone produced may be useful for the prognosis
level of specificity compared with NSE. (SOLCIA et al. 2000) and for the early recognition of
As well as endocrine differentiation markers, relapse or metastases.
these tumors are positive for cytokeratins 8, 18, Finally, the proliferation index (Ki -67), p53, human
and 19 (HOEFLER et al. 1986; HOORENS et al. 1998) chorionic gonadotropin, and the progesterone recep-
(Fig. 10.12). tor obtained with immunohistochemical methods
There are also specific antibodies for the nor- have shown prognostic significance in PETs (KAHN
mally produced pancreatic hormones or hormones et al. 1997; OBERG and WIDE 1981; PELOSI et al. 1996;
of ectopic origin, which can be used in classifying VIALE et al. 1992). The cells of these tumors are,
endocrine tumors because of their pathogenetic or however, negative for the markers which identify the
prognostic role (i.e., alpha-chain of human chorionic enzymes produced by the exocrine pancreas, such as
gonadotropin; KAHN et al. 1977; OBERG and WIDE trypsin, amylase, and lipase.

a b

Fig.l0.12a-c. Endocrine tumor, immunohistochemistry: cyto-


keratin (a), chromogranin (b), and PGP 9.5 (c) positivity
168 P. Capelli et al.

10.3.2 with a higher growth potential. This could explain why


Histochemistry most glucagon-producing tumors resulting in gluca-
gonoma syndrome are malignant in contrast to, for
The use of specific histochemical stains for endocrine example, insulinomas. The majority of glucagon-pro-
cells has almost been completely replaced by immuno- ducing tumors without syndrome are micro adenomas
histochemical markers. Grimelius silver staining was discovered by chance.
particularly diffuse; its role in identifying neurosecre- Insulinomas, glucagonomas, and VIPomas have a
tory cytoplasmic granules is analogous to that of the preference for the body-tail, while gastrinomas are
immunohistochemical expression of chromogranin A. mainly found in the head of the pancreas. Tumors in
Mucus staining (mucicarmine, PAS) is generally the body-tail preferably spread through blood ves-
negative, although mucus droplets can be identified sels, while pancreatic gastrinomas spread along the
inside a glandular or ductal type pattern. Glycogen, lymphatic route.
identified by PAS and PAS-diastasis, is usually absent.
Sometimes hyaline PAS-positive globules may be
identified inside or outside the cells (generally alpha- 10.4.1
I-antitrypsin positive). Insulinomas
Congo red staining and the subsequent observa-
tion under polarized light confirm the possible pres- Insulinomas are the most common functioning PET
ence of amyloid. and are, after gastrinomas, the tumors which are
most frequently found in patients with MEN I. The
clinical signs and symptoms are a direct result of
10.3.3 insulin -mediated hypoglycemia.
Electron Microscopy The tumors are exclusively located in the pancreas
and can be distributed throughout this organ. Most
Electron microscopy has long been used to show the insulinomas are benign (85%-99%), single, and less
neuroendocrine differentiation of the tumors since than 2.5 em in size (LIU et al. 1985; STEFANINI et al.
the cells contain electron-dense secretory granules, 1974). Malignant insulinomas are generally larger
which are similar to those of islet cells. The neurose- than 3 em, and about a third of them have metastases
cretory granules can vary in shape and size according at the time of diagnosis. In 2%-7% of patients, insu-
to the specific hormone that they store. This method linomas are multiple, synchronous or metachronous,
has been largely replaced by immunohistochemistry, and 6% of patients have MEN I syndrome (DONOW
at least at the diagnostic level. et al. 1990).
Immunohistochemically, well-differentiated insu-
linomas with a trabecular-type growth pattern are
characterized by considerable insulin positivity (at the
10.4 lower pole of the cell; ROTH et al. 1992). By contrast,
Functioning Endocrine Tumors insulinomas with a solid pattern show no positiv-
ity at all or only a very slight positivity. In 5% of the
These are tumors with an associated clinical syn- insulinom as, amyloid may be present in the stroma.
drome, and they represent the large majority of In the pancreatic parenchyma around the tumor, the
clinically significant PETs. islet content of immunoreactive insulin cells may
The different types of hormones or cells have a be decreased; this probably represents an adaptive
different potential in producing symptoms that are response by the beta-cells to the prolonged hypoglyce-
relevant from a clinical point of view. For example, mia (BANI SACCHI et al. 1989).
most patients with insulin-producing tumors develop
the insulinoma syndrome even when their tumor
is smaller than 1 em, while only a small percentage 10.4.2
of patients with glucagon-producing tumors, usu- Gastrinomas
ally very large, develop the glucagonoma syndrome
(BORDI et al. 1979; KLOPPEL et al. 1986; RUTTMAN et This is the second most common endocrine tumor
al. 1980). In the latter case, a more voluminous mass or of the pancreas. Most gastrinomas are located inside
a longer clinical course is probably necessary to pro- the anatomic region between the head of the pan-
duce symptoms, with the consequent selection of cells creas, the common bile duct, and the first and second
Pathology 169

portion of the duodenum, known as the 'gastrinoma 10.4.4


triangle' (STABILE et al. 1984). VIPomas
The excessive production of gastrin leads to the
onset of Zollinger-Ellison syndrome. This syndrome Most VIPomas arise in the pancreas and are malig-
may be sporadic or a manifestation of MEN I. About nant 80% of the time. They are usually large, solitary
50%-70% of gastrinomas associated with the spo- tumors with an average diameter at diagnosis of 4-5
radic form of Zollinger-Ellison syndrome are found cm and are often located in the tail of the pancreas
in the pancreas, especially in the head, while the rest (BLOOM et al. 1973; SOLCIA et al. 1997; VERNER and
are mainly found in the duodenum (DONOW et al. MORRISON 1974).
1991). Single gastrinomas have a diameter of 2 cm or The overproduction of VIP (vasoactive intestinal
more in the pancreas and usually of 1 cm or less when polypeptide), demonstrated immunohistochemi-
in the duodenum (DONOW et al. 1991; STAMM et al. cally, causes watery diarrhea, hypokalemia, and
1986; THOMPSON et al. 1989). Most of the gastrino- achlorhydria (BLOOM et al. 1973; HEITZ et al. 1982;
mas associated with MEN I are found in the duode- VERNER and MORRISON 1974).
num (PIPELEERS-MARICHAL et al.1990). In this case,
they are usually smaller than 1 cm, multicentric, and
difficult to identify. Pancreatic gastrinomas in MEN 10.4.5
I are rare (KLOPPEL et al. 1986; VELLA et al. 1988), Somatostatinomas
although the pancreas of these patients can be the
site of multiple endocrine tumors, which rarely pro- These are rare and generally malignant endocrine
duce appreciable amounts of gastrin (KLOPPEL et al. tumors of the pancreas. Only pancreatic somatostati-
1986; PIPELEERS-MARICHAL et al. 1990). nomas can be responsible for a hormonal syndrome.
At diagnosis, about 60% of gastrinomas already Histologically, they can be characterized by the pres-
have metastases in the peripancreatic lymph nodes ence of calcifications with the features of psammoma
and less frequently in the liver (DELCORE et al. 1988; bodies (SOLCIA et al. 1997; VINIK et al. 1987). The most
STABILE and PASSARO 1985). common site of extrapancreatic somatostatinoma is
Histologically, gastrinomas show a solid, trabecular the second portion of the duodenum around the
or pseudoglandular-type growth pattern, and immu- papilla of Vater, and some are associated with neuro-
nohistochemically almost all cases are gastrin positive, fibromatosis type 1 (Von Recklinghausen syndrome)
particularly in the forms with a trabecular growth pat- and pheochromocytomas (STAMM et al. 1986).
tern. In about 50% of cases, there is even positivity for
other hormones (PP, glucagon, insulin).
10.4.6
Functioning Endocrine Tumors
10.4.3 Producing Ectopic Hormones
Glucagonomas
This is a group of tumors associated with syndromes
These are malignant tumors in over 60% of patients caused by ectopic hormones. They appear as large
(RUTTMAN et al. 1980). The size at diagnosis varies masses and are malignant. Their microscopic pat-
between 2 and 3.5 cm, and they are generally located terns are usually the same as in other PETs. They
in the distal portion of the pancreas. Tumors char- can produce adrenocorticotropic hormone (ACTH;
acterized by glucagon-positive cells with no clinical Cushing's syndrome), serotonin (atypical carcinoid
symptoms are mostly benign and are either found by syndrome), growth hormone releasing factor (acro-
chance at autopsy or are small tumors in the course of megaly), or parathyroid hormone (PTH)-like hor-
MEN I syndrome (BORD! et al. 1979; KLOPPEL et al. mones (paraneoplastic hypercalcemia).
1986; RUTTMAN et al. 1980). Immunohistochemically,
glucagonomas often show weak glucagon positivity,
while there may be numerous PP cells.
The excessive amount of glucagon in the blood 10.5
results in a clinical syndrome characterized by Nonfunctioning Endocrine Tumors
migrant necrolytic erythema, slight intolerance to
glucose, depression, and a tendency to develop deep These are endocrine tumors not associated with any
venous thrombosis (MALLINSON et al. 1974). particular clinical syndrome. However, hormone
170 P. Capelli et al.

positivity can be shown on tissue with immunohis- multifocal. The pancreas is usually characterized by
tochemical methods. This functional 'silence' can be the presence of multiple endocrine tumors of vari-
due to: (1) the production and/or release of an insuf- ous sizes, which produce different hormones (PIPEL-
ficient amount of hormone; (2) the rapid degradation EERS-MARICHAL et al. 1990). In patients with MEN
of the hormone; (3) the processing of a hormone I, the Verner-Morrison syndrome, glucagon om a
which is unable to produce a definite clinical picture, syndrome, or acromegaly is very rare (KLOPPEL et al.
as is the case with PPomas; (4) the production of a 1986; 001 et a1.l985; STACPOOLE et al. 1981; WARNER
functionally inactive hormone. et al. 1983).
The symptoms are mostly linked to the presence of
metastases or the mass effect. At surgery, they appear
as large masses, usually with a diameter greater than
5 cm, and are malignant in the majority of cases 10.7
(ECKHAUSER et al.1986; SOLCIA et al.I997). However, Criteria of Malignancy
there are also small nonfunctioning tumors, discov- and Tumor Classification
ered by chance and generally benign.
There is no difference in the growth pattern Invasion of the adjacent organs and/or the pres-
between endocrine functioning and nonfunctioning ence of metastases within the lymph nodes, liver, or
tumors. other organs are unequivocal criteria of malignancy
About 10% of tumors have absent secretory (Fig. 10.13). With the exception of poorly differenti-
granules; this is reflected in the absence or slight ated tumors characterized by anaplastic cells (like
positivity of chromogranin, while the positivity of small-cell carcinoma), the traditional histopathologic
other differentiation markers, such as NSE, synapto- criteria of malignancy are poor predictors of the
physin, and PGP 9.5, is preserved. Among nonfunc- biological behavior of PETs. They are usually well-
tioning endocrine tumors, which are chromogranin differentiated and, at the time of diagnosis, display
A-positive and therefore have secreting granules, PP neither metastases nor macroscopic invasion of the
is the hormone which is more frequently identified adjacent organs, making prediction of their biologi-
(TOMITA et al. 1983), followed by glucagon and soma- cal behavior difficult. Metastases may appear years
tostatin (KLOPPEL and HEITZ 1988). after the operation. Numerous studies have been car-
ried out to determine the predictive value of several
indicators. The parameters considered are the size of
the tumor, microscopic identification of vascular inva-
10.6 sion, perineural invasion, mitosis, necrosis, expression
Endocrine Tumors in MEN I Syndrome of some immunophenotypic markers, and evaluation
of the DNA content.
The pancreatic involvement in MEN I syndrome is These studies demonstrate that the only reliable
characterized by a diffuse micro adenomatosis in histological marker of malignancy is invasion of the
association with one or several macrotumors (more blood vessels. Lymphatic vascular invasion, high pro-
than 0.5 cm in diameter) (KLOPPEL et al. 1986; KOM- liferative activity (Ki 67>2%-5%), high mitotic index
MINOTH et al. 1998; PADBERG et al. 1995). (>2 per 10HPF),and the presence of tumoral necrosis
Histologically, the majority of these small (LA ROSA et aI. 1996; PELOSI et al. 1996,1997; RIGAUD
tumors shows a characteristic trabecular pattern et al. 2001) are presumptively correlated to malignant
and can have a fibrous capsule. Immunohisto- behavior. Another prognostic factor seems to be the
chemical positivity for more than one hormone is size of the tumor. Well-differentiated endocrine non-
typical, even though one hormone usually prevails functioning tumors that are larger than 4 cm have a
(PP»glucagon>insulin; KLOPPEL et al. 1986). In greater risk of relapse and metastases (LA ROSA et al.
patients with MEN I and hypoglycemic syndrome, 1996); for functioning tumors, the cut-off is smaller
only one of the macrotumors generally produces (2 cm) (DONOW et al. 1990).
insulin (DONOW et al. 1990; KLOPPEL et al. 1986) On the whole, well-differentiated endocrine
despite the presence of multiple endocrine tumors. In tumors smaller than 2 cm are benign.
patients with MEN I and Zollinger-Ellison syndrome Ploidy (RIGAUD et al. 2001 ),expression of oncogenes/
(about 60% of the patients with MEN 1), gastrino- tumor suppressor genes (HOFLER et al. 1988; CHUNG et
mas are often located in the proximal portion of the al. 1997; BEGHELLI et al. 1998; MISSIAGLIA et al. 2002;
duodenum, are less than 1 cm in diameter, and are SCARPA et al. 2002), expression of progesterone recep-
Pathology 171

a b

Fig. 10.13a-c. Endocrine carcinoma: vascular invasion (a),


lymph node (b) and hepatic metastases (c)

tors (PELOSI et al. 1996; VIALE et al. 1992), and the pres- attempt to define the outcome of such lesions better.
ence of alpha-human chorionic gonadotropin (HCG) In this classification, PETs have been subdivided
(KAHN et al. 1977; OBERG and WIDE 1981; RUSCHOFF et into distinct categories according to their biological
al. 1993; SOLCIA et al. 2000), although important, seem behavior, considering the level of histological differ-
to be ofless prognostic value. entiation, eventual hormone secretion, size and other
The possible presence or absence of a clinical syn- parameters taken as prognostic indicators, such as
drome also seems to have a predictive value, with a angio-invasion, number of mitoses, and the prolif-
likelihood of metastases reaching 10% for clinically eration index (Table 10.1).
silent tumors discovered by chance and insulinomas,
and up to 60%-90% for glucagon om as, somatostatin-
omas, VIPomas, gastrinomas, tumors associated with
ectopic hormone production, and symptomatic non- 10.8
functioning endocrine tumors (SOLCIA et al. 1997). Differential Diagnosis
The poor survival rate of non functioning endo-
crine tumors compared with functioning ones The diagnosis of PET is usually made with conven-
should probably be considered secondary to the tional histology and confirmed by the immuno-
advanced stage at diagnosis. histochemical identification of endocrine markers.
A new classification of PET has recently been Problems of differential diagnosis may arise in
published by the World Health Organization (WHO) some patients with chronic pancreatitis with massive
(SOLCIA et al. 2000), based on a consensus of 12 endocrine hyperplasia and may occur with exocrine
leading pathologists. This classification takes into tumors of the pancreas, nonepithelial tumors, or
account the clinical and pathological features in an pancreatic metastases.
172 P. Capelli et al.

Table 10.1. Clinicopathological correlations of endocrine tumors of the pancreas (Functioning, associated with pertinent clinical
syndrome of endocrine hyperfunction; Nonfunctioning, not associated with pertinent clinical syndrome, irrespective of hormone
detection in blood or tumor tissue)
Well-differentiated endocrine tumor
1.1 Benign behavior: confined to the pancreas, non-angioinvasive, <2 cm in sizea, £2 mitoses, and £2 Ki 67-positive cells/lO HPF
1.1.1 Functioning insulinoma
1.1.2 Nonfunctioning
1.2 Uncertain behavior: confined to the pancreas, +2 cm in size, >2% Ki 67-positive cells/lO HPF, or angioinvasive
1.2.1 Functioning gastrinoma, insulinoma, VIPoma, glucagonoma, somatostatinoma, or inappropriate syndromeb tumor
1.2.2 Nonfunctioning
2 Well-differentiated endocrine carcinoma
2.1 Low-grade malignant with gross local invasion and/or metastases
2.1.1 Functioning gastrinoma, insulinoma, VIPoma, glucagonoma, somatostatinoma, or inappropriate syndromeb tumor
2.1.2 Nonfunctioning
3 Poorly differentiated endocrine small-cell carcinoma, high grade malignant

a <2 cm in size implies close to 100% probability of benign behavior, <3 cm corresponds to 90% probability
b Inappropriate hormone syndromes: Cushing (ACTH), acromegaly or gigantism (GRH), hypercalcemia, etc.

Endocrine hyperplasia may be observed in the ential diagnosis with other tumors. The presence of
course of obstructive or nonobstructive chronic endocrine markers solves the differential diagnosis
pancreatitis. In this case, endocrine elements are with poorly differentiated ductal adenocarcinoma;
grouped in vaguely insular structures characterized differential diagnosis with a malignant, small- or
by the persistence and normal distribution of all large-cell lymphoma is solved with the presence of
four cell types normally present in the pancreatic lymphoid markers. Differential diagnosis for the rare
islet. Moreover, the microadenomas generally show fused cell forms must be made with mesenchymal
signs of fibrosis or intratumoral sclerosis, unlike the tumors of low-level malignancy, such as gastroin-
hyperplastic insulae. testinal stromal tumor, highlighted by mesenchymal
The exocrine tumors that can be confused with markers. Among the pancreatic metastases which can
endocrine tumors are the solid pseudopapillary mimic an endocrine tumor are the clear-cell carcino-
tumor (KLIMSTRA et al. 2000) and less frequently mas of the kidney, small-cell carcinoma of the lung,
the acinar cell carcinoma (KLIMSTRA et al. 1992). and endocrine carcinoma originating in the ileum.
The solid pseudopapillary tumor mimics an endo-
crine neoplasm when it is solid and lacks regressive
phenomena usually seen in this tumor. Immuno-
histochemically, the cells show diffuse positivity for References
vimentin, neuron-specific enolase, focal but intense
Bani Sacchi T,Bani D,Biliotti G (1989) The endocrine pancreas
alpha-l antitrypsin positivity, and generally appear in patients with insulinomas. An immunocytochemical
negative for cytokeratin and other endocrine mark- and ultrastructural study of the nontumoral tissue with
ers (KLIMSTRA et al. 2000). morphometrical evaluations. Int J Pancreatol 5:11-28
The distinction between endocrine tumors and Beghelli S, Pelosi G, Zamboni G, Falconi M, Iacono C, Bordi C,
acinar cell carcinomas is very important because of Scarpa A (1998) Pancreatic endocrine tumors: evidence for
a tumor suppressor pathogenesis and for a tumor suppres-
the latter's worse prognosis. An acinar cell carcinoma sor gene on chromosome 17p. J PathoI186:41-50
with a solid-trabecular pattern, without an acinar Bloom SR, Polak JM, Pearse AG (1973) Vasoactive intestinal
pattern, may be confused with an endocrine tumor. peptide and watery-diarrhoea syndrome. Lancet 2:14-16
The presence of rare acinar structures and cells with Bordi C, Ravazzola M, Baetens D, Gorden P, Unger RH, Orci
abundant PAS-positive granular cytoplasm, together L (1979) A study of glucagonomas by light and electron
microscopy and immunofluorescence. Diabetes 28:925-936
with a large nucleus with a prominent nucleolus, are Brandi ML, Marx SJ, Aurbach GD, Fitzpatrick LA (1987)
suggestive of acinar differentiation. Confirmation is Familial multiple endocrine neoplasia type I: a new look
obtained by the immunophenotypic demonstration at pathophysiology. Endocr Rev 8:391-405
of pancreatic enzyme positivity (trypsin, lipase, Carney CN (1976) Congenital insulinoma (nesidioblastoma):
amylase) and the absence of endocrine differentia- ultrastructural evidence for histogenesis from pancreatic
ductal epithelium. Arch Pathol Lab Med 100:352-356
tion markers (KLIMSTRA et al. 1992). Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE,
Immunohistochemical investigations playa fun- Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z,
damental role in resolving the problems of differ- Lubensky lA, Liotta LA, Crabtree JS, Wang Y, Roe BA,
Pathology 173

Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim ate filament profile and Ki-ras mutations provides evidence
YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ of a ductal origin. J Pathol 185:53-60
(1997) Positional cloning of the gene for multiple endo- Kahn CR,Rosen SW, Weintraub BD,Fajans SS, Gorden P (1977)
crine neoplasia-type 1. Science 276:404-407 Ectopic production of chorionic gonadotropin and its sub-
Chung DC, Smith AP, Louis DN, Graeme-Cook F, Warshaw units by islet -cell tumors. A specific marker for malignancy.
AL, Arnold A (1997) A novel pancreatic endocrine tumor N Engl J Med 297:565-569
suppressor gene locus on chromosome 3p with clinical Klimstra DS, Heffess CS, Oertel JE, Rosai J (1992) Acinar cell
prognostic implications. J Clin Invest 100:404-410 carcinoma of the pancreas. A clinicopathologic study of 28
Davtyan H, Nieberg R, Reber HA (1990) Pancreatic cystic cases. Am J Surg Pathol16: 815-837
endocrine neoplasms. Pancreas 5:230-233 Klimstra DS, Wenig BM, Heffess CS (2000) Solid-pseudopapil-
Delcore R Jr, Cheung LY, Friesen SR (1988) Outcome ofiymph lary tumor of the pancreas: a typically cystic carcinoma of
node involvement in patients with the Zollinger-Ellison low malignant potential. Semin Diagn Pathol 17:66-80
syndrome. Ann Surg 208:291-298 Kloppel G, Heitz PU (1988) Pancreatic endocrine tumors.
Donow C, Pipeleers-Marichal M, Stamm B, Heitz PU, Kloppel Pathol Res Pract 183:155-168
G (1990) The pathology of insulinoma and gastrinoma. The Kloppel G, Willemer S, Stamm B, Hacki WH, Heitz PU
location, size, multicentricity, association with multiple (1986) Pancreatic lesions and hormonal profile of pan-
endocrine type-I neoplasms and malignancy. Dtsch Med creatic tumors in multiple endocrine neoplasia type 1. An
Wochenschr 115:1386-1391 immunocytochemical study of nine patients. Cancer 57:
Donow C, Pipeleers-Marichal M, Schroder S, Stamm B, Heitz PU, 1824-1832
Kloppel G (1991) Surgical pathology of gastrinoma. Site, size, Komminoth P, Heitz P, Kloeppel G (1998) Multiple endocrine
multicentricity, association with multiple endocrine neopla- disorders in molecular and cellular endocrine pathology.
sia type 1, and malignancy. Cancer 68:1329-1334 In: Stefaneanu L, Sasano T, Kovacs K (eds) Molecular and
Duh QY, Hybarger CP, Geist R, Gamsu G, Goodman PC, cellular endocrine pathology. Chapman and Hall, Phila-
Gooding GA, Clark OH (1987) Carcinoids associated with delphia
multiple endocrine neoplasia syndromes. Am J Surg 154: La Rosa S, Sessa F, Capella C, Riva C, Leone BE, Klersy C, Rindi
142-148 G, Solcia E (1996) Prognostic criteria in non-functioning
Eckhauser FE, Cheung PS, Vinik AI, Strodel WE, Lloyd RV, pancreatic endocrine tumors. Virchows Arch 429:323-333
Thompson NW (1986) Non-functioning malignant neuro- Larsson C, Skogseid B, Oberg K, Nakamura Y, Nordenskjold
endocrine tumors of the pancreas. Surgery 100:978-988 M (1988) Multiple endocrine neoplasia type 1 gene maps
Gotchall J, Traweek ST, Stenzel P (1987) Benign oncocytic to chromosome 11 and is lost in insulinoma. Nature 332:
endocrine tumor of the pancreas in a patient with polyar- 85-87
teritis no do sa. Hum PathoI18:967-969 Ligneau B, Lombard -Bohas C, Partensky C, Valette PJ, Calender
Gould VE, Wiedenmann B, Lee I, Schwechheimer K, Dock- A, Dumortier J, Gouysse G, Boulez J, Napoleon B, Berger F,
horn-Dworniczak B, Radosevich JA, Moll R, Franke WW Chayvialle JA, Scoazec JY (2001) Cystic endocrine tumors
(1987) Synaptophysin expression in neuroendocrine of the pancreas: clinical, radiologic, and histopathologic
neoplasms as determined by immunocytochemistry. Am features in 13 cases. Am J Surg Pathol 25:752-760
J PathoI126:243-257 Liu TH, Tseng HC, Zhu Y, Zhong SX, Chen J, Cui QC (1985)
Guarda LA, Silva EG, Ordonez NG, Mackay B, Ibanez ML (1983) Insulinoma. An immunocytochemical and morphologic
Clear cell islet cell tumor. Am J Clin Pathol 79:512-517 analysis of 95 cases. Cancer 56:1420-1429
Hagn C, Schmid KW, Fischer-Colbrie R, Winkler H (1986) Lloyd RV, Mervak T, Schmidt K, Warner TF, Wilson BS (1984)
Chromogranin A, B, and C in human adrenal medulla and Immunohistochemical detection of chromogranin and
endocrine tissues. Lab Invest 55:405-411 neuron-specific enolase in pancreatic endocrine neo-
Heitz PU, Kasper M, Polak JM, Kloppel G (1982) Pancreatic plasms. Am J Surg Pathol 8:607-614
endocrine tumors. Hum Pathol 13:263-271 Mallinson CN, Bloom SR, Warin AP, Salmon PR, Cox B (1974)
Hessman 0, Lindberg D, Skogseid B, Carling T, Hellman P, A glucagonoma syndrome. Lancet 2:1-5
Rastad J, Akerstrom G, Westin G (1998) Mutation of the Missiaglia E, Moore P, Williamson J, Lemoine N, Falconi M,
multiple endocrine neoplasia type 1 gene in nonfamilial, Zamboni G, Scarpa A (2002) Sex chromosome anomalies in
malignant tumors of the endocrine pancreas. Cancer Res pancreatic endocrine tumors. Int J Cancer 98: 532-538
58:377-379 Moore PS, Missiaglia E, Antonello D, Zamo A, Zamboni G, Cor-
Hoang MP, Hruban RH, Albores-Saavedra J (2001) Clear leto V, Falconi M, Scarpa A (2001a) Role of disease-causing
cell endocrine pancreatic tumor mimicking renal cell genes in sporadic pancreatic endocrine tumors: MEN1 and
carcinoma: a distinctive neoplasm of von Hippel-Lindau VHL. Genes Chromosomes Cancer 32:177-181
disease. Am J Surg Pathol 25:602-609 Moore PS, Orlandini S, Zamboni G, Capelli P, Rigaud G, Fal-
Hoefler H, Denk H, Lackinger E, Helleis G, Polak JM, Heitz PU coni M, Bassi C, Lemoine NR, Scarpa A (2001b) Pancreatic
(1986) Immunocytochemical demonstration of intermedi- tumors: molecular pathways implicated in ductal cancer
ate filament cytoskeleton proteins in human endocrine are involved in ampullary but not in exocrine non-ductal
tissues and (neuro-) endocrine tumors. Virchows Arch A or endocrine tumorigenesis. Br J Cancer 84:253-262
Pathol Anat Histopathol 409:609-626 Mukai K, Grotting JC, Greider MH, Rosai J (1982) Retrospec-
Hofler H, Ruhri C, Putz B, Wirnsberger G, Hauser H (1988) tive study of 77 pancreatic endocrine tumors using the
Oncogene expression in endocrine pancreatic tumors. Vir- immunoperoxidase method. Am J Surg PathoI6:387-399
chows Arch B Cell Pathol Incl Mol Pathol 55:355-361 Oberg K, Wide L (1981) hCG and hCG subunits as tumor
Hoorens A, Prenzel K, Lemoine NR, Kloppel G (1998) Undiffer- markers in patients with endocrine pancreatic tumors and
entiated carcinoma of the pancreas: analysis of intermedi- carcinoids. Acta Endocrinol (Copenh) 98:256-260
174 P. Capelli et al.

Ooi A, Kameya T, Tsumuraya M, Yamaguchi K, Abe K, Shimo- of the literature. Virchows Arch A Pathol Anat Histol 388:
sato Y, Yanaihara N (1985) Pancreatic endocrine tumors 51-67
associated with WDHA syndrome. An immunohistochemi- Samaan NA, Ouais S, Ordonez NG, Choksi UA, Sellin RV,
cal and electron microscopic study. Vir chows Arch A Pathol Hickey RC (1989) Multiple endocrine syndrome type 1.
Anat Histopathol 405:311-323 Clinical, laboratory findings, and management in five
Ordonez NG, Silva EG (1997) Islet cell tumor with vacuolated families. Cancer 64:741-752
lipid-rich cytoplasm: a new histological variant of islet cell Scarpa A, Orlandini S, Moore P, Lemoine N, Beghelli S, Baron
tumor. Histopathology 31:157-160 A, Falconi M, Zamboni G (2002) Dpc-4 is expressed in
Padberg B, Schroder S, Capella C, Frilling A, Kloppel G, Heitz virtually all primary and metastatic pancreatic endocrine
PU (1995) Multiple endocrine neoplasia type 1 (MEN 1) tumors. Virchows Arch 440:155-159
revisited. Virchows Arch 426:541-548 Schmechel D, Marangos PJ, Brightman M (1978) Neurone-
Pelosi G, Bresaola E, Bogina G, Pasini F, Rodella S, Castelli P, specific enolase is a molecular marker for peripheral and
Iacono C, Serio G, Zamboni G (1996) Endocrine tumors central neuroendocrine cells. Nature 276:834-836
of the pancreas: Ki-67 immunoreactivity on paraffin sec- Solcia E, Capella C, Fiocca R, Cornaggia M, Bosi F (1989)
tions is an independent predictor for malignancy: a com- The gastroenteropancreatic endocrine system and related
parative study with proliferating-cell nuclear antigen and tumors. Gastroenterol Clin North Am 18:671-693
progesterone receptor protein immunostaining, mitotic Solcia E, Capella C, Kloeppel G (1997) Tumors of the pancreas,
index, and other clinicopathologic variables. Hum Pathol vol fasc 20. AFIP atlas of tumor pathology, 3rd ser. Armed
27:1124-1134 Forces Institute of Pathology, Washington DC
Pelosi G, Pasini F, Bresaola E, Bogina G, Pederzoli P, Biolo S, Solcia E, Kloppel G, Sobin L (2000) Histological typing of
Menard S, Zamboni G (1997) High-affinity monomeric endocrine tumors. W.H.O. classification of endocrine
67-kD laminin receptors and prognosis in pancreatic tumors. Springer, Berlin Heidelberg New York
endocrine tumors. J Pathol 183:62-69 Speel EJ, Richter J, Moch H, Egenter C, Saremaslani P, Rutimann
Philippe J, Powers AC, Mojsov S, Drucker DJ, Comi R, Habener K, Zhao J, Barghorn A, Roth J, Heitz PU, Komminoth P
JF (1988) Expression of peptide hormone genes in human (1999) Genetic differences in endocrine pancreatic tumor
islet cell tumors. Diabetes 37:1647-1651 SUbtypes detected by comparative genomic hybridization.
Pipeleers-Marichal M, Somers G, Willems G, Foulis A, Imrie Am J PathoI155:1787-1794
C, Bishop AE, Polak JM, Hacki WH, Stamm B, Heitz PU et Stabile BE, Passaro E Jr (1985) Benign and malignant gastri-
al (1990) Gastrinomas in the duodenums of patients with noma. Am J Surg 149:144-150
multiple endocrine neoplasia type 1 and the Zollinger-Elli- Stabile BE, Morrow DJ, Passaro E Jr (1984) The gastrinoma
son syndrome. N Engl J Med 322:723- 727 triangle: operative implications. Am J Surg 147:25-31
Radi MJ, Fenoglio-Preiser CM, Chiffelle T (1985) Functioning Stacpoole PW, Jaspan J, Kasselberg AG, Halter SA, Polonsky
oncocytic islet-cell carcinoma. Report of a case with elec- K, Gluck FW, Liljenquist JE, Rabin D (1981) A familial
tron-microscopic and immunohistochemical confirma- glucagonoma syndrome: genetic, clinical and biochemical
tion. Am J Surg PathoI9:517-524 features. Am J Med 70:1017-1026
Rigaud G, Missiaglia E, Moore PS, Zamboni G, Falconi M, Tala- Stamm B, Hedinger CE, Saremaslani P (1986) Duodenal and
mini G, Pesci A, Baron A, Lissandrini D, Rindi G, Grigolato ampullary carcinoid tumors. A report of 12 cases with
P, Pederzoli P, Scarpa A (2001) High resolution allelotype pathological characteristics, polypeptide content and rela-
of non-functional pancreatic endocrine tumors: identifica- tion to the MEN I syndrome and von Recklinghausen's
tion of two molecular subgroups with clinical implications. disease (neurofibromatosis). Virchows Arch A Pathol Anat
Cancer Res 61:285-292 Histopathol 408:475-489
Rindi G, Villanacci V, Ubiali A, Scarpa A (2001) Endocrine Stefanini P, Carboni M, Patrassi N, Basoli A (1974) Beta-islet
tumors of the digestive tract and pancreas: histogenesis, cell tumors of the pancreas: results of a study on 1,067
diagnosis and molecular basis. Expert Rev Mol Diagn 1: cases. Surgery 75:597-609
323-333 Thompson NW, Vinik AI, Eckhauser FE (1989) Microgastrino-
Rode J, Dhillon AP, Doran JF, Jackson P, Thompson RJ (1985) mas of the duodenum. A cause of failed operations for the
PGP 9.5, a new marker for human neuroendocrine tumors. Zollinger-Ellison syndrome. Ann Surg 209:396-404
Histopathology 9:147-158 Tomita T, Bhatia P, Gourley W (1981) Mucin producing islet
Rosai J, Higa E, Davie J (1972) Mediastinal endocrine neoplasm cell adenoma. Hum Pathol 12:850-853
in patients with multiple endocrine adenomatosis. A previ- Tomita T, Friesen SR, Kimmel JR, Doull V, Pollock HG (1983)
ously unrecognized association. Cancer 29:1075-1083 Pancreatic polypeptide-secreting islet -cell tumors. A study
Roth J, Kloppel G,Madsen OD, Storch MJ, Heitz PU (1992) Dis- of three cases. Am J PathoI113:134-142
tribution patterns of proinsulin and insulin in human insu- Vella MA, Cowie AG, Gorsuch AN, Watson LC (1988) Giant gas-
linomas: an immunohistochemical analysis in 76 tumors. trinoma in a patient with multiple endocrine adenopathy
Virchows Arch B Cell Pathol Incl Mol PathoI63:51-61 (type 1). J R Soc Med 81:359-360
Ruschoff J, Willemer S, Brunzel M, Trautmann ME, Frank M, Verner JV, Morrison AB (1974) Endocrine pancreatic islet dis-
Arnold R, Kloppel G (1993) Nucleolar organizer regions ease with diarrhea. Report of a case due to diffuse hyper-
and glycoprotein-hormone alpha-chain reaction as mark- plasia of nonbeta islet tissue with a review of 54 additional
ers of malignancy in endocrine tumors of the pancreas. cases. Arch Intern Med 133:492-499
Histopathology 22:51-57 Viale G, Doglioni C, Gambacorta M, Zamboni G, Coggi G,
Ruttman E, Kloppel G, Bommer G, Kiehn M, Heitz PU (1980) Bordi C (1992) Progesterone receptor immunoreactivity
Pancreatic glucagonoma with and without syndrome. in pancreatic endocrine tumors. An immunocytochemi-
Immunocytochemical study of 5 tumor cases and review cal study of 156 neuroendocrine tumors of the pancreas,
Pathology 175

gastrointestinal and respiratory tracts, and skin. Cancer (1983) Glucagonomas. Ultrastructure and immunocyto-
70:2268-2277 chemistry. Cancer 51: 1091-1096
Vinik AI, Strodel WE, Eckhauser FE, Moattari AR, Lloyd R Warshaw AL, Compton CC, Lewandrowski K, Cardenosa G,
(1987) Somatostatinomas, PPomas, neurotensinomas. Mueller PR (1990) Cystic tumors of the pancreas. New
Semin OncoI14:263-281 clinical, radiologic, and pathologic observations in 67
Warner TF, Block M, Hafez GR, Mack E, Lloyd RV, Bloom SR patients. Ann Surg 212:432-443; discussion 444-435
11 Endocrine Hyperfunctioning Tumors
R. F. THOENI

CONTENTS 11.1
Introduction
ILl Introduction 177
11.2 Imaging of the Pancreas with CT 177 Radiologic imaging tests for suspected endocrine
11.2.1 Techniques 177
11.2.2 Appearance of the Pancreas on CT 179 hyperfunctioning tumors of the pancreas are usually
11.3 Imaging of the Pancreas with MR Imaging 179 performed to localize a lesion rather than to prove
1l.3.1 Spin-Echo Sequences 179 its existence. This is due to the fact that these lesions
1l.3.2 Fast MR Imaging 179 produce specific symptoms, and therefore the clini-
1l.3.2.1 Fast Spin-Echo and cal presentation, together with the laboratory tests,
Single-Shot Fast Spin-Echo Sequences 179
11.3.2.2 Gradient-Echo Sequences 180 is diagnostic of the disease. The role of radiologic
11.3.3 Appearance of the Pancreas on MR Imaging 180 imaging lies in determining the exact location of the
11.4 Imaging of the Pancreas with Ultrasound 181 neoplasm within the pancreas and/or outside of the
11.4.1 Endoscopic Ultrasound of the Pancreas 181 pancreas in cases with ectopic locations. Multiple
11.4.2 Intraoperative Ultrasound of the Pancreas 182 imaging techniques can be employed for detecting
11.4.3 Appearance of the Pancreas on Ultrasound 182
1l.S Somatostatin -Receptor these lesions as they are often quite small. We shall
Imaging of the Pancreas 182 describe the various techniques available in the
11.5.1 Techniques of armamentarium of imaging tests for assessing these
Somatostatin-Receptor Imaging 182 rare lesions and outline the specific features as well
11.5.2 Appearance of the Pancreas as the results with each one. Finally, we shall assess
on Somatostatin-Receptor Imaging 184
11.6 Other Radiologic Diagnostic Tests 184 the effectiveness and role of imaging for these hyper-
11.6.1 Arteriography of the Pancreas 184 functioning pancreatic tumors.
11.6.2 Portal Venous Sampling 184
11.6.3 Arterial Stimulation and Venous Sampling 185
11.7 Features and Results of Imaging Hyperfunctioning
Endocrine Tumors of the Pancreas 185
11.7.1 Insulinoma 185 11.2
11.7.2 Gastrinoma 188 Imaging of the Pancreas with (T
11.7.3 VIPoma 189
11.7.4 Glucagonoma 190 11.2.1
11.7.5 Somatostatinoma 191 Techniques
11.7.6 Other Hyperfunctioning Endocrine Tumors 192
11.8 Role of Imaging for Hyperfunctioning
Endocrine Tumors 193 Multirow helical computed tomography (CT) pro-
References 193 vides high image resolution, excellent contrast
enhancement, and very short examination times and
therefore is considered the primary imaging method
for the pancreas. In order to assess the pancreatic
parenchyma accurately by CT, intravenous iodinated
contrast material should be used. Initially, regardless
of the protocol employed, the patient is scanned
without an intravenous contrast agent in order to
determine where the pancreas is located.
R. F. THOEN!, MD
Professor of Radiology, University of California San Francisco, The protocol varies depending on the question at
Department of Radiology, Box 0628, San Francisco, hand. If the clinical question is to screen the pancreas,
CA 94143-0628, USA a single-phase protocol can be sufficient. However, for
178 R. F. Thoeni

diagnosing and staging a suspected hyperfunctioning low cardiac output or impaired bolus technique. Alter-
endocrine tumor (islet cell tumor) of the pancreas, a natively, some type of bolus tracking technique can be
dual-phase helical CT should be performed because used to optimize timing of the bolus.
these masses are predominantly hypervascular. This Whenever possible, oral contrast material should
protocol is similar to that commonly used for the be administered to avoid confusion of nonopacified
staging of pancreatic adenocarcinoma (TABUCHI et bowel loops with intraperitoneal fluid collections and
al. 1999; BOLAND et al. 1999). Hypervascular tumors to more clearly define the boundaries of the normal
are neoplasms that enhance significantly in the arte- and neoplastic pancreatic parenchyma. In our institu-
rial phase and appear as conspicuous high-density tion, the first cup of contrast material (500 ml of 2%
lesions (IGLESIAS et al. 2001; STAFFORD JOHNSON et al. iodinated contrast agent) is given 45 min before the
1998). In the portal venous phase, these tumors may examination together with an oral tablet of metoclo-
be isodense to the remaining pancreatic parenchyma pramide (Reglan, 10 mg,AH Robins, Philadelphia, PA)
(HOLLETT et al. 1995) and therefore could be missed, to facilitate filling of the distal small bowel and right
particularly if they are small. Large lesions can be par- colon (THOENI and FILSON 1988). The same amount
tially hypo dense in this phase. A dual-phase protocol of contrast agent is given 30 min before the start of the
is also needed for the liver as hepatic metastases have CT examination, which fills the proximal ileum and
the same enhancement characteristics as the primary jejunum. Finally, a cup of water is administered imme-
hypervascular tumor. diately before scanning to assure optimal filling of the
Using a single-row detector scanner and a dual- stomach and duodenum with a low-contrast medium
phase helical CT protocol of the pancreas, we initi- for better definition of the gastric and duodenal wall.
ate scanning with a 20 s delay. We obtain scans with With the advent of helical CT, 3D reconstruction of
3-5 mm slice thickness through the pancreas starting the celiac axis, superior mesenteric artery, and supe-
at the bottom (level of third portion of duodenum) rior mesenteric vein and their branches has become
and extending cranially to the dome of the liver. possible and has the potential of increasing the accu-
Contrast material is administered as a rapid bolus racy for staging pancreatic neoplasms (HONG and
(150 ml delivered at a rate of 3-4 mlls). We then FREENY 1999). Usually, any vascular findings can be
repeat this series at 55-65 s and start at the dome of identified on axial images, but surgeons often prefer
the diaphragm and continue through the pancreas the 3D display for surgical planning. It reduces the
and the remainder of the abdomen. Scans through number of images that need to be assessed. For the
the pancreas should be obtained in one breathhold best results, curvilinear reconstructions should be
with a second breathhold for the rest of the liver, and used. If needed, 3D images of the pancreatic and bili-
finally another one for the entire abdomen. The first ary duct can also be obtained (ZEMAN et al. 1995).
phase in the pancreas serves to define the arterial
anatomy and to detect hypervascular lesions in the
pancreas and liver, and the second phase serves to
define hypovascular lesions and parenchyma in the
pancreas, liver, and the rest of the abdomen.
With the multislice or multirow helical CT scanner,
a dual- or triple-phase study is used (McNULTY et al.
2001). The prospective slice thickness is thin (e.g., 2.5
mm on a GE LightSpeed scanner; General Electric,
Milwaukee, WI). For a dual-phase protocol, the first
scan series is obtained through the pancreas with a
scan delay of 40 s and a high pitch (e.g.,6:1) (Fig.ll.l).
The injection rate is increased to 4-5 mlls. The second
scan series is obtained from the diaphragm to the
symphysis with a scan delay of 60 s, a slice thickness of
3.75 mm, and a pitch of 6:1 or 3:1. With a triple-phase
protocol, scan series with a scan delay of 20, 40, and
60 s are obtained. The triple-phase protocol has the Fig. 11.1. Arterial phase of a dual-phase computed tomogra-
phy (CT). The late arterial phase shows an enhancing lesion in
advantage of demonstrating a hypervascular lesion the body of the pancreas measuring 1 cm in diameter (arrows).
on late arterial images (40 s) if the early (20 s) arterial The clinical picture suggested an insulin om a, and the lesion
phase did not demonstrate an enhancing lesion due to was surgically removed. L, liver; SP, spleen
Endocrine Hyperfunctioning Tumors 179

11.2.2 were available, which were relatively slow (4-6 min


Appearance of the Pancreas on CT for Tl-weighted and 13-16 min for T2-weighted
sequences). Artifacts from breathing often degraded
In the early or late arterial phase, hyperfunctioning these sequences. Fat suppression proved to be helpful
endocrine tumors of the pancreas appear as hyper- in eliminating some of the ghosting artifacts from
vascular masses that are round or oval in shape. respiration produced by the high signal intensity
Rapid washout is usually seen in the portal venous of fat on spin-echo sequences. For fat-suppression
phase. In lesions measuring 2 em or less, enhance- techniques, the triglycerides in an image volume
ment is usually homogeneous, but larger lesions may are initially excited and the resulting transverse
appear heterogeneous. Larger lesions tend to have magnetization of lipids dephased through a spoiling
areas of necrosis that can be quite large depending gradient pulse (MITCHELL et al. 1991). The remain-
on the overall size of the lesion. Hepatic metastases ing water magnetization is then excited to obtain
appear similar to the primary lesion in the pancreas. an image dominated by water. Because in spin-echo
Secondary phenomena such as bile duct dilation or techniques most signals are obtained from fat, sup-
vascular compromise may occasionally be seen. pression of this signal allows the receiver-gain to be
increased and the dynamic range to be improved.
Differences between tissues can thus be enhanced.
This latter effect is used for imaging the pancreas
11.3 with Tl-weighted breathhold sequences. For the
Imaging of the Pancreas with MR Imaging pancreas, fat suppression appears to be more useful
for imaging with Tl- than with T2-weighted series
Currently, magnetic resonance (MR) imaging is not (THOENI et al. 2001). Therefore at UCSF, we use fat
as widely used as CT for imaging hyperfunctioning suppression only for Tl-weighted sequences. With
endocrine tumors of the pancreas. This is in part the introduction of fast spin-echo, single-shot fast
due to the speed of the CT examination and its spin-echo, and gradient-recalled echo sequences,
excellent image resolution and in part due to the faster imaging and breathhold techniques became
ease with which reformations and reconstructions available that improved results.
in multiple planes can be obtained with multirow
helical CT. Nevertheless, results with MR imaging
of the pancreas are getting better. Intravenous 11.3.2
contrast material and fast Tl-weighted as well as Fast MR Imaging
T2-weighted sequences with breathholding have
improved MRI results in the pancreas. In addition, 11.3.2.1
MR cholangiopancreatography (MRCP) permits Fast Spin-Echo and Single-Shot Fast Spin-Echo
accurate assessment of the biliary and pancreatic Sequences
ducts. Presently in the USA, only gadopentetate
dimeglumine is approved for clinical use in assess- Fast spin-echo (FSE) and single-shot fast spin-
ing the pancreas; it acts similarly to iodinated con- echo (ssFSE) sequences are the main scan series
trast material for CT. Some reports indicate that used currently for imaging the pancreas with
mangafodipir trisodium (Teslascan, Mangafodipir T2-weighting (OUTWATER and MITCHELL 1996).
or Mn-DPDP, Amersham Health AS, Princeton, NJ) These fast sequences offer the advantage of rapid
also enhances the pancreatic tissue and postulate scanning with reduced or absent ghosting artifacts
that this enhancement could be used for demon- from breathing. FSE is a fast scanning method that
strating pancreatic pathology better (GEHL et al. uses spin-echoes and altered k-space filling. It is
1993). More work is needed in this area. designed to provide long TR/long TE images in scan
times that are about four to six times shorter than
those obtained with conventional spin-echo tech-
11.3.1 niques. For example, with FSE, using a TR of 4000
Spin-Echo Sequences ms and a TE of -100 ms, 18 slices can be obtained
during quiet breathing in 4.0 - 4.5 min depending
Imaging of the pancreas is performed with Tl- on the gradient performance and coils used. This
and T2-weighted sequences (TSCHOLAKOFF et al. technique is now mostly used with respiratory
1987). For a long time, only spin-echo sequences triggering, which further reduces artifacts from
180 R. F. Thoeni

breathing. ssFSE sequences are obtained with a of the contrast agent by the pancreatic tissue and
single 90 deg excitation pulse, and k-space filling is may possibly be caused by liberation of manganese
achieved through fast data sampling by using high from the complex. The exact mechanism is unknown.
receiver bandwidth and rapid switching of gradi- The advantage of this contrast agent is the fact that
ents. These breathhold sequences typically have the imaging window is much wider, and therefore, it
an infinite TR, a TE of 90-180 ms and last about appears more practical than gadopentetate if further
15-25 s. With axial and coronal ssFSE, images of studies confirm its usefulness. However, studies with
high quality can be obtained during breathhold- Mn-DPDP published thus far have only addressed its
ing. For the MRCP portion of the study, coronal use in patients with pancreatic adenocarcinoma.
and oblique coronal sequences without and with
fat suppression are employed. Sequences without
fat suppression provide a tomographic effect and 11.3.3
permit the evaluation of structures other than the Appearance of the Pancreas on MR Imaging
bright, fluid-filled ducts. Usually, the exact site and
cause of an obstruction are readily visible. On fat- On Tl-weighted images, the pancreas has medium
suppressed images with their cholangiographic signal intensity, similar to or slightly less than that of
effect, the location of the obstruction, but not the liver and lower than that of the fat surrounding the
actual cause, is apparent. pancreas. Fat is particularly helpful in the area of the
tail where the dark signal intensity of the pancreas is
77.3.2.2 contrasted against the signal intensity of fat. For the
Gradient-Echo Sequences best visualization of the pancreas on Tl-weighted
sequences, fat suppression (see previous paragraphs)
In recent years, gradient-echo sequences with breath- should be used. In this fashion, the pancreas assumes
holding have gained in popularity because of their a bright signal intensity that facilitates the detection
markedly reduced imaging time and associated of pathology (Fig. ll.2a). On T2-weighted images,
reduced motion artifacts. Gradient recalled echo the pancreas again demonstrates a signal intensity
sequences (GRE) demonstrate lower tissue contrast similar to that of the liver. T2-weighted images are
and, therefore, fat suppression, and administration of usually obtained without fat suppression for better
an intravenous bolus of gadopentetate dimeglumine definition of the remaining structures in the abdo-
is often needed for better definition of a mass in the men (Fig. ll.2b). The low signal intensity of the
pancreas (GABATA et al. 1994). This is more appli- splenic, mesenteric, and portal veins can be used as
cable to pancreas adenocarcinoma than to endocrine landmarks to identify the pancreas, and distension
tumors of the pancreas. These sequences offer strong of the stomach and duodenum with water can help
Tl-weighting, good temporal resolution for dynamic to define the head of the pancreas. In patients with
studies, and absence of respiratory artifacts. Rapid fatty involution of the pancreas, the signal intensity
bolus technique and short scan delays produce of this organ increases according to the amount of fat
improved tissue contrast between the non enhanced present within the parenchyma. However, on fat-sup-
or minimally enhanced pancreatic parenchyma pressed sequences, the pancreatic bed appears as an
and the markedly enhanced hyperfunctioning islet area of very low signal intensity. The surface of the
cell tumors. However, the imaging window for the normal pancreas is smooth or lobulated depending
pancreas using gadolinium is very narrow (less on the amount of fatty involution.
than 1.5 min and optimal for the arterial phase only The pancreatic duct appears as a low-signal inten-
between 20 and 35 s) due to the rapid enhancement sity tubular structure on Tl-weighted sequences
and washout effect of this compound in the pancreas and as a high-signal intensity structure on heavily
as well as in hypervascular metastases to the liver. T2-weighted scan series (Fig. ll.2c). During MRCP,
Similar to contrast-enhanced helical CT, enhance- heavy T2-weighting and fat suppression provide a
ment with gadolinium should be obtained in the cholangiogram effect where the pancreatic and bili-
arterial (scan delay 20 s) or late arterial (40 s) phase ary ducts appear similar to the images obtained with
(SEMELKA et al. 1993; THOEN! et al. 2000). Mn-DPDP endoscopic retrograde cholangiopancreatography
has been shown to increase the signal intensity of the (ERCP). On MRCP images without fat suppression,
pancreas and to improve the conspicuity of focal pan- both the bright duct and the dark parenchyma of the
creatic lesions (DIEHL et al. 1999; GEHL et al.1993).Itis pancreas are well visualized against a background of
assumed that this enhancement represents an uptake relatively bright fat.
Endocrine Hyperfunctioning Tumors 181

a b

Fig. 11.2a-c. Magnetic resonance (MR) image of an insuli-


noma. a Tl-weighted gradient recalled echo sequence with fat
suppression shows a low-signal intensity lesion in the head of
the pancreas (arrows) representing an insulinoma. A, aorta. b
T2-weighted fast spin-echo sequence demonstrates the lesion
(arrows) as a small high-signal intensity mass. L, liver. c T2-
weighted single-shot fast spin-echo sequence in the coronal
plane shows the same lesion (arrows). Note the normal main
pancreatic duct (PD). L, liver; ST, stomach c

11.4 is suspected clinically, a more focused examination


Imaging of the Pancreas with Ultrasound with endoscopic ultrasound is usually more appro-
priate if an ultrasonographic examination is desired
Correct interpretation of imaging and sampling (ROSCH et al. 1991). However, one has to bear in mind
methods depends on the expertise and skills of the that a complete evaluation of the abdomen is better
radiologist performing these procedures. This is obtained with CT in these cases.
particularly true for modalities such as ultrasound
(transabdominal, endoscopic, or intraoperative
ultrasound) or vascular procedures (arteriography, 11.4.1
portal venous sampling, or arterial stimulation and Endoscopic Ultrasound of the Pancreas
venous sampling) where the performance is highly
operator-dependent. Even if one of these procedures Patients undergoing endoscopic ultrasound are
is highly recommended, it serves little purpose if asked to fast starting at midnight of the night before
performed without the requisite expertise. Such the procedure. After the usual preprocedural inter-
limitations obviously become significant only in view, the relevant patient history and informed con-
cases where the pancreatic lesion is small or when sent are obtained. Intravenous access is established,
an appropriate differential diagnosis is needed. and following routine pre-endoscopic procedures,
Transabdominal ultrasound is only successful when the patient is sedated for the examination and vital
a pancreatic lesion is large enough to be visualized signs monitored.
and if the patient's body habitus or gas in bowel The actual endoscopic procedure is divided
does not prevent a complete examination of the into three parts. Initially, a preliminary endoscopic
pancreas (HESSEL et al. 1982). When the presence of examination of the upper gastrointestinal tract is
a hyperfunctioning endocrine tumor of the pancreas performed, and any abnormalities are noted. Then
182 R. F. Thoeni

the endoscopic examination is carried out. The in a single plane should be viewed with great cau-
conventional endoscope is removed and the endo- tion, as anatomic variations of the pancreas are
sonographic scope is introduced with a water-filled considerable. Commonly, the texture of the body of
balloon in place at its tip. The scope is advanced as the pancreas and the overlying left lobe of the liver
far into the duodenum as possible, and the duode- are compared. The fibrous pancreas usually shows
num, ampulla, and pancreas are visualized while the higher echogenicity than the liver. However, in many
instrument is slowly withdrawn into the duodenum instances, the texture of these two organs is identical.
and stomach. At times, difficulty can be encountered Numerous factors may influence the appearance of
in entering the duodenum, if peristalsis is excessive the pancreas including the state of hydration, fatty
or because of poor patient compliance. Muscle relax- involution, or fasting. Also, in many instances, the
ant and increased sedation may be needed. Ultraso- liver is not normal, which renders a comparison
no graphic images of the various portions of the pan- of the pancreas to the liver even more difficult.
creas obtained with the high-frequency probe can While vessels such as the splenic vein can be used
be recorded (SCHUMACHER et al. 1996). If necessary, as landmarks, it is often impossible to distinguish
biopsies are performed through a conventional endo- the contour of the gland from surrounding tissues
scope if a mass is present (GRESS et al. 2002).At times, in the retroperitoneum. Also, the depth of penetra-
it may not be possible to completely evaluate the tail tion is limited with ultrasound, and retroperitoneal
of the pancreas if it is located beyond the reach of the extension of disease such as invasive tumor cannot
endosonography probe (HAYAKAWA et al. 2000). be accurately assessed.

