Sei sulla pagina 1di 511

Liver Cancer

Methods of Cancer Diagnosis, Therapy, and Prognosis

Volume 5

For other titles published in this series, go to


www.springer.com/series/8172
Methods of Cancer Diagnosis,
Therapy, and Prognosis

Volume 5

Liver Cancer
Edited by

M.A. Hayat
Department of Biological Sciences,
Kean University, Union, NJ, USA
Editor
M.A. Hayat
Department of Biological Sciences
Kean University
Union, NJ, USA

ISBN 978-1-4020-9803-1 e-ISBN 978-1-4020-9804-8


Library of Congress Control Number: 2008930172

© 2009 Springer Science + Business Media B.V.


No part of this work may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic,
mechanical, photocopying, microfilming, recording or otherwise, without written permission from the Publisher, with the exception
of any material supplied specifically for the purpose of being entered and executed on a computer system, for exclusive use by the
purchaser of the work.

Printed on acid-free paper

springer.com
New technology, for better or for worse, will be used, as that is our nature.
Lewis Thomas

You have been given the key that opens the gates of heaven; the same key opens the
gates of hell.
Writing at the entrance to a Buddhist temple
Authors and Co-Authors of Volume 5

Maria I. Altbach Oliver F. Bathe


Department of Radiology, University Department of Surgery and Oncology,
of Arizona, 1501 N. Campbell Avenue, University of Calgary, Tom Baker Cancer
Tucson, AZ 85724, USA Center, 1331 29th Street, N.W., Calgary,
AB T2N 4N2, Canada
Tsutomu Araki
Department of Radiology, University Andrew K. Burroughs
of Yamanashi, 1110 Simokato, Royal Free Hospital, Liver
Chuo, Yamanashi, 409-3898, Japan Transplantation and Hepatobiliary
Medicine, Pond Street, London NW3
Rebecca Auer 2QG, UK
Centre for Cancer Therapeutics,
Ottawa Health Research Institute, Ann F. Chambers
501 Smyth Road, 7NE Department of Medical Biophysics,
Cancer Assessment Clinic, Ottawa, London Regional Cancer Program,
ON, K1H 8L6, Canada 790 Commissioners Road East,
London, Ontario, Canada, N6A 4L6
Kazuo Awai
Kumamoto University, Frédéric Charlotte
School of Medicine, Service d’Anatomie Pathologie,
Department of Radiology, GH La Pitié-Salpetrière,
1-1-1 Honjo, Kumamoto 860, Boulevard de l’hôpital, 75013 Paris,
Japan France
Yuji Baba Chiung-Nien Chen
Kumamoto University, School of Departments of Oncology and Internal
Medicine, Department of Radiology, Medicine, National Taiwan University
1-1-1 Honjo, Kumamoto 860, Hospital, 7, Chung-Shan South Road,
Japan Taipei, Taiwan, 10002

vii
viii Authors and Co-Authors of Volume 5

Ann-Lii Cheng Kevin C. Graham


Departments of Oncology and Internal Department of Medical Biophysics,
Medicine, National Taiwan University London Regional Cancer Program, 790
Hospital, 7, Chung-Shan South Road, Commissioners Road East, London,
Taipei, Taiwan, 10002 Ontario, Canada, N6A 4L6
Byung Ihn Choi Kenichi Harada
Department of Radiology, Seoul National Department of Human Pathology,
University College of Medicine 28, Kanazawa University Graduate School
Yongon dong Chongno-gu, Seoul, of Medicine, Takaramachi 13-1,
110-744, Korea Kanazawa 920-8640,
Japan
Eva Compérat
Service d’Anatomie Pathologie, Michiyo Higashi
GH La Pitié-Salpetrière, Boulevard Department of Human Pathology,
de l’hôpital, 75013 Paris, Field of Oncology, Kagoshima University
France Graduate School of Medical and
Dental Sciences, 8-35-1, Sakuragaoka,
N. Davies Kagoshima, 890-8544,
Royal Free Hospital, Liver Japan
Transplantation and Hepatobiliary
Medicine, Pond Street, Bert Hildebrandt
London NW3 2QG, UK Campus Virchow-Klinikum,
Charite-Universitatmedizin Berlin,
Timm Denecke Klinik fur Strahlenheilkonde und PET
Campus Virchow-Klinikum, Zentrum Berlin, Augustenburger Platz 1,
Charite-Universitatmedizin Berlin, Berlin 13353, Germany
Klinik fur Strahlenheilkonde und PET
Zentrum Berlin, Augustenburger Platz 1, Seung-Mo Hong
Berlin 13353, Department of Surgery and Cancer
Germany Biology, Vanderbilt University Medical
Center, 1255 Light Hall, 2215 Garland
Yuman Fong Avenue, Nashville, TN 37232,
Department of Surgery, Memorial USA
Sloan-Kettering Cancer Center, Weill
Cornell Medical Center 1275 York Avenue, Chiun Hsu
New York, NY 10021, USA Departments of Oncology and Internal
Medicine, National Taiwan University
Masamichi Goto Hospital, 7, Chung-Shan South Road,
Department of Human Pathology, Taipei, Taiwan, 10002
Field of Oncology, Kagoshima University
Graduate School of Medical and Damien Huglo
Dental Sciences, 8-35-1, Sakuragaoka, Service de Chirurgie Digestive
Kagoshima, 890-8544, et Transplantation, Hôpital Huriez,
Japan 59037 Lille Cedex, France
Authors and Co-Authors of Volume 5 ix

Tomoaki Ichikawa Amir H. Khandani


Department of Radiology, University Director, Nuclear Medicine Residency
of Yamanashi, 1110 Simokato, Chuo, and Fellowship Director, PET Services
Yamanhashi, 409-3898, Japan Department of Radiology UNC
School of Medicine, B113 POB,
Hiroko Ikeda CB7510 Chapel Hill,
Department of Human Pathology, NC 27599-7510
Kanazawa University Graduate School
of Medicine, Takaramachi 13-1, Se Hyung Kim
Kanazawa 920-8640, Japan Department of Radiology, Seoul National
University College of Medicine 28,
Kazuo Inui Yongon dong Chongno-gu, Seoul,
Department of Internal Medicine, 110-744, Korea
Second University Hospital, Fujita Health
University School of Medicine, 3-6-10, Shigeru Kiryu
Otobashi, Nakagawa-ku, Department of Radiology,
Nagoya 454-8509, Japan Institute of Medical Science,
University of Tokyo, 4-6-1
Masayuki Kanematsu Shirokanedai, Minato-ku, Tokyo,
Department of Radiology Services, 108-8639, Japan
Gifu University Hospital, 1-1 Yanagido,
Gifu 501-1194, Japan Yasuhiro Kuramitsu
Yamaguchi University Graduate
Koichi Kawanaka School of Medicine, Department
Kumamoto University, School of Biochemistry and Functional
of Medicine, Department of Radiology, Proteomics, 1-1-1 Minami-Kogushi,
1-1-1 Honjo, Kumamoto 860, Japan Ube, Yamaguchi 755-8505,
Kwong-Ming Kee Japan
Division of Hepatogastroenterology, James C. Lacefield
Department of Internal Medicine, Department of Medical Biophysics,
Kaohsiung Chang Gung Memorial London Regional Cancer Program,
Hospital, 123 Taipei Road, 790 Commissioners Road East, London,
Niaosung 833, Kaohsiung, Taiwan Ontario, Canada, N6A 4L6
Kengo Yoshimitsu Francesca Lodato
Department of Clinical Radiology, Royal Free Hospital, Liver
Graduate School of Medical Sciences, Transplantation and Hepatobiliary
Kyushu University, 3-1-1 Maidashi, Medicine, Pond Street, London NW3
Higashi-ku, Fukuoka 812-8582, Japan 2QG, UK
x Authors and Co-Authors of Volume 5

Enrique Lopez-Hänninen Isabella Mosberger


Campus Virchow-Klinikum, Department of Clinical Pathology,
Charite-Universitatmedizin Berlin, Klinik Medical University of Vienna,
fur Strahlenheilkonde und PET Zentrum Waehringer Guertel 18-20,
Berlin, Augustenburger Platz 1, 1090 Vienna, Austria
Berlin 13353, Germany
Thomas Moser
Sheng-Nan Lu Service de Radiologie, Pavillon Clovis
Division of Hepatogastroenterology, Vincent, Hôpital Civil, 1 Place de
Department of Internal Medicine, l’Hôpital, 6700 Strasbourg, France
Kaohsiung Chang Gung Memorial
Hospital, 123 Taipei Road, Joachim Mössner
Niaosung 833, Kaohsiung, Department of Internal Medicine II,
Taiwan University of Leipzig, Philipp-Rosenthal-
Str. 27, 04103, Leipzig,
Junji Machi Germany
405 N. Kuakini Street, Suite 601,
Honolulu, HI 96817, USA Takamichi Murakami
Department of Radiology, Osaka
Houman Mahallati University Graduate School of Medicine
Department of Surgery and Oncology, D1, 2-2, Yamadaoka, Suita-city, Osaka,
University of Calgary, Tom Baker Cancer 565 0871, Japan
Center, 1331 29th Street, N.W.,
Calgary, AB T2N 4N2, Canada Kohji Nagata
Department of Human Pathology,
Kwan Man Field of Oncology, Kagoshima
Center for the Study of Liver Disease and University Graduate School of
Department of Surgery, The University Medical and Dental Sciences,
of Hong Kong, Queen Mary Hospital, 8-35-1, Sakuragaoka, Kagoshima,
L9-55, Building 21, Sassoon Road, 890-8544, Japan
Pokfulam, Hong Kong, Republic of China
Hideji Nakamura
Vincenzo Migaleddu Division of Heptobiliary and Pancreatic
Sardinian Mediterranean Imaging Group, Medicine, Department of Internal
via Gorizia no. 11, Sassari 07100, Medicine, Hyogo College of Medicine,
Sardinia, Italy Mukogawa-cho, 1-Ishinomiya, Hyogo
663-8501, Japan
Hironao Miyoshi
Department of Internal Medicine, Second Hironobu Nakamura
University Hospital, Fujita Health Department of Radiology, Osaka
University School of Medicine, 3-6-10, University Graduate School of Medicine
Otobashi, Nakagawa-ku, D1, 2-2, Yamadaoka, Suita-city, Osaka,
Nagoya 454-8509, Japan 565 0871, Japan
Authors and Co-Authors of Volume 5 xi

Kazuyuki Nakamura Hiromitsu Onishi


Yamaguchi University Graduate School Department of Radiology, Osaka
of Medicine, Department of Biochemistry University Graduate School of Medicine
and Functional Proteomics, 1-1-1 D1, 2-2, Yamadaoka, Suita-city,
Minami-Kogushi, Ube, Yamaguchi Osaka, 565 0871, Japan
755-8505 Japan
Shinji Osada
Koichi Nakamura Department of Radiology Services,
Department of Radiology, Gifu University Hospital, 1-1 Yanagido,
Kanazawa University Graduate School of Gifu 501-1194, Japan
Medicine, Takaramachi 13-1, Kanazawa
920-8640, Japan François-René Pruvot
Service de Chirurgie Digestive et
Yasuni Nakanuma Transplantation, Hôpital Huriez,
Department of Human Pathology, 59037 Lille Cedex, France
Kanazawa University Graduate School
of Medicine, Takaramachi 13-1, Sóren R. Rafaelsen
Kanazawa 920-8640, Department of Radiology, Vejle Hospital,
Japan Kabbeltoft 25, DK-7100 Vejle, Denmark

Kevin Tak-Pan Ng Yasunori Sato


Center for the Study of Liver Disease Department of Human Pathology,
and Department of Surgery, Kanazawa University Graduate School
The University of Hong Kong, Queen of Medicine, Takaramachi 13-1,
Mary Hospital, L9-55, Building 21, Kanazawa 920-8640, Japan
Sassoon Road, Pokfulam, Hong Kong, Katharina Schmid
Republic of China Department of Clinical Pathology,
Medical University of Vienna, Waehringer
Akihiro Nishie
Guertel 18-20, 1090 Vienna, Austria
Department of Clinical Radiology,
Graduate School of Medical Sciences, Richard C. Semelka
Kyushu University, 3-1-1 Maidashi, Department of Radiology Services,
Higashi-ku, Fukuoka 812-8582, Gifu University Hospital, 1-1 Yanagido,
Japan Gifu 501-1194, Japan
Tereza S. Nogueira Hui-Chuan Sun
Department of Radiology, Hôpital de Liver Cancer Institute and Zhongshan
Hautepierre, CHU Strasbourg, Avenue Hospital, Fudan University,
Molière, Strasbourg 67000, France Shanghai 200032, China
Kuni Ohtomo Kenichi Takayasu
Department of Radiology, Institute of National Cancer Center Hospital,
Medical Science, University of Tokyo, Department of Diagnostic Radiology,
4-6-1 Shirokanedai, Minato-ku, Chuo ku, 5-1-1 Tsukiji, Tokyo 1040045,
Tokyo, 108-8639, Japan Japan
xii Authors and Co-Authors of Volume 5

Shugo Tamada Jing-Houng Wang


Department of Human Pathology, Field Division of Hepatogastroenterology,
of Oncology, Kagoshima University Department of Internal Medicine,
Graduate School of Medical and Kaohsiung Chang Gung Memorial
Dental Sciences, 8-35-1, Sakuragaoka, Hospital, 123 Taipei Road,
Kagoshima, 890-8544, Japan Niaosung 833, Kaohsiung, Taiwan
Bachir Taouli Marcus Wiedmann
NYU Medical Center, Department of Department of Internal Medicine II,
Radiology, MRI 530 First Avenue, University of Leipzig, Philipp-Rosenthal-
New York, NY, 10016, USA Str. 27, 04103, Leipzig, Germany
Yasuhiko Tomita Lauren A. Wirtzfeld
Division of Heptobiliary and Pancreatic Department of Medical Biophysics,
Medicine, Department of Internal London Regional Cancer Program, 790
Medicine, Hyogo College of Medicine, Commissioners Road East, London,
Mukogawa-cho, 1-Ishinomiya, Hyogo Ontario, Canada, N6A 4L6
663-8501, Japan
Helmut Witzigmann
Guido Torzilli Department of Surgery II, University
Chief Liver Surgery Unit, Department of Leipzig, Liebigstrasse 20a, 04103
of General Surgery 3, Instituto Clinico Leipzig, Germany
Humanitas IRCCS, Faculty of Medicine,
University of Milan, Via A. Manzoni, 56, Ching-Yee Oliver Wong
20089 Rozzano-Milano, Italy Positron Diagnostic Center and Medical
Cyclotron, Department of Nuclear
Stéphanie Truant Medicine, Michigan State University,
Service de Chirurgie Digestive et William Beaumont Hospital, 3601 West
Transplantation, Hôpital Huriez, 59037 Thirteen Mile Road, Royal Oak, MI
Lille Cedex, France 58073, USA
Po-Lin Tseng Fritz Wrba
Division of Hepatogastroenterology, Department of Clinical Pathology,
Department of Internal Medicine, Medical University of Vienna,
Kaohsiung Chang Gung Memorial Waehringer Guertel 18-20, 1090 Vienna,
Hospital, 123 Taipei Road, Niaosung 833, Austria
Kaohsiung, Taiwan
Yasuyuki Yamashita
Giuseppe Virgilio Kumamoto University, School of
Sardinian Mediterranean Imaging Group, Medicine, Department of Radiology,
via Gorizia no. 11, Sassari 07100, 1-1-1 Honjo, Kumamoto 860,
Sardinia, Italy Japan
Authors and Co-Authors of Volume 5 xiii

Suguru Yonezawa D. Yu
Department of Human Pathology, Field Royal Free Hospital, Liver
of Oncology, Kagoshima University Transplantation and Hepatobiliary
Graduate School of Medical and Medicine, Pond Street,
Dental Sciences, 8-35-1, Sakuragaoka, London NW3 2QG, UK
Kagoshima, 890-8544, Japan
Alex I. Zaika
Kenya Yoshida Department of Surgery and Cancer
Division of Heptobiliary and Pancreatic Biology, Vanderbilt University Medical
Medicine, Department of Internal Center, 1255 Light Hall, 2215 Garland
Medicine, Hyogo College of Medicine, Avenue, Nashville,
Mukogawa-cho, 1-Ishinomiya, Hyogo TN 37232, USA
663-8501, Japan
Yoh Zen
Junji Yoshino Division of Pathology,
Department of Internal Medicine, Second Kanazawa University Hospital,
University Hospital, Fujita Health Kanazawa, Japan
University School of Medicine, 3-6-10,
Otobashi, Nakagawa-ku, Nagoya
454-8509, Japan
Preface

This volume presents a detailed survey of imaging, multidetector-row computed tom-


various methodologies related to diagno- ography, helical computed tomography,
sis, therapy, and prognosis of liver cancer, ultrasonography, and power Doppler ultra-
including colorectal liver metastases and sound, for the prognosis and assessment of
biliary tract carcinomas, while the already liver cancer treatment (including HCC) and
published Volumes 1, 2, 3, and 4 detail liver metastases from colorectal cancer are
similar aspects of breast, lung, prostate, discussed in detail, as is the use of radiofre-
gastrointestinal, and colorectal cancer, quency ablation in hepatic tumors.
respectively. Approximately 50% of colorectal cancer
Surgical resection is the standard therapy (CRC) patients develop liver metastases
for resectable liver disease, resulting in during the course of their disease, and
5-year overall survival rates of 20–40%. more than 50% of patients who die of CRC
One the other hand, the median overall have liver metastases at autopsy. Regional
survival of patients with unresectable liver lymph node (RLN) involvement in patients
metastases does not exceed 18–20 months, with colorectal liver metastases is one
with a 5-year survival rate approaching of the worst prognostic factors. Recent
zero. In other words, there is virtually no studies indicate that for these patients,
long-term survival. Both resectable and combined liver resection and pedicular
unresectable liver cancers are discussed lymphadenectomy can be recommended,
in this volume. The method of selecting when RLN metastases respond to pre-
patients for resection of hepatic colorectal operative chemotherapy. In other words,
metastases is explained. The type of chem- combined hepatectomy and lymphadenec-
otherapy used for surgically unresectable tomy is justified for younger patients.
liver cancer is also presented. This volume includes explanations of liver
In the current volume, attention is also metastases from colorectal cancer using
given to the role of molecular genetics and ultrasound imaging, the methodology of
biomarkers in the initiation and progression radiofrequency ablation for unresectable
of liver cancer. The application of a vari- liver metastases from colorectal cancer,
ety of imaging modalities, including PET, and preoperative staging of patients with
SPECT, MRI, radial magnetic resonance colorectal liver metastases.

xv
xvi Preface

The hepatitis B and C viruses constitute their writings, which should build and fur-
a major global health problem. It is esti- ther the endeavors of the readers. The text
mated that 180 million people, ∼3% of is divided basically into seven subheadings
the world’s population are infected with for the convenience of the readers. It is my
these viruses (WHO). Most cases result in hope that the current volume will join the
chronic hepatitis, causing liver cirrhosis and preceding volumes of this series for assist-
HCC. Spontaneous eradication of the virus, ing in the more complete understanding of
however, does occur in ∼50% of acute globally relevant cancer syndromes. There
infections, which is associated with specific exists a tremendous public demand on
immune response to the virus. Because the scientific community to address cancer
HCC is the most frequent form of liver can- prevention, diagnosis, treatment and, hope-
cer, it is discussed extensively here. fully, cures.
Biliary tract carcinomas are not very I am grateful to the contributors for their
common tumors, but middle-aged or eld- promptness in accepting my suggestions,
erly people have poor prognosis. Although and I appreciate their dedication and dil-
palliative chemotherapy is commonly used igent work in sharing their invaluable
for these patients, there is no standard knowledge with the public through this
nor very effective therapy for advanced series. Each chapter provides unique indi-
disease. Commonly used therapy includes vidual, practical knowledge based on the
gemcitabine, mitomycin, fluoropyrimi- expertise and experience of the authors.
dines, and platinum compounds, used alone The chapters contain the most up-to-date
or in combination. Response rates with practical and theoretical information. It is
these treatments range from 10–35%, and my hope that these handbooks will assist
median overall survival times vary between in the more complete understanding of
5 and 12 months. Clinicopathological fea- at least come of the globally-encountered
tures of biliary cystic tumors are discussed cancer problems.
in this volume. The role of Mucin 4 as a I am thankful to Dr. Dawood Farahi
prognostic factor for extrahepatic bile duct and Dr. Kristie Reilly for recognizing
carcinoma is also presented. the importance of scholarship in an
As in the previous volumes of this series, institution of higher education and for
each chapter is written by distinguished, providing the resources for completing
practicing clinicians/surgeons/pathologists this project. I appreciate receiving expert
who provide detailed methodologies for help from Myrna Ortiz, Erin McNally
diagnosis and treatment of various forms and Lina Builes in preparing this volume.
of liver cancer. This volume was written
by 78 oncologists representing 16 coun- M.A. Hayat
tries. Their practical experience highlights November 2008
Introduction
M.A. Hayat

The enormous burden of liver cancer on society becomes clear by considering the fact
that approximately 625,000 new cases of this cancer are diagnosed globally each year.
Distressingly, the number of deaths is approximately the same at 598,000 per year. Liver
cancer, therefore, is the third most common cause of death from cancer. Survival rates
for liver cancer are only 3–5% globally. In the United States, 19,160 new cases of liver
cancer and 16,780 deaths were reported for 2007. The major risk factors for this cancer
include prior infection with hepatitis B and C viruses, with the former more prevalent.
Dietary exposure to fungus Aspergillus fumigatus (aflatoxins) also contributes to the
incidence of liver cancer in many parts of the world. Tobacco use is the most serious
preventable cause of cancer, as its use causes cancer of the lung, throat, mouth, liver,
pancreas, urinary bladder, stomach, kidney, as well as other types. Alcohol-induced liver
injury is another major risk factor for hepatocellular carcinoma (HCC).
In view of these devastating statistics, the urgency of deciphering the molecular mech-
anism underlying this disease, perfecting reliable diagnostic methods, understanding
risk factors, developing effective targeted drugs, improving other treatments, assessing
the effectiveness of therapies, and providing improved care for post-treatment patients,
becomes apparent. This volume provides up-to-date information on the above-mentioned
aspects of liver cancer; specifically, details of the methodologies used are included. The
other seven volumes in this series provide similar information on other types of can-
cers.
This series of handbooks has taken the unique approach of discussing cancer diagno-
sis, treatment, and prognosis in the same volume. It is pointed out that this vast subject
cannot be fully discussed by only one author. This is the primary reason for inviting a
large number of oncologists/clinicians/surgeons to write each of the eight volumes of
this series of handbooks. Another advantage of involving more than one author is to
present different points of view on a specific controversial aspect of cancer. I hope these
goals were accomplished in this and other published volumes of this series.

xvii
Contents of Volumes 1, 2, 3, and 4

Volume 1
1. Breast Cancer: An Introduction

2. Breast Cancer: Computer-Aided Detection

3. Sebaceous Carcinoma of the Breast: Clinicopathologic Features

4. Breast Cancer: Detection by In-Vivo Imaging of Angiogenesis

5. Breast and Prostate Biopsies: Use of Optimized High-Throughput


MicroRNA Expression for Diagnosis (Methodology)

6. Familial Breast Cancer: Detection of Prevalent High-Risk


Epithelial Lesions

7. Differentiation Between Benign and Malignant Papillary


Lesions of Breast: Excisional Biopsy or Stereotactic
Vacuum-Assisted Biopsy (Methodology)

8. Multicentric Breast Cancer: Sentinel Node Biopsy


as a Diagnostic Tool

9. Breast Cancer Recurrence: Role of Serum Tumor


Markers CEA and CA 15-3

10. Breast Cancer Patients Before, During or After Treatment:


Circulating Tumor Cells in Peripheral Blood Detected
by Multigene Real-Time Reverse Transcriptase-Polymerase
Chain Reaction

11. Breast Cancer Patients: Diagnostic Epigenetic


Markers in Blood

xix
xx Contents of Volumes 1, 2, 3, and 4

12. Breast Cancer Patients: Detection of Circulating Cancer


Cell-Related mRNA Markers with Membrane Array Method

13. Prediction of Metastasis and Recurrence of Breast Carcinoma:


Detection of Survivin-Expressing Circulating Cancer Cells

14. Node-Negative Breast Cancer: Predictive and Prognostic


Value of Peripheral Blood Cytokeratin-19 mRNA-Positive Cells

15. Breast and Colon Carcinomas: Detection with Plasma


CRIPTO-1

16. Breast Cancer Risk in Women with Abnormal Cytology


in Nipple Aspirate Fluid

17. Tissue Microarrays: Construction and Utilization


for Biomarker Studies

18. Systematic Validation of Breast Cancer Biomarkers


Using Tissue Microarrays: From Construction
to Image Analysis

19. Phyllodes Tumors of the Breast: The Role


of Immunohistochemistry in Diagnosis

20. Phyllodes Tumor of the Breast: Prognostic Assessment


Using Immunohistochemistry

21. Metaplastic Breast Carcinoma: Detection Using


Histology and Immunohistochemistry

22. Invasive Breast Cancer: Overexpression of HER-2 Determined


by Immunohistochemistry and Multiplex Ligation-Dependent
Probe Amplification

23. Operable Breast Cancer: Neoadjuvant Treatment


(Methodology)

24. Chemotherapy for Breast Cancer

25. Locally Advanced Breast Cancer: Role of Chemotherapy


in Improving Prognosis

26. Relevance of Dose-Intensity for Adjuvant Treatment


of Breast Cancer
Contents of Volumes 1, 2, 3, and 4 xxi

27. Advanced Breast Cancer: Treatment with Docetaxel/Epirubicin

28. Systemic Therapy for Breast Cancer: Using Toxicity


Data to Inform Decisions

29. Chemotherapy for Metastatic Breast Cancer Patients


Who Received Adjuvant Anthracyclines (An Overview)

30. Estrogen Receptor-Negative and HER-2/neu-Positive


Locally Advanced Breast Carcinoma: Therapy with
Paclitaxel and Granulocyte-Colony Stimulating Factor

31. Breast Cancer: Side Effects of Tamoxifen and Anastrozole

32. Breast Cancer: Expression of HER-2 and Epidermal


Growth Factor Receptor as Clinical Markers for Response
to Targeted Therapy

33. Young Breast Cancer Patients Undergoing


Breast-Conserving Therapy: Role of BRCA1 and BRCA2

34. Radiation Therapy for Older Women with Early Breast Cancer

35. Acute Side Effects of Radiotherapy in Breast Cancer


Patients: Role of DNA-Repair and Cell Cycle Control Genes
18
36. F-Fluorodeoxyglucose/Positron Emission Tomography
in Primary Breast Cancer: Factors Responsible for
False-Negative Results

37. Sentinel Lymph Node Surgery During Prophylactic


Mastectomy (Methodology)

38. Breast Conservation Surgery: Methods

39. Lymph Node-Negative Breast Carcinoma:


Assessment of HER-2/neu Gene Status as Prognostic Value

40. Multifocal or Multicentric Breast Cancer:


Understanding Its Impact on Management
and Treatment Outcomes

41. Are Breast Cancer Survivors at Risk for Developing


Other Cancers?
xxii Contents of Volumes 1, 2, 3, and 4

42. Distant Metastasis in Elderly Patients with


Breast Cancer: Prognosis with Nodal Status

43. Concomitant Use of Tamoxifen with Radiotherapy


Enhances Subcutaneous Breast Fibrosis in
Hypersensitive Patients

44. Malignant Phyllodes Tumor of the Breast:


Is Adjuvant Radiotherapy Necessary?

45. Locally Advanced Breast Cancer: Multidrug Resistance

46. Breast Cancer: Diagnosis of Recurrence


Using 18 F-Fluorodeoxyglucose-Positron Emission
Tomography/Computed Tomography

47. Role of Sentinel Lymph Node Biopsy in Ductal


Carcinoma In Situ: Diagnosis and Methodology

48. Breast Conservation Treatment of Early Stage Breast


Carcinoma: Risk of Cardiac Mortality

Volume 2
Part I General Methods and Overviews

1. Metabolic Transformations of Malignant Cells: An Overview

2. Detection of Recurrent Cancer by Radiological Imaging

3. Tumor Gene Therapy: Magnetic Resonance Imaging


and Magnetic Resonance Spectroscopy

4. Assessment of Gene Transfer: Magnetic Resonance Imaging


and Nuclear Medicine Techniques

5. Role of Mutations in TP53 in Cancer (An Overview)

6. Personalized Medicine for Cancer

7. Radiation Doses to Patients Using Computed Radiography,


Direct Digital Radiography and Screen-Film Radiography
Contents of Volumes 1, 2, 3, and 4 xxiii

8. Cancer Vaccines and Immune Monitoring (An Overview)

9. New Insights into the Role of Infection, Immunity, and Apoptosis


in the Genesis of the Cancer Stem Cell

10. Successful Cancer Treatment: Eradication of Cancer


Stem Cells

11. Overexposure of Patients to Ionizing Radiation: An Overview

Part II Lung Cancer

12. Lung Carcinoma

13. Extra-Pulmonary Small Cell Cancer: Diagnosis, Treatment,


and Prognosis

14. Magnetic Resonance Imaging of the Lung: Automated


Segmentation Methods

15. Peripheral Lung Lesions: Diagnosis Using Transcutaneous


Contrast-Enhanced Sonography

16. Small Pulmonary Nodules: Detection Using Multidetector-Row


Computed Tomography

17. Secondary Primary Cancer Following Chemoradiation


for Non-Small-Cell Lung Cancer

18. Advanced Non-Small Cell Lung Cancer: Second-Line


Treatment with Docetaxel

19. Non-Small Cell Lung Cancer with Brain Metastases:


Platinum-Based Chemotherapy

20. Non-Small Cell Lung Carcinoma: EGFR Gene Mutations


and Response to Gefitinib

21. Advanced Non-Small Cell Lung Carcinoma: Acquired


Resistance to Gefitinib

22. Prognostic Significance of [18F]-Fluorodeoxyglucose Uptake


on Positron Emission Tomography in Patients with Pathological
Stage I Lung Adenocarcinoma
xxiv Contents of Volumes 1, 2, 3, and 4

23. Non-Small Cell Lung Cancer: Prognosis Using


the TNM Staging System

24. Differentiation Between Malignant and Benign Pleural Effusions:


Methylation Specific Polymerase Chain Reaction Analysis

25. Pathological Distinction of Pulmonary Large Cell Neuroendocrine


Carcinoma from Small-Cell Lung Carcinoma Using
Immunohistochemistry

26. Differentiating Between Pleuropulmonary Desmoid Tumors


and Solitary Fibrous Tumors: Role of Histology and
Immunohistochemistry

27. Non-Small Cell Lung Cancer with Brain Metastasis:


Role of Epidermal Growth Factor Receptor Gene Mutation

Part III Prostate Cancer

28. Prostate Carcinoma

29. The Role of Intermediary Metabolism and Molecular Genetics


in Prostate Cancer

30. Array-Based Comparative Genomic Hybridization in


Prostate Cancer: Research and Clinical Applications

31. Prostate Cancer: Role of Vav3 Overexpression in


Development and Progression

32. Prostate Cancer: Detection and Monitoring Using Mitochondrial


Mutations as a Biomarker

33. Prognostic Markers in Prostate Carcinoma

34. Prostate Cancer: Detection of Free Tumor-Specific DNA


in Blood and Bone Marrow

35. Prostate Carcinoma: Evaluation Using Transrectal Sonography

36. Prostate Cancer: 16β-[18F]Fluoro-5α-Dihydrotesterone(FDHT)


Whole-Body Positron Emission Tomography

37. Effects of Standard Treatments on the Immune


Response to Prostate Cancer
Contents of Volumes 1, 2, 3, and 4 xxv

38. Vinorelbine, Doxorubicin, and Prednisone


in Hormone Refractory Prostate Cancer

39. Locally Advanced Prostate Cancer Biochemical Recurrence


After Radiotherapy: Use of Cyclic Androgen Withdrawal
Therapy

Volume 3

Part I Gastrointestinal Cancers

1. Introduction: Gastrointestinal Cancer

2. Metastatic Gastrointestinal Cancer: Safety of Cisplatin


Combined with Continuous 5-FU Versus Bolus 5-FU
and Leucovorin (Methodology)

3. Gastrointestinal Cancer: Endoscopic Submucosal


Dissection (Methodology)

4. Gastrointestinal Epithelial Neoplasms: Endoscopic


Submucosal Dissection (Methodology)

5. Inoperable Abdomino-Pelvic Tumors: Treatment


with Radio-Frequency Ablation and Surgical Debulking

6. Gastrointestinal Neuroendocrine Tumors:


Diagnosis Using Gastrin Receptor Scintigraphy

Part II Esophageal Cancer

7. Distal Esophagus: Evaluation with 18F-FDG PET/CT


Fusion Imaging

8. Endoscopic Ultrasound and Staging of Esophageal Cancer

9. Esophageal Cancer: Role of RNASEN Protein


and microRNA in Prognosis

10. Esophageal Cancer: Initial Staging


xxvi Contents of Volumes 1, 2, 3, and 4

Part III Gastric Cancer

11. Automated Disease Classification of Colon and Gastric


Histological Samples Based on Digital Microscopy
and Advanced Image Analysis

12. Early Gastric Cancer: Prediction of Metachronous


Recurrence Using Endoscopic Submucosal Dissection
(Methodology)

13. Helicobacter pylori-Infected Neoplastic Gastric Epithelium:


Expression of MUC2 as a Biomarker

14. Gastric Cancer: Role of Intestinal Metaplasia


by Histochemical Detection Using Biopsy Specimens

15. Gastric Cancer: Antitumor Activity of RUNX3

16. Early Gastric Cancer: Laparoscopic Gastrectomy (Methodology)

17. Gastric Cancer: Overexpression of Hypoxia-Inducible


Factor 1 as a Prognostic Factor

Part IV Pancreatic Cancer

18. Pancreatic Cancer: Hepatoma-Derived Growth Factor


as a Prognostic Factor

19. Pancreatic Cancer: 18F-Fluorodeoxyglucose Positron


Emission Tomography as a Prognostic Parameter

20. Imaging and Pathologic Findings of Peculiar Histologic


Variants of Pancreatic Endocrine Tumors

21. Periampullary Adenocarcinoma: Diagnosis and Survival


After Pancreaticoduodenectomy

22. Unresectable Locally Advanced Pancreatic Cancer:


Concurrent Chemotherapy

Index
Contents of Volumes 1, 2, 3, and 4 xxvii

Volume 4
Part I Colorectal Cancer

1. Introduction: Colorectal Cancer

2. Poorly Differentiated Colorectal Adenocarcinoma: (Methodology)

3. Colorectal Cancer: Immunohistochemical Diagnosis with Heterogenous


Nuclear Ribonucleoprotein K

4. Metastases and Recurrence of Colorectal Cancer: Diagnostic Role


of Immunoscintigraphy

5. Colorectal Cancer Diagnosis Using DNA Levels in Blood and Stool

6. Colorectal Carcinoma: Identification of MicroRNAs Using Real-Time


Polymerase Chain Reaction

7. Colorectal Cancer: Optimization of the Combination of 5-Flouroracil


and Irinotecan

8. Detection of Abdominal Abscesses After Colorectal Surgery:


Ultrasonography, Computed Tomography, and Gallium Scan

9. Antimetastatic Therapy in Colorectal Cancer: Role of Tumor Cell


Matrix Metalloproteinase 9 (Methodology)

10. Endoscopic Resection of Early Colorectal Tumours: Novel Diagnostic


and Therapeutic Techniques

11. Role of Stromal Variables in Development and Progression of Colorectal


Cancer

12. Quantitative Assessment of Colorectal Cancer Perfusion: Perfusion


Computed Tomography and Dynamic Contrast-Enhanced Magnetic
Resonance Imaging

13. Colorectal Cancer: Positron Emission Tomography

14. Prognostic Significance of Protein Markers in Colorectal Cancer


Stratified by Mismatch Repair Status

15. Colorectal Cancer: Lactate Dehydrogenase (LDH) Activity


as a Prognostic Marker
xxviii Contents of Volumes 1, 2, 3, and 4

Part II Colon Cancer

16. Detection of Tumor Cells in Lymph Nodes of Colon Cancer Patients Using
Real-Time Quantitative Reverse Transcription-Polymerase Chain Reaction

17. Colon Cancer: Laparoscopic Surgery

18. Sentinal Node-Based Immunotherapy of Colon Cancer

Part III Rectal Cancer

19. Rectal Cancer: Preoperative Staging Using Endorectal Ultrasonography


(Methodology)

20. Rectal Cancer: Spectral Imaging and Immunohistochemistry


of Thymidylate Synthase

21. Cancer of the Rectum: Abdominoperineal and Sphincter-Saving Resections

22. Chemoradiation for Rectal Cancer

23. Resectable Rectal Cancer: Preoperative Short-Course Radiation

24. Preoperative Chemoradiotherapy Allows for Local Control in


Rectal Cancer, but Distant Metastases Remain an Unsolved Problem

25. Locally Advanced Rectal Cancer: Combined Chemotherapy During


Preoperative Radiation Therapy

Part IV Colorectal Liver Metastases

26. Colorectal Cancer Liver Metastases: Neoadjuvant Therapy


with Bevacizumab

27. Colorectal Liver Metastases: Radiofrequency Ablation

Part V Anal Cancer

28. Anal Squamous Cell Carcinomas: Diagnosis Using p63


Immunohistochemistry

29. Anorectal Melanoma: Prediction of Outcome Based


on Molecular and Clinicopathologic Features
Contents

Authors and Co-Authors of Volume 5 .................................................................. vii

Preface ..................................................................................................................... xv

Introduction ............................................................................................................ xvii

Contents of Volumes 1, 2, 3, and 4 ........................................................................ xix

Part I Liver Cancer


A. Diagnosis
1. Applications of Positron Emission Tomography
in Liver Imaging: An Overview ..................................................................... 5
Amir H. Khandani
Introduction ................................................................................................... 5
Metastatic Liver Disease ............................................................................... 7
Cholangiocarcinoma ..................................................................................... 10
Gall Bladder Carcinoma ............................................................................... 11
Hepatocellular Carcinoma ............................................................................ 12
Therapy Monitoring ...................................................................................... 14
References ..................................................................................................... 15

2. Localized Fibrous Tumor of the Liver: Imaging Features .......................... 17


Thomas Moser and Tereza S. Nogueira
References ..................................................................................................... 19

xxix
xxx Contents

3. A Radial Magnetic Resonance Imaging Method


for Imaging Abdominal Neoplasms ............................................................... 21
Maria I. Altbach
Introduction ................................................................................................... 21
Techniques for T2-Weighted Imaging .......................................................... 22
Lesion Characterization with T2-Weighted Imaging .................................... 22
Radial Magnetic Resonance Imaging Methods:
An Alternative for Reducing Motion Artifacts
and Improving Image Quality ................................................................... 24
Radial Fast Spin-Echo, A New Alternative for T2
Mapping of the Liver ................................................................................ 27
Conclusions ................................................................................................... 30
References ..................................................................................................... 30

4. Liver: Helical Computed Tomography


and Magnetic Resonance Imaging................................................................. 33
Yuji Baba, Yasuyuki Yamashita, Kazuo Awai,
and Koichi Kawanaka
Introduction ................................................................................................... 33
Dynamic Computed Tomography Using Multi-Detector
Computed Tomography............................................................................. 33
Tumors Arising in Liver Cirrhosis ................................................................ 35
Tumor Arising in Noncirrhotic Liver or in Oncologic Patients .................... 36
Computed Tomography Angiography .......................................................... 36
Angiography-Assisted Computed Tomography........................................ 36
Computed Tomography During Arterial Portography .................................. 37
Computed Tomography During Hepatic Angiography ................................. 39
Magnetic Resonance Imaging ....................................................................... 39
Dynamic Magnetic Resonance Imaging ....................................................... 39
Superparamagnetic Iron Oxide ..................................................................... 40
References ..................................................................................................... 42

Part II Resectable Liver Cancer


A. Diagnosis
5. Selection of Patients for Resection of Hepatic Colorectal Metastases:
18F-Fluorodeoxyglucose/Positron Emission Tomography .......................... 49
Rebecca Auer and Yuman Fong
Introduction ................................................................................................... 49
Positron Emission Tomography Scanning as a Staging
Modality to Complement Conventional Imaging ..................................... 50
Positron Emission Tomography Scan for the Detection
of Extrahepatic Disease............................................................................. 50
Contents xxxi

Positron Emission Tomography Scan


for the Detection of Intrahepatic Disease.................................................. 51
Value of Positron Emission Tomography Correlated
to Prognostic Clinical Risk Score (CRS) .................................................. 52
Positive Impact of Positron Emission Tomography
on the Management of Patients with Liver Metastases ............................. 53
Negative Impact of Positron Emission Tomography
on the Management of Patients with Liver Metastases ............................. 54
Outcome of Patients Selected for Hepatic Resection Following
[18F]Fluorodeoxyglucose-Positron Emission Tomography
(FDG-PET) ............................................................................................... 54
Standard Uptake Value and Predicting Prognosis
or Response to Therapy ............................................................................ 55
Positron Emission Tomography Scanning
for Surveillance and Follow-Up ................................................................ 57
The Role of Positron Emission Tomography
in Evaluating an Elevated Carcinoembryonic Antigen (CEA) ................. 57
The Role of Positron Emission Tomography
in Follow-Up Post Hepatic Resection ....................................................... 58
Algorithm to Include Positron Emission Tomography
Scanning in Work-Up of Hepatic Colorectal Metastases.......................... 58
Conclusion .................................................................................................... 59
References ..................................................................................................... 59

B. Treatment
6. Ultrasonography During Liver Surgery ....................................................... 63
Guido Torzilli
Introduction ................................................................................................... 63
Technical Aspects ..................................................................................... 63
Liver Exploration ...................................................................................... 64
Resection Guidance ...................................................................................... 69
Hepatocellular Carcinoma ........................................................................ 69
Liver Metastases ....................................................................................... 73
References ..................................................................................................... 74

Part III Unresectable Liver Cancer


A. Treatment
7. Intraoperative Magnetic Resonance Imaging
for Radiofrequency Ablation of Hepatic Tumors......................................... 81
Oliver F. Bathe and Houman Mahallati
Introduction ................................................................................................... 81
Radiofrequency Ablation as a Treatment for Liver Tumors ......................... 81
xxxii Contents

Principles of Radiofrequency Ablation ..................................................... 81


Indications ................................................................................................. 83
Outcomes Associated with Radiofrequency Ablation .............................. 84
Alternative Ablative Techniques ............................................................... 85
Problems Associated with Monitoring the Ablative Endpoint ..................... 86
Impedence and Temperature ..................................................................... 86
Ultrasound ................................................................................................. 86
Defining the Extent of Thermal Injury by Magnetic
Resonance Imaging ................................................................................... 86
Magnetic Resonance Imaging Characteristics of Thermal Injury ............ 86
Enhancing the Definition of Extent of Thermal Injury ............................. 88
Potential Utility of Functional Magnetic Resonance Imaging.................. 89
Intraopertive Magnetic Resonance Imaging as an Adjunct
to Radiofrequency Ablation ...................................................................... 90
Rationale ................................................................................................... 90
Technical Developments ........................................................................... 90
Potential Indications.................................................................................. 92
Conduct of Intraoperative Magnetic Resonance Imaging
for Radiofrequency Ablation ................................................................ 92
Future Developments ................................................................................ 96
References ..................................................................................................... 98

8. Surgically Unresectable and Chemotherapy-Refractory


Metastatic Liver Carcinoma: Treatment with Yttrium-90
Microsphere Followed by Assessment with Positron
Emission Tomography .................................................................................... 103
Ching-Yee Oliver Wong
Introduction ................................................................................................... 103
Assessment Using Positron Emission Tomography...................................... 104
Y-90 Microsphere Radioembolization .......................................................... 105
Clinical Results ............................................................................................. 109
Illustrations ................................................................................................... 111
References ..................................................................................................... 112

B. Prognosis
9. Unresectable Liver Metastases from Colorectal Cancer:
Methodology and Prognosis with Radiofrequency Ablation ...................... 117
Junji Machi
Introduction ................................................................................................... 117
Methodology of Radiofrequency Ablation ................................................... 118
Prognosis Using Radiofrequency Ablation: Our Study ................................ 120
Patients and Methods ................................................................................ 120
Contents xxxiii

Unresectability of Tumors ........................................................................ 120


Preoperative and Intraoperative Evaluation .............................................. 121
Radiofrequency Ablation Methods ........................................................... 121
Postoperative Follow-Up........................................................................... 122
Statistical Analyses ................................................................................... 122
Short-Term Results ................................................................................... 123
Long-Term Results.................................................................................... 123
Current Role of Radiofrequency Ablation .................................................... 124
Future Perspective ......................................................................................... 128
References ..................................................................................................... 129

Part IV Hepatocellular Carcinoma


A. Diagnosis
10. Screening with Ultrasonography of Patients
at High-Risk for Hepatocellular Carcinoma:
Thrombocytopenia as a Valid Surrogate of Cirrhosis ............................... 137
Sheng-Nan Lu, Jing-Houng Wang, Kwong-Ming Kee,
and Po-Lin Tseng
Introduction ................................................................................................. 137
Epidemiology of Hepatocellular Carcinoma and Liver Cirrhosis .......... 137
Benefit of HCC Screening ...................................................................... 138
Surrogate Tests for Liver Cirrhosis and Fibrosis .................................... 139
Ultrasonographic HCC Screening on the Thrombocytopenic Adult ...... 141
References ................................................................................................... 142

11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography .................. 145


Byung Ihn Choi and Se Hyung Kim
Introduction ................................................................................................. 145
Physics of Microbubbles ............................................................................. 146
Contrast-Enhanced Color Doppler Sonography ......................................... 146
Contrast-Enhanced Power Doppler Sonography ........................................ 147
Contrast-Enhanced Harmonic Power Doppler Sonography ....................... 147
Contrast-Enhanced Pulse-Inversion Harmonic Sonography ....................... 148
Contrast-Enhanced Coded Harmonic Sonography ..................................... 150
Contrast-Enhanced Agent Detection Imaging ............................................ 151
Low Mechanical Index Imaging ................................................................. 153
Contrast-Enhanced Sonography in Assessing
the Therapeutic Response of Hepatocellular Carcinomas ...................... 153
References ................................................................................................... 156
xxxiv Contents

12. Focal Liver Lesion: Nonlinear Contrast-Enhanced


Ultrasound Imaging ...................................................................................... 159
Vincenzo Migaleddu and Giuseppe Virgilio
Introduction ................................................................................................. 159
Harmonic Imaging: Basic Principles .......................................................... 160
Physical and Technological Background .................................................... 162
Nonlinear Contrast Enhancement of Focal Liver Lesions .......................... 163
(A) Characterization of Focal Liver Lesions .......................................... 163
(B) Detection of Focal Liver Lesions...................................................... 171
(C) Monitoring of Percutaneous Ablative Treatment ............................. 174
Conclusion .................................................................................................. 176
References ................................................................................................... 177

13. Hepatocellular Carcinoma: Magnetic Resonance Imaging ...................... 183


Bachir Taouli
Introduction ................................................................................................. 183
Magnetic Resonance Imaging Technique ................................................... 183
Magnetic Resonance Imaging Appearance
of Hepatocellular Carcinoma .................................................................. 184
Gadolinium Enhancement of Hepatocellular Carcinoma ........................... 186
Magnetic Resonance Imaging Tissue-Specific Contrast Agents ................ 187
Combined Use of Superparamagnetic
Iron Oxide and Gadolinium .................................................................... 187
Atypical Appearances of Hepatocellular Carcinoma .................................. 188
New Perspectives ........................................................................................ 189
References ................................................................................................... 190

14. Expression of Vascular Endothelial Growth Factor


in Hepatocellular Carcinoma: Correlation
with Radiologic Findings .............................................................................. 193
Masayuki Kanematsu, Richard C. Semelka, and Shinji Osada
Introduction ................................................................................................... 193
History of Radiologic Diagnosis of Hepatocellular Carcinoma ................... 194
Development of Hepatocellular Carcinoma, Its Vasculature,
and Vascular Endothelial Growth Factor Expression ............................... 196
Correlation of Vascular Endothelial Growth Factor Expression
and Radiologic Findings in Hepatocellular Carcinoma
in Prior Reports ......................................................................................... 199
Our Previous Research with Immunohistochemistry.................................... 200
Magnetic Resonance and Computed Tomographic
Data Correlated to Vascular Endothelial Growth
Factor Measured Using Western Blotting ................................................. 202
Summary ....................................................................................................... 209
References ..................................................................................................... 210
Contents xxxv

15. Detection of Small Hepatic Lesions:


Superparamagnetic Oxide-Enhanced
Diffusion-Weighted T2 FSE Imaging .......................................................... 213
Shigeru Kiryu and Kuni Ohtomo
Introduction ................................................................................................. 213
Superparamagnetic Iron Oxide-Enhanced
Magnetic Resonance Imaging ................................................................. 213
Diffusion-Weighted Imaging for Suppression
of Signals from Hepatic Vessels ............................................................. 214
Periodically Rotated Overlapping Parallel Lines
with Enhanced Reconstruction Technique .............................................. 215
SPIO-Enhanced DWI T2 FSE Imaging Using Propeller ............................ 216
References ................................................................................................... 218

16. Diagnosis of Hepatocellular Carcinoma:


Multidetector-Row Computed Tomography
and Magnetic Resonance Imaging............................................................... 221
Hiromitsu Onishi, Takamichi Murakami, and Hironobu Nakamura
Introduction ................................................................................................. 221
Multidetector-Row Helical Computed Tomography................................... 221
Multidetector-Row Helical Computed Tomography
Scanning Technique ................................................................................ 222
Contrast Administration for the Dynamic
Multidetector-Row Helical Computed Tomography Study .................... 222
Administration Dose of Contrast Material.............................................. 222
Iodine Concentration of Contrast Material ............................................. 223
Injection Flow Rate and Duration of Contrast Material ......................... 223
Optimal Scanning Delay ............................................................................. 223
Scanning Timing ......................................................................................... 224
Image Processing ........................................................................................ 224
Magnetic Resonance Imaging ..................................................................... 225
Magnetic Resonance Scanning Technique.................................................. 225
Contrast-Enhanced Dynamic Magnetic Resonance Imaging ..................... 226
Tissue-Specific Contrast-Enhanced Magnetic Resonance Imaging............ 226
Computed Tomography Imaging Features
of Hepatocellular Carcinoma .................................................................. 227
Computed Tomographic Angiography........................................................ 228
Magnetic Resonance Imaging Features
of Hepatocellular Carcinoma .................................................................. 229
Superparamagnetic Iron Oxide-Enhanced Magnetic
Resonance Imaging Feature of Hepatocellular Carcinoma .................... 230
Staging of Hepatocellular Carcinoma ......................................................... 231
Accuracy for the Diagnosis of Hepatocellular Carcinoma ........................ 232
xxxvi Contents

Hepatocellular Carcinoma After Transcatherter


Arterial Chemoembolization................................................................... 232
Hepatocellular Carcinoma After Percutaneous
Radiofrequency Ablation Therapy .......................................................... 232
Limitations and Prospects ........................................................................... 233
Summary ..................................................................................................... 233
References ................................................................................................... 233

17. Hepatocellular Carcinoma: Effect of Injection


Rate/Injection Duration of Contrast Material
on Computed Tomography........................................................................... 237
Tomoaki Ichikawa and Tsutomu Araki
Introduction ................................................................................................. 237
Fixed Injection Rate and Injection Duration of Contrast Material ............. 237
Fixed Injection Rate of Contrast Material .............................................. 238
Fixed Injection Duration of Contrast Material........................................ 239
References ................................................................................................... 239

18. Detection of Combined Hepatocellular


and Cholangiocarcinomas: Enhanced Computed Tomography ............... 241
Akihiro Nishie and Kengo Yoshimitsu
Introduction ................................................................................................. 241
Pathogenesis ................................................................................................ 242
Enhanced Computed Tomographic Findings .............................................. 243
References ................................................................................................... 247

19. Hepatocellular Carcinoma and Adenomatous


Hyperplasia (Dysplastic Nodules): Dynamic Computed
Tomography and a Combination of Computed Tomography
and Angiography ........................................................................................... 249
Kenichi Takayasu
Introduction ................................................................................................. 249
Classification of Nodular Hepatocellular Lesions ...................................... 249
Multistep Progression of Hepatocarcinogenesis ......................................... 250
Needle Biopsy ............................................................................................. 252
Methods of Multidetector Computed Tomography .................................... 253
Methods of a Combination of Computed
Tomography and Angiography ............................................................... 253
CT Images of Advanced Hepatocellular Carcinoma .................................. 253
CT Images of Early Hepatocellular Carcinoma .......................................... 254
Computed Tomography Images of Adenomatous
Hyperplasia and Atypical Adenomatous Hyperplasia ............................ 255
Computed Tomography Images of Nodule-in-Nodule
Hepatocellular Carcinoma ...................................................................... 255
Contents xxxvii

Natural Outcome of Hypo-Attenuating Nodular Lesions ........................... 256


Critical Consideration to Treat Hypoattenuating Lesions........................... 257
References ................................................................................................... 257

20. Hepatocellular Cancer in Cirrhotic Patients:


Radiological Imaging .................................................................................... 261
Francesca Lodato, N. Davies, D. Yu, and Andrew K. Burroughs
Introduction ................................................................................................. 261
The EASL Consensus Statements and AASLD Guidelines ....................... 262
Surveillance for HCC: Radiological Techniques ........................................ 263
Ultrasonography (US), Doppler-Ultrasonography,
Power Doppler-Ultrasonography and
Contrast-Enhanced Ultrasonogrpahy ...................................................... 264
Spiral Computed Tomography .................................................................... 265
Magnetic Resonance Imaging ..................................................................... 267
The Problem of Small Nodules ................................................................... 268
Conclusions ................................................................................................. 270
References ................................................................................................... 271

B. Treatment
21. Treatment of Hepatocellular Carcinoma
with Thalidomide: Assessment with Power Doppler Ultrasound............. 277
Chiun Hsu, Chiung-Nien Chen, and Ann-Lii Cheng
Summary ..................................................................................................... 277
Introduction ................................................................................................. 277
Evaluation of Tumor Vascularity Using Power
Doppler Sonography ............................................................................... 279
Evaluation of Vascular Response of HCC to Thalidomide
by Power Doppler Ultrasound: A Prospective Study .............................. 279
Future Perspectives of Imaging Studies for Evaluation
of Anti-angiogenesis Therapy ................................................................. 282
References ................................................................................................... 284

22. Perfusion Scintigraphy with Integrated Single


Photon Emission Computed Tomography/Computed
Tomography in the Management of Transarterial
Treatment of Hepatic Malignancies ............................................................ 287
Timm Denecke, Bert Hildebrandt, and Enrique Lopez-Hänninen
Introduction ................................................................................................. 287
Current Status of Hepatic Arterial Chemotherapy
and Radioembolization ........................................................................... 287
Intraarterial Chemotherapy ..................................................................... 287
Transarterial Radioembolization ............................................................. 289
xxxviii Contents

Technique of Transarterial Treatment ......................................................... 289


Hepatic Arterial Infusion Chemotherapy ................................................ 289
Radioembolization .................................................................................. 290
Visualization of Perfusion Territories of Hepatic
Intraarterial Catheters in Planning and Control
of Transarterial Treatment................................................................... 291
Imaging Techniques ................................................................................ 291
Single Photon Emission Computed Tomography
with Integrated Computed Tomography for Port
Perfusion Scintigraphy ........................................................................ 291
Image Analysis........................................................................................ 293
Therapeutic Consequences...................................................................... 297
Discussion ................................................................................................... 298
References ................................................................................................... 300

23. Postoperative Interferon Alpha Treatment of Patients


with Hepatocellular Carcinoma: Expression of p48
Using Tissue Microarray .............................................................................. 303
Hui-Chuan Sun
Introduction ................................................................................................. 303
Materials and Methods............................................................................ 304
Tissue Microarray and Immunohistochemistry ...................................... 304
Scoring of P48 Immunohistochemistry .................................................. 304
Statistical Analysis .................................................................................. 305
Results ......................................................................................................... 305
Clinicopathological Data ........................................................................ 305
Survival ................................................................................................... 306
Prognostic Factors for Disease-Free Survival
and Overall Survival in Group 1 ......................................................... 306
Discussion ................................................................................................... 307
References ................................................................................................... 309

C. Prognosis
24. Hepatocellular Carcinoma: Overexpression
of Homeoprotein Six 1 as a Marker for Predicting Survival ..................... 313
Kevin Tak-Pan Ng and Kwan Man
Introduction ................................................................................................. 313
Materials and Methods................................................................................ 314
Cell Lines ................................................................................................ 314
Clinical Samples ..................................................................................... 314
Contents xxxix

Reverse Transcription-Polymerase Chain Reaction................................ 314


Western Blot............................................................................................ 315
Statistical Analysis .................................................................................. 315
Results ......................................................................................................... 315
Six1 Expression in HCC Cell Lines........................................................ 315
Six1 Expression in HCC Patients and Normal Donors ........................... 317
Six1 Protein Expression Correlated with Advanced Tumor Stage ......... 317
Six1 Protein Expression Correlated with Poor Survival ......................... 318
Discussion ................................................................................................... 319
References ................................................................................................... 322

25. Hepatocellular Carcinoma:


KiSS-1 Overexpression as a Prognostic Factor .......................................... 325
Katharina Schmid, Isabella Mosberger, and Fritz Wrba
Introduction ................................................................................................. 325
Materials ..................................................................................................... 327
Methods....................................................................................................... 327
Tissue Preparation ................................................................................... 327
Immunohistochemistry ........................................................................... 328
Evaluation of Immunohistochemical Result ........................................... 328
Results and Discussion ............................................................................... 328
Patients and Tissue Samples ................................................................... 328
References ................................................................................................... 330

26. Hepatocellular Carcinoma: Prognosis


Using Hepatoma-Derived Growth Factor
Immunohistochemistry ................................................................................. 333
Hideji Nakamura, Kenya Yoshida, and Yasuhiko Tomita
Hepatoma-Derived Growth Factor .............................................................. 333
Developmentally Regulated Expression of Hepatoma-Derived
Growth Factor ..................................................................................... 334
Role in Hepatocarcinogenesis ................................................................. 334
Role in Cancer Progression and Angiogenesis ....................................... 335
Immunohistochemical and Analytical Methods ......................................... 336
Materials ................................................................................................. 336
Method .................................................................................................... 337
Evaluation of Hepatoma-Derived Growth Factor
Expression in Hepatocellular Carcinoma................................................ 337
Prognostic Significance of Hepatoma-Derived
Growth Factor in Hepatocellular Carcinoma .......................................... 338
References ................................................................................................... 340
xl Contents

27. Hepatitis C Virus-Related Human


Hepatocellular Carcinoma: Predictive Markers
Using Proteomic Analysis (Methodology) ................................................... 343
Yasuhiro Kuramitsu and Kazuyuki Nakamura
Hepatitis C Virus-Related Human Hepatocellular
Carcinoma (HCV–HCC)......................................................................... 343
Proteomics................................................................................................... 343
Proteomics for Hepatocellular Carcinoma Tissues ..................................... 344
Proteomics for Sera from Hepatocellular Carcinoma Patients ................... 344
Proteomics for Hepatitis C Virus-Related Hepatocellular
Carcinoma Tissues .................................................................................. 345
Preparation of HCV–HCC Tissue Samples ............................................ 345
Two-Dimensional Gel Electrophoresis (2-DE) ....................................... 345
Sodium Dodecyl Sulfate-Polyacrilamide Gel
Electrophoresis (SDS-PAGE) ............................................................. 345
Image Analysis of the Gels ..................................................................... 346
Mass Spectrometry Analysis................................................................... 346
Amino Acid Sequencing by Liquid Chromatography–
Tandem Mass Spectrometry (LC-MS/MS) ......................................... 346
Proteins Whose Expression Was Increased in HCV–HCC Tissues ........ 346
Proteins With Decreased Expression ...................................................... 347
Proteomics for Auto-Antibodies in the Sera
of Hepatocellular Carcinoma Patients .................................................... 348
References ................................................................................................... 349

Part V Metastases
A. Diagnosis
28. Liver Metastases from Colorectal Cancer:
Ultrasound Imaging ...................................................................................... 355
Søren R. Rafaelsen
Introduction ................................................................................................. 355
Ultrasound Scanning Technique ................................................................. 356
Advantages of Diagnostic Hepatic Ultrasound ........................................... 357
Grayscale Echo Pattern ............................................................................... 357
Detection of Liver Metastases..................................................................... 358
Doppler Flow Pattern .................................................................................. 359
Ultrasound in Postoperative Follow-Up...................................................... 363
Future Potential Advancements .................................................................. 363
References ................................................................................................... 364
Contents xli

29. Preclinical Liver Metastases: Three-Dimensional


High-Frequency Ultrasound Imaging ......................................................... 369
Kevin C. Graham, Lauren A. Wirtzfeld, James C. Lacefield,
and Ann F. Chambers
Introduction ................................................................................................. 369
Method: High-Frequency Ultrasound ......................................................... 373
Three-Dimensional Imaging and Volume Calculation................................ 374
High-Frequency Ultrasound Imaging
of Preclinical Liver Metastases ............................................................... 376
Other Applications of High-Frequency Ultrasound
Imaging to Preclinical Cancer Research ................................................. 379
Future Developments – Ultrasound Contrast Agents ................................. 380
Conclusions ................................................................................................. 383
References ................................................................................................... 383

30. Colorectal Liver Metastases:


18
F-Fluorodeoxyglucose-Positron Emission Tomography ......................... 387
Stéphanie Truant, Damien Huglo, and François-René Pruvot
Introduction ................................................................................................. 387
Principle of FDG-PET and General Pitfalls................................................ 388
Colorectal Liver Metastases: Yield of 18F-Fluoro-
Deoxyglucose-Positron Emission Tomography ...................................... 389
Preoperative Staging of Patients
with Colorectal Liver Metastases ........................................................ 389
Detection of Liver Metastases................................................................. 389
Detection of Extrahepatic Metastases ..................................................... 391
Abdominal Cavity ................................................................................... 392
Extra-Abdominal Organs ........................................................................ 395
Impact of FDG-PET Findings on Patient’s Management ....................... 397
Additional Disease and Change of Management .................................... 398
Survival Impact ....................................................................................... 399
Postoperative Follow-Up After Resection of Colorectal Liver
Metastases and Rising Carcinoembryonic Antigen (CEA) ................ 400
FDG-PET for Monitoring the Response to Systemic
or Regional Therapy of Colorectal Liver Metastases ......................... 401
Anticancer Systemic Therapy ................................................................. 401
New Targeted Therapies (Monoclonal Antibodies) ................................ 402
Regional Therapies ................................................................................. 403
Hepatic Arterial Chemotherapy .............................................................. 403
Other Regional Therapies ....................................................................... 404
Conclusion .................................................................................................. 404
References ................................................................................................... 404
xlii Contents

Part VI Biliary Cancer


A. Diagnosis
31. Biliary Cystic Tumors: Clinicopathological
Features .......................................................................................................... 411
Yasuni Nakanuma, Hiroko Ikeda, Yasunori Sato,
Kenichi Harada, Koichi Nakamura, and Yoh Zen
Introduction ................................................................................................. 411
Anatomical Classification of the Biliary Tree............................................. 412
Non-Neoplastic Biliary Cystic Lesions ...................................................... 412
Peribiliary Cysts ...................................................................................... 412
Hepatic Foregut Cyst .............................................................................. 414
Biliary Hamartoma.................................................................................. 415
Biliary Cystic Neoplasm ............................................................................. 416
Hepatobiliary Cystadenoma and Cystadenocarcinoma .......................... 417
Intraductal Papillary Neoplasm of the Bile Duct (IPNB) ....................... 419
Other Types of Neoplastic Cystic Diseases ............................................ 426
References ................................................................................................... 426

32. Cholangiocarcinoma: Intraductal Sonography.......................................... 429


Kazuo Inui, Hironao Miyoshi, and Junji Yoshino
Introduction ................................................................................................. 429
Methods....................................................................................................... 429
Instruments.............................................................................................. 429
Insertion Methods ................................................................................... 430
Indications ............................................................................................... 431
Images and Clinical Utility ......................................................................... 432
Intraductal Sonography ........................................................................... 432
Three-Dimensional Intraductal Sonography ........................................... 432
Discussion ................................................................................................... 434
Conclusions ................................................................................................. 436
References ................................................................................................... 436

B. Prognosis
33. Extrahepatic Bile Duct Carcinoma: Role of the p53 Protein Family ....... 441
Alexander I. Zaika and Seung-Mo Hong
Introduction ................................................................................................. 441
Epidemiology .......................................................................................... 441
Etiology ................................................................................................... 441
Diagnosis................................................................................................. 442
Pathology ................................................................................................ 442
Prognosis ................................................................................................. 443
Contents xliii

Role of p53 in Malignant Tumors of the Bile Duct .................................... 443


Analysis of p53 Mutations ...................................................................... 444
Roles of p73 and p63 in Malignant Tumors of the Bile Duct ..................... 444
Analyses of p63 and p73 in Tumors ....................................................... 447
References ................................................................................................... 448

34. Extrahepatic Bile Duct Carcinoma:


Mucin 4, a Poor Prognostic Factor .............................................................. 451
Michiyo Higashi, Shugo Tamada, Kohji Nagata,
Masamichi Goto, and Suguru Yonezawa
Introduction ................................................................................................. 451
Mucin Characteristics ................................................................................. 451
MUC4 Mucin .............................................................................................. 452
Antibodies Against MUC4 ......................................................................... 453
MUC1 Mucin .............................................................................................. 453
Antibodies Against MUC1 ......................................................................... 455
Methods....................................................................................................... 455
Tissue Sample Management ................................................................... 455
Immunohistochemistry ............................................................................... 458
References ................................................................................................... 458

C. Treatment
35. Hilar Cholangiocarcinoma: Photodynamic
Therapy and Stenting ................................................................................... 463
Marcus Wiedmann, Joachim Mössner, and Helmut Witzigmann
Summary ..................................................................................................... 463
Introduction ................................................................................................. 463
Preoperative Biliary Drainage ..................................................................... 464
Palliative Bile Duct Stenting ....................................................................... 464
Palliative Stenting of Malignant Duodenal Obstruction ............................. 469
Photodynamic Therapy (PDT) .................................................................... 470
Mechanism of Action.............................................................................. 470
Contraindications for Photodynamic Therapy ........................................ 471
Photosensitizers....................................................................................... 472
Photodynamic Therapy for Palliation
of Hilar Cholangiocarcinoma .............................................................. 472
Photodynamic Therapy for Neoadjuvant
and Adjuvant Treatment of Hilar Cholangiocarcinoma ...................... 478
Future Directions of Photodynamic Therapy
for Hilar Cholangiocarcinoma ............................................................ 478
References ................................................................................................... 479
xliv Contents

Part VII Splenic Cancer


A. Diagnosis
36. Splenic Metastases: Diagnostic Methods .................................................... 489
Eva Compérat and Frédéric Charlotte
Introduction ................................................................................................. 489
Epidemiology .............................................................................................. 489
Pathologic Features ..................................................................................... 489
Pathogenesis ................................................................................................ 490
Clinical Features ......................................................................................... 491
Diagnostic Procedures ................................................................................ 493
Differential Diagnosis ................................................................................. 494
References ................................................................................................... 496

Index ........................................................................................................................ 499


A. Diagnosis
1
Applications of Positron Emission
Tomography in Liver Imaging:
An Overview
Amir H. Khandani

INTRODUCTION in different organs. Normal hepatocytes


express Glut2, Glut9, and Glut10 (Joost
While positron emission tomography (PET) and Thorens, 2001). Expression of Gluts,
has been used for several decades in the predominantly Glut1 and Glut3, is signifi-
research setting, its clinical use has grown cantly increased in most malignancies.
substantially in the past decade. This is a Once in the cell, glucose or FDG is
quantitative physiological imaging modal- phosphorylated by hexokinase to glucose-
ity utilizing positron emitters such as 6-phospate or FDG-6-phosphate, respec-
Fluorine-18, Oxygen-15, Nitrogen-13 and tively. Expression of hexokinase and its
Carbon-11. The fact that several of these affinity/functional activity for phosphory-
nuclides are components of common bio- lation of glucose or FDG is often higher in
logical molecules makes PET particularly cancer cells compared to normal cells; hex-
suitable to visually capture different bio- okinase II is predominantly expressed in
logic pathways; however, the short physical cancer cells. Glucose-6-phosphate travels
half-life of Oxygen-15 (2 min), Nitrogen-13 further down the glycolytic or oxidative
(10 min), and Carbon-11 (20 min) limits pathways to be metabolized, in contrast to
their utilization to the centers with an on- FDG-6-phosphate, which cannot go further,
site cyclotron. Fluorine-18 has a physical and cannot be metabolized. In normal cells,
half-life of 110 min and can be synthesized glucose-6-phospate or FDG-6-phosphate
in commercially operating cyclotrons and can be dephosphorylated and exit the cells.
transported to remote PET facilities. The In many cancer cells, however, expression
most commonly used radiotracer is 2-[18F] of glucose-6-phosphatase is often signifi-
fluoro-2-deoxy-D-glucose (FDG). cantly decreased, and therefore, glucose-
Tumor imaging with FDG is based on 6-phospate or FDG-6-phosphate is only
the principle of increased glucose metabo- minimally dephosphorylated and remains
lism of cancer cells. Like glucose, FDG in large part within the cell. On the other
is taken up by the cancer cells through hand, because FDG-6-phosphate cannot
facilitative glucose transporters (Glut). be metabolized, it is trapped in the cell as a
These transporters are glycoproteins, and polar metabolite, and can be visualized by
so far, 12 isoforms have been identified PET. In normal liver cells, the concentration

5
6 A.H. Khandani

of glucose-6-phosphatase is high, which the base of the skull through the upper
causes rapid clearance of FDG from the thighs. The CT portion (transmission scan)
liver. This may account for the mild inten- of the PET CT is acquired within seconds
sity of the normal liver on FDG-PET. while the PET acquisition time for each
Furthermore, the interindividual as well as bed position (~ 15 cm) is several minutes;
the intraindividual variation of the hepatic the total PET acquisition time in newer
uptake appears to be minimal (Rydberg machines is 10–25 min.
et al., 2005). Therefore, while comparing In addition to delivering anatomic infor-
PET scans of different patients or of the mation, the CT portion of PET CT is used
same patient at different occasions, liver to measure the attenuation of the X-ray
uptake can be used as the reference; first, photons traveling through the patient in
the PET scans to be compared should be order to produce the so-called “attenuation
displayed with the liver uptake on all the map” and correct the PET data for tissue
scans being the same, then the areas of attenuation (attenuation corrected PET).
interest can be compared to each other. During PET acquisition, photons from
The radioisotope portion of the molecule structures deep in the abdomen or pelvis
used in PET imaging emits a positron (a are attenuated more compared to those
positively charged electron), which travels from the superficial structures and the
a distance of a few millimeters in tissue chest. Therefore, the intensity of uptake
before it collides with a negatively charged in the deeper structures may be underesti-
electron. This collision annihilates the mated on the “non-attenuation corrected”
entire mass of the positron and electron into PET images. The intensity of uptake in
two photons (gamma rays) with energy of the deeper structures is normalized to the
511 KeV each. These two photons travel at intensity of uptake in the superficial struc-
the speed of light in exactly opposite direc- tures on the attenuation-corrected PET
tions (180° apart). Coincidental detection images. While correction of the PET data
of these two photons by two oppositely posi- for tissue attenuation is indispensable,
tioned detectors in the PET scanner results misalignment between PET and CT can
in images with a much higher resolution cause mislocalization of lesions on the
compared to the conventional, single pho- fused PET-CT images. This may be due
ton nuclear medicine studies and presents to changes in position of a body part (e.g.,
the possibility of quantitative measurement neck or legs) or due to physiologic changes
of the tracer uptake in a lesion of interest. in the position of an organ (i.e., respiratory
PET CT allows the fusion of the metabolic movement) between the transmission (CT)
information on PET with the anatomic and the emission scan (non-attenuation
information on CT. PET CT has been corrected PET). As the degree of misalign-
proven to increase the diagnostic accuracy ment and resulting mislocalization can be
compare to stand-alone PET. In PET CT, significant, one has to be cautious when
the patient undergoes a CT scan, followed interpreting the attenuation-corrected PET
by a PET scan without changing the patient CT images or using PET CT images for
position. PET for most oncologic indica- radiation therapy planning. The magnitude
tions (whole body PET) is acquired from of this misalignment can be assessed by
1. Applications of Positron Emission Tomography in Liver Imaging: An Overview 7

fusing the non-attenuation corrected PET squamous cell primaries, 1 of 3 biliary


images with CT, which can be performed tract primaries, and none of neuroendo-
on any PET review stations. In case of sig- crine primaries that were examined.
nificant misalignment, the non-attenuation Metastatic disease accounts for > 90% of
corrected PET images should be reviewed malignant liver lesions. Often, the presence
without fusion with CT and the metabolic of liver metastases is the main determi-
findings on PET should be correlated side nant of survival and guides the therapeutic
by side with the anatomic findings on CT. strategy, particularly in patients with color-
At its introduction, the CT portion of the ectal cancer. Among the colorectal cancer
PET CT was acquired without contrast and patients that die of the disease, 70% have
used solely for attenuation correction and liver metastases and a significant 10% have
anatomic localization of the PET findings. liver-only disease. Even in those patients
With the development of multislice CT with metastases to multiple organ sites, the
machines and their introduction to PET CT extent of liver disease remains the primary
scanners, the CT portion of the PET CT determinant of survival. Therefore, the
studies began to be performed as diagnostic timely diagnosis of the metastatic disease
CT with IV and oral contrast, as needed. to the liver is of paramount importance.
Because the density of IV and oral contrast The sensitivity of FDG-PET in detect-
used in the diagnostic CT can produce arti- ing hepatic metastases from different pri-
facts with overestimation of FDG uptake in maries is > 90%. Delbeke et al. (1998)
areas of high contrast density, the protocol studied the diagnostic value of FDG-PET
for CT has to be modified with the general in hepatic metastases and detected all 66
approach of dilution of the contrast medium. metastatic lesions originating from various
Alternatively, one can acquire a low or inter- primaries including the colon, pancreas,
mediate dose unenhanced CT first to be esophagus, sarcoma, and parotid. Similar
used as attenuation map, followed by emis- results with an overall greater sensitivity
sion scan; after which the IV enhanced CT of PET compared to spiral CT have been
should be performed. The IV enhanced CT reported by other groups (Hustinx et al.,
scan may be performed from the entire body 1998). Computed tomography can be read
or only from the area of interest in order to to achieve a higher sensitivity, but it will
limit the patient’s exposure to radiation. be at the expense of specificity, as many
CT positive lesions are false-positives.
This was shown in a study by Marom
METASTATIC LIVER DISEASE et al. (1999) in metastatic lung cancer,
where in a prospective study of 100 patients
Zimmerman et al. (2002a) studied expres- nearly twice as many lesions in the liver
sion of Glut1 in hepatic metastases origi- were identified by CT as by PET; however,
nating from different primaries, and found all of the incremental lesions identified
that Glut1 was overexpressed in hepatic by CT were false-positives. Comparing
metastases of 3 of 5 lung primaries, 7 magnetic resonance imaging (MRI) and
of 11 pancreatic primaries, 7 of 9 colon PET, Yang et al. (2003) reviewed studies
primaries, 2 of 7 breast primaries, 2 of 2 of 30 patients with histopathologically
8 A.H. Khandani

proven (# 27) or clinically suspected (# 3) detect additional hepatic metastases. This


hepatic metastases from non-hepatic pri- is of particular importance in preoperative
maries. The sensitivity, specificity, and evaluation of solitary hepatic metastasis
positive and negative predictive values on as detection of additional lesions often
MRI were 85.7%, 100%, 100%, and 89%, changes the management. In the case of
respectively, compared to 71%, 93.7%, suspected recurrent cancer based on rising
90.9%, and 79% on PET. The difference tumor markers or other parameters, FDG-
between the two methods was not sta- PET is more sensitive than CT or MRI for
tistically significant (Yang et al., 2003). discovering hepatic metastases with the
A meta-analysis of the literature on detec- potential of detecting disease earlier than
tion of hepatic metastases from colorectal, CT or MRI, so that metastatic disease is
gastric, and esophageal cancers by ultra more amenable to curative resection.
sound (US), CT, MRI, and PET revealed False-positive PET findings for malig-
that in studies with a specificity > 85%, the nancy in the liver can occur due to acute
mean weighted sensitivity was 55% (95% infection (intrahepatic abscess, penetrating
CI: 41, 68) for US, 72% (95% CI: 63, 80) gallbladder empyema) or chronic inflam-
for CT, 76% (95% CI: 57, 91) for MRI, matory processes (granulomas). Since most
and 90% (95% CI: 80, 97) for PET. Results of these conditions can be excluded by clin-
of pairwise comparison between imaging ical history and review of the transmission
modalities demonstrated a greater sensi- CT and/or available dedicated CT or MRI,
tivity of PET than US (P = .001), CT (P = a focal area of increased uptake in the liver
.017), or MRI (P = .055). The conclusion on PET that does not represent one of the
was that at an equivalent specificity, PET above conditions, has to be considered as
is the most sensitive noninvasive imag- malignant until proven otherwise. Positron
ing modality for the diagnosis of hepatic emission tomography is of particular ben-
metastases from colorectal, gastric, and efit in case of negative or indeterminate CT
esophageal cancers (Kinkel et al., 2002). or MRI findings for malignancy (Figure
In cases of known solitary hepatic metas- 1.1). Hemangiomas and adenomas of the
tasis diagnosed by CT or MRI, PET can liver are not FDG avid.

a b c

Figure 1.1. Patient with a history of ovarian cancer and rising ca-125 level. T2 (A) and postcontrast
fat-saturated T1 (B) MRI images were without any abnormal findings. PET was requested for further
evaluation and demonstrated focal uptake in the segment IV of the right hepatic lobe consistent with
metastatic disease (C, arrow head)
1. Applications of Positron Emission Tomography in Liver Imaging: An Overview 9

False-negative PET for hepatic metas- ment of the liver during the emission
tases can be due to limitations of spatial scan. The liver is an upper abdominal
resolution. It should be considered, how- organ that moves with respiratory move-
ever, that many of the PET publications ment of the diaphragm. Emission scans
were based on non-attenuation corrected are acquired over several minutes during
images, which may have lower sensitivity, which hepatic lesions are in a repetitive
especially for lesions deeper in the liver; cranio-caudal pendulous movement. The
such lesions generally appear much less respiratory excursion of the liver is
intense on non-attenuation corrected than ~ 10–30 mm. Therefore, it is conceivable
on attenuation corrected PET images. that cranial and caudal portions of small
Generally, the role of PET in detecting lesions are registered only during a portion
subcentimeter lesions should be redefined, of the acquisition time, and their uptake is
considering the higher spatial resolution underestimated, so that they appear less
and sophisticated image correction and intense on images than they really are.
reconstruction algorithms of current PET The degree of this underestimation is vari-
machines. Further improvements in image able, and particularly in the case of a sub-
resolution from currently 0.6–0.8 cm to a centimeter lesion, this may even lead to
few millimeters can be expected with the non-visualization of the lesion. One way
development of small surface area crystal to overcome this problem is to increase
elements in combination with alternative the target-to-background count ratio by
position-sensitive photomultiplier tubes, increasing the acquisition time. This can
and the implementation of depth of inter- be done by increasing the acquisition time
action measurements. in the mid- and upper-abdomen while
False-negative FDG-PET can occur also acquiring the whole body scan, if this is
due to underestimation of uptake, mislo- possible on the PET machine. Another
calization of foci as well as recent comple- solution would be to acquire a second
tion of chemotherapy. The latter is likely PET scan from mid- and upper-abdomen,
associated with significant decrease in the consisting of a transmission scan followed
number of the cells or their glucose metab- by an emission scan with longer acquisi-
olism after chemotherapy. No definitive tion time, once the whole body scan is
information is available in the literature on completed (“Liver View”). A different and
the time interval after completion of chem- preferable approach to solve this problem
otherapy during which PET can be false- is Respiratory Gating, in which case only
negative. Based on experience at large emission data collected in certain parts of
centers, this time interval is ~ 4–6 weeks the respiratory cycle are used for image
for high-grade lymphoma. However, no reconstruction, resulting in better visuali-
definite statement can be made regard- zation of small lesions with the disadvan-
ing the time interval after chemotherapy tage of longer acquisition time.
during which liver metastases may not be A changed position of the liver between
visualized on PET. the emission and transmission scan can cause
Underestimation of uptake of malignant misalignment between these two images,
lesions causing false-negative findings on resulting in mislocalization of pulmonary
PET can occur due to physiologic move- or colonic foci into the liver or vice versa.
10 A.H. Khandani

This is especially of concern when trans- mon in peripheral cholangiocarcinoma.


mission CT is acquired during full inspira- The diagnosis of cholangiocarcinoma has
tion and the PET scan is acquired during been based on clinical picture, laboratory
shallow breathing, but appears to be less values, radiologic imaging, and histol-
of a problem if the transmission CT is ogy, although the latter is often inconclu-
performed during shallow breathing or in sive in differentiating between this tumor
stand-alone PET units where a transmis- and metastatic adenocarcinoma. Work up
sion source such as Ge-68 is used for generally consists of MRI, magnetic reso-
attenuation correction. In the case of sus- nance cholangiopancreatography (MRCP),
pected misalignment, the non-attenuation CT, endoscopic retrograde cholangiopan-
corrected PET images should be reviewed creatography (ERCP), and percutaneous
for corrected localization of the lesion. transhepatic cholangiography (PTC).
Given the higher sensitivity of PET com- Overall, the prognosis of this tumor is
pared to CT or MRI in detecting hepatic dismal with 5-year survival as low as 15%;
metastasis, it is conceivable that PET will however, improved survival of 20–30% at 5
be increasingly employed for preoperative years are achievable with portal or arterial
staging of malignant tumors with poten- embolization followed by trisegmentec-
tial of hepatic metastasis. Presently, CT tomy. Therefore, preoperative assessment
with IV contrast or MRI is still needed for for hepatic and extra-hepatic metastases is
adequate evaluation of anatomic resecta- likely of prognostic value.
bility of liver metastases since an assess- Glut1 is not expressed in normal bile
ment of the venous anatomy in the liver is duct, but has been described to be strongly
critical for defining hepatic lobar anatomy. expressed in cholangiocarcinoma (Roh
However, with the availability of PET- et al., 2004; Zimmerman et al., 2002b).
multislice CT, one can expect that in the Overall, cholangiocarcinoma appears to
future, PET and contrast CT will be per- be highly FDG avid. Delbeke et al. (1998)
formed in one setting as the preoperative evaluated eight patients with cholangiocar-
imaging study of choice in patients under- cinoma: all lesions demonstrated intense
going resection of hepatic metastases. FDG uptake (Delbeke et al., 1998). Hilar
and extrahepatic cholangiocarcinoma,
however, appears to be less intense on
FDG-PET than the peripheral cholangi-
CHOLANGIOCARCINOMA ocarcinoma, which may be associated with
smaller size and/or higher mucin content
Cholangiocarcinoma originates from the of the hilar tumors compared to the periph-
epithelial cells of the biliary tract. After eral ones. Peripheral tumors accounts for
hepatocellular carcinoma, cholangiocarci- ~ 10% of all cholangiocarcinoma, and
noma is the second most common primary often has a characteristic central photo-
tumor of the liver, accounting for ~ 5–30% penia on FDG-PET, which corresponds
of the primary hepatic malignancies. Biliary to the central core of fibrotic tissue and
obstruction with jaundice is the most com- desmoplastic reaction provoked by the
mon presenting clinical feature in hilar neoplastic cells; on contrast enhanced CT
cholangiocarcinoma while it is uncom- or MRI, this is evident by early moderate
1. Applications of Positron Emission Tomography in Liver Imaging: An Overview 11

peripheral enhancement followed by pro- only ∼15% of all gallbladder carcinoma


gressive and concentric filling. cases. Patients with T1b tumors (tumor
FDG-PET is helpful in detecting unsus- invades muscularis) and beyond should
pected extra-hepatic metastatic disease be further evaluated for spread of disease
from cholangiocarcinoma. Kim et al. as T1b tumors are associated with lymph
(2003) discovered three unsuspected node involvement in 15% of cases vs.
extra-abdominal lymph nodes and three 2.5% of cases with T1a disease.
unsuspected lung metastases in a group Kim et al. (2002) examined 71 cases of
of 21 patients with this tumor. Kato et gallbladder carcinoma and demonstrated
al. (2002) reported 100% specificity for expression of Glut1 in 37 cases (52.1%).
regional nodal involvement on FDG-PET. Preoperatively, FDG-PET has been used
Especially in cases of peripheral cholan- to distinguish malignant from benign gall-
giocarcinoma, PET should be considered bladder lesions with a sensitivity of 75%
to evaluate the patient for extrahepatic (six of eight patients), and specificity of
metastases: peripheral cholangiocarcinoma 87.5% (seven of eight patients) (Koh et al.,
attains a large size before it becomes clini- 2003); however, this will likely be of lim-
cally apparent, since it does not obstruct ited clinical use, since most patients are
the central biliary system. Also, extrahe- status post cholecystectomy at the time of
patic metastases are commonly present at the diagnosis of gallbladder carcinoma.
the time of diagnosis. Also, if the histology Anderson et al. (2004) used PET to
is inconclusive in differentiating between assess possible local residual and meta-
cholangiocarcinoma and metastatic adeno- static disease after cholecystectomy. This
carcinoma from an extrahepatic primary, method detected local residual disease in
FDG-PET may help to make this dif- seven of nine cases (78%), and remote
ferentiation by detecting the extrahepatic metastases in two of three cases. Positron
primary site. emission tomography, however, detected
only three of six cases with carcinomatosis.
This imaging modality can potentially be
GALL BLADDER CARCINOMA used to assess for local residual and meta-
static disease within hours to days after
Gallbladder carcinoma is the most com- incidental detection of gallbladder carci-
mon malignant tumor of the biliary tract; noma at cholecystectomy. There has been
this is a highly fatal disease that presents some concern that non-specific uptake in
in 80–95% of cases as adenocarcinoma. the early post-surgical period may disturb
The diagnosis of gallbladder carcinoma image interpretation. Although generally
is rarely made preoperatively; in most surgical interventions can cause increased
cases, gallbladder carcinoma presents as FDG uptake in the surgical bed, there have
an incidental finding at the time of chole- been no systematic studies or publications
cystectomy for presumed benign etiol- concerning FDG uptake in a post-operative
ogy. In post-cholecystectomy setting, in setting. Additionally, one has to consider
patients with T1a (tumor invades mucosa), that the degree of local inflammatory reac-
simple cholecystectomy is regarded as tion varies between different tissues and
adequate treatment, but these represent also depends on the type of intervention.
12 A.H. Khandani

Generally, the decision to perform PET


after a surgical intervention, should strongly
take into account the clinical factors, such
as T-stage in case of gallbladder carcinoma,
and not merely the post-operative time
interval. Furthermore, the interpretation of
PET images should not be only based on the
intensity of uptake, but should also take into
account the pattern of uptake (round/local-
ized/multiple for malignancy vs. diffuse
for inflammation). Lastly, however, asses-
sing for distant metastases is likely to be
independent of the factors influencing the
FDG in the surgical bed. In author’s experi-
ence, FDG-PET is very useful for local and
distant staging of patients with gallblad-
der carcinoma detected at cholecystectomy
(Figure 1.2).

HEPATOCELLULAR
CARCINOMA
Hepatocellular carcinoma (HCC) or
hepatoma develops through malignant Figure 1.2. Patient status post laparoscopic chole-
transformation of hepatocytes and is com- cystectomy with T2 gallbladder cancer. PET was
mon in the setting of chronic liver changes requested for staging and indicated metastases in
and cirrhosis. This cancer is the most the porta hepatis, paraaortic (arrow heads) and par-
atracheal lymph nodes (arrow)
common primary epithelial malignancy of
the liver, accounting for ~ 80% of malig-
nant epithelial neoplasms of the liver. Facilitative glucose transporters do not
Hepatocellular carcinoma has been showing appear to be overexpressed in HCC as often
an increasing incidence in Western coun- as in other malignant tumors. Zimmerman
tries due to increasing frequency of hepa- et al. (2002b) and Roh et al. (2004) studied
titis B and C viral infection. The diagnosis expression of Glut1 in 35 and 22 cases of
is based on screening risk populations with HCC, respectively; the first group reported
measurements of serum AFP and liver two positive cases and the second group
ultrasound. Magnetic resonance imaging, reported one positive case. Delbeke et al.
CT, and lipiodol angiography with follow- (1998) examined a series of 23 patients
up CT are used in inconclusive cases to with HCC. In visual assessment, differ-
establish the diagnosis. Biopsy is only entiation of tumor from normal liver was
performed on patients in whom the radio- definitive in 13 patients, equivocal (mildly
logical diagnosis cannot be made. increased compared to normal liver) in
1. Applications of Positron Emission Tomography in Liver Imaging: An Overview 13

3 patients, and poor (same or less intense experience, whole body FDG-PET is very
compared to normal liver) in 7 patients. helpful to assess the malignant poten-
The sensitivity of FDG-PET for HCC is tial of a hepatic lesion of unknown pri-
~ 50–60%. There appears to be some asso- mary because FDG avidity of the lesion
ciation between histologic differentiation increases the likelihood of its malignant
of HCC and FDG uptake, with poorly dif- nature. Furthermore, simultaneous visu-
ferentiated tumors being more intense on alization of FDG avid foci outside of the
FDG-PET, likely explained by enzymology liver can be viewed as almost consistent
of HCC. Concentration of glucose-6- with malignant etiologyof the liver lesion.
phospatase is high in normal liver, causing Positron emission tomography may also
rapid clearance of glucose-6-phospate or visualize an easily accessible biopsy site
FDG-6-phosphate from hepatocytes with outside of the liver. Also in cases of known
consequent mildly intense appearance of HCC with clinically suspected extrahe-
liver on PET. The enzymology of well- patic metastasis, FDG-PET can be used
differentiated HCC resembles that of the to confirm this suspicion. Nonetheless, a
normal liver, possibly explaining mild FDG negative FDG-PET scan in such patients
uptake or non-visualization of these tumors does not exclude the possibility of HCC.
on PET. Less differentiated HCC tumors [1-11C] acetate (11-C Acetate) is a
have lower levels of glucose-6-phosphatase short-lived PET tracer with higher uptake
and higher levels of hexokinase, possi- in well-differentiated hepatomas compared
bly causing intense FDG uptake of these to poorly differentiated ones. The physical
tumors on PET (Torizuka et al., 1995). half-life of 11-C is 20 min. Several possible
Also, there appears to be some association pathways of acetate incorporation into
between FDG uptake and tumor-volume tumor metabolism have been postulated;
doubling time as well as between FDG the dominant one appears to be partici-
uptake and tumor size. Trojan et al. (1999) pation in free fatty acid synthesis. Ho
visualized all six tumors larger than 5 cm et al. (2003) studied a series of 57 patients
in a series of 14 HCC tumors. Therefore, with liver lesions, consisting of 39 cases
FDG PET could possibly be used to assess of HCC (group I), 13 cases of non-HCC
the effect of treatment in larger and less malignancies (three cases of cholangi-
differen-tiated HCC. High FDG uptake in ocarcinomas and ten cases of hepatic
HCC has been associated with overexpres- metastases from colon, breast, lung and
sion of mRNA levels for several markers, carcinoid primaries, group II) as well as
such as VEGF of aggressive tumor behavior 5 cases of benign liver lesions (2 cases of
(Lee et al., 2004). cavernous hemangioma, 2 cases of FNH
Detection of extrahepatic FDG avid and 1 case of adenoma, group III). In group
metastases originating from HCC has also I, there was a “complementing” sensitivity
been reported; especially in cases of less- for HCC on FDG and 11-C Acetate-PET,
differentiated HCC, metastases appear as these two tracers together gave a sensi-
to be more FDG avid (Ho et al., 2003; tivity of 100% for detecting HCC. Most of
Trojan et al., 1999). Although more data the well-differentiated HCC lesions were
are needed to establish the clinical role intense on 11-C Acetate scan with mild
of FDG-PET in HCC, based on author’s tracer uptake or not visualized on FDG
14 A.H. Khandani

scan, while this relationship was reversedproven to be more accurate than CT and
for poorly differentiated tumors. In groupMRI in assessing the effect of chemo-
II, all lesions showed intense FDG uptake therapy. In many malignancies such as
but no abnormal 11-C Acetate uptake. In high-grade lymphoma, early decrease of
group III, the uptake patterns were as fol-
uptake after the start of chemotherapy
lows: hemangiomas were isointense on indicates better prognosis and longer sur-
FDG and hypointense on 11-C Acetate vival. The role of FDG-PET in assessing
scan. Both FNH cases showed mild inten- the efficacy of chemotherapy in metastatic
sity uptake on 11-C Acetate scan, while liver disease, cholangiocarcinoma, and
one of them had mild and another one no other FDG avid hepatic malignancies has
visualized uptake on FDG scan. The only not yet been established. While increased
adenoma case showed no abnormal FDG intensity or number of liver foci under
or 11-C Acetate uptake. With an appar- ongoing chemotherapy indicates progres-
ently high specificity of 11-C Acetate forsion of disease, the pathologic and clini-
HCC, the authors concluded that when a cal correlates of decreased uptake in this
lesion is positive for both tracers or only
setting are not clearly understood. One
for 11-C Acetate, the likelihood of HCC has also to consider that in most cases of
is very high. On the other hand, when a metastatic disease to the liver and cholan-
liver lesion is positive only for FDG but giocarcinoma, chemotherapy has only a
is negative for 11-C Acetate, the possi- palliative effect and, therefore, although
bility of non-HCC malignancy or poorly disappearance of tumor on PET may indi-
differentiated HCC should be considered. cate effectiveness of the treatment and
In case both tracers are negative, benign better prognosis, it should not be under-
pathology is very likely. These conclu- stood as a sign of cure.
sions can be particularly helpful for eval- Due to the general assumption that tis-
uation of tumors < 2 cm in patients with sue injury causes inflammatory uptake,
low or intermediate likelihood of having FDG-PET has never been used to monitor
HCC (negative serum status for hepatitis the effect of invasive procedures such as
B or C, borderline or normal AFP). It radiofrequency ablation (RFA) of the liver.
must be considered that these very prom- However, there is no significant accumula-
ising data have not been reproduced yet tion of reactive cells at the ablation site in
in a large series and also that the tumorsthe liver after RFA (Tsuda et al., 2003)
in Asia, where this study was performed, and RFA of the liver does not appear to
may differ somewhat from the tumors cause false-positive findings on PET. This
seen in the US. is of great importance since capillaries
are leaky in the weeks and months after
RFA and, therefore, CT and MRI can-
THERAPY MONITORING not be used for several months after this
procedure to assess for residual tumor.
Effective chemotherapy is associated with In author’s experience, PET can be used
a decrease in the number and/or glucose to demonstrate complete ablation within
metabolism of cancer cells and decreased hours to days after RFA of liver metastases
FDG uptake on PET. FDG-PET has been (Figure 1.3).
1. Applications of Positron Emission Tomography in Liver Imaging: An Overview 15

a b c

Figure 1.3. Patient with solitary metastasis to the right lobe of the liver from an ocular malignant
melanoma, diagnosed on an outside PET scan (a, arrow head). PET was performed at our institution 16 h
after laparoscopic RFA and revealed complete ablation without any inflammatory uptake (b, arrow head).
This was confirmed on the 3-month follow-up PET scan (c, arrow head)

In conclusion, FDG-PET has an impor- Einstein College of Medicine, for critical


tant role in determining if there are metas- review of the manuscript.
tases to the liver and whether disease has
spread beyond the liver. This information REFERENCES
is critical for planning surgical resections
Anderson, C.D., Rice, M.H., Pinson, C.W.,
of liver metastases. While FDG-PET can Chapman, W.C., Chari, R.S., and Delbeke, D.
fail to detect many well differentiated 2004. Fluorodeoxyglucose PET imaging in
hepatocellular carcinomas, it does detect the evaluation of gallbladder carcinoma and
many of the poorly differentiated ones, cholangiocarcinoma. J. Gastrointest. Surg. 8:
and other PET tracers show promise for the 90–97.
detection of better differentiated hepato- Delbeke, D., Martin, W.H., Sandler, M.P.,
Chapman, W.C., Wright, J.K. Jr., and Pinson,
mas. While low volume cholangiocarci- C.W. 1998. Evaluation of benign vs malignant
nomas can escape detection by FDG-PET, hepatic lesions with positron emission tomogra-
higher volume lesions are well detectable. phy. Arch. Surg. 133: 510–515.
Similarly, gallbladder carcinoma is gener- Ho, C.L., Yu, S.C., and Yeung, D.W. 2003. 11C-
ally well detected by FDG-PET. The ability acetate PET imaging in hepatocellular carci-
of FDG-PET to quantitatively estimate noma and other liver masses. J. Nucl. Med. 44:
213–221.
metabolic rates makes it an important tool for Hustinx, R., Paulus, P., Jacquet, N., Jerusalem, G.,
therapy monitoring. Thus, with increas- Bury, T., and Rigo, P. 1998. Clinical evaluation
ingly broad applications for FDG-PET of whole-body 18F-fluorodeoxyglucose positron
imaging, it is expected that FDG-PET emission tomography in the detection of liver
(and PET/CT) of the liver will play a metastases. Ann. Oncol. 9: 397–401.
growing and increasingly important role Joost, H.G., and Thorens, B. 2001. The extended
GLUT-family of sugar/polyol transport facilita-
in detecting and monitoring treatment of tors: nomenclature, sequence characteristics, and
tumors involving the liver. potential function of its novel members. Mol.
Membr. Biol. 18: 247–256.
Kato, T., Tsukamoto, E., Kuge, Y., Katoh, C.,
Acknowledgement. The author would like Nambu, T., Nobuta, A., Kondo, S., Asaka, M.,
to thank M. Donald Blaufox, Chairman, and Tamaki, N. 2002. Clinical role of (18)
Department of Nuclear Medicine, Albert F-FDG PET for initial staging of patients with
16 A.H. Khandani

extrahepatic bile duct cancer. Eur. J. Nucl. Med. Rydberg, J., Khandani, A.H., Ivanovic, M., and
Mol. Imaging 29: 1047–1054. McCartney, W.H. 2005. Comparison of Liver,
Kim, Y.J., Yun, M., Lee, W.J., Kim, K.S., and Lee, Mediastinum, and Muscle as References for
J.D. 2003. Usefulness of 18F-FDG PET in intra- Background Activity in 18-F FDG PET-CT
hepatic cholangiocarcinoma. Eur. J. Nucl. Med. Imaging. RSNA Annual Meeting: Abstract # LPL
Mol. Imaging 30: 1467–1472. 15-04-P.
Kim, Y.W., Park, Y.K., Yoon, T.Y., and Lee, Torizuka, T., Tamaki, N., Inokuma, T., Magata, Y.,
S.M. 2002. Expression of the GLUT1 glu- Sasayama, S., Yonekura, Y., Tanaka, A.,
cose transporter in gallbladder carcinomas. Yamaoka, Y., Yamamoto, K., and Konishi, J.
Hepatogastroenterology 49: 907–911. 1995. In vivo assessment of glucose metabolism
Kinkel, K., Lu, Y., Both, M., Warren, R.S., and in hepatocellular carcinoma with FDG-PET.
Thoeni, R.F. 2002. Detection of hepatic metas- J. Nucl. Med. 36: 1811–1817.
tases from cancers of the gastrointestinal tract Trojan, J., Schroeder, O., Raedle, J., Baum, R.P.,
by using noninvasive imaging methods (US, CT, Herrmann, G., Jacobi, V., and Zeuzem, S. 1999.
MR imaging, PET): a meta-analysis. Radiology Fluorine-18 FDG positron emission tomography
224: 748–756. for imaging of Hepatocellular carcinoma. Am. J.
Koh, T., Taniguchi, H., Yamaguchi, A., Kunishima, S., Gastroenterol. 94: 3314–3319.
and Yamagishi, H. 2003. Differential diagnosis Tsuda, M., Rikimaru, H., Majima, K., Yamada, T.,
of gallbladder cancer using positron emission Saito, H., Ishibashi, T., Takahashi, S., Miyachi,
tomography with fluorine-18-labeled fluoro- H., Endoh, M., and Yamada, S. 2003. Time-
deoxyglucose (FDG-PET). J. Surg. Oncol. 84: related changes of radiofrequency ablation lesion
74–81. in the normal rabbit liver: findings of magnetic
Lee, J.D., Yun, M., Lee, J.M., Choi, Y., Choi, Y.H., resonance imaging and histopathology. Invest.
Kim, J.S., Kim, S.J., Kim, K.S., Yang, W.I., Radiol. 38: 525–531.
Park, Y.N., Han, K.H., Lee, W.J., Yoo, N., Lim, Yang, M., Martin, D.R., Karabulut, N., and Frick,
S.M., and Park, J.H. 2004. Analysis of gene M.P. 2003. Comparison of MR and PET imaging
expression profiles of hepatocellular carcinomas for the evaluation of liver metastases. J. Magn.
with regard to 18F-fluorodeoxyglucose uptake Reson. Imaging 17: 343–349.
pattern on positron emission tomography. Eur. J. Zimmerman, R.L., Burke, M., Young, N.A.,
Nucl. Med. Mol. Imaging 3: 1621–1630. Solomides, C.C., and Bibbo, M. 2002a.
Marom, E.M., McAdams, H.P., Erasmus, J.J., Diagnostic utility of Glut-1 and CA 15-3 in
Goodman, P.C., Culhane, D.K., Coleman, R.E., discriminating adenocarcinoma from hepatocel-
Herndon, J.E., and Patz, E.F. Jr. 1999. Staging lular carcinoma in liver tumors biopsied by fine-
non-small cell lung cancer with whole-body needle aspiration. Cancer 96: 53–57.
PET. Radiology 212: 803–809. Zimmerman, R.L., Fogt, F., Burke, M., and
Roh, M.S., Jeong, J.S., Kim, Y.H., Kim, M.C., and Murakata, L.A. 2002b. Assessment of Glut1
Hong, S.H. 2004. Diagnostic utility of GLUT1 expression in cholangiocarcinoma, benign
in the differential diagnosis of liver carcinomas. biliary lesions and hepatocellular carcinoma.
Hepatogastroenterology 5: 1315–1318. Oncol. Rep. 9: 689–692.
2
Localized Fibrous Tumor
of the Liver: Imaging Features
Thomas Moser and Tereza S. Nogueira

Also known as solitary fibrous tumors, the beginning of injection, respectively).


or localized fibrous mesothelioma, local- Occasionally, delayed images are also
ized fibrous tumors (LFTs) can occur obtained (at 5 min or more). Contrast uptake
in numerous different locations including within hepatic lesions is analyzed with
most organs and soft tissues. The arche- respect to these different phases, enabling
type of this entity has been described tumor characterization. For example, hyper-
in the pleura, where it accounts for 5% vascular tumors enhance simultaneously to
of tumors. On the other hand, LFT of the hepatic artery. Hypovascular tumors
the liver is a very infrequent tumor with disclose only mild or absent enhancement
no more than 30 cases reported in the during the portal phase. Lesions contain-
English language literature. Hepatic LFT ing a fibrous contingent may enhance only
usually manifests with vague abdominal during the delayed phase. Such delayed
complaints or even mildly abnormal liver enhancement is believed to correspond
blood tests. However, some patients remain to interstitial contrast diffusion through
asymptomatic and the tumor represents an collagen fibers.
incidental finding. It generally attains large On imaging studies, LFTs usually present
size before discovery. Patients may also as large well limited masses that could be
present with hypoglycemia as a paraneo- partially or entirely capsulated. Most are
plastic manifestation. Definitive diagnosis heterogeneous lesions, sometimes disclos-
of LFT reposes on histology coupled with ing cystic spaces. Calcification and necrosis
the characteristic immunological labeling are infrequent. On unenhanced CT, they are
for CD34. essentially hypodense to the surrounding
Imaging studies may sometimes suggest parenchyma. On unenhanced MRI, they
the diagnosis. As for any hepatic tumor, are hypointense on T1-weighted images,
multiphasic acquisition, either at computed whereas T2-weighted images show
tomography (CT) scan or magnetic reso- hypo- and hyperintense areas with some
nance imaging (MRI), is required. It means of them appearing fluid-like. After intra-
that after intravenous injection of contrast venous contrast injection, two principal
media, images are obtained during maxi- enhancement types have been described
mal hepatic artery and portal and hepatic so far.We summarize them as the hyper-
veins opacification (~30 and 70 s following vascular and delayed enhancement patterns.

17
18 T. Moser and T.S. Nogueira

In their observations, Fuksbrumer et al. portal phases and became marked on both
(2000) depicted large solitary lesions dem- CT and MRI delayed images. This large
onstrating early contrast uptake and het- tumor also showed multiple areas of cystic
erogeneous enhancement with areas of degeneration (Figure 2.1).
differential uptake and washout. These These two separate imaging patterns
findings suggestive of a hypervascu- are well correlated with histological find-
lar tumor are also consistent with most ings. Localized fibrous tumors are com-
descriptions of pleural LFTs. In the series posed of spindle cells intermixed with
of the Armed Force Institute of Pathology, collagen bundles. Most commonly, tumor
heterogeneous contrast enhancement was cells are disposed haphazardly in an
observed in 62% of cases (Rosado-de- arrangement known as the storiform pat-
Christenson et al., 2003). Conversely, we tern. Alternating hyper- and hypocellular
recently reported the case of a LFT of the areas are present in the same tumor.
liver presenting with delayed enhancement Prominent vessels are generally associated
pattern (Moser et al., 2005). After intrave- with the cellular proliferation, determining
nous contrast administration, a progressive the hemangiopericytoma-like vascular pat-
enhancement began during arterial and tern. Such an arrangement was observed

a b c

d e f

Figure 2.1. Hepatic LFT revealed by a hypoglycemic coma in a 73 year old woman. Transverse CT
images obtained before (a), 30 s (b), 1 min (c) and 5 min (d) after contrast injection. A well-demarcated,
hypodense lesion containing numerous prominent cystic areas is shown. The lesion enhances faintly at
arterial and portal phases. However, a marked contrast enhancement is seen 5 min after injection account-
ing for the fibrous content of the lesion. Delayed sagittal MR image obtained 8 min after contrast injection
also confirms the delayed enhancement pattern (e). View of the resected specimen (f). (Reprinted with
permission from the American Journal of Roentgenology.)
2. Localized Fibrous Tumor of the Liver: Imaging Features 19

in all nine cases reported by Moran et al. nosis of other rare mesenchymal hepatic
(1998) as well as in those cases reported by tumors, particularly fibrous tissue contain-
Fuksbrumer et al. (2000). In fact, it repre- ing sarcomas such as fibrosarcoma and
sents the most common microscopic pattern malignant fibrous histiocytoma. Hepatic
described so far and corresponds to the most fibrosarcoma also manifests as a large
common hypervascular pattern of contrast mass and possible hypoglycemia. It con-
uptake where heterogeneous enhancement tains an abundant collagenous stroma with
seems to result from the varying cellularity spindle cells, which are in this case char-
observed throughout the tumor. However, acteristically arranged in a herringbone
the hemangiopericytoma-like vascular pat- pattern. Moreover, immunological labe-
tern was absent in our study. Rather, it was ling for CD34 is negative.
a poorly vascularized tumor with an inter- To summarize, imaging can only sug-
stitium almost exclusively composed of gest the diagnosis of LFT in rare cases,
collagen fibers. Therefore, its enhancement but thorough analysis of vascularity often
dynamic at CT and MR was consistent with brings clues about histological content.
a collagen rich interstitium where contrast
gradually diffuses between the fibers. REFERENCES
Most hepatic LFTs are benign lesions.
However, a malignant variant with a more Chan, J.K. 1997. Solitary fibrous tumour–every-
where, and a diagnosis in vogue. Histopathology
aggressive behavior such as recurrence 31:568–576.
and distant metastasis has been described. Fuksbrumer, M.S., Klimstra, D., and Panicek, D.M.
Malignancy can be suspected solely in 2000. Solitary fibrous tumor of the liver: imaging
the presence of numerous cellular atypias findings. Am. J. Roentgenol. 175:1683–1687.
on histology, or with the identification of Moran, C.A., Ishak, K.G., and Goodman, Z.D.
distant metastases (Chan, 1997). Thus, 1998. Solitary fibrous tumor of the liver: a clin-
icopathologic and immunohistochemical study
from the imaging viewpoint, LFTs should of nine cases. Ann. Diagn. Pathol. 2:19–24.
be included in the differential diagnosis Moser, T., Nogueira, T.S., Neuville, A., Riehm, S.,
of any extensive and well limited hepatic Averous, G., Weber, J.C., and Veillon, F. 2005.
mass. A delayed enhancement pattern has Delayed enhancement pattern in a localized
been demonstrated for a wide range of fibrous tumor of the liver. Am. J. Roentgenol.
fibrous containing hepatic lesions, such 184:1578–1580.
Rosado-de-Christenson, M.L., Abbott, G.F.,
as fibrolamellar hepatocellular carcinoma, McAdams, H.P., Franks, T.J., and Galvin, J.R.
cholangiocarcinoma, or sclerosing heman- 2003. From the archives of the AFIP: local-
gioma. Localized fibrous tumor should ized fibrous tumor of the pleura. Radiographics
also be included in the differential diag- 23:759–783.
3
A Radial Magnetic Resonance Imaging
Method for Imaging Abdominal
Neoplasms
Maria I. Altbach

INTRODUCTION T1-weighted images acquired with lipid and


water in- and out-of-phase, respectively,
Abdominal magnetic resonance imaging are used primarily for the characteriza-
(MRI) has progressed rapidly during the tion of neoplasms such as adenomas,
last 12 years. The development of new focal nodular hyperplasia, regenerative
hardware and software has allowed the nodules, well-differentiated hepatocellular
implementation of faster and better pulse carcinoma, nodular regenerative hyperpla-
sequences, and MRI is now used routinely sia, renal clear cell carcinoma, and angi-
in the clinic as a complement to, or as omyolipoma (Ichikawa and Araki, 1999;
the method of choice over other imaging Outwater et al., 1998).
techniques such as ultrasound (US) and Metastatic lesions, however, cannot be
computed tomography (CT). Magnetic distinguished from the most common
resonance imaging combines good spatial benign lesions (cysts and hemangiomas)
resolution, lack of radiation, non-inva- with T1-weighted methods because the
siveness, and three-dimensional imaging two types of lesions have the same sig-
capability. This method provides useful nal characteristics (i.e., hypointense signal
information for the detection and charac- relative to liver). Differentiation of metas-
terization of focal liver lesions that cannot tasis, and other primary malignancies,
be diagnosed with US or CT (Robinson, from hemangiomas and cysts is based on
2000). In particular for small lesions T2-weighted images (Ito et al., 1997).
(< 1 cm2), studies have shown that the Because this is a crucial aspect of the
specificity and sensitivity of MRI is better diagnosis, T2-weighted imaging is a very
than US and CT (Ward et al., 2005). important part of the abdominal MRI
The standard MRI protocol for liver examination. Contrast-enhanced MRI is
and most abdominal imaging applications also important for characterization of liver
consists of acquiring T1-weighted, T2- lesions. It has been shown that the pattern
weighted, and contrast-enhanced images. of enhancement after the administration
All these methods are used for the detec- of a gadolinium-based contrast agent is
tion of lesions but each has a different different for malignancies, hemangiomas,
role in terms of lesion characterization. and cysts, and that the combined use of

21
22 M.I. Altbach

T2-weighted and contrast-enhancement data (Semelka et al., 1996). The speed of


data increases the sensitivity and spe- the method, however, is at the expense of
cificity for detecting and characterizing image bluriness and loss of small object
lesions compared to each technique alone detectability in soft tissue (Constable
(Pawluk et al., 1999). and Gore, 1992), which primarily affects
the detection of small malignant tumors
(Coates et al., 1998). Other fast imaging
TECHNIQUES FOR techniques such as single-shot echo pla-
T2-WEIGHTED IMAGING nar (SSEPI) and steady-state free pre-
cession (SSFP) have also been proposed
Initially, T2-weighted images of the abdo- for obtaining motionless images of the
men were acquired with two-dimensional abdomen. Single-shot echo planar offers
Fourier transform (2DFT) spin-echo a better lesion-to-liver contrast-to-noise
(SE) methods (McFarland et al., 1994). ratio (CNR) than SSFSE (Namimoto
Conventional T2-weighted SE imaging et al., 1997), but the technique is limited
suffers from long acquisition times (5– in spatial resolution and is affected by
10 min) and artifacts caused by respiratory artifacts caused by changes in magnetic
motion, cardiac motion, pulsatile flow, and susceptibility. Steady-state free preces-
peristalsis. Thus, the technqiue has been sion offers good spatial resolution but the
replaced by fast spin-echo (FSE) methods. CNR for malignant lesions is poor (Jung
Multi-shot 2DFT-FSE techniques are used et al., 2004; Herborn et al., 2003). Images
for data acquisition during quiet breath- acquire with SSFP are also affected by
ing (using respiratory trigerring) or during artifacts caused by magnetic field inho-
periods of suspended respiration. Both res- mogeneities (Bangerter et al., 2004).
piratory-trigerred and breath-hold 2DFT- An inherent problem of all T2-weighted
FSE methods yield images with good imaging methods is that the hyperintense
spatial resolution and signal-to-noise ratio signal from flow can mask or mimic the
(SNR) and the method was shown to be an signal of lesions.
improvement over SE acqusitions (Keogan
et al., 1996; Gaa et al., 1996). Despite
this improvement, however, the quality of LESION CHARACTERIZATION
images obtained with multi-shot 2DFT- WITH T2-WEIGHTED
FSE is still affected by ghosting and blur- IMAGING
ring caused by pulsatile flow and residual
breathing motion caused by improper res- Since the early developments of abdomi-
piratory trigerring or breath holding. nal MRI it was found that malignant
As an altenative to multi-shot 2DFT and benign lesions have different signal
FSE, single-shot fast spin-echo (SSFSE) characteristics in T2-weighted images.
is a faster imaging technique that has Numerous studies have been undertaken
become very popular in abdominal imag- to evaluate the best method to characterize
ing (Tang et al., 1997). With SSFSE, the liver neoplasms with T2-weighted imaging
acquisition of data for one section of the (Reimer et al., 1996; Yu et al., 1998). The
liver takes < 1 s providing motion-free performance of the various T2-weighted
3. A Radial Magnetic Resonance Imaging Method for Imaging Abdominal Neoplasms 23

techniques and imaging parameters was 100% accuracy. The SSEPI T2 measure-
investigated using qualitative approaches ment used data at five TE time points to
(i.e., observers visually comparing signal improve the accuracy of the T2 estimate,
intensities between a moderate and heavily but required the acquistion of data over
T2-weighted images) as well as quantita- several breath hold periods. This made
tive approaches (i.e., observers measuring the experiment both time-consuming and
signal intensity and/or CNR of lesions dependent on the ability of the subject
relative to liver). Although reports indi- to perform reproducible breath holds in
cated that the quantitative measurements order to avoid errors due to misregistra-
yielded a better characterization of liver tion between TE data sets. The low spatial
lesions, these measurements require resolution of SSEPI and its sensitivity to
significant operator interaction and are susceptibility artifacts limit the technique
not practical for clinical use (Ito et al., even further.
1997). Consequently, in current clinic Abe et al. (2000) used SSFSE to cal-
practice, lesion characerization based on culate T2 at two TE time points within a
T2-weighted imaging is performed by breath hold, but showed significant overlap
visually comparing the signal intensity between malignancies and benign lesions.
of lesions between moderate and heavily Olcott et al. (1999) measured T2 values in
T2-weighted images. the liver with a pulse sequence designed
Several studies have demonstrated that to acquire data from four TE time points
a better quantitative approach for liver with reduced errors due to radiofrequency
lesion charaterization is to directly meas- pulse imperfections. Although they dem-
ure the T2 value by acquiring data at two onstrated a good separation between
or more echo time (TE) points and fitting malignant and benign lesions for a set
the signal intensities to a single exponen- of 24 lesions, their method is also time-
tial decay. Initially, T2 measurements in consuming. Two recent studies based dual-
the upper abdomen were mainly obtained echo respiratory-trigerred 2DFT-FSE, a
with dual-echo 2DFT-SE methods with technique that is also sensitive to motion,
the drawback of long acquisition times reported that the accuracy of separating
(10–20 min) and errors due to motion malignancies from hemangiomas based
(i.e., signal displacement along the phase- on T2 is 92–94% (Cieszanowski et al.,
encoding direction and motion-induced 2002; Kim et al., 2005). In summary,
volume averaging). Thus, although one of the current methods for measuring T2
these studies reported a 97% accuracy for values in the body have at least one of
discriminating malignancies from heman- the following problems: long imaging
giomas (McFarland et al., 1994), the others times, motion-induced errors, misregistra-
show significant overlap between these two tion of images acquired in different breath
type of lesions (Ohtomo et al., 1988). To holds, low spatial resolution, and/or low
reduce the effect of motion in T2 measure- number of measured points on the relaxa-
ments, Goldberg et al. (1993) used SSEPI tion curve. These technical difficulties not
to measure T2 in 46 liver neoplasms and only reduce the accuracy of these methods
show that hemangiomas and cysts can but also make them impractical for routine
be discriminated from malignancies with clinical use.
24 M.I. Altbach

RADIAL MAGNETIC Recently, a multi-shot radial FSE tech-


RESONANCE IMAGING nique was developed for T2-weighted
METHODS: AN imaging of the liver (Altbach et al., 2002).
The technique yields motion-insensitive
ALTERNATIVE FOR abdominal images with high spatial reso-
REDUCING MOTION lution. One of the highligts of the tech-
ARTIFACTS AND IMPROVING nique is that a small amount of diffusion
IMAGE QUALITY weighting can be incorporated to sup-
press the bright signal from blood vessels
Motion-dependent phase shifts accumu- improving the visualization of lesions.
lated within the time domain data (i.e., the Examples of images acquired with the
Fourier or k-space data) cause artifacts in radial FSE and the conventional 2DFT-
MR images. If k-space data are sampled in FSE method are shown in Figure 3.1. The
a rectilinear grid, as in the case of 2DFT- images in Figure 3.1a were acquired dur-
MRI, motion artifacts are manifested as ing free breathing using respiratory-trig-
blurring and replicated ghost patterns along gered 2DFT-FSE and radial FSE methods.
one direction in the image, i.e., the direction Note that the two small metastatic lesions
of the phase encoding imaging gradient. seen in the radial FSE image (arrows) are
If k-space data are sampled on a polar grid, masked in the 2DFT-FSE image by flow
as in radial MRI, motion artifacts are reduced and motion artifacts. The images in Figure
due to two fundamental reasons (Glover 3.1b were acquired with 2DFT-FSE and
and Pauly, 1991). First, motion artifacts radial FSE in a breath hold. Again in this
are spread in two dimensions making the example, flow and motion artifacts impair
intensity of the artifact less severe. Second, the visualization of lesions in the 2DFT-
in polar data, there is a higher density of FSE image. Lesions are better depicted in
sampled points in the center of k-space the radial FSE image (arrow heads). The
(in Cartesian data the sampling density is importance of flow suppression is quite
uniform) which averages data and reduces evident in this example, where the small
phase errors due to motion. The insensitiv- metastatic lesion in the lower right portion
ity of radial MRI to motion has been widely of the liver is only distinguishable in the
demonstrated. Glover and Pauly (1991) radial FSE image due to improved flow
showed that radial SE methods produce suppression.
abdominal images with less motion-induced Examples of images acquired with radial
artifacts than 2DFT-SE methods. Rasche et FSE and the conventional SSFSE method
al. (1994) showed that radial FSE images of in a breath hold are shown in Figure 3.2.
the abdomen acquired within a breath hold The two small metastatic lesions indicated
have less ghosting and blurring than images by the arrows in Figure 3.2a are only seen
acquired with breath-hold 2DFT-FSE meth- in the radial FSE image but not seen in the
ods. The robustness of radial methods to SSFSE image due to the intrinsic bluriness
motion have also been demonstrated for of the latter. The effect of blurriness in
lung (Kauczor and Kreitner, 1999), cardiac SSFSE is also seen in the images shown
(Schaeffter et al., 2001), and diffusion- in Figure 3.2b where lesions are more
weighted MRI (Trouard et al., 1999). conspicuous in the radial FSE image than
3. A Radial Magnetic Resonance Imaging Method for Imaging Abdominal Neoplasms 25

Figure 3.1. A comparison of images acquired with the conventional 2DFT-FSE and the radial FSE method.
(a) Images acquired during free breathing using respiratory triggering. The two metastatic lesions (arrows)
seen in the radial FSE image are masked by flow artifacts in the 2DFT-FSE image. (b) Images acquired in
a breath hold. The image acquired with 2DFT-FSE has ghosting artifacts caused by flow. The signal from
flow is well suppressed in the radial FSE image by using a small amount of diffusion weighting (b = 1.2 s/
mm2), so the metastatic lesions (arrow heads) are clearly seen. Note that the small lesion in the lower right
portion of the liver is only seen in the radial FSE image

in the SSFSE image. An example showing These qualitative findings were cor-
the advantage of flow suppression via dif- roborated by a statistical evaluation of
fusion weighting in radial FSE is shown in image quality for a set of 16 patients with
Figure 3.2c for a patient with very small various types of liver neoplasms (Altbach
metastatic lesions (as reference the lesion et al., 2002). In the study, images were
highlighted by the arrow is 5 mm in diam- acquired with radial FSE and the conven-
eter). In the SSFSE image the signal from tional 2DFT-FSE and SSFSE methods.
lesions cannot be distinguished from the The results of the study indicated that
signal from blood vessels. This is not the radial FSE performed significantly better
case in the radial FSE image where the in terms of organ and lesion definition
signal from flow is suppressed leaving the than SSFSE or 2DFT-FSE, In particular,
bright signal from lesions present. it was demostrated that radial FSE was
26 M.I. Altbach

Figure 3.2. Comparison of images acquired with the conventional SSFSE and the radial FSE method. (a)
Images of a subject with two small metastatic lesions at the bottom right of the liver (arrows). The lesions
are clearly seen in the radial FSE image but not in the SSFSE image due to the intrinsic blurriness of the
latter. (b) Images of a patient with extensive metastatic invasion showing the higher lesion conspicuity
yielded by the radial FSE method. (c) Images of a subject with very small metastatic lesions. As a refer-
ence the lesion highlighted by the arrow is 5 mm in diameter. In the image acquired with SSFSE both
blurriness and the bright signal from blood vessels impairs the assignment of lesions. In the radial FSE
image the signal from blood vessels is suppressed via diffusion weighting which helps identifying the
signal from metastatic lesions
3. A Radial Magnetic Resonance Imaging Method for Imaging Abdominal Neoplasms 27

significantly better than the conventional equals the distance between k-space sam-
methods for malignant lesion definition. ples in the radial direction). In this man-
ner, high-resolution images at various TEeff
values as well as a T2 map are obtained
RADIAL FAST SPIN-ECHO, from a single radial FSE k-space data (Song
and Dougherty, 2000; Altbach et al., 2002,
A NEW ALTERNATIVE FOR 2005). This novel methodology provides a
T2 MAPPING OF THE LIVER new way to measure T2 without the prob-
lems typically encountered in the body. The
In conventional FT-MRI the measurement
T2 maps are generated from motion-insen-
of T2 requires the acquisition of two
sitive data acquired in a single breath hold.
or more k-space data sets with different
Thus, the errors due to motion or misreg-
T2 weighting. In SE acqusitions all the
istration of TE data sets are minimized.
k-space lines on each T2-weighted data Data have goodeffspatial resolution as well as
set are collected at a specific TE. In faster adequate TE time resolution (T2 values can
acqusition methods such as FSE, SSFSE, be calculated from 8 or 16 values of TE ).
eff
and EPI, the k-space lines on each data Moreover, the acquisition of data is fast (for
set have mixed TE values and the T2 abdominal imaging applications, it takes
weighting of the data set is approximated one breath hold to acquire data for several
to the TE of the lines in the central part of slices) and the post-processing algorithm
k-space (i.e., the so-called TEeff). For esti- is simple. Thus, the method can be easily
mating T2, it is necessary to acquire vari-
implemented as part of a regular clinical
ous data sets, each with a different TEeff.
MRI examination.
T2 is then calculated by fitting the MRI
Figure 3.4 shows the anatomical image
signal intensity, I, to:
(reconstructed from the full k-space data

TEeff set) as well as the T2-weighted images
I = Ioe T2
(3.1) and the T2 map (generated from the par-
tial k-space data sets) for a patient with
where Io, the equilibrium magnetization, is metastatic liver lesions and a cyst in the
a constant. kidney. All these information is obtained
In radial FSE, data at all TEs sample from a single k-space data set collected
the center of k-space (Figure 3.3a). Since in a breath hold. Note the differences in
the center of k-space is oversampled, contrast in the T2-weighted images and
T2-weighted contrast can be generated the T2 map for the various lesions. The
by partitioning a full k-space data set into signal intensity of the cyst (arrow head)
subsets that have in the center only the stays bright as TEeff increases, whereas the
data from lines acquired at a specific TE. signal intensity of the malignant lesions
Data at other TEs are added progressively (arrows) follows the signal intensity of the
from the center to the outer part of k-space kidney. The T2-weighted images and the
as indicated in Figure 3.3b. The radius on T2 map also reveal areas within two of
each tier of data satisfies the Nyquist crite- the metastatic lesions (long arrows) that
ria (i.e., the distance between two adjacent have a high T2. These correspond to
k-space samples in the azimuthal direction necrotic areas within the tumors. A study
28 M.I. Altbach

Figure 3.3. Schematic representation of the method used to generate multiple high-resolution images
with different T2-weighting and a T2 map from a single radial FSE data set. The method is demonstrated
for data acquired with an echo train length of four (i.e., four TE values)

on a set of 43 neoplasms which included 24 sented above, is the radius at which all TE
malignancies (22 metastases, 1 hepatoma, data are included. This radius determines
1 hepatocellular carcinoma) and 19 benign the accuracy of T2 estimates; the larger
lesions (6 hemangiomas and 12 cysts) cor- the radius the more accurate the estimates,
responding to 28 patients showed that the in particular for small lesions. This radius
T2 value determined from radial FSE data depends on the total number of radial lines
alone can discrimate hemangiomas and collected, and recently it was demon-
cysts from malignancies (Figure 3.5). strated that for a full data set of 512 radial
An important parameter in the determina- lines the error in T2 estimates for lesions
tion of T2, based on the radial method pre- with a diameter of 0.5–0.6 cm is only 7%.
3. A Radial Magnetic Resonance Imaging Method for Imaging Abdominal Neoplasms 29

Figure 3.4. Anatomical images at different TEeff, and the T2 map for a patient with metastatic liver lesions
(arrows) and a cyst (arrow head) in the kidney. All the images were reconstructed from a single radial FSE data
set acquired in a breath hold. The anatomical image is reconstructed using the full k-space data set. The images
at various TEeff were reconstructed using data at selected TE values in the center of k-space as illustrated in
Figure 3.3. The T2 map was obtained by fitting the pixel intensities to equation (3.1)

For lesions with a diameter greater than the other half is calculated and (Barger
0.8–1.0 cm the error is less than 4% (Altbach et al., 2002; Bydder et al., 2002) can be
et al., 2005). It should be pointed out that used to render the data needed for accurate
if the collection of sufficient radial lines is T2 estimation.
by the breath hold period, techniques were As any other technique that uses a single
only half of the radial lines collected and exponential fit to estimate T2, the method
30 M.I. Altbach

methods used for imaging the upper abdo-


men. The radial FSE method does not
suffer from the characteristic bluriness or
lack of spatial resolution of single-shot
methods. The radial FSE method does
not have the motion artifacts of multi-shot
2DFT-FSE methods. The robustness of
the technique to motion allows the use
of a small amount of diffusion weighting
to suppress the signal from flow, which
improves the visualization of lesions.
A major advantage of radial FSE is that
T2-weighted images as well as a T2 map
can be obtained from a single k-space data
set. The T2 maps obtained from radial
FSE data have high-spatial resolution as
well as adequate TE time resolution. T2
maps are generated from data acquired
in a single breath hold, thus errors due
Figure 3.5. T2 values for malignant and the most to motion-induced volume averaging
common benign liver lesions (i.e., hemangiomas and/or misregistration are minimized.
and cysts) obtained from radial FSE data using the The method is fast and can be implemented
algorithm presented in Figure 3.3. The T2 axis is as part of a regular clinical MR examina-
displayed on a logarithmic scale.
tion. This novel methodolgy may proved
to be very valuable for the detection and
presented here does not take into account characterization of liver neoplasms.
partial volume effects (i.e., when the signal
from liver is within the volume occupied
by the lesion). Partial volume may result Acknowledgement. This work was supported
in the misclassification of small hemangi- by NIH grant: CA099074.
omas and cysts as malignancies. However,
since the radial FSE method yields 8–16 REFERENCES
points along the T2 curve, it is possible to
use a double-exponential fit to calculate Abe, Y., Yamashita, Y., Tang, Y., Namimoto T., and
Takahashi, M. 2000. Calculation of T2 relaxa-
the correct T2 value when more than one tion time from ultrafast single shot sequences
type of tissue is present within a voxel. for differentiation of liver tumors: comparison of
echo-planar, HASTE, and spin-echo sequences.
Radiat. Med. 18:7–14.
Altbach, M.I., Outwater, E.K., Trouard, T.P.,
CONCLUSIONS Krupinski, E.A., Theilmann, R.J., Stopeck, A.T.,
Kono, M., and Gmitro, A.F. 2002. Radial fast
In summary, the radial FSE method over- spin-echo method for T2-weighted imaging
comes most of the major problems encoun- and T2 mapping of the liver. J. Magn. Reson.
tered with conventional T2-weighted MRI Imaging 16:179–189.
3. A Radial Magnetic Resonance Imaging Method for Imaging Abdominal Neoplasms 31

Altbach, M.I., Bilgin, A., Li, Z., Clarkson, E.W., Herborn, C.U., Vogt, F., Lauenstein, T.C., Goyen, M.,
and Gmitro, A.F. 2005. Processing of radial fast- Debatin, J.F., and Ruehm, S.G. 2003. MRI
spin echo data for obtaining T2 estimates from of the liver: can True FISP replace HASTE?
a single K-space data set. Magn. Reson. Med. J. Magn. Reson. Imaging 17:190–196.
54:549–559. Ichikawa, T., and Araki, T. 1999. Fast magnetic
Bangerter, N.K., Hargreaves, B.A., Vasanawala, resonance imaging of liver. Eur. J. Radiol.
S.S., Pauly, J.M., Gold, G.E., and Nishimura, 29:186–210.
D.G. 2004. Analysis of multiple-acquisition Ito, K., Mitchell, D.G., Outwater, E.K., Szklaruk, J.,
SSFP. Magn. Reson. Med. 51:1038–1047. and Sadek, A.G. 1997. Hepatic lesions: discrimi-
Barger, A.V., Block, W.F., Toropov, Y., Grist, T.M., nation of nonsolid, benign lesions from solid,
and Mistretta, C.A. 2002. Time resolved con- malignant lesions with heavily T2-weighted
trast-enhanced imaging with isotropic resolu- fast spin-echo MR imaging. Radiology 204:
tion and broad coverage using an undersampled 729–737.
3D projection trajectory. Magn. Reson. Med. Jung, B., Krombach, G.A., Gunther, R.W., and
48:297–305. Buecker, A. 2004. Is postcontrast trueFISP imag-
Bydder, M., Larkman, D.J., and Hajnal, J.V. 2002. ing advantageous? Invest. Radiol. 39:517–523.
Generalized SMASH imaging. Magn. Reson. Kauczor, H.U., and Kreitner, K.F. 1999. MRI
Med. 47:160–170. of the pulmonary parenchyma. Eur. Radiol.
Cieszanowski, A., Szeszkowski, W., Golebiowski, 9:1755–1764.
M., Bielecki, D.K., Grodzicki, M., and Keogan, M.T., Spritzer, C.E., Paulson, E.K., Paine,
Pruszynski, B. 2002. Discrimination of benign S.S., Harris, L., Dahlke, J.L., and MacFall, J.R.
from malignant hepatic lesions based on their 1996. Liver MR imaging: comparison of respira-
T2-relaxation times calculated from moderately tory triggered fast spin echo with T2-weighted
T2-weighted turbo SE sequence. Eur. Radiol. spin-echo and inversion recovery. Abdom.
12:2273–2279. Imaging 21:433–439.
Coates, G.G., Borrello, J.A., McFarland, E.G., Kim, Y.H., Saini, S., Blake, M.A., Harisinghani, M.,
Mirowitz, S.A., and Brown, J.J. 1998. Hepatic Chiou, Y.Y., Lee, W.J., Yu, J.S., and Hahn, P.F.
T2-weighted MRI: a prospective comparison of 2005. Distinguishing hepatic metastases from
sequences, including breath-hold, half-Fourier hemangiomas: qualitative and quantitative
turbo spin echo (HASTE). J. Magn. Reson. diagnostic performance through dual echo res-
Imaging 8:642–649. piratory-triggered fast spin echo magnetic reso-
Constable, R.T., and Gore, J.C. 1992. The loss of nance imaging. J. Comput. Assist. Tomogr. 29:
small objects in variable TE imaging: implica- 571–579.
tions for FSE, RARE, and EPI. Magn. Reson. McFarland, E.G., Mayo-Smith, W.W., Saini, S.,
Med. 28:9–24. Hahn, P.F., Goldberg, M.A., and Lee, M.J.
Gaa, J., Hatabu, H., Jenkins, R.L., Finn J.P., and 1994. Hepatic hemangiomas and malignant
Edelman, R.R. 1996. Liver masses: replace- tumors: improved differentiation with heavily
ment of conventional T2-weighted spin-echo T2-weighted conventional spin-echo MR imag-
MR imaging with breath-hold MR imaging. ing. Radiology 193:43–47.
Radiology 200:459–464. Namimoto, T., Yamashita, Y., Sumi, S., Tang, Y.,
Glover, G.H., and Pauly, J.M. 1992. Projection and Takahashi, M. 1997. Focal liver masses:
reconstruction techniques for reduction of characterization with diffusion-weighted echo-
motion effects in MRI. Magn. Reson. Med. planar MR imaging. Radiology 204:739–744.
28:275–289. Ohtomo, K., Itai, Y., Yoshikawa, K., Kokubo, T.,
Goldberg, M.A., Hahn, P.F., Saini, S., Cohen, and Iio M. 1988. Hepatocellular carcinoma and
M.S., Reimer, P., Brady, T.J., and Mueller, P.R. cavernous hemangioma: differentiation with MR
1993. Value of T1 and T2 relaxation times from imaging. Efficacy of T2 values at 0.35 and 1.5 T.
echoplanar MR imaging in the characterization Radiology 168:621–623.
of focal hepatic lesions. Am. J. Roentgenol. 160: Olcott, E.W., Li, K.C., Wright, G.A., Pattarelli,
1011–1017. P.P., Katz, D.S., Ch’en, I.Y., and Daniel, B.L.
32 M.I. Altbach

1999. Differentiation of hepatic malignancies Semelka, R.C., Kelekis, N.L., Thomasson, D.,
from hemangiomas and cysts by T2 relaxation Brown, M.A., and Laub, G.A. 1996. HASTE
times: early experience with multiply refocused MR imaging: description of technique and pre-
four-echo imaging at 1.5 T. J. Magn. Reson. liminary results in the abdomen. J. Magn. Reson.
Imaging 9:81–86. Imaging 6:698–699.
Outwater, E.K., Blasbalg, R., Siegelman, E.S., and Song, H.K., and Dougherty, L. 2000. K-space
Vala, M. 1998. Detection of lipid in abdominal weighted image contrast (KWIC) for contrast
tissues with opposed-phase gradient-echo images manipulation in projection reconstruction MRI.
at 1.5 T: techniques and diagnostic importance. Magn. Reson. Med. 44:825–832.
Radiographics 18:1465–1480. Tang, Y., Yamashita, Y., Namimoto, T., Abe, Y.,
Pawluk, R.S., Tummala, S., Brown, J.J., and and Takahashi, M. 1997. Liver T2-weighted
Borrello, J.A. 1999. A retrospective analysis MR imaging: comparison of fast and conven-
of the accuracy of T2-weighted images and tional half-Fourier single-shot turbo spin-echo,
dynamic gadolinium-enhanced sequences in the breath-hold turbo spin-echo, and respiratory-
detection and characterization of focal hepatic triggered turbo spin-echo sequences. Radiology
lesions. J. Magn. Reson. Imaging 9:266–273. 203:766–772.
Rasche, V., Holz, D., and Schepper, W. 1994. Trouard, T.P., Theilmann, R.J., Altbach, M.I., and
Radial turbo spin echo imaging. Magn. Reson. Gmitro, A.F. 1999. High-resolution diffusion
Med. 32:629–638. imaging with DIFRAD-FSE (diffusion-weighted
Reimer, P., Rummeny, E.J., Wissing, M., Bongartz, radial acquisition of data with fast spin-echo)
G.M., Schuierer, G., and Peters, P.E. 1996. MRI. Magn. Reson. Med. 42:11–18.
Hepatic MR imaging: comparison of RARE Ward, J., Robinson, P.J., Guthrie, J.A., Downing, S.,
derived sequences with conventional sequences Wilson, D., Lodge, J.P., Prasad, K.R., Toogood,
for detection and characterization of focal liver G.J., and Wyatt, J.I. 2005. Liver metastases in
lesions. Abdom. Imaging 21:427–432. candidates for hepatic resection: comparison of
Robinson, P.J. 2000. Imaging liver metastases: helical CT and gadolinium- and SPIO-enhanced
current limitations and future prospects, Br. J. MR imaging. Radiology 237:170–180.
Radiol. 73:234–241. Yu, J.S., Kim, K.W., Kim, Y.H., Jeong, E.K.,
Schaeffter, T., Weiss, S., Eggers, H., and Rasche, V. and Chien, D. 1998. Comparison of multishot
2001. Projection reconstruction balanced fast turbo spin echo and HASTE sequences for
field echo for interactive real-time cardiac imag- T2-weighted MRI of liver lesions. J. Magn.
ing. Magn. Reson. Med. 46:1238–1241. Reson. Imaging 8:1079–1084.
4
Liver: Helical Computed Tomography
and Magnetic Resonance Imaging
Yuji Baba, Yasuyuki Yamashita, Kazuo Awai, and Koichi Kawanaka

INTRODUCTION with MDCT is so rapid, the liver can


be imaged as a volume data during
Both computed tomography (CT) and mag- a single breath hold. Second, both the
netic resonance (MR) imaging have made hepatic arterial-dominant and portal
major contributions to the evaluation of venous-dominant phases of enhancement
liver morphology, blood flow and detection can be visualized. Numerous studies have
and characterization of liver neoplasms. demonstrated improved detection of hyper-
Studies in numerous institutions have vascular masses when such dual-phase
established that both CT and MR imag- hepatic CT is used (Baron et al., 1996;
ing are highly accurate in the diagnosis of Bonaldi et al.,1995).
hepatic pathology. In our current practice, multiple phases
In recent years, there has been a major of the whole liver are imaged, including
revolution in these imaging modalities, the precontrast, arterial, portal venous and
i.e., multi-detector computed tomography equilibrium phase, or some of these phases
and fast MR imaging. These advanced are selected for scanning. With the recent
imaging technologies as applied in liver development of MDCT technologies, we
imaging are discussed in this section. can obtain double arterial phases (early
arterial phases and late arterial phases) in
a single breath-hold. Hepatic scanning can
DYNAMIC COMPUTED be completed during the interval of peak
TOMOGRAPHY USING hepatic contrast enhancement. However,
MULTI-DETECTOR because the liver can generally be imaged
COMPUTED TOMOGRAPHY within a single breath-hold with MDCT,
respiratory misregistration between adja-
Ultrafast imaging has been made possi- cent scan slices is eliminated.
ble by the development of multi-detector In addition, partial volume averaging
computed tomography (MDCT). There of small lesions with the normal liver is
are many advantages of multi-detector reduced. This may improve the accuracy
computed tomography for the evaluation of the evaluation of small focal lesions.
of the liver disease. First, because imaging Isotropic volume data acquisition enables

33
34 Y. Baba et al.

multiplanar reconstruction of the image. liver contrast for hypervascular primary


With single-detector helical CT, the entire and metastatic neoplasms occurs with the
liver can be reliably imaged within 20-s. second pass. The first pass provides opti-
With MDCT, on the other hand, a true mum data for hepatic CT arteriography
hepatic arterial phase occurs in the 10-s in pretransplant and preresection patients.
time frame after the arrival of contrast in Multiplanar imaging is useful to evaluate
the abdominal aorta. A double-pass hepatic the relationship of hepatic tumors to major
scan is performed in a single breath-hold. vascular structures. The porta hepatis is
The first pass is a pure arterial phase and anatomically complex, and multiplanar
the second pass a late arterial or portal display of reconstructed images may also
venous inflow phase. Maximum tumor-to- be useful in this region (Figure 4.1).

Figure 4.1. A case of undifferentiated hepatocellular carcinoma. Upper left: Precontrast CT. Upper right:
Arterial-phase image. Lower left: Equilibrium-phase image. Lower right: Reconstructed coronal image.
Tumor is iso to hypointense relative to the surrounding liver on precontrast CT. In arterial-phase images,
irregularly enhanced tumor is seen. Tumor enhancement is not obvious compared to well-differentiated
tumor (Figure 4.3). In the equilibrium phase, the tumor is seen as iso to hypoattenuating mass relative
to the surrounding liver. The relationship between tumor and vascular/biliary structure is clearly demon-
strated in reconstructed coronal image
4. Liver: Helical Computed Tomography and Magnetic Resonance Imaging 35

TUMORS ARISING IN LIVER Many primary hepatic neoplasms and


CIRRHOSIS hypervascular metastases can become
isoattenuating during the portal venous
In patients with liver cirrhosis, dysplastic phase. Because of their hypervascularity,
nodules and hepatocellular carcinomas such neoplasms are better imaged during
frequently develop. The diagnosis of hepa- the arterial-phase of hepatic enhancement.
tocellular carcinoma is extremely impor- Multi-detector computed tomography pro-
tant in this subset of patients. The majority vides the important potential to image
of hepatocellular carcinomas are hyper- the entire liver in the multiple phases of
vascularbecause of the prominent blood enhancement. For visualization of hyper-
supply from the hepatic artery, and MDCT vascular hepatocellular carcinomas, a rapid
using a bolus injection of contrast material injection rate is preferred to maximize the
has played an important role in the detec- arterial-phase enhancement with appropri-
tion of hypervascular hepatocellular carci- ateadjustments made to the scan timing.
nomas (Hosoki et al., 1982; Bluemke et al., Tumors are usually more conspicuous in
1994). These lesions are typically rounded the arterial phase and equilibrium phase.
or oval in configuration and exhibit a mass Two other arterial-phase patterns are
effect on adjacent portal or hepatic veins in typically associated with hepatocellular
the portal venous phase. carcinoma. One is a mosaic pattern with

Figure 4.2. A case of hepatocellular carcinoma arising in a cirrhotic liver. Left: Arterial-phase image.
Right: Equilibrium-phase image. In arterial-phase image, well-enhanced tumor is seen. In the equilibrium
phase, the tumor is seen as hypoattenuating mass relative to the surrounding liver. A mosaic pattern is
well visualized in arterial phase image
36 Y. Baba et al.

alternating areas of hyperintense enhance- A subset of so-called hypervascular tumors


ment and areas of necrosis (Figure 4.2). in the liver behaves as though the arterial
These lesions may demonstrate a peripher- blood supply of the tumors was greater
ally enhancing pseudocapsule, often best than that of the liver.
visualized during the equilibrium phase. Computed tomography imaging dur-
Necrotic lesions, regardless of their histology, ing the equilibrium phase of enhance-
will demonstrate poor arterial-phase enhan- ment results in decreased tumor detection.
cement. They are best evaluated with portal Contrast from the interstitial spaces of the
venous-phase or equilibrium images. liver diffuses into the liver tumor, which
When we should start scanning? In our is enhanced (Figure 4.3). The combina-
institute, the start of arterial-phase scan is tion of declining hepatic attenuation and
determined by monitoring the density of increasing attenuation of the tumor mass
aorta. The peak enhancement time of aorta may result in the tumor becoming nearly
varies from patient to patient, and cannot isoattenuating to the normal liver and not
be estimated from body weight or other detectable.
physical data.

TUMOR ARISING IN COMPUTED TOMOGRAPHY


NONCIRRHOTIC LIVER OR ANGIOGRAPHY
IN ONCOLOGIC PATIENTS Accurate knowledge of the hepatic arterial
anatomy in a transplant recipient is helpful
Most metastatic tumors, having a relatively
to the surgeon before liver transplantation
poor vascular supply, are not well detected
(Winter et al., 1995). With a thin slice
during arterial-phase. Such tumors, termed
thickness MDCT data set during a single
hypovascular tumors, are among the most
breath fold period, CT angiography of the
common tumors metastatic to the liver,
hepatic artery can be reconstructed, with
including adenocarcinomas from primary
promising results (Figure 4.4). Computed
tumors such as colon, pancreas, and gastric
tomography portography can also be
carcinomas. A uniformly enhancing rim on
reconstructed with the data set obtained
the periphery of a lesion strongly suggests
during the portal venous phase.
metastatic disease. This finding is also
present with hepatic abscesses, although
Angiography-Assisted Computed
the clinical presentation of the two enti-
Tomography
ties will be quite different. Hypervascular
tumors are seen best during the hepatic Hepatic CT that uses contrast material
arterial phase of enhancement as hyperat- administered through an indwelling arte-
tenuating masses compared with the sur- rial catheter is the most sensitive technique
rounding liver (Hollett et al., 1995). for detecting liver lesions (Heiken et al.,
For most hypovascular tumors, the dif- 1989; Matsui et al., 1985; Matsui, 1988).
ference in attenuation between the liver However, because it is an invasive proce-
and the tumor is maximized during the dure, this technique is not used for routine
portal phase of hepatic enhancement. hepatic screening, but to evaluate selected
4. Liver: Helical Computed Tomography and Magnetic Resonance Imaging 37

Figure 4.3. A case of multiple metastatic liver tumors from colon cancer. Left: Portal-phase image.
Right: Equilibrium-phase image. Tumors are hypointense relative to the surrounding liver on portal-phase
CT. In the equilibrium phase, contrast enhancement is seen within tumors, resulting from poor contrast
between the tumor and surrounding liver

patients who are under consideration for agent to the portal venous system through
resection or interventional treatment of pri-
a catheter in the superior mesenteric or
mary or secondary liver neoplasms. There splenic artery. Although 70% of the hepatic
are two types of angiography-assisted CT blood supply is from the portal system,
techniques for the evaluation of liver neo- hepatic neoplasms are largely supplied
plasms: CT during hepatic arteriography by the hepatic artery. As a result, hepatic
(CTHA) and CT during arterial portogra- neoplasms are detected as hypoattenuat-
phy (CTAP). Combined use of CTHA with ing lesions compared with the normal
CTAP may be of value in differentiating contrast-enhanced parenchyma.
malignant from benign hepatic defects but Careful pretreatment evaluation of
does not appear to improve lesion detection.patients who are going to receive hepatic
resection or interventional treatment is
essential to determine the precise loca-
tion and number of metastatic lesions for
COMPUTED TOMOGRAPHY accurate surgical planning (Matsui et al.,
DURING ARTERIAL 1987) or embolization therapy. Computed
PORTOGRAPHY tomography during arterial portography is
a highly sensitive technique for detecting
Computed tomography during arterial liver lesions and is superior to intravenous
portography involves CT scanning during contrast-enhanced CT for detecting lesions
selective delivery of an iodinated contrast less than 2 cm in diameter (Heiken et al.,
38 Y. Baba et al.

Figure 4.4. CT angiography of a AV shunt in the liver. Three-dimensional vascular images can be recon-
structed from the data of multislice helical CT. Left: Arterial-phase image shows large abnormal vascular
structure in the liver. Upper right: CT arteriography reconstructed from the arterial-phase CT images.
Lower right: Celiac angiography. Enlarged celiac artery and dilated hepatic vein is demonstrated

1989; Matsui et al., 1988; Deflandre et al., scanning. Multi detector computed tomog-
1994). The reported sensitivity of CTAP raphy is an excellent method for perform-
for lesion detection is among the highest ing rapid hepatic scanning. This method
of the available radiologic techniques, results in high levels of liver parenchymal
ranging from 81% to 91% (Heiken et al., and vascular enhancement that are readily
1989; Deflandre et al., 1994; Bluemke maintained over the duration of the scan.
et al., 1995). Rapid scanning during injec- Computed tomography during arterial
tion is important because recirculation of portography may be particularly problem-
contrast to the systemic circulation results atic in patients with cirrhosis, not only
in diminished contrast differences between because of fibrosis and altered hepatic
the neoplasm and normal parenchyma. architecture but also due to altered hemo-
Recirculation of contrast to the aorta is dynamics.
evident 35-s following the start of the con- In patients with portal hypertension, portal
trast injection, indicating the need for rapid venous blood may be shunted away from the
4. Liver: Helical Computed Tomography and Magnetic Resonance Imaging 39

liver through portosystemic collateral ves- imaging capability. The presence of vari-
sels, limiting hepatic parenchymal enhance- ous kinds of contrast agents, including
ment. In such patients, CTHA may provide extracellular agents, hepatocyte-specific
more useful information than CTAP. agents, and reticuloendothelial system-
specific agents, also has some advantages.
Because of the numerous choices of imag-
ing sequences and parameters, selection
COMPUTED TOMOGRAPHY
and optimization of pulse sequences is a
DURING HEPATIC critical issue in MR imaging of the liver.
ANGIOGRAPHY The current standard MR imaging exam-
ination of the liver includes a T1-weighted
Computed tomography during hepatic sequence, a T2-weighted sequence and a
angiography is performed with the cath- contrast-enhanced dynamic sequence.
eter in the hepatic artery [13, 14]. This The most frequent contrast administration
technique is superb in analyzing neovascu- approach is the use of a nonspecific extra-
larity of hepatic neoplasms. On CT images cellular contrast agent, gadolinium chelate,
obtained with this technique, malignant as a rapid-bolus injection with serial imag-
hepatic neoplasms appear as peripherally ing using a gradient echo sequence as
or homogeneously enhancing hyperdense dynamic MR imaging. In several circum-
masses compared with the normal hepatic stances, tissue-specific contrast agents such
parenchyma (Irie et al., 1995; Chezmar as superparamagnetic iron oxide (SPIO)
et al., 1993), although these tumors may are used not only for tumor detection but
appear hypoattenuating on conventional also for tumor characterization.
liver CT. In early hepatocellular carcino-
mas developed in a precancerous adenom-
atous hyperplasia, enhancement is seen in
DYNAMIC MAGNETIC
most areas of the mass, indicating portal-
dominant supply of the lesion, consistent RESONANCE IMAGING
with precancerous atypical adenomatous
With recent advances in fast MR imaging
hyperplasia on CTAP images. Small foci
technology, the entire liver can be imaged
with absence of portal blood flow show
in a breath-hold with a spoiled gradient
arterial-dominant blood supply on CTHA.
echo sequence. The sequence is repeated
several times after administration of gado-
linium chelates (Figure 4.5). It is usually
MAGNETIC RESONANCE advisable to use an in-phase echo time on
IMAGING spoiled gradient echo images for contrast-
enhanced studies in order to avoid confus-
With the advent of the fast magnetic reso- ing variations in signal intensity based on
nance (MR) imaging technique, this has fatty infiltration of the liver and black-ring
become the modality of choice in some phase cancellation artifacts that will mask
patients for imaging of the liver. An advan- capsular-based disease.
tage of MR imaging compared with CT is In current clinical practice, gadolinium
its excellent tissue contrast and multiplanar chelates are frequently used for dynamic
40 Y. Baba et al.

Figure 4.5. A case of hepatocellular carcinoma after lipiodol TAE. Upper left: Precontrast CT. Upper right:
T1-weighted gradient echo image. Lower left: Arterial-phase MR imaging. Lower right: Equilibrium-
phase MR image. Tumor is visualized as significant hyperintense relative to the surrounding liver because
of lipiodol accumulation in the tumor. It is difficult to evaluate tumor vascularity using dynamic CT. The
tumor is seen as hypointense relative to the surrounding liver on T1-weighted MR images. Lipiodol in the
tumor does not affect signal intensity of the tumor. On arterial-phase image, well-enhanced tumor is seen.
Tumor is not as conspicuous in the equilibrium-phase image as in the arterial-phase image. Peripherally
enhancing pseudocapsule is well visualized

study as iodinated contrast agents on CT visually similar to iodine contrast enhance-


(Hamm et al., 1994; Yamashita et al., ment on CT images, and a similar phase of
1994, 1996). Gadolinium chelates, that enhancement of the liver is seen as in CT.
are nonspecific extracellular space agents,
are distributed initially within the intra-
vascular compartment and then diffuse SUPERPARAMAGNETIC
rapidly throughout the extracellular space IRON OXIDE
in a distribution similar to that of iodinated
contrast agents on CT. These agents cause Super paramagnetic iron oxide (e.g., feru-
T1 shortening of adjacent water molecules, moxides) is a crystalline compound that is
which results in an increase in signal inten- taken up selectively by the Kupffer cells
sity on T1-weighted images. Therefore, the of the liver. It markedly shortens the T2
pattern of enhancement with gadolinium is relaxation, resulting in a decrease in signal
4. Liver: Helical Computed Tomography and Magnetic Resonance Imaging 41

from normal liver tissue. Because the agent hemangioma, which also shows a decrease
is not taken up by tumors, hepatic masses in signal intensity on T2-weighted images.
stand out as hyperintense foci in the low- Some well-differentiated hepatocellular
signal intensity liver on proton density carcinomas and adenomatous hyperplasias
or T2/T2*-weighted images (Figure 4.6). may also show accumulation of this agent
Studies performed at 0.6 and 1.5 T have within the tumor. Therefore, in early hepa-
demonstrated an increased tumor detec- tocellular carcinomas that developed in a
tion rate (Tsang et al., 1988; Ros et al., precancerous adenomatous hyperplasia,
1995), and other studies have shown a signal decrease is seen in most areas of
increased lesion-to-liver contrast (Stark atypical adenomatous hyperplasia, indicat-
et al., 1988; Winter et al., 1993) compared ing the presence of Kupffer cells in this
with unenhanced imaging. Although most area. Small foci of signal hyperintensity
people may agree that this agent is effec- within the lesion may be present, indicat-
tive in detecting metastatic tumors, its role ing the absence of Kupffer cells, consistent
in detecting hepatocellular carcinomas is with malignant foci developed in atypical
questionable. The role of SPIO in char- adenomatous hyperplasia. Delineation of
acterizing focal liver lesions is limited. the clinical role of iron-containing hepatic
Some studies reported SPIO uptake in contrast agents for tumor characterization
lesions containing Kupffer cells, such as requires further testing.
focal nodular hyperplasia, and in lesions In conclusion, with modern imaging
in which blood pooling occurs, such as technologies, including MDCT and fast

Figure 4.6. A case of metastatic liver tumor from colon carcinoma. Left: Pre-SPIO T2-weighted fast spin
echo image. Right: Post-SPIO T2-weighted fast spin echo image. Tumor is hyperintense relative to the
surrounding liver on precontrast T2 image. After administration of SPIO, the signal intensity of the liver
parenchyma is decreased, resulting in better lesion-to-liver contrast on the postcontrast images
42 Y. Baba et al.

MR imaging, tumor detection is possible rial portography, and MR imaging. Radiology


in a noninvasive manner. Lesion charac- 171:47–51.
Hollett, M.D., Jeffrey, R.B.J., Nino-Murcia, M.,
terization is also possible in the majority
Jorgensen, M.J., and Harris, D.P. 1995. Dual-
of cases. Three-dimensional evaluation is phase helical CT of the liver: Value of arterial
feasible with fast imaging techniques such phase scans in the detection of small (1.5 cm)
as MDCT. With tissue-specific MR con- malignant hepatic neoplasms. Am. J. Roentgenol.
trast agents such as SPIO, our capability 164:879–884.
is enhanced not only with respect to lesion Hosoki, T., Chatani, M., and Mori, S. 1982. Dynamic
computed tomography of hepatocellular carci-
detection but also lesion characterization.
noma. Am. J. Roentgenol. 139:1099–1106.
We hope that these imaging techniques are Irie, T., Takeshita, K., Wada, Y., Kusano, S.,
properly applied in clinical practice. Terahata, S., Tamai, S., Hatsuse, K., Aoki, H.,
and Sugihara, Y. 1995. CT evaluation of hepatic
REFERENCES tumors: Comparison of CT with arterial portog-
raphy, CT with infusion hepatic arteriography,
Baron, R.L., Oliver, J., Dodd, G., Nalesnik, M., and simultaneous use of both techniques. Am. J.
Holbert, B.L., and Carr, B.V. 1996. Hepatocellular Roentgenol. 164:1407–1412.
carcinoma: Evaluation with biphasic, contrast- Matsui O. 1988. Hepatic metastasis detection:
enhanced, helical CT. Radiology 199:505–511. Comparison of three CT contrast enhancement
Bluemke, D.A., Urban, B., and Fishman, E.K. methods (letter). Radiology 168:283–284.
1994. Spiral CT of the liver: Current applica- Matsui, O., Takashima, T., Kadoya, M., Ida, M.,
tions. Semin. Ultrasound CT MR 15:107–121. Suzuki, M., Kitagawa, K., Kamimura, R., and
Bluemke, D.A., Soyer, P.A., Chan, B.W., Bliss, Inoue, K. 1985. Dynamic computed tomography
D.F., Calhoun, P.S., and Fishman, E.K. 1995. during arterial portography: The most sensitive
Spiral CT during arterial portography: Technique examination for small hepatocellular carcino-
and applications. Radiographics 15:623–637. mas. J. Comput. Assist. Tomogr. 9:19–24.
Bonaldi, V.M., Bret, P.M., Reinhold, C., and Matsui, O., Takahashi, S., Kadoya, M., Suzuki, M.,
Atri, M. 1995. Helical CT of the liver: Value Hirose, J., Kameyama, T., Choto, S., Konishi, H.,
of an early hepatic arterial phase. Radiology Ida, M., and Yamaguchi, A. 1987. Liver metas-
197:357–363. tases from colorectal cancers: Detection with
Chezmar, J.L., Bernardino, M.E., Kaufman, S.H., CT during arterial portography. Radiology 165:
and Nelson, R.C. 1993. Combined CT arterial 65–69.
portography and CT hepatic angiography for Ros, P.R., Freeny, P.C., Harms, S.E., Seltzer, S.E.,
evaluation of the hepatic resection candidate. Davis, P.L., Chan, T.W., Stillman, A.E., Muroff,
Work in progress. Radiology 189:407–410. L.R., Runge, V.M., and Nissenbaum, M.A. 1995.
Deflandre, M.F., Vilgrain, V., Zins, M., Najmark, D., Hepatic MR imaging with ferumoxides: A mul-
Arrive, L., Vullierme, M.P., and Nahum, H. ticenter clinical trial of the safety and efficacy in
1994. Nontumorous attenuation changes on CT the detection of focal hepatic lesions. Radiology
arterial portography. J. Comput. Assist. Tomogr. 196:482–488.
18:761–767. Stark, D.D., Weissleder, R., Elizondo, G., Hahn,
Hamm, B., Thoeni, R.F., Gould, R.G., Bernardino, P.F., Saini, S., Todd, L.E., Wittenberg, J., and
M.E., Luening, M., Saini, S., Mahfouz, A.E., Ferrucci, J.T. 1988. Superparamagnetic iron
Taupits, M., and Wolf, K.J. 1994. Focal liver oxide: Clinical application as a contrast agent
lesions: Characterization with nonenhanced and for MR imaging of the liver. Radiology 168:
dynamic contrast material-enhanced MR imag- 297–301.
ing. Radiology 190:417–423. Tsang, Y.M., Stark, D.D., Chen, M.C., Weissleder,
Heiken, J.P., Weyman, P.J., and Lee, J.K.T. 1989. R., Wittenberg, J., and Ferrucci, J.T. 1988.
Detection of focal hepatic masses: Prospective Hepatic micrometastases in the rat: Ferrite-
evaluation with CT, delayed CT, CT during arte- enhanced MR imaging. Radiology 167:21–24.
4. Liver: Helical Computed Tomography and Magnetic Resonance Imaging 43

Winter, T.C., Freeny, P.C., Nghiem, H.V., Mack, L.A., Yamashita, Y., Yamamoto, H., Arakawa, A.,
Patten, R.M., Thomas, C.R., and Elliott, S. 1993. Matsukawa, T., Hatanaka, Y., and Takahashi, M.
MR imaging with i.v. superparamagnetic iron 1994. Differential diagnosis of liver neoplasms:
oxide: Efficacy in the detection of focal hepatic Role of dynamic MR imaging. Radiology
lesions. Am. J. Roentgenol. 161:1191–1198. 193:59–65.
Winter, T.C., Freeny, P.C., Nghiem, H.V., Yamashita, Y., Mitsuzaki, K., Yi, T., Ogata, I.,
Hommeyer, S.C., Barr, D., Croghan, A.M., Nishiharu, T., Urata, J., and Takahashi, M. 1996.
Coldwell, D.M., Althans, S.J., and Mack, L.A. Small hepatocellular carcinoma in patients with
1995. Hepatic arterial anatomy in transplantation chronic liver damage: Prospective comparison of
candidates: Evaluation with three-dimensional detection with dynamic MR imaging and helical
CT arteriography. Radiology 195:363–370. CT of the whole liver. Radiology 200:79–84.
A. Diagnosis
5
Selection of Patients for Resection
of Hepatic Colorectal Metastases:
18F-Fluorodeoxyglucose/Positron
Emission Tomography
Rebecca Auer and Yuman Fong

INTRODUCTION hepatic resection for metastatic colorectal


cancer for three reasons. First, if preop-
The selection of appropriate patients for erative workup with PET scanning detects
surgical resection of colorectal liver metas- more extrahepatic disease than conven-
tases is a central clinical dilemma. Using tional imaging alone, surgery would not be
conventional imaging, up to one quarter undertaken in these patients, and the group
of patients scheduled for surgical resection of patients undergoing surgery would have
will be determined to have unresectable or a more favorable prognosis; a concept
metastatic disease at the time of surgery. similar to the Will-Rogers effect, which is
Of the patients who do undergo a resection used to describe stage migration. Second,
with curative intent, ~ 60% will develop a if hepatic recurrences are detected ear-
recurrence within 3 years, either within the lier in patients screened with PET scan-
liver or in an extrahepatic location (Bines ning as compared to conventional imaging
et al., 1996). It is clear that a more effective alone, surgery would be performed earlier
preoperative staging modality is needed to and survival, as defined from the time of
avoid unsuccessful or ineffective surgeries. metastasectomy, would be longer, a con-
Positron emission tomography (PET) cept termed lead-time bias. Third, if hepatic
using [18F]fluorodeoxyglucose (FDG) is recurrences are detected earlier with PET
emerging as a promising diagnostic modal- scan, surgery might be undertaken prior
ity in metastatic colorectal cancer. [18F] to the development of additional systemic
fluorodeoxyglucose-positron emission micrometastases. Based on tumor biology,
tomography (FDG-PET) has the poten- this results in a true improvement in survival.
tial to differentiate between patients The benefit of PET scanning is most
with resectable and unresectable hepatic likely related to the ability to find more
colorectal cancer metastases and therefore extrahepatic disease. This review will focus
improve patient selection for surgery with on the available evidence for the ability of
a positive effect on surgical treatment PET scanning to improve the selection of
outcomes, including survival. Positron patients for hepatic resection, and thereby
emission tomography may improve the improve the prognosis of resected patients.
survival time in patients undergoing Topics to be outlined include the sensitivity

49
50 R. Auer and Y. Fong

and specificity of PET scans to detect scanning (i.e., blinded to the CT result)
intrahepatic and extrahepatic lesions. The or the incremental value of PET scan fol-
correlation of PET scans with the preop- lowing CT. These studies have attempted
erative clinical risk score (CRS), and the to define a role for PET scanning in this
impact on surgical management. These specific cohort of patients. A meta-analysis
data will be used to argue that the ability of these studies has also been published
to detect and eliminate patients with unre- providing a concise summary of the data
sectable metastatic colorectal cancer may available to date (Wiering et al., 2005).
also translate into improved survival post- A more general overview on the use of PET
hepatic resection. The benefit of PET dur- scanning in screening patients with colon
ing follow-up, particularly in patients with cancer for local and distant recurrences can
an undiagnosed elevation in carcinoem- be found in other chapters in this volume.
bryonic antigen (CEA) or a suspected
recurrence post-hepatectomy will also be
discussed. Finally, a preoperative staging POSITRON EMISSION
algorithm that incorporates the available
TOMOGRAPHY SCAN
information regarding PET scanning will
be presented. FOR THE DETECTION OF
EXTRAHEPATIC DISEASE
Extrahepatic disease is the most significant
POSITRON EMISSION
operative finding related to adverse long-
TOMOGRAPHY SCANNING term survival (Rosen et al., 1992). The
AS A STAGING MODALITY existence of extrahepatic disease should
TO COMPLEMENT be considered a contraindication for hepa-
CONVENTIONAL IMAGING tectomy, aside from isolated portal lymph
node metastases (Jaeck et al., 2002) or
Although there are no published rand- limited resectable pulmonary metastases
omized controlled trials on the use of FDG- (DeMatteo et al., 1999).
PET in patients with metastatic colorectal The ability of PET scans to detect previ-
cancer to the liver, a number of prospective ously unrecognized extrahepatic disease is
and retrospective studies have attempted to the most compelling reason for use in the
evaluate the ability of PET scan to improve preoperative setting for patients with
the staging of patients in the preoperative hepatic metastases. Numerous studies have
workup for hepatic resection of colorectal demonstrated the superiority of PET scans
metastases. All of these studies included a (Wiering et al., 2005) as compared to
cohort of patients who presented for evalu- conventional CT scans in this setting. The
ation of suspected colorectal cancer recur- sensitivity of a PET scan is 91.5% (range
rence to the liver based on conventional from 63% to 100%) compared to 60.9%
imaging, such as computed tomography (range 25–86%) (Wiering et al., 2005).
(CT) or, in some cases, an elevated CEA Whereas CT scans have a specificity of
level. These studies compare the relative 95.4% as compared to 91.9% with PET
value of PET scans in staging a patient for (Wiering et al., 2005). [18F]fluorodeoxy-
hepatic resection; in the replacement of CT glucose-positron emission tomography
5. Selection of Patients for Resection of Hepatic Colorectal Metastases 51

(FDG-PET) is particularly helpful in within the liver compared to extrahepatic


nodal involvement, differentiating malig- disease as normal hepatocytes contain a
nant nodes from post-surgical change, and high hexokinase activity and therefore
evaluating malignancy of indeterminate incorporate a significant amount of 18FDG
pulmonary nodules (Truant et al., 2005). (Fong et al., 1999a). Conventional preop-
Evaluation of PET scan alone without a erative imaging to detect hepatic metas-
corresponding CT scan is associated with a tases includes CT and CT-portography.
number of drawbacks, particularly relating In prospective, blinded studies comparing
to the poor resolutions of PET imaging FDG-PET to conventional CT scans in
patients with colon cancer being evaluated
(Selzner et al., 2004). It is difficult to evalu-
ate the exact location of FDG uptake. This for hepatic resection, the detection of liver
may result in misinterpretation of a lesion metastases was equivalent or superior with
as intrahepatic, when it is actually extrahe- conventional CT scans (~ 80%) as com-
patic or vice-versa. In addition, obtaining pared to PET scans (65–80%) (Truant et al.,
histological confirmation of malignancy 2005). When directly compared to CT por-
in an area of increased FDG uptake may tography in a similar population, CT portog-
be impossible without anatomical infor- raphy had a higher or equivalent sensitivity
mation. In one study following blinded (97% versus 90% with PET), but a lower
interpretation of the CT and PET scans, a specificity, resulting in a lower accuracy
rereview of the CT scans was performed with CT portography (76–80%) as com-
for those patients with discordant and pared to PET scan (92–93%) (Fong et al.,
positive FDG-PET results. Of seven PET 1999b). When evaluated in conjunction
positive/CT negative lesions, four could with CT portography, PET scans appear
be detected on rereview, emphasizing the to be sensitive at distinguishing between
importance of combined reading of CT and benign and malignant intrahepatic lesions
PET images in daily practice (Ruers et al., based on the magnitude of the standard-
2002). The combined fusion technique of ized uptake value (SUV) (Figure 5.2). In
PET/CT will best address the shortcomings two separate analyses of the magnitude
of both modalities, however, at present the of SUV in hepatic colorectal metastases
CT resolution in the combined modality is (total of 144 lesions), the SUV for all but
not comparable to that of a standard CT. 1 malignant lesions was > 3.5, and only 2
benign lesion had an SUV > 3.5 (Delbeke
et al., 1997).
POSITRON EMISSION When compared directly to intraopera-
TOMOGRAPHY SCAN tive ultrasound and surgical pathology,
FOR THE DETECTION OF PET scans have a sensitivity of 71–78%
INTRAHEPATIC DISEASE with the probability of lesion detection
directly correlated with lesion size (Rohren
The utility of PET scans to evaluate the et al., 2002). Positron emission tomogra-
number and distribution of lesions in the phy scanning will detect between 25–40%
liver and therefore help select patients with of lesions ≤ 1 cm and 90% of lesions ≥
surgically resected disease has also been 3 cm (Rohren et al., 2002). The assess-
studied (Figure 5.1). Such scanning is gen- ment of lobar disease distribution was also
erally thought to be less sensitive for lesions found to be discordant between PET scans
52 R. Auer and Y. Fong

Figure 5.1. A patient with bilobar, multiple hepatic


metastases treated with neoadjuvant chemotherapy
of FOLFOX with good response and shrinking of
all the lesions. There are still residual tumors in the
liver by CT scan Figure 5.2. A patient with bilobar, multiple hepatic
metastases treated with neoadjuvant chemotherapy
of FOLFOX with good response and shrinking of
and surgical findings in 13% of patients, all the lesions. One lesion still took up a significant
frequently underestimating the extent of amount of FDG and this lesion was considered to be
disease (Rohren et al., 2002). The authors chemoresistant (black arrow). The prognostic value
of this study concluded that PET scans of a decrease in SUV following chemotherapy
is still being evaluated
lack the anatomic definition and sensi-
tivity, particularly with small lesions, to
replace surgical exploration when evaluat- clinical management of only one patient
ing resectability in patients with colorectal (2%) (Rydzewski et al., 2002).
liver metastases (Rohren et al., 2002).
The added value of intraoperative ultra-
VALUE OF POSITRON
sound may be limited when patients are
evaluated preoperatively with PET scan. EMISSION TOMOGRAPHY
In a study of 47 patients who underwent (PET) CORRELATED TO
surgical exploration for curative resec- PROGNOSTIC CLINICAL RISK
tion of hepatic colorectal metastases, the SCORE (CRS)
positive predictive value of FDG-PET was
93% as compared to 87% for intraopera- A number of clinical and pathological
tive sonography (Rydzewski et al., 2002). parameters have been found to be inde-
More importantly, the findings of the intra- pendent predictors of prognosis by robust
operative ultrasound led to a change in the multivariate analysis (Fong et al., 1999a).
5. Selection of Patients for Resection of Hepatic Colorectal Metastases 53

These parameters include: (1) the presence evidence of additional disease detected
of extrahepatic disease at laparotomy, (2) by PET (Schussler-Fiorenza et al., 2004).
a positive resection margin, (3) nodal The authors concluded that patients with a
metastases from primary cancer, (4) a short CRS of 0 should not undergo preoperative
disease-free interval, (5) largest tumor evaluation with a PET scan because the
> 5 cm, (6) more than one liver metasta- yield is too low to justify the expense and
sis, and (7) a CEA over 200 ng/ml. Using morbidity associated with false-positive
the last five criteria, a preoperative CRS results (Schussler-Fiorenza et al., 2004).
system was created with each positive
criterion counting as one point. This CRS
is a simple, easily remembered staging POSITIVE IMPACT OF
system for classifying patients with liver- POSITRON EMISSION
exclusive metastatic colorectal cancer. TOMOGRAPHY ON THE
The presence of any one of these charac- MANAGEMENT OF PATIENTS
teristics was still associated with a 5-year WITH LIVER METASTASES
survival of 24–34%, and therefore no single
criterion can be considered a contraindica- Positron emission tomography (PET)
tion to resection (Fong et al., 1999a). It is scans have repeatedly shown the ability to
the total score out of 5 that is highly predic- alter clinical treatment plans when used
tive of outcome. A score of 2 or less places for preoperative staging in patients with
a patient in a good prognostic group, for hepatic colorectal metastases. In a well
whom resection is ideal. For scores of 3 or designed prospective study evaluating the
4, outcome is less favorable and patients added benefit of CT/PET scan to CT scan
should be considered for aggressive tri- alone, 76 patients with hepatic colorectal
als of adjuvant therapy. For a score of 5, metastases were imaged preoperatively
long-term disease-free survivors are rarely (Selzner et al., 2004). In this study a pre-
encountered and resections in this high risk PET treatment plan was prospectively
group should be accompanied by trials of defined and compared to the treatment
adjuvant therapy (Fong et al., 1999a). plan outlined after consideration of the
This CRS is also now being used to results of the CT/PET scan (Selzner et al.,
help select the extent and sophistication 2004). In 60 patients (79%) PET/CT did
of preoperative assessment, including the not change the therapeutic strategy, and
use of FDG-PET. The ability of the CRS in 10 patients (13%) considered resect-
to predict the yield of a preoperative PET able by CT alone, the PET/CT demon-
scan was demonstrated in a subset of 63 strated extensive extrahepatic disease,
patients presenting with a first occur- representing a contraindication to surgery
rence of hepatic colorectal metastases (Selzner et al., 2004). In the remaining
(Schussler-Fiorenza et al., 2004). Among six patients (8%) the surgical strategy
patients with a CRS of 0, no patient and extent of resection was changed
had extrahepatic disease detected by PET because the PET/CT demonstrated pre-
and 57% showed a false-positive reading viously unrecognized positive nodes in
(Schussler-Fiorenza et al., 2004). Among the hepatoduodenal ligament (Selzner
patients with a CRS of ≥ 1, 14% had et al., 2004). Taken together, PET/CT
54 R. Auer and Y. Fong

provided information that significantly OUTCOME OF PATIENTS


altered the surgical treatment plan in SELECTED FOR HEPATIC
> 20% of the patients, including the RESECTION FOLLOWING [18F]
avoidance of morbidity associated with
an exploratory laparotomy/laparoscopy in
FLUORODEOXYGLUCOSE-
> 10% of patients (Selzner et al., 2004). POSITRON EMISSION
These results compare favorably to other TOMOGRAPHY (FDG-PET)
similarly designed studies where clinical
management was directly altered in Survival following hepatic resection for
20–58% of patients (Truant et al., 2005). colorectal liver metastases depends on the
stringency of the patient selection crite-
ria. In order to gain a long-term survival
NEGATIVE IMPACT OF benefit, surgical candidates must have
POSITRON EMISSION hepatic metastases that can be resected
TOMOGRAPHY ON THE with a negative margin and no evidence
MANAGEMENT OF PATIENTS of extrahepatic disease. As PET scanning
WITH LIVER METASTASES appears to be more sensitive at detect-
ing extrahepatic recurrences, this should
Although PET scans clearly have an advan- eliminate patients unlikely to benefit from
tage over conventional CT scans by avoid- surgery and improve the overall survival of
ing unnecessary laparotomy in unresectable patients who do undergo curative resection
patients, the potentially harmful impact of of hepatic colorectal metastases.
PET scans should not be overlooked. The Although there are no studies that
negative impact may result from a false- directly compare the survival results of
negative, or possibly more disconcerting, contemporary patients who underwent
a false-positive. The false-negative rate for resection with or without a preoperative
detection of extrahepatic disease appears to FDG-PET scan, one group published sur-
range from 0% to 17% (Truant et al., 2005), vival results in a cohort of patients who
and was generally explained by tumors < were evaluated with preoperative PET
1 cm in size, proximity of tumors to adja- scans and compared the results to his-
cent lesions, preoperative chemotherapy, or torical controls (Fernandez et al., 2004;
hypocellular mucinous tumors. The sensitiv- Strasberg et al., 2001). In their first pub-
ity of PET scans appears to be particularly lication in 2001, Strasberg et al. (2001)
limited for peritoneal metastases with a sen- found that 43 patients with metastatic
sitivity as low as 63% in one series (Truant colorectal cancer were referred for hepatic
et al., 2005). The false-positive rate ranged resection after conventional tumor stag-
from 0% to 6% (Truant et al., 2005) and was ing with a CT scan. FDG-PET identified
secondary to inflammation, infection or a additional cancer in ten patients, in whom
secondary malignancy, such as lymphoma. surgery was contraindicated in six (14%).
Although the false-positive rate is small, it Laparotomy was performed in 37 patients
is not negligible. In patients in whom this with a curative resection undertaken in all
information would represent a contraindica- but 2 patients (95%), each of whom had a
tion to surgery, a biopsy to confirm meta- single small focus of extrahepatic disease
static disease is usually warranted. (Strasberg et al., 2001). The disease-free
5. Selection of Patients for Resection of Hepatic Colorectal Metastases 55

survival estimate at 3 years was 40%, likely to have extrahepatic disease detected
higher than the previously reported results by FDG-PET and therefore considered
of 15–28% (Strasberg et al., 2001). unresectable (Strasberg et al., 2001). In
In a follow-up study, 100 patients who the subsequent analysis of survival follow-
underwent complete hepatic resection for ing hepatic resection among this selected
curative intent were evaluated by preoperative PET negative population, there is loss of
FDG-PET and CT scans, and were followed significance for the prognostic indicators,
for a median of 31 months (Fernandez et al., reflecting the added information obtained
2004). This study did not include patients by preoperative PET scanning.
who underwent an exploratory laparotomy
but were found to be unresectable; therefore,
it is not possible to comment on the ability of STANDARD UPTAKE
PET scans to prevent unnecessary laparoto- VALUE AND PREDICTING
mies from this study. The Kaplan-Meier esti- PROGNOSIS OR RESPONSE
mate of 5-year overall survival in this group TO THERAPY
of patients was 58.6% (95% CI of 46–72%)
which compares favorably to the results A fundamental metabolic difference
of 19 previously reported series where the between normal and malignant cells
estimated 5-year overall survival was 30% relates to the cells primary energy source.
(range 12–41%) (Fernandez et al., 2004). Whereas a normal cell derives most of its
One criticism of this study is that in com- energy from oxidative phosphorylation, a
paring to historical controls, the influences rapidly growing tumor cell derives more
of improved surgical techniques and new than half of its energy from glycolysis,
chemotherapeutic agents which might have with only a minor amount from the citric
contributed to the improved results are not acid cycle (Nakashima et al., 1984). The
considered. The authors acknowledge the glycolytic intermediates are used not only
potential for these confounding variables but to provide energy, but also to intermedi-
state that only 4% of the patients received ate in the synthesis of DNA/RNA and
irinotecan as adjuvant treatment and that membrane lipids. The glycolytic capacity
oxaliplatin was not yet available by the close of tumor cells has been demonstrated in a
of the study (Fernandez et al., 2004). number of studies to correlate with lack
It is interesting to note that in this study of differentiation of these cells (Pauwels
none of the previously established prog- et al., 2000), and when cells undergo
nostic factors relating to the hepatic metas- oncogenic transformation, their glycolytic
tasis, including the components of the activity increases. Many human tumors
CRS, were significant predictors of worse including colon cancers, have demon-
outcome (Fernandez et al., 2004). This strated increased expression and activity
suggests that PET scanning represents a of several glucose transporters (GLUT 1
surrogate for previously identified clinical and 3) and the key enzyme hexokinase
variables that make up the CRS. This (reviewed in Pauwels et al., 2000).
observation may be related to the fact that Positron emission tomography scan-
PET scans preferentially exclude patients ning has the ability to give functional
with multiple, bilateral, and synchronous information about a tumor and may be
tumors because such patients are more able to predict the prognosis or response
56 R. Auer and Y. Fong

to treatment on this basis. In order to of the pretreatment value) (Findlay et al.,


assess the response to 5-FU based chem- 1996). In patients receiving FOLFOX as
otherapy, FDG uptake on a PET scan was a second-line treatment of hepatic color-
measured in colorectal liver metastases ectal metastases, a reduction in SUV
before and at 2 and 4 weeks after treat- was similarly predictive of a > 1-year
ment and compared to tumor dimension survival (Dimitrakopoulou-Strauss et al.,
measured on a CT scan performed at 2003). These findings are in agreement
12 weeks (Findlay et al., 1996). In this with the ability of PET to predict patho-
study the pretreatment SUV did not cor- logical response to chemotherapy and
relate with response, but at 4 weeks the survival in patients receiving neoadju-
responding lesions had a significantly vant treatment in rectal cancer (Guillem
greater reduction in metabolism than et al., 2004) and in a number of other
non-responding lesions (99% versus 67% malignancies (Young et al., 1999).

a c

Figure 5.3. A patient with bilobar, multiple hepatic metastases treated with neoadjuvant chemotherapy
of FOLFOX with good response and shrinking of all the lesions. The lesions almost completely disap-
peared by PET scan (white arrow) suggesting a good response to chemotherapy. One lesion still took
up a significant amount of FDG and this lesion was considered to be chemoresistant (black arrow). The
prognostic value of a decrease in SUV following chemotherapy is still being evaluated
5. Selection of Patients for Resection of Hepatic Colorectal Metastases 57

The ability of chemotherapy to decrease THE ROLE OF POSITRON


tumor FDG uptake must be taken into EMISSION TOMOGRAPHY IN
consideration when evaluating a patient EVALUATING AN ELEVATED
for surgery following neoadjuvant treat-
ment (Figures 5.1–5.3). A recent pro-
CARCINOEMBRYONIC
spective study evaluating the detection of ANTIGEN (CEA)
colorectal hepatic metastases found that in
Elevation of serum CEA levels may be the
patients treated with neoadjuvant chemo-
earliest sign of colorectal cancer recur-
therapy, 37% of lesions were missed by
rence and, therefore, CEA is used for
FDG-PET, as compared to 23% of lesions
postoperative surveillance. Conventional
in patients who received no preoperative
diagnostic imaging does not lead to locali-
chemotherapy (Akhurst et al., 2005). In
zation of the site of recurrence in as many
addition, while no malignant hepatic lesion
as 8% of patients resulting in the delay of
> 1.2 cm was missed in the later group, a
a potentially curative surgery (Flamen et
lesion as large as 3.2 cm was missed in
al., 2001). Several studies have evaluated
the former group (Akhurst et al., 2005).
the additive value of PET scan in patients
The phosphorylating activity of hexoki-
with an increase in CEA during surveil-
nase in the tumor was also significantly
lance and normal or equivocal findings on
lower in patients treated preoperatively
CT scan (Flamen et al., 2001). In the larg-
with chemotherapy, providing a rationale
est study, 50 patients with elevated CEA
for the decrease in sensitivity of FDG-PET
during postoperative surveillance and a
(Akhurst et al., 2005). Although the disap-
normal or equivocal conventional diagnos-
pearance of a tumor by FDG-PET may
tic workup underwent FDG-PET imaging
indicate a good prognosis, it is clear from
(Flamen et al., 2001). A total of 15 patients
this study that it cannot be equated to a
were determined to have hepatic metas-
pathological complete response.
tases by histology or long-term follow-
up, and among this group PET detected
12 lesions, but only 5 were ultimately
POSITRON EMISSION resected for cure (Flamen et al., 2001).
TOMOGRAPHY SCANNING The PET scan also detected three lesions
FOR SURVEILLANCE AND in the liver that were not malignancies
FOLLOW-UP (20% false-positive) (Flamen et al., 2001).
These findings are in agreement with other
The role of PET scanning in screening all smaller studies and when taken together
colon cancer patients for recurrence and can indicate that in the subset of patients
metastases is discussed in more detail in with an elevated CEA and no or equivocal
other chapters in this volume. There are findings on CT scan, ~ 10% of the entire
two areas, however, unique to patients cohort will have resectable hepatic metas-
being evaluated for hepatic colorectal tases detected by PET (Flanagan et al.,
metastases that deserve mention; patients 1998). Overall, PET in this setting has a
with an elevated CEA and no lesion on positive predictive value of close to 90%
conventional imaging, and patients being with false-positives in 10–40% of patients
followed-up post-hepatic resection. (Flanagan et al., 1998). These studies
58 R. Auer and Y. Fong

however, fail to document the number of only one patient had a false-positive (PET
invasive procedures, such as biopsies that specificity 89%) (Selzner et al., 2004). In
PET scans either avoid or mandate, and the another blinded study, PET had a diag-
associated morbidity and costs. Moreover, nostic accuracy of 86% in differentiat-
these studies cannot claim that a PET scan ing intrahepatic postsurgical change from
is a superior test to evaluate for recurrent malignant recurrence, as compared to 45%
disease in the setting of an elevated CEA for CT portography (Delbeke et al., 1997).
during surveillance, because these studies The significant difference is the result of
evaluated PET with full knowledge of the more false-positives with CT portography.
results of conventional imaging.
ALGORITHM TO INCLUDE
THE ROLE OF POSITRON POSITRON EMISSION
EMISSION TOMOGRAPHY TOMOGRAPHY SCANNING
IN FOLLOW-UP POST IN WORK-UP OF HEPATIC
HEPATIC RESECTION COLORECTAL METASTASES
The most common site for recurrence after The preoperative staging prior to hepatic
resection of hepatic colorectal metastases resection of colorectal metastases must be
is the liver, and it is the sole site of recur- meticulous to avoid unexpected findings or
rence in up to 40% of cases (Taylor, 1996). obstacles at the time of laparotomy. A sys-
In the absence of extrahepatic disease and tematic approach to the colorectal cancer
in a patient with adequate hepatic reserve, patient who is referred for consideration of
a repeat hepatectomy may be considered. hepatic resection can facilitate a thorough
In approximately one third of patients the patient work-up and minimize unnecessary
recurrence will be amenable to further investigations. Patients should initially be
resection with a 5-year overall survival evaluated with a high quality CT scan of the
range from 31–41% (Bozzetti et al., 1992). chest abdomen and pelvis to identify hepatic
Positron emission tomography/com- metastases as well as local recurrence and
puted tomography (PET/CT) imaging may lung metastases. In patients with an equiv-
be superior to conventional CT in estab- ocal finding on CT scan or a persistently
lishing the diagnosis of intrahepatic recur- elevated CEA and a normal CT scan, PET
rence in patients with prior hepatectomy. scanning is warranted and may delineate
In a prospective study of the impact of the site of metastasis and/or demonstrate
FDG-PET on treatment in patients with additional extrahepatic disease. In patients
colorectal liver metastases, a subset of with a documented suspicious lesion in
18 patients with previous hepatic resec- the liver on a CT scan, a PET scan is
tion had a suspected local recurrence by best reserved for patients with a CRS ≥ 2
CT scan (Selzner et al., 2004). A total of because these patients will have the highest
nine patients had an intrahepatic recur- incidence of extrahepatic disease that may
rence, resulting in specificity of 50% for impact on the clinical management.
CT scans (Selzner et al., 2004). By com- As previously mentioned, PET scans may
parison, all nine patients were positive by have a false-negative rate as high as 17%
FDG-PET (PET sensitivity 100%) and (Truant et al., 2005), and this is frequently
5. Selection of Patients for Resection of Hepatic Colorectal Metastases 59

related to small peritoneal metastases. tion for recurrent metastases from colorectal
Laparoscopy is, therefore, still indicated in cancer. Br. J. Surg. 79:146–148.
Delbeke, D., Vitola, J.V., Sandler, M.P., Arildsen,
patients with a CRS of > 2 because these
R.C., Powers, T.A., Wright, J.K. Jr., Chapman,
patients have a high incidence of peritoneal W.C., and Pinson, C.W. 1997. Staging recurrent
metastases that would represent a contrain- metastatic colorectal carcinoma with PET. J.
dication to resection (Jarnagin et al., 2001). Nucl. Med. 38:1196–1201.
Finally, because false-positives can occur, DeMatteo, R.P., Minnard, E.A., Kemeny, N.,
a biopsy is warranted to confirm the pres- Downey, R., Burt, B.M., Fong, Y., and Blumgart,
L.H. 1999. Outcome after resection of both liver
ence of metastatic malignancy in patients
and lung metastases in patients with colorectal
with PET evidence of extrahepatic metas- cancer. ASCO Annual Meeting, abstract.
tases that would render them inoperable. Dimitrakopoulou-Strauss, A., Strauss, L.G., and
Rudi, J. 2003. PET-FDG as predictor of therapy
response in patients with colorectal carcinoma.
CONCLUSION Q. J. Nucl. Med. 47:8–13.
Fernandez, F.G., Drebin, J.A., Linehan, D.C.,
Dehdashti, B., Siegel, B.A., and Strasberg,
There is no doubt that FDG-PET has been
S.M. 2004. Five-year survival after resection
an important addition to our staging modal- of hepatic metastases from colorectal cancer in
ities for metastatic colorectal cancer. For patients screened by positron emission tomogra-
selection of patients for liver resection, this phy with F-18 fluorodeoxyglucose (FDG-PET).
test has become indispensable. The most Ann. Surg. 240:438–447.
exciting current area of research includes Findlay, M., Young, H., Cunningham, D., Iveson,
A., Cronin, B., Hickish, T., Pratt, B., Husband,
studies to correlate FDG uptake with tissue
J., Flower, M., and Ott, R. 1996. Noninvasive
markers of prognosis. Ongoing studies of monitoring of tumor metabolism using fluorode-
cost-effectiveness will likely decipher the oxyglucose and positron emission tomography
clinical parameters that should be used for in colorectal cancer liver metastases: correlation
selection of PET scanning in various clinical with tumor response to fluorouracil. J. Clin.
scenarios. Studies of the use of this test Oncol. 14:700–708.
Flamen, P., Hoekstra, O.S., Homans, F., Flamen,
as an early predictor of response to neo-
P., Hoekstra, O.S., Homans, F., Van Cutsem, E.,
adjuvant chemotherapy will likely further Maes, A., Stroobants, S., Peeters, M., Penninckx,
streamline and optimize patient care. F., Filez, L., Bleichrodt, R.P., and Mortelmans, L.
2001. Unexplained rising carcinoembryonic
antigen (CEA) in the postoperative surveillance
REFERENCES
of colorectal cancer: the utility of positron tom-
Akhurst, T., Kates, T.J., Mazumdar, M., Yeing, ography (PET). Eur. J. Cancer 37:862–869.
H., Riedel, E.R., Burt, B.M., Blumgart, L.H., Flanagan, F.L., Dehdashti, F., Ogunbiyi, O.A.,
Jarnagin, W., Larson, S.M., and Fong, Y. 2005. Kodner, I.J., and Siegel, B.A. 1998. Utility of
Recent chemotherapy reduces the sensitivity FDG-PET for investigating unexplained plasma
of [18F]fluorodeoxyglucose positron emission CEA elevation in patients with colorectal cancer.
tomography in the detection of colorectal metas- Ann. Surg. 227:319–323.
tases. J. Clin. Oncol. 23:8713–8716. Fong, Y., Fortner, J., Sun, R.L., Brennan, M.F.,
Bines, S.D., Doolas, L., Jenkins, L., Millikan, K., and Blumgart, L.H. 1999a. Clinical score for
and Roseman, D.L. 1996. Survival after repeat predicting recurrence after hepatic resection for
hepatic resection for recurrent colorectal hepatic metastatic colorectal cancer: analysis of 1001
metastases. Surgery 120:591–596. consecutive cases. Ann. Surg. 230:309–318.
Bozzetti, F., Bignami, R., Montalto, F., Doci, R., Fong, Y., Saldinger, P.F., Akhurst, T., Macaplinac,
and Gennari, L. 1992. Repeated hepatic resec- H., Yeung, H., Finn, R.D., Cohen, A., Kemeny,
60 R. Auer and Y. Fong

N., Blumgart, L.H., and Larson, S.M. 1999b. Oyen, W.J. 2002. Value of positron emission
Utility of 18F-FDG positron emission tom- tomography with [F-18]fluorodeoxyglucose in
ography scanning on selection of patients for patients with colorectal liver metastases: a pro-
resection of hepatic colorectal metastases. Am. spective study. J. Clin. Oncol. 20:388–395.
J. Surg. 178:282–287. Rydzewski, B., Dehdashti, F., Gordon, B.A.,
Guillem, J.G., Moore, H.G., Akhurst, T., Klimstra, Teefey, S.A., Strasberg, S.M., and Siegel, B.A.
D.S., Ruo, L., Mazumdar, M., Minsky, B.D., 2002. Usefulness of intraoperative sonography
Gollub, M.J., Saltz, L., Wong, W.D., and Larson, for revealing hepatic metastases from colorec-
S. 2004. Sequential preoperative fluorodeoxyglu- tal cancer in patients selected for surgery after
cose-positron emission tomography assessment undergoing FDG PET. AJR. Am. J. Roentgenol.
of response to preoperative chemoradiation: a 178:353–358.
means for determining long term outcomes of Schussler-Fiorenza, C.M., Mahvi, D.M.,
rectal cancer. J. Am. Coll. Surg. 199:1–7. Niederhuber, J., Rikkers, L.F., and Weber,
Jaeck, D., Nakano, H., Bachellier, P., Inoue, K., S.M. 2004. Clinical risk score correlates with
Weber, J.C., Oussoultzoglou, E., Wolf, P., and yield of PET scan in patients with colorec-
Chenard-Neu, M.P. 2002. Significance of hepatic tal hepatic metastases. J. Gastrointest. Surg.
pedicle lymph node involvement in patients with 8:150–157.
colorectal liver metastases: a prospective study. Selzner, M., Hany, T.F., Wildbrett, P., McCormack,
Ann Surg. Oncol. 9:430–438. L., Kadry, Z., and Clavien, P.A. 2004. Does the
Jarnagin, W.R., Conlon, J., Bodniewicz, J., novel PET/CT imaging modality impact on the
Dougherty, E., DeMatteo, R.P., Blumgart, L.H., treatment of patients with metastatic colorectal
and Fong, Y. 2001. A clinical scoring system cancer to the liver? Ann. Surg. 204:1027–1034.
predicts the yield of diagnostic laparoscopy Strasberg, S.M., Dehdashti, F., Siegel, B.A.,
in patients with potentially resectable hepatic Drebin, J.A., and Linehan, D. 2001. Survival
colorectal metastases. Cancer 91:1121–1128. of patients evaluated by FDG-PET before
Nakashima, R.A., Paggi, M.G., and Pederson, P.L. hepatic resection for metastatic colorectal car-
1984. Contributions of glycolysis and oxidative cinoma: a prospective database study. Ann.
phosphorylation to adenosine 5′-triphosphate Surg. 233:293–299.
production in AS-30D hepatoma cells. Cancer Taylor, I. 1996. Liver metastases from colorectal
Res. 44:5702–5706. cancer: lessons from past and present clinical
Pauwels, E.K., Sturm, E.J., Bombardieri, E., studies. Br. J. Surg. 83:456–460.
Cleton, F.J., and Stokkel, M.O. 2000. Positron- Truant, S., Huglo, D., Hebbar, M., Ernst, O.,
emission tomography with [18F]fluorodeoxy- Steinling, M., and Pruvot, F.R. 2005. Prospective
glucose. Part I. Biochemical uptake mechanism evaluation of the impact of [18F]fluoro-2-deoxy-
and its implications for clinical studies. J. Can. D-glucose positron emission tomography of
Res. Clin. Oncol. 126:549–559. resectable colorectal liver metastases. Br. J.
Rohren, E.M., Paulson, E.K., Hagge, R., Wong, Surg. 92:362–369.
T.Z., Killius, J., Clavien, P.A., and Nelson, R.C. Wiering, B., Krabbe, P.F., Jager, G.J., Oyen, W.J.,
2002. The role of F-18 FDG positron emission and Ruers, T.J. 2005. The impact of fluor-18-
tomography in preoperative assessment of the deoxyglucose-positron emission tomography in
liver in patients being considered for curative the management of colorectal liver metastases.
resection of hepatic metastases from colorectal Cancer 104:2658–2670.
cancer. Clin. Nucl. Med. 27:550–555. Young, H., Baum, R., Cremerius, U., Herholz,
Rosen, C.B., Nagorney, D.M., Taswell, H.F., K., Hoekstra, O., Lammertsma, A., Pruim, J.,
Helgeson, S.L., Illstrup, D.M., Van Heerden, and Price, P. 1999. Measurement of clinical
J.A., and Adson, M. 1992. Perioperative blood and subclinical tumour response using [18F]-
transfusion and determinants of survival after fluorodeoxyglucose and positron emission
liver resection for metastatic colorectal carci- tomography: review and 1999 ORTC recommen-
noma. Ann. Surg. 216:492–505. dations. European Organization for Research
Ruers, T.J., Langenhoff, B.S., Neeleman, N., Jager, and Treatment of Cancer (EORTC) PET Study
G.J., Strijk, S., Wobbes, T., Corstens, F.H., and Group. Eur. J. Cancer 35:1773–1782.
B. Treatment
6
Ultrasonography During Liver Surgery
Guido Torzilli

INTRODUCTION is mainly achievable because of IOUS


(Torzilli et al., 2005a, 2006). Recently, the
Since the late 1980s, it has been evident demonstration of feasibility and efficacy
that hepatic surgery performed using the of contrast-enhanced ultrasonography per-
conventional approach, carries a very high formed intraoperatively (CE-IOUS) has
risk for patients’ survivals due to the high furthermore stressed the relevance of IOUS
rate of postoperative liver failure (Lin, guidance during liver surgery (Torzilli
1979). In particular, the coexistence of et al., 2004a, b, 2005b).
liver cirrhosis in most cases with hepato-
cellular carcinoma (HCC) has a consider-
able adverse effect on the surgical results Technical Aspects
with 6% postoperative mortality, which is For a proper IOUS, high frequency echo-
not negligible (Liu et al., 2004). For this probes (7.5–10 MHz) are necessary and
reason and for the broadening of ultra- they should have a flat shape to allow their
sound-guided percutaneous therapies, the management in deep and narrowed spaces.
surgical treatment of HCC has loosen its For this purpose, T-shaped probes, inter-
role of first choice treatment. Furthermore, digital probes, and micro-convex probes
the good results with interstitial therapies are available. Main factors for probe
in the treatment of HCC has induced the selection are its volume, its stability and
extension of their use also for those patients the wideness of ultrasonographic scan-
with colorectal cancer (CRC) liver metas- ning window: the best probe should be
tases (Solbiati et al., 2001). However, the small, thin in width and short in transverse
imaging techniques have been introduced length, stable and with a wide ultrasono-
also as aids for surgeons in performing liver graphic scanning window. In this sense
resection. In fact, at the end of the 1970s, the micro-convex probe represents the
intraoperative ultrasonography (IOUS) in best compromise among all these require-
hepatic surgery in the case of liver cirrhosis ments. Indeed, the T-shaped probe is more
started (Makuuchi et al., 1980, 1998). Now, stable but has a lower ratio between lateral
liver resections can be carried out with no length and ultrasonographic scanning win-
mortality even if cirrhosis is associated dow than the micro-convex probe.
(Torzilli et al., 1999a), combining the For CE-IOUS we initially used a convex
needs for oncological radicality and 3–6 MHz frequency and 1.88–3.76 MHz
liver parenchyma sparing, and this goal harmonic frequency transducer was

63
64 G. Torzilli

adopted. In all patients 4.8 ml sulphur- detection of new lesions in around 30%
hexafluoride microbubbles (SonoVue®, of cases (Kokudo et al., 1996) (Figure
Bracco Imaging, Italy) was injected intra- 6.1). Conversely, in the case of CRC liver
venously through a peripheral vein by metastases 10–40% of cases do not have
the anaesthesiologist. Intraoperative high- palpable fore sites (Machi et al., 1991)
frequency probes have limited value for (Figure 6.2). As a consequence of this
CE-IOUS although they keep the advan- IOUS exploration of the liver has a great
tages in terms of volume, and stability; impact on the surgical strategy: Kane et al.
indeed, the effects of contrast enhancement (1994) experienced 51% of changed pro-
are less evident using these probes. More
recently, a micro-convex prototype with
the same frequency pattern of the per-
cutaneous probe used for CEIOUS was
released, and its adequacy during surgery
is under evaluation.
A background of perfect knowledge of
the liver anatomy surgically and ultra-
sonographically is requested to proceed.
For surgical anatomy, the Couinaud seg-
ments are considered here (Couinaud,
1957). After entering the abdominal cav-
ity, liver mobilisation dividing the round
and falciform ligaments, and division of
eventual adhesions to free the antero-
superior and inferior surfaces of the liver
are the steps which should precede the Figure 6.1. Millimetric hypoechogenic nodule
liver exploration with IOUS. Pulling the (arrow)
round ligament, the liver surface is widely
exposed and following the portal branches
and the hepatic veins, the liver can be
entirely studied.
The use of IOUS in liver resections can
be schematically divided into two prin-
cipal phases: the liver exploration for the
staging of the disease and planning of
the surgical strategy, and the guidance
of the surgical maneuvers.

Liver Exploration
The hard and irregular surface of cirrhotic
liver makes the detection of small nodules Figure 6.2. Millimetric hyperechogenic nodule
by palpation difficult; IOUS allows the (arrow)
6. Ultrasonography During Liver Surgery 65

cedures by IOUS in their series. However, even biopsy does not seem to be adequate.
the impact of IOUS on the operative deci- The only nodule which can be easily dif-
sion making, when compared with those ferentiated intraoperatively from a HCC
of preoperative imaging techniques, has or a liver metastases is the small hemangi-
decreased to 4–7% (Jarnagin et al., 2001; oma, which is often discovered primarily
Cerwenka et al., 2003; Sahani et al., 2004). at IOUS: it has a typical ultrasonographic
However, the problem of the impact of pattern and, moreover, when compressed
IOUS on the operative decision making changes its size and appearance. Further
depends on two main factors: the surgical improvement in differential diagnosis of
policy of each specific team, and the type liver nodules with IOUS may be expected
of tumor. Indeed, the low rates shown in by the introduction and diffusion of the
the latest reports (Jarnagin et al., 2001; intraoperative use of the last generation
Cerwenka et al., 2003; Sahani et al., contrast agents.
2004) are also partially motivated by the
surgeon’s surgical policy: in fact, since a Contrast Enhanced Intraoperative
considerable number of patients under- Ultrasonography
went major hepatectomies, new nodules More recently the introduction of CE-IOUS
detected by IOUS in the same hemiliver has set the rate of modified operative deci-
would not have modified the surgical strat- sion-making in 30–40% of cases (Torzilli
egy. In our experience major hepatectomies et al., 2004a, b, 2005b).
are carried out in the minority of patients Tumor vascularity as a criterion for dif-
(Torzilli et al., 2005a, 2006) because of the ferentiating the regenerative or dysplastic
extensive use of the IOUS-guidance for nodules from the HCC correlate well with
achieving parenchymal sparing resections, the histological evidence of a progressive
so that detection of new nodules is more increase in unpaired arteries from dys-
suitable for changing the surgical strategy. plastic to neoplastic nodules in a cirrhotic
Conversely, depending on the type of the liver (Roncalli et al., 1999). Certainly, the
tumor, liver exploration is focused mainly pattern of vascular enhancement is not suf-
on the detection of new lesions as in the ficient for differentiating malignant from
case of CRC liver metastases rather than non malignant nodules in a cirrhotic liver
both on the detection and on the differen- with 100% specificity. However, CE-US
tiation, as in the case of HCC. Indeed, for provides differential diagnosis of FLL
this last tumor, which generally develops with a 95% specificity (Quaia et al.,
in the cirrhotic liver the risk nowadays is 2004); of course, it must be considered
to overestimate the tumor stage with IOUS. that this last rate was referred to another
In fact, in the case of patients with HCC type of lesion in comparison to the target
in a cirrhotic liver, except for those nod- of the present study. Indeed, the intraop-
ules with mosaic ultrasonographic pattern erative exploration takes profit from the
which are malignant in 84% of cases, only higher resolution of the ultrasonography in
24–30% of hypoechoic nodules (Figure direct contact with the liver. Therefore, the
6.1), and 0–18% of those hyperechoic are need of differentiating nodules detected at
neoplasm (Kokudo et al., 1996; Takigawa IOUS is mostly focused on lesions smaller
et al., 2001). To overcome this problem than 1 cm: for these nodules, the vascularity
66 G. Torzilli

as criterion for differential diagnosis is These last were not removed, and
less specific. However, some improve- their benignity was confirmed after at
ments compared with conventional IOUS least 6 months of follow-up if there
could be expected. In our preliminary was no tumor growth or they were
experience, CE-IOUS provided remark- still not detectable at spiral CT.
able findings, either by adding information (C) If a new nodule showed a mosaic pat-
on nodular vascularity in patients with tern at IOUS, it was considered malig-
HCC, or by detecting nodules that were nant (Takigawa et al., 2001), and then
not visible at IOUS, in patients with CRC removed, independently from the pat-
liver metastases (Torzilli et al., 2004b, tern of enhancement at CE-IOUS.
2005b). Focusing the attention on patients
Specificity of this classification criteria
operated for HCC our preliminary experi-
is in our experience 69% (Torzilli et al.,
ence showed encouraging results (Torzilli
2007) which is certainly closer to the
et al., 2004a): however, they were related
true performance of CEIOUS, rather than
to a very limited number of patients, and
the results obtained at the initial experi-
data did not meet the expectancies shown
ence (Torzilli et al., 2004a). This value is
by optimistic findings with 100% specifi-
probably not that high, especially when
city. Therefore, we classified the CEIOUS
compared with that reported for CE-US
pattern based on our preliminary experi-
(Quaia et al., 2004). However, as we men-
ence and on what was reported in literature
tioned earlier the small size of the lesions
about the CE-US (Quaia et al., 2004).
targeted for CEIOUS study could explain
Classification was as follows:
this discrepancy: for these tiny nodules
(A) If an hypoechoic or hyperechoic nod- the neovascularity as criterion for differ-
ule showed at CEIOUS a full enhance- entiation between malignant and benign
ment (hyperechoic) in the arterial lesions has limits which are independ-
phase and became hypoechoic in the ent from the method we use for studying
delayed portal and late phases (A1) them. Therefore, CEIOUS can be helpful
or it remained hypoechoic with thin in a certain percentage of nodules but not
vessels detectable into the nodule in the in all. In this perspective the rate of 69%
arterial phase and delayed phase (A2) of specificity is encouraging as it means
or did not show any early enhance- that we can provide proper information
ment but remained hypoechoic in the with this new technique in seven out of ten
delayed phases with peripheral and/or lesions we detect at the time of laparotomy.
intralesional neovascularization (A3), Further improvements in this sense could
it was considered malignant and then come from the introduction of hepto-
removed. specific contrast spents for CE-IOUS.
(B) If an hypoechoic or hyperechoic
Colorectal Cancer Liver Metastases
nodule had no early enhancement at
CEIOUS and showed a hysoechoic Intraoperative ultrasonography is still
or slightly hypoechoic pattern in all the most accurate diagnostic technique
the phases in comparison with the for detecting focal liver lesions (FLLs)
surrounding liver parenchyma, it was (Sahani et al., 2004). However, during
considered a non neoplastic lesion. surgery for CRC liver metastases, IOUS
6. Ultrasonography During Liver Surgery 67

has a sensitivity of just 82% (Machi et al., out the risk of tumor exposure, and thus
1991), and as a consequence may miss enhancing the treatment radicality.
nodules less than 1 cm in diameter: this is
particularly evident in those patients who Tumor Location
undergo surgery after chemotherapy. In IOUS allows an accurate three-dimen-
these patients, liver fore sites have similar sional reconstruction of the relation-
echo-pattern with the surrounding liver ship between the tumor and the portal
parenchyma. branches, and hepatic veins: this is a fun-
In our experience, CE-IOUS confirmed damental step in the definition of the
its improvement of IOUS sensitivity in proper surgical strategy. Indeed, surgical
detecting new small fore sites reducing decision making should be obtained hav-
the risk of down-staging the disease and ing portal branches and hepatic veins as
enhancing the rate of treatment with cura- landmarks to reduce the risk of major
tive intent: indeed 9% of patients had morbidity and mortality.
new lesions discovered only by CEIOUS The definition of tumor-vessels relation-
(Figure 6.3a, b) (Torzilli et al., 2008a). ship is relevant for planning the type of
The association of IOUS and CEIOUS resection and for this purpose we classi-
increased the sensitivity from 88 to 93%. fied its appearance at IOUS. Based on this
Additionally, the better visualization of classification each category corresponds to
the margins of the main lesion in 19% of a specific operation.
patients with CRC liver metastases helps Based on IOUS findings, relationship with
the surgeon to better define the resection intrahepatic vascular structures were classi-
area and the proper dissection plane ruling fied as follows (Torzilli et al., 2005a):

a b

Figure 6.3. (a) The liver shows a inhomogeneous pattern at IOUS; (b) at CEIOUS is well evident black-
hole (arrow) which corresponds to a metastatic nodule
68 G. Torzilli

1. Portal branch or hepatic vein in contact with vessel wall discontinuation at


with a capsulated HCC without vessel IOUS: vascular resection is associated
wall discontinuation at IOUS: vascular (Figure 6.4b).
resection is not associated, and just 6. Portal branch in contact with a HCC or
enucleation at the level of the vascular CRC liver metastases with or without
contact is performed (Figure 6.4a). vessel wall discontinuation but with
2. Portal branch or hepatic vein separated proximal bile duct dilation at IOUS:
by a thin layer of liver parenchyma (at vascular resection is associated.
least 1 mm) from CRC liver metastases:
Extension of the hepatectomy is always
vascular resection is not associated.
considered for the parenchyma fed by an
3. Portal branch or hepatic vein in contact
infiltrated portal branch (categories 3–6).
with a HCC with undefined margins
Inversely, in case of infiltration of a hepatic
without vessel wall discontinuation
vein, an extension of the resection to the
at IOUS: vascular resection is associ-
whole liver parenchyma theoretically
ated.
drained by this vein is considered only
4. Portal branch or hepatic vein in contact
if there are no accessory hepatic veins
with a CRC liver metastases without
at IOUS, and color-Doppler IOUS shows
vessel wall discontinuation at IOUS:
hepatofugal blood flow in the feeding portal
vascular resection is associated.
branch once the hepatic vein is clamped.
5. Portal branch or hepatic vein in contact
IOUS tumor-vessel classification and
with HCC or CRC liver metastases
the related surgical policy has proven that

a b

HCC
RHV

HCC
RHV
IVC

Figure 6.4. (a) This patient was a carrier of HCC in contact without sign of infiltration with the right
hepatic vein (RHV) indicated by the arrows (pattern 1): operation consisted of a limited resection without
resection of the RHV; (b) this patient was a carrier of HCC in contact with sign of infiltration with the
right hepatic vein (RHV) indicated by the arrows (pattern 5): operation consisted of an extended Segment
7 resection including the RHV. IVC=Inferior Vena Cava
6. Ultrasonography During Liver Surgery 69

in selected patients it is possible to get (Makuuchi et al., 1991). As a consequence


close to the tumor burden without increas- of that, some authors consider that the
ing the risk of incomplete removal, and as resected specimen should comprehend at
a consequence of local recurrence (Torzilli least the portal area, which includes the
et al., 2005a, 2006). In practice, this evi- lesion (Makuuchi et al., 1991). This is
dence means that with IOUS guidance it is impossible to correctly identified without
possible to perform conservative but radi- the aid of IOUS, especially in a cirrhotic
cal hepatectomies also in complex pres- liver where there are generally wide varia-
entations, and then to enlarge the surgical tions and abnormalities in the distributions
indications. With this approach we have of the portal branches (Makuuchi et al.,
limited the rate of major hepatectomies to 1991). For this purpose the systematic seg-
8% in patients with tumors involving one mentectomy was devised in the early 1980s
or more hepatic veins close to their caval (Makuuchi et al., 1980), and its peculiar-
confluence, without performing any vas- ity consists originally in the dye injection
cular reconstruction (Torzilli et al., 2006). into the feeding portal branch. We have
These results not only underline how the recently established alternatives to this
IOUS guidance allows operation other- approach for tumors located in the left
wise not feasible, but also because this hemiliver which will be also described.
approach reduces the rate of major hepa-
tectomies, and in particular of right hemi- Puncture of the Portal Branch
hepatectomy (Torzilli et al., 2008b). The After the accomplishment of liver mobili-
real need for interventions such as preop- sation and exploration with IOUS, segmen-
erative portal vein embolization which are tal staining is carried out by puncturing
adopted to secure from liver failure after the portal branch near the target point to
major removal of liver parenchyma can be be divided during dissection (Makuuchi
discussed. et al., 1991). The portal branch is punctured
under echo-guidance, through a free-hand
technique or with a proper device, and
RESECTION GUIDANCE then 3–5 ml of indigo-carmine dye are
injected into the vessel. The stained area
Hepatocellular Carcinoma becomes evident on the liver surface and
it is marked with the electrocautery. If the
Systematic Segmentectomy
nodule is located between two adjacent
In a cirrhotic patient, the liver volume to segments, two portal branches afferent to
be resected must be determined with par- the area must be punctured and injected.
ticular care with the purpose to associate In this case, the deepest and most dorsal
surgical radicality and non-cancerous liver vessel must be punctured at first to prevent
parenchyma sparing. Liver function tests air bubbles contained in the dye from dis-
and liver volumetry on CT scans help in turbing the ultrasonographic detection and
this decision. Tumor dissemination from puncture of the other branch. The portal
the main lesion through the portal branches branch is punctured 1–2 cm distally from
cannot be detected with certainty by the its origin to prevent the dye from reflexing
pre- and intraoperative imaging modalities and the direction and velocity of the infusion
70 G. Torzilli

are controlled at IOUS. To prolong the Compression of the Portal Branch


staining, the hepatic artery at the hilum is In addition to the hooking technique we
clamped before the portal branch puncture. have proposed a new technique to perform
When there are numerous and thin vessels subsegmental resections, initially just for
which should be punctured or in the case segments 2 and 3, without clamping the
of tumor thrombus in the segmental portal hepatic artery and puncturing the feeding
branch of the segment that should be portal branches (Torzilli and Makuuchi,
removed, the dye is injected in the portal 2004). Indeed, once the feeding portal
branches afferent to the adjacent segment: branch has been identified at IOUS, it is
this is the so-called counterstaining tech- compressed using the IOUS probe by one
nique (Takayama et al., 1991). side of the liver and the finger by the oppo-
site side: in this way it is possible to induce
Hooking of the Portal Branch
a transient ischemia of the subsegmental
There are four segmental portal branches portion of the liver distally to the com-
to segment which are generally divided pression site. This portion can be marked
in two groups: those for the superior and with the electrocautery, the compression
those for the inferior portion but the most is released, and the subsegmentectomy is
common branching pattern can be rec- carried out. This technique is simple, fast,
ognized in just half of the cases. These not invasive and reversible. It is possible
branches rather than punctured under to modify the site of compression and the
IOUS guidance, can be approached by resection volume allows the sizing of the
dissecting the umbilical portion: once resection in function of the tumor features
exposed, the vessel can be encircled with and the status of the background liver.
a suture and pulled under IOUS control to Together with the S4 subsegmentectomies,
verify if it is the branch to S4 inferior or this operation represents an alternative to
not; then the proper portalbranch can be the puncture technique which is not as
ligated and divided and the discoloured reproducible as these two last methods
area which will appear on the liver sur- (Torzilli and Makuuchi, 2001, 2004).
face should correspond to the S4 inferior
Limited Resection
which can be marked with the electro-
cautery to proceed with the liver dissec- After the identification of the tumor, the
tion: this is a peculiar application of the surgeon under IOUS control can mark
so-called hooking technique (Torzilli and with the electrocautery the border of the
Makuuchi, 2001). Furthermore, the sub- lesion on the surface of the liver just above
segment 4 superior could be resected by the nodule. To carry out this maneuver the
clamping the portal branch to subseg- flat and thin tip of the electrocautery is
ment 4 inferior, as it is identified with the positioned between the probe and the liver
earlier described hooking technique; the surface: this maneuver results in a shadow
discoloured subsegment 4 inferior cau- at the IOUS image which runs deeply just
dally, the plan at IOUS which includes below the electrocautery. In this way it is
the middle hepatic vein laterally, and that possible to define the position of the elec-
marked by the falciform ligament medi- trocautery with the tumor edge and con-
ally delimitates the area to be resected. sequently to mark with the electrocautery
6. Ultrasonography During Liver Surgery 71

itself the nodule profile on the liver surface


and to select the safer edge for the inci-
sion. The adequacy of the marked edge HCC
can be furthermore checked with IOUS as
the air trapped between the probe and the
irregular surface of the demarcation line
drawn with the electrocautery on the liver L
surface can be visualized at IOUS.
Another way to draw precisely on the
liver surface with the aid of IOUS involves
the tumor edge carrying out using the finger
tips. With the probe positioned on the liver
surface the surgeon’s finger-tip pushes on
the opposite side and its profile is visual- Figure 6.5. At IOUS the dissection line can be
well visualized (arrows), and the planned direction
ized at IOUS: as a consequence relations
(transparent arrows) allow radical removel of the
between the finger-tip and the tumor edge HCC nodule
can be precisely estimated and the resection
area can be marked on the liver surface.
Parenchymal Dissection
L
The main advantage provided by the resec- MTX
tion guidance accomplished with the aid of
IOUS is the modification of the traditional
way to dissect the liver tissue which was
done on vertical planes to avoid the tumor
exposure on the cut surface. IOUS allows
to follow in real-time the dissection plane,
to put it constantly in relation with the
tumor edge, and to modify its direction
when needed: this is because it is possible
to visualize on the IOUS image the dis-
section plane which appears as an echoic
line due to the entrapment of air bubbles
and clots between the faced cut surfaces Figure 6.6. At IOUS the dissection plane (arrows)
(Figure 6.5). If the dissection plane is not has been modified, because its original direction
(transparent arrow) would have led to enter into the
clearly visible, it can be better visualized by
to metastatic nodule (MTX)
inserting a gauze or a specifically devised
silicon gauze between the faced cut sur-
faces. These techniques allow the surgeon (Figure 6.6). In this way it is possible to
to keep the proper dissection plane: an early carry out a rounded trajectory of the dis-
recognition of a wrong dissection plane section plane around the tumor avoiding its
permits the surgeon to modify it properly exposure, and allowing the surgeon to spare
and to avoid a possible tumor exposure important vascular structures: this results
72 G. Torzilli

in more conservative but radical treatments portal branch of segment 5 instead of the
and in a lower rate of major hepatectomies. planned subsegmental branch of segment
Furthermore, in those patients in which 8 and then, necrosis of the segment 5 may
major resections are needed, IOUS allows occur. The hooking technique under IOUS
better achievement of the proper dissection control enables the identification of the
plane, which should run along the hepatic branch, which was encircled, and then the
vein to be fully anatomic. surgeon can decide with certainty whether
The artifacts which allow the dissec- to ligate it. This is useful also in the case of
tion plan at IOUS can sometimes mask tumor thrombus in major portal branches.
structures such as portal branches which In this situation, once the portal branch is
should be ligated or conversely respected. skeletonized, it is encircled with a stitch
For this reason, to better visualise the and, under IOUS control, the stitch, is
targeted point where the portal branch gently pulled up: this traction stretches
should be divided, the so-called hooking the portal branch slightly and the traction
technique has been devised (Torzilli et al., point is demonstrated clearly by IOUS. If
1999b). When the Glissonian sheath is the traction point is not at the level of the
exposed and skeletonized, it is encircled tumor thrombus it is possible to ligate the
with a stitch, which is visualized by IOUS portal branch and proceed with the liver
as an echogenic spot with a posterior resection being sure that thrombus will not
shadow. Then under sonographic control, migrate because of surgical manipulation
the stitch, hooking the exposed vessel, is (Torzilli et al., 2005c).
gently pulled up, which stretches the portal During liver dissection the back-flow
branch slightly and the traction point is bleeding from the hepatic veins is an
demonstrated clearly by IOUS. If the important source of blood loss, and it
exposed portal branch is not clearly visible is one of the most important factors in
because it has collapsed, the portal triad is determining the short-term and long-term
unclamped to enable it to fill with blood patient’s outcome. Therefore, limiting
and then it is visualised better by IOUS. If the backflow bleeding from the hepatic
the target site is correct, the portal branch veins is a priority in liver resections. We
is ligated and divided and segmentec- described a ultrasound-guided technique
tomy is completed under IOUS guidance; for backflow bleeding control from the
conversely, if the exposed vessel was not RHV during right-sided liver resection.
the targeted one, it is spared and useless The technique is very simple: once the
sacrifice of further liver parenchyma is right surface of the extrahepatic RHV is
avoided. exposed to allow its compression by the
A practical example in which the hook- surgeon’s finger-tips, the effectiveness of
ing technique is used is during ventral or finger compression is checked by IOUS,
dorsal subsegmentectomy of segment 8. and color-Doppler.
The portal trunk to this segment may show
bifurcation in its dorsal branch and ventral Post-resectional Control
trunk just close to the origin of the portal There are possibilities given by IOUS after
vessel to segment 5. In this situation, there nodule removal: one is the “water bath”
is the risk of ligating and dividing the technique which consists of the real-time
6. Ultrasonography During Liver Surgery 73

control of the proper resection of the tar- resections, but it is mandatory to obtain the
geted nodule verifying its complete inclu- complete tumor clearance: for this purpose
sion in the specimen once removed from IOUS is essential.
the liver (Makuuchi, 1987); the second is
done by checking the cut surface refilled Definition of the Resection Area
with saline to avoid the artifacts generated Definition of the resection area once a non-
by the residual air bubbles and clots. anatomical resection has been chosen is
The aforementioned methods guarantee, similar for both resection for HCC and CRC
whenever possible, anatomical and limited liver metastases. In both these conditions,
resection: this has consequences either for the surgeon’s target should be to determine
the effectiveness of the surgical treatment a resection volume which could result in a
or for its safety. Patients with HCCs who flat and regular cut surface to minimize the
underwent IOUS-guided subsegmentec- risk of postoperative collections.
tomies had better survivals (Makuuchi
et al., 1991; Hasegawa et al., 2005), and Liver Dissection and Post-resectional Control
those with HCCs in contact with intrahe- The same concepts discussed for HCC
patic vessels, once removed under IOUS- regarding IOUS-guided dissection plane
guidance sparing the involved veins did maintenance and IOUS post-resectional
not show local recurrences (Torzilli et al., control are applied in the case of resection
2005a, 2006). Additionally, this kind of for liver metastases.
surgery allows radical surgical treatment The complete surgical clearance of the
of HCC without mortality (Torzilli et al., tumor tissue, even for multiple liver metas-
1999a). Procedures, which are not IOUS- tases, could be justified assuming that
guided, led to dangerous and useless major every single nodule is secondary to the
resection or incomplete operations. primary tumor rather than an expression
of the intrahepatic diffusion of the first
metastatic lesion. For this purpose several
Liver Metastases
authors showed that the occurrence of
At present, tumor clearance can be achieved satellite nodules around the main meta-
in most cases of patients undergoing liver static lesion are rare and wedge resection
resection for metastatic disease with sur- even with tumor-free margin less than
vival benefits even in the presence of 0.5–1 cm in thickness but without expo-
multiple fore sites or vascular infiltration sure of the tumor on the cut surface is jus-
(Minagawa et al., 2000). To achieve this tified (Kokudo et al., 2002; Pawlik et al.,
standard, most of the merits of conserva- 2005). Therefore, the goal to be achieved
tive and radical resections should prob- is the margin clearance: in this sense IOUS
ably be addressed to the extensive use of is fundamental to guarantee the complete
IOUS. In fact, similar to the case of HCC, removal of the lesions. The other goal
IOUS is not only a simple, accurate diag- is the conservativeness of the treatment
nostic modality, but it is a real surgical which should avoid useless parenchyma
device which allows a proper hepatic sacrifice and then safer procedures. Also
resection. In the case of CRC liver in this sense, IOUS with the techniques
metastases there is no need for anatomical described above allows the accomplishment
74 G. Torzilli

of radical but limited resection introduc- The American College of Surgeons has
ing the aforementioned concept of the recently recognized the need for surgeons
safe rounded dissection plane. With this of a specific training in US, meanwhile
approach, it has been possible to carry dedicated monographs have been pub-
out resections without mortality, with no lished almost simultaneously in America
residual gross tumor on the surgical stump and in Europe (Machi and Staren, 2004;
and with a relatively low rate of major Torzilli et al., 1997). The way for a wider
hepatectomies (21.3%) (Minagawa et al., diffusion of ultrasound in surgeons’ prac-
2000). These results were obtained in tice has definitely been opened.
spite of the high mean number of nodules
resected per patient (3.2) and its relatively
large mean size (4.5 cm). REFERENCES
This is a further confirmation that liver Cerwenka, H., Raith, J., Bacher, H., Werkgartner,
resection is an imaging-guided procedure G., el-Shabrawi, A., Kornprat, P., and Mischinger,
and as every interventional imaging-guided H.J. 2003. Is intraoperative ultrasonography
during partial hepatectomy still necessary
procedure, its features are the highest ther- in the age of magnetic resonance imaging?
apeutic efficacy combined with the mini- Hepatogastroenterology 50: 1539–1541.
mal invasiveness. With IOUS aid, it is now Couinaud, C. 1957. Le foie, etude anatomiques et
possible to carry out surgical procedures chirurgicales. Paris, Masson.
which are safe, oncologically radical and Hasegawa, K., Kokudo, N., Imamura, H.,
conservative for the liver function. Mostly Matsuyama, Y., Aoki, T., Minagawa, M., Sano,
K., Sugawara, Y., Takayama, T., and Makuuchi,
because of these reasons, surgery can be M. 2005. Prognostic impact of anatomic resec-
still considered the treatment of choice for tion for hepatocellular carcinoma. Ann. Surg.
most liver metastases, even in the case of 242: 252–259.
the diffusion of other treatments such as Jarnagin, W.R., Bach, A.M., Winston, C.B., Hann,
the thermal ablation which is impaired by L.E., Heffernan, N., Loumeau, T., DeMatteo,
a 39% tumor relapse rate in the first year of R.P., Fong, Y., and Blumgart, L.H. 2001. What is
the yield of intraoperative ultrasonography dur-
follow-up (Solbiati et al., 2001). ing partial hepatectomy for malignant disease? J.
In conclusion, IOUS has acquired pro- Am. Coll. Surg. 192: 577–583.
gressive importance in the field of sur- Kane, R.A., Hughes, L.A., Cua, E.J., Steele,
gical resection in cirrhotic and normal G.D., Jenkins, R.L., and Cady, B. 1994. The
liver, either in the diagnosis and staging impact of intraoperative ultrasonography on
or strictly in the surgical technique, and surgery for liver neoplasms. J. Ultrasound
Med. 13: 1–6.
now it can be considered a fundamental Kojiro, M. 2004. Focus on dysplastic nodules and
tool for hepatobiliary and other surgi- early hepatocellular carcinoma: an Eastern point
cal procedures (Makuuchi et al., 1998). of view. Liver Transpl. 10 (2 Suppl 1): 3–8.
Presently, we can affirm that liver resec- Kokudo, N., Bandai, Y., Imanishi, H., Minagawa,
tion is an imaging-guided procedure and M., Uedera, Y., Harihara, Y., and Makuuchi,
as every interventional imaging-guided M. 1996. Management of new hepatic nodules
detected by intraoperative ultrasonography during
procedure, its features include the highest hepatic resection for hepatocellular carcinoma.
therapeutic efficacy combined with mini- Surgery 119: 634–640.
mal invasiveness. IOUS should be a famil- Kokudo, N., Miki, Y., Sugai, S., Yanagisawa, A., Kato,
iar instrument for the hepatic surgeon. Y., Sakamoto, Y., Yamamoto, J., Yamaguchi, T.,
6. Ultrasonography During Liver Surgery 75

Muto, T., and Makuuchi, M. 2002. Genetic and Quaia, E., Calliada, F., Bertolotto, M., Rossi, S.,
histological assessment of surgical margins in Garioni, L., Rosa, L., and Pozzi-Mucelli, R.
resected liver metastases from colorectal carci- 2004. Characterization of focal liver lesions with
noma: minimum surgical margins for successful contrast-specific US modes and a sulfur hexaflu-
resection. Arch. Surg. 137: 833–840. oride-filled microbubble contrast agent: diag-
Lin, T.Y. 1979. Resectional therapy for primary nostic performance and confidence. Radiology
malignant hepatic tumors. In: International 232: 420–430.
Advances in Surgical Oncology. Murphy, GP. Roncalli, M., Roz, E., Coggi, G., Di Rocco, M.G.,
ed., New York, Alan R Liss, pp. 25–54. Bossi, P., Minola, E., Gambacorta, M., and
Liu, C.L., Fan, S.T., Lo, C.M., Wong, Y., Ng, I.O., Borio, M. 1999. The vascular profile of regen-
Lam, C.M., Poon, R.T., and Wong, J. 2004. erative and dysplastic nodules of the cirrhotic
Abdominal drainage after hepatic resection is liver: implications for diagnosis and classifica-
contraindicated in patients with chronic liver tion. Hepatology 30: 1174–1178.
diseases. Ann. Surg. 239: 194–201. Sahani, D.V., Kalva, S.P., Tanabe, K.K., Hayat,
Machi, J., and Staren, E.D. 2004. Ultrasound for S.M., O’Neill, M.J., Halpern, E.F., Saini, S.,
Surgeons. New York, Lippincott-Williams & and Mueller, P.R. 2004. Intraoperative US in
Wilkins, 2nd Edition. patients undergoing surgery for liver neoplasms:
Machi, J., Isomoto, H., Kurohiji, T., Yamashita, Y., comparison with MR imaging. Radiology 232:
Shirouzu, K., Kakegawa, T., Sigel, B., Zaren, 810–814.
H.A., and Sariego, J. 1991. Accuracy of intra- Solbiati, L., Livraghi, T., Goldberg, S.N., Ierace,
operative ultrasonography in diagnosing liver T., Meloni, F., Dellanoce, M., Cova, L., Halpern,
metastasis from colorectal cancer: evaluation E.F., and Gazelle, G.S. 2001. Percutaneous
with postoperative follow-up results. World J. radiofrequency ablation of hepatic metastases
Surg. 15: 551–556. from colorectal cancer in 117 patients. Radiology
Makuuchi, M. 1987. Abdominal Intraoperative 221: 159–166.
Utrasonography. Tokyo/New York, lgaku-Shoin. Takayama, T., Makuuchi, M., Watanabe, K.,
Makuuchi, M., Yamazaki, S., and Hasegawa, H. Kosuge, T., Takayasu, K., Yamazaki, S., and
1980. Ultrasonically guided liver surgery. Jpn. J. Hasegawa, H. 1991. A new method for mapping
Ultrasonics Med. 7: 45–49. hepatic subsegment: counterstaining identifica-
Makuuchi, M., Takayama, T., Kosuge, T., Yamazaki, tion technique. Surgery 109: 226–229.
S., Yamamoto, J., Hasegawa, H., and Takayasu, Takigawa, Y., Sugawara, Y., Yamamoto, J., Shimada,
K. 1991. The value of ultrasonography for hepatic K., Yamasaki, S., Kosuge, T., and Makuuchi,
surgery. Hepatogastroenterology 38: 64–70. M. 2001. New lesions detected by intraoperative
Makuuchi, M., Torzilli, G., and Machi, J. 1998. ultrasound during liver resection for hepatocellular
History of intraoperative ultrasonography. carcinoma. Ultrasound Med. Biol. 27: 151–156.
Ultrasound Med. Biol. 24: 1229–1242. Torzilli, G., and Makuuchi, M. 2001. Ultrasound-
Minagawa, M., Makuuchi, M., Torzilli, G., guided liver subsegmentectomy: the peculiarity
Takayama, T., Kawasaki, S., Kosuge, T., of segment 4. J. Am. Coll. Surg. 193: 706–708.
Yamamoto, J., and Imamura, H. 2000. Extension Torzilli, G., and Makuuchi, M. 2004. Ultrasound-
of the frontiers of surgical indications in the guided finger compression in liver subsegmen-
treatment of liver metastases from colorectal tectomy for hepatocellular carcinoma. Surg.
cancer: long-term results of our experience. Ann. Endosc. 18: 136–139.
Surg. 231: 487–499. Torzilli, G., Olivari, N., Livraghi, T., and Di Candio,
Pawlik, T.M., Scoggins, C.R., Zorzi, D., Abdalla, G. 1997. Ecografia in chirurgia: modalità diag-
E.K., Andres, A., Eng, C., Curley, S.A., Loyer, nostiche e terapeutiche. Milano, Poletto Editore.
E.M., Muratore, A., Mentha, G., Capussotti, L., Torzilli, G., Makuuchi, M., Inoue, K., Takayama,
and Vauthey, J.N. 2005. Effect of surgical mar- T., Sakamoto, Y., Sugawara, Y., Kubota, K.,
gin status on survival and site of recurrence after and Zucchi, A. 1999a. No-mortality liver resec-
hepatic resection for colorectal metastases. Ann. tion for hepatocellular carcinoma in cirrhotic
Surg. 241: 715–722. and non-cirrhotic patients: is there a way?
76 G. Torzilli

A prospective analysis of our approach. Arch. N., and Makuuchi, M. 2005c. Utility of the
Surg. 134: 984–992. hooking technique for cases of major hepatec-
Torzilli, G., Takayama, T., Hui, A.M., Kubota, K., tomy. Surg. Endosc. 19: 1156–1157.
Harihara, Y., and Makuuchi, M. 1999b. A new Torzilli, G., Montorsi, M., Del Fabbro, D.,
technical aspect of ultrasound-guided liver sur- Palmisano, A., Donadon, M., and Makuuchi
gery. Am. J. Surg. 178: 341–343. M. 2006. Ultrasonographically guided surgical
Torzilli, G., Olivari, N., Moroni, E., Del Fabbro, approach to liver tumours involving the hepatic
D., Gambetti, A., Leoni, P., Montorsi, M., and veins close to the caval confluence. Br. J. Surg.
Makuuchi, M. 2004a. Contrast-enhanced intra- 93: (10)1238–1246.
operative ultrasonography in surgery for hepato- Torzilli, G., Palmisano, A., Del Fabbro, D.,
cellular carcinoma in cirrhosis. Liver Transpl. 10 Marconi, M., Donadon, M., Spinelli, A., Bianchi,
(2 Suppl 1): 34–38. PP., Montorsi, M., 2007. Contrast-enhanced
Torzilli, G., Del Fabbro, D., Olivari, N., Calliada, intraoperative ultrasonograpy during surgery for
F., Montorsi, M., and Makuuchi, M. 2004b. hepatocellular carcinoma in liver cirrhosis: is it
Contrast-enhanced ultrasonography during liver useful or useless? A prospective cohort study
surgery. Br. J. Surg. 91: 1165–1167. of our experience. Ann. Surg. Oncol. 14(4):
Torzilli, G., Montorsi, M., Donadon, M., Palmisano, 1347–1355.
A., Del Fabbro, D., Gambetti, A., Olivari, N., Torzilli, G., Botes, F., Procopio, F., Donadon, M.,
and Makuuchi, M. 2005a. “Radical but con- Balzarini, L., Lutman, F., Calliada, F., Montorsi,
servative” is the main goal for ultrasonography- M. 2008a. Use of contrast-enhanced intraop-
guided liver resection: prospective validation of erative ultrasonograpy during liver surgery for
this approach. J. Am. Coll. Surg. 201: 517–528. colorectal cancer liver metastases. Its impact
Torzilli, G., Del Fabbro, D., Palmisano, A., on operative outcome. Analysis of prospective
Donadon, M., Bianchi, P., Roncalli, M., Balzarini, Cohort study. Eu. J. Cancer 6: 16–23.
L., and Montorsi, M. 2005b. Contrast-enhanced Torzilli, G., Donadon, M., Marconi, M., Botes,
intraoperative ultrasonography during hepatec- F., Palmisano, A., Del Fabbro, D., Procopio,
tomies for colorectal cancer liver metastases. J. F., Montorsi, M. 2008b. Systematic extended
Gastrointest. Surg. 9: 1148–1153. right posterior sectionectomy: a safe and effec-
Torzilli, G., Montorsi, M., Gambetti, A., Del tive alternative to right hepatectomy. Ann. Surg.
Fabbro, D., Donadon, M., Bianchi, P., Olivari, 247(4): 603–611.
A. Treatment
7
Intraoperative Magnetic Resonance
Imaging for Radiofrequency Ablation
of Hepatic Tumors
Oliver F. Bathe and Houman Mahallati

INTRODUCTION been completed. This further impairs the


surgeon’s ability to estimate the extent of
Curative resection represents the best thera- tissue necrosis. Thus, RFA, while a suitable
peutic alternative for colorectal liver metas- adjunct in some instances, has technical
tases and primary liver tumors (Abdalla limitations that limit its use in the setting
et al., 2004; Llovet, 2005). For functioning of more extensive lesions.
metastatic neuroendocrine tumors, resec- Intraoperative magnetic resonance imag-
tion for the purpose of debulking is also ing (iMRI) represents a potential solution
a therapeutic consideration (Veenendaal to the limitations of RFA. Magnetic reso-
et al., 2006). However, in some patients, nance imaging provides excellent soft tis-
resection is not feasible because of ana- sue contrast and resolution, and it provides
tomic constraints, insufficient hepatic func- clear visualization of vascular structures.
tional reserve, or other safety limitations. It detects hepatic tumors with high sensi-
Radiofrequency ablation (RFA) represents tivity, and is known to be useful for fol-
an alternative therapy, but there are limita- lowing lesions after RFA (Aliberti et al.,
tions to this treatment modality. It is best 2004; Limanond et al., 2003). In view of
for small lesions that can be fully ablated this, iMRI may be a useful tool for opti-
by a single placement of the probe (Berber mizing the application of RFA or other
et al., 2005; Komorizono et al., 2003). Larger similar ablative adjuncts.
lesions require multiple probe placements
to fully encompass the volume of the target
lesion (Figure 7.1). Unfortunately, ultra- RADIOFREQUENCY ABLATION
sound does not clearly delineate the full AS A TREATMENT FOR
extent of the burn and it cannot distinguish
necrotic from viable tissue (Solbiati et al.,
LIVER TUMORS
2004). In fact, bubbles occupy the general
region of the heated liver and, if multiple
Principles of Radiofrequency Ablation
heat applications are required, the bub-
bles produced by the first burns have often Radiofrequency ablation is a localized
dissipated by the time the last burns have thermal treatment that enables tumor

81
82 O.F. Bathe and H. Mahallati

Figure 7.1. To ablate larger lesions, the largest available RFA probe should be utilized (a). However, if
the lesion is larger than the probe, multiple deployments are required (b). Even when this approach is
taken, small regions of the tumor may still be insufficiently ablated (arrow, c)

destruction. During the application of radio- of the alternating current, causing fric-
frequency energy, a high frequency alter- tional heating of the tissue. At tempera-
nating current is passed through the tip of tures above 60°C, intracellular proteins
an electrode into the tissue surrounding become denatured and cell death occurs.
the electrode. The ions within the tissue Radiofrequency ablation treatments typi-
are compelled to move in the direction cally result in local tissue temperatures in
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 83

excess of 100°C, which produces coagu- RFA is typically best for smaller lesions
lative necrosis of the tumor tissue and sur- (i.e., ≤ 3 cm), in the absence of coagulopathy
rounding hepatic parenchyma. or thrombocytopenia.
The temperature is greatest near the tips In patients who can tolerate a general
of the electrode array and falls rapidly anesthetic, operative (open) RFA is cho-
with increasing distance from the elec- sen in some circumstances. Larger lesions
trode. Only small cylindrical regions of typically require multiple deployments
coagulative necrosis are possible when to ablate the entire tumor area and they
using simple monopolar needle electrodes. are frequently best treated by open RFA.
Modern RFA systems typically consist of Lesions that cannot be easily accessed
an array of electrodes that deploy in an percutaneously may be treated by an open
umbrella formation. The result is a sphere procedure. Finally, RFA may be utilized
of tissue destruction with some viable in conjunction with resection in instances
cells closer to the periphery of the sphere. of bilateral hepatic disease (Abdalla et al.,
Because the electrode shaft is insulated, 2004; Elias et al., 2005).
the tract is spared of thermal injury, mak- Hepatocellular carcinoma most fre-
ing transcutaneous administration of RFA quently occurs in patients with underlying
possible. cirrhosis. Therefore, in this situation, treat-
ment options available to any individual
depend on the degree of liver dysfunction.
Indications
The best outcomes have been reported in
In general, RFA is indicated in patients patients undergoing liver transplantation
with primary hepatic malignancies and (Llovet, 2005), but the limited supply of
metastatic liver tumors in the absence donor livers precludes its widespread use.
of extrahepatic disease, in whom resec- Therefore, liver transplantation is typi-
tion is not feasible. Reasons for ineligi- cally reserved for patients with impaired
bility for resection might include poor liver function (i.e., Child B or C) in
general medical condition (precluding a whom there is a limited disease burden
major operation attendant with potentially (Llovet, 2005). In practical terms, there
considerable blood loss, under general is frequently a delay before an organ is
anesthesia) or insufficient hepatic func- available, so (percutaneous) RFA can be
tional reserve. utilized as a temporizing measure, to avoid
Radiofrequency ablation can be admin- tumor growth beyond the limits of criteria
istered percutaneously if lesions are clearly for transplantation (Lu et al., 2005). Other
visible by transcutaneous ultrasound, ena- treatments that must also be considered
bling one to position the probe into the include transarterial chemoembolization
lesion. Another prerequisite for this approach and ethanol ablation.
is that the lesion is easily accessible by Colorectal liver metastases infreque-
that route. Lesions that are poorly situated ntly occur in the setting of cirrhosis,
for the percutaneous approach include so hepatic functional reserve is a lesser
those directly adjacent to the gallbladder or concern. However, in patients pre-
another hollow viscus, or those lying high treated with chemotherapy, some degree of
up near the dome of the liver. Percutaneous hepatic dysfunction may occur secondary
84 O.F. Bathe and H. Mahallati

to chemotherapy-associated steatohepatitis hepatologists, medical oncologists, and


(Karoui et al., 2006). Resection is the best interventional radiologists.
option if all disease can be removed, with
recent reports of 5-year survivals up to
58% (Pawlik et al., 2005). Patients typi- Outcomes Associated
cally selected for RFA include those who with Radiofrequency Ablation
cannot tolerate surgery; those who have There is a paucity of data on long-term
prohibitive anatomy or small hepatic rem- outcomes following RFA. There are few
nants from previous resections; and those randomized controlled trials that com-
with bilateral disease (in which case RFA pare long-term survivals following RFA
may be used in conjunction with resection) to those seen in resection or other ablative
(Abdalla et al., 2004; Elias et al., 2005). techniques. What has been the focus of
There are fewer data on the utility of studies on RFA is the local control rate.
RFA in non-colorectal liver metastases. Technical success rates and local recur-
The exception is in patients with liver rence rates vary widely in the literature
metastases from neuroendocrine tumors. for colorectal liver metastases and hepa-
These tumors are classically more indolent tocellular carcinoma (Tables 7.1 and 7.2).
than most malignancies. Moreover, if they Because technical factors are likely to be
are functional, greater disease burden is responsible for suboptimal local control,
attendant with worse symptoms that are it is this outcome that is most likely to be
more difficult to treat medically. Therefore, influenced by iMRI.
even if it is impossible to ablate all grossly A number of factors have been identified
visible disease, RFA is frequently consid- as important determinants of local control.
ered as a debulking procedure to provide Tumor size appears to be the most impor-
palliation (Veenendaal et al., 2006). tant factor associated with local recurrence
In each of the above situations, treat- following RFA, regardless of the histology
ment options are diverse and decisions (Bleicher et al., 2003; Bowles et al., 2001;
on the most appropriate treatments are Kosari et al., 2002; Machi et al., 2001;
complex. It is therefore imperative that all Mulier et al., 2005). The local recurrence
options be considered by a multidiscipli- rate following an open (surgical) approach
nary group that includes hepatic surgeons, is superior to that seen when lesions are

Table 7.1. Outcomes following radiofrequency ablation for hepatocellular carcinoma.

# Patients Incomplete Local


Series (# lesions) ablation (%) recurrence (%) Survival
Livraghi et al., 1999 42 (52) 10
Pearson et al., 1999 34 2.9
Machi et al., 2001 18 (70) 4.3
Bowles et al., 2001 25 14.7 24 months (median)
Kosari et al., 2002 (16) 6.3
Shibata et al., 2002 36 (48) 4 12
Bleicher et al., 2003 21 22
Lencioni et al., 2003 52 (69) 9 5.7 98% (2 years)
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 85

Table 7.2. Outcomes following radiofrequency ablation for colorectal liver metastases.
# Patients Incomplete Local
Series (# lesions) ablation (%) recurrence (%) Survival
Pearson et al., 1999 46 4.3
Solbiati et al., 2001 117 (179) 2 39.1 46% (3 years)
Machi et al., 2001 (130) 9.2
Bowles et al., 2001 39 30.8 25 months (median)
Kosari et al., 2002 76 6.6
Bleicher et al., 2003 59 18.3
Aloia et al., 2006 30 37 57% (3 years)
(all solitary) 27% (5 years)
Abdalla et al., 2004 57 (110) 9 22% (4 years)

ablated via a percutaneous route (Mulier et al., 2003). There are currently two
et al., 2005). In a meta-analysis, local con- competing thermal ablation technologies:
trol was best for neuroendocrine metastases microwave coagulation therapy (MCT)
(Mulier et al., 2005). In that same study, and laser-induced thermotherapy (LITT).
local recurrence rates for HCC and colorec- Microwave coagulation therapy involves
tal metastases were similar. While several ultrasound-guided placement of an elec-
investigators have demonstrated that prox- trode into a lesion, followed by microwave
imity of a lesion next to a large vessel is treatment. Laser-induced thermotherapy
associated with an increased risk of local involves placement of a laser catheter into
recurrence (Bowles et al., 2001; Machi et al., a lesion and heat photocoagulation using a
2001) due to a heat sink effect, a meta- Nd-YAG laser. Each of these technologies
analysis failed to demonstrate the predic- can be applied during laparotomy or by
tive significance of this factor (Mulier percutaneous techniques.
et al., 2005). As a whole, these data sug- In general, MCT and LITT are advanta-
gest that a percutaneous approach should geous in that multiple electrodes or cath-
be reserved for small, easily accessible eters can be simultaneously placed to ablate
lesions. Moreover, adjunctive techniques larger or more complex liver lesions. Success
should be considered when treating large rates (i.e., complete ablation rates and local
lesions near major blood vessels by RFA. recurrence rates in primary or metastatic
tumors) are reportedly similar with each of
these technologies and with RFA. However,
Alternative Ablative Techniques
there are few direct comparisons between
Cryoablation and ethanol injection are two RFA and LITT or MCT. In one randomized
alternatives that are becoming less fre- controlled trial, RFA and MCT were found
quently used. RFA is better tolerated than to be equally safe; RFA and microwave
cryoablation, as there are few if any sys- ablation were approximately equivalent in
temic side effects, and most complications their ability to completely ablate tumor, but
are minor (de Baere et al., 2003; Pearson microwave ablation on average took less
et al., 1999). In HCC, RFA outcomes time to administer (Shibata et al., 2002).
appear slightly superior to those seen in More such comparisons will be essential
percutaneous ethanol injection (Lencioni for progress in this field.
86 O.F. Bathe and H. Mahallati

PROBLEMS ASSOCIATED occupy the general region of the heated


WITH MONITORING liver. When ablating larger lesions, where
THE ABLATIVE ENDPOINT multiple deployments are necessary, the
utility of ultrasound for estimating extent
So far few clinically applicable and reli- of the thermal injury becomes even more
able methods have been devised to moni- limited. That is, the bubbles produced by
tor the effectiveness of RFA, as reflected the first burns have often dissipated by the
by the complete necrosis of a given lesion. time the last burns have been completed,
Conventional methods of evaluating the further impairing the surgeon’s ability
completion of a thermal treatment include to estimate the extent of tissue necrosis.
monitoring for target temperatures or sud- Ultrasound, therefore, represents a poor
den increases in impedance, as well as tool for estimating the extent of the RFA
estimating the extent of injury with ultra- thermal injury.
sound. While each of these methods of
estimating the adequacy of RFA is suf-
ficient for small lesions, their utility is DEFINING THE EXTENT
limited when treating larger lesions. OF THERMAL INJURY BY
MAGNETIC RESONANCE
Impedance and Temperature
IMAGING
Temperature and impedance have each
been used as surrogate endpoints for an Magnetic Resonance Imaging
ablation applied to any given region (Arata Characteristics of Thermal Injury
et al., 2001; Frich et al., 2004). Indeed, Magnetic resonance imaging is an excel-
for small lesions, a sudden increase in lent imaging modality for assessment
tissue impedance (“roll-off”) is a significant of hepatic lesions (Martin and Semelka,
predictor of local control after RF ablation 2001; Noone et al., 2004). It has a number
(Arata et al., 2001). However, when mul- of advantages over CT imaging, such as
tiple deployments are required to cover a lack of ionizing radiation, less toxicity
larger lesion (Figure 7.1), impedance and with commonly used contrast agents, and
temperature become less relevant end- greater tissue contrast resolution for liver
points, and it becomes important to be lesions (Kirchin and Runge, 2003; Martin
able to gauge whether the whole lesion and Semelka, 2001). Magnetic resonance
has been ablated. Anatomic correlates of imaging has proven to be particularly use-
tissue kill therefore become necessary. ful for the evaluation of the adequacy of
thermal ablation and for monitoring for
Ultrasound recurrence. Indeed, MR images reflect
Ultrasound is typically utilized to tar- complete histopathological destruction of
get a given lesion. Unfortunately, ultra- tissue, although tumor has not been spe-
sound does not clearly delineate the full cifically analyzed (Lee et al., 2001).
extent of the burn, and it cannot distin- Most modern MR hepatic imaging
guish necrotic from viable tissue (Solbiati protocols rely on a combination of T1
et al., 2004). Rather, hyperechoic bubbles and T2 weighted images, with or without
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 87

administration of contrast agents. Using


the combined information from noncon-
trast T1 and T2 signal intensities and the
dynamic enhancement pattern of lesions
over time, lesions can be characterized
with great accuracy (Noone et al., 2004).
Excellent image quality requires that
motion artefacts from breathing be mini-
mized. This is usually achieved by having
patients suspend respiration for 20–25 s
while image data are acquired. Recent
advances enable the acquisition of high
quality images with patients breathing
freely (Asbach et al., 2005).
Thermal ablative treatments have been
shown to change the MR properties (T1
and T2 relaxation times) of tissues. Hepatic
parenchyma surrounding a site of ther-
mal injury initially contains inflammatory
changes, which gradually subside to be
replaced by granulation tissue (Goldberg
et al., 2000; Kuszyk et al., 2000). These Figure 7.2. Post-ablation changes 2 months after
changes manifest as an ill-defined rim RFA. The T1 opposed phase image (a) and the STIR
of mild signal increase on T2 weighted (T2 weighted) image (b) demonstrate hemorrhagic
contents within the RFA bed (white arrows)
images and on arterial phase enhance-
ment, which can be intense (Dromain
et al., 2002; Kuszyk et al., 2000). Over
time, the intensity and thickness of rim images, and liquefactive necrosis appears
enhancement decrease; these changes usu- as high signal (Kuszyk et al., 2000).
ally disappear by 6 months (Braga and Defining complete responses follow-
Semelka, 2005; Goldberg et al., 2000; ing RFA is unlike the assessment of the
Kuszyk et al., 2000). degree of response for any other type of
The ablated region similarly shows dis- antineoplastic therapy. That is, WHO and
tinct changes (Figure 7.2). In the first RECIST criteria for response to cancer
week after therapy, it is typically filled therapy rely on a decrease in size of lesions
with hemorrhagic debris from liquefactive (Therasse et al., 2000). On the other hand,
or coagulative necrosis (Dromain et al., the aim of RFA and other thermal ablative
2002; Kuszyk et al., 2000). Non-contrast techniques is to produce an area of ther-
T1 weighted images typically show a mocoagulation greater than or equal to the
high signal area with some heterogeneity, size of the tumor. Therefore, conventional
depending on the stage of hemorrhage. size criteria are not appropriate measures
Coagulative necrosis typically appears as of response to RFA (Dromain et al., 2002).
low signal on non-contrast T2 weighted Early series showed that contrast enhanced
88 O.F. Bathe and H. Mahallati

CT and MRI are useful modalities for


assessing the size of the ablated area.
The non-enhancing region corresponded
to within 2 mm of the size of the area
of coagulative necrosis (Goldberg et al.,
2000). Therefore, MRI clearly defines the
completeness of necrosis and the extent of
the necrotic injury.
Subtraction images are also useful
adjuncts for MRI. Specifically, subtrac-
tion images in which precontrast (mask)
images are subtracted from postcontrast
(enhanced) images can better demonstrate
areas of enhancement. Such images have
been shown to be useful in assessing
hepatic lesions (Yu et al., 2005). In our
experience, application of subtraction
imaging greatly improves one’s ability to
determine the extent of thermal injury and
the characteristics of enhancement around
post-RFA beds.
Magnetic resonance imaging is similarly Figure 7.3. Local recurrence of hepatocellular car-
useful for monitoring lesions for local recur- cinoma following radiofrequency ablation. Axial
rence (Figure 7.3). It is slightly superior to and coronal reformatted post-contrast (Gd-DTPA)
CT in its sensitivity for detecting recur- subtraction images show nodular enhancement at
the superior peripheral margin of the RFA bed of a
rence (Dromain et al., 2002). In the first large lesion (white arrows)
few months after RFA, size criteria are not
particularly helpful. Rather, the margins of
the RFA bed must be evaluated. A smooth
internal contour and a smooth, thin rim-like Enhancing the Definition of Extent
enhancement of the margins are expected of Thermal Injury
around an ablated cavity. The presence of Magnetic resonance contrast agents are
nodular enhancement at the periphery of essential for the accurate definition of the
the ablation zone, or any protuberance dis- extent of thermal injury. However, conven-
torting the smooth inner margin are signs tional MR contrast agents are not ideal in
of tumor recurrence (Braga and Semelka, the setting of iMRI. Therefore, the utility
2005; Goldberg et al., 2000; Kuszyk et al., of newer contrast agents must be explored
2000). After the first few months, one may in the context of iMRI. Liver MRI is most
expect a progressive reduction in the size of often performed with contrast agents that
the ablative lesion (Dromain et al., 2002). are extracellular space agents (Hyslop
Any increase in size of the ablative zone is et al., 2005). These are comprised of solu-
considered a sign of recurrence (Braga and tions of gadolinium (Gd) chelates that
Semelka, 2005; Limanond et al., 2003). are rapidly and almost entirely cleared
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 89

by renal excretion. Enhancement with


such agents reflects tissue vascularity,
flow dynamics, and perfusion (Weinmann
et al., 1991). Liver MRI scans are typically
performed with three postcontrast phases:
arterial, portal venous, and equilibrium (or
interstitial) phases. Conventional extracel-
lular space agents are useful in determin-
ing enhancement of post-RFA beds and
they are helpful for the detection of tumor
recurrence (Dromain et al., 2002; Kuszyk
Figure 7.4. Hepatocellular carcinoma following
et al., 2000). administration of a hepatocyte-specific contrast
Tissue-specific agents for hepatic imag- agent. With Gd-EOB-DTPA (Primovist), hepato-
ing include hepatocyte-specific and retic- cytes are T1 hyperintense and tumor is hypointense
ulo-endothelial system-specific agents. on this opposed phase T1 weighted image
Early hepatocyte-specific agents were
based on manganese (e.g., MnDPDP),
have functioning hepatocytes (Huppertz
but they are not in widespread clinical use
et al., 2004). If a lesion does not contain
(Hyslop et al., 2005). While these agents
functioning hepatocytes, it will be hypoin-
provided excellent visualization of the bil-
tense on T1W images compared to func-
iary tree and hepatic parenchyma, the lack
tioning hepatocytes that have prolonged
of dynamic contrast enhancement infor-
signal increase because of uptake of Gd
mation limits their clinical utility.
compounds (Figure 7.4). Gd-BOPTA
Recently, a new class of contrast agents
results in maximal hepatic parenchymal/
has been introduced that has the properties
biliary enhancement at approximately
of both extracellular space agents as well as
1–2 h after injection and Gd-EOB-DTPA
hepatocyte-specific agents. These include
has the highest liver-to-lesion contrast at
gadobenate dimeglumine (Gd-BOPTA)
20–45 min after injection (Huppertz et al.,
and gadoxetic acid (Gd-ethoxybenzyl-
2004; Hyslop et al., 2005). In the case
diethylenetriaminepentaacetic acid;
of Gd-EOB-DTPA, despite the fact that
Gd-EOB-DTPA). Both contrast agents are
contrast between lesion and normal liver
distributed in the extracellular space in
is maximal at 20–45 min, this temporal
the early stages after injection and offer
window has been shown to persist for at
dynamic contrast enhancement data, and
least 90–120 min. The use of these contrast
both are ultimately taken up by function-
agents in the context of monitoring ther-
ing hepatocytes with subsequent biliary
mal damage from RFA has been relatively
excretion. Gd-BOPTA has 3–5% biliary
unexplored.
excretion and approximately 50% of
Gd-EOB-DTPA is excreted in the bile
(Hyslop et al., 2005). The hepatocyte- Potential Utility of Functional
specific uptake results in T1 shortening of Magnetic Resonance Imaging
the liver parenchyma signal and results in While there are good MRI criteria for
increased conspicuity of lesions that do not complete tumor ablation, functional MR
90 O.F. Bathe and H. Mahallati

imaging may also be useful in supporting recurrence rates should approach those
the lack of significant residual tumor after seen after resection.
ablation. The technology exists to monitor Lesions are often difficult to visualize
intratumoral temperature by MRI (Dick in their entirety with diagnostic modali-
et al., 2003; Puls et al., 2003; Weidens- ties other than MRI. This is especially
teiner et al., 2004). While functional MR true for some neuroendocrine tumors and
techniques have been used in neuroimaging some HCCs. Monitoring the extent of
for some time, it is only relatively recently the ablation is similarly difficult, as the
that these techniques have become appli- ablated region is immediately filled with
cable in abdominal imaging. Magnetic hyperechoic bubbles. The difficulty is
resonance spectroscopy has been used to compounded in large lesions, where mul-
evaluate patients with prostate cancer and tiple overlapping RFA applications must
renal cell carcinoma (Hyslop et al., 2005; be utilized to cover a lesion larger than
Katz-Brull et al., 2005). Kamel et al. the probe (Figure 7.1). In this instance,
(2006) used functional MR techniques as treatment is planned such that the zones
an adjunct to conventional MR imaging to of necrosis overlap, to ensure complete
assess response to chemotherapy (TACE) destruction of the tumor. However, during
in patients with HCC. Early results are the latter RFA applications, the bubbles
promising, and MR imaging has the unique disappear in the initially ablated regions,
advantage of offering a combination of making it impossible to keep track of the
anatomic as well biochemical informa- total extent of the burn. The area of the
tion in assessing tumor response. These burn is not always consistent, particularly
combined approaches may offer improved if there is a nearby large vessel which
sensitivity and specificity in assessing induces a heat sink. Finally, the surgeon
response. cannot always accurately reach the desired
target with the RFA probe. All of these
obstacles can limit the surgeon’s capability
INTRAOPERATIVE to completely ablate any lesion, particu-
MAGNETIC RESONANCE larly with adequate margins (Mulier et al.,
IMAGING AS AN ADJUNCT 2005). Improved intraoperative imaging
may help to promptly identify local treat-
TO RADIOFREQUENCY ment failures, reducing the need for repeat
ABLATION treatments and, perhaps, reducing local
recurrence rates.
Rationale
As described earlier, RFA is a promis-
ing technology that enables ablation of Technical Developments
lesions that may otherwise not be resect- The first intraoperative MRI systems were
able. However, technical limitations limit relatively recently described (Jolesz and
the efficacy of the treatment. Local failure Blumenfeld, 1994; Schenck et al., 1995),
rates and local recurrence rates are unac- and operative interventions guided by
ceptably high, particularly in larger lesions these systems were pioneered by the neu-
that are difficult to access. Ideally, local rosurgical community. The first systems
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 91

consisted of 2 weak magnets (i.e., ≤ 0.5 the 1.5T magnet remains retracted except
tesla (T) ), which were arranged in a verti- when it is utilized for imaging (Sutherland
cal biplanar donut configuration (Schenck et al., 2002). The use of a movable magnet
et al., 1995). The more recent iterations of is more “patient focused” than previously
these systems permitted excellent vertical developed systems. That is, the operating
and lateral exposure between magnets. table is fixed, the patient is positioned
Subsequently, horizontal low-field sys- once, and the magnet moves to the patient,
tems were developed (Steinmeier et al., as opposed to transporting the patient to
1998), but patients had to be transported the magnet. When the magnet is retracted,
between an operating theater and an imag- conventional surgical instruments can be
ing room. In this way, MR incompatibility utilized behind the 5-Gauss line. This
of surgical instruments was no longer an patient-focused approach is safer in that
issue, but patient safety was suboptimal it minimizes the anesthetic risk associated
due to the need for transportation during with patient transport, and it minimizes
the procedure. Using such systems, virtu- disruption of the surgical procedure.
ally any anatomical region could be easily Recently, we have demonstrated the fea-
reached for interventional procedures, as sibility of doing complex open abdominal
the patient could be placed supine, prone operations using this system (Bathe et al.,
or lateral along the longitudinal axis of 2006). Major hepatic surgery would not be
the magnets. However, such systems had readily feasible in a high-field open magnet
limited imaging capabilities and resolution system, given the equipment requirements
due to the low magnetic field strength. and access constraints. Our ability to do
Stronger magnetic field strength increa- surgery with a 1.5T magnet was facilitated
ses signal-to-noise ratio, enhances image by the portability of the magnet. The one
resolution, and accelerates image acquisi- drawback of a closed unit such as the one
tion. Therefore, “high-field” imaging sys- we use is that it does not allow real-time
tems incorporating stronger magnets were monitoring of the ablation or changes in
developed (Sutherland et al., 2002; Truwit intralesional temperature, which is fea-
and Hall, 2001). These 1.5T systems pro- sible for percutaneous procedures (Dick
duced image quality that was equivalent to et al., 2003; Puls et al., 2003). Therefore,
diagnostic scanners. Moreover, the higher a moveable, retractable system is essential
magnetic field strength enabled physi- for safely performing high-field imaging
ologic MR imaging, such as perfusion in the setting of open intracavitary surgery,
MR, spectroscopic MR, and functional but real-time information is not available.
MR. However, because of the enhanced Others have described minimally inva-
magnetic field strength, it was not possible sive intraabdominal and hepatic proce-
to perform procedures that were moni- dures in low magnetic field systems (i.e.,
tored “real-time” by MRI. Rather, the sys- 0.2–0.5T) (Dick et al., 2003; Morikawa
tems were limited to taking static images et al., 2002). Typically, these systems
before, during and after surgery. employ open magnets that enable access
At our institution, the neurosurgeons for percutaneous procedures. The imag-
developed a unique iMRI system that is ing limitations imposed by such low-field
comprised of an operating theatre in which systems limit their use in hepatic surgery.
92 O.F. Bathe and H. Mahallati

Moreover, with a 1.5T magnet temperature MRI are similarly not eligible for this type
mapping of the thermal burn is possible of approach. Finally, because intraopera-
(Weidensteiner et al., 2004), which poten- tive cardiac monitoring is impaired by the
tially provides additional important intra- iMRI system, patients with significant cor-
operative information. Recently, a report onary artery disease are currently excluded
demonstrated that percutaneous RFA was from these procedures.
possible through a closed high-field (1.5T)
system (Mahnken et al., 2004), but open
intracavitary surgery with such a strong Conduct of Intraoperative
magnet has not so far been reported out- Magnetic Resonance Imaging
side of our own experience. for Radiofrequency Ablation
The iMRI system that is used at the
University of Calgary has been described
Potential Indications
in previous reports (Bathe et al., 2006;
Patients currently selected for iMRI as an Sutherland et al., 2002). Briefly, the sys-
adjunct to surgery are those in whom it is tem is based on a moveable 1.5T mag-
anticipated that RFA (with or instead of net designed and constructed by Magnex
resection) is a likelihood and that iMRI UK. The system enables the positioning
might be helpful in making intraopera- of the patient and, because the magnet
tive decisions. Specific indications include enters and exits the room as required,
the following situations: (1) large lesions the patient remains the focal point. The
(≥ 3 cm) that will be treated by RFA; (2) intubated patient is positioned on a MR
recurrent lesions in which RFA will play compatible table that is bolted to the
an essential role; (3) lesions that are vis- floor, with the anesthetic machine at his/
ible on MRI; (4) lesions located at difficult her foot. The hands must be tucked to
sites, such as near potential heat sinks, the side to accommodate the patient in the
close to the gallbladder or any other hol- magnet. An inflatable positioning device
low viscus, or up near the diaphragm. is inserted under the right flank to elevate
The present closed iMRI systems utilize the patient’s right side slightly, enabling
magnets with a small bore. Therefore, a more complete right subcostal incision,
large patients who did not fit into a closed which is essential for open hepatic surgery.
magnet system cannot undergo this proce- The anesthetic tubes are passed along the
dure, until larger bore magnets are devel- left flank to the feet, where the anesthetic
oped. Open systems may be less limited in equipment resides as far from the magnet
this regard, although it is essential that any as feasible.
lesions targeted by this approach are vis- Following induction of anesthesia, base-
ible in the therapeutic magnet. Frequently, line images are obtained and compared to
in patients with a larger body habitus, a diagnostic MRI taken before the date of
lesions may be suboptimally visible if surgery. Sequence parameters are set to
they lie outside of the magnetic “sweet minimize breath-hold time and maximize
spot”. Patients who had ferrous prostheses coverage. Time to repetition (TR), echo
or other ferrous foreign bodies that would time (TE), flip angle, echo train lengths
normally be a contraindication to diagnostic (ETL), and slice thickness are all adjusted
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 93

to optimize imaging of the liver in each check of probe position is made with ultra-
patient. All patients are imaged with res- sound. The protocol for increases in elec-
piration suspended and monitored by the trical power delivered to the probe is set
attending anesthesiologist. Axial precon- by the manufacturer, and this is dependent
trast 2D T1 weighted gradient echo (GRE) on probe size. Larger probes require more
images are obtained with or without fat power to achieve the target temperature
saturation, and axial 2D fast spin echo throughout the volume of the burn region.
(FSE) T2 weighted images with fat satura- Where multiple deployments are required
tion are obtained. An intravenous bolus to cover the entire tumor volume, it is best
of 0.15 mmol/kg gadopentetate dimeg- to do the first deployments at the least
lumine (Gd-DTPA; Magnevist; Berlex accessible part of the tumor. Usually, that
Laboratories, Wayne, NJ) is then admin- means targeting the most posterior and
istered, and postcontrast T1 weighted superior aspects of the lesion. Thereafter,
fat saturated images are acquired, with the probe is pulled back 2 cm, the tines
parameters identical to the precontrast T1 are redeployed, and another thermal abla-
weighted fat saturated images. All target tion cycle is applied. This is repeated
lesions are visualized, measured, and until it is estimated that the entire lesion
localized relative to nonmobile extrahe- has been ablated. It is best if the regions
patic landmarks (e.g., vertebral body and ablated overlap, to ensure that no areas
liver capsule). In particular, it is important are exposed to insufficient heat to induce
to measure the distances from these non- coagulative necrosis. The latter ablations
mobile landmarks to the tumor edges. The are more difficult to target accurately with
mobile magnet is then retracted, allowing ultrasound, as the lesion fills with bubbles
the safe introduction of normal surgical after the first ablation. Therefore, if it is
instruments to the surgical field. anticipated that multiple deployments will
Following a laparotomy, the liver is be required, it is essential to plan probe
mobilized as necessary to permit access placements prior to initiating the ablation
to the targeted hepatic segments. An intra- for each lesion.
operative ultrasound is performed, and In general, we take an aggressive
decisions regarding the final operative approach to thoroughly burn each of the
approach are made. It is feasible using target lesions. For lesions that are close to
this system to do a resection, if that is the gallbladder, it is essential to remove
considered appropriate. RFA is performed the gallbladder to avoid perforation sec-
for any remaining lesions, using as large a ondary to thermal injury. For lesions that
probe as required to cover as much of the are located near the center of the liver, care
tumor field as possible. Currently, probes must be taken to avoid a thermal injury
up to 5 cm are available. If it is expected to the bile ducts. Injury to the bile ducts
that the probe will cover the entire diam- may be manifested postoperatively by a
eter of the tumor in a single deployment, bile leak, a bile duct stricture, or complete
the probe is initially guided with the aid of disruption of the bile ducts; such compli-
ultrasound to the center of the tumor. Once cations add considerable morbidity to the
the probe tip is at the desired region of the procedure. If necessary, the bile ducts can
tumor, the tines are deployed and a final be infused with cooled saline via the cystic
94 O.F. Bathe and H. Mahallati

duct, to reduce the chance of this type of Gd-EOB-DTPA allows visualization of


injury. All targeted lesions are resected the lesion prior to RFA and precise deline-
and/or ablated prior to MRI assessment of ation of RFA margins, even after multiple
the margins. time-consuming RFA deployments.
After resection and/or RFA, all instru- When using Gd-EOB-DTPA as the con-
ments are counted; metal objects are trast agent, it is administered 20–120 min
removed from the field, and the operating prior to taking intradissectional images.
field is covered with protective drapes. If possible, the same dose of contrast
An intradissectional image is obtained, is utilized for preoperative imaging and
to ensure complete disappearance of the intradissectional imaging. The preabla-
target lesion(s) with adequate margins. We tion size of the lesion is easily determined
strive to burn the entire area of the tumor on T1-weighted imaging by measuring
plus a margin of normal tissue of at least the hypo-intense lesion diameters against
5 mm. This can be estimated by measur- the hyperintense hepatic parenchyma. The
ing the distance from selected nonmo- ablation zone can then be measured on T1
bile neighboring structures to the various and T2-weighted images, ensuring that
margins of the burn and comparing these it exceeds the size of the original lesion.
distances to those measured before the In our limited experience, on delayed
surgery, where the edge of the tumor was (90 min) post-Gd-EOB-DTPA images, the
mapped. If necessary, more ablative treat- ablation bed is much more hyperintense
ments are applied, and further confirma- than hepatic parenchyma (Figure 7.5B).
tory images are taken. This corresponds to the hemorrhagic and
Digital subtraction images consisting of necrotic products that are known to fill
a mathematical subtraction of T1-weighted the recently ablated region (Braga and
data acquired before and after IV contrast Semelka, 2005). The use of Gd-EOB-
facilitate estimation of the burn extent DTPA eliminates the need for multiple
and determination of whether viable doses of contrast agent with multiple RFA
tumor remains. Such images pronounce attempts. In addition, subtraction images
enhanced structures like residual tumor: are no longer required to estimate the
when the center of the lesion is a signal extent of thermal injury.
void (i.e., black), this confirms that there Several technical limitations with our
is no residual tumor (Figures 7.5D, E). In system have become apparent during our
our early work, following administration development of this technology for hepatic
of Gd-DTPA, in cases where repeat burns surgery. First, since our iMRI system was
were needed, repeat imaging was limited primarily engineered for brain and spine
by dose (Bathe et al., 2006). surgery, the magnetic isocenter (“sweet
Most recently we have employed spot”) was posterior to the liver. Therefore,
Gd-EOB-DTPA as a contrast agent (Figure more anterior lesions were difficult to
7.5). The prolonged hepatocyte uptake of see because of poor signal-to-noise ratio.
this agent offers an extended temporal A modified RF coil is required to address
window which can be used to advan- this problem. Second, lesions in segment
tage in the intraoperative imaging. The IV of the liver can be obscured by the
90–120 min temporal window offered by aortic “ghost” artefact. If the lesion is veiled
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 95