11.4.2
Intraoperative Ultrasound of the Pancreas 11.5
Somatostatin-Receptor Imaging
Similar to endoscopic ultrasound, intraoperative of the Pancreas
ultrasound (lOUS) permits placement of the ultra-
sonographic probe very close to the area of interest, 11.5.1
unimpeded by fat, bone, or gas. Because of the short Techniques of Somatostatin-Receptor Imaging
distance of the probe from the pancreas, a high-
frequency transducer (7.5 or 10 MHz) can be used Somatostatin is a 14-amino-acid peptide hormone
which provides images with much greater spatial found on many cells of neuroendocrine origin. It
resolution than those obtainable in a transabdominal acts as a neurotransmitter in the central nervous
ultrasound study. system. Hormonally, it inhibits the release of growth
For IOUS, the pancreas is exposed, the head hormone, insulin, glucagon, and gastrin when it
mobilized, and the lesser sac opened. The peritoneal binds to cells. Somatostatin receptors have been
cavity is then filled with warm saline. The transducer demonstrated on the surface of human tumor cells
is enclosed in a sterile plastic sheath or drape and which includes cells with amine precursor uptake
the pancreas scanned in transverse and longitudinal and decarboxylation (APUD) properties such as pitu-
planes. Scanning is performed with the transducer itary tumors, endocrine pancreatic tumors (REUBI
held approximately 1 cm from the surface of the pan- et al. 1990), carcinoids, paragangliomas, small-cell
creas. This permits the visualization of superficial lung cancers, medullary thyroid carcinomas, and
lesions (GUNTHER et al. 1985). pheochromocytomas. Other non-APUD cells may
also bear somatostatin receptors, such as activated
lymphocytes, astrocytomas, and some breast carci-
11.4.3 noma. Animal studies have shown that somatostatin
Appearance of the Pancreas on Ultrasound analogues may inhibit the growth of many of these
tumors in vivo.
The normal dimensions of the pancreas are difficult Analogues of somatostatin were developed because
to establish and vary significantly among patients. human somatostatin has a very short half-life in
Some authors have accepted 3.5 cm as the maximal circulation (2-3 min) and is easily broken down
normal diameter of the head and tail of the pancreas, by endogenous peptidases (RENS-DoMIANO and
with smaller dimensions for the body (DE GRAAFF et REISINE 1992). The analogues preserved two impor-
al. 1978; HABER et al. 1976). Measurements obtained tant molecular features of somatostatin: its cyclic form
Endocrine Hyperfunctioning Tumors 183

and the four amino acids involved in the binding to pheochromocytoma) compared 1-123-Tyr3-octreo-
the receptor. One somatostatin analogue that has been tide and [In-111-DTPA-D-Phel]-octreotide scans and
studied extensively in vitro and in vivo is octreotide demonstrated a more rapid clearance of the former
(Sandostatin, Novartis Pharmaceuticals, East Hanover, (KRENNING et al. 1992). However, the iodine-labeled
N.J.). It also has been used as hormonal treatment in compound had a higher background because of deg-
patients with carcinoid syndrome. radation products and a higher intestinal background
Using a radiolabeled somatostatin analogue, because of biliary excretion. There appeared to be an
octreotide, tumors with somatostatin receptors can overall higher sensitivity of the indium-labeled com-
be detected by scintigraphy or positron emission pound in this small series. While results with indium-
tomography (PET) (KRENNING et al. 1992; LAM- 111-pentetreotide appear promising, insufficient data
BERTS et al. 1990). The incidence of somatostatin are currently available to conclusively compute the
receptors in hyperfunctioning endocrine tumors sensitivity, specificity, or accuracy (ELLISON et al.
of the pancreas was shown by in vivo scintigra- 1997; SCHIRMER et al. 1995). For the accurate inter-
phy with octreotide to be 100% for gastrinomas, pretation of the images, the normal tracer distribution
61% for insulinomas, 100% for glucagonomas, and needs to be understood, and knowledge of lesions
89% for unclassified APUDomas (REUBI et al. 1990; that are expected to accumulate the tracer is essential.
WEINEL et al. 1993). The numbers are generally More recently, other radioisotopes including Cu-64
slightly higher for in vitro receptor status. Failure to have been used for PET imaging and comparison to
demonstrate somatostatin receptors in some insuli-
nomas may be due to the existence of somatostatin
receptor subclasses that show high affinity binding
sites for native somatostatin but not for octreotide.
Somatostatin analogues also may be used therapeu-
tically to ablate lesions with somatostatin receptors
through radiation (UGUR et al. 2002). This method
is somewhat limited because many endocrine
tumors are quite small and located deep within the
abdomen.
In vivo studies with the radioiodinated (1-123)
derivative of octreotide have demonstrated the
visualization of somatostatin receptor-positive
tumors within minutes after the administration
of the tracer. However, 1-123 is expensive, not uni-
versally available, time consuming to prepare, has
a short half-life which does not allow for delayed
imaging, and its marked biliary excretion can
render interpretation of abdominal images dif-
ficult due to the large amounts of tracer in the
bowel. To avoid these problems, [In-ll1-DTPA-
D-Phe1]-octreotide was prepared (BAKKER et al.
1991; KRENNING et al. 1992). It was found to have a
high affinity for somatostatin receptors and similar
biological properties to octreotide. The compound,
also called OctreoScan, is easily labeled with In -Ill
(Fig. 11.3). Since this radiotracer is mainly elimi- Fig. 11.3. In-111-octreotide study of gastrinoma. The views
on the left demonstrate standard SPECT slice images in axial,
nated via the kidneys, intra-abdominal evaluation anteroposterior, and coronal planes. A lesion (thick arrows)
of somatostatin-receptor positive tumors could be is visualized adjacent to the upper pole of the kidney (thin
performed (DUNCAN et al. 1997). Usually, indium- arrows) on the right side. The anterior view of the whole body
111-0ctreoscan planar and SPECT images are on the right again shows the lesion (thick arrows) that proved
obtained at 4, 24, and 48 h following the injection of during endoscopic surgery to be located along the upper
common bile duct close to the origin of the cystic duct. CT
6.3 mCi of Octreoscan. and MRI did not visualize the gastrinoma. The patient has
One small series involving 6 patients (small-cell been symptom-free since the procedure. B, bladder (courtesy
lung cancer, gastrinoma, insulinoma, carcinoid, and of Dr. Robert Lull, SFGH Nuclear Medicine Services)
184 R. F. Thoeni

In -111-diethylenetriaminepenta-acetic acid (DTPA)- 11.6


D-Phel-octreotide showed favorable results (ANDER- Other Radiologic Diagnostic Tests
SON et al. 2001; DECRISTOFORO et al. 2000). However,
the small number of patients examined again does not 11.6.1
allow for definite conclusions. Arteriography of the Pancreas
Somatostatin-receptor imaging can be a useful
technique for the diagnosis of many tumors of Before noninvasive cross-sectional imaging methods
neuroendocrine and non-neuroendocrine origin. were introduced, arteriography was the principal
A positive finding may be predictive of the abil- technique for localizing these hypervascular endo-
ity of octreotide to suppress the neuroendocrine crine tumors of the pancreas. This technique includes
tumors. A lack of human antibody response allows selective arteriography of the proper or common
for repeated administration, and the entire body can hepatic artery, the gastroduodenal, splenic, superior
be imaged. Based on somatostatin-receptor imaging, mesenteric, and at times the dorsal pancreatic artery.
patient management decisions are more informed, The arteriographic appearance of all islet cell tumors
the therapy selection optimized based on tumor is similar. It is not possible to distinguish a functioning
biochemistry, and the success of the therapy readily from a nonfunctioning tumor or one type of function-
monitored. ing tumor from another. The marked hypervascularity
and intense homogeneous staining permit the distinc-
tion of these types of tumors from other tumors such
11.5.2 as pancreatic adenocarcinoma (DOPPMAN et al. 1990)
Appearance of the Pancreas on Somatostatin- (Fig. 11.4). Neovascularity and venous involvement
Receptor Imaging can often be demonstrated in larger lesions. The lower
limit of detectability of functioning islet cell tumors by
An 1-123-Tyr3-octreotide or In-111-pentetreotide arteriography is 5 mm (GUNTHER et al. 1985).
scintigram typically obtained at 4 and 24 h after
injection shows areas of increased tracer localization
in a hyperfunctioning endocrine mass or masses of 11.6.2
the pancreas. Other abnormal foci of uptake may be Portal Venous Sampling
present throughout the liver, adrenal gland, lymph
nodes, or bone consistent with metastases. Lesions Portal venous sampling (PVS), also called pancreatic
located adjacent to the liver and spleen may be venous sampling or transhepatic venous sampling,
missed because of the adjacent normal uptake. involves catheterization of the portal vein using a

a b

Fig. 11.4a,b. Arteriography of a glucagonoma. aArteriogram demonstrates a large enhancing lesion in the tail of the pancreas
(arrows). The clinical picture and surgery proved the presence of a glucagonoma. SA, splenic artery. Same patient as Fig. 11.11 a.
b The venous phase of the same patient shows a persistent blush (arrows). SV, splenic vein; PV, portal vein (courtesy of Dr.
Ernie Ring, UCSF)
Endocrine Hyperfunctioning Tumors 185

transhepatic approach and is not an imaging study by gastrinoma, glucagonoma, VIPoma, somatostati-
(VINIK et al. 1990). The branches of the extrahepatic noma, and other rarely encountered secretory tumors
portal venous system are selectively catheterized and of the pancreas. In functioning adenomas, the clinical
blood samples obtained. The hormone concentration data and laboratory tests often permit an accurate
in each sample is plotted on a diagram of the portal clinical diagnosis, but in most cases, cross-sectional
venous system. The sample with the highest concen- imaging can localize the tumor in the pancreas.
tration comes from the vein that drains the area of the Thus, radiography confirms a clinically suspected
tumor. The sensitivity of the procedure depends only lesion and can guide the surgeon to the appropriate
on hormone output by the tumor and not on the tumor location for tumor resection. Localization by imaging
size (VINIK et al. 1991). An extrapancreatic location of and treatment of patients with multiple endocrine
tumor such as seen in gastrinomas renders this test less adenomatosis (MEA) or multiple endocrine neo-
reliable for cure as the exact location of the tumor(s) plasia (MEN) are more difficult, and even venous
may not be established. This method requires thor- sampling does not always reveal the location of
ough knowledge of the anatomy of the portal venous additional tumors if the primary site is found in the
system. It also carries the disadvantages of being time pancreas. In MEN, islet cell tumors may be ectopic in
consuming, necessitating the administration of large location, such as in the duodenal or gastric wall, may
amounts of contrast material, exposure to a significant be multiple, and tend to be malignant. Many hyper-
radiation dose for patient and radiologist, and risk of functioning endocrine tumors of the pancreas may
serious complications (MILLER et al. 1992). be associated with MEN. MEN type I is an inherited
disease with an autosomal dominant trait and high
penetrance. It is a rare disease, and neoplasms involve
11.6.3 the pancreas, adrenal cortex, parathyroid, thyroid,
Arterial Stimulation and Venous Sampling and pituitary glands. The symptomatology depends
on the organs involved.
For this method, the tumor is stimulated to secrete
hormones by a drug called secretagogue, and samples
are obtained from the right and left hepatic veins 11.7.1
(DOPPMAN et al. 1990; IMAMURA et al. 1989). This Insulinoma
technique is done in conjunction with pancreatic
arteriography. Venous samples are obtained at 0.5, Insulinomas are generally small, measuring less than
1, and 2 min after each injection of secretagogue. 2 cm in diameter. This is due to the fact than even
If the hormone concentration increases twofold, the small tumors produce sufficient amounts of insulin to
arterial supply to the tumor can be identified and cause symptomatic hypoglycemia. Larger lesions or
therefore the region where the tumor is located. If the calcifications in the tumor mass suggest malignancy.
hormone concentration increases after the drug has Most insulin om as are solitary lesions, and 90%-95%
been injected into the proper hepatic artery, the pres- are benign. In the rare instance of malignant degen-
ence of hepatic metastases can be diagnosed. This eration of an insulinoma, the appearance of liver
technique is easier to perform than PVS and can be metastases is similar to that of the primary tumor,
done at the same time as the arteriogram, thereby and this feature is shared by all hyperfunctioning islet
avoiding an additional invasive procedure. Also, the tumors. The likelihood of having multiple tumors in
complication rate with this technique is lower than the pancreas is significantly increased in patients
with PVS. Today, this test is only used when morpho- with MEN type I, and presentation at a young age
logic studies are negative (PEREIRA et al. 1998). should suggest this syndrome. Patients with a spo-
radic insulinom a are predominantly women (60%)
and present in their fifth or sixth decade.
On CT, these hypervascular tumors appear as
11.7 brightly enhancing, round to oval masses that dem-
Features and Results of Imaging onstrate rapid washout on the portal venous phase
Hyperfunctioning Endocrine Tumors (Fig. 11.5). Over two-thirds are located to the left of
of the Pancreas the superior mesenteric artery (HOWARD et al. 1990).
On MR imaging, islet cell tumors of the pancreas are
Among the islet cell tumors of the pancreas, insulin- oflow signal intensity on Tl-weighted fat-suppressed
oma is the most common endocrine tumor, followed images and of high signal intensity on T2-weighted
186 R. F. Thoeni

a b

Fig. ll.5a,b. CT of an insulinoma. a A 1.5 cm lesion is seen in the neck of the pancreas which reveals marked enhancement
(black arrows) in the arterial phase. Of incidental note is a lesion in the medial segment of the left hepatic lobe with some
peripheral enhancement (white arrow). b The portal venous phase demonstrates rapid washout of the lesion (black arrows).
The liver lesion (white arrow) now demonstrates features of a hemangioma

images (LIESSE et al. 1992; MOORE et al. 1995). Occa- multirow helical CT have markedly improved the
sionally, an insulinoma can be of dark signal intensity detection of these small lesions compared with
on T2-weighted sequences due to the presence of conventional CT. This is due to faster scanning times
fibrous stroma (MORI et al. 1996) (Fig. 11.6a,b). This with earlier scan delays, super-thin slices and con-
was seen in 3 of 20 patients in our study (THOENI centrated bolus techniques that demonstrate these
et al. 2000). Some of the insulinomas show marked lesions well. This has been shown anecdotally in a
enhancement with gadolinium throughout the lesion, small series of patients, but presently no data in a
and some may demonstrate ring-like peripheral large group are available in the literature to prove
enhancement (KRAUS and Ros 1994). The presence of this point (VAN HOE et al. 1995; CHUNG et al. 1997;
fibrous tissue diminishes the degree of enhancement KING et al. 1998). One of those studies demonstrated
with gadolinium (Fig. 11.6c). a sensitivity of 82% (VAN HOE et al. 1995).
Transabdominal ultrasound and endoscopic We collected a series of 20 patients with small
ultrasound visualize the islet cell tumor as a rela- functioning adenomas which were missed by ultra-
tively hypo echoic area compared with the adjacent sonography and CT but readily detected by Tl-
pancreas, with smooth and at times slightly irregular weighted fat-suppression sequences (THOENI et al.
margins that are well defined. Intraoperative ultra- 2000). In our study, we achieved a MRI sensitivity of
sound can be used to confirm lesions identified prior 85% for detecting functioning islet cell tumors of 2
to surgery and detect small lesions that eluded detec- cm or less in diameter, which is similar to the sensi-
tion on other imaging studies. Octreotide studies tivity achieved by invasive procedures and is superior
demonstrate a hyperfunctioning endocrine tumor as to most of the MRI results reported in the literature.
a focal area of hyperactivity. On an arteriogram, insu- Some MRI series reported a sensitivity of 100%,
linomas appear as a round, intensely staining tumor but the number of patients examined was small
without visible vessels and with smooth borders. This (SEMELKA et al. 1993). It is very likely that MRI would
appearance is seen in functioning and nonfunction- detect lesions larger than 2 cm with a sensitivity of
ing tumors, and from the arteriogram it cannot be greater than 85%. While the specificity and positive
predicted what hormones the tumor is secreting. predictive value of MRI in our study were excellent
Neovascularity and portal vein invasion indicate that (each 100%), the negative predictive value was only
the tumor is malignant. 73%. However, these values should be viewed with
Based on results available from the literature, the caution, as the number of patients without tumors
sensitivity of nonhelical CT for determining the pres- (n=8) was relatively small in our series.
ence of an insulinoma ranges from 28% to 79%, with The superior MRI results in our study are based on
a mean of 38% (PAVONE et al. 1993; ASPESTRAND et improved sequence design and better gradients and
al. 1993). Single-row helical CT and more recently coils which enable faster imaging sequences, higher
Endocrine Hyperfunctioning Tumors 187

c _ __ ~

Fig.l1.6a-d. MR of an insulinoma. a Fat-suppressed Tl-weighted sequence demonstrates a low-signal intensity lesion (arrows)
in the body of the pancreas. ST, stomach (reprinted with permission from THOEN! et al. 2001). b The fast spin-echo sequence
demonstrates the same lesion as a low-signal intensity mass (arrows). The lack of bright signal is due to the fact that fibrous
stroma was present within the lesion. (Reprinted with permission from THOEN! et al. 2001). c Gadolinium-enhanced Tl-
weighted sequence demonstrates some enhancement (arrows) but not as pronounced as in Fig. ll.S due to the fibrous ele-
ments in the lesion. P, pancreas. d Insulinoma. The surgical specimen showed a well-defined lesion (arrows) in the body of the
pancreas. The patient has remained symptom-free since surgery

signal-to-noise ratios, and superior imaging of early poor evaluation of the tail of the pancreas (sensitiv-
gadolinium enhancement. The fact that the two read- ity 38%), because of its distance from the stomach
ers agreed on the diagnosis in 82% of cases (K=0.61) (SCHUMACHER et al. 1996). Therefore, it is of limited
shows that the detection of hyperfunctioning endo- use in this area. Another study that assessed the
crine tumors of the pancreas measuring <2 cm in use of the various imaging methods for detecting
diameter is reliably achieved by MRI. Nevertheless, insulinomas and gastrin om as found a sensitivity
errors in diagnosis occurred slightly more often with of 90% for endoscopic ultrasound and insulinomas
the less experienced reader. It appears likely that MRI (ZIMMER et al. 1995). This compares to a sensitivity
results in these patients are superior when a seasoned of only 20% for transabdominal ultrasound. For insu-
radiologist with special expertise in abdominal MR linomas, angiography is reported to reach a sensitiv-
imaging evaluates the images. ity between 59% and 80%, while venous sampling
The sensitivity of transabdominal ultrasound for reaches sensitivities between 77% and 94%, with
detecting insulinomas has been reported to range results slightly better for gastrinomas (GUNTHER et
from 19% to 60%, with a mean of 46% (GUNTHER al. 1985; DOHERTY et al. 1991; FRUCHT et al. 1989;
et al. 1985; DOHERTY et al. 1991; MORI et al. 1996). PISEGNA et al. 1993).
Endoscopic ultrasound provides excellent results At the present time, insufficient data are available to
in the head of the pancreas (sensitivity 83%), but determine whether somatostatin-receptor localization
188 R. F. Thoeni

of islet cell tumors with l11In-octreotide scintigraphy associated with MEN type I in 20%-60%. Approxi-
is a reliable method. Accurate numbers for hyper- mately 50% of patients with gastrinom a have metas-
functioning endocrine tumors of the pancreas are tases at the time of diagnosis. Most of these tumors
difficult to obtain from the various published studies are found in the so-called gastrin om a triangle
on radiolabeled somatostatin analogue scintigraphy. formed medially by the junction of head and neck of
These reports usually investigate the sensitivity of this the pancreas, inferiorly by the second and third por-
method in a wide variety of neuroendocrine tumors tion of the duodenum, and superiorly by the junction
including carcinoid, medullary carcinoma of the of the cystic duct and the common bile duct. These
thyroid gland, small-cell lung cancer, and neuroblas- tumors are slow growing and have a better prognosis
tomas. Preliminary data in 24 patients with biologic than pancreatic adenocarcinoma.
and/or histologic evidence of a neuroendocrine tumor Gastrinomas are hypervascular tumors that are
showed that lllIn-octreotide scintigraphy was positive best visualized with thin sections (FRUCHT et al.
in 23, but prospective surgical proof could be obtained 1989; TJON et al. 1989) and a dual-phase CT protocol
in only 8 of these patients (CORLETO et al. 1996). In using an arterial and portal venous phase (PISEGNA
another study of 28 patients with carcinoid and islet et al. 1993) similar to the technique used for insuli-
cell tumors, 22 of 28 were positive, and 3 were negative nomas. Most studies addressed the use of nonhelical
(KVOLS et al. 1993). False-positive and false-negative CT with a mean of over 38% for gastrinomas (ROSSI
results have been reported with this method (WESTLIN et al. 1989). Because of their larger size, helical CT
et al. 1993; ZIMMER et al. 1995). In one study that listed generally readily detects gastrinomas. However, ecto-
results for the various types of neuroendocrine tumors pic locations are more challenging. Metastases to the
separately, the sensitivity of somatostatin-receptor liver tend to have a similar appearance to the primary
scintigraphy for insulinomas was only 10% (ZIMMER tumor (DEBRAY et al. 2001) (Fig. 11.7). For gastrino-
et al. 1995). mas, some reports have shown MRI sensitivities of
Therefore, at present it appears that multirow heli- 20%-62% (PISEGNA 1993; FRucHT et al.1989; TJON et
cal CT and MRI are the best techniques for diagnos- al. 1989). In one study, somatostatin-receptor scintig-
ing small pancreatic insulinom as, but optimal CT raphy showed a sensitivity of 100% for gastrinomas
and MRI techniques and state-of-the-art equipment (ZIMMER et al. 1995), whereas EUS was 90%, and CT,
need to be used. In cases where CT, MRI, and EUS fail MRI, and transabdominal US combined were only
to demonstrate a lesion, radio labeled somatostatin 30%. The scintigraphic sensitivity for gastrinomas
analogue scintigraphy or arteriography could be was much higher than that for insulinomas (see
used to localize a lesion. However, for insulinomas, above). Scintigraphy was also useful in distinguish-
somatostatin-receptor scintigraphy has not proven ing hyperfunctioning endocrine tumors metastatic
to be sufficiently reliable. Arteriography and venous to the liver from hepatic hemangiomas, with an accu-
sampling are infrequently used today because of the racy of 96% (TERMANINI et al. 1997). Overall, EUS
invasiveness, the long procedure time, the special was shown to be cost -effective when compared with a
expertise needed for performance, and the higher control group employing venous sampling (BANSAL
cost of these tests. It is advisable to use intraopera- et al. 1999). Results with angiography vary greatly. In
tive ultrasound at the time of surgical resection (Fig. some studies, arteriography in patients with gastri-
11.6d) for the confirmation of lesions in patients with nomas was reported to achieve a sensitivity of about
a strong clinical suspicion and negative results from 65% (FRUCHT et al. 1989; WISE et al. 1989). At the
cross-sectional images and scintigraphy and/or to NIH, 86% of gastrinomas were correctly identified
find additional tumors. Intraoperative ultrasound with angiography, with a specificity of 100% (MATON
is particularly important in patients with multiple et al. 1987) (Fig. 11.8), but a later study of patients suf-
lesions and MEN as under these conditions, ectopic fering from gastrin om as without and with metastases
tumors that may be multiple are quite frequent and revealed a sensitivity of only 41 % (GIBRIL et al. 1996).
difficult to find with CT or MRI. In the same study, the sensitivity for CT and US was
64% and for MR and angiography, 77% (GIBRIL et al.
1996). In Maton's study (MATON et al. 1987), angiog-
11.7.2 raphy for extrahepatic gastrinomas had a specificity
Gastrinoma of 94% and a sensitivity of 68%, a positive predictive
value of 97%, and a negative predictive value of 53%.
Gastrinomas are usually larger than insulin omas (~.2 Comparison of CT and angiography revealed that
cm), multiple in 60%, malignant in 60%-65%, and for hepatic metastases, CT demonstrated 72% and
Endocrine Hyperfunctioning Tumors 189

a b

Fig. 1l.7a,b. CT of a gastrinoma. a A large enhancing lesion (white arrows) is seen in the head and body of the pancreas. The
common bile duct (CB) is dilated. Multiple liver metastases (black arrows) were present at the time of diagnosis. b In the portal
venous phase, the enhancement pattern of the hepatic metastases (arrows) changes. While in the arterial phase the entire lesion
opacifies, more enhancement is present in the periphery. In the portal venous phase, a reversal has occurred that is typical for
hypervascular metastases to the liver. SP, spleen

a _ _ _ _ _--';;:: b

Fig. 11.8a,b. Angiogram of a gastrinoma. a Arteriogram shows a large enhancing lesion (arrows) in the head of the pancreas
artery (courtesy of Dr. Ernie Ring, UCSF). b The venous phase demonstrates a persistent blush (arrows) in the area of the
pancreatic mass artery (courtesy of Dr. Ernie Ring, UCSF). SP, spleen

angiography 89% of tumors, and the combination of results with cross-sectional imaging and scintigra-
the two techniques detected all tumors with no false- phy, invasive techniques are not currently recom-
positive results. mended for this purpose.
From the available data on imaging of gastrino-
mas, it again appears that CT or MRI (Fig. 11.9) and
endoscopic ultrasound are the primary imaging 11.7.3
modalities. Both CT and MRI have the advantage of VIPoma
staging the entire abdomen and pelvis, which is not
possible with the limited depth penetration of endo- The tumor is usually larger than 3 cm in diameter
scopic ultrasound. In equivocal or negative cases and malignant in 60% or more (KLOPPEL and HEITZ
with a high clinical suspicion, somatostatin-receptor 1988; ROTHMUND et al. 1991; JAFFE 1987). Over two-
scintigraphy is very effective in assessing the primary thirds of the tumors are located in the body and tail
tumor in the pancreas or ectopic sites (Fig. 11.3) and of the pancreas (JAFFE 1987). Occasionally, tumors
metastatic lesions to the liver. Based on the improved producing the same symptomatology are located
190 R. F. Thoeni

a b

Fig. 11.9a,b. MRI of a gastrinoma. a On a Tl-weighted sequence, a large mass oflow-signal intensity (arrows) is seen in the head of
the pancreas. b A fat-suppressed T2-weighted sequence demonstrates the same lesion with high signal intensity (arrows). L, liver

in the adrenal gland, retroperitoneum, sympathetic (Fig. 11.10). A combination of CT or MRI with soma-
chain, lung, and intestinal carcinoids (KLOPPEL and tostatin-receptor scintigraphy currently appears to
HEITZ 1988; JAFFE 1987). Rarely, these tumors are be the most effective presurgical assessment.
associated with MEN type I (KREJS 1987).
Related to the fact that VIPomas are quite rare, sta-
tistical data on the use of imaging are not available. 11.7.4
Case reports or series with all types of hyperfunc- Glucagonoma
tioning endocrine tumors mixed together have been
published. CT, MR, angiography, and somatostatin- Glucagonomas are intrapancreatic and mostly occur
receptor scintigraphy have been used to localize the in the head and neck of the pancreas. The tumor
primary lesion and to identify metastases (SOFKA occurs slightly more often in women and is seen at
et al. 1997; MORTELE et al. 2001; THOMASON et al. a mean age of 55 years (BLOOM and POLAK 1987). In
2000). On MRI, metastatic lesions to the liver may rare instances, the tumor can cause obstructive pan-
show intense peripheral enhancement similar to the creatitis (PECH et al. 2000). The tumor is malignant
appearance of primary and metastatic lesions on CT in about 60%, and the 5-year survival is 50%. When

a b

Fig. 11.10a,b. CT of a VIPoma. a In the arterial phase, a markedly enhancing mass (white arrows) is present in the head of the
pancreas. Multiple liver metastases (black arrows) with a pattern similar to the primary tumor are also seen. b The portal venous
phase at a slightly higher level demonstrates that the mass also involves the body of the pancreas (white arrows). The washout
effect is similar for the pancreatic lesion and the hepatic metastases (black arrows)
Endocrine Hyperfunctioning Tumors 191

the tumor becomes symptomatic, its size usually imaging serves as an adjuvant to conventional imag-
exceeds 5 cm, it is invasive and has metastasized ing (JOHNSON et al. 2000). Current therapeutic appli-
to the regional lymph nodes (Fig. 11.1 1). Tumors cations of octreotide focus on stabilization of the
measuring 3 cm or less are often seen as incidental disease in tumors expressing somatostatin receptors,
findings. and tumor destruction using beta-emitting isotopes.
CT, MR, angiography, EUS, and F-18-fluorodeoxy-
glucose positron emission tomography (PET) have
been used with success, but all reports included only 11.7.5
anecdotal references to this type of tumor, largely Somatostatinoma
due to its rarity (SOLIVETTI et al. 2001; PHAN et al.
1998; ANDERSON et al. 2000; FERNANDEZ-REPRESA Somatostatinomas are usually solitary lesions and
et al. 2000; NISHIGUCHI et al. 2001). PET has been tend to be aggressive, with over 70% presenting
used more recently as a reliable tool to localize the with metastases, particularly if the primary tumor is
primary pancreatic mass and its metastases to the larger than 2 cm (GOWER and FABRI 1990; TANAKA
liver (FERNANDEZ-REPRESA et al. 2000; NISHIGU- et al. 2000) (Fig. 11.12). This tumor is usually located
CHI et al. 2001). Somatostatin analogue (octreotide) within the pancreas, but it may also arise from the
small bowel, the duodenal ampulla, or periampullary
mucosa. In men, the tumor is more frequently seen
in the duodenum, and in women more often in the
pancreas (PATEL et al. 1983). When the small bowel
is involved, the neoplasm is usually a carcinoid that
consists almost completely of somatostatin-contain-
ing cells but produces little somatostatin. If the neo-
plasm is outside the pancreas, it tends to be smaller
(0.5-4 cm). This is probably related to the fact that
it produces symptoms such as jaundice, bleeding,
and ulceration and therefore is discovered early. The
tumor occurs in the fourth to sixth decades of life,
and the prognosis is poor, with an average survival
time of only 1-2 years. The tumor tends to be large
a (2-1O cm in diameter), and 75% occur in the head of
the pancreas (HOWARD et al. 1990).

Fig. 11.11a,b. CT of a metastatic glucagonoma. a The patient is


status postresection of a glucagonoma in the tail and body of
the pancreas as demonstrated by the surgical clips (large white
arrow). Metastases to the liver (small white arrow) and a lymph Fig. 11.12. CT of a somatostatinoma. A very large, enhanc-
node (black arrow) are present. Note the dilation of the con- ing mass with areas of necrosis is demonstrated (large white
trast -filled jejunal loops (arrowheads) that is due to excessive arrows) that involves most of the pancreas. Encasement of the
glucagon in the circulation. b At a lower level, retroperitoneal splenic artery (small white arrows) is also seen. An enhancing
lymphadenopathy (black arrows) and multiple, dilated small- liver metastasis (black arrows) is noted in this female patient
bowel loops (arrowheads) without wall thickening are evident with a very aggressive lesion
192 R. F. Thoeni

The radiologic features of somatostatinomas Very rarely, a parathyrinoma may be present in


resemble those of other neuroendocrine tumors. patients with an islet cell tumor and hypercalcemia,
EUS, CT, MRI, angiography, and somatostatin-recep- but usually these patients have MEN type I with a
tor imaging have been used (ANDERSON et al. 2000; parathyroid adenoma. Parathyrinomas secrete a
SEMELKA et al. 2000; TJON et al. 1994; SCHILLACI et parathyroid hormone (PTH)-like protein (KLOPPEL
al. 1997). All studies reported only a few cases or and HEITZ 1988; FRIESEN 1982; BRESSLER et al. 1991).
included somatostatinomas in their series of neu- The pancreatic tumor is almost always metastatic to
roendocrine tumors. Lesions in the pancreas and the liver at the time of diagnosis. Only 3 cases have
liver are usually well shown. However, radiological been reported so far (SOLCIA et al. 1990).
techniques often fail to demonstrate tumors in the Some endocrine tumors of the pancreas can
duodenum but may show obstruction of pancreatic secrete serotonin and produce the typical features
or biliary ducts as indirect evidence. The diagnosis of a carcinoid tumor. These tumors may be benign
in cases of duodenal localization can be established or have features of a carcinoma with poorly differ-
by endoscopic techniques including biopsy (Guo et entiated cells. Over 50% of patients with this tumor
al. 2001). present with the carcinoid syndrome, but it is often
atypical. Patients tend to suffer from severer flash-
ing, hypotension, periorbital edema, and increased
11.7.6 lacrimation (FRIESEN 1982). Liver metastases do not
Other Hyperfunctioning Endocrine Tumors need to be present for the syndrome to occur. Nev-
ertheless, almost 90% of patients with the syndrome
There are some other hyperfunctioning endocrine have liver metastases (SOLCIA et al. 1990). Small
tumors of the pancreas worth mentioning that pro- tumors are likely to be homogeneous and hypervas-
duce an endocrine syndrome, but they are very rare. cular, whereas larger tumors are heterogeneous and
For each of them, no more than about 20 cases have hypovascular, with areas of cystic degeneration and
ever been reported. Nonfunctioning endocrine tumors necrosis (DAHAN et al. 2001).
belong to the group of islet cell tumors of the pancreas Like all the other hyperfunctioning endocrine
that are clinically not associated with a specific endo- tumors, these rare tumors demonstrate the features of
crine syndrome. Some of them have been reported a hypervascular mass without or with metastases. CT
to be positive on immunohistochemistry for various and MRI are used initially. EUS can be used for the pri-
ectopic hormones (VUOLTEENAHO et al. 1990). These mary tumor but cannot assess the frequent presence of
tumors will be discussed in another chapter. liver metastases. PET and somatostatin-receptor imag-
Corticotropinoma may secrete several different ing are also useful. Angiography and venous sampling
hormones. ACTH-producing pancreatic islet cell are rarely employed today for these rare lesions.
tumors are found in 4%-16% of patients with ecto-
pic ACTH syndrome. Corticotropinoma may also
secrete melanocyte-stimulating hormone (MSH),
corticotropin-releasing hormone (CRH), gastrin, or
insulin (FRIESEN 1982). ACTHomas must be distin-
guished from MEN type I that consists of an islet cell
tumor and an ACTH-secreting pituitary adenoma.
Corticotropin om as are large, malignant, and invari-
ably present with metastases at the time of the initial
diagnosis (DOPPMAN et al. 1989) (Fig.lLl3).
GRFoma secretes a growth hormone-releasing
factor, and patients with this tumor may develop
acromegaly (RIVIER et al. 1982; THORNER et al. 1982;
BERGER et al. 1984) (Table 12.1). Similar to corti-
cotropinoma, this tumor is large (>6 em) and has
metastasized at the time of diagnosis in over 30%
(JENSEN and NORTON 1991; SOLCIA et al. 1990). In Fig. 11.13. CT of an ACTHoma. A very large, heterogeneously
enhancing mass (white arrows) is seen in the pancreas. The
33% of these patients, MEN type I is present (TAKA- biliary ducts (white arrowheads) are dilated due to compres-
HASHI et al. 1988), and 40% also have Zollinger-Elli- sion by the pancreatic mass. Diffuse metastatic disease (black
son syndrome (see gastrinoma above). arrows) is shown in the liver
Endocrine Hyperfunctioning Tumors 193

11.8 Aspestrand F, Kolmannskog F, Jacobsen M (1993) CT, MR


Role of Imaging for Hyperfunctioning imaging and angiography in pancreatic APUDomas. Acta
Endocrine Tumors Radiol 34:468-473
Bakker WH, Krenning EP, Breeman WA, Kooij PP, Reubi JC,
Koper JW, de Jong M, Lameris JS, Visser TJ, Lamberts
Hyperfunctioning endocrine tumors of the pancreas SW (1991) In vivo use of radio iodinated somatostatin
represent a challenge to the radiologist. Earlier reports analogue: dynamics, metabolism, and binding to soma-
have shown that up to 27% of patients have tumors not tostatin receptor-positive tumors in man. J Nuc Med 32:
1184-1189
detected preoperatively. Helical CT and MR with fast
Bansal R, Tierney W, Carpenter S, Thompson N, Scheiman JM.
imaging and short breathhold techniques have mark- (1999) Cost effectiveness of EUS for preoperative localiza-
edly improved the preoperative results. Endoscopic tion of pancreatic endocrine tumors. Gastrointest Endosc
ultrasound often achieves sensitivities that approach 49:19-25
90%-95%, but lesions in the tail of the pancreas are Berger G, Trouillas J, Bloch B, Sassolas G, Berger F, Partensky
C, Chayvialle JA, Brazeau P, Claustrat B, Lesbros F (1984)
less well seen, and complete assessment of metastatic
Multihormonal carcinoid tumor of the pancreas. Secreting
disease cannot be achieved with this technique. Extra- growth hormone-releasing factor as a cause of acromegaly.
pancreatic islet cell tumors that are small and located Cancer 54:2097-2108
in the duodenal wall or other ectopic locations are Bloom SR, Polak JM (1987) Glucagonoma syndrome. Am J
the tumors least likely to be detected preoperatively Med 82:25-36
Boland GW, O'Malley ME, Saez M, Fernandez-del-Castillo C,
by any of the radiographic techniques, and even
Warshaw AL, Mueller PR (1999) Pancreatic-phase versus
intraoperative ultrasonography fails to detect some portal vein-phase helical CT of the pancreas: optimal
of these lesions. Overall, arteriography and venous temporal window for evaluation of pancreatic adenocar-
sampling are infrequently used today, even for these cinoma. AJR Am J Roentgenol 172:605-608
difficult cases, because of the inherent technical Bressler L, Boissel P, Conroy T (1991) Pancreatic islet cell
problems associated with the test and the higher cost. cafrcinoma with hypercalcemia: complete remission 5
years after surgical excision and chemotherapy. Am J Gas-
More recently, somatostatin-receptor scintigraphy troenterol 86:635-638
and PET have been used to establish or confirm the Chung MJ, Choi BI, Han JK, Chung JW, Han MC, Bae SH (1997)
presence of ectopic lesions or small masses that were Functioning islet cell tumor of the pancreas. Localization
only suspected on CT or MRI and to improve results with dynamic spiral CT. Acta Radiol 38:135-138
in assessing metastatic disease. However, while soma- Corleto VD, Scopinaro F, Angeletti S, Materia A, Basso N,
Polettini E, Annibale B, Schillaci 0, D' Ambra G, Marig-
tostatin-receptor scintigraphy is superb in evaluating nani M, Gualdi G, Bordi C, Passaro EJ, Delle Fave G (1996)
patients with gastrinomas, it is not sensitive in patients Somatostatin receptor localization of pancreatic endocrine
with insulinomas. Except in patients with insulinomas, tumors. World J Surg 20:241-244
these two low-resolution techniques should be used as Dahan H, Soyer P, Cochand-Priollet B, Abitbol M, Coumbaras
J, Pelage JP, Boudiaf M, Rymer R (2001) Imaging of primary
adjuvant to the other imaging techniques. A combi-
carcinoid tumor of the pancreas. J Radiol 82:987-990
nation of intraoperative palpation and intraoperative De Graaff CS, Taylor KJ, Simonds BD, Rosenfield AJ (1978)
ultrasonography was found to achieve the best results Gray-scale echography of the pancreas. Re-evaluation of
due to the complementary nature of these two tech- normal size. Radiology 129:157-161
niques during surgery. Somatostatin-receptor tech- Debray MP, Geoffroy 0, Laissy JP, Lebtahi R, Silbermann-Hoff-
man 0, Henry-Feugeas MC, Cadiot G, Mignon M, Schou-
niques can be used to treat and monitor the success
man-Claeys E (2001) Imaging appearances of metastases
of treatment in patients with hyperfunctioning tumors from neuroendocrine tumours of the pancreas. Br J Radiol
of the pancreas (UGUR et al. 2002). 74: 1065-1 070
Decristoforo C, Mather SJ, Cholewinski W, Donnemiller E,
Riccabona G, Moncayo R (2000) 99mTc-EDDA/HYNIC-
TOC: a new 99mTc-labelied radiopharmaceutical for
imaging somatostatin receptor-positive tumours; first
References clinical results and intra-patient comparison with
11 lin-labelled octreotide derivatives. Eur J Nucl Med 27:
Anderson CJ, Dehdashti F, Cutler PD, Schwarz SW, Laforest R, 1318-1325
Bass LA, Lewis JS, McCarthy DW (2001) 64Cu-TETA-octreo- Diehl SJ, Lehmann KJ, Gaa J, McGill S, Hoffmann V, Georgi M.
tide as a PET imaging agent for patients with neuroendo- (1999) MR imaging of pancreatic lesions. Comparison of
crine tumors. J Nucl Med 42:213-221 manganese-DPDP and gadolinium chelate. Invest Radiol
Anderson MA, Carpenter S, Thompson NW, Nostrant TT, Elta 34:589-595
GH, Scheiman JM. (2000) Endoscopic ultrasound is highly Doherty GM, Doppman JL, Shawker TH, Miller DL, Eastman
accurate and directs management in patients with neuro- RC, Gorden P, Norton JA (1991) Results of a prospective
endocrine tumors of the pancreas. Am J Gastroenterol 95: strategy to diagnose, localize, and resect insulinomas. Sur-
2271-2277 gery 110:989-997
194 R. F. Thoeni

Doppman JL, Nieman L, Miller DL, et al (1989) Ectopic adre- with three-dimensional volume rendering, maximum
nocorticotropic hormone syndrome: localization studies intensity projection, and shaded-surface display. AJR Am
in 28 patients. Radiology 172:115-124 J Roentgenol 172:925-931
Doppman JL, Miller Dl, Chang R, Maton PN, London JF, Howard TJ, Stabile BE, Zinner MJ, Chang S, Bhagavan BS,
Gardner JD, Jensen RT, Norton JA (1990) Gastrinomas: Passaro E Jr (1990) Anatomic distribution of pancreatic
localization by means of selective intraarterial injection of endocrine tumors. Am J Surg 159:258-264
secretin. Radiology 174:25-29 Iglesias A, Arias M, Casal M, Paramo C, Fiano C, Brasa J (2001)
Duncan JR, Stephenson MT, Wu HP, Anderson CJ (1997) Unusual presentation of a pancreatic insulin om a in heli-
Indium-111-DTPA-Octreotide is delivered to pancreatic, cal CT and dynamic contrast-enhanced MR imaging: case
tumor cell, renal and hepatocyte lysosomes. Cancer Res report. Eur Radiol11:926-930
57:659-671 Imamura M, Takahashi K, 1sobe Y, Hattori Y, Satomura K, Tobe
Ellison CE, Schirmer WJ, Olsen JO (1997) Localization of T. (1989) Curative resection of multiple gastrinomas aided
Neuroendrocrine Tumors Using Somatostatin Receptor by selective arterial secretin injection test and intraopera-
Imaging With Indium-111-Pentetreotide (OctreoScan). tive secretin test. Ann Surg 21O:71O-7l8
Cancer Control 4:35-39 Jaffe BM (1987) Surgery for gut hormone-producing tumors.
Fernandez-Represa JA, Fernandez Rodriguez D, Perez Contin Am J Med 82:68-76
MJ, Pena ML, Martinez-Sarmiento J, Carreras JL, Mayol Jensen RT, Norton JA (1991) Endocrine neoplasms of the
Martinez J (2000) Pancreatic glucagonoma: detection by pancreas. In: Yamada T, Alpers DH, Owyang C, Powell D,
positron emission tomography. Eur J Surg 166:175-176 Silverstein FE (eds) Textbook of gastroenterology. Lippin-
Friesen SR (1982) Tumors of the endocrine pancreas. N Engl cott, Philadelphia, pp 210-232
J Med 306:580-590 Johnson DS, Coel MN, Bornemann M (2000) Current imaging
Frucht H, Doppman JL, Norton lA, Miller DL, Dwyer AJ, Frank and possible therapeutic management of glucagonoma
JA, Vinayek R, Maton PN, Jensen RT (1989) Gastrinomas: tumors: a case report. Clin Nucl Med 25:120-122
comparison of MR imaging with CT, angiography, and US. King AD, Ko GT, Yeung VT, Chow CC, Griffith J, Cockram CS
Radiology 171:713-717 (1998) Dual phase spiral CT in the detection of small insu-
Gabata T, Matsui 0, Kadoya M (1994) Small pancreatic adeno- linomas of the pancreas. Br J Radiol 71:20-23
carcinomas: efficacy of MR imaging with fat suppression Kloppel G, Heitz PU (1988) Pancreatic endocrine tumors.
and gadolinium enhancement. Radiology 193:683-688 Pathol Res Pract 183:155-168
Gehl HB, Urhahn R, Bohndorf K, Klever P, Hauptmann S, Lode- Kraus BB, Ros PR (1994) 1nsulinoma: diagnosis with fat-sup-
mann KP, Matern S, Schumpelick V, Gunther RW (1993) pressed MR imaging. AJR 162:69-70
Mn-DPDP in MR imaging of pancreatic adenocarcinoma: Krejs GJ (1987) VIPoma syndrome. Am J Med 82:37-48
initial clinical experience. Radiology 186:795-798 Krenning EP, Bakker WH, Kooij PPM (1992) Somatostatin
Gibril F, Doppman JL, Chang R, Weber HC, Termanini B,Jensen receptor scintigraphy with [111In-DTPA-D-Phe1]-octreo-
RT (1996) Metastatic gastrinomas: localization with selec- tide in man: metabolism, dosimetry and comparison with
tive arterial injection of secretin. Radiology 198:77-84 [123I-Tyr-3]-octreotide. J Nucl Med 33:652-658
Gower WR Jr, Fabri BJ (1990) Endocrine neoplasms (non-gas- Kvols LK, Brown ML, O'Connor MR (1993) Evaluation of a
trin) of the pancreas. Semin Surg Oncol 6:98-109 radiolabeled somatostatin analog (1-123 Octreotide) in
Gress FG, Barawi M, Kim D, Grendel! JH (2002) Preoperative the detection and localization of carcinoid and islet cell
localization of a neuroendocrine tumor of the pancreas tumors. Radiology 187:129-133
with EUS-guided fine needle tattooing. Gastrointest Endosc Lamberts SWJ, Bakker WH, Reubi JC (1990) Somatostatin
55:594-597 receptor imaging in the localization of endocrine tumors.
Gunther RW, Klose KJ, Ruckert K, Beyer J, Kuhn FP, Klotter HJ. N Engl J Med 323:1246-1249
(1985) Localization of small islet-cell tumors. Preoperative Liessi G, Pasquali C, D'Andrea AA, Scandellari C, Pedrazzoli
and intraoperative ultrasound, computed tomography, S (1992) MRI in insulin om as: preliminary findings. Eur J
arteriography, digital subtraction angiography, and pan- Radiol 14:46-51
creatic venous sampling. Gastrointest Radiol1O:145-152 Maton PN, Miller DL, Doppman JL, Collen MJ, Norton JA,
Guo M, Lemos LB, Bigler S, Baliga M (2001) Duodenal Vinayek R, Slaff JI, Wank SA, Gardner JD, Jensen RT
somatostatinoma of the ampulla of vater diagnosed by (1987) Role of selective angiography in the management
endoscopic fine needle aspiration biopsy: a case report. of patients with Zollinger-Ellison syndrome. Gastroenter-
Acta Cytol 45:622-626 ology 92:913-918
Haber K, Freimanis A, Asher W (1976) Demonstration and McNulty NJ, Francis 1R, Platt JF, Cohan RH, Korobkin M,
dimensional analysis of the normal pancreas with gray Gebremariam A (2001) Multi-detector row helical CT of the
scale echography. AJR 126:624-628 pancreas: effect of contrast -enhanced multiphasic imaging
Hayakawa T, Jin CX, Hirooka Y (2000) Endoscopic ultrasonog- on enhancement of the pancreas, peripancreatic vasculature
raphy of the pancreas: new advances. JOP 1:46-48 and pancreatic adenocarcinoma. Radiology 220:97-102
Hessel SJ, Siegelman SS, McNeil BJ (1982) A prospective evalu- Miller Dl, Doppman JL, Metz D, Maton PN, Norton JA, Jensen
ation of computed tomography and ultrasound of the pan- RT (1992) Zollinger-Ellison syndrome: technique, results,
creas. Radiology 143:129-133 and complications of portal venous sampling. Radiology
Hollett MD, Jorgensen MJ, Jeffrey RB Jr (1995) Quantitative 182:235-241
evaluation of pancreatic enhancement during dual-phase Mitchell DG, Vinitski S, Saponaro S, Tasciyan T, Burk DL Jr,
helical CT. Radiology 195:359-361 Rifkin MD (1991) Liver and pancreas: improved spin-echo
Hong KC, Freeny PC (1999) Pancreaticoduodenal arcades and Tl contrast by shorter echo time and fat suppression at 1.5
dorsal pancreatic artery: comparison of CT angiography T. Radiology 178:67-71
Endocrine Hyperfunctioning Tumors 195

Moore NR, Rogers CE, Britton BJ (1995) Magnetic resonance Schirmer WJ, Melvin WS, Rush RM, O'Dorisio TM, Pozderac
imaging of endocrine tumours of the pancreas. Br Med J RV, Olsen JO, Ellison EC (1995) Ellison EC. Indium-111-
68:341-347 pentetreotide scanning versus conventional imaging
Mori M, Fukuda T, Nagayoshi K, Kohzaki S, Matsunaga N, techniques for the localization of gastrinoma. Surgery 118:
Hayashi K, Motojima K, Kanematsu T (1996) Insulinoma: llO5-1113
correlation of short-T1 inversion-recovery (STIR) imaging Schumacher B, Lubke HJ, Frieling T (1996) Prospective study
and histopathologic findings. Abdom Imaging 21:336-341 on the detection of insulinomas by endoscopic ultrasonog-
Mortele KJ, Oei A, Bauters W, Timmermans F, Cuvelier C, raphy. Endoscopy 28:273-276
Kunnen M, Ros PR (2001) Dynamic gadolinium-enhanced Semelka RC, Cumming MJ, Shoenut JP, Magro CM, Yaffe CS,
MR imaging of pancreatic VIPoma in a patient with Kroeker MA, Greenberg HM (1993) Islet cell tumors:
Verner-Morrison syndrome. Eur Radiolll:1952-1955 comparison of dynamic contrast-enhanced CT and MR
Nishiguchi S, Shiomi S, Ishizu H, Iwata Y, Kurooka H, Mina- imaging with dynamic gadolinium enhancement and fat
mitani S, Habu D, Kawabe 1, Ochi H (2001) A case of glu- suppression. Radiology 186:799-802
cagonoma with high uptake on F-18 fluorodeoxyglucose Semelka RC, Custodio CM, Cern Balci N, Woosley JT (2000)
positron emission tomography. Ann Nucl Med 15:259-262 Neuroendocrine tumors of the pancreas: spectrum of
Outwater EK, Mitchell DG (1996) MR imaging techniques for appearances on MRI. J Magn Reson Imaging 11:141-148
evaluation of the pancreas. Top Magn Reson Imaging 8: Sofka CM, Semelka RC, Marcos HB, Woosley JT (1997) MR
248-264 imaging of metastatic pancreatic VIPoma. Magn Reson
Patel YC, Ganda OP, Benoit R (1983) Pancreatic somatostati- Imaging 15:1205-1208
noma: abundance of somatostatin-28(1-12)-like immuno- Solcia E, Sessa F, Rindi G, Villani L, Riva C, Buffa R, Capella
reactivity in tumor and plasma. J Clin Endocrinol Metab C. (1990) Classification and histogenesis of gastroentero-
57:lO48-lO53 pancreatic endocrine tumours. Eur J Clin Invest 20 [Suppl
Pavone P, Mitchell DG, Leonetti F, Di Girolamo M, Cardone G, 1]: S72-S78
Catalano C, Tamburrano G, Passariello R (1993) Pancreatic Solivetti FM, Giunta S, Caterino M, De Majo A, Coscarella G,
B-cell tumors: MRI. J Comput Assist Tomogr 17:403-407 Carducci M (2001) CT findings in a case of glucagonoma
Pech 0, Lingenfelser T, Wunsch P (2000) Pancreatic gluca- with necrolytic migrating erythema. Radiol Med (Torino)
gonoma as a rare cause of chronic obstructive pancreatitis. 102:410-412
Gastrointest Endosc 52:562-564 Stafford Johnson DB, Francis IR, Eckhauser FE et al (199 8)
Pereira PL, Roche AJ, Maier GW, Huppert PE, Dammann F, Dual-phase helical CT of nonfunctioning islet cell tumors.
Farnsworth CT, Duda SH, Claussen CD (1998) Insulinoma J Comput Assist Tomogr 22:59-63
and islet cell hyperplasia: value of the calcium intraarterial Tabuchi T, Itoh K, Ohshio G, Kojima N, Maetani Y, Shibata T,
stimulation test when findings of other preoperative stud- Konishi J (1999) Tumor staging of pancreatic adenocarci-
ies are negative. Radiology 206:703-709 noma using early- and late-phase helical CT. Am J Roent-
Phan GQ, Yeo q, Hruban RH, Lillemoe KD, Pitt HA, Cameron genoI173:375-380
JL (1998) Surgical experience with pancreatic and peripan- Takahashi H, Nakano K, Adachi Y, Aoki N, Hajiro K, Yama-
creatic neuroendocrine tumors: review of 125 patients. J moto T, Higashizawa T, Chikugo T, Suzuki T (1988) Mul-
Gastrointest Surg 2:472-482 tiple nonfunctional pancreatic islet cell tumor in multiple
Pisegna JR, Doppman JL, Norton JA, Metz DC, Jensen RT endocrine neoplasia type I. A case report. Acta Pathol Jpn
(1993) Prospective comparative study of the ability of MR 38:667-668
imaging and other imaging modalities to localize tumors Tanaka S, Yamasaki S, Matsushita H, Kurosaki A, Takeuchi K,
in patients with Zollinger-Ellison syndrome. Dig Dis Sci Hoshihara Y, Doi T, Watanabe G, Kawaminami K (2000)
38:1318-1328 Duodenal somatostatinoma: a case report and review of
Rens-Domiano S, Reisine T (1992) Biochemical and func- 31 cases with special reference to the relationship between
tional properties of Somatostatin receptors. J Neurochem tumor size and metastasis. Pathol Int 50: 146-152
58: 1987 -1996 Termanini B, Gibril F Doppman JL, Reynolds JC, Stewart CA,
Reubi JC, Kvols E, Krenning E (1990) Distribution of soma- Sutliff VE, Venzon OJ, Jensen RT (1997) Distinguishing
tostatin receptors in normal and tumor tissue. Metabolism small hepatic hemangiomas from vascular liver metasta-
9:78-81 ses in gastrinoma: use of a somatostatin-receptor scinti-
Rivier J, Spiess 1, Thorner M et al (1982) Characterization of graphic agent. Radiology 202:151-158
a growth-hormone-releasing factor from a human pancre- Thoeni RF, Filson RG (1988) Abdominal and pelvic CT: Use
atic islet cell tumor. Nature 300:276-278 of oral metoclopramide to enhance bowel opacification in
Rosch T, Lorenz R, Braig C, Braig C, Dancygier H, Classen M abdominal and pelvic CT examinations. Radiology 169:
(1991) Endoscopic ultrasound in small pancreatic tumors. 391-393
Z GastroenteroI29:110-115 Thoeni RF, Mueller-Lisse UG, Chan R et al (2001) Detection of
Rossi P, Allison DJ, Bezzi M, Kennedy A, Maccioni F, Wynick small, functional islet cell tumors in the pancreas: selection
D, Maradei A, Bloom SR (1989) Endocrine tumors of the of MRI Imaging sequences for optimal sensitivity. Radiol-
pancreas. Radiol Clin North Am 27:129-161 ogy 214:483-490
Rothmund M, Stinner B, Arnold R (1991) Endocrine pancre- Thomason JW, Martin RS, Fincher ME (2000) Somatostatin
atic carcinoma. Eur J Surg OncoI17:191-199 receptor scintigraphy: the definitive technique for char-
Schillaci 0, Annibale B, Scopinaro F, Delle Fave G, Colella AC acterizing vasoactive intestinal peptide-secreting tumors.
(1997) Somatostatin receptor scintigraphy of malignant Clin Nucl Med 25:661-664
somatostatinoma with indium-111-pentetreotide. J Nucl Thorner MO, Perryman RL, Cronin MJ, Rogol AD, Draznin M,
Med 38:886-887 Johanson A, Vale W, Horvath E, Kovacs K (1982) Somato-
196 R. F. Thoeni

troph hyperplasia. Successful treatment of acromegaly by Vinik AI, Delbridge L, Moattari R, Cho K, Thompson N (1991)
removal of a pancreatic islet tumor secreting a growth Transhepatic portal vein catheterization for localization of
hormone-releasing factor. J Clin Invest 70:965-977 insulinomas: a ten year experience. Surgery 109:1-11
Tjon A, Tham RT, Falke THM (1989) CT and MR imaging of Vuolteenaho 0, Leppaeluoto J, Ying S-Y (1990) Isolation and
advanced Zollinger-Ellison syndrome. J Comput Assist characterization of human thyrotropin-releasing hormone
Tomogr 13:821-828 (TRH) from an endocrine pancreatic tumor. Regul Pept 31:
Tjon A, Tham RT, Jansen JB (1994) Imaging features of 33-40
somatostatinoma: MR, CT, US, and angiography. J Comput Weinel RJ, Neuhaus C, Strapp J (1993) Preoperative localiza-
Assist Tomogr 18:427-431 tion of gastrointestinal endocrine tumors using somatosta-
Tscholakoff D, Hricak H, Thoeni RF (1987) Magnetic reso- tin-receptor scintigraphy. Ann Surg 218:640-645
nance imaging in the diagnosis of pancreatic disease. AJR Westlin JE, Janson ET, Amberg H, Ahlstrom H, Oberg K, Nils-
148:703-709 son S. (1993) Somatostatin receptor scintigraphy of carci-
Ugur 0, Kothari PJ, Finn RD, Zanzonico P, Ruan S, Guenther noid tumours using the [1111n-DTPA-D-Phe1] octreotide.
I, Maecke HR, Larson SM (2002) Ga-66 labeled somatosta- Acta Oncol 32:783-786
tin analogue DOTA-DPhe1-Tyr3-octreotide as a potential Wise SR, Johnson J, Sparks J, Carey LC, Ellison EC (1989) Gas-
agent for positron emission tomography imaging and trinoma: the predictive value of preoperative localization.
receptor mediated internal radiotherapy of somatostatin Surgery 106:1087-1092
receptor positive tumors. Nucl Med Bioi 29:147-157 Zeman RK, Berman PM, Silverman PM, Cooper C, Garra BS,
Van Hoe L, Gryspeerdt S, Marchal G, Baert AL, Mertens L Patt RH,Ascher SM. (1995) Biliary tract: three-dimensional
(1995) Helical CT for the preoperative localization of islet helical CT without cholangiographic contrast material.
cell tumors of the pancreas: value of arterial and parenchy- Radiology 196:865-867
mal phase images. AJR Am J RoentgenoI165:1437-1439 Zimmer T, Stolzel U, Liehr RM, Bader M, Fett U, Hamm B,
Vinik AI, Moattari AR, Cho K, Thompson N (1990) Tran- Wiedenmann B, Riecken EO (1995) Somatostatin receptor
shepatic portal vein catheterization for localization of scintigraphy and endoscopic ultrasound for the diagnosis
sporadic and MEN gastrinomas: a ten-year experience. of insulinoma and gastrinoma. Dtsch Med Wochenschr
Surgery 107:246-255 120:87-93
12 Nonhyperfunctioning Endocrine Tumors
C. PROCACCI, M. FERDEGHINI, A. GUARISE, s. VASORI, D. COSER, and M. FERRARI

CONTENTS as loco regional infiltration and/or distant metastases,


are more common at presentation.
12.1 Introduction 197 The radiologist's task when faced with these
12.2 Imaging 197
12.2.1 Transabdominal Ultrasound 197 lesions is to avoid a misdiagnosis of ductal adeno-
12.2.2 Computed Tomography 199 carcinoma and, if possible, to suggest a NHFET. A
12.2.3 Magnetic Resonance Imaging 206 correct diagnosis influences the therapeutic strategy
12.2.4 Nuclear Medicine 209 and prognosis, which are significantly different from
12.3 Differential Diagnosis 213 those of ductal adenocarcinoma and other pancre-
12.4 Diagnostic Strategy 214
References 214 atic neoplasms. Moreover, imaging must determine
whether it is resectable or not, by visualizing signs
such as vascular invasion and hepatic metastases. In
these cases, the alternative to surgery is chemother-
apyor treatment with somatostatin analogues.
12.1
Introduction

Endocrine tumors of the pancreas are easily distin- 12.2


guishable from other pancreatic neoplasms when Imaging
specific clinical syndromes related to hypersecre-
tion of gastroenteropancreatic pep tides are present 12.2.1
(KITAJIMA et al. 1998). Imaging can identify the Transabdominal Ultrasound
single or multiple lesions that produce these clinical
findings. Unfortunately, this is not always possible Until recently, the characterization of endocrine
because of the small dimensions and the frequent tumors with ultrasound (US) was limited due to its
extrapancreatic location of the lesions. Sometimes inability to detect the hypervascularization of the
endocrine tumors can produce peptides but not in lesion, a decisive parameter. Recently, new contrast
sufficient amounts to result in a specific clinical syn- agents, consisting of microbubbles (i.e. Sonovue,
drome. These lesions are called nonhyperfunction- Bracco, Milan, Italy) allow for dynamic studies by
ing endocrine tumors (NHFET). From a pathological US of pancreatic masses during contrast medium
point of view, these tumors are indistinguishable from administration. The early and intense enhance-
hyperfunctioning ones. Their size is usually larger ment and the slow washout from hypervascularized
because of the delay in the diagnosis (KITAJIMA et al. tumors can be documented by monitoring the mass
1998). For the same reason, signs of malignancy, such (Fig. 12.1). US findings are completely in agreement
with those of CT. OSHIKAWA et al. (2002) have recently
reported the value of dynamic US using pulse-inver-
sion harmonic imaging after the administration of
C. PROCACCI, MD; M. FERDEGHINI, MD; S. VASORI, MD; Levovist (D-Galactose, Schering-Berlin, Germany) in
D. COSER, MD; M. FERRARI, MD the characterization of pancreatic tumors, including
Istituto di Radiologia, Dipartimento di Scienze Morfologico- three cases of endocrine tumors. In all three masses,
Biomediche, Universita degli Studi di Verona, Policlinico the contrast-enhanced US examination correlated well
'Giambattista Rossi', P.zza L.A. Scuro 10,37134 Verona, Italy
A. GUARISE, MD
with the spiral CT findings, showing a strong enhance-
Servizio di Radiologia, Ospedale S. Cuore-Don Calabria, Via D. ment in two cases and a moderate enhancement in one
Sempreboni, 37024 Negrar, Italy (OSHIKAWA et al. 2002). Our initial evaluation of con-
198 C. Procacci et al.

a b

c d

Fig. l2.la-d. Contrast-enhanced ultrasound (CEUS) of nonhyperfunctioning endocrine tumor (NHFET). a Unenhanced base-
line US shows a large, sharply outlined, inhomogeneous, hypoechoic mass at the pancreatic head with small internal lamellar
calcifications (arrow). The pancreatic gland appears atrophic upstream with dilatation of the main pancreatic duct (arrowheads).
b Enhanced US after microbubble injection in the arterial phase shows an intense enhancement of the lesion, appearing inhomo-
geneously hyperechoic due to the presence of small internal necrotic areas (arrows). c Enhanced US in the venous phase seconds
after contrast medium injection shows the persistent enhancement of the pancreatic mass. d Persistent weak enhancement of
the lesion in the delayed phase because of contrast medium microbubble 'stagnation' in the tumor circulation is seen

trast-enhanced ultrasound (CEUS) agrees with what gland is typically less echoic (Fig. 12.3). Occasionally,
others have said, that the contrast seems to have great the tumor can have a sonolucent appearance with
potential. In fact, we have been able to identify and enhanced posterior wall reflections when it contains
characterize all the tumors, making a correct evalu- fluid (KATO et al.2000).MostNHFETarelarge (Fig.12.1)
ation of the locoregional diffusion and the hepatic at presentation. The mass has a well-defined, multilob-
metastases at the same time. CEUS increases the ulated border, with compression of the adjacent struc-
diagnostic specificity but not the sensitivity because tures (digestive tract, vessels, etc.). Necrosis and hem-
contrast medium administration currently is only orrhage appear as central hypoechoic areas (Fig. 12.4)
performed if the lesion has already been identified (FUGAZZOLA et al. 1990; FURUKAWA et al. 1998). The
during a basic ultrasound examination (FURUKAWA lesion grows by expanding rather than encasing, and
et al. 1998). However, should the sensitivity of CEUS therefore the common bile duct and main pancreatic
become analogous to that of the other techniques, it duct are frequently normal. Encasement of the arterial
could be performed when the basic study is negative vessels, thrombosis, or neoplastic involvement of the
in order to identify even the smallest lesions. portal venous system may be present in association
A small NHFET, which is quite rare, appears as a with large masses. Peritumoral lymphadenopathy is
well-circumscribed nodule embedded in the pancre- rare, while metastatic disease in the liver occurs in
atic gland and is clearly hypoechoic when compared 30% of cases (OGAWA et al. 2000). The US appearance
with the normal tissue (Fig. 12.2a). This feature is also of the liver metastases is variable. Hyperechoic nodules
common to hyperfunctioning tumors (FUGAZZOLA et or 'target-like' appearances strongly suggest the endo-
al. 1990; FURUKAWA et al. 1998). The tumor can rarely crine nature of the primary tumor. In other cases, they
appear isoechoic compared to the normal gland. This show a nonspecific hypo echoic pattern (FUGAZZOLA
can occur in younger subjects, where the normal et al.I990).
Nonhyperfunctioning Endocrine Tumors 199

a b

Fig. 12.2a,b. Small NHFET, US evaluation. a Unenhanced US shows a well-outlined, small, hypoechoic lesion (circled) in the
pancreatic body. b Axial CT scan in arterial phase after contrast medium administration confirms the small hyperdense lesion
(circled) in the pancreatic body

12.2.2
Computed Tomography

The contribution of CT in identifying and char-


acterizing NHFET is almost always decisive, when
the properly timed spiral or multi slice scanners are
used. The short scanning time optimizes the dynamic
study of the pancreas after iodinated contrast mate-
rial administration and results in demonstration of
the typical enhancement of these tumors (BUETOW et
al. 1997; STAFFORD-JOHNSON et al.1998; ICHIKAWA et
al. 2000; ZEMAN et al. 1995; PROCACCI et al. 2001).
Larger tumors that distort the morphology of the
Fig. 12.3. US of small NHFET in a young patient. Basic US
shows a well-outlined lesion in the pancreatic head (arrow), gland can be demonstrated before contrast medium
displaying slight difference in echogenicity with respect to the administration (Figs. 12.5a, 12.6a, 12.7a) (MURASE et
normal parenchyma al. 2000). The mass usually develops along the major
axis of the gland (Figs. 12.5a, 12.6a); less frequently,
it has a rounded morphology and radial growth
(Fig. 12.7a). Sometimes dilation of the main duct and
atrophy of the parenchyma can be seen upstream of

a b

Fig. 12.4a,b. Color-Doppler US of NHFET. a Unenhanced US shows a large, hypoechoic mass in the pancreatic body with a
central, small, anechoic area (arrow). b Color-Doppler baseline US with pulse repetition frequency set at low flow of the lesion
demonstrates vessels within the lesion
200 C. Procacci et al.

a b

c d

Fig. 12.Sa-d. NHFET, CT evaluation. a Before iodinated contrast medium administration, the pancreatic neck appears larger in
volume compared with the remaining pancreatic parenchyma. The gastric antrum (asterisk) is displaced by the neoplasm. The
main duct is considerably dilated upstream (arrows). b, c During the pancreatic phase of contrast enhancement, the hyper-
vascularized tumor is visible, showing the same density as the renal cortex. There are hypodense necrotic gaps (black arrow)
within it. The gland is atrophic in the body-tail; the dilation of the main duct is more evident in this phase (arrows). A small
hyperdense metastasis (arrowhead) is evident in the right liver lobe (c). d In the venous phase, both the tumor and the liver
metastasis are almost isodense with the liver parenchyma

the lesion (Fig. Sa). Globular or lamellar calcifications delay). According to others (Lv et al. 1996), the pan-
may be present (Fig. 12.7a). The latter are more likely creatic phase (40-60 s scan delay) is more appropri-
to be found on the edge of a necrotic or cystic area ate to show the vascularization of the lesion. However,
(FVGAZZOLA et al.1990). The stomach and duodenum the majority of authors confirm that a preliminary
may appear compressed by the tumor (Fig. 12.5a), examination before contrast administration and a
but frank invasion into adjacent viscera is rare. dual-phase acquisition (arterial or pancreatic and
Enlargement of the liver, with irregular contours, sig- venous) during the dynamic study are needed (VAN
nals diffuse metastatic involvement (Fig. 12.8a). The HOE et al. 1995; ICHIKAWA et al. 2000; STAFFORD-
lesions are slightly hypo dense, compared with the JOHNSON et al. 1998).
normal parenchyma (Fig. 12.7a), appearing hyper- In the pancreatic phase, which is our personal
dense only in fatty liver (Fig. 12.8a). Calcifications are preference, the hypervascularized tumor shows
often present. intense enhancement, similar to that of the spleen
There are controversial opinions in the literature or the renal cortex, definitely greater than that of the
about the best dynamic phase, since some authors liver parenchyma (Figs. 12.5b,c, 12.6b,c, 12.7b, 12.8c,d,
(STAFFORD-JOHNSON et al. 1998; HOLLETT et al. 12.9,12.10,12.11, 12.12a,b, 12.13a,b). The lesion is also
1995) claim that they obtain the highest enhance- denser than the pancreatic parenchyma, especially if
ment of the pancreatic gland, and therefore of the obstruction of the main duct has resulted in chronic
tumor, during the early arterial phase (20-25 s scan obstructive pancreatitis upstream of the lesion. In
Nonhyperfunctioning Endocrine Tumors 201

a b

Fig. 12.6a-c. Typical NHFET, CT evaluation. a Before contrast


enhancement, the body of the pancreas has a slightly globular
appearance with convex posterior margins. A small, irregular,
hyperdense hepatic lesion is recognizable. b, c During the pan-
creatic phase following IV contrast administration, the tumor
is confirmed developing along the major axis of the pancreas.
It displays a slightly higher enhancement to the adjacent
parenchyma. The tumor displaces the splenic vessels. Metas-
c tasis shows the same density as the primary tumor

a b

Fig. 12.7a,b. NHFET with peripheral calcifications, CT evaluation. a Unenhanced scan shows small calcifications (arrow) along
the edge of the round mass in the tail of the pancreas. In the right hepatic lobe, there is a large, hypodense tumor. b During the
pancreatic phase, intense and inhomogeneous enhancement of the pancreatic mass is evident. The hepatic mass shows intense
and inhomogeneous enhancement, confirming its metastatic nature. Rich collateral networks are due to the compression of
the splenic vein by the mass
202 C. Procacci et al.

a b

c d

Fig. 12.8a-d. NHFET with diffuse metastatic involvement of the liver, CT evaluation. a Unenhanced scan reveals an increase
in liver volume from the presence of metastases. These appear slightly hyperdense due to the fatty liver. b In the pancreatic
phase, the multiple metastases show marginal enhancement with large, central, necrotic areas. c, d In the same pancreatic phase,
the primary tumor at the head of the pancreas shows inhomogeneous enhancement; the superior mesenteric vein (arrow) is
embedded by the tumor

a b

Fig. 12.9a,b. NHFET with mixed appearance, CT evaluation. a In the pancreatic phase, the mass of the head of the pancreas
shows homogeneous and intense enhancement. b In the same phase, a hypo dense cystic component can be seen in the inferior
portion of the mass
Nonhyperfunctioning Endocrine Tumors 203

a b

Fig. 12.10a,b. NHFET with vascular encasement, CT evaluation. a In the pancreatic phase, the large mass of the head and body
of the pancreas appears hyperdense. The obstruction of the hepatic artery (long arrow) and the encasement of the splenic
artery (arrowheads) are evident. b The occlusion of the splenic vein is demonstrated indirectly by the rich collateral network
(arrowheads) through the gastroepiploic veins

a b

Fig. 12.11a,b. NHFET with vascular encasement, CT evaluation. In the pancreatic phase, the large, inhomogeneously hyperdense
mass originating from the pancreatic body obstructs and obliterates the splenic vein, with resulting large collateral network
(arrows). The tumor displaces the stomach

this phase, necrotic areas or cystic degeneration, structures (Figs. 12.6b,c, 12.7b). Due to the unpredict-
if present, are characterized by clear hypodensity able growth of the tumor, some arterial vessels, like
compared with the hypervascularized neoplastic the superior mesenteric artery or the celiac axis, can
tissue (Figs. 12.5b,c, 12.7b, 12.9b, 12.11, 12.12b). Due be encased within the neoplastic mass (Fig. 12.10).
to the intense enhancement of the tumor, its contours The encasement (STAFFORD-JOHNSON et al. 1998) is
and real dimensions are defined (CHUNG et al.1997). better recognizable in the three-dimensional recon-
At the same time, the tumor's relationship with the struction of the peripancreatic vessels (CHUNG et
adjacent vascular structures are better depicted. The al. 1997). A rich collateral venous network bypasses
expansive growth and the considerable size of the the obstructed or thrombosed peripancreatic veins
tumor account for the compression of the vascular (Figs. 12.10b, 12.11). Neoplastic invasion into the
204 C. Procacci et al.

a b

c d

Fig. l2.l2a-d. Typical NHFET with metastasis, CT evaluation. a, b In the pancreatic phase, the primary pancreatic tumor
and the liver metastasis show intense and inhomogeneous enhancement. Central necrotic areas are present in both lesions.
c, d During the venous phase, the lesions have 'washed out'. The presence of hypodense central necrotic gaps can be better
appreciated (asterisk in d). The tumor causes obstruction of the main duct, which is dilated at the body-tail (arrows); despite
the considerable size, the primary tumor has well-defined margins, only displacing the stomach and the superior mesenteric
artery (arrowhead in a and b)

spleno-mesenteric-portal tree is seldom found. (FUGAZZOLA et al. 1990; PROCACCI et al. 2001). In the
When present, the neoplastic thrombus has the same other 30%, the pattern is nonspecific (Fig. 12.14) or
density as the mass from which it derives, so it dis- similar to that of ductal adenocarcinoma (Fig. 12.15a)
plays a slightly lower density than the vascular lumen (VAN HOE et al. 1995; KEOGAN et al. 1997) since the
(Fig. 12.13a,b). tumor is clearly hypo dense compared with the spleen
In the pancreatic phase, the hepatic metasta- or the renal cortex. When the mass is large and well-
ses have the same density as the primary tumor circumscribed, a ductal adenocarcinoma can be
(STAFFORD-JOHNSON et al. 1998; DEBRAY et al. 2001) excluded, but the problem of differential diagnosis
(Figs. 12.7b, 12.12a,b). The homogeneous or inho- from other rare solid tumors still remains. Small
mogeneous (target-like metastases) hyperdensity is tumors, especially those with infiltrating growth,
even more evident because of the low density of the mimic ductal adenocarcinoma. The liver metastases
hepatic parenchyma (Fig. 12.8b). show in virtually all cases the same enhancement as
The previously described features, essential for the the primary tumor (Fig. 12.15), also similar to ductal
diagnosis of NHFET, are present in 70% of patients adenocarcinoma.
Nonhyperfunctioning Endocrine Tumors 205

a b

c d

Fig. 12. 13a-d. NHFET with tumor thrombus in the portal vein, CT evaluation. a, b In the pancreatic phase, the portal mesenteric
axis is filled with a large neoplastic thrombus (arrowheads), which is in continuity with the primary tumor of the pancreatic
body (asterisk). c, d In the venous phase, both the neoplastic thrombus and the tumor have the same density

. .'

•.
( . '1

4 1
••. ~
..
.~ '.'

.: .

·~. ~·~~I
IlO
l40
'l:;J
... .."
a O.7~ b

Fig. 12.14a,b. Atypical NHFET, CT evaluation. In the pancreatic phase the normal parenchyma of the head (asterisk) appears
hyperdense compared with the tumor located at the uncinate process. The superior mesenteric vein (long arrow) is imprinted
and displaced

In the venous phase, there is a quick washout from (Fig. 12.12d). Some investigators reported that endo-
the hypervascularized tumor, whose density is anal- crine tumors, appearing as iso-attenuated in pan-
ogous to that of the normal pancreatic parenchyma creatic phase CT images, are sometimes detected in
(Figs. 12.5d, 12.12d, 12.13c,d). Possible calcifications, the portal venous phase (VAN HOE et al. 1995). The
necrotic or cystic areas are recognizable in the mass late enhancement of the tumor is common when the
206 C. Procacci et al.

a b

Fig. 12.1Sa,b. Atypical NHFET, CT evaluation. The mass in the tail of the pancreas remains hypodense in both the pancreatic
(a) and late (b) phases. This enhancement pattern is indistinguishable from ductal adenocarcinoma. The multiple hepatic
metastases show the same low enhancement

extensive necrosis results in a slower washout from tumors (SEMELKA and ASCHER 1993; PAVONE et al.
the mass, due to the reduced vascularization (KoITO 1993; ANGELI et al. 1997). With current MR imag-
et al. 1997). ing technology, the entire liver may be covered in a
In the venous phase, the tumor's contour and its breathhold or by single-shot techniques, allowing the
relationship with the surrounding arteries are less improved detection of hepatic metastases.
conspicuous, due to the rapid washout of contrast The elevated MR contrast resolution leads to the
from the mass. During this phase, the relationship with identification of very small primary tumors, which
the peripancreatic veins can be accurately evaluated appear hypointense in comparison with the normal
because the tumor thrombus is optimally displayed parenchyma on Tl-weighted sequences (Figs. 12.16a,
(Fig. 12.13c,d). The collateral network is also easily rec- 12.17a, 12.1Sa, 12.19a). This difference in signal is fur-
ognizable. The venous phase is less sensitive in depict- ther exaggerated when fat-suppressed Tl sequences
ing the typically hypervascular hepatic metastases, as are used with or without intravenous gadolinium
they become isodense or hypodense compared with (Fig. 12.17a,b). Some authors have pointed out that
the hepatic parenchyma (Fig. 12.12c,d). Sometimes gadolinium may actually decrease the ability to
only the larger lesions that displace portal branches or recognize the lesion (OWEN et al. 2001). The typical
lesions with the 'target -like' appearance can be seen in features of primary non-hyperfunctioning islet cell
this phase. Those masses which are relatively hypovas- tumors include a large pancreatic mass of low signal
cular tumors remain hypo dense in the venous phase, intensity (less than or equal to the normal pancreas)
as do their hepatic metastases (Fig. 12.1Sb). on Tl-weighted images (Figs. 12.16a, 12.17a,b, 12.1Sa,
12.19a) and of intermediate to high intensity on T2-
weighted images (Figs. 12.16b, 12.17c, 12.1Sb, 12.19b).
12.2.3 In a minority of patients, the mass may contain
Magnetic Resonance Imaging necrosis or hemorrhage, and is usually hyperintense
on Tl-weighted images (CARLSON et al.1993).
Magnetic resonance (MR) imaging gives similar Cystic changes within islet-cell tumors are more
information concerning NHFET to that displayed common than previously thought. Cystic islet-cell
by CT scanning. MR can be used for the character- tumors are usually unilocular lesions with hypoin-
ization and evaluation of the extent (Fig. 12.16) of tense contents on Tl-weighted and hyperintense
masses previously detected with US. The advantages contents on T2-weighted images (BUETOW et al.
of MR over CT include higher contrast resolution 1997) and a wall of variable thickness (BUETOW et
and therefore higher sensitivity of the dynamic con- al. 1997). The appearance is identical to other cystic
trast-enhanced MR study for detecting endocrine tumors of the pancreas, so that a definitive diagnosis
Nonhyperfunctioning Endocrine Tumors 207

a b

c d

Fig.12.16a-f. NHFET, MR evaluation. a, bTl gradient recalled echo with fat saturation (GRE FS) and T2 short time inversion
recovery (STIR) sequences: an ovoid mass growing along the major axis of the body and tail of the pancreas; it is slightly
hypointense compared with the normal parenchyma on TlW (a) and hyperintense on T2W (b). The hepatic metastases also
appear hypointense on TlW and slightly hyperintense on T2W. c, d Gadolinium (Gd)-DTPA opposed phase Tl GRE sequence
(venous phase) in axial (c) and coronal (d) planes: the mass demonstrates inhomogeneous enhancement in relation to the
presence of hypointense necrotic areas within it (arrow). The hepatic metastases are not recognizable. Accessory spleen at the
splenic hilum (arrowheads). e, f In the 3D angio-MR reconstructions, the celiac trunk with its branches and the mesenteric
artery show normal patency
208 C. Procacci et a1.

Fig. 12.17a-c. NHFET, MR evaluation. The mass of the pan-


creatic head is not recognizable with certainty in the II GRE
sequence in phase (a), while it is clearly evident on II GRE
using FS (b). The lesion typically appears slightly hyperintense
in the turbo spin-echo T2-weighted sequence (c)

can only be obtained by histological examination of est enhancement in the pancreatic phase (Fig. 12.18c)
the specimen. and remain hyperintense (Figs. 12.16c,d, 12.19c) even
The signal intensity and the morphological char- in the venous phase (STAFFORD-JOHNSON et al. 1998;
acteristics detected without intravenous gadolinium OWEN et al. 2001). Persistent hyperintensity is found
administration can only lead to the suspicion of mainly in the larger lesions (Fig. 12.19c), where the
NHFET, and not to a specific diagnosis. Moreover, neoplastic thrombosis of the draining veins results in
some islet tumors may have a low signal on T2- retention of contrast medium (STAFFORD-JOHNSON
weighted images due to the presence of abundant et al. 1998). According to KOITO et al., the delayed
fibrous tissue (OWEN et al. 2001), and therefore, they enhancement correlates with malignancy (KoITO
are indistinguishable from a ductal carcinoma. These et al. 1997). Sometimes, if quite large, these tumors
characteristics could justify the usefulness of gado- have central necrotic areas that remain hypointense
linium-enhanced Tl-weighted images obtained 5-10 on Tl-weighted images even after contrast medium
min following injection in detecting some endocrine administration.
tumors. In fact, according to IcHIKAWA, up to 35% The best dynamic phase for studying endocrine
of scirrhous NHFET at histopathological examina- tumors is still being debated. Some authors argue
tion show this delayed enhancement (IcHIKAWA et that the pancreatic phase is superior, similar to the
a1.2000) experience with CT, while others prefer the delayed
Nowadays, it is possible to carry out breathhold (5-10 min) Tl GRE sequence after contrast medium
dynamic studies of the abdomen, as with CT, but with administration in order to demonstrate also the late
a higher contrast resolution. The findings are the enhancement of tumors with an abundant scirrhous
same: the hypervascularized NHFET show the high- component (ICHIKAWA et al. 2000). All (DEBRAY et al.
Nonhyperfunctioning Endocrine Tumors 209

a b

c d

Fig.12.I8a-d. NHFET, MR evaluation. a, b II GRE in-phase sequence and T2 half-Fourier single-shot turbo spin-echo (HASTE)
images show a large, solid mass within the head of the pancreas displaying expansive growth with well-defined margins. The
mass is hypointense on II W image (a) and slightly hyperintense on HASTE (b). The HASTE sequence highlights the impressions
on the digestive tract. c On Gd-DPTA-enhanced, fat-suppressed, TI GRE sequence, the mass demonstrates marked enhancement
in the pancreatic phase. d At MRCP, the mass causes compression of the main duct, which is dilated upstream

2001; OWEN et al. 2001) agree on greater sensitivity small,hypointense nodules (Fig. 12.20). This particu-
during the pancreatic phase in identifying hypervas- lar capability of MR can be useful especially during
cularized hepatic metastases (Fig. 12.20). Metastases the follow-up of these patients.
rarely contain necrotic and hemorrhagic areas or
calcifications (DEBRAY et al. 2001). Because there is
no radiation risk, a multiphasic MR examination can 12.2.4
be safely performed. Nuclear Medicine
Using the 3D acquired Tl-weighted sequence, it
is possible to obtain a vascular map of the lesion, The first time that receptor scintigraphy was applied
together with information about its signal and to the study of endocrine tumors was in 1987 with
enhancement characteristics (MR angiography: the synthesis of 1231_Tyr3-octreotide, the radiola-
Figs. 12.16e,f, 12.1ge,f). Recently, some authors beled counterpart of the octapeptide somatostatin
(PETERSEIN et al. 2000) described the usefulness of analogue octreotide. Octreotide possesses a high
Gd-BOPTA where both the dynamic study and the affinity for somatostatin receptor subtype 2 (also for
late study can be carried out. In the delayed examina- subtypes 3 and 5) and an appropriate serum half-life
tion (after 2 h), small metastases that other sequences (KRENNING et al. 1992). Nevertheless, hepatobiliary
may have failed to demonstrate can be depicted as and intestinal excretion of this radiopharmaceutical
210 C. Procacci et al.

a b

c d

Fig. 12.19a-f. NHFET, MR evaluation: 'all in one' technique. Tl GRE FS and T2 HASTE sequences show a large, round tumor of
the body of the pancreas, hypointense on TlW (a) and inhomogeneously hyperintense on HASTE (b); a hypointense fibrous
component (arrowheads) is present. c Gd-DPTA enhanced Tl GRE FS sequence: in the late phase, the mass remains inhomo-
geneously enhanced. d On MRCP, the main duct is not recognizable in the body-tail due to compression by the mass. e The
arterial MR angiogram reveals regular patency of the celiac trunk and the superior mesenteric artery, which appear displaced
with a typical 'basket' pattern. f In the portal MR angiogram, compression and possible infiltration of the mesenteric-portal
confluence (arrows) can be seen
Nonhyperfunctioning Endocrine Tumors 211

a b

c d

Fig. 12.20a-e. Hypervascularized hepatic metastasis (MR


evaluation with gadolinium BOPTA, Multihance, Bracco,
Milan, Italy). Tl GRE FS and T2 STIR sequences: a tiny focus
of altered signal (arrow), hypointense on Tl W (a) and slightly
hyperintense on T2W (b) is recognizable. Gd-DPTA-enhanced
Tl GRE FS sequence reveals the lesion only in the arterial
phase (c), becoming isointense (arrows) with the liver in the
venous phase (d). e On a 2-h delayed image, the hypointense
e metastasis is again evident

affected the quality of the study of the abdomen. This high affinity receptors, which are overexpressed on
is overcome with D-Phel-octreotide peptide. When the cell membranes of many tumors, especially of
chelated with DTPA, and labeled with lllIn (>95%), endocrine origin.
the agent is mainly excreted by the kidneys ellln- After intravenous administration of 10 Ilg of pep-
pentetreotide, Octreoscan, Mallinckrodt Medical, tide labeled with at least 200 MBq of III In, images can
Petten, The Netherlands). The long half-life of lllIn be acquired with planar and single photon emission
allows for an accurate study of the lesion up to 48 h computed tomography (SPECT) technique after
after the injection. The basis for receptor scintigra- 4, 24, and/or 48 h, with a wide field-of-view tomo-
phy is receptor-mediated internalization and lyso- graphic gamma-camera equipped with medium
some entrapment of a radiolabeled peptide within energy collimators. In addition to octreotide scan-
212 C. Procacci et al.

ning, positron emission tomography (PET) utilizing the 60 NHFET of the pancreas. Conventional investi-
e
18F-Iabeled ftuorodeoxyglucose 8FDG) is used to gations, in contrast, could identify correctly 100% of
identify areas with high anaerobic metabolism, pres- the positive lesions at scintigraphy and demonstrated
ent in less differentiated endocrine tumors. These the primary in another nine patients, in whom SRS
anaplastic tumors tend not to express somatostatin was negative. Our personal experience matches
receptors and, therefore, are not visible on octreotide those of the literature: the sensitivity of scintigraphy
scintigraphy (SRS). Somatostatin analogues labeled for lesions expressing somatostatin receptors was
with other positron-emitting isotopes (86Y_DOTA_ 95.6%, while CT and MR sensitivity for pancreatic
Tyr3-octreotide, 68Ga-DFO-octreotide, 64CU_ TETA- masses was 100%. SRS therefore has a high sensitiv-
octreotide) can give more detailed images than ity with the possibility of also identifying small-sized
IllIn Octreoscan scintigraphy. Furthermore, unusual lesions (1-1.5 cm) (Fig. 12.21). False-negative scans
metabolic pathways, such as serotonin precursors in result from a paucity of somatostatin receptors on
carcinoids, can also be explored by PET with IIC_5_ an individual tumor. When studying NHFET, IIIIn_
HTP (5-hydroxytriptophane) (ERIKSSON et al. 2000). Octreoscan scintigraphy is not indicated for the
The majority of pancreatic endocrine tumors can identification but for the characterization of lesions.
be visualized by means of IIIIn-Octreoscan scintigra- In fact, in masses with uncertain or nonspecific fea-
phy. Nuclear medicine techniques have a well-defined tures on conventional imaging, scintigraphic uptake
role in localizing functioning endocrine tumors leads to a clear diagnosis of endocrine tumor.
(especially gastrinomas), particularly when small or The most significant contribution of receptor scin-
where other imaging methods have failed. However, tigraphy definitely lies in the possibility of staging
there are no reported studies comparing the different endocrine tumors. This is due to its high sensitivity
imaging techniques in pancreatic NHFET. and, particularly, in the possibility to perform a 'total
The European Multicenter Trial (KRENNING et al. body' exploration (Fig. 12.22).
1994) studied 350 patients who were submitted to In the identification of hepatic metastases, CHIT!
both somatostatin-receptor scintigraphy (SRS) and reports 90% sensitivity for SRS, slightly higher than
conventional radiological investigations. The overall that of CT (78%) and US (88%), with a similar speci-
sensitivity was 80% for SRS and 88% for the other ficity (CHIT! et al. 1998). The sensitivity was even
methods. Octreoscan scintigraphy identified 49 of greater for other soft-tissue lesions (90% with SRS vs

8
h

c
a

Fig.12.21a-c. Receptor scintigraphy with Octreoscan. a Planar image, 24 h post-injection: tracer uptake seen within
the pancreatic head. b SPECT image, transaxial projection, 4 h post-injection: intense uptake at the level of the
superior pole of the right kidney. c SPECT image, coronal projection, 4 h post-injection: intense uptake within the
pancreatic head
Nonhyperfunctioning Endocrine Tumors 213

a b c d

Fig.12.22a-d. Receptor scintigraphy with Octreoscan. a Planar image, anterior projection, 24 h post-injection: marked uptake
within the pancreatic region which corresponds to a 6 cm diameter pancreatic mass found with CT; hepatic lesions not found
on MR. b Planar image, anterior projection, 24 h post-injection uptake within the pancreatic region; large liver metastasis, bone
lesions. Planar images, anterior (c) and posterior (d) projections, 24 h post-injection: pancreatic lesion, mediastinal lymph
nodes, multiple bone metastases

66% with CT and 47% with US). In Chiti's large case and/or metastases. Further studies will focus on the
study (131 patients), which included both function- diagnostic value of 18FDG-PET in rapidly progress-
ing and nonhyperfunctioning tumors, Octreoscan ing, poorly differentiated endocrine tumors.
altered the therapy choice in 21%. The sensitivity
of SRS was also high in revealing bone metastases,
superior to that of bone scintigraphy (LEBTAHI et
al. 1999), and metastases in other sites (FRILLING 12.3
et al. 1998). Metastases taking up the radiopeptide Differential Diagnosis
can be treated with cold somatostatin analogues or
by radiometabolic therapy using analogues radiola- The problems of the differential diagnosis between
beled with beta-emitting isotopes. Nonfunctioning NHFET and the other pancreatic masses vary accord-
metastases by nuclear scan, however, need different ing to the macroscopic aspect of the tumor and its
treatments, often palliative (chemoembolization, vascularity.
chemotherapy). The long clinical history of non- The solid, hypervascularized variant is more easily
hyperfunctioning tumors accounts for their late categorized due to its conspicuous enhancement after
diagnosis and, therefore, for the frequent presence contrast medium administration on CT, MR, and,
of metastases at diagnosis. For this reason, NM can more recently, CEUS. The definite contours and expan-
be of critical importance for the characterization and sive growth pattern with the consequent compressive
staging of these tumors, leading to optimization of effect on the peripancreatic vessels and viscera con-
the subsequent therapeutic choice. tribute to the diagnosis. Arterial encasement and
If NM techniques demonstrate the overexpression venous thrombosis are rarely found. The characteriza-
of somatostatin receptors in the tumor at diagnosis, tion of these tumors is facilitated by the frequent pres-
the high sensitivity of this technique justifies its use ence of hypervascularized hepatic metastases. These
in the follow-up, for a rapid demonstration of relapse findings lead only to a suspicion of this type of tumor,
214 C. Procacci et al.

as the diagnosis is not certain since other neoplasms dyspepsia, etc.). The diagnosis is usually late, and
can show the same characteristics. either US or CT will be the first method to suggest
Acinar carcinoma, in its solid variant, can be well- the diagnosis. The most significant aim of imaging
vascularized and thus may, in the pancreatic phase, is characterization of the lesion, which is performed
mimic an endocrine tumor. It should be pointed out by CT or MR after contrast medium administration,
that also from a histological point of view, it is dif- demonstrating the high enhancement in the lesion.
ficult to distinguish the endocrine or acinar nature Since CT and MR findings are virtually identical,
of the mass. a choice has to be made as to which method to
Ductal adenocarcinoma, in rare cases character- adopt. Therefore, characterization could be made
ized by a poor endotumoral fibrous stromal com- by CEUS, but further studies are needed to deter-
ponent, can appear hyperintense in the pancreatic mine the effective accuracy of this technique. At
phase. Misdiagnosis can occur even when there are the moment, the routine use of nuclear medicine
clear signs of peripancreatic vessel infiltration, if the does not seem to be advisable. However, since a
mass is very large. definite characterization is not objectively possible
Finally, hypervascularized pancreatic metastases, with imaging, fine needle biopsy is always required
especially from renal tumors, must be taken into before treatment.
account, since they can have the same pattern. Usu- The prognosis of these tumors is much better
ally, the problem of differential diagnosis is extremely than that of ductal adenocarcinomas, and therefore
uncommon, since synchronous or metachronous a surgical attempt integrated with chemotherapy is
metastases always arise in a well-known history of a indicated, even in the more advanced stages of the
primary renal carcinoma. Moreover, pancreatic metas- disease. For this reason, accurate staging of the tumor
tases, which can appear many years after the identifi- can be obtained with CT or MR. Nuclear medicine
cation of the primary tumor, are frequently multiple. could improve the staging, being able to identify
The solid, hypovascularized variant can never be distant metastases. lllIn-Octreoscan scintigraphy
categorized by imaging, as it shows the same aspects is more accurate than bone scintigraphy in demon-
as other solid pancreatic tumors. If an infiltrating strating bone metastases.
growth pattern and/or hypovascularized hepatic Nuclear medicine investigation is essential to
metastases are already present, misdiagnosis of a assess the possibility of utilizing treatment with
ductal adenocarcinoma is almost unavoidable. When somatostatin analogues, either cold or radiolabeled.
the tumor, however, has expansive growth and well- The presence of somatostatin receptor subtype 2 is
defined contours, especially in women, a solid pseu- promising for the possible success of the treatment.
dopapillary tumor can be wrongly diagnosed. Finally, in the follow-up of these tumors, due to the
There is no possibility of primarily diagnosing further advantage of identifying distant metastases,
the cystic variant by imaging, since it has the same nuclear medicine techniques are advisable, given the
aspects as the mucinous cystic tumor (macro cystic actual difficulty of CT and MR in distinguishing scar
multilocular aspect) or the solid pseudopapillary tissue from the residual or relapsing tumor, especially
tumor in its cystic variant (mixed solid and liquid in operated patients.
aspect) or a completely nonspecific pattern (macro-
cystic unilocular aspect).
For these reasons, fine needle aspiration of all solid
hypervascularized or hypovascularized pancreatic References
masses is strongly recommended in order to achieve
a definite diagnosis. The cystic variant, however, can Angeli E, Van zulli A, Castrucci M, Venturini M, Sironi S, Zerbi
be characterized only by histological examination of A, Di Carlo V, Pozza G, Del Maschio A (1997) Value of
abdominal sonography and MR imaging at 0.5 T in preop-
the specimen.
erative detection of pancreatic insulinoma: a comparison
with dynamic CT and angiography. Abdom Imaging 22:
295-303
Buetow PC, Miller DL, Parrino TV, Buck JL (1997) Islet cell
12.4 tumors of the pancreas: clinical, radiologic, and pathologie
correlation in diagnosis and localization. Radiographies
Diagnostic Strategy
17:453-472
Carlson B, Johnson CD, Stephens DH, Ward EM, Kvols LK
NHFET are mainly found by chance when patients (1993) MRI of pancreatic islet cell carcinoma. J Comput
present with nonspecific symptoms (palpable mass, Assist Tomogr 17:735-740
Nonhyperfunctioning Endocrine Tumors 215

Chiti A, Fanti S, Savelli G et al (1998) Comparison of somatosta- Krenning EP, Kwekkeboom DJ, Oei HY et al (1994) Soma-
tin receptor imaging, computer tomography and ultrasound tostatin-receptor scintigraphy in gastroenteropancreatic
in the clinical management of neuroendocrine gastro- tumors. An overview of European results. Ann NY Acad
entero-pancreatic tumors. Eur J Nucl Med 25:l396-1403 Sci 733:416-424
Chung MJ, Choi BI, Han JK, Chung JW, Han MC, Bae SH (1997) Lebtahi R, Cadiot G, Delahaye N et al (1999) Detection of bone
Functioning islet cell tumor of the pancreas. Localization metastases in patients with endocrine gastroenteropancre-
with dynamic spiral CT. Acta Radiol 38:l35-l38 atic tumors: bone scintigraphy compared with somatosta-
Debray MP, Geoffroy 0, Laissy JP, Lebtahi R, Silbermann-Hoff- tin receptor scintigraphy. J Nucl Med 40:1602-1608
man 0, Henry-Feugeas MC, Cadiot G, Mignon M, Schou- Lu DS, Vedantham S, Krasny RM, Kadell B, Berger WL, Reber
man-Claeys E (2001) Imaging appearances of metastases HA (1996) Two-phase helical CT for pancreatic tumors:
from neuroendocrine tumours of the pancreas. Br J Radiol pancreatic versus hepatic phase enhancement of tumor,
74: 1065-1070 pancreas, and vascular structures. Radiology 199:697-701
Eriksson B, Bergstrom M, Orlefors H et al (2000) Use of PET in Murase J, Kubo S, Hirohashi K, Shuto T, Tanaka H, Hamba
neuroendocrine tumors. In vivo applications and in vitro H, Kinoshita H, Ikebe T, Wakasa K (2000) Resection of a
studies. Q J Nucl Med 44:68-76 non -functioning islet cell carcinoma occupying the entire
Frilling A, Malago M,Martin H et al (1998) Use of somatostatin pancreas. Hepatogastroenterology 47:1168-1169
receptor scintigraphy to image extrahepatic metastases of Ogawa T, Isaji S, Yabana T (2000) A case of mixed acinar-
neuroendocrine tumors. Surgery 124:1000-1004 endocrine carcinoma of the pancreas discovered in an
Fugazzola C, Procacci C, Bergamo Andreis lA, Iacono C, Por- asymptomatic subject. Int J Pancreatol 27:249-257
tuese A, Mansueto G, Residori E, Zampieri P, Jannucci A, Oshikawa 0, Tanaka S, Ioka T, Nakaizumi A, Hamada Y,
Serio G (1990) The contribution of ultrasonography and Mitani T (2002) Dynamic sonography of pancreatic
computed tomography in the diagnosis of nonfunction- tumors: comparison with dynamic CT. Am J Roentgenol
ing islet cell tumors of the pancreas.Gastrointest Radiol 178:1133-1137
15:l39-144 Owen NJ, Sohaib SA, Peppercorn PD, Monson JP, Grossman
Furukawa H, Mukai K, Kosuge T, Kanai Y, Shimada K, Yama- AB, Besser GM, Reznek RH (2001) MRI of pancreatic neu-
moto J, Mizuguchi Y, Ushio K (1998) Nonfunctioning islet roendocrine tumours. Br J Radiol 74:968-973
cell tumors of the pancreas: clinical, imaging and patho- Pavone P, Mitchell DG, Leonetti F, Di Girolamo M, Cardone
logical aspects in 16 patients. Jpn J Clin Oncol 28:255-261 G, Catalano C, Tamburrano G, Passariello R (1993) Pan-
Hollett MD, Jorgensen MJ, Jeffrey RB Jr (1995) Quantitative creatic beta-cell tumors: MRI. J Comput Assist Tomogr 17:
evaluation of pancreatic enhancement during dual-phase 403-407
helical CT. Radiology 195:359-361 Petersein J, Spinazzi A, Giovagnoni A, Soyer P, Terrier F, Len-
Ichikawa T, Peterson MS, Federle MP, Baron RL, Haradome cioni R, Bartolozzi C, Grazioli L, Chiesa A, Manfredi R,
H, Kawamori Y, Nawano S, Araki T (2000) Islet cell tumor Marano P, Van Persijn Van Meerten EL, Bloem JL, Petre C,
of the pancreas: biphasic CT versus MR imaging in tumor Marchal G, Greco A, McNamara MT, Heuck A, Reiser M,
detection. Radiology 216: 163-171 Laniado M, Claussen C, Oaldrup HE, Rummeny E, Kirchin
Kato K, Kondo S, Ambo Y, Omi M, Hirano S, Morikawa T, MA, Pirovano G, Hamm B (2000) Focal liver lesions: evalu-
Okushiba S, Katoh H, Fujita M, Shimizu M (2000) Non- ation of the efficacy of gadobenate dimeglumine in MR
functioning endocrine tumor of the pancreas with extra- imaging - a multicenter phase III clinical study. Radiology
pancreatic growth and cyst formation: report of a case. 215: 727-736
Surg Today 30:651-654 Procacci C, Carbognin G, Accordini S, Biasiutti C, Bicego E,
Keogan MT, McDermott VG, Paulson EK, Sheafor DH, Freder- Romano L, Guarise A, Minniti S, Pagnotta N, Falconi M
ick MG, de Long DM, Nelson RC (1997) Pancreatic malig- (2001) Nonfunctioning endocrine tumors of the pancreas:
nancy: effect of dual-phase helical CT in tumor detection possibilities of spiral CT characterization. Eur Radiol 11:
and vascular opacification. Radiology 205:5l3-518 1175-1183
Kitajima T, Tomioka T, Tajima Y, Yamamoto 0, Sasaki M, Semelka RC, Ascher SM (1993) MR imaging of the pancreas.
Kanematsu T (1998) Small non-functioning endocrine Radiology 188:593-602
tumor of pancreas: comparison with solid cystic tumor. J Stafford-Johnson DB, Francis IR, Eckhauser FE, Knol JA,
Gastroenterol 33: 129-l33 Chang AE (1998) Dual-phase helical CT of nonfunctioning
Koito K, Namieno T, Nagakawa T, Morita K (1997) Delayed islet cell tumors. J Comput Assist Tomogr 22:335-339
enhancement of islet cell carcinoma on dynamic computed Van Hoe L, Gryspeerdt S, Marchal G, Baert AL, Mertens L
tomography: a sign of its malignancy. Abdom Imaging 22: (1995) Helical CT for the preoperative localization of islet
304-306 cell tumors of the pancreas: value of arterial and parenchy-
Krenning EP, Bakker WH, Kooij PPM et al (1992) Somatostatin mal phase images. Am J RoentgenoI165:1437-1439
receptor scintigraphy with [111In-DTPA-DPhe1]-octreo- Zeman RK, Silverman PM, Ascher SM, Patt RH, Cooper C,
tide in man: metabolism, dosimetry and comparison with. al-Kawas F (1995) Helical (spiral) CT of the pancreas and
[123I-Tyr3]-octreotide. J Nucl Med 33:652-658 biliary tract. Radiol Clin North Am 33:887-902
Rare Tumors
13 Rare Solid Tumors:
Clinical Manifestations and Pathology
T. OWElTY and A. B. WEST

CONTENTS 13.5.1 Teratoma 233


13.5.2 Choriocarcinoma 233
13.1 Introduction 219 13.6 Mesenchymal Tumors 233
13.2 Tumors Peculiar to the Pancreas 220 13.6.1 Lymphovascular Tumors 233
13.2.1 Pancreatoblastoma 220 13.6.1.1 Lymphangioma 233
13.2.1.1 Epidemiology 220 13.6.1.2 Hemangioma 234
13.2.1.2 Clinical Features 220 13.6.1.3 Hemangioendothelioma 234
13.2.1.3 Pathology 220 13.6.1.4 Other Lymphovascular Tumors 234
13.2.1.4 Differential Diagnosis 222 13.6.2 Adipose Tissue Tumors 234
13.2.1.5 Molecular/Genetic Alterations 222 13.6.3 Myogenic Tumors 234
13.2.1.6 Natural History and Prognosis 222 13.6.3.1 Leiomyoma 234
13.2.2 Acinar Cell Carcinoma 223 13.6.3.2 Leiomyosarcoma 234
13.2.2.1 Clinical Features 223 13.6.4 Neurogenic Tumors 236
13.2.2.2 Pathology 223 13.6.4.1 Schwannoma 236
13.2.2.3 Differential Diagnosis 225 13.6.4.2 Primitive Neuroectodermal Tumor 236
13.2.2.4 Prognosis and Treatment 226 13.6.5 Other Mesenchymal Tumors 237
13.2.3 Mixed Acinar-Endocrine Carcinoma 226 13.7 Solid Tumor-like Lesions of the Pancreas 237
13.2.3.1 Clinical Features 226 13.7.1 Heterotopic (Ectopic) Spleen 237
13.2.3.2 Pathology 226 13.7.2 Hamartoma 237
13.2.3.3 Prognosis 227 13.7.3 Pseudolipomatous Hypertrophy 238
13.2.4 Solid Pseudopapillary Tumor 227 13.7.4 Inflammatory Pseudotumors 238
13.2.4.1 Pathology 227 13.7.5 Pseudolymphoma 239
13.2.4.2 Treatment and Prognosis 227 13.7.6 Chronic Pancreatitis 239
13.2.5 Osteoclast-like Giant-Cell Tumor 227 13.7.7 Miscellaneous Inflammatory Tumor-like
13.2.5.1 Clinical Picture 227 Masses 241
13.2.5.2 Pathology 227 References 241
13.2.5.3 Treatment and Prognosis 229
13.3 Other Epithelial Tumors 230
13.3.1 Clear-Cell Carcinoma 230
13.3.1.1 Pathology 230
13.3.1.2 Prognosis 230 13.1
13.3.1.3 Differential Diagnosis 230 Introduction
13.3.2 Oncocytic Tumors 230
13.3.2.1 Pathology 231
13.3.2.2 Prognosis 231 The rare solid tumors that arise in the pancreas
13.3.3 Minor Epithelial Tumors 231 can be divided conveniently into two major groups:
13.4 Lymphomas 232 those that are unique to the pancreas and those that
13.4.1 Incidence and Epidemiology 232 are found also in other locations. The former group
13.4.2 Clinical Manifestations 232 includes pancreatoblastoma, pancreatic acinar cell
13.4.3 Pathology 232
13.4.4 Treatment and Prognosis 232 carcinoma, mixed acinar-endocrine cell carcinoma,
13.5 Germ-Cell Tumors 233 solid-pseudopapillary tumor, and osteoclast-like
giant-cell tumor. These tumors are the major focus of
this chapter. The latter group includes some clear-cell
T. OWElTY, MD carcinomas, oncocytic tumors, lymphomas, germ -cell
Assistant Professor, Department of Pathology, New York Uni- tumors, and mesenchymal tumors. These entities are
versity, 560 First Avenue (TH461), New York, NY 10016, USA familiar because of their more common occurrence
A.B. WEST, MD, FRCPath
Professor and Director of Anatomic Pathology, Department
in other sites, and they are considered here in large
of Pathology, New York University Medical Center, 560 First part because of their importance in the differential
Avenue (TH461), NewYork,NY lO016, USA diagnosis. The literature relating to many of these
220 T. Oweity and A. B. West

lesions arising in the pancreas is scant and composed been reported, except for one patient with Cushing's
chiefly of case reports, in the absence of reports of syndrome with inappropriate antidiuretic hormone
series. A third group of entities is also considered (SIADH) (PASSMORE et al. 1988).
here because of their importance in the differential
diagnosis of solid pancreatic masses: These are the 13.2.1.3
tumor-like lesions such as ectopic spleen, inflamma- Pathology
tory pseudotumor, and chronic pancreatitis.
The pathology of pancreatoblastomas has been
reviewed in detail by KLIMSTRA et al. (1995).

13.2 Macroscopic Features. Pancreatoblastomas are usu-


Tumors Peculiar to the Pancreas ally well-demarcated to encapsulated, although gross
extension into the adjacent pancreas or surrounding
13.2.1 tissue does occur. The reported diameter ranges from
Pancreatoblastoma 1.5 to over 20 em (mean 10.6 em). There is no predi-
lection to a particular location in the pancreas. The
13.2.1.1 cut surfaces are yellow to tan, soft to firm, with promi-
Epidemiology nent lobulation (Fig. 13.1a). Areas of hemorrhage
and necrosis can be seen, and calcifications may be
Although rare, this histologically distinct pancre- prominent (KLIMSTRA et al. 1995; CHUN et al. 1997;
atic neoplasm represents the most common malig- KOHDA et al. 2000). Cystic change occurs occasionally,
nant pancreatic tumor in childhood. The reported particularly in patients with Beckwith-Wiedemann
incidence among all pancreatic malignancies in syndrome (PASSMORE et al. 1988; KLIMSTRA et al.
the Memorial Sloan-Kettering series was 0.16% 1995; KOHDA et al. 2000).
(CUBILLA and FITZGERALD 1984). First described by
BECKER in 1957, it was referred to as infantile pancre- Microscopic Features. Although the tumors are usu-
atic carcinoma until the term pancreatoblastoma was ally partially surrounded by a fibrous capsule, there
introduced by HORIE in 1977 (BECKER 1957; FRABLE is often invasion into the surrounding pancreatic,
et al. 1971; HORIE et al. 1977). duodenal, or peripancreatic tissue. Vascular and
Almost half of the reported cases have been in perineural invasion are common.
Asians (KLIMSTRA et al. 1995; CHUN et al. 1997). The
majority occur in the first decade of life, with a mean On microscopic examination, pancreatoblastomas
age of 4.1 years and a slight male predominance. usually appear lobulated with an organoid pattern
Occasional cases have been reported in teenagers and consist of mixed epithelial and mesenchymal
(IMAMURA et al. 1998) and adults (18-56 years) components. The epithelial component exhibits
(HOORENS et al. 1994; DUNN and LONGNECKER 1995; three different patterns in variable proportions: solid
KLIMSTRA et al. 1995; LEVEY and BANNER 1996; sheets, acinar formations, and squamoid corpuscles
ROBIN et al. 1997). Pancreatoblastoma may be con- (Fig. 13.1b). The cells in the solid and acinar areas
genital and is sometimes associated with Beckwith- are similar cytologically, with round nuclei and
Wiedemann syndrome (KOH et a1.l986; POTTS et al. finely granular cytoplasm. The nuclear chromatin
1986; DRUT and JONES 1988). is clumped, and the cytoplasmic borders are usu-
ally better defined in the acinar areas than in the
13.2.1.2 solid areas. Occasional mild pleomorphism and an
Clinical Features increased nuclear-to-cytoplasmic ratio may occur in
the solid areas. Mitoses vary from rare to abundant.
Clinical symptoms are nonspecific and are usually Squamoid corpuscles are present in every case and
referable to the presence of an abdominal mass, range in size from a few cells to geographic areas.
although pain, weight loss, diarrhea, and obstructive They vary from ill defined to sharply defined clusters
jaundice are present occasionally (FRABLE et al. 1971; with a whorling pattern, and may show keratiniza-
GRIFFIN et al.1987; KLIMSTRA et al.1995; CHUN et al. tion. The nuclei in these areas are larger (often with
1997). Elevation of alpha-fetoprotein (AFP) occurs in central clearing), and the cytoplasm is more eosino-
up to 68% of patients (KLIMSTRA et al. 1995; KOHDA philic. In most cases, these squamoid corpuscles are
et al. 2000). No evidence of hormone secretion has plentiful, but occasionally they require more than
Rare Solid Tumors: Clinical Manifestations and Pathology 221

ally appear neoplastic; occasionally, they may show


more nuclear atypia with architectural disarray and
even chondroid or osteoid metaplasia (CUBILLA and
FITZGERALD 1984; KLIMSTRA et al. 1995).

Histochemical and Immunohistochemical Features.