Figure 7.5. Intraoperative MRI using a hepatocyte-specific contrast agent. (a) A pre-ablation T1 weighted
image after Gd-EOB-DTPA (Primovist) shows tumor as hypointense against a hyperintense liver. (b) The
intradissectional image demonstrates the ablation zone relative to tumor. The thermal ablation zone is
more T1 hyperintense peripherally, compared to surrounding liver. The central hypointensity represents
tumor bed, which is excessively close to the ablative margin at the anterior aspect (white arrow). Further
deployments were required to improve this margin. (c, d) Images taken 2 weeks later confirmed that the
final ablation margin was sufficient. Following administration of Gd-EOB-DTPA, the T1 weighted image
(c) demonstrates the extent of the ablation bed. The subtraction image confirms the absence of residual
tumor. (e, f) Images taken 8 months later demonstrate no tumor recurrence. Post-contrast (Gd-DTPA)
3D T1 weighted images (VIBE) demonstrate the ablation bed (e). The subtraction images confirm no
enhancement in the RFA bed (f)
96 O.F. Bathe and H. Mahallati

Developments in Intraoperative
Magnetic Resonance Imaging
The magnet itself must be reengineered to
enhance the utility of MRI in a wide variety
of clinical circumstances. It will be essen-
tial to engineer such a magnet so that the
magnetic “sweet spot” is large enough to
encompass the targeted organ. Finally, the
magnet bore must be built so that it is large
enough to accommodate larger patients, as
the prevalence of obesity is increasing.
The ideal iMRI system would allow
Figure 7.6. Lesions in segment IV of the liver rapid, high quality imaging with combined
(white arrow) can be obscured by the aortic “ghost”
artefact. If the lesion is veiled by aortic ghost, then
morphologic and functional information.
tilting the patient effectively separates the target Systems must be built with a stronger
lesion from the artefact. magnetic field (e.g., ≥ 3.0T), as this ena-
bles faster image acquisition, greater reso-
lution, and better functional imaging. On
by aortic ghost, then tilting the patient the other hand, MRI using stronger mag-
effectively separates the target lesion from nets is associated with some disadvantages
the artefact (Figure 7.6). For this reason, such as increased susceptibility artefacts,
we routinely place air bags beneath each increased energy deposition, and altered
flank, enabling repositioning with less T1 properties of tissues (Hyslop et al.,
trouble. Third, we have found that image 2005; Lauzon et al., 2006). In the case of
quality is unacceptable in large patients. intraoperative MR, the increased suscep-
Not only is it difficult to fit these patients tibility artefacts warrant special attention.
into the magnet, it is impossible to image The presence of air/tissue interfaces in the
the anterior aspect of their liver. Each of surgical field can lead to signal loss from
these limitations can be addressed with susceptibility and may decrease the image
specific design changes to the magnet (see quality, negating the advantages of high
below). field imaging. Sequences and systems will
need to be optimized to minimize artefacts
in order to gain the maximum potential
Future Developments
from higher field intra-operative systems.
Image-guided ablation is an important Systems should ideally offer targeting
alternative to resection. However, to capabilities and real-time monitoring of
ensure that its therapeutic effectiveness therapies such as RFA. Magnetic resonance
better resembles that of resection, specific has been used to monitor RF ablation in
technological developments will be neces- real-time in animal models (Lepetit-Coiffe
sary. This is particularly important for the et al., 2006), but this is not yet feasible in
treatment of larger lesions, which have so high-field systems so far described in clin-
far infrequently been treated using these ical use. Real-time MR monitoring of RFA
techniques. would be greatly enhanced by robotics,
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 97

which would allow the remote insertion noise, impairing the acquisition of quality
of instruments into the lesion (McBeth images when monitoring the progression
et al., 2004). Robots are indefatigable, of the burn. To address the latter issue,
have superior spatial resolution and excel- Zhang et al. (1998) have described a
lent geometric accuracy. The development switching circuit to reduce this noise.
of an MR-compatible robot would there- While we have only had experience with
fore facilitate real-time monitoring of RFA RFA, MCT and LITT could be explored
of liver lesions. in the context of MR-guided ablation of
Finally, iMRI could benefit from the liver lesions. The MRI characteristics of
development of novel contrast agents. ablation zones produced by each of these
While conventional extracellular space methods are unlikely to differ significantly
agents are useful in determining enhance- from those produced by RFA (Braga and
ment of post-RFA beds and potential tumor Semelka, 2005). MR-guided insertion of
recurrence (Dromain et al., 2002; Kuszyk microwave electrodes and real-time moni-
et al., 2000), in our experience, the tem- toring of the ablation by MRI has success-
poral window offered by such agents and fully been performed (Morikawa et al.,
the dose limitations (Kirchin and Runge, 2002). Laser-induced thermotherapy may
2003) limit their utility in the intraopera- be particularly suited for MR imaging
tive setting. Ideally, a contrast agent that because laser fibers and catheter systems
has greater retention and better definition are not affected by and do not interfere with
of the target lesion relative to the extent of MR signal (Puls et al., 2003). Puls et al.
thermal injury would be useful. Gd-EOB- (2003) recently described a new miniatur-
DTPA fulfills these requirements, but it ized irrigated laser catheter system that
may not be as good for lesions containing can be inserted percutaneously under MR
functioning hepatocytes, such as hepatic guidance and that allows monitoring of
adenoma or well differentiated hepatocel- therapy in a closed high-field MR imager.
lular carcinoma. Tumor-specific contrast In conclusion, radiofrequency ablation
agents may be useful. Necrosis-avid agents may occasionally cause incomplete tumor
and high molecular weight intravascular necrosis. Moreover, the local recurrence
agents have been used in animal models rates following RFA of liver tumors are
to assess response to RFA therapy (Kim high, especially in larger tumors that are
et al., 2005; Ni et al., 2006), but we have difficult to access. One contributing fac-
not yet tested them in the clinical context. tor to this poor local control rate is the
difficulty in monitoring thermal ablations
Development of Alternative Ablative of larger tumors in which multiple RFA
Techniques deployments are required. Additionally,
Radiofrequency ablation has some dis- the margins of ablation may be inadequate.
advantages, when utilized in the context Intraoperative MRI enables the surgeon
of MRI. If real-time monitoring of the to better estimate the technical success of
ablation is to be done using MRI, then it RFA as well as the size of the ablative mar-
is essential to utilize MR-compatible tita- gins. However, it is still unknown whether
nium probes, which are quite expensive. iMRI changes local recurrence rates.
Moreover, RFA produces electromagnetic Complex hepatic surgeries (i.e., resections
98 O.F. Bathe and H. Mahallati

and/or RFA) are feasible and safe in the Radiofrequency ablation in 447 complex unre-
University of Calgary iMRI suite. The 1.5T sectable liver tumors: lessons learned. Ann. Surg.
Oncol. 10:52–58.
system enables high resolution imaging.
Bowles, B.J., Machi, J., Limm, W.M., Severino,
Our initial experience has identified some R., Oishi, A.J., Furumoto, N.L., Wong, L.L., and
technical limitations to our system. This Oishi, R.H. 2001. Safety and efficacy of radi-
experience will point the way to future ofrequency thermal ablation in advanced liver
system modifications. tumors. Arch. Surg. 136:864–869.
Braga, L., and Semelka, R. 2005. Magnetic reso-
nance imaging features of focal liver lesions
REFERENCES
after intervention. Top Magn. Reson. Imaging
Abdalla, E., Vauthey, J., Ellis, L., Ellis, V., Pollock, 16:99–106.
R., Broglio, K., Hess, K., and Curley, S. 2004. deBaere, T., Risse, O., Kuoch, V., Dromain, C.,
Recurrence and outcomes following hepatic Sengel, C., Smayra, T., Din, M.G.E., Letoublon,
resection, radiofrequency ablation, and com- C., and Elias, D. 2003. Adverse events during
bined resection/ablation for colorectal liver radiofrequency treatment of 582 hepatic tumors.
metastases. Ann. Surg. 239:818–825. Am. J. Roentgenol. 181:695–700.
Aliberti, C., Soriani, M., Tilli, M., Benea, G., Giorgi, Dick, E., Wragg, P., Joarder, R., Jode, M.D., Lamb,
U.D., and Fiorentini, G. 2004. Radiofrequency G., Gould, S., and Gedroyc, W. 2003. Feasibility
ablation of liver malignancies: MRI for evalua- of abdomino-pelvic T1-weighted real-time ther-
tion of response. J. Chemother. 16:79–81. mal mapping of laser ablation. J. Magn. Reson.
Aloia, T.A., Vauthey, J.N., Loyer, E.M., Ribero, D., Imaging 17:197–205.
Pawlik, T.M., Wei, S.H., Curley, S.A., Zorzi, D., Dromain, C., de Baere, T., Elias, D., Kuoch, V.,
and Abdalla, E.K. 2006. Solitary colorectal liver Ducreux, M., Boige, V., Petrow, P., Roche, A.,
metastasis: resection determines outcome. Arch. and Sigal, R. 2002. Hepatic tumors treated with
Surg. 141:460–466. percutaneous radio-frequency ablation: CT and
Arata, M., Nisenbaum, H., Clark, T., and Soulen, MR imaging follow-up. Radiology 223:255–262.
M. 2001. Percutaneous radiofrequency ablation Elias, D., Baton, O., Sideris, L., Boige, V., Malka,
of liver tumors with the LeVeen probe: is roll-off D., Liberale, G., Pocard, M., and Lasser, P. 2005.
predictive of response? J. Vasc. Interv. Radiol. Hepatectomy plus intraoperative radiofrequency
12:455–458. ablation and chemotherapy to treat technically
Asbach, P., Klessen, C., Kroencke, T.J., Kluner, C., unresectable multiple colorectal liver metas-
Stemmer, A., Hamm, B., and Taupitz, M. 2005. tases. J. Surg. Oncol. 90:36–42.
Magnetic resonance cholangiopancreatography Frich, L., Bjørnerud, A., Fossheim, S., Tilung, T.,
using a free-breathing T2-weighted turbo spin- and Gladhaug, I. 2004. Experimental application
echo sequence with navigator-triggered pro- of thermosensitive paramagnetic liposomes for
spective acquisition correction. Magn. Reson. monitoring magnetic resonance imaging guided
Imaging 23:939–945. thermal ablation. Magn. Reson. Imaging 52:
Bathe, O., Mahallati, H., Sutherland, F., Dixon, E., 1302–1309.
Pasieka, J., and Sutherland, G. 2006. Complex Goldberg, S.N., Gazelle, G.S., Compton, C.C.,
hepatic surgery aided by a 1.5-tesla moveable Mueller, P.R., and Tanabe, K.K. 2000. Treatment
magnetic resonance imaging system. Am. J. of intrahepatic malignancy with radiofrequency
Surg. 191:598–603. ablation: radiologic-pathologic correlation.
Berber, E., Pelley, R., and Siperstein, A. 2005. Cancer 88:2452–2463.
Predictors of survival after radiofrequency ther- Huppertz, A., Balzer, T., Blakeborough, A., Breuer,
mal ablation of colorectal cancer metastases to J., Giovagnoni, A., Heinz-Peer, G., Laniado, M.,
the liver: a prospective study. J. Clin. Oncol. Manfredi, R.M., Mathieu, D.G., Mueller, D.,
23:1358–1364. Reimer, P., Robinson, P.J., Strotzer, M., Taupitz,
Bleicher, R.J., Allegra, D.P., Nora, D.T., Wood, M., and Vogl, T.J. 2004. Improved detection of
T.F., Foshag, L.J., and Bilchik, A.J. 2003. focal liver lesions at MR imaging: multicenter
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 99

comparison of gadoxetic acid-enhanced MR radiofrequency ablation of hepatic malignancies.


images with intraoperative findings. Radiology J. Gastrointest. Surg. 6:255–263.
230:266–275. Kuszyk, B.S., Boitnott, J.K., Choti, M.A., Bluemke,
Hyslop, W.B., Balci, N.C., and Semelka, R.C. D.A., Sheth, S., Magee, C.A., Horton, K.M.,
2005. Future horizons in MR imaging. Magn. Eng, J., and Fishman, E.K. 2000. Local tumor
Reson. Imaging Clin. N. Am. 13:211–224. recurrence following hepatic cryoablation: radi-
Jolesz, F.A., and Blumenfeld, S.M. 1994. ologic-histopathologic correlation in a rabbit
Interventional use of magnetic resonance imag- model. Radiology 217:477–486.
ing. Magn. Reson. Q. 10:85–96. Lauzon, M.L., Mahallati, H., and Frayne, R. 2006.
Kamel, I.R., Bluemke, D.A., Eng, J., Liapi, E., Time-efficient breath-hold abdominal MRI at
Messersmith, W., Reyes, D.K., and Geschwind, 3.0 T. Am. J. Roentgenol. 187:649–657.
J.F. 2006. The role of functional MR imaging in Lee, J., Lee, J., Kim, S., Kim, C., and Mun, W.
the assessment of tumor response after chem- 2001. MR imaging-histopathologic correlation
oembolization in patients with hepatocellular of radiofrequency thermal ablation lesion in a
carcinoma. J. Vasc. Interv. Radiol. 17:505–512. rabbit liver model: observation during acute and
Karoui, M., Penna, C., Amin-Hashem, M., Mitry, chronic stages. Korean J. Radiol. 2:151–158.
E., Benoist, S., Franc, B., Rougier, P., and Lencioni, R., Allgaier, H.-P., Cioni, D., Olchewski, M.,
Nordlinger, B. 2006. Influence of preoperative Deibert, P., Crocetti, L., Frings, H., Laubenberger,
chemotherapy on the risk of major hepatec- J., Zuber, I., Blum, H., and Bartolozzi, C. 2003.
tomy for colorectal liver metastases. Ann. Surg. Small hepatocellular carcinoma in cirrhosis: ran-
243:1–7. domized comparison of radio-frequency thermal
Katz-Brull, R., Rofsky, N.M., Morrin, M.M., ablation versus percutaneous ethanol injection.
Pedrosa, I., George, D.J., Michaelson, M.D., Radiology 228:235–240.
Marquis, R.P., Maril, M., Noguera, C., and Lepetit-Coiffe, M., Quesson, B., Seror, O., Dumont,
Lenkinski, R.E. 2005. Decreases in free choles- E., Le Bail, B., Moonen, C.T., and Trillaud, H.
terol and fatty acid unsaturation in renal cell car- 2006. Real-time monitoring of radiofrequency
cinoma demonstrated by breath-hold magnetic ablation of rabbit liver by respiratory-gated
resonance spectroscopy. Am. J. Physiol. Renal quantitative temperature MRI. J. Magn. Reson.
Physiol. 288:F637–F641. Imaging 24:152–159.
Kim, T.J., Moon, W.K., Cha, J.H., Goo, J.M., Limanond, P., Zimmerman, P., Raman, S.S.,
Lee, K.H., Kim, K.H., Lee, J.W., Han, J.G., Kadell, B.M., and Lu, D.S. 2003. Interpretation
Weinmann, H.J., and Chang, K.H. 2005. VX2 of CT and MRI after radiofrequency ablation
carcinoma in rabbits after radiofrequency abla- of hepatic malignancies. Am. J. Roentgenol.
tion: comparison of MR contrast agents for 181:1635–1640.
help in differentiating benign periablational Livraghi, T., Goldberg, S.N., Lazzaroni, S., Meloni,
enhancement from residual tumor. Radiology F., Solbiati, L., and Gazelle, G.S. 1999. Small
234:423–430. hepatocellular carcinoma: treatment with radio-
Kirchin, M.A., and Runge, V.M. 2003. Contrast frequency ablation versus ethanol injection.
agents for magnetic resonance imaging: safety Radiology 210:655–661.
update. Top Magn. Reson. Imaging 14:426–435. Llovet, J.M. 2005. Updated treatment approach
Komorizono, Y., Oketani, M., Sako, K., Yamasaki, to hepatocellular carcinoma. J. Gastroenterol.
N., Shibatou, T., Maeda, M., Kohara, K., 40:225–235.
Shigenobu, S., Ishibashi, K., and Arima, T. Lu, D.S., Yu, N.C., Raman, S.S., Lassman, C.,
2003. Risk factors for local recurrence of small Tong, M.J., Britten, C., Durazo, F., Saab, S.,
hepatocellular carcinoma tumors after a single Han, S., Finn, R., Hiatt, J.R., and Busuttil, R.W.
session, single application of percutaneous radi- 2005. Percutaneous radiofrequency ablation of
ofrequency ablation. Cancer 97:1253–1262. hepatocellular carcinoma as a bridge to liver
Kosari, K., Gomes, M., Hunter, D., Hess, D.J., transplantation. Hepatology 41:1130–1137.
Greeno, E., and Sielaff, T.D. 2002. Local, intra- Machi, J., Uchida, S., Sumida, K., Limm, W.M.,
hepatic, and systemic recurrence patterns after Hundahl, S.A., Oishi, A.J., Furumoto, N.L.,
100 O.F. Bathe and H. Mahallati

and Oishi, R.H. 2001. Ultrasound-guided radi- Puls, R., Stroszczynski, C., Gaffke, G., Hosten, N.,
ofrequency thermal ablation of liver tumors: Felix, R., and Speck, U. 2003. Laser-induced
percutaneous, laparoscopic, and open surgical thermotherapy (LITT) of liver metastases:
approaches. J. Gastrointest. Surg. 5:477–489. MR-guided percutaneous insertion of an MRI-
Mahnken, A., Buecker, A., Spuentrup, E., compatible irrigated microcatheter system using
Krombach, G., Henzler, D., Gunther, R., and a closed high-field unit. J. Magn. Reson. Imaging
Tacke, J. 2004. MR-guided radiofrequency abla- 17:663–670.
tion of hepatic malignancies at 1.5 T: initial Schenck, J.F., Jolesz, F.A., Roemer, P.B., Cline,
results. J. Magn. Reson. Imaging 19:342–348. H.E., Lorensen, W.E., Kikinis, R., Silverman,
Martin, D.R., and Semelka, R.C. 2001. Imaging of S.G., Hardy, C.J., Barber, W.D., and Laskaris,
benign and malignant focal liver lesions. Magn. E.T. 1995. Superconducting open-configuration
Reson. Imaging Clin. N. Am. 9:785–802. MR imaging system for image-guided therapy.
McBeth, P.B., Louw, D.F., Rizun, P.R., and Radiology 195:805–814.
Sutherland, G.R. 2004. Robotics in neurosur- Shibata, T., Iimuro, Y., and Yamamoto, Y. 2002. Small
gery. Am. J. Surg. 188:68S–75S. hepatocellular carcinoma: comparison of radio-
Morikawa, S., Inubushi, T., Kurumi, Y., Naka, S., frequency ablation and percutaneous microwave
Sato, K., Tani, T., Yamamoto, I., and Fujimura, coagulation therapy. Radiology 223:331–337.
M. 2002. MR-guided microwave thermocoagu- Solbiati, L., Livraghi, T., Goldberg, S.N., Ierace,
lation therapy of liver tumors: initial clinical T., Meloni, F., Dellanoce, M., Cova, L., Halpern,
experiences using a 0.5 T open MR system. J. E.F., and Gazelle, G.S. 2001. Percutaneous
Magn. Reson. Imaging 16:576–583. radio-frequency ablation of hepatic metastases
Mulier, S., Ni, Y., Jamart, J., Ruers, T., Marchal, from colorectal cancer: long-term results in 117
G., and Michel, L. 2005. Local recurrence after patients. Radiology 221:159–166.
hepatic radiofrequency coagulation: multivariate Solbiati, L., Ierace, T., Tonolini, M., and Cova, L.
meta-analysis and review of contributing factors. 2004. Guidance and monitoring of radiofrequency
Ann. Surg. 242:158–171. liver tumor ablation with contrast-enhanced
Ni, Y., Chen, F., Mulier, S., Sun, X., Yu, J., ultrasound. Eur. J. Radiol. 51:S19–S23.
Landuyt, W., Marchal, G., and Verbruggen, Steinmeier, R., Fahlbusch, R., Ganslandt, O.,
A. 2006. Magnetic resonance imaging after Nimsky, C., Buchfelder, M., Kaus, M., Heigl,
radiofrequency ablation in a rodent model of T., Lenz, G., Kuth, R., and Huk, W. 1998.
liver tumor: tissue characterization using a Intraoperative magnetic resonance imaging with
novel necrosis-avid contrast agent. Eur. Radiol. the magnetom open scanner: concepts, neurosur-
16:1031–1040. gical indications, and procedures: a preliminary
Noone, T.C., Semelka, R.C., Chaney, and D.M., report. Neurosurgery 43:739–747.
Reinhold, C. 2004. Abdominal imaging studies: Sutherland, G., Kaibara, T., and Louw, D. 2002.
comparison of diagnostic accuracies resulting Intraoperative MR at 1.5 tesla - experience and
from ultrasound, computed tomography, and future directions. Acta Neurochir. 85:21–28.
magnetic resonance imaging in the same indi- Therasse, P., Arbuck, S.G., Eisenhauer, E.A.,
vidual. Magn. Reson. Imaging 22:19–24. Wanders, J., Kaplan, R.S., Rubinstein, L.,
Pawlik, T.M., Scoggins, C.R., Zorzi, D., Abdalla, Verweij, J., vanGlabbeke, M., van Oosterom, A.T.,
E.K., Andres, A., Eng, C., Curley, S.A., Loyer, Christian, M.C., and Gwyther, S.G. 2000. New
E.M., Muratore, A., Mentha, G., Capussotti, L., guidelines to evaluate the response to treatment in
and Vauthey, J.N. 2005. Effect of surgical mar- solid tumors. J. Natl. Cancer Inst. 92:205–216.
gin status on survival and site of recurrence after Truwit, C., and Hall, W. 2001. Intraoperative MR
hepatic resection for colorectal metastases. Ann. systems: high-field approaches. Neuroimaging
Surg. 241:715–724. Clin. N. Am. 11:645–650.
Pearson, A.S., Izzo, F., Fleming, R.Y., Ellis, L.M., Veenendaal, L.M., Rinkes, I.H., Lips, C.J., and van
Delrio, P., Roh, M.S., Granchi, J., and Curley, Hillegersberg, R. 2006. Liver metastases of neu-
S.A. 1999. Intraoperative radiofrequency abla- roendocrine tumours; early reduction of tumour
tion or cryoablation for hepatic malignancies. load to improve life expectancy. World J. Surg.
Am. J. Surg. 178:592–599. Oncol. 4:35.
7. Intraoperative Magnetic Resonance Imaging for Radiofrequency Ablation of Hepatic Tumors 101

Weidensteiner, C., Kerioui, N., Quesson, B., Yu, H., Cheng, J., Lai, K., Lin, C., Lo, G., Lin, C.,
Senneville, B.D., Trillaud, H., and Moonen, C. Hsu, P., Chan, H., Lo, C., Tsai, W., and Chen,
2004. Stability of real-time MR temperature W. 2005. Factors for early tumor recurrence
mapping in healthy and diseased human liver. J. of single small hepatocellular carcinoma after
Magn. Reson. Imaging 19:438–446. percutaneous radiofrequency ablation therapy.
Weinmann, H., Schuhmann-Giampieri, G., Schmitt- World J. Gastroenterol. 11:1439–1444.
Willich, H., Vogler, H., Frenzel, T., and Gries, H. Zhang, Q., Cheung, Y.-C., Lewin, J., and
1991. A new lipophilic gadolinium chelate as a Duerk, J. 1998. A method for simultaneous
tissue-specific contrast medium for MRI. Magn. RF ablation and MRI. J. Magn. Reson. Imaging
Reson. Med. 22:233–237. 8:110–114.
8
Surgically Unresectable and
Chemotherapy-Refractory Metastatic Liver
Carcinoma: Treatment with Yttrium-90
Microsphere Followed by Assessment
with Positron Emission Tomography
Ching-Yee Oliver Wong

INTRODUCTION for palliating unresectable hepatocellu-


lar carcinoma (Dancey et al., 2000; Ho
Detection of metastatic cancer to the liver et al., 1997). Their use in palliation of
with positron emission tomography (PET) unresectable liver metastases from solid
or computed tomography (CT) usually cancers has been shown to be promising
signifies an ominous sign for patients (Wong et al., 2004, 2005). Two commer-
with colorectal cancer (Fernandez et al., cial products of these microspheres are
2004; Arulampalam et al., 2001) and available in glass matrix (20–30 μm, 3.6 g/
other solid cancers (Silvestri et al., 2003; dl, 2,500 Bq/particle, THERA-Sphere®,
Eubank et al., 2002). Surgically unresectable MDS Nordion, Ottawa, Canada) or resin
and chemotherapy refractory metastatic bound (20–60 μm, 1.6 g/dl, 50 Bq/particle,
cancer to the liver often represents an SIR-Spheres®, Sirtex Medical, New South
end-stage event. For these patients, a wide Wales, Australia) forms (Wong et al.,
variety of local and regional therapies 2006). Both the glass or resin microspheres
have been developed from the experience are introduced via catheterization of the
of treating hepatocellular carcinoma for hepatic artery that supplies the majority of
the purpose of providing local control the blood. Tumors and the microspheres
of liver tumors while sparing essential are trapped in the tumor capillary bed
vascular structures, liver parenchyma, where they exert a local radio-therapeutic
and adjacent organs (Rilling and Drooz, effect. Yttrium-90, a pure beta-emitter
2002). Yttrium-90 glass and resin micro- with a physical half-life of 64.2 h, decays
spheres have been shown to be chemically to stable zirconium-90. The average energy
durable for in vivo delivery of β-radiation of the beta emissions from the Yttrium-90
(Drbe and Day, 1993). Recently hepatic is 0.9367 MeV, with an average penetra-
intraarterial infusion of Yttrium-90 glass tion range of 2.5 mm in tissue (Dancey
or resin microspheres has been introduced et al., 2000).

103
104 C.-Y.O. Wong

ASSESSMENT USING A standard uptake value (SUV) has been


POSITRON EMISSION introduced and defined by:
TOMOGRAPHY SUV = Tumor activity/Dose per body
mass
The traditional anatomic imaging by This SUV is often quoted for daily clini-
computed tomography (CT) or magnetic cal quantification of metabolic activity.
resonance imaging (MRI) is insensitive in Assuming negligible free 18F-FDG at the
monitoring response compared to meta- time of imaging, it has been theorized that
bolic imaging (Wong et al., 2002). Prior SUV is related to MRglc (Huang, 2000):
studies (Wong et al., 2002, 2004, 2005) MRglc = G x [Glc] x SUV
addressed the feasibility of using PET
for assessing and quantifying metabolic where G is a product of LC and a subject
response after intraarterial Y-90 micro- independent proportional constant relating
spheres. These studies justify the use of the term of dose per body weight to the
the novel technique by using the objective time integral of total plasma FDG activity
and quantitative property intrinsic to PET (Huang, 2000). Thus, the SUV is a semi-
imaging in evaluating tumor load reduc- quantitative measurement of tumor metab-
tion following Y-90 microsphere treatment olism from PET or PET/CT imaging. It has
(Wong et al., 2004, 2005). no dimensions when both the tumor activity
Positron emission tomography (PET) or and dose per body mass are expressed in
recently combined PET/CT imaging has the same unit as μCi/g. The percentage
been widely used in oncologic evaluation change in total liver SUV from all axial
by taking advantage of diverse capabilities slices of the liver has been shown to cor-
among cell types in metabolizing serum relate well with visual evaluations of meta-
glucose with 2-deoxy-2-[fluorine-18] bolic response (Wong et al., 2004, 2005).
fluoro-D-glucose (18F-FDG) as its analog The reduction of metabolism is related to
tracer. The modeling of glucose metabolic the tumor load reduction and correlates
rate (MRglc) with 18 F-FDG (Huang with reduction in tumor size measured by
et al., 1980; Phelps et al., 1979) follows CT or MRI (Wong et al., 2002, 2007). The
the solid foundation from C-14 deoxyglu- quantitative assessment total liver SUV
cose (Sokoloff et al., 1977): reduction after treatment has also been
shown to correlate with visual assessment
MRglc = {[Glc]/LC}{K1k3/(k2 + k3)} of decreased metabolism by PET in the
where the rate constants, K1 and k2 are tumor lesions, while the background
the forward and reversed transport across metabolism of the liver remains more
the capillary/cellular membrane, k3 is the or less the same (Wong et al., 2004). As
phosphorylation of FDG to FDG-6-P in the reduced tumor metabolism after Y-90
cells and LC is a lumped constant that treatment parallels the reduction of tumor
accounts for the transport and phospho- markers if available (Wong et al., 2004)
rylation difference between FDG and without significant permanent changes
glucose, assuming a small dephosphory- in transaminases reflecting liver injury
lation rate constant (k4) for FDG-6-P. (Wong et al., 2004, 2006), the reduction of
8. Surgically Unresectable and Chemotherapy-Refractory Metastatic Liver Carcinoma 105

metabolism assessed by functional FDG- that cannot be corrected by catheter tech-


PET or PET/CT imaging is related to niques. It is important that liver injection
tumor load reduction but not to the hepato- of macroaggregated albumin (MAA) is
cellular death. delivered with flow rates and catheter posi-
tion similar to the anticipated Y-90 infu-
sion rate and catheter position (Kennedy
Y-90 MICROSPHERE et al., 2007). Patients are also excluded
RADIOEMBOLIZATION from treatment if they are pregnant, had
limited hepatic reserve or irreversibly ele-
The radioisotope, Yttrium-90 (Y-90) is vated bilirubin levels (serum creatinine >
usually attached to some form of micro- 2.0 mg/dl or serum bilirubin > 3.0 mg/dl),
spheres for its brachytherapeutic and/or compromised portal vein (unless selective
radioembolising effects. The resin-bound or superselective radioembolization can
Y-90 microspheres have ~ 40–60 times be performed), prior radiation therapy
more particles than the glass matrix Y-90 involving the liver, hematologic disorders
microsphere per GBq of Y-90 (Wong (absolute granulocyte count < 1,500/ml,
et al., 2006). As there are differences platelet count < 75,000/ml), any con-
in the size and radioactivity per particle traindications to angiography or selective
among different forms of microspheres, visceral catheterization, significant extra-
the metabolic response of Y-90 micro- hepatic disease representing an immi-
spheres may be due to varying degrees of nent life-threatening outcome, pulmonary
the embolic effect of the particle number, insufficiency, active uncontrolled infec-
the local radiation effect that is imparted, tion or any other significant underlying
or both. Irrespective of the underlying medical or psychiatric illness.
tumor killing mechanism, PET will assess Treatment planning and administra-
the presence of viable tumor cells and thus tion of Yttrium-90 microspheres are per-
the final outcome of the treatment. formed generally according to standard
Candidates for radioembolization can published guidelines (Wong et al., 2004,
have either unresectable primary or meta- 2005; Kennedy et al., 2007). Dual phase
static hepatic disease with liver-dominant liver CT is first performed within 30
tumor burden and a life expectancy greater days before the embolization procedure
than 3 months (Kennedy et al., 2007). and 5-mm images are usually reviewed
Other eligibility criteria for Yttrium-90 with special attention to the volume
treatment are age older than 18 years of the liver, degree and distribution
and good (Eastern Cooperative Oncology of tumor infiltration by experienced
Group) performance status (less or equal interventional/body radiologists. Once
to 2) (Wong et al., 2004; Salem et al., a patient is deemed a suitable candidate
2005). Absolute contraindications to Y-90 for Yttrium-90 microsphere treatment, a
microsphere treatment include pretreat- visceral arteriogram with intra-hepatic
ment Tc-MAA scan demonstrating the arterial 99mTc-MAA (macroaggregated
potential of > 30 Gy radiation exposure to albumin) scan will be performed to
the lung or flow to the gastrointestinal tract define the intraarterial distribution of
106 C.-Y.O. Wong

particles and to evaluate the amount of shunting fraction (F) for the given physi-
pulmonary and gastrointestinal flow. ology and anatomy of the hepatic tumors.
All treated patients are then evaluated Prior to treatment, planar anterior 99mTc
to confirm an acceptable risk of shunting MAA scans (Figure 8.1) are obtained
of Yttrium-90 microsphere to the lungs within 30 min after intrahepatic arterial
(< 30 Gy) (Wong et al., 2004, 2005). In injection of 150 MBq 99mTc MAA during
patients with hepatic tumors, a portion of angiography to include the lungs, liver,
the arterial supply may bypass the capil- and stomach (Figure 8.2). Recently, single
lary bed and drain directly into venous photon emission computed tomography
system. The microspheres that are not (SPECT) or combined SPECT/CT is also
trapped in the liver will be shunted via the utilized after planar scintigraphic imag-
heart and deposited in the lungs. Thus, a ing to define more precisely the hepatic
lung shunting measurement is essential distribution of MAA (Figure 8.3), which
before the treatment to obtain a unique mimics the subsequent Y-90 microsphere

Figure 8.1. Planar Tc-99m MAA scan (left panel = anterior view; right panel = posterior view), showing
distribution to the right lobe only (expected from the injection into the right hepatic artery) and hetero-
geneity due to tumor uptake

Figure 8.2. The regions of interest for calculating lung shunting fraction (F)
8. Surgically Unresectable and Chemotherapy-Refractory Metastatic Liver Carcinoma 107

Figure 8.3. Tc-99m MAA SPECT images showing the localization of tracer in the tumor, which mirrors
the subsequent targeted distribution of Y-90 microspheres

Figure 8.4. Spreadsheet for calculating lung shunting fraction (F = 2.4%)

distribution within the normal liver and exposure to lung to be < 30 Gy. The treat-
metastatic tumor lesions. Lung shunting ment dose needs to be further verified by
(F) is assessed using intra-hepatic arterial the experienced physicists according to
99m
Tc MAA images [F = 100% × lung the formulae outlined later. The method
counts/(lung + liver + stomach counts)] of lobar treatment for Y-90 microsphere
(Wong et al., 2004). Lung activity < 10% radioembolization has recently been used
is considered insignificant for any dose by some investigators (Wong et al., 2004,
or activity adjustment (Figure 8.4). The 2005), which differs from the whole liver
lung fraction, even in the setting of F approach reported for hepatocellular
being > 10%, is used to adjust treatment carcinoma (Dancey et al., 2000; Herba
dose or activity to optimize the radiation et al., 1988) and metastatic colorectal
dose given to each lobe of liver or the cancer (Gray et al., 1992) by other inves-
whole liver while maintaining radiation tigators. In the lobar method, each lobe of
108 C.-Y.O. Wong

the liver is treated separately using a lobar Activity (GBq) = (BSA − 0.2) + (Tumor
arterial injection. volume/Total liver volume)
If there is no observable and uncor-
where BSA (m2) = 0.20247[height (m)]0.725
rectable gastrointestinal flow and if liver
[weight (kg)]0.425
reserve and tumor vascular anatomy are
With the calculated activity, the dose
judged to be suitable, a treatment plan
can be deduced from the activity – dose
employing either a lobar or whole liver
formula. Despite the different approach
treatment strategy, is constructed for each
that is used between Y-90 resin-bound
patient. As the treatment is a form of
and glass matrix microspheres, a level of
brachytherapy, the radiation is directly
radioactivity ranging from 0.5 to 2.5 GBq
imposed into the tumor by intraarterial
of Y-90 microspheres is usually pre-
delivery. Unlike conventional radiother-
scribed for left or right lobe or whole liver
apy, only target tumor lesions are identi-
based on the mass (Wong et al., 2004,
fied by CT and PET without the need of
2005; Stubbs et al., 2001). The activity is
drawing gross tumor volumes and their
generally higher for glass microspheres
margin. The targeted liver volume is,
than resin based microspheres due to
however, measured by CT. The amount
different activity uptake fractions to the
of activity required for the target dose of
tumor (TUF) (Wong et al., 2004, 2005).
Yttrium-90 microsphere is calculated (if
Using this technique, normal liver tis-
ignoring lung shunt) using the following
sue exposure will be less than the level
formula (Ho et al., 1997):
above which complications have been
Activity (GBq) = Tumor Dose (Gy) x Tumor Mass (kg) reported using external beam radiother-
50 x Tumor uptake fraction
apy (Campbell et al., 1997).
The prescribed dose of Y-90 will take
The tumor uptake fraction (TUF) depends
the amount of lung shunting into consider-
on the lung shunting fraction and tumor
ation. It has been previously estimated that
vascularity factor. For Y-90 glass micro-
the radiation dose to the lungs assuming a
spheres, TUF is assigned to be 1 while for
uniform microsphere distribution using the
Y-90 resin microspheres, TUF is adjusted
formula (Berger, 1971):
with lung shunting fraction (F) as: TUF = 1
− F. The mass of the targeted lobe was Lung Dose (Gy) = Activity (GBq) x F x 50
Mass of lungs (kg)
determined using CT images for volume
calculation with a conversion factor of
1.03 g/cm3. The target dose level for Y-90 when the total lung mass, including blood,
glass microsphere is set ~100–120 Gy to was assumed to be 1 kg (Snyder et al.,
balance between tumor response rates and 1975–1976), the lung radiation dose is
the risk of normal hepatocellular necrosis simply:
(Kennedy et al., 2007). The required activ- Lung Dose (Gy/kg) = Activity (GBq) x F x 50
ity of Y-90 can be calculated directly from
the above formula. An alternative approach, Patients are considered eligible for
which has been proposed for Y-90 resin Yttrium-90 microsphere treatment if the
microsphere, is to calculate the activity first lung shunt fraction can only result in
based on the body surface area: an estimated dose < 30 Gy to the lungs.
8. Surgically Unresectable and Chemotherapy-Refractory Metastatic Liver Carcinoma 109

To prevent any possible radiation pneu- be scheduled for treatment of the oppo-
monitis, the cumulative radiation dose to site lobe 30–60 days following initial
lungs is also limited to a maximum of treatment. To determine eligibility for
30 Gy. The activity prescribed may also retreatment, patients have repeat labora-
be reduced if the hepatic function is com- tory tests including hepatic function panel
promised by as much as 30% (Kennedy and cross-sectional imaging, similar to the
et al., 2007). All these considerations first treatment. At the time of retreatment
are balanced against the assessment of evaluation, consideration of the total dose
whether the final adjusted radiation dose to the lungs is based on the cumulative
or activity will be enough to deliver to dose over all prior treatments so as not to
treatment response on the target lesions exceed a total estimated cumulative dose
based on the following formula (Ho et al., of 30 Gy to the lungs.
1997; Wong et al., 2004).
Delivered Dose (Gy) =Delivered Activity (GBq) x (1 - F) x 50
Liver Lobe Mass (kg)
CLINICAL RESULTS
where the lung shunt fraction (F) is
obtained as the ratio of counts in the Prior data have suggested that PET is
lungs to the total counts in the abdo- useful in assessing therapy for hepatic
men and lungs, as measured by intra- colorectal metastases (Ho et al., 1997;
hepatic arterial 99mTc-MAA scan. After Selzner and Hany, 2004). Positron
the appropriate activity has drawn into emission tomography has changed the
the delivery device, Yttrium-90 micro- conventional management of patients with
spheres are injected into a percutaneous liver metastases due to its enhanced ability
catheter inserted via the femoral artery that to detect recurrent or metastatic lesions
directed to the targeted liver lobe or the over CT (Meta et al., 2001; Rohren et al.,
entire liver. The administration of Y-90 is 2002). The former has also been known to
done by slow intraarterial infusion at low be more accurate than the latter in detect-
pressure under strict aseptic conditions, ing liver metastases (Wong et al., 2002;
fluoroscopic guidance, institutional radia- Valk et al., 1999; Ogunbiyi et al., 1997).
tion safety guidelines, and the manufac- A prior prospective study on Y-90 glass
turer’s instructions. Treatment is usually microsphere has shown that it is feasible
delivered on an outpatient basis. Patients to quantify reduction of hepatic tumor
are observed in recovery room for 4–6 h metabolism objectively after Y-90 glass
for stability of vital signs, bleeding com- microsphere treatment for unresectable
plications or signs or symptoms of acute metastatic colorectal cancer to the liver
post-embolization syndrome. Patients can (Wong et al., 2004). The total SUV of the
be discharged home when they are clini- entire axial slices of liver agrees well with
cally stable and without complaints. Liver visual evaluation. The PET findings are
function tests are monitored on all patients, also correlated with serum tumor marker,
before and after treatment. suggesting that the reduction of metabo-
Patients with bilobar disease, if not lism is visually, quantitatively, and bio-
yet treated by whole liver approach, can logically related to the tumoricidal effects
110 C.-Y.O. Wong

of Y-90 rather than hepatocellular death metabolism or variable radiosensitivity


(Wong et al., 2004). of tumors, the preferential delivery of Y-90
The same technique of analysis of to the tumors suggests possible similar
PET or PET/CT can be used for the two results in treating these metastatic liver
different Y-90 agents for treating hepatic cancers by either of the two available
metastases from various solid cancers. agents at a comparable safety level (Wong
The resin-based agent contains more par- et al., 2006).
ticles per radioactivity of Y-90 (40–80 Anatomical imaging by CT is insensi-
microsphere than glass microspheres (1.2 tive in assessing tumor response by sim-
million/3 GBq or 50 times on average less ply measuring the change in the product
than resin microsphere), which causes or sum of orthogonal longest diameters
both radiation and embolization (radi- due to the presence of necrosis, edema,
oembolization) for its therapeutic effects hemorrhage, or cystic changes (Wong
(Wong et al., 2006; Kennedy et al., 2007). et al., 2002). Given the global nature of
The radioactivity of Y-90 glass micro- tumor markers in the presence of extra-
sphere is 2,500 Bq per particle, which is hepatic metastatic cancers, PET appears
50 times more than that of resin based to be an excellent adjunct in assessing
microsphere. Thus, more particles will be response following regional treatment of
required if the Y-90 resin microsphere is liver metastases (Wong et al., 2004, 2005;
used to achieve the same treatment dose. Barker et al., 2005). Therefore, the quan-
It is not currently well known whether the titative application of PET or combined
embolic effect will impede the local radia- PET/CT scan is useful for assessing the
tion effect of Y-90 by the potential reduc- response after Y-90 glass or resin-based
tion of oxygen free radicals (Koukourakis, microsphere treatment. Positron emission
2001). However, even though treatment tomography can assess residual hepatic
with Y-90 resin-based microspheres may disease and the status of extra-hepatic
involve the combination of significantly metastases for potential additional salvage
lower radiation and significantly greater chemotherapy, additional Y-90 intra-arte-
embolization when compared to glass rial brachytherapy or other interventional
microspheres, it is evident from a recent radiological procedures (radiofrequency
comparison that the radiation compo- ablation, chemo- or particle-embolization).
nent imparts a greater therapeutic effect Although a few patients may suffer from
than the embolization component and the permanent hyperbilirubinemia, the majority
embolic effect does not appear to impede of the treated patients generally feel better
significantly the local radiation effect after treatment, which parallels the find-
(Wong et al., 2006). ings of PET on the reduction in metastatic
One advantage rather than limitation of load in the liver (Wong et al., 2006).
the Y-90 microsphere treatment is related to The available results appear to be in
the heterogeneous presentation of patients. agreement with the survival bene-
By the nature of metastasis, no single fit imparted to patients with colorectal
patient will be identical. Even though it is metastases described previously by other
virtually impossible to quantify exactly the investigators (Van Hazel et al., 2004).
tumor burden in terms of number, size and Positron emission tomography allows the
8. Surgically Unresectable and Chemotherapy-Refractory Metastatic Liver Carcinoma 111

assessment of response following lobar or ILLUSTRATIONS


whole liver treatment, guiding the deci-
sion whether to proceed with treatment of Sixty-three year-old female had a history
the untreated lobe, retreatment or other of moderately differentiated infiltrating
additional treatments. Moreover, the per- adenocarcinoma of the cecum diagnosed
centage change of total liver SUV by PET 20 months ago. The tumor was removed
reflects the metastatic load reduction by by right hemicolectomy and was
Y-90 microspheres in a quantitative way. found to be 2.2 × 2.0 × 0.3 cm with
In conclusion, there is significant reduc- invasion through serosal surface, lymphatic
tion of hepatic metastatic tumor load involvement and extension into peri-
(metabolism) as evaluated objectively by nodal fat. Her baseline CEA was 1.4 ng/dl.
PET after Y-90 radioembolization for The patient also received adjuvant chemo-
the treatment of unresectable metastatic therapy. Thirteen months after surgery, the
disease to the liver. Y-90 microsphere CEA became 4.2 ng/dl, which gradually
therapy provides encouraging and safe increased to 30.1 and 70.2 ng/dl within
results by arresting progression of meta- the next 3 and 6 months, respectively. At
static cancer to the liver as evident by this point, she was found to have hepatic
decreasing tumor metabolism. While the metastasis, measuring 2.4 × 2.1 and 3.3 ×
cost-effectiveness of PET imaging has 2.9 cm by CT. She was not a surgical can-
been documented in colorectal cancers didate and underwent evaluation for Y-90
(Park et al., 2001), the application to this resin microsphere treatment (Figures 8.1–
type of liver-directed therapy will require 8.5). Post-operative CT did not show much
further investigation. change in the size, measuring respectively

Figure 8.5. Pre-treatment PET-CT scan showing liver metastasis. CT shows one of the two hypodense
lesions in the posterior segment of the right lobe of the liver. PET shows the two foci of high metabolic
activity within the liver metastases
112 C.-Y.O. Wong

Figure 8.6. Post-treatment PET-CT showing response to Y-90 therapy in the liver (with persistent
hypodensity in the posterior segment on CT but resolution of the foci with high metabolic activity seen
on the pre-treatment PET within the liver)

2.3 × 2.3 and 3.9 × 3.4 cm. But PET/CT Dancey, J.E., Shepherd, F.A., Paul, K., Sniderman,
showed drastic improvement in metabolism K.W., Houle, S., Gabrys, J., Hendler, A.L., and
Goin, J.E. 2000. Treatment of nonresectable
(Figure 8.6). She had transient twofold ele-
hepatocellular carcinoma with intrahepatic 90Y-
vation of transaminases. Her CEA decreased microspheres. J. Nucl. Med. 41:1673–1681.
to 5.8 ng/dl 3 months after Y-90 and the liver Drbe, E.M., and Day, D.E. 1993. Chemical durabil-
function tests returned to normal levels. ity of Y2O3-Al2O3-SiO2 glasses for the in vivo
delivery of beta radiation. J. Biomed. Mat. Res.
27:1301–1308.
Eubank, W.B., Mankoff, D.A., Vesselle, H.J., Eary,
REFERENCES
J.F., Schubert, E.K., Dunnwald, L.K., Lindsley,
Arulampalam, T.H.A., Costa, D.C., Loizidou, M., S.K., Gralow, J.R., Austin-Seymour, M.M., Ellis,
Visvikis, D., Ell, P.J., and Taylor, I. 2001. Positron G.K., and Livingston, R.B. 2002. Detection of
emission tomography and colorectal cancer. Br. J. locoregional and distant recurrences in breast can-
Surg. 88:176–189. cer patients by using FDG PET. Radiographics
Barker, D.W., Zagoria, R.J., Morton, K.A., 22:5–17.
Kavanagh, P.V., and Shen, P. 2005. Evaluation Fernandez, F.G., Drebin, J.A., Linehan, D.C.,
of liver metastases after radiofrequency ablation: Dehdashti, F., Siegel, B.A., and Strasberg,
Utility of 18F-FDG PET and PET/CT. Am. J. S.M. 2004. Five-year survival after resection
Roentgenol. 184:1096–1102. of hepatic metastases from colorectal cancer in
Berger, M.J. 1971. Distribution of absorbed dose patients screened by positron emission tomogra-
around point sources of electrons and beta par- phy with F-18 fluorodeoxyglucose (FDG- PET).
ticles in water and other media. J. Nucl. Med. Ann. Surg. 240:438–447.
12(Suppl 5):5–23. NM/MIRD Pamphlet No. 7. Gray, B.N., Anderson, J.E., Burton, M.A., van
Campbell, A.M., Bailey, I.H., and Burton, M.A. Hazel, G., Codde, J., Morgan, C., and Klemp, P.
1997. Tumour dosimetry in human liver follow- 1992. Regression of liver metastases following
ing hepatic Yttrium-90 microsphere therapy. treatment with Yttrium-90 microspheres. Aust.
Eur. J. Nucl. Med. 24:293–298. N. Z. J. Surg. 62:105–110.
8. Surgically Unresectable and Chemotherapy-Refractory Metastatic Liver Carcinoma 113

Herba, M.J., Illescas, F.F., Thirlwell, M.P., Boos, rate in man with (F-18) fluorodeoxyglucose:
G.J., Rosenthall, L., Atri, M., and Bret, P.M. Validation of method. Ann. Neurol. 6:371–388.
1988. Hepatic malignancies: Improved treat- Rilling, W.S., and Drooz, A. 2002. Multidisciplinary
ment with intraarterial Yttrium-90. Radiology management of hepatocellular carcinoma. J.
169:311–314. Vasc. Interv. Radiol. 13(Suppl 9):S259–S263.
Ho, S., Lau, W.Y., Leung, T.W.T., Chan, M., Rohren, E.M., Paulson, E.K., Hagge, R., Wong,
Johnson, P.J., and Li, A.K.C. 1997. Clinical T.Z., Killius, J., Clavien, P.A., and Nelson, R.C.
evaluation of the partition model for estimating 2002. The role of F-18 FDG positron emission
radiation doses from Yttrium-90 microspheres tomography in preoperative assessment of the
in the treatment of hepatic cancer. Eur. J. Nucl. liver in patients being considered for curative
Med. 24:293–298. resection of hepatic metastases from colorectal
Huang, S.C. 2000. Anatomy of SUV. Nucl. Med. cancer. Clin. Nucl. Med. 27:550–555.
Biol. 27:643–646. Salem, R., Lewandowski, R.J., Atassi, B., Gordon,
Huang, S.C., Phelps, M.E., Hoffman, E.J., Sideris, S.C., Gates, V.L., Barakat, O., Sergie, Z., Wong,
K., Selin, C.J., and Kuhl, D.E. 1980. Noninvasive C.Y., and Thurston, K.G. 2005. Treatment of
determination of local cerebral metabolic rate of unresectable hepatocellular carcinoma with use
glucose in man. Am. J. Physiol. 238:E69–E82. of 90Y microspheres (TheraSphere): Safety,
Kennedy, A., Nag, S., Salem, R., Murthy, R., McEwan, tumor response, and survival. J. Vasc. Interv.
A.J., Nutting, C., Benson, A., Espat, J., Bilbao, Radiol. 16:1627–1639.
J.I., Sharma, R.A., Thomas, J.P., and Coldwell, Selzner, M., and Hany, T.F. 2004. Does the novel
D. 2007. Recommendations for radioemboliza- PET/CT imaging modality impact on the treat-
tion of hepatic malignancies using Yttrium-90 ment of patients with metastatic colorectal cancer
microsphere brachytherapy: A consensus panel of the liver? Ann. Surg. 240:1027–1036.
report from the radioembolization brachytherapy Silvestri, G.A., Tanoue, L.T., Margolis, M.L., Barker,
oncology consortium. 1. Int. J. Radiat. Oncol. J., and Detterbeck, F. 2003. The noninvasive
Biol. Phys. 68:13–23. staging of non-small cell lung cancer: The guide-
Koukourakis, M.I. 2001. Tumour angiogenesis lines. Chest 123(Suppl 1):147S–156S.
and response to radiotherapy. Anticancer Res. Snyder, W.S., Ford, M.R., Warner, G.G., and
21:4285–4300. Watson, S.B. 1975–1976. Absorbed dose per unit
Meta, J., Seltzer, M., Schiepers, C., Silverman, D.H., cumulated activity for selected radionuclides and
Ariannejad, M., Gambhir, S.S., Phelps, M.E., organs. MIRD Pamphlet No. 11:S. New York:
Valk, P., and Czernin, J. 2001. Impact of 18F-FDG Society of Nuclear Medicine.
PET on managing patients with colorectal cancer: Sokoloff, L., Reivich, M., Kennedy, C., Des Rosiers,
The referring physician’s perspective. J. Nucl. M.H., Patlak, C.S., Pettigrew, K.D., Sakurada,
Med. 42:586–590. O., and Shinohara, M. 1977. The [C-14] deoxy-
Ogunbiyi, O.A., Flanagan, F.L., Dehdashti, F., glucose method for the measurement of local
Siegel, B.A., Trask, D.D., Birnbaum, E.H., cerebral glucose utilization: Theory, procedure,
Fleshman, J.W., Read, T.E., Philpott, G.W., and and normal values in the conscious and anesthe-
Kodner, I.J. 1997. Detection of recurrent and tized albino rat. J. Neurochem. 28:897–916.
metastatic colorectal cancer: Comparison of Stubbs, R.S., Cannan, R.J., and Mitchell, A.W.
positron emission tomography and computed 2001. Selective internal radiation therapy with
tomography. Ann. Surg. Oncol. 4:613–620. 90-Yttrium microspheres for extensive color-
Park, K.C., Schwimmer, J., Shepherd, J.E., Phelps, ectal liver metastases. J. Gastrointest. Surg.
M.E., Czernin, J.R., Schiepers, C., and Gambhir, 5:294–302.
S.S. 2001. Decision analysis for the cost-effective Valk, P.E., Abella-Columna, E., Haseman, M.K.,
management of recurrent colorectal cancer. Ann. Pounds, T.R., Tesar, R.D., Myers, R.W., Greiss,
Surg. 233:310–319. H.B., and Hofer, G.E. 1999. Whole-body PET
Phelps, M.E., Huang, S.C., Hoffman, E.J., Selin, imaging with F-18 fluorodeoxyglucose in man-
C.E., and Kuhl, D.E. 1979. Tomographic meas- agement of recurrent colorectal cancer. Arch.
urement of regional cerebral glucose metabolic Surg. 134:503–511.
114 C.-Y.O. Wong

Van Hazel, G., Blackwell, A., Anderson, J., Price, Nagle, C., and Salem, R. 2005. Reduction of
D., Moroz, P., Bower, G., Cardaci, G., and Gray, B. metastatic load to liver after intraarterial hepatic
2004. Randomised phase 2 trial of SIR-Spheres Yttrium-90 radioembolization as evaluated by
plus Fluorouracil/Leucovorin chemotherapy ver- [18F]fluorodeoxyglucose positron emission
sus Fluorouracil/Leucovorin chemotherapy alone tomographic imaging. J. Vasc. Interv. Radiol.
in advanced colorectal cancer. J. Surg. Oncol. 16:1101–1106.
88:78–85. Wong, C.Y., Savin, M., Sherpa, K.M., Qing, F.,
Wong, C.Y., Salem, R., Raman, S., Gates, V.L., and Campbell, J., Gates, V.L., Lewandowski, R.J.,
Dworkin, H.J. 2002. Evaluating 1 2 9 0 Y- g l a s s Cheng, V., Thie, J., Fink-Bennett, D., Nagle,
microsphere treatment response of unresectable C., and Salem, R. 2006. Regional yttrium-90
colorectal liver metastases by [18F]FDG PET: A microsphere treatment of surgically unresectable
comparison with CT or MRI. Eur. J. Nucl. Med. and chemotherapy refractory metastatic liver
Mol. Imaging 29:815–820. carcinoma: A quantitative comparison of tumor
Wong, C.Y., Salem, R., Qing, F., Wong, K.T., Barker, response with glass and resin based agents.
D., Gates, V., Lewandowski, R., Hill, E.A., Cancer Biother. Radiopharm. 21:305–313.
Dworkin, H.J., and Nagle, C. 2004. Metabolic Wong, C.Y., Schmidt, J., Bong, J.S., Chundru, S.,
response after intraarterial 90Y-glass micro- Kestin, L., Yan, D., Grills, I., Gaskill, M., Cheng,
sphere treatment for colorectal liver metastases: V., Martinez, A.A., and Fink-Bennett, D. 2007.
Comparison of quantitative and visual analyses Correlating metabolic and anatomic responses
by 18F-FDG PET. J. Nucl. Med. 45:1892–1897. of primary lung cancers to radiotherapy by
Wong, C.Y., Qing, F., Savin, M., Campbell, J., combined F-18 FDG PET-CT imaging. Radiat.
Gates, V.L., Sherpa, K.M., Lewandowski, R.J., Oncol. 2:18.
B. Prognosis
9
Unresectable Liver Metastases from
Colorectal Cancer: Methodology and
Prognosis with Radiofrequency Ablation
Junji Machi

INTRODUCTION not for metastatic tumors. Cryoablation


was extensively investigated in 1980s to
Liver metastases from colorectal cancer, early 1990s. Although less invasive than
although it is a stage 4 disease, has more resection and applicable for unresect-
treatment options and has better prognosis able metastases, cryoablation is associated
than liver metastases from other cancers. with various complications and usually
Multimodality treatment is required for requires laparotomy. Thermal ablation is a
colorectal liver metastases, including local newer modality, which can be achieved by
and systemic treatment. Systemic treatment various methods: microwave, laser, and
is mainly chemotherapy, that is described high-intensity ultrasound are under inves-
in detail in other chapters of this book. tigation, but sufficient clinical data are
Local treatment includes surgical resec- not available. On the other hand, radiofre-
tion, ablation, and hepatic artery infusion quency ablation (RFA) has been increas-
therapy. Surgical resection is the estab- ingly used during the last decade, and has
lished curative local therapy; however, almost replaced cryoablation.
only 10–20% of patients with colorectal Since its introduction, RFA has been
liver metastases have resectable tumors at widely investigated for both primary and
the time of diagnosis. Resection cannot metastatic liver tumors. For metastatic
be performed even in patients with liver tumors, RFA has been used mainly for
only metastases because of various reasons unresectable colorectal cancer metastases,
including multiple bilobar tumors, invasion although neuroendocrine or other meta-
or close proximity to major blood vessels static tumors have also been ablated. This
or bile ducts, limited liver function, or high is based on the assumption that RFA can
surgical risk due to comorbidity. provide a benefit to patients with unresect-
Ablation therapy was introduced initi- able colorectal metastases. A large volume
ally in the hope of achieving local control of literature on RFA for metastatic liver
of unresectable liver tumors; however, tumor is available, and its safety, repeat-
more lately it has been used for curative ability, and local control efficacy have
intent. Chemical ablation such as ethanol been well demonstrated for metastatic liver
injection is effective for hepatomas, but tumors (Berber et al., 2002; Bilchik et al.,

117
118 J. Machi

1999; Bowles et al., 2001; Curley et al., opposite first- and second-line chemo-
1999; Hellman et al., 2002; Iannitti et al., therapy, and to identify prognostic factors
2002; Jiao et al., 1999; Machi et al., 2001; associated with survival. We focused on
Parikh et al., 2002; Siperstein et al., 2000; colorectal cancer metastases, although
Solbiati et al., 1997). However, as described noncolorectal metastases were also treated
by Seidenfeld et al. (2002), many ques- by RFA during the study period. Finally,
tions remain unanswered regarding the I describe the current role and the future
current and future role of RFA in the treat- perspective of RFA.
ment of liver metastases: a comparison of
RFA to alternative methods, efficacy of
combining RFA with other methods such
METHODOLOGY OF
as chemotherapy, comparison of different RADIOFREQUENCY
RFA approaches (percutaneous, laparo- ABLATION
scopic, and open surgical), and long-term
outcome after RFA. To determine the role Most of RFA therapies are performed
of RFA for metastatic liver tumors, long- under ultrasound guidance. Computed
term follow-up data are essential but have tomography-guided RFA is possible, but
rarely been reported until recently. it is much cumbersome. Because of real-
Moreover, while series published to date time feature, ultrasound is a much better
have generally included patients with prog- imaging method to guide and monitor
ressive disease after chemotherapy, no one RFA. The basic ultrasound guidance tech-
has described and compared survival after nique for needle, cannula, or probe place-
RFA given prior to the first chemotherapy ment for RFA is the same as other needle
regimen for metastatic disease (i.e., “first- guidance under ultrasound.
line”), and survival after RFA when given Radiofrequency ablation treatment of
following disease progression subsequent abdominal organs such as tumors of the
to chemotherapy (i.e., “second-line”). liver can be performed percutaneously,
Failure to describe the impact of RFA in laparoscopically, or open surgically (Machi
relation to chemotherapy in distinct stages et al., 2001). Advantages and disadvan-
of disease progression has hampered the tages of each approach are as follows:
ability to estimate incremental survival 1. Percutaneous Advantages: least invasive,
benefit gained by RFA over the expected possible out-patient procedure, needle
survival on chemotherapy alone. guidance system (+), possible local
We have prospectively acquired data of anesthesia
patients undergoing RFA for unresectable
Disadvantages: less accurate cancer
metastatic liver tumors since 1997. In this
staging, some areas not accessible, risk of
chapter, first I describe the methodology
thermal injury to adjacent organs (bowel,
of RFA. Next, I describe our study, a part
diaphragm, gallbladder, etc.)
of which was recently published (Machi
et al., 2006). The purpose of this study 2. Laparoscopic Advantages: less invasive,
is to assess the long-term outcome of better cancer staging with laparoscopic
the patients based on a specific grade of ultrasound, short recovery, possible con-
unresectability and in relation to its timing current colorectal surgery
9. Unresectable Liver Metastases from Colorectal Cancer 119

Disadvantages: technically demanding, with multiple retractable curved electrode-


lack of needle guidance system needles (arrays), while Radionics uses a
3. Open Surgical Advantages: better cancer cool-tip single needle or a triple needle
staging with intraoperative ultrasound, cluster. The size of cannulas or needles
needle guidance system (+), inflow ranges from 18-gauge to 14-gauge.
occlusion possible, possible concurrent Under ultrasound guidance, cannulas
colorectal surgery or needles are inserted into the liver and
Disadvantages: most invasive, longer then into the tumor. It is important to
recovery visualize the tips of needles and to under-
Interventional radiologists can perform stand their locations before ablation starts
percutaneous RFA, while surgeons can in order to obtain an adequate margin of
use either approach. Different approaches ablation. Depending on the size of the
should be used appropriately depending tumor and the achievable size of abla-
on the condition of patients and tumors tion, a single ablation or multiple over-
to be ablated (also see the next section). lapping ablation sessions are performed.
Percutaneous ultrasound-guided RFA is The ablation process can be monitored
usually performed using a needle guid- by ultrasound imaging; the ablated lesion
ance system because the distance to the becomes hyperechoic because of outgas-
tumor is usually longer. Laparoscopic sing from heated tissues. However, the
ultrasound-guided RFA needs to be per- hyperechoic area does not always cor-
formed by a freehand technique because respond exactly to the ablated lesion.
of lack of a needle guidance system with In addition, the hyperechoic area will
laparoscopic ultrasound transducer. Open obscure the ultrasound images around
surgical RFA can be performed either by the tumor. Therefore, it is critical to
a freehand technique or using a needle make a good plan for multiple ablation
guidance system depending on the size, sessions before the first ablation starts.
depth, and location of tumors. Once open When the cannula is withdrawn at the
surgical approach is used, any tumors in completion of ablation, the cannula track
the liver can be ablated except for tumors is also ablated under ultrasound visuali-
abutting or in close proximity to the main zation to prevent bleeding and possible
right or left hepatic ducts. Ablation of tumor seeding.
these ducts causes a bile duct stricture or If color or power Doppler imaging dem-
obstruction, which is a major complica- onstrates intratumoral blood flow prior to
tion of RFA. Hand-assisted laparoscopic ablation, Doppler imaging can be repeated
ultrasound-guided RFA can be performed, after ablation to confirm loss of blood
taking advantages of both laparoscopic flow within the tumor. Contrast-enhanced
and open surgical approaches (Machi Doppler imaging is useful for this pur-
et al., 2002). pose, but intravenous ultrasound contract
In the United States, there are several agents are commercially not available yet
companies manufacturing RFA devices. in the United States. Preoperative tests or
RITA Medical System (AngioDynamics, preparation, postoperative follow-up, and
Inc.) and Radio Therapeutics (Boston complications of RFA are described in the
Scientific, Inc.) use electrode-cannulas following sections.
120 J. Machi

PROGNOSIS USING assess the survival of these patients,


RADIOFREQUENCY “unresectability” was classified into three
ABLATION: OUR STUDY grades, on the basis of oncological (cura-
tive or not), technical (tumor size, number,
Patients and Methods and location), and medical (surgical risk:
age, comorbidity, and liver dysfunction)
Patients undergoing RFA for unresect- factors. Three groups of patients were
able metastatic liver tumors from August defined:
1997 to July 2003 were followed. Data
were prospectively acquired, and were Grade 1: Relatively unresectable (not opti-
retrospectively analyzed. Metastases were mally resectable)
confirmed histologically by biopsy in all Patients with liver only metastases that
patients prior to RFA treatment. During were not optimally resectable or considered
this period of study, patients who had not resectable by the majority of surgeons
resectable metastatic tumors underwent due to oncological or medical reasons
surgical resection, while patients who were were included in this group although they
deemed to have “unresectable” tumors might have been technically resected by
were treated by RFA. aggressive approaches but with high risks.
Patients who were judged to be una- Most patients in this group had unfavorable
ble to tolerate RFA because of serious risk factors for recurrence: e.g., large size
comorbidity or because of limited liver tumors (> 5 cm), large number of tumors
function (e.g., underlying liver disease (> 4), bilobar tumors, high carcinoem-
such as cirrhosis, following prior hepatic bryonic antigen (CEA), short disease-free
resection) were excluded. Patients with interval from primary to metastases, node-
extensive extrahepatic metastases such as positive primary tumor, and/or moder-
advanced peritoneal carcinomatosis and ate to poorly differentiated tumor grade:
diffuse lung metastases who would not often two or more out of five clinical risk
benefit from RFA of liver metastases were scores deve-loped by Fong et al. (1999).
also excluded. On the other hand, patients Some patients in this group had high (but
who were considered likely to benefit from not prohibitive) surgical risks to perform
RFA for possible cure, possible prolon- resection, including certain comorbidity
gation of life, or symptomatic palliation (e.g., cardiovascular, respiratory, or renal
underwent RFA. Tumor number, tumor dysfunction) or limited liver function.
size, and invasion or proximity to major Grade 2: Absolutely unresectable
blood vessels were not considered for a Patients with liver only metastases that
contraindication to RFA. were absolutely unresectable due to tech-
nical, medical, or oncological reasons were
Unresectability of Tumors
included in this group. These patients freq-
As compared to patients with optimally uently had tumors invading major vascular
resectable liver metastases who underwent structures (such as main right and left
hepatic resection, patients with “unresect- portal veins, two or more major hepatic
able” metastases undergoing RFA included veins, or vena cava) and/or right and
a heterogeneous population. In order to left hepatic ducts or their confluence.
9. Unresectable Liver Metastases from Colorectal Cancer 121

Also included in this group were patients of metastatic tumors, visualization or acces-
who were judged not to be able to tolerate sibility on transabdominal ultrasound, need
resection. For example, some patients had for concurrent operations (i.e., synchronous
too many tumors and/or tumors too large colorectal or other organ resection), need
to resect by anatomical major hepatectomy for precise staging by laparoscopy or open
and/or by multiple metastasectomy, unless surgery, newly diagnosed tumors versus
two-stage hepatectomies combined with recurrent tumors following previous resec-
portal vein embolization were performed tion or ablation, and patient condition and
(Jaeck et al., 2004). Other patients had too liver function with regard to surgical and
high (prohibitive) surgical risks such as anesthesia risks (Bowles et al., 2001; Machi
significant comorbidity or liver dysfunction et al., 2000, 2001, 2002, 2006). The selec-
to undergo resection. Many of the patients tion of RFA approach was individualized,
in this group also had multiple risk factors and informed consent was obtained from
for recurrence as patients in grade 1. each patient after full explanation of ben-
Grade 3: Extrahepatic metastasis efits and risks of approaches.
Patients who had extrahepatic metastases During RFA operation, final number,
in addition to liver metastases were included size, and location of the tumors, vascular or
in this group. Extrahepatic metastases bile duct invasion were determined. When
(usually limited lung or peritoneal metas- laparoscopic or open surgical RFA was
tases) in these patients were not extensive performed, high-frequency (5–10 MHz)
as compared to liver metastases so that RFA laparoscopic or open intraoperative ultra-
would be considered to possibly provide sound was routinely performed to screen
benefits to patients. the entire liver for occult tumors. All occult
tumors were ablated when detected.
Preoperative and Intraoperative
Many patients had multiple tumors of
Evaluation
different sizes. In order to estimate the total
Patients had preRFA laboratory tests includ- (cumulative) tumor volume (sum of the
ing liver function tests and CEA. Patients volumes of all tumors to be ablated in a
had computed tomography (or magnetic given RFA session), “total tumor volume”
resonance imaging) and transabdominal was approximated by “total tumor size
ultrasound preoperatively. Positron emis- (sum diameter of tumors)”, which was
sion tomography (PET) was introduced calculated by adding the largest dimension
in October 1999; it was selectively used (in millimeter) of each tumor for all tumors
mainly to diagnose or exclude extrahepatic ablated in each treatment session.
metastases. The timing of offering RFA to
each patient in relation to chemotherapy Radiofrequency Ablation Methods
for metastases was individualized, and was The majority of patients had general anes-
determined by medical oncologists and thesia, although several patients underwent
surgeons together. RFA under local anesthesia. Radiofrequency
Radiofrequency ablation approaches ablation was performed under ultrasound
(percutaneous, laparoscopic, or open sur- guidance using a RFA system (RITA
gical) were determined by considering Medical System, Mountain View, CA) as
factors including number, size, and location described in the previous section. Initially, a
122 J. Machi

15-gauge, 3 cm ablation cannula was used. confirmed by biopsy prior to subsequent


From December 1999, a 14-gauge, 5 cm treatment including repeated RFA. When
ablation cannula was mainly used. In July local recurrence or new liver recurrence
2001, a new 7 cm ablation cannula with occurred, repeated RFA was considered and
saline infusion system was introduced and performed when indicated. When patients
used to ablate tumors 5 cm or larger. The with recurrent tumors were not candidates
average target temperature at the needle for repeated RFA (e.g., extensive recur-
tips was set at 100–110°C. The ablation was rence or extrahepatic recurrence), systemic
continued to achieve an ablation margin of or regional chemotherapy was considered
1 cm or more (Bowles et al., 2001; Machi and performed as indicated.
et al., 2000, 2001, 2002, 2006).

Postoperative Follow-Up Statistical Analyses


Patients were followed for at least 18 Statistical analyses were performed using
months or until death. Computed tomog- SAS®, version-8.2 for windows (SAS
raphy (or magnetic resonance imaging) Institute, Inc., Cary, North Carolina, statis-
was obtained 1 week postoperatively tical software). Continuous variables were
to determine the adequacy of RFA, and expressed as mean ± standard error (SE) or
then at 3-month intervals for 1 year, standard deviation (SD). The level of sig-
and 3–6-months intervals thereafter. nificance was set at p ≤ 0.05 for all tests.
Carcinoembryonic antigen, when elevated Survival was calculated using the Kaplan-
preoperatively, was measured a few weeks Meier method, and the median survival
postoperatively and then at 2–4-month between groups was first compared using
intervals. Whenever CEA increased during the log-rank test. Any factors that showed
follow-up, further studies were carried out a possible association with survival at p
using computed tomography routinely and ≤ 0.15 in the univariate Kaplan-Meier
PET selectively. analysis were then taken forward in a mul-
Postoperatively, systemic chemotherapy tivariate Cox proportional hazards method.
was initiated or resumed as indicated. The The results from this analysis included
nature of chemotherapy regimens received the hazard ratio and the corresponding
was variable, as irinotecan and oxaliplatin- 95% confidence interval (the detail is not
based regimens became available during reported in this chapter).
the study follow-up period. Hepatic artery Risk factors, which were prospectively
regional chemotherapy was used only in a thought to affect long-term survival out-
few selected patients. come, were analyzed. These included
Local recurrence at the site of RFA was patient characteristics (age, gender),
defined as increase in size of the ablated liver tumor factors (synchronous versus
lesion with change in the CT contrast- metachronous, CEA, number of tumors,
enhanced appearance and/or positive PET total tumor size), unresectability grade
activity at the site of RFA treatment. When (including extrahepatic metastasis), RFA
new tumors appeared away from the site of approaches, previous and subsequent treat-
RFA, they were judged to be new liver (int- ment, repeated RFA, complications, and
rahepatic) recurrences. Recurrences were recurrence patterns.
9. Unresectable Liver Metastases from Colorectal Cancer 123

Short-Term Results recurrence, using various agents, while


During the study period, 131 patients other therapies were also combined as
with a total of 653 metastatic liver tumors indicated: repeated RFA in 36 patients
underwent 187 RFA procedures. Of these, (for recurrence), lung metastasectomy in
100 patients with colon (n = 66) or rectal 6 patients, and hepatic infusion chemo-
(n = 34) cancers, 57 men and 43 women, therapy in 4 patients.
underwent 146 RFA procedures to ablate There were no surgical complications in
507 metastatic tumors. Age at the time 121 of 146 RFA procedures (82.9%). Eight
of the first RFA was 67.7 ± 10.1 (mean ± major complications occurred in seven pro-
SD), ranging from 46 to 88 years. Clinical cedures (4.8%): liver failure (n = 1), biliary
characteristics of tumors ablated in 146 stricture (n = 3), bile leakage (n = 2), liver
procedures of 100 patients were: abscess (n = 2). Nineteen minor complica-
tions occurred in 18 procedures (12.3%):
Number of tumors in each operation: ascites/pleural effusion (n = 4), wound
1–16, mean ± SD: 3.5 ± 2.8. infection (n = 3), myoglobinuria (n = 6),
Size of tumors: 3–174 mm, mean ± SD: thrombocytopenia (n = 1), pneumonia
30 ± 16 mm. (n = 2), and heart failure/arrhythmia (n =
Total tumor size: 11–418 mm, mean ± 3). One patient with limited liver function
SD: 96.4 ± 80.7 mm. (Child-Pugh Class B) from cirrhosis who
Unresectability: Grade 1: 40 (27.4%), had multiple metastases from a colon can-
Grade 2: 77 (52.7%), Grade 3: 29 cer had progression of liver dysfunction and
(19.9%). died of liver failure 2 months after RFA.
RFA approach: Percutaneous: 61 Local tumor recurrence (at the site of RFA)
(41.8%), Laparoscopic: 23 (15.8%), was diagnosed in 34 of 507 tumors (6.7%)
Open: 62 (42.4%). following 30 of 146 procedures (20.5%).
Eight patients had previous hepatic The interval between RFA and local tumor
resection for liver metastases. Fifty-five recurrence ranged from 1 to 23 months and
patients (55%) received RFA prior to ini- averaged 8.7 ± 0.9 months (mean ± SE).
tiation of therapeutic chemotherapy for New liver (intrahepatic) recurrence and/or
metastatic disease (first-line RFA); the extrahepatic recurrence (lung, bone, brain,
other 45 patients (45%) received RFA peritoneum/retroperitoneum, lymph node)
for residual or progressive disease after occurred following 87 of 100 first RFA
therapeutic chemotherapy for metastatic procedures (87.0%) and following 123 of
disease (second-line RFA). As this trial 146 procedures including repeated RFA
accrued patients largely prior to availabil- (84.2%). The interval between RFA and
ity of oxaliplatin, which became available new recurrence ranged from 1 to 40 months
during the follow-up period, initial chemo- and averaged 10.4 ± 0.6 months (mean ±
therapy consisted mostly of fluorouracil SE). Recurrence in the RFA cannula track
plus leucovorin and/or irinotecan. (“needle track seeding”) was not noted.
Eighty-five of 100 patients had subse-
quent treatment after RFA. Eighty-one Long-Term Results
had systemic chemotherapy as adjuvant The median and mean follow-up periods of
therapy of RFA and/or as treatment for the 100 patients were 24.5 months and 29.1
124 J. Machi

± 17.5 months (mean ± SD), respectively, Subsequent treatment affected recurrence-free


ranging from 2 to 84 months. At the time survival but not overall survival. The lower
of data analyses in 2005, 41 patients unresectability grade demonstrated better
(41.0%) were alive and 59 patients (59%) overall and recurrence-free survival. Extra-
were dead. Twenty-one of 41 alive patients hepatic metastasis (grade 3) was associated
were recurrence-free while 20 patients had with worst survival. Fifty five patients who
recurrent tumors. Causes of death were underwent RFA as a first-line treatment
liver tumors in 28 patients (47.5%), extra- prior to chemotherapy for metastatic dis-
hepatic metastases in 29 patients (49.2%), ease showed a better median survival of 48
and non-cancer in 2 patients (3.4%). months compared to a median survival of
The overall median and mean survival of 22 months among 45 patients who under-
the 100 patients was 28 months and 36.1 went RFA as second-line or salvage treat-
± 2.4 months (mean ± SE), and 1-, 3- and ment after chemotherapy.
5-year survival was 90.0%, 42.0% and Laparoscopic approach provided the best
30.5%, respectively. The median and mean survival, open surgical approach provided
of the 25 patients in the unresectability intermediate survival, and percutaneous
grade 1 group were 48 months, 50.9 ± 3.8 approach provided the worst survival. In
months, and the 1-, 3- and 5-year survival order to further assess differences among
were 100%, 83.6% and 49.4%, respectively. RFA approaches, tumor characteristics
The recurrence-free median and mean sur- stratified by approaches were evaluated and
vival of the 100 patients was 13 months and analyzed. Tumors treated by the laparo-
23.5 ± 2.2 months (mean ± SE), and 1-, scopic approach were smaller in size and
3- and 5-year recurrence-free survival was number and lower in unresectability grade,
53.0%, 23.2%, and 21.7%, respectively. whereas tumors treated by the percutaneous
Prognostic factors that were hypoth- approach were larger in size and higher in
esized to affect overall and recurrence-free unresectability grade. Such tumor character-
survival of 100 patients were analyzed first istics by RFA approach affected survival.
by univariate analysis. Carcinoembryonic In the multivariate analysis, only age (70
antigen level (200 ng/ml), total tumor size years) and unresectability grade including
(100 mm), RFA approach, previous thera- extrahepatic metastasis showed significant
peutic chemotherapy (not adjuvant but association with overall and recurrence-
therapy for metastases, namely first- versus free survival (p < 0.05). The median
second-line chemotherapy), local recur- survival of patients without extrahepatic
rence and new recurrence were signifi- metastasis was significantly better than
cantly associated with both overall and that of patients with extrahepatic metasta-
recurrence-free survival (p < 0.05), while sis (30 vs., 20 months).
age, gender, synchronous versus meta-
chronous, number of tumors, and repeated
RFA were not. Unresectability grade and CURRENT ROLE OF RADIO-
complications were significantly associated FREQUENCY ABLATION
with recurrence-free survival (p < 0.05) and
appeared to be associated with overall Hepatic resection is the most effective
survival approaching statistical significance established method for cure of colorectal
(p = 0.0576 and p = 0.0540, respectively). metastases to the liver. The 5-year overall
9. Unresectable Liver Metastases from Colorectal Cancer 125

survival rate after resection has been relative safety, less invasive nature and
reported in a range of 25–40% (Fong et al., patient willingness to undergo repeated
1999; Nordlinger et al., 1996; Registry procedures, as compared to liver resec-
of hepatic metastases, 1988; Scheele et al., tion. Approximately, 30% of treatments in
1995): the median 5-year survival of 19 our study were repeated RFA for recurrent
study series is 30% (Fernandez et al., metastatic tumors. Local tumor recurrence
2004). Recently, even higher 5-year sur- at the site of RFA occurred in 6.7% of
vival rates of 58% were achieved in three tumors, which is similar or superior to
studies (Abdalla et al., 2004; Choti et al., other studies (~ 5–15%). The diagnosis of
2002; Fernandez et al., 2004). This excel- local recurrence was made mostly within
lent outcome was attributed to improve- 1 year after RFA (mean 8.7 months). With
ment in patient selection (e.g., use of longer follow-up, new tumor recurrence
PET), perioperative care, multidisciplinary occurred much more frequently (higher
treatment including newer chemotherapy, than 80%) in other areas of the liver or
and aggressive surgical management. extrahepatic organs. The diagnosis of new
However, resection can be performed only tumor recurrence was made slightly later
in 10–20% of patients with colorectal (mean 10.4 months) than local tumor
metastases. Even with aggressive surgical recurrence, even as late as 40 months after
approaches such as a two-stage hepate- RFA.
ctomy (Jaeck et al., 2004), resectability In order to evaluate the effect of RFA as a
rates would still be ~ 30%. potentially curative treatment, a long-term
For unresectable metastatic liver tumors, follow-up of patients after RFA is essen-
ablation therapies including RFA were tial. However, the long-term outcome for
initially used for local control, but more liver metastases treated by RFA has rarely
recently its use was also aimed for curative been documented until recently. Most of
purpose. A number of reports documented the data on long-term survival have been
and confirmed the safety and local control reported by investigators in Europe, par-
ability of RFA for metastatic liver tumors ticularly Italy, where RFA was introduced
(Berber et al., 2002; Bilchik et al., 1999; in the early to mid-1990s. Initial survival
Bowles et al., 2001; Curley et al., 1999; outcomes for colorectal metastases were
Hellman et al., 2002; Iannitti et al., 2002; reported by Solbiati et al. (1997) and
Jiao et al., 1999; Machi et al., 2001; by Lencioni et al. (1998). Solbiati et al.
Parikh et al., 2002; Siperstein et al., (2003) updated their data, and Lencioni et
2000; Solbiati et al., 1997). Our study (as al. (2004) also updated data, which were
described above) demonstrated the safety, included in a long-term multicenter study
repeatability and local control efficacy of of 423 patients. Table 9.1 summarizes
RFA consistent with other studies. Major studies showing long-term prognosis of
complications occurred in fewer than 10% RFA. In these Italian and other European
of RFA procedures and were similar to studies with long-term follow-up (3–5-
those noted by others including Curley year survival), RFA was performed by the
et al. (2004) who recently reported early percutaneous approach by radiologists (De
complications in 7.1% and late complica- Baere et al., 2003; De Meijer et al., 2006;
tions in 2.4% of 608 patients. The repeat- Gillams and Lees, 2004; Lencioni et al.,
ability is one advantage of RFA due to its 2004; Solbiati et al., 2003).
126 J. Machi

Table 9.1. Literature review: long-term prognosis of radiofrequency ablation for colorectal cancer liver metastases.
Overall survival
Number of RFAa Follow-up 1-year 4-year 5-year
Author Year patients approach (mean) Median (%) 3-year (%) (%) (%)
(Italy/Europe)
Solbiati et al. 2003 166 P 41.6 Mo 33 Mo 96 46 37 22
Gillams and Lees 2003 167 P 22 Mo 71 21 – 14
De Baere et al. 2003 155 P 18 Mo 27 Mo 82 31
Oshowo et al. 2003 25 P 37 Mo 25 Mo 37 22
Lencioni et al. 2004 423 P 19 Mo 86 47 29 24
White et al. 2004 30 P 17 Mo 22 Mo 75 45
(United States)
Siperstein et al. 2000d 110 P, L, O 30 Mo 88 49 – –
Abdalla et al. 2004 57 O 21 Mob 37 22 –
Abdalla et al. 2004 101c O 21 Mob 43 36 –
Berber et al. 2005 135 L 28.9 Mo
Machi et al. 2006 100 P, L, O 29 Mo 28 Mo 90 42 31 31
(our study) 24.5 Mob
a
P = Percutaneous, L = Laparoscopic, O = Open surgical approach.
b
Median follow-up.
c
Patients underwent RFA combined with liver resection.
d
Study presented but not published.

In the United States, RFA was intro- 24.5-month median and 29-month mean
duced around 1996–1997. The first long- follow-up, which is the longest period ever
term follow-up outcome (3-year survival) reported in the United States (Machi et al.,
of a United States multicenter study was 2006). We used percutaneous, laparoscopic
presented by Siperstein et al. (2000) at and open surgical approaches to optimize
the congress of the American College of the potential benefit to each patient, reco-
Surgeons in October 2002; however, the gnizing the limitation of the percutaneous
data were not published. Curley et al. approach alone (Bowles et al., 2001; Machi
(1999), working with Italian investigators, et al., 2000, 2001, 2002, 2006).
have been demonstrating excellent results The long-term follow-up in our study has
of RFA for unresectable primary and met- demonstrated that RFA can contribute to
astatic hepatic malignancies, and their encouraging survival in patients with unre-
most recent report by Abdalla et al. (2004) sectable colorectal cancer liver metastases.
described long-term outcomes of open The survival outcome was similar or supe-
surgical RFA and combined RFA plus rior to those of Italian/European studies
resection (4-year survival) as compared to and similar to that reported by Berber et al.
resection for colorectal metastases (Table (2005) (Table 9.1). This may be due to
9.1). Berber et al. (2005), using exclusively our aggressive performance of RFA using
a laparoscopic RFA approach, reported laparoscopic and open approaches when
a median survival of 28.9 months in 135 the percutaneous approach is considered
patients with colorectal metastases. In our suboptimal. Additionally, the inclusion of
study (as described in the previous section), the unresectability grade 1 patients, who
we have reported follow-up results of 100 might have otherwise undergone hepatic
patients undergoing RFA for colorectal resection although not optimally, may have
metastatic tumors, up to 84 months, with skewed our survival rates. However, the
9. Unresectable Liver Metastases from Colorectal Cancer 127

overall 5-year survival rate of 30.5% was Radiofrequency ablation following pre-
comparable to those historically achieved vious chemotherapy for metastatic dis-
by hepatic resection (Fong et al., 1999; ease was associated with worse survival
Nordlinger et al., 1996; Registry of hepatic likely because patients who had pre-RFA
metastases, 1988; Scheele et al., 1995), treatment generally had more extensive
although lower than the 58% achieved in tumors, chemotherapy-refractory tumors
recent three reports of resection (Abdalla et or extrahepatic metastasis. However, the
al., 2004; Choti et al., 2002; Fernandez et median survival of 48 months among
al., 2004). patients receiving RFA prior to first-line
Prognostic factors such as the clinical chemotherapy compares favorably with
risk score for recurrence after hepatic resec- survival reported from various trials of
tion have been identified and are useful for first-line chemotherapy alone, which have
predicting post-resection survival outcome reported survival in the range of 19–22
in patients with colorectal metastatic liver months (Grothey et al., 2004; Hurwitz
tumors (Fong et al., 1999; Nordlinger et al., 2004). Similarly, the median survival
et al., 1996). In our study, we have iden- of 22 months in this study for RFA when
tified factors predictive of survival after given in the second-line setting compares
RFA for unresectable metastatic tumors. favorably with survival of 11–15 months
Among the factors not associated with reported from recent trials of second-line
overall and recurrent-free survivals was chemotherapy alone (Giantonio et al.,
the presence or absence of repeated RFA. 2005; Pitot et al., 2005; Rougier et al.,
Repeated RFA did not result in worse sur- 2002). In order to preserve the opportu-
vival, suggesting that RFA should be con- nity to measure secondary end points of
sidered for recurrent metastatic tumors. tumor response and response duration,
On the other hand, the factors associated many chemotherapy trial protocols pro-
with survival were CEA, total tumor size, hibit patients from receiving a potentially
unresectability grade including extrahe- curative resection or ablation after the
patic metastasis, RFA approach, previous start of chemotherapy, even though initial
therapeutic chemotherapy, complications, response to such therapy may move many
local tumor recurrence and new recur- of these initially unresectable tumors to a
rence. Many of these factors are related potentially resectable or ablatable status.
to one another. Complications and local Berber et al. (2005) have reported pre-
tumor recurrence are more frequent when dictors of survival after RFA: significant
total tumor size (volume) to be ablated is factors associated with overall survival
larger (Bowles et al., 2001; Machi et al., were CEA and tumor size, which are simi-
2001; Siperstein et al., 2000). Differences lar to our study. However, the presence of
in survival outcome among three RFA limited extrahepatic disease did not appear
approaches in our study are likely due to affect survival significantly in their study.
to selection of patients and tumors (e.g., This is in contradiction to patients with
unresectability grade) to be ablated: the extrahepatic metastasis in our study, prob-
laparoscopic approach was superior to ably due to differences in inclusion crite-
other approaches because patients in lower ria of patients for RFA depending on the
unresectability grade were selected. extent of extrahepatic metastases.
128 J. Machi

In our study, we have attempted to classify cancer recurrence. The precise monitoring
patients with unresectable liver metastatic of the size or extent of ablation intraopera-
tumors into groups in the hope of pre- tively by ultrasound is presently not pos-
dicting prognosis or outcome after RFA sible, and local cancer recurrence is higher
treatment. As compared to resectable meta- than resection. New ultrasound or other
static tumors for which easily applicable intraoperative technology such as 3-D
and valuable patient stratification methods (three-dimensional) ultrasound or contrast
such as the clinical risk score are avail- enhancement may improve the accuracy
able (Fong et al., 1999; Nordlinger et al., of monitoring RFA with improvement of
1996), unresectable liver tumors encom- ablation negative margin.
pass a wider variety of patient and tumor Hepatic resection is considered as the
characteristics, and thus stratification of gold standard for resectable colorectal
such heterogeneous patients by a straight- metastatic liver tumors. On the other hand,
forward scale is difficult. Unresectability the true role of RFA has not been determined.
is generally determined based on oncolo- Our study has demonstrated that RFA and
gical, technical and medical factors and, in repeated RFA can contribute to a relatively
a certain extent, the unresectability criteria good survival for patients with unresectable
differ among surgeons. Radiofrequency metastatic tumors, and has characterized
ablation can be used for the purpose survival following RFA in relation to first- and
of cure, palliation or life prolongation. second-line chemotherapy (Machi et al.,
Taking these factors and RFA purposes 2006). However, new liver or extrahepatic
into account, we have presented an “unre- recurrence is common after RFA; new liver
sectability grade” by defining three groups: tumors probably recur more frequently
grade 1 (relatively unresectable or not after RFA than after hepatic resection, as
optimally resectable), grade 2 (absolutely noted by Abdalla et al. (2004), probably
unresectable), and grade 3 (extrahepatic because larger liver parenchyma fields,
metastasis). This unresectability grade sys- which are at risk for recurrence remain
tem has demonstrated significant survival after RFA compared to resection.
differences among patients with colorec- To prevent and control such recurrences
tal metastatic liver tumors after RFA in and to achieve a long survival, multimo-
this study, and therefore may be a useful dality therapies, including locoregional
predictor in future studies. and systemic methods, are definitively
required. Because there is a limitation in
hepatic resection as a locoregional treat-
FUTURE PERSPECTIVE ment method, RFA is an additionally
valuable modality, particularly for unresect-
The current methodology of RFA is able metastatic tumors. Our study sug-
advancing, and a 5 cm ablation and even gests greater survival benefit when RFA
a 7 cm ablation can be performed. It is is used in the first-line setting than when
anticipated that further advances of RFA used in the second-line or salvage setting.
technology will improve the ablation size Further investigation is needed to evaluate
larger and the speed of ablation faster the role and timing of RFA in combination
with lower complications and lower local with systemic treatment, especially with
9. Unresectable Liver Metastases from Colorectal Cancer 129

the modern chemo-biological regimens. In conclusion, the safety, repeatability


The relatively longer survival provided by and local control efficacy of RFA have
RFA with chemotherapy in our study may been well documented. Furthermore, more
also suggest the value of prechemotherapy data have been lately reported, show-
cytoreduction by means of RFA for patients ing the long-term survival benefits of
with unresectable colorectal cancer. RFA. Radiofrequency ablation technology
We think that currently RFA cannot and methodology will continue evolving
completely replace hepatic resection, par- with provision of better ablation outcome.
ticularly for large tumors (e.g., > 7–8 cm) However, the exact role of RFA in the treat-
because of high local recurrence after RFA ment of colorectal cancer liver metastases
of large tumors. Radiofrequency ablation needs to be defined with future investiga-
can be combined with hepatic resection tions. Multi-disciplinary approach by more
for selected patients with multiple bilobar appropriate local therapy by resection and
metastases (Abdalla et al., 2004; Pawlik ablation or combination, together with
et al., 2003). This resection plus ablation advancement of systemic therapy, will fur-
method will likely increase the number of ther improve the prognosis of patients with
patients who are surgically treatable more liver metastases.
than resection alone. When patients have
synchronous liver metastases with primary REFERENCES
colorectal cancer, RFA can be used more
frequently than liver resection in conjunc- Abdalla, E.K., Vauthey, J.N., Ellis, L.M., Ellis, V.,
Pollock, R., Broglio, K.R., Hess, K., and Curley,
tion with colorectal resection because of S.A. 2004. Recurrence and outcomes following
less invasiveness and less complications hepatic resection, radiofrequency ablation, and
with RFA. This is currently performed by combined resection/ablation for colorectal liver
open surgery (Machi et al., 2000), but can metastases. Ann. Surg. 239: 818–827.
be performed more by laparoscopic surgery Berber, E., Flesher, N., and Siperstein, A.E. 2002.
in future because of anticipated increase in Laparoscopic radiofrequency ablation of neu-
roendocrine liver metastases. World J. Surg. 26:
laparoscopic colorectal cancer operations. 985–990.
The true impact of RFA in comparison to Berber, E., Pelley, R., and Siperstein, A.E. 2005.
resection for optimally and not-optimally Predictors of survival after radiofrequency ther-
resectable tumors cannot be determined mal ablation of colorectal cancer metastases to
unless a randomized study is performed. the liver: a prospective study. J. Clin. Oncol. 23:
We believe that there may be a role of RFA 1358–1364.
Bilchik, A.J., Rose, D.M., and Allegra, D.P. 1999.
for smaller resectable metastatic tumors Radiofrequency ablation: A minimally invasive
and that the long-term follow-up results of technique with multiple applications. Cancer
ours, especially in patients of unresectability J. Sci. Am. 5: 356–361.
grade 1 showing encouraging survival, and Bowles, B.J., Machi, J., Limm, W.M.L., Severino,
the results of others would support a pro- R., Oishi, A.J., Furumoto, N.L., Wong, L.L., and
posal for justification of such resection- Oishi, R.H. 2001. Safety and efficacy of radi-
ofrequency thermal ablation in advanced liver
versus-RFA randomization for resectable tumors. Arch. Surg. 136: 864–869.
metastatic liver tumors. This is particularly Choti, M.A., Sitzmann, J.V., Tiburi, M.F.,
true in face of newer chemo-biological Sumetchotimetha, W., Rangsin, R., Schulick,
systemic therapies. R.D., Lillemoe, K.D., Yeo, C.J., and Cameron,
130 J. Machi

J.L. 2002. Trends in long-term survival following Grothey, A., Sargent, D., Goldberg R.M., and
liver resection for hepatic colorectal metastases. Schmell, H.J. 2004. Survival of patients with
Ann. Surg. 235: 759–766. advanced colorectal cancer improves with avail-
Curley, S.A., Izzo, F., Delrio, P., Ellis, L.M., Granchi, ability of fluorouracil-leucovorin, irinotecan,
J., Vallone, P., Fiore, F., Pignata, S., Daniele, B., and oxaliplatin in the course of treatment. J. Clin.
and Cremona, F. 1999. Radiofrequency ablation Oncol. 22: 1209–1214.
of unresectable primary and metastatic hepatic Hellman, P., Ladjevardi, S., Skogseid, B., Akerstrom,
malignancies. Results in 123 patients. Ann. Surg. G., and Elvin, A. 2002. Radiofrequency tissue
230:1–8. ablation using cooled tip for liver metastases
Curley, S.A., Marra, P., Beaty, K., Ellis, L.M., of endocrine tumors. World J. Surg. 26: 1052–
Vauthey, J.N., Abdalla, E.K., Scaite, C., Rant, 1056.
C., Wolff, R., Choi, H., Loyer, E., Vallone, P., Hurwitz, H., Fehrenbacher, L., Novotny, W.,
Fiore, F., Scardino, F., DeRosa, V., Orlando, Cartwright, T., Hainsworth, J., Heim, W., Berlin,
R., Pignata, S., Daniele, B., and Izzo, F. 2004. J., Baron, A., Griffing, S., Holmgren, E., Ferrara,
Early and late complications after radiofre- N., Fyfe, G., Rogers, B., Ross, R., and Kabbinavar,
quency ablation of malignant liver tumors in 608 F. 2004. Bevacizumab plus irinotecan, fluorour-
patients. Ann. Surg. 239: 450–458. acil, and leucovorin for metastatic colorectal
De Baere, T.J., Elias, D., Ducreux, M., Kuoch, V., cancer. N. Engl. J. Med. 350: 2335–2342.
Boige, V., Dromain, C., Lasser, P., and Tursz, Iannitti, D.A., Dupuy, D.E., Mayo-Smith, W.W.,
T. 2003. Percutaneous radiofrequency of liver and Murphy, B. 2002. Hepatic radiofrequency
metastases: single center experience over a five ablation. Arch. Surg. 137: 422–427.
year period. Proc. Am. Soc. Clin. Oncol. 22: 345 Jaeck, D., Oussoultzoglou, E., Rosso, E., Greget,
(Abstract 1387). M., Weber, J.C., and Bachellier, P. 2004. A two-
De Meijer, V.E., Verhoef, C., Kuiper, J.W., Alwayn, stage hepatectomy procedure combined with
I.P.J., Kazemier, G., and Ijzermans, J.N.M. 2006. portal vein embolization to achieve curative
Radiofrequency ablation in patients with pri- resection for initially unresectable multiple and
mary and secondary hepatic malignancies. J. bilobar colorectal liver metastases. Ann. Surg.
Gastrointest. Surg. 10: 960–973. 240: 1037–1051.
Fernandez, F.G., Drebin, J.A., Linehan, D.C., Jiao, L.R., Hansen, P.D., Havlik, R., Mitry, R.R.,
Dehdashti, F., Siegel, B.A., and Strasberg, Pignatelli, M., and Habib, N. 1999. Clinical
S.M. 2004. Five-year survival after resection short-term results of radiofrequency ablation in
of hepatic metastases from colorectal cancer in primary and secondary liver tumors. Am. J. Surg.
patients screened by positron emission tomogra- 177: 303–306.
phy with F-18 fluorodeoxyglucose (FDG-PET). Lencioni, R., Crocetti, L., Cioni, D., Dellapina, C.,
Ann. Surg. 240: 438–450. and Bartolozzi, C. 2004. Percutaneous radiofre-
Fong, Y., Fortner, J., Sun, R.L., Brennan, M.F., quency ablation of hepatic colorectal metastases.
and Blumgart, L.H. 1999. Clinical score for Technique, indications, results, and new prom-
predicting recurrence after hepatic resection for ises. Invest. Radiol. 39: 689–697.
metastatic colorectal cancer. Analysis of 1001 Lencioni, R., Goletti, O., Armillotta, N., Paolicchi,
consecutive cases. Ann. Surg. 230: 309–321. A., Moretti, M., Cioni, D., and Donati, F. 1998.
Giantonio, B.J., Catalano, P.J., and Meropol, N.J. Radio-frequency thermal ablation of liver metas-
2005. High-dose bevacizumab improves survival tases with cooled-tip electrode needle: results of
when combined with FOLFOX 4 in previously a pilot clinical trial. Eur. Radiol. 8: 1205–1211.
treated advanced colorectal cancer: results from Machi, J., Oishi, A.J., Morioka, W.K., Yu, M.,
ECOG study E3200. Proc. Am. Soc. Clin. Oncol. Hundahl, S.A., Furumoto, N.L., and Oishi, R.H.
(Abstract 2). 23: Part 1. 2000. Radiofrequency thermal ablation of syn-
Gillams, A.R., and Lees, W.R. 2004. Radio- chronous metastatic liver tumors can be performed
frequency ablation of colorectal liver metastases safely in conjunction with colorectal cancer resec-
in 167 patients. Eur. Radiol. 14: 2261–2267. tion. Cancer J. 6(Suppl 4): S344–S350.
9. Unresectable Liver Metastases from Colorectal Cancer 131

Machi, J., Uchida, S., Sumida, K., Limm, W.M.L., (FOLFOX4) in patients with advanced colorectal
Hundahl, S.A., Oishi, A.J., Furumoto, N.L., cancer previously treated with 5FU. Proc. Am.
and Oishi, R.H. 2001. Ultrasound-guided radi- Soc. Clin. Oncol. 24 (Abstract 3506), page 3506.
ofrequency thermal ablation of liver tumors: Registry of hepatic metastases. 1988. Resection of
percutaneous, laparoscopic, and open surgical the liver for colorectal carcinoma metastases: a
approaches. J. Gastrointest. Surg. 5:477–489. multi-institutional study of indications for resec-
Machi, J., Oishi, A.J., Mossing, A.J., Furumoto, tion. Surgery 103: 278–288.
N.L., and Oishi, R.H. 2002. Hand-assisted laparo- Rougier, P., Lepille, D., Bennouna, J., Marre, A.,
scopic ultrasound-guided radiofrequency thermal Ducreux, M., Mignot, L., Hua, A., and Mery-
ablation of liver tumors: a technical report. Surg. Mignard, D. 2002. Antitumour activity of three
Laparosc. Endosc. Percutan. Tech. 12: 160–164. second-line treatment combinations in patients
Machi, J., Oishi, A.J., Sumida, K., Sakamoto, K., with metastatic colorectal cancer after optimal
Furumoto, N.L., Oishi, R.H., and Kylstra, J.W. 5-FU regimen failure: a randomized, multicentre
2006. Long-term outcome of radiofrequency phase II study. Ann. Oncol. 13: 1558–1567.
ablation for unresectable liver metastases from Scheele, J., Stang, R., Altendorf-Hofmann, A.,
colorectal cancer: evaluation of prognostic fac- and Paul, M. 1995. Resection of colorectal liver
tors and effectiveness in first- and second-line metastases. World J. Surg. 19: 59–71.
management. Cancer J. 12: 318–326. Seidenfeld, J., Korn, A., and Aronson, N. 2002.
Nordlinger, B., Guiguet, M., Vaillant, J.C., Balladur, Radiofrequency ablation of unresectable liver
P., Boudjema, K., Bachellier, P., and Jaeck, D. metastases. J. Am. Coll. Surg. 195: 378–386.
1996. Surgical resection of colorectal carcinoma Siperstein, A., Garland, A., Engle, K., Rogers,
metastases to the liver. A prognostic scoring S., Barbar, E., Foroutani, A., String, A., Ryan,
system to improve case selection, based on 1568 T., and Ituarte, P. 2000. Local recurrence after
patients. Cancer 77: 1254–1262. laparoscopic radiofrequency thermal ablation of
Oshowo, A., Gillams, A., Harrison, E., Lees, W.R., hepatic tumors. Ann. Surg. Oncol. 7: 106–113.
and Taylor, I. 2003. Comparison of resection Solbiati, L., Goldberg, S.N., Ierace, T., Livraghi, T.,
and radiofrequency ablation for treatment of Meloni, F., Dellanoce, M., Sironi, S., and Gazelle,
solitary colorectal liver metastases. Br. J. Surg. G.S. 1997. Hepatic metastases: Percutaneous
90: 1240–1243. radio-frequency ablation with cooled-tip elec-
Parikh, A.A., Curley, S.A., Fornage, B.D., and trodes. Radiology 205: 367–373.
Ellis, L.M. 2002. Radiofrequency ablation of Solbiati, L., Ierace, T., Tonolini, M., Bellobuono,
hepatic metastases. Semin. Oncol. 29: 168–182. A., and Cova, L. 2003. Long-term survival of
Pawlik, T.M., Izzo, F., Cohen, D.S., Morris, J.S., patients treated with radiofrequency ablation
and Curley, S.A. 2003. Combined resection and for liver colorectal metastases: improved out-
radiofrequency ablation for advanced hepatic come with increasing experience. Radiology
malignancies: results in 172 patients. Ann. Surg. 229: 411–412.
Oncol. 10: 1059–1069. White, T.J., Roy-Choudhury, S.H., Breen, D.J.,
Pitot, H.C., Rowland, K.M., Sargent, D.J., Philip, Cast, J., Maraveyas, A., Smyth, E.F., Hartley,
P.A., Mitchell, E.P., Goldberg, R.M., and Alberts, J.E., and Monson, J.R.T. 2004. Percutaneous
S.R. 2005. N9841: a randomized, phase III radiofrequency ablation of colorectal hepatic
equivalence trial of irinotecan (CPT-11) ver- metastases – initial experience. Dig. Surg.
sus oxaliplatin/5-fluorouracil (FU)/leucovorin 21: 314–320.
A. Diagnosis
10
Screening with Ultrasonography
of Patients at High-Risk for Hepatocellular
Carcinoma: Thrombocytopenia as a Valid
Surrogate of Cirrhosis
Sheng-Nan Lu, Jing-Houng Wang, Kwong-Ming Kee, and Po-Lin Tseng

INTRODUCTION HBV-related HCC is a common cause


of HCC in Asia and sub-Saharan Africa.
Sequelae of chronic hepatitis B virus (HBV) The major transmission of HBV infection
and hepatitis C virus (HCV) infection, in these countries is vertical transmission
including hepatocellular carcinoma (HCC), from carrier mother to newborn at the
liver cirrhosis (LC) and their complications, time of birth. The majority of infected
are health burdens of the world. Improving newborns will develop chronic and sub-
treatment results for early stage HCC clinical hepatitis, subsequently leading to
makes the screening meaningful. Two- LC and HCC. This transmission route is
staged HCC screening identifying high risk rare in Western countries, where sexual
subjects in the first stage and screening for transmission or intravenous drugs use are
HCC by ultrasonography (US) in the second the major causes of HBV infections during
stage is a practical and workable concept. adolescences, and which mean infected
The benefit of US screening on high risk individuals are at lower risk of develop-
groups has been well documented, but “who ing into chronic infection. Consequently,
should be included in the US screening” HBV-related HCC in Western countries is
depends on cost-benefits and considera- relatively low.
tions of feasibility. On the other hand, HCV infections in
adults have high rates (~ 75%) of chronic
hepatitis and liver disease. Approximately
Epidemiology of Hepatocellular
25% of patients with chronic HCV infec-
Carcinoma and Liver Cirrhosis
tion progress to LC within 20 years
There are different etiologies of HCC (Fattovich et al., 1997). Among those
around the world, such as chronic HBV and patients with cirrhosis, 1–4% develop HCC
HCV infections, which lead to advanced each year. The burden of HBV-related
liver disease and eventual development of HCC will decrease in the future because
LC and HCC. These two viral infections HBV mass vaccination programs will con-
are the major risk factors for HCC. sequently reduce the incidence of HCC.

137
138 S.-N. Lu et al.

However, HCV-related HCC is increasing Toyoda et al., 2006; Leykum et al., 2007;
in some countries such as Taiwan (Lu et al., Chen et al., 2002). A community study
2006a), Japan (Kiyosawa et al., 2004), and with a cohort of 4,843 Taiwanese at risk
the United States (Wong et al., 2000). In for HCC has demonstrated that the morta-
the United States, after incidents of acute lity was 41% lower in the screening group
HCV infection peaked in the mid-1980s, compared to the control group (Chen et al.,
increasing numbers of future cases of 2002). To assess the value of screening,
HCV-related HCC can be expected. Until one randomized controlled study with
an HCV vaccination is developed and 18,816 Chinese at risk for HCC indicated
become readily available, chronic HCV that biannual screening reduces HCC mor-
infection will remain a major cause of LC tality by 37%. Compared with the control
and HCC. group, the screened group had a higher
number of HCC patients diagnosed at an
earlier stage, with better overall survival
Benefit of HCC Screening
rates (Zhang et al., 2004). It is obvious that
Individuals at risk for developing HCC surveillance of high risk groups for HCC
usually present with clinical symptoms has a sound scientific basis.
when HCC is in a far advanced stage, Alpha-fetoprotein (AFP) and/or ultra-
without effective therapeutic options. sonography (US) are the most widely used
There is clearly a need for early diagnosis tools for surveillance. For HCC screening,
of HCC. Thus, screening and surveillance AFP has a sensitivity of 17–65% and a
appear to be very appropriate. Surveillance specificity of 80–99%, depending on the
programs are ideal for a disease such as prevalence of HCC in the screened popula-
HCC, because it is relatively common tion as well as the cut-off level for the diag-
in patients with chronic liver disease, nosis (Daniele et al., 2004). The results for
has readily identifiable risk factors, high US examination depend on the expertise of
mortality rates, and potentially curative operator and the equipment available. In a
therapy (Di Bisceglie, 2004). However, systemic review, the pooled estimated sen-
there is considerable controversy regard- sitivity and specificity for US in diagnosing
ing the role of screening and surveillance HCC is 60% and 97%, respectively (Colli
in HCC management. The value of screen- et al., 2006). Although neither AFP nor US
ing and surveillance for HCC should be alone is an effective surveillance tool, their
assessed by its impact on detecting earlier combination increases the sensitivity of
stage of the disease, increasing the effec- screening by 5–10% (Daniele et al., 2004;
tive application of curative treatment, and Colli et al., 2006). The optimal surveil-
thus reducing mortality. Although there lance interval for HCC has yet to be deter-
are inconsistent conclusions, most cohort mined. However, based on the estimated
studies from Western and Eastern countries growth rate of HCC, the suggested interval
have shown that surveillance of patients for surveillance in patients has been set at
with viral hepatitis or cirrhosis increases 6 months. Although there are no firm data
early detection and effective application to support the hypothesis that 6 months
of curative treatments (Zoli et al., 1996; is better than 12 months, recent practice
Yuen et al., 2000; Bolondi et al., 2001; guideline on HCC management recommend
10. Screening with Ultrasonography of Patients at High-Risk for Hepatocellular Carcinoma 139

US surveillance of patients at high risk of are an ideal approach to detect LC in


developing HCC within an interval of 6–12 the first stage screening. Direct fibrosis
months (Bruix and Sherman, 2005). markers include collagens, glycoproteins,
Cost-effectiveness analyses of various polysaccharides, metalloproteinases, tissue
HCC screening strategies differ widely. inhibitors of metalloproteinases, and cyto-
The cost per treatable HCC and per year kines. None of these biomarkers is liver
of life saved is US$1,667–17,934 and specific, and alterations in the levels of
US$25,232–284,000, respectively. Such these markers depend on the etiology of
varied results might be due to differences underlying liver disease. Due to lack of
in the incidence of HCC in the screening assay standardization, it is difficult to
population, screening intervals, diagnostic draw strong conclusions regarding which
modalities, available treatment methods, assays have better performance. Indirect
and tumor recurrence rates (Daniele et al., markers of liver fibrosis include aspar-
2004). New markers in identifying patients tate transaminase (AST)/ alanine transam-
at high risk of developing HCC, curative inase (ALT) ratio (AAR), prothrombin
and effective HCC treatment modalities time, platelet counts, and AST to platelet
and better strategies might make HCC ratio index (APRI). More sophisticated
surveillance more cost-effective and acce- indices, including cirrhosis discriminant
ptable in the future. score (CDS), PGA index, PGAA index,
Fibrotest, AcitTest, and Forns index have
also been developed. The cost of these pro-
Surrogate Tests for Liver Cirrhosis
prietary tests is similar to that of a biopsy,
and Fibrosis
and is consequently not suitable for large-
Many tools are used to evaluate the sever- scale community screening. PGA index has
ity of liver fibrosis, such as liver biopsy, shown greater accuracy in detecting LC
image modalities, and laboratory tests. (66–72%) in alcoholic liver disease than
Liver biopsy has been the gold stand- viral liver disease (Kelleher and Afdhal,
ard for evaluating liver fibrosis, but has 2005). Fibrotest (Imbert-Bismut et al.,
been limited by interobserver and intraob- 2001) was used for correctly identifying
server variability and sampling error. the stage of disease in 46% of all patients.
Furthermore, the invasive procedure is However, false-positives may be caused by
not suitable for community screening. Gilbert’s syndrome, cholestasis and acute
Fibroscan, which measures the stiffness liver inflammation. ActiTest is a modifica-
of hepatic tissue, is sensitive for stag- tion of Fibrotest, with the addition of ALT
ing liver fibrosis. It is a reliable tool for to reflect both liver fibrosis and necroin-
detecting LC with areas under ROC curves flammatory activity. Major criticisms of
(AUC) of 0.97 (Kelleher and Afdhal, the Forns index include concerns regarding
2005). Although it is quick, painless, and the impact of lipid abnormality and cho-
reproducible, the equipment is not readily lesterol altering medications (Afdhal and
available worldwide. Nunes, 2004; Kelleher and Afdhal, 2005).
On the other hand, blood samples are AAR ≥ 1 had wide range of sensiti-
easily collected in large scale community vity (31.5–77.8%) and PPV (13.5–100%),
screening. Hence, serum markers (Table 10.1) based on differing LC prevalences.
140 S.-N. Lu et al.

Table 10.1. Non-invasive diagnostic surrogate tests for liver fibrosis and cirrhosis.
Sensitivity Specificity
Tests Cutoff Disease (%) (%) Comments References
Direct
markers
HA 30 ug/l HCV 90 73 Boeker et al., 2002
100 ug/l Viral 84 87 Murawaki et al., 1996
110 ug/l HCV 79.2 89.4 Guechot et al., 1996
183.5 ug/l HCV 80 80 Saitou et al., 2005
Laminin 1.26 U/ml HCV 80 83 Severe liver injury Walsh et al., 2000
Type IV C 148 ng/ml HCV 73 85 Severe liver injury Walsh et al., 2000
160 ng/ml Viral 80 81 Murawaki et al., 1996
6.55 ng/ml HCV 60 61 Saitou et al., 2005
PIIINP 1.0 U/ml HCV 60 74 Guechot et al., 1996
0.995 U/ml HCV 77 66 Saitou et al., 2005
Pro 1,500 ug/l HCV 74 100 Boeker et al., 2002
MMP-2
TIMP1 950 ug/l HCV 100 75 Boeker et al., 2002
YKL-40 284.8 ng/ml HCV 80 71 Saitou et al., 2005
Indirect
markers
AST/ALT ratio ≥1 HCV 77.8 96.9 Giannini and Testa, 2003
APRI ≤1 HCV 89 75 Wai et al., 2003
≤2 HCV 75 93 Wai et al., 2003
Platelet <130 × 109/l HCV 91.1 88.3 Giannini and Testa, 2003
<150 × 109/l HCV 68.2 76.4 Lu et al., 2006b
<150 × 109/l HBV 52.5 78 Lu et al., 2006b
API ≥6 HCV 67 87 Borroni et al., 2006
CDS ≥7 HCV 17 100 Borroni et al., 2006
PGA index >3 Mixed 91 81 Liver fibrosis Teare et al., 1993
Forns <4.2 HCV 94 51 Significant fibrosis Forns et al., 2002
index
Fibrotest <0.1 HCV 100 22 Imbert-Bismut et al., 2001
>0.6 HCV 70 95 Imbert-Bismut et al., 2001
<0.1 HCV 92 29 Rossi et al., 2003
>0.6 HV 42 94 Rossi et al., 2003
Abbreviations: AST: aspartate transaminase; ALT: alanine transaminase; HA: hyaluronic acid; PIIINP: procollagen type III aminot-
erminal peptide; MMP: matrix metalloproteinase TIMP: tissue inhibitor of metalloproteinase; APRI: AST to platelet ratio index;
API: age-platelet index; CDS: cirrhosis discriminate score.

The efficacy is not appropriate for all forms A blood test for platelet count is the
of liver diseases, especially alcoholic liver simplest, most inexpensive, and easily
disease and autoimmune hepatitis. APRI is available serum marker for predicting LC.
a simple test for predicting cirrhosis with The possible causes of thrombocytopenia
an AUC of 0.94 (Wai et al., 2003). When are decreased thrombopoietin production,
using upper (1.0) and lower (2.0) as cut-off antibody mediated platelet destruction,
values, 81% of the patients with or without hypersplenism, and myelotoxic effects.
cirrhosis were characterized. Its perform- Confounding factors are infectious disease
ance for evaluating cirrhosis is similar and hematological disease. The study by
to other, more complex indexes, such as Lu et al. (2006b) investigated the diagnos-
Forns index and Fibrotest. However, APRI tic performance of platelet counts in pre-
cannot be completely standardized due to dicting pathological LC in chronic HBV
the variability of AST ranges considered and HCV patients and US LC in HCV
normal in different laboratories. endemic community residents. Platelet
10. Screening with Ultrasonography of Patients at High-Risk for Hepatocellular Carcinoma 141

counts decrease while pathological fibro- percent of all participants were identified
sis stages increase in chronic HCV, but as belonging to the high risk group, and
not in HBV patients. AUC was larger were scheduled for US (Chen et al., 2002).
in HCV (0.799) than HBV (0.672) and Although more than 95% of HCC cases
the validity was acceptable when cutoff could be identified in this manner, the cost
was set at 150 × 109/l in cases of chronic of screening was high, and it is a daunting
hepatitis B. At this cutoff level, sensitivity task to perform US on 29% of the adult
and specificity were 52.5% and 78% in population in a community.
HBV patients, and 68.2% and 76.4% in Among 4,042 proven HCC cases at a
HCV patients, respectively. In a small medical center in Taiwan, 2,032 (50.3%)
scale HCV endemic community (Lu et al., were HBVAg positive, 1,151 (28.5%)
2006b), the prevalence of HBsAg, anti- were anti-HCV positive, 449 (11.1%)
HCV, both, and neither were 9.0%, 37.3%, were positive for both, and 410 (10.1%)
3.5% and 50.2%; the patients’ prevalence were negative for both. Using 150 × 109/l
of thrombocytopenia were 5.6%, 26.7%, as the cutoff, 1,926 (48%) of the HCC
42.9%, and 3.0%, respectively. The mean cases were found to be thrombocyto-
platelet counts decreased as the US paren- penic. Thrombocytopenic patients made
chyma score increased. Using this cutoff up 42% (860/2,032) of the HBV-related
to identify US cirrhosis, sensitivity was HCC cases, 63% (720/1,151) of the HCV-
76.2%, specificity was 87.8%, and accu- related cases, 50% (225/449) of cases
racy was 86.6%. For cases with chronic positive to both, and 30% (121/410) of the
HCV infection, platelet counts inversely cases tested negative for both. In short,
correlated to both histological and US HCV-related HCC cases had a higher rate
scores for hepatic fibrosis. Platelet count of thrombocytopenia than HBV-related
is cheap, easily interpreted and not inferior cases (Lu et al., 2006b).
to others tests, and should be one of most In an HBsAg (12.4%) and HCV
feasible surrogates. Platelet counts can be (40.8%)-endemic area, 825 (17.9%) of
employed not only for the diagnosis of 4,616 participants were thrombocyto-
hepatic fibrosis and cirrhosis, but also for penic. Of 586 (71%) participants in HCC
identification of the population at high risk screening by US and AFP, 116 (19.8%)
of developing HCC. were diagnosed as LC. Forty-two cases
had suspected liver nodules, and 25 (4.3%)
cases were confirmed as HCC. Only 13
Ultrasonographic HCC Screening on the
of 25 HCC cases had elevated AFP. In
Thrombocytopenic Adult
the low HCV prevalence area (HBsAg
Two-staged community-based HCC 12%, anti-HCV 2%), only 104 throm-
screening seems reasonable. The first bocytopenic cases (6.1%) were found
stage is identification of high risk groups, among 1,694 participants. Forty-four
and the second stage is US screening of patients (42.3%) participated in US and
the high risk group only. In a community AFP screening, but only two (4.5%)
study in Taiwan, HBsAg, anti-HCV, AST, were diagnosed at an early stage of liver
ALT, AFP, and a family history of HCC cirrhosis and no incidence of HCC was
were selected as markers for the identifi- found (Lu et al., 2006b). Among the 25
cation of high risk groups. Twenty-nine detected HCC cases, tumor diameters
142 S.-N. Lu et al.

were < 3 cm in 13 cases, 3–5 cm in REFERENCES


6 cases, 5–10 cm in 3 cases, and > 10 cm Afdhal, N.H., and Nunes, D. 2004. Evaluation of liver
in 3 cases. fibrosis: a concise review. Am. J. Gastroenterol.
In 2004 a community-based HCC screen- 99: 1160–1174.
ing was conducted and 56,702 adults parti- Boeker, K.H., Haberkorn, C.I., Michels, D.,
cipated. Subjects with thrombocytopenia Flemming, P., Manns, M.P., and Lichtinghagen,
(platelet counts < 150 × 109/l) or with AFP R. 2002. Diagnostic potential of circulating
TIMP-1 and MMP-2 as markers of liver fibrosis
> 20 ng/ml were enrolled for the second in patients with chronic hepatitis C. Clin. Chim.
stage US screening. Three thousand two Acta 316: 71–81.
hundred and thirty four subjects (5.7%) Bolondi, L., Sofia, S., Siringo, S., Gaiani, S., Casali,
were candidates for US screening, and 2,983 A., Zironi, G., Piscaglia, F., Gramantieri, L.,
(92.2%) responded. Of 132 suspected cases, Zanetti, M., and Sherman, M. 2001. Surveillance
123 (89.8%) complied with referral for con- programme of cirrhotic patients for early detec-
tion and treatment of hepatocellular carcinoma:
firmation, and 72 (58.5%) were confirmed a cost effectiveness analysis. Gut 48: 251–259.
to be HCC cases. Among the confirmed Borroni, G., Ceriani, R., Cazzaniga, M., Tommasini,
patients, only five were too advanced to be M., Roncalli, M., Maltempo, C., Felline, C.,
actively treated. In addition to thrombocyto- and Salerno, F. 2006. Comparison of simple
penia, AFP was added as a marker of high risk tests for the non-invasive diagnosis of clinically
identification, with a sensitivity estimated silent cirrhosis in chronic hepatitis C. Aliment
Pharmacol. Ther. 24: 797–804.
at 70%. That is, 70% of HCC cases among Bruix, J., and Sherman, M. 2005. Management of hepa-
the population had either thrombocytopenia tocellular carcinoma. Hepatology 5: 1208–1236.
or AFP > 20 ng/ml. Among 3,234 high risk Chen, T.H., Chen, C.J., Yen, M.F., Lu, S.N., Sun,
subjects, the 42% without chronic HBV C.A., Huang, G.T., Yang, P.M., Lee, H.S., and
or HCV infections should not be at high Duffy, S.W. 2002. Ultrasound screening and risk
risk for HCC. Therefore, before entering of factors for death from heptocellular carci-
noma in a high risk group in Taiwan. Int. J.
a long-term surveillance program, their Cancer 98: 257–261.
HBsAg and anti-HCV should be checked Colli, A., Fraquelli, M., Casazza, G., Massironi,
(Lu et al., 2008). S., Colucci, A., Conte, D., and Duca, P. 2006.
In conclusion, for cases of chronic HCV Accuracy of ultrasonography, spiral CT, magnetic
infection, platelet counts are inversely cor- resonance, and alpha-fetoprotein in diagnosing
related to both histological and US scores hepatocellular carcinoma: a systemic review. Am.
J. Gastroenterol. 101: 513–523.
for hepatic fibrosis. This method is cheap, Daniele, B., Bencivenga, A., Megna, A.S., and
easily interpreted, and not inferior to other Tinessa, V. 2004. α-fetoprotein and ultrasonog-
diagnostic methods, and should be consid- raphy screening for hepatocellular carcinoma.
ered one of the most feasible surrogates. Gastroenterology 127: 108–112.
Platelet counts can be employed not only Di Bisceglie, A.M. 2004. Issues in screening
for the diagnosis of hepatic fibrosis and and surveillance for hepatocellular carcinoma.
Gastroenterology 127: S104–107.
cirrhosis, but also for identification of the Fattovich, G., Giustina, G., Degos, F., Tremolada, F.,
population groups at high risk of develop- Diodati, G., Almasio, P., Nevens, F., Solinas, A.,
ing HCC. Due to economic considera- Mura, D., Brouwer, J.T., Thomas, H., Njapoum,
tions, this design is especially feasible in C., Casarin, C., Bonetti, P., Fuschi, P., Basho, J.,
HCV and HCC endemic areas with limited Tocco, A., Bhalla, A., Galassini, R., Noventa, F.,
medical resources. Schalm, S.W., and Realdi, G. 1997. Morbidity
10. Screening with Ultrasonography of Patients at High-Risk for Hepatocellular Carcinoma 143

and mortality in compensated cirrhosis type C: high-risk for hepatocellular carcinoma. Cancer
a retrospective follow-up study of 384 patients. 107: 2212–2222.
Gastroenterology 112: 463–472. Lu, S.N., Wang, J.H., Chen, P.F., Tung, H.D.,
Forns, X., Ampurdanes, S., Llovet, J.M., Aponte, Tseng, P.L., Hung, C.H., Kee, K,M., Chen,
J., Quintó, L., Martínez-Bauer, E., Bruguera, C.H., Chang, K.C., Lee, C.M., Changchien,
M., Sánchez-Tapias, J.M., and Rodés, J. 2002. C.S., Chen, Y.D., Tsai, L.S., and Chen, T.H.
Identification of chronic hepatitis C patients 2008. Community-based mass ultrasonographic
without hepatic fibrosis by a simple predictive screening of hepatocellular carcinoma among
model. Hepatology 36: 986–992. thrombocytopenic adults. Cancer Epidemiol.
Giannini, E., and Testa, R. 2003. The metabolic Biomarkers Prev. 17: 1813–1821.
syndrome: all criteria are equal, but some criteria Murawaki, Y., Ikuta, Y., Koda, M., Yamada, S., and
are more equal than others. Arch. Intern. Med. Kawasaki, H. 1996. Comparison of serum 7S
163: 2787–2788. fragment of type IV collagen and serum central
Guechot, J., Laudat, A., Loria, A., Serfaty, L., triple-helix of type IV collagen for assessment of
Poupon, R., and Giboudeau, J. 1996. Diagnostic liver fibrosis in patients with chronic viral liver
accuracy of hyaluronan and type III procollagen disease. J. Hepatol. 24: 148–154.
amino-terminal peptide serum assays as markers of Rossi, E., Adams, L., Prins, A., Bulsara, M., de
liver fibrosis in chronic viral hepatitis C evalu- Boer, B., Garas, G., MacQuillan, G., Speers, D.,
ated by ROC curve analysis. Clin. Chem. 42: and Jeffrey, G. 2003. Validation of the FibroTest
558–563. biochemical markers score in assessing liver
Imbert-Bismut, F., Ratziu, V., Pieroni, L., Charlotte, fibrosis in hepatitis C patients. Clin. Chem. 49:
F., Benhamou, Y., Poynard, T. and MULTIVIRC 450–454.
Group. 2001. Biochemical markers of liver fibro- Saitou, Y., Shiraki, K., Yamanaka, Y.,
sis in patients with hepatitis C virus infection: a Yamaguchi, Y., Kawakita, T., Yamamoto, N.,
prospective study. Lancet 357: 1069–1075. Sugimoto, K., Murata, K., and Nakano, T.
Kelleher, T.B., and Afdhal, N. 2005. Noninvasive 2005. Noninvasive estimation of liver fibrosis
assessment of liver fibrosis. Clin. Liver Dis. 9: and response to interferon therapy by a serum
667–683. fibrogenesis marker, YKL-40, in patients
Kiyosawa, K., Umemura, T., Ichijo, T., Matsumoto, with HCV-associated liver disease. World J.
A., Yoshizawa, K., Gad, A., and Tanaka, E. Gastroenterol. 11: 476–481.
2004. Hepatocellular carcinoma: recent trends in Teare, J.P., Sherman, D., Greenfield, S.M.,
Japan. Gastroenterology 127: S17–26. Simpson, J., Bray, G., Catterall, A.P., Murray-
Leykum, L.K., El-Serg, H.B., Cornell, J., and Lyon, I.M., Peters, T.J., Williams, R., and
Papadopoulos, K.P. 2007. Screening for hepato- Thompson, R.P. 1993. Comparison of serum
cellular carcinoma veterans with hepatitis C on procollagen III peptide concentrations and PGA
disease stage, treatment received, and survival. index for assessment of hepatic fibrosis. Lancet
Clin. Gastroenterol. Hepatol. 5: 508–512. 342: 895–898.
Lu, S.N., Su, W.W., Yang, S.S., Chang, T.T., Toyoda, H., Kumada, T., Kiriyama, S., Sone, Y.,
Cheng, K.S., Wu, J.C., Lin, H.H., Wu, S.S., Tanikawa, M., Hisanaga, Y., Yamaguchi, A.,
Lee, C.M., Changchien, C.S., Chen, C.J., Sheu, Isogai, M., Kaneoka, Y., and Washizu, J. 2006.
J.C., Chen, D.S., and Chen, C.H. 2006a. Secular Impact of surveillance on survival of patients
trends and geographic variations of hepatitis B with initial hepatocellular carcinoma: a study
virus and hepatitis C virus-associated hepatocel- from Japan. Clin. Gastroenterol. Hepatol. 4:
lular carcinoma in Taiwan. Int. J. Cancer 119: 1170–1176.
1946–1952. Wai, C.T., Greenson, J.K., Fontana, R.J.,
Lu, S.N., Wang, J.H., Liu, S.L., Hung, C.H., Chen, Kalbfleisch, J.D., Marrero, J.A., Conjeevaram,
C.H., Tung, H.D., Chen, T.M., Huang, W.S., Lee, H.S., and Lok, A.S. 2003. A simple noninvasive
C.M., Chen, C.C., and Changchien, C.S. 2006b. index can predict both significant fibrosis and
Thrombocytopenia as a surrogate for cirrhosis cirrhosis in patients with chronic hepatitis C.
and a marker for the identification of patients at Hepatology 38: 518–526.
144 S.-N. Lu et al.