Periodic acid-Schiff (PAS}-positive, diastase-resistant
granules can be seen in the acinar cells and squamoid
corpuscles. Mucicarmine positivity is detectable in
more than half of the patients (BUCHINO et al. 1984;
OHAKI et al. 1987; MOROHOSHI et al. 1987; KLIMS-
TRA et al. 1995). Pancreatoblastomas are thought to
a
originate from totipotent stem cells capable of dif-
ferentiating towards all three major cell types of the
adult pancreas: acinar, endocrine, and ductal, with
acinar differentiation being the one most consis-
tently present. These lines of differentiation can be
detected by routine light microscopy, by histochemi-
cal and immunohistochemical stains, and by electron
microscopy. The tumor cells usually express trypsin,
chymotrypsin, alpha-I-antitrypsin, and lipase, and
are also positive for cytokeratin (OHAKI et al. 1987;
MOROHOSHI et al. 1987; KISSANE 1994; KLIMSTRA et
al. 1995). Occasionally, a proportion of the cells will
stain for neuroendocrine markers such as neuron-
specific enolase (NSE), synaptophysin, and chromo-
granin, usually as scattered clusters or single cells
Fig. 13. 1a,b. Pancreatoblastoma. a The tumor has a fleshy, (KLIMSTRA et al. 1995; IMAMuRA et al. 1998). Rarely,
tan, lobulated cut surface with focal areas of hemorrhage some cells may stain for pancreatic hormones such as
and degeneration. The spleen is at the right. The patient was glucagon, (KLIMSTRA et al.1995) and insulin (IMAM-
a 6-year-old boy. At surgery, metastases were present in the
peritoneum and lungs. b The tumor cells display focal acinar URA et al.1998) or for nonpancreatic hormones (TSH,
arrangements, solid areas, squamoid corpuscles, and a mes- FSH) (HuA et al. 1996). Ductal differentiation is evi-
enchymal component that is present around the epithelial dent on routine histology and by mucicarmine stain-
clusters (H&E). Photograph in a by courtesy of Dr KATHLEEN ing, which usually highlights the luminal surfaces of
PATTERSON and Dr MIGUEL REYES-MUGICA. Case in b cour- the acinar/glandular structures with rare intracyto-
tesy of Dr DAVID KLIMSTRA
plasmic positivity. Carcinoembryonic antigen (CEA)
positivity is also commonly encountered and may
be seen in the squamous corpuscles, while CA 19.9
one histologic section to find them. Ductal structures or DUPAN-2 staining may occur focally in scattered
are usually inconspicuous, with a rare case showing clusters of cells. Double immunostaining for trypsin
abundant apical cytoplasm and ectatic lumens with and chromogranin reveals an intimate admixture of
tall columnar lining cells containing intracellular the two cell types with rare cells expressing both pro-
mucin. Occasionally, the growth pattern is more dif- teins. Staining for p53 is usually negative (KLIMSTRA
fuse than organoid, with increased pleomorphism, et al. 1995).
larger nuclei, higher nuclear-to-cytoplasmic ratios
and necrosis, and without squamoid corpuscles. The Ultrastructural Findings. Acinar differentiation
stromal component varies in quantity and appear- can be seen by demonstrating polygonal cells with
ance. It may be scant and fibrotic, particularly in easily identifiable desmosomes, abundant rough
adults, but usually it is composed of broad bands endoplasmic reticulum, and Golgi complexes, form-
that are hypercellular, especially in infants. The ing acinar/glandular structures with a surrounding
stromal cells generally resemble plump fibroblasts basement membrane, and luminal spaces exhibiting
with the nuclei arranged parallel to the long axis of apical tight junctions and blunt microvilli. Homog-
the band. Although hypercellular, they do not gener- enous, electron-dense, 400-800 nm, zymogen-like
222 T. Oweity and A. B. West

granules are seen in variable numbers and distribu- mones. Ductal adenocarcinomas are extremely rare
tion, but are most numerous in the apical cytoplasm in childhood and lack the characteristic acinar and
(FRABLE et al. 1971; BUCHINO et al. 1984; KLIMSTRA squamoid areas.
et al. 1995). Occasional, irregular, elongated, fibril- In general, pancreatoblastoma is the most likely
lary granules, mucin-like granules, and dense-core, diagnosis in a child presenting with a pancreatic
125-215 nm, neuroendocrine-like granules may be mass in the first decade of life without clinical hor-
present. However, none of the cells contain more monal abnormalities, particularly when the tumor is
than one specific granule type (BUCHINO et al. 1984; hypervascular, encapsulated, with calcifications and
KLIMSTRA et al. 1995). necrosis, and associated with raised alpha-fetopro-
tein (KOHDA et al. 2000).
Cytology. Fine needle aspirates of pancreatoblastoma
have rarely been described (SILVERMAN et al. 1990; 13.2.1.5
HENKE et al. 2001). The smears are cellular, with large Molecular/Genetic Alterations
clusters of cells and occasional single cells. The cells are
intermediate to large in size, round to spindle-shaped, ABRAHAM et al. (2001) analyzed nine pancreato-
some with a moderate amount of granular cytoplasm blastomas for alterations that are usually found in
and others with a high nuclear-to-cytoplasmic ratio, pancreatic ductal adenocarcinomas (K-ras oncogene
scant cytoplasm, and centrally placed nuclei. The chro- and p53 and DPC 4 tumor suppressor genes) and
matin is finely granular, with frequent inconspicuous for mutations common to other embryonic malig-
nucleoli. Mesenchymal fragments vary from rare to nancies [somatic alterations in the adenomatous
abundant in the background. Squamoid corpuscles polyposis coli (APC) /beta-catenin pathway and
have not been identified with certainty in smears, chromosome l1p]. Allelic loss on chromosome l1p
but sections of cell blocks show the characteristic was the most common genetic alteration, present in
histological features of solid, focally acinar cellular 8 of the reviewed cases (89%). Molecular alterations
arrangements and squamoid corpuscles with inter- in the APC/beta-catenin pathway were detected in
vening, small, round to spindle cells and scant stroma. 6 cases (67%). No alterations in the K-ras gene or
Ultrastructural preparations and immunohistochemi- p53 expression were detected. Loss of DPC 4 protein
cal stains help to confirm the diagnosis. expression was seen in 2 cases. These findings prove
the distinctive genetic nature of pancreatoblastoma,
13.2.1.4 which appears to be closer to hepatoblastoma in
Differential Diagnosis pathogenesis than to ductal adenocarcinomas of
the pancreas. Other abnormalities, including those
Acinar cell carcinoma may be difficult to differenti- involving chromosome 1, have been reported in
ate from pancreatoblastoma histologically, but usu- 2 cases (WILEY et al. 1995; NAGASHIMA et al. 1999).
ally occurs in adults and rarely in children, contrary In a separate study, KERR et al. (2002) confirmed the
to pancreatoblastoma. Squamoid corpuscles are the loss of heterozygosity for chromosome 11 p.
most distinctive morphologic feature of pancreato-
blastoma, as both tumors otherwise show acinar 13.2.1.6
differentiation and may show some degree of neu- Natural History and Prognosis
roendocrine marker positivity. Mixed acinar-endo-
crine tumors are also distinguished by the lack of Pancreatoblastomas are malignant neoplasms with
squamoid corpuscles. Solid-pseudopapillary tumor frequent local invasion, recurrence, and metastases.
usually shows papillary-pseudopapillary arrange- Lymph nodes, liver, and lung are the most common
ments and is positive for vimentin but negative sites of metastasis (KLIMSTRA et al. 1995; CHUN
for cytokeratin, in contrast to pancreatoblastoma. et al. 1997; IMAMURA et al. 1998). Of the patients
Although this tumor may occur at a younger age, reviewed by KLIMSTRA et al. (1995), 35% presented
it usually presents in the teenage years or later, and with metastases, and some patients developed them
in women. Neuroendocrine/islet-cell tumors may subsequently. In this series, 42% of the patients died
occur in childhood. They are usually functional, and of disease within 3.5 years of presentation, with a
the age of onset is the second decade (KOHDA et al. mean survival of 17 months. The prognosis in adults
2000). They lack acinar differentiation and squamoid appears worse than in children.
corpuscles, and are usually more diffusely positive The treatment of choice for pancreatoblastoma
for general neuroendocrine markers or specific hor- is complete surgical resection; resection of hepatic
Rare Solid Tumors: Clinical Manifestations and Pathology 223

metastases is sometimes performed also (KUMSTRA nated (mostly subcutaneous) fat necrosis, and occa-
et al. 1995; MURAKAMI et al. 1996; BENOIST et al. sionally fever with peripheral eosinophilia (GOOD et
2001). Neoadjuvant and postoperative chemotherapy al. 1976; KLIMSTRA et al. 1992). These features are
have been applied with good results, with radiation mostly associated with, and probably related to, high
for unresectable local recurrences (GRIFFIN et al. serum lipase (KUMSTRA et al. 1992) or trypsin (VAN
1987; KUMSTRA et al. 1995; MURAKAMI et al. 1996; KLAVEREN et al. 1990), while alpha-amylase is rarely
IMAMURA et al. 1998). Serum alpha -fetoprotein, when elevated (GOOD et al. 1976). An increase in serum
elevated, is helpful in monitoring patients following lipase, amylase, or elastase-l without concomitant
surgery (KUMSTRA et al. 1995; CHUN et al. 1997; symptoms has been reported (ISHIHARA et al. 1989).
OGAWA et al. 2000). The prognosis is generally favor- With rare exceptions (HORlE et al. 1984), serum CEA
able in resectable cases, and a substantial number of and CA 19.9 are normal, and there are reported
patients have survived long enough to be considered cases of elevated alpha-fetoprotein (ITOH et al. 1992;
cured (KUMSTRA et al. 1995; IMAMURA et al. 1998; KAWAMOTO et al. 1992). Cases associated with hypo-
OWAGA et al. 2000; BENOIST et al. 2001; DEFACHELLES glycemia (MIZUTA et al. 1998), myeloma-like cast
et al. 2001). nephropathy (REDUCKA et al. 1988), and thrombotic
endocarditis (WEBB 1977) have been described.
13.2.2
Acinar Cell Carcinoma 13.2.2.2
Pathology
Acinar cell carcinomas are rare tumors of the exo-
crine pancreas with evidence of acinar cell differenti- Macroscopic Features. There is no clear preferential
ation and occasional endocrine components (SOLCIA localization in the pancreas, although more are found
et al. 1997). The majority are solid lesions and will in the head. The tumor is usually large, ranging in
be covered in this chapter. Cystic variants, includ- diameter from 2 to 30 em (mean 10 em) and occasion-
ing acinar cell cystadenocarcinoma and acinar cell ally appears attached to, rather than arising within, the
cystadenoma, are discussed in the chapter on cystic pancreas. It consists of well circumscribed to partially
tumors. Acinar cell carcinoma accounts for 1%-2% of encapsulated masses. The cut surface is soft to friable,
all primary nonendocrine pancreatic tumors (MoRO- yellow to red, separated into lobules by fine fibrous
HOSHI et al. 1983; CUBILLA and FITZGERALD 1984; strands, and often showing necrosis and hemorrhage
CHEN et al. 1985). and rarely focal cystic change (Fig. 13.2a). Invasion of
adjacent organs is seen in some cases (MOROHOSHI et
13.2.2.1 al. 1987; KUMSTRA et al. 1992; HOORENS et al. 1993).
Clinical Features
Microscopic Features. Most acinar cell carcinomas
Acinar cell carcinomas can occur at any age (range appear well circumscribed at low power and often sur-
3-90 years), but they are more common during rounded by a partial or complete capsule, but nearly all
the 5th through the 7th decades of life. They are show evidence of at least focal capsular invasion, with
very rare in children, with few examples recorded broad nodules of tumor invading into adjacent tissue.
in the literature (OSBORNE et al. 1977; LACK et al. Broad fibrous bands divide the tumors into lobules
1983; MOROHOSHI et al. 1987; KUMSTRA et al. 1992; that are markedly cellular with scant stroma. A vari-
HOORENS et al. 1993). Male predominance was noted ety of growth patterns may be seen, most commonly
in two series (KLIMSTRA et al. 1992; HOORENS et al. mixed, including acinar, solid, glandular, and trabecu-
1993). Although the majority of cases in KUMSTRA'S lar patterns (KUMSTRA et al. 1992) (Fig. 13.2b).
series affected Caucasian patients, overall, no racial
predominance has been noted (ORDONEZ 2001; The acinar pattern is the most characteristic pat-
SOLCIA et al. 1997). tern of growth. It is present at least focally in most
Symptoms at presentation are nonspecific and tumors. It consists of cells growing in well-formed
related to local expansion or metastases (CUBILLA acini, having frequently identifiable small lumina
and FITZGERALD 1984). The ones most commonly with sharp borders. The solid pattern of growth is
described are abdominal pain, loss of weight and also common, with sheets and cords of cells sepa-
appetite, and vomiting. Jaundice is rare. Some rated by a thin fibrovascular stroma. Less commonly,
patients (16% in KUMSTRA'S series) present with a a glandular pattern is seen, probably reflecting
syndrome characterized by polyarthralgia, dis semi- dilated acinar structures, with eosinophilic secretory
224 T. Oweity and A. B. West

material within the lumen. A trabecular pattern with Histochemical Stains. Periodic acid-Schiff (PAS)
double rows of cells is infrequently seen. The cells staining after diastase digestion (PASD) character-
of acinar cell carcinomas are cuboidal or columnar istically demonstrates variably positive cytoplasmic
with eosinophilic granular cytoplasm, which is most granules which are best seen in the apical cytoplasm
abundant in the acinar areas, while scant in the solid of cells in the acinar areas. Occasionally, PAS-positive
areas (Fig. l3.2c). The nuclei are usually uniform, material is seen in the glandular/acinar lumina, and
normochromatic, round to oval, with rare pleomor- rarely mucin, although no intracytoplasmic mucin is
phism which occurs most often in the solid areas. In present. Butyrase stains, which define lipase activity,
acinar areas, the nuclei are polarized basally, result- are positive in more than 70% of cases (KLIMSTRA
ing in a palisading pattern bordering the stroma or et al. 1992).
the capsule. The nuclei are usually centrally located
in the solid areas. Single, small, prominent nucleoli Immunostains. A variety of pancreatic enzymes are
are commonly seen, centrally or peripherally. Mitotic expressed in acinar cell carcinomas and may be
counts are variable between different tumors and dif- detected immunohistochemically. Trypsin is the one
ferent areas, ranging from none to more than 50110 most commonly expressed, occurring in up to 97% of
high power fields, and occasionally atypical forms are patients, and the one most useful in making a diag-
present (KLIMSTRA et al. 1992; ORDONEZ 2001).

a b

c .... -...:;_~_

Fig. l3.2a-d. Acinar cell carcinoma. a The tumor is circumscribed and solid with a tan to red cut surface and foci of hemor-
rhage. b The tumor cells are arranged in solid masses with scattered acini, most conspicuous here at the top right of the field.
A duct or dilated acinar structure is seen on the left (H&E, low power). c The acini are lined by cuboidal cells with granular,
eosinophilic apical cytoplasm and uniform round nuclei, some with small prominent nucleoli (H&E, high power). d The cells
are arranged singly and in acinar groups, with eosinophilic cytoplasm, uniform round nuclei, and small punctate nucleoli (fine-
needle aspirate, H&E). Case courtesy of Dr PETER ILLEI
Rare Solid Tumors: Clinical Manifestations and Pathology 225

nosis (MOROHOSHI et al. 1987; KUMSTRA et al. 1992; elongated in shape. Some have granular or flocculent
ORDONEZ 2001). Lipase has been reported to occur contents, while others are pleomorphic and may con-
in 85% of patients, chymotrypsin in 66%, and amy- tain fibrillary internal structures resembling zymo-
lase inl4% (ORDONEZ 2001). In general, enzymes are gen granules of fetal pancreas. They are commonly
more frequently recognized in areas of tumor having oriented toward the lumen in the acinar areas and
an acinar growth pattern and are frequently local- are scanty in the solid areas (KUMSTRA et al. 1992;
ized to the apical cytoplasm (KUMSTRA et al. 1992). TUCKER et al. 1994; ORDONEZ 2001).
Alpha -I-antitrypsin and alpha-l-antichymotrypsin
are expressed by some acinar cell carcinomas, but Molecular Analysis. It appears that acinar cell carci-
they appear to be neither sensitive nor specific as they noma is genetically distinct from ductal adenocarci-
also occur in other pancreatic and extrapancreatic noma. No overexpression of p53 protein or molecular
tumors (ORDONEZ and MANNING 1984; LEADER et al. evidence of p53 mutation is seen (TERHUNE et al.
1987; MOROHOSHI et al. 1987; KUMSTRA et al. 1992; 1998; HOORENS et al. 1993; ABRAHAM et al. 2002),
NOJIMA et al. 1992). only rare cases show the characteristic ductal Ki-ras
mutation at codon 12 (HOORENS et al.1993; MOORE et
Acinar cell carcinomas react with the broad-spec- al. 2001),and no loss ofDPC4 or p16 is found (MOORE
trum anti-cytokeratin antibodies AEl! AE3 and CAM et al. 2001). In contrast, allelic loss on chromosome
5.2, and some express epithelial membrane antigen 11 p occurs in up to 50% of patients, and molecular
(EMA). Markers expressed commonly by ductal alterations in the APC/ beta-catenin pathway are
carcinoma, such as CEA and CA 19.9, may occasion- present in approximately 25% of patients, genetic
ally be positive (KUMSTRA et al. 1992; HOORENS alterations held in common with pancreatoblastoma
et al. 1993). Vimentin staining in the tumor cells is (ABRAHAM et al. 2002).
negative (ORDONEZ 2001). Alpha-fetoprotein (AFP)
expression, which is rare in pancreatic tumors other Cytology. Few reports describe the fine-needle
than pancreatoblastoma, has been reported in some aspirate features of these tumors. Cytomorphologi-
cases (KUMSTRA et al.1992; HOORENS et al.1992) and cally, the smears show loose clusters or single cells
is occasionally associated with elevated serum AFP with granular eosinophilic cytoplasm and eccentric
levels (NOJIMA et al. 1992). Scattered, nondescript nuclei with prominent nucleoli, some forming acinar
endocrine cells expressing chromogranin A and syn- structures (ISHIHARA et al. 1989; LABATE et al. 1997)
aptophysin occur in about 40% of acinar cell carcino- (Fig. l3.2d).
mas and occasionally may be shown to contain pan-
creatic hormones such as glucagon (KUMSTRA et al. 13.2.2.3
1992; HOORENS et al. 1993). Only rarely are exocrine Differential Diagnosis
and endocrine markers demonstrable in the same
cells by double staining (HOORENS et al. 1993). The main tumors in the differential diagnosis are
ductal adenocarcinoma, endocrine tumors, solid-
Electron Microscopy. Ultrastructural features of pseudopapillary tumor, and pancreatoblastoma.
acinar cell carcinomas resemble those of normal Ductal adenocarcinoma is easily separated from
acinar cells, particularly in the areas with acinar acinar cell carcinoma (SOLCIA et al. 1997). Macro-
patterns where the cells possess microvilli at the scopically, those tumors are usually firm, ill defined,
apical surfaces with tight junctions and desmosomes predominantly occurring in the head of the pancreas.
joining adjacent cells (KLIMSTRA et al. 1992). Abun- Histologically, they consist of duct-like glands embed-
dant rough endoplasmic reticulum, well developed ded in desmoplastic stroma. The cells stain for mucin
Golgi complexes, some mitochondria, and a few and for CEA and CA19.9 immunohistochemically.
lysosomes are seen, in addition to the characteristic They are usually negative for pancreatic enzymes
zymogen granules (CUBILLA and FITZGERALD 1984; (MOROHOSHI et a1.1983; HOORENS et al.1993).
MOROHOSHI et al. 1987; DI SANT'AGNESE 1991; Endocrine tumors have a better prognosis than
KUMSTRA et al. 1992; HOORENS et al. 1993). These acinar cell carcinoma, and it is important to distin-
electron-dense granules lack a halo between their guish them. This distinction may be difficult at the
homogenous dark-staining contents and their limit- light microscopic level when acinar cell carcinoma
ing membranes, and resemble zymogen granules of displays a solid and trabecular pattern.
the normal pancreas but with a wider size range (75 Histologic features favoring endocrine tumors
to > 1000 nm). They may be round, ovoid, fusiform, or include: absence of foci of acinar differentiation,
226 T. Oweity and A. B. West

more regular arrangement of the nuclei in the solid 13.2.3


areas, absence of intracellular PAS-positivity, and Mixed Acinar-Endocrine Carcinoma
more distinct demarcation from the surrounding
normal tissue along with denser, unevenly distrib- Although scattered endocrine cells may be seen in
uted fibrous stroma. Immunohistochemically, endo- up to 40% of acinar cell carcinomas (KUMSTRA et
crine tumors are diffusely positive for markers such al. 1992; HOORENS et al. 1993), truly mixed acinar-
as synaptophysin and chromogranin A, and negative endocrine tumors are extremely rare. They are
for pancreatic enzymes such as trypsin and lipase. defined as tumors in which the acinar or endocrine
Ultrastructurally, they have typical dense core neuro- components are significant, constituting over 25%
secretory granules, 100-200 nm in diameter, and they (KUMSTRA et al. 1994) or 30%-50% of the tumor,
lack zymogen granules and acinar differentiation. with the two cell populations intimately admixed
Solid-pseudopapillarytumors have a favorable prog- with each other (SOLCIA et al. 1997). Only rare cases
nosis and a predilection for young women, although have been reported, and terms such as mixed acinar-
they can occur at any age. Macroscopically, they often endocrine carcinoma (KUMSTRA et al. 1994), mixed
show cystic degenerative changes, and microscopically, exocrine-endocrine tumor (HASSAN and GOGATE
they are characterized by a solid-pseudopapillary 1993), and acinar-endocrine cell tumor (UUCH et
pattern of growth with distinct fibrovascular myxoid al. 1982) have been used. These tumors provide fur-
stroma and monomorphic cells, which have clear ther evidence of the close histogenetic relationship
to faintly eosinophilic cytoplasm and often nuclear between the endocrine and exocrine components of
grooves. Immunohistochemically, these tumors are the pancreas. Their existence suggests that neoplastic
positive for vimentin and negative for cytokeratin cells of the pancreas may have the potential for dual
CAM 5.2. They are also diffusely positive for NSE and differentiation, reflecting the common embryologic
CDs6, with strong focal positivity for alpha-I-antitryp- origin of the endocrine and exocrine components
sin. They are usually negative for chromogranin and (KLOPPEL 2000).
mostly negative for pancreatic enzymes (MIETTINEN
et al.1987; MOROHOSHI et al.1987). They are discussed 13.2.3.1
in detail in the chapter on cystic tumors. Clinical Features
Pancreatoblastomas enter the differential diagnosis
with acinar cell carcinomas when they occur in adults In the small series of five patients reported by
or when acinar cell carcinomas occur in children. KUMSTRA, the tumors occurred in adult men and
women, age range 48-81 years (mean 68 years). None
13.2.2.4 presented with symptoms related to either enzyme or
Prognosis and Treatment hormone production (KUMSTRA et al. 1994).

Acinar cell carcinomas are aggressive tumors 13.2.3.2


that have a poor prognosis. More than half of the Pathology
patients present with metastases at the time of the
diagnosis, mostly to regional lymph nodes or liver, The tumors were fairly well circumscribed and histo-
with rare extraabdominal spread in advanced cases logically very cellular, with various combinations of
(WEBB 1977; MOROHOSHI et al. 1987; KUMSTRA et solid, trabecular, acinar, and glandular growth pat-
al. 1992; HOORENS et al. 1993). The reported overall terns, the solid pattern predominating. Only in one
mean survival is 18 and 42 months in different series, patient were the endocrine and exocrine components
patients who are younger at presentation enjoying distinct under light microscopy, while in the others,
longer survival periods, the longest reported survival the cells were morphologically uniform with round
being 90 months (KUMSTRA et al. 1992; HOORENS to oval nuclei and prominent nucleoli, with divergent
1993). Among all pathological parameters, only a differentiation detected only immunohistochemically.
tumor diameter less than 10 cm was found to influ- The cells contained diastase-resistant PAS-positive
ence survival in KUMSTRA'S series (KUMSTRA et al. granules and showed immunohistochemical positivity
1992). Along with surgical resection, radiation and for pancreatic enzymes (trypsin, chymotrypsin, and
chemotherapy have a palliative role, with an occa- focally lipase) and endocrine markers (chromogranin
sional good response to chemotherapy in patients and synaptophysin) with specific endocrine hormones
with liver metastases (VAN KLAVEREN et al. 1990; (glucagon, somatostatin, gastrin, vasoactive intestinal
KUMSTRA et al. 1992). polypeptide, VIP) found in two cases. Double immu-
Rare Solid Tumors: Clinical Manifestations and Pathology 227

no staining showed only one line of differentiation to chemically, the tumor cells are positive for vimen-
(acinar or endocrine) in most cells, with few cells tin (Fig. 13.3c), NSE, alpha-I-antitrypsin, CD10 and
showing both reactions (amphicrine). CD56, and negative for cytokeratin.
Ultrastructurally, there was an admixture of cells Cytologically, fine-needle aspirate smears show
showing acinar and endocrine differentiation. Two single cells and papillary fragments with fibrovas-
populations of granules were detected: neurosecre- cular stroma, which appears metachromatic on Diff-
tory dense core type, 100-130 nm in diameter, and Quik stain, surrounded by cells with uniform, often
zymogen granules, 250-525 nm in diameter. Many of grooved nuclei, and wispy, ill-defined to eosinophilic
the cells showed intermediate features with frequent cytoplasm (WILSON et al.1992) (Fig. 13.3d-f).
variably sized granules of indeterminate type.
13.2.4.2
13.2.3.3 Treatment and Prognosis
Prognosis
Surgery is the treatment of choice, and experience
The prognosis appears to be similar to that of acinar with adjuvant therapy is limited. Complete resection
cell carcinoma. Four of the five patients developed should be attempted, even if this requires resection
metastases within 1 year, and 2 died of their disease of metastases, as long-term survival can be achieved
(KUMSTRA et al. 1994). even following the development of metastatic disease.
At a median follow-up of 8 years, only one recurrence
was seen in 18 patients who underwent complete
13.2.4 resection (MARTIN et al. 2002).
Solid-Pseudopapillary Tumor

These uncommon tumors are frequently cystic and 13.2.S


are reviewed in greater detail in the chapter on cystic Osteoclast-like Giant-Cell Tumor
tumors. First described by FRANTZ in 1959 (FRANTZ
1959), they are tumors of low malignant potential Also known as giant-cell tumor of the pancreas,
that may occur at any age, but primarily affect young giant-cell carcinoma of the pancreas osteoid type,
women (KLOPPEL et al.1981). They have low metastatic giant-cell tumor (osteoclastic) of the pancreas, these
potential, metastases most commonly occurring in the are rare tumors of the pancreas that simulate giant-
liver. They have been reported under various names cell tumors of bone. First described by ROSAI in 1968,
describing their variable gross and microscopic fea- they are defined histologically by the presence of
tures, which include: solid and cystic (KLOPPEL et al. multinucleate osteoclast-like giant cells admixed with
1981), papillary-cystic (BOOR and SWANSON 1979), and proliferating oval to spindle-shaped, mononuclear
solid and papillary (CUBILLA and FITZGERALD 1984). cells (ROSAI 1968).
The name solid-pseudopapillary tumor encompasses
the most characteristic histologic features: the solid 13.2.5.1
and pseudopapillary regions (SOLCIA et al. 1997). Clinical Picture

13.2.4.1 The patients' age range is 3-82 years with a mean


Pathology age of 60 years. Women are more frequently affected
than men. The main symptoms are abdominal pain,
Macroscopically, the tumor varies from solid to weight loss, jaundice, and a palpable mass (MANCI et
frequently cystic due to extensive hemorrhage and al. 1985; SOLCIA et al. 1997).
degeneration (Fig. 13.3a).
Microscopically, it is characterized by uniform 73.2.5.2
cells, with often indented, hypo chromatic nuclei, and Pathology
clear to faintly eosinophilic cytoplasm, arranged in
solid and pseudopapillary patterns. In the latter, the Macroscopic Features. Most tumors appear in the
cells tend to degenerate away from the fibrovascular head of the pancreas and present as a large mass,
stroma and gradually form cystic spaces with foamy averaging 11 cm in diameter, with a rubbery, firm
macrophages and cholesterol clefts (SOLCIA et al. consistency and a lobulated, white to yellow, focally
1997; KUMSTRA et al. 2000) (Fig. 13.3b). Immunohis- necrotic cut surface. Invasion into adjacent organs is
228 T. Oweity and A. B. West

a b

c d

e f
Fig.13.3a-f. Solid-pseudopapillary tumor. a This tumor is mostly solid with a tan, granular surface, but some areas of degenera-
tion and cystic change (right and left). b The tumor cells are arranged around fibrovascular cores with a papillary appearance.
The cells away from the cores degenerate and falloff (H&E). c The tumor cells are diffusely positive for vimentin (brown),
highlighting the papillary arrangement (vimentin immunostain). d Papillary fragments have fibrovascular cores surrounded
by tumor cells, which are also present dissociated in the background (fine-needle aspirate, Papanicolaou stain, low power). e
The tumor cells are bland without significant atypia and with occasional nuclear grooves (fine-needle aspirate, Papanicolaou
stain, high power). f The fibrovascular cores appear metachromatic, with and without surrounding tumor cells (fine-needle
aspirate, Diff-Quik stain)
Rare Solid Tumors: Clinical Manifestations and Pathology 229

common, but metastases are found in only 50% of


patients (MANCI et al. 1985; SOLCIA et al. 1997).

Microscopic Features. The tumors are composed of


two populations of cells. The first population is a
proliferation of mononuclear cells of oval to spindle
shape, with pleomorphic-hyperchromatic nuclei,
distinct nucleoli, and faintly eosinophilic cytoplasm.
Mitoses can be abundant. The second distinctive
population is of multinucleate giant cells containing
abundant dense eosinophilic cytoplasm and 20 to
100 small, uniform nuclei with prominent nucleoli,
resembling osteoclasts (Fig. 13.4). In some of the
cases, pleomorphic, malignant-appearing, bizarre,
multinucleated giant cells may also be present
Fig. 13.4. Osteoclast-like giant-cell tumor. Osteoclast-like cells
(LEWANDROWSKI et al. 1990; MULLICK and MOODY with numerous nuclei are disposed among spindle-shaped
1996; DECKARD-JANATPOUR et al. 1998). mononuclear cells with pleomorphic hyperchromatic nuclei
and occasional bizarre giant cells. The pigment is hemosiderin
Some osteoclast-like giant-cell tumors are associ- (H&E). Case courtesy of Dr DIVA SALOMAO
ated with nearby areas of ductal hyperplasia or atypia
(GOLDBERG et al. 1991), others contain foci of ductal
adenocarcinoma (CUBILLA and FITZGERALD 1984), as monohistocytic!mesenchymal cells for vimentin,
and yet others lack neoplastic glands (ROSAI 1968; leukocyte common antigen (LCA), lysosomes, CD-68,
SUSTER et al.1989; NEWBOULD et al.1992; WESTRA et al. alpha-1-antichymotrypsin, and occasionally smooth
1998; MACHADO et al. 2001). A few have been reported muscle actin, and mostly lack staining for epithelial
arising in the wall of a mucinous cystic neoplasm markers (LEWANDROWSKI et al. 1990; DECKARD-
(POSEN 1981; AOKI et al.1989; MENTES and YUCE 1993). JANATPOUR et al. 1998; SAKAI et al. 2000; MACHADO
In some, foci of osteoid formation are present (CUBILLA et al. 2001), although the mononuclear and other
and FITZGERALD 1984; KAY and HARRISON 1969). pleomorphic giant cells in the same tumors may show
epithelial differentiation (DECKARD-JANATPOUR et al.
Electron Microscopy, Immunohistochemistry, and 1998; IMAI et al. 1999; SAKAI et al. 2000), suggesting
Histogenesis. Conflicting results have been published that osteoclast-like giant cells may represent a reactive
in the literature demonstrating epithelial and/or rather than a neoplastic component of the tumor.
mesenchymal features of this tumor and reflecting
the uncertainty about its histogenesis, which could Molecular Analysis. K-ras mutation involving codon
theoretically be explained by divergent differentiation 12.13 has been detected in osteoclast-like giant-cell
from a common precursor cell (LEWANDROWSKI et al. tumors, supporting a ductal origin (DECKARD-
1990). Epithelial acinar differentiation was suggested JANATPOUR et al. 1998; IMAI et al. 1999; SAKAI et al.
initially by ROSAI (1968), based on the findings of 2000). In some reports, these mutations were localized
microvilli, desmosomes, and zymogen-like granules. specifically to the osteoclastic giant cells, suggesting
This was supported later by immunohistochemical that this may be due to phagocytosis of mononuclear
data showing positivity for epithelial markers such as tumor cells (WESTRA et al. 1998), while in another
cytokeratin, epithelial membrane antigen (EMA), CEA, report, the mutation was demonstrated to occur in
and CA 19.9 (TREPETA et al.1981; BERENDT et al.1987; the other neoplastic cells but not in the osteoclastic
NOJIMA et al. 1993; DECKARD-JANATPOUR et al. 1998; giant cells (SAKAI et al. 2000). Whether these cells are
IMAI et al. 1999; LEIGHTON and SHUN 2001). Mesen- neoplastic due to the fusion of mononuclear cells or
chymal differentiation was suggested by some reports reactive is still a matter of debate.
documenting positivity for vimentin in the absence
of keratin and of ultrastructural epithelial features in 13.2.5.3
the mononuclear and osteoclast-like cells (SUSTER et Treatment and Prognosis
al. 1989; LEWANDROWSKI et al. 1990; GOLDBERG et al.
1991; DWORAK et al. 1993; MACHADO et al. 2001). The Complete surgical resection is the treatment of
osteoclast-like giant cells have been found to stain choice; experience with radiotherapy or chemo-
230 T. Oweity and A. B. West

therapy has not been reported (LEIGHTON and tion of a K-ras mutation at codon 12, which is seen
SHUN 2001). Although many of these tumors have in more than 90% of pancreatic ductal carcinomas,
been found to be aggressive with a behavior similar is further supporting evidence of a ductal phenotype
to ductal adenocarcinoma and expected survival of (LUTTGES et al. 1998).
less than 1 year (DWORAK et al. 1993; LEIGHTON and
SHUN 2001), occasional cases with long survival (up 13.3.1.2
to 15 years) have been reported (JEFFREY et al. 1983; Prognosis
CUBILLA and FITZGERALD 1984; DWORAK et al.1993;
MACHADO et al. 2001). Prolonged survival has been The few reported cases of clear-cell carcinoma of the
related to a total absence of epithelial/ductal dif- pancreas have metastasized and behaved poorly, sim-
ferentiation on both routine light microscopy and ilar to other pancreatic ductal carcinomas (CUBILLA
immunohistochemistry (MACHADO et al. 2001). This and FITZGERALD 1984; KANAI et al. 1987; LUTTGES
probably reflects the presence of a spectrum of more et al. 1998).
and less aggressive tumors displaying osteoclast-like
giant cells, including at one end the highly aggres- 13.3.1.3
sive pleomorphic giant-cell carcinoma (CUBILLA and Differential Diagnosis
FITZGERALD 1984; MACHADO et al. 2001).
The first consideration on encountering a clear-cell
carcinoma in the pancreas is a metastasis from an
extrapancreatic primary tumor, particularly renal
13.3 cell carcinoma which has a tendency to develop
Other Epithelial Tumors solitary late metastases (THOMPSON and HEFFESS
2000). The presence of an intraductal component
13.3.1 is helpful in excluding metastasis. Immunostaining
Clear-Cell Carcinoma positivity for cytokeratins and CEA, and negativity
for vim en tin and for other renal cell carcinoma mark-
Focal clear-cell change is occasionally seen in ductal ers (RCC, CD 10) help in establishing the diagnosis of
or anaplastic pancreatic carcinomas (URBANSKI and primary pancreatic carcinoma, supported by clinical
MEDLINE 1982), but tumors composed predomi- and radiologic findings.
nantly of clear cells are rare (CUBILLA and FITZGER- Other pancreatic neoplasms with clear-cell fea-
ALD 1984; KANAI et al. 1987; LUTTGES et al. 1998). tures include endocrine tumors (GUARDA et al. 1983;
SOLCIA et al. 1997), solid-pseudopapillary tumor,
13.3.1.1 solid serous cystadenoma (PEREZ-ORDONEZ et al.
Pathology 1996), and sugar tumor (ZAMBONI et al. 1996). The
last of these has been described only once in the
Macroscopically, these are solid tumors; microscopi- pancreas. Solid serous cystadenoma is negative for
cally, they consist of sheets and nests of cells with CEA; sugar tumor (a mesenchymal tumor with epi-
slightly pleomorphic nuclei and clear glycogen-rich thelioid cells characterized by an abundant vascular
cytoplasm. These tumors resemble the more com- sinusoidal network) is positive for vimentin and
monly encountered clear-cell tumors of the kidney, HMB 45. Negativity for neuroendocrine markers
adrenals, and lung. No sinusoidal/vascular pattern, such as chromogranin and synaptophysin excludes
necrosis, or distinct desmoplastic reaction are noted a neuroendocrine tumor, and negativity for NSE,
(LUTTGES et al. 1998). Evidence of ductal origin has alpha-I-antitrypsin, and vimentin excludes solid-
been found in some cases, including intracytoplas- pseudopapillary tumor.
mic mucin, elevated serum levels of CEA and CA 19.9,
and focal positive staining for CEA (CUBILLA and
FITZGERALD 1984; KANAI et al. 1987; LUTTGES et al. 13.3.2
1998). Additionally, an intraductal papillary compo- Oncocytic Tumors
nent was detected in the case described by LUTTGES
(LUTTGES et al. 1998). In that case, the tumor cells Tumors with oncocytic change in the pancreas are
showed positive staining for p53 and cytokeratins 7, otherwise well-defined entities such as endocrine
8, 18, and 19, but were negative for neuroendocrine neoplasms (GOTCHALL et al. 1987; PACCHIONI et al.
markers and vimentin. This, along with documenta- 1996), solid-pseudopapillary tumor (LEE et al. 1993),
Rare Solid Tumors: Clinical Manifestations and Pathology 231

or acinar cell carcinoma packed with zymogen gran- reported by NOZAWA et al. (1990) showed perineural!
ules (PAPOTTI et al. 1999). Pure oncocytic tumors in vascular invasion but lacked mitoses or nuclear pleo-
the pancreas are extremely rare (HUNTRAKOON morphism and was stable in size without metastases
1983; CHEN and BAIT HUN 1985; BONDE SON et al. at the 3-year follow-up.
1990; NOZAWA et al. 1990; SIRONI et al. 1991; ZERBI
et a1.1993). They are considered to be of ductal origin
(PAPOTTI et al. 1999). 13.3.3
Minor Epithelial Tumors
73.3.2.7
Pathology Other rare tumors have been described, most of
which represent variants of ductal carcinoma, e.g.,
Macroscopically, they may be large and encapsulated mucoepidermoid carcinoma (ONODA et al. 1995),
with a white-tan firm cut surface (Fig. 13.5a) and may ciliated carcinoma (SOMMERS and MEISSNER 1954;
exhibit cystic change (HUNTRAKOON 1983; NOZAWA MORINAGA et al. 1986), and carcinosarcoma (MILLIS
et al. 1990; THOMPSON et al. 1998). et al. 1994).
Microscopically, the tumor cells are large with
abundant, finely granular eosinophilic cytoplasm, and
uniform nuclei with prominent nucleoli (Fig. 13.5b).
The growth pattern is solid (HUNTRAKOON 1983;
NOZAWA et al. 1990), nested (CHEN and BAIT HUN
1985), or papillary (THOMPSON et al. 1998). Intra-
ductal papillary carcinomas with predominantly
oncocytic features have also been described (ADSAY
et al. 2000) where arborizing papillae lined by strati-
fied oncocytic cells may be confluent, forming large
sheets and imparting a pseudosolid pattern of
growth.
Ultrastructurally, the cells are packed with mito-
chondria but lack zymogen and neurosecretory
granules. Immunohistochemistry confirms the lack of
endocrine and acinar markers, and positivity for cyto-
keratin (CAM 5.2) and CEA further supports a ductal
origin (NOZAWA et al.1990; THOMPSON et al. 1998).
Cytologically, the cells obtained by fine-needle aspi- a
rates are uniform, epithelioid, clustered, disassociated,
or in papillary fragments. They have abundant well-
defined cytoplasm, which appears tan, gray-blue on
the May-Grunwald-Giemsa and Diff-Quik stains, and
granular eosinophilic on H&E stains. The nuclei are
eccentric, round, regular with single, prominent nucle-
oli (BONDESON et al. 1990; THOMPSON et al. 1998).
The differential diagnosis includes other acinar,
endocrine, and solid-pseudopapillary tumors of the
pancreas. It also includes extrapancreatic tumors
which are much more likely to have oncocytic features
such as those arising in the kidney or thyroid.

73.3.2.2
Prognosis Fig.13.5a,b. Oncocytic tumor. a The tumor has a yellowish-tan
cut surface with a central scar resembling renal oncocytoma.
b The tumor has a solid lobular architecture and is composed
The behavior of oncocytic tumor is uncertain although of cells with abundant, granular, eosinophilic cytoplasm,
usually malignant (HUNTRAKOON 1983; SIRONI et al. uniform nuclei, and prominent nucleoli (H&E). Photographs
1991; ZERBI et al. 1993; THOMPSON et al. 1998). A case courtesy of Dr RICHARD EISEN
232 T. Oweity and A. B. West

13.4 The diagnosis of lymphoma in most cases can be


Lymphomas established without surgery, using a percutaneous or
endoscopic, CT- or ultrasound -guided, fine- needle or
13.4.1 core biopsy (CARTER et al.1988; ROBBINS et al.1995).
Incidence and Epidemiology Immediate assessment of the aspirate at the time of
the procedure is critical to ensure proper handling
Although secondary involvement of the pancreas of the specimen. When a preliminary diagnosis of
is seen in more than 30% of patients with non- lymphoma is formed, sufficient sample for ancillary
Hodgkin's lymphoma (BEHRNS et al. 1994), primary studies should be obtained for confirmation. These
pancreatic lymphoma is rare, comprising less than may include immunophenotyping by flow cytometry
2% of extranodal non-Hodgkin's lymphomas, though or immunohistochemistry, and gene rearrangement
it is more common in HIV-positive patients (RADIN et studies, particularly in certain rare cases of T-cell
al. 1993; JONES et al. 1997). The diagnostic criteria for lymphoma (KATZ et al. 1991; ROBBINS et al. 1995).
primary pancreatic lymphoma include a predominant The majority of cases are of diffuse large-celllym-
pancreatic mass with gross involvement of only the phoma, predominantly of B-cell immunophenotype
peripancreatic lymph nodes, in the absence of hepatic, (EZZAT et al. 1996;, BOUVET et al. 1998), although low
splenic, mediastinal, or superficial lymphadenopathy, or intermediate grade tumors (small or mixed small
and along with a normal leukocyte count (BEHRNS et and large cells) have been reported in about 20% of
al. 1994). Primary pancreatic lymphoma represents cases (PRAYER et al. 1992; KONIARIS et al. 2000), in
1%-3% of pancreatic malignancies (REED et al. 1979; addition to occasional T-cell lymphomas (NISHIMURA
MANSOUR et al. 1989; TUCHECK et al. 1993). et al. 2002). The aspirate smears commonly show large,
atypical lymphocytes, distributed singly, in a back-
ground of lymphoglandular bodies, which represent
13.4.2 small fragments of cytoplasm that have detached from
Clinical Manifestations lymphoid cells; these are specific for lymphoma and
cannot be seen on histology sections (BOUVET et al.
The patients' age range is 23-89 years (mean 56 years), 1998). Additional findings that support the diagnosis
and this tumor is slightly more common in men than of lymphoma include a monomorphic appearance of
women (MERKLE et al. 2000). Primary pancreatic lym- the lymphoid cells (DEMAY 1996). The chromatin can
phoma presents with nonspecific symptoms similar to range from fine to coarse, with variable numbers and
those of adenocarcinoma. The duration of symptoms prominence of nucleoli (Fig. 13.6b).
ranges from 2 days to 32 years (average 19 weeks). The Cytologically, the differential diagnosis includes
most common symptoms reported are: abdominal undifferentiated carcinoma, particularly small-cell
pain (73%), weight loss (51 %),jaundice (42%), nausea carcinoma. The latter is more likely to show nuclear
(34%), vomiting (18%), and fatigue (9%). Fever, chills, molding, with a lack of lymphoglandular bodies. In
and night sweats, symptoms classic of non-Hodgkin's contrast to lymphomas, small-cell carcinomas are usu-
lymphoma elsewhere, are rarely reported (9%). The ally positive for keratin and some endocrine markers,
only clinical presentation that suggests the possibility and negative for leukocyte common antigen (ACKER-
of pancreatic lymphoma is a large, palpable pancre- MAN et al.1976). Other rare tumors that may enter the
atic mass, homogenous on imaging, presenting with differential diagnosis include peripheral neuroecto-
abdominal pain and without jaundice, particularly dermal tumor (PNET), which should be considered
when lactate dehydrogenase (LDH) levels are elevated especially in children and can be distinguished by posi-
(BOUVET et al. 1998; MERKEL et al. 2000). tivity for CD99 and negativity for leukocyte common
antigen (LUTTGES et al. 1997; MOVAHEDI-LANKARANI
et al. 2000), and desmoplastic small-cell tumor of the
13.4.3 abdomen.
Pathology

Resected primary pancreatic lymphomas appear 13.4.4


grossly as large, soft, nodular masses, ranging in size Treatment and Prognosis
from 4 to 14 cm, most commonly occurring in the
head of the organ (SATAKE et al.1991). The pancreatic The primary treatment for pancreatic lymphoma is
ductal system may be displaced but is not usually combined chemotherapy with involved-field radio-
destroyed (SOLCIA et al. 1997) (Fig. 13.6a). therapy, achieving cure rates of about 46% (KONIARIS
Rare Solid Tumors: Clinical Manifestations and Pathology 233

et al. 2000). The role of surgery is more controversial.


While some believe that the only role for surgery is in
establishing the diagnosis when percutaneous or endo-
scopic biopsies fail to do so (WEBB et al. 1989), others
recommend a more aggressive approach in resectable
cases (BEHRNS et al. 1994), suggesting a higher cure rate
for surgically treated patients (KONIARIS et al. 2000).

13.5
Germ-Cell Tumors

13.5.1 a
Teratoma

Intrapancreatic teratomas usually occur in patients


less than 25 years old (mean 20 years, median 11
years) (MESTER et al. 1990). Symptomatic patients
present generally with abdominal-lumbar pain and
have a palpable left upper quadrant mass. Radiologi-
cally and macroscopically, these tumors are charac-
terized by a solid-cystic appearance with a distinct
capsule (BOWEN et al. 1987). Hair, mucoid and fatty
areas, bone, or teeth are usually seen. Histologically,
teratomas consist of variably differentiated cells (usu-
ally well differentiated) from the three germ layers
including cartilage, bone, fat, teeth, and columnar or b
squamous epithelium, similar to mature teratomas
elsewhere. They likewise probably originate from Fig. 13.6a,b. Lymphoma. a Neoplastic lymphocytes diffusely
germ cells arrested in migration to the gonads early infiltrate and replace part of the pancreas (H&E, low power). b
The tumor is composed of a monomorphic population of dys-
in embryonic life (GONZALEZ-CRUSSI 1982).
cohesive lymphoid cells with prominent nucleoli (fine-needle
aspirate, ultrafast Papanicolaou stain)

13.5.2
Choriocarcinoma

Two cases of primary pancreatic choriocarcinoma (KLOPPEL and MAILLET 1989; FER ROZZI et al. 2000).
have been reported. Both tumors were cystic due to They derive from various connective tissue compo-
necrosis and hemorrhage. They consisted microscop- nents and can be classified according to their histo-
ically of typical choriocarcinoma, with undifferenti- logic origin into lymphovascular, myogenic, fatty, or
ated cytotrophoblastic cells and syncytiotrophoblast neurogenic categories.
which stained for HCG. They lacked gland forma-
tion and keratinization. Serum beta-HCG levels were
elevated, and dropped following the initiation of che- 13.6.1
motherapy (CHILDS et al. 1985; COSKUN et al. 1998). Lymphovascular Tumors

13.6.1.1
Lymphangioma
13.6
Mesenchymal Tumors Lymphangiomas are rare benign tumors, of which
only a few cases have been reported in the literature.
Mesenchymal tumors of the pancreas are rare, In a review of 10 cases from the Endocrine Pathology
accounting for 1%-2% of all pancreatic tumors Registry of the Armed Forces Institute of Pathology,
234 T. Oweity and A. B. West

8 cases occurred in female patients, two in males, growth pattern may raise a suspicion of malignancy,
age range 2-61 years (mean 28.9 years) at initial hemangioendotheliomas have a favorable prognosis
presentation (PAAL et al. 1998). Macroscopically, the and spontaneous regression can be expected, even
tumors were well circumscribed, and 6 occurred in without resection or treatment. Thus, a conservative
the tail of the pancreas. They can grow within the approach to treatment is generally recommended
parenchyma or can be connected to the organ by a (TUNELL 1976).
pedicle (FERROZZI et aI. 2000). They are multicystic,
containing serous or chylous fluid, and range in size 13.6.1.4
between 3 and 20 cm in greatest dimension (PAAL Other Lymphovascular Tumors
et al. 1998). Histologically, lymphangiomas consist
of multilocular cystic spaces of various sizes rang- Other vascular tumors reported include hemangio-
ing from microscopic to as large as 10 cm, lined by pericytoma (BARDAXOGOU et al. 1995), which may
endothelial cells. The stroma contains smooth muscle behave aggressively and metastasize, and angiosar-
and mature lymphocytes. A thin capsule of fibrous coma (BANCU et al. 1971).
tissue is present (ABE et al. 1997). Calcifications are
rare (HANELIN and SCHIMMEL 1977). Immunohis-
tochemically, the lining endothelial cells stain posi- 13.6.2
tively for factor VIII-R antigen and CD-31, but are Adipose Tissue Tumors
CD-34 negative in all cases.
Lymphangiomas have an excellent prognosis. In In the pancreas, tumors of adipose tissue are
the series of PAAL et al. (1998), following surgery, all extremely rare (BIGARD et al. 1989; DI MAGGIO et al.
patients were alive without disease with a median 1996; BARUTCU et al. 2002). Lipomas consist of lob-
follow-up of 7.2 years from diagnosis. ules of mature adipose tissue surrounded by a thin
capsule presenting as a sharply defined mass. They
13.6.1.2 are usually recognized radiologically and should be
Hemangioma differentiated from localized lipomatosis which is
connected to the peripancreatic fat and does not have
Rare cases of hemangioma have been reported a capsule (FERROZZI et al. 2000; BARUTCU et al. 2002),
(MANGIN et al. 1985). Most have a characteristic and from the rare liposarcomas (ELLIOTT et al. 1980;
radiological appearance and are easily diagnosed MILANO et al. 1988), fibrolipoma, and lipoblastoma.
(KOBAYASHI et al. 1991). Macroscopically, hemangio-
mas appear red, resembling a hematoma. Histologi-
cally, they consist of cavernous vascular spaces lined 13.6.3
by endothelial cells and filled with blood (KOBAYASHI Myogenic Tumors
et al. 1991) and may be a cause of retroperitoneal or
intraabdominal hemorrhage. 13.6.3.1
Leiomyoma
13.6.1.3
Hemangioendothelioma Primary leiomyoma of the pancreas has occasionally
been described (NAKAMURA et al. 2000; BAKOLAS et
Few cases of hemangioendothelioma have been al. 2001). Radiologically and macroscopically, the
reported, mainly in infants and children (HORIE et tumor has a sharp, well defined/encapsulated margin
al. 1985). Jaundice or pain was the presenting symp- without invasion of surrounding structures. It is com-
tom. Macroscopically, hemangioendotheliomas may posed of bland, spindle-shaped, smooth muscle cells
be poorly demarcated, white, and hard. Histologically, in interweaving fascicles. However, the differential
the tumor cells infiltrate the pancreas, compressing diagnosis from leiomyosarcoma can be exceedingly
adjacent tissue. They are spindled or round, arranged difficult (see below).
in rows and short strands in a fibrous stroma. Some
of the tumor cells line vascular channels and blood- 13.6.3.2
filled spaces, with moderate atypia and rare mitoses. Leiomyosarcoma
Focal fibrin thrombi are seen in the vascular spaces.
The cells stain positively for factor VIII (HoRIE et al. Leiomyosarcomas of the pancreas primarily affect
1985). Although high cellularity and an infiltrative adults in the 5th decade of life or older (mean age
Rare Solid Tumors: Clinical Manifestations and Pathology 235

53.6 years, range 14-80 years), with a nearly equal


male to female ratio (NESI et al. 2001). The present-
ing symptoms are nonspecific including abdominal
mass, pain, and weight loss. The tumors range in
size from 3 to 25 cm (median 11 cm), larger ones
frequently undergoing cystic degeneration and mim-
icking pancreatic pseudo cysts radiologically. The cut
surface is fleshy with variable areas of necrosis and
degeneration. Microscopically, the tumors have an
expansile growth pattern and may be encapsulated
and entrap residual normal pancreatic elements
(DE ALAVA et al. 1993; FERLAN-MARLoT et al. 2000;
MACHADO et al. 2000; NESI et al. 2001) (Fig. 13.7a).
The tumor cells are spindle-shaped, arranged in com- a
pact bundles and fascicles which may display herring
bone or storiform patterns in areas. The nuclei are
generally elongated, blunt ended, and cigar shaped,
surrounded by ill-defined eosinophilic cytoplasm
(Fig. 13.7b). Foci of round cells with an epithelioid
appearance may be present (FERLAN-MARLoT et al.
2000; NESI et al. 2001). Nuclear atypia and pleomor-
phism are variable depending on the grade of the
tumor: While these can be minimal in better differen-
tiated tumors (ISHIKAWA et al. 1981), they tend to be
prominent features in higher grade tumors with giant
and bizarre cells occasionally present (ZALATNAI et
al. 1998). Mitotic counts are variable also: Cases with
rare mitoses (0 to 1 per 10 high power fields) have b
been reported to metastasize, while mitotic counts of
more than 10 per 10 high power fields are associated
with an adverse outcome (ISHIKAWA et al. 1981; NESI
et al. 2001). Tumor necrosis is common.
Leiomyosarcomas are thought to originate from
smooth muscle cells of either the pancreatic ducts
or the intrapancreatic small vessels (ZALATNAI et al.
1998). The tumor cells are usually positive for actin
(muscle-specific actin and smooth muscle actin)
(Fig. 13.7c) with more variable staining for desmin,
consistent with a myogenic phenotype. This along
with positive vimentin staining and negative stain-
ing for epithelial markers (cytokeratins, EMA, CEA),
helps in excluding the more commonly encountered
sarcomatoid carcinoma, although focal keratin posi-
tivity in an otherwise typical leiomyosarcoma has
Fig. 13.7a-c. Leiomyosarcoma. a The tumor is cellular with an
been reported (SOLCIA et al. 1997). Ultrastructural expansile growth pattern (H&E, low power). b Spindle-shaped
studies are supportive of smooth muscle differentia- neoplastic cells exhibit pleomorphism and abundant mitoses
tion (ISHIKAWA et al. 1981; DE ALAVA et al. 1993). (H&E, high power). c The tumor cells express smooth muscle
The differential diagnosis of primary pancreatic actin (brown), supporting a myogenic phenotype (actin
immunostain). Case courtesy of Dr DIVA SALOMAO
leiomyosarcoma includes sarcomatoid carcinoma,
other poorly differentiated sarcomas, and leiomyo-
sarcoma metastatic from other organs. The clinical
and immunohistochemical features generally help
in making a definitive diagnosis. Distinguishing
236 T. Oweity and A. B. West

leiomyoma from leiomyosarcoma is a more difficult and negative for epithelial and myogenic markers.
problem: However, since leiomyomas have been Electron microscopy shows long spaced collagen
very rarely reported in the pancreas, and as smooth (Luse bodies), numerous long, interdigitating cel-
muscle tumors with as few as one mitosis per 10 high lular processes, and abundant intercellular basement
power fields in a moderately cellular background membrane material (BROWN et al. 1998).
with minimal pleomorphism have been reported to Surgical resection is the treatment of choice and
metastasize (ISHIKAwA et al. 1981), a diagnosis of may involve enucleation for the benign tumors or
primary pancreatic leiomyoma should be made with pancreatic resection. Preoperative or intraoperative
caution. diagnosis aids in choosing the appropriate surgery
Although the number of reported cases is small, (MORITA et al. 1999). Following surgery, the progno-
leiomyosarcomas appear to be highly malignant, sis is good, with no postoperative death reported.
usually pursuing an aggressive course. They are often
widely metastatic at the time of the presentation, par- 13.6.4.2
ticularly affecting the liver and sparing lymph nodes. Primitive Neuroectodermal Tumor
Surgical resection offers the best hope for prolonged
survival in localized cases (NESI et al. 2001). Primitive neuroectodermal tumors (PNETs, neu-
roepitheliomas) are very rare, small, round-cell
tumors that are part of the Ewing sarcoma family
13.6.4 of lesions. Patients range in age from 6 to 25 years
Neurogenic Tumors (mean 18 years) with an almost equal sex incidence
(BULCHMANN et al. 2000; MOVAHEDI-LANKARANI
13.6.4.1 et al. 2002). They usually present with jaundice or
Schwannoma abdominal pain.
These tumors are typically located in the head of
Schwannomas (neurinomas, neurilemmomas) are the pancreas and range in diameter from 3.5 to 9 cm.
thought to derive from Schwann cells enveloping Microscopically, they have the typical histology of
peripheral nerves. They generally affect older adults PNETs elsewhere and are composed of sheets and lob-
(average 60 years), with equal sex incidence, and are ules of small cells with round to oval nuclei and scant
often incidental findings (BROWN et al. 1998). The cytoplasm. Nuclear molding may be prominent with a
most common presenting symptom is pain (ALMO mitotic rate of 3-5 per 10 high power fields. Infiltration
and TRAVERSO 2001). Some arise in a setting of Von of peripancreatic soft tissue is a consistent finding. No
Recklinghausen's disease (WALSH and BRANDSPIGEL Homer-Wright rosettes are identified. Necrosis may be
1989; ALAMO and TRAVERSO 2001). Although the present focally (MOVAHEDI-LANKARANI et al. 2002).
majority of the reported cases have been benign, a Immunohistochemically, there is strong cell mem-
few malignant schwannomas (malignant peripheral
nerve sheaths tumors) have been reported (LUTT-
GES et al. 1997; MORITA et al. 1999), occasionally in
patients with Von Recklinghausen's disease. These
also tend to be cystic. Microscopically, they are often
poorly differentiated sarcomas in which the neural
phenotype may be difficult to demonstrate (BROWN
et al. 1998).
Schwannomas can be located in any part of
the pancreas (MORITA et al. 1999). They tend to
be encapsulated with a cystic cut surface, varying
from tan firm to yellow gelatinous depending on the
proportions of their cellular elements. They range
in diameter from 2 to 26 cm, average 7.4 cm. Histo-
logically, they are typical schwannomas, composed of
spindle cells with areas of cellular palisading (Antoni
Fig. 13.S. Schwannoma. The neoplastic spindle cells of this
A) and ofloose myxoid appearance (Antoni B). Peri- tumor show prominent palisading. A hyalinized large vessel is
vascular hyalinization may be prominent (Fig. 13.8). present on the left of the field (H&E,low power). Case courtesy
The cells are immunoreactive for S-l 00 and vimentin of Dr DIVA SALOMAO
Rare Solid Tumors: Clinical Manifestations and Pathology 237

brane positivity for CD-99, and most tumors are dif- 13.7
fusely positive for keratin AE-l/AE-3. NSE is positive Solid Tumor-like Lesions of the Pancreas
in the majority of patients, and focal chromogranin or
synaptophysin positivity may be seen. The tumor cells A variety of nonneoplastic conditions may present
are negative for desmin, actin, S-100 protein, insulin, with clinical and radiological features mimicking
glucagon, and somatostatin. Ultrastructurally, they pancreatic cancer.
display epithelial features including numerous desmo-
somes, and both neurosecretory granules and tonofila-
ments have been reported. Some 85% of patients show 13.7.1
the classic translocation t(11:22) (q24;q12), resulting Heterotopic (Ectopic) Spleen
in the EWS-FLIl fusion gene.
The histologic differential diagnosis of PNET Accessory spleens occur in about 10% of the popula-
includes all small, round-cell tumors such as lym- tion. Most are located around the splenic hilum, but
phoblastic lymphoma, neuroblastoma, rhabdo- in 16% of patients, they are located in the tail of the
myosarcoma, desmoplastic small round-cell tumor, pancreas (HAYWARD et al. 1992), and they rarely
pancreatoblastoma, small-cell undifferentiated carci- occur in the head of the pancreas (LANDRY and
noma, and endocrine tumors. The pattern of positive SARMA 1989). Although most of them are asymp-
immunostaining for CD-99 (the MIC-2 gene product), tomatic, they may come to medical attention as a
NSE, and keratin is characteristic and should confirm hypervascular pancreatic mass which can mimic an
the diagnosis. Although this profile may be seen in endocrine (islet-cell) tumor, metastasis, or pancreatic
desmoplastic small round-cell tumor, the latter is less carcinoma (SUZUKI et al. 1994; HARRIS et al. 1994;
frequently positive for CD-99 and always positive for CHUREI et al. 1998; BARAWI et al. 2000). Elevation
desmin, in contrast to PNETs. Additionally, desmoplas- of CA19.9 and CEA serum levels has been reported
tic small round-cell tumor displays the characteristic (CHUREI et al. 1998; LAUFFER et al. 1999).
t(11;22) translocation involving fusion of EWS gene Macroscopically, accessory spleens appear as well-
and WTl gene, different from the EWS-FLI 1 fusion demarcated red masses, 0.5-4 em in size, usually in
gene of PNETs. Small-cell undifferentiated carcinomas the tail of the pancreas (SOLCIA et al. 1997). Micro-
appear in the adult population with a high mitotic rate scopically, they consist of lymphoid follicles and
of more than 10 per high power field and karyorrhexis splenic pulp. They may give rise to one or multiple
similar to small-cell carcinomas of the lung. Endocrine epidermal inclusion cysts which can mimic other
neoplasms of the pancreas usually display more diffuse pancreatic neoplastic or nonneoplastic cysts (DAVID-
strong staining with synaptophysin or chromogranin SON et al. 1980; TANG et al.1994; TSUTSUMI et al. 2000;
and may stain for islet-cell hormones and, unlike PNETs, HORIBE et al. 2001; SONOMURA et al. 2002; YOKOMIZO
are predominantly tumors of adults. They display fine, et al. 2002; FINK et al. 2002). These are usually lined
evenly distributed chromatin patterns and lack nuclear by keratinizing or nonkeratinizing squamous epi-
molding. They also lack the EWS-FLI 1 fusion gene. thelium, surrounded by splenic pulp tissue with a
Neuroblastomas occur in young adults but lack the fibrous capsule within the pancreatic parenchyma
characteristic translocation of PNETs. PNETs can be (MOROHOSHI et al. 1991). They may be up to 15 em
distinguished from lymphoblastic lymphomas and in diameter (CHO! et al. 2000). The cystic part con-
rhabdomyosarcomas by the lack of leukocyte common tains yellow-white caseous material, and the solid
antigen and muscle differentiation, respectively. part appears red-brown granular, similar to splenic
Although these tumors appear to be highly aggres- parenchyma. No hair or skin appendages are seen.
sive, some patients treated surgically or with chemo-
therapy have a reported survival of up to 4 years.
13.7.2
Hamartoma
13.6.5
Other Mesenchymal Tumors Pancreatic hamartomas are extremely rare lesions
consisting of normal pancreatic tissues in abnormal
Other miscellaneous mesenchymal tumors have been relationship to one another. They may be solid or
reported such as solitary fibrous tumor (LUTTGES et cystic. In one case, in a premature infant with refrac-
al. 1999) and the so-called malignant fibrous histio- tory hypoglycemia and hypocalcemia, the entire
cytoma (PASCAL et al. 1989; BALEN et al. 1993). pancreas consisted of non cystic ductal elements in
238 T. Oweity and A. B. West

groups of varying sizes within connective tissue, with Several cases isolated to the pancreas have been
a minority of well-organized islets and acinar tissue described (JOHNSON et al. 1983; ABREBANEL et al.
(BURT et al. 1983). ANTHONY et al. (1977) described 1984; SCOTT et al. 1988; PALAZZO and CHANG 1993;
pancreatic 'pseudotumors' in three patients com- MORRIS-STIFF et al. 1998; SHANKAR et al. 1998;
posed oflobulated connective tissue enclosing acinar PETTER et al. 1998), and a few have been described
cells, endocrine cells, and dilated pancreatic ducts in in association with retroperitoneal fibrosis (CLARK
complete disarray, without malignant features. Large et al. 1988; CHUTAPUTTI et al. 1995). The most
multicystic pancreatic hamartomas, composed of common presentations are abdominal pain and bili-
disorganized lobules of exocrine pancreatic acinar ary obstruction secondary to local extension of the
tissue surrounding pancreatic ducts and cyst-like lesion. Constitutional symptoms such as fever, weight
spaces of variable diameter, lined by cuboidal to loss, and malaise have also been reported (PALLAZZO
columnar epithelial cells, admixed with clusters of and CHANG 1993),in addition to asymptomatic cases.
adipocytes and with diffusely scattered or clustered Hypergammaglobulinemia, anemia, and an elevated
islet cells, have been reported in an infant (FLAHERTY sedimentation rate have also been reported in a child
and BENJAMIN 1992) and an adult (IZBICKI et al. (SCOTT et al. 1988). The age at presentation ranges
1994). Patients with Brunner's gland hamartoma from 2.5 to 65 years (Lm and CONSORTI 2000), but
mimicking carcinoma of the head of the pancreas the majority present in childhood or early adulthood
have also been described (SKELLENGER et al. 1983). (MCCLAIN et al. 2000). Given the radiologic similari-
ties to a malignant neoplasm, the low index of clinical
suspicion, and the variability of histologic appear-
13.7.3 ance, a misdiagnosis of malignancy may be readily
Pseudolipomatous Hypertrophy made (WALSH et al. 1998).
Macroscopically, the tumors are firm, poorly
Pancreatic lipomatosis is uncommon as an isolated circumscribed, yellow-white with indurated areas
abnormality, but it can be seen in a variety of con- (WREESMANN et al. 2001), or well-circumscribed,
ditions such as chronic pancreatitis, obesity, aging, nodular, gelatinous to firm, and mostly solid (WALSH
cystic fibrosis, alcoholism, and Cushing's syndrome et al. 1998) but occasionally cystic (Lm and CON-
(NAKAMURA et al.1979; PATEL et al.1980; THAM et al. SORTI 2000), up to 12 cm in size.
1991; ITAI et al. 1995; MATSUMOTO et al. 1995; KATZ Microscopically, these tumors are composed
et al. 1999). It may be associated with pancreatic exo- of variable amounts of proliferating spindle cells,
crine insufficiency (LOZANO et al.1988) and, particu- admixed with a variety of inflammatory cells, pro-
larly when localized, may mimic pancreatic tumors, ducing a wide spectrum of histopathologic appear-
especially lipoma (BARUTCU et al. 2002). However, as ances. This has led to the diverse confusing nomen-
opposed to lipoma, it is usually unencapsulated and clature of this lesion.
shows direct contact with the peripancreatic fat (ITAI Three basic histologic patterns have been recog-
et al. 1995; KATZ et al. 1998; FERROZZI et al. 2000). nized in extrapulmonary inflammatory pseudotumor,
Grossly, the pancreas is moderately to markedly of which myofibroblasts are the main proliferating
enlarged, maintaining its usual shape. Histologi- cell (COFFIN et al. 1995). These include a myxoid
cally, the parenchyma consists of mature fatty tissue pattern with a loose arrangement of plump spindle
separated by thin fibrous septa, containing isolated to stellate cells in a vascular, edematous background
clusters of normal islets (SOLCIA et al. 1997). resembling granulation tissue, containing a polymor-
phous inflammatory infiltrate. This pattern resembles
nodular fasciitis, and the stromal cells have vesicular
13.7.4 nuclei and abundant eosinophilic cytoplasm, reminis-
Inflammatory Pseudotumors cent of rhabdomyoblasts but without cross-striations.
Numerous mitoses may be seen. The second pattern
Inflammatory pseudotumors are tumor-like lesions is characterized by compact spindle cell proliferation
of uncertain pathogenesis, also known as plasma- which may have a vascular arrangement or a storiform
cell granuloma, inflammatory myofibroblastic tumor, pattern with areas of variable cellularity and variable
and inflammatory myofibrohistocystic proliferation mitotic activity resembling a myogenic or fibrohistio-
(Lm and CONSORTI 2000). Although the lung is the cytic neoplasm. Plasma cells often predominate in the
most common site, these lesions occur in a variety background and may form aggregates. Lymphoid fol-
of extrapulmonary locations (COFFIN et al. 1995). licles may be present, and growth into or around ves-
Rare Solid Tumors: Clinical Manifestations and Pathology 239

sels may be seen. Obliterative phlebitis and perineural aged by surgical resection, and some associated
accentuation of inflammation have been described as with retroperitoneal fibrosis have regressed with
distinctive features of the entity (WREESMANN et al. the use of corticosteroids (CHUTAPUTTI et al. 1995).
2001). The third histologic pattern is the least prolif- Recurrence rates of up to 25% have been reported,
erative, with extensive collagenous, sparsely cellular however (COFFIN et al. 1998). Cases of synchronous
areas resembling fibromatosis or myofibromatosis, or metachronous distant tumors with a similar his-
with scattered lymphocytes and plasma cells. Calcifi- tology have been described, and it is uncertain if
cation and ossifications may be seen. These three his- these represent multifocal disease or distant metas-
tologic patterns may be equally represented in a single tases (Voss et al. 1999). Cases of malignant trans-
tumor, or one or two may predominate. formation have been described in recurrent tumors
The myofibroblastic nature of the proliferating (COFFIN et al. 1998). There are no distinguishing
spindle cells is confirmed by diffuse immunoreactiv- pathologic features predictive of aggressive behav-
ity for vimentin, muscle-specific actin, and smooth ior (COFFIN et al. 1998). Based on that, and as it is
muscle actin with more variable and focal des min difficult to distinguish this tumor from inflammatory
positivity. Focal cytokeratin positivity occurs in up fibrosarcoma, the prognosis of this tumor should be
to 35% of extrapulmonary myofibroblastic tumors guarded. Long-term follow-up is mandatory because
(COFFIN et al. 1995). The cells are negative for S- recurrences and possibly distant tumors may develop
100 protein and CI;! 35 (the follicular dendritic cell (WALSH et al. 1998).
marker), and the pl'asma cell infiltrate is polyclonal
(PALAZZO and CHANG 1993). Few B-Iymphocytes are
seen, but CD-4- and CD-8-positive T-Iymphocytes 13.7.5
are numerous (WALSH et al. 1998). Pseudolymphoma

Electron Microscopy. Ultrastructural examination Pseudolymphoma is characterized by a predomi-


shows spindle cells with abundant endoplasmic nant nonneoplastic proliferation of lymphoid tissue.
reticulum, pinocytotic vesicles and subplasmalem- Only rare cases have been reported in the pancreas
mal plaques, and extracellular collagen. The dif- (NAKASHIRO et al. 1991; HATZITHEOKLITOS et al.
ferential diagnosis includes a variety of spindle-cell 1994; PETTER et al. 1998), where it may be diffuse
tumors, benign and malignant, and the diagnosis is or localized. One localized lesion presented with
based on finding the characteristic myofibroblastic obstructive jaundice and appeared grossly as a soft,
proliferation with a mixed inflammatory back- yellow nodule measuring 2 cm in size (NAKASHIRO
ground. Differentiation between this tumor and et al. 1991). Microscopically, it showed massive pro-
inflammatory fibrosarcoma may be more difficult as liferation of variably sized lymphoid follicles, not
the two entities overlap, and they have been used syn- closely packed, with prominent, well polarized ger-
onymously (MEIS and ENZINGER 1991), although no minal centers, rimmed by a mantle of small round
cases of so-called inflammatory fibrosarcoma have lymphocytes, surrounded by variable amounts
been described in the pancreas. of fibrous connective tissue in the interfollicular
areas. Tingible body macrophages and mitoses were
Etiology. The etiology of inflammatory pseudotumor noted in the germinal centers, and plasma cells and
is unclear. Autoimmune mechanisms have been impli- eosinophils were scattered in the interfollicular area.
cated. This is supported by its association with reactive The adjacent pancreatic parenchyma was generally
hyperplastic lymphadenopathy (Voss et al. 1999), and intact. Immunostaining revealed a polyclonal het-
with other autoimmune diseases such as retroperito- erogeneous population of lymphocytes and plasma
neal fibrosis (CHUTAPUTTI et al. 1995) and Sjogren's cells. Another case mimicked lymphoplasmacytic
syndrome (ECKSTEIN et al. 1995). Infectious causes lymphoma (PETTER et al. 1998).
have also been proposed with emphasis on Epstein
Barr virus (EBV) in inflammatory pseudotumors of
other organs (ARBER et al. 1995), but this has been 13.7.6
found to be negative in the pancreatic cases tested Chronic Pancreatitis
(WALSH et al. 1998).
Chronic pancreatitis is the condition that most often
Treatment and Prognosis. Most extrapulmonary mimics pancreatic carcinoma clinically, radiologi-
inflammatory pseudotumors are successfully man- cally, and even morphologically (SOLCIA et al. 1997).
240 T. Oweity and A. B. West

There are two major forms: chronic calcifying pan- mental sclerosis and enlargement of the affected area.
creatitis, the main type, and obstructive pancreatitis The involved ducts may be distorted and occasion-
(BARTHET et al.1999). In Western countries, it is most ally contain calculi. Pseudocysts may be present. In
commonly due to chronic alcoholism with a relapsing the advanced stages, the entire pancreas is rock hard,
acute autodigestive pancreatitis. Hereditary forms irregular, and shrunken. The main duct shows irregular
and others related to malnutrition or autoimmune dilatation and distortion, usually impacted by calculi.
origin have also been described (KLOPPEL 1999), the In chronic obstructive pancreatitis, there is marked
last in association with other autoimmune diseases duct dilatation upstream of the stenotic or occluded
such as primary sclerosing cholangitis, Sjogren's dis- duct in the head of the pancreas. There is fibrotic
ease (MONTEFUSCO et al. 1984), multifocal idiopathic atrophy of the parenchyma, and no calculi are usually
fibrosclerosis (LEVEY and MATHAI 1988; CLARK et seen. Microscopically, fibrosis progresses from inter-
al. 1988), and inflammatory bowel disease (BARTHET lobular to intralobular distribution in the later stages,
et al. 1999). The diffuse atrophy and fibrosis of the against a background of lymphocytes, plasma cells,
pancreas following obstruction of the main duct in and macrophages (Fig. 13.9a, b). The intralobular
the head of pancreas by tumor, scar, or stone is called ducts are distorted and focally ectatic, embedded in
chronic obstructive pancreatitis. fibrous tissue. The epithelium may show hyperplastic
Macroscopically, in early chronic pancreatitis, the changes initially and become atrophic later on, while
gland is unevenly affected, with focal nodular or seg- lacking significant cellular atypia. Eosinophilic pro-

Fig.13.9a-d. Chronic pancreatitis. a There is extensive destruction of pancreatic acini by chronic inflammation and replacement
by fibrosis. Surviving ducts are present on the right and left of the field (H&E, low power). b The inflammatory infiltrate consists
of plasma cells and lymphocytes, admixed with collagen bundles (H&E, high power). c Large blood vessels and nerve bundles are
usually seen, surrounded by marked inflammation (H&E, medium power). d In the late stage, only islets survive in the hyalinized
fibrous tissue. The lobular arrangement of the islets distinguishes this from an islet-cell tumor (H&E, medium power)
Rare Solid Tumors: Clinical Manifestations and Pathology 241

teinaceous material develops into plugs and calculi. Abraham SC, Wu T-T, Hruban RH, et al (2002) Genetic and
The fibrous tissue contains thick-walled arteries and immunohistochemical analysis of pancreatic acinar cell
nerve trunks (Fig. 13.9c), and over time the inflam- carcinoma: frequent allelic loss on chromosome 11 p and
alterations in the APC/beta-catenin pathway. Am J Pathol
matory infiltrate becomes scantier, predominantly 160:953-962
lymphocytic, and periductal. In the late stages, the Abrebanel P, Sarfaty S, Gal R, et al (1984) Plasma cell granu-
pancreatic acini are replaced by fibrous tissue, and loma of the pancreas. Arch Pathol Lab Med 108:531-532
only irregular, variably sized clusters of islet cells Ackerman NB, Aust JC, Bredenberg CE, et al (1976) Problems
remain (Fig. 13.9d), along with dilated interlobular in differentiating between pancreatic lymphoma and ana-
plastic carcinoma and their management. Ann Surg 184:
ducts. In obstructive chronic pancreatitis, the fibrotic 705-708
process is usually more evenly distributed and more Adsay NV, Adair CF, Heffess CS, Klimstra DS (1996) Intraductal
pronounced centrally around the pancreatic duct oncocytic papillary neoplasms of the pancreas. Am J Surg
rather than peripherally. Pathol 20:980-994
Almo KM, Traverso LW (2001) Pancreatic schwannoma: an
uncommon but important entity. J Gastrointest Surg 5:
Differential Diagnosis. Chronic pancreatitis can be 359-363
mistaken on frozen sections and in small biopsies Alvarez M, Rodriguez A, Zabaleta S, et al (1994) [Pseudotumor
for ductal adenocarcinoma. However, the lobular pancreatic tuberculosisJ. Rev Esp Enferm Dig 86:625-626
arrangement of the remaining ducts, even when dis- Anthony PP, Faber RG, Russel RC (1977) Pseudotumors of the
torted, is usually maintained, and significant atypia Pancreas. Br Med J 26:814
Aoki Y, Tanimura H, Mori K, et al (1989) Osteoclast-like giant
is not seen. No perineural ductal invasion is present. cell tumor of the pancreas associated with cystadenocar-
The remaining disorganized islets may be mistaken cinoma. Nippon Geka Hokan 58:452-460
for endocrine tumor, but they are usually arranged in Arber DA, Kamel OW, Van de Rijn M, et al (1995) Frequent
clusters and rarely show a diffuse irregular infiltration presence of Epstein-Barr virus in inflammatory pseudotu-
mor. Hum PathoI26:1093-1098
of the fibrous tissue. The presence of calculi helps to
Bakolas I, Sofos AG, Athanassoulias V, et al (2001) [A volumi-
establish the diagnosis of chronic pancreatitis (SOLCIA nous vascular leiomyoma of the pancreas treated by per-
et al. 1997). It is important to remember that chronic sonal technique of resection of the pancreatic head without
pancreatitis may coincide with ductal carcinoma. duodenectomyJ. Minerva Chir 56:175-181
Balen EM, De Villa VH, Cienfuegos JA, et al (1993) [Malignant
fibrous histiocytoma of the pancreas J. Rev Esp Enferm Dig
83:475-480
13.7.7 Bancu VE, Vincze L, Kesztenbaum E, et al (1971) [Angioplastic
Miscellaneous Inflammatory Tumor-like Masses sarcoma of the body of the pancreas, subtotal splenopan-
createctomyJ. Chirurgia (Burcur) 20:927-930
Tuberculosis (ALVAREZ et al. 1994), fungal infec- Barawi M, Bekal P, Gress F (2000) Accessory spleen: a poten-
tion (LUDWIG et al. 1999), and parasites (HUAROTO tial cause of misdiagnosis at EUS. Gastrointest Endosc 52:
769-772
SEDDA et al. 1989) have been reported to present Bardaxogou E, Manganas D, Landen S, et al (1995) Hemangio-
as pseudotumors. Sarcoidosis (MAHER et al. 1981) pericytoma of the pancreas: a report of a case and review of
and malakoplakia (ZUK et al. 1990) of the pancreas the literature. Hepatogastroenterology 42:172-174
have been described also. Noninflammatory lesions Barthet M, Hastier P, Bernard JP, et al (1999) Chronic pancre-
presenting as tumors include traumatic neuroma atitis and inflammatory bowel disease: true or coincidental
association? Am J Gastroenterol 94:2141-2148
(GEDDY and VENABLES 1991), peripancreatic fat Barutcu 0, Cihangiroglu M, Yildirim T, et al (2002) Fat contain-
necrosis (THURNHER et al. 2001), and pancreatic ing unusual tumor of the pancreas. Eur RadioI12:770-773
artery aneurysm (ITOH et al. 2002). Becker WF (1957) Pancreatoduodenectomy for carcinoma of
the pancreas in an infant: report of a case. Ann Surg 145:
864-872
Behrns KE, Sarr MG, Strickler JG (1994) Pancreatic lym-
phoma: is it a surgical disease? Pancreas 9:662-667
References Benoist S, Penna C, Julie C, et al (2001) Prolonged survival
after resection of pancreatoblastoma and synchronous
Abe H, Kubota K, Noie T, et al (1997) Cystic lymphangioma of liver metastases in an adult. Hepatogastroenterology 48:
the pancreas: a case report with special reference to embry- l340-l342
ological development. Am J Gastrenterol 92:1566-1567 Berendt RC, Shnitka TK, Wiens E, et al (1987) The osteoclast-
Abraham SC, Wu TT, Klimstra DS, et al (2001) Distinctive type giant cell tumor of the pancreas. Arch Pathol Lab Med
molecular genetic alterations in sporadic and familial 111:43-48
adenomatous polyposis-associated pancreatoblastomas: Bigard MA, Boissel P, Regent D, et al (1989) Intrapancreatic
frequent alterations in the APC/beta-catenin pathway and lipoma. First case in the literature. Gastroenterol Clin Bioi
chromosome IIp. Am J PathoI159:1619-1627 l3:505-507
242 T. Oweity and A. B. West

Bondeson L, Bondeson AG, Grimelius L, et al (1990) Oncocytic carcinoma of the stomach and pancreas: CT findings. Eur
tumor of the pancreas. Report of a case with aspiration RadioI8:1425-1428
cytology. Acta Cytol 34:425-428 Cubilla AL, Fitzgerald PJ (1984) Tumors of the exocrine pan-
Boor PI, Swanson MR (1979) Papillary-cystic neoplasm of the creas. Atlas of Tumor Pathology, 2nd series, fascicle 19.
pancreas. Am J Surg Pathol 3:69-75 Armed Forces Institute of Pathology, Washington DC
Bouvet M, Staerkel GA, Spitz FR, et al (1998) Primary pancre- Davidson ED, Campbell WG, Hersh T (1980) Epidermoid
atic lymphoma. Surgery 123:382-390 splenic cyst occurring in an intra pancreatic accessory
Bowen B, Ros PR, McCarthy MJ, et al (1987) Gastrointestinal spleen. Dig Dis Sci 25:964-967
teratomas: CT and US appearance with pathologic correla- De Alava E, Torramade J, Vazquez H (1993) Leiomyosarcoma of
tion. Radiology 162:431-433 the pancreas. Virchows Archiv [AJ 422:419-422
Brandt KR, Charboneau JW, Stephens DH, et al (1993) CT - and Deckard-Janatpour K, Kragel S, Teplitz RL, et al (1998) Tumors
US-guided pancreatic biopsy. Radiology 187:99-lO4 of the pancreas with osteoclast-like and pleomorphic giant
<refBrown SZ, Owen D, O'Connel JX, et al (1998) Schwannoma cells: an immunohistochemical and ploidy study. Arch
of the pancreas: a report of two cases and a review of the Pathol Lab Med 122:266-272
literature. Mod Pathol11:1178-1182 Defachelles AS, Martin De Lassalle E, Boutard P, et al (2001)
Buchino H, Castello FM, Nagaraj HS (1984) Pancreatoblas- Pancreatoblastoma in childhood: clinical course and thera-
toma. A histochemical and ultrastructural analysis. Cancer peutic management of seven patients. Med Pediatr Oncol
53:963-969 37:47-52
Bulchmann G, Schuster T, Haas RJ, et al (2000) Primitive DeMay RM (1996) The art and science of cytopathology. ASCP
neuroectodermal tumors of the pancreas. An extremely Press, Chicago
rare tumor. Case report and review of the literature. Klin Di Maggio EM, Solcia M, Dore R, et al (1996) Intrapancreatic
Pediatr 212:185-188 lipoma: first case diagnosed with CT. AJR Am J Roentgenol
Burt TB, Condon VR, Matlak ME (1983) Fetal pancreatic ham- 167:56-57
artoma. Pediatr Radiol13:287-289 Di Sant'Agnese PA (1991) Acinar cell carcinoma of the pan-
Carter TR, Feldman PS, Innes DJ, et al (1988) The role of fine creas. Ultrastruct PathoI15:573-577
needle aspiration cytology in the diagnosis of lymphoma. Drut R, Jones MC (1988) Congenital pancreatoblastoma in
Acta Cytol 32:848-853 Beckwith-Wiedemann syndrome: an emerging associa-
Chang KJ,Nguyen P, Erickson RA,et al (1997) The clinical util- tion. Pediatr PathoI8:331-339
ity of endoscopic ultrasound-guided fine-needle aspiration Dunn JL, Longnecker DS (1995) Pancreatoblastoma in an older
in the diagnosis and staging of pancreatic carcinoma. Gas- adult. Arch Pathol Lab Med 119:547-551
trointest Endoscop 45:387-393 Dworak 0, Wittekind C, Koerfgen HP, et al (1993) Osteoclastic
Chen J, Baithun SI (1985) Morphological study of 391 cases of giant cell tumor of the pancreas. An immunohistological
exocrine pancreatic tumors with special reference to the study and review of the literature. Pathol Res Pract 189:
classification of exocrine pancreatic carcinoma. J Pathol 228-231
146:17-29 Eckstein RP, Hollings RM, Martin PA, et al (1995) Pancreatic
Chen J, Baithun ST, Ramsay MA (1985) Histogenesis of pan- pseudotumor arising in association with Sjogren's syn-
creatic carcinomas: a study based on 248 cases. J Pathol drome. Pathology 27:284-248
146:65-76 Elliott TE, Albertazzi VJ, Danto LA (1980) Pancreatic liposar-
Childs CC, Korsten MA, Choi HS, et al (1985) Pancreatic coma: case report with review of retroperitonealliposarco-
choriocarcinoma presenting as inflammatory pseudocyst. mas. Cancer 45:1720-1723
Gastroenterology 89:426-431 Ezzat A, Jamshed A, Khafaga Y, et al (1996) Primary pancre-
Choi SK, Ahn SI, Hong KC, et al (2000) A case of epidermoid atic non-Hodgkin's lymphomas. J Clin Gastroenterol 23:
cyst of the intrapancreatic accessory spleen. J Korean Med 109-112
Sci 15:589-592 Fabre A, SauvanetA, Flejou JF, et al (2001) Intraductal acinar cell
Chun Y, Kim W, Park K, et al (1997) Pancreatoblastoma. J carcinoma of the pancreas. Virchows Arch 438:312-315
Pediatr Surg 32:1612-1615 Faulkner JE, Gaba CE, Powers JD, et al (1998) Diagnosis of
Churei H, Inoue H, Nakajo M (1998) Intrapancreatic accessory primary pancreatic lymphoma by fine needle aspiration.
spleen: case report. Abdom Imaging 23:191-193 Acta Cytol 42:834-836
Chutaputti A, Burrel MI, Boyer JL (1995) Pseudotumor of Ferlan-Marlot V, Vladislav P, Alojz P (2000) Pancreatic leio-
the pancreas associated with retroperitoneal fibrosis. A myosarcoma: clinicohistopathological presentation of a
dramatic response to corticosteroid therapy. Am J Gastro- rare tumor. Hepatogastroenterology 47:556-559
enteroI90:1155-1158 Ferrozzi F, Zuccoli G, Bova D, et al (2000) Mesenchymal tumors
ClarkA, Zeman RK, Choyke PL, et al (1988) Pancreatic pseudo- of the pancreas: CT findings. J Com put Assist Tomogr 24:
tumors associated with multifocal idiopathic fibrosclerosis. 622-627
Gastrointest Radiol 13:30-32 Fink AM, Kulkarni S, Crowley P, et al (2002) Epidermoid cyst
Coffin CM, Watterson J, Priest JR, et al (1995) Extrapulmonary in a pancreatic accessory spleen mimicking an infected
inflammatory myofibroblastic tumor (inflammatory pseu- abdominal cyst in a child. Am J RoentgenoI179:206-208
dotumor). A clinicopathologic and immunohistochemical Fischer HP, Altmannsberger M, Kracht J (1988) Osteoclast-
study of 84 cases. Am J Surg PathoI19:859-872 type giant cell tumor of the pancreas. Virchows Arch [AJ
Coffin CM, Humphrey PA, Dehner LP (1998) Extrapulmonary 412:247-253
inflammatory myofibroblastic tumor: a clinical and patho- Flaherty MJ, Benjamin DR (1992) Multicystic pancreatic ham-
logical survey. Semin Diagn Pathol 15:85-101 artoma: a distinctive lesion with immunohistochemical
Coskun M,Agildere AM, Boyvat F, et al (1988) Primary chorio- and ultrastructural study. Hum PathoI23:1309-1312
Rare Solid Tumors: Clinical Manifestations and Pathology 243

Frable WI, Still WJ, Kay S (1971) Carcinoma of the pancreas, Horie H, Iwasaki I, Iida H, et al (1985) Benign hemangioen-
infantile type. A light and electron microscopic study. dothelioma of the pancreas with obstructive jaundice. Acta
Cancer 27:667-673 Pathol Jpn 35:975-979
Frantz VK (1959) Tumors of the pancreas. Atlas of Tumor Pathol- Horie Y, Gomyoda M, Kishimoto Y, et al (1984) Plasma car-
ogy. Armed Forces Institute of Pathology, Washington DC cinoembryonic antigen and acinar cell carcinoma of the
Geddy PM, Venables CW (1991) Traumatic neuroma at the pancreas. Cancer 53:1137-1142
tail of the pancreas following splenectomy. Postgrad Med Hua C, Shu XK,Lei C (1996) Pancreatoblastoma: a histochemi-
J 67:90-91 cal and immunohistochemical analysis. J Clin Pathol 49:
Goldberg RD, Michelassi F, Montag AG (1991) Osteoclast-like 952-954
giant cell tumor of the pancreas: immunophenotypic simi- Huaroto Sedda M, Cisneros E, Huaman A (1989) [Severe stron-
larity to giant cell tumor of bone. Hum Pathol 22:618-622 gyloidiasis with pseudotumor pancreatic involvement.
Gonzalez-Crussi F (1982) Extragonadal teratomas. Atlas of Clinical casel. Rev Gastroenterol Peru 9:146-152
Tumor Pathology, 2nd series, fascicle 18. Armed Forces Huntrakoon M (1983) Oncocytic carcinoma of the pancreas.
Institute of Pathology, Washington DC Cancer 51:332-336
Good AE, Schnitzer B, Kawanishi H, et al (1976) Acinar pan- Imai Y, Morishita S, Ikeda Y, et al (1999) Immunohistochemi-
creatic tumor with metastatic fat necrosis: report of a case cal and molecular analysis of giant cell carcinoma of the
and review of rheumatic manifestations. Am J Dig Dis 21: pancreas: a report of three cases. Pancreas 18:308-315
978-987 Imamura A, Nakagawa A, Okuno M, et al (1998) Pancreato-
Gotchall J, Traweek T, Stenzel P (1987) Benign oncocytic endo- blastoma in an adolescent girl: case report and review of
crine tumor of the pancreas in a patient with polyarthritis 26 Japanese cases. Eur J Surg 164:309-312
nodosa. Hum PathoI18:967-969 Ishihara A, Sanda T, Takanari H, et al (1989) Elastase-I-secret-
Griffin BR, Wisbeck WM, Schaller RT, et al (1987) Radiother- ing acinar cell carcinoma of the pancreas. A cytologic,
apy for locally recurrent infantile pancreatic carcinoma electron microscopic and histochemical study. Acta Cytol
(pancreatoblastoma) Cancer 60: 1734-1736 33:157-163
Guarda LA, Silva EG, Ordonez NG, et al (1983) Clear cell islet Ishikawa 0, Matsui Y, Aoki Y, et al (1981) Leiomyosarcoma of
cell tumor. Am J Clin Pathol 79:512-517 the pancreas. Report of a case and review of the literature.
Hanelin LG, Schimmel DH (1977) Lymphangioma of the Am J Surg Pathol 5:597-602
pancreas exhibiting an unusual pattern of calcification. Itai Y, Saida Y, Kurosaki Y, et al (1995) Focal fatty masses of the
Radiology 122:636 pancreas. Acta Radiol 36:178-181
Harewood GC, Wiersema MJ (2002) Endosonography-guided Itoh K, Kamiya Y, Ohno N, et al (2002) A case of pancreatodu-
fine needle aspiration biopsy in the evaluation of pancre- odenal artery aneurysm causing pancreatic pseudotumor
atic masses. Am J Gastroenterol 97:1386-1391 and duodenal obstruction. Eur J Gastroenterol Hepatol14:
Harris GN, Kase DJ, Bradnock H, et al (1994) Accessory spleen 457-461
causing a mass in the tail of the pancreas: MR imaging Itoh T, Kishi K, Tojo M, et al (1992) Acinar cell carcinoma of
findings. Am J RoentgenoI163:1120-1121 the pancreas with elevated serum alpha-fetoprotein levels:
Hart MJ, White TT, Brown PC, et al (1987) Potentially curable a case report and review of 28 cases reported in Japan.
masses in the pancreas. Am J Surg 154: 134-136 Gastroenterol Jpn 27:785-791
Hassan MO, Gogate PA (1993) Malignant mixed exocrine- Izbicki JR, Knoefel WT, Muller-Hocker J, et al (1994) Pancreatic
endocrine tumor of the pancreas with unusual intracyto- harmartoma: a benign tumor of the pancreas. Am J Gas-
plasmic inclusions. Ultrastruct Pathol 17:483-493 troenteroI89:1261-1262
Hatzitheoklitos E, Buchler MW, Friess H, et al (1994) Pseudo- Jeffrey I, Crow J, Ellis BW (1983) Osteoclast-type giant cell
lymphoma of the pancreas mimicking cancer. Pancreas 9: tumour of the pancreas. J Clin PathoI36:1165-1170
668-670 Johnson RL, Page DL, Dean RH (1983) Pseudotumor of the
Hayward I, Mindelzun RE, Jeffrey RB (1992) Intrapancreatic pancreas. South Med J 76:647-649
accessory spleen mimicking pancreatic mass on CT. J Jones WF, Sheikh MY, McClave SA (1997) AIDS-related non-
Comput Assist Tomogr 16:984-985 Hodgkin's lymphoma of the pancreas. Am J Gastroenterol
Henke AC, Kelley CM, Jensen CS, et al (2001) Fine needle 92:335-338
aspiration cytology of pancreatoblastoma. Diagn Cytopa- Kanai N, Nagaki S, Tanaka T (1987) Clear cell carcinoma of the
thoI25:118-121 pancreas. Acta Pathol Jpn 37:1521-1526
Hoorens A, Lemoine NR, McLellan E, Lemoine NR, Kloppel G Katz DS, Nardi PM, Hines J, et al (1998) Lipomas of the pan-
(1993) Pancreatic acinar cell carcinoma: an analysis of cell creas. Am J Roentgenol 170: 1485-1487
lineage markers, p53 expression and Ki-ras mutation. Am Katz DS, Hines J, Math KR, et al (1999) Using CT to reveal fat-
J PathoI143:685-698 containing abnormalities of the pancreas. Am J Roentgenol
Hoorens A, Gebhard F, Kraft K, et al (1994) Pancreatoblastoma 172:393-396
in an adult: its separation from acinar cell carcinoma. Vir- Katz RL, Hirsch-Ginsberg C, Childs C, et al (1991) The role of
chows Arch 424: 485-490 the gene rearrangement for antigen receptors in the diag-
Horibe Y, Murakami M, Yamao K, et al (2001) Epithelial inclu- nosis of lymphoma obtained by fine-needle aspiration. A
sion cyst (epidermoid cyst) formation with epithelioid cell study of 63 cases with concomitant immunophenotyping.
granuloma in an intrapancreatic accessory spleen. Pathol Am J Clin Pathol 96:479-490
lnt 51 :50-54 Kawamoto S, Hiraoka T, Kanemitsu K, et al (1992) Alpha-feto-
Horie A, Yano Y, Kotoo Y, et al (1977) Morphogenesis of pan- protein-producing pancreatic cancer: a case report and
creatoblastoma, infantile carcinoma of the pancreas: report review of 28 cases. Hepatogastroenterology 39:282-286
of two cases. Cancer 39:247-254 Kay S, Harrison JM (1969) Unusual pleomorphic carcinoma
244 T. Oweity and A. B. West

of the pancreas featuring production of osteoid. Cancer the pancreas: case report and literature review. Am J Clin
23:1158-1162 OncoI24:77-80
Kerr NJ, Chun YH, Yun K, et al (2002) Pancreatoblastoma is Levey JM, Banner BF (1996) Adult pancreatoblastoma: a case
associated with chromosome 11 p loss of heterozygosity report and review of the literature. Am J Gastroenterol 91:
and IGF2 overexpression. Med Pediatr Oncol 39:52-54 1841-1844
Kissane JM (1994) Pancreatoblastoma and solid and cystic Levey JM, Mathai J (1988) Diffuse pancreatic fibrosis: an
papillary tumor: two tumors related to pancreatic ontog- uncommon feature of multifocal idiopathic fibrosclerosis.
eny. Semin Diagn Pathol 11:152-164 Am J Gastroenterol 93:640-642
Klimstra D, Heffess CS, Oertel JE, et al (1992) Acinar cell car- Lewandrowski KB, Weston L, Dickersin GR, et al (1990) Giant
cinoma of the pancreas. A clinicopathologic study of 28 cell tumor of the pancreas of mixed osteoclastic and
cases. Am J Surg Pathol 16:815-837 pleomorphic cell type: evidence for a histogenetic rela-
Klimstra DS, Rosai J, Heffess CS (1994) Mixed acinar-endo- tionship and mesenchymal differentiation. Hum Pathol
crine carcinomas of the pancreas. Am J Surg Pathol 18: 21:1184-1187
765-778 Liu TH, Consorti ET (2000) Inflammatory pseudotumor
Klimstra DS, Wenig BM, Adair CF, et al (1995) Pancreatoblas- presenting as a cystic tumor of the pancreas. Am Surg 66:
toma. A clinicopathologic study and review of the litera- 993-997
ture. Am J Surg Pathol 19: 13 71-1389 Lozano M, Navarro S, Perez- Ayuso R, et al (1988) Lipomatosis
Klimstra DS, Wenig BM, Heffess CS (2000) Solid-pseudopapil- of the pancreas: an unusual cause of massive steatorrhea.
lary tumor of the pancreas: a typically cystic carcinoma of Pancreas 3:580-582
the low malignant potential. Semin Diagn Pathol 17:66-80 Ludwig DJ, Morimoto K, Traverso LW (1999) Fungal pseudotu-
Kloppel G (1999) Progression from acute to chronic pancreati- mor masquerading as pancreatic cancer - a sequela of new
tis. A pathologist's view. Surg Clin North Am 79:801-814 technology? J Gastrointest Surg 3:123-126
Kloppel G (2000) Mixed exocrine-endocrine tumors of the Luttges J, Pierre E, Zamboni G, et al (1997) [Malignant non-
pancreas. Semin Diagn Pathol 17:104-108 epithelial tumors of the pancreas.] Pathologe 18:233-237
Kioppel G, Maillet B (1989) Classification and staging of Luttges J, Vogel I, Menke M, et al (1998) Clear cell carcinoma
pancreatic nonendocrine tumors. Radiol Clin North Am of the pancreas: an adenocarcinoma with ductal phenotype
27:105-119 . Histopathology 32:444-448
Kloppel G, Morohoshi T, John HD, et al (1981) Solid and cystic Luttges J, Mentzel T, Hubner G, et al (1999) Solitary fibrous
acinar cell carcinoma tumour of the pancreas. A tumour in tumor of the pancreas: a new member of the small group
young women with favourable prognosis. Virchows Arch of mesenchymal pancreatic tumors. Virchows Arch 435:
[A] 392:171-183 37-42
Kobayashi H, Itoh T, Murata R, et al (1991) Pancreatic cavern- Machado MA, Herman P, Montagnini AL, et al (2001) Benign
ous hemangioma: CT, MRI, US, and angiography charac- variant of osteoclast-type giant cell tumor of the pancreas:
teristics. Gastrointest Radiol 16:307-310 importance of the lack of epithelial differentiation. Pan-
Koh TH, Cooper JE, Newman CL, et al (1986) Pancreatoblas- creas 22:105-107
toma in a neonate with Wiedemann-Beckwith syndrome. Machado MC, Cunha JE. Penteado S, et al (2000) Preoperative
Eur J Pediatr 145:435-438 diagnosis of pancreatic leiomyosarcoma. Int J Pancreatol
Kohda E, Iseki M, Ikawa H, et al (2000) Pancreatoblastoma. 28:97-100
Three original cases and review of the literature. Acta Maher L, Choi H, Dodds WJ (1981) Noncaseating granulomas
RadioI41:334-337 of the pancreas. Probable sarcoidosis. Am J Gastroenterol
Koniaris LG, Lillemore KD, Yeo CJ, et al (2000) Is there a role 75:222-225
for surgical resection in the treatment of early-stage pan- Manci EA, Gardner LL, Pollock WJ, et al (1985) Osteoclastic
creatic lymphoma? J Am CoIl Surg 190:319-330 giant cell tumor of the pancreas. Aspiration cytology, light
Labate AM, Klimstra DL, Zakowski MF (1997) Comparative microscopy, and ultrastructure with review of the litera-
cytologic features of pancreatic acinar cell carcinoma and ture. Diagn Cytopathol 1:105-110
islet cell tumor. Diagn CytopathoI16:112-116 Mangin P, Perret M, Ronjon A (1985) Hemangioma du pan-
Lack EE, Cassady JR, Levey R, et al (1983) Tumors of the creas. J Radiol 66:381-384
exocrine pancreas in children and adolescents. A clinical Mansour GM, Gucchiaro G, Niotis MT, et al (1989) Surgical
and pathologic study of eight cases. Am J Surg Pathol 7: management of pancreatic lymphoma. Arch Surg 124:
319-327 1287-1289
Landry MM, Sarma DP (1989) Accessory spleen in the head of Martin RC, Klimstra DS, Brennan MF, et al (2002) Solid-pseu-
the pancreas. Hum Pathol 20:497 dopapillary tumor of the pancreas: a surgical enigma? Ann
Lauffer JM, Baer HU, Maurer CA, et al (1999) Intrapancreatic Surg Oncol 9:35-40
accessory spleen. A rare cause of a pancreatic mass. Int J Matsumoto S, Mori H, Miyake H, et al (1995) Uneven fatty
Pancreatol 25:65-68 replacement of the pancreas: evaluation with CT. Radiol-
Leader M. Patel J, Collins M, et al (1987) Anti-alphal-antichy- ogy 194:453-458
motrypsin staining of 194 sarcomas, 38 carcinomas, and McClain MB, Burton EM, Day DS (2000) Pancreatic pseudo-
17 malignant melanomas: its lack of specificity as a tumor tumor in an 11-year-old child: imaging findings. Pediatr
marker. Am J Surg Pathol 11:133-139 RadioI30:610-613
Lee WY, Tzeng CC, Jin YT et al (1993) Papillary cystic tumor Meis JM, Enzinger FM (1991) Inflammatory fibrosarcoma
of the pancreas: a case indistinguishable from oncocytic of the mesentery and retroperitoneum: a tumor closely
carcinoma. Pancreas 8:127-132 simulating inflammatory pseudotumor. Am J Surg Pathol
Leighton CC, Shun DT (2001) Osteoclastic giant cell tumor of 15:1146-1156
Rare Solid Tumors: Clinical Manifestations and Pathology 245

Mentes A, Yuce G (1993) Osteoclast-type giant cell tumor of Nakamura M, Katada N, Sakakibara A, et al (1979) Huge lipo-
the pancreas associated with mucinous cytoadenoma. Eur matous pseudohypertrophy of the pancreas. Am J Gastro-
J Surg OncoI19:84-86 enterol72:171-174
Merkle EM, Bender GN, Brambs HJ (2000) Imaging findings Nakamura Y, Egami K, Maeda S, et al (2000) Primary leio-
in pancreatic lymphoma: differential aspects. Am J Roent- myoma of the pancreas. Int J Pancreatol 28:235-238
genoI174:671-675 Nakashiro H, Tokunago 0, Watanabe T, et al (1991) Localized
Mester M, Trajber HJ, Compton CC, et al (1990) Cystic terato- lymphoid hyperplasia (pseudolymphoma) of the pancreas
mas of the pancreas. Arch Surg 125:1215-1218 presenting with obstructive jaundice. Hum Pathol 22:
Miettinen M, Partanen S, Fraki 0, et al (1987) Papillary cystic 724-726
tumor of the pancreas. An analysis of cellular differentia- Nesi G, Pantalone D, Ragionieri I, et al (2001) Primary leio-
tion by electron microscopy and immunohistochemistry. myosarcoma of the pancreas. A case report and review of
Am J Surg Pathol11:855-865 the literature. Arch Pathol Lab Med 125:152-155
Milano C, Columbato LA, Fliescher I, et al (1988) [Liposarcoma Newbould MJ, Benbow EW, Sene A, et al (1992) Adenocarci-
of the pancreas: report of a clinical case and review of the noma of the pancreas with osteoclast-like giant cells: a case
literature]. Acta Gastroenterol Latinoam 18:133-138 report with immunohistochemistry. Pancreas 7:611-615
Millis JM, Chang B, Zinner MJ, et al (1994) Malignant mixed Nishimura R, Takakuwa T, Hoshida Y, et al (2002) Primary
tumor (carcinosarcoma) of the pancreas: a case report pancreatic lymphoma: clinicopathological analysis of 19
supporting organ-induced differentiation of malignancy. cases from Japan and review of the literature. Oncology
Surgery 115:132-137 60:322-329
Mizuta Y, Isomoto H, Futuki Y, et al (1998) Acinar cell car- Nojima T, Kojima T, Kato H, et al (1992) Alpha-fetoprotein-
cinoma of the pancreas associated with hypoglycemia: producing acinar cell carcinoma of the pancreas. Hum
involvement of 'big' insulin -like growth factor II. J Gastro- Pathol 23:828-830
enteroI33:761-765 Nojima T, Nakamura F, Ishikura M, et al (1993) Pleomorphic
Montefusco PP, Geiss AC, Bronzo et al (1984) Sclerosing chol- carcinoma of the pancreas with osteoclast-like giant cells.
angitis, chronic pancreatitis, and Sjogren's syndrome: a Int J PancreatolI4:275-281
syndrome complex. Am J Surg 147:822-826 Nozawa Y, Abe M, Sakuma H, et al (1990) A case of pancreatic
Moore PS, Orlandini S, Zamboni G, et al (2001) Pancreatic oncocytic tumor. Acta Pathol Jpn 40:367-370
tumours: molecular pathways implicated in ductal cancer Ohaki Y, Misugi K, Fukuda J, et al (1987) Immunohisto-
are involved in ampullary but not exocrine nonductal or chemical study of pancreatoblastoma. Acta Pathol Jpn 37:
endocrine tumorigenesis. Br J Cancer 84:253-262 1581-1590
Morinaga S, Tsumuraya M, Nakajima T, et al (1986) Ciliated- Onoda N, Kang SM, Sugano S, et al (1995) Mucooepidermoid
cell adenocarcinoma of the pancreas. Acta Pathol Jpn 36: carcinoma of the pancreas: report of a case. Surg Today
1905-1910 25:843-847
Morita S, Okuda J, Sumiyoshi K, et al (1999) Pancreatic Ordonez NG (2001) Pancreatic acinar cell carcinoma. Adv
schwannoma: report of a case. Surg Today 29:1093-1097 Anat Patho18:144-59
Morohoshi T, Held G, Kloppel G (1983) Exocrine pancreatic Ordonez NG, Manning JT Jr (1984) Comparison of alpha-
tumors and their histological classification. A study based on I-antitrypsin and alpha-l-antichymotrypsin in hepato-
167 autopsy and 97 surgical cases. Histopathology 7:645-661 cellular carcinoma: an immunoperoxidase study. Am J
Morohoshi T, Kanda M, Horie A, et al (1987) Immunocyto- Gastroenterol 79:959-963
chemical markers of uncommon pancreatic tumors. Acinar Osborne BM, Culbert SJ, Cangir A, et al (1977) Acinar cell carci-
cell carcinoma, pancreatoblastoma, and solid cystic (papil- noma of the pancreas in a 9 year old child: case report with
lary-cystic) tumor. Cancer 59:739-747 electron microscopic observations. South Med J 70:370-372
Morohoshi T, Hamamoto T, Kunimura T, et al (1991) Epi- Owaga B, Okinaga K, Obana K, et al (2000) Pancreatoblastoma
dermoid cyst derived from an accessory spleen in the treated by delayed operation after effective chemotherapy.
pancreas. A case report with literature survey. Acta Pathol J Pediatr Surg 35:1163-1165
Jpn 41:916-921 Paal E, Thompson LD, Heffess CS (1998) A clinicopatho-
Morris-Stiff G, Vujanic GM, Al-Wafi A, Klimstra DS (1998) logic and immunohistochemical study of ten pancreatic
Pancreatic inflammatory pseudo tumor: an uncommon lymphangiomas and review of the literature. Cancer 82:
childhood lesion mimicking a malignant tumor. Pediatr 2150-2158
Surg Int 13:52-54 Pacchioni D, Papotti M, Macri L, et al (1996) Pancreatic ono-
Movahedi-Lankarani S, Hruban RH, Westra WH, et al (2000) cytic endocrine tumors. Cytologic features of two cases.
Primitive neurectodermal tumors of the pancreas: a report Acta Cyto140:742-746
of seven cases of a rare neoplasm. Am J Surg Pathol 26: Palazzo JP, Chang CD (1993) Inflammatory pseudotumor of
1040-1047 the pancreas. Histopathology 23:475-457
Mullick SS, Mody DR (1996) 'Osteoclastic' giant cell carcinoma Papotti M, Cassoni P, Taraglio S, et al (1"999) Oncocytic and
of the pancreas. Report of a case with aspiration cytology. oncocytoid tumors of the exocrine pancreas, liver and gas-
Acta Cytol 40:975-979 trointestinal tract. Semin Diagn Pathol 16: 126-134
Murakami T, Ueki K, Kawakami H, et al (1996) Pancreatoblas- Pascal RR, Sullivan L, Hauser L, et al (1989) Primary malig-
toma: case report and review of treatment in the literature. nant fibrous histiocytoma of the pancreas. Hum Pathol 20:
Med Pediatr Onco127:193-197 1215-1217
Nagashima Y, Misugi K, Tanaka Y, et al (1999) Pancreatoblas- Passmore SJ, Berry pJ, Oakhill A (1988) Recurrent pancre-
toma: a second report on cytogenetic findings. Cancer atoblastoma with inappropriate adenocorticotrophic hor-
Genet Cytogenet 109:178-179 mone secretion. Arch Dis Child 63:1494-1496
246 T. Oweity and A. B. West

Patel S, Bellon EM, Haaga J, et al (1980) Fat replacement of the cyst originating from an intrapancreatic accessory spleen.
exocrine pancreas. Am J Roentgenol 135:843-845 Abdom Imaging 27:560-562
Perez-Ordonez B, Naseem A, Lieberman PH, et al (1996) Solid Suits J, Frazee R, Erickson RA (1999) Endoscopic ultrasound
serous adenoma of the pancreas. The solid variant of and fine needle aspiration for the evaluation of pancreatic
serous cystadenoma? Am J Surg PathoI20:1401-1405 masses. Arch Surg 134:639-642
Petter LM, Martin JK Jr, Menke DM (1998) Localized lympho- Suster S, Phillips M, Robinson MJ (1989) Malignant fibrous
plasmacellular pancreatitis forming a pancreatic inflam- histocytoma (giant cell type) of the pancreas. A distinctive
matory pseudotumor. Mayo Clin Proc 73:447-50 variant of osteoclast-type giant cell tumor of the pancreas.
Posen JA (1981) Giant cell tumor of the pancreas of the osteo- Cancer 64:2303-2308
clastic type associated with a mucous secreting cystoande- Suzuki Y, Veda E, Takada K (1994) Intrapancreatic accessory
no carcinoma. Hum PathoI12:944-947 spleen mimicking islet cell tumor. Nichidokurihov 39:
Potts SR, Brown S, O'Hara MD (1986) Pancreatobastoma in a 34-35
neonate associated with Beckwith-Weidemann syndrome. Tang X, Tanka Y, Tsutsumi Y (1994) Epithelial inclusion cysts in
Z Kinderchir 41:56-57 an intrapancreatic accessory spleen. Pathol Int 44:652-654
Prayer L, Schurawitzki H, Mallek R, et al (1992) CT in pancre- Terhune PG, Memoli VA, Longnecker DS (1998) Evaluation
atic involvement of non -Hodgkin's lymphoma. Acta Radiol of p53 mutation in pancreatic acinar cell carcinomas of
33:123-127 humans and transgenic mice. Pancreas 16:6-12
Radin DR, Esplin JA, Levine AM, et al (1993) AIDS-related Tham RT, Heyerman HG, Falke TH, et al (1991) Cystic fibrosis:
non-Hodgkin's lymphoma: abdominal CT findings in 112 MR imaging of the pancreas. Radiology 179: 183-186
patients. Am J RoentgenoI160:1133-1139 Thompson K, Castelli MJ, Gattuso P (1998) Metastatic pap-
Reducka K, Gardiner GW, Sweet J, et al (1988) Myeloma-like illary oncocytic carcinoma of the pancreas to the liver
cast nephropathy associated with acinar cell carcinoma of diagnosed by fine needle aspiration. Diagn Cytopathol 18:
the pancreas. Am J Nephrol 8:421-424 291-296
Reed K, Vose PC, Jarstfer BS (1979) Pancreatic cancer: 30 year Thompson LD, Heffess CS (2000) Renal cell carcinoma to the
review (1947 to 1977). Am J Surg 138:929-933 pancreas in surgical pathology material. A clinicopatho-
Robbins DB, Katz RL, Evans DB, et al (1995) Fine-needle logic study of 21 cases with a review of the literature.
aspiration of the pancreas. In quest of accuracy. Acta Cytol Cancer 89:1076-1088
39:1-10 Thurnher MM, Schima W, Turetschek K, et al (2001) Peri-
Robin E, Terris B, Valverde A, et al (1997) [Pancreatoblastoma pancreatic fat necrosis mimicking pancreatic cancer. Eur
in adults J. Gastroenterol Clin BioI 21 :880-883 Radiol 11:922-925
Rosai J (1968) Carcinoma of pancreas simulating giant cell Tikkakoski T, Siniluoto T, Paivansalo M, et al (1992) Diagnos-
tumor of bone. Electron-microscopic evidence of its acinar tic accuracy of ultrasound -guided fine-needle pancreatic
cell origin. Cancer 22:333-344 biopsy. Rofo Fortschr Geb Rontgenstr Neuen Bildgeb Ver-
Sakai Y, Kupelioglu AA, Yanagisawa A, et al (2000) Origin of fahr 156:178-181
giant cells in osteoclast-like giant cell tumors of the pan- Trepeta RW, Mathur B, Lagin S, et al (1981) Giant cell tumor
creas. Hum Pathol 31:1223-1229 (,osteoclastoma') of the pancreas: a tumor of epithelial
Salvatore JR, Cooper B, Shah I, et al (2000) Primary pancreatic origin. Cancer 48:2022-2028
lymphoma: a case report, literature review, and proposal Tsutsumi S, Kojima T, Fukai Y, et al (2000) Epidermoid cyst of
for nomenclature. Med OncoI17:237-247 an intrapancreatic accessory spleen - a case report. Hepa-
Satake K, Arimoto Y, Fujimoto Y, et al (1991) Malignant T-cell togastroenterology 47: 1462-1464
lymphoma of the pancreas. Pancreas 6:120-124 Tuchek JM, De Jong SA, Pickleman J (1993) Diagnosis, sur-
Scott L, Blair G, Taylor G, et al (1988) Inflammatory pseudotu- gical intervention, and prognosis of primary pancreatic
mors in children. J Pediatr Surg 23:755-758 lymphoma. Am Surg 59:514-518
Shankar KR, Losty PD, Khine MM, et al (1998) Pancreatic Tucker JA, Shelburne JD, Benning TL, Yacoub L, Federman M
inflammatory tumor: a rare entity in childhood. JR ColI (1994) Filamentous inclusions in acinar cell carcinoma of
Surg Edinb 43:422-423 the pancreas. Ultrastructural PatholI8:279-286
Silverman JF, Holbrook CT, Pories WJ, et al (1990) Fine needle Tunell WP (1976) Hemangioendothelioma of the pancreas
aspiration cytology of pancreatoblastoma with immuno- obstructing the common bile duct and duodenum. J Pedi-
cytochemical and ultrastructural studies. Acta Cytol 34: atr Surg 11:827-830
632-640 Ulich T, Cheng L, Lewin KJ (1982) Acinar-endocrine cell tumor
Sironi M, Radice F, Taccagni GL, et al (1991) Fine needle of the pancreas. Report of a pancreatic tumor containing
aspiration of a pancreatic oxyphilic carcinoma with pul- both zymogen and neuroendocrine granules. Cancer 50:
monary and subcutaneous metastases. Cytopathology 2: 2099-2105
303-309 Urbanski SJ, Medline A (1982) Giant cell carcinoma of pan-
Skellenger ME, Kinner BM, Jordan PH Jr (1983) Brunner's creas with clear cell pattern in metastases. Hum Pathol 13:
gland harmartomas can mimic carcinoma of the head of 1047-1049
the pancreas. Surg Gynecol Obstet 156:774-776 Van Klaveren RJ, de Mulder PH, Boerbooms AM, et al (1990)
Solcia E, Capella C, Kloppel G (1997) Tumors of the pancreas. Pancreatic carcinoma with polyarthritis, fat necrosis, and
Atlas of Tumor Pathology, 3rd series, fascicle 20. Armed high serum lipase and trypsin activity. Gut 31 :953-955
Forces Institute of Pathology, Washington DC Voss M, Hammel P, Molas G, et al (2000) Value of endoscopic
Sommers SC, Meissner WA (1954) Unusual carcinomas of the ultrasound guided fine-needle aspiration biopsy in the
pancreas. Arch Pathol 58: 10 1-111 diagnosis of solid pancreatic masses. Gut 46:244-249
Sonomura T, Kataoka S, Chikugo T, et al (2002) Epidermioid Voss SD, Kruskal JB, Kane RA (1999) Chronic inflammatory
Rare Solid Tumors: Clinical Manifestations and Pathology 247

pseudotumor ansmg in the hepatobiliary-pancreatic cytometric analysis of a pancreatoblastoma. Cancer Genet


system: progressive multisystemic organ involvement in Cytogenet 79: 115-118
four patients. Am J Roentgenol 173:lO49-lO54 Wilson MB, Adams DB, Garen PD, et al (1992) Aspiration
Walsh MM, Brandspigel K (1989) Gastrointestinal bleeding cytologic, ultrastructural, and DNA cytometric findings
due to pancreatic schwannoma complicating von Reck- of solid and papillary tumor of the pancreas. Cancer 69:
linghausen's disease. Gastroenterology 97:1550-1551 2235-2243
Walsh SV, Evangelista F, KhettryV (1998) Inflammatory myo- Wreesmann V, Van Eijck CHJ, Naus DCWH, et al (2001)
fibroblastic tumor of the pancreatobiliary region: morpho- Inflammatory pseudotumor (inflammatory myofibroblas-
logic and immunocytochemical study of three cases. Am J tic tumor) of the pancreas: a report of six cases associated
Surg Pathol 22:412-418 with obliterative phlebitis. Histopathology 38:105-110
Watanabe M, Miura H, Inoue H, et al (1997) Mixed osteoclastic/ Yokomizo H, Hifumi M, Yamane T, et al (2002) Epidermoid
pleomorphic-type giant cell tumor of the pancreas with cyst of an accessory spleen at the pancreatic tail: diagnostic
ductal adenocarcinoma: histochemical and immunohisto- value of MRI. Abdom Imaging 27:557-559
chemical study with review of the literature. Pancreas 15: Zalatnai A, Kovacs M, Flautner L, et al (1998) Pancreatic leio-
201-208 myosarcoma. Case report with immunohistochemical and
Webb IN (1977) Acinar cell neoplasms of the exocrine pan- flow cytometric studies. Virchows Arch 432:469-472
creas. J Clin Pathol 30:lO3-112 Zamboni G, Pea M, Martignoni G, et al (1996) Clear cell 'sugar'
Webb TH, Lillemoe KD, Pitt HA, et al (1989) Pancreatic lym- tumor of the pancreas. A novel member of the family of
phoma. Is surgery mandatory for diagnosis or treatment? lesions characterized by the presence of perivascular epi-
Ann Surg 209:25-30 theloid cells. Am J Surg Pathol 20:722-730
Westra WH, Sturm P, Drillenburg P, et al (1998) K-ras onco- Zerbi A, De Nardi P, Braga M, et al (1993) An oncocytic carci-
gene mutations in osteoclast-like giant cell tumors of the noma of the pancreas with pulmonary and subcutaneous
pancreas and liver: genetic evidence to support origin from metastases. Pancreas 8: 116-119
the duct epithelium. Am J Surg PathoI22:1247-1254 Zuk RJ, Neal JW, Baithun SI (1990) Malakoplakia of the pan-
Wiley J, Posekany K, Riley R, et al (1995) Cytogenetic and flow creas. Virchows Arch [A] 417:181-184
14 Unusual Pancreatic Neoplasms: Imaging
A. J. MEGIBOW and I. R. FRANCIS