Walsh, K.M., Fletcher, A., MacSween, R.N., and treatment: Hong Kong experience. Hepatology
Morris, A.J. 2000. Basement membrane peptides 31: 330–335.
as markers of liver disease in chronic hepatitis C. Zhang, B.H., Yang, B.H., and Tang, Z.Y. 2004.
J. Hepatol. 32: 325–330. Randomized controlled trial of screening for
Wong, J.B., McQuillan, G.M., McHutchison, J.G., hepatocellular carcinoma. J. Cancer. Res. Clin.
and Poynard, T. 2000. Estimating future hepatitis Oncol. 130: 417–422.
C morbidity, mortality, and costs in the United Zoli, M., Magalotti, D., Bianchi, G., Gueli, C.,
States. Am. J. Public Health 90: 1562–1569. Marchesini, G., and Pisi, E. 1996. Efficacy
Yuen, M.F., Cheng, C.C., Lauder, I.J., Lam, S.K., of surveillance program for early detection
Ooi, C.G., and Lai, C.L. 2000. Early detection of of hepato-cellular carcinoma. Cancer 78:
hepatocellular carcinoma increases the chance of 977–985.
11
Hepatocellular Carcinoma:
Contrast-Enhanced Sonography
Byung Ihn Choi and Se Hyung Kim

INTRODUCTION Choi, D. et al., 2000; Ding et al.,


2 0 0 1 ; Meloni et al., 2001; Solbiati
In patients with chronic liver disease, sono- et al., 1999; Youk et al., 2003b) as well
graphy and measurements of serum alpha- as for making a correct diagnosis of HCC
fetoprotein (αFP) levels are frequently used by observing tumoral vascularity (Choi
to screen for hepatocellular carcinoma et al., 2000a; Dill-Macky et al., 2002; Kim
(HCC). The sensitivity of sonography for et al., 1998; Wen et al., 2004). Indeed,
HCC detection, however, in the end-stage there is an increasing consensus that the
cirrhotic liver is only 50% (Dodd et al., use of contrast agents improves the ability
1992). Furthermore, sonographic find- of sonography to detect and characterize
ings of HCC are variable and frequently focal hepatic lesions. Levovist, one of the
nonspecific. Hepatocellular carcinoma is most popular sonographic contrast agents,
highly vascularized and often shows irreg- is a suspension of galactose microparticles
ular neovascularization within the tumor. in sterile water. The microbubbles (2–8 μm
Therefore, detection and characterization in diameter, with a mean of 3 μm), which
of tumor vascularity are important in the are stabilized in the microparticle suspen-
differential diagnosis, the choice of treat- sion, can traverse the pulmonary capillary
ment method, and assessment of the thera- bed and diverse into whole blood circula-
peutic response for HCC. Although color tion (Kim et al., 1998).
and power Doppler sonography are helpful In this chapter, the physics of micro-
in detecting vascularity of hepatic tumors, bubbles is briefly reviewed, and various
it has not proved satisfactory. contrast-enhanced sonographic techniques
Since intravenous microbubble contrast for HCC in sequence of their develop-
agents including SH U 508A (Levovist; ments and their strengths and weaknesses
Schering AG, Berlin, Germany) for are discussed: contrast-enhanced color
sonography have become available, many Doppler sonography, contrast-enhanced
researchers have reported that contrast- power Doppler sonography, contrast-
enhanced sonography could be a promis- enhanced harmonic power Doppler sonog-
ing technique for assessing post-treatment raphy, contrast-enhanced pulse-inversion
response of HCC (Bartolozzi et al., 1998; harmonic sonography, contrast-enhanced

145
146 B.I. Choi and S.H. Kim

coded harmonic sonography, contrast- and the subsequent ones are the second,
enhanced agent detection imaging, and low third, and fourth harmonics.
mechanical index imaging. Finally, the Bubbles in a liquid tend to diffuse and
usefulness of contrast-enhanced sonogra- disappear unless they are stabilized by
phy in assessing the therapeutic response some form of a shell. Once the shell is dis-
of HCCs is reviewed. rupted, the gas inside will diffuse into the
surrounding fluid. The mechanical index
(MI), defined originally to predict the onset
PHYSICS OF MICROBUBBLES of cavitation in fluids, also gives an indica-
tion of the likelihood of bubble destruction.
Microbubble contrast agents with diagnos- It has been well established that the acous-
tic ultrasound have been an active area of tic power level used during routine exami-
research since Gramiak and Shah (1968) nations destroys the contrast microbubbles.
observed opacification of the right ventri- The blood flow in a normal capillary bed is
cle after an injection of saline. The earliest
on the order of 1 mm/s, and a typical capil-
microbubbles were unable to pass through lary is ~ 1 mm long. Thus, if the contrast
the lungs, and so were only able to opacify within a capillary is destroyed, it will take
the right ventricle. But, during the past 2 ~ 1 s or more to refill the capillary. Given the
decades, stabilized microbubbles capable of branching structure of the microvasculature
transpulmonary passage for left-side blood and the thickness of a typical scan plane,
pool enhancement were developed by sev- it can take several seconds to replenish the
eral major pharmaceutical companies. contrast in the scan plane, depending on
An acoustic wave generated by an the flow rate to the organ. During real-time
ultrasonic system consists of alternating scanning at normal output power levels, the
high and low pressures at frequencies contrast is never given a chance to fill the
of 1.5–10 MHz. When an acoustic wave microvasculature. This was first observed
encounters a microbubble, it alternately by Porter et al. (1996) when they found
compresses the microbubble on the posi- that triggered imaging allows much better
tive pressure and expands it on the nega- visualization of contrast within the myo-
tive pressure. On the positive portion of cardium. This led to the widespread use
the wave the microbubbles are compressed of electrocardiographic triggering during
in a different fashion than the way they myocardial contrast echo, users often trig-
expand in the negative portion. This resultsgering only once every four or more cardiac
in an asymmetric, nonlinear bubble oscil- cycles. Similar techniques have been used
lation. Instead of producing a sinusoidal to image flow in the hepatic parenchyma.
echo with a clean frequency spectrum
like the transmitted signal, it produces an
odd-looking echo with an asymmetric top CONTRAST-ENHANCED
and bottom. This asymmetry produces COLOR DOPPLER
harmonics and can be used to enhance the SONOGRAPHY
signals from the bubbles. In the frequency
spectrum of the bubble echoes, the first Doppler sonography can provide informa-
major hump is the fundamental component tion on blood flow that is useful in the
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 147

differential diagnosis of a focal hepatic of several studies have shown that power
lesion. Tanaka et al. (1990) have described Doppler sonography is more sensitive than
a color Doppler pattern of intra-lesional color Doppler US in the depiction of vas-
flow which may be helpful in differen- cular flow in focal hepatic lesions, includ-
tiating HCC from other hepatic tumors. ing HCC (Choi B.I. et al., 1996; Lencioni
A basket pattern, fine blood flow net- et al., 1996). Even using the optimized
work around the HCC nodule, and vessels parameters for power Doppler sonography,
within the tumor are seen only in HCC. however, the ability to detect low-velocity
This contrasts with the detour or dot pat- blood flow in very small intratumoral
tern seen in metastases or hemangioma, vessels, particularly in small HCCs and
respectively. However, on conventional hemangioma, is still limited.
color Doppler sonography, the ability to According to the previous report by Kim
evaluate low-velocity blood flow in very et al. (1998), intra- and peritumoral flow
small intratumoral vessels, in particular in signals increased in most HCCs (95%)
small hepatic lesions and lesions located after administration of contrast agent and
deep within hepatic parenchyma, is less enhanced vascular flow signals in the tumor
impressive. Attempts to improve color changed from an internal flow pattern or a
Doppler imaging led to the development of basket pattern to a prominent mixed pattern.
ultrasound contrast agents. Consequently, Contrast enhancement of the Doppler signal
contrast-enhanced color Doppler sono- in the tumor began ~ 15–20 s after injection
graphy demonstrated more intratumoral of contrast agent and reached a maximum
vascularity in HCCs than did conventional at 30–60 s. This study concluded that con-
color Doppler sonography (Ernst et al., trast-enhanced power Doppler sonography
1996; Strobel et al., 2000). Tanaka et al. was superior to unenhanced power Doppler
(1998) have also shown an improvement sonography in the demonstration and char-
in the detection rate of intratumoral vascu- acterization of tumor vascularity in nodular
larity. In a study on contrast-enhanced or massive HCC.
color Doppler sonography, the majority of
HCCs showed extensive or moderate intra-
tumoral vascularity (Strobel et al., 2000). CONTRAST-ENHANCED
HARMONIC POWER
DOPPLER SONOGRAPHY
CONTRAST-ENHANCED
POWER DOPPLER Even though contrast-enhanced power
SONOGRAPHY Doppler sonography showed a good per-
formance to depict tumor vascularity in
Unlike color Doppler sonography, which HCC, it still has several limitations related
emphasizes velocity, variance informa- to intrinsic weaknesses of power Doppler
tion, or both, power Doppler sonography technique. First, high susceptibility to tissue
is a technique based on the total integrated motion limits the application of this tech-
power of the Doppler spectrum, which is nique in patients with poor breath-holding
related to the number of red blood cells and those with hepatic masses near the heart
that produce the Doppler shift. Findings or pulsatile great vessels. Second, color
148 B.I. Choi and S.H. Kim

blooming artifact, which can degrade image sonography. Similarly, the sizes of intrahe-
quality, is frequently encountered immedi- patic and intratumoral vessels likely were
ately after bolus injection of contrast agent overestimated. Enhanced vessels adjacent
(Kim et al., 1998). In one study, the size to a liver tumor can be seen as if they are
of the vessel enhanced with contrast agent located within the tumor on conventional
frequently appeared larger than its actual power Doppler sonography. This can be an
size, even though reduction of Doppler important problem when a physician needs
gain was done until artifacts outside the to know whether any residual viable tumor
vessel nearly disappeared (Choi et al., is present after local treatment for a malig-
2000a). Third, power Doppler artifacts pro- nant tumor. In that case, harmonic power
duced from areas with high echogenicity Doppler sonography might be superior to
frequently mimic intratumoral vessels and conventional power Doppler sonography
make assessing intratumoral vascularity (Choi et al., 2000b).
difficult (Kim et al., 1999). To circumvent
such limitations, the harmonic techniques
have been introduced and merged into con- CONTRAST-ENHANCED
trast-enhanced power Doppler sonography. PULSE-INVERSION
The use of harmonic mode can effectively HARMONIC SONOGRAPHY
reduce artifacts including blooming and
motion-related artifacts. Like tissue harmonic imaging, pulse-
In our previous study, we performed a inversion harmonic imaging (PIHI) displays
prospective study to compare contrast- the amplitude of the harmonic signals result-
enhanced harmonic power Doppler sonog- ing from non-linear echoes. However, in
raphy with conventional power Doppler PIHI, two identical pulses with reversed
sonography in the assessment of tumor polarity which are transmitted down each
vascularity in HCC (Choi et al., 2000b). ray line were used. When these result-
In most cases, the intensity of enhanced ant returned waveforms are added, the
signals in the tumor and hepatic paren- harmonic component gives the multiplied
chyma in conventional mode was stronger harmonic level of a single waveform,
than that in harmonic mode (Choi et al., while all linear fundamental components
2000b). This is not surprising when consid- are canceled out more effectively than
ering the relatively low intensity of second those on tissue harmonic imaging using a
harmonic echoes compared with funda- simple bandpass filter (Hohl et al., 2004;
mental echoes (Burns, 1996). However, Jang et al., 2000). This technique, instead
harmonic mode was superior to conven- of using a narrow receiver filter tuned
tional mode in terms of power Doppler around harmonic frequency components
artifacts at any imaging time. In harmonic which is used in a simple bandpass fil-
power Doppler mode, continuous scan- tering method, allows the use of broader
ning during a patient’s quiet breathing was transmit and receive bandwidths to achieve
possible without producing considerable improved resolution. Therefore, PIHI can
motion-related artifacts. Overestimation of overcome the bandwidth limitations of
the size of contrast-enhanced vessel was filtering harmonic imaging (Hohl et al.,
evident on conventional power Doppler 2004; Jang et al., 2000).
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 149

Although PIHI produces significantly bet- motion artifacts are seen when a transducer
ter image quality for focal hepatic lesions, moves between frames.
it has several limitations that should be In contrast to power Doppler sonography,
overcome for it to replace conventional contrast-enhanced PIHI with interval delay
sonography in routine practice. Because scanning can provide strong gray-scale
multiple pulses are used, the frame rate is enhancement by microbubble contrast agents
relatively low compared with that of con- and detect signals from microbubbles in very
ventional sonography or tissue harmonic slow flow without Doppler-related arti-
imaging. Although we often found that PIHI facts (Figure 11.1). It has been shown that
improves the conspicuousness of a HCC this technique can depict the enhancement
barely recognizable on conventional US, patterns of various hepatic tumors includ-
we cannot use PIHI during biopsy because ing HCC. Kim et al. (2000) performed a
of its slow frame rate. In addition, the fact prospective study with contrast-enhanced
that multiple pulses are used to form each PIHI in various liver tumors. Hepatocellular
ray line, makes it susceptible to motion arti- carcinomas with or without irregular intra-
facts. Anything that moves between pulses tumoral enhancing vessels show findings
is not completely cancelled, so some tissue similar to those on hepatic angiography

Figure 11.1. Contrast-enhanced pulse-inversion harmonic sonography of hepatocellular carcinoma.


Gray-scale sonogram before contrast administration (left upper) shows a heterogeneous hyperechoic mass
(arrows) in the liver. Contrast-enhanced pulse-inversion harmonic sonograms obtained 33 s (right upper)
and 59 s (left lower) after injection shows heterogeneous enhancement of the tumor (arrows). A tubular
structure with intense enhancement (arrowhead) that represents an irregular intratumoral vessel is seen.
On contrast-enhanced pulse-inversion harmonic sonogram (right lower) obtained 213 s after injection
reveals mild wash-out of contrast material within the tumor (arrows). (Reproduced with permission from
Kim et al., 2000.)
150 B.I. Choi and S.H. Kim

(Figure 11.1). The pattern of enhancement (Lee et al., 2002). It combines PIHI with
on PIHI in that study was thought to be a coded excitation technique, which trans-
useful in the differential diagnosis of liver mits coded pulse sequence, decoding them
tumors (Kim et al., 2000). As compared on receipt. Coded technology uses a code
with computed tomography (CT) or mag- and decode pulse sequence system, so-
netic resonance imaging (MRI), there were called digitally encoded sonographic tech-
a few limitations of the sonographic tech- nology, to enhance the signals from the
nique used in that study. The first is that contrast agent and, at the same time, sup-
dynamic sonographic scanning is possible press unwanted background, fundamental,
in only one scanning plane; thus, it is not and harmonic signals from the tissue. This
possible to characterize all lesions simul- technique can be paired with the benefits
taneously in patients with multiple lesions. of B-flow. B-flow optimizes direct visuali-
Second, interval delay scanning in the same zation of blood cells on gray-scale imag-
area is not easy for unskilled examiners. ing. Because blood echoes are very weak,
It may be difficult even for experienced it uses codes to boost weak blood echoes
examiners to scan the lesion in the liver and equalize non-moving tissue signal.
region that is visible only during a patient’s For that reason, B-flow technique does
deep inspiration. Sufficient practice of inter- not cause Doppler artifact. Ultimately,
val delay scanning at the same area, includ- contrast-enhanced CHA can provide
ing the tumor, must be done before injection extremely high quality and detailed vas-
of the contrast agent in such cases. However, cular information without the artifacts
sonography is superior to CT or MRI in associated with contrast agent as well as
that immediate, accurate characterization preservation of wideband resolution of
of a newly detected focal hepatic lesion on harmonic signals.
sonographic examination is possible (Kim In a previous study using contrast-
et al., 2000). enhanced CHA, the typical hemodynamic
pattern of HCCs was shown as abundant
tumor vessels supplied from the periphery
CONTRAST-ENHANCED to the center of the tumor and dense paren-
CODED HARMONIC chymal tumor staining with fast washout
SONOGRAPHY (Figure 11.2) and the sensitivity and
specificity of those findings were 92.8%
In general, transmitted sonographic pulse and 92.3%, respectively (Wen et al., 2004).
is distorted in shape and loses energy Our previous study showed similar results
as it travels through tissue. It is true for that contrast-enhanced CHA was an effec-
contrast-enhanced signals as well as for tive sonographic technique in evaluating
fundamental sonic waves. To improve the the vascularity of HCCs because it has the
detectability of the enhanced microbub- advantages such as the depiction of excel-
ble signals, researchers have studied the lent dynamic enhancement of tumors, high-
behavior of microbubbles in blood flow resolution imaging of detailed intratumoral
and developed newer sonographic tech- vessels, and no sonographic artifacts (Lee et
nology. Coded harmonic angio (CHA) al., 2003) (Figure 11.2). However, despite
is one of the new imaging techniques many advantages of CHA, CHA had several
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 151

Figure 11.2. Contrast-enhanced coded harmonic sonography of hepatocellular carcinoma. Unenhanced


transverse sonogram (upper left) shows a heterogeneous hypochoic mass (arrow) in the liver. The image
obtained at 30 s (upper right) after contrast injection reveals multiple feeding vessels and intratumoral ves-
sels (arrows) within the tumor. Strong and homogeneous enhancement of the tumor is seen on the image
obtained at 50 s (lower left) after injection of the contrast agent. These enhanced flow signals within the
tumor markedly decrease and disappear at 100 s (lower right) after injection

drawbacks. First, the stationary tissue back-


technique, is a multi-pulse technique that
ground is severely suppressed. Therefore,
provides very high-resolution images,
the quality of unenhanced CHA is inferior to
which detail the effect of microbubble
conventional gray-scale images, particularly
emission, and uses the property of bubble
in a deep-seated lesion (Lee et al., 2003).
emission to dramatically show regions
Second, interval delay scanning in the same
where the contrast agent has localized.
area is not easy for unskilled examiners.
Pulse inversion harmonic imaging uses
Sufficient practice of interval delay scan-
two alternately phased pulses and adds
ning in the area that includes the tumor must
echoes from both pulses together. On the
be done before injection of contrast agent.
contrary, ADI uses two pulses with the
same polarity and subtracts the signals
from the two pulses; consequently, only
CONTRAST-ENHANCED fundamental and harmonic contrast agent
AGENT DETECTION signals remain. Therefore, ADI may be
IMAGING referred to as contrast-only imaging. As
a result, ADI may depict signals from
Agent detection imaging (ADI), a most microbubbles better than those from PIHI.
recently developed high-MI harmonic When an intermittent imaging strategy is
152 B.I. Choi and S.H. Kim

used with ADI, it may effectively reveal 2005b; Youk et al., 2003a). According to
the vascularity of focal liver lesions and our recent report, contrast-enhanced ADI
may also help in lesion characterization. sonography can be used to characterize
Furthermore, ADI may separate the tis- focal hepatic lesions with a higher diag-
sue image from the contrast-only image nostic confidence than baseline sonogra-
(Figure 11.3). Because the tissue image phy and furthermore, it reduced the need
and contrast-only image can be shown sep- for further diagnostic workups for focal
arately, the positional relationship between hepatic lesion characterization (Kim et al.,
the tumor and other structures can be accu- 2005b) (Figure 11.3). Our another recent
rately assessed (Figure 11.3). report showed that single-level dynamic
Several reports have suggested that sonography with ADI was potentially
contrast-enhanced ADI sonography has useful in diagnosing HCC, with excellent
improved the detection and characteriza- visibility of intra- and peri-tumoral vascu-
tion of focal liver lesions (Kim et al., larity, early tumor staining, and sometimes

Figure 11.3. Contrast-enhanced agent detection imaging of hepatocellular carcinoma. Images obtained
at 20 s (upper) and 30 s (lower) after injection of the contrast agent shows a small hepatocellular carci-
noma (arrows) with hyperechoic contrast enhancement compared with hepatic parenchyma. Comparison
of combined (left), contrast (middle), and tissue (right) modes of display enables precise location of the
distribution of the microbubble; therefore, the hypervascularity of the tumor during the arterial phase can
be assessed confidently
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 153

delayed capsular enhancement (Kim et al., contrast-enhanced perfusional sonography


2003). for HCC will be introduced below.

LOW MECHANICAL INDEX CONTRAST-ENHANCED


IMAGING SONOGRAPHY
Until recently, visualization of flow in IN ASSESSING THE
the microcirculation has required some THERAPEUTIC RESPONSE
form of triggering or low-frame rate imag- OF HEPATOCELLULAR
ing. To detect the bubbles, the ultrasound CARCINOMAS
had to be strong enough to destroy them.
However, the increased sensitivity pro- Sonography or CT is most often used as
vided by newer imaging techniques makes the primary guidance technique in local
it possible to image contrast microbubbles ablation therapy such as percutaneous
relatively nondestructively in real time at ethanol injection (PEI) and radiofrequency
very low acoustic pressures. Therefore, ablation (RFA) for HCC. Of these modali-
low MI scanning technique makes it ties, gray-scale sonography is preferred
possible to perform contrast-enhanced because it is more widely available, less
perfusional sonography for various hepatic expensive, and allows real-time monitor-
lesions (Schneider et al., 1995; Schneider, ing, which facilitates placement of the
1999; von Herbay et al., 2004). electrode needle. However, it is of little
Low MI scanning is important for two help in determining the completeness of
reasons. First, at a low MI, bubble destruc- the procedure or in long-term follow-up
tion is avoided. Although microbubbles of therapeutic efficacy (Lencioni et al.,
differ in their shell composition, previous 1995). Postablation gray-scale sonogra-
work indicates that at an MI of ~ 0.15, the phy cannot help differentiate viable tumor
microbubbles examined are not destroyed from necrotic tissue because of variable
markedly, yet give a good harmonic con- echogenicity and because postablation
trast signal. The second major reason for sonographic findings do not correlate well
low MI scanning is the reduction of the with the overall shape and volume of the
harmonic component in the tissue echoes necrosis (Goldberg et al., 2000). Although
relative to bubble echoes. Although tissue several reports have shown that color
harmonics have benefited routine diagnos- Doppler sonography is somewhat useful
tic scanning, it is the background “noise” in the assessment of therapeutic efficacy
signal above which the contrast signal must after local ablation therapy for HCC, nei-
rise. Because tissue is less nonlinear than ther color nor power Doppler sonography
bubbles, it requires a higher MI than the has proved entirely satisfactory because of
contrast microbubbles for a certain har- low sensitivity to microvasculature within
monic response. Therefore, at a low MI, the residual viable tumor (Choi et al., 2000b;
contrast-to-tissue ratio is higher than at a Rossi et al., 1996).
high MI, helping to remove the tissue signal Because blood pool microbubble con-
and leaving only the contrast. The details of trast agents have become available for use
154 B.I. Choi and S.H. Kim

with Doppler sonography, there have been tic response to RF ablation therapy (Choi
a lot of researches underway in the use- et al., 2003; Kim et al., 2005b) (Figures
fulness of contrast-enhanced sonography 11.5 and 11.6). This technique allows
for assessing the therapeutic responses of detection of residual tumor after ablation
HCC after treatment. Several reports have without the blooming and motion artifacts
suggested that contrast-enhanced color and seen at contrast-enhanced color or power
power Doppler sonography can be useful Doppler sonography. Another advantage
in detecting residual or recurrent tumors of contrast-enhanced gray-scale harmonic
after local ablation of HCC (Goldberg sonography over CT and MR imaging
et al., 2000) (Figure 11.4). According to is that it can directly show persistent
a preliminary report, contrast-enhanced fine vascular flow signals within the
power Doppler sonography can be used tumor during a period of effective con-
as an alternative to immediate follow-up trast enhancement that lasts for more than
CT for early assessment of the therapeu- 10 min. Consequently, contrast-enhanced
tic efficacy of RF ablation of HCC (Choi gray-scale harmonic sonography could be
et al., 2000b) (Figure 11.4). helpful in detecting residual or recurrent
More recently, contrast-enhanced gray- tumor after local ablation of HCC. These
scale harmonic sonography has been modalities may also be helpful in identify-
developed for the evaluation of therapeu- ing residual tumor when performed during

Figure 11.4. Successful radiofrequency ablation of hepatocellular carcinoma. Unenhanced, transverse,


gray-scale (upper left) and power Doppler (upper right) sonograms before radiofrequency ablation (RFA)
demonstrate a large heterogeneous mass (arrows) with hypervascularity in right hepatic lobe. Transverse
gray-scale (lower left) and contrast-enhanced power Doppler (lower right) sonograms obtained 1 day after
RFA show no flow signals within the ablated area (arrows), a finding that is consistent with complete
tumor necrosis
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 155

Figure 11.5. Contrast-enhanced coded harmonic sonography for evaluation of local recurrence after
radiofrequency ablation (RFA) of HCC. Unenhanced, oblique coronal sonogram (upper left) obtained 1
year after RFA shows a heterogeneous high echoic nodule (arrow) in right lobe of the liver. On contrast-
enhanced coded harmonic sonogram (upper middle) obtained 42 s after injection of the contrast agent, a
nodular enhancing focus (arrow) is noted in the anterior side of the lesion. This represents the recurred
tumor. During RFA (upper right), a large echogenic lesion (arrow) suggesting ablated zone is seen. Portal
phase contrast CT scan (lower left) before second RFA shows a nodular enhancing focus (arrow) of
the residual tumor. On CT scan (lower right) obtained immediate after second RFA, the ablated lesion
remains unenhanced, which indicates a successful RFA

repeat ablation, when accurate localization to the risk of toxicity. However, real-time
of viable tumor is needed. This real-time sonography and the ability to administer
confirmation of the accurate localization repeated doses of sonographic contrast
of viable tumor and of correct placement material may, in some cases, allow better
of the electrode within the tumor cannot detection of residual tumor.
be expected at CT, and may reduce the In conclusion, many recent advances
number of treatment sessions without fur- in contrast-enhanced sonography imag-
ther use of contrast-enhanced CT or MR ing have occurred, making it a strong
imaging. Furthermore, contrast-enhanced contender for several problem-solving
CT is less than ideal because errors in applications. Admittedly, CT and MR
contrast bolus timing cannot always be cor- imaging continue to have complimentary
rected immediately with repeat scanning due roles. However, sonographic examinations
156 B.I. Choi and S.H. Kim

Figure 11.6. Contrast-enhanced agent detection imaging for evaluation of therapeutic response after
transarterial chemoembolization (TACE) of HCC. Unenhanced, sagittal sonogram (upper left) shows a
1.5 cm-size, hypoechoic nodule (arrow) in the left hepatic lobe. Contrast-enhanced sonograms obtained
at 30 s (upper right), 40 s (lower left), and 120 s (lower right) after injection of the contrast agent reveal
focal enhancing portion (arrows) that represents residual viable tumor. The residual tumor was treated
with second TACE later on the same day

with reproducibly excellent quality are in the depiction of vasculature in liver tumors.
expected with the aid of newer imaging Radiology 200:55–58.
Choi, B.I., Kim, T.K., Han, J.K., Kim, A.Y., Seong,
techniques combined with the use of more
C.K., and Park, S.J. 2000a. Vascularity of hepa-
stable, second generation contrast agents. tocellular carcinoma: assessment with contrast-
Contrast-enhanced sonography will con- enhanced second-harmonic versus conventional
tinue to have a major role in liver imaging power Doppler US. Radiology 214:381–386.
into the foreseeable future. Choi, D., Lim, H.K., Kim, S.H., Lee, W.J., Jang,
H.J., Lee, J.Y., Paik, S.W., Koh, K.C., and Lee,
J.H. 2000b. Hepatocellular carcinoma treated
REFERENCES
with percutaneous radio-frequency ablation: use-
Bartolozzi, C., Lencioni, R., Ricci, P., Paolicchi, A., fulness of power Doppler US with a microbubble
Rossi, P., and Passariello, R. 1998. Hepatocellular contrast agent in evaluating therapeutic response-
carcinoma treatment with percutaneous etha- preliminary results. Radiology 217:558–563.
nol injection: evaluation with contrast-enhanced Choi, D., Lim, H.K., Lee, W.J., Kim, S.H., Kim,
color Doppler US. Radiology 209:387–393. Y.H., Kim, S.H., and Lim, J.H. 2003. Early
Burns, P.N. 1996. Harmonic imaging with ultrasound assessment of the therapeutic response to radio
contrast agents. Clin. Radiol. 51: Suppl. 1:50–55. frequency ablation for hepatocellular carcinoma:
Choi, B.I., Kim, T.K., Han, J.K., Chung, J.W., Park, utility of gray scale harmonic ultrasonography
J.H., and Han, M.C. 1996. Power versus conven- with a microbubble contrast agent. J. Ultrasound
tional color Doppler sonography: comparison Med. 22:1163–1172.
11. Hepatocellular Carcinoma: Contrast-Enhanced Sonography 157

Dill-Macky, M.J., Burns, P.N., Khalili, K., and utility of contrast-enhanced agent detection
Wilson, S.R. 2002. Focal hepatic masses: imaging. Eur. J. Radiol. 56:66–73.
enhancement patterns with SH U 508A and Kim, K.W., Choi, B.I., Park, S.H., Kim, H.C.,
pulse-inversion US. Radiology 222:95–102. Lee, M.W., Kim, S.H., Lee, K.H., Park, C.H.,
Ding, H., Kudo, M., Onda, H., Suetomi, Y., Kim, J.S., Won, H.J., and Han, J.K. 2003.
Minami, Y., Chung, H., Kawasaki, T., and Hepatocellular carcinoma: assessment of vascu-
Maekawa, K. 2001. Evaluation of posttreatment larity with single-level dynamic ultrasonography
response of hepatocellular carcinoma with con- during the arterial phase. J. Ultrasound Med.
trast-enhanced coded phase-inversion harmonic 22:887–896.
US: comparison with dynamic CT. Radiology Kim, S.H., Lee, J.M., Lee, J.Y., Han, J.K., An,
221:721–730. S.K., Han, C.J., Lee, K.H., Hwang, S.S., and
Dodd, G.D. III, Miller, W.J., Baron, R.L., Skolnick, Choi, B.I. 2005b. Value of contrast-enhanced
M.L., and Campbell, W.L. 1992. Detection of sonography for the characterization of focal
malignant tumors in end-stage cirrhotic livers: hepatic lesions in patients with diffuse liver dis-
efficacy of sonography as a screening technique. ease: receiver operating characteristic analysis.
AJR Am. J. Roentgenol. 159:727–733. AJR Am. J. Roentgenol. 184:1077–1084.
Ernst, H., Hahn, E.G., Balzer, T., Schlief, R., and Kim, T.K., Han, J.K., Kim, A.Y., Park, S.J., and
Heyder, N. 1996. Color Doppler ultrasound of Choi, B.I. 1999. Signal from hepatic hemangi-
liver lesions: signal enhancement after intrave- omas on power Doppler US: real or artefactual?
nous injection of the ultrasound contrast agent Ultrasound Med. Biol. 25:1055–1061.
Levovist. J. Clin. Ultrasound 24:31–35. Kim, T.K., Choi, B.I., Han, J.K., Hong, H.S.,
Goldberg, S.N., Gazelle, G.S., Compton, C.C., Park, S.H., and Moon, S.G. 2000. Hepatic
Mueller, P.R., and Tanabe, K.K. 2000. Treatment tumors: contrast agent-enhancement patterns
of intrahepatic malignancy with radiofrequency with pulse-inversion harmonic US. Radiology
ablation: radiologic-pathologic correlation. 216:411–417.
Cancer 88:2452–2463. Lee, J.Y., Choi, B.I., Han, J.K., Kim, A.Y., Shin,
Gramiak, R., and Shah, P.M. 1968. Echocardiography S.H., and Moon, S.G. 2002. Improved sono-
of the aortic root. Invest. Radiol. 3:356–366. graphic imaging of hepatic hemangioma with
Hohl, C., Schmidt, T., Haage, P., Honnef, D., contrast-enhanced coded harmonic angiography:
Blaum, M., Staatz, G., and Guenther, R.W. 2004. comparison with MR imaging. Ultrasound Med.
Phase-inversion tissue harmonic imaging com- Biol. 28:287–295.
pared with conventional B-mode ultrasound in Lee, J.Y., Choi, B.I., Han, J.K., Kim, A.Y., Shin,
the evaluation of pancreatic lesions. Eur. Radiol. S.H., and Park, C.M. 2003. Assessment of
14:1109–1117. hepatocellular carcinoma vascularity: compari-
Jang, H.J., Lim, H.K., Lee, W.J., Kim, S.H., Kim, son of contrast-enhanced coded harmonic ultra-
K.A., and Kim, E.Y. 2000. Ultrasonographic sound with harmonic power Doppler ultrasound.
evaluation of focal hepatic lesions: comparison J. Med. Ultrasound 11:147–155.
of pulse inversion harmonic, tissue harmonic, and Lencioni, R., Caramella, D., and Bartolozzi, C.
conventional imaging techniques. J. Ultrasound 1995. Hepatocellular carcinoma: use of color
Med. 19:293–299. Doppler US to evaluate response to treatment
Kim, A.Y., Choi, B.I., Kim, T.K., Han, J.K., with percutaneous ethanol injection. Radiology
Yun, E.J., Lee, K.Y., and Han, M.C. 1998. 194:113–118.
Hepatocellular carcinoma: power Doppler Lencioni, R., Pinto, F., Armillotta, N., and
US with a contrast agent–preliminary results. Bartolozzi, C. 1996. Assessment of tumor vascu-
Radiology 209:135–140. larity in hepatocellular carcinoma: comparison
Kim, C.K., Choi, D., Lim, H.K., Kim, S.H., Lee, of power Doppler US and color Doppler US.
W.J., Kim, M.J., Lee, J.Y., Jeon, Y.H., Lee, J., Radiology 201:353–358.
Lee, S.J., and Lim, J.H. 2005a. Therapeutic Meloni, M.F., Goldberg, S.N., Livraghi, T., Calliada,
response assessment of percutaneous radiofre- F., Ricci, P., Rossi, M., Pallavicini, D., and
quency ablation for hepatocellular carcinoma: Campani, R. 2001. Hepatocellular carcinoma
158 B.I. Choi and S.H. Kim

treated with radiofrequency ablation: compari- contrast-enhanced color Doppler sonography in


son of pulse inversion contrast-enhanced har- the differential diagnosis of liver tumors. J. Clin.
monic sonography, contrast-enhanced power Ultrasound 28:1–13.
Doppler sonography, and helical CT. AJR Am. J. Tanaka, S., Kitamra, T., Fujita, M., and Yoshioka, F.
Roentgenol. 177:375–380. 1998. Value of contrast-enhanced color Doppler
Porter, T.R., Xie, F., Kricsfeld, D., and Armbruster, sonography in diagnosing hepatocellular carci-
R.W. 1996. Improved myocardial contrast with noma with special attention to the “color-filled
second harmonic transient ultrasound response pattern”. J. Clin. Ultrasound 26:207–212.
imaging in humans using intravenous perfluor- Tanaka, S., Kitamura, T., Fujita, M., Nakanishi,
ocarbon-exposed sonicated dextrose albumin. J. K., and Okuda, S. 1990. Color Doppler flow
Am. Coll. Cardiol. 27:1497–1501. imaging of liver tumors. AJR Am. J. Roentgenol.
Rossi, S., Di, S.M., Buscarini, E., Quaretti, P., 154:509–514.
Garbagnati, F., Squassante, L., Paties, C.T., von Herbay, A., Vogt, C., Willers, R., and
Silverman, D.E., and Buscarini, L. 1996. Haussinger, D. 2004. Real-time imaging with
Percutaneous RF interstitial thermal ablation the sonographic contrast agent SonoVue: differ-
in the treatment of hepatic cancer. AJR Am. J. entiation between benign and malignant hepatic
Roentgenol. 167:759–768. lesions. J. Ultrasound Med. 23:1557–1568.
Schneider, M. 1999. SonoVue, a new ultrasound con- Wen, Y.L., Kudo, M., Zheng, R.Q., Ding, H., Zhou,
trast agent. Eur. Radiol. 9: Suppl. 3:S347–S348. P., Minami, Y., Chung, H., Kitano, M., Kawasaki,
Schneider, M., Arditi, M., Barrau, M.B., Brochot, T., and Maekawa, K. 2004. Characterization
J., Broillet, A., Ventrone, R., and Yan, F. 1995. of hepatic tumors: value of contrast-enhanced
BR1: a new ultrasonographic contrast agent coded phase-inversion harmonic angio. AJR Am.
based on sulfur hexafluoride-filled microbub- J. Roentgenol. 182:1019–1026.
bles. Invest. Radiol. 30:451–457. Youk, J.H., Kim, C.S., and Lee, J.M. 2003a.
Solbiati, L., Goldberg, S.N., Ierace, T., Dellanoce, Contrast-enhanced agent detection imaging:
M., Livraghi, T., and Gazelle, G.S.1999. Radio- value in the characterization of focal hepatic
frequency ablation of hepatic metastases: post- lesions. J. Ultrasound Med. 22:897–910.
procedural assessment with a US microbubble Youk, J.H., Lee, J.M., and Kim, C.S. 2003b.
contrast agent–early experience. Radiology Therapeutic response evaluation of malignant
211:643–649. hepatic masses treated by interventional proce-
Strobel, D., Krodel, U., Martus, P., Hahn, E.G., dures with contrast-enhanced agent detection
and Becker, D. 2000. Clinical evaluation of imaging. J. Ultrasound Med. 22:911–920.
12
Focal Liver Lesion: Nonlinear
Contrast-Enhanced Ultrasound Imaging
Vincenzo Migaleddu and Giuseppe Virgilio

INTRODUCTION lesions may be detected that are some-


times missed with CT or MR imaging.
Ultrasound (US) is the most widespread Positron emission tomography (PET)
imaging modality for screening focal liver is proving to be a molecular imaging
lesions. Noninvasive diagnosis (detection modality with very high sensitivity, but
and characterization) of focal liver lesions not competitive as the advantages do not
is still carried out in many centers by com- outweigh the high cost of the exam.
puterized tomography (CT) and magnetic Indeed, color Doppler is a method that
resonance (MR) using contrast agents. can separate echoes from blood and tissue.
Hepatic lesions have in effect charac- This is possible because of the relatively
teristic perfusion and enhancement pat- high velocity of blood in comparison to
terns during the various vascular phases, that of the surrounding tissue. This dis-
which assist characterization and in many tinction is valid for flow in large vessels
cases make definite diagnosis possible. where the Doppler signals of blood flow
Conventional and Doppler US are often may be separated from those of the tissue
limited in diagnosing this kind of lesion as clutter using a wall filter. This fails for
color Doppler vascular information of the the parenchymal micro-vessel flow where
lesion is limited (see chapter 11). blood velocity is similar or slower than
Recently, the detection and characteri- that in tissue. In fact in the latter case the
zation of focal hepatic lesions by US has Doppler shift frequency coming from the
increased considerably due to the devel- moving tissue is comparable or higher
opment of new nonlinear (harmonic and (up to a thousand times) than that of the
nonlinear fundamental) technologies that blood which perfuses it. Thus, when we
optimize detection of contrast agents. analyze together tissue and the blood in
Furthermore, small lesions may not be it, wall filters eliminate both the flow
well defined by CT or MR imaging and the clutter echoes. “Flash” artifact
because real-time visualization of lesion in color and “thump” artifact in spectral
vascularization is not possible. Because Doppler represent overwhelming signals
contrast-enhanced US (CEUS) is a from tissue movements which corrupt
dynamic analysis that allows this, small image information.

159
160 V. Migaleddu and G. Virgilio

Ultrasound contrast media are sus- US beam, depends on the density of the
pensions containing air or gas micro- tissue. The compression part of the US
bubbles that increase reflection of the wave increases by fractions the density of
US beam used during the investigation. the tissue, and thus this part of the wave
Contrast agents were previously used as travels marginally faster than the rarefac-
echo-enhancers as they amplified blood tion part of the wave where the density is
reflectivity and, therefore, increased the lower. Thus, over distance, the shape of
signal-to-noise ratio for Doppler signals the waveform becomes distorted and its
amplifying blood reflectivity and increas- angular components represent the over-
ing the signal-to-noise ratio. This enhances tones or harmonics. These can be filtered
the echogenicity of the blood but creates and used for imaging (Ward et al., 1997).
more artifacts due to blooming and tissue The advantage is that they depend on a
motion (Powers et al., 1997). Another, high acoustic pressure for their formation,
more efficient way to avoid artifacts would and therefore are not produced as much
be to obtain a contrast B-mode image by the weaker unwanted side and grat-
that suppresses Doppler artifacts without ing lobes or by reverberations than by the
the use of a velocity-dependent filter. main lobe. Thus, tissue harmonic images
Contrast- enhanced ultrasound, defined as are cleaner with fewer side lobe and rever-
nonlinear or harmonic imaging, aims to beration artifacts.
provide such a method. The harmonics are generated from within
the tissue and only at the center portion
of the main US beam. This results in higher
HARMONIC IMAGING: tissue contrast. The disadvantage is that
BASIC PRINCIPLES the harmonic echoes are some 20 dB less
intense than the fundamental echoes with
Harmonics are additional frequencies that the risk of reducing the signal-to-noise
are multiples of the fundamental (transmit- ratio. The disadvantage is that the harmonic
ted) frequency and are commonly found in echoes are some 20 dB less intense than
acoustics. Double and fourfold overtones fundamental echoes with a corresponding
(harmonics) give character to the different reduction of the signal-to-noise ratio. This
musical instruments. The same pheno- is why harmonics are better handled by
menon occurs in ultrasound (Hamilton and newer generation systems powered with a
Blackstock, 1998). We need to distinguish high dynamic range than those with older
two different modes of harmonic gene- technology-designed scanners. When it
ration: those produced by transmission works well, harmonic imaging is spec-
through the tissue and those produced tacularly useful in sharpening up images
by interaction with microbubbles. Tissue since it increases contrast by eliminating
harmonics are derived from the nonlin- noise, especially in the evaluation of the
ear distortion of the incident beam as it fluid-filled structures such as the biliary
passes through the tissues. The pheno- tree, gallbladder and vascular structures
menon occurs for US beams of a sufficiently (Ortega et al., 2001). In clinical practice,
high power. A well-known basic principle for most scanners the benefit in abdomi-
of US, the speed of propagation of an nal and general work is so great that most
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 161

users now set the systems to default in a cardiac evaluations, are effectively one-
tissue harmonic mode; however, in our pass-only agents and have been replaced
opinion, harmonic mode is more useful by new-generation agents such as Optison,
in the evaluation of fluid-filled structures Levovist and SonoVue.
positioned in the middle field because For radiology, the two most important
a penetration loss occurs in the far field are Levovist (Schering) and SonoVue
with the poor echo intensity of deeper (Bracco). Levovist is a first-generation
tissue second harmonics (Migaleddu contrast medium of air containing micro-
et al., 2002). bubbles made by shaking galactose micro-
Microbubble nonlinear (harmonic) particles with water. The galactose
responses can be produced in two dif- micro-particles contain micro-defects,
ferent ways: microbubble oscillations or which force the attached air microbubbles
disruption. Microbubbles are air or gas into the required size. They are stabilized
bodies ranging from 1 to 7 μm in diameter, by a monomolecular layer of a surfactant,
enclosed by some sort of membrane. Two palmitic acid (Nanda and Carstensen,
important characteristics of microbubbles, 1997). The second-generation contrast
stability and diameter, must be optimized medium SonoVue uses an inert high-
in order to have efficient contrast media. density gas (sulfur hexafluoride), which
Stability means that microbubbles of con- improves the longevity of the microbub-
trast medium must dwell in the blood bles because of its high molecular weight,
stream long enough to allow the comple- which slows diffusion. Microbubble shell
tion of an US investigation with contrast is constituted by a phospholipid layer
enhancement. Microbubbles must also be which is similar to that of cell membranes
of a diameter necessary to obtain contrast (Schneider et al., 1995; Morel et al.,
medium that can pass through the pul- 2000). The essence of their function
monary circulation and that, once in the as contrast agents is that microbubbles
systemic circulation, can act as a contrast- behave quite differently from solid or
enhancing medium in all organs (http:// watery tissues in that they can be comp-
www.smirg.org/.contrast.php). Several ressed and expanded much more readily.
cardiac and vascular US contrast agents Thus, they change their diameter from
are available on the American, Canadian, two to tenfold, depending on the power
European or worldwide market; Echovist of the incident US and, like all reac-
and Levovist (Schering), SonoVue (Bracco) tive processes, have a natural resonant
are available in a number of European frequency at which they respond most
countries. Other agents that underwent or actively. It happens that the resonant fre-
are still undergoing American regulatory quency for 1–7 μm microbubbles lies in
approval include Optison (Amersham/GE the 2–10 MHz range that is used for diag-
Healthcare), Definity (BMS), Imagent nostic imaging. This lucky coincidence
(Alliance Pharmaceutical), Sonazoid explains the remarkable reflectivity of
(Amersham/GE Healthcare), Quantison microbubbles which are many more times
(Quadrant Healthcare), SonoVue (Bracco), echogenic than comparable tissue elements
Biosphere (Ponit Biomedical), and AI-700. such as red blood cells (Cosgrove in
Albunex and Echovist, used mainly in http://www.smirg.org/lectures.php).
162 V. Migaleddu and G. Virgilio

PHYSICAL AND in limited stability and shortened time


TECHNOLOGICAL available for examination. They behave
BACKGROUND like soap bubbles and the greatest amount
of high intensity information in response is
When an US beam encounters microbub- obtained the moment they break. Second-
bles present in an insonated field, different generation contrast agents (SonoVue) are
information is acquired depending on the made up of microbubbles with notable
pressure exerted by the US waves, which elasticity, containing a gas (sulfur hex-
in turn is related to the mechanical index afluoride or similar), and are wrapped
(MI). The mechanical index is the tool in a shell of phospholipids and/or albu-
which expresses the acoustic pressure of min. This enables real-time continuous-
the US beam produced on insonated struc- mode software to be used, exploiting the
tures; it enables the power of the US beam information from the resonance of the
to be quantified and is directly propor- microbubbles that are maintained in time,
tionate to the peak of maximum negative though their duration is not unlimited and
pressure at the point of focalization of the they break spontaneously after 8–10 min.
beam (P), and is inversely proportionate to Microbubbles like any gas particle resist
the mean frequency of the US beam (V) in compression more than they respond to
the following expansion so that they may respond asym-
P metrically to symmetrically transmitted
ratio: M.I. = US waves. This asymmetry may take
V either the form of a change in the bubbles’
This quantity is related to the mechanical shape whereby they become geometrically
work that can be carried out on the micro- distorted or of an asymmetry in the temporal
bubble during the negative cycle of US response. The latter is perhaps more usual
propagation (Apfel and Holland, 1991). because the bubbles expand more quickly
With very low mechanical indexes reflec- than they compress. In either case the result
tion of ultrasounds occurs. For slightly is that the returning wave contains differ-
higher mechanical indexes (up to 0.2) a ent frequencies from those of the insonant
nonlinear signal is emitted in relation to wave and if these can be extracted, images
the resonance of the microbubbles that favoring the contribution of the microbub-
are not destroyed, and with a mechanical bles can be created (Greis, 2004).
index greater than 1.2 the microbubbles A variety of algorithms that have been
break. This creates transitory echoes in marketed involve enhancement of the sig-
second harmonics with a high signal inten- nal coming from the microbubbles and
sity (Chin and Burns, 1997). cancellation of signals from tissues. They
First-generation contrast agents exploit favor a reading of the echoes with specific
the high MI and corresponding microbub- frequency of contrast agent resonance in
bles rupture by using intermittent software second harmonics, and at the same time
imaging. Fragile microbubbles that con- cancel the fundamental echoes coming
tain air and have a galactose shell stabi- from the tissues. In pulse inversion, also
lized by palmitic acid (such as Levovist known as phase inversion, two pulses in
or first-generation contrast agent) result rapid succession are emitted into the tissue;
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 163

the second pulse undergoes a 180° phase signal intensity that is greater than that of
change so to be a mirror image of the first. the second harmonics alone of the contrast
The fundamental response produced in agent. The identification of this nonlinear
the tissues which will be phase-inverted response requires the use of an algorithm
compared with the fundamental pulses is capable of modulating the pulses so that
erased because the sum of two inverted fundamental responses are elicited from
pulse is zero. In this way the echoes from the tissue and from the contrast agent.
the microbubbles are enhanced so as to Such an algorithm requires perfect pulse
differentiate them from those coming from modulation both in phase and in ampli-
stationary tissues (Burns et al., 2000). tude. It has been marketed with the name
A great spatial and details resolution (capa- cadence Contrast Pulse Sequencing (CPS
bility to identify macro and micro vessels) by Acuson Sequoia, Siemens) and it works
is achieved by intermittent imaging and spe- in real-time with continuous observation,
cific software – Agent Detection Imaging using second-generation contrast agents.
(ADI by Acuson Sequoia, Siemens) – with Previous algorithms exploit only the non-
a high MI and first-generation contrast linear harmonic response of contrast agent.
agent. The ADI can be used for a short These require cancellation of the funda-
time either with static techniques or in mental echoes coming from the tissue
real-time (Migaleddu et al., 2004). forcing examination to be done blindly
Compared with the intermittent imaging (without an image) until the contrast agent
characteristic of the reconstruction algo- arrives. Contrast Pulse Sequencing software
rithms used in breaking first-generation has the immediate advantage of enabling
contrast agents, the algorithms based on observation of the fundamental signals
the use of a low mechanical index enable coming from the tissue at the same time
signals from the resonance of second- as signals coming from the nonlinear
generation microbubbles to be detected fundamental response of the contrast agent
in real-time; this permits enhancement (Phillips and Gardner, 2004).
of the contrast resolution of the vascular
phenomena present in an organ or in the
lesions it contains. These methods, however, NONLINEAR CONTRAST
show low spatial and details resolution,
useful for detecting peri- and intra-lesional
ENHACEMENT OF FOCAL
macro- and micro-circulation due to the LIVER LESIONS
low intensity of signal coming from micro-
(A) Characterization of Focal Liver
bubble resonance.
Lesions
The observation that there is also a non-
linear response coming from the contrast Due to the dual blood supply of the liver
agent in the proximity of the fundamental tissue by the hepatic artery (25–30%) and
frequency has enabled a new algorithm the portal vein (70–75%), three vascular
to be developed, which permits the non- phases can be defined in all the contrast
linear fundamental echo to be detected. imaging modalities and even in CEUS.
The advantage of detecting this nonlinear The arterial phase starts from 10–20 s after
fundamental component is an increase in contrast administration into a peripheral
164 V. Migaleddu and G. Virgilio

vein and persists for ∼10–15 s; followed by related to the perfusion behavior of the
the portal vein phase which usually lasts lesions which shows the evolution of lesion
2 min after i.v. contrast injection. The last vascular volume over time.
phase (parenchymal or sinusoidal) persists
until clearance of the contrast medium Enhancement Patterns of Benign Focal
from the hepatic parenchyma. It has two Lesions
different lengths: 4–6 min for SonoVue
Hemangioma
and 15–20 min for Levovist, which sug-
gests several mechanisms including sinu- Hemangioma is the most frequent solid
soid pooling and RES/Kupffer’s cells focal lesion in the liver from 1% to
uptake. This differs from the extracellular 20% in the general population (Ros
equilibrium phase observed in contrast et al., 2001). These can be divided into
agent CT and MR imaging (EFSUMB two different types: capillary and cavern-
Study Group. Guidelines for the Use of ous hemangioma. The first are relatively
Contrast Agents in Ultrasound 2004). In small networks of vascular spaces
our opinion the evaluation criteria of the (< 3 cm). Some of them can show an
US and CEUS examinations should be early hypervascular pattern in the arterial
specified; meaning that the terms Hypo-, phase due to concentrated arterial micro-
Iso- and Hyper-echogenic should be used vessels – high-flow hemangioma (Jang
in the description of the conventional US et al., 1998). These are generally charac-
examinations; basket patterns, peripheral terized by the presence of small arterial
or central, regular or irregular vessels for branches, venous lakes and fibrous areas,
color Doppler examinations. and absence of capsule. The latter, cav-
Terms to be used for CEUS should be: ernous hemangiomas (> 3 cm) and giant
hemangiomas (> 10 cm) may show an
1. Type of enhancement: Rim, Marginal, inhomogeneous structure composed of
Nodular; Centripetal, Centrifugal filling venous lakes, dense connective tissue and
2. Degree of enhancement or enhancing: thrombosed areas (Unal et al., 2002).
Hypo-iso- and Hyper-enhancement or Arterial portal micro-fistulas can be
enhancing – Lower, Equal or Higher identified with CEUS (http://www.smirg.
than surrounding parenchymal it/research/avi-shunt.php?sl=13). On con-
3. Time of washout of contrast in the differ- ventional US, hemangiomas are often inci-
ent phases – portal and parenchymal dental findings since they are asymptomatic
4. Lack of enhancement in the different except for the relatively rare giant type.
phases – arterial portal and parenchymal Conventional US capillary hemangiomas
5. Sustained enhancement in the different have a typical homogeneous hyper-echoic
phases – arterial portal and parenchymal appearance in comparison with surround-
Hypo-, Iso- and Hyper-echoic terms might ing hepatic parenchyma with well defined
be useful to describe static phenomena margins; in a fatty liver it may be found as
underlying the conventional US imaging a hypo-echoic lesion. Cavernous heman-
but are ambiguous for explaining dynamic giomas can present an inhomogeneous
phenomena underlying the CEUS imaging. aspect with hyper-echoic and hypo-echoic
It might be appropriate to uniform terms areas (Moody and Wilson, 1993). In all
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 165

cases conventional US findings cannot be it is smaller than 5 cm (Ros et al., 2001).


considered specific particularly in onco- On conventional US iso-, hypo-, hyper-
logic and cirrhotic patients. Intralesional echoic appearances may be found. A
blood flow Doppler signals may be identi- hypo-echoic stellate central scar can be
fied in high-flow capillary hemangioma identified in 18% of cases. Color Doppler
(Tanaka et al., 1990). can depict flow signals within the central
Contrast-enhanced ultrasound can show and radiating vessels (Tanaka et al., 1990).
several appearances (Bleuzen and Tranquart, Contrast-enhanced ultrasound may show
2004; Catalano et al., 2005). In fact cont- typical and atypical features as described
rast material uptake may be fast or slow below (Migaleddu et al., 2004; Catalano
depending on intralesional circulation speed. et al., 2005).
Typical and atypical features are summa-
Typical features:
rized as follows:
Arterial phase: hyper-enhancing complete
Typical features: Portal vein phase: hyper-enhancing
Arterial phase: peripheral-nodular Parenchymal phase: iso- hyper-enhancing
enhancement, no central enhancement, rim Additional features:
enhancement Arterial phase: early “central spider” or
Portal vein phase: partial/complete cen- “spoke wheel” arteries and centrifugal fill-
tripetal filling ing, feeding artery
Parenchymal phase: complete enhance- Portal vein phase: hypo-enhanced cen-
ment tral scar
Atypical features: Parenchymal phase: hypo-enhanced cen-
Arterial phase: (small lesions) complete tral scar.
rapid centripetal enhancement; arterial–
portal fistula and arterial inflow vessel. Fatty Liver
Parenchymal phase: non enhanced cen- Hepatic steatosis is due to decreased liver
tral areas (partial thrombosis, fibrosis). clearance of fatty acids. It is caused by
hepatocellular injury and increased pro-
Focal Nodular Hyperplasia duction or mobilization of fatty acids.
Focal nodular hyperplasia is the second Alcohol, diabetes, obesity, and drugs are
most common benign solid hepatic focal the most common etiologies. Fatty changes
lesion. It occurs more commonly in young may present several morphologic and his-
women than men (two–four females to tological appearances according to diffuse
one male) in the third and fifth decade or focal involvement. A pseudo-nodular
(Volk et al., 2001), and is a hypervascular pattern might lead to a questionable diag-
hyperplasic lesion caused by preexisting nosis particularly in oncologic patients.
vascular malformation. It is character- Also spared areas can show ambiguous US
ized by abnormally arranged hepatocytes, findings (Kroncke et al., 2000).
Kupffer’s cells, bile duct elements and Focal fatty sparing:
vascularized fibrous septae. A peripheral In diffuse fatty liver focal spared areas are
pseudo capsule and a central or eccentric frequently encountered; these are typi-
fibrous scar radiating to the periphery and cally located along the liver hilum or
containing arteries can be found. Usually around the gallbladder. On conventional
166 V. Migaleddu and G. Virgilio

US they appear as hypo-echoic areas with Cyst


well-defined margins. Color Doppler Simple bile cysts are most common liq-
does not reveal irregular vessels due to uid benign lesions (1–18%), and are
mass effect within the lesions. Contrast- spread throughout the world (Mirilas and
enhanced ultrasound reveals typical vas- Skandalakis, 2002). Hydatid cysts are
cular behavior similar to the surrounding notuncommon in countries where sheep-
parenchyma. breeding is practiced. Conventional US
Typical features: findings (typical anechoic lesions with
Arterial phase: iso-enhancing posterior enhancement) are specific for
Portal vein phase: iso-enhancing diagnosis. A nonvital parasitic cyst with
Parenchymal phase: iso-enhancing solid aspect may be mistaken for tumor;
Focal fatty change: on color Doppler flow signals are absent.
Focal areas of fatty infiltration can be Contrast-enhanced ultrasound findings are
found mimicking true focal lesions; on as follows:
conventional US they appear as typically
round-shaped hyper-echoic lesion. Color Typical features:
Doppler and CEUS findings are similar to Arterial phase: non-enhancing
those of fatty spared areas. Portal vein phase: non-enhancing
Typical features: Parenchymal phase: non-enhancing
Arterial phase: iso-enhancing Additional features:
Portal vein phase: iso-enhancing Parenchymal phase: increased ultra-
Parenchymal phase: iso-enhancing sound transmission

Adenoma
Regenerative Nodule
Adenoma is an uncommon primary benign
Regenerative nodules are found in liver
tumor that has hepatocellular origin. Young
cirrhosis surrounded by fibrous septae.
females who use hormonal medication
The nodular dimension characterizes the
are the most frequently affected patients
micro-macro or mixed type of cirrhosis
(Karhunen, 1986). An adenoma may be
evolution. Large regenerative nodules from
asymptomatic or may present abdominal
5 to 20 mm can be detected on conven-
pain. This tumor has a marked tendency
tional US as hypo-echoic nodular lesions;
to intra-lesional or intra-abdominal high
hyper-echoic patterns due to fatty change
risk bleeding, although very rare potential
are rarer. Color Doppler or CEUS real-
malignant transformation could be consid-
time vascular evaluation show no apparent
ered. Hepatic adenoma presents (80% of
vessels in lesions (Migaleddu et al., 2004;
cases) as a solitary well-defined or encap-
Nicolau et al., 2006).
sulated tumor. This is a hypervascular
Typical features: lesion composed of cords of proliferating
Arterial phase: iso-enhancing hepatocytes without portal venous tracts,
Portal vein phase: iso-enhancing terminal hepatic veins, and bile ductules,
Parenchymal phase: iso-enhancing but with typical large sub-capsular tribu-
Additional features: tary arteries originating from the hepatic
Arterial phase: hypo- or hyper-enhancing arterial system. In larger masses necrotic
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 167

and hemorrhagic changes are frequently Typical features:


observed (Ros et al., 2001, Bartolozzi Arterial phase: rim enhancement, no
et al., 2001). On conventional US, depend- enhancement
ing on lipid contents in the hepatocytes, Portal vein phase: hyper-/iso-enhancing
HA appears as iso or Hyper-echoic a rea- rim, no central enhancement
sonably large, well-circumscribed focal Parenchymal phase: hypo-enhancing
lesion. When intralesional hemorrhages or rim, no central enhancement
necrosis occur, an inhomogeneous pattern Additional features:
is prevalent (Grazioli et al., 2000). Color Arterial phase: enhanced septa, hyper-
Doppler demonstrates large peripheral sub- enhanced liver segment
capsular arteries and veins (Golli et al., Portal vein phase: hypo-enhancing rim,
1994). Contrast-enhanced ultrasound can enhanced septae
be summarized as follows (Bleuzen and Tuberculosis is an uncommon liver infec-
Tranquart, 2004; Catalano et al., 2005): tion which occurs in the miliary form. On
Typical features: conventional ultrasound single or multiple
Arterial phase: hyper-enhancing, com- hypo-echoic, well-defined nodules spread
plete throughout the liver are generally observed
Portal vein phase: hypo-enhancing (Brauner et al., 1989; Levine 1990). Contrast-
Parenchymal phase: iso-enhancing enhanced ultrasound reveals hypo-enhancing
Atypical features: well-defined nodules, especially in the later
Arterial phase: non-enhancing areas phase. Clinical findings are mandatory for
(hemorrhage) the diagnosis (Akahn and Pringot, 2002)
Portal vein phase: hyper-enhancing, non- and differentiation from hepatic metastases
enhancing areas (hemorrhage) is necessary.
Parenchymal phase: non-enhancing
areas (hemorrhage) Inflammatory Pseudo Tumors
The inflammatory pseudo tumor of the
Abscess liver is very uncommon, and is character-
Abscesses of different etiology, pyogenic ized by a well-defined mass consisting
or amoebic, may present different features of chronic inflammatory cell infiltration
due to the varying degree of liquefac- and fibrosis (Ishak et al., 1994). Prevalent
tion and location. Furthermore, vascular- necrotic nodules are also reported (Iwase
ity depends on evolution stage; usually it et al., 2002; Abbey-Toby et al., 2003).
is located at the periphery of the lesion, The etiology of this disease is unclear:
whereas liquefied areas are positioned in micro-organisms from food, cholangitis
the central zone. Conventional US can or hepatic involvement from chronic intra-
demonstrate an inhomogeneous echo- abdominal infection with ascending infec-
structure with peripheral hyper-echoic tion via the portal blood stream could
aspects and central hypo-echoic features. explain the inflammatory hepatic lesion
Color Doppler may detect vascular signals due to oblitering phlebitis (Horiuchi et al.,
at periphery or in the septa of the lesion. 1990). On conventional ultrasound large,
Contrast-enhanced ultrasound shows as inhomogeneous, hypo-echoic masses
described below (Catalano et al., 2005): with multiple thin septae are identified
168 V. Migaleddu and G. Virgilio

(Nam et al., 1996). In our experience small vascular enhancement behavior is related
encapsulated nodules may be observed. to the evolution of the lesion. Thus, dur-
Color Doppler presents some flow sig- ing the arterial phase dysplastic nodules
nals in the periphery of the lesion in large or early HCC appear usually hypo- iso-
masses and no flow signal in the small vascularized, while advanced HCCs are
nodules. Contrast-enhanced ultrasound hyper-vascularized. On conventional US,
shows marginal enhancement in the arte- findings are non-specific; usually small
rial phase, while hypo enhancement areas HCCs (< 3 cm) are hypo-echoic and homo-
are demonstrated in the portal and paren- geneous but may appear hyper-echoic due
chymal phases. Small nodules present to fatty content (Kim et al., 2001; Ward and
hypo or lack of enhancement especially in Robinson, 2002). Color Doppler or power
the late phase. This ambiguous behavior Doppler may detect characteristic hyper-
may mimic metastases. vascularity; the “basket” pattern of vessels
surrounding and penetrating or “vessel-
Enhancement Patterns of Malignant Focal within the tumor” pattern may be visual-
Lesions
ized in HCC as has been reported (Tanaka
Dysplastic Nodule, Early and Advanced et al., 1990). Contrast-enhanced ultrasound
Hepatocellular Carcinoma may show any selective enhancement in
Hepatocellular carcinoma (HCC) is the the arterial phase differentiating HCCs
most common primary hepatic malignancy. from regenerative nodules and dysplastic
Virus infections (B and particularly C), nodules (Migaleddu et al., 2004; Nicolau
genetic hemochromatosis, primary bil- et al., 2006) (Figure 12.1a–d). The most
iary cirrhosis, environmental and dietary common features are described below:
factors such as persistent organic pol-
Typical features:
lutant substances, alcohol and aflotoxins
Arterial phase: hyper-enhancing, com-
are the most important causes. Regional
plete, non-enhancing areas (necrosis)
distribution worldwide is related to the
Portal vein phase: iso-, hypo-enhancing,
different etiologies. Chronic liver diseases
non-enhancing areas (necrosis)
correlated with C virus infection and alco-
Parenchymal phase: hypo-enhancing or
hol abuse are becoming the most diffuse
wash-out
causes of HCC. Dysplastic nodules occupy
an intermediate position between regenera- Additional features:
tive nodules and well-differentiated HCC. Arterial phase: “chaotic” vessels, enhanc-
Dysplastic nodules are characterized by ing tumor thrombus in Portal Vein + HCC/
cellular atypia without the development portal vein: hypo-enhancing with intral-
of small arteries with a size ranging from esional vessels
8–10 to 15 mm. Their pre-malignant nature Portal vein phase: hypo-iso-enhancing
has been well demonstrated. Hepatic car- Parenchymal phase: hypo-iso-enhancing
cinogenesis is described as a multi-step
process where the progressive arterializa- Liver Metastases
tion and gradual loss of portal vessels are Metastatic liver disease is the most fre-
the principal features (Ueda et al., 1992; quent malignant hepatic affliction, the liver
Matsui et al., 1991). It is evident that the being the most common site for secondary
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 169

Figure 12.1. (a–d): Well-differentiated hepatocellular carcinoma in 80-year-old man. (a) On base line
conventional US, hyper-echoic lesion is identified (arrow). (b) On CEUS in the early arterial phase, “cha-
otic” micro-vessels within the lesion are observed (arrow). (c) On CEUS in arterial phase, hyper-enhanced
feature is evident (arrow). (d) On CEUS in late phase, a poor wash-out is demonstrated (arrow)

malignant involvement. Depending on the surrounding parenchyma but with a loss of


original organ, the liver metastases may portal vessels so at least the blood volume
be hyper-vascular or hypo-vascular, small is inferior. On conventional US, metastases
or large; the latter can present roughly may present hypo-, iso- or hyper-echoic
central necrotic areas. All metastases are patterns matching the primary tumor or
characterized by some degree of arte- necrosis or calcification. Usually small
rial neoangiogenesis (macro-vessels in lesions appear as homogeneous hypo- or
peripheral margins and micro-vessels in iso-echoic solid nodules, while the larger
the central zone) and lack or reduced lesions frequently show an inhomogene-
portal supply. The total hematic volume ous aspect with hypo-echoic areas due
of the lesion is inferior to the surrounding to necrosis and iso-, hyper-echoic areas
healthy parenchyma. Some metastases like related to fibrosis and calcification. The
those from neuro-endocrine, thyroid, lym- margins may be reasonably well-defined,
phoma, plasmacytoma, and kidney tumors and sometimes a halo sign or target pat-
may have a superior arterial supply to the tern can be found. Cystic appearances may
170 V. Migaleddu and G. Virgilio

be related to ovarian and neuro-endocrine Hypo-vascular Metastases


primary tumors (Cosgrove, 2001; Harvey Typical features:
and Albrecht, 2001). Color Doppler may Arterial phase: rim or marginal enhancing
detect flow signals in the outer zone of Portal vein phase: hypo-enhancing
the lesions, whereas this finding in the Parenchymal phase: hypo-, lack of enhan-
inner zone is relatively rare (Numata et al., cement
1997). Depending on whether the lesion Additional features:
is hyper- or hypo-vascularized CEUS Arterial phase: complete enhancing,
may present different aspects (Albrecht et non-enhancing areas (necrosis)
al., 2004; Catalano et al., 2005) (Figure Portal vein phase: non-enhancing areas
12.2a–d). (necrosis)

Figure 12.2. (a–d): Multiple Hypo-vascular liver metastases from pancreas carcinoma in 75-year-old
man. (a) On base line conventional US: a large hypo-echoic mass is well detectable. (b) On CEUS in the
early arterial phase, minimal vessels within the lesions are visible. (c) On CEUS in the arterial phase, mul-
tiple Hypo-vascular lesions show a prevalent marginal enhancement. (d) On CEUS in late phase, multiple
lacks of enhancement are evident; metastases less than 10 mm are proved (arrows)
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 171

Hyper-vascular Metastases who were exposed to Thorotast vinyl


Typical features: chloride or other toxic agents (Bartolozzi
Arterial phase: hyper-enhancing, complete et al., 2001; Ros et al., 2001). Other
Portal vein phase: hypo-enhancing less common sarcomas such as leiomy-
Parenchymal phase: hypo-, lack of enhan- oma, fibrous, histiocytoma can be found.
cement Sarcomas usually appear as large size
Additional features: uncapsulated lesions at imaging detec-
Arterial phase: “chaotic” vessels tion, but very small multifocal nodules
may also be seen (Ros et al., 2001). On
Cholangiocarcinoma conventional US mixed echogenicity pre-
vails due to hemorrhagic or necrotic areas.
Cholangiocarcinoma is a malignant tumor
Color Doppler might capture minimal int-
originating from the bile duct epithelium.
ralesional flow signals. Contrast-enhanced
Peripheral cholangiocarcinoma arises from
ultrasound presents hypo-enhancement in
the small intra-hepatic bile duct. It con-
all the vascular phases.
sists of multiple nodules or a large lobu-
Lymphoma as a primary hepatic involve-
lated mass without capsule. It usually has
ment is very rare; however, it is possible to
hypo-vascularization regarding the arterial
find liver involvement during the outcome
vessels and the portal supply also is lim-
of Hodgkin’s or non Hodgkin’s systemic
ited. Arterial hyper-vascularization is less
lymphoma. Immune deficiency patients
common (Ros et al., 2001). On conven-
are mainly concerned. On conventional
tional US the small lesions are hypo- or
US hypo-echoic nodules or masses may
iso-echoic, whereas the larger lesions are
be found. The diffuse hepatic involvement
mixed-hyper-hypo-echoic. Color Doppler
appears as a diffused alteration of the
reveals the absence of intralesional blood
normal liver echo-texture. Lymphomatous
flow. During color or power Doppler evalu-
nodules present on CEUS iso- or hyper-
ation it is easy to detect bile duct dilatation
enhancement in the arterial phase but
and portal venous encasement (Harvey
hypo- or lack of enhancement in the fol-
and Albrecht, 2001). Contrast-enhanced
lowing phases (Catalano et al., 2005).
ultrasound reveals features close to those
of metastasis.
Typical features: (B) Detection of Focal Liver Lesions
Arterial phase: marginal enhancing Conventional US is the most widely dif-
Portal vein phase: hypo-, lack of enhan-fused cross-sectional imaging procedure for
cement the first approach in diagnosis of abdominal
Parenchymal phase: hypo-, lack of enhan-organs and liver diseases due to its rela-
cement tive low cost, its availability, safety, and
high patient acceptance. In the detection
Rare Malignant Neoplasm and staging of a liver lesion conventional
Angiosarcoma is the most frequent malig- US is less accurate than contrast-enhanced
nant mesenchymal liver malignancy, but spiral CT and MR because of the difficulty
proves to be a very rare tumor prevalently in detecting small size and/or iso-echoic
diagnosed in 60–80 year old male patients lesions, deep lesions especially in difficult
172 V. Migaleddu and G. Virgilio

anatomical areas (sub-capsular or sub- measuring 10 mm or less were found,


diaphragmatic areas). The recent advances among which 59 (15.6%) were considered
in nonlinear imaging and the introduction metastases; while 303 (80.2%) were clas-
of new contrast agents permit a large sified as benign lesions, 16 (4.2%) were
number of limitations to be overcome, but not classified (Schwartz et al., 1999).
the role of CEUS is under debate. Current US, CT and MR technological
Clinical investigations evaluating CEUS advances improve detection of incidental
have demonstrated that the accuracy in liver focal lesions. Differentiation between
detecting liver metastases is improved the benign or malignant lesions is diffi-
and may be comparable to spiral CT cult in small lesions that do not always
Additionally, others have reported that show enough characteristic features on
CEUS can detect lesions not visible on static cross-sectional imaging as CT or
CT (Harvey et al., 2000; Albrecht et al., MR; besides, depending on dimension
2003a). Data on diagnostic performance and position, it may also be difficult to
have also been published using the much perform a biopsy. In these cases CEUS,
simpler real-time method with low MI and being a dynamic real-time imaging modal-
SonoVue (Albrecht et al., 2003b). Even if ity with good detail resolution, may be
further studies are required, current data used to play a crucial role (Bleuzen and
from single center and multicenter clini- Tranquart, 2004).
cal trials are becoming more robust. It is
not expected that US will replace CT or Staging and Follow-Up of Oncologic Patients
MR; nevertheless, US remains useful as a For optimal management of cancer patients
complementary procedure in some clinical with suspected metastases we need accu-
situations. rate tumor staging. Molecular imaging
with PET and cross-sectional imaging
Incidental Detection of Hepatic Focal Lesion
with CT and MR prove that they have an
The widespread use of a new generation of important role in total body evaluation.
imaging modalities has permitted a higher However, the liver is the most common
frequency in detecting incidental focal target organ for metastases of many pri-
liver lesions in patients without symp- mary tumors, especially for those arising
toms or evidence of liver disease (Ros from abdominal organs and structures.
and Davis, 1998). Benign hepatic lesions Conventional US being readily available
are very frequent. In an autoptic series of and cheap remains in many European
95 patients, benign lesions were detected centers the first-line imaging modality in
in 52% of cases; the most common were detecting hepatic metastases. Ultrasound
bile duct tumor in 27% and hemangiomas sensitivity has been reported to be as high
in 20% (Karhunen, 1986). In ∼50% of as 85% (Clarke et al., 1989); especially
the cases multiple lesions were found. It for rectal-colon tumor, intra-operative US
has also been reported that small hepatic is the most sensitive imaging modality
lesions are frequently detected on routine compared to current multi-phasic contrast
imaging evaluations. In a non-spiral con- CT and RM. Thus, it is usually performed
trast enhanced CT review of 2,978 cancer before liver resection to identify synchro-
patients, 378 (12.7%) small hepatic lesions nous metastases (Conlon et al., 2003).
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 173

Data related to the use of intra-operative blood-pool contrast agent without any
CEUS are still scarce (Siösteen and interstitial equilibrium phase. The appear-
Elvin, 2004). ance of metastases in the arterial phase is
In rectal-colon cancer, patients who variable. Hypo-vascular metastases show
underwent liver resection range between up in CEUS as hypo-enhanced lesions
30–40% in terms of 5 years survival at with and without an additional marginal
best because other metastases at the time enhancement, while hyper-vascular metas-
of resection were undetectable. To detect tases appear as brightly hyper-enhanced
metachronous metastases, a 3-monthly more or less homogeneous lesions. Hyper-
conventional US and yearly CT follow-up vascular metastases occur most often from
program may identify 88% of patients who primary tumors of neuro-endocrine origin
will develop liver metastases in an asympto- or from renal or breast cancer.
matic stage in order to plan liver resection, A common pitfall is that small cysts are
chemotherapy or percutaneous treatments sometimes detected on late phase scanning.
(Howell et al., 1999). Additionally, it has These can usually be distinguished from
been reported that patients with diffuse metastases as they characteristically show
metastatic diseases receiving systemic a higher US beam transmission (Bleuzen
chemotherapy at an asymptomatic stage and Tranquart, 2004). Differential diag-
have higher response rates, better quality nosis may be difficult for metastases with
of life, and survival, than those who begin dominant cystic component, as in ovarian
the chemotherapy at a symptomatic stage metastases. Contrast-enhanced ultrasound
(Nordic Gastrointestinal Tumor Adjuvant could help to observe enhanced septae
Therapy Group, 1992). In these follow-up or nodules that are very frequent in such
programs CEUS results as the more cost- metastases. Thrombosed hemangiomas
effectiveness imaging procedure in the may mimic the hypo enhancing behav-
detection of liver metastases. ior of the metastases. Contrast-enhanced
Evaluation in the late parenchymal phase ultrasound follow-up could identify the
with high M.I., using a first-generation modification of lesion vascularity due to
contrast agent (Levovist) has been recom- intralesional thrombus changes over time.
mended in some reports (Albrecht et al.,
Surveillance for Hepatocellular Carcinoma
2003b). A long liver-specific late phase
of up to 20 min has been described due Surveillance means repeated application
to adherence of the microbubbles to the of screening tests. Conventional US and
hepatic sinusoid or to phagocytosis by serum alpha-fetoprotein are the most
Kupffer’s cells (Albrecht et al., 2001). widespread screening tests for hepatocel-
The late phase is the most useful time lular carcinoma particularly in European
to detect metastases that usually appear countries; a biannual interval between US
as hypo-enhanced or lack enhancement examinations is generally accepted. A ran-
areas. In comparison, most benign lesions domized study demonstrated the benefit of
show uptake at this time and are therefore such a program considering that in the sur-
not likely to be confused with metastases. veillance arm the HCC-related mortality
Second-generation gas filled microbub- was reduced by 37% (Zhang et al., 2004).
bles as in SonoVue are considered a truly During the Consensus Conference of
174 V. Migaleddu and G. Virgilio

European Association for the study of the low M.I., 100 primary tumors (96.2%)
liver (held in Barcelona in the year 2000), demonstrated hyper-enhancement in the
a biannual US and AFL level evaluations arterial phase but 4 did not (3.8%) (Nicolau
were recommended as the minimal follow up. et al., 2004). One study did not find arterial
A spiral CT investigation is suggested for enhancement in three well differentiated
patients who do not have nodules on US HCCs out of 60 cases consequently high-
but an increasing AFP level. For patients lighted the relationship between absence
who have a US finding of a nodule of of enhancement in the arterial phase and
10 mm or less in diameter, 3 monthly US histological differentiation (Tanaka et al.,
is suggested taking into account that these 2001). After diagnosis of HCC, the pres-
lesions are too small to be accurately char- ence and degree of cirrhosis, as well as
acterized and at least 50% of these lesions the tumor extension, need to be evalu-
will not be HCC. In patients with a nod- ated for staging and management. In fact,
ule of > 20 mm in diameter HCC can be the presence of satellite nodules, biliary
confirmed if the AFP level is over 400 ng/ extension, vascular, and extra-hepatic
ml and there is contrast-enhanced CT, involvement modify therapeutic planning.
MR, or angiographic proof of the hyper- Entire hepatic evaluation during the arte-
vascularity of the lesion. At least two pieces rial phase (20–30 s) might be difficult, also
of positive imaging evidence are required because the sweeping scanning might fail
for HCC diagnosis (Bruix et al., 2001). to detect some lesions. The administration
Biopsy is a controversial option espe- of several doses of contrast agents and
cially for small nodules as it has a poten- scanning of different hepatic segments
tial risk of bleeding and tumor seeding in are suggested as well as the use of a high
the needle track, whereas it is not always M.I. at intervals to evaluate the destruction
useful due to the heterogeneity of vari- and parenchyma replenishment in order to
ous types of hepatic nodular lesions (Brú detect hyper-enhancing foci. To date, there
et al., 1989). Microbubble contrast as echo- are no data enabling comparison between
enhancer permits useful information to be CEUS and spiral CT MRI in this chal-
obtained for diagnosis if prevalent hyper- lenging field. The extra-hepatic staging
vascularity of the lesion is demonstrated favors total body cross-sectional imaging.
(Lencioni et al., 1996; Fracanzani et al., The evaluation of portal tumor invasion is
2001). These imaging modalities are asso- obviously easier with US examination.
ciated with motion and blooming artifacts.
With first and second-generation con-
trast media, CEUS reveals a well defined (C) Monitoring of Percutaneous Ablative
hyper-enhancement in arterial phase and Treatment
prevalent washout in the late phase. For In the last 10 years numerous changes
real-time evaluation of all three phases, the have occurred in therapeutic approach
use of second-generation contrast medium for primary and metastatic hepatic
and low M.I., is more suitable (Nicolau malignancies. Many minimally invasive
et al., 2006). therapies as the percutaneous ethanol
In a series of 104 patients, using a second- injection (PEI) and the thermal ablation
generation of contrast media SonoVue and using different energy sources (radio
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 175

frequencies, laser or micro-wave) Livraghi et al., 2004). Comparative


are proposed (Livraghi et al., 1998; data between surgical and percutaneous
Goldberg et al., 2000; Dodd et al., 2000; treated patients show advantages for the
De Baere et al., 2000; Lencioni et al., latter (Livraghi in http://www.smirg.org/
2003). Percutaneous ablation therapies lectures.php).
play a key role in the management of As previously described, US, CEUS,
patients with liver malignancies (HCC contrast-enhanced CT, and MRI play a
and metastases). Numerous reports have key role in the diagnosis of liver malignan-
shown the efficacy and safety of these cies in patients that are candidates for any
ablating therapies especially in non surgi- treatment plan. For the safety, low cost,
cal candidates. The possibility to repeat and absence of radiation and iodine con-
treatment sessions in the case of recur- trast administration, CEUS play a crucial
rent or new lesions, the lower morbidity role in the early and long-term follow-up
and mortality as well as the low costs are of percutaneous treated patients, usually
notable advantages (Solbiati et al., 2001; in the first week post treatment and after

Figure 12.3. (a–d): Hepatocellular carcinoma: pre and post radio-frequency ablative treatment features.
(a) and (b): Pre; on CEUS in the early and arterial phase: typical hyper-vascular pattern is identified
(arrows). (c) and (d): Post; on CEUS in the early and arterial phase: no vessels within the lesion and no
enhancement are present. 100% of necrosis is achieved (arrows)
176 V. Migaleddu and G. Virgilio

1, 3, 6 months and then yearly. No reli- CONCLUSION


able information about the outcome of
ablation treatments is provided by con- Conventional US, being readily available,
ventional US, color Doppler, and power economical and safe, remains the imaging
Doppler. In fact, the assessment of vas- modality most widespread throughout the
cularization and tissue perfusion is essen- world in detecting hepatic focal lesions.
tial to distinguish necrosis from residual In detection with CEUS, the number of
viable tumor. Triphasic helical CT or confirmed metastases increase from 3.06
dynamic gadolinium-enhanced MR can to 5.42 in base-line comparison. After con-
predict the extent of the coagulation area trast medium administration the sensitivity
to within 2–3 mm. for detecting individual liver metastases
The disappearance, if any was previously improves dramatically from 63% to 91%.
visualized, of intralesional enhancement Furthermore, the capability to detect
on contrast-enhanced imaging is the most very small metastases (> 10 mm) passes
important finding that suggests complete with CEUS from 54% to 92% (Albrecht
ablation (Figure 12.3a–d). While persist- et al., 2001). Comparing conventional US
ence of hyper-enhancing in the arterial and CEUS in a multicenter series of 123,
phase or clearly enhancing in the portal sensitivity in the detection of single metas-
phase suggest suspicious residual viable tasis improved from 71% to 87%, while
tumor. Conatrast-enhanced ultrasound the specificity increased from 60% to 88%
can provide any kind of vascular related following contrast injection (Albrecht
information. Comparing pre- and post et al., 2003).
treatment findings, the size modification Moreover, contrast-enhanced intra-
of the lesions permits a good evaluation operative US (CEIOUS) also seems to
of the necrotic coagulation area and of improve these results in a recent report.
treatment results. The efficacy of treat- In a series of 24 patients CEIOUS found
ment in the hypo-enhanced lesions, as lesions missed at preoperative imaging
for most liver metastases, can be identi- and at intra-operative US in four patients
fied when the lack of enhanced area is and confirmed all of the new findings of
increased determining in this manner the intra-operative US in four patients. In
sufficiency of the perilesional “safety” addition CEIOUS helped to define the
margin achieved. tumor margins of the main lesion in 29%
Contrast-enhanced ultrasound and con- of patients with rectal colon liver metas-
trast-enhanced CT, within the first 30 tases (Torzilli et al., 2005). This intra-
days, post-ablative evaluation may reveal operative approach is suitable for the
a thin and uniform enhancing rim of synchronous metastases of rectal-colon
hyper-vascularity along the periphery of or other abdominal tumors. Contrast
the necrotic area. Misinterpretation of this sectional imaging (US, CT and RM)
perilesional hyperemic halo as residual conserve a central role for pre-treatment
viable tumor can be avoided by comparing diagnosis of other kinds of secondary
post-ablation with pre-ablation findings tumor. Surgeons’ and physicians’ interest
(Solbiati et al., 2004). in CEUS is rapidly increasing.
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 177

Earlier we have described the guide- useful to read together fundamental echoes
lines for primary tumor management pro- from tissue as well as nonlinear fundamental
duced by the European Association for the echoes from the contrast agent; the CPS
Study of Liver Disease (EASLD) (Bruix (the most recent technological approach)
et al., 2001). The American Association for permits this also with second-generation
the Study of Liver Disease (AASLD) has contrast media in real-time. The possibility
recently published “Practice Guidelines of aiming at the lesion, and in particular
for the management of HCCs”. The use seeing both extra- and intra-lesional vascular
of only one contrast enhanced imaging contribution in the arterial phase, consid-
modality including CEUS to demonstrate erably assists early phase characteriza-
hyper-vascularization and diagnose a HCC tion of HCC. Using second-generation
was suggested for a lesion larger than of contrast media, nonlinear fundamental
20 mm (Bruix and Sherman, 2005). (CPS) and “conventional” harmonic imag-
The characterization of the focal liver ing findings proves to be equivalent in
lesions by evaluation of hemodynamic the study of the late phase useful for
contrast enhancement behavior is con- metastases diagnosis. This is true except
firmed by an important number of studies, for the lesions positioned in the distal
using both first or second-generation con- field because there is poor intensity of the
trast media (Leen et al., 2002; Migaleddu contrast harmonic echoes in comparison
et al., 2004; Wen et al., 2004; Quaia et al., with contrast nonlinear fundamental ones
2004; Catalano et al., 2005). (Migaleddu et al., 2005). Excellent details
Whereas some authors have emphasized resolution especially in the arterial phase
the importance of observing enhancement is also valuable in outcome assessment of
at a late time, in our experience it is also ablative therapy.
important to observe arterial and early
portal enhancement. Other experiences REFERENCES
seem to confirm this (Nicolau et al.,
Abbey-Toby, A., Cazals-Hatem, D., Colombat, M.,
2006). In particular, detection through Belghiti, J., Vilgrain, V., and Degott, C. 2003.
good details resolution of peri- and intra- Inflammatory pseudo-tumor of the liver: is pre-
lesional macro- and micro-vessels con- operative diagnosis possible? Gastroenterol.
tributes to the diagnosis of focal liver Clin. Biol. 27: 883–890.
lesions, above all in the cirrhotic liver and Akahn, O., and Pringot, J. 2002. Imaging of abdom-
inal tubercolosis. Eur. Radiol. 12: 321–323.
therefore in differential diagnosis between
Albrecht, T., Hoffmann, C.W., Schmitz, S.A.,
Regenerative macro-nodules, Dysplastic Schettler, S., Ovemberg, A., Germen, C.T., and
nodules in the transformation phase and Wolf, K.J. 2001. Phase-inversione sonography
well-differentiated HCC; as well, early during the liver-specific late phase of contrast
rim enhancement can suggest diagnosis enhancement: improved detection of liver metas-
of high flow hemangioma. The study of tases. Am. J. Roentgenol. 176 (5): 1191–1198.
Albrecht, T., Blomley, M.J., Burns, P.N., Wilson, S.,
the only late phase is more useful in the
Harvey, C.J., Leen, E., Claudon, M., Calliada,
diagnosis of metastases. F., Correas, J.M., LaFortune, M., Campani,
In order to obtain excellent details reso- R., Hoffmann, C.W., Cosgrove, D.O., and
lution, especially in the arterial phase, it is LeFevre, F. 2003a. Improved detecion of hepatic
178 V. Migaleddu and G. Virgilio

metastases with pulse-inversion US during the Chin C.T., and Burns P.N. 1997. Predicting the
liver-specific phase of SHU 505A: multicenter acoustic response of a microbubble population
study. Radiology 227: 361–370. for contrast imaging. In: IEEE Ultrason. Symp.
Albrecht, T., Oldenburg, A., Hohmann, J., Skrok, J., 22: 1557–1560.
Hoffmann, C.W., Schettler, S., and Wolf, K.J. Clarke, M.P., Kane, R.A., Steele, G. Jr., Hamilton,
2003b. Imaging of liver metastases with contrast- E.S., Ravikumar, T.S., Onik, G., and Clouse,
specific low-MI real-time ultrasound and SonoVue. M.E. 1989. Prospective comparision of pre-
Eur. Radiol. 13 (Suppl 3): 79–86. operative imaging and intraoperative ultrasonog-
Albrecht, T., Hohmann, J., Oldenburg, A., Skrok, J., raphy in the detecion of liver tumours. Surgery
and Wolf, K.J. 2004. Detection and characterisa- 106: 849–855.
tion of liver metastases. Eur. Radiol. 14 (Suppl 8): Conlon, R., Jacobs, M., Dasgupta, D., and Loge,
25–33. J.P. 2003. The value of intraoperative ultra-
Apfel, R.E., and Holland, C.K. 1991. Gauging the sound during hepatic resection compared with
likelihood of cavitation from short-pulse, low improved preoperative magnetic resonace imag-
duty cycle diagnostic ultrasound. Ultrasound ing. Eur. Ultrasound 16: 211–216.
Med. Biol. 17: 175–185. Cosgrove, D.O. 2001. Malignant liver disease. In:
Bartolozzi, C., Cioni, D., Donati, F., and Lencioni, R. Meire, H.B., Cosgrove, D.O., Dewbury, K.C.,
2001. Focal liver lesions: MR imaging-patho- and Farrant, P. (eds). Clinical Ultrasound: A
logic correlation. Eur. Radiol. 11: 1374–1388. Comprehensive Text. Abdominal and General
Bleuzen, A., and Tranquart, F. 2004. Incidental Ultrasound, vol. 1, 2nd ed. Churchill Livingstone,
liver lesions: diagnostic value of cadence con- London, 211–231.
trast pulse sequencing (CPS) and SonoVue. Eur. Cosgrove, D.O. 2005. Harmonic imaging: tissue
Radiol. 14 (Suppl 8): 53–62. and microbubbles in http://www.smirg.org/lec-
Brauner, M., Buffard, M.D., Jaentils, V., Legrand, I., tures.php.
and Gotheil, C. 1989. Sonography and computed De Baere, T., Elias, D., Dromain, C., Din, M.G.,
tomography of mocroscopic tuberculosis of the Kuoch, V., Ducreux, M., Boige, V., Lassau, N.,
liver. J. Clin. Ultrasound 17: 563–568. Marteau, V., Lasser, P., and Roche, A. 2000.
Brú, C., Marotto, A., Bruix, J., Faus, R., Biachi, L., Radiofrequency ablation of 100 hepatic metas-
Calvet, X., Ayuso, C., Vilana, R., Gilalbert, R., tases with a mean follow-up of more than 1 year.
and Rodes, J. 1989. Diagnostic accurancy of Am. J. Roentgenol. 175: 1619–1625.
fine-needle biopsy in patients with hepatocellular Dodd, G.D. 3rd., Soulen, M.C., Kane, R.A.,
carcinoma. Dig. Dis. Sci. 34: 1765–1769. Livraghi, T., Lees, W.R., Yamashita, Y.,
Bruix, J., and Sherman, M., 2005. Management Gillams, A.R., Karahan, O.I., and Rhim, H.
of Hepatocellular carninoma. J. Hepatol. 42: 2000. Minimally invasive treatment of malig-
1208–1236. nant hepatic tumors: at the threshold of a major
Bruix, J., Sherman, M., Llovet, J.M., Beaugrand, M., breakthrough. Radiographics 20: 9–21.
Lencioni, R., Burroughs, A.K., Christensen, E., EFSUMB Study Group Albrecht, T., Blomley, M.,
Pagliaro, L., Colombo, M., and Rodes, J. 2001. Bolondi, L., Claudon, M., Correas, J.M.,
Clinical management of hepatocellular carci- Cosgrove, D., Greiner, L., Jager, K., Jong, N.D.,
noma. Conclusions of the Barcelona. EASL Leen, E., Lencioni, R., Lindsell, D., Martegani,
Conference. J. Hepatol. 35: 421–430. A., Solbiati, L., Thorelius, L., Tranquart,
Burns, P.N., Wilson, S.R., and Simpson, D.H. F., Weskott, H.P., and Whittingham, T.
2000. Pulse inversion imaging of liver blood 2004. Guidelines for the Use of Contrast
flow: an improved method for characterization Agents in Ultrasound. Ultraschall. Med. 25:
of focal masses with microbubble contrast. 249–256.
Invest. Radiol. 35: 58–71. Fracanzani, A.L., Burdick, L., Borzio, M., Roncalli, M.,
Catalano, O., Nunziata, A., Lobianco, R., and Siani, A. Bonelli, N., Borzio, F., Maraschi, A., Fiorelli,
2005. Real-time harmonic contrast material- G., and Fargion, S. 2001. Contrast-enhanced
specific US of focal liver lesions. RadioGraphics Doppler ultrasonography in the diagnosis of
25: 333–349. hepatocellular carcinoma and premalignant
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 179

lesions in patients with cirrhosis. Hepatology 34: sign at two-phase spiral CT. Radiology 208:
1109–1112. 543–548.
Goldberg, S.N., Gazelle, C.S., Compton, C.C., Karhunen, P.J. 1986. Benign hepatic tumours and
Mueller, P.R., and Tanabe, K.K. 2000. Treatment tumour-like conditions in men. Clin. Pathol. 39:
of intrahepatic malignancy with radiofrequency 183–188.
ablation: radiologic-pathologic correlation. Kim, C.K., Lim, J.H., and Lee, W.J. 2001. Detection
Cancer 88: 2452–2463. of hepatocellular carcinomas and dysplastic nod-
Golli, M., Van Nhieu, J.T., Mathieu, D., Zafrani, ules in cirrhotic liver: accuracy of ultrasonogra-
E.S., Cherqui, D., Dhumeaux, D., Vasile, N., and phy in transplant patients. J. Ultrasound Med.
Rahmouni, A. 1994. Hepatocellular adenoma: 20: 99–104.
Color Doppler US and pathologic correlations. Kroncke, T.J., Taupitz, M., Kivelitz, D., Scheer,
Radiology 190: 741–744. I., Daberkow, U., Rudolph, B., and Hamm, B.
Grazioli, L., Federle, M.P., Ichikawa, T., Balzano, E., 2000. Multifocal nodular fatty infiltration of the
Nalesnik, M., and Madariaga, J. 2000. Liver liver mimicking metastatic disease on CT: imag-
adenomatosis: clinical, histopathologic and ing findings and diagnosis using MR imaging.
imaging findings in 15 patients. Radiology 216: Eur. Radiol. 10: 1095–1100.
395–402. Leen, E., Angerson, W.J., Yarmenitis, S.,
Greis, C. 2004. Technology overview: SonoVue Bongartz, G., Blomley, M., Del Maschio, A.,
(Bracco, Milan). Eur. Radiol. 14 (Suppl 8): 11–15. Summaria, V., Maresca, G., Pezzoli, C., and
Hamilton, M.F., and Blackstock, D.T. 1998. Llul, J.B. 2002. Multi-centre clinical study
Nonlinear Acoustics. Academic, San Diego, CA. evaluating the efficacy of SonoVue (BR1),
Harvey, C.J., and Albrecht, T. 2001. Ultrasound of a new ultrasound contrast agent in Doppler
focal liver lesions. Eur. Radiol. 11: 1578–1593. investigation of focal hepatic lesions. Eur. J.
Harvey, C.J., Blomley, M.J., Eckersley, R.J., Radiol. 42: 200–206.
Cosgrove, D.O., Patel, N., Heckemann, R.A., and Lencioni, R., Pinto, F., Armillota, N., and
Butler-Barnes, J. 2000. Hepatic malignancies: Bartolozzi, C. 1996. Assessment of tumor vasco-
improved detection with pulse-inversion US in larity in hepatocarcinoma: comparison of power
late phase of enhancement with SH U 508A-early Doppler and Color Doppler US. Radiology 201:
experience. Radiology 216: 903–908. 353–358.
Horiuchi, R., Uchida, T., Kojima, T., and Shikata, T. Lencioni, R.A., Allgaier, H.P., Cioni, D.,
1990. Inflammatory pseudotumor of the liver. Olschewski, M., Deibert, P., Crocetti, L., Frings,
Clinicalpathological study and review of litera- H., Laubenberger, J., Zuber, I., Blum, H.E.,
ture. Cancer 65: 1583–1590. and Bartolozzi, C. 2003. Small hepatocellular
Howell, J.D., Wotherspoon, H., Leen, E., Cooke, carcinoma in cirrhosis: randomized compari-
T.C., and MacArlde, C.S. 1999. Evaluation of son of radiofrequency thermal ablation versus
a follow up programme after curative resec- percutanous ethanol injection. Radiology 228:
tion for colorectal cancer. Br. J. Cancer 79: 235–240.
308–310. Levine, C. 1990. Primary macronodular hepatic
Ishak, K.G., Anthony, P.P., and Sobin, L.H. 1994. tuberculosis: US and CT appearances. Gastro-
World Health Organization: International Histo- intest. Radiol. 15: 307–309.
logic Classification of Tumors, 2nd ed. Berlin, Livraghi, T. 2006. Role of image-guided thera-
Springer. pies in hepatocellular carcinoma in http://www.
Iwase, K., Higaki, J., Yoon, H.E., Mikata, S., smirg.org/lectures.php.
Miyazaki, M., Torikai, K., Shirai,Y., Awai, K., Livraghi, T., Benedini, V., Lazzaron, S., Meloni, F.,
Imakita, M., and Kamiike, W. 2002. Solitary Torzilli, G., and Vettori, C. 1998. Long term
necrotic nodule of the liver. J. Hepatobiliary results of single session PEI in patients with large
Pancreat. Surg. 9: 120–124. hepatocellular carcinoma. Cancer 83: 48–57.
Jang, H.J., Choi, B.I., Kim, T.K., Jun, A.J., Kim., Livraghi, T., Meloni, F., Morabito, A., and Vettori,
K.W., Han, J.K., and Han, M.C. 1998. Atypical C. 2004. Multimodal image-guided tailored
small hemangiomas of the liver: “bright dot” therapy of early and intermediate hepatocellular
180 V. Migaleddu and G. Virgilio

carcinoma: long-term survival in the experi- management of the cirrothic patient and for detec-
ence of a single radiologic referral center. Liver tion of HCC. Eur. Radiol. 14 (Suppl 8): 63–71.
Transpl. 10: 98–106. Nicolau, C., Vilana, R., Catalá, V., Bianchi, L.,
Matsui, O., Kadoja, M., Kamejama, T., Takashima, T., Gilabert, R., Carcía, A., and Brú, C. 2006.
Nakanuma, J., Unoura, M., Kobayashi, K., Importance of evaluating all vascular phases on
Izumi, R., and Ida, M. 1991. Benign and malig- contrast enhanced sonography in the diferentia-
nant nodules in cirrhotic patients: distinction on tion of benign from malignant focal lesions. Am.
blood supply. Radiology 178: 493–497. J. Roentgenol. 186: 158–167.
Migaleddu, V., Virgilio, G., Campisi, G., Sirigu, D., Nordic Gastrointestinal Tumour Adjuvant Therapy
and Canalis, G.C. 2002. Conventional ultra- Group, 1992. Expectancy or primary chemo-
sonography versus tissue harmonic imaging for therapy in patients with advanced asymptomatic
the assessment of the common bile duct in chole- colorectal cancer; a randomisede trial. J. Clin.
cystectomized patients. Radiol. Med. 103: 1–6. Oncol. 10: 904–911.
Migaleddu, V., Virgilio, G., Turilli, D., Conti, M., Numata, K., Tanaka, K., Kiba, T., Norimoto, M.,
Campisi, G., Canu, M., Sirigu, D., and Arata, S., Kondo, M., and Sekihara, H. 1997.
Vincentelli, I. 2004. Charaterization of focal Use of hepatic tumor index on color Doppler
liver lesions in real-time using harmonic sonography for differentiating large hepatic
imaging with high mechanical index and con- tumors. Am. J. Roentgenol. 168: 991–995.
trast agent levovist. Am. J. Roentgenol. 182: Ortega, D., Burns, P.N., Simpson, D.H., and
1505–1512. Wilson, S.R. 2001. Tissue harmionic imaging:
Migaleddu, V., Manca, A., Turilli, D., Luciano, P., is it a benefit for bile duct sonography? Am. J.
Canu, N., Campisi, G., and Virgilio, G. 2005. Roentgenol. 176: 653–659.
Fundamental nonlinear imaging versus harmonic Phillips, P., and Gardner, E. 2004. Contrast-agent
imaging with contrast medium sonovue in the detection and quantification. Eur. Radiol. 14
evaluation of the hepatic focal lesions: prelimi- (Suppl 8): 4–10.
nary results. Eur. Radiol. 14 (Suppl. 1): B-126. Powers, J.E., Burns, P.N., and Souquet, J. 1997.
Mirilas, P., and Skandalakis, J.E. 2002. Benign Imaging instrumentation for ultrasound con-
anatomical mistakes: incidentaloma. Am. Surg. trast agents. In: Nanda, N.C., Schlief, R., and
68: 1026–1028. Goldberg, B.B. (eds). Advances in Echo Imaging
Moody, A.R., and Wilson, S.R. 1993. Atypical Using Contrast Enhancement, 2nd, ed. Kluwer,
hepatic hemangioma: a suggestive sonographic Dordrecht, 139–170.
morphology. Radiology 188: 413–417. Quaia, E., Calliada, F., Bertolotto, M., Rossi, S.,
Morel, D.R., Schwieger, I., Hohn, L., Terrettaz. J., Garioni, L., Rosa, L., and Pozzi-Mucelli, R.
Llull, J.B., Cornioley, Y.A., and Schneider, M. 2004. Characterization of focal liver lesions with
2000. Human pharmacokinectics and safety eval- contrast-specific US modes and a sulfur hexaflu-
uation of SonoVueTM, a new contrast agent for oride-filled microbubble contrast agent: diag-
ultrasound imaging. Invest. Radiol. 35 (Suppl 1): nostic performance and confidence. Radiology
80–85. 232: 420–430.
Nam, K.J., Kang, H.K., and Lim, J.H. 1996. Ros, P., and Davis, G. 1998. The incidental focal liver
Inflammatory pseudotumor of the liver: CT and lesion: photon, proton, or needle? Hepatology
sonographic features. Am. J. Roentgenol. 167: 98: 1183–1190.
485–487. Ros, P.R., Menu, Y., Vilgrain, V., Mortele, K.J., and
Nanda, N.C., and Carstensen, E.L. 1997. Echo- Terris, B. 2001. Liver neoplasms and tumor-like
enhancing agents: safety. In: Nanda N.C., conditions. Eur. Radiol. 11 (Suppl 2): 145–165.
Schlief, R., and Goldberg, B.B. (eds). Advances Schneider, M., Arditi, M., Barrau, M.B., Brochot,
in Echo Imaging Using Contrast Enhancement, J., Broillet, A., Ventron, R., and Yan, F. 1995.
2nd, ed.Kluwer, Dordrecht, 139–170. BR1: a new ultrasonographic contrast agent
Nicolau, C., Vilana, R., and Brù, C. 2004. The based on sulfur hexafluoride-filled microbub-
use of contrast-enhanced ultrasound in the bles. Invest. Radiol. 30: 451–457.
12. Focal Liver Lesion: Nonlinear Contrast-Enhanced Ultrasound Imaging 181

Schwartz, L.H., Gandras, E.J., Colangelo, S.M., Ueda, K., Terada, T., Nakanuma, Y., and Matsui, O.
Ercolani, M.C., and Panicek, D.M. 1999. 1992. Vascular supply in adenomatous hyperplasia
Prevalence and importance of small hepatic of the liver and hepatocellular carcinoma: a mor-
lesions found at CT in patients with cancer. phometric study. Hum. Pathol. 23: 619–626.
Radiology 210: 71–74. Unal, O., Sakarya, M.E., Arslan, H., Tuncer, I.,
Siösteen, A.K., and Elvin, A. 2004. Intra-operative and Etlik, O. 2002. Hepatic cavernous hemangi-
uses of contrast-enhanced ultrasound. Eur. omas: patterns of contrast enhancement on MR
Radiol. 14 (Suppl 8): 87–95. fluoroscopy imaging. Clin. Imaging 26: 39–42.
Solbiati, L., Livraghi, T., Golberg, S.N., Ierace, T., Volk, M., Strotzer, M., Lenhart, M., Techert, J.,
Meloni, F., Dellanoce, M., Cova, L., Halpern, Seitz, J., and Feuerbach, S. 2001. Frequency of
E.F., and Gazelle, G.S. 2001. Percutaneous benign hepatic lesions incidentally detected with
radio-frequency ablation of liver metastase contrast-enhanced section portal venous pahase
from colon rectal cancer: long term risults 117 spiral CT. Acta Radiol. 42: 172–175.
patients. Radiology 221: 159–166. Ward, B., Baker, A.C., and Humprey, V.F. 1997.
Solbiati, L., Tonolini, M., and Cova, L. 2004. Non linear propagation applied to the improve-
Monitoring RF ablation. Eur. Radiol. 14 (Suppl 8): ment of resolution in diagnostic medical ultra-
34–42. sound. J. Acoust. Soc. Am. 101: 143–154.
Tanaka, S., Kitamura, T., Fuijta, M., Nakanishi, K., Ward, J., and Robinson, J. 2002. How to detect
and Okuda, S. 1990. Color Doppler flow imag- hepatocellular carcinoma in chirrhosis. Eur.
ing of liver tumors. Am. J. Roentgenol. 154: Radiol. 12: 2258–2272.
509–514. Wen, Y.L., Kudo, M., Zheng, R.Q., Ding, H., Zhou, P.,
Tanaka, S., Ioka, T., Oshikawa, O., Hamada, Y., and Minami, Y., Chung, H., Kitano, M., Kawasaki, T.,
Yoshioka, F. 2001. Dynamic sonography of hepatic and Maekawa, K. 2004. Characterizazione of
tumors. Am. J. Roentgenol. 177: 799–805. hepatic tumors: value of contrast-enhanced
Torzilli, G., Del Fabro, D., Palmisano, A., Donadon, M., coded phase-inversion harmonic angio. Am. J.
Bianchi, P., Roncalli, M., Balzarini, L., and Roentgenol. 182: 1019–1026.
Montorsi, M. 2005. Contrast-enhanced intraop- Zhang, B.H., Yang, B.H., and Tang, Z.Y. 2004.
erative ultrasonography during hepatectomies Randomized controll trial of screening for hepa-
for Colon rectal metastaes. J. Gastrointest. Surg. tocellular carcinoma. J. Cancer Res. Clin. Oncol.
9: 1148–1154. 130: 417–422.
13
Hepatocellular Carcinoma: Magnetic
Resonance Imaging
Bachir Taouli

INTRODUCTION MAGNETIC RESONANCE


IMAGING TECHNIQUE
Hepatocellular carcinoma (HCC) is the
most common primary hepatic malig- There is a considerable overlap in the
nancy, accounting for 90% of primary imaging appearances of the different types
liver neoplasms and is responsible for up of cirrhotic nodules. Because it provides
to 1 million deaths annually worldwide excellent contrast resolution and mul-
(Fung and Marsh, 2002). The major risk tiplanar evaluation, magnetic resonance
factor for HCC is cirrhosis. All types imaging (MRI) is the examination of
of cirrhoses predispose to HCC, but the choice for detecting and characterizing
incidence is particularly high in patients cirrhotic nodules. Compared to ultrasound
with chronic hepatitis B (HBV), chronic and computerized tomography (CT), MRI
hepatitis C (HCV), and in alcoholic liver is superior in detecting and characterizing
disease. HCC develops by sequential steps HCC in the cirrhotic liver (Yamashita
from low grade dysplastic nodules to et al., 1996; Aschoff et al., 2001). The use
frank malignant HCC. The recognition of of intravenous contrast material with mul-
HCC is critical because curative treatment tiphasic imaging (arterial, portal venous,
(including surgical resection and liver and equilibrium) is essential to accurately
transplantation) and prognosis depend on identify focal hepatic lesions. However,
early diagnosis. The 5-year survival rate MRI remains less accurate for lesions
for untreated, symptomatic HCC is < 5% < 2 cm in diameter. Recent advances in MRI
(El-Serag and Mason, 1999), whereas the software and hardware, including the use of
5-year survival rate in patients with cirrhosis parallel imaging and surface phased-array
who have a small (< 2 cm) HCC lesion and coils, provide faster sequences that can be
undergo liver transplantation is 80%; there- acquired within a breath-hold, decreasing
fore, the detection of small HCC is highly motion and respiratory artifacts.
critical to patient outcome (Mazzaferro In our institution, we use breath-hold seq-
et al., 1996). uences for routine liver MRI examination

183
184 B. Taouli

(whole acquisition time < 30 min). The degree of differentiation (Ebara et al.,
protocol includes: T1-weighted in- and out- 1999; Amano et al., 2003). In-phase and
of-phase gradient-recalled echo (GRE) to opposed-phase images can demonstrate
evaluate for microscopic fat content, Turbo microscopic fat components within HCC,
Spin Echo T2-weighted with spectral fat areas of fat accumulation showing a signal
saturation or T2-fast STIR (short tau inver- drop on opposed-phase images.
sion recovery), T2-weighted single-shot On T2-weighted images, HCCs are
RARE (rapid acquisition with relaxation classically hyperintense, whereas regener-
enhancement) and 3D T1-weighted fat ative nodules (Figure 13.1) and most dys-
suppressed GRE before and after dynamic plastic nodules are hypointense (Earls et al.,
injection of 15–20 ml of gadolinium 1996; Matsui et al., 1989; Krinsky et al.,
chelates using an automatic injector, with 2001) (Figures 13.2 and 13.4). However,
acquisitions at the arterial, portal venous, well-differentiated HCC lesions can be
and equilibrium phases. We use the short- iso- or rarely hypointense on T2-weighted
est possible TR (repetition time) in order images (Earls et al., 1996; Hecht et al.,
to minimize acquisition time and allow 2006). A focus of high signal intensity on
sequential acquisitions through the entire T2 within a hypo- or isointense nodule
liver during breath-holding. Fat suppres- (“nodule within a nodule” sign) is highly
sion improves the conspicuity of contrast suggestive of HCC developing within a
enhancement, and eliminates chemical- dysplastic nodule (Mitchell et al., 1991;
shift artifacts at fat-water interfaces. van den Bos et al., 2006). Ebara et al.
(1999) have shown that the signal inten-
sity of HCC on T1- and T2-weighted
MAGNETIC RESONANCE images is related to the degree of his-
IMAGING APPEARANCE tologic differentiation and intratumoral
OF HEPATOCELLULAR copper content, whereas the signal inten-
CARCINOMA sity on T2-weighted images is related to
the degree of histologic differentiation.
The macronodular architecture of the However, a relationship between copper
cirrhotic liver and the morphology of content and signal intensity of HCC was
HCC are usually well demonstrated on T1- not confirmed by other studies (Nakakoshi
and T2-weighted images. On T1-weighted et al., 1996). Hemosiderin deposition is
images, HCC can be hypo-, iso- or hyper- common in regenerative nodules (called
intense, compared to the surrounding liver siderotic nodules), producing specific imag-
(Aschoff et al., 2001; Honda et al., 1997). ing features on MRI, such as hypointensity
Most well differentiated HCCs and high- on T1-GRE images and T2-weighted GRE
grade dysplastic nodules have high signal images (Krinsky et al., 2001). Dysplastic
intensity on T1-weighted images, and T1 nodules are usually homogeneously hyper-
hyperintensity can be related to copper, intense on T1-weighted images, but small
Fe3+, glycogen deposition, or high pro- HCC nodules may exhibit similar findings
tein or lipid content, and possibly to the (Krinsky and Israel, 2003).
13. Hepatocellular Carcinoma: Magnetic Resonance Imaging 185

Figure 13.1. (a–c): Regenerative nodule and HCC in the same patient. (a): T2-weighted images show
that the regenerative nodule is hypointense (left arrow) and the HCC is hyperintense (right arrow). (b):
Post-contrast images at the arterial phase show that the regenerative nodule is not enhancing (left arrow)
whereas the HCC is hypervascular (right arrow). (c): Post-contrast images at the portal venous phase
show that the regenerative nodule is hypovascular (left arrow) and the HCC has washout with a capsule
(right arrow)
186 B. Taouli

GADOLINIUM ENHANCEMENT retraction. In the portal venous phase,


OF HEPATOCELLULAR HCC appears hyper-, iso- or hypointense
CARCINOMA relative to the background liver (Figures
13.1, 13.2). A minority of well-differ-
The overlapping signal intensities of regen- entiated HCC lesions are hypovascular
erative nodules, dysplastic nodules, and (Matsui et al., 1991) and best seen at the
HCC on T1- and T2-weighted images portal venous phase, because liver paren-
necessitate the use of contrast media, includ- chymal enhancement peaks at this phase.
ing non-specific extracellular gadolinium Delayed images can show late enhancement
chelates or tissue-specific contrast media. of the fibrous capsule which is typically a
Gadolinium-DTPA is an extracellu- feature of HCC with a size > 2 cm, but not
lar contrast medium that produces T1 of regenerative or dysplastic nodules. Both
shortening and enhancement of HCC on necrosis and hemorrhage are common in
T1-weighted images. HCC obtains its large tumors. Regenerative and dysplastic
blood supply almost exclusively from the nodules may be difficult to identify on
hepatic artery, and most HCCs are best post-gadolinium images because these
seen on arterial phase images (Figures are typically hypovascular lesions with
13.1–13.3) (Kim et al., 1995; Murakami predominantly portal blood supply, with
et al., 1995; Oi et al., 1996), allow- an enhancement similar to that of the
ing differentiation from regenerative and adjacent parenchyma, although increased
dysplastic nodules, as well as from focal arterial flow has been described in a small
fibrosis, which enhances during the portal minority of dysplastic nodules (Matsui
and equilibrium phases, with typical capsular et al., 1991).

Figure 13.2. Dynamic post-contrast T1-weightedimages at the arterial (left) and portal venous (right)
show a segment 4 hypervascular HCC (arrow) with washout at the portal venous phase
13. Hepatocellular Carcinoma: Magnetic Resonance Imaging 187

Figure 13.3. Dynamic post-contrast T1-weighted images at the arterial phase (left: source image, right:
coronal maximum intensity projection reformat) show multifocal HCCs (arrows). The largest lesion is
located in the posterior right hepatic lobe, with disseminated nodules in the rest of the liver

field inhomogeneities, causing a marked


MAGNETIC RESONANCE
decrease in the signal intensity of liver tis-
IMAGING TISSUE-SPECIFIC sue, particularly on T2-weighted images.
CONTRAST AGENTS A breath-hold T2-weighted gradient-echo
sequence is generally used after SPIO
The possibility of improving the diagnosis injection, because the susceptibility effects
of HCC by the use of contrast agents spe- are maximized and respiratory artifacts are
cifically targeted to the liver cells has been eliminated. Because of the decrease of the
evaluated in multiple studies. signal intensity of the background liver,
Tissue-specific MR contrast media SPIO particles increase the detection and
which produce T1 enhancement in hepa- the characterization of focal liver lesions
tocytes include mangafodipir (MnDPDP), (benign lesions will show uptake of SPIO).
gadobenate (Gd-BOPTA), and gadoxetic Kupffer cells are rarely present in HCC,
acid (Gd-EOB-DTPA). All of these agents and therefore the tumor will show little or
show uptake in well-differentiated HCC, as no uptake of SPIO, and will appear more
well as in benign hepatocellular nodules, conspicuous after SPIO injection.
so their value in discriminating between
benign and malignant lesions is limited
Combined Use of Superparamagnetic
(Rofsky et al., 1993; Murakami et al.,
Iron Oxide and Gadolinium
1996). Results with these agents have not
been shown to be superior to those with Using both gadolinium and ferumoxides
gadolinium for the detection of HCC. (an SPIO agent) MR agents, Bhartia
Superparamagnetic iron oxide (SPIO) et al. (2003) demonstrated a 78% sen-
particles are taken up by Kupffer cells of the sitivity for detection of HCC in 31
reticuloendothelial system. They enhance patients transplanted between 3 and 245
T2 and T2* relaxation by increasing local days after MR imaging. The sensitivity
188 B. Taouli

dropped to 38% for lesions smaller than intense pseudo-lesions and HCCs is
1 cm. However, these authors did not difficult with MRI, and follow-up is
evaluate dysplastic nodules. In a previ- often required. Hyperintense pseudo-
ous study (Ward et al., 2000), using lesions should be recognized by their
SPIO and dynamic gadolinium injection lack of interval growth or frank dis-
in 27 patients (with explant correla- appearance (Shimizu et al., 2003;
tion available in 15 patients), the same Jeong et al., 2002), whereas HCC will
authors were able to detect significantly have a tendency to grow. Jeong et al.
more HCC nodules (particularly HCCs (2002) have calculated the doubling
< 1 cm), although few lesions were vis- time of small enhancing lesions, and
ible only on SPIO-enhanced images. showed that all nodules with interval
Dysplastic nodules were also character- growth and/or change in MR signal are
ized with greater confidence on the basis HCCs (mean doubling time of HCCs,
of their combined enhancement charac- 2 months 2 weeks). The same authors
teristics (no enhancement with gadolin- reported positive and negative predic-
ium, enhancement with SPIO). However, tive values of 100% and 98% respec-
this approach is more complex and adds tively, on the basis of nodule growth,
to the cost and duration of MRI. Another and suggested a close interval follow-up
study found that the benefit of the com- in such patients.
bined use of SPIO and gadolinium is 2. Large HCC: A large HCC can have
negligible (Halavaara et al., 2002). a number of distinctive features, such
as a mosaic pattern, tumor capsule,
satellite nodules, vascular invasion,
ATYPICAL APPEARANCES and metastasis. The mosaic pattern is
OF HEPATOCELLULAR related to confluent nodules separated
CARCINOMA by areas of necrosis and thin septa,
best seen on T2-weighted images and
1. Small HCC: A small HCC is defined post-contrast images (Kadoya et al.,
as a lesion < 2 cm in size. Most small 1992). A large HCC often has a fibrous
HCC will show intense arterial phase tumor capsule (seen in 60–82% of
enhancement. The widespread use of cases) (Kadoya et al., 1992; Choi et
arterial phase CT and MRI in cir- al., 1993), appearing hypointense on
rhotic patients has shown the existence T1- and T2-weighted images, rarely
of enhancing “pseudo-lesions” (small hyperintense on T2-weighted images,
hepatic nodules with arterial phase with delayed enhancement. Vascular
enhancement, not seen on the portal invasion is often present histologically
venous phase images), that may be in the HCC, affecting the portal vein
wedge-shaped, geographic, oval or and the hepatic veins, particularly in a
round in shape (Shimizu et al., 2003). large non-encapsulated HCC. On MRI,
These lesions may represent arterioportal vascular invasion appears as non-
shunts, but the underlying structure is visualization of the portal branches or
still not clear (Yu et al., 1997). The as an intraluminal mass, with enhance-
differentiation between small hyper- ment at the arterial phase, and a filling
13. Hepatocellular Carcinoma: Magnetic Resonance Imaging 189

defect on later phases. Extrahepatic NEW PERSPECTIVES


dissemination of HCC can occur in
advanced HCC, and has been described Diffusion-weighted MRI: Diffusion-weighted
in 37% of a series of 403 patients with MRI represents a potential new tool for the
HCC evaluated with CT (Katyal et al., detection and characterization of HCC, and
2000), occurring mostly as lung and differentiation of benign from malignant
lymph node metastases. lesions in the cirrhotic liver. Diffusion-
3. Diffuse type HCC: Infiltrative or dif- weighted MRI is noninvasive, requires no
fuse type HCC represented 13% of contrast injection, and can be performed
HCCs in a recent series, showing ill- within a breath-hold. Previous studies
defined margins with extensive portal have shown that benign lesions (cysts
venous tumor thrombosis, variable and hemangiomas) show higher appar-
enhancement, and markedly elevated ent diffusion coefficients (ADCs) than
alpha-fetoprotein levels in the blood malignant lesions (HCCs and metastases)
(Kanematsu et al., 2003) (Figure 13.4). (Kim et al., 1999; Taouli et al., 2003).

Figure 13.4. Diffuse type HCC with portal vein invasion. Pre-contrast T2- (left upper row), pre- (right
upper row) and post-contrast (lower image) T1-weighted images show an infiltrative HCC of the right
hepatic lobe (arrows) invading the portal vein (small arrow)
190 B. Taouli

This is likely related to free water motion in In conclusion, although MRI with
benign lesions, and restricted water motion dynamic gadolinium enhancement appears
in the presence of tumor. We showed in to be superior to helical CT for detecting
a previous study (Taouli et al., 2003) a HCC, its sensitivity for the detection of
significant difference between ADCs of small lesions is still low, and improvement
benign and malignant liver lesions (2.45 ± in accuracy is still needed. The use of new
0.96 × 10−3 and 1.08 ± 0.50 × 10−3 mm2/s contrast media, high field (3T) magnets
for b = 0 and 500 s/mm2; respectively, and new MRI techniques such as perfusion
p < 0.001). The mean ± SD ADCs (x10−3 and diffusion MRI will likely improve the
mm2/s) of the different groups of lesions image quality and the accuracy of MRI in
were: HCCs 1.33 ± 0.13, hemangiomas the future.
2.95 ± 0.67 and cysts 3.63 ± 0.56 (Taouli
et al., 2003).
REFERENCES
Perfusion-weighted MRI: Perfusion-weighted
MRI provides the ability to detect regional Amano, S., Ebara, M., Yajima, T., Fukuda, H.,
Yoshikawa, M., Sugiura, N., Kato, K., Kondo, F.,
and global alterations in liver blood flow, Matsumoto, T., and Saisho, H. 2003. Assessment
and could be performed with CT or MRI. of cancer cell differentiation in small hepatocel-
Whole-liver CT imaging has the potential lular carcinoma by computed tomography and
to provide both high-temporal-resolution magnetic resonance imaging. J. Gastroenterol.
and high-spatial-resolution imaging of the Hepatol. 18:273–279.
entire liver for HCC detection; however, Aschoff, A.J., Merkle, E.M., Wong, V., Zhang,
Q., Mendez, M.M., Duerk, J.L., and Lewin,
the radiation dose must be considered. J.S. 2001. How does alteration of hepatic blood
Perfusion-weighted MRI represents a flow affect liver perfusion and radiofrequency-
promising technique for HCC surveillance induced thermal lesion size in rabbit liver? J.
(Pandharipande et al., 2005). The closest Magn. Reson. Imaging 13:57–63.
technique to perfusion-weighted MRI has Bhartia, B., Ward, J., Guthrie, J.A., and Robinson,
been use of a double-arterial phase MR P.J. 2003. Hepatocellular carcinoma in cirrhotic
livers: double-contrast thin-section MR imaging
imaging technique enabled by a rapid par- with pathologic correlation of explanted tissue.
allel imaging method; one group reported AJR Am. J. Roentgenol. 180:577–584.
sensitivity of HCC detection of 91.7% Choi, B.I., Takayasu, K., and Han, M.C. 1993.
using double arterial phase vs. 76.3% with Small hepatocellular carcinomas and associ-
a single arterial phase (Yoshioka et al., ated nodular lesions of the liver: pathology,
2002). These findings suggest the poten- pathogenesis, and imaging findings. AJR Am. J.
Roentgenol. 160:1177–1187.
tial of high-temporal-resolution perfusion- Earls, J.P., Theise, N.D., Weinreb, J.C., DeCorato,
weighted MRI to improve detection of D.R., Krinsky, G.A., Rofsky, N.M., Mizrachi, H.,
HCC. and Teperman, L.W. 1996. Dysplastic nodules
3T MRI: Advantages of imaging at 3T and hepatocellular carcinoma: thin-section MR
include higher signal-to-noise ratio, better imaging of explanted cirrhotic livers with patho-
image contrast, particularly in gadolinium- logic correlation. Radiology 201:207–214.
Ebara, M., Fukuda, H., Kojima, Y., Morimoto, N.,
enhanced applications, which translate to Yoshikawa, M., Sugiura, N., Satoh, T., Kondo, F.,
higher spatial resolution images and faster Yukawa, M., Matsumoto, T., and Saisho, H. 1999.
imaging. This could potentially improve Small hepatocellular carcinoma: relationship of
HCC detection in the future. signal intensity to histopathologic findings and
13. Hepatocellular Carcinoma: Magnetic Resonance Imaging 191

metal content of the tumor and surrounding ing for liver disease. AJR Am. J. Roentgenol.
hepatic parenchyma. Radiology 210:81–88. 173:393–398.
El-Serag, H.B., and Mason, A.C. 1999. Rising inci- Krinsky, G.A., and Israel, G. 2003. Nondysplastic
dence of hepatocellular carcinoma in the United nodules that are hyperintense on T1-weighted
States. N. Engl. J. Med. 340:745–750. gradient-echo MR imaging: frequency in cir-
Fung, J., and Marsh, W. 2002. The quandary over rhotic patients undergoing transplantation. AJR
liver transplantation for hepatocellular carcinoma: Am. J. Roentgenol. 180:1023–1027.
the greater sin? Liver Transpl. 8:775–777. Krinsky, G.A., Lee, V.S., Nguyen, M.T., Rofsky,
Halavaara, J., Tervahartiala, P., Isoniemi, H., and N.M., Theise, N.D., Morgan, G.R., Teperman,
Hockerstedt, K. 2002. Efficacy of sequential use L.W., and Weinreb, J.C. 2001. Siderotic nodules
of superparamagnetic iron oxide and gadolinium in the cirrhotic liver at MR imaging with explant
in liver MR imaging. Acta. Radiol. 43:180–185. correlation: no increased frequency of dys-
Hecht, E.M., Holland, A.E., Israel, G.M., Hahn, W.Y., plastic nodules and hepatocellular carcinoma.
Kim, D.C., West, A.B., Babb, J.S., Taouli, B., Lee, Radiology 218:47–53.
V.S., and Krinsky, G.A. 2006. Hepatocellular car- Matsui, O., Kadoya, M., Kameyama, T., Yoshikawa,
cinoma in the cirrhotic liver: gadolinium-enhanced J., Arai, K., Gabata, T., Takashima, T., Nakan-uma,
3D T1-weighted MR imaging as a stand-alone Y., Terada, T., and Ida, M. 1989. Adenomatous
sequence for diagnosis. Radiology 239:438–447. hyperplastic nodules in the cirrhotic liver: differ-
Honda, H., Kaneko, K., Kanazawa, Y., Hayashi, entiation from hepatocellular carcinoma with MR
T., Fukuya, T., Matsumata, T., Maeda, T., and imaging. Radiology 173:123–126.
Masuda, K. 1997. MR imaging of hepatocellular Matsui, O., Kadoya, M., Kameyama, T., Yoshikawa,
carcinomas: effect of Cu and Fe contents on sig- J., Takashima, T., Nakanuma, Y., Unoura, M.,
nal intensity. Abdom. Imaging 22:60–66. Kobayash, K., Izumi, R., Ida, M., et al. 1991.
Jeong, Y.Y., Mitchell, D.G., and Kamishima, T. Benign and malignant nodules in cirrhotic livers:
2002. Small (<20 mm) enhancing hepatic nod- distinction based on blood supply. Radiology
ules seen on arterial phase MR imaging of the 178:493–497.
cirrhotic liver: clinical implications. AJR Am. J. Mazzaferro, V., Regalia, E., Doci, R., Andreola,
Roentgenol. 178:1327–1334. S., Pulvirenti, A., Bozzetti, F., Montalto, F.,
Kadoya, M., Matsui, O., Takashima, T., and Ammortuna, M., Morabito, A., and Gennari, L.
Nonomura, A. 1992. Hepatocellular carci-noma: 1996. Liver transplantation for the treatment of
correlation of MR imaging and histopathologic small hepatocellular carcinomas in patients
findings. Radiology 183:819–825. with cirrhosis. N. Engl. J. Med. 334:693–699.
Katyal, S., Oliver, J.H. 3rd, Peterson, M.S., Ferris, Mitchell, D.G., Kim, I., Chang, T.S., Vinitski, S.,
J.V., Carr, B.S., and Baron, R.L. 2000. Extrahe- Consigny, P.M., Saponaro, S.A., Ehrlich, S.M.,
patic metastases of hepatocellular carcinoma. Rifkin, M.D., and Rubin, R. 1991. Fatty liver.
Radiology 216:698–703. Chemical shift phase-difference and suppres-
Kanematsu, M., Semelka, R.C., Leonardou, sion magnetic resonance imaging techniques in
P., Mastropasqua, M., and Lee, J.K. 2003. animals, phantoms, and humans. Invest. Radiol.
Hepatocellular carcinoma of diffuse type: MR 26:1041–1052.
imaging findings and clinical manifestations. J. Murakami, T., Kim, T., Oi, H., Nakamura, H.,
Magn. Reson. Imaging 18:189–195. Igarashi, H., Matsushita, M., Okamura, J., and
Kim, T., Murakami, T., Oi, H., Matsushita, M., Kozuka, T. 1995. Detectability of hypervascu-
Kishimoto, H., Igarashi, H., Nakamura, H., and lar hepatocellular carcinoma by arterial phase
Okamura, J. 1995. Detection of hypervascular images of MR and spiral CT. Acta Radiol.
hepatocellular carcinoma by dynamic MRI and 36:372–376.
dynamic spiral CT. J. Comput. Assist. Tomogr. Murakami, T., Baron, R.L., Peterson, M.S., Oliver,
19:948–954. J.H. 3rd, Davis, P.L., Confer, S.R., and Federle,
Kim, T., Murakami, T., Takahashi, S., Hori, M., M.P. 1996. Hepatocellular carcinoma: MR imag-
Tsuda, K., and Nakamura, H. 1999. Diffusion- ing with mangafodipir trisodium (Mn-DPDP).
weighted single-shot echoplanar MR imag- Radiology 200:69–77.
192 B. Taouli

Nakakoshi, T., Kajiyama, M., Fujita, N., Jong- diffusion isotropy and characterization of focal
Hon, K., Takeichi, N., and Miyasaka, K. 1996. hepatic lesions with two single-shot echo-planar
Quantitative analyses of correlations of signal MR imaging sequences: prospective study in 66
intensity on T1-weighted images and T1 relaxa- patients. Radiology 226:71–78.
tion time with copper concentration in the rat van den Bos, I.C., Hussain, S.M., Terkivatan, T.,
liver. Acad. Radiol. 3:36–39. Zondervan, P.E., and de Man, R.A. 2006. Stepwise
Oi, H., Murakami, T., Kim, T., Matsushita, M., carcinogenesis of hepatocellular carcinoma in the
Kishimoto, H., and Nakamura, H. 1996. Dynamic cirrhotic liver: demonstration on serial MR imag-
MR imaging and early-phase helical CT for ing. J. Magn. Reson. Imaging 24:1071–1080.
detecting small intrahepatic metastases of hepa- Ward, J., Guthrie, J.A., Scott, D.J., Atchley, J.,
tocellular carcinoma. AJR Am. J. Roentgenol. Wilson, D., Davies, M.H., Wyatt, J.I., and
166:369–374. Robinson, P.J. 2000. Hepatocellular carcinoma
Pandharipande, P.V., Krinsky, G.A., Rusinek, H., in the cirrhotic liver: double-contrast MR imag-
and Lee, V.S. 2005. Perfusion imaging of the ing for diagnosis. Radiology 216:154–162.
liver: current challenges and future goals. Yamashita, Y., Mitsuzaki, K., Yi, T., Ogata, I.,
Radiology 234:661–673. Nishiharu, T., Urata, J., and Takahashi, M. 1996.
Rofsky, N.M., Weinreb, J.C., Bernardino, M.E., Small hepatocellular carcinoma in patients with
Young, S.W., Lee, J.K., and Noz, M.E. 1993. chronic liver damage: prospective comparison of
Hepatocellular tumors: characterization with detection with dynamic MR imaging and helical
Mn-DPDP-enhanced MR imaging. Radiology CT of the whole liver. Radiology 200:79–84.
188:53–59. Yoshioka, H., Takahashi, N., Yamaguchi, M., Lou,
Shimizu, A., Ito, K., Koike, S., Fujita, T., Shimizu, D., Saida, Y., and Itai, Y. 2002. Double arterial
K., and Matsunaga, N. 2003. Cirrhosis or chronic phase dynamic MRI with sensitivity encoding
hepatitis: evaluation of small (< or =2-cm) early- (SENSE) for hypervascular hepatocellular carci-
enhancing hepatic lesions with serial contrast- nomas. J. Magn. Reson. Imaging 16:259–266.
enhanced dynamic MR imaging. Radiology Yu, J.S., Kim, K.W., Sung, K.B., Lee, J.T., Yoo,
226:550–555. and H.S. 1997. Small arterial-portal venous
Taouli, B., Vilgrain, V., Dumont, E., Daire, J.L., shunts: a cause of pseudolesions at hepatic imag-
Fan, B., and Menu, Y. 2003. Evaluation of liver ing. Radiology 203:737–742.
14
Expression of Vascular Endothelial Growth
Factor in Hepatocellular Carcinoma:
Correlation with Radiologic Findings
Masayuki Kanematsu, Richard C. Semelka, and Shinji Osada

INTRODUCTION VEGF appears to play an important role.