CONTENTS 14.1
Introduction
14.1 Introduction 249
14.2 Solid-Pseudopapillary Tumor of the Pancreas 250
14.2.1 Clinical Considerations 250 'Unusual' pancreatic neoplasms can be thought of
14.2.2 Imaging Features 250 as a heterogeneous group of tumors that are infre-
14.2.3 Treatment and Prognosis 251 quently seen in any given individual clinical prac-
14.3 Primary Pancreatic Lymphoma 252 tice. Even large referral centers may see fewer than
14.3.1 Clinical Considerations 252 10 cases over a decade. There is no strict definition
14.3.2 Imaging Features 252
14.3.3 Treatment and Prognosis 252 as to what tumors comprise this group of neoplasms.
14.4 Adenosquamous Carcinoma 253 Probably the single most important reason why these
14.4.1 Clinical Considerations 253 lesions are being seen with any frequency is directly
14.4.2 Imaging Features 254 related to the widespread use of imaging (STEPHENS
14.4.3 Treatment and Prognosis 254 1997). These lesions display clinical, demographic,
14.5 Acinar Cell Carcinoma 254
14.5.1 Clinical Considerations 254 and imaging features that are sufficiently different
14.5.2 Imaging Features 255 from pancreatic adenocarcinoma, islet-cell neo-
14.5.3 Treatment and Prognosis 257 plasms, or cystic tumors that an 'unusual' histology
14.6 Osteoclast-like Giant-Cell Tumor 257 is anticipated when they are encountered on imaging
14.6.1 Clinical Considerations 257
studies.
14.6.2 Imaging Features 257
14.6.3 Treatment and Prognosis 257 Table 14.1 is derived from the Armed Forces Insti-
14.7 Pancreatoblastoma 258 tute of Pathology (AFIP) classification of pancreatic
14.7.1 Clinical Considerations 258 tumors and contains the histological type of those
14.7.2 Imaging Features 258 lesions which may be thought of as 'unusual' tumors
14.7.3 Treatment and Prognosis 258 of the exocrine pancreas (SOLCIA et al. 1997). The
14.8 Mesenchymal and Ectopic Tumors 258
14.8.1 Mesenchymal Tumors - Introduction 258 list as published in the AFIP fascicle is by no means
14.8.1.1 Mature Teratoma 259 complete. Omitted from the list are the secondary
14.8.1.2 Lymphangioma 260 tumors (considered in Chapter 17), the tumor-like
14.8.1.3 Pancreatic Lipoma 260 conditions of the pancreas (e.g., chronic pancreati-
14.8.1.4 Other Mesenchymal Tumors 260 tis), and several varieties of inflammatory cysts. No
14.8.2 Ectopic Tumors - Introduction 261
14.8.2.1 Intrapancreatic Accessory Spleen 262 consideration will be given to unusual tumor-like
References 263 conditions of the endocrine pancreas. Because of
the rare occurrence of these lesions, we are unable
to provide imaging examples of all of the entities
listed. This chapter will consider imaging features
of solid-pseudopapillary tumor, primary pancreatic
A. J. MEGIBOW, MD,MPH, FACR lymphoma, adenosquamous tumor, acinar cell carci-
Professor of Radiology, NYU Medical Center, New York, NY, noma, osteoclastic giant-cell tumor, pancreatoblas-
USA
1. R. FRANCIS, MD
toma, and mesenchymal tumors including mature
Professor of Radiology, University of Michigan School of Medi- teratoma, lymphangioma, lipoma, and heterotopic
cine, Ann Arbor, Michigan, USA spleen.
250 A. J. Megibow and I. R. Francis

Table 14.1. Entities from Armed Forces Institute of Pathology tumors. The term 'Frantz's tumor' refers to the initial
(AFIP) classification of pancreatic tumors (SOLCIA et al. 1997) description of this lesion in the 1959 AFIP classifica-
considered as 'unusual' tumors
tion (FRANTZ 1959). The term solid-pseudopapillary
Ductal adenocarcinoma variants tumor has been proposed as the single best term for
Mucinous noncystic adenocarcinoma this lesion in both the AFIP classification of 1997 and
Signet-ring-cell carcinoma the World Health Organization (WHO) classification
Adenosquamous carcinoma
Undifferentiated (anaplastic) carcinoma of 1996 (KLOPPEL et al. 1996).
Osteoclast-like giant-cell tumor The female predominance of this lesion is well
Acinar cell carcinoma documented. BUETOW et al. (1996) reviewed the
Acinar cell carcinoma variants AFIP experience, finding 53 female patients out of 56
Acinar cell cystadenocarcinoma (94.6%). MARTIN et al. (2002) reviewed the experi-
Mixed acinar-endocrine carcinoma
Pancreatoblastoma ence at Memorial Sloan-Kettering Cancer Center in
Solid-pseudopapillary tumor New York. documenting 20 female patients out of 24
Miscellaneous carcinomas (83.3%). The reason for the female predominance is
Oncocytic carcinoma unclear. Despite sophisticated pathologic, genetic,
Choriocarcinoma and immunohistochemical analysis, no consistent
Nonmucinous, glycogen-poor cystadenocarcinoma
Mature teratoma evidence of hormone receptors has been found.
Nonepithelial tumors The age of these patients is considerably less than
Benign soft-tissue tumors typically seen in patients with pancreatic adenocar-
Malignant soft-tissue tumors cinoma. Mean ages reported range from 25 years
Malignant lymphoma (range 10-74 years) (BUETOW et al. 1996), 27 years
Cysts
Congenital cyst (LAM et al. 1999),32 years (range 16-66 years) (YOON
Lymphoepithelial cyst et al. 2001), to 39 years (range 12-79 years) (MARTIN
Endometrial cyst et al. 2002). A few elderly patients are encountered in
Heterotopic pancreas all series. Individual case reports in patients as young
Heterotopic (ectopic) spleen as 10 years old also appear in the literature (YAG I et
Hamartoma and pseudotumor
Pseudolipomatous hypertrophy
al. 1994). Metastatic disease has been reported in an
Pseudolymphoma 11-year-old patient (HORISAWA et al. 1995), although
there is evidence that the tumor is more aggressive
in elderly patients based on the higher prevalence of
14.2 metastases and microscopic evidence of angio- and
Solid-Pseudopapillary Tumor perineural invasion (MATSUNOU and KONISHI 1990;
of the Pancreas PIATEK et al. 2000). These authors stress that even in
the presence of these markers of advanced disease,
14.2.1 the prognosis is extremely favorable.
Clinical Considerations

This neoplasm is considered as either a benign tumor 14.2.2


or a tumor with low malignant potential occurring Imaging Features
predominantly in young women (MAO et al. 1995;
MARTIN et al. 2002; YAMAGUCHI and TANAKA 2001). Because of their large size and heterogeneous
A review of clinical experience with this tumor pub- morphology, these lesions are easily recognized on
lished in 1999 documents 452 cases described in the imaging studies. Most imaging descriptions of this
literature. Microscopic-based descriptions stress the lesion are based on the computed tomography (CT)
papillary features (hence the 'solid-papillary' nomen- appearance (Figs. 14.1, 14.3) On imaging studies, this
clature), whereas macroscopic-based descriptions lesion appears as a round, encapsulated mass with
emphasize the gross pathologic appearance (hence variable amounts of soft tissue and necrosis. The
the 'solid-cystic' nomenclature). Therefore, it is not appearance of the cystic regions suggests hemor-
surprising that this lesion has been referred to by a rhage as the major factor in producing the necrosis
variety of names including: benign or malignant pap- (CHO! et al. 1988; DONG et al. 1996). The lesions may
illary neoplasms, papillary cystic tumors of the pan- appear anywhere within the pancreas, although there
creas, papillary epithelial neoplasms, solid and papil- appears to be a slight predilection for the pancreatic
lary epithelial neoplasms, solid and cystic acinar cell tail. When detected, lesions are often quite large:
Unusual Pancreatic Neoplasms: Imaging 251

Fig. 14.1. Solid-pseudopapillary pancreatic neoplasm. A large Fig. 14.3. Solid-pseudopapillary pancreatic neoplasm. Low
mass is noted in the head of the pancreas. Note the well-defined attenuation lesion present in the pancreatic tail. The diagnosis
capsule. The mass contains a greater amount of soft-tissue atten- was suggested preoperatively based on the fact that the patient
uating (solid) tumor. Low attenuation necrotic regions are pres- was a 17-year-old woman
ent. Note the high attenuation material in the superior portion of
the mass (asterisks) compatible with hemorrhagic debris

1996). A frequently described feature is a well devel-


oped capsule resulting in a sharply circumscribed
lesion. The cystic regions within the tumor contain
variable amounts of soft tissue and liquefaction. The
presence of blood products within the mass produces
hyperintense signal on Tl-weighted MR images
(SAVCI et al. 1996). Fluid-debris levels resulting from
prior hemorrhage (recognizable by MR) have been
noted (BUETOW et al. 1996). There is a conspicuous
absence of internal septations, which may aid in the
differential diagnosis from serous and mucinous
cystic tumors (CHOI et al. 1988). BUETOW and col-
leagues found calcifications in 16 of 56 (29%) patients
(BUETOW et al. 1996), with both central and rim calci-
fication being reported (DONG et al. 1996).

Fig. 14.2. Solid-pseudopapillary pancreatic neoplasm in a 19-


year-old woman. A large mass is seen in the pancreatic head. A
well-defined capsule defines a predominantly fluid attenuating
14.2.3
mass. Several poorly defined densities of slightly higher atten- Treatment and Prognosis
uation are seen in the dependent portion. This mass could be
confused with a pseudocyst or cystic pancreatic neoplasm The biological behavior of solid-pseudopapillary
tumor of the pancreas is variable. Most authors con-
sider these lesions to be low-grade malignancies and
mean sizes reported are 7.5 cm (YAMAGUCHI et al. suggest aggressive, curative resection when discov-
1990},9 cm (BUETOW et al. 1996), and 11 cm (MAO ered (YOON et al. 2001; ZINNER 1995). In a review of
et al. 1995). 292 cases reported in the world literature, 43 (14.7%)
The imaging appearances are cataloged as solid were recognized as malignant (MAO et al. 1995). The
(Fig. 14.1), partially or completely cystic (Figs. 14.2, likelihood of malignancy increases with the patient's
14.3). However, even in the 'solid' tumors, some age (MATSUNOU and KONISHI 1990). Despite malig-
degree of cystic change and/or hemorrhagic degen- nant epithelial changes and even the presence of
eration (Fig. 14.1) can be identified (BUETOW et al. liver metastases, the long-term prognosis is good
252 A. J. Megibow and 1. R. Francis

for those patients in whom the total disease burden cm in diameter (PRAYER et al. 1992; TEEFEY et al.
can be resected (even with resectable extrapancre- 1986; VAN BEERS et al. 1993). The masses can extend
atic disease, the prognosis is excellent) (MARTIN et and infiltrate beyond the pancreas to encase major
al. 2002). Mean survival rates of 5.2 years are reported peripancreatic vascular structures similar to ductal
following successful resection (YOON et al. 2001). adenocarcinoma (BOUVET et al. 1998). However, the
degree of pancreatic ductal dilatation is minimal,
even with ductal invasion. This lesser degree of
ductal dilatation in the presence of a bulky homo-
14.3 geneous tumor mass should suggest the possibility
Primary Pancreatic Lymphoma of pancreatic lymphoma (Fig. 14.4). Alternatively,
diffuse infiltration and enlargement of the pancreas
14.3.1 without clinical signs of acute pancreatitis should
Clinical Considerations alert one to the possibility of pancreatic lymphoma
(Fig. 14.5). Finally, lymph node involvement below
Primary pancreatic lymphoma is an extremely rare the level of the renal veins, which can be seen in lym-
neoplasm. Eleven patients were seen at the M.D. phoma, is unusual in ductal adenocarcinoma. These
Anderson Cancer Center between 1980 and 1995 findings may prompt fine needle aspiration biopsy
(BOUVET et al. 1998); 5 of a total of 1212 patients (FNAB) of the pancreatic mass to obtain a diagnosis
with adult non-Hodgkin's lymphoma (NHL) were (MERKLE et al. 2000).
seen at the King Faisal Specialist Hospital between The classic ultrasound appearance of a lymphoma
1987 and 1994 (EZZAT et al. 1996). Seven patients anywhere in the body is that of a hypoechoic mass
with primary pancreatic lymphoma were treated at with poor through transmission (KAUDE and JOYCE
the Loyola Medical Center between 1976 and 1991 1980). Therefore, pancreatic lymphoma can be poten-
(TUCHEK et al. 1993). Pancreatic involvement as part tially confused with a pseudocyst, and thus color
of disseminated disease is more frequent but still Doppler may be useful to help distinguish between
relatively unusual (BORROWDALE and STRONG 1994). these diagnostic possibilities (LEVIN et al. 1993). The
Involvement of the pancreas was present in 9 of 402 availability of high frequency, longer focal length US
patients (2.2%) with NHL and 4.9% of all patients probes has shown that, in fact, many lymphomatous
presenting with pancreatic malignancies (9 of 182) at masses will display some degree of echogenicity.
the Johns Hopkins Medical Institutions between 1982 US should be thought of as an index examination,
and 1986 (WEBB et al. 1989). There is one reported bringing the patient to the attention of the physician.
case of non-cleaved small B-cell lymphoma in an Diagnosis will be obtained by FNAB of the pancreas
HIV-seropositive patient (JONES et al. 1997). or biopsy of peripheral lymph nodes. Some have
Patients present most commonly with abdominal reported that endoscopic ultrasound (EUS) is useful
pain of a severity engendering an imaging examina- for detecting pancreatic lymphoma, and EUS-guided
tion, such as US, CT or MR, with CT scanning being biopsy can minimize sampling errors that can occur
preferred in the USA and US in Europe and Asia. Labo- when 'blind' open biopsies are performed (HAR-
ratory values are nonspecific. Lactate dehydrogenase RISON et al. 1999). Most oncologists prefer to use
(LDH) levels may be elevated (BOUVET et al. 1998). conventional cross-sectional imaging techniques that
can survey the entire abdominal and pelvic cavities
(and thorax, if indicated).
14.3.2
Imaging Features
14.3.3
The lesion presents most commonly as a focal mass Treatment and Prognosis
and less frequently as diffuse pancreatic enlarge-
ment. When the lesion presents as a focal mass, Primary pancreatic lymphoma has a significantly
there is little to distinguish it from primary ductal better prognosis than adenocarcinoma. When che-
adenocarcinoma (TEEFEY et al. 1986; VAN BEERS et motherapy is instituted, complete remission can be
al. 1993). The mass is most frequently hypo dense and expected in the majority of patients (EZZAT et al.
slightly heterogeneous on dynamic CT (VAN BEERS 1996; WEBB et al. 1989). Significant morbidity and
et al. 1993). They are usually quite large at presenta- mortality are associated with postoperative com-
tion; reported sizes typically range between 5 and 7 plications that occur when patients are operated on
Unusual Pancreatic Neoplasms: Imaging 253

a b

c d

Fig.14.4a-d. Primary pancreatic lymphoma. a CT scan shows low attenuation mass in the pancreatic neck (arrow). The common
bile duct (CBD) is minimally enlarged. b Same patient, slightly more caudad level. A low attenuation mass is present in the
pancreatic head (arrow) . There is no pancreatic duct nor CBD dilatation. It would be difficult to exclude pancreatic ductal
adenocarcinoma on the basis of the CT appearance. c Arterial phase image from gadolinium-enhanced, Tl-weighted gradi-
ent recalled echo (GRE) sequence reveals the tumor as a hypointense mass in the pancreatic head. Compare with the brightly
enhanced pancreatic parenchyma. d Image at a level slightly caudad to c shows the hypointense mass against the brightly
enhanced pancreas. As in other images, note the lack of pancreatic and biliary dilatation. As on CT, it would be impossible to
differentiate this case from primary ductal adenocarcinoma. (Courtesy of Dr. Donald Hulnick, MD, Danbury, CT, USA)

for suspected adenocarcinoma of the pancreas, but tures characterized by the differentiation of both glan-
surgical pathology ultimately proves the lesion to be dular and squamous elements. Although few reports of
pancreatic lymphoma (UEDA et al. 2000). this lesion have appeared in the literature, all authors
agree that the lesion is highly aggressive, with a prog-
nosis actually worse than ductal adenocarcinoma.
The lesion is extremely rare, with 134 cases
14.4 reported between 1907 and 1999. The clinical symp-
Adenosquamous Carcinoma toms are similar to those associated with ductal
adenocarcinoma (MADURA et al. 1999). A series of
14.4.1 25 patients from the AFIP collected between 1961 and
Clinical Considerations 1994 represents the largest reported patient cohort.
The mean age was 65 years (range 28-82 years).
Adenosquamous carcinoma of the pancreas is a rare Weight loss and painless jaundice were the most fre-
exocrine neoplasm, with distinctive histological fea- quent symptoms. The head of the gland was the most
254 A. J. Megibow and 1. R. Francis

Fig. 14.5. Pancreatic lymphoma. Diffuse enlargement. Superfi- Fig. 14.6. Adenosquamous carcinoma of the pancreas. A bulky
cially analyzed, this image could be interpreted as acute pancre- mass occupies the body and tail of the pancreas. The mass
atitis. However, on the arterial phase image, there is no necrosis. is heterogeneous but predominantly of low attenuation. The
Note diffuse enhancement of the gland. The splenic vein is lesion invades the splenic hilum. The splenic vein is encased,
engulfed, and the tumor extends to involve the left kidney and a prominent gastroepiploic collateral vein is present. A
metastasis is present in the greater omentum (arrow)

frequent site, and multiple areas were involved in 5 14.4.3


cases (KARDON et al. 2001). Treatment and Prognosis
No specific serum tests are available, but in one
report, marked elevation of serum CA 19-9 was As difficult as it is to imagine, the prognosis for patients
found. In one patient, the level was 354.8 Ulml, and with adenosquamous carcinoma is significantly worse
in the second 2010 Ulml (NABAE et al. 1998). than for patients with ductal carcinoma. In the AFIP
series, the average survival was 5.8 months (KARDON
et al. 2001). For 8 patients with this lesion, the cumula-
14.4.2 tive I-year survival was 21.4% compared with 42.1%
Imaging Features for patients with ductal adenocarcinoma (YAMAGU-
CHI and ENJOJI 1991). In a series of 6 patients seen at
Imaging studies cannot differentiate adenosqua- Indiana University who underwent either pancreati-
mous carcinoma from ductal adenocarcinoma. The co duodenectomy or distal pancreatectomy, only one
most common presentation is that of a bulky mass was alive 9 months after surgery with residual tumor
extending through multiple portions of the pancreas around the portal vein seen on follow-up imaging. It
(Fig. 14.6). The typical low attenuation associated with is striking that all of the lesions were poorly differen-
squamous cancers is not evident in these tumors, and tiated and 5 of 6 were locally metastatic to nodes or
they simulate ductal adenocarcinoma in that the contiguous structures (MADURA et al. 1999).
masses are predominantly lower in attenuation than
the normal pancreatic parenchyma. Masses may result
in upstream duct dilatation (Fig. 14.7}. Involvement of
the large peripancreatic veins, particularly the splenic 14.5
and portal veins, is characteristic, although arterial Acinar Cell Carcinoma
invasion is not usually seen (KOMATSUDA et al. 2000;
MAKIYAMA et al. 1995). Tumoral calcification has been 14.5.1
reported as well (MAKIYAMA et al. 1995). As opposed Clinical Considerations
to other pancreatic tumors, adenosquamous cancers
appear to be Ga-67 avid (KuJI et al. 1997). Acinar cell carcinoma is a well described, albeit
rare pancreatic neoplasm, and accounts for 1% of
pancreatic exocrine neoplasms. The tumor occurs in
Unusual Pancreatic Neoplasms: Imaging 255

sized hypoechoic regions suggesting necrosis may


be seen (RADIN et al. 1986). Dual-phase helical CT
with bolus contrast enhancement will demonstrate
the mass as hyperdense related to the pancreas
during the arterial phase and isodense in the portal
venous phase (Fig. 14.8). The brisk enhancement of
the tumor results in an imaging appearance which
mimics a neuroendocrine neoplasm (MUSTERT et al.
1998). Central low attenuation representing necrosis
and or hemorrhage may be visualized (Fig. 14.9a,b).
Punctate calcifications have been reported in a few
cases (LIM et al. 1990; RADIN et al. 1986). These
imaging features support the similarity of acinar cell
tumors to neuroendocrine tumors by light micros-
copy (ORDONEZ and MACKAY 2000). Peripancreatic
venous invasion is common with tumor thrombus
found growing into the portal and or splenic veins
(UEDA et al. 1996). The degree of thrombosis can
be so prominent that the splenoportal system can
appear enlarged. Recently, a single case of an acinar
tumor arising within a branch pancreatic duct, mim-
icking intraductal papillary mucinous tumor (IPMT),
was reported (FABRE et al. 2001).
Fig. 14.7a,b. Adenosquamous tumor of pancreas. The upstream On MRI, the lesion appears markedly hyperintense
pancreatic duct is dilated (a). The mass (arrow) is seen in (b).
This lesion is indistinguishable from adenocarcinoma
on T2-weighted images (Fig. 14.9c) and hyperintense
on enhanced Tl-weighted images (Fig. 14.9d,e).After
the administration of mangafodopir trisodium (Mn-
adults in the 5th-6th decades with white men being DPDP; Teslascan; Nycomed, Amersham, Princeton,
predominantly affected and has been also reported NJ), the lesion enhances brightly. This brisk enhance-
in children (KLIMSTRA et al. 1992). Clinical presen- ment is seen in functioning pancreatic parenchyma
tations are nonspecific, and jaundice is rare even in and in functioning pancreatic tumors (SAHANI et al.
tumors within the pancreatic head.
A widely reported clinical syndrome manifested
by any or all of the symptoms including subcuta-
neous or interosseous fat necrosis, panniculitis,
polyarthralgias, and eosinophilia has been reported
(KUERER et al. 1997; MACMAHON et al. 1965; RADIN
et al. 1986). The syndrome occurs in association with
elevated serum lipase levels. Despite the widespread
knowledge of this association, this has been reported
in approximately 16% of cases (KLIMSTRA et al.
1992). CEA and or CA 19-9 levels may remain normal
(CHEN et al. 2001). Elevation of serum alpha-fetopro-
tein has been reported in tumors characterized by
acinar cell differentiation (CINGOLANI et al. 2000).

14.5.2
Imaging Features
Fig. 14.8. Acinar cell carcinoma. Arterial phase CT image
reveals hyperdense mass in the pancreatic body. Low attenu-
The masses are usually large, encapsulated, and ation clefts centrally presumably represent necrotic regions.
heterogeneous as seen on CT (Figs. 14.8, 14.9a,b) or The similarity to neuroendocrine tumor is evident (© 1998
US. US through transmission is good, and variably American Roentgen Ray Society, used with permission)
256 A. J. Megibow and 1. R. Francis

a b

c d

Fig. 14.9a-e. Acinar cell carcinoma. a Noncontrast CT reveals


bulky mass in pancreatic head. There was no evidence of
biliary or pancreatic duct obstruction. b Same patient as in a.
Portions of the mass enhance during the arterial phase. Large
areas of low attenuation are consistent with necrosis andlor
hemorrhage. c Fat-suppressed T2-weighted MR reveals the
mass to be hyperintense. d Gadolinium-DTPA (Gd-DTPA)-
enhanced, fat-suppressed Tl-weighted GRE MR image in late
arterial phase. The contrast enhancement is more apparent
than on the CT scan. The necrotic region is clearly delineated.
e Contrast-enhanced, fat-suppressed, Tl-weighted GRE MR
image in portal venous phase. The mass enhances brightly.
e The characteristic capsule is evident as is the necrotic center
Unusual Pancreatic Neoplasms: Imaging 257

2002). Mn-DPDP MR would therefore be useful in the of osteoclast-like giant cells is extremely rare (ROSAI
subset of acinar tumors which are 'functioning'. 1968; SCOTT et al. 1993). This tumor is characterized
as a malignant neoplasm of the pancreas contain-
ing osteoclast-like giant cells. The tumor has been
14.5.3 reported most frequently in the pancreas, but it also
Treatment and Prognosis occurs in the parotid, thyroid, skin, orbit, kidney, and
breast. Clinically, patients present with an abdominal
Acinar cell carcinoma is an aggressive lesion. Some mass which immediately results in an imaging study
50% of patients present with metastases, which are (SCOTT et al. 1993; LEIGHTON and SHUM 2001).
commonly seen in the liver, local lymph nodes or the
adjacent peripancreatic fat, while 25% of patients will
develop metastases while under treatment. Mean sur- 14.6.2
vival time in 28 cases reviewed by Klimstra and associ- Imaging Features
ates was 18 months, with 57% alive at 1 year and 26%
alive at 3 years. Survival was better in younger patients These lesions present as large cystic masses with
with localized disease (KLIMSTRA et al. 1992), but sur- focal hemorrhage or necrosis (LEIGHTON and SHUM
vival times of longer than 6 years have been reported 2001). The lesion has a predilection for local invasion
in patients presenting with widespread metastatic and lymph node metastases (GIL-GARCIA et al. 1992;
disease (KUOPIO et al. 1995). Surgery with attempted SHINDOH et al. 1998). The mass may contain septa with
removal of all of the tumor is the best chance for sur- multiple loculations with mural nodules simulating a
vival, but on the whole, the prognosis is similar to that cystadenocarcinoma (Fig. 14.10). Without careful
of ductal adenocarcinoma. scrutiny of the imaging findings, the mass could be
confused with a pseudocyst (OEHLER et al. 1997).

14.6 14.6.3
Osteoclast-like Giant-Cell Tumor Treatment and Prognosis

14.6.1 In all cases, resection should be attempted, but the


Clinical Considerations local invasiveness of the lesion may make total resec-
tion impossible. Assuming complete excision is pos-
While the presence of giant cells in ductal adenocar- sible and no lymph node metastases are present, no
cinoma is a frequent finding at histology, the presence benefit accrues in using adjuvant radiation. Although

a b

Fig. 14.10a,b. Osteoclast-like giant -cell tumor. A large, multiseptate mass is present in the pancreatic tail. Several soft-tissue
attenuation regions are present. b Same patient as in a. Note the irregularly thickened septa and the soft-tissue elements located
inferiorly within the tumor. This lesion is easily confused with mucinous cystic tumor. The septa and mural nodules should
eliminate pancreatic pseudocyst as a diagnostic consideration
258 A. J. Megibow and 1. R. Francis

there are reports of patients surviving up to 10 years Other findings included calcification (2 of 10), biliary
from the time of diagnosis, the overall prognosis is and pancreatic ductal dilatation (1 of 10), and ascites
poor with a median survival of 11 months, similar to (3 of 10). Hepatic (2 patients) and pelvic (2 patients)
survival times with ductal adenocarcinoma (LEIGH- metastases were present in a small number of these
TON and SHUM 2001). patients. Adenopathy (2 patients) and vascular invasion
(1 patient) which were seen at surgery and pathology
were not identified radiologically (MONTEMARANO et
al. 2000). The mass will be hypoechoic on US (ROE-
14.7 BUCK et al. 2001). Tumors had low to intermediate
Pancreatoblastoma signal intensity on Tl-weighted images and high signal
intensity on T2-weighted images (MONTEMARANO et
14.7.1 al. 2000). ROEBUCK and colleagues emphasized the pre-
Clinical Considerations dilection for origin in the body and tail of the pancreas,
with a tendency toward involvement of the entire gland.
Pancreatoblastoma is a malignant pancreatic neoplasm Although one report comments on the frequency of
that displays a bimodal peak frequency, occurring in vascular invasion (GUPTA et al. 2000), most others
children (mean age 2.4 years) and, less frequently, in indicate that vascular invasion is, in fact, infrequent
adults (mean age 40 years). Boys are affected more fre- (ROEBUCK et al. 2001).
quently than girls (KLIMSTRA et al. 1995; LEVEY and
BANNER 1996). The tumor is second in frequency to
solid-pseudopapillary pancreatic tumors in children 14.7.3
with pancreatic neoplasms (SHORTER et al. 2002). Treatment and Prognosis
Patients most frequently present with an abdominal
mass and consequent vomiting, constipation, or early The biological behavior of the tumor is variable. The
satiety. Pain, weight loss, and jaundice occur less fre- tumor is thought to be slow growing because of the
quently (KLIMSTRA et al. 1995; ROEBUCK et al. 2001). frequency of presentation as a large mass. Metastases
Patients with this neoplasm may demonstrate elevated occur in 36% of patients, most frequently in the liver
serum levels of alpha-fetoprotein (particularly when (KLIMSTRA et al. 1995). The best treatment option
there are liver metastases), alpha-I-antitrypsin, and appears to be complete surgical excision, and the
LDH levels. Elevated levels of adrenal corticotrophic tumors in children appear to be more frequently
hormone (ACTH) are thought to be responsible for resectable than those in adults (SHORTER et al. 2002).
the Cushing-like syndrome or the syndrome of inap- Recurrence rates are high, but survival is improved
propriate ADH secretion. The tumor has also been by aggressive resection of the recurrent masses.
associated with the Beckwith-Wiedemann syndrome Unresectable disease has a poor prognosis (CHUN
(ROEBUCK et al. 2001). et al. 1997). The role of adjuvant radiation and che-
Pathologically, these encapsulated tumors contain motherapy has not been established (KLIMSTRA et
tissue closely resembling the fetal pancreas. Fre- al. 1995; MONTEMARANO et al. 2000; ROEBUCK et al.
quently, one sees small areas of squamoid corpuscles 2001; SHORTER et al. 2002). When the tumor presents
(MONTEMARANO et al. 2000; ROEBUCK et al. 2001) in adults, the prognosis is significantly worse, with a
mean survival of 18 months (KLIMSTRA et al. 1995).

14.7.2
Imaging Features
14.8
The imaging features of pancreatoblastoma are well Mesenchymal and Ectopic Tumors
described. The masses are large (reported mean size
approximately 10 cm) and lobulated (LEE et al. 1996). 14.8.1
MONTEMARANO and colleagues reviewed the imaging Mesenchymal Tumors - Introduction
and surgical findings in 10 patients (age range 2-20
years; mean age 6.8years).Well-definedmargins (90f10) Mesenchymal tissues, supporting connective tissue,
correlated with the gross, morphologically evident vessels, nerves, and lymphatics provide the stroma
capsules (Fig. 14.11). The texture was heterogeneous for the specialized epithelial cells in all organs of the
(9 of 10), and enhancement was seen in all tumors. body. These elements are derived from the meso-
Unusual Pancreatic Neoplasms: Imaging 259

Fig. 14.1 la-d. Pancreatoblastoma in an adult man. a Gross specimen showing solid well-demarcated mass. b Microscopic section
of the tumor displays large nodules separated by thin fibrous strands similar to acinar cell carcinoma. c Contrast -enhanced CT
examination shows the mass appearing to be well-demarcated, hypodense, and nonhomogeneous. d Gd-DTPA-enhanced GRE
Tl-weighted MR image shows minimal enhancement

dermal cells in the embryo. Anyone of a number CEBRIAN et al. 1998). The cases that appear to be true
of elements can give rise to a benign or malignant dermoid cysts occur in a younger age group (mean
neoplasm. It is estimated that these lesions account age 23 years, range 2-53 years), with no gender pre-
for 1%-2% of all pancreatic neoplasms (FERROZZI dominance. The cysts are lined with a squamous epi-
et al. 2000). This next section describes the imaging thelium, but mucinous epithelium, respiratory-type
findings in several of these lesions. mucosa, and sebaceous units are readily identifiable
in dermoid cysts, and they may contain hair (ADSAY
74.8.7.7 et al. 2000).
Mature Teratoma The imaging appearance of pancreatic teratoma is
similar to those seen elsewhere in the body (such as
These are extremely rare tumors arising from intra- in the adnexa) and is related to the amount of pre-
pancreatic embryonic rests. Fewer than 20 cases ponderant tissue within the teratoma. Fat with fat/
have been described in the literature (FERNANDEZ- fluid levels are diagnostic (Fig. 14.12), and septations
260 A. J. Megibow and I. R. Francis

a _~a.....J b

Fig. 14.12a,b. Mature teratoma. a A fat density mass is seen in the pancreatic body. A soft-tissue nodule (arrow) is seen in the
inferior portion, adjacent to dense calcification. b A fat attenuation mass is seen in the pancreatic tail. The fat is not homoge-
neous, with lower attenuation at the superior portion of the mass

and calcifications have been reported (FERROZZI et injection (BISHOP and STEER 2001; GRAY et al. 1998;
al. 2000; JACOBS and DINSMORE 1993). KOENIG et al. 2001; PANDOLFO et al. 1985; VIOLA et al.
1997). The differential diagnosis includes mucinous
74.8.7.2 cystic tumor and pancreatic pseudocyst. Aspiration
Lymphangioma of the cyst content may provide the diagnosis when
the fluid is chylous (PAAL et al. 1998).
Lymphangiomas are congenital abnormalities of Lymphangiomas are benign but can be locally inva-
the lymphatics that occur predominantly in the sive. Complete surgical excision is curative. Incomplete
pancreatic head and neck, frequently in children excision results in local recurrence (PAAL et al. 1998).
(KHANDELWAL et al. 1995). Abdominal sites account
for 1% of all lymphangiomas, with mesentery and 74.8.7.3
retroperitoneum (perirenal) accounting for the vast Pancreatic Lipoma
majority (ABE et al. 1997). Pancreatic lymphan-
giomas are extremely rare (BISHOP and STEER 2001; These are extraordinarily rare tumors. The first case
GRAY et al. 1998; VIOLA et al. 1997). on CT was described in 1996 (DI MAGGIO et al. 1996).
The clinical symptoms are nonspecific and include They are identical to lipomas arising in other loca-
abdominal pain and a palpable mass. All ages are tions. Their capsule allows for relatively easy enucle-
affected, and a female predominance is noted. Symp- ation (FERROZZI et al. 2000). The tumors are detected
toms evolve slowly in adults but may present acutely as incidental findings on imaging studies performed
in children (KOENIG et al. 2001). The tumor is con- for other reasons. CT scans are diagnostic, show-
sidered to arise from the pancreas if it is within the ing well-circumscribed masses within the pancreas
parenchyma, adjacent to the gland, or attached by a composed almost entirely of fat, with a few scattered
pedicle (PAAL et al. 1998). vessels or septa or both {Fig. 14.14) (KATZ et al. 1998).
At imaging, the tumor appears as a homogeneous The presence of a capsule allows for differentiation
cystic mass (Fig. 14.13). Following IV contrast, there from pancreatic fatty infiltration (JACOBS et al. 1994).
may be mural enhancement and visualization of fine Conservative management is indicated.
septa. When pedunculated, the mass may appear as
a juxtapancreatic cyst. Local mass effect on adjacent 74.8.7.4
organs is present when the masses achieve a large Other Mesenchymal Tumors
size. The masses are hypoechoic on US. MRI will
confirm a hyperintense cyst content on T2-weighted As noted above, any connective tissue element can
sequences with low signal on Tl-weighted sequences. give rise to a mesenchymal neoplasm. Schwannoma
The wall and the septa enhance following gadolinium may be characterized by two distinct architectural
Unusual Pancreatic Neoplasms: Imaging 261

a b

Fig. 14.13a. Cystic lymphangioma, pancreatic tail. A low attenuation mass (M) grows between the pancreatic body (B) and the
tail (T). The features are nonspecific, and the differential diagnosis could be that of any 'macrocystic' pancreatic neoplasm or
a pseudocyst. b Cystic lymphangioma, pancreatic body. A unilocular cystic mass projects into the lesser sac from the ventral
portion of the pancreatic body

a b

Fig. 14.14a,b. Pancreatic lipoma: a sharply circumscribed, fat attenuation mass is present in the pancreatic head. The mass dis-
places adjacent parenchyma and does not alter the contour of the superior mesenteric vein as it travels adjacent to the mass

patterns: (a) the highly organized cellular Antoni-A Sarcomas are extremely rare, accounting for 0.6%
component and (b) the loose hypo cellular Antoni- of all pancreatic neoplasms. Malignant fibrous his-
B component. The Antoni-A predominant tumor tiocytoma, Kaposi sarcoma, and leiomyosarcoma
is hypervascular, and tumors with this architecture have been described. More frequently, the pancreas is
appear as hyperdense lesions with central necrosis secondarily involved by invasion from the retroperi-
simulating islet-cell tumors. In those lesions with an toneum or adjacent portion of the gastrointestinal
Antoni-B predominance, the lesion appears more tract (FERROZZI et al. 2000).
cystic. Neurofibromas in the pancreas are most often
seen in patients with neurofibromatosis as opposed
to isolated lesions. In these patients, there is a high 14.8.2
frequency of neurofibrosarcoma. Neurofibromas are Ectopic Tumors - Introduction
frequently low attenuating masses, probably related to
the lipid content of the Schwann cells composing them. A variety of ectopic tissues may arise within the pan-
Larger, more heterogeneous lesions are more likely to creas and locally grow to simulate a mass. When the
be malignant. Frequently, neurofibromas encase the mass is composed of disordered normal tissue, it can
celiac axis, simulating locally invasive pancreatic ductal be defined as a hamartoma. In other cases, the mass
adenocarcinoma (FERROZZI et al. 2000). can be composed of tissues which are histologically
262 A. J. Megibow and I. R. Francis

identical to those seen within the normal organ, such spleen (SICA and REED 2000), it is not seen in clinical
as endometrial rests. practice with this frequency.
When present on imaging, the lesion appears as a
14.8.2.1 well circumscribed mass with enhancement charac-
Intrapancreatic Accessory Spleen teristics paralleling the enhancement of the normal
spleen and can be confused for a hypervascular
An accessory spleen is a common finding on CT pancreatic mass arising from the tail of the pancreas
imaging. These are usually not even subjected to (CHUREI et al. 1998; HARRIS et al. 1994; HAYWARD et
further imaging because of their spherical shape al. 1992). The mass may be most frequently confused
and attenuation identical to splenic tissue. Acces- with islet-cell tumor or metastatic tumor such as
sory spleens are estimated to occur in 30% of autopsy from renal cell cancer (Fig. 14.15). If the diagnosis
cases (SICA and REED 2000). They originate embryo- is considered, MR can be useful in confirming that
logically from the dorsal mesentery of the stomach the lesion 'tracks' with splenic signal on a variety of
and or pancreas (DODDS et al. 1990), or following unenhanced and enhanced sequences (CHUREI et al.
disruption of the spleen by surgery or trauma (sple- 1998). The diagnosis can be proven with scintigraphy
nosis). Although intrapancreatic accessory spleen using 99ffiTc_sulfur colloid or lllIn-labeled damaged
is estimated to occur in 17% of cases of accessory red blood cells (SICA and REED 2000).

a b

Fig.14.15a-c. Intrapancreatic accessory spleen. a Mass of soft-


tissue attenuation is present (arrow) in the splenic tail. b Arte-
rial phase imaging shows significant enhancement of the mass
(arrow). c The mass (arrow) continues to remain hyperdense.
Note the similar attenuation to the spleen. (Courtesy of Dr.
c Dennis Balfe, MD, St. Louis, MO, USA)
Unusual Pancreatic Neoplasms: Imaging 263

References

Abe H, Kubota K, Noie T, Banda Y, Makuuchi M (1997) Cystic Gray G, Fried K, Iraci J (1998) Cystic lymphangioma of the
lymphangioma of the pancreas: a case report with special pancreas: CT and pathologic findings. Abdom Imaging
reference to embryological development. Am J Gastroen- 23:78-80
terol92:1566-1567 Gupta AK, Mitra DK, Berry M, Dinda AK, Bhatnagar V (2000)
Adsay NV, Hasteh F, Cheng JD, Klimstra DS (2000) Squamous- Sonography and CT of pancreatoblastoma in children. AJR
lined cysts of the pancreas: lymphoepithelial cysts, der- Am J RoentgenoI174:1639-1641
moid cysts (teratomas), and accessory-splenic epidermoid Harris GN, Kase DJ, Bradnock H, MCKinley MJ (1994) Acces-
cysts. Semin Diagn PathoI17:56-65 sory spleen causing a mass in the tail of the pancreas: MR
Bishop MD, Steer M (2001) Pancreatic cystic lymphangioma imaging findings. AJR Am J RoentgenoI163:1120-1121
in an adult. Pancreas 22:lO1-lO2 Harrison JL, Millikan KW, Prinz RA, Zaidi S (1999) Endoscopic
Borrowdale R, Strong RW (1994) Primary lymphoma of the ultrasound for diagnosis and staging of pancreatic tumors.
pancreas. Aust N Z J Surg 64:444-446 Am Surg 65:659-664; discussion 664-665
Bouvet M, Staerkel GA, Spitz FR, Curley SA, Charnsangavej C, Hayward I, Mindelzun RE, Jeffrey RB (1992) Intrapancreatic
Hagemeister FB, Janjan NA, Pisters PW, Evans DB (1998) accessory spleen mimicking pancreatic mass on CT. J
Primary pancreatic lymphoma. Surgery 123:382-390 Comput Assist Tomogr 16:984-985
Buetow PC, Buck JL, Pantongrag-Brown L, Beck KG, Ros PR, Horisawa M, Niinomi N, Sato T, Yokoi S, Oda K, Ichikawa M,
Adair CF (1996) Solid and papillary epithelial neoplasm of Hayakawa S (1995) Frantz's tumor (solid and cystic tumor
the pancreas: imaging-pathologic correlation on 56 cases. of the pancreas) with liver metastasis: successful treatment
Radiology 199:707-711 and long-term follow-up. J Pediatr Surg 30:724-726
Chen JD, Wu MS, Tien YW, Kuo KT, Chang MC, Lin JT (2001) Jacobs JE, Dinsmore BJ (1993) Mature cystic teratoma of the
Acinar cell carcinoma with hypervascularity. J Gastroen- pancreas: sonographic and CT findings. AJR Am J Roent-
terol HepatoI16:107-111 genoI160:523-524
Choi BI, Kim KW, Han MC, Kim YI, Kim CW (1988) Solid and Jacobs JE, Coleman BG,Arger PH, Langer JE (1994) Pancreatic
papillary epithelial neoplasms of the pancreas: CT findings. sparing of focal fatty infiltration. Radiology 190:437-439
Radiology 166:413-416 Jones WF, Sheikh MY, McClave SA (1997) AIDS-related non-
Chun Y, Kim W, Park K, Lee S, Jung S (1997) Pancreatoblas- Hodgkin's lymphoma of the pancreas. Am J Gastroenterol
toma. J Pediatr Surg 32:1612-1615 92:335-338
Churei H, Inoue H, Nakajo M (1998) Intrapancreatic accessory Kardon DE, Thompson LD, Przygodzki RM, Heffess CS (2001)
spleen: case report. Abdom Imaging 23:191-193 Adenosquamous carcinoma of the pancreas: a clinicopath-
Cingolani N, Shaco-Levy R, Farruggio A, Klimstra DS, Rosai J ologic series of 25 cases. Mod PathoI14:443-451
(2000) Alpha-fetoprotein production by pancreatic tumors Katz DS, Nardi PM, Hines J, Barckhausen R, Math KR, Fruauff
exhibiting acinar cell differentiation: study of five cases, one AA, Lane MJ (1998) Lipomas of the pancreas. AJR Am J
arising in a mediastinal teratoma. Hum PathoI31:938-944 Roentgenol 170: 1485-1487
Di Maggio EM, Solcia M, Dore R, Preda L, La Fianza A, Rodino Kaude JV,Joyce PH (1980) Evaluation of abdominal lymphoma
C, Camp ani R (1996) Intrapancreatic lipoma: first case by ultrasound. Gastrointest Radiol 5:249-254
diagnosed with CT. AJR Am J Roentgenol 167:56-57 Khandelwal M, Lichtenstein GR, Morris JB, Furth EE, Long WB
Dodds WJ, Taylor AJ, Erickson SJ, Stewart ET, Lawson TL (1995) Abdominal lymphangioma masquerading as a pan-
(1990) Radiologic imaging of splenic anomalies. AJR Am creatic cystic neoplasm. J Clin GastroenteroI20:142-144
J RoentgenoI155:805-810 Klimstra DS, Heffess CS, Oertel JE, Rosai J (1992) Acinar cell
Dong PR, Lu DS, Degregario F, Fell SC, Au A, Kadell BM (1996) carcinoma of the pancreas. A clinicopathologic study of 28
Solid and papillary neoplasm of the pancreas: radiological- cases. Am J Surg PathoI16:815-837
pathological study of five cases and review of the literature. Klimstra DS, Wenig BM, Adair CF, Heffess CS (1995) Pancre-
Clin Radiol 51:702-705 atoblastoma. A clinicopathologic study and review of the
Ezzat A, Jamshed A, Khafaga Y, Rahal M, Linjawi T, Martin J, literature. Am J Surg Pathol 19: 13 71-1389
Taha I (1996) Primary pancreatic non-Hodgkin's lympho- Kloppel G, Solcia E, Longnecker DS, Capella C, Sobin LH
mas. J Clin GastroenteroI23:109-112 (1996) World Health Organization International Classifica-
Fabre A, Sauvanet A, Flejou JF, Belghiti J, Palazzo L, Ruzniewski tion of tumors. Histological Classification of tumors of the
P, Degott C, Terris B (2001) Intraductal acinar cell carci- exocrine pancreas. Springer, Berlin Heidelberg New York
noma of the pancreas. Virchows Arch 438:312-315 Koenig TR, Loyer EM, Whitman GJ, Raymond AK, Charnsan-
Fernandez-Cebrian JM, Carda P, Morales V, Galindo J (1998) gavej C (2001) Cystic lymphangioma of the pancreas. AJR
Dermoid cyst of the pancreas: a rare cystic neoplasm. Am J Roentgenol177:1090
Hepatogastroenterology 45: 1874-1876 Komatsuda T, Ishida H, Konno K, Sato M, Watanabe S, Furuya
Ferrozzi F, Zuccoli G, Bova D, Calculli L (2000) Mesenchymal T, Ishida J (2000) Adenosquamous carcinoma of the pan-
tumors of the pancreas: CT findings. J Comput Assist creas: report of two cases. Abdom Imaging 25:420-423
Tomogr 24:622-627 Kuerer H, Shim H, Pertsemlidis D, Unger P (1997) Functioning
Frantz V (1959) Tumors of the pancreas. In: Stout A (ed) pancreatic acinar cell carcinoma: immunohistochemical
Tumors of the pancreas, vol VII. Armed Forces Institute of and ultrastructural analyses. Am J Clin Oncol 20: 101-1 07
Pathology, Washington DC, pp 32-33 Kuji I, Sumiya H, Taki J, Nakajima K, Yokoyama K, Kinuya S,
Gil-Garcia I, Valls C, Sanchez-Marquez A, Catala I (1992) Kinuya K, Ichikawa A, Konishi S, Michigishi T, Tonami N
Osteoclast-type giant-cell tumor of the pancreas. AJR Am (1997) Intense Ga-67 uptake in adenosquamous carcinoma
J Roentgenol159:1l28 of the pancreas. Ann Nucl Med 11:41-43
264 A. J. Megibow and I. R. Francis

Kuopio T, Ekfors TO, Nikkanen V, Nevalainen TJ (1995) Acinar lymphangiomas and a review of the literature. Cancer 82:
cell carcinoma of the pancreas. Report of three cases. 2150-2158
Apmis 103:69-78 Pandolfo I, Scribano E, Gaeta M, Fiumara F, Longo M (1985)
Lam KY, Lo CY, Fan ST (1999) Pancreatic solid-cystic-papil- Cystic lymphangioma of the pancreas: CT demonstration.
lary tumor: clinicopathologic features in eight patients J Comput Assist Tomogr 9:209-210
from Hong Kong and review of the literature. World J Surg Piatek S, Manger T, Rose I, Schulz HU, Lippert H (2000) Solid
23:1045-1050 pseudopapillary tumor of the pancreas. Int J Pancreatol
Lee JY, Kim 10, Kim WS, Kim CW, Yeon KM (1996) CT and us 27:77-81
findings of pancreatoblastoma. J Comput Assist Tomogr Prayer L, Schurawitzki H, Mallek R, Mostbeck G (1992) CT in
20:370-374 pancreatic involvement of non-Hodgkin lymphoma. Acta
Leighton CC, Shum DT (2001) Osteoclastic giant cell tumor of RadioI33:123-127
the pancreas: case report and literature review. Am J Clin Radin DR, Colletti PM, Forrester DM, Tang WW (1986)
OncoI24:77-80 Pancreatic acinar cell carcinoma with subcutaneous and
Levey JM, Banner BF (1996) Adult pancreatoblastoma: a case intraosseous fat necrosis. Radiology 158:67-68
report and review of the literature. Am J Gastroenterol 91: Roebuck DJ, Yuen MK, Wong YC, Shing MK, Lee CW, Li CK
1841-1844 (2001) Imaging features of pancreatoblastoma. Pediatr
Levin MF, Bach DB, Vellet AD, Munk PL, Downey DB, Reitz WG RadioI31:501-506
(1993) Sonolucent peripancreatic masses: differential diag- Rosai J (1968) Carcinoma of pancreas simulating giant cell
nosis and related imaging. Can Assoc Radiol J 44:168-175 tumor of bone. Electron-microscopic evidence of its acinar
Lim JH, Chung KB, Cho OK, Cho KS (1990) Acinar cell car- cell origin. Cancer 22:333-344
cinoma of the pancreas. Ultrasonography and computed Sahani D, Prasad SR, Maher M, Warshaw AL, Hahn PF, Saini S
tomography findings. Clin Imaging 14:301-304 (2002) Functioning acinar cell pancreatic carcinoma: diag-
MacMahon HE, Brown PA, Shen EM (1965) Acinar cell car- nosis on mangafodipir trisodium (Mn-DPDP)-enhanced
cinoma of the pancreas with subcutaneous fat necrosis. MRI. J Comput Assist Tomogr 26:126-128
Gastroenterology 49:555-559 Savci G, KiJicturgay S, Sivri Z, Parlak M, Tuncel E (1996) Solid
Madura JA, Jarman BT, Doherty MG, Yum MN, Howard TJ and papillary epithelial neoplasm of the pancreas: CT and
(1999) Adenosquamous carcinoma of the pancreas. Arch MR findings. Eur Radiol 6:86-88
Surg 134:599-603 Scott R, Jersky J, Hariparsad G (1993) Case report: malignant
Makiyama K, Takuma K, Zea-Iriarte WL, Ikuno N, Kawatomi giant cell tumour of the pancreas presenting as a large
M, Mori N, Ishino T, Yonemitsu N (1995) Adenosquamous pancreatic cyst. Br J RadioI66:1055-1057
carcinoma of the pancreas. J Gastroenterol 30:798-802 Shindoh N, Ozaki Y, Kyogoku S, Nakanishi A, Sumi Y, Katayama
Mao C, Guvendi M, Domenico DR, Kim K, Thomford NR, H (1998) Osteoclast-type giant cell tumor of the pancreas:
Howard JM (1995) Papillary cystic and solid tumors of helical CT scans. AJR Am J Roentgenol 170:653-654
the pancreas: a pancreatic embryonic tumor? Studies of Shorter NA, Glick RD, Klimstra DS, Brennan MF, Laquaglia
three cases and cumulative review of the world's literature. MP (2002) Malignant pancreatic tumors in childhood and
Surgery 118:821-828 adolescence: the Memorial Sloan-Kettering experience,
Martin RC, Klimstra DS, Brennan MF, Conlon KC (2002) Solid- 1967 to present. J Pediatr Surg 37:887-892
pseudopapillary tumor of the pancreas: a surgical enigma? Sica GT, Reed MF (2000) Case 27: intrapancreatic accessory
Ann Surg Oncol 9:35-40 spleen. Radiology 217:134-137
Matsunou H, Konishi F (1990) Papillary-cystic neoplasm of SoJcia E, Capella C, Kloppel G (1997) Tumors of the pancreas.
the pancreas. A clinicopathologic study concerning the In: Rosai J (ed) Atlas of tumor pathology, fasicle 20. Armed
tumor aging and malignancy of nine cases. Cancer 65: Forces Institute of Pathology, Washington DC, pp 1-292
283-291 Stephens DH (1997) CT of pancreatic neoplasms, part II. The
Merkle EM, Bender GN, Brambs HJ (2000) Imaging findings unusual tumors. Curr Prob Diagn RadioI26:81-106
in pancreatic lymphoma: differential aspects. AJR Am J Teefey SA, Stephens DH, Sheedy PF Jr (1986) CT appearance
RoentgenoI174:671-675 of primary pancreatic lymphoma. Gastrointest Radiol 11:
Montemarano H, Lonergan GJ, Bulas DI, Selby DM (2000) 41-43
Pancreatoblastoma: imaging findings in 10 patients and Tuchek JM, De Jong SA, Pickleman J (1993) Diagnosis, sur-
review of the literature. Radiology 214:476-482 gical intervention, and prognosis of primary pancreatic
Mustert BR, Stafford-Johnson DB, Francis IR (1998) Appear- lymphoma. Am Surg 59:513-518
ance of acinar cell carcinoma of the pancreas on dual- Ueda K, Nagayama Y, Narita K, Kusano M, Mernyei M, Kamiya
phase CT. AJR Am J Roentgenol171 M (2000) Pancreatic involvement by non-Hodgkin's lym-
Nabae T, Yamaguchi K, Takahata S, Utsunomiya N, Matsunaga phoma. J Hepatobiliary Pancreat Surg 7:610-613
H, Sumiyoshi K, Chijiiwa K, Tanaka M (1998) Adenosqua- Ueda T, Ku Y, Kanamaru T, Hasegawa Y, Kuroda Y, Saitoh Y
mous carcinoma of the pancreas: report of two cases. Am (1996) Resected acinar cell carcinoma of the pancreas
J GastroenteroI93:1167-1170 with tumor thrombus extending into the main portal vein:
Oehler U, Jurs M, Kloppel G, Helpap B (1997) Osteoclast-like report of a case. Surg Today 26:357-360
giant cell tumour of the pancreas presenting as a pseudo- Van Beers B, Lalonde L, Soyer P, Grandin C, Trigaux JP, De
cyst-like lesion. Virchows Arch 431:215-218 Ronde T, Dive C, Pringot J (1993) Dynamic CT in pancreatic
Ordonez NG, Mackay B (2000) Acinar cell carcinoma of the lymphoma. J Comput Assist Tomogr 17:94-97
pancreas. Ultrastruct PathoI24:227-241 Viola G, Frontera D, Bellantone R, Doglietto GB, Crucitti F
Paal E, Thompson LD, Heffess CS (1998) A clinicopatho- (1997) Lymphangiomas of the pancreas: a case report.
logic and immunohistochemical study of ten pancreatic Pancreas 14:207-210
Unusual Pancreatic Neoplasms: Imaging 265

Webb TH, Lillemoe KD, Pitt HA, Jones RJ, Cameron JL (1989) Yamaguchi K, Tanaka M (2001) Atlas of cystic neoplasms of
Pancreatic lymphoma. Is surgery mandatory for diagnosis the pancreas. Karger, Basel
or treatment? Ann Surg 209:25-30 Yamaguchi K, Hirakata R, Kitamura K (1990) Papillary cystic
Yagi M, Shiraiwa K, Abiko M, Miura N, Sakuma H, Naito M neoplasm of the pancreas: radiological and pathological
(1994) A solid and cystic tumor of the pancreas in a 10- characteristics in 11 cases. Br J Surg 77: 1000-1003
year-old girl: report of a case and review of the literature. Yoon DY, Hines OJ, Bilchik AJ, Lewin K, Cortina G, Reber HA
Surg Today 24:826-828 (2001) Solid and papillary epithelial neoplasms of the pan-
Yamaguchi K, Enjoji M (1991) Adenosquamous carcinoma of creas: aggressive resection for cure. Am Surg 67:1195-1199
the pancreas: a clinicopathologic study. J Surg Oncol 47: Zinner MJ (1995) Solid and papillary neoplasms of the pan-
109-116 creas. Surg Clin North Am 75:17-24
Secondary Tumors
15 Secondary Tumors:
Clinical Manifestations and Pathology
T. OWElTY and A.B. WEST

CONTENTS in Willis's study of 500 autopsies, whereas Cubilla


identified this route of involvement in 6% of autopsy
15.1 Introduction 269 cases with secondary pancreatic tumors (WILLIS
15.2 Tumors Directly Invading
the Pancreas 269 1975; CUBILLA and FITZGERALD 1984). Some 17%
15.3 Lymphomas and Leukemias 269 of secondary pancreatic tumors were directly inva-
15.4 Tumors with Distant Metastases sive in Nakamura's Japanese series (NAKAMURA
to the Pancreas 271 et al. 2001). While the first two series identified
15.4.1 Solitary Metastases 271
carcinomas of the stomach and colon as the most
15.4.2 Metastatic Renal Cell Carcinoma 271
15.4.2.1 Clinical Features 271 common directly invasive tumors of the pancreas,
15.4.2.2 Pathology 272 no colonic cases were found in the Japanese series,
15.4.2.3 Differential Diagnosis 272 which identified extrahepatic bile duct carcinoma
15.5 Role of Fine Needle Aspiration and gastric carcinoma as the first and second most
Biopsy 273
common directly invasive tumors to the pancreas,
References 275
respectively.
Pathologically, it may be difficult to distinguish
primary pancreatic ductal carcinoma from invasive
gastrointestinal malignancies as both are adeno-
15.1 carcinomas that may display similar cytological!
Introduction architectural features and tumor markers (SESSA
et al. 1990). Finding in situ areas helps to estab-
Secondary tumors of the pancreas are uncommon. lish the site of tumor origin (SOLCIA et al. 1997).
The reported incidence varies widely within dif- Extensive lymphatic invasion is a peculiar feature
ferent series. Autopsy series describe pancreatic of involvement by gastric signet-ring cell carci-
involvement in 3%-15% of patients with general- noma (Fig. 15.1). Positivity for cytokeratin 7 is more
ized malignancies, secondary tumors being nearly likely seen in pancreatic carcinoma compared with
four times as common as primary pancreatic malig- colorectal adenocarcinomas, which are frequently
nancies (WILLIS 1975; CUBILLA and FITZGERALD negative (TOT 1999). In addition, morphologically,
1984). This most likely reflects the fact that pancre- colonic cancers tend to have more necrosis, mucin,
atic metastases from other organs are usually part of and tall columnar cells than primary pancreatic ade-
widespread preterminal metastatic disease. nocarcinoma (Fig. 15.2). Some patients with locally
advanced primary tumors invading the pancreas are
considered for surgical resection (HARRISON et al.
1997).
15.2
Tumors Directly Invading the Pancreas

Direct invasion of the pancreas by extension from 15.3


neighboring organs was the most common mode Lymphomas and Leukemias
of involvement by tumors of extrapancreatic origin
Secondary involvement of the pancreas by advanced
T. OWElTY, A.B. WEST
Hodgkin's disease, non-Hodgkin's lymphoma, mul-
Department of Pathology, New York University, 560 First tiple myeloma, and granulocytic sarcoma is not
Avenue (TH461), New York, NY 10016, USA uncommon (BANKS et al. 1975; BELL et al. 1982;
270 T. Oweity and A. B. West

a
Fig.lS.la, b. Gastric carcinoma directly invading the pancreas (H&E). a Signet-ring cell adenocarcinoma infiltrates the pancreas
(center) between residual islets (upper left) and pancreatic acini (lower right). b Lymphatic invasion in the pancreas is often a
prominent feature of gastric carcinoma

a
Fig.lS.2a, b. Cecal adenocarcinoma metastatic to the pancreas on fine needle aspiration biopsy. a Atypical intestinal-type
epithelium in a background of extensive necrosis (ultrafast Papanicolaou stain). b Tumor cells are columnar, with elongated
stratified nuclei (Diff-Quik)

EHRLICH et al. 1968; FISCHER et al. 1991; FRANTZ


1959; SATAKE et al. 1991; MWANDA and RAJAB 1999;
KAPLANSKI et al. 1994} and was noted in 1.9% of
all autopsies performed in the Memorial Hospital
series (CUBILLA and FITZGERALD 1984). They may
extend to the pancreas from the peripancreatic
lymph nodes. Leukemia, including chronic myelo-
cytic (CML), acute lymphocytic (ALL), chronic
lymphocytic (CLL), and acute myelocytic (AML)
forms, was present in 0.7% of the autopsies in
that series (CUBILLA and FITZGERALD 1984)
(Fig. 15.3}.