The expression of this factor in HCC tis-
Angiogenesis is the process whereby new sues has been considered to be associated
blood vessels develop from the preexisting with tumor size or histological tumor
vasculature. It takes place physiologically grade (Yamaguchi et al., 1998). Even in
during embryonic development, during the the nontumorous liver parenchyma, VEGF
normal growth of tissues and the wound is expressed by sinusoidal endothelial cells
healing, and during the female reproduc- and hepatocytes (Yamane et al., 1994).
tive cycle (i.e., ovulation, menstruation, Although radiologic examinations such
and placental development) as well as dur- as conventional angiography, computed
ing the pathologic growth and metastatic tomography (CT), and magnetic resonance
spread of malignant neoplasms (Folkman, imaging (MRI) have been widely used as
1971). A variety of humoral agents need tools for the diagnosis of HCCs, relations
to be activated to generate a neovascular between the imaging findings and biomo-
blood supply or angiogenesis in the human lecular angiogenetic activities in HCC and
body, and vascular endothelial growth fac- in the surrounding liver have yet to be ascer-
tor (VEGF) is one of the most important tained. Investigations of this relation may
humoral agents to be activated to ensure help radiologists understand radiologic
the growth of the vascular endothelium. imaging findings related to molecular bio-
Various types of hepatocellular nodules logical treatments using an antiangiogenic
with malignant potential are known to agent such as bevacizmab, an antibody that
develop in livers with chronic liver dam- binds to VEGF, which has been approved
age: they are large regenerative nodules, by the US Food and Drug Administration
low-grade dysplastic nodules, high-grade for the treatment of metastatic colorectal
dysplastic nodules, well-differentiated cancer. Furthermore, PTK787/ZK 222584
hepatocellular carcinoma (HCC), and (PTK/ZK) is a potent, orally active and
moderately or poorly differentiated HCC selective inhibitor of the VEGF-receptor
(International Working Party, 1995). In the tyrosine kinases VEGFR-1 (Flt-1) and
process of HCC development, the expres- VEGFR-2 (KDR) under development. The
sion of proangiogenetic factors such as anti-angiogenetic effects of PTK/ZK in

193
194 M. Kanematsu et al.

several advanced cancers, including color- the first or second most common cause
ectal cancer, breast cancer, glioblastoma of cancer death worldwide, and in 1990
multiforme, prostate, and renal cancer the World Health Organization estimated
where VEGF is known to play a role, that there were ∼430,000 new cases of
have been expected. Most interestingly, liver cancer worldwide, and a similar
previous results have demonstrated that number of patients died as a result of this
gadolinium-enhanced MRI findings may disease. The most common risk factors for
be useful biomarkers for defining the HCC include hepatitis B or C viral infec-
pharmacological response of colorectal tion, aflatoxins, iron overload, and alcohol
cancer and liver metastases and for dose abuse. This disease is particularly preva-
of angiogenesis inhibitors, such as PTK/ lent in parts of Asia, Africa, and Europe.
ZK (Morgan et al., 2003). However, rela- Eighty percent of HCC patients have cir-
tions between VEGF activity in tumors rhosis of the liver, and 70% have elevated
and radiologic findings have yet to be serum alpha fetoprotein levels. Treatment
ascertained. options include liver transplantation, par-
We previously assessed the correla- tial hepatectomy, transcatheter arterial
tion between radiologic findings in ang- embolization, percutaneous ablation, and
iographically-assisted CT and MRI and radiation therapy. Reported 5-year survival
angiogenetic activities determined by rates range from 10% to 60%, depend-
immunohistochemistry (Kanematsu et al., ing on the tumor stage and the treatment
2004a) and Western blotting (Kanematsu method. The number of hepatitis virus
et al., 2004b, 2005) for VEGF in vari- carriers in Japan is estimated to be > 3
ous hepatocellular nodules in cirrhosis, million (1.6% of the population), and of
HCCs, and the surrounding liver. They these 1 million develop chronic hepati-
have described the potential usefulness tis and 30,000 HCC. A national survey
of radiologic imaging in the evaluation of (the third National Health and Nutrition
angiogenetic activity of hepatic nodules Examination Survey [NHANES III]) of the
and the surrounding liver. We summa- civilian, non-institutionalized U.S. popula-
rize the results and discuss the relations tion found that 1.8% of Americans (3.9
between radiologic findings and VEGF million) have been infected with hepatitis
expression in this chapter. C virus (HCV), of whom most (2.7 mil-
lion) are chronically infected with HCV.
One interesting footnote is that tattooing,
which is gaining in popularity, increases
HISTORY OF RADIOLOGIC the risk of HCV infection by ninefold.
DIAGNOSIS Hepatocellular carcinoma may become
OF HEPATOCELLULAR the most common malignant tumor in
CARCINOMA North America in a decade or two, and is
the only major cancer with a substantial
Hepatocellular carcinoma is a malignant increase in incidence.
neoplasm that arises from hepatocytes A pathology-based study from 1984
chiefly in patients with chronic liver dam- reported that the common major gross
age. Hepatocellular carcinoma is either patterns were expanding, spreading, and
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 195

multifocal by means of aggregating a total or borderline hepatocellular nodules in


of 529 HCC cases from Japan, the United cirrhosis were found on sonography, dif-
States, and South Africa. Further modifica- ficulty in the differentiation of HCCs
tions have divided HCC into infiltrative or from those nodules was a significant
expansive, single or multinodular, and mixed problem.
types, using observations on encapsulation In the early 1980s, radiologists began
and intrahepatic venous spread. Recently, using early-generation CT for diagnos-
however, with the advent of high-perform- ing HCCs. Detection was not satisfactory
ance radiologic diagnosis equipment such because 10-mm slices were incrementally
as sonography, CT and MRI, small HCCs, obtained after intravenous drip infusion
of 10 mm in size or less, have been preoper- of iodinated contrast material in a lengthy
atively diagnosed in greater numbers, which examination time, and small lesions were
has impacted upon improvement in early obscured in the thick slice acquisition, or
detection of HCCs. missed due to slice misregistration caused
In the mid 1970s, selective hepatic by irregular patient breath holding, or
angiography was introduced as an inva- were isodense due to poor contrast admin-
sive diagnostic tool of importance in the istration dynamics, which did not allow
preoperative evaluation of the extent and detection of small hypervascular HCCs. In
the vascular supply of HCCs and the liver. the mid 1980s, an incremental CT acqui-
Many tumors were readily diagnosed on sition during conventional trans-superior
angiograms, but there were limitations mesenteric arterial portography (CT during
which included: invasiveness of the pro- arterial portography) was introduced as a
cedure, difficulty distinguishing multi- mean to examine small HCCs. This exami-
centric HCCs from hepatic metastases nation was very sensitive but suffered from
and poorly vascularized lesions from a variety of false-positive lesions due to
primary cholangiocarcinoma, and inabil- portal venous obstruction, arterioportal
ity to detect iso- or hypovascular HCCs. shunting, non-portal splanchnic venous
During the same period, liver scintig- return, or benign focal hepatic lesions. As
raphy using 99mTc-sulfur colloid was a further development in the mid 1990s, a
introduced as a technique to detect focal combination method of CT during arterial
abnormalities, but it was nonspecific, portography and CT during conventional
since most focal hepatic lesions from hepatic arteriography was reported, and
metastases to hepatic cysts appeared as found to have high diagnostic accuracy
cold defects. In the early 1980s, transab- (Kanematsu et al., 1997). This procedure
dominal ultrasonography was developed did not gain widespread utilization because
as a noninvasive tool in the diagnosis of of its invasiveness, and technical challeng-
HCCs. Ultrasound experienced difficulty ing nature. We have employed combined
in visualization the entire liver due to CT during arterial portography and CT
acoustic blockage by ribs or intestinal or hepatic arteriography for the preoperative
pulmonary air, and sound attenuation due detection and evaluation of vascularity
to thick truncal fat in obese patients, and of HCCs and of surrounding liver in our
these represented significant limitations. clinical and research practice, and have cor-
Although a variety of benign, premalignant related VEGF expression and vascularity
196 M. Kanematsu et al.

of HCCs (Kanematsu et al., 2004b). Although CT will also continue to shorten


In the early 1990s, helical or spiral CTs its acquisition time and reduce radiation
were introduced into clinical routine and exposure, MRI will likely remain advanta-
have undergone continued evaluation since geous because of the superior capability of
then. The most up-to-date 64-row multi- describing histopathologic components in
detector CT scanner completes whole liver tumors, detection and characterization of
scanning within no more than 1.8 s, and lesions, and safety.
hepatic arterial dominant-phase CT images
obtained immediately after intravenous
bolus injection of iodinated contrast mate- DEVELOPMENT OF
rial permit detection of tiny hypervascular HEPATOCELLULAR
HCC foci in the liver.
In the mid 1980s, MRI had been reported
CARCINOMA, ITS
as an efficient diagnostic tool for HCCs, VASCULATURE, AND
but only conventional spin-echo sequences VASCULAR ENDOTHELIAL
with lengthy acquisition time were avail- GROWTH FACTOR
able in the clinical setting. By the early EXPRESSION
1990s, fast gradient-echo sequences that
allowed whole liver scanning during a Large regenerative nodule is the initial
breath-hold, became available and gado- primary form of nodular hepatocellular
linium-enhanced dynamic MRI of the lesions in cirrhosis, which has blood sup-
liver began to be used in daily radiologic ply from hepatic arteries and portal veins
practice. Magnetic resonance imaging has in similar proportion to normal liver tis-
continued to evolve in speed of data acqui- sue. Dysplastic nodule is a nodule more
sition and in contrast agent design, and progressed towards HCC than a large
it has maintained its status as the most regenerative nodule, and dysplastic nodule
accurate examination in the diagnosis of develops clone-like populations or nuclear
HCCs, which it initially acquired in the hyperchromasia. Dysplastic nodules are
mid 1990s (Semelka et al., 1992). By further categorized as low-grade and
allowing evaluation of T1 and T2 relaxa- high-grade dysplasias, with high-grade
tion, protein, hemorrhage, fat deposition, dysplasia showing histopathologic find-
metal deposition, blood flow, perfusion, ings similar to those of well-differentiated
and diffusion, MRI provides more abun- HCCs. High-grade dysplastic nodule dis-
dant information on underlying pathology plays cell density more than twice normal,
than CT for the diagnosis of diffuse and irregular nuclear contour, and invasion of
focal hepatic diseases. stroma or portal tracts, which are features
In the near future, MRI will continue to of HCC. The pathologic differentiation
improve in temporal and spatial resolu- of high-grade dysplasias from well-dif-
tions, develop and refine contrast agents ferentiated HCCs is often difficult, and
including liver-specific contrast agents, as a result lesions are occasionally called
develop diffusion-weighted imaging, and “borderline lesions” in the clinical setting.
allow performing perfusion and spectro- Large dysplastic nodules are perfused by
scopic studies on a routine clinical basis. both hepatic arteries and portal veins.
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 197

Researchers using combined CT during be highly promoted at this time, due to


arterial portography and CT hepatic arte- hypoxia. By this upregulation mechanism
riography to evaluate blood supply to (Figure 14.1A) (Viglietto et al., 1995),
hepatocellular nodules in cirrhosis, have early-stage HCC begins to create newly
suggested that hepatic arterial blood sup- developed arteries, referred to as “unpaired
ply was decreased in large regenerative
and dysplastic nodules compared with
that in background liver tissue (Lim et al.,
1999). Our current belief is that oxygen
supply to such nodules might be somewhat
decreased. We have found that VEGF
expression, determined by immunohisto-
chemistry, was moderately increased in
large regenerative nodules and high-grade
dysplastic nodules, which might reflect a
moderate hypoxic status of these nodules
(Kanematsu et al., 2004a).
Early-stage well-differentiated HCC
retains histologic structures similar to nor-
mal liver, they harbor portal triads as in nor-
mal liver, and are perfused by both hepatic
arteries and portal veins, although the
relative proportions of arterial and portal
venous blood supply to well-differentiated
HCCs is highly variable. In the beginning
phases, small well-differentiated HCCs, in
which cancer cells replace noncancerous
hepatocytes, are commonly hypovascu-
lar and the proportion of arterial blood Figure 14.1. Flow charts showing mechanisms of
supply is somewhat decreased. As can- VEGF upregulation and downregulation. A, Inefficient
cer cells invade into sinusoids, which is vascular supply and the resultant decrease in tissue
referred to as “stromal invasion”, portal oxygen tension lead to neovascularization to sat-
triads decrease in number to no more than isfy the needs of tissue. In human tissues, hypoxia
induces upregulation of VEGF gene expression by
one fourth of those in normal liver. In this biomolecular mechanism, i.e., HIF-1α and HIF-2α,
early stage, well-differentiated HCC lacks produced by the signal of transformation from oxy-
both hepatic arterial and portal venous hemoglobin into deoxyhemoglobin induce VEGF
blood supply, and is in a hypoxic status peptides synthesis via mRNA, and hyperoxia is
in the liver. Consequently, early-stage, brought about by way of newly developed blood
hypovascular well-differentiated HCCs vessels by angiogenesis. B, Hyperoxia eventually
brought about in previously hypoxic tissues by
appear hypovascular on both CT during angiogenesis transforms deoxyhemoglobin back to
arterial portography and CT hepatic arte- oxyhemoglobin, which suppresses VEGF peptide
riography (Hayashi et al., 1999). Vascular synthesis via HIF-1 α/2α and mRNA, which is
endothelial growth factor expression may referred to as downregulation
198 M. Kanematsu et al.

arteries”, by recruiting sinusoidal endothe- defined nodular appearance of the tumor.


lial cells to supply oxygen to the tumor. In hypervascular nodular HCCs with
Angiogenesis may be vigorous until the fibrous pseudocapsules, blood entering
lesion acquires abundant oxygen by way the tumor through hepatic arteries circu-
of unpaired arteries. This phase occurs late in sinusoids or capillarized vessels in
when lesion size is ∼10–15 mm. However, the tumor, then outflow into portal veins
the lesion may at this time still not appear or sinusoids in the surrounding noncan-
hypervascular on imaging studies. cerous liver tissue through portal venules
As early-stage HCC grows up to 15 mm penetrating the fibrous pseudocapsules,
in size, the number of portal triads sub- and finally drain into the hepatic veins.
stantially decreases, and a greater number This distinctive blood flow pathway in
of small unpaired arteries develop from hypervascular nodular HCCs was well-
the residual portal triads in the tumor. demonstrated by Ueda et al. (1998) using
At this stage of development, sinusoid- single-level cine CT during conventional
like vascular spaces contribute to tumor hepatic arteriography. Ring enhancement
vascularity, and they develop vigorously, that is seen in the late hepatic arterial phase
which is referred to as “capillarization”. was termed ‘corona enhancement’, which
Once lesions exceed 15 mm in size, HCCs may represent summation of enhance-
often acquire increased hepatic arterial ment of pseudocapsules with high vessel
vascularity, which is observed on conven- density and enhancement of peritumoral
tional angiograms, contrast-enhanced CT, sinusoids. The development of ‘corona
and MRI, as hypervascular enhancement. enhancement’ can also be observed on
When an HCC is shown as a hypervascular intravenously contrast-enhanced CT and
lesion on radiographic imaging such as CT MRI if two serial scans are run during
or MRI, it usually no longer has appreci- hepatic arterial-dominant phase, using
able portal venous perfusion, and they are high-speed CT (Murakami et al., 2001)
referred to as “overt or frank HCC”. or MRI (Shimizu et al., 2003).
Once a small well-differentiated HCC The growth of HCCs from 15 mm to
acquires ample hepatic arterial blood sup- 3 cm in size, is usually accompanied
ply with concomittent high oxygen tension, by marked hypervascularity. The tumor
expression of VEGF may be suppressed by grade of these very hypervascular HCCs
downregulation (Figure 14.1B). The HCC with fibrous pseudocapsules is often
continues to grow in size owing to its bio- moderately-differentiated. The HCC is
logical malignant behavior and still creates well supplied with oxygen because of
more tumor arteries, thereby maintaining the abundant hepatic arterial vascularity
oxygen supply. Well-differentiated HCC and this situation may be allegorically
progressively dedifferentiates to moderately explained as “satisfactory status”. Such
differentiated HCC, at which stage they do an HCC often is in a hyperoxic status,
not harbor portal triads and are almost and therefore, does not need to further
exclusively perfused by hepatic arteries. generate tumor arteries by angiogenesis,
Generally, most HCCs 2 cm in size with the result that VEGF activity may
or greater have macroscopic fibrous be suppressed due to downregulation
pseudocapsules that result in a clearly mechanism (Figure 14.1B).
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 199

As the HCC grows beyond 3 cm in size, ing in angiograms, and VEGF protein was
the distance between tumor vessels and substantial in HCC cells on immunohis-
cancer cells tends to increase. As shown by tochemical observation. In the same year,
Folkman (1971), oxygen diffuses no more Suzuki et al. (1996) studied vascular per-
than 150 μm in tissue, thus some parts of meability factor (VPF)/VEGF transcript
the enlarged HCC develop hypoxia and expression using reverse-transcription
resultant internal necrosis. The hypoxia polymerase chain reaction (PCR) analy-
induces the generation of VEGF peptides sis, and reported that 16 (69.6%) cases
once again. However, the generation of showed VEGF transcript expression in
tumor vessels is physically limited when the tumor, whereas only 9 (39%) showed
the HCC reaches 5–15 cm in size, due to it in adjacent liver parenchyma. Vascular
tissue pressure and cell crowding, and large endothelial growth factor mRNA expres-
HCCs may require anaerobic metabolism sion in HCCs in their study was associated
with glycolytic pathway with hexokinase with fibrous capsule formation and septal
II to sustain themselves (Gwak et al., formation. Vascular endothelial growth
2005). This is observed on imaging stud- factor activity was not correlated with the
ies as HCCs measuring 5–15 cm in size vascularity of HCCs as determined by con-
often do not enhance well on contrast- ventional angiography.
enhanced hepatic arterial-phase CT or MRI, El-Assal et al. (1998) performed quantita-
despite highly activated VEGF expression. tive reverse-transcription PCR and immu-
Yamaguchi et al. (1998) described that sar- nostaining in 71 patients with HCCs to
comatous HCCs, the final form of dediffer- determine microvessel density (MVD) and
entiation with highly malignant behavior, VEGF expressions in HCCs and in the sur-
showed low VEGF expression in immuno- rounding liver. They reported that tumor vas-
histochemistry studies. cularity on angiography was not correlated
with the MVD, and neither VEGF mRNA
levels nor protein expression in HCCs was
CORRELATION OF VASCULAR correlated with the MVD or any histopatho-
ENDOTHELIAL GROWTH logical features of HCCs, although they did
FACTOR EXPRESSION AND not directly correlate VEGF expression and
angiographic findings.
RADIOLOGIC FINDINGS Kwak et al. (2001) attempted to deter-
IN HEPATOCELLULAR mine whether VEGF is a histopathological
CARCINOMA IN PRIOR factor influencing contrast enhancement of
REPORTS HCC by qualitatively evaluating 22 nodular
HCCs on intravenously contrast-enhanced
Mise et al. (1996) performed Northern CT in correlation with the qualitative
blot analysis and immunohistochemistry to extent of anti-VEGF antibody expres-
semi-quantify and localize the expression sion determined by immunohistochemical
of VEGF in 20 HCCs and nine metastatic staining. They reported that the degree of
tumors, and reported that the degree of enhancement on the hepatic arterial-phase
VEGF mRNA expression was significantly CT had a significant positive correlation
correlated with the intensity of tumor stain- with VEGF expression in HCCs.
200 M. Kanematsu et al.

OUR PREVIOUS 4 were moderately-differentiated HCCs.


RESEARCH WITH The one large regenerative nodule and one
IMMUNOHISTOCHEMISTRY dysplastic nodule showed immunoreactiv-
ity for VEGF of grade 2. No significant
We evaluated 16 patients with HCC, 1 correlation was found between the tumor
with a large regenerative nodule, and 1 grade and the grade of immunoreactiv-
with a high-grade dysplastic nodule, who ity for VEGF. No correlation was found
underwent surgical resection of lesions between the lesion size and the grade of
at University of North Carolina from immunoreactivity for VEGF. The contrast-
December 1999 to April 2002 (Kanematsu to-noise ratios on T2-weighted images
et al., 2004a). Magnetic resonance imaging were higher (P < 0.05) with immunore-
included a breath-hold T1-weighted in- activity for VEGF of grade 4 than with
phase spoiled gradient-echo and a breath- those of grades 1, 2, and 3, and were
hold T2-weighted short-tau inversion marginally higher (P < 0.07) than those
recovery echo-train spin-echo imaging. of grade 0. No correlation was found
Spoiled gradient-echo images were acquired between contrast enhancement indexes on
before and after intravenous bolus injection postcontrast early and late-phase images
of gadolinium chelate. Postcontrast early- and immunoreactivity for VEGF.
phase imaging was initiated at 18 s, and A previous pathologic study by Yamaguchi
late-phase imaging was at 2 min. Lesion-to- et al. (1998) reported that in HCCs of
liver contrast-to-noise ratio was measured. single histologic grade, VEGF expres-
Contrast enhancement index was calculated sion was the highest in well-differentiated
by subtracting the contrast-to-noise ratio HCCs, followed by moderately differenti-
on precontrast images from that on post- ated HCCs, and then poorly differentiated
contrast images. Two blinded radiologists HCCs. In our study, although no statisti-
reviewed the images for signal intensity. cal correlation was found between tumor
Immunohistochemical staining was per- grade and immunoreactivity for VEGF,
formed using the standard avidin-biotin immunoreactivity for VEGF was the high-
peroxidase complex technique. An expe- est in moderately-differentiated HCCs:
rienced liver pathologist evaluated immu- five (83%) of six well-differentiated HCCs
noreactivity for VEGF semi-quantitatively: showed weak to moderate immunoreactiv-
grade 0 stood for virtually no immunore- ity for VEGF and none of them showed
activity, grade 1 for patchy to diffuse weak strong or intense immunoreactivity, but
immunoreactivity, grade 2 for patchy to four (67%) of six moderately differen-
diffuse moderate immunoreactivity, grade tiated HCCs showed strong to intense
3 for patchy to diffuse strong immunore- immunoreactivity for VEGF. The reason
activity, and grade 4 for diffuse intense for the discrepancy between our study
immunoreactivity. The histologic diagnosis and that of Yamaguchi et al. (1998) is
of hepatic nodules was based on the crite- unclear. There might be a difference in
ria described by the International Working subjective pathologic evaluation of tumor
Party (1995). grades or in composition of tumor grades:
All four hepatic nodules that showed Yamaguchi’s study included ten small
immunoreactivity for VEGF of grade 3 or well-differentiated HCCs of 13.0 ± 9.8 mm
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 201

in diameter, in our study six well-differen- activity upregulated by hypovascularity or


tiated HCCs measured slightly larger 22 ± hypoxia, there may be a future propensity
10 mm. It may be that Yamaguchi’s popu- to increased vascularity and development
lation included more small hypovascular into a hypervascular HCC.
well-differentiated HCCs, which would In two HCCs with diffuse intense immu-
predictably have higher VEGF expression noreactivity for VEGF the lesion-to-liver
than tumors of a larger size as observed contrast-to-noise ratios on T2-weighted
in our study. Another reason might be images were significantly higher than
epidemiologic differences associated with those of HCC with no, mild, moderate,
underlying liver diseases between North and strong immunoreactivity for VEGF.
American and Asian populations. This observation suggests that the HCCs
Our study included a large regenerative with intense immunoreactivity for VEGF
nodule and a high-grade dysplastic nodule, contained a greater amount of free water
and both expressed patchy to diffuse mod- in the extracellular or interstitial spaces,
erate immunoreactivity for VEGF. To our reflected by higher T2 signal, possibly due
knowledge, immunoreactivity of VEGF in to increased vascular permeability caused
large regenerative or dysplastic nodules in by activated VEGF. Vascular permeability
cirrhosis has not been assessed in previous is known to be increased by activation
studies. Although the few number of these of VEGF peptides (Shweiki et al., 1992;
hepatic nodules precluded statistical power, Monacci et al., 1993).
our results suggest that benign or border- In our study using immunohistochemis-
line hepatic nodules in cirrhosis, such as try, we found no correlation between arterial
large regenerative or dysplastic nodules, vascularity observed on postcontrast early-
may show an increased immunoreactivity phase images and VEGF activity. Our belief
for VEGF, and therefore, may have angio- is that small, well-differentiated HCCs may
genetic potential prefiguring further devel- well have increased VEGF activity that
opment of well-differentiated HCCs. serves to promote vascularity and tumor
It is believed that an HCC develops in a growth, but when well-differentiated HCCs
background of cirrhosis frequently by means are well vascularized and have increased in
of a multi-step dedifferentiation processes size, VEGF activity decreases, perhaps by
that progress from large regenerative to dys- downregulation mechanism (Figure 14.1B).
plastic nodule and to HCC (International Vascular endothelial growth factor activ-
Working Party, 1995). Regarding vascu- ity may occur temporally as a precursor to
larity in precancerous hepatic nodules in actual vessel presence, and therefore may
cirrhosis, one report described that large not correlate directly with vascularity as
regenerative nodules in cirrhosis were mini- reflected by intensity of early enhancement.
mally enhancing nodules surrounded by We thought this might explain the poor
enhancing fibrous septa at CT hepatic correlation between early enhancement
arteriography in 15 of 20 patients with on gadolinium-enhanced MRI and VEGF
cirrhosis (Lim et al., 1999). We believe activity that we observed, and determined
that most of large regenerative and dys- to perform further studies to elucidate these
plastic nodules are iso- or hypovascular, questions. Signal intensities on unenhanced
and if they have an increased VEGF T1- and T2-weighted images were more
202 M. Kanematsu et al.

closely associated with immunoreactivitytumorous liver parenchyma resected at


for VEGF than vascularity evaluated on Gifu University Hospital. Among them,
gadolinium-enhanced MRI, which may 22 patients underwent preoperative MRI
suggest that these features, reflecting of the liver within 2 weeks of surgery, and
higher fluid content, occur before vessel
20 patients underwent preoperative angi-
development. ographically assisted CT with combined
There were some limitations to our CT during arterial portography and CT
study: first, the study population was small
hepatic arteriography. Of the 28 patients, 6
reflecting that we excluded many patients,
patients had type B viral hepatitis, and 22
including those in whom pathologic diag-had type C viral hepatitis. No patient had
nosis was established by biopsy, becausea history of alcohol abuse. The underlying
VEGF activity could not be determined. liver disease documented by histopatho-
This study was retrospective, indicatinglogic study was chronic hepatitis due to
that this was a new concept to us to mild cirrhosis in 7 patients, moderate cir-
evaluate the relationship between VEGF rhosis in 16, and severe cirrhosis in 5.
activity and MRI findings of several histo-For immunoblotting techniques, the
logic types of hepatic nodules. A prospec-
specimen was sectioned through the tumor
tive, randomized, multi-institutional study
center to ensure correlation with CT and
with a larger population would be neces-MRI. Five-gram samples were dissolved in
sary. Although we evaluated the VEGF 1 ml of radioimmunoprecipitation buffer.
expression only in hepatic nodules, the Cell lysates were subjected to 10% sodium
background liver parenchyma or large dodecyl (lauryl) sulfatepolyacrylamide gel
regenerative nodules in cirrhosis also show
electrophoresis. Proteins were transferred
varying degree of VEGF activity. Although
to polyvinylidene difluoride membranes.
none of the hepatic nodules or background
After blocking membranes with tris-buff-
livers had significantly decreased signal
ered saline containing polysorbate 20 and
intensity due to iron deposition, iron depo-
5% skim milk, membranes were incubated
sition would affect the MR signal intensi-
with anti-VEGF monoclonal antibody and
ties and expected results. then with antimouse IgG coupled with
horseradish peroxidase. Detection was
performed using enhanced chemilumines-
MAGNETIC RESONANCE cence. Vascular endothelial growth factor
AND COMPUTED expression, observed as electrophoretic
bands, was quantified using image analysis
TOMOGRAPHIC DATA software, which calculated the area under
CORRELATED TO VASCULAR the histograms. Each pair of HCC sam-
ENDOTHELIAL GROWTH ples and surrounding liver was examined
FACTOR MEASURED USING using recombinant human VEGF solution
WESTERN BLOTTING (1.25 mg/ml) for calibration purpose. The
VEGF expression index (VEGFIND) was
In a second study using Western blot- calculated by dividing the area under the
ting to determine VEGF expression, we histogram for the specimen band by that
evaluated 28 HCCs with surrounding non- of the calibration band.
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 203

Magnetic resonance imaging was per- fat deposition in HCCs, the phase-shift
formed in a similar fashion to our prior index was calculated as follows: phase-
study obtaining T1-weighted gradient- shift index = (SI in-phase – SI opposed-
echo, T2-weighted echo-train spin-echo, phase)/SI in-phase, where SI in-phase and
and gadolinium-enhanced multiple-phase SI opposed-phase are the signal intensi-
gradient-echo images in the hepatic arte- ties of HCC on in-phase and opposed-
rial-dominant, portal venous, and equilib- phase T1-weighted gradient-echo images,
rium phases. Computed tomography was respectively.
performed using a combination of unen- To measure CT values in HCCs and in
hanced CT, CT during arterial portogra- the surrounding liver, a circular region
phy, and CT hepatic arteriography. On of interest was drawn to encompass as
CT portography, scan was started 25–35 s much of the HCC as possible, and another
after the initiation of a transcatheter supe- was drawn in the surrounding liver paren-
rior mesenteric arterial injection of 95 ml chyma. Quantitative degree of contrast
of nonionic contrast material containing enhancement was expressed as contrast
150 mg iodine/ml at 3 ml/s, and on CT enhancement index, which was calculated
arteriography scan was started 5 s after by subtracting the CT values on unenhanced
the initiation of a transcatheter common CT from those on contrast-enhanced CT.
hepatic arterial injection of 30 ml of the Two experienced gastrointestinal radi-
same contrast at 1.5 ml/s. ologists independently reviewed MRI in
This study evaluated MRI data using a retrospective manner. They subjectively
a quantitative approach. Our technique evaluated the signal intensity in HCC and
was as follows: to determine MR signal in the surrounding liver using a seven-point
intensity values and their standard devia- scale from “strong hypointensity (−3)” to
tions (SDs) in HCCs and in the surround- “marked hyperintensity (+3)”. Intensity of
ing liver, a circular region of interest was enhancement and degree of heterogeneity
drawn to encompass as much of the lesion in HCCs were determined on postcontrast
as possible, and another circular region of images using four-point scales from “vir-
interest was drawn in a region of the sur- tually no enhancement (0)” to “marked
rounding liver. The SD of the background enhancement (+3)” and from “virtually no
noise, SDB, was measured in the phase- heterogeneity (0)” to “marked heterogene-
encoding direction outside the anterior ity (+3)”, respectively.
abdominal wall to calculate the following: The two radiologists independently
lesion-to-liver contrast-to-noise ratio = (SI reviewed CT findings in a retrospective
lesion – SI liver)/SDB, where SI lesion manner. They evaluated the contrast-
and SI liver are the signal intensities of enhanced CT with reference to unen-
the HCC and of the surrounding liver, hanced CT to determine the degree of
respectively. As a quantitative parameter contrast enhancement in HCC and in the
of signal intensity heterogeneity, the sig- surrounding liver using a four-point scale
nal intensity SD ratio was calculated as from “virtually no enhancement (0)” to
follows: SD ratio = SD lesion/SDB, where “intense enhancement (+3)”. The radiolo-
SD lesion is the signal intensity SD of gists evaluated the degree of heterogeneity
the HCC. As a quantitative parameter of of hepatic arterial contrast enhancement in
204 M. Kanematsu et al.

HCC on CT hepatic arteriography using The qualitative degree of signal inten-


a four-point scale from “homogeneous sity heterogeneity of HCCs correlated
enhancement (0)” to “strong heterogeneity directly with the VEGFINDs of HCCs on
(+3)”. Statistical correlations were deter- opposed-phase T1-weighted gradient-echo
mined using simple regression analysis (0.64, P = 0.016), T2-weighted echo-train
for continuous data and the Spearman rank spin-echo (0.52, P = 0.038), postcontrast
correlation test for categorical data. hepatic arterial-phase gradient-echo (0.48,
The VEGFINDs of HCCs and of the sur- P = 0.045), and equilibrium-phase gradi-
rounding liver ranged from 0.46 to 9.27 ent-echo (0.56, P = 0.018) images.
(mean ± SD, 3.28 ± 2.29) and from 0.44 to On CT hepatic arteriography, the con-
5.91 (2.86 ± 1.49), respectively. In 14 (50%) trast enhancement indices of HCCs
of the 28 HCCs, the VEGFIND of the sur- showed moderate inverse correlation with
rounding liver was greater than that of the the VEGFINDs of HCCs (−0.57, P = 0.017)
corresponding HCC. Pathologic tumor size (Figures 14.2 and 14.3). The contrast
showed moderate direct correlation with enhancement indices of the surrounding
the VEGFINDs of HCCs (correlation coef- liver showed marginal, moderate direct
ficient: 0.40, P = 0.035). No correlation was correlation with the VEGFINDs of the sur-
found between tumor sizes and VEGFINDs rounding liver (0.44, P = 0.076). On CT
of the surrounding liver. The 28 HCCs were during arterial portography, no significant
predominantly well-differentiated in 3, correlation was found between the con-
moderately differentiated in 20, and poorly trast enhancement index and VEGFIND.
differentiated in 5. The mean VEGFIND of Qualitatively, the degree of heterogeneity
HCCs tended to be higher with poorly dif- of hepatic arterial contrast enhancement
ferentiated HCCs than with the other histo- in HCCs on CT hepatic arteriography
logic tumor grades, although no statistical showed moderate direct correlation with
significance was found. the VEGFINDs of HCCs (0.55, P = 0.033).
The contrast-to-noise ratios correlated Half of the 28 HCCs in our series had
inversely with the VEGFINDs of HCCs on a VEGFIND lower than in the surrounding
opposed-phase T1-weighted gradient-echo liver (Figure 14.3), which is comparable
images (−0.46, P = 0.038), and correlated to other reports that described 40–59% of
directly with the VEGFINDs of HCCs on HCCs have VEGF activity that is either
T2-weighted echo-train spin-echo images equal to or less than that in the surround-
(0.49, P = 0.025) (Figure 14.2). The ing liver (Suzuki et al., 1996, El-Assal
contrast-to-noise ratios on gadolinium- et al., 1998). Some of these HCCs showed
enhanced hepatic arterial-phase gradient- considerably greater arterial enhancement
echo images were marginally and inversely compared to the surrounding liver (Figure
correlated with the VEGFINDs of HCCs 14.3). This emphasizes that presence of
(−0.39, P = 0.081) (Figure 14.2). The SD VEGF does not correlate with the pres-
ratios of HCCs correlated directly with the ence of vascularity; rather it is a precursor
VEGFINDs of HCCs on T2-weighted echo- to vascularity. The biological interaction
train spin-echo images (0.44, P = 0.044). between the VEGF activity in an HCC
No correlation was found between phase- and in the surrounding liver has yet to be
shift indices of HCCs and VEGFINDs. determined.
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 205

a b

c d

Figure 14.2. A 57-year-old man with chronic type C viral hepatitis and a poorly differentiated 5.8-cm
HCC showing high VEGF expression. Child-Pugh grade was A. a, Fat-suppressed T2-weighted echo-
train spin-echo (repetition time [TR] of 4,286 ms, echo time [TE] of 80 ms) axial image shows the HCC
(arrow) as a moderately hyperintense lesion with internal areas of higher signal intensity (arrowheads)
due to internal necrosis. b, On gadolinium-enhanced hepatic arterial-phase T1-weighted gradient-echo
(TR of 150 ms, TE of 1.6 ms) axial image, the HCC is totally hypovascular other than its peripheries,
which are slightly enhanced (arrows). Central areas corresponding to hyperintense internal areas on a
remain unenhanced (arrowheads). c, CT hepatic arteriogram shows weak hepatic arterial enhancement
with moderate heterogeneity in the HCC (arrow). Note that central areas of the HCC exhibit water density
with no enhancement due to necrosis. d, Scheme shows electrophoretic bands and corresponding histo-
grams in this patient; 1.25 mg/ml of VEGF solution was used for calibration. Areas of the histogram were
376 pixels for the calibration band, 3,485 pixels for the HCC band, and 1,395 pixels for the surrounding
liver band. The VEGFIND values were 9.27 in the HCC and 3.71 in the surrounding liver. Note that electro-
phoretic peaks adjacent to those of HCC and liver are due to expression of irregular protein. (Permission
to reproduce granted by the American Journal of Roentgenology.)

Results of correlation between VEGF employed in VEGF activity evaluation


expression in HCC tissues and tumor (Yamaguchi et al., 1998; Kwak et al.,
size have varied among researchers, partly 2001; Jeng et al., 2004; Ng et al., 2001).
this may reflect different methodologies Although we found a positive correlation
206 M. Kanematsu et al.

a b

Figure 14.3. A 74-year-old woman with chronic type C viral hepatitis and a moderately differentiated
2-cm HCC showing weak VEGF expression. Child-Pugh grade was A. a, CT hepatic arteriogram shows
very intense, homogeneous hepatic arterial enhancement in the HCC (arrow). b, Scheme shows elec-
trophoretic bands and their corresponding histograms in this patient; 1.25 mg/ml of VEGF solution was
used for calibration. Areas of histogram were 480 pixels for the calibration band, 761 pixels for the HCC
band, and 2,375 pixels for the surrounding liver band. The VEGFIND values were 1.59 in the HCC and
4.95 in the surrounding liver. Note that electrophoretic peak adjacent to that of liver is due to expression
of irregular protein. (Permission to reproduce granted by the American Journal of Roentgenology)

between VEGF expression in HCCs and level of VEGF mRNA in HCC tissues did
tumor size, Yamaguchi et al. (1998), who not significantly correlate with tumor size,
immunohistochemically investigated the cellular differentiation, capsule, daughter
expression of VEGF in HCCs, found that nodules, vascular permeation, necrosis and
VEGF positivity gradually decreased with hemorrhage of tumors. Ng et al. (2001)
an increase in tumor size. It may be similarly reported that there was no sta-
that because the population they evalu- tistically significant association between
ated included a greater number and per- tumor VEGF mRNA levels and tumor size
centage of small (15 mm or less) HCCs or other pathologic features. Nevertheless,
than other reports, that they might have our opinion is that positive correlation
observed VEGF activity changes during between VEGF expression and tumor
the continuous dedifferentiation in small size is reasonable, because intratumoral
HCCs. Kwak et al. (2001) also observed hypoxia or necrosis induced by tumor
that tumor size correlated negatively with growth may induce VEGF protein synthe-
immunohistochemical VEGF expression. sis (Shweiki et al., 1992; Mukhopadhyay
While, Jeng et al. (2004) found that the et al., 1995; von Marschall et al., 2001).
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 207

Hepatocellular carcinomas are often vascular permeability regulated by VEGF


hypo- to isovascular on radiologic images peptides results in increased free water in
during the early stage of development, the extracellular or interstitial spaces of
typically during the stage of small well- HCCs and thus causes T1- and T2 relaxa-
differentiated HCC (Hayashi et al., 1999), tion time prolongation in HCCs.
and intratumoral oxygen tension at this time The contrast-to-noise ratios on gado-
is believed to be low. Such a hypoxic state linium-enhanced hepatic arterial-phase
may induce neovascularization in HCCs, MRI were marginally and inversely cor-
which would result in an increased supply related with the VEGFINDs, and the con-
of arterial blood and concomitant oxygen trast enhancement indices of HCCs on
supply to the HCC, which then leads to the CT hepatic arteriography were moderately
development of a hypervascular HCC that inversely correlated with the VEGFINDs
is typically referred to as a moderately to of HCCs, which indicated that the more
poorly differentiated HCC. Once a hyper- hypervascular is an HCC, the weaker is the
vascular HCC has developed, VEGF gene VEGF expression in the tumor (Figures
expression may be suppressed due to the 14.2 and 14.3). It is known that an inef-
downregulation of VEGF (Figure 14.1B). ficient vascular supply and the resultant
This might explain prior observations that reduction in tissue oxygen tension, often
the intensity of VEGF expression in HCCs leads to neovascularization in order to
is related to the histological tumor grade, maintain the nutrition of tissue. In human
i.e., highest in well-differentiated HCCs, HCC, it has been suggested that hypoxia
and lowest in poorly differentiated HCCs induces an upregulation of VEGF gene
(Yamaguchi et al., 1998). expression by biomolecular mechanism
In our study the HCC-to-liver contrast- (Figure 14.1A) (Shweiki et al., 1992), i.e.,
to-noise ratios on T1-weighted gradient- hypoxia inducible factor (HIF)-1α and
echo images inversely correlated with the HIF-2α are upregulated by hypoxia and
VEGFINDs of HCCs, and contrast-to-noise induce pro-angiogenic peptide formation
ratios on T2-weighted echo-train spin- and VEGF expression (Mukhopadhyay
echo images directly correlated with the et al., 1995).
VEGFINDs of HCCs, which indicates that Heterogeneity of HCC on CT or MRI
the stronger the VEGF expression is in correlated directly with the VEGFINDs of
HCCs, the more prolonged are the T1- HCCs, which indicated that the more het-
and T2-relaxation times of HCCs (Figure erogeneous the tumor signal or enhance-
14.2). T1- and T2-relaxation time prolon- ment, the stronger the VEGF expression
gation commonly occurs in tissues con- (Figures 14.2 and 14.3). Heterogeneity
taining increased amounts of free water in of HCC on unenhanced MRI may be
extracellular or interstitial spaces, typically explained by an uneven distribution of
as seen in malignant tumors. Moreover, it extracellular free water or more com-
is known that VEGF, which was previ- monly by the presence of intratumoral
ously termed vascular permeability factor necrosis. Heterogeneity of HCC on gado-
(VPF), raises the permeability of blood linium-enhanced MRI and on CT hepatic
vessels (Shweiki et al., 1992; Monacci arteriography may also be indicative of
et al., 1993). We suspect that increased unevenness of vascularity which results
208 M. Kanematsu et al.

in regions of tumor with decreased oxy- microvascular density and VEGF expres-
gen tension. Previous research indicated sion than noncirrhotic livers. The reason
that the hypoxic regions of solid tumors why in our study the correlations between
produced powerful and directly acting the intensity of hepatic arterial enhance-
angiogenic proteins such as VEGF, or that, ment and of VEGF expression differed
in an experimental model using rat livers, between HCCs and the surrounding liver
VEGF peptides were produced by non- was unclear, this discrepancy in part may
parenchymal and parenchymal cells after be associated with the difference in mag-
necrosis (Mukhopadhyay et al., 1995). nitude between hepatic arterial perfusion
Fat deposition is observed in various in a HCC and in the surrounding liver: the
types of HCCs. Kutami et al. (2000) contrast enhancement indices of HCCs
reported that fatty changes in small HCCs ranged from 33 to 236 Hounsfield unit
are closely related to tumor size, histologic (HU) (mean, 105.3 ± 51.7 HU), whereas
grade, and the insufficient development the contrast enhancement indices of the
of arterial tumor vessels. Previous stud- surrounding liver ranged from 9 to 68 HU
ies have demonstrated that lipid bodies (mean, 28.9 ± 15.7 HU). The increased
in endothelial cells are induced during oxygen supply to the HCC secondary to
hypoxia in any cell type (Scarfo et al., neovascularization was sufficiently great
2001). Based on these previous results, to suppress VEGF expression via VEGF
we suspect that VEGF expression, hepatic downregulation (Figure 14.1B), but the
arterial perfusion, hypoxia, and fat deposi- increased oxygen supply in the surround-
tion in HCCs are closely related. However, ing liver might not be sufficient to cause
in our study, we did not observe a cor- VEGF downregulation (Figure 14.3).
relation between fat deposition of HCCs The most contentious aspect of VEGF
and VEGF expression, where phase shift correlation is the relationship with the con-
imaging accurately reflects intratumoral comitant presence of increased vascularity.
fat deposition. Kwak et al. (2001) correlated tumor atten-
The contrast enhancement index uation qualitatively, and determined on
of the surrounding liver on CT hepatic intravenously contrast-enhanced CT scans
arteriography showed moderate direct with immunoreactivity for anti-VEGF
correlation with the VEGFINDs of the sur- antibody, which was qualitatively deter-
rounding liver with marginal statistical mined by immunohistochemical staining.
significance, which suggested that the They concluded that the degree of VEGF
greater the hepatic arterial enhancement expression in HCC was directly correlated
of the surrounding liver, the stronger is the with the degree of contrast enhancement
VEGF expression in the liver. Rosmorduc during the hepatic arterial phase. The
et al. (1999) described experimental bil- exact reasons why their results differ from
iary cirrhosis, indicating that there was ours (Kanematsu et al., 2004b, 2005) in
evidence that angiogenesis was stimulated terms of the correlation between the inten-
primarily by VEGF in response to hepa- sity of hepatic arterial tumor enhancement
tocellular hypoxia caused by liver dam- and VEGF expression in HCC is unclear.
age. El-Assal et al. (1998) reported that Eighteen (82%) of 22 patients had type-B
cirrhotic livers had a significantly higher hepatitis and 1 (5%) had type-C hepatitis
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 209

in their study, whereas in our study only histologic tumor grades was uneven.
6 (21%) of 28 patients had type-B hepa- Multi-institutional studies will be needed
titis and 22 (79%) had type-C hepatitis. to confirm our results. Also, although
This substantial difference in the patient we employed Western blotting to semi-
populations and in the underlying hepatic quantify VEGF peptides, this technique is
disease might affect the results. The sensi- limited in terms of its ability to differenti-
tivity for enhancement and timing of data ate the multiple types of VEGF peptides
acquisition of single detector row spiral that exist in cell membranes, cytoplasm,
CT, and subjective ratings of contrast and interstitial spaces.
enhancement on CT images and of VEGF
expression may also affect statistical cor-
relations. SUMMARY
Some researchers have reported that
VEGF activity does not correlate with We have reviewed the historical devel-
the vascularity of HCCs as determined opment of various diagnostic imaging
by conventional angiography (Suzuki modalities for the evaluation of HCC.
et al., 1996; El-Assal et al., 1998), while We further described the research pub-
others have reported that VEGF activity lications on the correlation of VEGF
correlates directly with the intensity of and imaging findings. The apparently
tumor stain on angiography (Mise et al., discrepant observations of the extent of
1996). All these reports focused on the correlation of VEGF activity and HCC,
clinical significance of VEGF expression we believe, reflects that different studies
in HCC and in the surrounding liver, the contained patients with HCCs of different
evaluation of VEGF activity by means evolution. Based on our research studies,
of VEGF mRNA quantification with we have observed that VEGF appears
Northern blotting, and VEGF localiza- to correlate with increased tissue fluid,
tion by immunohistochemical staining, which on MRI appears as low signal on
while the radiological assessment of the T1-weighted images and high signal on
degree of tumor staining by angiogra- T2-weighted images. It is our impres-
phy was performed by subjective rat- sion that VEGF increases as a precursor
ings using a two- or three-point scale. to increased vascularity, as vascularity
We believe that our study methodology increases, seen in small HCCs, VEGF
more accurately clarified the relation- is downregulated and as tumor increases
ship between the vascularity of HCCs to larger size, > 3 cm, VEGF may again
and liver and VEGF activity, as contrast increase in response to hypoxia.
enhancement was quantitatively deter- The future clinical impact of VEGF
mined using region-of-interest measure- measurements and correlation of VEGF
ments and VEGF was determined using values with imaging results have yet to
Western blotting technique. be determined. Our hope is that VEGF
Limitations of our studies have included measurements will provide insight into the
that the study populations were small, angiogenic potential of tumors, and also
reflecting single institution accrual, predict or reflect response to therapeutic
and distribution of varying types of interventions.
210 M. Kanematsu et al.

REFERENCES Yokoyama, R., Hoshi, H., and Moriyama, N.


2004b. Expression of vascular endothelial
El-Assal, O.N., Yamanoi, A., Soda, Y., Yamaguchi, M., growth factor in hepatocellular carcinoma and
Igarashi, M., Yamamoto, A., Nabika, T., and the surrounding liver: correlation with angi-
Nagasue, N. 1998. Clinical significance of ographically assisted CT. Am. J. Roentgenol.
microvessel density and vascular endothelial 183: 1585–1593.
growth factor expression in hepatocellular car- Kanematsu, M., Osada, S., Amaoka, N., Goshima, S.,
cinoma and surrounding liver: possible involve- Kondo, H., Kato, H., Nishibori, H., Yokoyama,
ment of vascular endothelial growth factor in the R., Hoshi, H., and Moriyama, N. 2005.
angiogenesis of cirrhotic liver. Hepatology 27: Expression of vascular endothelial growth factor
1554–1562. in hepatocellular carcinoma and the surrounding
Folkman, J. 1971. Tumor angiogenesis: therapeutic liver and correlation with MRI findings. Am. J.
implications. N. Engl. J. Med. 285: 1182–1186. Roentgenol. 184: 832–841.
Gwak, G.Y., Yoon, J.H., Kim, K.M., Lee, H.S., Kutami, R., Nakashima, Y., Nakashima, O., Shiota, K.,
Chung, J.W., and Gores, G.J. 2005. Hypoxia and Kojiro, M. 2000. Pathomorphologic study
stimulates proliferation of human hepatoma cells on the mechanism of fatty change in small hepa-
through the induction of hexokinase II expres- tocellular carcinoma of humans. J. Hepatol. 33:
sion. J. Hepatol. 42: 358–364. 282–289.
Hayashi, M., Matsui, O., Ueda, K., Kawamori, Y., Kwak, B.K., Shim, H.J., Park, E.S., Kim, S.A.,
Kadoya, M., Yoshikawa, J., Gabata, T., Choi, D., Lim, H.K., Park, C.K., Chung, J.W.,
Takashima, T., Nonomura, A., and Nakanuma, and Park, J.H. 2001. Hepatocellular carcinoma:
Y. 1999. Correlation between the blood supply correlation between vascular endothelial growth
and grade of malignancy of hepatocellular nod- factor level and degree of enhancement by mul-
ules associated with liver cirrhosis: evaluation tiphase contrast-enhanced computed tomogra-
by CT during intraarterial injection of contrast phy. Invest. Radiol. 36: 487–492.
medium. Am. J. Roentgenol. 172: 969–976. Lim, J.H., Kim, E.Y., Lee, W.J., Lim, H.K., Do, Y.S.,
International Working Party. 1995. Terminology of Choo, I.W., and Park, C.K. 1999. Regenerative
nodular hepatocellular lesions. Hepatology 22: nodules in liver cirrhosis: findings at CT during
983–993. arterial portography and CT hepatic arteriogra-
Jeng, K.S., Sheen, I.S., Wang, Y.C., Gu, S.L., Chu, phy with histopathologic correlation. Radiology
C.M., Shih, S.C., Wang, P.C., Chang, W.H., and 210: 451–458.
Wang, H.Y. 2004. Is the vascular endothelial Mise, M., Arii, S., Higashituji, H., Furutani, M.,
growth factor messenger RNA expression in Niwano, M., Harada, T., Ishigami, S., Toda,
resectable hepatocellular carcinoma of prognos- Y., Nakayama, H., Fukumoto, M., Fujita, J.,
tic value after resection? World J. Gastroenterol. and Imamura, M. 1996. Clinical significance
10: 676–681. of vascular endothelial growth factor and basic
Kanematsu, M., Hoshi, H., Imaeda, T., Murakami, T., fibroblast growth factor gene expression in liver
Inaba, Y., Yokoyama, R., and Nakamura, H. tumors. Hepatology 23: 455–464.
1997. Detection and characterization of hepatic Monacci, W.T., Merrill, M.J., and Oldfield, E.H.
tumors: value of combined helical CT hepatic 1993. Expression of vascular permeability fac-
arteriography and CT during arterial portogra- tor/vascular endothelial factor in normal rat tis-
phy. Am. J. Roentgenol. 168: 1193–1198. sues. Am. J. Physiol. 264: 995–1002.
Kanematsu, M., Semelka, R.C., Leonardou, P., Morgan, B., Thomas, A.L., Drevs, J., Hennig, J.,
Mastropasqua, M., Armao, D., Vaidean, G., Buchert, M., Jivan, A., Horsfield, M.A., Mross,
Firat, Z., and Woosley, J.T. 2004a. Angiogenesis K., Ball, H.A., Lee, L., Mietlowski, W., Fuxuis, S.,
in hepatocellular nodules: correlation of MR Unger, C., O’Byrne, K., Henry, A., Cherryman,
imaging and vascular endothelial growth factor. G.R., Laurent, D., Dugan, M., Marme, D., and
J. Magn. Reson. Imaging 20: 426–434. Steward, W.P. 2003. Dynamic contrast-enhanced
Kanematsu, M., Osada, S., Amaoka, N., Goshima, S., magnetic resonance imaging as a biomarker for
Kondo, H., Nishibori, H., Kato, H., Matsuo, M., the pharmacological response of PTK787/ZK
14. Expression of Vascular Endothelial Growth Factor in Hepatocellular Carcinoma 211

222584, an inhibitor of the vascular endothe- Shweiki, D., Itin, A., Soffer, D., and Keshet, E.
lial growth factor receptor tyrosine kinases, in 1992. Vascular endothelial growth factor induced
patients with advanced colorectal cancer and by hypoxia may mediate hypoxia-initiated ang-
liver metastases: results from two phase I stud- iogenesis. Nature 359: 843–845.
ies. J. Clin. Oncol. 21: 3955–3964. Suzuki, K., Hayashi, N., Miyamoto, Y., Yamamoto, M.,
Mukhopadhyay, D., Tsiokas, L., Zhou, X.M., Ohkawa, K., Ito, Y., Sasaki, Y., Yamaguchi, Y.,
Foster, D., Brugge, J.S., and Sukhatme, V.P. Nakase, H., Noda, K., Enomoto, N., Arai, K.,
1995. Hypoxic induction of human vascular Yamada, Y., Yoshihara, H., Tujimura, T.,
endothelial growth factor expression through Kawano, K., Yoshikawa, K., and Kamada, T.
c-Src activation. Nature 375: 577–581. 1996. Expression of vascular permeability fac-
Murakami, T., Kim, T., Takamura, M., Hori, M., tor/vascular endothelial growth factor in human
Takahashi, S., Federle, M.P., Tsuda, K., Osuga, hepatocellular carcinoma. Cancer Res. 56:
K., Kawata, S., Nakamura, H., and Kudo, M. 3004–3009.
2001. Hypervascular hepatocellular carcinoma: Ueda, K., Matsui, O., Kawamori, Y., Nakanuma,
detection with double arterial phase multi-detector Y., Kadoya, M., Yoshikawa, J., Gabata, T.,
row helical CT. Radiology 218: 763–767. Nonomura, A., and Takashima, T. 1998.
Ng, I.O., Poon, R.T., Lee, J.M., Fan, S.T., Ng, M., Hypervascular hepatocellular carcinoma: evalu-
and Tso, W.K. 2001. Microvessel density, vas- ation of hemodynamics with dynamic CT during
cular endothelial growth factor and its receptors hepatic arteriography. Radiology 206: 161–166.
Flt-1 and Flk-1/KDR in hepatocellular carci- Viglietto, G., Maglione, D., Rambaldi, M., Cerutti,
noma. Am. J. Clin. Pathol. 116: 838–845. J., Romano, A., Trapasso, F., Fedele, M.,
Rosmorduc, O., Wendum, D., Corpechot, C., Ippolito, P., Chiappetta, G., and Botti, G. 1995.
Galy, B., Sebbagh, N., Raleigh, J., Housset, C., Upregulation of vascular endothelial growth
and Poupon, R. 1999. Hepatocellular hypoxia- factor (VEGF) and downregulation of placenta
induced vascular endothelial growth factor growth factor (PlGF) associated with malig-
expression and angiogenesis in experimental bil- nancy in human thyroid tumors and cell lines.
iary cirrhosis. Am. J. Pathol. 155: 1065–1073. Oncogene 11: 1569–1579.
Scarfo, L.M., Weller, P.F., and Farber, H.W. 2001. von Marschall, Z., Cramer, T., Hocker, M.,
Induction of endothelial cell cytoplasmic lipid Finkenzeller, G., Wiedenmann, B., and Rosewicz S.
bodies during hypoxia. Am. J. Physiol. Heart 2001. Dual mechanism of vascular endothelial
Circ. Physiol. 280: 294–301. growth factor upregulation by hypoxia in human
Semelka, R.C., Shoenut, J.P., Kroeker, M.A., hepatocellular carcinoma. Gut 48: 87–96.
Greenberg, H.M., Simm, F.C., Minuk, G.Y., Yamaguchi, R., Yano, H., Iemura, A., Ogasawara,
Kroeker, R.M., and Micflikier, A.B. 1992. Focal S., Haramaki, M., and Kojiro, M. 1998.
liver disease: comparison of dynamic contrast- Expression of vascular endothelial growth factor
enhanced CT and T2-weighted fat-suppressed, in human hepatocellular carcinoma. Hepatology
FLASH, and dynamic gadolinium-enhanced MR 28: 68–77.
imaging at 1.5 T. Radiology 184: 687–694. Yamane, A., Seetharam, L., Yamaguchi, S., Gotoh, N.,
Shimizu, A., Ito, K., Koike, S., Fujita, T., Shimizu, K., Takahashi, T., Neufeld, G., and Shibuya, M.
and Matsunaga, N. 2003. Cirrhosis or chronic 1994. A new communication system between
hepatitis: evaluation of small (< or = 2-cm) early- hepatocytes and sinusoidal endothelial cells in
enhancing hepatic lesions with serial contrast- liver through vascular endothelial growth factor
enhanced dynamic MR imaging. Radiology 226: and Flt tyrosine kinase receptor family (Flt-1
550–555. and KDR/Flk-1). Oncogene 9: 2683–2690.
15
Detection of Small Hepatic Lesions:
Superparamagnetic Oxide-Enhanced
Diffusion-Weighted T2 FSE Imaging
Shigeru Kiryu and Kuni Ohtomo

INTRODUCTION SUPERPARAMAGNETIC
IRON OXIDE-ENHANCED
The liver is a common site for metastases
MAGNETIC RESONANCE
from colorectal carcinoma and other neo-
plasms. Most colorectal carcinoma deaths IMAGING
are attributable to hepatic metastases, There are several methods such as com-
and the number of these metastases is puted tomography (CT), magnetic reso-
increasing (Ballantyne and Quin, 1993). nance imaging (MRI), and ultrasonography
The prognosis of patients without treat- for preoperative imaging assessment in
ment is dismal because most cases die detecting liver metastases. Presently, super-
within a few years after discovering the paramagnetic iron oxide (SPIO)-enhanced
liver lesion. Hepatic resection is the sole MRI is being widely used to detect liver
treatment that regularly leads to long- metastases. This oxide is taken up by
term survival with a possible chance of phagocytosis of the reticuloendothelial
cure in patients with hepatic metastases, system, including Kupffer cells of the liver,
especially those of colorectal origin. substantially shortening T2 of liver tissue
With techniques such as intraoperative (Stark et al., 1988). The hepatic metastases
ultrasonography (Makuuchi et al., 1991), that contain negligible or a few Kupffer
which improves the management of peri- cells remain largely nonenhanced, while
operative patients, hepatic resection has the normal liver is enhanced and shows
extended the possibility of liver surgery in a low signal intensity on T2-weighted
patients with advanced metastatic tumors images, with the result that the lesion-to-
(Kawasaki et al., 1994). Also, hepatic liver signal intensity ratio is improved on
resection has been proved to improve SPIO-enhanced MRI, as compared with
the prognosis of patients (Penna and nonenhanced T2-weighted images.
Nordlinger, 2002). Therefore, accurately In a multicenter trial, SPIO-enhanced MRI
detecting small lesions is indispensable revealed additional lesions not seen on
in preoperative imaging assessment when unenhanced images in 27% of cases and
planning hepatic resection. additional lesions not seen by conventional

213
214 S. Kiryu and K. Ohtomo

CT in 40% (Ros et al., 1995). In a mul- parenchyma; therefore, the small vessels
tiobserver study that compared the sen- and lesions exhibit similar signal intensities.
sitivities of dual phase spiral CT with Furthermore, SPIO-enhanced MRI dis-
SPIO-enhanced MRI for detecting metas- played more peripheral vascular structures
tases, both techniques showed high sen- than contrast enhanced CT (Fretz et al.,
sitivities for detecting lesions larger than 1990). Distinguishing small lesions from
1 cm at 94% vs. 99%, whereas the sen- the signal intensities from hepatic vessels
sitivities of dual phase spiral CT and is the key to detect small hepatic lesions.
SPIO-enhanced MRI for detecting lesions
smaller than 1 cm were 52% and 47%,
respectively (Ward et al., 1999). Computed
tomography during arterial portography DIFFUSION-WEIGHTED
(CTAP) has been regarded as a technique IMAGING FOR SUPPRESSION
that is more sensitive at detecting small OF SIGNALS FROM HEPATIC
lesions than dual phase spiral CT and VESSELS
SPIO-enhanced MRI, but CTAP is inva-
sive and has a greater risk of providing Diffusion-weighted imaging (DWI) is
false-positive results than other techniques sensitive to molecular diffusion, which
(Soyer et al., 1993). CTAP is also insensi- is the thermally induced motion of water
tive to lesions smaller than 1 cm (Young molecules in biological tissues called
et al., 1997). Therefore, the strategy to Brownian motion. The changes in proton
improve the detection rate of small lesions self-diffusion are an early indicator of
is anticipated. alterations of cellular homeostasis in acute
SPIO-enhanced MRI increases the rate ischemic stroke (Moseley et al., 1990);
of false-positive findings compared to therefore, DWI has a significant impact
spiral-CT (Muller et al., 1998), and these in the detection of early ischemic change
false-positive results are responsible for the and has played an important role in the
difficulty in distinguishing small lesions. clinical setting in the field of neuroradiol-
One of the common false-positive lesions is ogy. DWI is able to suppress the signals
a small hepatic cyst (Ward et al., 1999). Some of vessels and has been used to improve
techniques are recommended for detecting the sensitivity of the detection of small
hepatic cysts, such as T1-weighted breath- hepatic lesions (Okada et al., 1998).
hold GRE imaging after SPIO enhance- In the DWI pulse sequence, motion-
ment and short- and long-TE SSFSE probing gradient (MPG) pulses are
(Oudkerk et al., 1997; Kiryu et al., 2002). placed before and after a 180° RF pulse,
Adding these techniques to routine SPIO- which comprises the spin echo method
enhanced MRI, hepatic cysts can be distin- along with a 90° pulse. The protons
guished from true hepatic lesions. Another that have moved during or after the
cause for false-positives is the signals first MPG randomly change their spin
from hepatic vessels. SPIO-enhanced MRI phase as they move from one region
increases the signal of cross-sectioned of the magnetic field to another. Thus,
vessels in the low signal background liver the signal intensity of diffusion protons
15. Detection of Small Hepatic Lesions: Superparamagnetic Oxide-Enhanced 215

decreases whereas the slow diffusion PERIODICALLY ROTATED


appears as a retention of signal intensity OVERLAPPING PARALLEL
(Mosley and Butts, 1999). LINES WITH ENHANCED
The strength of MPG is represented by
the b-value (measured in s/mm2). Using a
RECONSTRUCTION
strong MPG with a high b-value provides TECHNIQUE
images contrasted by proton diffusion,
whereas via DWI pulse a small MPG Fast spin echo (FSE), characterized by a
with a small b-value provides images series of 180° rephasing pulses and multiple
contrasted by perfusion, mainly micro- echoes, is a rapid scanning technique com-
circulation of blood. Using this feature, pared with conventional T2-weighted spin
Okada et al. (1998) reported the use- echo imaging. Conventional T2-weighted
fulness of MPG with small a b-value spin echo imaging requires quite a long
in the suppression of signals from ves- scanning time (at least 5 min), whereas data
sels. Patients with focal liver lesions were acquisition with FSE can be completed
evaluated with DWI with a small b-value. within a few minutes or less. Moreover,
They found that the signals from hepatic the breathhold FSE technique shows bet-
vessels were suppressed and the con- ter lesion conspicuity than conventional
spicuity of small lesions was improved. T2-weighted imaging on liver imaging
The lesion-to-liver signal intensity ratio of (Gaa et al., 1996). FSE can be com-
DWI images was significantly better than bined with MPG, and FSE DWI has far
that of T2-weighted fast spin echo images. less B0-related artifacts (e.g., from metal,
However, some problems were mentioned sinuses, and eddy currents) and can be
and these were mainly due to the EPI tech- implemented with a less intensive gradient
nique, which was combined with MPG in requirement than EPI. However, FSE DWU
DWI and widely used in most DWI stud- faces a major challenge because the signal
ies in clinical settings. EPI samples all the will not meet Carr-Purcell-Meiboom-Gill
data necessary for reconstruction of an conditions, leading to unstable echoes
image, and images can be acquired within (Bammer et al., 2002)
a second. EPI utilizes gradient rephrasing, Periodically Rotated Overlapping
which results in spatial distortions from ParallEL Lines with Enhanced Recon-
magnetic susceptibility effects, especially struction (PROPELLER) is a radial scan-
at the diaphragm near the air interface. The ning variation based on the FSE. MRI data
spatial resolution is also limited (Farzaneh are collected in a series of rotating blades
et al., 1990). These are inevitable prob- containing phase encoding lines, with each
lems associated with the EPI technique blade sampling a common central k-space
and are disadvantageous to detect small data set, providing most of the image’s sig-
hepatic lesions. To overcome these prob- nal intensity and determining gross tissue
lems, MPG should be combined with other contrast throughout the image (Pipe, 1999)
suitable pulse sequences, which provide (Figure 15.1). PROPELLER has inherent
high spatial resolution images with less 2D navigator information in each FSE echo
susceptibility-induced distortion. train, and the basic idea of PROPELLER is
216 S. Kiryu and K. Ohtomo

efficacy of SPIO DWI PROPELLER in


the detection of hepatic metastases was
evaluated in a recent study (Kiryu et
al., 2006). Fourteen patients with hepatic
metastases were examined with a 1.5 T
MR scanner. After the infusion of SPIO
(Ferumoxides), SPIO-enhanced FSE
(SPIO FSE) and SPIO DWI PROPELLER
were acquired with a respiratory trigger
using a body coil. The b-value of MPG for
SPIO DWI PROPELLER was 10 s/mm2
to suppress signals from vessels. Hepatic
resection was carried out on all patients,
and all resections were performed with an
ultrasound-guided procedure. Among the
Figure 15.1. Illustration of PROPELLER k-space
acquisition. MRI data are collected in a series
38 metastases 1 cm or larger, 37 (97.4%)
of rotating blades containing phase encoding were detected with SPIO FSE and 36
lines, with each blade sampling a common central (94.7%) were detected with SPIO DWI
k-space data set PROPELLER. There was no significant
difference between both pulse sequences.
Among the 30 metastases smaller than
that this oversampled region in the center 1 cm, 16 (53.3%) were detected with SPIO
of the k-space can be compared between FSE and 24 (80.0%) were detected with
blades to correct for inconsistencies prior SPIO DWI PROPELLER. The detectability
to combining the data. PROPELLER pro- of metastases by SPIO DWI PROPELLER
vides far greater immunity against geomet- was significantly higher than that of SPIO
ric distortion than that obtained with EPI FSE. As expected, the small hepatic metas-
sequences. It also mitigates signal instabil- tases near to the hepatic vessels were dif-
ity, present in DWI FSE (Bammer, 2003). ficult to detect by SPIO FSE. SPIO DWI
Diffusion-weighted PROPELLER has been PROPELLER provided images with a
introduced and successful in high-resolution high spatial resolution and the signals
diffusion-weighted tensor imaging of the from the vessels were suppressed by MPG
brain (Pipe et al., 2002). (Figure 15.2).
Although a respiratory trigger was
applied, motion artifacts were noted on
SPIO-ENHANCED DWI T2 FSE SPIO FSE in some cases whereas no
IMAGING USING PROPELLER motion artifacts were found on the SPIO
DWI (Figure 15.3).
Based on the hypothesis that SPIO- Patient movement also introduces
enhanced diffusion-weighted PROPEL- ghosting, blurring, and the appearance
LER MRI (SPIO DWI PROPELLER) of mottled bands parallel to the phase-
of the liver has advantages in terms of encoding direction of the image (Bellon
detecting small hepatic metastases, the et al., 1986). As Pipe (1999) showed that
15. Detection of Small Hepatic Lesions: Superparamagnetic Oxide-Enhanced 217

Figure 15.2. A 63-year-old man with hepatic metastases. a: SPIO-enhanced T2-FSE. A small metastasis
(arrow) is indistinguishable from the bright signals from vessels. b: SPIO-enhanced diffusion-weighted
PROPELLER. The lesion is clearly noted (arrow)

Figure 15.3. A 38-year-old man with hepatic metastases. a: SPIO-enhanced T2-FSE. Motion artifacts
are shown. b: SPIO-enhanced diffusion-weighted PROPELLER. Metastases are clearly shown without
motion artifacts

PROPELLER MRI was useful for cor- artifacts is an advantage of PROPELLER


recting bulk motion in head images and in detecting such small lesions.
respiratory motion in nongated cardiac On PROPELLER images, a streak arti-
images, such artifacts were not found on fact occasionally appears around structures
SPIO DWI PROPELLER. Motion artifacts with high signal intensity (Pipe, 1999). The
have a risk of obscuring small hepatic bright signal, which moved with cardiac
lesions; therefore, the correction of motion pulsation, produced a spatially and rapidly
218 S. Kiryu and K. Ohtomo

varying phase, which could not be cor- Gaa, J., Hatabu, H., Jenkins, R.L., Finn, J.P., and
rected by the PROPELLER method. Large Edelman, R.R. 1996. Liver masses: replace-
ment of conventional T2-weighted spin-echo
high signal intensity structures caused a
MR imaging with breath-hold MR imaging.
severe streak artifact, and small lesions Radiology 200: 459–464.
near these structures may be obscured. Kawasaki,S.,Makuuchi,M.,Kakazu, T., Miyagawa, S.,
This is a pitfall of PROPELLER imaging. Takayama, T., Kosuge, T., Sugihara, K., and
It is concluded that SPIO-enhanced MRI Moriya, Y. 1994. Resection for multiple meta-
is widely used to detect hepatic metastases; static liver tumors after portal embolization.
Surgery 115: 674–677.
however, a problem arises in the detection
Kiryu, S., Okada, Y., and Ohtomo, K. 2002.
of small lesions. The signal from hepatic Differentiation between hemangiomas and cysts
vessels mimicking small lesions causes of the liver with single-shot fast-spin echo image
false-positive lesions in SPIO-enhanced using short and long TE. J. Comput. Assist.
MRI. MPG with a small b-value is a fea- Tomogr. 26: 687–690.
sible technique to suppress such signals Kiryu, S., Watanabe, M., Kabasawa, H., Akahane, M.,
Aoki, S., and Ohtomo, K. 2006. Evaluation of
from hepatic vessels. The combination of
super paramagnetic iron oxide-enhanced diffu-
MPG and an appropriate pulse sequence, sion-weighted PROPELLER T2-fast spin echo
such as PROPELLER, provides high spa- magnetic resonance imaging: preliminary expe-
tial resolution SPIO images and is useful rience. J. Comput. Assist. Tomogr. 30: 197–200.
for detecting small hepatic lesions. Makuuchi, M., Takayama, T., Kosuge, T.,
Yamazaki, S., Yamamoto, J., Hasegawa, H., and
REFERENCES Takayasu, K. 1991. The value of ultrasonogra-
phy for hepatic surgery. Hepatogastroenterology
Ballantyne, G.H., and Quin, J. 1993. Surgical treat- 38: 64–70.
ment of liver metastases in patients with colorectal Mosley, M.E., and Butts, K. 1999. Diffusion and
cancer. Cancer 71: 4252–4266. perfusion. In: Stark, D. D., Bradley, W. G., Jr.,
Bammer, R. 2003. Basic principles of diffusion- eds. Magnetic Resonance Imaging. St. Louis,
weighted imaging. Eur. J. Radiol. 45: 169–184. MO: Mosby, pp1515–1538.
Bammer, R., Augustin, M., Prokesch, R.W., Moseley, M.E., Cohen, Y., Mintorovitch, J.,
Stollberger, R., and Fazekas, F. 2002. Diffusion- Chileuitt, L., Shimizu, H., Kucharczyk, J.,
weighted imaging of the spinal cord: interleaved Wendland, M.F., and Weinstein, P.R. 1990.
echo-planar imaging is superior to fast spin- Early detection of regional cerebral ischemia in
echo. J. Magn. Reson. Imaging 15: 364–373. cats: comparison of diffusion- and T2-weighted
Bellon, E.M., Haacke, E.M., Coleman, P.E., Sacco, MRI and spectroscopy. Magn. Reson. Med. 14:
D.C., Steiger, D.A., and Gangarosa, R.E. 1986. 330–346.
MR artifacts: a review. Am. J. Roentgenol. 147: Muller, R.D., Vogel, K., Neumann, K., Hirche, H.,
1271–1281. Barkhausen, J., Stoblen, F., Henrich, H., and
Farzaneh, F., Riederer, S.J., and Pelc, N.J. 1990. Langer, R. 1998. MRI with supermagnetic iron
Analysis of T2 limitations and off-resonance particles versus double-spiral CT in identifi-
effects on spatial resolution and artifacts in cation of malignant liver lesions. Rofo 168:
echo-planar imaging. Magn. Reson. Med. 14: 436–443.
123–139. Okada, Y., Ohtomo, K., Kiryu, S., and Sasaki, Y.
Fretz, C.J., Stark, D.D., Metz, C.E., Elizondo, G., 1998. Breath-hold T2-weighted MRI of hepatic
Weissleder, R., Shen, J.H., Wittenberg, J., Simeone, J., tumors: value of echo planar imaging with
and Ferrucci, J.T. 1990. Detection of hepatic diffusion-sensitizing gradient. J. Comput. Assist.
metastases: comparison of contrast-enhanced CT, Tomogr. 22: 364–371.
unenhanced MR imaging, and iron oxide-enhanced Oudkerk, M., van den Heuvel, A.G., Wielopolski,
MR imaging. Am. J. Roentgenol. 155: 763–770. P.A., Schmitz, P.I., Borel Rinkes, I.H., and
15. Detection of Small Hepatic Lesions: Superparamagnetic Oxide-Enhanced 219

Wiggers, T. 1997. Hepatic lesions: detection Soyer, P., Lacheheb, D., and Levesque, M. 1993.
with ferumoxide-enhanced T1-weighted MR False-positive CT portography: correlation with
imaging. Radiology 203: 449–456. pathologic findings. Am. J. Roentgenol. 160:
Penna, C., and Nordlinger, B. 2002. Colorectal 285–289.
metastasis (liver and lung). Surg. Clin. North. Stark, D.D., Weissleder, R., Elizondo, G., Hahn, P.F.,
Am. 82: 1075–1090, x–xi. Saini, S., Todd, L.E., Wittenberg, J., and Ferrucci,
Pipe, J.G. 1999. Motion correction with J.T. 1988. Superparamagnetic iron oxide: clinical
PROPELLER MRI: application to head motion application as a contrast agent for MR imaging of
and free-breathing cardiac imaging. Magn. Reson. the liver. Radiology 168: 297–301.
Med. 42: 963–969. Ward, J., Naik, K.S., Guthrie, J.A., Wilson, D.,
Pipe, J.G., Farthing, V.G., and Forbes, K.P. and Robinson, P.J. 1999. Hepatic lesion detec-
2002. Multishot diffusion-weighted FSE using tion: comparison of MR imaging after the
PROPELLER MRI. Magn. Reson. Med. 47: administration of superparamagnetic iron oxide
42–52. with dual-phase CT by using alternative-free
Ros, P.R., Freeny, P.C., Harms, S.E., Seltzer, S.E., response receiver operating characteristic analy-
Davis, P.L., Chan, T.W., Stillman, A.E., Muroff, sis. Radiology 210: 459–466.
L.R., Runge, V.M., Nissenbaum, M.A., et al. Young, N., Sing, T., Wong, K.P., Hollands, M., and
1995. Hepatic MR imaging with ferumoxides: Tait, N. 1997. Use of spiral and non-spiral com-
a multicenter clinical trial of the safety and puted tomography arterial portography in the
efficacy in the detection of focal hepatic lesions. detection of potentially malignant liver masses.
Radiology 196: 481–488. J. Gastroenterol. Hepatol. 12: 385–391.
16
Diagnosis of Hepatocellular Carcinoma:
Multidetector-Row Computed Tomography
and Magnetic Resonance Imaging
Hiromitsu Onishi, Takamichi Murakami, and Hironobu Nakamura

INTRODUCTION current multidetector-row helical com-


puted tomography (MDCT) technique
Diagnostic imaging plays an important including the underlying principles of
role in the diagnosis of early-stage hepato- contrast injection and MR techniques,
cellular carcinoma (HCC); with contrast- including recent advanced technology
enhanced computed tomography (CT) and for the diagnosis of HCC.
magnetic resonance (MR) imaging exami-
nation being especially important for the
precise and objective evaluation of HCC. MULTIDETECTOR-ROW
At present, both imaging techniques are HELICAL COMPUTED
widely used for diagnosis of HCC. For TOMOGRAPHY
the CT examination, a contrast-enhanced
dynamic study consisting of an arterial Multidetector-row helical computed tom-
phase and a portal venous phase is useful ography scanners were introduced to clinical
for detecting HCC, while multidetector- practice in 1998 and currently are in use in
row helical CT (MDCT) scanner offers many clinical institutes. The MDCT scanner
major advantages for the evaluation of has an X-ray detector divided into multiple
HCC due to its faster acquisition capabil- rows along the longitudinal axis direction
ity and higher spatial resolution. of patient’s body, and can simultaneously
For contrast-enhanced MR imaging, acquire imaging data sets from multiple
a dynamic study using gadolinium- rows. The number of the rows from which
based contrast material is also useful for imaging data can be acquired simultane-
evaluating HCC. In addition, a tissue- ously depends on the number of channels
specific contrast material such as super- of the scanner’s data acquisition systems.
paramagnetic iron oxide (SPIO), which As the number of channels increases, the
is available only for MR imaging is a scanner can acquire imaging data sets faster
valuable aid for the diagnosis of HCC. and provide images with thinner slice thick-
This chapter presents an overview of nesses and shorter scanning times.

221
222 H. Onishi et al.

MULTIDETECTOR-ROW the tube current to attain an acceptable


HELICAL COMPUTED noise level across the region of interest
TOMOGRAPHY SCANNING (3D Auto mA; GE Healthcare, or Real
EC; Toshiba,). For the X-ray tube voltage,
TECHNIQUE 120 kVp is invariably employed.
The selection of scanning parameters
depends largely on the number of chan-
nels of the data acquisition system. At our CONTRAST ADMINISTRATION
institute, two 64-channel MDCT scanners FOR THE DYNAMIC
and two 4-channel MDCT scanners were MULTIDETECTOR-ROW
in operation at the end of 2005. For heli- HELICAL COMPUTED
cal scanning with a 64-channel MDCT,
a detector configuration of 0.625 mm ×
TOMOGRAPHY STUDY
64 = 40 mm (LightSpeed VCT64; GE Contrast enhancement is essential for the
Healthcare, Milwaukee, WI) or 0.5 mm × evaluation of various diseases, especially
64 = 32 mm (Aquilion 64; Toshiba, Nasu, tumorous ones. In addition, a dynamic
Tochigi) is invariably used because the study that consists of an arterial phase
number of channels of the data acquisition and a portal venous phase after contrast
system is equal to the number of detector enhancement has proven to be useful for
rows in the 64-channel MDCT scanner. the evaluation of tumorous diseases of
For the helical pitch, GE Medical Systems the liver, especially hypervascular tumors
scanners offer a choice of 0.6, 0.984, 1.375 such as HCC (Murakami et al., 2001). Our
or 1.75 and Toshiba scanners provide a basic dynamic CT scanning procedure for
choice of 0.641, 0.828 and 1.484. Because the evaluation of HCC using 64-channel
the scanning speed of the 64-channel MDCT scanners consists of precontrast
MDCT scanner is sufficiently fast, an images, late arterial phase images and
extremely large helical pitch may not be portal venous phase images. Early arterial
required. The helical pitch from 0.8 to and/or equilibrium phase images can be
1.5 is considered appropriate for contrast- added as an option depending on the clinical
enhanced dynamic studies of the upper requirements. For an effective dynamic
abdomen, while a helical pitch of 1.375 study, however, an optimal contrast injection
(GE) or 0.828 (Toshiba) is employed for protocol is most important.
the abdominal region. As for the rotation
speed of the X-ray tube, a lower rotation
Administration Dose of Contrast Material
speed of 0.5 or 0.6 s per rotation may be
used for the abdominal region, so that the Heiken et al. (1995) and Brink et al. (1995)
scanning period of the upper abdomen reported that a maximum hepatic enhance-
ranges from 2 to 5 s. ment of at least 50 Hounsfield Units (HU) is
For the X-ray tube current, the automatic needed to perform high-diagnostic-quality
tube current modulation technique should hepatic CT and that 521 mgI/kg of iodine
be employed in order to optimize the is necessary to achieve this level of hepatic
radiation dose (Kalra et al., 2004). Based enhancement. This dose corresponds to
on a single scout scan, the system adjusts ~ 1.73 ml/kg of contrast material with
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 223

an iodine concentration of 300 mgI/ml. tion rate method is employed, however,


Yamashita et al. (2000) reported that for the scan delay after the contrast arrival
abdominal helical CT the dose of intravenous should be varied for each phase accord-
contrast medium should be 2.0–2.5 ml/kg ing to the injection duration. Although
of body weight with an iodine concentration a high injection rate with short injection
of 300 mgI/ml. duration is considered to be advantageous
for obtaining strong enhancement of the
Iodine Concentration of Contrast Material arteries or hypervascular lesions in arterial
phase imaging, it may increase the risk of
Iodine concentration of contrast materials
extravasation or other complications. A
for clinical CT examinations is available
fixed injection duration of 25–30 s, result-
at 300, 320, 350 and 370 mgI/ml. Awai
ing in an injection flow rate of 4–5 ml/s, is
et al. (2002) reported that the high iodine
therefore generally employed for contrast-
concentration of 370 mgI/ml was more
enhanced dynamic studies.
effective than the moderate iodine concen-
At our institute, contrast material at
tration of 300 mgI/ml for the detection of
2.0 ml/kg of 300 mgI/ml or 1.7 ml/kg of
hypervascular HCC when the same injec-
370 mgI/ml is injected intravenously by
tion flow rate and iodine dose were used,
power injectors equipped with a double
both of which were adapted to the patient’s
injection drive system (Dual Shot; Nemoto
body weight.
Kyorindo Co., Ltd., Tokyo, Japan) in 25 s
through a 20-gauge plastic catheter placed
Injection Flow Rate and Duration of in an antecuvital vein. However, the limi-
Contrast Material tation for the contrast amount is 150 ml
When the administered dose is adapted and that for the injection flow is 5 ml/s.
to the patient’s weight, the injection flow
rate and/or injection duration will vary.
There are two mainstream approaches OPTIMAL SCANNING DELAY
to determine these parameters, the fixed
injection duration method, which involves There are two ways to determine scan
adjustment of the injection flow rate and timing. One uses a fixed scan delay and
the fixed injection rate method, in which the other a scan delay adjusted to the
the injection duration is adjusted. Awai patient’s parameters by means of a test
et al. (2004) reported that the fixed injec- bolus method or an automatic bolus track-
tion duration method is more effective ing system. The contrast arrival time var-
for determining the time needed to reach ies even with normal cardiac function,
peak aortic enhancement after initiation and it can be protracted to 30 s or more
of the contrast injection in the abdominal in patients with heart failure. These inter-
aorta because this time span is equal to patient differences cannot be ignored for
the duration of the injection time (Bae, a good dynamic study because the images
2003). With the fixed injection duration acquired with incorrect timing can affect
method, constant scan delay after contrast the clinical diagnosis, especially in terms
arrival can be employed for each phase in of arterial phase imaging. Although the
a dynamic study. When the fixed injec- fixed scan delay method may actually be
224 H. Onishi et al.

preferable for the majority of institutes, CT scan is triggered automatically (Real


to minimize scanning at the incorrect Prep) or manually (SmartPrep). Because
enhancement time, inclusion of the test the mini bolus injection is not necessary
bolus method (Murakami et al., 2001) or with the automatic bolus tracking method,
the automatic bolus tracking system (Kim the total dose of contrast material required
et al., 2002) is recommended. is less than that for the test bolus method.
Determination of the optimal scan delay
with the test bolus method is performed
after precontrast imaging. A mini bolus SCANNING TIMING
(e.g., 15 ml at 5 ml/s) of contrast mate-
rial is injected intravenously and flushing The early arterial phase images which are
with a saline chaser (e.g., 25 ml at 5 ml/s) useful for the precise evaluation of the
is recommended. A series of single-level arteries are acquired 10–15 s after the con-
CT scans with low dose exposure (e.g., trast arrival (Murakami et al., 2006). At
120 kVp, 10 mA) at the abdominal aorta our institute, the early arterial scanning is
is acquired every 2 s from 8 to 40 s after started 10 s after the triggering (the point
the initiation of the mini bolus injection. that the time-enhancement curve raises to
A region-of-interest cursor is then placed the threshold value) using the bolus track-
within the aorta and a time-enhancement ing system. The threshold is set at 50 HU
curve of the aorta is generated on the of enhancement value.
scanner console. Contrast arrival corre- The late arterial phase images which are
sponds to the beginning of the upslope of useful for visualization of the hypervascu-
the time-enhancement curve, and contrast lar HCC or for the precise evaluation of
arrival time (AT) to the span between the the intrahepatic portal veins are acquired
initiation of the mini bolus injection and 20–25 s (22 s at our institute) after trig-
the contrast arrival at the abdominal aorta. gering. For this timing, maximal tumor-
Although the test bolus method can to-liver contrast can be achieved in most
be used with any MDCT scanner, the cases. The portal venous phase images
automatic bolus tracking method can be which are useful for evaluation of the por-
used only with a scanner equipped with tal venous system including tumor throm-
a system such as Real Prep (Toshiba) or bus are acquired ~ 60 s after the triggering.
SmartPrep (GE Healthcare). Capturing the The equilibrium phase images which are
optimal scan delay using the automatic useful for the detection of the hypovascu-
bolus tracking system is done as follows. lar HCC are acquired 180 s after the initia-
On the precontrast CT image, a region-of- tion of contrast injection.
interest cursor is placed within the aorta
and the threshold value for enhancement
(e.g., 50 HU) or the CT value is input to the IMAGE PROCESSING
scanner. The monitoring scan is started 8 s
after the initiation of the contrast injection, Multidetector-row helical computed tom-
and when the enhancement of the aorta ography can generate not only 5 mm-slice
reaches the threshold on the monitoring thickness images but also thin-slice images
scan images, initiation of the diagnostic with the same scanning. Usually, 5 mm-
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 225

slice thickness transverse images are used imaging techniques are employed. The
for the evaluation of HCC (Kawata et al., main advantage of MR imaging is its
2002), while the thin-slice images are used higher sensitivity for contrast, which plays
for detailed diagnosis or postprocessing. a critically important role in accurate
Truly isotropic voxel data consisting of evaluation of HCC.
submillimeter-slice thickness images can
be acquired easily when using an MDCT
scanner with ³ 16 channels. Multiplanar MAGNETIC RESONANCE
reformatted (MPR) images reconstructed SCANNING TECHNIQUE
from isotropic voxel data have the same
spatial resolution as the original transverse Our standard technique for evaluation
images, and three-dimensional (3D) of HCC using the 1.5-Tesla unit (Signa
CT-angiography and maximum intensity Excite HD; GE Healthcare) employs the
projection (MIP) images can visualize array spatial sensitivity encoding tech-
even small branching arteries. These imaging nique (ASSET; GE Healthcare), which is
techniques are thus useful for acquiring one of the parallel imaging techniques.
the clinical information needed to select For signal reception, an 8-channel phased
the appropriate treatment course and to array coil system for the abdomino-pelvic
plan the treatment procedure including region is used. The parallel imaging tech-
surgical resection or transcatheter arterial nique is based on the fact that receiver sen-
chemoembolization of HCC. To gener- sitivity generally has an encoding effect
ate effective MPR, 3D, and MIP images, complementary to Fourier preparation by
however, we need a clear understanding of linear field gradients. In this technique the
the features and management of the disease. spatial information related to the spatially
varying sensitivity of receiver coils is used
to reduce the number of k-space lines
MAGNETIC RESONANCE needed to form an image. By using mul-
IMAGING tiple receiver coils in parallel, scan time
for Fourier imaging can be considerably
Magnetic resonance imaging has also been reduced.
used for the evaluation of HCC, and faster After localizer scanning, T2-weighted
imaging techniques have been developed, single shot first spin echo (SSFSE)
such as the gradient echo imaging and images are obtained during breath-hold.
three-dimensional Fourier transformation The sequence parameters are: repeti-
techniques. Recently, even faster imaging tion time [TR]: 900 ms, echo time [TE]:
was achieved with the parallel imaging 90 ms, band width: 62.5 kHz, matrix size:
technique. In addition, a sequence dedi- 320 × 160, field of view [FOV]: 320
cated for dynamic studies of the abdomi- × 320 mm, slice thickness: 5.0–6.0 mm,
nal region, especially the liver, was newly slice interval: 2.0–2.5 mm, number of
developed. However, MR imaging used for excitations [NEX]: 0.6. The slice thick-
liver dynamic studies is inferior to MDCT ness and interval are increased accord-
imaging in terms of spatial and temporal ing to the size of the liver to include the
resolution, even when these faster MR whole liver. After calibration scanning
226 H. Onishi et al.

for parallel imaging, T1-weighted fast High Resolution Interpolated Volume


spoiled gradient recalled acquisition in Examination [THRIVE]; Philips Medical
steady state (FSPGR) images are obtained Systems, Best, The Netherlands). The
during breath-hold. The images consist of parameters of the LAVA sequence that is
in-phase and opposed-phase echoes. The employed at our institute are, TR: 4.5 ms,
sequence parameters are, TR: 175 ms, TE: 2.2 ms, flip angle: 12°, inversion
TE: 2.2 ms for opposed-phase imaging time: 7 ms, band width: 62.5 kHz, matrix
and 4.5 ms for in-phase imaging, flip size: 320 × 192, FOV: 320 × 320 mm,
angle: 90°, band width: 62.5 kHz, matrix slice thickness: 4.0 mm, slice interval:
size: 256 × 160, FOV: 320 × 320 mm, 0 mm, NEX: 0.73. The dynamic study
slice thickness: 5.0–6.0 mm, slice interval: consists of precontrast, arterial-phase,
2.0–2.5 mm, NEX: 1. T2-weighted fast portal venous-phase and equilibrium-
spin echo images (TR: 3,750–8,000 ms, phase images.
TE: 90 ms, echo train length: 8, band After the precontrast imaging, 20 ml
width: 62.5 kHz, matrix size: 512 × 160, of gadolinium chelate contrast material
FOV: 320 × 320 mm, slice thickness: 5.0– is injected intravenously with a power
6.0 mm, slice interval: 2.0–2.5 mm, NEX: 3) injector at a flow rate of 2 ml/s through
were obtained with the respiratory trigger- a 22-gauge plastic catheter placed in an
ing technique, in which TR depends on antecubital vein. This is followed by a
the patient’s respiratory interval. After the 20-ml saline chaser at a flow rate of 2 ml/s.
precontrast scanning mentioned earlier, The scan delay for the arterial phase imag-
contrast-enhanced imaging is performed. ing is determined by means of the bolus
tracking system, and the arterial scanning
is started 10 s after the triggering. The
CONTRAST-ENHANCED portal venous phase and the equilibrium
DYNAMIC MAGNETIC phase scanning are started 60 and 180 s,
RESONANCE IMAGING respectively, after the initiation of the con-
trast injection.
The “Liver Acquisition with Volume
Acceleration” (LAVA; GE Healthcare,)
sequence is employed for the contrast- TISSUE-SPECIFIC CONTRAST-
enhanced dynamic study. The sequence ENHANCED MAGNETIC
employed is the fast gradient echo sequence RESONANCE IMAGING
combined with the three-dimensional
Fourier transformation technique, which Superparamagnetic iron oxide is a liver-
uses the fat suppression sequence by specific particulate MR contrast material
means of the improved spectral inver- that is taken up by the reticuloendothe-
sion recovery method. Other manu- lial system of the liver and improves
facturers also offer fast gradient echo hepatic tumor detection by means of
sequences for the dynamic study such as increasing the focal hepatic lesion-to-
the Volumetric Interpolated Breath-hold liver contrast. Two SPIO contrast materi-
Examination [VIBE]; Siemens Medical als are commercially available in Japan:
Systems, Erlangen, Germany, or the T1 ferumoxides (Feridex; Eiken and Tanabe
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 227

Pharmaceutical, Osaka, Japan) and fer- COMPUTED TOMOGRAPHY


ucarbotoran (Resovist; Nihon Schering, IMAGING FEATURES
Osaka, Japan). While the ferumoxide is OF HEPATOCELLULAR
administrated with an intravenous drip, the
ferucarbotran can be injected as a bolus.
CARCINOMA
Superparamagnetic iron oxide contrast On nonenhanced CT images, HCCs can
materials have a T2 shortening effect and appear as slightly less attenuated than
reduce the signal intensity on T2-weighted adjacent liver parenchyma (Figure 16.1a).
images of the normal liver parenchyma, It often happens that small HCCs are
where SPIO are mainly distributed. In not detected on noncontrast CT images.
addition, there are other liver-specific MR In HCC, however, there may be met-
contrast materials such as gadoxetic acid amorphosis, necrosis, periotic changes
disodium (Gd-EOB-DTPA), gadobenate or hemorrhage; therefore, it is usually
dimeglumine (Gd-BOPTA) and manga- heterogeneous attenuation, especially
fodipir trisodium (Mn-DPDP), which are in large HCCs. Fatty metamorphism is
adsorbed into the liver cells and excreted often observed especially in the well-
into the bile (Murakami et al., 1996). differentiated type of HCC and is shown
The scan protocol for SPIO-enhanced as a low-attenuation area. Hepatocellular
MR imaging at our institute is as follows. carcinoma develops tumor vessels, thus
After the precontrast imaging, 0.7–1.4 ml increasing the arterial blood supply of the
of ferucarbotran is injected intravenously tumor. Conversely, in most HCCs, except
and manually through a 22-gauge plas- for the well-differentiated type, portal
tic catheter placed in an antecubital vein, venous blood flow in the tumor disap-
which is followed by a 20-ml saline chaser. pears. Generally, therefore, hypervascular
Post-contrast images are obtained > 10 min HCC is strongly enhanced in the arterial
after the SPIO injection. One is a heavily dominant phase while the attenuation of
T1-weighted gradient echo with minimum HCC decreases in the portal venous domi-
echo time (TR: 150 ms, TE: 1.4 ms, flip nant phase as seen in contrast-enhanced
angle: 90°, band width: 62.5 kHz, matrix dynamic studies. Meanwhile, the liver
size: 256 × 160, FOV: 320 × 320 mm, slice parenchyma that is supplied predominantly
thickness: 5.0–6.0 mm, slice interval: 2.0– by the portal vein is strongly enhanced in
2.5 mm, NEX: 1) The second is T2-weighted the portal venous dominant phase.
fast spin echo (TR: 3,750–8,000 ms, TE: In the early arterial phase, the aorta and
90 ms, flip angle: 90°, echo train length: 8, hepatic arteries are enhanced strongly and
band width: 62.5 kHz, matrix size: 512 × HCCs moderately, so that the early arte-
224, FOV: 320 × 320 mm, slice thickness: rial images are useful for accurate evalu-
5.0–6.0 mm, slice interval: 2.0–2.5 mm, ation of the arteries, especially by means
NEX: 2). The third is T2*-weighted gradi- of 3D-CT angiography or MIP images.
ent echo with a long echo time (TR: 200 ms, The early arterial phase imaging can be
TE: 8.5 ms, flip angle: 60°, band width: skipped to reduce the radiation exposure
62.5 kHz, matrix size: 512 × 160, FOV: 320 dose when there is no clinical need for
× 320 mm, slice thickness: 5.0–6.0 mm, evaluation of the arteries by means of CT
slice interval: 2.0–2.5 mm, NEX: 1). angiography.
228 H. Onishi et al.

a b c

d e f

Figure 16.1. Hepatocellular carcinoma in a 71-year-old man with severe liver cirrhosis. Precontrast (a),
late arterial phase (b), portal venous phase (c), and equilibrium phase (d) dynamic MDCT images. The
HCC (arrowheads) is visualized as an enhanced nodule in the late arterial phase and as a hypo-attenuated
nodule in the equilibrium phase. Three-dimensional CT angiography of hepatic arteries (e) created with
early arterial phase data by means of the color-coded volume rendering method shows that the right hepatic
artery (arrow in e) branches from the supermesenteric artery. Maximum intensity projection image of the
portal venous system (f) created with portal venous phase data clearly depict the varices (arrow in f)

Because most HCCs are strongly ally hypovascular and are not visualized
enhanced and tumor-to-liver contrast is as enhanced nodules in the arterial phase.
maximal in the late arterial phase, the Computer tomography images obtained
images obtained during this phase are during the portal venous and equilib-
essential to detect the hypervascular HCCs rium phases have proven to be useful for
(Figure 16.1b). The intrahepatic vein the detection of less vascularized tumors
and tumor thrombus are also thoroughly such as well-differentiated or early HCCs.
enhanced during this phase. Iannaccone et al. (2005) reported that
In the portal venous and equilibrium the equilibrium phase images that were
phases, the contrast medium is moving acquired 180 s after initiation of the con-
from the intravascular to the intercellular trast injection were a useful adjunct to the
space. The washout of contrast material biphasic images (arterial and portal venous
from the tumor is rapid because the HCC phase) for the detection of HCC.
contains less intercellular space than the
surrounding liver parenchyma. Although
most HCCs are not supplied by the portal COMPUTED TOMOGRAPHIC
vein, the liver parenchyma is maximally ANGIOGRAPHY
enhanced in the portal venous phase, so
that low attenuation is typically observed in The hepatic arteries are depicted with
these phases (Figures 16.1c, d) (Kim et al., CT angiography using the thin-slice data
1995). Well-differentiated HCCs are usu- obtained from early arterial phase imaging
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 229

(Figure 16.1e). It has been reported that useful for detection of tumor invasion to
100% of major arterial trunks and ~ 90% the portal or the hepatic vein. Patients
or more of the small branches of the with HCC usually have cirrhosis as an
hepatic arteries were detected on partial underlying disease and have collateral
MIP images and original thin-slice cross- vessels and varices due to portal hyper-
sectional images in the early arterial phase tension. Computed tomographic angi-
(Takahashi et al., 2002). Visualization of ography can also accurately show the
the anatomy of hepatic arteries is essen- anatomy of these vessels.
tial for planning not only surgical resec-
tion but also the transcatheter arterial
chemoembolization or reservoir infusion MAGNETIC RESONANCE
system. The portal and hepatic veins can IMAGING FEATURES
be visualized by CT angiography from OF HEPATOCELLULAR
late arterial and portal venous phases CARCINOMA
imaging (Figure 16.1f). Hepatocellular
carcinomas often invade the portal vein On nonenhanced MR imaging, 35% of HCC
and/or hepatic vein and form the tumor cases show high intensity on T1-weighted
thrombus. Computed tomographic por- images, 25% show isointensity and 40%
tography and CT venography are thus show low intensity (Figure 16.2a), while

a b c

d e f

Figure 16.2. Hepatocellular carcinoma in a 69-year-old man. Precontrast (a), arterial phase (b), portal
venous phase (c), and equilibrium phase (d) dynamic MR images obtained with the LAVA sequence
before therapeutic treatment. The HCC (arrow) is seen adjacent to the hepatic vein as an enhanced nodule
in the late arterial phase and as a hypo-attenuated nodule in the portal venous and equilibrium phases.
T1-weighted gradient echo image (e) and spin echo T2-weighted image (f) 2 days after radiofrequency
ablation therapy for HCC. The completely ablated area shows as hyperintense on T1-weighted images
and as hypointense on T2-weighted images (arrowheads)
230 H. Onishi et al.

~ 90% show high intensity on T2-weighted mal liver parenchyma. In most tumors,
images (Kadoya et al., 1992). However, however, uptake of SPIO declines with a
well-differentiated HCC often shows iso- decrease in Kupffer cells. This contrast
intensity or low intensity on T2-weighted material has a relatively strong T2 and
images. Fatty deposition which is some- moderate T1 shortening effect. Therefore,
times seen in well-differentiated HCC on images obtained with SPIO-enhanced
T2-weighted spin echo and T2*-weighted
results in high-signal intensity on in-phase
T1-weighted images, which decreases gradient echo with long echo time, the
on opposed-phase T1-weighted images. tumor is visualized as an area with a higher
Contrast-enhanced dynamic studies with signal intensity than the surrounding liver
gadolinium chelate materials show HCC as parenchyma (Figure 16.3). Furthermore,
a hypervascular tumor in the arterial phaseHCC which has lost its hepatocellular
function, including its reticuloendothelial
and the washout of contrast materials in the
portal venous phase and the equilibrium function, is visualized in most cases as
phase (Figures 16.2b–d). higher signal intensity. However, SPIO
is sometimes taken up by the tumor of
well-differentiated HCC, adenomatous
SUPERPARAMAGNETIC hyperplasia, regenerating nodules and
focal nodular hyperplasia, which have
IRON OXIDE-ENHANCED maintained some degree of hepatocel-
MAGNETIC RESONANCE lular function (Imai et al., 2000). It may
IMAGING FEATURE thus be possible to differentiate benign
OF HEPATOCELLULAR and malignant tumors by using SPIO-
CARCINOMA enhanced MR imaging. Moreover, since
the SPIO agent ferucarbotran can be
Superparamagnetic iron oxide is taken injected as a bolus, contrast-enhanced
up by reticuloendothelial cell such as dynamic studies can be performed by
Kupffer cells that are present in the nor- using T1-weighted or T2-weighted imaging

a b c

Figure 16.3. Hepatocellular carcinoma in an 85-year-old man. Transverse (a) and sagittal (b) long TE
gradient echo images and transverse fast spin echo T2-weighted image (c) obtained more than 10 min
after SPIO administration. The HCC (arrow) is clearly seen as a hyperintense mass on these images
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 231

for the evaluation of the tumor hemody- systems can also be evaluated by both of
namics (Hori et al., 2006). Hepatobiliary these imaging techniques.
contrast agents enhance normal liver
parenchyma on T1-weighted images due
to the T1 shortening effect. The tumor is ACCURACY FOR THE
seen as a low-intensity area because the DIAGNOSIS OF
tumor cell does not take up the agents. HEPATOCELLULAR
With certain agents that can be injected
in bolus form, such as Gd-EOB-DTPA
CARCINOMA
and Gd-BOPTA (Kuwatsuru et al., 1999), The accuracy of tumor detection is often
contrast-enhanced dynamic studies can evaluated by means of receiver oper-
also be performed for the evaluation of ating characteristic (ROC) analyses or
tumor hemodynamics. alternative-free response ROC (AFROC)
analyses as well as in terms of sensitiv-
ity and positive predictive values. While
STAGING OF the conventional ROC method allows for
HEPATOCELLULAR only one response per image, the AFROC
CARCINOMA method allows for multiple responses per
image (Chakraborty and Winter, 1990).
Several staging systems for HCC are cur- Several studies have demonstrated the Az
rently in use. The Union Interbationale value, which is the area under the ROC
Contre le Cancer [UICC], the General curve, and sensitivity for the detection of
Rules for the Clinical and Pathological HCC. Az value and sensitivity determined
Study of Primary Liver Cancer [Liver by means of contrast-enhanced dynamic
Cancer Study Group of Japan classifi- MDCT imaging ranged from 0.82 to 0.99
cation system], and the American Joint and from 77% to 93%, respectively (Kim
Committee on Cancer [AJCC] have estab- et al., 2006; Iannaccone et al., 2005).
lished staging systems that are based on When determined by means of gadolin-
the results of pathologic analyses, which ium-enhanced dynamic MR imaging, Az
incorporate anatomic and histologic find- value and sensitivity ranged from 0.68 to
ings obtained at tumor resection. The 0.97 and from 65% to 91%, respectively
number and size of the primary tumors as (Mori et al., 2005; Kim et al., 2004), while
well as the presence or absence of portal the corresponding values ranged from 0.76
or hepatic vein involvement are included to 0.98 and from 56% to 95%, respec-
in the staging of the primary lesion tively, when SPIO-enhanced MR imaging
because HCC is frequently associated was used (Ward et al., 2000; Kang et al.,
with portal vein or hepatic vein tumor 2003).
thrombus. Both MDCT imaging and MR Contrast-enhanced dynamic CT imag-
imaging are also useful for the evalua- ing using a 16-channel MDCT scanner
tion of the tumor thrombus. Lymph node and SPIO-enhanced MR imaging showed
metastasis and distant metastasis which similar diagnostic accuracy, sensitivity,
are taken into consideration in the staging and positive predictive value for the
232 H. Onishi et al.

detection of HCC (Kim et al., 2006). the viable portion of the tumor in the
However, gadolinium-enhanced dynamic arterial phase.
MR imaging proved to be significantly more
accurate than SPIO-enhanced MR imaging
(Pauleit et al., 2002; Kim et al., 2004). HEPATOCELLULAR
CARCINOMA AFTER
PERCUTANEOUS
HEPATOCELLULAR RADIOFREQUENCY
CARCINOMA AFTER ABLATION THERAPY
TRANSCATHERTER ARTERIAL
CHEMOEMBOLIZATION Percutaneous radiofrequency ablation
therapy can be used for local treatment
Transcatheter arterial chemoemboliza- of HCC, with coagulation necrosis gener-
tion (TACE) with iodized oil is often ally occurring after the treatment and the
performed for treatment of HCC, after ablated area often showing a zonal pattern
which the tumor is seen as a nodule with on both T1- and T2-weighted images (Lee
markedly high attenuation on nonen- et al., 2001). The completely ablated area
hanced CT images, since high concentra- appears as hyperintense on T1-weighted
tions of iodized oil remain in the tumor. images and as hypointense on T2-weighted
Because the portion of the tumor where images, reflecting coagulation necrosis or
iodized oil is retained at high density is hemorrhage (Figures 16.2e, f). On the
considered to be necrotic, CT is suit- other hand, the incompletely ablated area
able for the evaluation of the therapeutic that is often seen around the completely
efficacy of TACE with iodized oil for ablated area appears as hypointense on
HCC. However, incomplete retention of T1-weighted images and as hyperin-
iodized oil corresponds to a wide vari- tense on T2-weighted images, reflecting
ety of tumor necroses, from completely fibrous change or sinusoidal congestion
necrotic to viable cancer tissue (Choi (Onishi et al., 2004). These zonal pat-
et al., 1992). Although contrast-enhanced terns on non-enhanced MR imaging may
dynamic CT may be helpful to evaluate be important for the assessment of the
viability for HCC with incomplete reten- effect of ablation therapy on HCC soon
tion after the TACE treatment, the high after completion of the therapy, since
level of attenuation of iodized oil in the these signal changes are seldom affected
tumor can negatively affect the evalua- by the arterio-portal venous shunt or por-
tion by dynamic CT. On the other hand, tal thrombus that is often associated with
as iodized oil does not have much effect percutaneous ablation therapy. In clinical
on the intensity observed on MR images, practice, contrast-enhanced dynamic CT
MR images can clearly show intratu- and contrast-enhanced dynamic MR imag-
moral signal changes (Murakami et al., ing are used as the standard techniques to
1993a, b). Contrast-enhanced dynamic evaluate the therapeutic effect of ablation
MR imaging is thus useful for evaluating therapy, with the necrotic area shown as a
viability for HCC after TACE treatment nonenhanced area and the residual viable
because it shows enhancement only in lesion as an enhanced area. However, it is
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 233

often difficult to distinguish whether the SUMMARY


enhanced area is a residual viable lesion or
an arterio-portal venous shunt (Dromain Multidetector-row helical computed tom-
et al., 2002). ography and MR imaging are powerful
tools for the accurate diagnosis of HCC.
Both techniques are important not only for
LIMITATIONS AND PROSPECTS detection but also for staging. Moreover,
CT angiography using MDCT is helpful
The most serious drawbacks of MDCT when performing pre-interventional radio-
imaging may be ionized radiation exposure. logical or surgical treatment for HCC. For
Although the scanning time is markedly useful and accurate clinical evaluation, it is
reduced by means of 64-channel MDCT essential to understand the theories behind
scanners, it is difficult to increase the number imaging techniques as well as the clinical
of phases for the dynamic study because of pathology of HCC and to employ optimal
the need to limit the radiation exposure scanning and contrast injection protocols.
dose. One solution for reducing the radia- Because MDCT and MR imaging have
tion exposure dose is the post-processing their respective advantages and some limi-
technique, while the CT images obtained tations, it is essential in clinical practice to
by scanning with a low exposure dose can make the best use of each modality so that
be improved by means of noise reduction they can complement each other.
filters (Kalra et al., 2003). It can be expected
that the number of detector rows in MDCT REFERENCES
scanners will continue to increase and the Awai, K., Takada, K., Onishi, H., and Hori, S.
scanning time will be reduced much further. 2002. Aortic and hepatic enhancement and
In addition, perfusion imaging for the whole tumor-to-liver contrast: analysis of the effect of
liver may become available in the near different concentrations of contrast material at
future when a wide detector covering the multi-detector row helical CT. Radiology 224:
757–763.
entire liver has been developed. Awai, K., Hiraishi, K., and Hori, S. 2004. Effect
In comparison with MDCT imaging, of contrast material injection duration and rate
the spatial and temporal resolution of on aortic peak time and peak enhancement at
MR imaging may currently be considered dynamic CT involving injection protocol with
a drawback. However, boosting of the dose tailored to patient weight. Radiology 230:
acceleration factor of the parallel imaging 142–150.
Bae, K.T. 2003. Peak contrast enhancement in CT
technique is expected to overcome this and MR angiography: when does it occur and
disadvantage and to give MR imaging an why? Pharmacokinetic study in a porcine model.
edge for the evaluation of HCC. Recently, Radiology 227: 809–816.
MR scanners equipped with an ultra-high Brink, J.A., Heiken, J.P., Forman, H.P., Sagel, S.S.,
magnetic field such as 3.0-Tesla have Molina, P.L., and Brown, P.C. 1995. Hepatic spi-
become commercially available for use ral CT: reduction of dose of intravenous contrast
material. Radiology 197: 83–88.
in the abdominal region as well. Their Chakraborty, D.P., and Winter L.H. 1990. Free-
higher signal-to-noise ratio represents response methodology: alternate analysis and a
a significant advantage for high spatial new observer-performance experiment. Radiology
resolution imaging. 174: 873–881.
234 H. Onishi et al.

Choi, B.I., Kim, H.C., Han, J.K., Park, J.H., Kim, with use of noise reduction filters pilot study.
Y.I., Kim, S.T., Lee, H.S., Kim, C.Y., and Han, Radiology 228: 251–256.
M.C. 1992. Therapeutic effect of transcatheter Kalra, M.K., Maher, M.M., Toth, T.L., Schmidt, B.,
oily chemoembolization therapy for encapsu- Westerman, B.L., Morgan, H.T., and Saini, S.
lated nodular hepatocellular carcinoma: CT and 2004. Techniques and applications of automatic
pathologic findings. Radiology 182: 709–713. tube current modulation for CT. Radiology 233:
Dromain, C., de Baere, T., Elias, D., Kuoch, V., 649–657.
Ducreux, M., Boige, V., Petrow, P., Roche, A., Kang, B.K., Lim, J.H., Kim, S.H., Choi, D., Lim,
and Sigal, R. 2002. Hepatic tumors treated H.K., Lee, W.J., and Lee, S.J. 2003. Preoperative
with percutaneous radio-frequency ablation: CT depiction of hepatocellular carcinoma: ferumox-
and MR imaging follow-up. Radiology 223: ides-enhanced MR imaging versus triple-phase
255–262. helical CT. Radiology 226: 79–85.
Heiken, J.P., Brink, J.A., McClennan, B.L., Sagel, Kawata, S., Murakami, T., Kim, T., Hori, M.,
S.S., Crowe, T.M., and Gaines, M.V. 1995. Federle, M.P., Kumano, S., Sugihara, E.,
Dynamic incremental CT: effect of volume and Makino, S., Nakamura, H., and Kudo, M. 2002.
concentration of contrast material and patient Multidetector CT: diagnostic impact of slice
weight on hepatic enhancement. Radiology 195: thickness on detection of hypervascular hepa-
353–357. tocellular carcinoma. AJR. Am. J. Roentgenol.
Hori, M., Murakami, T., Kim, T, Iannaccone, R., 179: 61–66.
Abe, H., Onishi, H., Tomoda, K., Catalano, C, Kim, T., Murakami, T., Oi, H., Matsushita, M.,
Passariello, R., and Nakamura, H. 2006. Kishimoto, H., Igarashi, H., Nakamura, H., and
Hemodynamic characterization of focal hepatic Okamura, J. 1995. Detection of hypervascular
lesions: role of ferucarbotran-enhanced dynamic hepatocellular carcinoma by dynamic MRI and
MR imaging using T2-weighted multishot spin- dynamic spiral CT. J. Comput. Assist. Tomogr.
echo echo planar sequence. J. Magn. Reson. 19: 948–954.
Imag. 23: 509–519. Kim, T., Murakami, T., Hori, M., Takamura, M.,
Iannaccone, R., Laghi, A., Catalano, C., Rossi, P., Takahashi, S., Okada, A., Kawata, S., Cruz, M.,
Mangiapane, F., Murakami, T., Hori, M., Federle, M.P., and Nakamura, H. 2002. Small
Piacentini, F., Nofroni, I., and Passariello, R. hypervascular hepatocellular carcinoma revealed
2005. Hepatocellular carcinoma: role of unen- by double arterial phase CT performed with
hanced and delayed phase multi-detector row single breath-hold scanning and automatic bolus
helical CT in patients with cirrhosis. Radiology tracking. AJR Am. J. Roentgenol. 178: 899–904.
234: 460–467. Kim, Y.K., Kim, C.S., Kwak, H.S., and Lee, J.M.
Imai, Y., Murakami, T., Yoshida, S., Nishikawa, M., 2004. Three-dimensional dynamic liver MR
Ohsawa, M., Tokunaga, K., Murata, M., Shibata, K., imaging using sensitivity encoding for detection
Zushi, S., Kurokawa, M., Yonezawa, T., Kawata, S., of hepatocellular carcinomas: comparison with
Takamura, M., Nagano, H., Sakon, M., superparamagnetic iron oxide-enhanced MR
Monden, M., Wakasa, K., and Nakamura, H. 2000. imaging. J. Magn. Reson. Imag. 20: 826–837.
Superparamagnetic iron oxide-enhanced magnetic Kim, Y.K., Kwak, H.S., Kim, C.S., Chung, G.H.,
resonance images of hepatocellular carcinoma: Han, Y.M., and Lee, J.M. 2006. Hepatocellular
correlation with histological grading. Hepatology Carcinoma in Patients with Chronic Liver
32: 205–212. Disease: Comparison of SPIO-enhanced MR
Kadoya, M., Matsui, O., Takashima, T., and Imaging and 16-Detector Row CT. Radiology
Nonomura, A. 1992. Hepatocellular carcinoma: 238: 531–541.
correlation of MR imaging and histopathologic Kuwatsuru, R., Kadoya, M., Ohtomo, K., Tanimoto, A.,
findings. Radiology 183: 819–825. Hirohashi, S., Murakami, T., Tanaka, Y.,
Kalra, M.K., Maher, M.M., Sahani, D.V., Blake, Yoshikawa, K., and Katayama, H. 1999. Clinical
M.A., Hahn, P.F., Avinash, G.B., Toth, T.L., late phase II trials of MultiHance (Gd-BOPTA) for
Halpern, E., and Saini, S. 2003. Low-dose CT of the magnetic resonance imaging of liver tumors in
the abdomen: evaluation of image improvement Japan. J. Comput. Assist. Tomogr. 23: 65–74.
16. Diagnosis of Hepatocellular Carcinoma: Multidetector-Row Computed Tomography 235

Lee, J.D., Lee, J.M., Kim, S.W., Kim, C.S., and Mun, Murakami, T., Onishi, H., Mikami, K., Iannaccone, R.,
W.S. 2001. MR imaging-histopathologic correla- Federle, M.P., Kim, T., Hori, M., Okada, A.,
tion of radiofrequency thermal ablation lesion in Marukawa, T., Higashihara, H., Passariello, R.,
a rabbit liver model: observation during acute and and Nakamura, H. 2006. Determining the optimal
chronic stages. Korean J. Radiol. 2: 151–158. timing for early arterial phase hepatic CT imaging
Mori, K., Yoshioka, H., Takahashi, N., Yamaguchi, by measuring abdominal aortic enhancement in
M., Ueno, T., Yamaki, T., and Saida, Y. 2005. variable contrast injection protocols. J. Comput.
Triple arterial phase dynamic MRI with sensitiv- Assist. Tomogr. 30: 206–211.
ity encoding for hypervascular hepatocellular car- Onishi, H., Matsushita, M., Murakami, T., Tono, T.,
cinoma: comparison of the diagnostic accuracy Okamoto, S., Aoki, Y., Iannaccone, R., Hori,
among the early, middle, late, and whole triple M., Kim, T., Osuga, K., Tomoda, K., Passariello,
arterial phase imaging. AJR Am. J. Roentgenol. R., and Nakamura, H. 2004. MR appearances
184: 63–69. of radiofrequency thermal ablation region: his-
Murakami, T., Nakamura, H., Hori, S., Tomoda, K., topathologic correlation with dog liver models
Mitani, T., Nakanishi, K., Hashimoto, T., Tsuda, K., and an autopsy case. Acad. Radiol. 11: 1180–
Kozuka, T., Monden, M., and Wakasa, K. 1993a. 1189.
Detection of viable tumor cells in hepatocellular Pauleit, D., Textor, J., Bachmann, R., Conrad, R.,
carcinoma following transcatheter arterial chem- Flacke, S., Layer, G., Kreft, B., and Schild, H.
oembolization with iodized oil. Pathologic cor- 2002. Hepatocellular carcinoma: detection with
relation with dynamic turbo-FLASH MR imaging gadolinium- and ferumoxides-enhanced MR
with Gd-DTPA. Acta. Radiol. 34: 399–403. imaging of the liver. Radiology 222: 73–80.
Murakami, T., Nakamura, H., Tsuda, K., Nakanishi, K., Takahashi, S., Murakami, T., Takamura, M., Kim, T.,
Hori, S., Tomoda, K., Mitani, T., Kozuka, T., Hori, M., Narumi, Y., Nakamura, H., and Kudo, M.
Monden, M., and Wakasa, K. 1993b. Treatment of 2002. Multi-detector row helical CT angiog-
hepatocellular carcinoma by chemoembolization: raphy of hepatic vessels: depiction with dual-
evaluation with 3DFT MR imaging. AJR Am. J. arterial phase acquisition during single breath
Roentgenol. 160: 295–299. hold. Radiology 222: 81–88.
Murakami, T., Baron, R.L., Peterson, M.S., Oliver, Ward, J., Guthrie, J.A., Scott, D.J., Atchley, J.,
J.H. 3rd, Davis, P.L., Confer, S.R., and Federle, Wilson, D., Davies, M.H., Wyatt, J.I., and
M.P. 1996. Hepatocellular carcinoma: MR imag- Robinson, P.J. 2000. Hepatocellular carcinoma
ing with mangafodipir trisodium (Mn-DPDP). in the cirrhotic liver: double-contrast MR imag-
Radiology 200: 69–77. ing for diagnosis. Radiology 216: 154–162.
Murakami, T., Kim, T., Takamura, M., Hori, M., Yamashita, Y., Komohara, Y., Takahashi, M.,
Takahashi, S., Federle, M.P., Tsuda, K., Osuga, K., Uchida, M., Hayabuchi, N., Shimizu, T., and
Kawata, S., Nakamura, H., and Kudo, M. 2001. Narabayashi, I. 2000. Abdominal helical CT:
Hypervascular hepatocellular carcinoma: detec- evaluation of optimal doses of intravenous con-
tion with double arterial phase multi-detector row trast material -a prospective randomized study.
helical CT. Radiology 218: 763–767. Radiology 216: 718–723.
17
Hepatocellular Carcinoma: Effect
of Injection Rate/Injection Duration
of Contrast Material on Computed
Tomography
Tomoaki Ichikawa and Tsutomu Araki

INTRODUCTION that higher injection rates of contrast mate-


rial can improve hypervascular hepatic
Multiphasic, contrast-enhanced computed lesion detection (Kim et al., 1995).
tomography (CT) scanning has improved There has been another controversy that the
the detection and characterization of focal detection rate of hypervascular false-positive
hepatic lesions, such as hepatocellular lesions, for which small arterial-portal venous
carcinomas (HCC) (Ohashi et al., 1993; shunts (A-P shunts) may play a major role,
Hollett et al., 1995; Baron et al., 1996). may increase on HAP images in cirrhotic
It has been well established that hepatic liver (Yu et al., 1997). Frequent visualization
arterial-dominant phase (HAP) scanning of A-P shunts on HAP images may dimin-
is essential to detect hypervascular HCC. ish lesion specificity, which in turn, may
Hepatic arterial-dominant phase images diminish overall diagnostic performance for
can detect a larger number of HCC (67/81, detecting hypervascular HCC underly-
83%) than any other phases including ing cirrhotic liver. Considering the possible
unenhanced, portal venous phase (PVP), increase of A-P shunts as well as hypervas-
and delayed phase (DP) images (Hwang cular HCC on HAP images obtained with
et al., 1997). In addition, some hypervas- a higher injection rate of contrast material,
cular HCC (26–32%) are only seen or are little agreement has been found with regard
most conspicuous on HAP images (Hwang to the use of higher injection rates of contrast
et al., 1997; Mitsuzaki et al., 1996). material for the evaluation of hypervascular
The injection rate of the contrast mate- HCC underlying liver cirrhosis.
rial is one of the most important technique-
related factors for detecting hypervascular
HCC on HAP images. The enhancement FIXED INJECTION RATE
of the aorta (including hepatic artery) on AND INJECTION DURATION
HAP images significantly increases at OF CONTRAST MATERIAL
higher injection rates of contrast material
(Tublin et al., 1999; Kim et al., 1998). There are two methods to administer
Based on these results, it was suggested patients’ body-weight-tailored dose of
in recent HAP imaging studies of the liver contrast material for individual patients:

237
238 T. Ichikawa and T. Araki

(1) the administration of contrast material controversial, considering the possible


with a fixed injection rate, which pres- increase of detection rate of hypervascular
ently is the common method, and (2) with false-positive lesions represented by A-P
a fixed injection duration. In one injection shunts with a higher injection rate of con-
protocol, when the alternative of injection trast material (Yu et al., 1997).
rate or injection duration is fixed, the other In our study with a comparison of 3
factor should be variable by depending on and 5 ml/s of injection rates for detecting
the patients’ body-weight-tailored dose hypervascular HCC underlying cirrhotic
of contrast material. However, it should liver, the sensitivity values for detecting
be recognized that the injection duration hypervascular HCC tended to be higher
should be fixed in an injection protocol with 5 ml/s (mean, 36/41 [88%]) than
rather than the injection rate to establish an with 3 ml/s of contrast material (mean,
optimal multiphasic contrast-enhanced CT 28/35 [80%]), although there was no
protocol of the liver because the injection significant difference (Ichikawa et al.,
duration is the most important and the only 2006b). While our study also showed
factor of all technical factors to predict that the detection rate of the false-posi-
the scan timing for each phase (Ichikawa tive lesions on HAP images significantly
et al., 2006a). increased with 5 ml/s than with 3 ml/s
of contrast material, the lesion specifi-
city was significantly lower with 5 ml/s
Fixed Injection Rate of Contrast Material
than with 3 ml/s. In conclusion, it can
The injection rate of the contrast material be stated that a high injection rate of
is the most important technical factor for contrast material, such as 5 ml/s, is not
determining the amplitude of contrast- always better than medium injection rate
enhancement in hypervascular HCC on of contrast material, such as 3 ml/s, for
HAP images. The enhancement of the the purpose of evaluating hypervascu-
aorta (including hepatic artery) on HAP lar HCC in patients with cirrhotic liver.
images significantly increases at higher Finally, we recommend that one should
injection rates of contrast material (Bae use a medium injection rate of contrast
et al., 1998; Awai and Hori, 2003). Because material for hepatic CT imaging in cir-
hypervascular HCC is always contrast- rhotic patients because the specificity
enhanced by contrast material supplied and the positive predictive values in the
from the hepatic artery, higher injection detection of hypervascular HCC are sig-
rates of contrast material may increase the nificantly higher with a medium injec-
maximum enhancement value of hyper- tion rate (3 ml/s) of contrast material
vascular HCC. Such a hypothesis has been (mean, 236/250 [95%] and 28/42 [67%],
supported by a previous report that 4 ml/s respectively) than with a high injection
injection rate significantly improved the rate (5 ml/s) of contrast material (mean,
detection of small hypervascular HCC 227/265 [86%] and 36/73 [49%], respec-
(< 3 cm) compared with 2 ml/s (Mitsuzaki tively). Our recommendation seems to be
et al., 1996). However, the determina- reliable, especially for general institutes
tion of the best injection rate of contrast involving non-expert radiologists for the
material underlying cirrhotic liver is still liver into hepatic CT image interpretation
17. Hepatocellular Carcinoma: Effect of Injection Rate/Injection Duration 239

because of the definite increasing detec- for individual patients. When a fixed injec-
tion rate of the false-positive lesions tion duration is employed in a hepatic
with a high injection rate of contrast CT protocol, injection rates should be
material. Considering the balance of the variable depending on the patient’s body-
sensitivity and the specificity, we finally weight-tailored dose of contrast material.
recommend 4 ml/s as a medium injection Therefore, the concept of an effective
rate for multiphasic contrast-enhanced injection rate (an injection rate applied
hepatic CT imaging in cirrhotic patients. for a patient with mean body weight in
a patients’ group) is needed. An effec-
Fixed Injection Duration of Contrast tive injection rate can be established by
Material refining the optimal fixed injection rate
(4 ml/s) derived from our previous study.
At present, it has started to be recognized
For example, under the assumption of
that a use of a fixed injection duration can
the use of 2.0 ml/kg with 300 mgl/ml
minimize the patients’ variables in the scan
(600 mgI/kg) of contrast material and the
timing for each phase among all patients
mean patient’s body weight of 60 kg in a
(Bae, 2003; Awai et al., 2004). Such effect
patients’ group, a fixed injection duration
of the fixed injection duration depends on
should be established so that the effective
the fact that the peak enhancement time of
injection rate becomes 4 ml/s by following
the aorta (pET-Ao) may be strictly defined
the equation (Ichikawa et al., 2006b);
by the injection duration. Based on our
experience, the mean time delay from the a fixed injection duration = 120 ml (2 ml/
completion of any injection durations to kg × 60 kg)/4 ml/s = 30 s
pET-Ao was ∼10 s regardless of patients’ In conclusion, we recommend the use of
body-weight. In conclusion, pET-Ao, can 30 s as an optimal injection duration in a
be calculated with a simple equation as practical multiphasic contrast-enhanced CT
follows (Ichikawa et al., 2006b); protocol of the liver. This recommendation
pET − Ao = an injection duration + 10 s might be reliable based on our previous
results that the peak enhancement values of
In general, peak enhancement time for
the aorta with injection duration of < 30 s
hypervascular HCC (pET-HCC) might
were significantly higher than those with
occur several seconds (∼5 s based on our
the injection duration of > 30 s (Ichikawa
experience) after pET-Ao because it sup-
et al., 2006b).
plies arterial flow from hepatic artery to
the aorta. Therefore, pET-HCC can be cal-
culated with the following equation; REFERENCES
Awai, K., and Hori, S. 2003. Effect of contrast
pET − HCC = an injection duration + 15 s injection protocol with dose tailored to patient
Thus, use of fixed injection duration can weight and fixed injection duration on aortic and
hepatic enhancement at multidetector-row heli-
minimize the differences in HAP scan
cal CT. Eur. Radiol. 13: 2155–2160.
duration for individual patients and there- Awai, K., Hiraishi, K., and Hori, S. 2004. Effect
fore, this method might become standard of contrast material injection duration and rate
rather than the use of fixed injection rate on aortic peak time and peak enhancement at
that makes a difference in injection duration dynamic CT involving injection protocol with
240 T. Ichikawa and T. Araki

dose tailored to patient weight. Radiology 230: weight-tailored dose of contrast material. Eur. J.
142–150. Radiol. 58: 165–176.
Bae, K.T. 2003. Peak contrast enhancement in CT Ichikawa, T., Nakajima, H., Nanbu, A., Hori, M.,
and MR angiography: when does it occur and and Araki, T. 2006b. Effect of injection rate of
why? Pharmacokinetic study in a porcine model. contrast material on CT of hepatocellular carci-
Radiology 227: 809–816. noma. Am. J. Radiol. 186: 1413–1418.
Bae, K.T., Heiken, J.P., and Brink, J.A. 1998. Aortic Kim, T., Murakami, T., and Oi, H. 1995. Detection
and hepatic contrast medium enhancement at of hypervascular hepatocellular carcinoma by
CT. Part II. Effect of reduced cardiac output in a dynamic MRI and dynamic spiral CT. J. Comput.
porcine model. Radiology 207: 657–662. Assist. Tomogr. 19: 948–954.
Baron, R.L., Oliver, J.H., Dodd, G.D., Nalesnik, Kim, T., Murakami, T., and Takahashi, S. 1998. Effects
M., Holbert, B.L., and Carr, B. 1996. of injection rates of contrast material on arterial
Hepatocellular carcinoma: Evaluation with phase hepatic CT. Am. J. Radiol. 171: 429–432.
biphasic, contrast-enhanced, helical CT. Mitsuzaki, K., Yamashita, Y., Ogata, I., Nishiharu,
Radiology 199: 505–511. T., Urata, J., and Takahashi, M. 1996. Multiple-
Hollett, M.D., Jeffrey, R.B., Nino-Murcia, M., phase helical CT of the liver for detecting small
Jorgensen, M.J., and Harris, D.P. 1995. hepatomas in patients with liver cirrhosis: con-
Dual-phase helical CT of the liver: value of arterial trast-injection protocol and optimal timing. Am.
phase scans in the detection of small (<1.5 cm) J. Radiol. 167: 753–757.
malignant hepatic neoplasms. Am. J. Radiol. Ohashi, I., Hanafusa, K., and Yoshida, T. 1993.
164: 879–884. Small hepatocellular carcinomas: two-phase
Hwang, G.J., Kim, M.J., Yoo, H.S., and Lee, dynamic incremental CT in detection and evalu-
J.T. 1997. Nodular hepatocellular carcinomas: ation. Radiology 189: 851–855.
detection with arterial-, portal-, and delayed- Tublin, M.E., Tessler, F.N., Cheng, S.L., Peters,
phase images at spiral CT. Radiology 202: T.L., and McGovern, P.C. 1999. Effect of injec-
383–388. tion rate of contrast medium on pancreatic and
Ichikawa, T., Erturk, S.M., and Araki, T. 2006a. hepatic helical CT. Radiology 210: 97–101.
Multiphasic contrast-enhanced multidetector- Yu, J.S., Kim, K.W., Sung, K.B., Lee, J.T., and
row CT of liver: contrast-enhancement theory Yoo, H.S. 1997. Small arterial-portal venous
and practical scan protocol with a combination shunts: a cause of pseudolesions at hepatic imag-
of fixed injection duration and patients’ body- ing. Radiology 203: 737–742.
18
Detection of Combined Hepatocellular
and Cholangiocarcinomas: Enhanced
Computed Tomography
Akihiro Nishie and Kengo Yoshimitsu

INTRODUCTION chronic liver disease in the cHCC-CC


group is also intermediate between that in
Most primary liver cancers are grossly the HCC group and in the CC group (Yano
divided into two histologic categories: et al., 2003; Liu et al., 2003), or similar to
hepatocellular carcinoma (HCC), derived that in the HCC group (The liver cancer
from hepatocytes, and cholangiocellular study group of Japan, 1990). On the other
carcinoma (CC), arising from the intra- hand, Jarnagin et al. (2001) reported that
hepatic bile duct epithelium. Combined the incidence of positive hepatitis B or
hepatocellular and cholangiocarcinoma C serology and liver cirrhosis in patients
(cHCC-CC) is a rare form of primary liver with cHCC-CC was similar to that in CC
cancer presenting histologic evidence of patients. The reasons for the difference
both hepatocellular and biliary epithelial in this clinicopathological feature may
differentiation, and is seen in 1.0–3.3% include the difference between Western
of surgically treated patients with pri- and Asian societies in the cause of liver
mary liver tumors (The liver cancer study cancer or the difference in the selection
group of Japan, 1990; Aoki et al., 1993). criteria of patients for hepatic resection
Comparing the clinicopathological fea- (Liu et al., 2003). The survival rate in
tures of patients with cHCC-CC with patients with cHCC-CC has been reported
those of HCC or CC patients has yielded to be worse than that in patients with HCC
somewhat conflicting results in several or CC (Jarnagin et al., 2001; Yano et al.,
studies. In many studies, cHCC-CC was 2003; Liu et al., 2003), while Koh et al.
found to be more prevalent in males (Lee (2005) concluded that the cHCC-CC group
et al., 2002). The incidence of positive had a prognosis that was better than the
hepatitis B or C serological test results CC group but worse than the HCC group.
in the cHCC-CC group is reported to be We have not come to a conclusion about
intermediate between that in the HCC clinicopathological features of cHCC-CC
group and in the CC group (Liu et al., at the present time, and more cases must
2003; Koh et al., 2005), or to be similar to be analyzed to elucidate them.
that in the HCC group (Yano et al., 2003). In our daily practice we tend to be
Furthermore, the incidence of underlying distressed at the difficulty of making a

241
242 A. Nishie and K. Yoshimitsu

preoperative diagnosis of this type of modalities, small HCC and CC can now be
tumor because it is rarely encountered. In diagnosed with relative ease (Honda et al.,
an examination performed by The liver 1993a, b), while a prospective diagnosis of
cancer study group of Japan (1990), the cHCC-CC on enhanced CT remains diffi-
frequency of lymph node metastases in cult, although several reports describe CT
patients with cHCC-CC was 76.2% and findings of this entity (Aoki et al., 1993;
was as high as that in patients with CC, Fukukura et al., 1997; Nishie et al., 2005).
while that in patients with HCC is 30.3%. We herein describe enhanced CT findings
Some of the literature suggests that there of cHCC-CC and discuss the possibility of
was a need for a lymph node resection dur- making a diagnosis of cHCC-CC based on
ing a hepatectomy for cHCC-CC (Uenishi enhanced CT.
et al., 2000; Sasaki et al., 2001; Eguchi
et al., 2002). Therefore, it is important to
make a diagnosis of cHCC-CC preopera- PATHOGENESIS
tively.
Fine needle aspiration biopsy (FNAB) In general, the histopathological definition
is one of the possible methods of diagnos- of cHCC-CC is based on criteria proposed
ing this entity. However, making a reliable by the World Health Organization: a tumor
diagnosis of cHCC-CC based on cytologic that contains unequivocal elements of both
preparations alone is very difficult because HCC and CC. If bile production, intercel-
HCC and CC have a wide spectrum of lular bile canaliculi, trabecular growth
morphologic features (Dusenbery, 1997). pattern or all three are recognized, the
Wee and Nilsson (1999) mentioned that, at epithelium is defined as a hepatocellular
the very least, there should be a high index element; if mucin production, definite
of suspicion, especially when the clinical gland formation, or both are noted, the
history is highly suggestive of HCC, and epithelium is defined as a cholangiocel-
FNAB reveals only an adenocarcinoma or lular component (Gibson, 1978). Allen
tumor cells which could be interpreted as and Lisa (1949) first defined three types
intermediate cells. They also suggested that of cHCC-CC: (1) HCC and CC originating
the FNAB diagnosis of cHCC-CC may be from different sites in the liver, but consist-
possible if the clinical, cytohistologic, and ing of a uniform cell type (double cancer);
immunohistochemical findings support (2) contiguous HCC and CC originating
the presence of two cellular components, independently, but intermingling as they
HCC and adenocarcinoma. However, the grow (combined type); and (3) completely
usefulness of FNAB in diagnosing cHCC- intermingled HCC and CC (mixed type).
CC has been previously discussed in only On the other hand, Goodman et al.
a few studies, and presently we have not (1985) also defined criteria for a patho-
come to a conclusion about it. morphological diagnosis of cHCC-CC
On the other hand, sonography, computed that were different from those proposed
tomography (CT), and magnetic resonance by Allen and Lisa (1949). According
imaging (MRI) are now routinely used to this classification, type I, “collision
to detect liver tumors and evaluate their tumors”, contain separate and apparently
characteristics. With the advent of these coincidental components of HCC and CC,
18. Detection of Combined Hepatocellular and Cholangiocarcinomas 243

a b c

Figure 18.1. A 73-year-old man with the mixed type of cHCC-CC. (a) Early phase of enhanced CT. A
tumor with a diameter of 3.5 cm is located in the right lobe. The left-sided main component shows high
density, and the right-sided component under the hepatic capsule shows low density (arrow). (b) Delayed
phase of enhanced CT. The left-sided component shows low density with mild ring-enhancement, and the
right-sided component shows low density (arrow); they are recognized as an HCC-pattern component and
a CC-pattern component, respectively. (c) Macroscopic finding. The inner part of the tumor is a yellowish
mass with partial necrosis and a capsule, representing HCC, and the outside part is a white mass without
expansion, representing CC (arrow). The enhancing pattern of the two components was consistent with
the histological findings. (Nishie et al., 2005; with permission.)

including tumors in the different sites; (Taguchi et al., 1996; Kim et al., 2004). It
however, no transitional forms between is supposed that the latter type of tumor
HCC and CC are observed; type II, “tran- may originate from hepatic progenitor
sitional tumors”, exhibit hepatocellular cells. Another pathological characteris-
differentiation with a transition area to tic of this tumor is a broad desmoplastic
CC or to a mixed hepatocellular glan- stroma encasing the strands and trabecu-
dular tumor; and type III, “fibrolamellar lae of the tumor cells (Kim et al., 2004).
tumors”, resemble a fibrolamellar vari- It is very important to be familiar with the
ant of HCC but contain mucin-producing known histopathological characteristics
glands. Goodman’s type I tumor is equiva- when radiological imaging of cHCC-CC
lent to Allen and Lisa’s double cancer and is analyzed.
combined type, and Goodman’s type II is
similar to Allen and Lisa’s mixed type.
However, Goodman’s type III tumor does ENHANCED COMPUTED
not correspond to any categories in Allen TOMOGRAPHIC FINDINGS
and Lisa’s classification. Furthermore,
another type of cHCC-CC is also present. Investigations into the imaging character-
This type is composed of intermediate istics of cHCC-CC appear to be somewhat
tumor cells that show morphological fea- limited to case reports and small series using
tures intermediate between HCC and CC imaging techniques that are not up to current
244 A. Nishie and K. Yoshimitsu

standards because of the rarity of this entity. That is to say, these results suggest that
Aoki et al. (1993) reported on the CT find- cHCC-CCs tend to show CT findings charac-
ings of 14 lesions in 12 cases with cHCC- terized by a predominant component identi-
CC. Enhanced CT findings were divided into fied in the mass and that it is difficult to make
three types with reference to differences in a diagnosis of cHCC-CC based on enhanced
the enhancement pattern. Type A (n = 9) was CT alone. However, Aoki et al. (1993) evalu-
defined as a mass in which only the periph- ated the values of alpha-fetoprotein (AFP)
eral portion with a wide to narrow width was and carcinoembryonic antigen (CEA), which
variously enhanced as a lesion that remained are tumor markers characteristic to HCC
isodense or as a lesion in which the central and CC, respectively. The AFP level was
portion was of low density or isodense on positive in 65% of patients with cHCC-CC.
the early phase. On the late phase the central Furthermore, 47% of patients had abnormal
portion showed high density or isodensity. CEA values. They concluded that cHCC-CC
We can guess that viable cancer cells are might be suspected when CT findings and
located in the periphery of a mass, and are AFP or CEA values are at variance.
enhanced on the early phase but fade on Fukukura et al. (1997) also analyzed the
the late phase; the central portion of a mass, CT findings of 15 cases with cHCC-CC.
which mainly comprises fibrous tissue and They compared the CT findings on the
scant tumor cells, is enhanced only on the basis of the gross appearance of resected
late phase. These findings are consistent with specimens. The tumors were classified
those of liver metastasis and CC (Muramatsu into two groups: HCC type which is green-
et al., 1986; Honda et al., 1993b). In fact, ish or yellow in color, has hemorrhagic
there were viable cancer cells consisting of necrosis in varying degrees and is sharply
HCC-predominant components in the mar- demarcated with a fibrous capsule; and CC
ginal portion, and the central portion of the type which is whitish in color and shows
tumor was composed of abundant fibrous an infiltrative growth with an irregular mar-
stroma containing CC-predominant compo- gin. In the HCC type (n = 6), most parts of
nents. In the midzone between the peripheral the masses comprised HCC components,
and central areas, HCC and CC components and CC components were observed only
intermingled. Type B (n = 4) was defined as sporadically or focally within the mass.
a mass in which the entire portion was well On the other hand, in the CC type (n = 9),
enhanced on the early phase and faded to four masses had a large volume of HCC
low density on the late phase. This finding is components. In the two masses the ratio
identical to that for HCC. These masses were of HCC to CC components was almost
mainly composed of HCC with CC compo- equal, and three masses consisted of pre-
nents scattered in the mass or of a main mass dominant CC components. All HCC type
of HCC with a small CC area attached to it. masses were entirely enhanced at the early
Type C (n = 1) showed low density on the late phase and faded into low density at the late
phase as well as on the early phase, and this phase. This result is quite consistent with
finding is one of the characteristic CT find- that reported by Aoki et al. (1993). Two
ings in CC (Honda et al., 1993b). The main of the masses also had a ring enhancement
lesion was histopathologically composed of representing a fibrous capsule, which is
CC and an attached small HCC component. also one of the characteristics of HCC
18. Detection of Combined Hepatocellular and Cholangiocarcinomas 245

(Itai et al., 1986). However, eight of nine Twenty-seven patients with cHCC-CCs his-
CC type masses were enhanced only at the topathologically proven by surgical resec-
peripheral portion at the early phase and tion were enrolled in our study. Before the
changed to low density areas or had only evaluation of enhanced CT findings, the
central portions enhanced at the delayed pathologist identified all areas of HCC or
phase; in other words, they showed CT CC cell type and subclassified cHCC-CC
findings compatible with those of CC, according to the criteria of Allen and Lisa
although a sufficient volume of HCC com- (1949). Then, we focused on the enhanc-
ponents was present in at least six cases. ing pattern of a part or component of the
This was because these HCC components lesions. Based on the previous reports, we
presented remarkable fibrosis along the defined the components showing high den-
blood space, which is also seen in scle- sity on the early phase and low density on
rosing HCC (Yamashita et al., 1993). the late phase (high–low pattern) as HCC-
Therefore, these HCC components were pattern component (Honda et al., 1993a),
considered to show CT findings similar to and we defined the components showing
those seen in CC. The volume of fibrous low density on the early phase and low,
stroma in a mass is one of the factors that iso, or high density on the delayed phase
can change enhanced CT findings, and we (low–low, low-iso, or low–high patterns)
are sometimes puzzled when diagnosing as CC-pattern components (Honda et al.,
a liver mass. Fukukura et al. (1997) con- 1993b). We correlated the radiologically
cluded that this CC type can be diagnosed determined components to the histological
as cHCC-CC by evaluating virus markers areas of each cell type. When a complete
and serum AFP levels, which may suggest concordance between the component
the presence of HCC components. detected radiologically and the histological
Choi et al. (1994) reported iodized-oil finding was obtained in terms of site, size,
CT findings in six cases with cHCC-CC. and enhancing pattern, we determined this
On iodized-oil CT scans, a partial reten- to be a correct concordance (Figure 18.1).
tion of iodized oil was observed in the The diagnostic rate (DR) was defined as the
mass in all six cases. HCC portions of ratio of patients in whom CT (HCC- and
pathologic specimens showed compact CC-pattern components) correctly depicted
retention of iodized oil, whereas CC por- HCC/CC to the total number of patients.
tions did not show compact retention. According to the criteria of Allen and
However, it is usually impossible to make Lisa (1949), histological evaluation deter-
a diagnosis of cHCC-CC based only on mined the presence of double cancer,
an iodized-oil CT result because scleros- combined type, and mixed type in 2, 9 and
ing HCC or necrotic portions of HCC also 16 patients, respectively. In the two cases
show no retention of iodized oil. of double cancer, two distinct masses were
How frequently can we diagnose cHCC- described as HCC-pattern and CC-pattern
CC based on enhanced CT alone? We components, respectively. The DR was
evaluated the performance of enhanced 100% (2/2) in double cancer.
CT in making a diagnosis of cHCC-CC On the other hand, the DR was 44.4%
by closely comparing CT findings with (4/9) in the combined type. Why could we
histologic findings (Nishie et al., 2005). not diagnose at a high rate? The reasons
246 A. Nishie and K. Yoshimitsu

for discordance included the different carcinoma with abundant fibrous stroma.
enhancing pattern from the typical pat- Either component was very small (< 1 cm
terns as defined earlier and the small size in diameter) in nine patients and could not
of each component. The CC in one patient be detected on CT. Another reason for the
showed a high–low (HCC) pattern on CT discordance was the presence of tumors
and was composed of poorly differentiated composed of intermediate cells as men-
adenocarcinoma cells with a small amount tioned earlier (Taguchi et al., 1996; Kim
of desmoplastic stroma. Yoshida et al. et al., 2004). Two different components
(1999) also reported two cases of small int- are not reasonably detected in this type
rahepatic CCs with marked hypervascular- of tumor, which is constructed of single
ity, composed of a large number of tumor tumor cells. In our series, three patients
cells and little interstitial fibrous tissue. had mixed type tumors that were shown
Honda et al. (1993b) reported that only 5% to be totally composed of intermediate
of intrahepatic peripheral cholangiocarci- cells, with one showing an HCC pattern
noma showed the high–low pattern. The and the other two showing a CC pattern
HCC in another patient showed a low–low on CT. The two intermediate cell tumors
(CC) pattern on CT and was composed showing a CC pattern had a large amount
of well-differentiated hepatocellular car- of interstitial fibrosis, which is one of the
cinoma cells. Fujita et al. (1996) reported characteristics of this type (Kim et al.,
that 17.6% of well-differentiated HCC 2004) and is histopathologically similar
showed a low–low pattern. That is to say, to CC. However, the third tumor showing
these results suggest that it becomes dif- the HCC pattern was composed of malig-
ficult to diagnose cHCC-CC on CT when nant cells proliferating in a trabecular,
one component of cHCC-CC shows an solid, and tubular pattern. Some of these
atypical enhancing pattern. Furthermore, cells reacted with CA19-9, and only a
another reason for the discordance is that small amount of fibrous tissue was seen.
either component was very small (<1 cm This result suggests that all tumors com-
in diameter) and could not be detected on posed of intermediate cells do not show
CT. This discordance was observed in the the CC enhancement pattern. As a result,
other three patients. the total DR on enhanced CT was 33.3%
The DR in the mixed type was 18.8% (9/27), which was not very high. Variable
(3/16), and even lower than that for the factors such as an atypical enhancement
combined type. Three masses composed pattern, the small size of the HCC or CC
of intermediate cells were included in this components, and the presence of a tumor
group. The main reasons for discordance composed of intermediate cells led to a
included the different enhancing pattern misreading interpretation.
from the typical patterns as defined ear- The main differential diagnosis of
lier, and the small size of each component. cHCC-CCs on enhanced CT is consid-
In one patient the CC-pattern component ered to be HCC or CC containing various
on CT was histologically proven to be kinds of differentiation. An early advanced
HCC. This HCC showed a low-iso (CC) HCC, which is a representative of such
pattern on CT and was composed of tumors, may be differentiated from cHCC-
moderately differentiated hepatocellular CCs by delayed enhancement of a low
18. Detection of Combined Hepatocellular and Cholangiocarcinomas 247

attenuation component seen on the early Dusenbery, D. 1997. Combined hepatocellular-


phase, because most CCs had increased cholangiocarcinoma. Cytologic findings in four
cases. Acta Cytol. 41:903–909.
CT values on the delayed phase compared
Eguchi, H., Nagano, H., Sakon, M., Miyamoto, A.,
with the early phase (Honda et al., 1993b). Kondo, M., Arai, I., Morimoto, O., Dono, K.,
However, it may be very difficult to dif- Umeshima, K., Nakamori, S., Wasaka, K., and
ferentiate such tumors from cHCC-CCs Monden, M. 2002. A successful resection and
on enhanced CT alone when HCC or CC long-term survival of a patient with intrahepatic
components with atypical enhancing pat- recurrences of combined hepatocellular-cholan-
giocarcinoma: report of a case. Surg. Today
terns may be contained in the tumor.
32:742–746.
In conclusion, currently it is not easy Fujita, M., Kuroda, C., Inoue, E., Kumatani, T.,
to make a diagnosis of cHCC-CC cor- Hosomi, N., Narumi, Y., Kuriyama, K., Kadota,
rectly on the basis of enhanced CT alone. T., Kasugai, H., Sasaki, Y., and Ishiguro, S.
However, this type of tumor may be diag- 1996. Usefulness of spiral CT in the detection
nosed by referring to clinical data such as of hypovascular, well-differentiated hepatocel-
lular carcinoma [in Japanese]. Nippon Igaku
tumor markers. Cases with cHCC-CC had
Hoshasen Gakkai Zasshi 56:155–159.
to be recruited over 10 years in most of Fukukura, Y., Taguchi, J., Nakashima, O., Wada,
the previous reports because of the rarity Y., and Kojiro, M. 1997. Combined hepatocellu-
of this entity. Therefore, most of the CT lar and cholangiocarcinoma: correlation between
images were scanned by the incremental CT findings and clinicopathological features. J.
dynamic technique. Multi-detector row Comput. Assist. Tomogr. 21:52–58.
Gibson, J. 1978. Histological typing of tumors of
technology, which presents high spatial
the liver, biliary tract, and pancreas. Geneva,
resolution, may be able to improve the Switzerland: World Health Organization.
detection of small components that are Goodman, Z. D., Ishak, K. D., Langloss, J. M.,
< 1 cm in diameter, and may lead to an Sesterhenn, I. A., and Rabin, L. 1985. Combined
improvement in the diagnostic rate. hepatocellular-cholangiocarcinoma. A histo-
logic and immunohistochemical study. Cancer
55:124–135.
REFERENCES Honda, H., Ochiai, K., Adachi, E., Yasumori, K.,
Allen, R. A., and Lisa, J. R. 1949. Combined liver Hayashi, T., Kawashima, A., Fukuya, T., Gibo,
cell and bile duct carcinoma. Am. J. Pathol. M., Matsumata T., Tsuneyoshi M., and Masuda,
25:647–655. K. 1993a. Hepatocellular carcinoma: correlation
Aoki, K., Takayasu, K., Kawano, T., Muramatsu, of CT, angiographic, and histopathologic find-
Y., Moriyama, N., Wakao, F., Yamamoto, ings. Radiology 189:857–862.
J., Shimada, K., Takayama, T., Kosuge, T., Honda, H., Onitsuka, H., Adachi, E., Ochiai,
Yamasaki, S., Sakamoto, M., and Hirohashi, K., Gibo, M., Yasumori, K., Matsumata, T.
S. 1993. Combined hepatocellular carcinoma Sugimachi, K., and Masuda, K. 1993b.
and cholangiocarcinoma: clinical features and Intrahepatic peripheral cholangiocarcinoma:
computed tomographic findings. Hepatology two-phased dynamic incremental CT and patho-
18:1090–1095. logic correlation. J. Comput. Assist. Tomogr.
Choi, B. I., Han, J. K., Kim, Y. I., Kim, H. C., 17:397–402.
Park, J. H., Kim, C. W., and Han, M. C. 1994. Itai, Y., Ohtomo, K., Kokubo, T., Yamaushi, T.,
Combined hepatocellular and cholangiocarci- Minami, M., Yashiro, N., and Araki, T. 1986.
noma of the liver: sonography, CT, angiography, CT of hepatic masses: significance of prolonged
and iodized-oil CT with pathologic correlation. and delayed enhancement. Am. J. Roentgenol.
Abdom. Imaging 19:43–46. 146:729–733.
248 A. Nishie and K. Yoshimitsu

Jarnagin, W. R., Weber, S., Tickoo, S. K., Koea, hepatocellular and cholangiocellular carcinoma:
J. B., Obiekwe, S., Fong, Y., DeMatteo, R. modes of spreading and choice of surgical treat-
P., Blumgart, L. H., and Klimstra, D. 2001. ment by reference to macroscopic type. J. Surg.
Combined hepatocellular and cholangiocarcinoma: Oncol. 76:37–46.
demographic, clinical, and prognostic factors. Taguchi, J., Nakashima, O., Tanaka, M., Hisaka, T.,
Cancer 94:2040–2046. Takazawa, T., and Kojiro, M. 1996. A clinico-
Kim, H., Park, C., Han, K., Choi, J., Kim, Y. B., pathological study on combined hepatocellu-
Kim, J. K., and Park, Y. N. 2004. Primary liver lar and cholangiocarcinoma. J. Gastroenterol.
carcinoma of intermediate (hepatocyte-cholangi- Hepatol. 11:758–764.
ocyte) phenotype. J. Hepatol. 40:298–304. The liver cancer study group of Japan. 1990.
Koh, K. C., Lee, H., Choi, M. S., Lee, J. H., Paik, Primary liver cancer in Japan: clinicopatho-
S. W., Yoo, B. C., Rhee, J. C., Cho, J. W., Park, logical features and results of surgical treatment.
C. K., and Kim, H. J. 2005. Clinicopathological Ann. Surg. 221:277–287.
features and prognosis of combined hepatocellular Uenishi, T., Hirohashi, K., Shuto, T., Yamamoto, T.,
cholangiocarcinoma. Am. J. Surg. 189:120–125. Kubo, S., Tanaka, H., Ikebe, T., and
Lee, C. C., Wu, C. Y., Chen, J. T., and Chen, Kinoshita, H. 2000. Surgery for mixed hepa-
G. H. 2002. Comparing combined hepatoc- tocellular and cholangiocellular carcinoma.
cellular-cholangiocarcinoma and cholan- Hepatogastroenterology 47:832–834.
giocarcinoma: a clinicopathological study. Wee, A., and Nilsson, B. 1999. Combined hepa-
Hepatogastroenterology 49:1487–1490. tocellular-cholangiocarcinoma: diagnostic chal-
Liu, C. L., Fan, S., Lo, C., Ng, I. O., Lam, C., Poon, lenge in hepatic fine needle aspiration biopsy.
R. T., and Wong J. 2003. Hepatic resection for Acta Cytol. 43:131–138.
combined hepatocellular and cholangiocarci- Yamashita, Y., Fan, Z. M., Yamamoto, H.,
noma. Arch. Surg. 138:86–90. Matsukawa, T., Arakawa, A., Miyazaki, T.,
Muramatsu, Y., Takayasu, K., Moriyama, N., Harada, M., and Takahashi, M. 1993. Sclerosing
Shima, Y., Goto, H., Ushio, K., Yamada T., hepatocellular carcinoma: radiologic findings.
Hasegawa, H., Koyama, Y., and Hirohashi, S. Abdom. Imaging 18:347–351.
1986. Peripheral low-density area of hepatic Yano, Y., Yamamoto, J., Kosuge T., Sakamoto, Y.,
tumors: CT-pathologic correlation. Radiology Yamasaki, S., Shimada, K., Ojima, H.,
160:49–52. Sakamoto, M., Takayama, T., and Makuuchi,
Nishie, A., Yoshimitsu, K., Asayama, Y., Irie, H., M. 2003. Combined hepatocellular and
Aibe, H., Tajima, T., Shinozaki, K., Nakayama, cholangiocarcinoma: a clinicopathologic
T., Kakihara, D., Shimada, M., Aishima, S., study of 26 resected cases. Jpn. J. Clin. Oncol
Yoshida, K., and Honda, H. 2005. Detection of 33:283–287.
combined hepatocellular and cholangiocarcino- Yoshida, Y., Imai, Y., Murakami, T., Nishikawa,
mas on enhanced CT: comparison with histologic M., Kurokawa, M., Yonezawa, T., Tokunaga, K.,
findings. Am. J. Roentgenol. 184:1157–1162. Fukushima, Y., Wakasa, K., Kim, T., Nakamura,
Sasaki, A., Kawano, K., Aramaki, M., Ohno, T., H., Sakon, M., and Monden, M. 1999. Intrahepatic
Tahara, K., Takeuchi, Y., Yoshida, T., and Kitano, cholangiocarcinoma with marked hypervascular-
S. 2001. Clinicopathological study of mixed ity. Abdom. Imaging 24:66–68.
19
Hepatocellular Carcinoma
and Adenomatous Hyperplasia
(Dysplastic Nodules): Dynamic
Computed Tomography and a Combination
of Computed Tomography and Angiography
Kenichi Takayasu

INTRODUCTION hepatic nodular lesions as well as small


HCC (< 3 cm) have been frequently found
Hepatocellular carcinoma (HCC) is the in chronic hepatic disease. They include
fifth most common cancer in the world and adenomatous hyperplasia (AH) corre-
responsible for 500,000 deaths globally every sponding to low grade dysplastic nod-
year (Parkin et al., 2001). Approximately, ule (DN) proposed by the International
80% of the HCC is related to hepatitis B Working Party (1995), atypical AH to high
and/or C virus infection. The prognosis grade dysplastic nodule, and early HCC
remains poor because of advanced stage of (Sakamoto et al., 1991).
cancer and associated hepatic impairment
at diagnosis and high recurrence rate of ∼
80% after surgery (Imamura et al., 2003) or CLASSIFICATION
percutaneous ethanol injection (PEI) (Koda OF NODULAR
et al., 2000) in a different segment of the HEPATOCELLULAR LESIONS
liver. Therefore, early diagnosis is impera-
tive to enable patients to undergo curable To differentiate various small nodular
therapies. According to the nation-wide lesions (< 2 cm) developed in chronic injured
biannual report of the Liver Cancer Study liver, histological criteria were proposed
Group of Japan, HCC accounted for 95% by the Liver Cancer Study Group of Japan
of 19,920 patients with primary liver cancer in 1992, 2003, for early HCC, AH, atypical
(Ikai et al., 2005). AH, and large regenerative nodule. A new
With the advancement of imaging classification system of nodular hepato-
modalities, such as helical computed tom- cellular lesions was introduced by the
ography (CT) (Murakami et al., 2001), International Working Party in 1994. It
and high-quality ultrasonography and consists of low grade DN, high grade
magnetic resonance (MR) imaging, small DN, and HCC. Subsequently, a new guide

249
250 K. Takayasu

book was published by the World Health that of surrounding hepatic parenchyma
Organization (Hirohashi et al., 2000), (Sakamoto et al., 1991), irregular thin
which mentioned that the morphological trabecular pattern, acinar and/or pseu-
criteria for differential diagnosis among doglandular pattern, fatty change and/or
AH, atypical AH and early HCC was still clear cell changes of cancer cells (Kojiro,
under discussion mainly due to the lack of 1997), and stromal invasion (Kondo et al.,
objective phenotypic or genotypic markers. 1994). Early HCC is deemed to be a well-
Recently, the discrepancy between differentiated HCC, namely cancer in
Japanese (Sakamoto et al., 1991) and non- situ. Meanwhile, AH shows a moderate
Japanese (International Working Party, increase in cell density compared with
1995) investigators regarding dysplastic the surrounding tissue but no structural
nodule and early HCC was reported to abnormalities. The nuclear cytoplasmic
reflect differences in interpretation and ratio is slightly increased due to a slight
applications of nomenclature and not due decrease in the amount of cytoplasm (The
to different biological pathways (Hirohashi liver cancer study group of Japan, 1992,
et al., 2000; Theise et al., 2002): 2003). Atypical AH presents with inter-
Pathology of adenomatous hyperplasia, mediate findings between AH and early
atypical adenomatous hyperplasia, early HCC. Nodule-in-nodule HCC consists of
hepatocellular carcinoma, and nodule-in- progressed HCC within an early HCC;
nodule hepatocellular carcinoma. emergence of less differentiated HCC
Macroscopically, the first three entities within a very well differentiated one.
such as AH (Figure 19.1c), atypical AH, Immunohistochemical examination with
and early HCC (Figure 19.2c) are difficult heat-shock protein 70 was reported to be a
to differentiate. However, mean diameters sensitive molecular marker to differentiate
of AH, atypical AH and early HCC were early HCC from precancerous lesion or
0.8, 1.2 and 1.4 cm, respectively, with a sig- noncancerous liver (Chuma et al., 2003).
nificant difference between AH and atypi- The frequency of vascular invasion and/
cal AH and between AH and early HCC or intrahepatic metastases observed micro-
(Takayasu et al., 1989). Nodule-in-nodule scopically in early HCC, nodule-in-nodule
type HCC, composed of internal develop- type HCC, and progressed HCC (Figure
ment of dedifferentiated HCC within early 19.1c) was 5%, 26%, and 35%, respec-
HCC, can be differentiated from the other tively (Watanabe et al., 1990). In our
three entities when the internal nodule hospital, early HCC and nodule-in-nodule
is large enough to be visualized grossly. HCC accounted for 14% and 5.1%, respec-
The boundary of early HCC as well as tively, of surgically resected 980 HCCs in
nodule-in-nodule type HCC is equivocal 664 patients (Oikawa et al., 2005).
to recognize on both intraoperative and
extracorporeal ultrasonography. Thus, care-
ful attention is needed not to leave positive MULTISTEP PROGRESSION OF
margins especially at surgical resection HEPATOCARCINOGENESIS
and/or local ablation therapy.
Microscopically, early HCC presents Clinical and pathological studies have
focal structural abnormalities of increased elucidated multistep progression of hepa-
cell density which are more than twice tocarcinogenesis (Sakamoto et al., 1991).
19. Hepatocellular Carcinoma and Adenomatous Hyperplasia 251

a b

Figure 19.1. A small advanced HCC associated with adenomatous hyperplasia (dysplastic nodule).
Computed Tomography hepatic arteriography (a) shows a small hyperattenuating nodular lesion (arrow)
close to a simple cyst (arrowhead) in segment 3, the lesion was demonstrated as a hypoattenuating one
(arrow) on CT arterial portography (b). Cut surface of resected specimen (c) disclosing two lesions with
almost the same size measuring 1 cm; a yellow-colored lesion was advanced HCC with satellite lesions
or intrahepatic metastases around it (arrow) and a tan-colored one was adenomatous hyperplasia (arrow-
heads), the latter of which was found only by this cut slice

Adenomatous hyperplasia emerges within differentiated HCC. As another pathway,


chronic injured liver, which progresses to de novo hepatocarcinogenesis is also pre-
early HCC. Subsequently, less differenti- sumed to occur.
ated HCC develops within an early HCC, The size of early HCCs is usually < 2 cm
namely nodule-in-nodule type HCC, which in diameter; however, large-size early
finally develops to overt HCC which is HCC and nodule-in-nodule HCC measur-
usually occupied by moderately to poorly ing 5 cm have been rarely encountered.
252 K. Takayasu

a b

Figure 19.2. An early HCC located in segment 8. The dynamic CT in the arterial phase (a) discloses a
faintly hypoattenuating lesion (arrow), which was demonstrated as hypoattenuation in the delayed phase
(b) as well as on unenhanced CT (not shown). Cut surface of resected specimen (c) shows a slightly
yellow-colored equivocal lesion (arrows) with several portal tracts (arrowheads). The intratumoral archi-
tecture is preserved and similar to the surrounding parenchyma. Microscopically, the portal tract and
increased cell density with fatty change are recognized (not shown)

NEEDLE BIOPSY ficiently accurate imaging technique to


distinguish HCC from other benign or
Histopathologic diagnosis is still the gold malignant conditions. However, the diag-
standard, especially for nonenhanced nod- nosis of the small amount of specimen
ular lesions by dynamic CT. According obtained by needle biopsy is not always
to the Barcelona-2000 EASL conference easy due to sampling error and intratu-
(Bruix et al., 2001), needle biopsy is moral heterogeneity. One study reported
recommended for the patient whose hepatic that 9.7% of 99 histologically proven
lesion does not exceed 2 cm due to insuf- regenerative nodules obtained by needle
19. Hepatocellular Carcinoma and Adenomatous Hyperplasia 253

aspiration biopsy showed malignant trans- the other was CT hepatic arteriography
formation at 3 years (Kobayashi et al., (CTHA) in which the catheter tip is
2006). This might evoke a critical problem advanced into the superior mesenteric
of whether it suggests limitation of needle artery or the proper hepatic artery, respec-
biopsy or whether regenerative nodule tively. The former was started 20 s after
actually develops to carcinoma. On the the injection of 90 ml of Ioversol (Optiray;
other hand, needle tract seeding and intra- 350 mgI/ml, Yamanouchi, Tokyo) diluted
peritoneal hemorrhage are still possible with saline (1:3 ratio; iodine, 87.5 mgI/
risks of needle biopsy. ml) at a speed of 3 ml/s after the adminis-
tration of 10 μg of prostaglandin E1 into
the superior mesenteric artery. The latter
METHODS OF was commenced 10 s after the injection of
MULTIDETECTOR 60 ml of the same diluted contrast medium
COMPUTED TOMOGRAPHY into the proper, right or left hepatic artery
at a speed of 1.3–2 ml/s based on the
Helical CT with two (arterial and delayed variation of arterial anatomy. Both the
phases) or three phases (arterial, portal, beam collimation and image reconstruc-
and delayed phases) is widely used by tion was 7 mm. Usually CTAP was first
injecting 100–120 ml of nonionic contrast performed to obtain unequivocal contrast
medium into antecubital veins at a speed of image, followed by celiac arteriography
3 ml/s. Computed tomography is started at to know the variation of artery, and next
35–40 s (arterial phase), 70 s (portal phase), CTHA, and superselective arteriography,
and 3 min (delayed phase) after the start of if necessary.
the injection with contrast medium. Entire
livers are scanned within one breath-hold CT IMAGES OF ADVANCED
for ∼ 8–20 s, which depends on liver size. HEPATOCELLULAR
Scanning parameters are as follows: axial
single- or 4-slice mode, 5–10 mm beam
CARCINOMA
collimation, 0.5–1.0 s/rotation, 0.7–3.0 of In general, advanced HCC is demon-
helical pitch (0.7–1.0 of pitch factor), strated as a hyper-attenuating lesion in
120–150 kVp, and 200–250 mAs. Image arterial phase and hypo-attenuating in
reconstructions are 5–7 mm in thickness. portal and/or delayed phases of dynamic
CT (Takayasu et al., 1990). The washout
of contrast medium from a lesion is more
METHODS OF A clearly seen in delayed phase (3–5 min
COMBINATION OF after contrast injection). The small HCC
COMPUTED TOMOGRAPHY (< 3 cm) is homogeneously enhanced by
AND ANGIOGRAPHY contrast medium (Takayasu et al., 1995a),
and faint ring enhancement is recognized
As a further examination of hemodynamics if it is encompassed by fibrous capsule.
of lesions, a combination study of helical Large HCC (∼ 10 cm) is inhomogeneously
CT and angiography was performed. One enhanced mainly due to associated necro-
was CT arterial portography (CTAP) and sis and hemorrhage. Arterioportal shunt is
254 K. Takayasu

also recognized to show a dilated arterial The delayed phase CT showed the highest
branch followed by hepatopetal (periph- detection rate. Only 5% of early HCC pre-
eral) and/or hepatofugal (retrograde) por- sented hyper-attenuation of lesion in the arte-
tal blood flow in the arterial phase. The rial phase CT. Lim et al. (2000) reported that
filling defect of large vessels secondary to the sensitivity of three-phase helical CT for
portal and/or hepatic vein tumor thrombi DN was 39% in transplant patients. Another
could be seen more clearly in the portal study showed that 20% of 55 lesions of DN
phase. On unenhanced CT, small HCC is (13 high grade DNs and 42 low grade DNs),
usually shown as an iso-attenuating lesion which were initially found by ultrasonogra-
and a large HCC is shown as a hypo- phy, could be detected by helical dynamic
attenuating lesion. The hypo-attenuating CT (Kobayashi et al., 2006). However, these
area appears in hepatic lobar, segmental, results do not always suggest that ultra-
or hepatic hilar area due to the obstruc- sonography is superior to CT for detect-
tion of the portal vein on unenhanced CT. ing DN because ultrasonography generally
The hypo-attenuation area changes due shows high sensitivity and low specificity
to hyper-attenuation in the arterial phase rates as compared to dynamic CT.
secondary to increased arterial blood flow The most common CT pattern among
as a compensation. An advanced HCC is detected lesions was iso-iso-hypo-attenuated
shown as hyper-attenuated on CTHA and in 22%, followed by hypo-hypo-hypo pat-
hypo-attenuated on CTAP due to 100% tern in 19% (Figure 19.2A, B), and several
nutritional supply from the arterial blood different patterns in the remaining 13%.
flow (Figure 19.1A, B). For small HCC The reason for hypo- or iso-attenuation
(< 3 cm), 93% of lesions were hyper- of early HCC is presumed to be mainly
attenuated and 7% were iso-attenuated on immature neovascularization (Takayasu
CTHA, whereas 97% were hypo-attenuated et al., 1989, 1995a). The hypoattenuat-
and 3% were iso-attenuated on CTAP ing lesions on unenhanced CT showed
(Takayasu et al., 1995b). microscopically the tendency of moderate
to high grade of fatty change within the
cytoplasm (Takayasu et al., 1995a).
CT IMAGES OF EARLY CT hepatic arteriography of early HCC
HEPATOCELLULAR was hypo-attenuated in 55%, iso- in 30%,
CARCINOMA and hyper- in 15%. Twenty-seven percent
of early HCCs were hypo-attenuated on
The overall sensitivity of conventional CT both CTAP and CTHA, which suggests
for 39 resected early HCCs was 56%; the that they receive less blood flow from both
remaining 44% could not be detected due the portal and arterial vessels than the sur-
to isoattenuation throughout all phases of rounding parenchyma (Takayasu et al.,
dynamic CT and unenhanced CT. Twenty- 1995b). On the other hand, overall sensitiv-
four percent of the lesions were detected as ity of CTAP for early HCC was 66% shown
hypo-attenuation on unenhanced CT, 31% as hypo-attenuation, and the remaining
as hypo-attenuation in the arterial phase of 34% was iso-attenuated. As an unusual
dynamic CT, and 51% as hypo-attenuation in case, hyper-attenuating early HCC and/or
the delayed phase (Takayasu et al., 1995a). AH shown on CTAP has been reported.
19. Hepatocellular Carcinoma and Adenomatous Hyperplasia 255

To understand the serial change of atten- 33.3% on CTHA, respectively (Takayasu


uation of the nodular lesions on dynamic et al., 1989). Even CTAP and CTHA often
CT, both pathologic (Ueda et al., 1992) failed to detect AH (Figure 19.1A, B).
and hemodynamic studies with CTAP and When the lesions of AH and/or atypical
CTHA (Hayashi et al., 1999) have been AH were detected on CT and/or CTHA,
performed by focusing on multistep pro- they were usually shown as hypoattenu-
gression of hepatocarcinogenesis. At the ated ones which is similar to early HCC.
initial stage of multistep hepatocarcino- AH is more difficult to detect on various
genesis, AH receives almost similar blood images due to its smaller size as compared
supply to nontumorous parenchyma from to atypical AH and early HCC (Takayasu
both arterial and portal blood flow; subse- et al., 1989).
quently a gradual decline of blood supply
from both vessels occurs mainly due to the
decrease in the number of the portal tracts COMPUTED TOMOGRAPHY
such as in atypical AH and/or early HCC. IMAGES OF NODULE-IN-
However, arterial blood flow only continu- NODULE HEPATOCELLULAR
ously increases following the temporary CARCINOMA
decrease secondary to the development of
unpaired artery which might occur during When a dedifferentiated component (mod-
the process from early HCC to nodule-in- erately or poorly differentiated HCC)
nodule HCC. Finally, arterial blood flow emerges within a well-differentiated HCC,
alone nourishes the lesion of advanced the lesion presents with a nodule-in-nodule
HCC. The portal blood supply shows one- appearance. The arterial phase CT dis-
way decrease during the multistep pro- closes the central area hyperattenuated
gression, resulting in complete depletion and the peripheral area remains iso- or
in advanced HCC. The arterial blood flow hypoattenuated. In the delayed phase, the
into the lesions draws the inverted para- entire lesion becomes hypoattenuated
bolic curve during the process from AH because the central enhanced area changes
to advanced HCC which might explain the to hypoattenuation (Winter et al., 1994). In
unstable state of attenuation of lesions onother words, the CT attenuation behavior
dynamic CT and CTHA. of the central and peripheral portions of
nodule-in-nodule type HCC is similar to
that of isolated advanced HCC and iso-
COMPUTED TOMOGRAPHY lated early HCC, respectively. CT arterial
IMAGES OF ADENOMATOUS portography and CT hepatic arteriography
HYPERPLASIA AND could show more precise hemodynamics
ATYPICAL ADENOMATOUS of this entity (Winter et al., 1994).
HYPERPLASIA A lesion requires differential diagnosis
from early HCC, complicated cyst (hemor-
The detection rate of 14 AHs and 12 atypi- rhagic cyst), atypical hemangioma, which
cal AHs surgically resected was 0% and is hardly enhanced due to entirely hyali-
50% on US, 0% and 9.1% on dynamic nized degeneration, necrotic regenerative
CT, 50% and 33.3% on CTAP, and 0% and nodule and hypovascular metastasis.
256 K. Takayasu

NATURAL OUTCOME OF (1998) studied the natural outcome of


HYPO-ATTENUATING hypovascular nodules on angiography and
NODULAR LESIONS well-differentiated HCC histologically
with ultrasonography; as a result, 13 (72%)
It is important to elucidate the natural out- and 1 (6%) of 18 AH nodules developed to
come of hypo-attenuating nodules mainly early HCC and overt HCC, respectively;
corresponding to early HCC as well as while, 4 (33%) of 12 early HCCs pro-
atypical AH in order to know the bio- gressed to overt HCC without observation
logical behavior, to determine the optical of the stage of early HCC on ultrasonogra-
treatment, and to predict risk factors of phy. Borzio et al. (2003) reported that 28
malignant transformation. Therefore, we (31%) ultrasound-detected macronodules
studied the natural outcome of 60 hypo- (including large regenerative nodule, low
attenuating nodular lesions in chronic liver grade DN and high grade DN) in cirrhosis
disease using follow-up CT. As a result, 36 transformed into HCC at 3-years. Recently,
(60%) lesions showed attenuation conver- Kobayashi et al. (2006) reported that the
sion from hypo- to hyper-attenuation sug- malignant transformation was recognized
gesting malignant transformation occurred. in 29 of 154 (18.8%) nodular lesions. The
Twenty-one hypo-attenuating lesions were biopsy showed high or low grade DN and
unchanged in attenuation, and the remain- regenerative nodules which were initially
ing three disappeared spontaneously found by ultrasonography. In addition,
(Takayasu et al., 2006). Overall cumulative 3-year malignant transformation rate of
attenuation conversion rates from hypo- to high grade DN was 61.5% that was very
hyper-attenuation were 15.8% at 1-year, similar to ours of 58.7% studied by follow-
44.3% at 2-years, and 58.7% at 3-years. up dynamic CT (Takayasu et al., 2006). The
Thirty-six lesions, converted to hyper- annual rate of malignant transformation
attenuation, consisted of two subtypes; (Kobayashi et al., 2006) and conversion
one was hyper-in-hypoattenuating lesion attenuation (Takayasu et al., 2006) were
suggesting nodule-in-nodule type HCC 20% and 15.8%, respectively. Their data
in 25 lesions and the other was entirely were higher than those of occurrence
hyperattenuating nodule corresponding to of HCC ranging 4–7% in patients with
overt HCC in 11 lesions. Between the two chronic viral hepatic disease (Ikeda et al.,
subtype groups, the attenuation conversion 1993; Tsai et al., 1997).
rate from hypo- to hyper-in-hypo or from A hyper-in-hypoattenuating nodular
hypo- to entirely hyperattenuation was not lesion suggesting nodule-in-nodule HCC
significantly different. showed rapid progression to an entirely
Takayama et al. (1990) reported that hyper-attenuating one (overt HCC)
histologically proven AH developed to (Takayasu et al., 2006). Sadek et al. (1995)
advanced HCC with a malignant transfor- and Hayashi et al. (2002) also reported
mation of 22% (4 of 18 lesions) at 1-year, similar findings in their follow-up with
50% (5 of 10) at 2-years, and 80% (4 of MRI and a combination study of CT and
5) at 3-years. Sakamoto and Hirohashi angiography, respectively.
19. Hepatocellular Carcinoma and Adenomatous Hyperplasia 257

CRITICAL CONSIDERATION TO in order to reveal the intratumoral vas-


TREAT HYPOATTENUATING culature by separately evaluating portal
LESIONS and arterial blood supply (Takayasu et al.,
1995b; Hayashi et al., 1999). However, it
Consensus has not been reached regard- is invasive and periodical follow-up is not
ing the management of hypo-attenuating adequate. Instead, dynamic MR imaging or
nodular lesion in virus-related chronic multidetector CT is an alternative. Because
liver disease. Until now, several treat- radiation hazard of CT is problematic,
ments have been performed. However, contrast enhanced ultrasonography should
they have some limitations; surgery is replace dynamic CT (Ding et al., 2001).
invasive (Takayama et al., 1998), transar- In conclusion, early HCC derived from
terial chemoembolization (TACE) is not chronic injured liver was mainly demon-
effective (Takayasu et al., 1993), and local strated as hypo- or iso-attenuating nodule
ablation therapy sometimes has severe in both arterial and delayed phases of
complications such as needle tract implan- dynamic CT. A hyper-attenuating early
tation in PEI (Ishii et al., 1998), and RFA HCC was only 5% on arterial phase of
related death (de Baere et al., 2003). dynamic CT and 15% on CT hepatic
There is no evidence for early treatment arteriography. Sixty percent of 60 hypo-
of hypo-attenuating nodular lesions to attenuating lesions developed to hyper-
provide benefit to patients with chronic in-hypo (nodule-in-nodule type HCC) or
injured liver. Some researchers do not entirely enhanced lesion (overt HCC) with
agree with early treatment of such lesions cumulative attenuation conversion rates
because good survival could be obtained of 15.8% at 1-year, 44.3% at 2-years,
mainly due to lead-time bias. There is a and 58.7% at 3 years. Thus, careful and
belief that the real critical factor that deter- periodic follow-up helical CT is needed to
mines the prognosis is not a hypo-vascular survey early detection of attenuation con-
lesion but a hyper-vascular advanced HCC version or malignant transformation.
which abruptly appears as a multicentric
fashion of occurrence in the different seg- REFERENCES
ment of the liver during the follow-up of
hypo-vascular lesion. The uncertainness Borzio, M., Fargion, S., Borzio, F., Fracanzani,
A.L., Croce, A.M., Stroffolini, T., Oldani, S.,
of natural history of hypovascular lesion is Cotichini, R., and Roncalli, M. 2003. Impact
another problem; spontaneous disappear- of large regenerative, low grade and high grade
ance was reported with a wide range from dysplastic nodules in hepatocellular carcinoma
5% (Takayasu, in press) to 40% (Tochio development. J. Hepatol. 39: 208–214.
et al., 1994) when studied by CT and ultra- Bruix, J., Sherman, M., Llovet, J.M., Beaugrand, M.,
sonography, respectively. Lencioni, R., Burroughs, A.K., Christensen, E.,
Pagliaro, L., Colombo, M., and Rodes, J. 2001.
The combination of CT hepatic arteri- Clinical management of hepatocellular carcinoma.
ography and CT arterial portography is Conclusions of the Barcelona-2000 EASL confer-
a precise examination to determine the ence. European Association for the Study of the
malignancy of the hypo-vascular lesion Liver. J. Hepatol. 35: 421–430.
258 K. Takayasu

Chuma, M., Sakamoto, M., Yamazaki, K., Ohta, observation of 795 patients with viral and alco-
T., Ohki, M., Asaka, M., and Hirohashi, S. holic cirrhosis. Hepatology 18: 47–53.
2003. Expression profiling in multistage hepa- Imamura, H., Matsuyama, Y., Tanaka, E., Ohkubo, T.,
tocarcinogenesis: identification of HSP70 as a Hasegawa, K., Miyagawa, S., Sugawara, Y.,
molecular marker of early hepatocellular carci- Minagawa, M., Takayama, T., Kawasaki, S., and
noma. Hepatology 37: 198–207. Makuuchi, M. 2003. Risk factors contributing to
de Baere, T., Risse, O., Kuoch, V., Dromain, C., early and late phase intrahepatic recurrence of
Sengel, C., Smayra, T., Gamal El Din, M., hepatocellular carcinoma after hepatectomy.
Letoublon, C., and Elias, D. 2003. Adverse events J. Hepatol. 38: 200–207.
during radiofrequency treatment of 582 hepatic International Working Party. 1995. Terminology of
tumors. AJR Am. J. Roentgenol. 181: 695–700. nodular hepatocellular lesions. Hepatology 22:
Ding, H., Kudo, M., Onda, H., Suetomi, Y., Minami, Y., 983–993.
and Maekawa, K. 2001. Hepatocellular carci- Ishii, H., Okada, S., Okusaka, T., Yoshimori, M.,
noma: depiction of tumor parenchymal flow Nakasuka, H., Shimada, K., Yamasaki, S.,
with intermittent harmonic power Doppler US Nakanishi, Y., and Sakamoto, M. 1998. Needle
during the early arterial phase in dual-display tract implantation of hepatocellular carcinoma
mode. Radiology 220: 349–356. after percutaneous ethanol injection. Cancer 82:
Hayashi, M., Matsui, O., Ueda, K., Kawamori, Y., 1638–1642.
Kadoya, M., Yoshikawa, J., Gabata, T., Kobayashi, M., Ikeda, K., Hosaka, T., Sezaki, H.,
Takashima, T., Nonomura, A., and Nakanuma, Y. Someya, T., Akuta, N., Suzuki, F., Suzuki, Y.,
1999. Correlation between the blood supply and Saitoh, S., Arase, Y., and Kumada, H. 2006.
grade of malignancy of hepatocellular nodules Dysplastic nodules frequently develop into hepa-
associated with liver cirrhosis: evaluation by CT tocellular carcinoma in patients with chronic viral
during intraarterial injection of contrast medium. hepatitis and cirrhosis. Cancer 106: 636–647.
AJR Am. J. Roentgenol. 172: 969–976. Koda, M., Murawaki, Y., Mitsuda, A., Ohyama, K.,
Hayashi, M., Matsui, O., Ueda, K., Kawamori, Y., Horie, Y., Suou, T., Kawasaki, H., and Ikawa, S.
Gabata, T., and Kadoya, M. 2002. Progression to 2000. Predictive factors for intrahepatic recur-
hypervascular hepatocellular carcinoma: corre- rence after percutaneous ethanol injection ther-
lation with intranodular blood supply evaluated apy for small hepatocellular carcinoma. Cancer
with CT during intraarterial injection of contrast 88: 529–537.
material. Radiology 225: 143–149. Kojiro, M. 1997. Pathology of hepatocellular
Hirohashi, S., Ishak, K., Kojiro, M., Wanless, I., carcinoma. In: Okuda, K., Tabor, E., editors.
Theise, N., Tsukuma, H., Blum, H., Deugnier, Y., Liver Cancer. New York: Churchill Livingstone.
Laurent Puig, P., Fischer, H., and Sakamoto, M. pp. 165–187.
2000. Hepatocellular carcinoma. In: Hamilton, S., Kondo, F., Kondo, Y., Nagato, Y., Tomizawa, M.,
Aaltonen, L., editors. World Health Organization and Wada, K. 1994. Interstitial tumour cell
Classification of Tumours: Pathology and invasion in small hepatocellular carcinoma.
Genetics of Tumours of the Digestive System. Evaluation in microscopic and low magnification
Lyon: International Agency for Research on views. J. Gastroenterol. Hepatol. 9: 604–612.
Cancer Press. pp. 159–172. Lim, J.H., Kim, C.K., Lee, W.J., Park, C.K., Koh,
Ikai, I., Arii, S., Ichida, T., Okita, K., Omata, M., K.C., Paik, S.W., and Joh, J.W. 2000. Detection
Kojiro, M., Takayasu, K., Nakanuma, Y., of hepatocellular carcinomas and dysplastic nod-
Makuuchi, M., Matsuyama, Y., and Yamaoka, Y. ules in cirrhotic livers: accuracy of helical CT in
2005. Report of the 16th follow-up survey of pri- transplant patients. AJR Am. J. Roentgenol. 175:
mary liver cancer. Hepatol. Res. 32: 163–172. 693–698.
Ikeda, K., Saitoh, S., Koida, I., Arase, Y., Tsubota, A., Murakami, T., Kim, T., Takamura, M., Hori, M.,
Chayama, K., Kumada, H., and Kawanishi, M. Takahashi, S., Federle, M.P., Tsuda, K., Osuga,
1993. A multivariate analysis of risk factors K., Kawata, S., Nakamura, H., and Kudo, M.
for hepatocellular carcinogenesis: a prospective 2001. Hypervascular hepatocellular carcinoma:
19. Hepatocellular Carcinoma and Adenomatous Hyperplasia 259

detection with double arterial phase multi-detec- and Hirohashi, S. 1990. The diagnosis of small
tor row helical CT. Radiology 218: 763–767. hepatocellular carcinomas: efficacy of various
Oikawa, T., Ojima, H., Yamasaki, S., Takayama, T., imaging procedures in 100 patients. AJR Am. J.
Hirohashi, S., and Sakamoto, M. 2005. Multistep Roentgenol. 155: 49–54.
and multicentric development of hepatocellular Takayasu, K., Wakao, F., Moriyama, N., Muramatsu, Y.,
carcinoma: histological analysis of 980 resected Sakamoto, M., Hirohashi, S., Makuuchi, M.,
nodules. J. Hepatol. 42: 225–229. Kosuge, T., Takayama, T., and Yamazaki, S.
Parkin, D.M., Bray, F., Ferlay, J., and Pisani, P. 1993. Response of early-stage hepatocellular
2001. Estimating the world cancer burden: carcinoma and borderline lesions to therapeutic
Globocan 2000. Int. J. Cancer 94: 153–156. arterial emboli-zation. AJR Am. J. Roentgenol.
Sadek, A.G., Mitchell, D.G., Siegelman, E.S., 160: 301–306.
Outwater, E.K., Matteucci, T., and Hann, H.W. Takayasu, K., Furukawa, H., Wakao, F., Muramatsu, Y.,
1995. Early hepatocellular carcinoma that devel- Abe, H., Terauchi, T., Winter, T.C., 3rd,
ops within macroregenerative nodules: growth Sakamoto, M., and Hirohashi, S. 1995a. CT
rate depicted at serial MR imaging. Radiology diagnosis of early hepatocellular carcinoma:
195: 753–756. sensitivity, findings, and CT-pathologic correla-
Sakamoto, M., and Hirohashi, S. 1998. Natural tion. AJR Am. J. Roentgenol. 164: 885–890.
history and prognosis of adenomatous hyperpla- Takayasu, K., Muramatsu, Y., Furukawa, H., Wakao, F.,
sia and early hepatocellular carcinoma: multi- Moriyama, N., Takayama, T., Yamasaki, S.,
institutional analysis of 53 nodules followed up Sakamoto, M., and Hirohashi, S. 1995b. Early
for more than 6 months and 141 patients with hepatocellular carcinoma: appearance at CT during
single early hepatocellular carcinoma treated by arterial portography and CT arteriography with
surgical resection or percutaneous ethanol injec- pathologic correlation. Radiology 194: 101–105.
tion. Jpn. J. Clin. Oncol. 28: 604–608. Takayasu, K.M., Mizuguchi, Y., Okusaka, T., Shimada,
Sakamoto, M., Hirohashi, S., and Shimosato, Y. K., Takayama, T., and Sakamoto, M. 2006. CT
1991. Early stages of multistep hepatocarcino- evaluation of the progression of hypoattenuating
genesis: adenomatous hyperplasia and early nodular lesions in virus related chronic liver dis-
hepatocellular carcinoma. Hum. Pathol. 22: ease. AJR Am. J. Roentgenol. 187: 454–463.
172–178. Theise, N.D., Park, Y.N., and Kojiro, M. 2002.
Takayama, T., Makuuchi, M., Hirohashi, S., Dysplastic nodules and hepatocarcinogenesis.
Sakamoto, M., Okazaki, N., Takayasu, K., Clin. Liver Dis. 6: 497–512.
Kosuge, T., Motoo, Y., Yamazaki, S., and The liver cancer study group of Japan. 1992. The
Hasegawa, H. 1990. Malignant transformation General Rules for the Clinical and Pathological
of adenomatous hyperplasia to hepatocellular Study of Primary Liver Cancer, 3rd ed. Tokyo:
carcinoma. Lancet 336: 1150–1153. Kanehara.
Takayama, T., Makuuchi, M., Hirohashi, S., The liver cancer study group of Japan. 2003.
Sakamoto, M., Yamamoto, J., Shimada, K., General Rules for the Clinical and Pathological
Kosuge, T., Okada, S., Takayasu, K., and Study of Primary Liver Cancer, 2nd English ed.
Yamasaki, S. 1998. Early hepatocellular carci- Tokyo: Kanehara.
noma as an entity with a high rate of surgical Tochio, H., Tomita, S., Kudo, M., Okabe, Y.,
cure. Hepatology 28: 1241–1246. Kashida, H., Hamada, M., Yamamoto, K., Terai,
Takayasu, K., Makuuchi, M., Hirohashi, S., Y., Itani, T., Mimura, J., Hirasa, M., Ibuki, Y.,
Okazaki, N., Muramatsu, Y., Moriyama, N., Komori, H., Orino, A., and Todo, A. 1994.
Takayama, T., and Hasegawa, H. 1989. Imaging Clinical tracing of small hypovascular hepatic
of adenomatous hyperplastic lesions containing nodules associated with chronic liver disease.
and not containing hepatocellular carcinoma in Acta Hepatol. Jpn. 35: 333–346.
the liver. Jpn. J. Gastroenterol. 86: 2404–2412. Tsai, J.F., Jeng, J.E., Ho, M.S., Chang, W.Y.,
Takayasu, K., Moriyama, N., Muramatsu, Y., Hsieh, M.Y., Lin, Z.Y., and Tsai, J.H. 1997.
Makuuchi, M., Hasegawa, H., Okazaki, N., Effect of hepatitis C and B virus infection on risk
260 K. Takayasu

of hepatocellular carcinoma: a prospective study. small hepatocellular carcinoma -common type in


Br. J. Cancer 76: 968–974. early type- and histological consideration of the
Ueda, K., Terada, T., Nakanuma, Y., and Matsui, multistep progression of HCC. Acta Hepatol.
O. 1992. Vascular supply in adenomatous hyper- Jpn. 31: 1267–1273.
plasia of the liver and hepatocellular carci- Winter, T.C., 3rd, Takayasu, K., Muramatsu, Y.,
noma: a morphometric study. Hum. Pathol. 23: Furukawa, H., Wakao, F., Koga, H., Sakamoto,
619–626. M., Hirohashi, S., and Freeny, P.C. 1994. Early
Watanabe, H.H., Sakamoto, M., Hasegawa, H., advanced hepatocellular carcinoma: evaluation
Yamazaki, S., Makuuchi, M., Moriyama, N., and of CT and MR appearance with pathologic cor-
Takayasu, K. 1990. New macroscopic subtype of relation. Radiology 192: 379–387
20
Hepatocellular Cancer in Cirrhotic
Patients: Radiological Imaging
Francesca Lodato, N. Davies, D. Yu, and Andrew K. Burroughs

INTRODUCTION a potential curative therapy, but all thera-


pies have best results with single nodules
Hepatocellular carcinoma (HCC) is the £ 3 cm (Bruix et al., 2001).
major cause of death in cirrhotic patients. This clinical scenario has important
It is the fifth most common neoplasm implications for strategies for early diag-
and the third most common cause of nosis of HCC. However, there are no
death related to cancer in the world randomized controlled trials concerning
(Parkin et al., 2001; Llovet et al., 2003). surveillance for HCC in cirrhosis, but
Moreover, data on the epidemiology and only cohort studies. Two studies show a
natural history of chronic hepatitis C higher chance of receiving treatment at
virus (HCV) infection, suggest that its an earlier stage (Sangiovanni et al., 2004)
frequency has been increasing over the and strongly support current practice of
past 20–30 years in the U.S (El Serag and surveillance in most centres, despite the
Mason, 1999) and in Europe (Deuffic et lack of formal evidence. The knowledge
al., 1998), and it is the leading cause of that all treatments yield better results
death in patients with cirrhosis in Europe when used for smaller and unifocal HCC
(Fattovich et al., 1997). (Bruix et al., 2001) is the linchpin to cur-
There are several potential therapies for rent practice. Therefore, it is important
HCC including resection, liver transplan- to be accurate in both the diagnosis and
tation and several percutaneous as well staging of HCC as this has prognos-
as transarterial ablative techniques, but tic significance and helps programming
their applicability is limited by several potential treatments. For both orthotopic
anatomical factors, such as the size and liver transplantation (OLT) and resection
number of nodules, their location and also the survival is higher for patients with a
the patients liver reserve. Unfortunately single nodule smaller than 5 cm or with
only a minority of cirrhotic patients diag- three nodules each 3 cm in diameter or
nosed with HCC have tumor amenable to less (Adam and Del Gaudio, 2003).

261
262 F. Lodato et al.

THE EASL CONSENSUS months. Surveillance is recommended also


STATEMENTS AND AASLD by AASLD guidelines.
GUIDELINES Secondly, the EASL consensus state-
ments categoriz cirrhotic patients on the
An expert panel on HCC from the European basis of the size of the single nodule. For
Association for the Study of the Liver tumors > 2 cm, diagnosis can be made
(EASL) formulated a consensus state- using two noninvasive radiological tech-
ment in 2001 to standardize the approach niques demonstrating arterial hypervas-
on HCC diagnosis and treatment (Figure cularity (Lim et al., 2000; Nino-Murcia
20.1) (Bruix et al., 2001). These have et al., 2000). When alfa-fetoprotein is
evolved, and recent practice guidelines > 400 ng/ml, only one imaging technique
from the American Association for the is considered sufficient to confirm the
Study of Liver Diseases (AASLD), have diagnosis. It should be remembered that
included data from later studies and tech- a biopsy for a nodule > 2 cm in a cirrhotic
nical advances (Figure 20.2) (Bruix and liver which does not confirm malignancy
Sherman, 2005). From the diagnostic point cannot be considered definitive; using
of view, EASL recommendations define only radiological diagnostic techniques
histological and radiological criteria for the and clinical findings, the positive predictive
identification of HCC. Firstly, surveillance value is higher than 95% (Torzilli et al.,
is recommended for cirrhotics in whom 1999; Levy et al., 2001). However there is
potentially effective treatment for HCC no mention of discordant vascular patterns
can be offered if tumors can be detected in the radiological imaging. The American
early. The available data on tumor growth Association for the Study of Liver Diseases
suggest that the interval from undetectable Guidelines revised these statements, giv-
lesions to 2 cm diameter is about 4–12 ing diagnostic importance to venous/portal
months (Ebara et al., 2005). Thus, with the wash-out together with arterial hypervas-
aim of detecting HCC smaller than 3 cm, cularization, for the diagnosis of HCC.
the suggested interval for surveillance is 6 Moreover, it is suggested that contrast-en-
hanced ultrasonography (US) can be used
as a non-invasive additional diagnostic
technique to spiral Computed Tomography
(CT), Magnetic Resonance Imaging (MRI)
and angiography. However, standard arte-
riography and CT arteriography are not
recommended for routine diagnosis. In
cases of an atypical vascular pattern, a
liver biopsy is recommended but no men-
tion is made of the diagnostic accuracy
of histology for these nodules or the
applicability of biopsy, e.g., difficult loca-
Figure 20.1. European association for the study of tion, poor coagulation. Based on AASLD
the liver consensus algorithm for the imaging path- Guidelines with nodules > 2 cm, only one
way in cirrhotic patients to detect HCC (2001) dynamic technique showing a typical
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 263

Figure 20.2. American association for the study of liver diseases practice guidelines for the algorithm to
be followed for a suspicious nodule detected on US (2005)

vascular pattern or atypical vascular pat- to perform two imaging techniques, and
tern with alfa-fetoprotein > 200 ng/ml is to proceed to liver biopsy only in cases of
sufficient to make a diagnosis of HCC. discordant vascular pattern or atypical vas-
When the nodule is less than 2 cm in cular pattern between the two techniques.
diameter, diagnosis is more difficult and However, no mention is made of the prob-
may require needle liver biopsy. For nod- lems of biopsy, as detailed above, with the
ules smaller than 1 cm, the EASL consensus added consideration of operator error or
recommends serial US examinations every difficulty of being accurate in placing the
3 months until the nodule size exceeds biopsy needle in a lesion < 2 cm.
1 cm, and the same approach is suggested
by AASLD guidelines. When the nodule
is between 1 and 2 cm, EASL consensus SURVEILLANCE FOR
suggests proceeding to needle liver biopsy HCC: RADIOLOGICAL
to confirm the diagnosis, irrespective of the TECHNIQUES
vascular profile at imaging. The American
Association for the Study of Liver Diseases Radiological imaging of cirrhotic patients is
Guidelines have refined this, suggesting not easy and requires particular training and
264 F. Lodato et al.

expertise of the operator. Hepatocellular variable sensitivity from 35% to 84%


carcinoma can exhibit different echo- depending mostly on the expertise of
graphic features reflecting the variability of the operator and the equipment avail-
the macroscopic and microscopic findings. able (Peterson and Baron, 2001). When
Cirrhosis itself is a disease that produces the gold standard for diagnosis was the
pathological changes, such as regenerative explanted liver, the sensitivity of US was
and dysplastic nodules (DN), so that some- quite disappointing ranging from 33% to
times it is impossible to discriminate these 72% (Kim et al., 2001; Rode et al., 2001;
from a small HCC. During the evolution Gambarin-Gelwan et al., 2000; Teefey
from benign regenerative nodule, to low et al., 2003) in part because the livers
and high grade dysplastic nodule and finally were explanted months after diagnosis.
to HCC, the portal venous supply progres- Only one article (Teefey et al., 2003)
sively disappears so that new arterial ves- provides information on the size of the
sels constitute the main blood supply for false negative nodules, showing that all
the HCC. Therefore, differences in focal lesions which were missed were smaller
lesion perfusion can help to characterize than 2.5 cm. Ultrasonography specificity
the nodules in a cirrhotic liver (Lencioni ranged from 92% to 100%.
et al., 1996). Following the detection of At US examination, HCC may appear as
a suspicious nodule ultrasonographically, round or oval lesions, small tumors usually
both EASL statements and AASLD guide- have smooth and sharp margins. Nodular-
lines require confirmation using dynamic type HCC with extranodular growth and
techniques as contrast-enhanced US, spiral multinodular-type HCC may present with
CT or dynamic magnetic resonance (MR). irregular margins. Small HCC may be
However the single best technique fol- hypoechoic, hyperechoic or even isoechoic
lowing ultrasound is not well established with the surrounding liver. Approximately
(Fung et al., 2004). 75% of small HCC (< 2 cm) are hypoe-
choic. The hyperechoic appearance usu-
ally reflects fatty changes of the tumor and
ULTRASONOGRAPHY its differential diagnosis may be difficult
(US), DOPPLER- with respect to a hemangioma. As HCC
nodules grow they tend to develop hyp-
ULTRASONOGRAPHY, oechoic peripheral rims and become more
POWER DOPPLER- heterogeneous.
ULTRASONOGRAPHY AND The use of Doppler US or power Doppler
CONTRAST-ENHANCED may help in the diagnosis. Hepatocellular
ULTRASONOGRAPHY carcinoma usually has a florid arterial
vascularization, and this is seen as int-
Ultrasonography is a rapid and non inva- ralesional flow signal with an arterial
sive evaluation although it may be hin- spectrum. Large HCC may have a basket
dered by factors such as the patients body pattern with a fine blood network sur-
mass or intestinal interposition. rounding the nodule and flowing into it.
Hepatocellular carcinoma surveillance Doppler examination shows the presence
studies based on the use of US showed of pulsatile flow with arterial waveform
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 265

and high resistive and pulsality indexes. the gold standard to identify hypervascular
Large regenerative nodules and DN do not lesions, the sensitivity of contrast enhanced
have arterial intralesional vessels, so these US was 97% for nodules > 3 cm in diam-
findings support the diagnosis of HCC. eter, 92% for nodules between 2 and 3 cm,
The problem is that in small HCC the and 67% for nodules < 1 cm (Gaiani et al.,
sensitivity of Doppler techniques is low 2004). Another disadvantage is that only
and a typical pulsatile flow arterial aspect one lesion or lesions close together can be
can be shown in less than 50% of nodules examined at one time. Therefore, a cost-
(Lencioni et al., 1996). effective algorithm, of which technique to
The newest microbubble contrast agents use following routine US still needs to be
and the development of specific contrast determined (Fung et al., 2004).
techniques have opened a new era in liver
ultrasonography. The AASLD Guidelines
consider contrast enhanced US as a con- SPIRAL COMPUTED
firmatory technique when a suspicion of TOMOGRAPHY
HCC is made from standard US. Images
are based on non linear acoustic effects The introduction of spiral CT in the early
of bubbles enhancing grey scale, with 1990s has dramatically improved the accu-
high contrast and spatial resolution. This racy of CT in the diagnosis of HCC.
technique improves the accuracy of US Computed Tomography data acquisition
in the diagnosis of focal liver lesions speed is greatly increased so that it is pos-
(Lencioni et al., 2002; Wen et al., 2004). sible to scan the whole liver during a single
Contrast agents are safe and well tolerated. breath hold, allowing a satisfactory evalu-
The advent of second generation contrast ation of different contrast enhancement
agents together with low mechanical index phases. Moreover, 3-D reconstructions are
scanning have led the European Federation available with faster data acquisition. The
for Ultrasound in Medicine and Biology, to standard spiral CT evaluation for HCC
define the guidelines for its use (Albrecht should include unenhanced and contrast
et al., 2004). Contrast-enhanced US is rec- enhanced phase of the arterial phase (25–
ommended to characterize any suspected 30 s after the contrast injection), of the
lesion detected at baseline US in cirrho- portal venous phase (70–80 s after contrast
sis. At contrast enhanced-US examination, injection), and a late phase (180–210 s
HCC typically shows a strong enhancement after contrast injection). As HCC is a
in the arterial phase and a rapid washout vascular tumor that receives almost all
during the portal and late phases in which its blood supply from arterial vessels, it
the nodule remains iso or hypoechoic. is well seen during the arterial phase; the
Regenerative nodules and DN usually do rest of the liver receives the majority of
not show any uptake during the arterial blood supply from the portal vein, and will
phase and their appearance resembles the not enhance until the contrast reaches the
surrounding liver. However, as for non portal system. Fast scans during the dif-
contrast-enhanced US, the sensitivity of ferent phases of contrast flow through the
using ultrasound contrast agents decreases liver help in increasing the differentiation
with smaller lesions. Defining spiral CT as between the tumor and the liver.
266 F. Lodato et al.

The technique can be improved by arterial-portal shunting. However these


using multidetector-row scan technology images are often peripheral, wedge shaped
which due to the rapid acquisition time, and not round and sometimes disappear
allows visualization of two different arte- in follow up examination. These features
rial phases, early and late. The first arte- help to exclude HCC. The portal phase
rial phase is a CT arteriography that may acquisition is particularly important as
be of some help depending on the aim of HCCs have a rapid wash out during this
the examination. Some studies show that a phase, whereas hemangiomas or vascular
double arterial phase did not improve the malformation show contrast enhancement.
detection rate of HCC compared to a single The most problematic lesions mimicking
arterial phase (Laghi et al., 2003; Francis HCC are dysplastic nodules that may show
et al., 2003), whereas it did improve with arterial phase enhancement, a problem that
the addition of a third arterial phase as the cannot be resolved even when using the
middle phase is claimed to be the most most modern imaging technologies.
sensitive (Murakami et al., 2003). Another technique that could improve
However, despite the tremendous tech- CT performance is the injection of contrast
nical progress in this area, CT is still through the hepatic artery while perform-
insensitive for detecting small HCC. When ing CT. This method has shown 66% more
considering studies on explanted livers, tumors in patients with HCC compared to
the sensitivity of spiral CT ranged between triphasic helical contrast CT (Oliver et al.,
54% and 79% (Lim et al., 2000; Rode et al., 1997), but its use is limited by cost and
2001; Burrel et al., 2003; Valls et al., the mortality and morbidity related to the
2004) mostly due to the lack of detection angiographic procedure. However, it can
of smaller lesions (< 2 cm). be easily performed during chemoemboli-
Another important issue is the specificity zation or embolization therapy when the
of this technique. It is well recognized that catheter in the hepatic artery is needed to
a number of benign lesions can simulate perform the therapeutic procedure.
HCC on CT. The advent of arterial phase Lipiodol CT is another technique that
imaging brought about a series of new requires the administration of contrast
“HCC pseudolesions” that could be mis- media through the hepatic artery. It is lim-
taken for enhancing tumors. Specificity ited by the need to perform angiography
of spiral CT ranges between 93% and and requires the patient to be rescanned
97% in explant studies (Rode et al., 2001; at least 14 days after the injection. This
Lim et al., 2000; Burrel et al., 2003). allows the normal liver to wash-out the
Among the hypervascular lesions that can contrast while the HCC retains it for a
simulate HCC are pseudoaneurysms, arte- longer period. However retention of lipi-
riovenous malformations, hemangiomas odol is not specific for HCC alone, with
and dysplastic nodules; while hypovascu- only one study reporting that lipiodol-
lar lesions simulating HCC are hemangi- CT is more specific than spiral CT in
omas, confluent fibrosis and regenerative detecting small, less differentiated nodules
nodules. Moreover, cirrhotics may have (Nakayama et al., 2001). Moreover, it can
transient focal enhancement of the liver hinder assessment of residual vascularity
during the arterial phase, mostly due to following loco-regional therapies.
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 267

MAGNETIC RESONANCE lar components, such as fat, glycogen, or


IMAGING ions which may cause different appearance
in T1-weighted and T2-weighted images.
The use of MRI in the diagnosis of HCC The signal may appear hypo, iso or hyper-
has expanded greatly over the last few years intense in T1-weighted images, and iso or
due to the development of new hardware hyperintense in T2-weighted images.
and software that have allowed the rapid Gadolinium chelates are distributed in
acquisition of very detailed images, reduc- extracellular space and their enhancing
ing the artefact secondary to respiratory effect depends on the blood supply dur-
activity. Volumetric sequences allow the ing the early dynamic phase and on the
reconstruction of 3-D images with very high interstitial matrix during the delayed phase.
spatial and temporal resolution. Moreover, The new fast MRI techniques allow the
the introduction of liver-specific contrast acquisition of multiple intravenous contrast
agents into clinical practice, has enabled a enhanced dynamic images of the liver,
perfusional investigation of liver nodules. including the arterial phase, the portal peak
The standard examination for the detection phase and an delayed phase. As already
of HCC should include: mentioned, HCC is usually a hypervascular
tumor, so that most HCC are characterized
 T1-weighted fat spoiled gradient-echo
by an enhancement in the arterial phase,
sequences with fat suppression
and rapid washout during the portal phase
 Respiration triggered or breath hold T2
(Figures 20.3a–c). Regenerative nodules
weighted fat spin echo sequences with
and DNs are usually not hypervascular.
fat suppression
As described for spiral CT, some benign
 Serial dynamic T1-weighted fast spoiled
lesions, especially high grade DN, may
gradient-echo sequences after injection
have increased arterial blood supply that
of a gadolinium chelate (Lencioni et al.,
may render diagnosis difficult, and in cir-
2004)
rhotic livers there are often arterioportal
Once again, as HCC is a very heterogeneous shunts that may produce false positive
tumor, these characteristics are reflected in interpretations (Ito et al., 2004).
an MRI examination. For example, HCC Dynamic MRI can also be performed
may have variable stromal and intracellu- by using other tissue specific agents, for

a b c

Figure 20.3. MRI of 76 year old man with HBV cirrhosis and 2 HCC. (a) T2 weighted image showing two
high signal lesions. (b) Arterial phase T1 image revealing arterialisation of these lesions. (c) Portal phase T1
image showing washout of the lesions. The signal characteristics of these two lesions are typical of HCCs
268 F. Lodato et al.

example, superparamagnetic iron oxide the nature of these small nodules and,
(SPIO) particles are taken up by Kuppfer most importantly, the relationship between
cells of the reticuloendothelial system these nodules and the development of
(RES-targeted contrast agents) which are HCC is uncertain. It is well accepted that
absent in HCC. They produce a decrease cirrhosis itself represents a preneoplastic
in the intensity of the liver tissue espe- condition, but in carcinogenesis the exact
cially on T2-weighted images as HCC point in time between a lesion that still
nodules show little or no uptake of the has some growth regulation and one that
contrast. Gadolinium and SPIO agents has lost growth regulation is difficult to
could be coupled (Ward and Robinson, define. The only morphological change
2002), but the usefulness of this technique that is considered as intermediate to HCC
is debated (Halavaara et al., 2002). is dysplasia (International Working Party,
Despite substantial progress in MRI still 1995), although foci of dysplasia may
fails in detecting small HCC; the sensitiv- take place within benign regenerative nod-
ity of dynamic MRI ranges from 33% to ules. Moreover, it is not clear whether all
77% in explant studies (Rode et al., 2001; dysplastic nodules will inevitably evolve
Burrel et al., 2003; Krinsky et al., 2002; to HCC. Some reports suggest that high
Teefey et al., 2003) and 78% when using grade dysplastic nodules (HGDN) should
dynamic imaging plus RES-targeted agents be considered as HCC precursors in most
(Bhartia et al., 2003). When considering cases (Borzio et al., 2003). In clinical
only HCC smaller than 1 cm, sensitivity practice a histological diagnosis of HGDN
drops to 4–71%. Nevertheless, a study poses several difficulties in the interpre-
showed that dynamic MRI is superior to tation of this finding, as a conservative
helical CT in the detection of nodules therapeutic approach could affect the out-
between 1 and 2 cm (Burrel et al., 2003). come. More difficulties arise considering
In this study, 50 patient with HCC under- that in the context of a dysplastic nodule,
going liver transplantation were analyzed. foci of HCC can exist but biopsy may
Twenty-nine of them had HCC; the patho- miss the sampling of the malignant por-
logic examination was the gold standard. tion of the lesion. In the setting of cir-
On a per nodule basis, sensitivity of MRI rhosis the picture is also complicated by
was superior to triphasic helical CT (76% vascular lesions that are not malignant
vs. 61%, p = 0.001). Sensitivity for detec- e.g., arteroportal shunts or other perfusion
tion of additional nodules decreased with abnormalities.
size and was superior to CT for nodules 10 The introduction of radiological tech-
to 20 mm (84% vs. 47%, p = 0.016). niques that study the perfusional pattern
of nodules has enormously improved the
accuracy of diagnosis, and the vascular
THE PROBLEM OF SMALL study of lesion has become the main
NODULES parameter used for the radiological diag-
nosis of HCC. Consequently the use of
The improvements in imaging techniques liver biopsy has reduced during the past
have allowed the identification of smaller few years reflecting the EASL recom-
nodules within the cirrhotic liver, but often mendations. This is clearly an advantage
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 269

considering that liver biopsy can also be performing biopsy in all lesions > 1 cm and
related to a 0–5% of seeding (Stigliano < 2 cm without a typical vascular pattern.
and Burroughs, 2005) even if associ- However, these recommendations for
ated with percutaneous ablative tech- biopsy open up a problem of the clinical
niques (Stigliano and Burroughs, 2005; applicability to perform biopsy for such
Ebara et al., 2005; Shiina et al., 2005). small nodules, as three factors affect this:
Moreover, when a biopsy is negative for the reliability of obtaining tissue from the
HCC its presence cannot be excluded, right place, the uncertainty of the diagno-
as there is a 10% risk of false negativity sis histologically and the risk of seeding.
especially in the presence of small nodules The consequences of seeding can be dev-
(< 2 cm) (Caturelli et al., 2004). astating. Surgery may no longer be fea-
Bolondi et al. (2005) showed that 78.5% sible because of the spread of neoplastic
of nodules between 1 and 3 cm diameter disease, which has been reported or seed-
with only one positive imaging technique ing may occur after potentially definitive
in terms of hypervascularity suggestive for therapy including liver transplantation. Its
HCC were found to be true HCC at biopsy. occurrence is reported in 0–5.1% of cases
When looking at the rate of hypervascular- following liver biopsy but may also occur
ity demonstrated by two separate radio- following percutaneous treatment for
logical techniques leading to the diagnosis HCC. Percutaneous ethanol injection (PEI)
of HCC, this rate was significantly lower has a risk of 0.13–6.8%. Radiofrequency
in the group of nodules between 1 and Ablation (RFA) has a reported risk of
2 cm than in the group of nodules between 0–3%, that increases to 12.5% in the
2 and 3 cm (44% vs. 84%, p = 0.001). case of superficial nodules (Stigliano and
Amongst the group of nodules between Burroughs, 2005). Prospective studies of
1 and 2 cm with discordant detection of loco-regional therapy that might be con-
hypervascularity by two techniques, a defi- sidered for reducing the risk of seeding
nite diagnosis of HCC by histology was show a seeding rate of 1.96% with PEI
made in 48%. The American Association (Ebara et al., 2005; Shiina et al., 2005) and
for the Study of Liver Diseases Guidelines 2.55% with RFA (Shiina et al., 2005).
recommend performing liver biopsy in In the setting of liver transplantation, the
cases of discordant imaging technique for need of a precise diagnosis can be dictated
nodules > 1 and < 2 cm. Based on the data by the allocation system adopted. In the
by Bolondi et al. (2005) this would be USA and in some European liver trans-
the case for almost half of these nodules plantation centres, the Model for End-
as 44% of small nodules had discordant Stage Liver Disease-HCC scoring system
imaging hypervascularity. The problem is used, in which a single nodule ≥ 2 cm
of hypovascular nodules (20% of cases or multiple nodules increases the priority
in this study) is another important one; on the waiting list (Sharma et al., 2004;
all cases occurred in the group of nodules Hayashi et al., 2004). Here the issue is
between 1 and 2 cm, and 36% were later the need of a precise diagnosis to justify
proven to be HCC. These data support the increased priority, regardless of baseline
approach suggested by the most recent liver function. It is interesting to note that
AASLD guidelines, which recommend explant studies in the USA have shown the
270 F. Lodato et al.

lack of histological confirmation in one these techniques is poor when nodules


third of patients diagnosed with HCC pre- are smaller than 2 cm, which may also
transplant (Wiesner et al., 2004), which be hypovascular.
appears higher than European studies. In the era of liver transplantation, the
If the diagnosis will not change the need for a correct diagnosis has become
priority for liver transplantation, nodules imperative in most cases particularly for
between 1 and 2 cm without confirmatory prioritisation of patients for transplanta-
hypervascularity by two separate tech- tion. Although liver biopsy may not be
niques can be treated by locoregional avoided there is a risk of seeding and this
treatment even without “a priori” histolog- must be taken into account during the
ical diagnosis, associated with performing diagnostic pathway. Diagnostic algorithms
biopsy at the time of the procedure. It is need to be worked out considering that
not clear if the seeding risk is increased by single nodules < 2 cm have in general a far
performing both biopsy and locoregional better prognosis and virtually no risk of
therapy at the same time but this could be recurrence after liver transplantation.
possible. For single nodules smaller than In cirrhotic patients nodules suspected
3 cm among various techniques PEI may of being malignant that are smaller than
be preferable, with fewer major compli- 1 cm should be followed up at short term
cations, and possibly less risk of seeding intervals of no more than 3 months, as
(Ebara et al., 2005; Shiina et al., 2005); the use of dynamic techniques is only
PEI is also cheaper than radiofrequency sufficiently accurate when the nodules
ablation (RFA) and the size of the nee- are larger. With nodules between 1 and
dle is smaller. However, beyond 1 year a 2 cm, if two dynamic techniques are not
recent randomized trial has shown higher concordant in demonstrating hypervascu-
survival rates with RFA when compared to larity, liver biopsy should be performed
PEI (Shiina et al., 2005). when a precise diagnosis would change
Each centre should follow an established the immediate therapeutic option for the
protocol and audit their decision making patient, but could be delayed or avoided
as there is insufficient evidence to select depending on the timing of potential ther-
one approach over another. apy such as liver transplantation (e.g., list
and reimage). However, another therapeu-
tic option is to treat the nodule as if it is
CONCLUSIONS HCC, even in the absence of a histological
diagnosis, as the risk of seeding may not
Current imaging techniques have sub- be increased by adding ablative therapy
stantially improved the diagnostic to the biopsy (Stigliano and Burroughs,
accuracy for HCC in cirrhotic patients, 2005) at least for PEI. For nodules larger
reducing the need of liver biopsy and its than 2 cm, a single dynamic technique is
attendant immediate complications, as sufficient if the hypervascular pattern is
well as the risk of seeding. This is mostly typical, to diagnose HCC. In the pres-
due to the development of dynamic tech- ence of an atypical vascular pattern the
niques that study the vascular pattern of presence of alfa-fetoprotein > 200 ng/ml is
nodules. However, the performance of diagnostic.
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 271

REFERENCES C., and De Sio, I. 2004. Ultrasound guided fine


needle biopsy of early hepatocellular carcinoma
Albrecht, T., Blomley, M., Bolondi, L., Claudon, M., complicating liver cirrhosis: a multicentre study.
Correas, J.M., Cosgrove, D., Greiner, L., Jager K., Gut 53: 1356–1362.
Jong, N.D., Leen, E., Lencioni, R., Lindsell, D., Deuffic, S., Poynard, T., Buffat, L., and Valleron,
Martegani, A., Solbiati, L., Thorelius, L., A.J. 1998. Trends in primary liver cancer. Lancet
Tranquart, F., Weskott, H.P., Whittingham, T., 17;351: 214–215.
and EFSUMB Study Group. 2004. Guidelines Ebara, M., Okabe, S., Kita, K., Sugiura, N., Fukuda,
for the use of contrast agents in ultrasounds. H., Yoshikawa, M., Kondo, F., and Saisho, H.
Ultrashall. Med. 25: 249–256. 2005. Percutaneous ethanol injection for small
Adam, R., and Del Gaudio, M. 2003. Evolution hepatocellular carcinoma: therapeutic efficacy
of liver transplantation for hepatocellular carci- based on 20-year observation. J. Hepatol. 43:
noma. J. Hepatol. 39: 888–895. 377–380.
Bhartia, B., Ward, J., Guthrie, J.A., and Robinson, El Serag, H.B., and Mason, A.C. 1999. Rising inci-
P.J. 2003. Hepatocellular carcinoma in cirrhotic dence of hepatocellular carcinoma in the United
livers: double-contrast thin section – MR imag- States. N. Engl. J. Med. 340: 745–750.
ing with pathologic correlation of explanted tis- Fattovich, G., Giustina, G., Degos, F., Tremolada, F.,
sue. AJR Am. J. Roentgenol. 180: 577–584. Diodati, G., Almasio, P., Nevens, F., Solinas, A.,
Bolondi, L., Gaiani, S., Celli, N., Golfieri, R., Mura, D., Brouwer, J.T., Thomas, H., Njapoum, C.,
Grigioni, W.F., Leoni, S., Venturi, A.M., and Casarin, C., Bonetti, P., Fuschi, P., Basho, J.,
Piscaglia, F. 2005. Characterization of small Tocco, A., Bhalla, A., Galassini, R., Noventa, F.,
nodules in cirrhosis by assessment of vascular- Schalm, S.W., and Realdi, G. 1997. Morbidity
ity: the problem of hypovascular hepatocellular and mortality in compensated cirrhosis type C:
carcinoma. Hepatology 42: 27–34. a retrospective follow-up study of 384 patients.
Borzio, M., Fargion, S., Borzio, F., Fracanzani, Gastroenterology 112: 463–472.
A.L., Croce, A.M., Stroffolini, T., Oldani, S., Francis, I.R., Cohan, R.H., McNulty, N.J., Platt, J.F.,
Cotichini, R., and Roncalli, M. 2003. Impact Korobkin, M., Gebremariam, A., and Ragupathi,
of large regenerative, low grade and high grade K.I. 2003. Multidetector CT of the liver and
dysplastic nodules in hepatocellular carcinoma hepatic neoplasm: effect of multiphasic imaging
development. J. Hepatol. 39: 209–214. on tumor conspicuity and vascular enhancement.
Bruix, J., Sherman, M., Llovet, J.M., Beaugrand, M., AJR Am. J. Roentgenol. 180: 1217–1224.
Lencioni, R., Burroughs, A.K., Christensen, E., Fung, K.T., Li, F.T., Raimondo, M.L., Maudgil, D.,
Pagliaro, L., Colombo, M., Rodes J, and EASL Mancuso, A., Tibballs, J.M., Watkinson, A.A.,
Panel of Experts on HCC. 2001. Clinical manage- Patch, D., and Burroughs, A.K. 2004. Systematic
ment of hepatocellular carcinoma. Conclusion review of radiological imaging for hepatocellu-
of the Barcelona – 2000 EASL Conference. J. lar carcinoma in cirrhotic patients. Br. J. Radiol.
Hepatol. 35: 421–430. 77: 633–640.
Bruix, J., and Sherman, M. 2005. Management Gaiani, S., Celli, N., Piscaglia, F., Cecilioni, L.,
of hepatocellular carcinoma. Hepatology 42: Losinno, F., Giangregorio, F., Mancini, M., Pini,
1208–1236. P., Fornari, F., and Bolondi, L. 2004. Usefulness
Burrel, M., Llovet, J.M., Ayuso, C., Iglesias, C., of contrast enhanced computed sonography in
Sala, M., Miquel, R., Caralt, T., Ayuso, J.R., the assessment of hepatocellular carcinoma
Sole, M., Sanchez, M., Bru, C., Bruix, J., and hypervascular at spiral computed tomography. J.
Barcelona Clinic Liver Cancer Group. 2003. Hepatol. 41: 421–426.
MRI angiography is superior to helical CT for Gambarin-Gelwan, M., Wolf, D.C., Shapiro, R.,
detection of HCC prior to liver transplanta- Schwartz, M.E., and Min, A.D. 2000. Sensitivity
tion: an explant correlation. Hepatology 38: of commonly available screening tests in detect-
1034–1042. ing hepatocellular carcinoma in cirrhotic
Caturelli, E., Solmi, L., Anti, M., Fusilli, S., Roselli, patients undergoing liver transplantation. Am.
P., Andriulli, A., Fornari, F., Del Vecchio Blanco, J. Gastroenterol. 95: 1535–1538.
272 F. Lodato et al.

Halavaara, J., Tervahartiala, P., Isoniemi, H., and Levy, I., Greig, P.D., Gallinger, S., Langer, B., and
Hockerstedt, K. 2002. Efficacy of sequential use Sherman, M. 2001. Resection of hepatocellular
of superparamgnetic iron oxide and gadolinium carcinoma without preoperative tumor biopsy.
in liver MR imaging. Acta. Radiol. 43: 180–185. Ann. Surg. 234: 206–209.
Hayashi, P. H., Trotter, J.F., Forman, L., Kugelmas, Lim, J.H., Kim, C.K., Lee, W.J., Park, C.K., Koh,
M., Steinberg, T., Russ, P., Wachs, M., Bak, T., K.C., Paik, S.W., and Joh, J.W. 2000. Detection
Kam, I., and Everson, G.T. 2004. Impact of pre- of hepatocellular carcinomas and dysplastic nod-
transplant diagnosis of hepatocellular carcinoma ules in cirrhotic livers: accuracy of helical CT in
on cadveric liver allocation in the era of MELD. transplant patients. AJR Am. J. Roentgenol. 175:
Liver Transplant. 10: 42–48. 693–698.
International Working Party. 1995. Terminology of Llovet, J.M., Burroughs, A, and Bruix, J. 2003.
nodular hepatocellular lesions. Hepatology 22: Hepatocellular carcinoma. Lancet 363: 1907–1917.
983–989. Murakami, T., Kim, T., Kawata, S., Kanematsu, M.,
Ito, K., Fujita, T., Shimizu, A., Koike, S., Sasaki, K., Federle, M.P., Hori, M., Okada, A., Kumano, S.,
Matsunaga, N., Hibino S., and Yuhara, M. 2004. Sugihara, E., Tomoda, K., and Nakamura, H.
Multiarterial phase dynamic MRI of small early 2003. Evaluation of optimal timing of arterial
enhancing hepatic lesions in cirrhosis or chronic phase imaging for the detection of hypervas-
hepatitis: differentiating between hypervascular cular hepatocellular carcinoma by using triple
hepatocellular carcinomas and pseudolesions. arterial phase imaging with multidetector-row
AJR Am. J. Roentgenol. 183: 699–705. helical computed tomography. Invest. Radiol.
Kim, C.K., Lim, J.H., and Lee, W.J. 2001. Detection 38: 497–503.
of hepatocellular carcinoma and dysplastic nod- Nakayama, A., Imamura, H., Matsuyama, Y., Kitamura,
ules in cirrhotic liver: accuracy of ultrasonogra- H., Miwa, S., Kobayashi, A., Miyagawa, S., and
phy in transplant patients. J. Ultrasound. Med. Kawasaki, S. 2001. Value of lipiodol computed
20: 99–104. tomography and digital subtraction angiography
Krinsky, G.A., Lee, V.S., Theise, N.D., Weinreb, in the era of helical biphasic computed tomogra-
J.C., Morgan, G.R., Diflo, T., John, D., phy as preoperative assessment of hepatocellular
Teperman, L.W., and Goldenberg, A.S. 2002. carcinoma. Ann. Surg. 234: 56–62.
Transplantation for hepatocellular carcinoma Nino-Murcia, M., Olcott, E.W., Jeffrey, R.B.,
and cirrhosis: sensitivity of magnetic resonance Lamm, R.L., Beaulieu, C.F., and Jain, K.A.
imaging. Liver Transplant. 8: 1156–1164. 2000. Focal liver lesions: pattern-based classifi-
Laghi, A., Iannaccone, R., Rossi, P., Carbone, I., cation scheme for enhancement at arterial phase
Ferrari, R., Mangiapane, F., Nofroni, I., and CT. Radiology 215: 746–751.
Passariello, R. 2003. Hepatocellular carcinoma: Oliver, J.H., Baron, L.L., and Carr, B.E. 1997.
detection with triple-phase multidetector row CT imaging of hepatocellular carcinoma:
helical CT in patients with chronic hepatitis. CT-arteriography versus triphasic helical con-
Radiology 226: 543–549. trast CT. Radiology 205: 144 A.
Lencioni, R., Cioni, D., Crocetti, L., Donati, F., Parkin, D.M., Bray, F., Ferlay, J., and Pisani, P. 2001.
Franchini, C., Giusti, S., and Bartolozzi, C. Estimating the world cancer burden: Globocan
2002. Ultrasound imaging of focal liver lesion 2000. Int. J. Cancer 15;94: 153–156.
with a second generation contrast agent. Acad. Peterson, M.S., and Baron, R.L. 2001. Radiologic
Radiol. 9: S 371–374. diagnosis of hepatocellular carcinoma. Clin.
Lencioni, R., Cioni, D., Crocetti, L., Della Pina, C., and Liver Dis. 5: 123–144.
Bartolozzi, C. 2004. Magnetic resonance imag- Rode, A., Bancel, B., Douek, P., Chevallier, M.,
ing of liver tumors. J. Hepatol. 40: 162–171. Vilgrain, V., Picaud, G., Henry, L., Berger, F.,
Lencioni, R., Pinto, F., Armillotta, N., and Bizollon, T., Gaudin, J.L., and Ducerf, C. 2001.
Bartolozzi, C. 1996. Assessment of tumor vas- Small nodule detection in cirrhotic livers: evalu-
cularity in hepatocellular carcinoma: compari- ation with US, spiral CT, and MRI and correla-
son of power-doppler US and color-doppler US. tion with pathologic examination of explanted
Radiology 201: 353–358. liver. J. Comput. Assist. Tomogr. 25: 327–336.
20. Hepatocellular Cancer in Cirrhotic Patients: Radiological Imaging 273

Sangiovanni, A., Del Ninno, E., Fasani, P., De candidates: Prospective comparison of CT, MR
Fazio, C., Ronchi, G., Romeo, R., Morabito, imaging, US and PET. Radiology 226: 533–542.
A., De Franchis, R., and Colombo, M. 2004. Torzilli, G., Minagawa, M., Takayama, T., Inoue, K.,
Increased survival of cirrhotic patients with a Hui, A.M., Kubota, K., Ohtomo, K., and
hepatocellular carcinoma detected during sur- Makuuchi, M. 1999. Accurate preoperative eval-
veillance. Gastroenterology 126: 1005–1014. uation of liver mass lesions without fine-needle
Sharma, P., Balan, V., Hernandez, J.L., Harper, biopsy. Hepatology 30: 889–893.
A.M., Edwards, E.B., Rodriguez-Luna, H., Valls, C., Cos, M., Figueras, J., Andia, E., Ramos, E.,
Byrne, T., Vargas, H.E., Mulligan, D., Rakela, J., Sanchez, A., Serrano, T., and Torras, J. 2004.
and Wiesner, R.H. 2004. Liver transplantation Pretransplantation diagnosis and staging of
for hepatocellular carcinoma: the MELD impact. hepatocellular carcinoma in patients with cir-
Liver Transpl. 10: 36–41. rhosis: value of dual phase helical CT. AJR Am.
Shiina, S., Teratani, T., Obi, S., Sato, S., Tateishi, R., J. Roentgenol. 182: 1011–1017.
Fujishima, T., Ishikawa, T., Koike, Y., Yoshida, H., Ward, J., and Robinson, P.J. 2002. How to detect
Kawabe, T., and Omata, M. 2005. A randomized hepatocellular carcinoma in cirrhosis. Eur
controlled trial of radiofrequency ablation with Radiol. 12: 2258–2272.
ethanol injection for small hepatocellular carci- Wen, Y.L., Kudo, M., Zheng, R.Q., Ding, H.,
noma. Gastroenterology 129: 122–130. Zhou, P., Minami, Y., Chung, H., Kitano,
Stigliano, R., and Burroughs, A.K. 2005. Should M., Kawasaki, T., and Maekawa, K. 2004.
we biopsy each liver mass suspicious for HCC Charaterzation of hepatic tumors: value of
before liver transplantation? – No, please don’t. contrast enhanced coded phase-inversion har-
J. Hepatol. 43: 563–568. monic angio. AJR Am. J. Roentgenol. 182:
Teefey, S.A., Hildeboldt, C.C., Dehdashti, F., 1019–1026.
Siegel, B.A., Peters, M.G., Heiken, J.P., Wiesner, R.H., Freeman, R.B., and Mulligan, D.C.
Brown, J.J., McFarland, E.G., Middleton, W.D., 2004. Liver transplantation for hepatocellular
Balfe, D.M., and Ritter, J.H. 2003. Detection of cancer: the impact of the MELD allocation
primary hepatic malignancyn in liver transplant policy. Gastroenterology 127: S261–267.
B. Treatment
21
Treatment of Hepatocellular Carcinoma
with Thalidomide: Assessment with Power
Doppler Ultrasound
Chiun Hsu, Chiung-Nien Chen, and Ann-Lii Cheng

SUMMARY INTRODUCTION
Hepatocellular carcinoma (HCC) is typically a Hepatocellular carcinoma (HCC) is the
hypervascular tumor and anti-angiogenesis fifth most common cancer in the world,
therapy may be effective for the treatment with an estimated incidence of 1 million
of HCC. Thalidomide has been shown to new cases per year (Parkin, 2001). Most
inhibit angiogenesis induced by various of the patients with HCC are diagnosed at
proangiogenic factors and may produce advanced stage and not eligible for cura-
objective tumor response in various can- tive therapy. However, there is no standard
cers. In HCC, thalidomide may produce systemic therapy that has proven efficacy
complete or partial remission in ~ 4–7% in patients with advanced HCC. HCC
of patients and disease stabilization in patients usually tolerate conventional cyto-
10–20% patients with advanced disease. toxic chemotherapy poorly because of the
We have performed a prospective study underlying cirrhosis and the accompanying
using power Doppler ultrasound to evalu- hypersplenism and peripheral cytopenia
ate the vascular response to thalidomide (Hsu et al., 2004). Therefore, novel thera-
treatment in patients with advanced HCC. peutic agents must be developed to treat
Patients who achieved objective tumor this difficult disease.
response to thalidomide tended to have Molecular targeted therapy, which aims
a higher pretreatment vascularity index, at specific molecular derangements in
defined as the number of colored (vas- cancer cells or their microenvironment,
cular) pixels within a well-demarcated represents breakthrough over conven-
tumor area divided by the number of total tional cytotoxic therapy (Gschwind et
pixels in that area, than patients who did al., 2004). A rapidly expanding list of
not. The pretreatment vascularity index cancers, including non-small cell lung
also correlated with levels of circulating cancer, breast cancer, gastrointestinal stro-
angiogenic factors. The value of power mal tumor, colorectal cancer, and chronic
Doppler ultrasound in future clinical trials myelogeneous leukemia, are now treatable
of anti-angiogenesis therapy of HCC is by agents targeting at specific signaling
discussed. pathways (Krause and Van Etten, 2005).

277
278 C. Hsu et al.

In addition to targeting the cancer cells per decrease the transcription of proangiogenic
se, anti-angiogenic/antivascular therapy factors such as, VEGF, and insulin-like
is another promising approach to treat growth factor (Stephens et al., 2000; Vacca
cancers that are usually refractory to con- et al., 2005). Its potential therapeutic
ventional cytotoxic therapy. Development efficacy for neoplastic disorders has been
of novel molecular targeting agents has reported in patients with refractory multiple
thus become a key endeavor for the global myeloma and human immunodeficiency
pharmacological industry and numerous virus-associated Kaposi’s sarcoma (Singhal
agents are already in the development et al., 1999; Little et al., 2000).
pipeline. Several single-arm clinical trials to test
HCC is typically a hypervascular tumor. the antitumor efficacy of thalidomide for
HCC cells and their surrounding stroma advanced HCC have been published, and
cells have been found to express various their results are summarized in Table 21.1
proangiogenic factors, and these proangio- (Hsu et al., 2003; Wang et al., 2004a, b;
genic factors may be closely associated Lin et al., 2005; Patt et al., 2005; Chen
with high histological grade, portal venous et al., 2005; Schwartz et al., 2005). The
thrombosis, and tumor capsular invasion results are similar among different studies,
(Torimura et al., 1998; Chow et al., 1997, with objective response rate of ~ 4–7% and
1998; Harada et al., 1998). It appears that overall survival of 4–7 months. Dose esca-
a proangiogenic milieu exists to facilitate lation of thalidomide was planned in most
tumor proliferation and invasion in HCC. studies, but the patients usually cannot
It is thus reasonable to hypothesize that tolerate a daily dose of > 300 mg because
anti-angiogenesis therapy may be able to of treatment-related toxicity, including
control this tumor. fatigue, constipation, and neurotoxicity.
The anti-angiogenic activity of thalidomide The short survival and poor tolerance
was first demonstrated in the 1990s. It mostly reflect the advanced disease status
can inhibit in vivo angiogenesis induced and compromised liver function reserves
by vascular endothelial growth factor in this group of patients.
(VEGF) and basic fibroblast growth factor Despite its unsatisfactory overall efficacy
(D’Amato et al., 1994; Kenyon et al., in the treatment of advanced HCC, thali-
1997). Thalidomide has also been shown to domide does produce tumor stabilization

Table 21.1. Clinical trials of thalidomide for patients with advanced HCC.
No. of Daily dose of Objective Disease Overall survival
Author patients thalidomide response rate stabilization rate (months)
Hsu et al. 68 200 mg 6.3% (1 CR, 3 PR) 15.9% 4.3
Wang et al. 99 150–300 mg 7.0% (6 PR) NA NA
Lin et al. 26 200 mg 3.9% (1 PR) 11.5% 4.0
Patt et al. 37 400 mg 5% (1 PR) NA 6.8
Chen et al. 42 200 mg 7% (3 PR) 23.8% 4.0
Schwartz et al. 38 200 mg 5% (1 CR, 1 PR) 18% 5.5
Disease control rate: complete response + partial response + stable disease > 2 months
CR, complete response; PR, partial response
21. Treatment of Hepatocellular Carcinoma with Thalidomide 279

in ~ 10–25% of HCC patients. In our to measure the biological efficacy of antian-


study, the ten patients who had either an giogenesis therapy has also been suggested
objective tumor response (four patients) in preclinical studies (Gee et al., 2001;
or a > 50% decrease in α-fetoprotein Goertz et al., 2002).
levels (six patients) had a median time Power (amplitude mode) Doppler ultra-
to disease progression of 23 weeks (95% sound, in which the color map displays the
CI 19.5–26.5) and a median overall sur- integrated power of the Doppler signals,
vival of 62.4 weeks (95% CI 31.2–93.6) has been shown to be superior to conven-
(Hsu et al., 2003). Thalidomide and other tional color Doppler ultrasound, which
biological agents are most likely to exert estimates the mean Doppler frequency
cytostatic rather than cytotoxic effect shift (Choi et al., 1996; Lencioni et al.,
on established tumors. Therefore, tumor 1996). Power Doppler ultrasound has the
shrinkage may not be a good surrogate advantages of low noise variance, rela-
marker of efficacy for these agents. In tive angular independence, and increased
addition, the optimal biological dose of dynamic signal range, which make it more
these agents may be significantly lower sensitive and specific than the conven-
than the maximum tolerated dose defined tional Doppler ultrasound (Rubin et al.,
by conventional phase I trials. Therefore, 1994). Therefore, it may provide a conve-
new endpoints are needed to provide nient, real-time method for observing the
proof-of-principle evidence for the biolo- biological effects of novel agents targeted
gical efficacy of molecular targeted therapy at tumor vasculature.
(Korn et al., 2001).

EVALUATION OF VASCULAR
EVALUATION OF TUMOR RESPONSE OF HCC TO
VASCULARITY USING POWER THALIDOMIDE BY POWER
DOPPLER ULTRASOUND DOPPLER ULTRASOUND:
A PROSPECTIVE STUDY
Many methods have been developed to
evaluate tumor vascularity (Choyke et al., We have performed a prospective study of
2002). Among these methods, Doppler thalidomide for the treatment of advanced
ultrasound has the advantage of non- HCC and power Doppler ultrasound was
invasiveness and ease to use for serial used to evaluate the vascular response to
follow-up. Tumor vascularity measured thalidomide treatment (Hsu et al., 2005).
by Doppler ultrasound has been shown to Patients with advanced HCC that were not
provide important prognostic information suitable for curative surgery or other local
for patients with colon, gastric, ovarian, treatment were enrolled to receive thali-
and cervical cancers and has shown good domide treatment. The starting dose was
correlation with vessel density determined 200 mg per day and the dose was escalated
by immunohistochemical staining (Wu by 100-mg increments every 2–3 weeks
et al., 1994; Cheng et al., 1999, 2000, 2002). if no grade 2 or greater treatment-related
The potential of using Doppler ultrasound toxicity developed. Clinical response to
280 C. Hsu et al.

thalidomide treatment was evaluated based stored for later analysis. Each tumor was
on computed tomography (CT) scan find- scanned three times. The stored images
ings and the clinical response. The patients were then retrieved and the examiner
were considered to have response to thali- contoured the tumor margin by using
domide treatment if complete or partial a cursor. Quantification of the vascular
response by WHO criteria (Miller et al., color signals within the demarcated area
1981) or a clinical benefit response was doc- was performed by software (Encomate;
umented. The clinical benefit response was Electronic Business Machine Co., Ltd.,
achieved when all the following three criteria Taipei, Taiwan). The vascularity index (VI)
were met: (1) stable disease on CT imaging, was defined as the number of colored
(2) a more than 50% decrease of alpha- (vascular) pixels within a well-demarcated
fetoprotein level for more than 4 weeks, and tumor area divided by the number of total
(3) improved performance status. pixels in that area. For each tumor the
Sonographic examination was performed mean VI of the three representative tumor
every 1–2 weeks during thalidomide sections, one from each scan, was used for
treatment. The HDI5000 power Doppler statistical analysis.
ultrasound unit (Advanced Technology From April 2000 to March 2003, 144
Laboratories, Bothell, WA) was used. A patients with advanced HCC were screened
2–5 MHz curved array was used for evalu- for enrollment into this study. Tumor vas-
ation of liver tumors and a 5–10 MHz cularity was evaluable by power Doppler
broad-band linear array transducer for ultrasound in only 56 of these patients.
superficial tumors, such as lymph nodes Reasons for nonevaluability included (1)
metastasis. For examination of the liver huge tumor size that could not be encom-
tumors, routine abdominal ultrasound was passed within the field of the sonographic
first performed to identify the index lesion, examination; (2) indiscrete tumor margin
followed by power Doppler evaluation. resulting from diffuse tumor infiltration,
For examination of the superficial tumors, severe cirrhosis, or prior local therapy;
power Doppler ultrasound was directly (3) location of the index tumor that could
used. The power Doppler ultrasound set- not be approached by sonographic exami-
ting was standardized using a medium nation, such as lung or intra-abdominal
wall filter, a color gain of 79%, a pulse lymph nodes. Of the 56 patients with
repetition frequency of 1,000 Hz with evaluable tumor vascularity, 47 (38 men,
moderate-to-long persistence, and a slow 9 women, median age 62.2 years) were
sweep technique to achieve the highest enrolled into this study. The index lesion
sensitivity without apparent background was a liver tumor for 44 patients, a meta-
noise. Focusing depth was set between 1 static neck lymph node for 2 patients,
and 12 cm. Only one representative lesion and a metastatic skin tumor for 1 patient.
was chosen for follow-up power Doppler Patients who completed at least 1 month
examination. of thalidomide treatment were considered
The tumor was scanned carefully in evaluable for response. Forty-four of these
all directions, and the tumor section with patients were evaluable for response to
maximal color signals was captured and thalidomide treatment and all 47 patients
21. Treatment of Hepatocellular Carcinoma with Thalidomide 281

were evaluable for toxicity. Three of the by liver biopsy. He had received chemo-
47 enrolled patients did not complete 1 therapy with doxorubicin before the start
month of treatment because of subjective of thalidomide treatment but progressive
intolerance of side effects, mainly fatigue. disease after chemotherapy was docu-
During thalidomide treatment, the pati- mented. The patient took thalidomide up
ents were followed regularly with physical to 600 mg per day and had stable disease
examination, hematology, serum biochem- for 2 months. Figure 21.1a was from the
istry and alpha-fetoprotein tests. Computed baseline examination and Figure 21.1b
tomography scan was done before the start from examination done at the documenta-
and after 1 month of thalidomide therapy tion of progressive disease to thalidomide
and then every 2 months. Sonographic treatment. An increase in both tumor area
examination was performed before and and vascularity index was noted. Figure
every 1–2 weeks during thalidomide treat- 21.1c and d were from a 72-year-old
ment. Five patients were considered to have woman with hepatitis C-related cirrhosis.
response to thalidomide treatment, includ- Hepatocellular carcinoma was diagnosed
ing one complete and one partial response by compatible imaging findings and high
based on CT scan findings. Another three alpha-fetoprotein levels. She had received
patients fulfilled the criteria of clinical multiple sessions of trans-arterial chemo-
benefit response described above. The total embolization before enrollment into this
response rate was 11.4% (95% C.I.: 1.6– study. The patient received thalidomide
21.2%). Fifteen patients had stable disease 200 mg per day and stable disease was
and 24 patients had progressive disease. achieved for 23 weeks. Figure 21.1c was
The median pretreatment value of VI from the baseline examination and Figure
was 2.73 (range 0–25.36). Pretreatment VI 21.1d from examination done at the docu-
was significantly higher in patients with an mentation of stable disease to thalidomide
objective tumor response to thalidomide treatment. The tumor area remained sta-
(median 7.42 vs. 2.15, p = 0.03). After tha- tionary while a decrease of vascularity
lidomide treatment, four of the five patients index was noted.
with response had a decrease in VI, but 20 In addition to power Doppler ultra-
of the 36 patients without response had sound, we also checked the levels of cir-
also a decrease of VI. The tumors of three culating proangiogenic factors, including
patients became non-evaluable for tumor vascular endothelial growth factor, basic
vascularity during follow-up because of fibroblast growth factor, and placental
progressive tumor infiltration and blurring growth factor. The pretreatment VI was
of the tumor margins. All of them had pro- significantly correlated with pretreatment
gressive disease. levels of basic fibroblast growth factor and
Representative images of power Doppler placental growth factor, suggesting that VI
ultrasound in our patients are shown in may reflect the activity of tumor angiogen-
Figure 21.1. Figure 21.1a and b were esis. However, none of the proangiogenic
from a 51-year-old man who presented factors we measured differed significantly
with multiple liver and lung tumors. between responders and non-responders to
Hepatocellular carcinoma was diagnosed thalidomide treatment.
282 C. Hsu et al.

Figure 21.1. Power Doppler ultrasound findings of HCC patients treated with thalidomide. a and b
were from a 51-year-old man with progressive disease after thalidomide treatment. c and d were from a
72-year-old woman with stable disease for 23 weeks after thalidomide treatment. a and c, before start of
thalidomide treatment; b and d, at documentation of response to thalidomide treatment

FUTURE PERSPECTIVES OF tumor vascularity in clinical trials of anti-


IMAGING STUDIES FOR angiogenic therapy. Our study indicates
EVALUATION OF ANTI- that power Doppler ultrasound may pro-
ANGIOGENESIS THERAPY vide a convenient, real-time method for
observing the biological effects of agents
Although conventional Doppler ultrasound targeted at tumor vasculature. New tech-
has been extensively studied for evaluation niques are being developed to improve the
of tumor vascularity, the low flow rate in sensitivity and resolution of conventional
tumor microvessels makes this method ultrasound. Of these techniques, micro-
not sensitive enough to detect changes of bubble-based, contrast-enhanced ultrasound
21. Treatment of Hepatocellular Carcinoma with Thalidomide 283

is the most extensively studied. The intra- other molecular targeted therapy for HCC
venously injected microbubbles can signi- is ~ 5% (Philip et al., 2005; Schwartz et al.,
ficantly enhance the Doppler signals of 2006; Abou-Alfa et al., 2006). Therefore, it
blood flow and help differential diagnosis is difficult to find a clear-cut threshold VI
of liver tumors (Kim et al., 1998; Vilana value that may predict the response to anti-
et al., 2003; Quaia et al., 2004). The use- angiogenesis therapy. Besides, in our study
fulness of contrast-enhanced ultrasound decrease in VI after thalidomide treatment
in evaluation of tumor blood flow after was also found in patients with progressive
thalidomide treatment has been suggested disease, suggesting that tumors in these
in a pilot study for patients with advanced patients have surpassed the angiogenesis
HCC (Bertolotto et al., 2006). New imag- regulatory mechanisms that may be inhib-
ing techniques, such as pulse inversion ited by thalidomide. The third limitation
harmonic ultrasound and agent detection is the inherent dependence of ultrasound
imaging, may further improve the image on individual examiner’s technique. Repro-
resolution and reduce artifacts (Choi et al., ducibility of these new sonography tech-
2002; Lee et al., 2006). The potential of niques must be validated before they can
these new techniques in the monitoring of be widely used in the clinical settings.
anti-angiogenesis therapy for HCC should Other functional imaging modalities
be further investigated. have also been extensively studied for
Despite these technical advances, there their use in evaluation of anti-angiogenesis
are still limitations in establishing the therapy. Dynamic contrast-enhanced mag-
role of ultrasound for evaluation of anti- netic resonance imaging (DCE-MRI) has
angiogenesis therapy in HCC. The first been widely used to evaluate the vascular
limitation is the patient population. Most of response, including change in vascular
the HCC patients have underlying cirrhosis perfusion and permeability, after antiang-
and had received various local treatments, iogenic therapy (Padhani and Leach, 2005;
such as trans-arterial embolization or per- Liu et al., 2005). Guidelines for its use
cutaneous ethanol injection, before they in early-phase clinical trials have been
started empirical antiangiogenesis therapy. proposed (Leach et al., 2005; Evelhoch
The margins of their tumors are often et al., 2005). DCE-MRI is relatively non-
indiscrete and calculation of VI is there- invasive and can be objectively measured
fore difficult. Besides, patients with large serially. However, detailed definition of
or deep-seated tumors in the liver may not the examination protocols is needed to
be assessable by ultrasound. In our study improve reproducibility. The potential
only ~ 40% of patients with advanced HCC anti-angiogenesis effects of thalidomide
who were otherwise eligible for thalido- in HCC have been evaluated by DCE-
mide therapy were considered evaluable MRI. Patients with stable disease after
by power Doppler ultrasound. The second thalidomide treatment tended to have
limitation is the difficulty in correlating greater reduction of perfusion in their
the vascular response with other clinical tumors, as indicated by a greater decrease
outcomes, such as tumor shrinkage and in peak and maximal signal enhancement,
patient survival. The response rate of curr- than patients with progressive disease (Wang
ently available anti-angiogenesis therapy or et al., 2004b). Other endpoints of vascular
284 C. Hsu et al.

response, such as the transfer constant Cheng, W.F., Lee, C.N., Chu, J.S., Chen, C.A.,
(Ktrans) and the initial area under the gado- Chen, T.M., Shau, W.Y., Hsieh, C.Y., and Hsieh,
F.J. 1999. Vascular index as a novel parameter
linium concentration time curve, should
for the in vivo assessment of angiogenesis in
be evaluated for their validity in future patients with cervical carcinoma. Cancer 85:
clinical trials of anti-angiogenesis therapy 651–657.
for HCC. Choi, B.I., Kim, T.K., Han, J.K., Chung, J.W.,
In conclusion, evaluation of tumor vas- Park, J.H., and Han, M.C. 1996. Power versus
cularity by power Doppler ultrasound conventional color Doppler sonography: com-
parison in the depiction of vasculature in liver
is feasible in a subset of patients with
tumors. Radiology 200: 55–58.
advanced HCC. The usefulness of power Choi, B.I., Kim, A.Y., Lee, J.Y., Kim, K.W.,
Doppler ultrasound and other functional Lee, K.H., Kim, T.K., and Han, J.K. 2002.
imaging modalities should be explored in Hepatocellular carcinoma: contrast enhancement
future clinical trials of anti-angiogenesis with Levovist. J. Ultrasound Med. 21: 77–84.
therapy for HCC. Chow, N.H., Hsu, P.I., Lin, X.Z., Yang, H.B., Chan,
S.H., Cheng, K.S., Huang, S.M., and Su, I.J.
1997. Expression of vascular endothelial growth
factor in normal liver and hepatocellular carci-
REFERENCES
noma: an immunohistochemical study. Human
Abou-Alfa, G.K., Schwartz, L., Ricci, S., Amadori, D., Pathol. 28: 698–703.
Santoro, A., Figer, A., De Greve, J., Douillard, J.Y., Chow, N.H., Cheng, K.S., Lin, P.W., Chan, S.H.,
Lathia, C., Schwartz, B., Taylor, I., Moscovici, M., Su, W.C., Sun, Y.N., and Lin, X.Z. 1998.
and Saltz, L.B. 2006. Phase II study of sorafenib Expression of fibroblast growth factor-1 and
in patients with advanced hepatocellular carcinoma. fibroblast growth factor-2 in normal liver and
J. Clin. Oncol. 24: 4293–4300. hepatocellular carcinoma. Digest. Dis. Sci. 10:
Bertolotto, M., Pozzato, G., Croce, L.S., Nascimben, F., 2261–2266.
Gasparini, C., Cova, M.A., and Tiribelli, C. 2006. Choyke, P.L., Knopp, M.V., and Libutti, S.K. 2002.
Blood flow changes in hepatocellular carcinoma Special techniques for imaging blood flow to
after the administration of thalidomide assessed tumors. Cancer J. 8: 109–118.
by reperfusion kinetics during microbubble infu- D’Amato, R.J., Loughnan, M.S., Flynn, E., and
sion: preliminary results. Invest. Radiol. 41: Folkman, J. 1994. Thalidomide is an inhibitor of
15–21. angiogenesis. Proc. Natl. Acad. Sci. U.S.A. 91:
Chen, C.N., Cheng, Y.M., Liang, J.T., Lee, P.H., 4082–4085.
Hsieh, F.J., Yuan, R.H., Wang, S.M., Chang, Evelhoch, J., Garwood, M., Vigneron, D., Knopp, M.,
M.F., and Chang, K.J. 2000. Color Doppler Sullivan, D., Menkens, A., Clarke, L., and
vascular index can predict distant metastasis and Liu, G. 2005. Expanding the use of magnetic
survival in colon cancer patients. Cancer Res. resonance in the assessment of tumor response
60: 2892–2897. to therapy: workshop report. Cancer Res. 65:
Chen, C.N., Cheng, Y.M., Lin, M.T., Hsieh, F.J., 7041–7044
Lee, P.H., and Chang, K.J. 2002. Association of Gee, M.S., Saunders, H.M., Lee, J.C., Sanzo,
color Doppler vascularity index and microves- J.F., Jenkins, W.T., Evans, S.M., Trinchieri,
sel density with survival in patients with gastric G., Sehgal, C.M., Feldman, M.D., and Lee,
cancer. Ann. Surg. 235: 512–518. W.M. 2001. Doppler ultrasound imaging detects
Chen, L.T., Liu, T.W., Chao, Y., Shiah, H.S., changes in tumor perfusion during antivascu-
Chang, J.Y., Juang, S.H., Chen, S.C., Chuang, lar therapy associated with vascular anatomic
T.R., Chin, Y.H., and Whang-Peng, J. 2005. alterations. Cancer Res. 61: 2974–2982.
α-fetoprotein response predicts survival benefits Goertz, D.E., Yu, J.L., Kerbel, R.S., Burns, P.N.,
of thalidomide in advanced hepatocellular carci- and Foster, F.S. 2002. High-frequency Doppler
noma. Aliment. Pharmacol. Ther. 22: 217–226. ultrasound monitors the effects of antivascular
21. Treatment of Hepatocellular Carcinoma with Thalidomide 285

therapy on tumor blood flow. Cancer Res. 62: Workman, P., and Pharmacodynamic/Pharma-
6371–6375. cokinetic Technologies Advisory Committee,
Gschwind, A., Fischer, O.M., and Ullrich, A. 2004. Drug Development Office, Cancer Research
The discovery of receptor tyrosine kinases: UK. 2005. The assessment of antiangiogenic
targets for cancer therapy. Nat. Rev. Cancer 4: and antivascular therapies in early-stage clini-
361–370. cal trials using magnetic resonance imaging:
Harada, T., Arii, S., Mise, M., Imamura, T., issues and recommendations. Brit. J. Cancer 92:
Higashitsuji, H., Furutani, M., Niwano, M., 1599–1610.
Ishigami, S., Fukumoto, M., Seiki, M., Sato, H., Lee, J.Y., Choi, B.I., Han, J.K., Lee, J.M., and Kim,
and Imamura, M. 1998. Membrane-type matrix S.H. 2006. State-of-the-art ultrasonography of
metalloproteinase-1 (MT1-MMP) gene is over- hepatocellular carcinoma. Eur. J. Radiol. 58:
expressed in highly invasive hepatocellular 177–185
carcinomas. J. Hepatol. 28: 231–239. Lencioni, R., Pinto, F., Armillotta, N., and
Hsu, C., Chen, C.N., Chen, L.T., Wu, C.Y., Yang, Bartolozzi, C. 1996. Assessment of tumor vascu-
P.M., Lai, M.Y., Lee, P.H., and Cheng, A.L. larity in hepatocellular carcinoma: comparison
2003. Low-dose thalidomide treatment for of power Doppler US and color Doppler US.
advanced hepatocellular carcinoma. Oncology Radiology 201: 353–358.
65: 242–249. Lin, A.Y., Brophy, N., Fisher, G.A., So, S., Biggs, C.,
Hsu, C., Cheng, J.C., and Cheng, A.L. 2004. Yock, T.I., and Levitt, L. 2005. Phase II
Recent advances in non-surgical treatment for study of thalidomide in patients with unre-
advanced hepatocellular carcinoma. J. Formos. sectable hepatocellular carcinoma. Cancer 103:
Med. Assoc. 103: 483–495. 119–125.
Hsu, C., Chen, C.N., Chen, L.T., Wu, C.Y., Hsieh, Little, R.F., Wyvill, K.M., Pluda, J.M., Welles, L.,
F.J., and Cheng, A.L. 2005. Effect of thalidomide Marshall, V., Figg, W.D., Newcomb, F.M.,
in hepatocellular carcinoma: assessment with Tosato, G., Feigal, E., Steinberg, S.M.,
power Doppler US and analysis of circulating Whitby, D., Goedert, J.J., and Yachoan, R. 2000.
angiogenic factors. Radiology 235: 509–516. Activity of thalidomide in AIDS-related Kaposi’s
Kenyon, B.M., Browne, F., and D’Amato, R. 1997. sarcoma. J. Clin. Oncol. 18: 2593–2602.
Effects of thalidomide and related metabolites Liu, G., Rugo, H.S., Wilding, G., McShane, T.M.,
in a mouse corneal model of neovascularization. Evelhoch, J.L., Ng, C., Jackson, E., Kelcz, F.,
Exp. Eye Res. 64: 971–978. Yeh, B.M., Lee, F.T. Jr., Charnsangavej, C.,
Kim, A.Y., Choi, B.I., Kim, T.K., Han, J.K., Park, J.W., Ashton, E.A., Steinfeldt, H.M.,
Yun, E.J., Lee, K.Y., and Han, M.C. 1998. Pithavala, Y.K., Reich, S.D., and Herbst, R.S.
Hepatocellular carcinoma: power Doppler 2005. Dynamic contrast-enhanced magnetic
US with a contrast agent: preliminary results. resonance imaging as a pharmacodynamic measure
Radiology 209: 135–140. of response after acute dosing of AG-013736,
Korn, E.L., Arbuck, S.G., Pluda, J.M., Simon, R., an oral angiogenesis inhibitor, in patients with
Kaplan, R.S., and Christian, M.C. 2001. advanced solid tumors: results from a phase I
Clinical trial designs for cytostatic agents: are study. J. Clin. Oncol. 23: 5464–5473.
new approaches needed? J. Clin. Oncol. 19: Miller, A.B., Hoogstraten, B., Staquet, M., and
265–272. Winkler, A. 1981. Reporting results of cancer
Krause, D.S., and Van Etten, R.A. 2005. Tyrosine treatment. Cancer 47: 207–214.
kinases as targets for cancer therapy. N. Engl. J. Padhani, A.R., and Leach, M.O. 2005. Antivascular
Med. 353: 172–187. cancer treatments: functional assessments by
Leach, M.O., Brindle, K.M., Evelhoch, J.L., dynamic contrast-enhanced magnetic resonance
Griffiths, J.R., Horsman, M.R., Jackson, A., imaging. Abd. Imaging 30: 324–341
Jayson G.C., Judson, I.R., Knopp, M.V., Maxwell, Parkin, D.M. 2001. Global cancer statistics in the
R.J., McIntyre, D., Padhani, A.R., Price, P., year 2000. Lancet Oncol. 2: 533–543.
Rathbone, R., Rustin, G.J., Tofts, P.S., Tozer, Patt, Y.Z., Hassan, M.M., Lozano, R.D., Nooka,
G.M., Vennart, W., Waterton, J.C., Williams, S.R., A.K., Schnirer, I.I., Zeldis, J.B., Abbruzzese, J.L.,
286 C. Hsu et al.

and Brown, T.D. 2005. Thalidomide in the treat- Stephens, T.D., Bunde, C.J., and Fillmore, B.J.
ment of patients with hepatocellular carcinoma: 2000. Mechanism of action in thalidomide tera-
a phase II trial. Cancer 103: 749–755. togenesis. Biochem. Pharmacol. 59: 1489–1499.
Philip, P.A., Mahoney, M.R., Allmer, C., Thomas, J., Torimura, T., Sata, M., Ueno, T., Kin, M., Tsuji, R.,
Pitot, H.C., Kim, G., Donehower, R.C., Fitch, T., Suzaku, K., Hashimoto, O., Sugawara, H., and
Picus, J., and Erlichman, C. 2005. Phase II Tanikawa, K. 1998. Increased expression of
study of erlotinib (OSI-774) in patients with vascular endothelial growth factor is associated
advanced hepatocellular cancer. J. Clin. Oncol. with tumor progression in hepatocellular carci-
23: 6657–6663. noma. Human Pathol. 29: 986–991.
Quaia, E., Calliada, F., Bertolotto, M., Rossi, S., Vacca, A., Scavelli, C., Montefusco, V., Di Pietro, G.,
Garioni, L., Rosa, L., and Pozzi-Mucelli, R. 2004. Neri, A., Mattioli, M., Bicciato, S., Nico, B.,
Characterization of focal liver lesions with Ribatti, D., Dammacco, F., and Corradini, P. 2005.
contrast-specific US modes and a sulfur Thalidomide downregulates angiogenic genes in
hexafluoride-filled microbubble contrast agent: bone marrow endothelial cells of patients with
diagnostic performance and confidence. Radiology active multiple myeloma. J. Clin. Oncol. 23:
232:420–430. 5334–5346.
Rubin, J.M., Bude, R.O., Carson, P.L., Bree, Vilana, R., Llovet, J.M., Bianchi, L., Sanchez, M.,
R.L., and Adler, R.S. 1994. Power Doppler Pages, M., Sala. M., Gilabert, R., Nicolau, C.,
US: a potentially useful alternative to mean- Garcia, A., Ayuso, C., Bruix, J., and Bru, C.
frequency-based color Doppler US. Radiology 2003. Contrast-enhanced power Doppler sonog-
190: 853–856. raphy and helical computed tomography for
Schwartz, J.D., Sung, M., Schwartz, M., Lehrer, D., assessment of vascularity of small hepatocellular
Mandeli, J., Liebes, L., Goldenberg, A., and carcinomas before and after percutaneous abla-
Volm, M. 2005. Thalidomide in advanced hepa- tion. J. Clin. Ultrasound 31: 119–128.
tocellular carcinoma with optional low-dose Wang, J., Chen, L.T., Tsang, Y.M., Liu, T.W., and
interferon-α2a upon progression. Oncologist 10: Shih, T.T. 2004a. Dynamic contrast-enhanced
718–727. MRI analysis of perfusion changes in advanced
Schwartz, J.D., Schwartz, M., Lehrer, D., Cohen, E., hepatocellular carcinoma treated with an antian-
Sung, M., Kinkhabwala, M., Siegel, A., giogenic agent: a preliminary study. Am. J.
Holloway, S., Ocean, A., and Wadler, S. 2006. Roentgenol. 183: 713–719.
Bevacizumab in unresectable hepatocellular Wang, T.E., Kao, C.R., Lin, S.C., Chang, W.H.,
carcinoma (HCC) for patients without metastasis Chu, C.H., Lin, J., and Hsieh, R.K. 2004b.
and without invasion of the portal vein. Proc. Salvage therapy for hepatocellular carcinoma
Am. Soc. Clin. Oncol. 24: abstr 4144. with thalidomide. World J. Gastroenterol. 10:
Singhal, S., Mehta, J., Desikan, R., Ayers, D., 649–653.
Roberson, P., Eddlemon, P., Munshi, N., Anaissie, Wu, C.C., Lee, C.N., Chen, T.M., Shyu, M.K.,
E., Wilson, C., Dhodapkar, M., Zeddis, J., Hsieh, C.Y., Chen, H.Y., and Hsieh, F.J. 1994.
and Barlogie, B. 1999. Antitumor activity of Incremental angiogenesis assessed by color
thalidomide in refractory multiple myeloma. N. Doppler ultrasound in the tumorigenesis of ovarian
Engl. J. Med. 341: 1565–1571 neoplasm. Cancer 73: 1251–1256.
22
Perfusion Scintigraphy with Integrated
Single Photon Emission Computed
Tomography/Computed Tomography
in the Management of Transarterial
Treatment of Hepatic Malignancies
Timm Denecke, Bert Hildebrandt, and Enrique Lopez-Hänninen

INTRODUCTION the use of different imaging modalities,


especially single photon emission com-
During the recent years it has been shown puted tomography (SPECT) and SPECT
that transarterial applications of antican- with integrated computed tomography
cer agents are promising approaches in (SPECT-CT), for planning and control of
the treatment of hepatic malignancies. treatment will be discussed.
While new techniques led to a reappraisal
of hepatic arterial infusion chemotherapy
(HAIC) over implanted port systems, CURRENT STATUS OF HEPATIC
there has also been a growing interest in
ARTERIAL CHEMOTHERAPY
the use of transarterial radioembolization
(RE). The concept of transarterial regional AND RADIOEMBOLIZATION
oncologic treatment of hepatic malignan-
Because liver tumors are supplied prima-
cies presupposes that the entire liver can
rily by the hepatic artery, direct intraarte-
be exclusively treated over the supplying
rial infusion of chemotherapeutic agents
arteries. Several imaging modalities are
can expose the tumor to a much higher
in use for prediction and control of per-
drug concentration than intravenous therapy.
fusion territories of intraarterial catheters.
For transarterial RE, a semiselectivity of
Besides digital subtraction angiography
drug delivery is given by the low (∼ 30% or
(DSA), computed tomography (CT) and
less) arterial blood supply of normal liver
magnetic resonance imaging (MRI), scin-
tissue with the portal vein as the main sup-
tigraphic imaging is being employed to
plying vessel.
visualize the intrahepatic perfusion pat-
tern, to exclude extrahepatic perfusion,
and to assess an intrahepatic arteriovenous Intraarterial Chemotherapy
shunting to the lung. In the following, the The role of HAIC in patients with cancers
current status of HAIC and RE in cancer confined to the liver has been controver-
therapy, the application techniques and sially discussed for many years, whereby

287
288 T. Denecke et al.

most experiences with this approach has advantage of systemic drug concentrations
been gained with the treatment of “liver provided by the intraarterial application of
only”-metastases of colorectal can- 5-FU (Lorenz and Muller, 2000; Sadahiro
cer (Cohen and Kemeny, 2003; MAGC, et al., 2004).
1996). In addition, limited data are avail- Unlike hepatic arterial FUDR, which
able for other liver neoplasms such as is usually delivered through surgically
hepatocellular (Tanaka et al., 2005) or implanted infusion pumps, regional appli-
cholangiocellular carcinoma (Cantore cations of 5-FU demand the use of external
et al., 2005). Recently, the introduction of pumps, due to the maximal drug concen-
minimal invasively implanted port systems tration of 25 mg/ml. Therefore, hepatic
led to a reappraisal of this approach (Ricke arterial 5-FU can only be delivered by
et al., 2004). intermittent percutaneous access, or
For colorectal cancer patients with unre- through port systems. There is extensive
sectable liver metastases or after hepatic experience with the use of port catheters in
metastasectomy, more than a dozen of ran- a number of specialized treatment centers
domized trials have compared HAIC using worldwide, but this technique is associ-
fluoropyrimidines floxuridine (FUDR) or ated with a considerable proportion of
5-flourouracil (5-FU) with intravenous primary failure which has been reported to
drug application (Cohen and Kemeny, approximate one third of patients sched-
2003; Nelson and Freels, 2004). Summing uled for 5-FU-based HAIC in recent mul-
up those results, HAIC proved to improve ticenter trials (Lorenz and Muller, 2000;
response rates in patients with unresect- Kerr et al., 2003). Once HAIC has been
able disease and time to hepatic progres- initiated, port catheters are associated with
sion for both indications, but results were higher complication rates than surgically
counterbalanced by the lack of a survival implanted pumps, whereas secondary fail-
benefit in most of the studies. ure rates have been reported to be similar
The reasons preventing a clear-cut sur- (Heinrich et al., 2003). Presently, it is
vival benefit for HAIC have been dis- accepted that the low performance level of
cussed in detail (Cohen and Kemeny, surgically implanted port catheters repre-
2003; MAGC, 1996; Nelson and Freels, sents one major reason why the theoretical
2004). Particularly in the earlier trials, advantage of 5-FU-based HAIC could
small patient numbers and inappropriate not be transferred to convincing clinical
control groups may have led to mislead- improvements.
ing interpretations. In addition, exces- In this context, interventionally
sively high rates of extrahepatic failure implanted port catheter systems (IIPCS)
were observed in studies on intraarte- have evolved a promising alternative to
rial FUDR. Adjacent trials completed in surgically implanted devices. The use of
North America suggested that systemic IIPCS enables initiation of HAIC with-
control rates of intraarterial FUDR can be out laparotomy, and available data sug-
improved by combinations with intrave- gest favorable complication and failure
nous 5-FU (Kemeny et al., 1999). In addi- rates with this approach. However, studies
tion, European and Asian investigators published so far mainly focused on either
aimed to optimize results by taking the technical or clinical aspects of treatment
22. Perfusion Scintigraphy with Integrated Single Photon Emission 289

(Ricke et al., 2004). Further trials are nec- of these patients resection was enabled by
essary to more clearly define the value of therapy related shrinkage of the tumors.
this promising novel technique. Concerning colorectal cancer metas-
tases, the most relevant (randomized phase
Transarterial Radioembolization III) trial so far included 71 patients who
received intraarterially either FUDR alone
As another innovative modality, the arte-
or FUDR in combination with Y90 resin
rial administration of Y90 loaded glass
microspheres. There was a significant dif-
and resin microspheres to the liver for
ference observed concerning the mean
treatment of hepatic malignancies was
time to disease progression in both groups,
currently approved by the North American
which were favorable for those patients
Food and Drug Administration (FDA) for
who were treated with radioembolization
specific indications, the stand alone or pre-
(9.7 vs. 15.9 months, p = 0.001) (Gray
surgical or pretransplant radiation therapy
et al., 2001). Similar survival benefits
of irresectable hepatocellular carcinoma
were demonstrated in other trials for RE
(glass device, 2000) and the treatment of
plus chemotherapy vs. chemotherapy alone
irresectable colorectal metastases in com-
with a mean survival time of 29.4 vs. 12.8
bination with intraarterial FUDR (resin
months (Van Hazel et al., 2004). Studies
device, 2002). Currently, the widest experi-
employing the glass microsphere device
ence comprises colorectal liver metastases
also achieved encouraging results with sta-
and hepatocellular carcinoma, and there
ble disease or partial response in more than
is evidence that both devices are suitable
50% of cases (Andrews et al., 1994).
for the treatment of hepatocellular carci-
noma and colorectal metastases (Murthy
et al., 2005). However, radioembolization TECHNIQUE OF
of other tumor entities in the liver (e.g.,
breast cancer, gall bladder cancer, thyroid
TRANSARTERIAL
cancer, non-small cell lung cancer) has TREATMENT
also shown encouraging results concern-
Hepatic Arterial Infusion Chemotherapy
ing local control and may prove beneficial
in the future. While intraarterial application of FUDR is
In hepatocellular carcinoma, a phase II exceptionally performed using surgically
trial has shown improved mean survival implanted infusion pumps, 5-FU can be
in those patients receiving a higher dose administered intermittently via a percutane-
of Y90 glass microspheres (> 104 Gy, 635 ous access. This can be done over a subcu-
days; < 104 Gy, 323 days) (Dancey et al., taneous port reservoir linked to a catheter
2000). Data of another trial, summarizing which is inserted into the arterial system.
80 patients with hepatocellular carcinoma Surgical intraarterial port systems are usually
treated with different stages of disease implanted within the bounds of a laparotomy
and doses of 47–270 Gy showed similar with hepatic resection or resection of the
survival times as reported for transarterial primary. A subcutaneous port reservoir is
chemoembolization. The response rate in placed ventrally at the upper abdomen with
a trial including 71 patients was 27%, in 4 the catheter inserted retrogradely into the
290 T. Denecke et al.