Fig. lS.3. Resected pancreatic mass in a patient with a long


history of CLL. Photograph shows eLL (bottom left) with large
cell transformation (top right) (H&E)
Secondary Tumors: Clinical Manifestations and Pathology 27l

15.4 al. 1994), thyroid and bone (SWENSON et al. 1980;


Tumors with Distant Metastases SUGIMURA et al. 1991), medulloblastoma (KRouwER
to the Pancreas et al. 1991), Merkel cell carcinoma (RUMANCIK et al.
1984), and soft-tissue sarcomas, including synovial
When direct pancreatic invasion from carcinoma sarcoma, mesenchymal chondrosarcoma, and leio-
of adjacent organs and systemic malignancies myosarcoma (YAMAMOTO et al. 2001; KOMATSU et
(lymphomas/leukemias) are excluded, the truly al. 1999; HOLMES and ALI 1997).
metastatic tumors to the pancreas usually arise
from the lung, breast, melanoma, colon, stomach,
ovary, or kidney (CUBILLA and FITZGERALD 1984; 15.4.2
WILLIS 1975). Moreover, when clinical series are Metastatic Renal Cell Carcinoma
reviewed, rather than autopsy series, metastatic
tumors comprise only 2%-3% of pancreatic tumors 15.4.2.1
(ROLAND and VAN HEERDEN 1989; KLUGO et al. 1977; Clinical Features
STANKARD and KARL 1992).
The scarcity of premortem evidence of meta- The most commonly reported solitary metastasis
static disease probably relates to the lack of symp- to the pancreas in recent series has been renal cell
toms, or their nonspecific nature. Initial symptoms carcinoma (ROBBINS et al. 1996; WHITTINGTON et al.
include abdominal pain, weight loss, fatigue, anemia, 1982; HARRISON et al. 1997). Renal cell carcinoma can
diarrhea, bleeding, jaundice, and the presence of a recur at any time after nephrectomy. In the majority
mass lesion. Metastatic foci were discovered in the of cases, pancreatic metastases occur early with a
pancreas in 14%-24% of asymptomatic patients tumor-free interval of often less than 1 year. How-
during radiographic follow-up for primary renal cell ever, late metastasis is not uncommon (KASSABIAN
carcinoma (THOMPSON and HEFFESS 2000). Intra- et al. 2000): Among 15 patients reported by Robbins
pancreatic metastases can be identified by various to have renal cell carcinoma developing solitary
radiologic imaging studies. They can be solitary, pancreatic metastases, the mean disease-free interval
multifocal, or diffuse (MERKLE et al. 1998). Multi- was 11 years (ROBBINS et al. 1996), with the longest
focal disease favors a metastatic malignancy over reported interval being 32.7 years (THOMPSON and
primary pancreatic disease, although multifocal HEFFESS 2000). The frequency of reports suggests a
pancreatic carcinoma occurs in up to 30% of cases recent increase in the detection of metastatic renal
(WITTENBERG et al. 1982; MURR et al. 1994). With cell carcinoma, perhaps reflecting an improvement
improvement in cross-sectional imaging, more cases in cross-sectional imaging that has allowed for the
of metastasis to the pancreas are being described. Of detection of indolent, discrete metastases in the pan-
1357 patients with solitary pancreatic masses, 2% creas which are characteristic of renal cell carcinoma.
were found at exploration to be secondary rather These tumors are usually depicted as one or more,
than primary pancreatic tumors (ROLAND and VAN well-defined, soft -tissue masses, with similar imaging
HEERDEN 1989). features to the primary renal cell carcinoma.
For isolated pancreatic metastases, an aggressive
surgical approach is recommended (Z'GRAGGEN et
15.4.1 al. 1998; ROBBINS et al. 1996). This is particularly
Solitary Metastases true for renal cell carcinoma metastases, for which
marked palliation or even potential cure is possible
Of patients presenting with a pancreatic mass who with resection (STANKARD and KARL 1992; MELO et
had a history of previous nonpancreatic malignancy, al. 1992; SAHIN et al. 1998; JINGU et al. 1998; HIROTA
42% had metastatic disease (WHITTINGTON et al. et al. 1996; HASHIMOTO et al. 1998; GHAVAMIAN et
1982). The most common sites of primary tumor al. 2000). In a Mayo clinic series of 23 patients with
that present as solitary pancreatic lesions are: breast metastatic renal cell carcinoma, approximately
(ODZAK et al. 2001), lung (JOHNSON et al. 1985), 4.6 years of palliation was achieved with surgery
and kidney. Other reported primary tumor sites (GHAVAMIAN et al. 2000). The grade of the metastatic
include stomach, ovary, melanoma (BRODISH and lesion correlated with the grade of the primary tumor
McFADDEN 1993; SMITH et al. 1994), duodenum, in most cases (96%). In the same series, survival was
esophagus, uterus and salivary gland (ROLAND and shown to be worse with higher tumor grade (mean
VAN HEERDEN 1989), prostate, liver, testis (ZUGEL et 41 months and 10 months in patients with grade
272 T. Oweity and A. B. West

2 and 3 lesions, respectively). Other features that uncommon. A sarcomatous pattern was seen in one
may be associated with a favorable prognosis after case. The cells showed variable amounts of glycogen.
resection of renal cell carcinoma metastases include No stainable mucin was found. Immunohistochemi-
a long interval between the primary tumor resec- cal staining showed about 50% of the cases to be
tion and the development of metastasis, evidence positive for cytokeratin (AE1/AE3) and epithelial
of a solitary or isolated lesion in the pancreas, and membrane antigen, while 9 of 21 cases were positive
lack of clinical symptoms (THOMPSON and HEFFEss for cytokeratin 7 (focally) and vimentin. Stains for
2000). In the 21 surgical pathology cases from the CA19-9 and CEA were negative. That for GP200AB-1,
file of the Endocrine Registry of the Armed Forces a marker of distal kidney tubule, was positive in 9 of
Institute of Pathology, the mean overall survival rate 21 cases, usually along the cell membrane and lumi-
from the date of nephrectomy was 19.8 years (range nal border. The surrounding pancreas in the majority
0.7-39.6 years) with a 6.2 years mean overall survival of cases showed chronic pancreatitis. Of 21 patients
from the date of detection of pancreatic metastasis who had undergone pancreatic resections for meta-
(range 1 month to 21.8 years) (THOMPSON and HEF- static renal cell carcinoma, 17 had been treated ini-
FESS 2000). This excellent prognosis for renal cell tially by nephrectomy 1 week to 32.7 years before the
carcinomas with solitary metastases, along with the development of pancreatic metastases. In 4 patients,
general lack of effective adjuvant therapy, empha- the pancreatic mass was the initial manifestation of
sizes the necessity for complete resection of primary the disease: a nephrectomy was performed 2 months
and metastatic lesions when possible (KASSABIAN et to 13.2 years after the diagnosis of metastatic clear-
al. 2000; TOLIA and WHITMORE 1975). cell carcinoma to the pancreas, which was not evident
by imaging during the interim (dormant).
15.4.2.2 There are several immunohistochemical features
Pathology that are helpful in differentiating renal cell carcinoma
from other neoplasms in the pancreas. Antibodies to
Of the 21 patients who underwent resection of CD10 stain up to 89% of renal cell carcinomas (CHU
metastatic renal cell carcinoma in the pancreas and ARBER 2000), while pancreatic carcinomas show
described by Thompson, 9 were women and 12 were less frequent and more apical positivity. Renal Cell
men, with an age varying between 47 and 67 years Carcinoma Marker, a monoclonal antibody against a
(mean 64.4 years) (THOMPSON and HEFFESS 2000). normal human proximal tubular brush border anti-
More patients presented with a solitary nodule in gen, has also been found to be highly specific for renal
the pancreas (12) than with multifocal disease (9). cell carcinomas (98% specificity), although not as
The tumors occurred in the head of the pancreas sensitive in metastases (67% sensitivity) (MCGREGOR
only (6), tail only (7), or randomly throughout the et al. 2001). Renal cell carcinoma also shows a positive
pancreas, and ranged in size from 1.5 to 12 cm oil-red-O reaction and strong vimentin and GP200
(mean 4.0 cm). Macroscopically, the neoplasms were immunoreactivity, accompanied by negative mucin,
usually well defined; the cut surfaces were brightly cytokeratin 20, CEA, and CA19.9 staining (LUTTGES
yellow/orange to white-gray and sharply circum- et al. 1998; MURAISHI et al. 1996; KAUFMANN et al.
scribed. A few were partially necrotic, containing 1996; ECHENIQUE and GRAHAM 1988; TOT 2002,
cysts filled with hemorrhagic fluid. Microscopically, SHEAHAN et al. 1990; Loy et al. 1993).
although the neoplastic deposits were usually well
circumscribed or even encapsulated, tumor cells 15.4.2.3
were occasionally identified invading the surround- Differential Diagnosis
ing pancreas. The predominant histologic pattern
was characterized by the presence of sheets, small The differential diagnosis of metastatic renal cell
nests, and cords of neoplastic cells separated by a carcinoma to the pancreas includes micro cystic
prominent vascular stroma. The neoplastic deposits adenoma, clear cell adenocarcinoma, islet cell tumor,
were composed of polygonal or elongated cells with and sugar tumor. Histologically, both metastatic
clear cytoplasm, distinct cytoplasmic membranes, renal cell carcinoma and micro cystic adenoma can
and small, compact, eccentric nuclei showing mostly have small, gland-like structures and a low nuclear:
minimal pleomorphism. Eosinophilic cytoplasm cytoplasmic ratio with clear cytoplasm surrounding
was present focally in three cases. No papillary nonatypical hyperchromatic nuclei. A rich vascular
features were detected. Hemorrhage was seen in all pattern can be seen in both, but renal cell carcinoma
cases, while necrosis and lymphoid infiltrates were tends to form sheets of cells separated by fibrovas-
Secondary Tumors: Clinical Manifestations and Pathology 273

cular septa with a thin sinusoidal vascular pattern. 15.5


Microcystic adenomas generally have fibrotic bands Role of Fine Needle Aspiration Biopsy
separating cells that are arranged around a larger
cyst-like lumen. Cytoplasmic glycogen is found in As the preoperative distinction between primary and
both lesions. The cells of clear cell adenocarcinoma secondary pancreatic neoplasms is almost impossible,
of the pancreas have abundant clear cytoplasm sur- particularly in solitary masses, diagnostic biopsies are
rounding atypical nuclei, similar to those of renal cell commonly performed, especially in those patients with
carcinoma although the latter's vascular sinusoidal a history of malignancy. Although a surgical biopsy of
pattern is lacking (LUTTGES et al. 1998). Pancreatic the pancreatic tumor may be obtained in cases where
ductal tumors are usually negative for vimentin and relief of obstructive symptoms mandates surgery, in
the renal cell carcinoma marker, but positive for cyto- most other cases, fine needle aspiration (FNA) of the
keratin. Some are positive for CEA or CA19.9. Clear- pancreas (percutaneous or endoscopic) is the pro-
cell islet-cell tumors are positive for chromogranin cedure of choice (BENNING et al. 1992; FRITSCHER-
and islet-cell hormones, while renal cell carcinoma is RAVENS et al. 2001; DI STASI et al. 1998; LJUBICIC et
negative for chromogranin (CUBILLA and FITZGER- al. 1992; WILLIAMS et al. 1999; ERICKSON et al. 1997;
ALD 1984; SOLCIA et al. 1997). Sugar tumors of the CARSON et al. 1995). This rapid, inexpensive, accurate,
pancreas are extremely unusual: Although the sinu- and safe procedure with minimal complications can
soidal pattern is present, the tumor cells are negative have a decisive influence on the selection of appropri-
for keratin and positive for HMB-45 (ZAMBONI et al. ate therapeutic strategies. Selected patients with iso-
1996) (Fig. 15.4). lated pancreatic metastases from tumors of relatively
better prognosis can undergo resection, achieving a
5-year survival of as much as 35% (Z'GRAGGEN et
al. 1998), which is much better than that of primary
pancreatic carcinoma (HARRISON et al. 1997), while
other patients who might benefit from alternative
treatments such as chemotherapy or radiotherapy will
avoid superfluous surgery. When cytology reveals cells
that are unusual for the pancreas (e.g., lymphoid cells,
small cells, or squamous cells), a systemic or a second
malignancy, with pancreatic metastases, is still a dis-
tinct possibility (BENNING et al. 1992). The clinical his-
tory and a variety of immunohistochemical stains may
aid in establishing and refining this diagnosis. This
may include cytokeratins 7 and 20 in cases of adeno-
carcinoma, CD20 and CD3 in lymphoma (Fig. 15.5),
HMB-45 and S100 protein in melanoma (Fig. 15.6),
estrogen and progesterone receptors and gross cystic
disease fluid protein (GCDFP-15) in mammary carci-
noma, TTF-1 (thyroid transcription factor) in pulmo-
nary carcinoma, CD 10 and RCC (renal cell carcinoma
marker) in renal cell carcinoma (Table 15.1).
Table 15.1 lists the most characteristic immuno-
histochemical stains for each tumor as seen in the
majority of cases. However, the specificity and sensi-
tivity of these antibodies vary depending on the type
and differentiation of tumors and the type of com-
mercial antibodies used. They should be interpreted
Fig. 15.4a, h. Patient with a history of renal cell carcinoma, in the context of the cytomorphological findings and
presenting with a pancreatic mass (H&E). a Low power the clinical/radiological setting (Tot 1999,2000,2002;
view showing the fairly well circumscribed clear-cell tumor,
Jang et al. 2001; Lagendijk et al. 1999; Castro et al.
with pancreatic islets at the periphery. b Higher power view.
Cords of renal cell carcinoma cells displace the remaining 2001; Sheahan et al. 1990; Loy et al. 1993; Clarkson
pancreatic islets. The cells have clear cytoplasm and irregular et al. 2001; Chu and Arber 2000; Moritani et al. 2002;
nuclei (H&E) Kristiansen et al. 2002).
274 T. Oweity and A. B. West

a a

Fig. 15.5a, b. Non-Hodgkin's lymphoma involving the pan- Fig. 15.6a, b. Malignant melanoma metastatic to the pan-
creas on fine needle aspiration biopsy. a The tumor cells are creas on fine needle aspiration biopsy. a The cells are char-
dissociated, with irregular chromatin and prominent nucleoli. acteristically discohesive, binucleate or multinucleate, with
Lymphoglandular bodies (small, gray, circular structures char- intranuclear cytoplasmic inclusions (ultrafast Papanicolaou
acteristic of lymphoid cells) are abundant in the background stained smear). b The tumor cells are immunoreactive for
(Diff-Quik stained smear). b The cells are positive (brown) HMB-45 (brown), confirming the diagnosis of melanoma
for the B-cell marker CD20 (immunoperoxidase with diami- (immunoperoxidase with diaminobenzidine on cell block
nobenzidine on a cell block section) section)

Table 15.1. Immunohistochemistry in differentiating primary pancreatic ductal carcinoma from metastases (+ positive in
majority of cases, - negative in majority of cases, ± positive or negative)
Tumor type CK-20 CK-7 CA19.9 CEA TTF-l GCDFP-15 ER SlOO/HMB-45 CDIO Vimentin

Pancreatic ductal ± + + + ±
adenocarcinoma
Colonic adeno- + + + ±
carcinoma
Gastric carcinoma ± ± + + ±
Pulmonary carcinoma - + ± ± + ±
Breast carcinoma + + +
Renal cell carcinoma +a +
Melanoma + ± +

aDiffuse positivity
Secondary Tumors: Clinical Manifestations and Pathology 275

References Hashimoto M, Watanabe G, Matsuda M, Dohi T, Tsurumaru


M (1998) Management of the pancreatic metastasis from
Banks PM, Arseneau JC, Gralnick HR, Canellos GP, DeVita renal cell carcinoma: report of four resected cases. Hepa-
VT Jr, Berard CW (1975) American Burkitt's Lymphoma. to gastroenterology 45:1150-1154
A clinicopathologic study of 30 cases. II. Pathologic corre- Hirota T, Tomida T, Iwasa M, Takahashi K, Kaneda M, Tamaki
lations. Am J Med 58:322-329 H (1996) Solitary pancreatic metastasis occurring eight
Bell HG, David R, Shamsuddin AM (1982) Extrahepatic biliary years after nephrectomy for renal cell carcinoma: a case
obstruction and liver failure secondary to myeloma of the report and surgical review. Int J PancreatoI19:145-153
pancreas. Hum Pathol 13:940-942 Holmes GF, Ali SZ (1997) Fine needle aspiration of leio-
Benning TL, Silverman JF, Borns I, Geisinger KR (1992) Fine myosarcoma metastatic to the pancreas. Diagn Cytopathol
needle aspiration of metastatic and hematologic malig- 16:189-190
nancies clinically mimicking pancreatic carcinoma. Acta Jang KY, Kang MJ, Lee DG, Chung MJ (2001) Utility of thyroid
CytoI36:471-476 transcription factor-1 and cytokeratin 7 and 20 immunos-
Brodish RJ, McFadden DW (1993) The pancreas as the solitary taining in the identification of origin of malignant effu-
site of metastasis from melanoma. Pancreas 8:276-278 sions. Anal Quant Cytol Histol 23:400-404
Carson HJ, Green LK, Castelli AJ, Reyes Cv, Prinz RA, Gattuso Jingu K, Watanabe K, Yamamoto H, Fujita Y, Honda I, Wata-
P (1995) Utilization of fine-needle aspiration biopsy in nabe S, Nagata M, Sugimoto K, Watanabe Y (1998) Surgi-
the diagnosis of metastatic tumors to the pancreas. Diagn cal treatment of a solitary pancreatic metastasis from renal
CytopathoI12:8-13 cell carcinoma: report of a case. Surg Today 28:91-94
Castro CY, Moran CA, Fleider DG, Suster S (2001) Primary Johnson DH, Hainsworth JD, Greco FA (1985) Extrahepatic
signet ring cell adenocarcinomas of the lung: a clinico- biliary obstruction caused by small cell lung cancer. Ann
pathological study of 15 cases. Histopathology 39:397-401 Intern Med 102:487-490
Chu P, Arber DA (2000) Paraffin-section detection of CD 10 in Kaplanski G, Kloeppel MC, Deharo C, Durand JM, Andrac
505 nonhematopoietic neoplasms. Frequent expression in L, Sayage J (1994) Secondary pancreatic involvement of
renal cell carcinoma and endometrial stromal sarcoma. Am mycosis fungoides detected by a clinically palpable mass.
J Clin PathoI113:374-382 Dermatology 189:406-408
Clarkson KS, Sturdgess IC, Molyneux AJ (2001) The use- Kassabian A, Stein J, Jabbour N, Parsa K, Skinner D, Parekh D,
fulness of tyrosinase immunohistochemical assessment Cosenza C, Selby R (2000) Renal cell carcinoma metastatic
of melanocytic lesions: a comparison of the novel T311 to the pancreas: a single-institution series and review of
antibody (anti-tyrosinase) with S-100, HMB45, and A103 the literature. Urology 56:211-215
(anti-melan-A). J Clin Pathol 54:196-200 Kaufmann 0, Deidesheimer T, Muehlenberg M, Deicke P,
Cubilla A, Fitzgerald PJ (1984) Tumors of the exocrine pan- Dietel M (1996) Immunohistochemical differentiation
creas. Atlas of tumor pathology, 2nd ser, vol 19. Armed of metastatic breast carcinomas from metastatic adeno-
Forces Institute of Pathology, Washington DC carcinomas of other common primary sites. Histopathol-
Di Stasi M, Lencioni R, Solmi L, Magnolfi F, Caturelli E, DeSio I, ogy 29:233-240
Salmi A, Buscarini L (1998) Ultrasound-guided fine-needle Klugo RC, Detmers M, Stiles RE, Talley RW, Cerny JC (1977)
biopsy of pancreatic masses: results of a multicenter study. Aggressive versus conservative management of stage IV
Am J GastroenteroI93:1329-1333 renal cell carcinoma. J Urol 118:244-246
Echenique JE, Graham SD Jr (1988) Significance oflipid-asso- Komatsu T, Taira S, Matsui 0, Takashima T, Note M, Fugita H
ciated sialic acid and CA19-9 as tumor markers for renal (1999) A case of ruptured mesenchymal chondrosarcoma
cell carcinoma. Urology 32:397-400 of the pancreas. Radiat Med 17:239-241
Ehrlich A, Stalder G, Geller W, Sherlock P (1968) Gastrointes- Kristiansen G, Schluns K, Yong wei Y, Dietel M, Petersen I
tinal manifestations of malignant lymphoma. Gastroen- (2002) CD10 expression in non-small lung cancer. Anal
terology 54:1115-1121 Cell Pathol 24:41-46
Erickson RA, Sayage-Rabie L, Avots-Avatins A (1997) Clinical Krouwer HG, Vollmerhausen J, White J, Prados MD (1991)
utility of endoscopic ultrasound-guided fine-needle aspi- Desmoplastic medulloblastoma metastatic to the pancreas:
ration. Acta CytoI41:1647-1653 case report. Neurosurgery 29:612-616
Fischer A, Suhrland MJ, Vogl SE (1991) Myeloma of the head of <refLangendijk JH, Mullink H, Van Diest PJ, Meijer GA,
the pancreas. A case report. Cancer 67:681-683 Meijer CJLM (1999) Immunohistochemical differentiation
Frantz VK (1959) Tumors of the pancreas. Atlas of tumor between primary adenocarcinoma of the ovary and ovar-
pathology, 1st ser, fasc 27 and 28. Armed Forces Institute ian metastases of colonic and breast origin. Comparison
of Pathology, Washington DC between a statistical and an intuitive approach. J Clin
Fritscher-Ravens A, Sriram PV, Krause C, Atay Z, Jaeckle S, Pathol 52:283-290
Thonke F, Brand B, Bohnacker S, Soehendra N (2001) Ljubicic N, Bilic A, Lang N, Bakula B (1992) Ultrasonically-
Detection of pancreatic metastases by EOS-guided fine- guided percutaneous fine-needle aspiration biopsy of the
needle aspiration. Gastrointest Endosc 53:65-70 hepatic and pancreatic focal lesions; accuracy of cytology
Ghavamian R, Klein KA, Stephens DH, Welch TJ, LeRoy AJ, in the diagnosis of malignancy. J R Soc Med 85:139-141
Ritchardson RL, Burch PA, Zinake H (2000) Renal cell Loy TS, Sharp SC, Andershock CJ, Craig SB (1993) Distribution
carcinoma metastatic to the pancreas: clinical and radio- of CA19.9 in adenocarcinomas and transitional cell carci-
logical features. Mayo Clin Proc 75:581-585 nomas. An immunohistochemical study of 527 cases. Am J
Harrison LE, Merchant N, Cohen AM, Brennan MF (1997) Clin Pathol 99:726-728
Pancreaticoduodenectomy for nonperiampullary primary Luttges J, Vogel I, Monke M, Henne-Bruns D, Kremer B, Klop-
tumors. Am J Surg 174:393-395 pel G (1998) Clear cell carcinoma of the pancreas: an
276 T. Oweity and A. B. West

adenocarcinoma with ductal phenotype. Histopathology Solcia E, Capella C, Kloppel G (1997) Non-epithelial and sec-
32:444-448 ondary tumors. Tumors of the pancreas, chap 6. Atlas of
McGregor DK, Khurana KK, Cao C, Tsao CC, Ayala G, Krishnan tumor pathology, 3rd ser, vol 20. Armed Forces Institute of
B, Ro JY, Lechago J, Truong LD (2001) Diagnosing primary Pathology, Washington DC, pp 211-214
and metastatic renal cell carcinoma. The use of monoclonal Stankard CE, Karl RC (1992) The treatment of isolated pan-
antibody 'Renal Cell Carcinoma Marker'. Am J Surg Pthol creatic metastases from renal cell carcinoma: a surgical
25: 1485-1492 review. Am J Gastroenterol 87:1658-1660
Melo CR, Melo IS, Monteiro AZ, de Mello ES (1992) Pancreatic Sugimura H, Tamura S, Kodama T, Kakitsubata Y, Asada K,
metastasis from renal cell carcinoma. Arg Gastroenterol29: Watanabe K (1991) Metastatic pancreas cancer from the
110-112 thyroid: Clinical imaging mimicking non-functioning islet
Merkle EM, Boaz T, Kolokythas 0, Haaga JR, Lewin JS, Brambs cell tumor. Radiat Med 9:167-169
HJ (1998) Metastases to the pancreas, pictorial review. Br J Swenson T, Osnes M, Serck-Hansson A (1980) Endoscopic
RadioI71:1208-1214 retrograde cholangiopancreatography in primary and sec-
Moritani S, Kushima R, Sugihara H, Bamba M, Kobayashi ondary tumors of the pancreas. Br J Radiol 53:760-764
TK, Hattori T (2002) Availability of CD10 immunohis- Thompson LD, Heffess CS (2000) Renal cell carcinoma to the
tochemistry as a marker of breast myoepithelial cells on pancreas in surgical pathology material. A clinicopatho-
paraffin sections. Mod PathoI4:397-405 logic study of 21 cases with a review of the literature.
Muraishi 0, Kinebuchi Y, Ogawa A (1996) CA19-9 negative pan- Cancer 89:1076-1088
creatic cancer mimicking renal cancer. Br J Urol 77:751-758 Tolia BM, Whitmore WF Jr (1975) Solitary metastasis from
Murr MM, Sarr MG, Oishi AJ, van Heerden JA (1994) Pan- renal cell carcinoma. J Urol 114:836-838
creatic Cancer. CA Cancer J Clin 44:304-18 Tot T (1999) Adenocarcinomas metastatic to the liver: the
Mwanda WO, Rajab JA (1999) Granulocytic sarcoma: report of value of cytokeratins 20 and 7 in the search for unknown
three cases. East Afr Med J 76:594-596 primary tumors. Cancer 85:171-177
Nakamura, Etsuko, Michio Shimizu, Tomoo Itoh and Toshiaki Tot T (2000) The role of cytokeratins 20 and 7 and estrogen
Manabe (2001) Secondary tumours of the pancreas: receptor analysis in separation of metastatic lobular
clinicopathological study of 103 autopsy cases of Japanese carcinoma of the breast and metastatic signet ring cell
patients. Pathol Int 51 :686-690 carcinoma of the gastrointestinal tract. APMIS 108:
Odzak A, Geliberti F, Farace G, Benitez S, Bistoletti R, Kozima 467-472
S, Frider B (2001) Pancreatic tumor: an unusual presen- Tot T (2002) Cytokeratins 20 and 7 as biomarkers: useful-
tation of an occult breast carcinoma. Acta Gastroenterol ness in discriminating primary from metastatic adeno-
Latinoam 31:395-398 carcinoma. Eur J Cancer 38:758-763
Robbins EG, Franceschi D, Barkin JS (1996) Solitary metastatic Whittington R, Moylan DJ, Dobelbower RR, Kramer S (1982)
tumors to the pancreas: a case report and review of the Pancreatic tumors in patients with previous malignancy.
literature. Am J GastroenteroI91:2414-2417 Clin Radiol 33:29-79
Roland CF, Van Heerden JA (1989) Nonpancreatic primary Williams DB, Sahai AV, Aabakken L, Penman ID, Van Velse A,
tumors with metastases to the pancreas. Surg Gynecol Webb J, Wilson M, Hoffman BJ, Hawes RH. (1999) Endo-
Obstet 168:345-347 scopic ultrasound-guided fine-needle aspiration biopsy: a
Rumancik WM, Megibow AJ, Bosniak MA, Hilton S (1984) large single center experience. Gut 44:720-726
Metastatic disease to the pancreas: evaluation by computed Willis RA (1975) The spread of tumours in the human body.
tomography. J Comp Assist Tomogr 8:829-834 Butterworth, New York, pp 216-217
Sahin M, Foulis AA, Poon FW, Imrie CW (1998) Late focal Wittenberg J, Simeone JF, Ferrucci JT Jr, Mueller PR, vanSon-
pancreatic metastasis of renal cell carcinoma. Dig Surg nenberg E, Neff CC (1982) Non-focal enlargement in pan-
15:72-74 creatic carcinoma. Radiology 144:l31-l35
Satake K, Arimoto Y, Fujimoto Y, Nishiwaki H, Nishino H, Yamamoto H, Watanabe K, Nagata M, Honda I, Watanabe S,
Umeyama K, Inoue T, Mitsuhashi T (1991) Malignant T- Soda H, Tatezaki S (2001) Surgical treatment for pancreatic
cell lymphoma of the pancreas. Pancreas 6:120-124 metastasis from soft-tissue sarcoma: report of two cases.
Sessa F, Bonato M, Frigerio B, Capella C, Solcia E, Prat M, Am J Clin Oncol 24:198-200
Bara J, Sanoff 1M (1990) Ductal cancer of the pancreas Zamboni G, Pea M, Martignoni G, Zancanaro C, Facciali G,
frequently expresses markers of gastrointestinal epithelial Pederzoli P (1996) Clear cell "sugar" tumor of the pancreas.
cells. Gastroenterology 98:165-165 A novel member of the family of lesions characterized by
Sheahan K, O'brien MJ, Burke B, Dervan PA, O'Keane JC, Got- the presence of perivascular epithelioid cells. Am J Surg
tlieb LS, Zamcheck N (1990) Differential reactivities of PathoI20:722-730
Carcinoembryonic antigen (CEA) and CEA-related mono- Z'graggen K, Fernandez-del Castillo C, Rattner DW, Sigala H,
clonal and polyclonal antibodies in common epithelial Washaw AL (1998) Metastases to the pancreas and their
malignancies. Am J Clin PathoI94:157-164 surgical extirpation. Arch Surg l33:4l3-417
Smith CD, Behrns KE, Van Heerden JA, Sarr MG (1994) Rad- Zugel N, Leipprand E, Weckermann D, Witte J (1994) Solitary
ical pancreatoduodenectomy for misdiagnosed pancreatic metastasis to the head of the pancreas in hypernephroid
mass. Br J Surg 81:585-589 carcinoma. Fortschr Med 112:388-390
16 Secondary Pancreatic Tumors: Imaging
A. J. MEGIBOW

CONTENTS enlarge the pancreas - in other words they mimic


primary pancreatic neoplasms of a variety of mor-
16.1 Introduction 277 phologic appearances. Because these masses signal
16.2 Clinical Considerations 277
16.3 Imaging Features - Hematogenous Metastases 278 advanced disease, their presence is usually docu-
16.3.1 General Features 278 mented on a single imaging examination, almost
16.3.2 Renal Cell Carcinoma 278 exclusively computed tomography (CT). Correlative
16.3.3 Other Hematogenous Metastases 279 imaging is not available in most cases; therefore, as
16.3.4 Complications and Treatment 283
distinct from other sections of this volume, the orga-
16.4 Secondary Pancreatic Involvement
by Tumor in Adjacent Lymph Nodes 284 nization of the chapter will be based on pathology
16.5 Secondary Pancreatic Involvement instead of the imaging modality.
by Tumors in Adjacent Organs 285 For the purposes of this discussion, we shall divide
References 287 the description of the imaging findings based on the
pathophysiologic mechanism by which the abnor-
mality may affect the pancreas. Three pathways by
which metastases can arrive at the pancreas include:
16.1 (1) hematogenous spread, (2) secondary involvement
Introduction of contiguous lymphoid tissue, (3) direct extension
from adjacent viscera.
The estimated prevalence of metastases to the pan-
creas at autopsy is between 3% and 10% (CUBILLA
and FITZGERALD 1980; SCATARIGE et al. 2001;
WILLIS 1973). Because pancreatic metastases occur 16.2
in patients with advanced disease, the presence of Clinical Considerations
the lesions themselves rarely cause 'pancreas related'
symptoms; rather, they are discovered during surveil- Autopsy studies reveal that metastases to the pan-
lance imaging in oncology patients. creas were found in 3% of 5000 patients (WILLIS
Beyond the detection of a pancreatic mass, the 1973) and 10.6% of 2587 patients (CUBILLA and
imaging features are nonspecific. The diagnosis is FITZGERALD 1980). From these data, one might
suspected when a pancreatic mass is detected in a conclude that pancreatic metastases should be seen
patient with a known primary tumor and confirmed more frequently in an individual clinical practice.
by biopsy. On imaging studies, pancreatic metas- Most imaging is performed at the time of diagnosis,
tases can appear as localized (50%-73% of cases), and pancreatic metastases often appear as a manifes-
multifocal masses (15%-44% of cases) or result in tation of advanced disease. Conversely, the presence
diffuse enlargement of the gland (5%-10% of cases) of pancreatic metastases may be underestimated by
(MERKLE et al.1998; SCATARIGE et al. 2001). Enhance- imaging studies. In 18/34 (53.8%) cases of autopsy-
ment characteristics vary with the primary tumor. confirmed metastatic tumors of the pancreas includ-
The lesions may appear as solid masses, hyperdense ing secondary malignant lymphoma, the pancreas
masses, cystic masses, or even diffusely infiltrate and appeared abnormal on CT (8 diffuse enlargement, 9
localized mass, 1 multiple low attenuation nodules)
. In the remaining 16 patients, histologic investiga-
A. J. MEGIBOW, MD, MPH, FACR
tion revealed that metastatic carcinoma resulted
Professor of Radiology, NYU School of Medicine, New York, in diffuse infiltration of the pancreatic lobules, not
NY, USA macroscopically visualized by CT (MURANAKA et al.
278 A. J. Megibow

1989). Although this study (from 1989) utilized less 16.3


sophisticated technology than is presently available, Imaging Features - Hematogenous
it is evident that a significant number of cases may Metastases
go unrecognized.
KLEIN et al. (1998) reviewed a 10-year experience 16.3.1
in 66 patients seen at the Mayo Clinic between 1984 General Features
and 1994. Of these, 13 patients already had pancre-
atic metastases at the time of initial CT, whereas the Virtually any primary neoplasm may metastasize to
remainder developed them in the follow-up period the pancreas. However, from multiple reports, certain
(range 2-295 months, mean 57.2 months). In more cell types have a marked predilection to deposit in this
than one-third of patients, the metastases appeared location. RCC and lung carcinoma are by far the most
after more than 60 months. The variability could be commonly reported sources (FERROZZI et al. 1997;
correlated with the cell type of the primary neoplasm. KLEIN et al. 1998; RUMANCIK et al. 1984; SCATARIGE
Patients with pancreatic metastases from renal cell et al. 2001). Other frequently reported primary lesions
carcinoma (RCC) demonstrated the longest interval include melanoma (BIANCA et al.1992), plasmacytoma
between the detection of tumor and the development (MITCHELL and HILL 1985; RICE et al. 1981; TWOMEY
of pancreatic metastases. In Klein's series, 85% of and KATZ 1983; WILSON et al. 1989), carcinoid tumor
patients with RCC who developed metastases mani- (AKERSTROM 1996; VILLANUEVA et al.1994), and breast
fested the masses more than 60 months after diag- cancer (MouNTNEY et al.1997). This list is by no means
nosis, followed by melanoma, breast cancer, and lung complete; disparate sources such as hepatocellular car-
cancers. In another series of 10 patients, the mean cinoma (TEXLER et al. 1998) and tonsillar carcinoma
time from nephrectomy for resection of the primary (McLATCHIE and IMRIE 1981) have been reported.
tumor to re-operation for periampullary recurrence As stated in Section 16.2, three broad morphologi-
in 10 patients with RCC was 9.8 years (median 8.5 cal patterns have been described: (1) solitary mass,
years, range 0-28 years) (SOHN et al. 2001). (2) multiple nodules; (3) diffuse micronodular pat-
The pancreas is rarely the only site of metastatic tern with gland enlargement (KLEIN et al. 1998; SCA-
dissemination. In Klein's series, 45.5% of patients TARIGE et al. 2001). The metastases tend to mimic the
had demonstrable extrapancreatic abdominal sites of enhancement characteristics of the primary tumor
metastases. The most frequent sites were liver, lymph (FERROZZI et al.1997).
nodes, and adrenal glands (KLEIN et al. 1998). In
another series, 19120 patients with pancreatic metas-
tases had lesions in other anatomic sites (FERROZZI et 16.3.2
al. 1997). When the pancreas is the sole site of metasta- Renal Cell Carcinoma
sis, differentiation from the primary pancreatic tumor
is difficult without tissue confirmation by biopsy. The In our initial publication (RUMANCIK et al. 1984), and
long-term survival of these patients is considerably subsequently borne out in many series, RCC seems to
improved over those who might have primary pan- be the most common primary tumor to metastasize
creatic cancer, and therefore, in appropriate situations to the pancreas (GHAVAMIAN et al. 2000; KLEIN et
(when imaging fails to define other foci of disease), al. 1998; RIVOIRE and VOIGLIO 1996; RUMANCIK et
aggressive surgical therapy may provide survival ben- al. 1984; WEERDENBURG and JURGENS 1984). A large
efit (HASHIMOTO et al.1998; Z'GRAGGEN et al.1998). percentage of patients will harbor metastases in
Clinical symptoms directly and solely related other sites, most frequently the lungs. In an analysis
to the metastatic disease are unusual when the of 392 autopsy cases with metastatic RCC, 27% of
tumor reaches the pancreas by hematogenous dis- patients with metastatic renal cancer did not have
semination. Mild liver function test abnormalities lung metastases, whereas 73% of cases without lung
or hyperamylasemia may be observed. New onset metastases had no evidence of tumor elsewhere. In
diabetes is rare (GHAVAMIAN et al. 2000). Acute pan- 84% of patients with lung metastases, there were
creatitis may occur as a result of ductal obstruction other metastases in the body. These observations
(McLATCHIE and IMRIE 1981). When the pancreas are concordant with the fact that the pancreas may
is secondarily involved by adjacent invasive lymph- be secondarily seeded from other metastatic sites, so-
adenopathy, there is a higher frequency of symptoms called 'metastatic cascades' (WEISS et al. 1988).
such as biliary obstruction and bowel obstruction Most descriptions of the imaging features of these
(CHARNSANGAVEJ and WHITLEY 1993). lesions are derived from CT experience. RCC metas-
Secondary Pancreatic Tumors: Imaging 279

tases to the pancreas are most frequently solitary on Tl-weighted fat-suppressed SE images and high
(Fig. 16.1), although multiple masses are not unusual in signal intensity on T2-weighted images. They
(Figs. 16.2, 16.3). Utilizing thin-section, bolus con- demonstrate rapid enhancement on the immediate
trast-enhanced CT, the lesions rapidly enhance postgadolinium Tl-weighted gradient recalled echo
during the arterial (25 s following injection) and (GRE) images. Larger tumors peripherally enhance
early portal phases to levels between 50 and 100 HU due to central necrosis (KELEKIS et al. 1996; MERKLE
over the normal pancreatic parenchymal enhance- et al.1998).
ment. On delayed (greater than 60 s) phase imaging, Although studies have shown ultrasound (US)
this difference decreases to between 5 and 45 HU (NG (WERNECKE et al. 1986) to be capable of detecting
et al. 1999). The pattern of enhancement may show these lesions, most agree that transabdominal US is
preferential peripheral enhancement or be uniform inferior compared with CT or MRI (BISET et al. 1991).
(GHAvAMIAN et al. 2000). In one series, the metastases appear as solitary or
On magnetic resonance imaging (MRI), the lesions multiple hypo echoic nodules (Fig. 16.3). US is most
are hypointense compared with normal pancreas useful in detecting complications such as biliary dila-
tation and pancreatitis (SATO et al. 2001). A new class
of intravenously administered ultrasound contrast
agents, stabilized microbubbles of sulfur hexafluo-
rine gas (Sonovue, Bracco Laboratories), have been
introduced which increase the echogenicity in hyper-
vascular masses (LEEN et al. 2002) (Fig. 16.4). While
frequently utilized in Europe, there is little US experi-
ence with these agents in abdominal imaging.

16.3.3
Other Hematogenous Metastases

As stated previously, any primary tumor could metas-


tasize to the pancreas. In our practice, melanoma and
lung cancer are the two most frequent sources. Mel-
anoma surpasses RCC in frequency in our practice
Fig. 16.1. Renal cell carcinoma (RCC) metastatic to pancreas. A because our institution is a regional referral center
hyperdense (hypervascular) mass is present in the pancreatic
body. Note the upstream dilatation of the pancreatic duct. for patients with this disease.
Surgical clips and the position of the spleen are evidence of Lung cancer (bronchogenic carcinoma) is the most
prior left nephrectomy common malignant tumor in the USA, and therefore

Fig.16.2a. Multiple RCC metastases to the pancreas. There are multiple hyperdense nodules throughout the entire pancreatic paren-
chyma. Notice the primary tumor in the left kidney. b RCC metastatic to pancreas. Celiac axis angiogram of patient in a. The patient
presented with a nodule in her thyroid approximately 10 years prior to the CT examinations. The nodule was metastatic RCC
280 A. J. Megibow

a b

Fig. 16.3a,b. RCC metastatic to the pancreas. a Transabdominal ultrasound reveals a hypoechoic mass delimited by markers (+)
in the pancreatic body. b Solitary hyperdense mass is visualized during the arterial phase of helical/spiral CT scan

a b

Fig. 16.4a-c. RCC metastatic to pancreas. a Transabdominal


ultrasound reveals a sharply defined mass in the pancreatic
body (arrow) . SMA, superior mesenteric artery. b Ultrasound
following intravenous injection of Sonovue (stabilized micro-
bubbles of sulfur hexafiuorine gas) documents significant
increase in echogenicity within the lesion (arrow) and sug-
gests multiple smaller nodules. The increased echogenicity
correlated with a hypervascular lesion. c CT scan in arterial
phase shows typical hypervascular mass with several smaller
c nodules

one is not surprised that pancreatic metastases are (n=9) over non-smaIl-cell carcinoma (n=6). In other
encountered in clinical practice. In Klein's series, bron- series, bronchogenic carcinoma is the most common
chogenic carcinoma (small-cell and non-smaIl-cell source of metastatic disease (FERROZZI et al. 1997;
types) was the second most frequent source of metas- ROLAND and VAN HEERDEN 1989). In our experience,
tasis (15 of 66 patients) (KLEIN et al. 1998). There was lung cancer metastases exhibit anyone of the three
a slight predilection of metastases in small-cell cancer broad morphologic patterns enumerated previously
Secondary Pancreatic Tumors: Imaging 281

(Fig. 16.5). Most of our cases have demonstrated low The high signal on Tl-weighted images and low signal
attenuation masses, but the number is too few to make on T2-weighted images result from the paramagnetic
a generalized statement (Fig. 16.6). effects of melanin (DEJORDY et al. 1992).
Melanoma metastases present with solitary or Pancreatic tumors with serotonin immunoreactiv-
multifocal lesions (Figs. 16.7, 16.8). In most cases, ity may be classified as carcinoids and may result in an
extensive metastatic disease will be present in many unusual cause of the carcinoid syndrome (AKERSTROM
sites not commonly seen in primary adenocarcinoma, 1996). In our practice, we have seen two patients with
although, unlike RCC, the pancreas has been reported carcinoid tumors in the pancreas, neither of whom
as the sole site of metastasis (BIANCA et al. 1992). Con- demonstrated carcinoid syndrome. In both, the pan-
trast enhancement will vary in relation to the size or the creas was involved as part of multiorgan dissemina-
treatment history of the individual patient. Melanoma tion. In one, the lesion was barely visible on a CT scan
metastases demonstrate low signal on T2-weighted performed with 'pancreatic technique', but MR in this
images and rapid enhancement following intravenous patient showed the lesion to much greater advantage
gadolinium administration (SOLOMONS et al. 2000). (Fig. 16.9). We have seen pancreatic metastases in

a b

Fig.16.5a,b. Non-small-celliung cancer metastatic to pancreas. a There is a unifocallow attenuation mass in the pancreatic head.
The lesion is indistinguishable from other forms of cystic pancreatic tumors. b A fusiform low attenuation mass is present in
the pancreatic tail. The similarity in appearance to primary pancreatic adenocarcinoma is evident. Multiple hepatic metastases
are present as well

a b

Fig. 16.6a. Small-cell carcinoma metastatic to the pancreas. Multiple low attenuation masses are dispersed throughout the
pancreatic body. b Non-small-cell carcinoma metastatic to pancreas. Multiple nodules have coalesced to replace and enlarge
the entire gland. Notice the encasement of the splenic vein by the tumor. Vascular encasement is uncommon but reported in
patients with pancreatic metastases
282 A. J. Megibow

a b

Fig. 16.7a. Metastatic melanoma, tail of pancreas. A mass is present in the pancreatic tail. Notice the hepatic metastases and the
subcutaneous lesions in the fat in the left posterior chest wall. b Melanoma metastatic to the pancreas: follow-up after chemo-
therapy. The lesion has diminished in size. There appears to be a decrease in attenuation possibly due to central necrosis as a
response to the therapy. Note the decrease in size of the hepatic metastasis in segment 2. The subcutaneous lesions persist

a .....
a

b b

Fig. 16.8a,b. Metastatic melanoma to pancreas: Fig. 16.9a,b. Metastatic carcinoid to pancreas. a
mass in body of pancreas results in pancreatic There is a dilated main pancreatic duct to the mid-
duct obstruction (b). Notice the adenopathy in body. A poorly visualized low attenuation region is
the porta hepatis (b) seen at the point of obstruction (arrow). b Gado-
linium (GD}-enhanced Tl-weighted fat-suppressed
image reveals extensive replacement of neck and
head of the gland. The extent of disease is more
clearly depicted than on CT. Notice the multiple
liver metastases shown on both studies
Secondary Pancreatic Tumors: Imaging 283

patients with hepatocellular carcinoma (Fig. 16.10)


and breast carcinoma (Fig. 16.11).
Scattered case reports of pancreatic masses in
patients with multiple myeloma or plasmacytoma
have appeared in the literature (MITCHELL and
HILL 1985; OLSON et al. 1993; SCHEIMAN et al. 1987).
Should a pancreatic mass develop in a patient with
these diagnoses, needle aspiration is recommended.
These lesions are responsive to chemotherapy and
may be temporarily palliated by biliary stenting
during the period of treatment.