Radioembolization
dissected gastroduodenal artery (GDA).
The perfusion territory is controlled ini- Radioembolization (RE) is a one-time
tially by injection of e.g., methylene blue. procedure and is usually not thought to be
The surgical placement of intraarterial port repeated. The therapeutic agent consists
systems is definite. In case of system dys- of microspheres, 20–40 nm in diameter,
function, e.g., catheter or artery occlusion, which contain a radionuclide emitting
a revision is difficult. Thus, dysfunction of beta-radiation (e.g., Yttrium90) (Gray
surgical ports usually leads to a replace- et al., 2001). The microspheres are deliv-
ment of HAIC by systemic treatment ered into the hepatic artery and embolize
(Heinrich et al., 2003). the microvessels and capillaries of tissues.
Employing novel catheterization tech- Again, the dominance of arterial supply
niques, minimally invasive interventional of hepatic malignancies in contrast to the
implantations of intraarterial port systems normal liver is used for a semiselectivity
became possible (Ricke et al., 2004). For of drug delivery. The typical vascular
this alternative approach, port catheters are architecture of liver metastases leads to a
usually positioned through a femoral artery predominant seeding of the microspheres
access under DSA guidance to position the into the outer regions of the tumors, which
catheter tip in the hepatic artery. The subcu- are hypervascularized and thus hyperoxy-
taneous port reservoir is placed just inferior genated, making radiation in these areas
to the groin. A subclavicular approach is also more effective (Campbell et al., 2001).
practicable. In principle, minimal invasively The transarterial access is similar to that
implanted port systems appear to be superior described for interventional radiological
to surgically implanted port catheters with port placement. Alternatively, the injection
respect to primary and secondary failure and can be performed over an intraarterial port
the possibility to easily repair or replace the system supplying the liver. For the procedure
systems in case of dysfunction (Ricke et al., of drug application, radiation protection
2004; Sadahiro et al., 2004). An additional is mandatory. Therefore, specific devices
advantage is the minimal risk of the proce- for application were developed in order
dure as compared with the mortality rate of to avoid direct and prolonged contact of
surgical port placement (Kerr et al., 2003). the radiologist to the therapeutic agent. To
However, the correct position of intraar- preserve a hepatic reserve, a fractionized
terial catheters regarding hepatic and application selectively to one and sub-
extrahepatic distribution of therapeuti- sequently to the other liver lobe is under
cally applied drugs, unlike in surgically evaluation.
implanted catheters, cannot be confirmed Avoidance of extrahepatic implantation of
by intra-operative application of e.g., microspheres, potentially causing gastritis,
methylene blue, and dislocations of the ulcers, pancreatitis and other gastrointestinal
catheter after initial placement and dur- morbidities, is even more decisive than in
ing the therapy course have to be consid- HAIC (Murthy et al., 2005). Therefore,
ered (Ricke et al., 2004). Therefore, valid a sufficient and careful planning of the
imaging procedures are mandatory to pre- procedure is mandatory. The interven-
serve the advantages of minimal-invasive tion planning includes an arterial selec-
catheter implantation in clinical practice. tive catheterization with application of
22. Perfusion Scintigraphy with Integrated Single Photon Emission 291

Tc99m-MAA and subsequent planar scin-


extrahepatic perfusion can be best
tigraphy to predict the distribution of
diagnosed by port scintigraphy with
microspheres, subsequently released at the
Tc-99m labeled macroaggregated albu-
same intraarterial location, with respect
min (MAA) employing planar imaging
to shunting to the lung and gastrointestinal
and SPECT (Cohen and Kemeny, 2003;
accumulations (Murthy et al., 2005). The
Lehner et al., 1987; Lubin et al., 1987).
most exact detection of extrahepatic accu-
Further techniques such as perfusion CT,
mulation is desirable in order to avoid
and slow-infusion magnetic resonance
unnecessary treatment complications. This
arteriography (MRA) have been used to
and the control of microsphere distribution
evaluate the intrahepatic arterial flow in
by Bremsstrahlung scintigraphy after RE
patients with hepatic intraarterial ports
are valuable indications for SPECT-CT.
(Morimoto et al., 1999; Seki et al., 2003),
The issues concerning the avoidance of
but did not gain broad clinical accept-
inadvertent deposition of therapeutic agent
ance. In the routine surveillance during
into non-target arteries are the same as
treatment, both scintigraphy with Tc-99m
discussed for HAIC. The quantification of
labeled MAA and DSA are suitable to
pulmonary shunting is more important for
check for patency of catheters and hepatic
RE than for HAIC, because the complica-
arteries as well as system leaking, but may
tions after RE with pneumonitis and pul-
lack valid information about completeness
monary insufficiency are life threatening,
and homogeneity of hepatic perfusion. A
while pulmonary complications in HAIC
combination of perfusion imaging with
are controversially discussed. However,
sufficient anatomic information may be
planar scintigraphy is sufficient for dosing
useful to ensure homogenous perfusion of
microspheres. the entire liver in patients treated with HAI
via intra-arterial port systems. This espe-
cially holds true for those patients with
VISUALIZATION aberrant hepatic arteries and/or who previ-
OF PERFUSION TERRITORIES ously underwent hepatic resection (Ricke
OF HEPATIC INTRAARTERIAL et al., 2004; Denecke et al., 2005).
CATHETERS IN PLANNING
AND CONTROL Single Photon Emission Computed
OF TRANSARTERIAL Tomography with Integrated Computed
TREATMENT Tomography for Port Perfusion
Scintigraphy
Imaging Techniques
A novel tool of nuclear medicine imaging,
There are several issues to be addressed by a hybrid SPECT-CT camera, is potentially
imaging techniques employed for the rou- able to show both parenchymal perfusion
tine control of intraarterial port systems over the port system and anatomic rela-
before HAIC. Before initiation of treat- tion for correct assignment of intra- and
ment, leaking of the port system and alter- extrahepatic tracer accumulation (Denecke
ations of supplying arteries and catheters et al., 2005). This device combines a
may be easily detected by DSA, whereas dual head gamma camera with a rotating
292 T. Denecke et al.

low dose X-ray tube enabling both X-ray tracer retrogradely into arteries originating
based attenuation correction for enhanced proximally from the tip of the port line,
image quality of SPECT and anatomic suggesting a catheter dislocation. Just after
mapping on inherently fused SPECT-CT sequential imaging, static planar acquisi-
images with sufficient spatial resolution tion is started. Anterior, right oblique and
to delineate the liver silhouette (Bocher right lateral planar image acquisitions of
et al., 2000). Other comparable devices, the upper abdomen (each up to 800,000
which became available more recently, counts) as well as anterior projections of
consist of a dual head gamma camera and the lower abdomen to rule out leaking
an integrated diagnostic multirow detector and of the thorax to assess shunting to the
CT scanner. lung are obtained. In patients with two
A recommendable scintigraphy proto- port systems, the activity can be divided
col for a low dose SPECT-CT includes by two and is injected into the port sys-
planar scintigraphy covering pelvis, abdo- tems, each injection followed by dynamic
men, and thorax, as well as tomographic and static planar imaging as described
imaging of the relevant organs in the above. Then (∼ 20 min after tracer injec-
upper abdomen with image fusion for ana- tion) a SPECT-CT of the upper abdomen
tomical mapping. The patient is prepared is performed using a dual-head gamma
with a thyroid blockage (e.g., perchlorate, camera (LEHR collimators; FOV, 540 ×
potassium iodide). This is important to 400 mm; matrix, 128 × 128; 120 frames;
avoid active Tc-99m uptake of the gastric 3° steps; 15 s/frame) with an integrated
mucosa to enable the identification of rotating X-ray tube (fixed tube current,
accidental embolization of Tc-99m-MAA 2.5 mA; 140 kV; slice thickness, 10 mm;
into the stomach wall. After testing the matrix, 128 × 128; rotation time, 13.5 s;
port system for patency with a bolus of step and shoot technique) for attenuation
isotonic saline (0.9%), 200 MBq Tc-99m- correction (“Hawkeye”Millenium VG, GE
MAA in 2 ml saline (the syringe must Medical Systems). With this device, total
be shook before injection to optimize acquisition time is ∼ 27 min including the
the distribution) is injected into the port initial transmission scan using the low-
capsule followed by another saline flush dose X-ray tube (9 min for 40 cm scan
(∼ 5–10 ml) under dynamic anterior pla- length) and the subsequent SPECT result-
nar image acquisition of the abdomen (1 ing in a delay of SPECT acquisition from
image/5 s over 1 min) with a low energy tracer injection of ∼30 min. After iterative
high resolution (LEHR) collimator to rule reconstruction (OSEM algorithm; number
out tracer extravasation. The injection has of iterations, 2; number of subsets, 10;
to be performed carefully and slowly. prefiltering, Butterworth (frequency, 0.25;
This is important because of two major order, 10); postfiltering, Butterworth (fre-
problems: (1) the viscosity of MAA needs quency, 0.5; order, 10) and attenuation
a relatively high injection pressure (espe- correction (attenuation map derived from
cially when thin supraselective catheters the low dose CT), the reconstructed data
are used as port lines), which may cause are visualized in sagital, coronal, and axial
rupture of the port system, and (2) a high slices, with the SPECT, the CT and the
injection rate can cause a reflux of the fusion images side by side. Inherent image
22. Perfusion Scintigraphy with Integrated Single Photon Emission 293

fusions are generated from the coregis- While calculation of an intrahepatic


tered SPECT and low-dose CT images. shunting to the lung can be assessed
With a SPECT-CT unit consisting of visually and semiquantitatively on planar
a diagnostic dedicated CT scanner, the images, the intrahepatic perfusion terri-
imaging protocol would be similar. In tory of the port can be best evaluated on
this case, an unenhanced scan of the area SPECT images with anatomic mapping.
covered by SPECT would be acquired in For readings of port perfusion scintigraphy
low dose technique (∼ 40 mAs) which is obtained with integrated SPECT-CT, the
enough for anatomic mapping. Regular physician should be aware of the patients
CT scans should only be performed if clinical history. Knowledge of previous
restaging is necessary. The patient is asked hepatic surgery, or other interventional
to hold the breath in mild expiration for a procedures (e.g., radiofrequency ablation)
better congruency of the diaphragm posi- and other conditions (e.g., portal vein
tion in SPECT and CT. This is not required thrombosis, bile duct obstruction, tumor
when a low-dose device is used as the burden) potentially affecting the liver sil-
X-ray tube takes ∼ 13 s for one rotation houette is important. Previously performed
covering 1 cm scan length, resulting in an imaging procedures such as MRI or CT
averaged image of the diaphragm move- are helpful to gather information about
ment comparable to that of SPECT. the liver configuration and localization of
hepatic metastases. Additionally, it is nec-
essary to know the arterial anatomy in the
Image Analysis
upper abdomen and the (desired) location
Image interpretation includes several issues: of the port line tip.
(1) the patency of the port line, (2) the If the tracer is injected slowly, extra-
assessment of intrahepatic arteriovenous hepatic accumulation can occur only in
shunting to the lung, (3) tracer extravasation perfusion territories of arteries originating
indicative of leakage of the port system, (4) distally from the port line tip. The desired
the presence and location of intraabdominal position of the catheter tip in the case of
extrahepatic tracer embolization, and (5) a regular arterial anatomy is the proper
the intrahepatic perfusion pattern especially hepatic artery, distally of origins of arte-
regarding the perfusion intensity of tumor rial branches supplying organs other then
baring segments (Ziessman et al., 1985; the liver. However, there are numerous
Denecke et al., 2005). variants of both the hepatic arteries and
Patency of the port system is assessed by the arterial branches to the gastrointesti-
the performance of injection and describes nal tract. These normal variants may be
the line as blocked or smooth running. A relevant for transarterial treatments and
leakage of the system is rarely present and the interpretation of planning and control
usually occurs as a disconnection at the imaging. They have recently been exten-
junction of the catheter to the reservoir as sively reviewed by Liu et al., (2005).
a weak point. This is indicated by a tracer A few arteries, which are potentially
retention at the groin without activity reach- included in the perfusion territory of
ing the liver or upper abdomen on initial intraarterial liver catheters, surgically and
dynamic images obtained during injection. minimal invasively implanted systems
294 T. Denecke et al.

alike, are to be mentioned. The cystic artery implantation, the incidence of endoscopi-
arises usually from the right hepatic artery cally confirmed mucosal lesions is 36%,
(71%). This implies the risk of therapy compared to 3% in cases with sufficient
related cholecystitis. If the gall bladder has coiling. Thus, it is important to exclude
not been resected previously, intraarterial insufficient coiling with recanalization of
drug delivery should be performed distally the vessel (4% of cases), subsequent open-
from the origin of the cystic artery. The ing collateral vessels of the right gastric
right gastric artery is a minor contributor artery before application of therapeutic
to the gastric blood supply. Variation of agents proximally to its origin. The GDA
the right gastric artery is common. It may and its branches may be responsible for
arise from the proper hepatic artery (51%), duodenal ulceration or pancreatitis dur-
the left (23%) or the right (3%) hepatic ing regional therapy, when coiled insuffi-
artery, the origin of the GDA (3%) or the ciently or subsequently opening collaterals
common hepatic artery (9%). The iden- were initially not visible during coiling
tification of this vessel is imperative for and MAA test injection (Liu et al., 2005).
regional treatment, as inflammation with If extrahepatic perfusion of non-target ves-
gastric necrosis, mucosal ulceration, and sels is detected before therapy application
perforation are potential complications (Figure 22.1), a reintervention or a change
of inadvertent delivery of RE similarly of the therapeutic management becomes
as in intraarterial infusion chemotherapy. necessary (Denecke et al., 2005).
Here, insufficient coiling of the right gas- Tc-99m-MAA deposition to non-target
tric artery during minimally invasive port vessels can easily be seen when there

Figure 22.1. Port perfusion SPECT-CT of a 64-year-old male who recieved coil embolization of the
right gastric artery and the gastroduodenal artery before transarterial radioembolisation with Y90 micro-
spheres. The red target demonstrates tracer accumulation in the gastric wall. In a second angiography
session, a collateral vessel supplying the gastric wall was identified and occluded
22. Perfusion Scintigraphy with Integrated Single Photon Emission 295

is accumulation in the spleen, indicating are change of the liver silhouette due to
reflux or dislocation of port lines into the surgical treatment (e.g., hemihepatectomy,
celiac trunk. Accumulation in the gastric enucleation) or tumor and treatment related
wall in relevant amounts can be depicted dystrophy. To distinguish sufficient supply
on planar images, if a focal or longitudinal with heterogeneous accumulation (that
tracer retention occurs clearly separated are not affecting therapy efficacy) from
from the liver silhouette. Discrete accumu- true hypoperfusion or perfusion defects
lations in the right gastric wall close to the on scintigraphic images, it is necessary
liver, however, can be obscured by the inten- to have anatomic images for correlation.
sive uptake of the adjacent hepatic paren- Ideally, image fusion helps to compare
chyma. These subtle findings can usually the scintigraphically visualized perfusion
be depicted on axial and coronal SPECT territory and the liver outlines, which is
images. This, as well as accumulation in important in cases with atypical liver con-
the gall bladder or the GDA bed, is best figuration. Additionally, heterogeneous
diagnosed on SPECT-CT fusion images MAA accumulations on SPECT images
(Figure 22.1). The clear outline of the liver can be correlated to the size, shape, and
on the anatomic images help to discriminate distribution of hepatic tumor lesions on
extrahepatic foci from hepatic uptake. This CT or MRI to clearly identify unsup-
especially holds true in the cases with an plied liver segments, especially those with
untypical configuration of the liver or a het- tumor involvement.
erogeneous intrahepatic perfusion pattern. A lack of supply can be caused by sev-
For an effective regional chemotherapy eral problems. A (subtotal) port line occlu-
of liver metastases, the intrahepatic tran- sion is diagnosed easily by unsuccessful
sarterial supply with chemotherapy agents saline injection and requires recanaliza-
should address the entire liver or at least tion. System leakage and disconnection is
reach a sufficient level in those segments discussed above. Occlusion of the arterial
with tumor burden. Heterogeneous per- vessels supplied by the port catheter can
fusion detected on Tc-99m-MAA SPECT also occur. This can be due to intima irri-
can be due to several reasons, such as tation by the catheter or the chemotherapy
hyper- or hypoperfusion of tumors or agent, resulting in dissection and/or throm-
vascular alterations. Heterogeneous int- bosis. Tracer reflux to proximal non-target
rahepatic perfusion patterns that do not vessels is seen in the case of a total occlu-
represent malsupply of tumor baring seg- sion of the hepatic bed. This is sometimes
ments and do not require an alteration of accompanied by a prominent focal tracer
the therapeutic management, can poten- retention in the region of the catheter tip,
tially be misinterpreted on scintigraphic which, however, is not a pathognomonic
images because of the lack of anatomical sign for arterial occlusion. A partial lack
background information. These are for of hepatic supply can be observed in the
example, necrotic areas within hepatic case of occlusion of the right or left liver
tumors and areas previously treated with artery only. Just as perfusion of non-
chemoembolization or interstitial tumor target vessels, these are important findings
ablation (e.g., radiofrequency ablation). and require recanalization efforts before
Other potential causes of misinterpretation resumption of therapy infusion.
296 T. Denecke et al.

Another reason for hypoperfusion or is possible by placing the catheter tip into
perfusion defects within the liver are aber- the proper hepatic artery. In patients with
rant hepatic arteries. There exist numer- aberrant hepatic arteries, however, a tactic
ous variants of the anatomy of hepatic is needed, which allows for perfusion of the
arteries. The relevant variants for minimal entire liver without changing the location
invasive implantation and scintigraphic of agent release. There are different strate-
control of intraarterial liver ports are listed gies in use: (1) catheter implantation into
in the following. An aberrant or accessory the major feeder and neglect of the minor
right hepatic artery occurs in 10–31% hepatic arteries (Figure 22.2); (2) catheter
of cases, with 96% of these originating implantation into the dominant vessel and
from the superior mesenteric artery (Liu proximal coil embolization of the minor
et al., 2005). The caliber of accessory hepatic artery; (3) Catheter implantation
right hepatic arteries varies as well as the into the splenic artery with the therapeutic
supplied volume of liver. An accessory or agent entering the liver over the portal
replaced left hepatic artery is present in vein; or (4) double port implantation into
12–21% of cases. It usually arises from the hepatic arteries of separate origin with a
left gastric artery and thus communicates port reservoir in each groin are not to be
with the gastric and esophageal bed. Other favored as the supply of hepatic malignan-
relevant variants are double hepatic arter- cies is predominantly arterial not portal,
ies with separate origin from the aorta or and a double port system poses a doubled
an accessory middle hepatic artery (Liu risk of dysfunction or complication (Ricke
et al., 2005). et al., 2004).
Regarding the application of therapeutic To ignore a minor accessory vessel results
agents for HAIC as a continuous proce- in a parallel blood flow. Macroaggregated
dure or for RE as a one-time procedure, albumine (MAA) as well as therapeutic
specific considerations have to be made agents injected in one hepatic artery reaches
for both intervention and scintigraphic only this territory, while the neighboring
control. For a one-time procedure, it is territory is fed by the ignored vessel. This
possible to divide the amount of therapeutic is represented by a signal defect on per-
agents into two or more portions and to fusion SPECT (Denecke et al., 2005). To
infuse them over an angiographic cath- interpret this defect correctly, the outline
eter selectively into the supplying arteries of the liver (visualized on SPECT-CT)
of the liver or single liver lobes. This is as well as the arterial anatomy and the
advantageous not only because this tech- desired location of the port line are needed
nique is independent from anatomic vari- (Figure 22.2).
ants with separate hepatic arteries but also To avoid hypoperfusion in tumor bearing
for fractionation of the therapy into two segments which are supplied by accessory
single lobe treatment sessions in order to vessels, a different strategy is needed. It is
preserve a hepatic reserve. In contrast, con- proposed that proximal coil embolization
tinuing infusions of chemotherapy require of one liver artery induces intrahepatic
a single fix location of the port line, flood- collateralization from the remaining feeder
ing the arterial bed of the entire liver. In by lowering the filling pressure in the
patients with regular arterial anatomy, this depending arterial bed. Coil embolization
22. Perfusion Scintigraphy with Integrated Single Photon Emission 297

a b

Figure 22.2. Fifty-one-year-old male with bilobar liver metastases from colorectal cancer: CT-angiography
(Figure 22.2a, volume rendering reconstruction) shows the port line (5) inside the abdominal aorta and the
celiac trunc with its tip (4) in the common hepatic artery just proximal from the origin of the coil embol-
ized gastroduodenal (3) artery. The port system supplies the right hepatic artery (2), while the replaced
left hepatic (1) artery arises separately from the left gastric artery. Consecutively, the port perfusion
SPECT-CT (Figure 22.2c) shows sufficient supply of the right liver lobe and a hypoperfused area in the
left lateral segments. No accumulation of tracer is seen outside the liver, stating the sufficient emboliza-
tion of the gastroduodenal artery. As a consequence, proximal coiling of the left hepatic artery should be
performed to induce intrahepatic collateralization from the right lobe to improve the therapeutic access to
the metastases in the left lateral segments as seen on the CT scan (Figure 22.2b)

of accessory arteries causes some col- interpretation of scintigraphic findings,


lateralization from the remaining artery, knowledge about the arterial anatomy and
and also increased portal vein supply. the shape of the liver are required.
Even though the arterial supply and thus
the supply with therapeutic agents in the
embolized territory is reduced, it is still Therapeutic Consequences
the best achievable result. On perfusion A diagnostic modality has to be judged
SPECT, this imposes a hypoperfused area by the potential impact on the therapeutic
(Denecke et al., 2005). Again, for correct management. More recently published
298 T. Denecke et al.

studies have shown a relevant impact of correct placement and homogenous and
port perfusion scintigraphy with SPECT exclusive supply of the liver (Lehner et al.,
and SPECT-CT on the therapeutic strategy 1987; Lubin et al., 1987; Morimoto et al.,
in 33% (8/24) of patients. In two of these 1999; Seki et al., 2003; Pelosi et al., 1999;
patients, the relevant finding leading to anSuzuki et al., 1991; Denecke et al., 2005).
alteration of the therapeutic management In transarterial RE with Y90-microspheres,
was only depicted on SPECT-CT fusion intraaterial hepatic perfusion scintigraphy
images. The consequences were repre- is mandatory to evaluate the shunt volume
sented by repositioning of the port lines, to the lung and, as in HAIC, to exclude
recanalization therapy of port lines and/ extrahepatic intraabdominal accumulation.
or vessels or coil embolization of non-target
For perfusion scintigraphy of intraarte-
vessels, not visualized by initial DSA. The rial port systems promising results have
opportunity of repositioning is, compared been reported (Lubin et al., 1987; Pelosi
with the formally common surgical tech- et al., 1999). These evaluations have
nique of port placement, a characteristic focused specifically on the exclusion
advantage of angiographically implanted of extrahepatic perfusion to avoid addi-
ports. This even further strengthens the tional morbidity of regional chemotherapy.
value of port perfusion scintigraphy, since With the employment of SPECT it is
the consequence of inadequate perfusion possible to get additional information
territories is not to stop HAIC and start about the intrahepatic distribution of
systemic chemotherapy instead, but to tracer injected over the intra-arterial port
optimize drug delivery. (Suzuki et al., 1991).
In RE patients, the therapeutic impact Many patients receiving HAIC or RE
of perfusion scintigraphy is 100% as the have unusual anatomic configurations of
modality is mandatory as a planning tool the liver (e.g., owing to surgical or locally
in every single patient. This, however, is to
ablative therapy of hepatic metastases,
assess the hepatopulmonary shunt volume excessive tumor burden, dysmorphic liver).
than to visualize the hepatic perfusion pat-Hence, an estimation of hepatic supply
tern. With SPECT-CT as a highly accurate with chemotherapy agents over intraarterial
tool, obviously superior to SPECT alone port systems is difficult, if planar images
(Denecke et al., 2005), for identifica- or SPECT is used only, particularly in
tion of extrahepatic tracer accumulations patients with inhomogeneous hepatic tracer
and intrahepatic malperfusions, it is ide- accumulation. In these cases, the reviewer
ally suited to improve safety and efficacy extrapolates the imaging findings to a nor-
of liver directed administration of Y90- mal liver silhouette without morphologic
microspheres. correlation. However, this interpretation
may be unreliable. In this context, CT with
intraport injection of contrast agent has
DISCUSSION been reported to be superior compared to
scintigraphy including SPECT (Morimoto
Several imaging modalities have been et al., 1999). In assessing the intrahepatic
employed for control of intra-arterial port perfusion pattern, slow infusion MRA is
systems for HAIC with respect to patency, considered even superior to infusion CT
22. Perfusion Scintigraphy with Integrated Single Photon Emission 299

(Seki et al., 2003). In contrast to scintigra- Therefore, it is possible to accurately


phy, both techniques provide a high level assess the perfusion territory of intra-
of anatomic information, but depend on arterial port systems with SPECT-CT and
dynamic contrast media infusion, whereas Tc99m-MAA. This enables not only exclu-
Tc-99m-MAA enables static imaging of sion of extrahepatic accumulation but also
the perfusion pattern. In addition, contrast evaluation regarding catheter dislocation
revealed by scintigraphic imaging in dif- and occlusion of hepatic arteries. Indeed,
ferentiating perfused from non-perfused the impact on the therapeutic management
tissue is higher compared with CT and of 24 patients observed in a recent study
MRI. Thus, a combination of morphologi- was considered relevant (33%) (Denecke
cal and scintigraphically acquired func- et al., 2005). This was largely due to
tional information is warranted. subsequently dislocated port catheters
Correlation of SPECT images with sep- and extrahepatic perfusion which can be
arately generated CT images to assign treated by reintervention before initiating
perfusion patterns to anatomic structures the next cycle of chemotherapy. In another
using a simple side by side analysis is patient in whom chemotherapy application
not promising, especially when inhomo- was stopped because of abdominal pain,
geneous tracer distributions are present extrahepatic perfusion could be excluded
(Amthauer et al., 2005). For sufficient with SPECT-CT.
retrospective fusion, one would need pro- In addition, mapping of intrahepatic
spective planning of both examinations for tracer distribution with visualization of
optimal image fusion, despite the problems lobes and single segments is possible on
of breathing motion of the liver which are SPECT-CT images. When correlated to
hard to overcome. Additionally, the lack of the initial CT scan it can be estimated if
anatomic information in the SPECT data metastases are located in sufficiently sup-
set is problematic for fusion algorithms plied regions, or if reduced chemotherapy
based on mutual information (Amthauer supply has to be suggested. The latter was
et al., 2005). observed predominantly in patients with
To overcome these problems, a prom- abnormal anatomy of the hepatic arteries
ising approach is the combination of and consecutive problematic port place-
scintigraphic perfusion imaging with ment (Denecke et al., 2005). Whether a
morphologic correlation in one exami- reduced supply of segments with metas-
nation and inherent fusion of data sets. tases can be tolerated with the assump-
These requirements are ideally fulfilled tion of increasing intrahepatic arterial
by hybrid SPECT-CT systems. Breathing collateralization over time is not clear to
excursions of the liver are averaging dur- date. Further follow up and evaluation of
ing the relatively long acquisition times response rates are required.
of both SPECT (30 min) and low dose CT Regarding RE, in our opinion the
(9 min) (Amthauer et al., 2005; Denecke employment of SPECT-CT for perfusion
et al., 2005). The anatomic resolution of scintigraphy is decisive. The risk of gas-
the integrated low dose CT is sufficient trointestinal morbidity is more severe
for delineation of liver silhouette and than in HAIC. Once implanted, the pro-
neighboring organs (Denecke et al., 2005). gression of embolic and radiation effects
300 T. Denecke et al.

on the tissues hardly be stopped. For safety Cantore, M., Mambrini, A., Fiorentini, G., Rabbi, C.,
reasons, the best possible modality for Zamagni, D., Caudana, R., Pennucci, C.,
Sanguinetti, F., Lombardi, M., and Nicoli, N.
exclusion of inadvertent gastrointestinal
2005. Phase II study of hepatic intraarterial
implantation of diagnostic and therapeutic epirubicin and cisplatin, with systemic 5-fluor-
agents should be used, which is currently ouracil in patients with unresectable biliary tract
perfusion scintigraphy and Bremsstrahlung tumors. Cancer 103: 1402–1407.
scintigraphy with SPECT-CT. Cohen, A.D., and Kemeny, N.E. 2003. An update
In conclusion, SPECT-CT improves on hepatic arterial infusion chemotherapy for
colorectal cancer. Oncologist 8: 553–566.
identification and localization of areas
Dancey, J.E., Shepherd, F.A., Paul, K., Sniderman,
with reduced chemotherapy agent supply K.W., Houle, S., Gabrys, J., Hendler, A.L.,
and eventual extrahepatic perfusion in and Goin, J.E. 2000. Treatment of nonresect-
patients receiving HAIC. This has a rele- able hepatocellular carcinoma with intra-
vant impact on the therapeutic strategy and hepatic 90Y-microspheres. J. Nucl. Med. 41:
enables treatment optimization. In patients 1673–1681.
Denecke, T., Hildebrandt, B., Lehmkuhl, L., Peters, N.,
scheduled for RE with Y90-microspheres,
Nicolaou, A., Pech, M., Riess, H., Ricke, J.,
SPECT-CT is a very valuable tool to detect Felix, R., and Amthauer, H. 2005. Fusion imag-
or exclude inadvertent deposition of tracer ing using a hybrid SPECT-CT camera improves
into gastrointestinal non-target vessels, port perfusion scintigraphy for control of hepatic
and thus helps to increase the safety of this arterial infusion of chemotherapy in colorectal
procedure. cancer patients. Eur. J. Nucl. Med. Mol. Imaging
32(9): 1003–1010.
Gray, B., Van Hazel, G., Hope, M., Burton, M.,
REFERENCES
Moroz, P., Anderson, J., and Gebski, V. 2001.
Amthauer, H., Denecke, T., Rohlfing, T., Ruf, J., Randomised trial of SIR-Spheres plus chemo-
Bohmig, M., Gutberlet, M., Plockinger, U., therapy vs. chemotherapy alone for treating
Felix, R., and Lemke, A.J. 2005. Value of image patients with liver metastases from primary large
fusion using single photon emission computed bowel cancer. Ann. Oncol. 12: 1711–1720.
tomography with integrated low dose computed Heinrich, S., Petrowsky, H., Schwinnen, I., Staib-
tomography in comparison with a retrospective Sebler, E., Gog, C., El-Ganainy, A., Gutt, C.,
voxel-based method in neuroendocrine tumours. Muller, H.H., and Lorenz, M. 2003. Technical
Eur. Radiol. 15: 1456–1462. complications of continuous intra-arterial chem-
Andrews, J.C., Walker, S.C., Ackermann, R.J., otherapy with 5-fluorodeoxyuridine and 5-fluor-
Cotton, L.A., Ensminger, W.D., and Shapiro, B. ouracil for colorectal liver metastases. Surgery
1994. Hepatic radioembolization with yttrium-90 133: 40–48.
containing glass microspheres: preliminary Kemeny, N., Huang, Y., Cohen, A.M., Shi, W.,
results and clinical follow-up. J. Nucl. Med. 35: Conti, J.A., Brennan, M.F., Bertino, J.R.,
1637–1644. Turnbull, A.D., Sullivan, D., Stockman, J.,
Bocher, M., Balan, A., Krausz, Y., Shrem, Y., Blumgart, L.H., and Fong, Y. 1999. Hepatic
Lonn, A., Wilk, M., and Chisin, R. 2000. arterial infusion of chemotherapy after resection
Gamma camera-mounted anatomical X-ray of hepatic metastases from colorectal cancer. N.
tomography: technology, system characteris- Engl. J. Med. 341: 2039–2048.
tics and first images. Eur. J. Nucl. Med. 27: Kerr, D.J., McArdle, C.S., Ledermann, J., Taylor, I.,
619–627. Sherlock, D.J., Schlag, P.M., Buckels, J., Mayer,
Campbell, A.M., Bailey, I.H., and Burton, M.A. D., Cain, D., and Stephens, R.J. 2003. Medical
2001. Tumour dosimetry in human liver fol- Research Council’s colorectal cancer study group;
lowing hepatic yttrium-90 microsphere therapy. European Organisation for Research and Treatment
Phys. Med. Biol. 46: 487–498. of Cancer colorectal cancer study group.
22. Perfusion Scintigraphy with Integrated Single Photon Emission 301

Intrahepatic arterial versus intravenous fluor- 1999. Hepatic arterial infusion chemotherapy
ouracil and folinic acid for colorectal cancer for unresectable confined liver metastases: pre-
liver metastases: a multicentre randomised trial. diction of systemic toxicity with the applica-
Lancet 361: 368–373. tion of a scintigraphic and pharmacokinetic
Lehner, K., Reiser, M., Gebhardt, U., Heuck, A., approach. Cancer Chemother. Pharmacol. 44:
and Schaff, J. 1987. DSA – control of implanted 505–510.
devices for arterial hepatic perfusion. Cardiovasc. Ricke, J., Hildebrandt, B., Miersch, A., Nicolaou, A.,
Intervent. Radiol. 10: 71–74. Warschewske, G., Teichgraber, U., Lopez
Liu, D.M., Salem, R., Bui, J.T., Courtney, A., Hanninen, E., Riess, H., and Felix, R. 2004.
Barakat, O., Sergie, Z., Atassi, B., Barrett, K., Hepatic arterial port systems for treatment of
Gowland, P., Oman, B., Lewandowski, R.J., liver metastases: factors affecting patency and
Gates, V.L., Thurston, K.G., and Wong, C.Y. adverse events. J. Vasc. Interv. Radiol. 15:
2005. Angiographic considerations in patients 825–833.
undergoing liver-directed therapy. J. Vasc. Interv. Sadahiro, S., Suzuki, T., Ishikawa, K., Yasuda, S.,
Radiol. 16: 911–935. Tajima, T., Makuuchi, H., Saitoh, T., and
Lorenz, M., and Muller, H.H. 2000. Randomized, Murayama, C. 2004. Prophylactic hepatic arte-
multicenter trial of fluorouracil plus leucov- rial infusion chemotherapy for the prevention
orin administered either via hepatic arterial or of liver metastasis in patients with colon carci-
intravenous infusion versus fluorodeoxyurid- noma: a randomized control trial. Cancer 100:
ine administered via hepatic arterial infusion 590–597.
in patients with nonresectable liver metastases Seki, H., Ozaki, T., Takaki, S., Ooi, H., Oda, J.,
from colorectal carcinoma. J. Clin. Oncol. 18: and Shiina, M. 2003. Using slow-infusion MR
243–254. arteriography and an implantable port system
Lubin, E., Cyjon, A., and Neuman, M. 1987. to assess drug distribution at hepatic arterial
Technetium-99m macroaggregates for the study infusion chemotherapy. AJR Am. J. Roentgenol.
of the distribution of arterial infusion chemother- 180: 681–686.
apy in the liver. Clin. Nucl. Med. 12: 385–388. Suzuki, Y., Kobayashi, S., and Yasuda, S. 1991.
Meta-Analysis Group in Cancer (MAGC). 1996. Application of single-photon emission com-
Reappraisal of hepatic arterial infusion in the puted tomography (SPECT) with 99mTc-MAA
treatment of nonresectable liver metastases from in evaluation of perfusion patterns during hepatic
colorectal cancer. J. Natl. Cancer Inst. 88: infusion chemotherapy. Ann. Nucl. Med. 5:
252–258. 123–126.
Morimoto, M., Satake, M., Sekiguchi, R., Haruno, M., Tanaka, S., Shimada, M., Shirabe, K., Maehara, S.,
and Moriyama, N. 1999. Harimoto, N., Tsujita, E., Sugimachi, K.,
Optimal injection protocol for CT evaluation during and Maehara, Y. 2005. A novel intrahepatic
hepatic arterial infusion chemotherapy. Invest. arterial chemotherapy after radical resec-
Radiol. 34: 744–750. tion for advanced hepatocellular carcinoma.
Murthy, R., Nunez, R., Szklaruk, J., Erwin, W., Hepatogastroenterology 52: 862–865.
Madoff, D.C., Gupta, S., Ahrar, K., Wallace, Van Hazel, G., Blackwell, A., Anderson, J., Price, D.,
M.J., Cohen, A., Coldwell, D.M., Kennedy, A.S., Moroz, P., Bower, G., Cardaci, G., and Gray, B.
and Hicks, M.E. 2005. Yttrium-90 microsphere 2004 Randomised phase 2 trial of SIR-Spheres
therapy for hepatic malignancy: devices, indica- plus fluorouracil/leucovorin chemotherapy ver-
tions, technical considerations, and potential sus fluorouracil/leucovorin chemotherapy alone
complications. Radiographics 25(1): S41–S55. in advanced colorectal cancer. J. Surg. Oncol.
Nelson, R.L., and Freels, S. 2004. A systematic 88: 78–85.
review of hepatic artery chemotherapy after Ziessman, H.A., Gyves, J.W., Juni, J.E., Wahl,
hepatic resection of colorectal cancer metastatic F.L., Thrall, J.H., Ensminger, W.D., Goldstein,
to the liver. Dis. Colon Rectum. 47: 739–745. H.A., and Dubiansky, V. 1985. Atlas of hepatic
Pelosi, E., Bar, F., Battista, S., Bello, M., Bucchi, arterial perfusion scintigraphy. Clin. Nucl. Med.
M.C., Alabiso, O., Molino, G., and Bisi, G. 10: 675–681.
23
Postoperative Interferon Alpha
Treatment of Patients with Hepatocellular
Carcinoma: Expression of p48 Using
Tissue Microarray
Hui-Chuan Sun

INTRODUCTION C virus (HCV) infection (Lin et al., 2007;


Yu et al., 2006).
Hepatocellular carcinoma (HCC) is a major The consensus is that recurrence of HCC
health problem in China and the third most after resection may originate from intra-
common cause of cancer death in the world hepatic metastasis of the primary tumor,
(Parkin et al., 2005). Surgical resection or from de novo carcinogenesis in the
can have a curative outcome, but > 50% remnant liver (Sun et al., 2005). A number
of patients will have recurrence in the of randomized controlled studies have
first 5 years after resection (Poon, 2007), demonstrated the effect of postoperative
and the recurrence usually results in death IFN-α therapy on the prevention of tumor
(Regimbeau et al., 2004). No standard post- recurrence. Ikeda et al. (2000) and Lin
operative adjuvant therapy has yet been et al. (2004) concluded that the preventive
established (Sun and Tang, 2003). effect of IFN-α therapy can be attributed
In a previous study, we demonstrated to the clearance of HBV or HCV, result-
that interferon-α (IFN-α) delays tumor ing in inhibition of hepatocarcinogenesis.
growth and inhibits metastasis in a nude However, other investigators concluded
mouse model (Wang et al., 2000). Further that the direct antitumor effect of IFN-α
studies revealed that both antiangio- plays an important role in the prevention of
genesis and antiproliferative effects are tumor recurrence (Kubo et al., 2001; Sun
involved in the antitumor effect in the et al., 2006). Furthermore, our study did
HCC model (Wang et al., 2003; Wu et al., not find a relationship between conversion
2004, 2005). In addition to the direct of hepatitis B e antigen (HBeAg) and out-
antitumor effect, the antivirus effect may come of IFN-α therapy (Sun et al., 2006).
also be involved in the prevention of In our previous randomized control-
recurrence of HCC. Some reports have led trial, 28% of IFN-α-treated patients
indicated that long-term IFN-α reduces experienced recurrence within 1 year after
the incidence of HCC in patients with resection (Sun et al., 2006). Clearly, IFN-α
chronic hepatitis B virus (HBV) or hepatitis therapy was not effective in every patient.

303
304 H.-C. Sun

On the other hand, IFN-α therapy pro- surgical team at the Liver Cancer Institute
duced some side effects in patients who (Fudan University, Shanghai, China) were
did not respond therapeutically to IFN-α. enrolled to test the effect of postoperative
Therefore, a predictive marker of response IFN-α therapy on the prevention of recur-
to IFN-α is needed to avoid unnecessary rence. All patients were randomized into
treatment costs and side effects. either the IFN-α therapy group (5 MU three
A wealth of information is available times a week for 18 months or until recur-
on the molecular processes underlying rence was diagnosed) or the control group
IFN-α-induced signaling. Binding of (no antitumor treatment, until recurrence
IFN-α to the type I receptor brings the was diagnosed). The details of this trial
receptor-associated tyrosine kinases Tyk2 have been reported elsewhere (Sun et al.,
and Jak1 in close proximity, which allows 2006).
the kinases to phosphorylate, leading to In this study, 80 samples from the IFN-α
their activation and the generation of recep- group (group 1) and 75 samples from the
tor docking sites for the latent cytoplasmic control group (group 2) were retrieved from
protein STAT (Wu et al., 2004). Tyrosine- the Department of Pathology, Zhongshan
phosphorylated STAT1 and STAT2 also Hospital. All samples were from patients
associate as heterodimers to form IFN- who followed the treatment protocol.
α-stimulated gene factor 3 (ISGF3α) and
translocate to the nucleus, where they asso- Tissue Microarray and
ciate with the DNA-binding protein ISGF3γ Immunohistochemistry
(P48) to form ISGF3, which then binds After careful screening of hematoxylin-
to the IFN-stimulated response element eosin-stained slides for optimal tumor con-
to induce transcription of several IFN- tent, we constructed tissue microarray slides
inducible genes (Ghislain et al., 2001). Our in collaboration with Shanghai Biochip
previous study showed that transduction Co. Briefly, two 0.8-mm-diameter punch
of P48 in resistant HCC cells can restore cores were taken from each formalin-fixed,
sensitivity to IFN-α, a finding that further paraffin-embedded HCC sample. Samples
confirms the importance of Jak/STAT sig- were obtained from nonnecrotic tumor foci.
naling in the antiproliferative effects of Immunohistochemistry for P48 was per-
IFN-α in HCC cells (Wu et al., 2005). We formed using the avidin-biotin complex
believe that P48 expression in tumor cells (Vector Laboratories, Burlingame, CA),
is related to the effect of IFN-α. Therefore, including heat-induced antigen retrieval
we hypothesized that P48 expression would procedures. Incubation with polyclonal anti-
predict outcome in the patients receiving bodies against P48 (P48:C20, 1:100; Santa
IFN-α therapy who were enrolled in the Cruz Biotechnology, Santa Cruz, CA) was
randomized controlled trial presented here. carried out at 4°C for 18 h (Clifford et al.,
2000). Negative controls were treated identi-
cally but with the primary antibody omitted.
MATERIALS AND METHODS
In this randomized controlled trial, 232 Scoring of P48 Immunohistochemistry
patients who underwent curative resection Three pathologists who were blinded to
of HCC (pathologically proven) by the same patient data independently evaluated
23. Postoperative Interferon Alpha Treatment of Patients with Hepatocellular Carcinoma 305

P48 immunoreactivity. The percentage of tial prognostic significance, which were


positive tumor cells was determined by each all categorized as binary variables. These
observer, and the average of three scores included four clinical factors (age < or >
was calculated. We randomly selected 10 60 years, gender, HBeAg status, and serum
high-power fields (400 ×, 100 cells per α-fetoprotein level ≤ or > 20 ng/ml) and 9
high-power field); 1,000 cells were counted pathological factors (cirrhotic or noncir-
in each core. In this study, P48 expression rhotic liver, tumor size ≤ or > 5 cm, single
was graded as follows: tumors with posi- or multiple tumor nodules, degree of tumor
tive cytoplasm staining in < 20% of tumor encapsulation, Edmondson’s grade I/II or
cells were graded as negative, and tumors III/IV, presence or absence of microvascu-
with positive cytoplasm staining in ≥ 20% lar invasion, pTNM stage I/II or III, CLIP
of tumor cells were graded as positive. 0 or 1/2 (Yan and Yan, 2003), and P48
staining). Receiver operating characteristic
Statistical Analysis (ROC) analysis was used to assess the pre-
dictive accuracy of prognostic factors; we
Continuous variables were expressed as
used the free software developed by Metz at
mean ± standard deviation and were com-
the University of Chicago (http://xray.bsd.
pared between groups using the Student t
uchicago.edu/krl/roc_soft.htm). All statis-
test. Categorical variables were compared
tical analyses were performed using statistical
using the chi-square test. Survival curves
software (SPSS Inc., Chicago, IL). Statistical
were computed using the Kaplan-Meier
significance was defined as P < 0.05.
method and were compared between groups
using the log-rank test. To elucidate factors
that could have contributed to improved sur-
vival results, we performed an analysis to RESULTS
identify the prognostic factors for disease-
Clinicopathological Data
free survival (DFS) and overall survival.
Univariate and then multivariate analyses Positive P48 expression was found in 81
using the Cox proportional hazards model patients, and negative P48 expression was
were performed on 13 factors with poten- found in 74 patients (Figure 23.1A, B).

a b c

Figure 23.1. Immunohistochemistry of P48. a: Overview of tissue dot with positive staining of P48 in
hepatocellular carcinoma (avidin-biotin complex [ABC], 40×). b: Overview of tissue dot with negative
staining of 48 in hepatocellular carcinoma (40×). c: Positive staining of P48 in cytoplasm in hepatocellular
carcinoma (ABC, 400×)
306 H.-C. Sun

P48 was stained mainly in the cytoplasm group 2 (hazard ratio, 3.9; 95% CI, 1.7–9.2;
of HCC cells but occasionally in the P = 0.001). The mean overall survival was
nucleus of HCC cells and in the bile duct 57.3 months (95% CI, 53.0–61.5 months)
(Figure 23.1C). There was no statistically in group 1 and 41.5 months (95% CI,
significant difference in clinicopatholog- 34.0–49.0 months) in group 2, but the differ-
ical data, except gender, between patients ence was not statistically significant (hazard
with positive and those with negative P48 ratio, 1.5; 95% CI, 0.8–2.9; P = 0.246).
expression. There was no statistically
significant difference from patients who
received IFN-α (group 1) and those who Prognostic Factors for Disease-Free
did not (group 2) between patients with Survival and Overall Survival
positive and those with negative P48 in Group 1
expression. Univariate analysis of prognostic factors for
DFS revealed that of all evaluated factors,
only liver cirrhosis (P = 0.000) and P48
Survival
expression (P = 0.011) had a significant prog-
In group 1 (IFN-α therapy group), DFS nostic influence. After multivariate analysis,
was significantly better in patients with only liver cirrhosis (present vs. absent: risk
positive P48 expression compared with ratio [RR] 4.9; 95% CI, 1.4–16.9; P = 0.012)
those with negative P48 expression (haz- and pattern of P48 staining (negative vs. pos-
ard ratio, 1.8; 95% CI, 1.0–3.2; P = 0.036). itive: RR, 2.0; 95% CI, 1.1–3.8; P = 0.028)
The cumulative 1-, 3-, and 5-year DFS remained significant prognostic factors for
rates were 84%, 55%, and 36% among DFS . Receiver operating characteristic anal-
patients with positive P48 expression and ysis showed that these two prognostic factors
53%, 34%, and 29% among those with have good predictive accuracy with regard
negative P48 expression, respectively. In to 1-year DFS (area under the curve [AUC]
the control group, however, there was no = 0.649; 95% CI, 0.5–0.8; P < 0.01) and
significant difference in DFS between acceptable accuracy in terms of 2-year DFS
patients with positive and those with nega- (AUC = 0.604; 95% CI, 0.5–0.7; P = 0.05).
tive staining (hazard ratio, 1.4; 95% CI, Univariate analysis of prognostic fac-
0.8–2.6; P = 0.208). tors for overall survival revealed that of
In group 1, overall survival was signifi- all evaluated factors, liver cirrhosis (P =
cantly better in patients with positive P48 0.032), HBeAg positivity (P = 0.009),
expression than in those with negative P48 and P48 expression (P = 0.014) had a sig-
expression (hazard ratio, 2.7; 95% CI, nificant prognostic influence. After multi-
1.2–6.2; P = 0.014). Again, in group 2, variate analysis, only Edmondson’s stage
there was no significant difference in over- (high grade [III/IV] vs. low grade [I/II]:
all survival between patients with positive RR, 5.6; 95% CI, 2.0–15.7; P = 0.001),
and those with negative staining (hazard and pattern of P48 staining (negative vs.
ratio, 1.2; 95% CI, 0.6–2.3; P = 0.545). positive: RR, 3.8; 95% CI, 1.4–10.3; P =
When we analyzed data from patients 0.008) were significant prognostic factors
with positive P48 expression, we found that for overall survival. ROC analysis showed
overall survival in group 1 was better than in that these two prognostic factors have a
23. Postoperative Interferon Alpha Treatment of Patients with Hepatocellular Carcinoma 307

impressive predictive accuracy for overall also show that patients with poorly dif-
survival in the first 4 years (AUCs for 1-, ferentiated HCC and/or liver cirrhosis may
2-, 3-, and 4-year survival were 0.883 [P < have a poorer outcome. We noticed that the
0.01], 0.737 [P < 0.01], 0.646 [P = 0.01], predictive accuracy of P48 expression in
and 0.634 [P = 0.03], respectively). terms of DFS and overall survival dimin-
ished over time. The reason is that IFN-α
was administered for 18 months to patients
DISCUSSION in the treatment group, and P48 expression
was used to predict the effect of IFN-α
A gene expression profile and IFN-α/type therapy, so the predictive accuracy of P48
2 interferon receptor (IFNAR2) have been expression was good in the first several
reported to predict response of HCC to the years and diminished after IFN-α therapy
combination of IFN-α and 5-fluorouracil was stopped. As we found in our clinical
(Kurokawa et al., 2004; Ota et al., 2005). trial (Sun et al., 2006), 18 months of IFN-α
In melanoma and transitional cell carci- therapy delayed tumor recurrence but did
noma, a decrease in P48 expression was not reduce the incidence of recurrence over
associated with poor response to IFN-α 5 years; a number of patients developed
(Matin et al., 2001). Other molecular tumor recurrence after IFN-α therapy was
markers, such as SOCS-1 and SOCS-3, stopped. However, overall survival within
acted as negative feedback regulators of the 5-year period was improved by IFN-α
Jak/STAT and determined sensitivity to therapy, because of delayed recurrence in
IFN (Takayama et al., 2000). However, the treatment group.
until now there have been no reports on a Tissue microarray analysis was first
predictive marker for outcome after post- introduced by Kononen et al. in 1998. It
operative IFN-α therapy in patients with is a high-throughput approach for histo-
HBV-related HCC. logic examination of tissue for the pur-
In the present tissue microarray analy- pose of determining protein expression.
sis, we found no significant differences However, high-throughput platforms are
in DFS and in overall survival between compromised by a loss of completeness of
control subjects (patients not treated with data. Therefore, several important issues
postoperative IFN-α) with positive and need to be considered in conducting tissue
those with negative P48 expression; how- microarrays.
ever, significant differences were found First, adequate sample size is important.
between treated patients with positive Tumor heterogeneity is of major concern in
and those with negative P48 expression. interpreting results from tissue microarrays
Furthermore, overall survival in patients (Rubin et al., 2002). In well-established
with positive P48 expression who were methods of immunohistochemistry, about
treated with IFN-α was better than over- 0.05% of tissue in a routine section rep-
all survival in patients with positive P48 resents 1 cc of tumor tissue (Camp et al.,
expression who were not treated with IFN- 2000), whereas a tissue microarray section
α. These data clearly demonstrate that P48 represents a greatly reduced amount of tis-
expression is associated with outcome after sue. Obtaining more tissue microarray cores
postoperative IFN-α therapy. Our results should yield a more accurate result with
308 H.-C. Sun

less variability, but this is not always pos- Tissue microarray analysis is the best tool
sible, especially when the archived block for evaluating the clinical significance
has been repeatedly used or when a fine- of a specific gene expression in a larger
needle biopsy sample is used. Furthermore, number of specimens with different sub-
obtaining too many cores reduces the value types of tumor.
of tissue microarray analysis. Rubin et It is also notable that approximately
al. (2002) analyzed the results of a tis- 16% of patients with positive P48 expres-
sue microarray test for Ki67 expression sion received a diagnosis of recurrence
in prostate cancer and found that three within 1 year. One explanation for this
0.8-mm-diameter cores are necessary and may be that expression of P48 is not suf-
enough to obtain accurate results, but other ficient for antiproliferation in Jak/STAT
researchers have reported that one or two signaling. A previous study found that lack
cores are enough (Camp et al., 2000; Lugli of STAT1 expression also led to resistance
et al., 2004). Currently, there is no hard- to growth inhibitory effects of IFN-α in
and-fast guideline for determining optimal a liver cancer cell line (Murphy et al.,
sample size (Hewitt, 2006). In this study, 2001). In addition to antiproliferation,
two 0.8-mm-diameter cores were taken IFN-α has other antitumor mechanisms,
from each specimen. such as immunomodulation and antian-
Second, nonrepresentative samples should giogenesis (Jonasch and Haluska, 2001).
be avoided. The location of sampling sites Our previous study showed that the antian-
may play an important role. In this study, the giogenesis mechanism involved another
location of each core was decided in col- signal transduction pathway, despite P48
laboration with an experienced pathologist. deficiency in the HCC cell line (Wu et
The principle is to take cores from the non- al., 2005). Meanwhile, the natural limita-
necrotic portion of the sample and to collect tions of tissue microarray analysis, such as
as many intact tumor cells as possible. tumor heterogeneity, nonuniform tissue-
The third challenge is to establish a fixation processes, or even interpretation
scoring system for evaluation of P48 of immunoactivity, may also compromise
expression. In the present study, three predictive accuracy.
pathologists reviewed every slide and As pointed out by Kononen et al. (1998),
noted a mean of 348, 338, and 361 positive the tissue microarray test can be used with
tumor cells per patient. Standard devia- a large number of samples to profile more
tions were 355, 348 and 359, and standard general trends related to the tissue type
deviation did not increase as the corre- being studied; the test is not geared toward
sponding mean increased. determining the exact protein expression
An implication of this study is that tissue of an individual case. Tissue microarrays
microarray analysis could be used to deter- are designed to analyze a large number
mine the significance of gene expression of samples from archived paraffin blocks
in a larger sample size. Although we found but are not suitable for routine clinical
P48 expression to be associated with sen- analysis. For the routine clinical access of
sitivity of liver cancer cells to IFN-α, we P48 expression as a screening tool, a pro-
are not sure about the significance of P48 spective study with a standard section and
expression in patients who receive IFN-α. staining protocol is still needed.
23. Postoperative Interferon Alpha Treatment of Patients with Hepatocellular Carcinoma 309

In conclusion, the results of this study put molecular profiling of tumor specimens. Nat.
indicate that P48 assessed immunohisto- Med. 4: 844–847.
Kubo, S., Nishiguchi, S., Hirohashi, K., Tanaka, H.,
chemically from surgical samples may be
Shuto, T., Yamazaki, O., Shiomi, S., Tamori, A.,
a useful marker for predicting patient out- Oka, H., Igawa, S., Kuroki, T., and Kinoshita, H.
come after postoperative IFN-α therapy. 2001. Effects of long-term postoperative inter-
Postoperative IFN-α therapy should be feron-alpha therapy on intrahepatic recurrence
administered on the basis of tumor P48 after resection of hepatitis C virus-related hepa-
expression status, the presence of cirrhosis, tocellular carcinoma. A randomized, controlled
trial. Ann. Intern. Med. 134: 963–967.
and Edmondson’s grade, to provide maxi-
Kurokawa, Y., Matoba, R., Nagano, H., Sakon, M.,
mal benefit and avoid IFN-α toxicities. Takemasa, I., Nakamori, S., Dono, K., Umeshita, K.,
Ueno, N., Ishii, S., Kato, K., and Monden, M. 2004.
REFERENCES Molecular prediction of response to 5-fluorouracil
and interferon-alpha combination chemotherapy in
Camp, R.L., Charette, L.A., and Rimm, D.L. 2000. advanced hepatocellular carcinoma. Clin. Cancer
Validation of tissue microarray technology in Res. 10: 6029–6038.
breast carcinoma. Lab. Invest. 80: 1943–1949. Lin, S.M., Lin, C.J., Hsu, C.W., Tai, DI., Sheen,
Clifford, J.L., Menter, D.G., Yang, X., Walch, E., I.S., Lin, D.Y., and Liaw, Y.F. 2004. Prospective
Zou, C., Clayman, G.L., Schaefer, T.S., El-Naggar, randomized controlled study of interferon-alpha
A.K., Lotan, R., and Lippman, S.M. 2000. in preventing hepatocellular carcinoma recur-
Expression of protein mediators of type I inter- rence after medical ablation therapy for primary
feron signaling in human squamous cell carci- tumors. Cancer 100: 376–382.
noma of the skin. Cancer Epidemiol. Biomarkers Lin, S.M., Yu, M.L., Lee, C.M., Chien, R.N., Sheen,
Prev. 9: 993–997. I.S., Chu, C.M., and Liaw, Y.F. 2007. Interferon
Ghislain, J.J., Wong, T., Nguyen, M., and Fish, therapy in HBeAg positive chronic hepatitis
E.N. 2001. The interferon-inducible Stat2:Stat1 reduces progression to cirrhosis and hepatocel-
heterodimer preferentially binds in vitro to a con- lular carcinoma. J. Hepatol. 46: 45–52.
sensus element found in the promoters of a sub- Lugli, A., Tornillo, L., Mirlacher, M., Bundi, M.,
set of interferon-stimulated genes. J. Interferon Sauter, G., and Terracciano, L.M. 2004.
Cytokine Res. 21: 379–388. Hepatocyte paraffin 1 expression in human
Hewitt, S.M. 2006. The application of tissue normal and neoplastic tissues: tissue microarray
microarrays in the validation of microarray analysis on 3,940 tissue samples. Am. J. Clin.
results. Meth. Enzymol. 410: 400–415. Pathol. 122: 721–727.
Ikeda, K., Arase, Y., Saitoh, S., Kobayashi, M., Matin, S.F., Rackley, R.R., Sadhukhan, P.C., Kim,
Suzuki, Y., Suzuki, F., Tsubota, A., Chayama, K., M.S., Novick, A.C., and Bandyopadhyay, S.K.
Murashima, N., and Kumada, H. 2000. Interferon 2001. Impaired alpha-interferon signaling in
beta prevents recurrence of hepatocellular car- transitional cell carcinoma: lack of p48 expres-
cinoma after complete resection or ablation of sion in 5637 cells. Cancer Res. 61: 2261–2266.
the primary tumor – a prospective randomized Murphy, D., Detjen, K.M., Welzel, M., Wiedenmann, B.,
study of hepatitis C virus-related liver cancer. and Rosewicz, S. 2001. Interferon-alpha delays
Hepatology 32: 228–232. S-phase progression in human hepatocellular
Jonasch, E., and Haluska, F.G. 2001. Interferon carcinoma cells via inhibition of specific cyclin-
in oncological practice: review of interferon dependent kinases. Hepatology 33: 346–356.
biology, clinical applications, and toxicities. Ota, H., Nagano, H., Sakon, M., Eguchi, H, Kondo, M.,
Oncologist 6: 34–55. Yamamoto, T., Nakamura, M., Damdinsuren, B.,
Kononen, J., Bubendorf, L., Kallioniemi, A., Wada, H., Marubashi, S., Miyamoto, A., Dono, K.,
Bärlund, M., Schraml, P., Leighton, S., Torhorst, J., Umeshita, K., Nakamori, S., Wakasa, K., and
Mihatsch, M.J., Sauter, G., and Kallioniemi, Monden, M. 2005. Treatment of hepatocellular
O.P. 1998. Tissue microarrays for high-through- carcinoma with major portal vein thrombosis by
310 H.-C. Sun

combined therapy with subcutaneous interferon- Takayama, T., Sekine, T., Makuuchi, M., Yamasaki,
alpha and intra-arterial 5-fluorouracil; role of S., Kosuge, T., Yamamoto, J., Shimada, K.,
type 1 interferon receptor expression. Br. J. Sakamoto, M., Hirohashi, S., Ohashi, Y., and
Cancer 93: 557–564. Kakizoe, T. 2000. Adoptive immunotherapy to
Parkin, D.M., Bray, F., Ferlay, J., and Pisani, P. lower postsurgical recurrence rates of hepatocel-
2005. Global cancer statistics. 2002. CA Cancer lular carcinoma: a randomised trial. Lancet 356:
J. Clin. 55: 74–108. 802–807.
Poon, R.T. 2007. Optimal initial treatment for Wang, L., Tang, Z.Y., Qin, L.X., Wu, X.F., Sun,
early hepatocellular carcinoma in patients H.C., Xue, Q., and Ye, S.L. 2000. High-dose and
with preserved liver function: transplantation or long-term therapy with interferon-alfa inhibits
resection? Ann. Surg. Oncol. 14: 541–547. tumor growth and recurrence in nude mice bear-
Regimbeau, J.M., Abdalla, E.K., Vauthey, J.N., ing human hepatocellular carcinoma xenografts
Lauwers, G.Y., Durand, F., Nagorney, D.M., Ikai, I., with high metastatic potential. Hepatology 32:
Yamaoka, Y., and Belghiti, J. 2004. Risk factors 43–48.
for early death due to recurrence after liver resec- Wang, L., Wu, W.Z., Sun, H.C., Wu, X.F., Qin,
tion for hepatocellular carcinoma: results of a L.X., Liu, Y.K., Liu, K.D., and Tang, Z.Y. 2003.
multicenter study. J. Surg. Oncol. 85: 36–41. Mechanism of interferon alpha on inhibition of
Rubin, M.A., Dunn, R., Strawderman, M., and metastasis and angiogenesis of hepatocellular
Pienta, K.J. 2002. Tissue microarray sampling carcinoma after curative resection in nude mice.
strategy for prostate cancer biomarker analysis. J. Gastrointest. Surg. 7: 587–594.
Am. J. Surg. Pathol. 26: 312–319. Wu, W.Z., Sun, H.C., Gao, Y.Q., Li, Y., Wang, L.,
Sun, H.C., and Tang, Z.Y. 2003. Preventive treat- Zhou, K., Liu, K.D., Iliakis, G., and Tang, Z.Y.
ments for recurrence after curative resection of 2004. Reduction in p48-ISGFgamma levels con-
hepatocellular carcinoma – a literature review of fers resistance to interferon-alpha2a in MHCC97
randomized control trials. World J. Gastroenterol. cells. Oncology 67: 428–440.
9: 635–640. Wu, W.Z., Sun, H.C., Shen, Y.F., Chen, J., Wang, L.,
Sun, H.C., Tang, Z.Y., Ma, Z.C., Qin, L.X., Wang, L., Tang, Z.Y., Iliakis, G., and Liu, K.D. 2005.
Ye, Q.H., Fan, J., Wu, Z.Q., and Zhou, X.D. Interferon alpha 2a down-regulates VEGF
2005. The prognostic factor for outcome follow- expression through PI3 kinase and MAP kinase
ing second resection for intrahepatic recurrence signaling pathways. J. Cancer Res. Clin. Oncol.
of hepatocellular carcinoma with a hepatitis B 131: 169–178.
virus infection background. J. Cancer Res. Clin. Yan, P., and Yan, L.N. 2003. Staging of hepatocel-
Oncol. 131: 284–288. lular carcinoma. Hepatobiliary Pancreat. Dis.
Sun, H.C., Tang, Z.Y., Wang, L., Qin, L.X., Ma, Int. 2: 491–495.
Z.C., Ye, Q.H., Zhang, B.H., Qian, Y.B., Wu, Yu, M.L., Lin, S.M., Chuang, W.L., Dai, C.Y.,
Z.Q., Fan, J., Zhou, X.D., Zhou, J., Qiu, S.J., and Wang, J.H., Lu, S.N., Sheen, I.S., Chang, W.Y.,
Shen, Y.F. 2006. Postoperative interferon alpha Lee, C.M., and Liaw, Y.F. 2006. A sustained
treatment postponed recurrence and improved virological response to interferon or interferon/
overall survival in patients after curative resec- ribavirin reduces hepatocellular carcinoma and
tion of HBV-related hepatocellular carcinoma: improves survival in chronic hepatitis C: a
a randomized clinical trial. J. Cancer Res. Clin. nationwide, multicentre study in Taiwan. Antivir.
Oncol. 132: 458–465. Ther. 11: 985–994.
C. Prognosis
24
Hepatocellular Carcinoma:
Overexpression of Homeoprotein
Six1 as a Marker for Predicting Survival
Kevin Tak-Pan Ng and Kwan Man

INTRODUCTION Fan, 2004; Llovet et al., 2005). For bet-


ter stratification and management of HCC
Hepatocellular carcinoma (HCC) which patients, identification of potential mark-
occurs in 80% of all primary liver is the ers for accurate prognosis of HCC patients
fifth most common cancer and the third in terms of malignancy and survival rate is
most common cause of death from cancer indispensable.
in the world (Parkin et al., 2001; Befeler Molecular studies of hepatocarcinogen-
and Di Bisceglie, 2002). The incident esis have identified a number of molecular
rate of HCC, which is highest in Asia and biomarkers conferring prognostic signifi-
Africa, is increasing in Western countries. cance on prognosis and survival prediction
In the United States, 19,160 new cancers of HCC patients. One of the approaches is
of the liver and intrahepatic bile duct to identify genes specifically or excessively
were estimated in 2007, with an estimated expressed in HCC tissue as potential tumor
16,780 deaths (Jemal et al., 2007). Chronic markers. For instance, overexpression of
hepatitis B infection is the most common proto-oncogene c-met in HCC patients is
cause of HCC in China and Southeast Asia significantly associated with intrahepatic
(Muller, 2006), while hepatitis C infection metastasis and a short 5-year survival rate
is of high importance in Europe, Japan (Ueki et al., 1997). Survivin has been found
and North America (Bosch et al., 1999). to be associated with disease-free survival
Other common risk factors for hepatocar- and a higher rate of tumor recurrence
cinogenesis include alcohol, aflatoxin β1, (Fields et al., 2004; Ikeguchi et al., 2002).
cirrhosis, α-1-antitrypsin deficiency and Specific overexpression of Stat5b is sig-
hereditary hemochromatosis. Currently, nificantly correlated with poor disease-free
there is no standard treatment for advanced and overall survival of HCC patients (Lee
HCC. Surgical treatments, including hepa- et al., 2006). However, HCC is heterogenous
tectomy and liver transplantation, are and different etiological factors may lead
major curative options for HCC. However, to diverse molecular pathways involved
long-term survival rate remains unsat- in hepatocarcinogenesis. Current knowl-
isfactory due to tumor recurrence and edge on prediction of survival rate of
metastasis (Poon and Fan, 2004; Lo and HCC patients is still insufficient so that

313
314 K. Tak-Pan Ng and K. Man

continuous investigation of novel potential (Manassas, VA), and metastatic human


prognostic markers is urgently needed. HCC cell lines MHCC97L and MHCC97H
Vertebrate Six1 gene is a homolog of the were obtained from the Liver Cancer
Drosophila sine oculis (so) gene, which Institute and Zhongshan Hospital of Fudan
belongs to a subfamily of the Six class University, Shanghai, the People’s of
of homeodomain-containing transcription Republic of China (Li et al., 2001). All
factors characterized with a lysine resi- cells were cultured in DMEM high glucose
due within the DNA-binding helix of the medium with 10% FBS (Gibco, Grand
homeodomain (Oliver et al., 1995). The Island, NY) and 1% penicilium and strep-
functions of vertebrate Six1 are involved tomycin at humidified 37°C incubator sup-
in diverse early organ developments of plied with 5% CO2.
the brain, ear, eye, muscle and kidney
(Relaix and Buckingham, 1999; Laclef Clinical Samples
et al., 2003; Ozaki et al., 2004; Zheng et al.,
2003; Xu et al., 2003). Interestingly, Six1 One hundred and three HCC patients who
gene has been found to be deregulated in underwent liver resection between March
human cancers. Overexpression of Six1 1999 and June 2004 were recruited from
occurs in a large percentage of primary Department of Surgery, Queen Marry
breast cancers and strongly correlates with Hospital, The University of Hong Kong
metastatic breast lesions (Ford et al., 1998). contributed 103 pairs of protein extracts
Moreover, upregulation of Six1 in human and 72 pairs of RNA extracts from tumor
rhabdomyosarcoma (RMS) is significantly tissues and adjacent nontumor tissues in
correlated with progression and metastatic the study. There were 87 men and 16
potential (Yu et al., 2004). In this study, women. The age of the patients ranged
we provided the first evidence of Six1 from 33 to 75 years, with a median age of
expression in HCC patients after liver 55 years. Eighty-six HCC patients (83.5%)
resection to investigate its correlation with were positive for hepatitis B surface anti-
clinicopathological significance and sur- gen, whereas only three (2.9%) were posi-
vival rate of HCC patients. We use reverse tive for hepatitis C virus antibody. Twenty
transcription-polymerase chain reaction normal liver tissues were recruited from
(RT-PCR) and Western blot analysis to living donors at the same hospital. All
study both the mRNA and protein level of donors were examined to ensure that they
Six1 gene in HCC clinical cases aiming were free of liver diseases and hepatitis B
to identify a novel prognostic marker for infection. The follow-up duration for the
predicting survival of HCC patients. HCC patients ranged from July 1999 to
November 2005. The study was approved
by the Ethics Committee of the University
MATERIALS AND METHODS of Hong Kong.

Cell Lines Reverse Transcription-Polymerase


Nonmetastatic human HCC cell lines Chain Reaction
Hep3B, Huh7 and PLC were purchased Total RNA from liver tissues and cell
from the American Type Culture Collection lines were extracted by TRIZol regent
24. Hepatocellular Carcinoma: Overexpression of Homeoprotein Six1 315

according to the manufacturer’s instruc- TBS/T for 10 min each and incubated
tions (Invitrogen, Carlsbad, CA). Each with secondary antibody at room tempera-
complementary DNA was synthesized ture for 1 h. Protein signal was detected by
from 1 μg of RNA extract using the High ECL Plus system (Amersham Biosciences,
capacity cDNA Kit (Applied Biosystems, Piscataway, NJ). Antibodies Six1 and
Foster City, CA) under the condition of β-Actin were purchased from Santa Cruz
25°C for 5 min followed by 37°C for 2 h. Biotechnology (Santa Cruz, CA).
For clinical samples, PCR reaction for
Six1 was performed using the Taq PCR Statistical Analysis
Kit (Promega, Madison, WI) under the
following PCR cycles: 95°C for 5 min, Statistical analysis was carried out using
40 cycles at 95°C for 1 min, 57°C for SPSS 11.5 for Windows (SPSS Inc.,
1 min and 72°C for 1 min. For internal Chicago, IL). The association of Six1
control 18S, 30 PCR cycles were used. protein expression and clinicopathological
For cell line samples, 30-cycle multiplex parameters including sex, age, pTNM
PCR reaction was performed by combin- staging, venous infiltration, encapsulation,
ing Six1 and 18S primers. PCR products tumor size, alpha-fetoprotein level, and
were visualized by 2% agarose gel electro- hepatitis B surface antigen was analyzed
phoresis stained with ethidium bromide. by Chi-square test. The prognostic value
Primer sets used were as follows: for Six1, of Six1 protein for predicting the overall
sense 5′ AAG GAG AAG TCG AGG survival of HCC patients after hepatic
GGT GT-3′, antisense 5′-TGC TTG TTG resection was calculated by Kaplan-Meier
GAG GAG GAG TT-3′; for 18S ribosomal analysis with the log-rank test. Cox pro-
RNA, sense 5′-CTC TTA GCT GAG TGT portional hazard regression model was
CCC GC-3′, antisense 5′-CTG ATC GTC performed with univariable and multi-
TTC GAA CCT CC-3′. variable analyses to test factors that were
significantly associated with the overall
Western Blot survival of the HCC patients. Logistic
regression analysis was also performed
Proteins from clinical specimens and HCC
to compare those factors for predicting
cell lines were prepared by using Urea
1- and 5-year overall survival of the HCC
buffer (8M Urea, pH 8.0). The amount
patients. A P value of < 0.05 was consid-
of protein lysates used for Western blot
ered to be statistically significant.
analysis was 100 and 50 mg for clini-
cal HCC samples and HCC cell lines,
respectively. Protein extracts were sepa-
rated by 12% SDS-PAGE and transferred RESULTS
to PDMF membrane (Millipore, Billerica,
Six1 Expression in HCC Cell Lines
MA) according to the standard proto-
col. After blocking with 5% nonfat milk Three nonmetastatic HCC cell lines
at room temperature for 1 h, Six1 anti- (Hep3B, Huh7 and PLC) and two meta-
body with 1:1,000 dilution was hybridized static HCC cell lines (MHCC97L and
with the membrane at 4°C overnight. The MHCC97H) were checked for Six1 gene
membrane was washed three times with expression in terms of mRNA and protein
316 K. Tak-Pan Ng and K. Man

levels (Figure 24.1A). Using multiplex Hep3B, MHCC97L, and MHCC97H when
RT-PCR analysis, Six1 mRNA was compared with the mRNA level in Huh7
detected with a relatively higher level in and PLC. The metastatic HCC cell line

18S

18S

18S

Figure 24.1. (a) mRNA and protein expression analysis of Six1 gene among nonmetastatic (Huh7, 3B and
PLC) and metastatic (MHCC97L and MHCC97H) HCC cells. (b) mRNA and protein expression pattern of
Six1 gene in HCC patients. T, tumor tissue; NT, paired nontumor tissue. (c) mRNA and protein expression
analysis of Six1 genes in normal liver tissue. 1–8, liver tissues from different normal donors; C, positive
control. Ribosomal RNA 18S and β-actin were internal control for RT-PCR and Western blot
24. Hepatocellular Carcinoma: Overexpression of Homeoprotein Six1 317

MHCC97H with the highest metastatic Six1 protein overexpression (p = 0.000, r


potential expressed the highest level of = 0.438). All tumor tissues with positive
Six1 mRNA. Western blot analysis showed Six1 protein expression could match with
that Six1 protein was only expressed in the their positive mRNA expression except
metastatic cells and expressed the highest for one case.
level in MHCC97H. Reverse transcription-polymerase chain
reaction and Western blot were also used
Six1 Expression in HCC Patients to analyze the Six1 mRNA and protein
and Normal Donors expression patterns in normal liver tissues
(Figure 24.1C, Table 24.1). Most of the
Seventy-two pairs of tumor and non-
normal liver tissues did not show Six1
tumor liver tissues were subjected for
mRNA expression (90%), whereas only a
detection of Six1 mRNA by RT-PCR
few of them (2 of 20) showed weak posi-
(Figure 24.1B). A single-pair primer
tive signal. Moreover, Six1 protein was not
with 40-PCR-cycle amplification was
expressed in normal liver tissues.
employed because Six1 mRNA in clinical
samples could not be detected by using
multiplex PCR condition. Overexpression Six1 Protein Expression Correlated with
of Six1 mRNA was found in ∼ 85% (61 Advanced Tumor Stage
of 72) of tumor tissues compared with Six1 protein expression pattern in HCC
nontumor tissues (Table 24.1). Most of tumor tissues was compared with their
the nontumor liver tissues (91.7%) did clinicopathological features (Table 24.2).
not show Six1 mRNA expression. A few Overexpression of Six1 protein was sig-
nontumor tissues (6 of 72) expressed Six1 nificantly correlated with pTNM stage
mRNA, but all the expression levels were (p = 0.002, r = 0.312) and venous infiltra-
lower than those in the matched tumor tion (p = 0.004, r = 0.282). Six1 protein
tissues. Western blot analysis showed expression was detected in 71% (45 of 63)
that near 60% (61 of 103) of tumor tis- of advanced stage HCC patients and in
sues expressed Six1 protein while no 71% (42 of 59) HCC patients with venous
nontumor tissue expressed Six1 protein infiltration. No significant association was
(Figure 24.1B, Table 24.1). Using Chi- found between Six1 protein and sex, age,
squared test with Fisher’s exact test, Six1 cirrhosis, encapsulation, tumor size, alpha-
mRNA overexpression in HCC patients fetoprotein level of hepatitis B surface
was significantly correlated with their antigen.

Table 24.1. Summary of Six1 expression in liver tissues from HCC patients and
normal donors.
HCC patients
Six1 expression Tumor Nontumor Normal
RNA Positive 61 (84.7%) 6 (8.3%) 2 (10%)
Negative 11 (15.3%) 66 (91.7%) 18 (90%)
Protein Positive 61 (59.2%) 0 (0%) 0 (0%)
Negative 42 (40.8%) 103 (100%) 20 (100%)
318 K. Tak-Pan Ng and K. Man

Table 24.2. Correlation of Six1 protein expression and clinicopathological features of HCC patients.
Six1 protein expression (n)
Clinicopathological features Number (n) Negative Positive P
Sex
Male 87 35 52
Female 16 7 9 0.792
Age
≤55 years 59 25 34
>55 years 44 17 27 0.703
pTNM stage
Early stage (I–II) 40 24 16
Advanced stage (III–IV) 63 18 45 0.002*
Venous infiltration
Absent 44 25 19
Present 59 17 42 0.004*
Cirrhosisa
Absent 33 13 20
Present 69 29 40 0.800
Encapsulationa
Absent 48 22 26
Present 25 11 14 0.881
Tumor size
<5 cm 26 13 13
≥5 cm 77 29 48 0.268
AFP level
≤20 ng/ml 41 20 21
>20 ng/ml 62 22 40 0.179
Hepatitis B surface antigena
Negative 16 8 8
Positive 86 34 52 0.520

P
TNM = pathologic tumor-node-metastasis;
AFP = alpha-fetoprotein.
a
Total number less than 103 due to missing data.
*
Significant difference.

Six1 Protein Expression Correlated


with Poor Survival
Kaplan-Meier analysis was employed to
analyze the overall survival rate of HCC
patients in correspondence to the Six1 protein
expression pattern. The result showed that
HCC patients who overexpressed Six1
protein were significantly associated with
poor overall survival (Log rank = 4.12,
P = 0.0423, Figure 24.2). Kaplan-Meier
analysis of pTNM staging, venous infiltra-
tion and alpha-fetoprotein level were also
significantly associated with the overall
survival of HCC patients (data not shown). Figure 24.2. Kaplan-Meier overall survival curve
To further examine which factors were of HCC patients correlated with Six1 protein
the independent predictors, univariate expression
24. Hepatocellular Carcinoma: Overexpression of Homeoprotein Six1 319

and multivariate Cox proportional hazard the 1- and 5-year overall survival showed
regression analyses of these factors that Six1 protein was the most influential
corresponding to the overall survival of factor for predicting the 1-year overall sur-
HCC patients were performed. Univariate vival (OR = 5.405, 95% CI 1.427–20.474,
Cox proportional hazard regression analy- p = 0.013) while pTNM staging was the
sis showed that Six1 protein (HR = 1.956, most important factor for predicting the
95% CI 1.011–3.784, p = 0.046) and other 5-year overall survival (OR = 12.152, 95%
factors were significantly associated with CI 1.652–89.378, p = 0.004, Table 24.4).
the overall survival of HCC patients after
hepatic resection (Table 24.3). However,
multivariate analysis indicated that pTNM DISCUSSION
staging was the only independent factor
for predicting the overall survival of HCC Homeobox transcription factor Six1 located
patients (HR = 7.698, 95% CI 1.891–31.33, at 14q23 of the chromosome is involved in
p = 0.004, Table 24.3). Logistic regression the early development of many organs
analysis of these factors associated with including the brain, ear, eye, muscle and

Table 24.3. Cox proportional hazard regression analysis of Six1 protein expression and clinico-
pathological parameters in relation to the overall survival of HCC patients.
Univariate analysis Multivariate analysis
HR (95% CI) P HR (95% CI) P
Six1 protein
Positive vs. negative 1.956 (1.011–3.784) 0.046 1.29 (0.624–2.503) NS
pTNM stage
Advanced vs. early 4.952 (2.077–11.808) 0.000 7.698 (1.891–31.33) 0.004
Venous infiltration
Presence vs. absence 3.302 (1.572–6.934) 0.002 0.493 (0.136–1.780) NS
AFP level
> 20 ng/ml vs. ≤ 20 ng/ml 3.062 (1032–4.118) 0.04 1.735 (0.783–3.844) NS
HR = hazard ratio; CI = confidence interval; NS = not significant

Table 24.4. Logistic Regression Analysis of Six1 Protein and Clinicopathological Parameters on
Predicting the 1- and 5-year Overall Survival of HCC Patients.
1-year survival 5-year survival
OR (95% CI) P OR (95% CI) P
Six1 protein
Positive vs. negative 5.405 (1.427–20.474) 0.013 1.044 (0.401–2.719) NS
pTNM stage
Advanced vs. early 2.914 (0.245–34.63) NS 12.152 (1.652–89.378) 0.004
Venous infiltration
Presence vs. absence 1.557 (0.155–15.636) NS 0.478 (0.072–3.19) NS
AFP level
>20 ng/ml vs. <20 ng/ml 1.754 (0.554–5.552) NS 2.239 (0.846–5.929) NS
OR = odd ratio; CI = confidence interval; NS = not significant.
320 K. Tak-Pan Ng and K. Man

kidney (Relaix and Buckingham, 1999; expected to be tumor-specific as indicated


Laclef et al., 2003; Ozaki et al., 2004; by the evidence that no Six1 protein was
Zheng et al., 2003; Xu et al., 2003). expressed in both normal liver tissues and
Alteration of Six1 expression takes place HCC nontumor liver tissues (Table 24.1).
in human breast and Wilms’ cancer as well The tumor-specific characteristic of Six1 in
as rhabdomyosarcoma (RMS), indicating HCC may thus provide a useful implication
its possible contribution in the tumorigenic- for the therapeutic application of Six1 in
ity of different cancers (Ford et al., 1998; HCC by suppression strategy.
Yu et al., 2004; Li et al., 2002). Six1 is Overexpression of Six1 is potentially
overexpressed in 44% of primary tumors related to metastatic status of breast cancer
and 90% of metastatic tumors in breast and RMS (Ford et al., 1998; Yu et al., 2004).
cancer (Ford et al., 1998). Moreover, ele- MHCC97H and MHCC97L are human
vation of Six1 in human RMS patients is HCC cell lines with the same genetic back-
significantly associated with a later tumor ground but with different local and distant
stage (Yu et al., 2004). To investigate metastatic potentials (Li et al., 2001).
whether Six1 is also deregulated in HCC Orthotopic implantation of the MHCC97H
patients, both mRNA and protein levels cell into nude mice results in 100% of lung
of Six1 were examined in this study. In metastasis, whereas MHCC97L exhibits
agreement with other studies, we found 40% of lung metastasis (Li et al., 2001).
that overexpression of Six1 frequently Our data showed that Six1 protein was
occurred in tumor tissues of human HCC only expressed in metastatic HCC cell
patients at ~ 85% in mRNA level and 60% lines (MHCC97L and MHCC97H) but not
in protein level (Table 24.1). Although in nonmetastatic HCC cell lines (Hep3B,
overexpression of Six1 mRNA in tumor Huh7 and PLC), suggesting that Six1
tissues of HCC patients was significantly may be a metastasis-associated oncogene.
correlated with its positive Six1 protein Statistical analysis of the clinicopatho-
expression, the percentage of Six1 protein logical features of HCC patients revealed
overexpression in tumor tissues was lower that overexpression of Six1 protein was
than that of Six1 mRNA overexpression. significantly correlated with pTNM stag-
This phenomenon was also observed in ing (p = 0.002) and venous infiltration
HCC cell lines, in which Hep3B expressed (p = 0.004). Approximately, 71% of HCC
Six1 mRNA but not Six1 protein (Figure patients recognized at the advanced pTNM
24.1A). Furthermore, a small portion of stage (45 of 63) or with venous infiltra-
HCC nontumor liver tissues (8.3%) and tion (42 of 59) were found to overexpress
normal liver tissues (10%) showed posi- Six1 protein, suggesting that Six1 may
tive Six1 mRNA expression rather than play an important role in HCC progres-
protein expression (Table 24.1). All these sion and invasion. Although Six1 protein
data suggested that the post-transcriptional did not show significant association with
regulation of Six1 among HCCs may be tumor size (p = 0.268), > 60% (48 of 77)
different, and Six1 protein level, rather of HCC patients with tumors larger than
than mRNA level, is more likely to reflect 5 cm showed positive Six1 protein expres-
its real expression status. In addition, sion (Table 24.2), indicating its probable
expression of Six1 protein in liver is function in tumor progression.
24. Hepatocellular Carcinoma: Overexpression of Homeoprotein Six1 321

Hepatic resection is one of the major HCC patients may still provide a valuable
curative options for HCC patients. Different index for effective management of HCC
centers have reported achieving different patients after hepatectomy especially during
survival rates (Llovet et al., 2005). Identifi- early year.
cation of novel risk markers after hepa- Up to now, the understanding of the
tectomy is beneficial for the manage- mechanism of Six1 in pathogenesis of can-
ment of HCC patients and improvement of cers is still limited. Overexpression of Six1
their survival. Our results showed that in breast cancer cells can promote the can-
overexpression of Six1 protein in HCC cer cells to escape from the G2 cell cycle
patients was significantly associated with checkpoint after X-ray irradiation (Ford
poor overall survival after hepatectomy et al., 1998). The cell cycle regulatory
(p = 0.0423, Figure 24.2), suggesting that activity of Six1 in breast cancer is regu-
this protein may be a novel marker related lated by casein kinase II which inactivates
to poor overall survival of HCC patients Six1 through phosphorylation (Ford et al.,
after hepatectomy. 2000). Cyclin A1 is a downstream effector
Although multivariate Cox proportional for Six1 in breast cancer where over-
hazard regression analysis showed that expression of Six1 promotes cyclin A1
Six1 protein was not an independent factor expression and subsequently increases
for predicting the overall survival, logistic cell proliferation and progression (Coletta
regression analysis indicated that Six1 pro- et al., 2004). Gene amplification of
tein was the most influential factor asso- Six1 is a probable mechanism which
ciated with poor 1-year overall survival contributes to tumorigenesis in breast
(p = 0.013, Table 24.4) rather than 5-year cancer (Reichenberger et al., 2005).
overall survival (Table 24.4), proposing Overexpression of Six1 in RMS cells
that Six1 may be regarded as a predictor can boost their pulmonary metastasis
for short-term overall survival of HCC potential, whereas downregulation of
patients after hepatectomy. Hepatocellular Six1 suppresses their metastatic abil-
carcinoma patients with a higher rate of ity (Yu et al., 2004). Yu et al. (2006)
early-year death after hepatectomy may be also demonstrated that Six1 potentially
caused by higher malignancy of HCC that activates several oncogenes including
pathologically is, in part, determined to be cyclin D1, c-Myc and Ezrin. Moreover, the
at the advanced pTNM stage or the presence ability of Six1 to promote metastasis of
of venous infiltration. Overexpression of RMS requires the function of Ezrin. The
Six1 protein in HCC patients was deter- above research in breast cancer and RMS
mined to be significantly associated with provides important clues to clarify the
these malignant factors indicating the pos- functional roles of Six1 in HCC because
sible role of Six1 on promoting HCC different cancers may have different regu-
malignancy. Even though pTNM staging latory mechanisms. In fact, Cyclin A is
was determined to be the most important overexpressed in HCC and its overexpres-
factor in predicting long-term overall sur- sion is associated with poor survival of
vival of HCC patients after hepatectomy HCC patients (Chao et al., 1998; Ohashi
(Tables 24.3 and 24.4), assessment of Six1 et al., 2001). Casein kinase II shows
protein level of resected tumor tissues of an important involvement in transforming
322 K. Tak-Pan Ng and K. Man

growth factor-beta1-induced HCC (Gavin Fields, A.C., Cotsonis, G., Sexton, D., Santoianni, R.,
et al., 2003). The relationship between Six1 and Cohen, C. 2004. Survivin expression in
hepatocellular carcinoma: correlation with pro-
and Cyclins as well as casein kinase II in
liferation, prognostic parameters, and outcome.
regulating the tumorigenesis and metastasis Mod. Pathol. 17: 1378–1385.
of HCC thus needs further clarification. Ford, H.L., Kabingu, E.N., Bump, E.A., Mutter,
The specificity of Six1 protein expression G.L., and Pardee, A.B. 1998. Abrogation of the
in HCC metastatic cells may also provide a G2 cell cycle checkpoint associated with over-
good bridge to study the functions of Six1 expression of HSIX1: a possible mechanism
of breast carcinogenesis. Proc. Natl. Acad. Sci.
involved in metastasis of HCC through
USA 95: 12608–12613.
suppression strategy such as using antisense Ford, H.L., Landesman-Bollag, E., Dacwag, C.S.,
or RNA interference means. Stukenberg, P.T., Pardee, A.B., and Seldin, D.C.
In conclusion, our data indicated that 2000. Cell cycle-regulated phosphorylation of
Six1 protein was specifically and fre- the human SIX1 homeodomain protein. J. Biol.
quently expressed in HCC tumor tissues. Chem. 275: 22245–22254.
Gavin, L.G., Romieu-Mourez, R., Panta, G.R., Sun,
Overexpression of Six1 protein in HCC
J., Factor, V.M., Thorgeirsson, S.S., Sonenshein,
patients was significantly associated with G.E., and Arsura, M. 2003. Inhibition of CK2
advanced pTNM stage and venous infiltra- activity by TGF-beta1 promotes IkappaB-alpha
tion. In addition, Six1 protein could be a protein stabilization and apoptosis of immortal-
novel marker for predicting the short-term ized hepatocytes. Hepatology 38: 1540–1551.
overall survival of HCC patients after Ikeguchi, M., Ueda, T., Sakatani, T., Hirooka, Y.,
and Kaibara, N. 2002. Expression of survivin
hepatic resection. Further functional stud-
messenger RNA correlates with poor progno-
ies are worthwhile to explore the precise sis in patients with hepatocellular carcinoma.
mechanism and eventually develop potential Diagn. Mol. Pathol. 11: 33–40.
therapies targeting Six1 in liver cancer Jemal, A., Siegel, R., Ward, E., Murray, T., Xu, J.,
recurrence and metastases. and Thun, M.J. 2007. Cancer statistics 2007. CA.
Cancer J. Clin. 57: 43–66.
Laclef, C., Hamard, G., Demignon, J., Souil, E.,
REFERENCES Houbron, C., and Maire, P. 2003. Altered myo-
Befeler, A.S., and Di Bisceglie, A.M. 2002. genesis in Six1-deficient mice. Development
Hepatocellular carcinoma: diagnosis and treat- 130: 2239–2252.
ment. Gastroenterology 122: 1609–1619. Lee, T.K., Man, K., Poon, R.T., Lo, C.M., Yuen,
Bosch, F.X., Ribes, J., and Borras, J. 1999. A.P., Ng, I.O., Ng, K.T., Leonard, W., and Fan,
Epidemiology of primary liver cancer. Semin. S.T. 2006. Signal transducers and activators of
Liver Dis. 19: 271–285. transcription 5b activation enhances hepatocellu-
Chao, Y., Shih, Y.L., Chiu, J.H., Chau, G.Y., Lui, lar carcinoma aggressiveness through induction
W.Y., Yang, W.K., Lee, S.D., and Huang, T.S. of epithelial-mesenchymal transition. Cancer
1998. Overexpression of cyclin A but not Skp 2 Res. 66: 9948–9956.
correlates with the tumor relapse of human hepa- Li, C.M., Guo, M., Borczuk, A., Powell, C.A.,
tocellular carcinoma. Cancer Res. 58: 985–990. Wei, M., Thaker, H.M., Friedman, R., Klein, U.,
Coletta, R.D., Christensen, K., Reichenberger, Tycko, B. 2002. Gene expression in Wilms’
K.J., Lamb, J., Micomonaco, D., Huang, L., tumor mimics the earliest committed stage in the
Wolf, D.M., Muller-Tidow, C., Golub, T.R., metanephric mesenchymal-epithelial transition.
Kawakami, K., and Ford, H.L. 2004. The Six1 Am. J. Pathol. 160: 2181–2190.
homeoprotein stimulates tumorigenesis by reac- Li, Y., Tang, Z.Y., Ye, S.L., Liu, Y.K., Chen, J., Xue,
tivation of cyclin A1. Proc. Natl. Acad. Sci. USA Q., Chen, J., Gao, D.M., and Bao, W.H. 2001.
101: 6478–6483. Establishment of cell clones with different
24. Hepatocellular Carcinoma: Overexpression of Homeoprotein Six1 323

potential from the hepatocellular carcinoma and postoperative outcome. Liver Transpl. 10:
cell line MHCC97. World J. Gastroenterol. 7: S39–S45.
630–636. Reichenberger, K.J., Coletta, R.D., Schulte, A.P.,
Llovet, J.M., Schwartz, M., and Mazzaferro, V. 2005. Varella-Garcia, M., and Ford, H.L. 2005. Gene
Resection and liver transplantation for hepatocel- amplification is a mechanism of Six 1over-
lular carcinoma. Semin. Liver Dis. 25: 181–200. expression in breast cancer. Cancer Res. 65:
Lo, C.M., and Fan, S.T. 2004. Liver transplanta- 2668–2675.
tion for hepatocellular carcinoma. Br. J. Surg. Relaix, F., and Buckingham, M. 1999. From insect
91: 131–133. eye to vertebrate muscle: redeployment of a
Muller, C. 2006. Hepatocellular carcinoma – rising regulatory network. Genes Dev. 13: 3171–3178.
incidence, changing therapeutic strategies. Wien. Ueki, T., Fujimoto, J., Suzuki, T., Yamamoto, H.,
Med. Wochenshr. 156: 404–409. and Okamoto, E. 1997. Expression of hepatocyte
Ohashi, R., Gao, C., Miyazaki, M., Hamazaki, K., growth factor and its receptor c-met proto-onco-
Tsuji, T., Inoue, Y., Uenura, T., Hirai, R., gene in hepatocellular carcinoma. Hepatology
Shimizu, N., and Namba, M. 2001. Enhanced 25: 619–623.
expression of cyclin E and cyclin A in human Xu, P.X., Zheng, W., Huang, L., Maire, P., Laclef, C.,
hepatocellular carcinomas. Anticancer Res. 21: and Silvius, D. 2003. Six1 is required for
657–662. the early organogenesis of mammalian kidney.
Oliver, G., Wehr, R., Jenkins, N.A., Copeland, Development 130: 3085–3094.
N.G., Cheyette, B.N., Hartenstein, V., Zipursky, Yu, Y., Davicioni, E., Triche, T.J., and Merlino, G.
S.L., and Gruss, P. 1995. Homeobox genes and 2006. The homeoprotein six1 transcriptionally
connective tissue patterning. Development 121: activates multiple protumorigenic genes but
693–705. requires ezrin to promote metastasis. Cancer
Ozaki, H., Nakamura, K., Funahashi, J., Ikeda, Res. 66: 1982–1989.
K., Yamada, G., Tokano, H., Okamura, H.O., Yu, Y.L., Khan, J., Khanna, C., Helman, L.,
Kitamura, K., Muto, S., Kotaki, H., Sudo, K., Meltzer, P.S., and Merlino, G. 2004. Expression
Horai, R., Iwakura, Y., and Kawakami, K. 2004. profiling identifies the cytoskeletal organ-
Six1 controls patterning of the mouse otic vesi- izer ezrin and the developmental homeopro-
cle. Development 131: 551–562. tein Six-1 as key regulators. Nat. Med. 10:
Parkin, D.M., Bray, F., Ferlay, J., and Pisani, P. 2001. 175–181.
Estimating the world cancer burden: Globocan Zheng, W., Huang, L., Wei, Z.B., Silvius, D.,
2000. Int. J. Cancer 94: 153–156. Tang, B., and Xu, P.X. 2003. The role of Six1
Poon, R.T., and Fan, S.T. 2004. Hepatectomy in mammalian auditory system development.
for hepatocellular carcinoma: patient selection Development 130: 3989–4000.
25
Hepatocellular Carcinoma:
KiSS-1 Overexpression
as a Prognostic Factor
Katharina Schmid, Isabella Mosberger, and Fritz Wrba

INTRODUCTION a predominately hydrophilic, 164 amino


acid protein, serves as the natural ligand
Knowing about reliable prognostic markers of an orphan G-protein-coupled receptor,
is of great clinical importance to predict termed as AXOR12 (hOT7T175, human
relapse and dissemination of hepatocellular GPR54), a 398-amino acid protein encoded
carcinoma (HCC), one of the most common by the AXOR12 gene. In healthy individuals
malignant diseases worldwide. It is well the highest expression levels of KiSS-1
known that surgery serves as the only mRNA were observed in placenta, and in
curative therapy; by the end of the last testis, whereas moderate levels of KiSS-1
century, orthotopic liver transplantation mRNA were found in pancreas, liver, and
had become an optional treatment for a small intestine. For AXOR12 transcripts,
subgroup of patients with low tumor stage, the highest expression levels were seen
resulting in better prognosis with regard to in placenta and pancreas, with moderate
tumor recurrence (Island et al., 2005). expression in spleen, peripheral blood
Lee et al. (1996) reported for the first leukocyte, testis, and lymph nodes.
time that the KiSS-1 gene, mapped to During the following years, only a few
chromosome 1q32–q41, functions as a clinical data on KiSS-1 and AXOR12 gene
metastasis suppressor gene. This working expression in human malignancies were
group found that KiSS-1 mRNA expres- published. The majority reported that down-
sion occurred only in nonmetastatic, but regulation of KiSS-1 mRNA correlated with
not in metastatic melanoma. Lee and Welch tumor progression: Shirasaki et al. (2001)
(1997) also observed that the expression of found that KiSS-1 mRNA expression
KiSS-1 reduced the metastatic potential was lost in advanced stage melanomas.
of human breast carcinoma cells by 95%, Sanchez-Carbayo et al. (2003) described
while tumorigenicity was not affected. lower transcript levels of KiSS-1 in invasive
The study groups of Kotani et al. (2001), bladder carcinoma compared with superfi-
Muir et al. (2001), and Ohtaki et al. (2001) cial tumors and normal urothelium. Masui
reported that the product of KiSS-1 gene, et al. (2004) reported that pancreatic can-
termed as KiSS-1 (metastin, kisspeptin-54), cer showed significantly lower expression

325
326 K. Schmid et al.

levels of KiSS-1 mRNA than normal pan- (2003) reported that overexpression of
creas tissue. Ikeguchi et al. (2004) observed KiSS-1 correlated with tumor progression
that the loss of KiSS-1 gene expression in in HCC. Also, Martin et al. (2005) found
esophageal squamous carcinoma was a sig- that the expression of KiSS-1 increased in
nificant predictor of lymph node metastasis. patients with aggressive breast cancer.
According to Dhar et al. (2004), down- In order to investigate a possible correla-
regulation of KiSS-1 expression was the tion between KiSS-1 expression in HCC
strongest independent prognostic factor for and patient prognosis, we performed an
gastric carcinoma in multivariate analysis. immunohistochemical study: expression
Kostadima et al. (2007) found that the levels of the KiSS-1 gene product and its
KiSS-1 gene was silent in the vast majority cognate receptor AXOR12 were analysed
of resected node-positive breast carcino- in HCC specimens on the protein level,
mas, strongly supporting the antimetastatic and results were correlated with clinical
role of the gene. Conversely, Ikeguchi et al. follow-up (Figure 25.1).

a b

c d

Figure 25.1. Cases with high immunoreactivity of (a) KiSS-1 and (b) AXOR12 in HCC (both magnifi-
cation x200); (c) shows KiSS-1 (magnification x100) and (d, right side) AXOR12 immounoreactivity in
cirrhotic liver tissue, and (d, left side) negative immunostaining of tumor cells (magnification x200)
25. Hepatocellular Carcinoma: KiSS-1 Overexpression as a Prognostic Factor 327

MATERIALS 13. Levamisole (Sigma-Aldrich, St. Louis,


MO, USA)
1. SuperFrost Plus slides (silane coated) 14. Mayer’s Hematoxylin
2. Xylene 15. For mounting sections: Aquatex (Merck
3. Ethanol: anhydrous denaturated, 100%, Pharma, Darmstadt, Germany)
96%, 80%, and 70% 16. Placenta tissue slides for positive control
4. Distilled water (dH2O), physiologi-
cal saline (Merck Pharma, Darmstadt,
Germany)
5. Wash buffer: Tris-HCl-buffer stemsolu- METHODS
tion (TBS): Add 60.57 g Tris (Biomol,
Hamburg, Germany) to 500 ml dH2O This report presents a step-by-step proto-
and 400 ml HCl, adjust pH to 7.5, bring col for immunohistochemical staining of
to 1,000 ml with dH2O. Working solu- KiSS-1 and AXOR12 in HCC. Paying atten-
tion: add 900 ml physiological saline tion to correct tissue processing, including
to 100 ml stemsolution a reliable method and adequate duration of
6. Antigen retrieval citrate buffer: fixation, is essential for a good final result.
stemsolution A (10 mM citric acid): To choose the right tissue block containing
bring 21.06 g citric acid-1-hydrate to sufficient vital tumor tissue is indispensable
1,000 ml dH2O; stemsolution B: bring to obtaining reliable results. By incubating
29.41 g sodium citrate-dihydrate to the tissue sections with primary antibody
1,000 ml dH2O (both Merck Pharma, at 4°C for 20 h before storing them at room
Darmstadt, Germany). Working solu- temperature for another hour, a strong reac-
tion: 9 ml stemsolution A, and 41 ml tion of KiSS-1 in tumor cells will be found,
stemsolution B are brought to 500 ml and possible background staining is neg-
with dH2O ligible. Antibody binding was visualized
7. Blocking solution: Avidin/Biotin- with Supersensitive Alkaline Phosphatase
Blocking Kit (Vector, Burlingame, Complex. For visualizing the antibody bind-
CA, USA) ing the New Fuchsin Kit is applied, adding
8. Antibodies: KISS-1 and AXOR12, levamisole to block endogenous alkaline
both polyclonal rabbit antibodies phosphatase. Negative and positive controls
(Phoenix Pharmaceuticals, Belmont, should be used to ensure correct results:
CA, USA) for negative controls we recommend sub-
9. Antibody dilution: Albumine from stituting primary antibodies with irrelevant
Bovine Serum (BSA) (Sigma-Aldrich, rabbit serum, and for positive controls, pla-
St. Louis, MO, USA) centa tissue is of proper value.
10. Biotinylated secondary antibody: Goat
Anti-Rabbit IgG (Biocare Medical,
Tissue Preparation
San Diego, CA, USA)
11. Supersensitive Alkaline Phosphatase To obtain representative paraffin-embed-
Complex (Biocare Medical, San Diego, ded tissue blocks, tumor tissue pieces of
CA, USA) no more than 1 cm in diameter should be
12. New Fuchsin Kit (Nichirei Corporation, fixed in 7.5% formalin for 24 h before
Tokyo, Japan) embedding in paraffin.
328 K. Schmid et al.

Immunohistochemistry 9. Remove secondary antibody solution


1. Cut 4 μm thick sections of paraffin- and wash sections three times in TBS
embedded tumor tissue specimens, for 5 min each.
transfer the sections onto silane coated 10. Add 100–200 μl Supersensitive Alkaline
SuperFrost Plus slides, and dry the Phosphatase Complex (diluted 1:3 in
sections at room temperature. TBS) to each section and incubate for
2. Before deparaffinizing, heat the sections 30 min at room temperature.
for 30 min at 55°C to prevent detachment 11. Remove the complex and wash sections
of tissue from slides during the staining three times in TBS for 5 min each.
procedure. Deparaffinize and rehydrate 12. Visualize the antibody binding using
the sections by submerging the slides in the New Fuchsin Kit according to the
xylene (2 × 5 min), then in 100% ethanol manufacturer’s instructions: incubate
for 5 min twice, followed by rinsing the for at least 10 min and at a maximum
tissue sections in 96%, 80%, 70% ethanol of 30 min to obtain an optimal staining
and in dH2O. result (check under the microscope).
3. For antigen retrieval, apply autoclave 13. Wash sections three times in TBS for
pretreatment in sodium citrate buffer 5 min each.
(10 mM, pH 6.0) at 1 bar for 20 min; 14. Counterstain the sections in Mayer’s
afterwards cool slides for 40 min in the hematoxylin for 3 min.
autoclave. 15. Wash sections in tap water (10–20
4. Wash sections three times in TBS for times).
5 min each. 16. Mount sections using Aquatex.
5. To avoid unspecific staining, block
Evaluation of Immunohistochemical
each section with 100–200 μl blocking
Result
solution: first apply avidin solution for
15 min, rinse briefly with TBS, fol- Count immunopositive tumor cells in at
lowed by incubation with biotin solu- least ten consecutive high-power fields
tion for another 15 min as outlined by (× 400), and give number of positive cells
the manufacturer’s guideline. as a percentage (use a system of 10%
6. Remove blocking solution, and after steps, range 0–100%). Only samples that
washing in TBS, apply 100–200 μl exhibited 50% or more intensely stained
diluted primary antibody to each tumor cells should be classified as posi-
section (dilute KiSS-1 1:150 and tive (weakly stained samples and samples
AXOR12 1:400 in 1% BSA in TBS), showing less than 50% intensely stained
and incubate for 20 h (overnight) at tumor cells are considered negative).
4°C, followed by an additional hour
at room temperature.
7. Remove antibody solution and wash sec- RESULTS AND DISCUSSION
tions in TBS three times for 5 min each.
Patients and Tissue Samples
8. Apply 100–200 μl biotinylated goat
anti-rabbit IgG as secondary antibody This immunohistochemical study was
to each section, and incubate for 30 min undertaken to elucidate a possible link
at room temperature. between KiSS-1 and AXOR12 expression
25. Hepatocellular Carcinoma: KiSS-1 Overexpression as a Prognostic Factor 329

levels in HCC and patient prognosis (Schmid protein level by immunohistochemistry.


et al., 2007). All patients included in the study This difference might reflect an additional
underwent orthotopic liver transplantation as regulation of KiSS-1/AXOR12 expression
a curative therapeutic approach. Underlying at the translational level or by different
liver diseases were mostly alcoholic and cleavage variants of the KiSS-1 protein.
hepatitis C cirrhosis, less frequently hepatitis The KiSS-1 investigated in our study is
B cirrhosis, cryptogenic cirrhosis and other the major cleavage product, consisting of
rare causes of cirrhosis (hemochromatosis, 54 amino acids (kisspeptin-54). Kotani
primary biliary cirrhosis, primary sclerosing et al. (2001) also discovered two other
cholangitis) as well as few cases of non- naturally occurring smaller peptides in
cirrhotic liver. Before surgery was carried the human placenta, termed kisspeptin-14
out, a few patients received percutaneous, and kisspeptin-13. It was reported by Muir
endotumoral alcohol instillation, or were et al. (2001) that KiSS-1 the main cleavage
treated by transarterial chemoembolization product has greater stability than kisspep-
of the tumors. After liver transplantation, tin-14 and kisspeptin-13, which exhibit
adjuvant chemotherapy (doxorubicin) was stronger receptor binding activity.
administered to a subgroup of the patients In our study we also demonstrated by
(Pokorny et al., 2005). multivariate analysis that positive immuno-
The results of our study revealed a gen- reactivity of KiSS-1 in HCC is associated
erally diffuse, cytoplasmatic staining pat- with poorer clinical outcome concerning
tern of both, KiSS-1 and AXOR12 antigen disease-free (i.e., early local recurrence
showing a heterogenic reaction from focal of HCC, lymph node, and distant metas-
up to > 80% of tumors cells. Nonneoplastic tases) as well as overall survival, whereas
heptocytes in cirrhotic liver tissue also AXOR12 expression is not associated
expressed KiSS-1 and AXOR12 strongly with these parameters. Ikeguchi et al.
in a focal pattern; reactivity could be (2003) also reported a worse prognosis
observed mainly at the margin of regenera- for HCC patients with increased KiSS-1
tive nodules (Figure 25.1). The epithelium and AXOR12 mRNA levels. Martin et al.
of bile ducts and hepatocytes of normal (2005) described higher mRNA transcripts
liver tissue showed only weak KiSS-1 but in advanced stages of breast carcinoma.
no AXOR12 expression. No correlation On the contrary, the remainders of clini-
with underlying liver disease was seen. cal data on KiSS-1 and/or AXOR12 gene
Immunoreactivity of KiSS-1 and expression in human malignancies have
AXOR12 in HCC showed statistically shown a strong correlation between KiSS-1
significant correlation, which confirms and/or AXOR12 gene overexpression and
previous studies reporting a coincidence improved clinical course (Shirasaki et al.,
expression of KiSS-1 receptor and its 2001; Dhar et al., 2004).
cognate ligand in various tissues (Lee et Reasons why the expression of KiSS-1
al., 1996; Muir et al., 2001; Ohtaki et al., gene product in HCC and advanced stages
2001). Ikeguchi et al. (2003) investigated of breast cancer had a converse effect on
KiSS-1 and AXOR12 mRNA expression clinical course compared to other malig-
in HCC, and found a higher percentage of nant tumors remain to be clarified. Ikeguchi
mRNA transcripts in HCC as we did on et al. (2003) postulated that the estrogen
330 K. Schmid et al.

hormone might influence the expression REFERENCES


patterns of KiSS-1 gene products. Patients Chow, P.K., Tai, B.C., Tan, C.K., Machin, D., Win, K.M.,
with HCC generally suffer from liver cir- Johnson, P.J., and Soo, K.C. 2002. High-dose
rhosis, which leads to a hyperestrogenic tamoxifen in the treatment of inoperable hepa-
state. The fact that the KiSS-1/AXOR tocellular carcinoma: a multicenter randomized
system in HCC might be influenced by a controlled trial. Hepatology 36:1221–1226.
Dhar, D.K., Naora, H., Kubota, H., Maruyama, R.,
hormonal disorder is supported by the find-
Yoshimura, H., Tonomoto, Y., Tachibana, M.,
ings of Muir et al. (2001) and Horikoshi Ono, T., Otani, H., and Nagasue, N. 2004.
et al. (2003), who found significantly higher Downregulation of KiSS-1 expression is respon-
plasma levels of KiSS-1 in pregnant women sible for tumor invasion and worse prognosis in
than in nonpregnant women. Interestingly, gastric carcinoma. Int. J. Cancer 111:868–872.
in breast carcinoma, an association between Horikoshi, Y., Matsumoto, H., Takatsu, Y., Ohtaki,
T., Kitada, C., Usuki, S., and Fujino, M. 2003.
the increase of KiSS-1 and tumor progres-
Dramatic elevation of plasma metastin concen-
sion was found (Martin et al., 2005). It is trations in human pregnancy: metastin as a novel
well known that one of the most important placentaderived hormone in humans. J. Clin.
systemic therapies in breast cancer is the Endocrinol. Metab. 88:914–919.
blockade of the estrogen receptor path- Ikeguchi, M., Hirooka, Y., and Kaibara, N. 2003.
way. Although Chow et al. (2002) found Quantitative riverse transcriptase polymer-
ase chain reaction analysis for KiSS-1 and
that antiestrogenic therapy did not prolong
orphan G-protein-coupled receptor (hOT7T175)
survival in patients with inoperable HCC, gene expression in hepatocellular carcinoma.
further studies are warranted to investigate J. Cancer Res. Clin. Oncol. 129:531–535.
whether patients with overexpression of Ikeguchi, M., Yamaguchi, K., and Kaibara, N. 2004.
KiSS-1 in HCC improve their clinical out- Clinical significance of the loss of KiSS-1 and
come with antiestrogenic therapy. orphan G-protein-coupled receptor (hOT7T175)
gene expression in esophageal squamous cell
In conclusion, KiSS-1 and AXOR12
carcinoma. Clin. Cancer Res. 10:1379–1383.
are expressed in a subset of human HCC. Island, E.R., Pomposelli, J., Pomfret, E.A., Gordon,
High KiSS-1 expression is an independ- F.D., Lewis, W.D., and Jenkins, R.L. 2005.
ent prognostic factor for disease free and Twenty-year experience with liver transplanta-
overall survival in patients with HCC. tion for hepatocellular carcinoma. Arch. Surg.
Thus, KiSS-1 could serve as an addi- 140:353–358.
Kostadima, L., Pentheroudakis, G., and Pavlidis, N.
tional molecular marker for assessing
2007. The missing kiss of life: transcriptional
the malignant potential of HCC. Former activity of the metastasis suppressor gene KiSS1 in
reports have shown that estrogen levels early breast cancer. Anticancer Res. 27:2499–2504.
might modulate the expression patterns Kotani, M., Detheux, M., Vandenbogaerde, A.,
of KiSS-1 gene products; therefore, we Communi, D., Vanderwinden, J.M., Le Poul, E.,
speculate that patients with overexpression Brézillon, S., Tyldesley, R., Suarez-Huerta, N.,
Vandeput, F., Blanpain, C., Schiffmann, S.N.,
of KiSS-1 in HCC might benefit from a
Vassart, G., and Parmentier, M. 2001. The
antiestrogenic therapy. However, further metastasis suppressor gene KiSS-1 encodes
studies are needed to clarify a possible link kisspeptins, the natural ligands of the orphan G
of estrogen signalling and KiSS-1 expres- proteincoupled receptor GPR54. J. Biol. Chem.
sion in HCC tumors. 276:34631–34616.
25. Hepatocellular Carcinoma: KiSS-1 Overexpression as a Prognostic Factor 331

Lee, J.H., Miele, M.E., Hicks, D.J., Phillips, K.K., Takatsu, Y., Masuda, Y., Ishibashi, Y., Watanabe, T.,
Trent, J.M., Weissman, B.E., and Welch, D.R. Asada, M., Yamada, T., Suenaga, M., Kitada, C.,
1996. KiSS-1, a novel human malignant Usuki, S., Kurokawa, T., Onda, H., Nishimura, O.,
melanoma metastasis-suppressor gene. J. Natl. and Fujino, M. 2001. Metastasis suppressor
Cancer Inst. 88:1731–1737. gene KiSS-1 encodes peptide ligand of
Lee, J.H., and Welch, D.R. 1997. Suppression a G-proteincoupled receptor. Nature 411:
of metastasis in human breast carcinoma 613–617.
MDA-MB-435 cells after transfection with the Pokorny, H., Gnant, M., Rasoul-Rockenschaub, S.,
metastasis suppressor gene, KiSS-1. Cancer Gollackner, B., Steiner, B., Steger, G., Steininger,
Res. 57:2384–2387. R., and Muehlbacher, F. 2005. Does additional
Martin, T.A., Watkins, G., and Jiang, W.G. 2005. doxorubicin chemotherapy improve outcome in
KiSS-1 expression in human breast cancer. Clin. patients with hepatocellular carcinoma treated
Exp. Metastasis 22:503–511. by liver transplantation? Am. J. Transplant
Masui, T., Doi, R., Mori, T., Toyoda, E., Koizumi, M., 5:788–794.
Kami, K., Ito, D., Peiper, S.C., Broach, J.R., Sanchez-Carbayo, M., Capodieci, P., and Cordon-
Oishi, S., Niida, A., Fujii, N., and Imamura, M. Cardo, C. 2003. Tumor suppressor role of KiSS-1
2004. Metastin and its variant forms suppress in bladder cancer. Loss of KiSS-1 expression is
migration of pancreatic cancer cells. Biochem. associated with bladder cancer progression and
Biophys. Res. Commun. 315:85–92. clinical outcome. Am. J. Pathol. 162:609–617.
Muir, A.I., Chamberlain, L., Elshourbagy, N.A., Schmid, K., Wang, X., Sieghart, W., Peck-
Michalovich, D., Moore, D.J., Calamari, A., Radosavljevic, M., Bodingbauer, M., Rasoul-
Szekeres, P.G., Sarau, H.M., Chambers, J.K., Rockenschaub, S., and Wrba, F. 2007. KiSS-1
Murdock, P., Steplewski, K., Shabon, U., Miller, J.E., overexpression as an independent prognostic
Middleton, S.E., Darker, J.G., Larminie, C.G., marker in hepatocellular carcinoma: an immu-
Wilson, S., Bergsma, D.J., Emson, P., Faull, nohistochemical study. Virchows Arch. 450:
R., Philpott, K.L., and Harrison, D.C. 2001. 143–149.
AXOR12, a novel human G protein-coupled Shirasaki, F., Takata, M., Hatta, N., and Takehara,
receptor, activated by the peptide KiSS-1. K. 2001. Loss of expression of the metastasis
J. Biol. Chem. 276:28969–28975. suppressor gene KiSS-1 during melanoma pro-
Ohtaki, T., Shintani, Y., Honda, S., Matsumoto, H., gression and its association with LOH of chromo-
Hori, A., Kanehashi, K., Terao, Y., Kumano, S., some 6q16.3–q23. Cancer Res. 61:7422–7425.
26
Hepatocellular Carcinoma: Prognosis
Using Hepatoma-Derived Growth Factor
Immunohistochemistry
Hideji Nakamura, Kenya Yoshida, and Yasuhiko Tomita

HEPATOMA-DERIVED Erk phosphorylation in endothelial cells


GROWTH FACTOR (Everett et al., 2004).
Hepatoma-derived growth factor is a
Hepatoma-derived growth factor (HDGF) major member of the HDGF family of
is a heparin-binding protein purified from proteins which consists of HDGF and five
the conditioned medium of the human HDGF-related proteins (HRP) (Izumoto
well-differentiated hepatocellular carci- et al., 1997; Dietz et al., 2002). The
noma (HCC) cell line, HuH-7, which can N-terminal region of HDGF is highly con-
proliferate autonomously in a serum-free served among the other five HDGF-related
chemically-defined medium; this factor proteins (HRP), and this region is called
is highly expressed in several cancer cells hath (homologous to the amino terminus of
(Nakamura et al., 1989, 1994; Mori et al., HDGF) region, which contains the PWWP
2004). Hepatoma-derived growth factor is domain (Izumoto et al., 1997). The HDGF
an acidic 26 kDa protein consisting of 230 family members are characterized based on
amino acids with no hydrophobic signal whether they contain the hath region and
sequence in its N-terminus, and it has a nuclear localization signals (NLS) in their
high affinity to the glycosaminoglycans gene-specific regions and are targeting to
heparin and heparan sulphate (Nakamura et the nucleus (Nakamura and Hada, 2004).
al., 1994; Dietz et al., 2002). Exogenously Hepatoma-derived growth factor contains
supplied HDGF stimulates the prolifera- two nuclear localization signals (NLS) in
tion of fibroblasts, endothelial cells, vas- the molecule of HDGF; the first func-
cular smooth muscle cells, pulmonary tional nuclear localization signal (NLS1)
epithelial cells and hepatocytes, as well is in the hath region and the second NLS
as HCC, lung cancer, and colon cancer (NLS2) is in gene-specific regions of the
cells. A possible receptor-binding site is C-terminal region (Kishima et al., 2002a).
estimated to be residing at amino acid Hepatoma-derived growth factor can traf-
residues 81–100 within the hath region fic to the nucleus using these NLSs, espe-
(described below) (Abouzied et al., 2005). cially the NLS2 in its gene-specific region,
Indeed, exogenous HDGF stimulates the and this factor is dominantly localized in

333
334 H. Nakamura et al.

the nucleus rather than in the cytoplasm HDGF is highly expressed in oval cells,
(Everett et al., 2001). The gene-specific that are progenitor cells with bipotential
region of HDGF, at least the bipartite NLS activity for differentiating into hepatocytes
sequence and both the N-and C-terminal and bile duct cells. These findings sug-
neighboring portions, is essential for the gest that HDGF plays important roles in
mitogenic activity (Kishima et al., 2002a). the proliferation of immature hepatocytes
Thus, HDGF exerts its proliferating activity and hepatic progenitor cells including oval
via two different pathways: (1) via a putative cells, showing significant involvement of
plasma membrane-located HDGF receptor HDGF in liver development.
for which signaling depends on the hath In the fetus, HDGF was also abundantly
region, resulting in MAP kinase activation, expressed in other organs. Hepatoma-
and (2) via targeting to the nucleus by NLS. derived growth factor is highly expressed
in fetal rat aorta, conotruncus, and heart
Developmentally Regulated Expression of (Everett et al., 2000, 2001), and is widely
Hepatoma-Derived Growth Factor distributed in the renal anlage at the early
Cancers develop by the accumulation of stages of renal development and disap-
dysregulated gene expression from multistep peared from the adult kidney (Oliver and
genetic mutations of oncogenes and/or Al-Awqati, 1998). Hepatoma-derived
suppressor genes. These oncogenic proteins growth factor is highly expressed in the
and tumor suppressor proteins generally endothelial cells of non-muscularized
include growth factors, their receptors, forming blood vessels of the fetal lung
intracellular signal transduction molecules, (Everett et al., 2004), and its expression is
and transcriptional regulatory factors. enhanced dominantly in the bronchial and
Genes that are reduced significantly during alveolar epithelial cells including type II
organ development are frequently absent pneumocytes by bleomycin-instillation in
or little expressed in the adult tissues, mice (Mori et al., 2004). This growth fac-
and these developmentally regulated genes tor is also highly expressed in hind gut epi-
may be reactivated in human cancers. In thelia as well as atrial myocytes in the fetal
the fetus, HDGF is abundantly expressed stage. Furthermore, HDGF is expressed in
in various organs including liver. the nucleus of the colonic epithelial cells,
Hepatoma-derived growth factor is dominantly in the bottom of the intestinal
highly expressed in fetal liver of the mid- crypts by immunohistochemical analysis
gestation stage, and is markedly decreased (Nakamura et al., 2002). Thus, HDGF
near birth. The expression of this factor was is one of the developmentally regulated
significantly decreased with differentiation genes which are abundantly expressed in
in fetal hepatocytes induced by oncostatin cancer cells.
M treatment in in vitro primary culture dif-
ferentiation system (Enomoto et al., 2002). Role in Hepatocarcinogenesis
Adenoviral introduction of HDGF antisense The Fatty Liver Shionogi (FLS) mouse is
cDNA into the fetal hepatocytes suppresses an inbred strain that develops spontaneous
their proliferation, and the inhibitory effects fatty liver without obesity, and results in
of the HDGF antisense virus were recov- liver tumor development to ~ 45% in 52
ered by exogenous HDGF. Furthermore, weeks and 90% at 72 weeks after birth
26. Hepatocellular Carcinoma: Prognosis Using Hepatoma-Derived Growth Factor 335

in male mice; these tumors have been was dramatically increased in human colorec-
histologically diagnosed as hepatocellular tal cancers, especially in tumors proficient
adenoma and carcinoma. In the liver of in DNA mismatch repair (Lepourcelet et
FLS mice, HDGF expression has already al., 2005). Conversely, down regulation of
increased at an early stage before the HDGF by use of HDGF-siRNA had mini-
tumors developed microscopically in the mal effect on anchorage-dependent growth
liver (Yoshida et al., 2003). Hepatoma- but significantly reduced anchorage-
derived growth factor may be induced and independent growth of non-small cell lung
secreted or released from hepatocytes and cancer (NSCLC) cells in soft agar (Zhang
macrophages, enhancing the cell cycle et al., 2006). Thus, these findings suggest
progression of hepatocytes, inducing their that HDGF is an oncogenic protein, and
transformation, and promoting the pro- plays a role in cancer development.
liferation of HCC cells in an intracrine,
autocrine and/or paracrine manner. Role in Cancer Progression and
Furthermore, HDGF expression was Angiogenesis
strongly detected in an oval cell line, Hepatoma-derived growth factor protein
Oc 15–5, by immunohistochemistry, which is abundantly expressed in various human
is established from the liver of Long- cancers. In liver, HDGF expression is
Evans-Cinnamon rats. Hepatocellular car- higher in HCC than in the adjacent tissues
cinoma is considered to be developed (Yoshida et al., 2003, 2006). The HDGF-
from oval cells induced in the regen- over-expressing hepatoma cell line HepG2
eration process after liver injury. By dif- proliferates more rapidly and produces
ferential subtractive chain reaction from larger tumors, showing more rapid growth
strong anchorage-independent growth to in nude mice than neomycin-resistant cells
its negative HCC cells, HDGF is cloned in vivo. Recombinant HDGF stimulates
as one of the genes related to anchorage- the growth of cancer cells, while antisense
independency (Huang et al., 2004). These HDGF oligonucleotides or polyclonal anti-
findings strongly suggest that HDGF poten- HDGF antibody suppress their growth
tially participates in hepatocarcinogenesis (Kishima et al., 2002b; Nakamura et al.,
and in the early stage of HCC. 2002). Exogenously supplied HDGF pro-
Hepatoma-derived growth factor-over- motes the proliferation of bronchial squa-
expressing NIH3T3 cells generate sarco- mous cell carcinoma cell line, A549 cells
matous tumors in nude mice. In our study, (Mori et al., 2004). Knock-down expres-
HDGF-over-expressing NIH3T3 cells did sion of HDGF by small interfering RNA
not show significant anchorage-independ- (siRNA) in NSCLC cells significantly
ent growth in soft agar assay; however, shows slower growth, less colony for-
HDGF-over-expressing NIH3T3 cells mation in soft agar, and lesser in vitro
developed more small colonies in soft agar invasion activity (Zhang et al., 2006).
than parent or neomycin-resistant cells By proteomic differential display analysis
(Okuda et al., 2003). Hepatoma-derived and mass spectrometry, HDGF is down-
growth factor was more highly expressed regulated in regressive cancer cells as
in colon cancer cells than non-transformed compared with that in inflammatory cell-
intestinal epithelial cells, and its expression promoting progressive cells of the murine
336 H. Nakamura et al.

fibrosarcoma cell line (Hayashi et al., and through the induction of VEGF in the
2005). The higher expression of HDGF nucleus (Okuda et al., 2003).
shows more malignant potentials, suggest-
ing that HDGF is a candidate factor for IMMUNOHISTOCHEMICAL
cancer progression.
Hepatocellular carcinoma is a hyper-
AND ANALYTICAL
vascular tumor. Hepatoma-derived growth METHODS
factor is intrinsically related to angiogen-
Materials
esis and vasculogenesis and its protein
expression is induced in the regenerating 1. Rabbit polyclonal antibody raised
process of vascular vessels in wound repair against C-terminal amino acids (amino
(Everett et al., 2000). Additionally, HDGF acids 231–240) of the human HDGF.
is a candidate endothelial growth factor 2. Dulbecco’s phosphate-buffered saline
for renal glomerulus formation (Oliver and without magnesium and calcium – Tween
Al-Awqati, 1998). 20 (PBS-T): 200 mg potassium chloride,
Tumors developed from HDGF-over- 200 mg monobasic potassium phosphate,
expressing NIH3T3 cells inoculated 8 g sodium chloride, and 2.9 g dibasic
in nude mice are macroscopically red- sodium phosphate·12 H2O, bring volume
colored and histologically rich in vascu- to 11 ml with deionized distilled water, pH
lature, and immunohistochemical analysis 7.4. Add 0.1 ml Tween 20 and mix well.
by anti-CD31 antibody shows prominent 3. Fixative: 10% formalin in distilled water.
new vessel formation (Okuda et al., 2003). 4. 10 mM citric acid buffer (pH 6.0): 294.10 g
Hepatoma-derived growth factor protein trisodium citrate·2 H2O, bring volume to
stimulates the proliferation and tubule for- 11 ml with deionized distilled water to
mation of human umbilical vein endothe- prepare 1 M trisodium citrate. 210.14 g
lial cells and the migration of human citric acid·H2O, bring volume to 11 ml
pulmonary microvascular endothelial cells. with deionized distilled water to prepare
Using chick chorioallantoic membrane 1 M citric acid. Add 5 ml of 1 M trisodium
(CAM) assay, recombinant HDGF stimu- citrate and 0.8 ml of 1 M citric acid to
lates blood vessel formation, and stimu- 494.2 ml of deionized distilled water
lates cellular reorganization within the to prepare 10 mM citric acid, pH 6.0.
CAM from a loose network into a more 5. Methanol containing 0.2% hydrogen
compact, linear alignment reminiscent peroxide: 9 ml of 31% hydrogen perox-
of tube formation (Everett et al., 2004). ide in 150 ml methanol.
The more potent growth stimulating activ- 6. Blocking serum (goat serum) diluted at
ity of HDGF in vivo than in vitro must 1/50 in PBS-T.
be brought on by both the direct cell 7. Primary antibody (rabbit polyclonal
growth activity and its angiogenic activity. anti-HDGF antibody) diluted at 1/5,000
Furthermore, HDGF induces vascular in PBS-T.
endothelial growth factor (VEGF) gene 8. Secondary antibody (biotinylated goat
expression, and is a potent angiogenic fac- polyclonal anti-rabbit antibody) diluted
tor that functions by its own direct stimu- at 1/100 in PBS-T.
lation of the proliferation of endothelial 9. Avidin–biotin-complex solution: Add
cells and vascular smooth muscle cells 20 μl of VECTASTAIN Elite reagent
26. Hepatocellular Carcinoma: Prognosis Using Hepatoma-Derived Growth Factor 337

A and B to 1 ml PBS-T. Mix well and 13. Incubate the slides with secondary
let it ,stand for more than 30 min. antibody diluted in PBS-T for 30 min
10. 3,3 -Diaminobenzidine, tetrahydro- in a humidity chamber at 37°C.
chloride (DAB) solution: 75 mg of 14. Rinse the slide three times in PBS-T
DAB powder in 150 ml PBS-T. Stir for 5 min each.
well. Add 1 ml of 31% hydrogen per- 15. Incubate the slides with Avidin–
oxide to 150 ml of DAB solution. biotin-complex solution for 45 min in
a humidity chamber at 37°C.
16. Rinse the slide three times in PBS-T
Method
for 5 min.
1. Fix samples in 10% formalin, dehy- 17. Prepare DAB solution.
drated, and embedded in paraffin. 18. Immerse the slide in DAB solution for
2. Cut the paraffin block at 4 μm thick- 1 min.
ness and mount sections on poly-L- 19. Wash the slide in distilled water five
lysine-coated glass slides. times.
3. Deparaffinize the slide in xylene three 20. Immerse the slide in methyl green for
times for 15 min each, and in 100% 10 min.
ethanol three times for 5 min each. 21. Wash the slide in distilled water five
4. Immerse the slide in PBS-T. times.
5. For antigen retrieval, heat the slide 22. Dehydrate with 100% ethanol three
in 10 mM citrate buffer (pH 6.0) for times for 5 min each, and xylene three
15 min using microwave at 600 W. times for 5 min each, then mount with
6. Rinse the slide three times in PBS-T mounting medium.
for 5 min.
7. Immerse the slide in methanol con-
taining 0.2% hydrogen peroxide for
EVALUATION OF HEPATOMA-
20 min to block the endogenous per- DERIVED GROWTH
oxide activity at room temperature. FACTOR EXPRESSION
8. Rinse the slide three times in PBS-T IN HEPATOCELLULAR
for 10 min. CARCINOMA
9. Incubate the slides with 2% goat serum
in PBS-T for 30 min in a humidity Observe the slide under light microscope.
chamber at room temperature to block Bile ducts in the noncancerous lesions
the nonspecific binding of the primary uniformly show positive HDGF stain-
antibody. ing both for the nucleus and cytoplasm
10. Lightly tap off excess serum from each (Yoshida et al., 2006). Cancer cells showing
slide, and apply anti-HDGF primary staining similar to or stronger than the
antibody diluted in PBS-T. Incubate noncancerous bile ducts can be regarded
the slides overnight in a humidity as positive. Carefully examine the positivity
chamber at 4°C. ratio of all the cancer cells in the slide
11. Rinse the slide three times in PBS-T for both the nucleus and the cytoplasm.
for 5 min each. The HDGF labeling index (LI) can be
12. Prepare Avidin–biotin-complex solu- determined as follows: samples with more
tion. than 90% of cancer cells showing positive
338 H. Nakamura et al.

staining both for nucleus and cytoplasm nucleus and cytoplasm of HCC cells
were regarded as HDGF LI Level 2, and than in the adjacent normal hepatocytes
others as Level 1 (Figure 26.1). (Figure 26.2) (Yoshida et al., 2003, 2006).
Hepatocellular carcinomas with HDGF
LI Level 2 were regarded as having a
PROGNOSTIC SIGNIFICANCE positive HDGF index, and the remaining
OF HEPATOMA-DERIVED HCC with HDGF LI Level 1 as a nega-
GROWTH FACTOR IN tive HDGF index. Rate of positive HDGF
index was 27% in all HCC, and 29.9%
HEPATOCELLULAR among HCC with Edmondson’s differ-
CARCINOMA entiation grade I, II, and III. The HDGF
expression level by immunohistochemis-
Immunohistochemical analysis using try was significantly correlated to the dif-
specific anti-C terminus of HDGF anti- ferentiation of HCC, but it was not shown
body revealed that HDGF was more in other types of cancers. Hepatoma-
strongly and frequently expressed in the derived growth factor expression was

Figure 26.1. The immunohistochemical staining of hepatoma-derived growth factor. Representative


pictures of HDGF immunostaining for HDGF L1 level 1 and HDGF L1 level 2 are shown. In HDGF L1
level 1, as a negative HDGF index, most HCC cells do not express HDGF. In HDGF L1 level 2, as a positive
HDGF index, more than 90% of HCC cells show positive staining both for nucleus and cytoplasma
26. Hepatocellular Carcinoma: Prognosis Using Hepatoma-Derived Growth Factor 339

Figure 26.2. Hepatoma-derived growth factor immunohistochemistry in hepatocellular carcinoma cells


and normal hepatocytes. HDGF is more strongly and frequently expressed in the nucleus and cytoplasma
of HCC cells than in the adjacent normal hepatocytes, by using specific anti-C terminus of HDGF
antibody at a dilution of 1:5,000

higher in well-differentiated carcinomas HDGF expression in HCC showed an


than in poorly-differentiated carcinomas earlier recurrence and a poorer over-
in our study (Yoshida et al., 2006). In all survival rate than those with lower
contrast, HDGF was reported to be more expression after hepatectomy for HCC
highly expressed in poorly-differentiated (Hu et al., 2003; Yoshida et al., 2006).
HCC than in well-differentiated HCC In patients with differentiated HCC except
(Hu et al., 2003). One possible explana- for Edmondson’s grade IV, the prognostic
tion for the discrepancy between the two significance of the HDGF index was
groups may be due to the specificity of strong. By multivariate analysis, HDGF
the anti-HDGF antibody used for immu- expression was an independent prog-
nostaining. However, a more satisfactory nostic factor for disease-free and overall
explanation will be shown by a larger survival in patients who underwent a
scale study. Conversely, in both our and hepatectomy for HCC. These findings
their studies, the patients with higher suggest that HDGF is a candidate for use
340 H. Nakamura et al.