16.3.4
Complications and Treatment

Metastatic masses in the pancreatic head can obstruct


both the common bile duct and the pancreatic duct,
resulting in a 'double-duct sign', an imaging feature
which potentially could be confused with primary
adenocarcinoma (Figs. 16.11, 16.12) (SMITH et al.
1994). We have seen two patients who developed
clinical acute pancreatitis related to ductal obstruc-
tion from pancreatic metastases. In one patient
with melanoma, a rapidly growing lesion within the
pancreatic body resulted in central cavitary necro-
sis, destruction of the main pancreatic duct, and the Fig. 16.11. Metastatic breast cancer to pancreas. Images reveal
rapid development of a pseudocyst (Fig. 16.13) asso- obstruction of the pancreatic duct and biliary tree by a hetero-
ciated with the rapidly growing tumor mass. geneous soft-tissue mass (asterisk) in the pancreatic head

Fig. 16.10. Metastatic hepatocellular carcinoma. A low attenu- Fig. 16.12. Metastatic lung carcinoma to pancreas. Dilated
ation mass with hyperdense rim is present in the pancreatic intrahepatic bile ducts and a portion of the dilated pancreatic
body (arrow). The lesion developed 2 years following the duct are visualized. The appearance is indistinguishable from
initial diagnosis primary adenocarcinoma or cystadenocarcinoma
284 A. J. Megibow

c-----.
Fig. 16.13a-d. Central cavitary necrosis - metastatic melanoma. a A low attenuation mass grows across the main pancreatic
duct, resulting in minimal obstruction. b Scan 7 months later. The mass has grown and is associated with the development of
a unilocular fluid collection. c ERCP at same time of examination as b. There is disruption of the main pancreatic duct in the
d

region of the mass. The duct is not entirely destroyed, as evidenced by the filling of the upstream duct. d Contrast injected at
the site of duct disruption demonstrates extravasation into the tumor mass

Several reports have advocated aggressive surgical 16.4


treatment in selected cases. If en-bloc resection is pos- Secondary Pancreatic Involvement
sible, the median overall survival after metastatic dis- by Tumor in Adjacent Lymph Nodes
ease to the pancreas has been reported to be 56 months
(PINGPANK et al. 2002). Others report 5-year survival Multiple lymph node groups cluster around the
rates, in heterogeneous patient populations, ranging pancreas. While they may harbor metastases from
between 17% and 24% (LE BORGNE et al. 2000; WOOD primary pancreatic carcinoma, tumors of the gas-
et al. 2001). In one series of patients with RCC, aggres- trointestinal (GI) tract and lung can also spread
sive resection resulted in 5-year survival rates as high to these lymph nodes. Once a tumor is established
as 75% (SOHN et al. 2001). All reports stress the proper within these nodes, growth directly into the pancre-
selection of patients - those with total resectable dis- atic parenchyma can occur, producing an imaging
ease - as the criteria for this selection. Survival may be picture identical to a primary pancreatic neoplasm.
improved in patients with multiple lesions as long as Metastatic involvement in either the hepatoduode-
the total tumor burden can be resected. nal nodes or the peripancreatic nodes (EFREMIDIS
Secondary Pancreatic Tumors: Imaging 285

Fig. 16.15. Gastric carcinoma invading the pancreas. A soft-


tissue mass (asterisk) extends from the posterior gastric wall
across the lesser sac. The residual, normally enhanced pan-
creas (p) is seen. Note the irregular margin of the advancing
tumor mass against the normally enhanced gland

pancreas and were indistinguishable from peripan-


Fig. 16.14. Metastatic gastric carcinoma to peripancreatic
lymph nodes. A multiseptate mass is seen in the pancreatic
creatic nodal disease (CHARNSANGAVEJ and WHIT-
head. The lesion could easily be mistaken for a cystic pancre- LEY 1993). These observations have been made in
atic neoplasm. However, the extension into the porta hepatis patients with other than colon cancer (Fig. 16.15).
reveals the true identity Lymphoma can affect the peripancreatic nodes,
and this is usually easily recognized in the context of
et al. 1999) may break through the lymph node(s) other imaging findings in the individual patient. The
and invade the pancreas. Peripancreatic lymph node imaging findings of primary pancreatic lymphoma
metastases from inframesocolic lesions follow the will be considered elsewhere. Tuberculous peripan-
distribution of regional lymph nodes, which follow creatic nodes can invade the pancreas, simulating a
the vascular distributions of the major abdomi- pancreatic mass (HULNICK et al. 1985).
nal vessels and mesenteric supporting structures
(McDANIEL et al. 1993) (Fig. 16.14).
Tumors which predictably metastasize to these
lymph nodes include carcinomas of the esophagus, 16.5
stomach, duodenum, and right colon. Carcinoma of Secondary Pancreatic Involvement
the lung and melanoma are also frequent. Lymph by Tumors in Adjacent Organs
nodes cluster beneath the transverse duodenum and
are contiguous with the pancreas. These lymph nodes The pancreas may be a site of secondary tumors
may drain tumors at a considerable distance, most resulting from tumor extension across soft-tissue
frequently located in the cecum or ascending colon. supporting structures between it and adjacent vis-
In 12 patients with ascending colon cancer and pan- cera. Most frequently, these tumors arise from the
creatic metastases, excluding direct extension of the stomach, gallbladder, and proximal colon. The spread
tumor, 7 (58%) had obstruction of the bile duct and/ of tumor is directly through the serosa of the organ
or pancreatic duct, 4 others had symptoms related and into the closely applied pancreas.
to the mass, including pain and gastrointestinal Primary tumors of the posterior wall of the stomach
obstruction. In 8 patients (67%), metastatic tumors (Fig. 16.16) or metastases to this portion of the stomach
involved the pancreas as a focal mass; in the other 4 (Fig. 16.17) can traverse the lesser sac and adhere to
(33%), the masses were lobulated and engulfed the and/or invade the pancreas. In 282 patients with gas-
286 A. J. Megibow

a ..... b

Fig. 16.16a,b. Metastatic breast carcinoma to gastric antrum with secondary pancreatic invasion. a The antral wall is thickened
by a hypoattenuating soft-tissue mass (asterisks). b Scan slightly superior reveals a dilated main pancreatic duct (arrow) in the
tail and body

a b

Fig. 16.17a,b. Gallbladder cancer invading the pancreas. a The gallbladder is replaced by a soft-tissue mass. The mass extends across
the local fat and invades the gastric antrum. Note that the common bile duct is dilated. b The head of the pancreas is engulfed by
the extensive soft-tissue mass. Morrison's pouch is filled with tumor, and greater omental seeding is present as well

tric cancer invading adjacent organs, 150 (53.2%) had of the pancreatic surface, pancreatic invasion can be
tumors invading the pancreas. In cases of pancreas inva- excluded with certainty (BANBA et al.1998).
sion, the extent of lymph node metastasis was severer, Gallbladder carcinoma can be locally invasive
vascular involvement was more frequent, and the rate of and metastasize to regional lymph nodes along the
concomitant liver metastasis was higher. The prognosis hepatoduodenalligament, which can also be a source
was better for patients treated with curative surgery and for invasion of the pancreas (EFREMIDIS et al. 1999).
pancreas resection. Pancreas-invasive gastric cancer The lesion can extend directly across the fat planes
cells are likely to advance via lymphatic and vascular between the gallbladder and biliary tree and pan-
routes (MAEHARA et al. 2000). Cross-sectional imaging creas. Secondary pancreatic involvement should be
has performed poorly in detecting or excluding gastric suspected when the biliary tree is dilated below the
cancer extension into the pancreas (DAVIES et al. 1997; level of the cystic duct and/or pancreatic duct dilata-
TSUBURAYA et al. 1994). In the absence of irregularity tion is present.
Secondary Pancreatic Tumors: Imaging 287

References

Akerstrom G (1996) Management of carcinoid tumors of Maehara Y, Oiwa H, Tomisaki S, Sakaguchi Y, Watanabe A, Anai
the stomach, duodenum, and pancreas. World J Surg 20: H, Sugimachi K (2000) Prognosis and surgical treatment of
173-182 gastric cancer invading the pancreas. Oncology 59:1-6
Banba Y, Kanazawa T, Seto H (1998) CT evaluation of gastric McDaniel KP, Charnsangavej C, DuBrow RA, Varma DG, Gran-
cancer: depth of tumor invasion and pancreas invasion. field CA, Curley SA (1993) Pathways of nodal metastasis in
Radiat Med 16:161-166 carcinomas of the cecum, ascending colon, and transverse
Bianca A, Carboni N, Di Carlo V, Falleni M, Ferrero S, Liverani colon: CT demonstration. AJR Am J RoentgenoI161:61-64
C, Staudacher C, Turra G, Vergani D, Zerbi A (1992) Pan- McLatchie GR, Imrie CW (1981) Acute pancreatitis associ-
creatic malignant melanoma with occult primary lesion. A ated with tumour metastases in the pancreas. Digestion
case report. Pathologica 84:531-537 21:13-17
Biset JM, Laurent F, de Verbizier G, Houang B, Constantes G, Merkle EM, Boaz T, Kolokythas 0, Haaga JR, Lewin JS,
Drouillard J (1991) Ultrasound and computed tomographic Brambs HJ (1998) Metastases to the pancreas. Br J Radiol
findings in pancreatic metastases. Eur J RadioI12:41-44 71:1208-1214
Charnsangavej C, Whitley NO (1993) Metastases to the pan- Mitchell DG, Hill MC (1985) Obstructive jaundice due to
creas and peri pancreatic lymph nodes from carcinoma of multiple myeloma of the pancreatic head: CT evaluation.
the right side of the colon: CT findings in 12 patients. AJR J Comput Assist Tomogr 9:1118-1119
Am J RoentgenoI160:49-52 Mountney J, Maury AC, Jackson AM, Coleman RE, Johnson AG
Cubilla A, Fitzgerald P (1980). Surgical pathology of exocrine (1997) Pancreatic metastases from breast cancer: an unusual
tumors of the pancreas. In: Moosa A (ed) Tumors of the cause of biliary obstruction. Eur J Surg OncoI23:574-576
pancreas. Williams and Wilkins, Baltimore, pp 159-193 Muranaka T, Teshima K, Honda H, Nanjo T, Hanada K,
Davies J, Chalmers AG, Sue-Ling HM, May J, Miller GV, Martin Oshiumi Y (1989) Computed tomography and histologic
IG, Johnston D (1997) Spiral computed tomography and appearance of pancreatic metastases from distant sources.
operative staging of gastric carcinoma: a comparison with Acta Radiol 30:615-619
histopathological staging. Gut 41:314-319 Ng CS, Loyer EM, Iyer RB, David CL, DuBrow RA, Charnsan-
DeJordy JO, Bendel P, Horowitz A, Salomon Y, Degani H (1992) gavej C (1999) Metastases to the pancreas from renal cell
Correlation of MR imaging and histologic findings in carcinoma: findings on three-phase contrast-enhanced
mouse melanoma. J Magn Reson Imaging 2:695-700 helical CT. AJR Am J Roentgenoll72:1555-1559
Efremidis SC, Vougiouklis N, Zafiriadou E, Sofianou F, Sba- Olson MC, Kalbhen CL, Posniak HV (1993) Pancreatic plas-
rounis C, Fardellas Y, Patsiaura K, Leontsini M (1999) macytomas in a patient with multiple myeloma: CT and
Pathways of lymph node involvement in upper abdominal ultrasound findings. Abdom Imaging 18:323-324
malignancies: evaluation with high-resolution CT. Eur Pingpank JF Jr, Hoffman JP, Sigurdson ER, Ross E, Sasson
RadioI9:868-874 AR, Eisenberg BL (2002) Pancreatic resection for locally
Ferrozzi F, Bova D, Campodonico F, Chiara FD, Pas sari A, Bassi advanced primary and metastatic nonpancreatic neo-
P (1997) Pancreatic metastases: CT assessment. Eur Radiol plasms. Am Surg 68:337-340, discussion 340-341
7:241-245 Rice NT, Woodring JH, Mostowycz L, Purcell M (1981) Pan-
Ghavamian R, Klein KA, Stephens DH, Welch TJ, LeRoy AJ, creatic plasmacytoma: sonographic and computerized
Richardson RL, Burch PA, Zincke H (2000) Renal cell carci- tomographic findings. J Clin Ultrasound 9:46-48
noma metastatic to the pancreas: clinical and radiological Rivoire M, Voiglio EJ (1996) Late pancreatic metastases from
features. Mayo Clin Proc 75:581-585 renal cell carcinoma. Surgery 119:240
Hashimoto M, Watanabe G, Matsuda M, Dohi T, Tsurumaru Roland CF, van Heerden JA (1989) Nonpancreatic primary
M (1998) Management of the pancreatic metastases from tumors with metastasis to the pancreas. Surg Gynecol
renal cell carcinoma: report of four resected cases. Hepa- Obstet 168:345-347
togastroenterology 45:1150-1154 Rumancik WM, Megibow AJ, Bosniak MA, Hilton S (1984)
Hulnick DH, Megibow AJ, Naidich DP, Hilton S, Cho KC, Metastatic disease to the pancreas: evaluation by computed
Balthazar EJ (1985) Abdominal tuberculosis: CT evalua- tomography. J Comput Assist Tomogr 8:829-834
tion. Radiology 157:199-204 Sato M, Ishida H, Konno K, Hamashima Y, Naganuma H, Kom-
Kelekis NL, Semelka RC, Siegelman ES (1996) MRI of pancre- atsuda T, Ishida J, Watanabe S (2001) Pancreatic metastasis:
atic metastases from renal cancer. J Comput Assist Tomogr sonographic findings. Abdom Imaging 26:72-75
20:249-253 Scatarige JC, Horton KM, Sheth S, Fishman EK (2001) Pancre-
Klein KA, Stephens DH, Welch TJ (1998) CT characteristics atic parenchymal metastases: observations on helical CT.
of metastatic disease of the pancreas. Radiographics 18: AJR Am J RoentgenoI176:695-699
369-378 Scheiman J, Elta G, Francis I (1987) Biliary obstruction sec-
Le Borgne J, Partensky C, Glemain P, Dupas B, de Kerviller B ondary to an extramedullary plasmacytoma of the pan-
(2000) Pancreaticoduodenectomy for metastatic ampullary creas: confusion with pancreatitis on computed tomogra-
and pancreatic tumors. Hepatogastroenterology 47:540-544 phy. Pancreas 2:237-239
Leen E, Angerson WJ, Yarmenitis S, Bongartz G, Biomley M, Smith CD, Behrns KE, van Heerden JA, Sarr MG (1994) Radi-
Del Maschio A, Summaria V, Maresca G, Pezzoli C, Uull JB cal pancreatoduodenectomy for misdiagnosed pancreatic
(2002) Multi-centre clinical study evaluating the efficacy of mass. Br J Surg 81:585-589
SonoVue (BR1), a new ultrasound contrast agent in Dop- Sohn TA, Yeo CJ, Cameron JL, Nakeeb A, Lillemoe KD (2001)
pler investigation of focal hepatic lesions. Eur J Radiol 41: Renal cell carcinoma metastatic to the pancreas: results of
200-206 surgical management. J Gastrointest Surg 5:346-351
288 A. J. Megibow

Solomons G, Gibson RN, Tello RJ (2000) Pancreatic head mela- Weiss L, Harlos JP, Torhorst J, Gunthard B, Hartveit F, Svend-
noma producing obstructive jaundice: magnetic resonance sen E, Huang WL, Grundmann E, Eder M, Zwicknagl M
characteristics aiding differentiation from adenocarci- et al (1988) Metastatic patterns of renal carcinoma: an
noma. Aust Radiol 44:471-473 analysis of 687 necropsies. J Cancer Res Clin Oncol 114:
Texler ML, Pierides J, Maddern GJ (1998) Case report: a hepa- 605-612
tocellular carcinoma metastasis in the distal pancreas. J Wernecke K, Peters PE, Galanski M (1986) Pancreatic metas-
Gastroenterol Hepatol13:467-470 tases: US evaluation. Radiology 160:399-402
Tsuburaya A, Noguchi Y, Matsumoto A, Kobayashi S, Masu- Willis R (1973) The Spread of tumors in the human body. But-
kawa K, Horiguchi K (1994) A preoperative assessment of terworth, London, pp 216-217
adjacent organ invasion by stomach carcinoma with high Wilson TE, Korobkin M, Francis IR (1989) Pancreatic
resolution computed tomography. Surg Today 24:299-304 plasmacytoma: CT findings. AJR Am J Roentgenol 152:
Twomey BP, Katz D (1983) Computed tomography in the diag- 1227-1228
nosis of extramedullary plasmacytoma. J Comput Tomogr Wood TF, DiFronzo LA, Rose DM, Haigh PI, Stern SL, Wanek
7:407-408 L, Essner R, Morton DL (2001) Does complete resection
Villanueva A, Perez C, Llauger J, Traid C, Parellada JA, Puig of melanoma metastatic to solid intra-abdominal organs
P (1994) Carcinoid tumors of the pancreas: CT findings. improve survival? Ann Surg Oncol 8:658-662
Abdom Imaging 19:221-224 Z'Graggen K, Fernandez-del Castillo C, Rattner DW, Sigala H,
Weerdenburg JP, Jurgens PJ (1984) Late metastases of a hyper- Warshaw AL (1998) Metastases to the pancreas and their
nephroma to the thyroid and the pancreas. Diagn Imaging surgical extirpation. Arch Surg 133:413-417; discussion
Clin Med 53:269-272 418-419
Subject Index

A Behavior 231
Accessory spleens 237 Beta-emitting isotopes 191
Acinar cell 23,221 Beta-HCG 233
- markers 24 Biliary (enteric) bypass 7, 158
Acinar cell carcinoma 22,162,214,222,224,255-257 - obstruction 37
Acinar cell cystadenocarcinoma 9,18,23,24,223 - stasis 146
Acinar differentiation 225 Biliary duct 238
Acinar growth pattern 225 Biochemical and immunological analysis 13
Acinar markers 231 Biological behavior 6,10,97,119,131,170,251,258
Acinar pattern 223 Biopsy 111
Acinar-endocrine cell tumor 226 Biphasic tumor 17
Acini 24 Blood products 251
Acoustic characteristics 58 Bone metastases 213
ACTH (adrenal corticotrophic hormone) 258 Breath-hold sequences 106,180
ACTH-producing pancreatic islet cell tumor 192 Brunner's gland hamartoma 238
Acute pancreatitis 78
Adenosquamous carcinoma 16,254
Adenosquamous tumor of pancreas 255 C
Adrenal corticotrophic hormone (ACTH) 258 C peptide 146
AFIP (Armed Forces Institute of Pathology) 249 CA 19.9 5,12,19,225,229,254,255
AFP (alpha-fetoprotein) 225 CA 72-4 5
Alanine aminotransferase (ALT) 80 CA 125 5,13
Alcohol abuse 78 Calcification/s 11,15,23,33-35,39,58,59,64, 7l, 101, 115, 129,
Alkaline phosphatase (ALP) 88 133,164,200,201,205,234,239,251,254,260
All-in-one technique 210 Calcifying pancreatitis, chronic 239
Alpha cells 145 Carcinoid 143, 191, 281
Alpha-fetoprotein (AFP) 225 Carcinoid syndrome 281
Alpha-inhibin 17 Carcinomatous invasion (in IPMT) 87,90
ALT (alanine aminotransferase) 80 Cardiovascular manifestations 144
Amine precursor uptake and decarboxylation (APUD) 182 CEA 5,12,17,19,50,70,225,229,255
Anaplastic pancreatic carcinoma 230 Cecal adenocarcinoma 270
Angiographic evaluation 48 Celiac axis 203
Angiography 69,187,188 Cellular palisading 236
Angiosarcoma 234 Central (fibrous) scar 11,12,35,37
Antoni-A component 261 Centro acinar cells 11
Antoni-B component 261 Cephalic resection 6
APUD (amine precursor uptake and decarboxylation) 182 CEUS (contrast-enhanced US) 198,213,214
Armed Forces Institute of Pathology (AFIP) classification 249 Chemo-embolization 157
Arteriogram 186 Chemotheraphy 158
Arteriography 46,184,188 Cholangiopancreatography (MRCP) 41,43,52,64,106,107,
Ascites 123, 127 119,124,129,133,209,210
Aspirate smears 232 - imaging (MR) 57
Asthenia 79,154 Choriocarcinoma 16
Chromogranin A 155
Chromosome 10q 10
B Chronic calcifying pancreatitis 239
B-cell 25, 144 Chronic pancreatitis (CP) 72,77,80,129,133
- hyperplasia 141 Clear cell adenocarcinoma 272,273
Basal acid output (BAO) 147 Clear cell carcinoma 33
Basket-like imprint/pattern 46,210 Clear cell change 230
Beckwith-Wiedemann syndrome 220,258 Clear cell 'sugar' tumor 13
290 Subject Index

Clinical information 81 Dorsal pancreatic anlage 18


Clinical presentation 15,81 Double-duct sign 283
- (ofNFET) 154 DPC4 91
Clinical symptoms 220, 260 Dual-phase acquisition (CT) 200
Collateral branches IPMT 69,70, 129 Dual-phase CT protocol 188
Collateral network 206 Dual-phase helical CT 255
Colloid carcinoma 90 Duct ectatic mucinous cystadenoma 85
Colonic cancer 269 - obstruction 63
Color-doppler (US) 199 Ductal adenocarcinoma 9, 15,214,222,225,253
Common bile duct 123 - dilation 100, 102
Communicating duct 43,49,70,114,115,119,122 Duodenal papilla 100
Compact spindle cell proliferation 238 Duodenectomy 148
Computed tomography (CT) 33,57,250 Dynamic phase (CT) 200,208
- dual-phase 178
- multirow helical 177
Congenital cysts 9 E
Contrast-enhanced US (CEUS) 198,213,214 EBV (Epstein Barr virus) 239
Conventional resection 156 Electron microscopy 225,229,239
Corticotropinoma 192 EMA (epithelial membrane antigen) 12,17,229
Corticotropin-releasing hormone (CRH) 192 En-bloc resection 284
CT (computed tomography) 177,186,188,193,250 Encasement (vascular) 203
- dual-phase helical 255 Endocrine cells 16
- dynamic 252 - cystic tumor 71
Cushing's syndrome 146,220,238 - insufficiency 154
Cushing-like syndrome 258 - markers 25,171
Cystic change/degeneration 203,206,231,235,251 - tumor 9,39,154
- ectasia 99-101,105,123 Endocrine hormone 226
- fibrosis 129,133 Endocrine tumors 225,230
- islet cell (endocrine) Tumor 18,25,64,206 Endoluminal papillary growth 81
- pancreatic lesions 9 - vegetation 81
- solid pseudopapillary tumor 71 Endometrial cysts 9
Cystic fibrosis 238 Endoscopic procedure 181
Cystic lymphangioma 261 Endoscopic Retrograde Cholangiopancreatography (ERCP)
Cystojejunostomy 18 46,52,57,122,123,124,129,180
Cytokeratin 17 - ultrasonography (EUS) 51,53,57,70,123,252
Cytokeratin markers 24 Endoscopic ultrasound (EUS) 51,53,57,70,123,186,187,252
Cytological smears 21 Endoscopically directed FNAB 70, 113
- evaluation 162 Endosonographic scope 182
Cytology 222,225 Enterogenous cysts 9
Enucleation 149
Epithelial markers 235
D Epithelial membrane antigen (EMA) 12, 17,229
3D reconstruction 178 Epstein Barr virus (EBV) 239
Debulking 157 Estrogen receptors (ER) 17,22
Delayed enhancement 63 Etiology 239
Demographic data 80 European Multicentric Trial 212
Derivative surgery 129 EUS (Endoscopic ultrasound) 51,53,57,70,123,186,187,252
Dermoid cyst, true 259 - guided biopsy 252
Diabetes 5,79, 146,278 Exocrine acini 12
- value of 79 Exocrine insufficiency 238
Diabetes mellitus 142 Extraductal proliferation 113
Diagnostic criterion (in cystic tumors) 9 Extra-pancreatic islet cell tumors 193
Diarrhea 144
Diarrheogenic substances 147
Differential diagnosis 21,22,25,31,33,80,204,213,231,232, F
235,237,240 F-18-fluorodeoxy-glucose positron emission tomography
Diff-Quik stain 231 (PET) 191,193
Diffuse intraductal papillary adenocarcinoma 85 Factor VIII 234
Diffusion-weighted echo-planar imaging 108 Fast sequences 179
Dilatation of the main duct (ofWirsung) 52,98,99,111,199 Fast T2-weighted sequences (FSE) 106
Dilated papilla 103 Fat 180
Display collimation 34 Fat necrosis 255
Distal pancreatectomy 156 Fat-suppressed TI-weighted images 56
Subject Index 291

Fat supression 180 HCG (human chorionic gonadotropin) 155


- techniques 179 Helical CT 193
Fatlfluid levels 259 Hemangiopericytoma 234
Fatty involution 180 Hemorrhage 24,34,58,59,64,71,198,220,250,255,272
Fatty liver 200 Hemorrhagic degeneration 251
Fibrolipoma 234 Hemosiderin-laden macrophages 12
Fibrosis 163 Hepatic metastases 7,82,123,149,155,185,204,206,209,212,
Fibrotic atrophy 239 213
Fibrous capsule 237 - transplant 157
Fibrous pancreas 182 Hepatobiliaryanlage 18
Fibrous pseudocapsule 15,23 High frequency miniprobes (EUS) 113
- tissue 63, 208 Histochemical stains 224
Fibrovascular stroma 227 Histogenesis 11,22,229
Findings (correlating to malignancy) 113 Histologic pattern 238,272
Fine-needle aspiration biopsy (FNAB) 5,6,13,19,21,57,64, Histological type 10
71,155,161,214,252,270 Hodgkin's disease 269
Fistulas (in IPMT) 86 Homer-Wright rosettes 236
Fistulous tract 11 0 Honeycomb (architecture) 32,37,43,49,50,113,114
Fluid -debris levels 251 Hormonal hyperproduction 154
Fluid-fluid level 59,60 Human chorionic gonadotropin (HCG) 155
FNAB 270 Hypervascular endocrine tumors 184
FNAB (Fine needle aspiration biopsy) 5,6,13,19,21,57,64, Hypervascular lesion 44
71,155,161,214,252,270 Hypervascular metastases 180
Follow-up 82,83 Hypervascularization 46, 197
Frantz's tumor 250 Hypervascularized tumor 200
Functional secreting neuroendocrine tumors 141 Hypoglicemia 144
Functioning endocrine tumors 212
Fungal infection 240

IDUS (intraductal ultrasonography) 88,92,113


G Image guided FNAB 6
Gadolinium-BOPTA 209,211 Imaging findings 98
Gallbladder carcinoma 286 Immunocytochemical method 166
Gamma-glutamyl transferase (GGT) 80 Immunohistochemical features 221,272
Gastric carcinoma 270 Immunoreactivity 17
Gastric signet-ring cell carcinoma 269 Immunostains 224
Gastric-type mucin markers (PGII and Ml) 17 Imunohistochemistry 229
Gastrin 192 Infantile pancreatic carcinoma 220
Gastrinoma 141,142,145,183,185,189,212 Inflammatory myofibroblastic tumor 238
Gastrinoma triangle 188 Inflammatory myofibrohistocystic proliferation 238
Gastrointestinal hemorrhages 145 In-octreotide scintigraphy 188
G-cells 144 Insulin 192
Genetic screening 4 Insulinoma 141,142,181,185,187
Germ cells 233 Intermediate resection 6
Germ-cell tumors 219 Intraductal component 230
GGT (gamma-glutamyl transferase) 80 Intraductal mucin-hypersecreting neoplasm 85
Giant-cell carcinoma 227 - carcinoma and invasive carcinoma, IPMC/IC 77,89
Giant-cell tumor 227 - ductectatic mucin-hypersecreting variant 86
Glandular-like or acinar pattern (of growth) 164 - IPMA/B 77,89
Glucagonoma 141,142,168,184,185 - IPMT of the main duct 80,81,86,130
Gradient recalled echo (GRE), Tl-weighted (FS) sequences - IPMT of the peripheral (branch)(collateral) ducts 43,81,87,
42,67,106,108,119,128,253,279 131
GRFoma 192 - oncocytic variant 89
Growth pattern 164,223,226 - operability 82
- papillary (mucinous) neoplasms (IPMNs) 14,15,85
- papillary-villous variant 86
H - pathological hallmarks of 85
Half Fourier single-shot turbo spin echo (HASTE) 64,106, - prognosis 93
107,119,209 - resectability 82
Hamartoma 261 - tumor, IPMT 41,57,77,80,85
Harmonic imaging 197 - types of infiltration 82
HASTE (Half Fourier single-shot turbo spin echo) 64, 106, Intraductal papillary carcinoma 231
107,119,209 Intraductal papillary mucinous tumor (rPMT) 255
292 Subject Index

Intraductal ultrasonography (lDUS) 88,92, 113 Macroscopic feature 34,220,223,227


Intraluminal (filling) defects 110,114,122,129 Magnetic resonance angiography 209
Intraoperative examination 146 Magnetic resonance (MR) 179
- ultrasound 148 Main duct IPMT 124
Intraoperative palpation 193 Major papilla 86,125,127
Intraoperative ultrasonography 193 Malignancy 16,208
Intraoperative ultrasound (lOUS) 182,188 - indications 115
Intrapancreatic embryonic rests 259 Malignant cystic tumors 37
Intrapancreatic metastases 271 - degeneration 6,59,100,102,130,131
Intrapancreatic teratoma 233 - potential 162
Invasive mucinous cystadenocarcinoma 16 Malignant degeneration 185
Iodine-labeled -compound 183 Malignant fibrous histiocytoma 237
IPMT (intraductal papillary mucinous tumor) 255 Malignant melanoma 274
Islet cell tumor 178,185,222,262,272 Management (therapeutic) 98,129,156
Islets of Langerhans 12 Mangafodopir trisodium (Mn-DPDP) 179,255
Markers 225
- of malignancy 170
Mature teratoma 25
Jaundice 79,223 May-Grunwald-Giemsa 231
MDCT (multislice (multidetector-row) CT) 33,34,60,64,114
Medical therapy 158
K Melanin 281
Kaposi sarcoma 261 Melanocyte-stimulating hormone (MSH) 192
Ki-67 21,155 Melanoma metastases 281
K-ras (mutation) 90,91,222 MEN - 1 (syndrome) 72,153-155,161,163
MEN type I 185,188,192
Mesenchymal tumors 219
L Metachronous metastases 214
Laboratory 80 Metastases 63,156
Lactate dehydrogenase (LDH) 232,252 - hepatic 178, 179
Leiomyoma, primary 234 - intrapancreatic 271
Leiomyosarcoma 235 - of the liver 226, 251
Lifestyle habits 80 - pancreatic 272,277,278
Lipoblastoma 234 Metastatic breast cancer to pancreas 283
Lipoma 234 Metastatic carcinoid to pancreas 282
Liposarcoma 234 Metastatic cascades 278
Liver mestastases 156, 185, 191, 192, 198, 204, 226, 251 Metastatic clear-cell carcinoma 13
Loading tests 147 Metastatic glucagonoma 191
Local (tumor) recurrence 82 Metastatic hepatocellular carcinoma 283
- in IPMT 92 Metastatic melanoma 282
Location ofNFET 154 Metastatic tumors of the pancreas 271
Lung cancer 279 Microcystic (or glicogen-rich) cystadenoma 10,57
- metastases 280 - appearance/architecture 33,49
Lymph node involvement 252 - (or classic) type (SCA)(SCT) 11,32,47
Lymph node metastases 87,257,286 - tumor 46
Lymphangioma/s 13,129,234 Microcystic adenoma 272,273
Lymphatic invasion 269 Microscopic features 227,229
Lymphoepithelial cyst/s 9,25 Minor papilla 86
Lymphoid infiltration 272 Mitotic activity 164
lymphoma 219,285 Mixed acinar-endocrine cell carcinoma 219
Lymphoplasmacytic lymphoma 239 Mixed appearance (ofNHFET) 202
- pattern 32
- tumor 33
M Mixed exocrine-endocrine tumors 226
Macrocystic cystadenoma/cystadenocarcinoma 57 MNCC (mucinous noncystic carcinoma) 18,85,90,91
- multilocular (appearance) pattern (MCT) 58,60,61,62,64, Mn-DPDP (mangafodopir trisodium) 180,255
71 Molecular analysis 225,229
- multilocular mass with thick wall 71 Morphological patterns 278
- multilocular mass with thin wall 71 MR(magnetic resonance) 179
- oligo cystic type (SCA) 11,33 MR (evaluation) 53,64,207
- unilocular pattern 59,63,64,72 - sequences 41
Macrocysts 11,37,43,46,50 MR cholangiopancreatography (MRCP) 179,180
Macroscopic characteristics 97 MRCP 179, 180
Subject Index 293

MRI 188,279 Obstructive jaundice 239


MSH (melanocyte-stimulating hormone) 192 Obstructive symptoms 7
Mucin 78,101,106,110,119 Octreoscan 158,183,211-214
- globules 100 Octreotide 183, 209
- hyperproduction 78 - scintigraphy (SRS) 212
- leakage 124 Ohhashi triad 77
- like carcinoma -associated antigen 19 Oligo cystic SCT 46,48, 7l
- plugs 86 Omental masses 127
- producing tumor 85 Oncocytic change 230
- secreting epithelium 14,15 Oncocytic tumor 219,231
Mucinous cystadenoma (MCA) 16,19 Operability 82
- cystadenocarcinoma (MCC) 4,16,19,60 Oral contrast (agent) 34,116
- cystic neoplasm of borderline malignant potential (MCB) - medium 101
16,19 Osteoclast-like giant cell tumor 16,219,227,229,257
- cystic neoplasms (MCN) 9,13-15,17-19 Ovarian (like) type stroma 3,14-17,88
- tumor, MCT 5,81,70,93,129,214
Mucinous cystic neoplasm 229
Mucinous cystic tumor 251,260 P
Mucinous neoplasms 10 Pain 78
Mucinous noncystic carcinoma (MNCC) 18,85,90,91 Pancreas, metastatic tumors 27l
Muconodular infiltration 82 Pancreatic acinar cell carcinoma 219
Mucous-hypersecreting tumor 85 Pancreatic carcinoma, primary 284
Mucous-secreting cancer 85 Pancreatic cholera 145
Multicentric (multifocal) IPMT 121,122 - endocrine tumor (PET) 161
Multilocular cystic mass 41 - insufficiency 154
Multiple endocrine adenomatosis (MEA) 185 - intraepithelial neoplasia (PanIN) 85
Multiple endocrine neoplasia (MEN) 161,185 - parenchyma 35,60,98
Multirow helical CT 186,188 - phase 60,115,200,208
- scanner 178 - polypeptide (PP) 155
Multislice (multidetector-row) CT (MDCT) 33,34,60,64,114 - secretion 146
Mural nodules 15,257 - signal (MR) 119
Myxoid apperance 236 - stone protein (PSP) 24
Myxoid pattern 238 - technique (CT) 101
- type pain 5
Pancreatic choriocarcinoma 233
N
Pancreatic ductal tumors 273
Necrosis 24, 7l, 179, 198,220,236,250,272,283
Pancreatic fatty infiltration 260
Neoplastic thrombus 204,205
Pancreatic hamartoma 238
Neovascularity 184,186
Pancreatic invasion, direct 271
Nesidioblastosis 142
Pancreatic lipoma 261
Neuroblastoma 142
Pancreatic lipomatosis 238
Neuroendocrine markers 230
Pancreatic lymphangioma 260
Neuroepithelioma 236
Pancreatic lymphoma 252,254
Neurofibroma 261
- primary 232,253
Neurofibromatosis of von Recklinghausen (type 1) 146,169
Pancreatic metastases 272,277,278
Neurological alterations 146
Pancreatic pseudo tumors 238
Neuron specific enolase (NSE) 21,155,221
Pancreatic technique 281
Neurovegetative disorders 144
Pancreatic teratoma 259
NHFET (nonhyperfunctioning endocrine tumor) 197
Pancreatic venous sampling 184
- atypical 205
Pancreaticoduodenectomy 31,156
NHL (non-Hodgkin'S lymphoma) 232,252,269,274
Pancreatitis-like pain 81
Nodular fasciitis 238
Pancreatoblastoma 22,219,222,226,259
Nodules 60
Pancreatogram 110
Non-functioning endocrine tumors (lesions) 52,154,155,
Pancreatojejunostomy 129
157,192
Pancreoduodenectomy 82
Nonfunctioning islet cell tumors 22
Papilla major 86
Non-Hodgkin's lymphoma (NHL) 232,252,269,274
- minor 86
Nonhyperfunctioning endocrine tumor (NHFET) 197
- patulous orifice of 86
Nuclear Medicine 214
Papillary cystic neoplasm (tumor) 20,250
Nuclear-to-cytoplasmic ratio 221,222
- growth 81,99,101,110,125,126,131
- projections 15,16,23,60
o - proliferations 59,68,88-90,101,102,104,106,108,110,111,
Obstrucitve pancreatitits 239 115,119,122
294 Subject Index

Papillary epithelial neoplasm 250 - markers 230,272


Parameters (in evaluating neoplasms) 81 Resectability 82, 156
Paraneoplastic hypercalcemia 169 Resection 6, 7
Parathyrinoma 192 Residual disease 157
Parenchymal atrophy 100,101,104,106,128 Respiratory triggering 179
- calcifications 102 Retention cyst 129
- thickness 101,102 Retroperitoneal fibrosis 238,239
Parietal nodules 63 Retroperitoneallymphadenopathy 191
PAS (periodic acid-schiff) 10,12,224 Ring-like peripheral enhancement 186
Pathway 277 Risk of misdiagnosing 105
Patient's position 102, 115
Pattern of enhancement 279
Periodic acid-Schiff (PAS) (stain) 10,12,224 S
Peripancreatic fat necrosis 240 Salt and pepper pattern 162
Peripancreatic veins 203,206 Sampling 19
- vessels 60, 69 Santorini (duct) 99
Peripheral cystic tumors 3 Sarcoidosis 240
Peripheral eosinophilia 223 Sarcoma 261
Peritoneal carcinomatosis 123 Schwann cells 236,261
Peritumoral infiltration (extension) 7, 17 Schwannoma 260
-lymphadenopathy 198 Scintigraphy 262
Perivascular epithelioid cells (PEC) 13 Scirrous tissue 208
Peroral pancreatoscopy (POPS) 88 Secondary pancreatic involvement 286
PET (F-18-fluorodeoxy-glucose positron emission tomogra- Secretagogue 185
phy) 191,293 Secretin 69
Plain film (examination) 44,64 SegmentalIP11T 98,105,106,111,129
Plasma cell granuloma 238 Selective arteriography 46
Pleomorphic giant-cell carcinoma 230 Sensitivity of transabdominal ultrasound 187
PNETs 236 Septa/septations 60
Portal vein invasion 186 Serous adenoma 12
Positron emission tomography (PET) 183,212 Serous cystadenoma (SCA) 10,18,25,80,81,129
Posterior shadow/ing (US) 33,101 - cyst 4
Power doppler 123,126 - cystadenocarcinoma (SCAC) 10,31
Predictors (of malignancy) 71 Serous cystic tumor (SCT) 5,41,70
Premalignant lesion 57 - micro cystic tumor (adenoma) 9,13
Preoperative diagnosis 19,21 - oligocystic (macrocystic) tumor (adenoma) 9,10
Primary hyperparathyroidism 153 Serum tumor markers 19
Primary leiomyoma 234 Signal intensity 208
Primary pancreatic carcinoma 284 Signs of malignancy 197
Primary pancreatic lymphoma 232,253 Single photon emission computed tomography (SPECT) 211
Primordial gonad 18 Single-row helical CT 186
Progesterone receptors (PR) 17,22 Sjogren's syndrome 239
Prognosis 14,19,20,98,149,191,214 Skip lesions 135
Prognostic criteria 71 Small cell carcinoma metastatic to the pancreas 281
Pro-insulin 146 Small-cell cancer 280
Protruding papilla 77 Solid and cystic acinar cell tumors 250
Psammoma bodies 164 Solid and papillary cystic (epithelial) tumor 9,20
Pseudocysts 13,18,21,37,59,64,71,78,252,283 Solid and papillary epithelial neoplasm 250
Pseudopapillae 21, 24 Solid pseudopapillary pancreatic neoplasm 251
Solid pseudopapillary tumor (SPT) 9,13,18,20,21,162,214,
219,222,226,228,230
R - nodular (insular) pattern (of growth) 164
Radical resection (in functioning endocrine tumors) 148 Solid serous adenoma 12
Radiologicfeatures 192 Solid serous cystadenoma 230
Radiometabolic therapy 158,213 Solitary fibrous tumor 237
Rapid acquisition relaxation enhancement (RARE) 64 Somatostatin 191
RCC (renal cell carcinoma) 271,278 Somatostatin analogue 149,158,183
- metastases 279 - receptors 213, 214
Receptor scintigraphy 209,211 Somatostatin receptors 182, 183
Reconstruction interval (CT) 34 Somatostatin-receptor imaging 184
Recurrence 156 Somatostatin-receptor localization 187
Recurrent pain 78 Somatostatin-receptor scintigraphy 188,189,193
Renal cell carcinoma (RCC) 271,278 Somatostatinoma 143,147,185,191
Subject Index 295

Sonovue 279,280 Tubular type (of infiltration) 82


Spectrum of pancreatic tumors 9 Tumor
Spindle cell tumors 239 - hyperfunctioning endocrine 179
Spiral CT 60 - hypervascular 178
Splenic infarcts 163 Tumor thrombus 255
Splenopancreasectomy 82 Tumor type 6
Sponge-like (appearance) (mass) (SCT) 33,39,42,43,46,113 Tumoral markers 49
Squamoid corpuscules 220,258 Tumor-like lesions 220
Stromal luteinization 17 Type of infiltration 82
Structural findings 60
Sugar tumor 272,273
Superior mesenteric artery 203 U
Superparamagnetic contrast media 106 Ultrasonography (US) 57,197
Surgery 227 Ultrasound contrast agents 279
- indications 82 Ultrastructural findings 221
Surgical biopsy 273 Unilocular mass 59
Surgical indication 6 - with thick wall 71
- resection 7,14,71,98,129,156,157 - with thin wall 71
- treatment 149
Surgical resection 229,236
Survival rate 149,156,157 v
Suspicion of malignancy 102 Vascular encasement 46
Symptom complex 154 - resection 156
Symptoms 10,32,80,81,214,191,235 - thrombosis 163
Synchronous (metastases) 214 Vascular invasion 258
Syndrome of inappropriate ADH -secretion 258 Vasoactive intestinal polypeptide (VIP) 142,145,226
Venous involvement 184
Venous phase 63,189,206
T - sampling 148
Tl-weighted fat-suppression sequences 186 - thrombosis 63
Tl-weighted gradient recalled echo 253,279 Venous sampling 187,188
T2 short time inversion recovery (STIR) 207 Verner-Morrison Syndrome 145,170
TZ-W fast (turbo) spin-echo (TSE) 42 VHLgene 10
T2-weighted images 180,255 Villous adenoma of the main pancreatic duct 85
Target -like metastases 204 - adenomatosis of duct ofWirsung 85
Therapeutic management/option(s) 31,77 VIP (vasoactive intestinal polypeptide) 142, 145, 226
- approach 71 VIPoma 142,185,190
Thickness (of the septa/walls) 58,81 Von Hippel-Lindau disease 4,33,39,40,43,153
Three-dimensional reconstruction 203 - syndrome (VHL) 10
Tissue harmonics 58 Von Recklinghausen's disease 236
Total pancreatectomy 131,133
Trabecular pattern 164,168
Transabdominal ultrasonography (US) 123 W
Transabdominal ultrasound 181,186 Watchful monitoring 135
Transduodenal FNA 122 WDHA 144
Transhepatic venous sampling 184 Weight loss 79
Trans-illumination 148 WHO (World Health Organization) 250
Traumatic neuroma 240 - (2000) classification 10,89,156,158,161
Treatment 222,232 Wilkinson migrant necrolitic erythema 144
Treatment and prognosis 239
Triple-phase 178
Tuberculosis 240 Z
Tuberculous peripancreatic nodes 285 Zollinger-Ellison syndrome 144,168,192
List of Contributors

CLAUDIO BASSI, MD LUCA CASETTI, MD


Department of Surgical-Gastroenterological Department of Surgical-Gastroenterological
Endocrine and Pancreatic Unit Endocrine and Pancreatic Unit
Hospital "GB Rossi" Hospital "GB Rossi"
University of Verona University of Verona
37134 Verona 37134 Verona
Italy Italy

ROSSELLA BETTINI, MD FERNANDO CIRILLO, MD

Department of Surgical-Gastroenterological Department of General Surgery II


Endocrine and Pancreatic Unit "Istituti Ospitalieri"
Hospital "GB Rossi" Hospital- Cremona and Post-Graduate School
University of Verona of Endocrinology and Metabolic Diseases
37134 Verona Viale Concordia, 1
Italy 26100 Cremona
Italy
CARLO BIASIUTTI, MD
DANIELA COSER, MD
Istituto di Radiologia
Istituto di Radiologia
Policlinico Universitario "GB Rossi"
Policlinico Universitario "GB Rossi"
P.le LA Scuro, 10
P.le LA Scuro, 10
37134 Verona
37134 Verona
Italy
Italy

GIACOMO BOGINA,MD EMILIANO DELLA CHIARA, MD


Servizio di Anatomia Patologica Istituto di Radiologia
Ospedale S. Cuore-Don Calabria Policlinico Universitario "GB Rossi"
Via D. Sempreboni P.le LA Scuro, 10
37024 Negrar (VR) 37134 Verona
Italy Italy

ANTONIETTA BRIGHENTI, MD MASSIMO FALCONI, MD


Servizio di Anatomia Patologica Department of Surgical-Gastroenterological
Ospedale S. Cuore-Don Calabria Endocrine and Pancreatic Unit
Via D. Sempreboni Hospital "GB Rossi"
37024 Negrar (VR) University of Verona
Italy 37134 Verona
Italy
PAOLA CAPELLI, MD
MARCO FERDEGHINI, MD
Istituto di Anatomia Patologica Istituto di Radiologia
Policlinico Universitario "GB Rossi" Policlinico Universitario "GB Rossi"
P.le LA Scuro, 10 P.le LA Scuro, 10
37134 Verona 37134 Verona
Italy Italy

GIOVANNI CARBOGNIN, MD MAURO FERRARI, MD


Istituto di Radiologia Istituto di Radiologia
Policlinico Universitario "GB Rossi" Policlinico Universitario "GB Rossi"
P.le LA Scuro, 10 P.le LA Scuro, 10
37134 Verona 37134 Verona
Italy Italy
298 List of Contributors

FRANCESCA FORNASA, MD ENRICO MOLINARI, MD


Istituto di Radiologia Department of Surgical-Gastroenterological
Policlinico Universitario "GB Rossi" Endocrine and Pancreatic Unit
P.le LA Scuro, lO Hospital "GB Rossi"
37134 Verona University of Verona
Italy 37134 Verona
Italy
ISAAC R. FRANCIS, MD
Professor Radiology and Associate Chair for Research THAIRA OWElTY, MD
Department of Radiology Department of Pathology
University of Michigan Medical Center NYU School of Medicine
RmBID502 560 First Avenue (TH 461)
1500 East Medical Center Dr., Box 0300 New York, NY lO016
Ann Arbor, MI 48lO9 USA
USA

NICOLETTA PAGNOTTA, MD
ISABELLA FRIGERIO, MD Istituto di Radiologia
Department of Surgical-Gastroenterological Policlinico Universitario "GB Rossi"
Endocrine and Pancreatic Unit P.le LA Scuro, lO
Hospital "GB Rossi" 37134 Verona
University of Verona Italy
37134 Verona
Italy
PAOLO PEDERZOLI, MD
Dipartimento di Scienze Chirurgiche
ARNALDO FUINI, MD Policlinico Universitario "GB Rossi"
Servizio di Gastroenterologia P.le LA Scuro, 1
Ospedale Civile Maggiore 37134 Verona
P.le Stefani, 1
Italy
37124 Verona
Italy
ANNA PESCI, MD
Servizio di Anatomia Patologica
ALESSANDRO GUARISE, MD
Ospedale S. Cuore-Don Calabria
Servizio di Radiologia
Via D. Sempreboni
Ospedale S. Cuore-Don Calabria
37024 Negrar (VR)
Via D. Sempreboni
Italy
37024 Negrar (VR)
Italy
ENRICO PETRELLA, MD
WILLIAM MANTOVANI, MD Istituto di Radiologia
Policlinico Universitario "GB Rossi"
Department of Surgical-Gastroenterological
P.le LA Scuro, 10
Endocrine and Pancreatic Unit
37134 Verona
Hospital "GB Rossi"
Italy
University of Verona
37134 Verona
Italy CARLO PROCACCI, MD
Professor, Istituto di Radiologia
GUIDO MARTIGNONI, MD Dipartimento di Scienze Morfologico-Biomediche
Istituto di Anatomia Patologica Universita degli Studi di Verona
Policlinico Universitario "GB Rossi" Policlinico "Giambattista Rossi"
P.le LA Scuro, lO P.zza L. A. Scuro 10
37134 Verona 37134 Verona
Italy Italy

ALEC J. MEGIBOW, MD, MPH, FACR ROBERTO SALVIA, MD


Department of Radiology Department of Surgical-Gastroenterological
NYU School of Medicine Endocrine and Pancreatic Unit
Department of Radiology Hospital "GB Rossi"
560 First Avenue University of Verona
New York, NY 10016 37134 Verona
USA Italy
List of Contributors 299

ALDO SCARPA, MD SIMONE VASORI, MD


Istituto di Anatomia Patologica Istituto di Radiologia
Policlinico Universitario "GB Rossi" Policlinico Universitario "GB Rossi"
P.le LA Scuro, 10 P.le LA Scuro, 10
37134 Verona 37134 Verona
Italy Italy

SILVIA VENTURINI, MD
GIACOMO SCHENAL, MD Istituto di Radiologia
Istituto di Radiologia Policlinico Universitario "GB Rossi"
Policlinico Universitario "GB Rossi" P.le LA Scuro, 10
P.le LA Scuro, 10 37134 Verona
37134 Verona Italy
Italy
A. BRIAN WEST, MD, FRCPath
MARG HERITA TAPPARELLI, MD Professor and Director of Anatomic Pathology
NYU Medical Center
Istituto di Radiologia
Policlinico Universitario "GB Rossi" Department of Pathology
560 First Avenue, (TH 461)
P.le LA Scuro, 10
New York, NY 10016
37134 Verona
USA
Italy

GIUSEPPE ZAMBONI, MD
RUEDI F. THOENI, MD Universita di Verona
Professor of Radiology Servizio di Anatomia-Istologia Patologica
University of California San Francisco Ospedale S. Cuore-Don Calabria
Campus Box 0628 Via don Sempreboni, 5
San Francisco, CA 94143-0628 37024 Negrar-Verona
USA Italy
MEDICAL RADIOLOGY Diagnostic Imaging and Radiation Oncology
Titles in the series already published

DIAGNOSTIC IMAGING

Innovations in Diagnostic Imaging Functional MRI 3D Image Processing


Edited by J. H. Anderson Edited by c. T. W. Moonen and P. A. Bandettini Techniques and Clinical Applications
Edited by D. Caramella and C. Bartolozzi
Radiology of the Upper Urinary Tract Radiology of the Pancreas
Edited by E. K. Lang 2nd Revised Edition Imaging of Orbital and Visual Pathway Pathology
Edited by A. 1. Baert Edited byW. S. Muller-Forell
The Thymus - Diagnostic Imaging, Functions,
Co-edited by G. Delorme and 1. Van Hoe
and Pathologic Anatomy Pediatric ENT Radiology
Edited by E. Walter, E. Willich, and W. R. Webb Emergency Pediatric Radiology Edited by S. J. King and A. E. Boothroyd
Edited by H. Carty
Interventional Neuroradiology Radiological Imaging ofthe Small Intestine
Edited by A. Valavanis Spiral a of the Abdomen Edited by N. C. Gourtsoyiannis
Edited by F. Terrier, M. Grossholz,
Radiology of the Pancreas Imaging of the Knee
and C. D. Becker
Edited by A. 1. Baert, co-edited by G. Delorme Techniques and Applications
Liver Malignancies Edited by A. M. Davies
Radiology of the Lower Urinary Tract
Diagnostic and Interventional Radiology and v. N. Cassar-Pullicino
Edited by E. K. Lang
Edited by C. Bartolozzi and R. Lencioni
Magnetic Resonance Angiography Perinatal Imaging
Medical Imaging of the Spleen From illtrasound to MR Imaging
Edited by 1. P. Arlart, G. M. Bongartz,
Edited by A. M. De Schepper Edited by Fred E. Avni
and G. Marchal
and F. Vanhoenacker
Contrast-Enhanced MRI of the Breast Radiological Imaging ofthe Neonatal Chest
Radiology of Peripheral Vascular Diseases Edited by V. Donoghue
S. Heywang-Kobrunner and R. Beck
Edited by E. Zeitler
Spiral a ofthe Chest Diagnostic and Interventional Radiology
Diagnostic Nuclear Medicine in Liver Transplantation
Edited by M. Remy-Jardin and J. Remy
Edited by C. Schiepers Edited by E. Bucheler, V. Nicolas,
Radiological Diagnosis of Breast Diseases Radiology of Blunt Trauma of the Chest C. E. Broelsch, X. Rogiers, and G. Krupski
Edited by M. Friedrich and E.A. Sickles
P. Schnyder and M. Wintermark Radiology of Osteoporosis
Radiology of the Trauma Portal Hypertension Edited by S. Grampp
Edited by M. Heller and A. Fink Diagnostic Imaging-Guided Therapy Imaging and Intervention in Abdominal Trauma
Biliary Tract Radiology Edited by P. Rossi Edited by R. F. Dondelinger
Edited by P. Rossi Co-edited by P. Ricci and 1. Broglia
Imaging of the Foot and Ankle
Radiological Imaging of Sports Injuries Recent Advances in Diagnostic Neuroradiology Techniques and Applications
Edited by C. Masciocchi Edited by Ph. Demaerel Edited by A. M. Davies, R. W. Whitehouse,
Modern Imaging of the Alimentary Tube Virtual Endoscopy and Related 3D Techniques and J. P. R. Jenkins
Edited by A. R. Margulis Edited by P. Rogalla, Interventional Radiology in Cancer
J. Terwisscha Van Scheltinga, and B. Hamm Edited by A. Adam, R. F. Dondelinger,
Diagnosis and Therapy of Spinal Tumors
Edited by P. R. Algra, J. Valk, and J. J. Heimans Multislice a and P. R. Mueller
Edited by M. F. Reiser, M. Takahashi, Imaging of the Pancreas
Interventional Magnetic Resonance Imaging M. Modic, and R. Bruening
Edited by J. F. Debatin and G. Adam Cystic and Rare Tumors
Pediatric Uroradiology Edited by C. Procacci and A. J. Megibow
Abdominal and Pelvic MRI Edited by R. Fotter
Edited by A. Heuck and M. Reiser Intracranial Vascular Malformations
Transfontanellar Doppler Imaging in Neonates and Aneurysms
Orthopedic Imaging A. Couture and C. Veyrac From Diagnostic Work-Up
Techniques and Applications to Endovascular Therapy
Edited by A. M. Davies and H. Pettersson Radiology of AIDS Edited by M. Forsting
A Practical Approach
Radiology of the Female Pelvic Organs Edited by J.W.A.J. Reeders and P.C. Goodman Imaging Pelvic Floor Disorders
Edited by E. K.Lang Edited by C. 1. Bartram and J. o. 1. DeLancey
Magnetic Resonance of the Heart
a of the Peritoneum Associate Editors: S. Halligan, F. M. Kelvin,
Armando Rossi and Giorgio Rossi and J. Stoker
and Great Vessels
Clinical Applications Magnetic Resonance Angiography High Resolution Sonography
Edited by J. Bogaert, A. J. Duerinckx, 2nd Revised Edition of the Peripheral Nervous System
and F. E. Rademakers Edited by 1. P. Arlart, G. M. Bongratz, Edited by S. Peer and G. Bodner
and G. Marchal
Modern Head and Neck Imaging Radiology Imaging of the Ureter
Edited by S. K. Mukherji and J. A. Castelijns Pediatric Chest Imaging Edited by F. Joffre, ph. Otal, and M. Soulie
Edited by Javier Lucaya and Janet 1. Strife
Radiological Imaging of Endocrine Diseases
Edited by J. N. Bruneton Applications of Sonography
in collaboration with B. Padovani in Head and Neck Pathology
and M.-Y. Mourou Edited by J. N. Bruneton in collaboration with
C. Raffaelli and o. Dassonville
Trends in Contrast Media
Edited by H. S. Thomsen, R. N. Muller, Imaging of the Larynx
and R. F. Mattrey Edited by R. Hermans Springer
ME DIe A L R A D I 0 LOG Y Diagnostic Imaging and Radiation Oncology
Titles in the series already published

RADIATION Non-Disseminated Breast Cancer Radiation Therapy of Benign Diseases


ONCOLOGY Controversial Issues in Management A Clinical Guide
Edited by G. H. Fletcher and S.H. Levitt 2nd Revised Edition
S. E. Order and S. S. Donaldson
Current Topics in Clinical Radiobiology
ofTumors Carcinoma of the Kidney and Testis, and Rare
Edited by H.-P. Beck-Bornholdt Urologic Malignancies
Innovations in Management
Practical Approaches to Cancer Invasion Edited by Z. Petrovich, L. Baert,
Lung Cancer and Metastases and L.w. Brady
Edited by C.W. Scarantino
A Compendium of Radiation
Oncologists' Responses to 40 Histories Progress and Perspectives in the Treatment
Innovations in Radiation Oncology Edited by A. R. Kagan with the of Lung Cancer
Edited by H. R. Withers and L. J. Peters Assistance of R. J. Steckel Edited by P. Van Houtte, J. Klastersky,
and P. Rocmans
Radiation Therapy of Head and Neck Cancer Radiation Therapy in Pediatric Oncology
Edited by G. E. Laramore Combined Modality Therapy of
Edited by J. R. Cassady
Central Nervous System Tumors
Gastrointestinal Cancer - Radiation Therapy Radiation Therapy Physics Edited by Z. Petrovich, L. W. Brady,
Edited by R.R. Dobelbower, Jr. Edited by A. R. Smith M. L. Apuzzo, and M. Bamberg
Radiation Exposure and Occupational Risks Late Sequelae in Oncology Age-Related Macular Degeneration
Edited by E. Scherer, C. Streffer, Edited by J. Dunst and R. Sauer Current Treatment Concepts
and K.-R. Trott
Edited by W. A. Alberti, G. Richard,
Mediastinal Tumors_ Update 1995 and R. H. Sagerman
Radiation Therapy of Benign Diseases Edited by D. E. Wood and C. R. Thomas, Jr.
A Clinical Guide Radiotherapy of Intraocular
S.E. Order and S. S. Donaldson Thermoradiotherapy and Orbital Tumors
and Thermochemotherapy 2nd Revised Edition
Interventional Radiation Therapy Techniques Edited by R. H. Sagerman, and w. E. Alberti
- Brachytherapy Volume 1:
Edited by R. Sauer Biology, Physiology, and Physics
Clinical Target Volumes in Conformal and
Volume 2: Intensity Modulated Radiation Therapy
Radiopathology of Organs and Tissues Clinical Applications A Clinical Guide to Cancer Treatment
Edited by E. Scherer, C. Streffer, Edited by M.H. Seegenschmiedt, Edited by V. Gregoire, P. Scalliet,
and K.-R. Trott P. Fessenden, and C.C. Vernon and K. K.Ang
Concomitant Continuous Infusion Carcinoma of the Prostate Biological Modification of Radiation Response
Chemotherapy and Radiation Innovations in Management Edited by C. Nieder, L. Milas, and K. K. Ang
Edited by M. Rotman and C. J. Rosenthal Edited by Z. Petrovich, L. Baert,
and L.W. Brady Palliative Radiation Oncology
Intraoperative Radiotherapy - R. G. Parker. N.A. Janjan, and M. T. Selch
Clinical Experiences and Results Radiation Oncology of Gynecological Cancers
Edited by F. A. Calvo, M. Santos, Edited by H.W. Vahrson
and L.W. Brady
Carcinoma ofthe Bladder
Radiotherapy of Intraocular Innovations in Management
and Orbital Tumors Edited by Z. Petrovich, L. Baert,
Edited by W. E. Alberti and R. H. Sagerman and L.W. Brady

Interstitial and Intracavitary Blood Perfusion and Microenvironment


Thermoradiotherapy of Human Tumors
Edited by M. H. Seegenschmiedt Implications for Clinical Radiooncology
and R. Sauer Edited by M. Molls and P. Vaupel Springer

Potrebbero piacerti anche