as a prognostic factor for disease-free and nostic marker for cancer patients. It is
overall survival of patients with HCC. expected in the future that any tool regu-
Besides HCC, HDGF is a prognsostic lating HDGF expression or HDGF signal
factor of pancreatic ductal cancer, lung pathways may be a useful candidate for
cancer, gastric cancer, and esophageal the suppression of carcinogenesis and can-
cancer. In patients with HCC or early cer progression.
stage NSCLC who underwent curative sur-
gery, the disease-free and disease-specific REFERENCES
survival and overall survival were lower
Abouzied, M.M., El-Tahir, H.M., Prenner, L.,
in those with higher HDGF expression Haberlein, H., Gieselmann, V., and Franken, S.
indexes than in those with lower HDGF 2005. Hepatoma-derived growth factor.
indexes (Ren et al., 2004; Iwasaki et al., Significance of amino acid residues 81–100 in
2005). The high expression of HDGF cell surface interaction and proliferative activity.
showed distant metastasis and shortened J. Biol. Chem. 280: 10945–10954.
survival time in patients with NSCLC. In Dietz, F., Franken, S., Yoshida, K., Nakamura, H.,
Kappler, J., and Gieselmann, V. 2002. The family of
gastric cancer, the patients with abundant hepatoma-derived growth factor proteins:
expression of HDGF showed significantly characterization of a new member HRP-4 and
higher rates of infiltrative tumor growth classification of its subfamilies. Biochem. J.
and vascular and lymphatic invasion, as 366: 491–500.
compared to those with lower expression Enomoto, H., Yoshida, K., Kishima, Y., Kinoshita, T.,
(Yamamoto et al., 2006). Pancreatic ductal Yamamoto, M., Everett, A.D., Miyajima, A., and
Nakamura, H. 2002. Hepatoma-derived growth
cancers with higher HDGF expression in factor is highly expressed in developing liver
the nucleus showed a poorer 5-year sur- and promotes fetal hepatocyte proliferation.
vival rate than those with lower expression Hepatology 36: 1519–1527.
(Uyama et al., 2006). These clinical find- Everett, A.D., Lobe, D.R., Matsumura, M.E.,
ings and in vitro data of pleiotropic HDGF Nakamura, H., and McNamara, C.A. 2000.
activities described above suggest that the Hepatoma-derived growth factor stimulates smooth
muscle cell growth and is expressed in vascular
higher expression of HDGF shows more development. J. Clin. Invest. 105: 567–575.
malignant potential including invasive and Everett, A.D., Stoops, T., and McNamara, C.A.
metastatic activity. 2001. Nuclear targeting is required for hepato-
In conclusion, HDGF is a unique nuclear ma-derived growth factor-stimulated mitogen-
targeting growth factor, which is expressed esis in vascular smooth muscle cells. J. Biol.
abundantly in cancer cells, promotes their Chem. 276: 37564–37568.
Everett, A.D., Narron, J.V., Stoops, T., Nakamura, H.,
malignant potential, and generates tumors and Tucker, A. 2004. Hepatoma-derived growth
and promotes their growth in vivo via its factor is a pulmonary endothelial cell-expressed
mitogenic activity and angiogenic activity angiogenic factor. Am. J. Physiol., Lung Cell.
deriving from both its own direct ang- Mol. Physiol. 286: L1194–1201.
iogenic activity and the induction of VEGF. Hayashi, E., Kuramatsu, Y., Okada, F., Fujimoto, M.,
Multivariate analysis of the relationship Zhang, X., Kobayashi, M., Iizuka, N., Ueyama,
Y., and Nakamura, K. 2005. Proteomic profil-
of HDGF expression and recurrence-free ing for cancer progression: differential display
and overall survival in patients with HCC analysis for the expression of intracellular pro-
confirmed that HDGF has the potential to teins between regressive and progressive cancer
become a significantly efficacious prog- cell lines. Proteomics 5: 1024–1032.
26. Hepatocellular Carcinoma: Prognosis Using Hepatoma-Derived Growth Factor 341

Hu, T.H., Huang, C.C., Liu, L.F., Lin, S.Y., epithelial growth. Am. J. Respir. Cell Mol. Biol.
Chang, H.W., Changchien, C.S., Lee, C.M., 30: 459–469.
Chuang, J.H., and Tai, M.H. 2003. Expression Nakamura, H., Kambe, H., Egawa, T., Kimura,
of hepatoma-derived growth factor in hepatocel- Y., Ito, H., Hayashi, E., Yamamoto, H., Sato, J.,
lular carcinoma. Cancer 98: 1444–1456. and Kishimoto, S. 1989. Partial purification and
Huang, J.S., Chao, C.C., Su, T.L., Yeh, S.H., Chen, characterization of hepatoma-derived growth
D.S., Chen, C.T., Chen, P.J., and Jou, Y.S. 2004. factor. Clin. Chim. Acta 183: 273–284.
Diverse cellular transformation capability of Nakamura, H., Izumoto, Y., Kambe, H., Kuroda,
overexpressed genes in human hepatocellular T., Mori, T., Kawamura, K., Yamamoto, H.,
carcinoma. Biochem. Biophys. Res. Commun. and Kishimoto, T. 1994. Molecular cloning
315: 950–958. of complementary DNA for a novel human
Iwasaki, T., Nakagawa, K., Nakamura, H., hepatoma-derived growth factor. J. Biol. Chem.
Takada, T., Matsui, K., and Kawahara, K. 2005. 269: 25143–25149.
Hepatoma-derived growth factor as a prognostic Nakamura, H., Yoshida, K., Ikegame, K., Kishima,
marker in completely resected non-small-cell Y., Uyama, H., and Enomoto, H. 2002.
lung cancer. Oncol. Rep. 13: 1075–1080. Antibodies against hepatoma-derived growth
Izumoto, Y., Kuroda, T., Harada, H., Kishimoto, T., and factor and mucosal repair in ulcerative colitis.
Nakamura, H. 1997. Hepatoma-derived growth J. Gastroenterol. 37 (Suppl. 14): 8–14.
factor belongs to a gene family in mice showing Nakamura, H., and Hada, T. 2004. Hepatoma-
significant homology in the amino terminus. derived growth factor in ontogeny and tumori-
Biochem. Biophys. Res. Commun. 238: 26–32. genesis. Recent Res. Devel. Biophys. Biochem.
Kishima, Y., Yamamoto, H., Izumoto, Y., Yoshida, K., 4: 17–27.
Enomoto, H., Yamamoto, M., Kuroda, T., Ito, H., Okuda, Y., Nakamura, H., Yoshida, K., Enomoto, H.,
Yoshizaki, K., and Nakamura, H. 2002a. Uyama, H., Hirotani, T., Funamoto, M., Ito,
Hepatoma-derived growth factor stimulates cell H., Everett, A.D., Hada, T., and Kawase, I.
growth after translocation to the nucleus by 2003. Hepatoma-derived growth factor induces
nuclear localization signals. J. Biol. Chem. 277: tumorigenesis in vivo through both direct
10315–10322. angiogenic activity and induction of vascular
Kishima, Y., Yoshida, K., Enomoto, H., endothelial growth factor. Cancer Sci. 94:
Yamamoto, M., Kuroda, T., Okuda, Y., Uyama, 1034–1041.
H., and Nakamura, H. 2002b. Antisense oli- Oliver, J.A., and Al-Awqati, Q. 1998. An endothe-
gonucleotides of hepatoma-derived growth lial growth factor involved in rat renal develop-
factor (HDGF) suppress the proliferation of ment. J. Clin. Invest. 102: 1208–1219.
hepatoma cells. Hepatogastroenterology 49: Ren, H., Tang, X., Lee, J.J., Feng, L., Everett, A.D.,
1639–1644. Hong, W.K., Khuri, F.R., and Mao, L. 2004.
Lepourcelet, M., Tou, L., Cai L., Sawada, J., Expression of hepatoma-derived growth factor
Lazar, A.J.F., Glickman, J.N., Williamson, is a strong prognostic predictor for patients with
J.A., Everett, A.D., Redston, M., Fox, E.A., early-stage non-small cell lung cancer. J. Clin.
Nakatani, Y., and Shivdasani, R.A. 2005. Oncol. 22: 3230–3237.
Insights into development mechanisms and can- Uyama, H., Tomita, Y., Nakamura, H., Nakamori, S.,
cers in the mammalian intesitine derived from Zhang, B., Hoshida, Y., Enomoto, H., Okuda, Y.,
serial analysis of gene expression and study of Sakon, M., Aozasa, K., Kawase, I., Hayashi, N.,
the hepatoma-derived growth factor (HDGF). and Monden, M. 2006. Hepatoma-derived
Development 132: 415–427. growth factor is a novel prognostic factor for
Mori, M., Morishita, H., Nakamura, H., Matsuoka, H., patients with pancreatic cancer. Clin. Cancer
Yoshida, K., Kishima, Y., Zhou, Z., Kida, H., Res. 12: 6043–6048.
Funakoshi, T., Goya, S., Yoshida, M., Kumagai, Yamamoto, S., Yasuhiko, T., Hoshida, Y., Takiguchi,
T., Tachibana, I., Yamamoto, Y., Kawase, I., and S., Fujiwara, Y., Yasuda, T., Doki, Y., Yoshida, K.,
Hayashi, S. 2004. Hepatoma-derived growth fac- Aozasa, K., Nakamura, H., and Monden, M.
tor is involved in lung remodeling by stimulating 2006. Expression of hepatoma-derived growth
342 H. Nakamura et al.

factor is correlated with lymph node metastasis and Aozasa, S., Nagano, H., Sakon, M., Kawase, I.,
prognosis of gastric carcinoma. Clin. Cancer Monden, M., and Nakamura, H. 2006. Hepatoma-
Res. 12: 117–122. derived growth factor is a novel prognostic fac-
Yoshida, K., Nakamura, H., Okuda, Y., Enomoto, tor for hepatocellular carcinoma. Ann. Surg.
H., Kishima, Y., Uyama, H., Ito, H., Hirasawa, Oncol. 13: 159–167.
T., Inagaki, S., and Kawase, I. 2003. Expression Zhang, J., Ren, H., Yuan, P., Lang, W., Zhang, L.,
of hepatoma-derived growth factor in hepato- and Mao, L. 2006. Down-regulation of
carcinogenesis. J. Gastroenterol. Hepatol. 18: hepatoma-derived growth factor inhibits
1293–1301. anchorage-independent growth and invasion
Yoshida, K., Tomita, T., Okuda, Y., Yamamoto, S., of non-small cell lung cancer cells. Cancer
Enomoto, H., Uyama, H., Ito, H., Hoshida, Y., Res. 66: 18–23.
27
Hepatitis C Virus-Related Human
Hepatocellular Carcinoma: Predictive
Markers Using Proteomic Analysis
(Methodology)
Yasuhiro Kuramitsu and Kazuyuki Nakamura

HEPATITIS C VIRUS-RELATED Therefore, it is important to identify the


HUMAN HEPATOCELLULAR predictive marker proteins by which we
CARCINOMA (HCV–HCC) can predict the carcinogenesis of HCC in
HCV-infected patients.
It was reported by El-Serag and Mason In a recent study, we analyzed proteome
(1999) that hepatocellular carcinoma in HCV–HCC tissues and identified many
(HCC) is one of the most common fatal proteins whose expression in cancer tis-
cancers world-wide. From the repots by sues were different from corresponding
Okuda (1992, 2000) and Seow et al. noncancerous tissues by using two-dimen-
(2001), the most apparent risk factor for sional gel electrophoresis (2-DE) and
HCC is thought to be the chronic infec- mass spectrometry (MS). Furthermore, we
tion with hepatitis B or C virus (HBV identified the autoantibodies recognizing
or HCV). Although in the other Asian HSP70.1, Mn-SOD or peroxiredoxin from
countries HCCs developed from HBV- the sera of HCC-patients.
infection, the characteristic of HCCs in
Japan is that they develop from the liver
tissues infected with HCV or of liver cir- PROTEOMICS
rhosis. From the reports by Yano et al.
(2003) and Yatsuhashi (2004), 80% of the The term of ‘proteome’ was named
death cases of HCC patients have HCV- by the group of Ian Humphery-Smith
infection, and the number of HCV-carriers (Wasinger et al., 1995). ‘Proteome’ was
in Japan is 2 million. The number of defined as the total protein complement
patients with HCC is still on the increase, of a genome, and ‘proteomics’ means the
although diagnostic and therapeutic tech- analysis for the total protein complement
niques have become remarkably advanced. of a genome. The combination of two-
It was reported by Yatsuhashi (2004) that dimensional gel electrophoresis (2-DE)
the incidence of HCC is increasing in and mass spectrometry (MS) is a popular
many developed countries in parallel to method of ‘proteomics’. By means of
an increase in chronic HCV infection. 2-DE we can separate many proteins

343
344 Y. Kuramitsu and K. Nakamura

according to their charges and their in cancerous tissues compared to non-


size in isoelectoric focusing (IEF) gels cancerous tissues. Kim et al. (2003)
and sodium dodecyl sulfate (SDS) gels, performed proteomics for HCC tissues
respectively. With 2-DE we can exam- including HBV–HCC patients, HCV–
ine the expressions of a huge number of HCC patients and NBNC-HCC patients,
proteins simultaneously. There are many and reported upregulated and downregu-
reports of cancer proteomics obtained by lated proteins in HCC cancerous tissues.
means of 2-DE. Simpson et al. (2000) In HCV–HCC tissues, both 4-aminobu-
reported the proteomic analysis of the tyrate aminotransferase and aflatoxin B1
human colon carcinoma cell line LIM aldehyde reductase were upregulated,
1215 by means of 2-DE. Ha et al. (2002) and cytosol aminopeptidase or acyl-CoA
reported the proteome analysis of human dehydrogenase, or glutamate dehydroge-
stomach tissues. They separated soluble nase was downregulated.
proteins by 2-DE and identified by mass
spectrometry. Steel et al. (2001) reported
a proteomic approach for the discovery of PROTEOMICS FOR SERA
early detection markers of hepatocellular FROM HEPATOCELLULAR
carcinoma by using 2-DE. Presently, MS CARCINOMA PATIENTS
has become the first choice for the deter-
mination of protein sequences. We can Although we can understand the patho-
determine the masses of peptides with genesis of HCC from the proteomics
much accuracy by MS, and we can iden- for HCC tissues, it is not expected to
tify the protein from the huge data base discover biomarkers for HCC from the
of genomics. results of proteomics for HCC tissues.
To discover predictive biomarkers, we
need to perform the proteomics for sera
PROTEOMICS FOR from HCC patients. Some proteomic studies
HEPATOCELLULAR for sera from HCC patients have been
CARCINOMA TISSUES reported. Paradis et al. (2005) reported
proteomics for sera from patients with
Using the proteomic technique many pro- cirrhosis without HCC and patients with
teins have been reported as candidates for cirrhosis without HCC, and identified a
new diagnostic biomarkers for HCCs. Li catalytic fragment of vitronectin as a new
et al. (2005) reported on proteomic analysis marker of HCC. Steel et al. (2003) ana-
of HBV–HCCs without HCV infection in lyzed sera from patients with HBV–HCC,
ten patients, and identified many upregu- and identified down regulated carboxyl
lated (enolase1, GRP 75, galactokinase1, terminal fragment of complement C3 and
HSP27, HSP70.5, PCNA, stathmin, etc.) an isoform of apolipoprotein A1 in sera
and downregulated proteins (aldolase B, from HBV–HCC patients. It is neces-
annexin V, fructose-bisphosphatase, glutath- sary to repeat these types of studies and
ione peroxidase, nucleophosmin, SOD 1, confirm the results for the clinical trial as
triosephosphate isomerase, vimentin, etc.) biomarkers.
27. Hepatitis C Virus-Related Human Hepatocellular Carcinoma: Predictive Markers 345

PROTEOMICS FOR HEPATITIS Two-Dimensional Gel Electrophoresis


C VIRUS-RELATED (2-DE)
HEPATOCELLULAR Isoelectric focusing (IEF)
CARCINOMA TISSUES The first-dimensional isoelectric focusing
(IEF) on 7-cm, immobilized, pH 3–10
Preparation of HCV–HCC Tissue linear gradient (IPG) strips (Bio-Rad) was
Samples performed with 300 μg of each tissue
Cancerous and corresponding noncan- sample at 20°C and 50 μA/strip. Isoelectric
cerous liver tissues were obtained from focusing was run in three steps: 500 V for
patients who were diagnosed with HCV– 1 h, 1,000 V for 1 h, and 8,000 V for 3 h.
HCC and underwent surgical liver resec-
tion. The liver tissues were suspended Sodium Dodecyl Sulfate-Polyacrilamide
and homogenized in lysis buffer (50 mM Gel Electrophoresis (SDS-PAGE)
Tris-HCl, pH 7.5, 165 mM NaCl, 10 mM Figure 27.2 shows the experimental
NaF, 1 mM sodium vanadate, 1 mM phe- design of 2-DE. The second-dimensional
nylmethylsulfonyl fluoride, 10 mM EDTA, run was performed on precast polyacry-
10 μg/ml aprotinin, 10 μg/ml leupeptin, lamide gels (2-D homogeneous 12.5;
and 1% NP40). Suspensions were soni- GE Healthcare) in two steps: 600 V,
cated and centrifuged with 15,000 g for 20 mA for 30 min and 600 V, 50 mA for
30 min at 4°C. The supernatants were 70 min. After electrophoresis the gels
taken as samples. Figure 27.1 shows a were stained with CBB R250 or SYPRO
workflow of our experiment. Ruby (Invitrogen) for 24 h. Gels stained

Figure 27.1. Workflow of proteomics for HCV–HCC


346 Y. Kuramitsu and K. Nakamura

Mass Spectrometry Analysis


In-gel Digestion
For staining gels, CBB dye was removed
by rinsing three times in 60% methanol,
50 mM ammonium bicarbonate, and 5 mM
dithiothreitol (DTT) for 15 min and twice
in 50% acetonitrile, 50 mM ammonium
bicarbonate, and 5 mM DTT for 10 min.
Gel pieces were dehydrated three times
in 100% acetonitrile for 30 min and then
Figure 27.2. Scheme of two-dimensional gel
rehydrated in an in-gel digestion reagent
electrophoresis. The technique of 2-DE is able to containing 10 μg/ml sequencing-grade
differentiate proteins according to both their charge modified trypsin in 30% acetonitrile,
in isoelectoric focusing (IEF) gels and their size in 50 mM ammonium bicarbonate, and 5 mM
sodium dodecyl sulfate (SDS) gels DTT. In-gel digestion was performed over-
night at 30°C. The samples were rinsed
in 30% acetonitrile, 50 mM ammonium
with CBB R250 were destained with bicarbonate, and 5 mM DTT for 2 h and
10% acetic acid in water containing lyophilized overnight at −30°C.
30% methanol for 30 min and then with
7% acetic acid for 3 h and used for in-
Amino Acid Sequencing by Liquid
gel digestion. Gels stained with SYPRO
Chromatography–Tandem Mass
Ruby were washed with 7% acetic acid
Spectrometry (LC-MS/MS)
in water containing 10% methanol for
30 min and washed twice with mili-Q Lyophilized samples were dissolved in
water for 5 min, and then they were gen- 20 μl 0.1% formic acid and centrifuged
erated by scanning of gels at suitable at 15,000 g for 5 min. Peptide sequenc-
excitation and emission wavelengths. ing of identified protein spots was per-
formed by LC-MS/MS with a Spectrum
Image Analysis of the Gels Mill MS Proteomics Workbench (Agilent
Technologies).
The positions of the protein spots The different expression levels of identi-
were recorded using an image scan- fied proteins were confirmed by Western
ner (ProXPRESS 2D Proteomic Imaging blotting using specific antibodies.
System, Perkin-Elmer), and the posi-
tions and intensities of the spots were
Proteins Whose Expression Was
analyzed individually with computer
Increased in HCV–HCC Tissues
software (Progenesis, Perkin-Elmer).
Spots stained at different intensities were By means of the analysis with 2-DE, 11
excised from the gels and identified by spots, whose expression intensity was
peptide sequence trap (PST) with liquid increased more than twofold in cancerous
chromatography-tandem mass spectro- tissues than in noncancerous tissues, were
metry (LC-MS/MS). detected, and identified as GRP75, GRP78,
27. Hepatitis C Virus-Related Human Hepatocellular Carcinoma: Predictive Markers 347

HSC71, HSP70.1, HSP60, glutamine syn- three protein spots of Mr 42,000 and pI
thetase, triosephosphate isomerase, ATP 6.4–6.8 on the 2-DE gel.
synthetase beta chain and alpha-enolase (7) Triosephosphate isomerase
(Takashima et al., 2003, 2005; Kuramitsu Triosephosphate isomerase is an enzyme of
et al., 2006). The functions of these protein glycolytic pathway, and catalyzes the reac-
spots are described below. tion between D-glyceraldehyde-3-phosphate
(1) GRP75 and dihydroxyacetone phosphate.
Glucose-regulated 75 kDa protein (GRP75) (8) Alpha-enolase
is located in the mitochondria, and inhibits Alpha-enolase is an isoenzyme of eno-
cell division and controls cell proliferation lase, a key protein that catalyzes the
and cellular aging. conversion of 2-phosphoglycerate to
(2) GRP78 phosphoenolpyruvate in the glycolytic
Glucose-regulated 78 kDa protein (GRP78) pathway. Three enolase isoenzymes
is a molecular chaperone. This molecule is have been identified. The alpha form is
involved in the unfolded protein response, present in most embryonic tissues, the
and plays a role in facilitating the assembly beta form is expressed in muscle tissues,
of multimeric protein complexes inside the and the gamma form is found only in
endoplasmic reticulum. neuronal tissues. Alpha enolase has been
(3) HSC70 detected not only in the cytoplasm but also
Heat shock cognate 71 kDa protein at the membrane surface. Although the
(HSC70) is a constitutively expressed mechanism of surface expression and the
chaperone that is also inducible by heat orientation on the membrane are not well
shock stress. It binds to nascent polypep- understood, it is known that the C-terminal
tides to facilitate correct folding and also amino acid, lysine, is exposed at the cell
participates in the formation and repair surface and is involved in binding to
of higher order protein structures such as plasminogen, which is then activated and
that of ATPase. converted to plasmin. Plasmin, stabilized
(4) HSP70.1 at the cell surface, induces fibrinolysis.
Heat shock 70 kDa protein 1 (HSP70.1) (9) Adenosine triphosphate (ATP) syn-
stabilizes preexistent proteins against thase beta chain
aggregation and mediates the folding of Adenosine triphosphate synthase is an
newly translated polypeptides within the enzyme that synthesizes ATP from adeno-
cytosol as well as within in cooperation sine diphosphate (ADP) and phosphate.
with other chaperones. Beta chain is a subunit of this enzyme.
(5) HSP60
Although heat shock 60 kDa protein
(HSP60) is one of the molecular chaper- Proteins With Decreased Expression
ones, this molecule does not belong to the By means of the analysis with 2-DE, 11
HSP70 family. spots with expression intensity decreased
(6) Glutamine synthetase less than half in cancerous tissues than
Glutamine synthetase is an enzyme to noncancerous tissues were detected, and
synthesize glutamine from glutamate and identified as aldolase, arginase 1, enoyl-CoA
NH3. This protein was identified from the hydratase, ketohexokinase, smoothelin,
348 Y. Kuramitsu and K. Nakamura

tropomyosin beta chain, ferritin light PROTEOMICS FOR AUTO-


chain, and serum albumin (Yokoyama ANTIBODIES IN THE SERA
et al., 2004). The functions of these protein OF HEPATOCELLULAR
spots are described below.
CARCINOMA PATIENTS
(1) Aldolase B (liver type)
This protein was identified from the four Serum samples. The autologous serum
protein spots with different pI on the 2-DE samples were obtained from each patient
gel. Aldolase is an enzyme to perform and sera from patients with normal liver
the sixth step of glycolysis, and has three tissues were also obtained as control sera.
isozymes. Aldolase A exists in muscle, Two-dimensional immunoblot analy-
aldolase B exists in liver, and aldolase C sis. To detect autoantibodies expected as
exists in brain. cancer biomarkers for HCC, we tried to
(2) Arginase 1 identify serum autoantibodies against pro-
Arginase 1 is an enzyme that performs the teins in HCC cancer tissue. Tissue proteins
first step of urea cycle, and is rich in liver. were separated by 2-DE, transferred onto
(3) Enoyl-CoA hydratase PVDF membranes, and immunoblotted
Enoyl-CoA hydratase is an enzyme that with autologous sera. For the blotting-
performs the second step of beta oxidation gels, separated proteins were transferred
pathway of fatty acid. onto PVDF membranes (Immobilon-P;
(4) Ketohexokinase Millipore) electrophoretically, and the
Ketohexokinase is an enzyme that performs membranes were blocked for 1 h at 4°C
the first step of fructose metabolic pathway. with TBS containing 1% skimmed milk.
(5) Smoothelin They were subsequently incubated over-
Smoothelin is a cytoskeletal protein exist- night at 4°C with autologous serum (1:50
ing in visceral and vascular smooth muscle dilution), washed four times with TBS
cells as 59 kDa smoothelin-A and 110 kDa containing 0.05% Tween 20, and incu-
smoothelin-B. bated for 1 h at 4°C with horseradish per-
(6) Tropomyosin beta chain oxidase-conjugated secondary antibody
Tropomyosin is a heterodimer of alpha and (1:1,000). The reaction was visualized
beta chains. Tropomyosin binds to actin with a chemiluminescence reagent (ECL;
filaments and regulates muscle contraction GE Healthcare).
in conjunction with troponin. Tropomyosin By comparing each immunoblot pattern,
stabilizes of actin filaments. we identified spots with stronger staining
(7) Ferritin light chain intensity in cancer tissues than in non-
Ferritin is composed of light chain and cancerous tissues. Matched proteins
heavy chain, and an iron-storage molecule, on 2-DE gels were identified by MS.
which acts as an internal iron reserve and Immunoreactivity of autologous serum
protects against iron toxicity. autoantibodies to tissue proteins was
(8) Serum albumin assessed in samples of HCC cancer tissues
Serum albumin is the most abundant plasma and noncancerous tissues. Four immuno-
protein produced in hepatocytes, and main- reactive spots were detected that showed
tains the osmotic pressure of body fluid. increased intensity in cancer tissues
27. Hepatitis C Virus-Related Human Hepatocellular Carcinoma: Predictive Markers 349

Figure 27.3. Detection for autoantibodies from HCC patients.


Experimental procedure for the detection of autoantibodies against the proteins in HCC cancerous
tissues.

compared to that of noncancerous tis- Kim, W., Lim, S.O., Kim, J-S., Ryu, Y.H., Byeon,
sues. Each of the spots was matched to an J.Y., Kim, H.J., Kim, Y.I., Heo, J.S., Park,
equivalent spot on staining gels. To iden- Y.M., and Jung, G. 2003. Comparison of pro-
tify these four immunoreactive proteins, teome between hepatitis B virus- and hepatitis C
the spots were digested, and were identi- virus-associated hepatocellular carcinoma. Clin.
Cancer Res. 9: 5493–5500.
fied by MS as HSP70.1, glyceraldehyde
Kuramitsu,Y.,Harada,T.,Takashima,M.,Yokoyama,Y.,
3-phosphate dehydrogenase (GAPDH), Hidaka, I., Iizuka, N., Toda, T., Fujimoto, M.,
peroxiredoxin, and Mn-SOD (Takashima Zhang, X., Sakaida, I., Okita, K., Oka, M. and
et al., 2006). Figure 27.3 shows a scheme Nakamura, K. 2006. Increased expression, and
of the detection system for autoantibodies phosphorylation of liver glutamine synthetase
from HCC-patients sera. in well-differentiated hepatocellular carcinoma
tissues of patients infected with hepatitis C virus.
Electrophoresis 27: 1651–1658.
REFERENCES
Li, C., Tan, Y-X., Zhou, H. Ding, S.J., Li, S.J.,
El-Serag, H. B., and Mason, A. C. 1999. Rising Ma, D.J., Man, X.B., Hong, Y., Zhang, L.,
incidence of hepatocellular carcinoma in the Li, L., Xia, Q.C., Wu, J.R,, Wang, H.Y., and
United States. N. Eng. J. Med. 340: 745–750. Zeng, R. 2005. Proteomic analysis of hepa-
Ha, G.H., Lee, S.U., Kang, D.G., Ha, N.Y., Kim, titis B virus-associated hepatocellular carci-
S.H., Kim, J., Bae, J.M., Kim, J.W., and Lee, noma: Identification of potential tumor markers.
C.W. 2002. Proteome analysis of human stom- Proteomics 5: 1125–1139.
ach tissue: separation of soluble proteins by Okuda, K. 1992. Hepatocellular carcinoma: recent
two-dimensional polyacrylamide gel electro- progress. Hepatology 15: 948–963.
phoresis and identification by mass spectrom- Okuda, K. 2000. Hepatocellular carcinoma. J.
etry. Electrophoresis 23: 2513–2524. Hepatol. 32: 225–237.
350 Y. Kuramitsu and K. Nakamura

Paradis, V., Degos, F., Dargere, D., Pham, N., shock protein 70 family members as biomarkers
Belghiti, J., Degott, C., Janeau, J.L., Bezeaud, for hepatitis C virus-related hepatocellular carci-
A., Delforge, D., Cubizolles, M., Laurendeau, noma. Proteomics 3: 2487–2493.
I., and Bedossa, P. 2005. Identification of a new Takashima, T., Kuramitsu, Y., Yokoyama, Y.,
marker of hepatocellular carcinoma by serum Iizuka, N., Fujimoto, M., Nishisaka, T., Sakaida,
profiling of patients with chronic liver diseases. I., Okita, K., Oka, M., and Nakamura, K. 2005.
Hepatology 41: 40–47. Overexpression of alpha enolase in hepatitis C
Seow, T. K., Liang, R. C. Y., Leow, C. K., and virus-related hepatocellular carcinoma: asso-
Chung, M. C. M., 2001. Hepatocellular carci- ciation with tumor progression as determined by
noma: from bedside to proteomics. Proteomics proteomic analysis. Proteomics 5: 1686–1692.
1: 1249–1263. Takashima, T., Kuramitsu, Y., Yokoyama, Y.,
Simpson, R.J., Connolly, L.M., Eddes, J.S., Iizuka, N., Fujimoto, M., Sakaida, I., Okita, K.,
Pereira, J.J., Moritz, R.L., and Reid, G.E. 2000. Oka, M., and Nakamura, K. 2006. Proteomic
Proteomic analysis of the human colon carci- analysis of autoantibodies in patients with hepa-
noma cell line (LIM 1215): development of a tocellular carcinoma. Proteomics 6: 3894–3900.
membrane protein database. Electrophoresis 21: Wasinger, V.C., Cordwell, S.J., Cerpa-Poljak,
1707–1732. A., Yan, J.X., Gooley, A.A., Wilkins, M.R.,
Steel, L.F., Mattu, T.S., Mehta, A., Hebestreit, Duncan, M.W., Harris, R., Williams, K.L., and
H., Dwek, R., Evans, A. A., London, W.T., and Humphery-Smith, I. 1995. Progress with gene-
Block, T. 2001. A proteomic approach for the product mapping of the Mollicutes: Mycoplasma
discovery of early detection markers of hepato- genitalium. Electrophoresis 16: 1090–1094.
cellular carcinoma. Dis. Markers 17: 179–189. Yano, K., Yatsuhashi, H., and Yano, M., 2003.
Steel, L.F., Shumpert, D., Trotter, M., Seeholzer, Hepatitis C. Nippon Rinsho 61: 241–244.
S.H., Evans, A.A., London, W.T., Dwek, R., and Yatsuhashi, H., 2004. General remarks on chronic
Block, T.M. 2003. A strategy for the compara- hepatitis C. Nippon Rinsho 62: 400–404.
tive analysis of serum proteomes for the discov- Yokoyama, Y., Kuramitsu, Y., Takashima, T., Iizuka,
ery of biomarkers for hapatocellular carcinoma. N., Toda, T., Terai, S., Sakaida, I., Oka, M.,
Proteomics 3: 601–609. Nakamura, K., and Okita, K. 2004. Proteomic
Takashima, T., Kuramitsu, Y., Yokoyama, Y., Iizuka, profiling of proteins decreased in hepatocellular
N., Toda, T., Saiakida, I., Okita, K., Oka, M., and carcinoma from patients infected with hepatitis
Nakamura, K. 2003. Proteomic profiling of heat C virus. Proteomics 4: 2111–2116.
A. Diagnosis
28
Liver Metastases from Colorectal
Cancer: Ultrasound Imaging
Søren R. Rafaelsen

INTRODUCTION curative intent increased over time from


6% (1976–1984) to 30% (1994–2003).
Colorectal cancer is the second largest No existing form of systemic therapy can
cause of cancer-related deaths in the United effectively eradicate diffuse metastatic
States. disease, but a partial response to chemothe-
Approximately 15–20% of the patients rapy can be achieved to downstage the liver
with cancer of the colon and rectum have disease, which would then be amenable to a
liver metastases at the time of primary sur- surgical resection. Neoadjuvant chemothe-
gery (Faivre et al., 2003). Assessment of rapy is able to increase the tumor resectability
liver metastases is essential for the appro- by 10%. This increased number of curative
priate management of colorectal cancer. resections was accompanied by a relatively
Surgical resection is an effective treatment low complication rate and a 5-year survival
for hepatic metastases of colorectal cancer. that is within the range of the survival rates
However, this option is available only with initially resectable lesions (Adam et al.,
to a small group of patients. Preoperative 2001). Patients with advanced rectal cancer
portal vein embolization for hypertrophy as well as limited liver metastases may ben-
of future hepatic resection and other pio- efit from a combination of radio frequency
neering multidisciplinary modalities and ablation (RFA) and local radiotherapy prior
the aggressive surgical approach has been to rectal surgery. In patients with limited but
adopted to extend the frontiers of surgi- inoperable colorectal metastases RFA has
cal therapy (Khatri et al., 2005). For the shown a 5-year survival up to 30%. Radio
majo-rity of patients surgery is not a cura- frequency ablation provides the opportunity
tive option, and only palliative therapy is for localized tissue destruction of limited
available resulting in no long-term survi- volumes of tumor; it can be offered to non-
vors. If left untreated the median survival surgical candidates and used in conjunc-
is 6 months, but with modern combina- tion with systemic therapy and/or surgery
tion chemothe-rapy a median survival of (Gillams, 2005).
24 month is possible. In a French study Presently, multiple imaging modalities
(Guyot et al., 2005) the proportion of are available to evaluate liver metastases.
patients with liver metastases resected with Despite the powerful imaging modalities,

355
356 S.R. Rafaelsen

such as computed tomography (CT), mag- lobes provides a better signal-to-noise ratio
netic resonance imaging (MRI) and positron and reduces artifacts. Destructive sound-
emission tomography (PET), ultrasound wave effects from the body wall are also
has continued to play an important role. reduced. Choudhry et al. (2000) found that
Ultrasound has no ionizing radiation and in patients with a body mass index of 30 or
can be performed at low cost compared more tissue harmonic imaging was clearly
to the former modalities. With the advent better for lesion visibility and confidence
of intraoperative- and larparoscopic ultra- of diagnosis. The authors recommend rou-
sound, tissue harmonic imaging, color-, tine use of harmonic imaging for abdominal
power Doppler, contrast enhancement, pulse ultrasound studies in all adult patients.
inversion imaging, microflow imaging, 3D, However, for patients with diffuse fatty
and 4D ultrasound still have an important infiltration in the liver, penetration of the
role in diagnosing liver metastases as well low frequency ultrasound waves is often
as response evaluation, ultrasound guided better on conventional ultrasound images
biopsy and RFA. than when using tissue harmonic imaging.
Ultrasound guided biopsy is often used
to obtain cytological and histological veri-
ULTRASOUND SCANNING fication of liver metastases from colorectal
TECHNIQUE cancer prior to chemotherapy. Because of
the risk of needle-track tumor seeding,
Hepatic ultrasound is usually performed liver surgeons advise against percutane-
with the patient in the supine position. ous biopsy. A recent study has described
Turning the patient with the right side a negative effect on patient long-term
up may also be helpful in some cases. survival after liver resection. The 4-year
Images are obtained in sagital, transverse, survival was 32% in the group of patients
and oblique planes. The transducer is who had a preoperative liver biopsy versus
positioned in the right subcostal margin 47% in those without (Jones et al., 2005).
and by angling or moving the transducer In addition to biopsy assistance, ultrasound
it is possible to obtain images in various is used as needle guidance of percutaneous,
planes. Deep inspiration is often helpful intraoperative and larparoscopic RFA.
by displacing the liver caudally inferior Realtime virtual sonography enables
to the curvature. Intercostal scanning is the realtime demonstration of multiplanar
also used as a visualization technique. reconstructed CT imaging corresponding
Harmonic waves are generated from non- to the realtime ultrasound image side-by-
linear distortion of an acoustic signal as an side on the screen. The system uses an
ultrasound wave insonates tissues in the electromagnetic position sensor mounted
body. These harmonic ultrasound waves on the transducer. This can be helpful
are numeral multiples of a fundamental in ultrasound-guided RFA and biopsies.
transmitted frequency. Potential advan- The sonographical visualization of focal
tages of harmonic imaging include better lesions in a steatotic liver can be difficult.
axial resolution due to higher frequencies The association between hepatic steatosis
and improved lateral resolution due to nar- and oral 5-fluorouracil (5-FU) agents
rower beams. Decreasing noise from side is well described (Miyake et al., 2005).
28. Liver Metastases from Colorectal Cancer 357

5-fluorouracil is frequently used in CRC GRAYSCALE ECHO PATTERN


patients and “realtime virtual sonography”
is helpful in ultrasound guided interven- Isoechoic and hyperechoic metastases are
tions within steatotic livers. often surrounded by a low echoic halo,
which is highly predictive of malignancy,
(Figure 28.1 [Bruneton et al., 1996]). There
ADVANTAGES OF DIAGNOSTIC is a considerable difference in the size of
HEPATIC ULTRASOUND the metastases. A mean size of 4.5 cm is
reported. The mean thickness of the halo
Although non-enhanced ultrasound has in colorectal liver metastases is publicized
lower sensitivity compared to CT espe- to 2.6 mm, range: 1–5 mm (Rafaelsen and
cially in obese patients, ultrasound can be Solvig, 2004).
performed bedside, is low cost and has no Calcification is described in 12–27% of
radiation. hepatic colorectal metastases. Calcification
Small liver cysts may be mistakenly occurred independent of the degree of tumor
diagnosed as metastatic disease on diag- differentiation, the presence of mucinous
nostic CT and screening CT colonogra- adenocarcinoma, or the hepatic tumor burden.
phy. Sonography may be useful in cancer The presence of calcification within a
patients with average body habitus to char- colorectal liver metastasis appears to imply
acterize small (0.6–1.5-cm) indeterminate a significantly better prognosis (Easson
liver lesions detected on CT (Eberhardt et al., 1996). Overall survival difference was
et al., 2003). The partial volume effect on compared between hyper- and hypoechoic
CT scan can cause small benign cysts to metastases on an intention-to-treat basis
mimic solid liver metastases. Since the dis-
tinction between cysts and metastases has
important clinical implications, sonography
should be used whenever the true nature of
these lesions cannot be determined by CT.
Hemangiomas can also be clarified by con-
trast enhanced sonography, showing centrip-
etal progression of the enhancement. There is
no contraindication using contrast-enhanced
ultrasound in patients with renal failure or
Metformin medication, as is the case in
contrast-enhanced CT scanning. Gallstones
can easily be detected using ultrasound and
evidence for an association between gall-
stones and colorectal cancer is confirmed.
This group of cancer patients had a signifi-
cantly higher prevalence of gallstone disease
than the control population (odds ratio = Figure 28.1. Ultrasound image showing a charac-
1.59; 95% confidence limits: 1.04–2.45) teristic hepatic metastasis. The thin halo is clearly
(Jorgensen and Rafaelsen, 1992). visible
358 S.R. Rafaelsen

in a study of 212 consecutive patients 295 consecutive patients with colorectal


with unresectable hepatic metastases from cancer showed a sensitivity of 70% versus
colorectal carcinoma. A significant survival 97% with intraoperative ultrasound. Sub-
benefit was observed in patients having analysis on a segmental level showed a
hyperechoic lesions compared to hypoe- high sensitivity of preoperative ultrasound
choic lesions (16 vs. 12 months median of metastases in segments III and VI, but
survival) (Gruenberger et al., 2002). in the other liver segments intraoperative
ultrasound had statically higher sensitivity
(Rafaelsen et al., 1995). An earlier study
DETECTION OF LIVER also found preoperative ultrasonography
METASTASES less powerful for examination of the pos-
terior segment of the liver (Gunven et al.,
In an early study, high-end ultrasound scan- 1985). A comparative study of preopera-
ning in experienced hands equalled the tive ultrasound, CT, and CT portography at
sensitivity of CT in detecting small metas- the time of primary resection of colorectal
tases (Green et al., 1977). Ultrasound is cancer showed that preoperative percuta-
still more operator-dependent than CT and neous ultrasound was the best non-invasive
yields results that are only as good as the imaging method. The accuracy of CT porto-
capabilities of the sonographer and has lim- graphy was worst because of many false
itations due to body habitus and overly-ing positive results (Nies et al., 1996).
gas. The diagnostic accuracy increased with Intraoperative ultrasonography (IOUS)
an increase in the number of metastases reduces the number of patients with liver
and with an increase in size of the lesions. metastases from being subjected to super-
A prospective comparative investigation of fluous or even harmful liver surgery and
71 colorectal patients demonstrated a sensiti- it may increase the number in whom liver
vity of 71% using ultrasound and 82% using surgery will prolong life. IOUS accu-
CT scanning (Lundstedt et al., 1987). racy may be further improved by con-
The clinical value of preoperative ultra- trast enhanced -IOUS with an impact on
sonography in screening for synchronous surgical strategy for metastatic tumors.
liver metastases was prospectively evalu- However, the new technique has a learn-
ated in 338 patients with colorectal cancer. ing curve and requires additional costs
Synchronous liver metastases were observed (Torzilli et al., 2005). Because conven-
at laparotomy in 11.5% (39/338) of the tional palpation of the liver is not possible
patients. The sensitivity of preoperative during laparoscopic surgery, the routine
ultrasound was 76%. In detecting liver use of laparoscopic ultrasound should also
metastases the results were superior to those be considered for laparoscopic oncologic
of biochemical blood tests and measure- colorectal surgery. This method enables
ments of carcinoembryonic antigen serum an earlier detection of liver metastases and
levels. The accuracy of the ultrasonography may have an impact on the survival rate
was also superior to that of these other tests of a limited subgroup of colorectal cancer
combined (Yoshida et al., 1989). patients (Milsom et al., 2000).
In a prospective blinded patient by Using contrast enhanced sonography,
patient study of preoperative ultrasound, an increased sensitivity is reported. Pulse
28. Liver Metastases from Colorectal Cancer 359

inversion technique at low mechanical to the hepatic vein seems to be able to


index performed with microbubble con- discriminate between patients with and
trast agents shows that noninvasive char- without liver metastases. In a small series,
acterization of liver masses is possible. the transit times in 14 patients with liver
A hypoechoic mass seen during the portal metastases were ≤ 10 s, while in 50 patients
venous phase, regardless of its appearance without metastases the times were ≥ 12 s
during the arterial phase, is suspicious for (Bernatik et al., 2004).
malignancy. Metastases have consistently
shown less enhancement compared to the
liver during the portal venous phase of DOPPLER FLOW PATTERN
contrast-enhanced ultrasound (Brannigan
et al., 2004). Several studies have shown The hepatic metastases from colorectal
that the use of contrast agents increases cancer often have a characteristic sono-
the sensitivity and specificity of liver graphic appearance, typically showing a
metastases. One study showed an increase hypoechoic halo. The nature of this halo
in sensitivity from 69% on non-enhanced is poorly understood and opinions differ.
ultrasound to 90% using contrast enhance- Some authors have suggested that the halo
ment (Oldenburg et al., 2005). Missed represents an oedema or compressed peri-
metastases using conventional ultrasound tumoral liver tissue (Marchal et al., 1985),
is of small size or isoechoic. while others have shown active tumor tissue
In an animal study no significant differ- in an in-vitro study (Wernecke et al., 1992).
ence was found between contrast-enhanced Color Doppler can visualize blood flow
ultrasound and MR imaging in the detec- in tissue but the technique is not particu-
tion of hepatic tumors, whereas contrast- larly efficient for detection of slow flow
enhanced ultrasound had the highest in small vessels. Power Doppler outper-
accuracy (92%) of the modalities studied forms color Doppler for the detection of
(Forsberg et al., 2002a). However, these blood flow. Although echo-enhancers have
results have to be proven in a clinical set- been reported to improve the signal-to-
ting, taking into consideration that the blind noise-ratio, these agents have only been
angles and subcutaneous fat can hamper used for evaluation of liver metastases
diagnostic hepatic ultrasound scanning. from colorectal cancer in patients using
In another animal study the size of the color Doppler. Power Doppler in com-
implanted tumor nodules measured by his- bination with ultrasound contrast agents
topathology correlated closely (correlation has been found most useful for detection
coefficient: 0.996) with those measured by of vascular flow (Goldberg et al., 1996).
contrast enhanced sonography (Maruyama To our knowledge, a systematic, contrast-
et al., 2005). enhanced power Doppler study of liver
A short hepatic contrast transit time metastases from colorectal cancer has not
(< 27 s) appears to be found only in patients previously been conducted. An under-
with liver disease shown in a small series standing of the tumor margin is essential
by Bang et al. (2001). A sonographic in relation to the increasing use of thermal
measurement of the transit time of an ablation. The vascular nature of the metas-
echo enhancer from the hepatic artery tases also has definite clinical implications
360 S.R. Rafaelsen

because anti-angiogenic tumor therapy, An in vivo study of eight colon tumors


i.e., bevacizumab, represents a promising compared contrast-enhanced Doppler
new strategy for cancer treatment, and ultrasonography with histological slides
the side-effect profile makes it a suitable for evaluation of angiogenesis and found
adjunct to standard chemotherapy in set- that the detection threshold after Levovist
tings where efficacy has been demonstrated. allowed visualization of small vessels
Bevacizumab is now approved for use in (Lassau et al., 2001). Leen et al. (1994)
the USA and the European Union (Gordon have previously described that Levovist
et al., 2005). improved the sensitivity of color Doppler
Rafaelsen and Solvig (2004) described flow imaging of the vascular appear-
the ability of color Doppler, power Doppler ance of colorectal cancer metastases. A
and contrast-enhanced flow imaging to study deploying contrast-enhanced power
detect Doppler signals in liver metastases Doppler in the examination of heterogene-
from colorectal cancer and to evaluate a ous liver lesions in 32 patients detected an
possible connection between local recur- increased number of intra-tumoral vessels
rence and the vascular architecture in liver in focal lesions with power Doppler upon
metastases as shown by power Doppler. administration of contrast medium (Hosten
The superiority of contrast-enhanced et al., 1999). This also applied to liver
power Doppler imaging over native power metastases from colorectal cancer. The
Doppler examination of liver metastasis vascularity of the halo in liver metastases
halos from colorectal cancer (P < 0.005) has also been demonstrated in an animal
supports the assumption of peripheral vas- study of hepatic colon cancer metastases,
cularity. However, it is emphasized that the which reported distended sinusoidal spaces
technique could not differentiate between that gave rise to new tumor-penetrating
tumor and non-tumor vascularization, vessels as well as tumor cells (Kruskal
Figure 28.2. et al., 2000). This supports the assumption
that the peritumoral vascularity represents
neoangiogenesis rather than simply dis-
placed liver vessels. A peripheral arterial
ring enhancement of liver metastases from
colorectal cancer has also been described
using contrast agent and pulse inversion
harmonic imaging (Tanaka et al., 2001).
Detection of flow signal in the center was
possible using contrast-enhanced power
Doppler in 21 of 43 of the metastases
(Rafaelsen and Solvig, 2004). The study
seems to be the first reporting this observa-
tion in liver metastases. None or few signals
were observed in the core of the lesions in
the above-mentioned study (Leen et al.,
Figure 28.2. Power Doppler image showing flow 1994) using enhanced color Doppler. This
within the halo finding could be explained by the increased
28. Liver Metastases from Colorectal Cancer 361

ability of power Doppler to detect flow Doppler signals tended to be detected


in comparison with color Doppler. In our more frequently in patients with multiple
flow study larger metastases were found liver metastases than in patients with fewer
more frequently to have a central Doppler than five lesions (50% vs. 23%), which
signal than smaller lesions. This supports may also signal a more aggressive beha-
the hypothesis that there is an associa- vior. Synchronous liver disease and lymph
tion between metastases showing a central node metastases seemed to be associated
power Doppler flow and local tumor recur- with a slight increase in Doppler signals
rence. However, the chance of detecting a (Rafaelsen and Solvig, 2004). These find-
signal is greater in a large volume than in ings need to be further confirmed in future
a small volume. Moreover, the larger the large-scale studies. Patients with synchro-
metastasis, the more advanced the disease, nous disease have been shown to do signifi-
and therefore the higher the likelihood of cantly worse than those with metachronous
disease with recurrence elsewhere. Thus, hepatic resections (Jenkins et al., 1997).
it is implied that central signal detection is Important new information may be derived
merely a function of larger masses. from studying the relationship between
Among 12 patients with power Doppler, flow pattern and tumor grade.
flow signals within the center eight were A recent study of 152 colorectal cancer
observed in association with a local recur- patients showed a significant correlation
rence, whereas none without central flow between a poorer 5-year survival rate and
had local recurrence, which indicates that vascular endothelial growth factor C and
a central flow may be associated with histological microvessel density (Furudoi
even more advanced disease (Figure 28.3). et al., 2002). Tumor necrosis factor has

Figure 28.3. Power Doppler image showing a central Doppler signal


362 S.R. Rafaelsen

exhibited specific tumoricidal activity and At present the measurements of the size
may damage the blood vessels within the of the metastases should incorporate the
tumor (Smyth et al., 2003). However, halo in the total diameter. It is essential
it might be too early to conclude that a to include the halo in resection of liver
natural tumor necrotic factor could be an metastases. In addition, the halo must be
explanation for a correlation between the encompassed in the ablation field in inter-
registered few central Doppler signals and stitial therapy studies. The neovascularity
a low incidence of local recurrence. in solid tumors can hinder thermal ablation
A large-scale follow-up study is required as well as large vessels surrounding the
to answer the important question of a pos- lesion. The hypervascularity present in the
sible relationship between flow pattern and margin of the metastasis can act as a heat
growth and response to chemotherapy. sink that limits the heating of tissue used in
In the future, power Doppler examina- radiofrequency ablation. In the flow study
tion of neovascularity of hepatic metastases local recurrence was observed in 66% of
could be a possible adjunct in the clinical the patients with a power Doppler signal in
evaluation of antiangioneogenic therapy the center but in none of the patients who
(Du et al., 2005). Contrast-enhanced ultra- had no such power Doppler signal. The
sound has currently been described as a tool clinical conclusion of this could be that
for assessing differences in the microcircu- attention should be paid to the possibility
lation of animal tumors in studies of antian- of local recurrence where hepatic resection
giogenic agents (Pollard et al., 2002). or tissue ablation is considered.
Bevacizumab (Avastin), the first approved It is concluded that the peripheral halo
therapy designed to inhibit tumor angio- in hepatic metastases indicates tissue with
genesis, has significant clinical benefits in increased vascularity. A central power
the management of colorectal cancer (Diaz- Doppler signal within the liver metas-
Rubio and Schmoll, 2005). tases implies the co-existence of more
With the advent of three-dimensional advanced disease. Further survival studies
power Doppler ultrasound it is now pos- are needed to assess the prognostic value
sible to visualize all detectable vessels of of this new finding and its usefulness as a
the metastasis in one 3-D image and thus tool for monitoring the possible effect of
reduce operator dependability at the same anti-angiogenic therapy.
time using contrast media (Forsberg et al., An in vitro study showed that ultra-
2002b). Furthermore, new contrast specific sound contrast microbubbles targeted
methods and contrast agents are now avail- via linkage with the tripeptide arginine-
able. Phase inversion imaging and flash- arginine-leucine could specifically adhere
echo imaging with intravenously infused to tumor angiogenic endothelium (Weller
microbubble contrast have shown promis- et al., 2005). Endothelial cells of angiogenic
ing results in measuring vascular flow and tumor vasculature are characterized by
tissue perfusion (Lucidarme et al., 2004). altered expression of molecular markers on
The new perfluro gasses replacing air and their surface. Targeted microbubbles may
multipulse scanning technique are very thus offer a noninvasive contrast-enhanced
sensitive in detecting the microbubble sig- ultrasound imaging technique for the func-
nals (Cosgrove and Blomley, 2004). tional imaging of tumor neovascularization
28. Liver Metastases from Colorectal Cancer 363

and may have further implications for thera- FUTURE POTENTIAL


peutic tumor targeting. ADVANCEMENTS
New results have shown that, rather than
ULTRASOUND IN measuring the largest diameter, three-
POSTOPERATIVE FOLLOW-UP dimensional high-frequency ultrasound
imaging may be particularly well-suited for
The aim of follow-up programs in cancer the quantitative assessment of metastatic
patients is to detect relapse or metastases progression and the evaluation of chemo-
in an early asymptomatic and potential therapeutics in preclinical liver metastasis
curable stage. The 5-year cumulative risk models. The traditional two-dimensional
of metachronous liver metastases is 16% method often gives large overestimation or
(Faivre et al., 2003). Computed tomog- underestimation of the tumor volume com-
raphy scanning is used in follow-up pared to the three-dimensional method
programs. Ultrasound of the liver has also (Graham et al., 2005).
been suggested to detect metacroneous Conventional ultrasound systems acquire
liver metastases in patients with high risk ultrasound data sequentially one image
of recurrence (de Goede et al., 1998). The line at a time. The configuration of these
median time to development of liver metas- systems is, therefore, also sequential in
tases detected by prospective serial ultra- nature and processes most of the data
sound scanning of the liver at 3-monthly in a sequential pipeline. The possibility
intervals of ultrasound scanning was 59 of sending out arbitrary signals and the
weeks in the control group and 91 weeks storage of data from all transducer ele-
in the group treated with chemotherapy ments for 5–10 s allows clinical evaluation
(Taylor et al., 1984). Little is known of of synthetic aperture and 3-D imaging.
the frequency of follow-up, and presently Jensen et al. (2005) described a new high-
there are no controlled trials randomising speed real-time ultrasound system with
patients to high- or low-volume control 16 Gbytes RAM especially designed for
programs. However, follow-up programs research purposes.
for patients with curatively resected color- Tele-ultrasound consultation systems
ectal cancer do improve survival but it is via high-speed commercial telecommuni-
not clear which type of imaging or frequency cation lines are presently being tested, and
of visits is optimal (Figueredo et al., 2003). Yoo et al. (2004) suggested that a rate of
Doppler perfusion index (DPI) measure- more than 0.6 Mbit/s at 30 frames/s is suffi-
ments for an early identification of patients cient for the maintenance of diagnostic
at high risk for recurrent disease have been image quality. This will make it possible to
suggested by Leen et al. (1993). Patients send “cine clips” via teleradiology and ask
with normal DPI remained disease-free an ultrasound expert for a second opinion.
and patients with an abnormally high DPI With a new tele-operated robotic chain for
at the time of primary resection later devel- real-time ultrasound image, it might be
oped liver metastases. A recent study could possible to hold and move a real probe on
not confirm the usefulness of the method a distant patient according to the expert ges-
(Roumen et al., 2005). ture and permit an image acquisition using
364 S.R. Rafaelsen

a standard ultrasound device. The remote Brannigan, M., Burns, P.N., and Wilson, S.R.
ultrasound scanning system agreed in at 2004. Blood flow patterns in focal liver lesions
at microbubble-enhanced US. Radiographics
least 80% of the 52 cases with the diagnosis
24:921–935.
obtained from conventional scanning Bruneton, J.N., Raffaelli, C., Balu-Maestro, C.,
(Courreges, 2005). Padovan I, B., Chevallier, P., and Mourou, M.Y.
Computer-aided detection may have the 1996. Sonographic diagnosis of solid liver nodules
potential to increase the accuracy of diag- in cancer patients. Eur. Radiol. 6:439–442.
nosis of focal liver lesions in ultrasound Choudhry, S., Gorman, B., Charboneau, J.W.,
Tradup, D.J., Beck, R.J., Kofler, J.M., and Groth,
images, although the literature is very
D.S. 2000. Comparison of tissue harmonic imag-
scarce (Yoshida, 2003). ing with conventional US in abdominal disease.
The development of new contrast agents Radiographics 20:1127–1135.
and ultrasound software still continues, Cosgrove, D., and Blomley, M. 2004. Liver tumors.
i.e., micro flow imaging. In the future, Abdom. Imaging 29:446–454
ultrasound might be adopted in various Courreges, F., Vieyres, P., Istepanian, R.S., Arbeille, P.,
and Bru C. 2005. Clinical trials and evaluation
drug delivery and other therapeutic appli-
of a mobile, robotic tele-ultrasound system. J.
cations. Microspheres and nanoparticles of Telemed. Telecare 11 Suppl 1:46–49.
different sizes with or without drugs, stem Diaz-Rubio, E., and Schmoll, H.J. 2005. The future
cells, and/or radioisotopes, can be used development of bevacizumab in colorectal can-
for drug delivery or diagnostic purposes. cer. Oncology 69 Suppl 3:34–45.
Ultrasound will then facilitate the deli- Du, W.H., Yang, W.X., Xiong, X.Q., Wang, X.,
Zhou, Y., and Wang H. 2005. Contrast-enhanced
very of chemotherapeutic drugs into liver
ultrasonographic imaging diagnosis on assess-
metastases and promote gene therapy to ment of vascularity in liver metastatic lesions.
targeted tissues under real-time imaging World J. Gastroentol. 11:3610–3613.
(Mitragotri, 2005). Ultrasound will pro- Easson, A.M., Barron, P.T., Cripps, C., Hill, G.,
vide information that will benefit patient Guindi, M., and Michaud, C. 1996. Calcification
management now and in the future. in colorectal hepatic metastases correlates with
longer survival. J. Surg. Oncol. 63:221–225.
Eberhardt, S.C., Choi, P.H., Bach, A.M., Funt,
REFERENCES
S.A., Felderman, H.E., and Hann, L.E. 2003.
Adam, R., Avisar, E., Ariche, A., Giachetti, S., Utility of sonography for small hepatic lesions
Azoulay, D., Castaing, D., Kunstlinger, F., found on computed tomography in patients with
Levi, F., and Bismuth, F. 2001. Five-year survival cancer. J. Ultrasound Med. 22:335–343.
following hepatic resection after neoadjuvant Faivre, J., Manfredi, S., and Bouvier, A.M. 2003.
therapy for nonresectable colorectal cancer. Ann. Epidemiology of colorectal cancer liver metas-
Surg. Oncol. 8:347–353. tases. Bull. Acad. Nath. Med. 187:815–823.
Bang, N., Nielsen, M.B., Rasmussen, A.N., Figueredo, A., Rumble, R.B., Maroun, J., Earle,
Osterhammel, P.A., and Pedersen, J.F. 2001. C.C., Cummings, B., McLeod, R., Zuraw, L.,
Hepatic vein transit time of an ultrasound con- and Zwaal, C. Gastrointestinal Cancer Disease
trast agent: simplified procedure using pulse Site Group of Cancer Care Ontario’s Program
inversion imaging. Br. J. Radiol. 74:752–755. in Evidence-based Care. 2003. Follow-up of
Bernatik, T., Becker, D., Neureiter, D., Hansler, patients with curatively resected colorectal can-
J.H., Frieser, M., Schaber, S., Wein, A., Hahn, cer: a practice guideline. Bio. Med. Central.
E.G., and Strobel, D. 2004. Hepatic transit Cancer 6;3:26
time of an echo enhancer: an indicator of meta- Forsberg, F., Piccoli, C.W., Liu, J.B., Rawool, N.M.,
static spread to the liver. Eur. J. Gastroenterol. Merton, D.A., Mitchell, D.G., and Goldberg, B.B.
Hepatol. 16:313–317. 2002a. Hepatic tumor detection: MR imaging
28. Liver Metastases from Colorectal Cancer 365

and conventional US versus pulse-inversion Guyot, F., Faivre, J., Manfredi, S., Meny, B.,
harmonic US of NC100100 during its reticu- Bonithon-Kopp, C., and Bouvier, A,M. 2005.
loendothelial system-specific phase. Radiology Time trends in the treatment and survival of
222:824–829. recurrences from colorectal cancer. Ann. Oncol.
Forsberg, F., Rawool, N.M., Merton, D.A., Liu, J.B., 16:756–761.
and Goldberg, B.B. 2002b. Contrast enhanced Hosten, N., Puls, R., Lemke, A.J., Steger, W.,
vascular three-dimensional ultrasound imaging. Zendel, W., Zwicker, C., and Felix, R. 1999.
Ultrasonics 40:117–122. Contrast-enhanced power Doppler sonography:
Furudoi, A., Tanaka, S., Haruma, K., Kitadai, Y., improved detection of characteristic flow pat-
Yoshihara, M., Chayama, K., and Shimamoto, F. terns in focal liver lesions. J. Clin. Ultrasound
2002. Clinical significance of vascular endo- 27:107–115.
thelial growth factor C and angiogenesis at the Jenkins, L.T., Millikan, K.W., Bines, S.D., Staren,
deepest invasive site of advanced colorectal E.D., and Doolas, A. 1997. Hepatic resec-
carcinoma. Oncology 62:157–166. tions for metastatic colorectal cancer. Am. Surg.
Gillams, A.R. 2005. The use of radiofrequency in 63:605–610.
cancer. Br. J. Cancer 92:1825–1829. Jensen, J.A., Holm, O., Jensen, L.J., Bendsen, H.,
de Goede, E., Filez, L., Janssens, J., and Van Nikolov, S.I., Tomov, B.G., Munk, P., Hansen, M.,
Cutsem, E. 1998. Follow-up of colon cancer: Salomonsen, K., Hansen, J., Gormsen, K.,
detection of liver metastases: benefit and perio- Pedersen, H.M., and Gammelmark, K.L. 2005.
dicity. Acta Gastroenterol. Belg. 61:8–10. Ultrasound research scanner for real-time synthetic
Goldberg, B.B., Merton, D.A., Forsberg, F., Liu, aperture data acquisition. IEEE. Trans. Ultrason.
J.B., and Rawool, N. 1996. Color Amplitude Ferroelectr. Freq. Control 52:881–891.
Imaging: Preliminary results using vascular Jones, O.M., Rees, M., John, T.G., Bygrave, S., and
sonographic contrast agents. J. Ultrasound Med. Plant, G. 2005. Biopsy of resectable colorectal
15:127–134. liver metastases causes tumour dissemination
Gordon, M.S., and Cunningham, D. Managing and adversely affects survival after liver resec-
patients treated with bevacizumab combination tion. Br. J. Surg. 92:1165–1168.
therapy. 2005. Oncology 69 Suppl 3:25–33. Jorgensen, T., and Rafaelsen, S. 1992. Gallstones
Graham, K.C., Wirtzfeld, L.A., MacKenzie, L.T., and colorectal cancer – there is a relationship,
Postenka, C.O., Groom, A.C., MacDonald, I.C., but it is hardly due to cholecystectomy. Dis.
Fenster, A., Lacefield, J.C., and Chambers, A.F. Colon Rectum 35:24–28.
2005. Three-dimensional high-frequency ultra- Khatri, V.P., Petrelli, N.J., and Belghiti, J. 2005.
sound imaging for longitudinal evaluation of Extending the frontiers of surgical therapy for
liver metastases in preclinical models. Cancer hepatic colorectal metastases: is there a limit?
Res. 65:5231–5237. J. Clin. Oncol. 23:8490–8499.
Green, B., Bree, R.L., Goldstein, H.M., and Stanley, Kruskal, J.B., Thomas, P., Nasser, I., Cay, O., and
C. 1977. Gray scale ultrasound evaluation of Kane, RA. 2000. Hepatic colon cancer in mice:
hepatic neoplasms: patterns and correlations. Dynamic in vivo correlation with hypoechoic
Radiology 124:203–208. rims visible at US. Radiology 215:852–857.
Gruenberger, T., Zhao, J., King, J., Chung, T, Clingan Lassau, N., Koscielny, S., Opolon, P., De Baere, T.,
P.R., and Morris, D.L. 2002. Echogenicity of Peronneau, P., Leclere, J., and Roche, A. 2001.
liver metastases from colorectal carcinoma is Evaluation of contrast-enhanced color Doppler
an independent prognostic factor in patients ultrasound for the quantification of angiogenesis
treated with regional chemotherapy. Cancer in vivo. Invest. Radiol. 36:50–55.
94:1753–1759. Leen, E., Angerson, W.J., Warren, H.W., O’Gorman, P.,
Gunven, P., Makuuchi, M., Takayasu, K., Moule, B., Carter, E.C., and Mc Ardel, C.S.
Moriyama, N., Yamasaki, S., and Hasegawa, H. 1994. Increased sensitivity of color Doppler
1985. Preoperative imaging of liver metastases. flow imaging of colorectal hepatic metastases
Comparison of angiography, CT scan, and ultra- using galactose microparticles: a preliminary
sonography. Ann. Surg. 202:573–579. report. Br. J. Surg. 8:252–254.
366 S.R. Rafaelsen

Leen, E., Goldberg, J.A., Robertson, J., Angerson, with low MI real time contrast enhanced sonogra-
W.J., Sutherland, G.R., Cooke, T.G., and phy and SonoVue. Ultraschall Med. 26:277–284.
McArdle, C.S. 1993. Early detection of occult Pollard, R.E., Sadlowski, A.R., Bloch, S.H., Murray,
colorectal hepatic metastases using duplex colour L., Wisner, E.R., Griffey, S., and Ferrara, K.W.
Doppler sonography. Br. J. Surg. 80:1249–1251. 2002. Contrast-assisted destruction-replenishment
Lucidarme, O., Nguyen, T., Kono, Y., Corbeil, J., ultrasound for the assessment of tumor microvas-
Choi, SH., Varner, J., and Mattrey, R.F. 2004. culature in a rat model. Technol. Cancer Treat.
Angiogenesis model for ultrasound contrast 1:459–470.
research. Acad. Radiol. 11:4–12. Rafaelsen, S.R., Kronborg, O., Larsen, C., and
Lundstedt, C., Ekberg, H., Hederstrom, E., Fenger, C. 1995. Intraoperative ultrasonography
Stridbeck, H., Torfason, B., and Transberg, K.G. in detection of hepatic metastases from colorec-
1987. Radiologic diagnosis of liver metastases tal cancer. Dis. Colon Rectum 38:355–360.
in colo-rectal carcinoma. Prospective evaluation Rafaelsen, S.R. and Solvig, J. 2004. Ultrasound
of the accuracy of angiography, ultrasonography, imaging of flow patterns in liver metastases
computed tomography and computed tomo- from colorectal cancer. Scand. J. Gastroenterol.
graphic angiography. Acta Radiol. 28:431–438. 39:761–765.
Marchal, G.J., Plyser, K., Tshibwabwa-Tumba, Roumen, R.M., Scheltinga, M.R., Slooter, G.D.,
E.A., Verbeken, E.K., Oyen, R.H., Baert, A.L., and van der Linden, A.W. 2005. Doppler per-
and Lauweryns, J.M. 1985. Anechoic halo in fusion index fails to predict the presence of
solid liver tumors. Radiology 156:479–483. occult hepatic colorectal metastases. Eur. J. Surg.
Maruyama, H., Matsutani, S., Saisho, H., Mine, Y., Oncol. 31:521–527.
Kamiyama, N., Hirata, T., and Sasamata, M. Smyth, M.J., Takeda, K., Haykawa, Y., Peschon,
2005. Real-time blood-pool images of contrast J.J., van den Brink, M.R., and Yagita, H. 2003.
enhanced ultrasound with Definity in the detec- Nature´s TRAIL – on a path to cancer immuno-
tion of tumour nodules in the liver. Br. J. Radiol. therapy. Immunity 18:1–6.
78:512–518. Tanaka, S., Ioka, T., Oshikawa, O., Hamada, Y.,
Milsom, J.W., Jerby, B.L., Kessler, H., Hale, and Yoshioka, F. 2001. Dynamic sonography of
J.C., Herts, B.R., and O’Malley, C.M. 2000. hepatic tumors. Am. J. Radiol. 177:799–805.
Prospective, blinded comparison of laparoscopic Taylor, R.H., Gilbert, J.M., Evans, M.G., Lane,
ultrasonography vs. contrast-enhanced com- H.G., and Cassell, P.G. 1984. Prospective serial
puterized tomography for liver assessment in liver ultrasound scanning in resectable colorec-
patients undergoing colorectal carcinoma surgery. tal cancer treated with adjuvant razoxane. Clin.
Dis. Colon Rectum 43:44–49. Exp. Metastasis 2:321–331.
Mitragotri, S. 2005. Healing sound: the use of Torzilli, G., Del Fabbro, D., Palmisano, A., Donadon,
ultrasound in drug delivery and other therapeutic M., Bianchi, P., Roncalli, M., Balzarini, L.,
applications. Nat. Rev. Drug Discov. 4:255–260. and Montorsi, M. 2005. Contrast-enhanced
Miyake, K., Hayakawa, K., Nishino, M., intraoperative ultrasonography during hepatec-
Morimoto, T., and Mukaihara, S. 2005. Effects tomies for colorectal cancer liver metastases.
of oral 5-fluorouracil drugs on hepatic fat con- J. Gastrointest. Surg. 9:1148–1154.
tent in patients with colon cancer. Acad. Radiol. Weller, G.E., Wong, M.K., Modzelewski, R.A.,
12:722–727. Lu, E., Klibanov, A.L., Wagner, W.R., and
Nies, C., Leppek, R., Sitter, H., Klotter, H.J., Riera, J., Villanueva, F.S. 2005. Ultrasonic imaging
Klose, K.J., Schwerk, W.B., and Rothmund, M. of tumor angiogenesis using contrast micro-
1996. Prospective evaluation of different diag- bubbles targeted via the tumor-binding pep-
nostic techniques for the detection of liver metas- tide arginine-arginine-leucine. Cancer Res.
tases at the time of primary resection of colorectal 65:533–539.
carcinoma. Eur. J. Surg. 162:811–816. Wernecke, K., Henke, L., Vassaloo, P., von
Oldenburg, A., Hohmann, J., Foert, E., Skrok, J., Bassewitz, D.B., Diederich, S., Peters, P.E., and
Hoffmann, C.W., Frericks, B., Wolf, K.J., and Edel, G. 1992. Pathologic explanation for hyp-
Albrecht, T. 2005. Detection of hepatic metastases oechoic halo seen on sonograms of malignant
28. Liver Metastases from Colorectal Cancer 367

liver tumors: An in vitro correlative study. Am. packet-based texture analysis for differentiation
J. Radiol. 159:1011–1116. between benign and malignant liver tumours
Yoo, S.K., Kim, D.K., Jung, S.M., Kim, E.K., in ultrasound images. Phys. Med. Biol. 48:
Lim, J.S., and Kim, J.H. 2004. Performance of a 3735–753.
web-based, realtime, tele-ultrasound consultation Yoshida, T., Matsue, H., Suzuki, M., Okazaki, N.,
system over high-speed commercial telecommu- Yoshino, M., Moriya, Y., and Hojo, K. 1989.
nication lines. J. Telemed. Telecare 10:175–179. Preoperative ultrasonography screening for liver
Yoshida, H., Casalino, D.D., Keserci, B., Coskun, metastases of patients with colorectal cancer.
A., Ozturk, O., and Savranlar, A. 2003. Wavelet- Jpn. J. Clin. Oncol. 19:112–115.
29
Preclinical Liver Metastases:
Three-Dimensional High-Frequency
Ultrasound Imaging
Kevin C. Graham, Lauren A. Wirtzfeld, James C. Lacefield, and Ann F. Chambers

INTRODUCTION new blood vessels to allow for continued


development. Although many cells initiate
Metastasis, the spread of a primary can- this sequence of events by gaining access
cer to distant organs, continues to be the to the vascular system, < 1% of these cells
most significant cause of cancer mortality. are able to complete all of the steps to
Isolated primary tumors can often be treated form overt metastases (Chambers et al.,
surgically with a relatively high success rate. 2002). The multi-step nature and bio-
However, if the primary tumor has invaded logical and molecular complexity of the
the surrounding tissue and metastasized metastatic process have necessitated that a
to secondary sites in the body, treatment variety of research tools be used to effec-
options are often limited to systemic chemo- tively model this process. In vitro models
therapies with much lower success rates and have allowed for a greater understanding
greater toxicity. Thus, it is imperative that a of how tumor cells circumvent normal cell
greater understanding of the biology of the growth and survival regulations. In vitro
metastatic process be acquired in order to models are essential to isolate the contri-
achieve a significant reduction in the mor- bution of specific molecular pathways to
bidity and mortality associated with cancer the development of a metastatic cell, but
diagnosis. fail to capture the complexity of the entire
The process of metastasis consists of metastatic process that exists in the in vivo
multiple biological steps governed by situation. Thus, in order to study the com-
a wide range of molecular processes plete metastatic process it is necessary to
(Chambers et al., 2002). The cells in a develop and utilize animal models. Animal
developing primary tumor must invade models are used to study the interactions
the surrounding tissue and gain access of a tumor cell with a changing micro-
to a blood or lymphatic vessel to facili- environment as it progresses through the
tate dissemination. Once the metastatic metastatic process, and are often used to
cell has arrived at a secondary site, the evaluate novel therapeutics and treatment
cell must arrest in the vascular system, strategies.
survive, undergo cell division in the new Historically, preclinical mouse mod-
microenvironment, and eventually recruit els of cancer have been developed from

369
370 K.C. Graham et al.

transplantable tumors of mouse origin in Transgenic animals produce tumors that


immune competent (syngeneic) animals, develop over a time scale and in a pro-
or of human origin (xenograft) in immune gression series that more closely resem-
deficient animals. In both xenograft and bles the clinical situation (Khanna and
syngeneic models the method of inoculat- Hunter, 2005). Transgenic models are
ing the cancer cells into the mice is cho- expensive to produce and maintain, and
sen based on the biological question that because of variable tumor incidence
is being asked. The tumor cells may be and infrequent development of metas-
introduced into the mice through surgical tases these models are often limited to
implantation of cells or a tumor tissue frag- studies with specific objectives that pre-
ment into the orthotopic site. The orthotopic clude the use of transplantable or experi-
site is the natural site of development for a mental models.
tumor type. For instance, the orthotopic One feature that all metastasis models
site for implanting colon tumor cells would have in common is that the development
be into a mouse colon. This method reca- of the metastases is difficult to quantify
pitulates many steps of metastatic develop- non-invasively. Traditionally, metastasis
ment including primary tumor growth and models have been studied by using the
invasion of the surrounding tissue and vas- simple approach of analyzing groups of
cular system. A drawback of this system experimental animals at a common end-
is that the primary tumors often develop point. To study the process of metastasis a
rapidly, inducing animal morbidity or mor- large group of animals was used and then
tality that obscures the study of secondary sub-groups were sacrificed and analyzed at
metastases or requires the surgical resec- specific timepoints throughout the experi-
tion of the primary tumor. Furthermore, ment. This method is prohibitively expen-
there is a lack of control over the number sive and relies on the assumption that the
of metastases that form and the timing of variability in tumor development is small
their development in orthotopic models. enough that an accurate impression of
For these reasons, experimental metastasis the kinetics of metastatic progression can
models are often preferred (Welch, 1997). be gleaned by analyzing small groups of
In experimental metastasis models cancer mice. In many models, particularly geneti-
cells are grown in culture and then injected cally engineered models, the variability in
intravenously to form metastases at specific metastasis development is sufficiently high
sites. In this procedure, the primary tumor to prevent statistically meaningful results
growth and invasion stages are bypassed, from the evaluation of small subgroups of
but the researcher has greater control over animals (Khanna and Hunter, 2005). To
the number of potential metastatic clones address this problem, non-invasive imag-
in an organ and the timing in which overt ing technologies have been developed spe-
metastases may form. cifically for small animal research. The
Recent advances in the development ability to non-invasively image metasta-
of transgenic animals have created ani- sis models would decrease experimental
mal models that carry specific genetic variability by allowing individual mice,
defects that increase their suscep- and the individual metastases developing
tibility to developing particular cancers. within them, to be studied longitudinally.
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 371

The variability in the timing of metastatic needs of each research project when select-
development, while still present, is no ing an imaging modality. Criteria specific
longer a factor in a longitudinal experiment to each research project include the type of
because the researcher can study metastatic metastasis model being used, the anatomical
progression within each individual mouse location at which metastases are expected,
and gather data or initiate a treatment the desired resolution, the availability of
based on the unique stage of progression equipment and funding, the necessity and
in each mouse. By decreasing the impact availability of contrast agents, and access
of experimental variability, smaller groups to personnel skilled in the acquisition and
of animals are required to achieve a statis- interpretation of the images.
tically significant result, thereby reducing For researchers who have access to MRI
both the cost and time required to perform facilities, it offers high-resolution imag-
an experiment. ing, inherent soft tissue contrast, and the
The importance of small animal imaging availability of a variety of contrast agents
in metastasis research is reflected by the for vascular imaging (Evelhoch et al.,
number of technologies dedicated to this task. 2000; Gillies et al., 2000). These features
Many of the technologies commonly used have led to the selection of MRI for pre-
clinically, such as magnetic resonance clinical studies involving the quantifica-
imaging (MRI), x-ray computed tomogra- tion of tumor burden in liver metastasis
phy (CT), and positron emission tomogra- models (Cai et al., 2005; Wu et al., 2003)
phy (PET), have been redesigned to give and in the measurement of tumor blood
spatial resolutions that are adequate for volume and vascular permeability through
small animal imaging (Weissleder, 2002). dynamic contrast enhanced (DCE) MRI
Other imaging technologies, because of (Leach, 2001). Current studies are extend-
their inherent limitations in imaging depth ing the use of MRI to study metastasis to
or their requirement for the genetic mani- the brain (Heyn et al., 2006). Widespread
pulation of the cell line being studied, are use of MRI to noninvasive study of cancer
not widely used clinically, but have been models is largely hampered by long image
developed for and utilized by small animal acquisition and processing times, and the
researchers. These technologies include high costs associated with equipment pur-
many of the optical techniques based on chase and operation (Weissleder, 2002).
fluorescent or bioluminescent proteins. X-ray CT offers high-resolution imag-
The general attributes that make an ing and inherent contrast between bone
imaging modality attractive for metastasis and soft tissue, but poor contrast within
research include high spatial resolution to the various soft tissues themselves. Due
allow metastatic progression to be tracked to these characteristics, CT has been pre-
from early stages, fast image acquisition dominately selected for studies of bone
to increase animal throughput, affordable metastasis (Paulus et al., 2000). The devel-
capital and operating costs, and inherent opment of novel iodinated contrast agents
contrast between the metastases and the which provide liver parenchyma – liver
normal parenchyma. No single imaging metastases contrast has introduced the
modality can fulfill all of these criteria, so possibility of using CT to study liver
the researcher must evaluate the particular metastasis models (Weber et al., 2004).
372 K.C. Graham et al.

This development may make CT useful morbidity and mortality in cancer patients.
for endpoint studies of tumor burden, but Preclinical models of liver metastasis are
longitudinal imaging may be limited due invaluable for our understanding of the
to repeated ionizing radiation exposure metastatic process and in the development
during imaging. of novel therapeutics to target this process.
Positron emission tomography is We have developed an experimental
extremely sensitive, yet suffers from poor metastasis model, injecting cultured tumor
resolution. The strength of PET imag- cells into the mesenteric vein, to produce
ing lies in the use of the specific radi- metastases in the liver of mice (Morris
oisotope-labeled tracers that can serve et al., 1993). This method can be utilized
as reporters for specific molecular proc- with a variety of cell lines, of both mouse
esses (Gambhir, 2002). As opposed to and human origin, from a variety of tumor
the anatomical imaging of MRI and CT, types. We opted to pursue an experimental
PET is able to image biological processes metastasis model to be able to control the
such as glucose metabolism or cellular number and timing of cells delivered to
proliferation by linking the radioisotope the liver. Since the timing of delivery was
to the appropriate tracer. By combining known, we were able to study the earliest
PET and CT, imaging information can be steps in the metastatic process. This led us
gleaned on both the anatomical location to identify a number of key biological and
and the biological activity of tumor cells molecular processes that govern metastatic
(Gambhir, 2002). efficiency in a secondary site (Chambers et
Similar to PET imaging, fluorescent al., 1995, 2002). The resolution required
and bioluminescent imaging have limited to study the early stages of metastasis was
spatial resolution, but are extremely ver- on the single cell level, which necessitated
satile in that the reporter gene or protein the use of high resolution in vivo videomi-
can be manipulated to provide a signal croscopy (IVVM). This is an inherently
in response to a variety of biological invasive procedure that is terminal for the
processes. Fluorescent and bioluminescent animal being studied.
imaging technologies are widely available Expanding on these studies, we are work-
and relatively inexpensive to purchase and ing to identify key biological and molecu-
operate. For these reasons, optical imag- lar processes that govern tumor growth at
ing has found a wide range of applications later stages of metastasis, with the ultimate
in the study of cancer models from the goal of using these discoveries to help
quantification of tumor burden to monitor- identify effective therapeutic strategies.
ing the delivery of cancer gene therapies With the focus on following the develop-
(Choy et al., 2003; Lyons, 2005). ment of micrometastases into tumors of a
A large portion of the metastasis research clinically relevant size, our studies require
in our laboratory has focused on the biology an imaging modality capable of longitu-
of the metastatic process with the liver as dinal imaging at sub-millimeter resolu-
our model organ (Chambers et al., 1995, tions. The imaging modality that best fit
2002). Liver metastases are common from the biological requirements of these studies
primary tumors in the colon and breast, was high-frequency ultrasound. High-
and are a frequent contributor to both frequency ultrasound offers cost effective,
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 373

high-resolution imaging with rapid image the 20–60 MHz range. Increasing the fre-
acquisition and processing, and allows for quency restricts imaging penetration to
the non-invasive longitudinal study of liver 5–15 mm, which is sufficient for many
metastasis models without the requirement preclinical applications in mouse models.
for contrast agents. Imaging penetration does limit clinical
use of high-frequency ultrasound to very
specialized applications, such as ocular
METHOD: HIGH-FREQUENCY or intravascular imaging, where the ultra-
ULTRASOUND sound transducer can be placed in close
proximity to the tissue of interest (Foster
Ultrasound images are produced by send- et al., 2000, 2002).
ing short duration acoustic pulses from The exact specifications of a high-fre-
the ultrasound transducer into the tissue of quency ultrasound system will depend on
interest. When the sound wave encounters the design and vary slightly between sys-
an interface between two tissues hav- tems. The system that we have utilized for
ing different acoustic properties, some of preclinical cancer studies is the Vevo 660
the energy is reflected back towards the produced by VisualSonics Inc. The design
transducer. The time it takes for the echo and performance of this scanner has been
to return to the transducer allows for the previously reviewed (Foster et al., 2002).
calculation of the distance to this interface. In brief, the Vevo 660 transducer operates
The resulting image displays the location at a 40 MHz center frequency with a 6 mm
and magnitude of sound reflection at these focal distance. The spatial resolution at the
interfaces. focus is 40 × 80 × 80 μm3.
In ultrasound imaging there is an Similar to the clinical situation, the
inherent tradeoff between the resolution acquisition of high quality preclinical
and depth of penetration. As the ultra- images with high-frequency ultrasound
sound frequency increases, the resolution depends on appropriate preparation of the
increases, while the imaging penetration subject to be imaged and the technical
decreases. For this reason, clinical ultra- experience of the sonographer. In order
sound is often performed in the range of to image the mouse liver, the abdomen
3–12 MHz, allowing for a resolution of must first be depilated with commercial
0.5–2.0 mm and an imaging depth in the hair removal cream. The mouse must be
range of 5–10 cm. For preclinical imag- lightly anesthetized to keep the animal
ing, mouse organs have linear dimen- immobile during imaging, a task most
sions that are approximately tenfold easily accomplished with an inhaled anes-
smaller than humans (Foster et al., 2002). thetic such as 1.5% isoflurane in oxygen.
To resolve the same tissue characteristics Unlike clinical imaging, which uses a
seen in the clinical setting, the preclinical very thin layer of ultrasound contact gel
ultrasound system must achieve a resolu- to allow transmission of sound waves into
tion that is approximately tenfold greater the body, when imaging a small animal a
than the clinical systems. This increase 0.5 cm thick layer of ultrasound contact
in resolution is achieved by increasing gel is used. This allows the transducer
the frequency of the sound waves into to slide easily over the curvature of the
374 K.C. Graham et al.

abdomen without losing contact with the shape and thus would be expected to give
gel. Ultrasound is strongly reflected by a more accurate measurement of tumor
bone and air-filled structures, which hin- volume. Indeed, previous work with a
ders imaging of any tissues located below clinical ultrasound system has shown that
bony structures or the lungs. In the evalu- three-dimensional imaging is more accu-
ation of the mouse liver, we have found rate than two-dimensional imaging for
that the liver is partially obscured by the phantoms of regular and irregular shapes
ribcage, but significant portions of the left (Tong et al., 1998; Xu et al., 2003).
lateral, left medial, and right medial liver To acquire three-dimensional ultrasound
lobes are accessible for imaging (Graham images with the Vevo 660, the transducer
et al., 2005). The liver lobes move in rela- is fixed to a stepper motor and acquires
tion to the bony anatomy and in relation two-dimensional images at 30 μm inter-
to each other over time. This movement vals as it moves in the out-of-plane dimen-
necessitates landmarks internal to each sion. The two-dimensional images are
liver lobe to be noted during longitudinal then interpolated and a three-dimensional
imaging. For liver metastases, landmarks image is reconstructed on-line (Fenster
such as the specific liver lobe in which et al., 2001). A typical 3-D image of
the metastasis develops, tumor shape, and the mouse liver containing a metastasis
proximity to readily identifiable structures of interest would take ∼20 s to acquire
such as major blood vessels and liver lobe and reconstruct. This rapid processing of
edges may be used to reliably identify an the imaging data is advantageous to the
individual metastasis over multiple imag- researcher as it allows the acquired image
ing sessions. to be evaluated and approved before mov-
ing on to the next subject. Such rapid
analysis is not possible with many other
THREE-DIMENSIONAL imaging modalities.
IMAGING AND VOLUME We have compared the calculated volume
CALCULATION of preclinical liver metastases after two-
dimensional measurement and assumption
A calculation of the volume of a region of an ellipsoid volume with the volume
of interest from two-dimensional ultra- calculated after segmentation of three-
sound images involves an assumption of dimensional images. We have shown that,
a defined three-dimensional shape. For for murine B16F1 melanoma liver metas-
instance, when calculating the volume of tases, the two-dimensional method on
a tumor in preclinical models, the tumor is average yields a smaller value than the
often assumed to have an ellipsoid volume three-dimensional method with a mean
that can be calculated after measurement percent difference of 8.8 ± 23.5% (Graham
of the length and diameter of the tumor. et al., 2005). The large standard deviation
The accuracy of this method is dependent indicates that the two-dimensional method
on how closely the tumor of interest can yield large overestimations or under-
matches the assumed shape. The ability to estimations of tumor volume in compari-
acquire three-dimensional images elimi- son to the three-dimensional method. To
nates the need to assume a predetermined determine which method is more accurate,
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 375

the true tumor volume of each metastasis variation (COV), the standard deviation
would have to be known. There is currently divided by the mean volume measure-
no method of measuring tumor volume that ment, for each set of volume measure-
is accepted as the “gold standard” to which ments. For B16F1 liver metastases with
other methods can be compared. Tumor volumes less than 2 mm3, the COV was
dimensions seen through non-invasive 6.8 ± 2.6% (Hastie et al., 2004). For
imaging may be compared to serial histo- HT-29 liver metastases, with volumes
logical sections to gain a sense of whether less than 2 mm3, the COV was 13 ± 5.7%
the two methods yield similar results, but (Hastie et al., 2004). The larger measure-
values obtained through histology cannot ment variability in the HT-29 model, in
be considered the true values because the comparison to the B16F1 model, may
extent of tissue distortion during histologi- reflect the more complex geometries of
cal processing is unknown. Without such the HT-29 metastases and the superior
a “gold standard”, the accuracy of any delineation of tumor boundaries in the
volume measurement cannot be known, but B16F1 model (Hastie et al., 2004).
it is reasonable to assume that the three- Since a measured change in tumor vol-
dimensional method is more accurate in ume is only significant if the change is
measuring tumor volume. The improved greater than the measurement variation,
accuracy of the three-dimensional method knowledge of measurement variability and
reflects the ability to measure complex tumor growth rate can be used to ensure
shapes, and the three-dimensional segmen- that imaging is only performed when
tation is done offline, providing more time significant changes in tumor volume are
for the operator to evaluate the images. In expected. In the case of B16F1 metastases,
the two-dimensional method, the operator with an average doubling time of 1.2 days
must quickly decide which imaging plane (Graham et al., 2005) and a measure-
contains the maximum tumor diameter, a ment variability of 6.8 ± 2.6%, signifi-
decision that may change between observers cant changes in tumor volume could be
or even for the same observer over multiple detected with daily imaging sessions. The
imaging sessions. HT-29 metastases show variable growth
For a three-dimensional imaging modal- rates, with doubling times between 3.7 and
ity to be useful for longitudinal studies 4.8 days (Graham et al., 2005) and a meas-
of tumor progression, the tumor volume urement variation of 13 ± 5.7%. In this
measurement must be accurate and con- case, daily imaging may be unnecessary,
sistent. The variability in repeated volume with more time necessary to ensure sig-
measurements of the same tumor will nificant changes in tumor volume between
dictate the minimum change in volume imaging sessions.
that can be measured. We have measured In studies of tumor growth or treatment
the intraobserver variability in repeated response, it is advantageous to use an
volume measurements of murine B16F1 imaging modality and volume measure-
melanoma and human HT-29 colon car- ment method with the smallest possible
cinoma cells growing as liver metastases. measurement variability so that small
Intraobserver measurement variability is changes in volume can be distinguished.
frequently reported by the coefficient of The reported coefficients of variation for
376 K.C. Graham et al.

tumor volume measurement with high- on the ultrasound images. The minimum
frequency ultrasound, between 6.8% and metastasis size that has been detected
13% depending on the tumor model, are by ultrasound imaging, and confirmed
similar to the clinical results obtained for through histological methods is (maxi-
the measurement of liver tumor volumes mum diameter → volume) 0.22 mm →
with MRI. It has been reported that quan- 0.01 mm3, 0.47 mm → 0.03 mm3, 0.66 mm
tification of tumor volumes from MRI by → 0.08 mm3 and 0.78 mm → 0.17 mm3
manual segmentation had an intraobserver for B16F1, HT-29, MDA-MB-435/HAL
COV of 9.3%, while volume measurement and PAP2 tumor, respectively (Graham
by point counting and stereological cor- et al., 2005). For the B16F1 model a
rection had a COV of 8.4% (Mazonakis large number of mice have been imaged
et al., 2004). Studies of measurement vari- throughout the early stages of metastasis
ation among preclinical imaging modali- development, thus the reported detection
ties are notably absent from the literature, threshold may be at or near the true detec-
which is an issue that needs to be addressed tion limit. For the other three models the
to ensure the proper use of these emerging absolute detection limit may be less than
technologies. the reported values, because only a limited
number of animals were imaged over vary-
ing time frames.
The experimental detection limits of
HIGH-FREQUENCY high-frequency ultrasound compare favo-
ULTRASOUND IMAGING rably to many of the other non-invasive
OF PRECLINICAL LIVER imaging modalities. It has been reported
METASTASES that contrast-enhanced micro-CT and
contrast-enhanced micro-MRI may detect
Using experimental metastasis models, we liver metastases as small as 0.3 mm in
have shown that high-frequency ultrasound diameter (Kobayashi et al., 2001; Weber
can reliably detect murine liver metastases et al., 2004). Non-invasive optical imaging
(Graham et al., 2005). We have shown that with green fluorescent protein may detect
ultrasound detection of liver metastases extremely small tumors, but imaging sen-
shows excellent agreement with post-mor- sitivity is dependent on tissue depth. For
tem analysis, including gross pathology liver metastasis models, we identified the
and histological sections (Figure 29.1). majority of the liver metastases at the
Ultrasound detection of liver metastases 2–3 mm depth range. It has been reported
was demonstrated with four cell lines of that for green fluorescent protein (GFP)-
different primary tumor origin, includ- labeled tumors at a depth of 2.2 mm, the
ing B16F1 (murine melanoma), HT-29 minimum tumor size detected is 1.86 mm
(human colon carcinoma), MDA-MB-435/ (Yang et al., 2000).
HAL (human breast carcinoma), and PAP2 It is important to note that unlike micro-
(murine oncogene-transformed fibrob- CT, micro-MRI or optical methods, the
lasts). With all four models, liver metas- imaging protocol to identify liver tumors
tases were hypoechoic, appearing darker by high-frequency ultrasound does not
than the surrounding liver parenchyma, involve the use of genetically encoded or
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 377

Figure 29.1. Identification of B16F1 (a) and HT-29 (b) liver metastases by non-invasive ultrasound
imaging. Two-dimensional ultrasound images show excellent agreement with corresponding histologi-
cal sections. The maximum diameter of the metastasis shown in the ultrasound image is 1.56 mm (a)
and 0.47 mm (b). Bar on the ultrasound images, 1.00 mm. Bar on histological sections, 1.00 mm (a) and
0.10 mm (b). (Reprinted with permission from Graham et al., 2005.)

exogenously given contrast agents. The contrast agent may also limit longitudi-
inherent tumor to liver parenchyma con- nal imaging. In the case of genetically
trast seen in ultrasound imaging offers encoded marker proteins, such as GFP, the
significant advantages in terms of animal necessity of introducing a foreign reporter
throughput and in the number of animal gene into each cell line to be studied may
models able to be used. For ultrasound limit the number of animal models that
imaging, each animal can be anesthetized, can be utilized. Additionally, the expres-
prepared, imaged, the images reconstructed sion of a foreign reporter protein can lead
and the content reviewed, and the animal to increased immunogenicity of the tumor
recovered in a time window of 15 min cell line being studied (Steinbauer et al.,
per animal. The use of a contrast agent, 2003).
usually delivered intravenously, may be The high-throughput, detection sensitivity
technically difficult and time consum- and the inherent tumor to liver parenchyma
ing, which decreases animal throughput. contrast makes high-frequency ultrasound
The clearance kinetics and toxicity of a uniquely suited for studying the longitudinal
378 K.C. Graham et al.

growth of preclinical liver metastases. volume doubling time between 4 and 5


We have demonstrated the longitudinal days (Graham et al., 2005). A third liver
tracking of individual metastases in the metastasis, imaged during the same 10-day
B16F1, HT-29 and MDA-MB-435/HAL time period, did not appreciably increase in
models (Graham et al., 2005). Individual volume, remaining at a constant 0.03 mm3
metastases were not monitored in the (Figure 29.2). The longitudinal study of
PAP2 model since in this highly aggres- individual metastases allows the identifi-
sive model, individual metastases grow cation of metastases with variable growth
and fuse very rapidly. In this case, tumor patterns for further investigation.
growth may be evaluated by assessing From a clinical perspective, the identi-
total tumor burden at various time points. fication and study of dormant metastases,
It is reasonable to speculate that the tumor as defined by their constant volume, would
burden seen in the percentage of the liver be of great interest. The mechanisms by
visible for ultrasound imaging reflects which metastatic growth is controlled to
the status of the entire liver, but this has produce dormant metastases could poten-
not yet been shown. For studies in which tially be harnessed to produce a clinically
analysis of individual metastases is dif- relevant treatment to slow or halt meta-
ficult or not required, it may be more static progression. Alternatively, the trig-
appropriate to use an imaging modality gers and mechanisms which lead a once
that gives a rapid quantification of total dormant metastasis to reinitiate progres-
tumor burden, such as bioluminescence or sive growth are also of great clinical interest.
one that gives three-dimensional images In some cases metastases have been
unimpeded by the ribcage, such as con- known to occur many years after initial
trast-enhanced CT. diagnosis, with tumor dormancy being
Although imaging modalities that give a a likely explanation for the large time
rapid assessment of tumor burden, such as interval (Demicheli et al., 1998; Karrison
bioluminescence, are invaluable for many et al., 1999). It has been proposed that
high throughput studies, they are unable
to resolve individual metastases in close
proximity. The ability to image and study
individual liver metastases through high-
frequency ultrasound imaging is a power-
ful new tool that can be used to address
many unanswered questions regarding the
metastatic process. For example, through
longitudinal study of liver metastases we
have shown that individual metastases
within a single animal may have widely
varied growth rates. The most striking Figure 29.2. Tracking the growth of individual
liver metastases by non-invasive ultrasound imag-
example of this is with the HT-29 (colon
ing. Two HT-29 liver metastases are rapidly grow-
carcinoma) liver metastasis model. In this ing, as a third metastasis, in the same animal,
model a single animal developed two liver remains at a constant volume. (Modified with
metastases that were proliferating with a permission from Graham et al., 2005.)
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 379

tumor dormancy may be the result of a be used to image tumor development in


dynamic balance between apoptosis and any anatomical site that is not obscured by
proliferation in preangiogenic microme- air-filled structures, such as lungs, or by
tastases (Holmgren et al., 1995; Udagawa bony structures, such as the rib cage.
et al., 2002). In this theory, a micrometas- High-frequency ultrasound has been
tasis is able to escape dormancy and switch used to identify and track the longitudi-
to progressive growth by recruiting a func- nal growth of prostate tumors in a trans-
tional blood supply. Longitudinal imaging is genic model of prostate adenocarcinoma
imperative in order to evaluate this theory in (Wirtzfeld et al., 2005). Non-invasive
the context of metastatic models. imaging is particularly useful for trans-
Apart from the issue of tumor dormancy, genic prostate tumor models because the
the study of treatment response and treatment study of tumor progression or therapeutic
resistance would also benefit from non-inva- response in these models is hindered by
sive imaging. The ability to analyze growth the wide variability in the timing of tumor
rates of individual metastases, with image- development. This variability in tumor
guided isolation of the tumors of interest, development necessitates repeated and
would enable many studies on the molecular long-term imaging of the experimental
basis of tumor response and tumor resistance, animals, which in turn makes ultrasound
including an assessment of heterogeneity imaging, with rapid image acquisition and
in responsiveness of individual metastases. non-ionizing radiation, particularly suited
Although the biological question may vary, for the evaluation of these models.
the unifying theme in all of these studies Non-invasive imaging is the only method
is that non-invasive longitudinal imaging, that enables longitudinal study of tumor
capable of resolving individual micrometas- growth in internal organs. This is not the
tases, is imperative for preclinical study of case in preclinical models that utilize
many clinically relevant issues. tumors grown subcutaneously, intrader-
mally, or in the mammary fat pad, for
which calipers may be used to acquire
longitudinal measurements. Although con-
OTHER APPLICATIONS venient, the ease of caliper measurement
OF HIGH-FREQUENCY must be balanced against the volume over-
ULTRASOUND IMAGING estimation that arises from including the
TO PRECLINICAL CANCER skin and other tumor surrounding tissue
RESEARCH layers in the caliper measurement and
against the fact that many tumors do not
The characteristics of high-frequency conform to the assumed ellipsoid shape
ultrasound imaging – high spatial resolu- for volume calculation. For these reasons,
tion, rapid image acquisition, economical high-frequency ultrasound has been pro-
capital and maintenance costs, and good posed as a time efficient imaging modality
soft tissue contrast without the requirement to increase the accuracy of these preclini-
for contrast agents – make ultrasound an cal studies (Cheung et al., 2005; Turnbull
appealing modality for the study of many et al., 1996). With detection of tumors as
preclinical cancer models. Ultrasound may small as 0.25 mm in width, ultrasound can
380 K.C. Graham et al.

also detect and track tumor development (Goertz et al., 2003), as this technique is
from a much earlier stage than caliper less sensitive to image artifacts than color
measurement (Cheung et al., 2005). The Doppler (Lencioni et al., 1996).
continued development of tumors past Many cancer therapies induce apoptosis,
a size of 1 mm is thought to rely on the an active response to cellular injury that
formation of a neovasculature through the results in characteristic changes to the cell
process of angiogenesis. Thus, the ability nucleus and membrane, eventually leading
to detect and track small tumors may be to cell death. The ability to non-invasively
useful for studying biological processes, quantify apoptosis would be useful in pre-
such as angiogenesis, that occur early in clinical studies to assess the efficacy of
tumor development. novel cancer therapeutics. To date, quan-
Beyond the study of tumor growth, tification of apoptosis is largely limited to
angiogenesis research would benefit from biochemical analysis on harvested tissues,
non-invasive methods of quantifying vas- although single photon emission computed
cular parameters. For determination of tomography with radiolabeled annexin V
red blood cell velocity, Doppler ultra- has shown promise for non-invasive in vivo
sound may be used. Doppler ultrasound is analysis in the clinical setting (Blankenberg
based on the principle that when incident et al., 1998; Narula et al., 2001). For preclin-
ultrasound waves hit a moving object the ical research, the low resolution of SPECT
frequency of the return signal is shifted in imaging may be limiting, so alternative
proportion to the velocity of the object. In methods of quantifying apoptosis may be
color Doppler ultrasound, the frequency required. In a series of in vitro experiments,
shift in the return signal from moving red utilizing cell pellets of human acute myeloid
blood cells is displayed by mapping the leukemia cells, it has been demonstrated
frequency shifts to a blue-red color map that the structural changes associated with
with positive frequency shifts being red apoptosis may be detectable by analyzing
and negative frequency shifts being blue the radiofrequency backscatter signal in
in the resulting image. In an alternative high-frequency ultrasound (Kolios et al.,
method of processing Doppler frequency 2002; Tunis et al., 2005). It is not yet known
shift data, termed Power Doppler, the if the in vitro results will be translatable to
magnitude of frequency shift, and hence the more complex in vivo model, where the
red blood cell velocity, is ignored. In this presence of tissue structures other than the
method, all frequency shifts contribute tumor cells may affect the method of analysis
equally to the display data, meaning the (Kolios et al., 2002).
displayed value is related to the number
of red blood cells moving through a given
area. The utility of high-frequency power FUTURE DEVELOPMENTS –
Doppler ultrasound in the non-invasive ULTRASOUND CONTRAST
evaluation of anti-vascular therapies has AGENTS
been demonstrated in an intradermal
melanoma model (Goertz et al., 2002). It The use of high-frequency ultrasound in
is likely that Power Doppler will be fur- the identification of primary and metastatic
ther developed for high-frequency systems tumors, and in the longitudinal evaluation
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 381

of tumor progression, is well documented between 250 and 1,000 nm, have also been
(Graham et al., 2005; Wirtzfeld et al., investigated (Couture et al., 2006; Lanza
2005). These studies utilized the inherent et al., 1997).
ultrasound contrast, present between tumor Sensitivity of microbubble detection
and normal tissue, to delineate the tumors. may be further enhanced based on the
As outlined in this review, the longitudinal non-linear reflection of ultrasound by
evaluation of tumor growth allows for the microbubbles. This characteristic allows
study of a variety of processes, including the echoes produced from microbubbles
tumor dormancy and treatment response, to be differentiated from those of normal
and allows the researcher greater con- tissue using appropriate signal processing
trol when using models that exhibit large techniques. On clinical ultrasound systems
variability in the timing of tumor develop- this is often accomplished by transmitting
ment. Although the ability to study tumor at one frequency and receiving signals
growth based on inherent ultrasound con- at integer multiples of the transmit fre-
trast is clearly a significant development quency, referred to as harmonic imaging.
in its own right, the current development The strength of a nonlinear echo at a given
and application of novel contrast agents is frequency depends on the transmitted fre-
poised to expand the utility of preclinical quency and on the size of the microbubble.
ultrasound imaging in the fields of vas- For preclinical high-frequency ultrasound,
cular imaging, molecular imaging, and with a 40 MHz transmitting frequency, a
targeted drug delivery. microbubble will produce a much stronger
Inherent ultrasound contrast can be nonlinear echo at one-half of the trans-
enhanced by adding an agent to the imag- mit frequency, the subharmonic, than at
ing field of view that has an acoustic the higher harmonic frequencies. For this
impedance that differs from the tissue reason, subharmonic imaging may be
of interest. Intravenously injected micro- more sensitive than harmonic imaging
bubbles are commonly used to achieve for detecting microbubble contrast agents
this objective. Microbubbles are typically with preclinical imaging systems (Goertz
composed of a gaseous perfluorocarbon et al., 2005).
core surrounded by an outer phospholipid While Doppler ultrasound can provide
or albumin shell. The large difference in valuable information about tumor blood
acoustic impedance between the perfluor- flow, the tissue motion artifacts created
ocarbon gas and the surrounding tissue by the movement of internal organs cur-
leads to greatly enhanced ultrasound signal rently limit the utility of Doppler sys-
reflection and the ability to detect small tems. This limitation may be overcome
numbers of microbubbles in the ultrasound by the development of array transducers
image. Microbubbles are commonly pro- for preclinical systems or through the use
duced with diameters between 1 and 5 μm of intravenously injected microbubbles.
to allow them to pass freely through the Microbubbles enhance the signal from
circulatory system without being arrested blood in B-mode images, and hence are
by size restriction. Recently, the prop- less susceptible to artifacts from tissue
erties of much smaller contrast agents, motion. In addition, continuous imaging
liquid filled nanoparticles with diameters after the contrast agent is administered
382 K.C. Graham et al.

can provide information about perfusion ligand echistatin to target microbubbles


dynamics, including blood volume and to the neovasculature specific integrin
flow rates, that is not available from con- receptor αvβ3 (Ellegala et al., 2003), and
ventional Doppler imaging. Perfusion to image the vasculature in subcutane-
dynamics have been used clinically to ous tumors with a cyclic peptide ligand,
differentiate between different types of arginine–arginine–leucine, which binds to
liver lesions (Albrecht et al., 2004), while a yet unidentified receptor on angiogenic
preclinically, where the lesion is purposely endothelium (Ellegala et al., 2003; Weller
produced, perfusion dynamics hold prom- et al., 2005).
ise as a method of evaluating the efficacy Microbubbles have the potential to be
of novel anti-angiogenic therapeutics (Krix used for both therapeutic and diagnostic
et al., 2003). applications. As toxicity is a major prob-
After intravenous injection, microbub- lem limiting the dose of chemotherapeutic
bles are eventually sequestered in the liver agents, the ability to specifically target
by functional hepatocytes. This results in chemotherapy delivery to the tumor, thereby
enhancement of the normal liver tissue, limiting systemic exposure, is a promising
but not primary liver tumors or metas- avenue for research. Chemotherapeutic
tases, thereby providing enhanced liver compounds can be attached to the outer
parenchyma to liver tumor contrast. This membrane of the microbubble, imbedded
contrast enhancement may be useful for within the membrane, or loaded into the
the delineation of small or poorly defined interior of the microbubble, preventing
tumors (Albrecht et al., 2004). the activity of the compound when the
Ultrasound contrast agents may extend microbubble is in the systemic circula-
the utility of ultrasound from anatomical tion (Unger et al., 2002). Focused high-
imaging to targeted or molecular imag- pressure ultrasound waves can be used
ing (Dayton and Ferrara, 2002; Lindner, to rupture the microbubbles, a process
2004). As with all targeted contrast agents, called cavitation, and thereby release the
the first step is the identification of cell chemotherapeutic. There is emerging evi-
surface receptors that are specific to the dence that microbubble cavitation can lead
tissue of interest. Once a unique receptor to local damage of cellular membranes
is identified, targeted microbubbles may and disrupt capillary integrity (Miller and
be created by conjugation of the receptor- Quddus, 2000; Price et al., 1998). The cel-
specific ligand to the outer surface of the lular damage that results from cavitation
microbubble. After intravenous injection may be detrimental for some applications
the microbubbles will circulate the body of contrast-enhanced ultrasound, but for
and adhere to their target receptors or be targeted cancer therapy this effect may
cleared by the liver. Once the majority of increase the interstitial concentration of
free microbubbles are cleared from the the chemotherapeutic within the tumor,
circulation, the presence of the cell recep- while sparing the normal tissue that is not
tor can be imaged indirectly by detect- exposed to the ultrasound pulse (Unger
ing the presence of the microbubbles. et al., 2002).
This approach has been used to image The principles that are being developed
angiogenesis in rat gliomas by using the for targeted chemotherapeutic delivery
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 383

may also be applied to targeted gene Anwer, K., Kao, G., Proctor, B., Anscombe, I.,
delivery. For gene therapy, the applica- Florack, V., Earls, R., Wilson, E., McCreery,
T., Unger, E., Rolland, A., and Sullivan, S.M.
tion of ultrasound is known to increase
2000. Ultrasound enhancement of cationic
the transfection efficiency of traditional lipid-mediated gene transfer to primary tumors
cationic lipid based transfection complexes following systemic administration. Gene Ther.
(Anwer et al., 2000), although performing 7:1833–1839.
ultrasound in the presence of microbubbles, Blankenberg, F.G., Katsikis, P.D., Tait, J.F., Davis,
causing cavitation and disruption of cellular R.E., Naumovski, L., Ohtsuki, K., Kopiwoda,
S., Abrams, M.J., Darkes, M., Robbins, R.C.,
membranes may be more effective than
Maecker, H.T., and Strauss, H.W. 1998. In vivo
ultrasound alone (Lawrie et al., 2000). In detection and imaging of phosphatidylserine
both of these procedures the application of expression during programmed cell death. Proc.
ultrasound can be localized to a target site Natl. Acad. Sci. U S A 95:6349–6354.
to enhance therapeutic effect, while mini- Cai, S.R., Garbow, J.R., Culverhouse, R., Church,
mizing systemic toxicities. R.D., Zhang, W., Shannon, W.D., and McLeod,
H.L. 2005. A mouse model for developing treat-
ment for secondary liver tumors. Int. J. Oncol.
27:113–120.
CONCLUSIONS Chambers, A.F., MacDonald, I.C., Schmidt, E.E.,
Koop, S., Morris, V.L., Khokha, R., and Groom,
High-frequency ultrasound offers cost A.C. 1995. Steps in tumor metastasis: new con-
effective, non-invasive, high-resolution cepts from intravital videomicroscopy. Cancer
Metastasis Rev. 14:279–301.
imaging with rapid image acquisition and
Chambers, A.F., Groom, A.C., and MacDonald,
processing. These attributes make high- I.C. 2002. Dissemination and growth of can-
frequency ultrasound an attractive option cer cells in metastatic sites. Nat. Rev. Cancer
for the preclinical study of primary and 2:563–572.
metastatic cancer models. The longitudi- Cheung, A.M., Brown, A.S., Hastie, L.A., Cucevic,
nal study of primary tumor development V., Roy, M., Lacefield, J.C., Fenster, A., and
Foster, F.S. 2005. Three-dimensional ultrasound
and metastatic progression may provide
biomicroscopy for xenograft growth analysis.
further insight into dynamic processes Ultrasound Med. Biol. 31:865–870.
such as tumor dormancy and angiogen- Choy, G., Choyke, P., and Libutti, S.K. 2003.
esis. With the development of ultrasound Current advances in molecular imaging: non-
contrast agents for diagnostic and thera- invasive in vivo bioluminescent and fluores-
peutic purposes, high-frequency ultra- cent optical imaging in cancer research. Mol.
Imaging 2:303–312.
sound will be an invaluable tool in the
Couture, O., Bevan, P.D., Cherin, E., Cheung, K.,
preclinical characterization and evalua- Burns, P.N., and Foster, F.S. 2006. Investigating
tion of these agents, eventually leading to perfluorohexane particles with high-frequency
the clinical application of these emerging ultrasound. Ultrasound Med. Biol. 32:73–82.
technologies. Dayton, P.A., and Ferrara, K.W. 2002. Targeted
imaging using ultrasound. J. Magn. Reson.
Imaging 16:362–377.
REFERENCES Demicheli, R., Terenziani, M., and Bonadonna,
Albrecht, T., Hohmann, J., Oldenburg, A., Skrok, G. 1998. Estimate of tumor growth time for
J., and Wolf, K.J. 2004. Detection and charac- breast cancer local recurrences: rapid growth
terisation of liver metastases. Eur. Radiol. 14 after wake-up? Breast Cancer Res. Treat. 51:
Suppl. 8:P25–P33. 133–137.
384 K.C. Graham et al.

Ellegala, D.B., Leong-Poi, H., Carpenter, J.E., Fenster, A., Lacefield, J.C., and Chambers, A.F.
Klibanov, A.L., Kaul, S., Shaffrey, M.E., Sklenar, 2005. Three-dimensional high-frequency ultra-
J., and Lindner, J.R. 2003. Imaging tumor ang- sound imaging for longitudinal evaluation of
iogenesis with contrast ultrasound and micro- liver metastases in preclinical models. Cancer
bubbles targeted to alpha(v)beta3. Circulation Res. 65:5231–5237.
108:336–341. Hastie, L.A., Graham, K.C., Groom, A.C.,
Evelhoch, J.L., Gillies, R.J., Karczmar, G.S., MacDonald, I.C., Chambers, A.F., Fenster, A.,
Koutcher, J.A., Maxwell, R.J., Nalcioglu, O., and Lacefield, J.C. 2004. Variability of three-
Raghunand, N., Ronen, S.M., Ross, B.D., and dimensional high-frequency ultrasound measure-
Swartz, H.M. 2000. Applications of magnetic ments of small tumor volumes. IEEE Ultrason.
resonance in model systems: cancer therapeu- Sympos. Proc. 3:2185–2188.
tics. Neoplasia 2:152–165. Heyn, C., Ronald, J.A., Mackenzie, L.T.,
Fenster, A., Downey, D.B., and Cardinal, H.N. MacDonald, I.C., Chambers, A.F., Rutt, B.K.,
2001. Three-dimensional ultrasound imaging. and Foster, P.J. 2006. In vivo magnetic resonance
Phys. Med. Biol. 46:R67–R99. imaging of single cells in mouse brain with opti-
Foster, F.S., Pavlin, C.J., Harasiewicz, K.A., cal validation. Magn. Reson. Med. 55:23–29.
Christopher, D.A., and Turnbull, D.H. 2000. Holmgren, L., O’Reilly, M.S., and Folkman,
Advances in ultrasound biomicroscopy. J. 1995. Dormancy of micrometastases: bal-
Ultrasound Med. Biol. 26:1–27. anced proliferation and apoptosis in the pres-
Foster, F.S., Zhang, M.Y., Zhou, Y.Q., Liu, G., Mehi, ence of angiogenesis suppression. Nat. Med. 1:
J., Cherin, E., Harasiewicz, K.A., Starkoski, 149–153.
B.G., Zan, L., Knapik, D.A., and Adamson, S.L. Karrison, T.G., Ferguson, D.J., and Meier, P. 1999.
2002. A new ultrasound instrument for in vivo Dormancy of mammary carcinoma after mastec-
microimaging of mice. Ultrasound Med. Biol. tomy. J. Natl. Cancer Inst. 91:80–85.
28:1165–1172. Khanna, C., and Hunter, K. 2005. Modeling metas-
Gambhir, S.S. 2002. Molecular imaging of cancer tasis in vivo. Carcinogenesis 26:513–523.
with positron emission tomography. Nat. Rev. Kobayashi, H., Saga, T., Kawamoto, S., Sato, N.,
Cancer 2: 683–693. Hiraga, A., Ishimori, T., Konishi, J., Togashi,
Gillies, R.J., Bhujwalla, Z.M., Evelhoch, J., K., and Brechbiel, M.W. 2001. Dynamic micro-
Garwood, M., Neeman, M., Robinson, S.P., magnetic resonance imaging of liver micrometas-
Sotak, C.H., and Van Der Sanden, B. 2000. tasis in mice with a novel liver macromolecular
Applications of magnetic resonance in model sys- magnetic resonance contrast agent DAB-Am64-
tems: tumor biology and physiology. Neoplasia (1B4M-Gd)(64). Cancer Res. 61:4966–4970.
2:139–151. Kolios, M.C., Czarnota, G.J., Lee, M., Hunt,
Goertz, D.E., Yu, J.L., Kerbel, R.S., Burns, P.N., J.W., and Sherar, M.D. 2002. Ultrasonic spec-
and Foster, F.S. 2002. High-frequency Doppler tral parameter characterization of apoptosis.
ultrasound monitors the effects of antivascu- Ultrasound Med. Biol. 28:589–597.
lar therapy on tumor blood flow. Cancer Res. Krix, M., Kiessling, F., Vosseler, S., Farhan, N.,
62:6371–6375. Mueller, M.M., Bohlen, P., Fusenig, N.E.,
Goertz, D.E., Yu, J.L., Kerbel, R.S., Burns, P.N., and Delorme, S. 2003. Sensitive noninvasive
and Foster, F.S. 2003. High-frequency 3-D monitoring of tumor perfusion during antiang-
color-flow imaging of the microcirculation. iogenic therapy by intermittent bolus-contrast
Ultrasound Med. Biol. 29:39–51. power Doppler sonography. Cancer Res. 63:
Goertz, D.E., Cherin, E., Needles, A., Karshafian, 8264–8270.
R., Brown, A.S., Burns, P.N., and Foster, F.S. Lanza, G.M., Wallace, K.D., Fischer, S.E., Christy,
2005. High frequency nonlinear B-scan imag- D.H., Scott, M.J., Trousil, R.L., Cacheris, W.P.,
ing of microbubble contrast agents. IEEE Trans. Miller, J.G., Gaffney, P.J., and Wickline, S.A.
Ultrason. Ferroelectr. Freq. Control 52:65–79. 1997. High-frequency ultrasonic detection
Graham, K.C., Wirtzfeld, L.A., MacKenzie, L.T., of thrombi with a targeted contrast system.
Postenka, C.O., Groom, A.C., MacDonald, I.C., Ultrasound Med. Biol. 23:863–870.
29. Preclinical Liver Metastases: Three-Dimensional High-Frequency Ultrasound Imaging 385

Lawrie, A., Brisken, A.F., Francis, S.E., Cumberland, created by targeted microbubble destruction with
D.C., Crossman, D.C., and Newman, C.M. 2000. ultrasound. Circulation 98:1264–1267.
Microbubble-enhanced ultrasound for vascular Steinbauer, M., Guba, M., Cernaianu, G., Kohl,
gene delivery. Gene Ther. 7:2023–2027. G., Cetto, M., Kunz-Schughart, L.A., Geissler,
Leach, M.O. 2001. Application of magnetic reso- E.K., Falk, W., and Jauch, K.W. 2003. GFP-
nance imaging to angiogenesis in breast cancer. transfected tumor cells are useful in examining
Breast Cancer Res. 3:22–27. early metastasis in vivo, but immune reaction
Lencioni, R., Pinto, F., Armillotta, N., and precludes long-term tumor development studies
Bartolozzi, C. 1996. Assessment of tumor vascu- in immunocompetent mice. Clin. Exp. Metastasis
larity in hepatocellular carcinoma: comparison 20:135–141.
of power Doppler US and color Doppler US. Tong, S., Cardinal, H.N., McLoughlin, R.F.,
Radiology 201:353–358. Downey, D.B., and Fenster, A. 1998. Intra- and
Lindner, J.R. 2004. Microbubbles in medical imag- inter-observer variability and reliability of pros-
ing: current applications and future directions. tate volume measurement via two-dimensional
Nat. Rev. Drug Discov. 3:527–532. and three-dimensional ultrasound imaging.
Lyons, S.K. 2005. Advances in imaging mouse Ultrasound Med. Biol. 24:673–681.
tumour models in vivo. J. Pathol. 205:194–205. Tunis, A.S., Czarnota, G.J., Giles, A., Sherar, M.D.,
Mazonakis, M., Damilakis, J., Mantatzis, M., Hunt, J.W., and Kolios, M.C. 2005. Monitoring
Prassopoulos, P., Maris, T., Varveris, H., and structural changes in cells with high-frequency
Gourtsoyiannis, N. 2004. Stereology versus ultrasound signal statistics. Ultrasound Med.
planimetry to estimate the volume of malignant Biol. 31:1041–1049.
liver lesions on MR imaging. Magn. Reson. Turnbull, D.H., Ramsay, J.A., Shivji, G.S.,
Imaging 22:1011–1016. Bloomfield, T.S., From, L., Sauder, D.N., and
Miller, D.L., and Quddus, J. 2000. Diagnostic Foster, F.S. 1996. Ultrasound backscatter micro-
ultrasound activation of contrast agent gas bod- scope analysis of mouse melanoma progression.
ies induces capillary rupture in mice. Proc. Natl. Ultrasound Med. Biol. 22:845–853.
Acad. Sci. U S A 97:10179–10184. Udagawa, T., Fernandez, A., Achilles, E.G.,
Morris, V.L., MacDonald, I.C., Koop, S., Schmidt, Folkman, J., and D’Amato, R.J. 2002. Persistence
E.E., Chambers, A.F., and Groom, A.C. 1993. of microscopic human cancers in mice: altera-
Early interactions of cancer cells with the tions in the angiogenic balance accompanies loss
microvasculature in mouse liver and mus- of tumor dormancy. FASEB J. 16:1361–1370.
cle during hematogenous metastasis: videomi- Unger, E.C., Matsunaga, T.O., McCreery, T.,
croscopic analysis. Clin. Exp. Metastasis 11: Schumann, P., Sweitzer, R., and Quigley, R.
377–390. 2002. Therapeutic applications of microbubbles.
Narula, J., Acio, E.R., Narula, N., Samuels, Eur. J. Radiol. 42:160–168.
L.E., Fyfe, B., Wood, D., Fitzpatrick, J.M., Weber, S.M., Peterson, K.A., Durkee, B., Qi,
Raghunath, P.N., Tomaszewski, J.E., Kelly, C., C., Longino, M., Warner, T., Lee, F.T., Jr.,
Steinmetz, N., Green, A., Tait, J.F., Leppo, and Weichert, J.P. 2004. Imaging of murine
J., Blankenberg, F.G., Jain, D., and Strauss, liver tumor using microCT with a hepatocyte-
H.W. 2001. Annexin-V imaging for noninvasive selective contrast agent: accuracy is dependent
detection of cardiac allograft rejection. Nat. on adequate contrast enhancement. J. Surg. Res.
Med. 7:1347–1352. 119:41–45.
Paulus, M.J., Gleason, S.S., Kennel, S.J., Hunsicker, Weissleder, R. 2002. Scaling down imaging:
P.R., and Johnson, D.K. 2000. High resolution molecular mapping of cancer in mice. Nat. Rev.
X-ray computed tomography: an emerging tool Cancer 2:11–18.
for small animal cancer research. Neoplasia Welch, D.R. 1997. Technical considerations for
2:62–70. studying cancer metastasis in vivo. Clin. Exp.
Price, R.J., Skyba, D.M., Kaul, S., and Skalak, T.C. Metastasis 15:272–306.
1998. Delivery of colloidal particles and red Weller, G.E., Wong, M.K., Modzelewski, R.A.,
blood cells to tissue through microvessel ruptures Lu, E., Klibanov, A.L., Wagner, W.R., and
386 K.C. Graham et al.

Villanueva, F.S. 2005. Ultrasonic imaging of efficacy of the cytosine analogue 1-[2-C-cyano-
tumor angiogenesis using contrast microbubbles 2-deoxy-beta-D-arabino-pentofuranosyl]-N(4)-
targeted via the tumor-binding peptide arginine- palmitoyl cytosine (CS-682) in a liver-metastasis
arginine-leucine. Cancer Res. 65:533–539. athymic nude mouse model. Cancer Res.
Wirtzfeld, L.A., Wu, G., Bygrave, M., Yamasaki, 63:2477–2482.
Y., Sakai, H., Moussa, M., Izawa, J.I., Downey, Xu, H.X., Yin, X.Y., Lu, M.D., Liu, G.J., and Xu,
D.B., Greenberg, N.M., Fenster, A., Xuan, Z.F. 2003. Estimation of liver tumor volume
J.W., and Lacefield, J.C. 2005. A new three- using a three-dimensional ultrasound volumetric
dimensional ultrasound microimaging technol- system. Ultrasound Med. Biol. 29:839–846.
ogy for preclinical studies using a transgenic Yang, M., Baranov, E., Jiang, P., Sun, F.X., Li, X.M.,
prostate cancer mouse model. Cancer Res. 65: Li, L., Hasegawa, S., Bouvet, M., Al-Tuwaijri,
6337–6345. M., Chishima, T., Shimada, H., Moossa, A.R.,
Wu, M., Mazurchuk, R., Chaudhary, N.D., Spernyak, Penman, S., and Hoffman, R.M. 2000. Whole-
J., Veith, J., Pera, P., Greco, W., Hoffman, R.M., body optical imaging of green fluorescent pro-
Kobayashi, T., and Bernacki, R.J. 2003. High- tein-expressing tumors and metastases. Proc.
resolution magnetic resonance imaging of the Natl. Acad. Sci. U S A 97:1206–1211.
30
Colorectal Liver Metastases:
18
F-Fluorodeoxyglucose-Positron
Emission Tomography
Stéphanie Truant, Damien Huglo, and François-René Pruvot

INTRODUCTION findings was also variable from one study


to another, including histopathologic
During the last years, 18F-fluorodeoxyglucose- analy-sis (performed at surgery, biopsy,
positron emission tomography (FDG-PET) and autopsy), intraoperative observation
has been advocated as a useful tool in the (e.g., manual palpation or ultrasonogra-
management of recurrent colorectal can- phy) and/or 6-months or 1-year follow-
cer, mainly in liver and pelvic recurrences, up. The rate of histological confirmation
whereas regarding the primary tumor, that is the gold standard particularly in
FDG-PET has proven to show very poor the evaluation of false-positive and false-
yield in comparison with conventional negative readings varied from < 10% to
diagnostic methods. In the first meta- 95% (Truant et al., 2005). A non-standard
analysis of the literature for whole-body method of image analysis, as well as
FDG-PET detection of recurrent colorectal some degree of inequivalence in FDG-
cancer, FDG-PET had a positive impact PET and computed tomography (CT) per-
on surgical decision-making in 29% of formance, may have led to a misjudgement
patients (Huebner et al., 2000). On the of their relative diagnostic performance,
other hand, in the beginning of FDG-PET, as reflected by high rates of FDG-PET/
most studies on colorectal liver metastases CT discordances (range, 10–31%). FDG-
have shown limitations. Survey of previ- PET was performed prospectively and the
ous reports shows indeed heterogeneous interpretation of the FDG-PET images
study designs that were mostly retrospec- was generally made with a priori knowl-
tive and simultaneously included various edge of results of conventional diagnostic
subsets of circumstances, e.g., preopera- procedures. In contrast, CT was reviewed
tive staging of liver metastases together retrospectively and performed at various
with locoregional recurrence, unexplained institutions, resulting in its sensitivity and
elevated serum carcinoembryonic antigen specificity rates being among the worst,
(CEA) levels and/or symptoms with nega- which were reported in general literature
tive imaging. The methodology to define regarding colorectal liver metastases. In
the true lesion status of the FDG-PET the two meta-analyses performed in the

387
388 S. Truant et al.

beginning of the use of FDG-PET, only host cell and tumor glucose metabolism
few studies fulfilled the inclusion cri- may alter appearance of tumor on FDG-
teria and the guidelines established by PET. Patients with diabetes mellitus may
the authors, reflecting the limitations of have an increased accumulation of FDG in
published reports (Huebner et al., 2000; skeletal muscle related to insulin adminis-
Kinkel et al., 2002). Over time, however, tration and a subsequent decreased uptake
studies on FDG-PET have focused on in malignant tissue, affecting both sensi-
more definite settings and have included tivity and specificity of FDG-PET imaging
more systematically histological verifica- in this patient population. In overweight
tions of FDG-PET findings, resulting in a patients, the significant amount of soft tis-
better assessment of FDG-PET accuracy sue can cause excessive scattering, and it
and impact. In this chapter, the yield of is recommended to increase the injected
FDG-PET in colorectal liver metastases activity and/or the acquisition time for
will be preferentially analyzed through both emission and transmission scans
these latter studies. to improve the image quality. Systemic
therapy such as chemotherapy may also
deteriorate FDG-PET accuracy by signifi-
PRINCIPLE OF FDG-PET cantly decreasing the activity of the key
AND GENERAL PITFALLS glycolytic enzyme hexokinase (Akhurst
et al., 2005). It is, therefore, recommended
FDG-PET is based on the increased uptake to delay FDG-PET scanning for 2–3 weeks
of glucose and, therefore, of FDG in after achieving chemotherapy. Mucinous
cancer cells. Many factors are involved adenocarcinoma and thus mucinous-
in FDG accumulation within a tumor or differentiated metastases use 18F-FDG at
metastases, such as the number of viable the same rate as normal surrounding tis-
tumor cells within a lesion, the amount of sues owing to the relative abundance of
necrotic tissue as well as regional blood mucin and the subsequent relative hypocel-
flow, and tumor hypoxia has proven to lularity of such tumors. The sensitivity of
increase FDG uptake. Besides, FDG is FDG-PET for detection of mucinous ade-
not a specific tracer of malignant cells. nocarcinoma is significantly lower than
The uptake of this tracer may occur in the nonmucinous ones (Whiteford et al.,
any condition associated with increased 2000). Finally, the risk of FDG-PET mis-
tissue metabolism, such as inflammatory interpretation may be aggravated by the
cells – as those observed in nonneoplas- lack of anatomic information. It has been
tic stroma tissues within tumor tissue shown that the recently-developed hybrid
– infection or healing. There also exist PET/CT machines, by better localizing
many sources of physiologic increased FDG uptakes, were able to reduce the rate
activity at FDG-PET, such as uptake in of inaccurate readings of FDG-PET alone.
brain, muscle, thyroid, digestive or urinary The knowledge of these pitfalls empha-
tract. This lack of FDG specificity can sizes the importance of correlating FDG
lead to its false-positive uptake in cancer uptake with clinical findings and results of
patients. Moreover, conditions that alter conventional imaging studies.
30. Colorectal Liver Metastases 389

COLORECTAL LIVER hepatic disease can be achieved. If isolated


METASTASES: YIELD hepatic metastases are suspected, patients
OF 18F-FLUORO- are screened with a CT of the chest and
abdomen to assess resectability and to iden-
DEOXYGLUCOSE-POSITRON tify any extrahepatic disease. Despite this
EMISSION TOMOGRAPHY extensive work-up, 20% patients prove to
have unresectable disease during laparot-
Approximately 50% of patients with color-
omy. Moreover, up to 60% of patients have
ectal cancer eventually develop liver metas-
cancer recurrence within 1 year of curative
tases. In 25% of cases liver metastases are
liver resection, with an equal distribution
synchronous and a further 25% will develop
between liver and extra-hepatic recurrences.
metachronously within 5 years. In 30–40%
This indicates the presence of hepatic and
of those patients, the metastases are still
extrahepatic tumor foci that were not identi-
confined to the liver at the time of detec-
fied by conventional diagnostic methods or
tion, but will be resectable in only 15–20%
at the time of liver surgery. In this setting, it
of patients, with partial hepatectomy being
was hoped that the application of FDG-PET
the only chance for cure. Postoperative
would lead to improved selection of patients
follow-up in association with CEA is man-
for partial hepatic resection by optimizing
datory afterwards to detect recurrences at
the detection of unsuspected metastases
an early and potentially curable stage. In
throughout the body. Contribution of FDG-
patients with nonresectable liver metas-
PET in the preoperative staging of colorectal
tases, systemic chemotherapy with irinote-
liver metastases is successively detailed for
can and/or oxaliplatin regimen, although
the liver, the abdominal cavity and the extra-
never curative, offers a median overall
abdominal organs, as well as specific pitfalls
survival of 2 years in responders or even
encountered in each site. Subsequent impact
more in secondarily-resectable patients. All
on patient’s management is then correlated
of these treatments can be associated with
with survival analysis.
a curative or palliative intent with local
ablative therapy such as radiofrequency
ablation and cryotherapy. The yield of Detection of Liver Metastases
FDG-PET in the management of colorectal
Interesting results have been obtained for
liver metastases has been assessed in these
the detection of metastases in the liver, with
different settings.
sensitivity and specificity of FDG-PET
ranging from 65% to 100% and from 57%
Preoperative Staging of Patients with to 100%, respectively. In the beginning of
Colorectal Liver Metastases FDG-PET use, the first meta-analysis of
A 35–40% 5-year survival rate can be 11 articles and 577 patients with recur-
anticipated after partial hepatectomy in rent colorectal cancer showed an average
patients with colorectal liver metastases, lesion-based sensitivity and specificity of
providing that a complete resection of FDG-PET for detection of liver metastases
liver disease with tumor-free margins and of 91% and 97%, respectively (Huebner
with absence of residual or metastatic extra- et al., 2000). Studies further reported a
390 S. Truant et al.

higher accuracy for FDG-PET compared metastatic lesions were identified by the
with CT or other conventional diagnostic former. Accordingly, another meta-analy-
methods in detecting liver metastases in sis came to the conclusion that FDG-PET
recurrent colorectal cancer (Vitola et al., had a significantly higher sensitivity for
1996a, b). In a second meta-analysis, the detection of colorectal liver metastases on
mean sensitivity of FDG-PET (90%) was a per-patient basis compared with that of
significantly higher than that of ultrasound other modalities such as CT or MRI, but
(55%), CT (72%), and MRI (76%) in the showed comparable sensitivity on a per-
detection of liver metastases from color- lesion basis (Bipat et al., 2005).
ectal, gastric, and esophageal cancer, but This poor sensitivity of the liver is partly
any conclusion was difficult to draw due a result of the relatively high hexokinase
to comparison of 15-year range of publi- activity and thereby high FDG uptake
cation for CT with 3-year range for FDG- of normal hepatocytes. The liver usually
PET (Kinkel et al., 2002). The sensitivity displays a faint and homogeneous FDG
of FDG-PET was comparable to that of the uptake that is worsened by respiratory
most sensitive test for detection of hepatic motion leading to FDG smearing. Large
metastases, i.e., CT portography (90% vs. metastases are generally readily identified,
97%), with a much higher specificity – appearing as discrete foci of increased
expected by the high rate of false-positive activity in the liver or rings of increased
findings of CT portography (100% vs. 9%) activity in the case of necrotic centers.
(Vitola et al., 1996a, b). The adjunctive In contrast, small lesions < 10 mm can be
of intraoperative sonography also limited missed by FDG-PET due to its limited res-
impact on treatment selection in patients olution. The sensitivity of FDG-PET in the
screened preoperatively by FDG-PET. detection of liver metastases was confirmed
Over time however, with improve- to be related to lesion size: only 14–25%
ment in study designs, the superiority of hepatic lesions < 1 cm were detected
of FDG-PET over three phase CT or by FDG-PET in contrast with 80–100%
contrast-enhanced MRI in the detection of lesions > 1 cm, resulting in an overall
of colorectal liver metastases was not rate of undetected lesions of 30% (Fong
confirmed (Truant et al., 2005). This et al., 1999). Preoperative chemotherapy
was particularly true in studies using has proven to significantly alter tumor FDG
a systematic histological verification of uptake, resulting in rates of undetected
PET/CT discordances and an analysis lesions as high as 37% and possible missed
by lesion that appeared to be more lesions > 3 cm (Akhurst et al., 2005).
accurate than an analysis by patient. In FDG-PET is also unable to detect muci-
lesion-by-lesion analyses of resected liver nous-differentiated metastases owing to
specimens, FDG-PET demonstrated only the relative hypocellularity of such tumors.
70–79% of liver metastases, namely not Another major drawback of FDG-PET
better than conventional diagnostic meth- regarding liver metastases detection is the
ods (Truant et al., 2005). Rohren et al. lack of anatomic details, such as anatomi-
(2002), in comparing FDG-PET with sur- cal landmarks and relationship to hepatic
gical inspection and intraoperative ultra- vasculature. This method is often unhelp-
sound, found that only 79% of hepatic ful in evaluating anatomical resectability of
30. Colorectal Liver Metastases 391

Detection of Extrahepatic Metastases


liver metastases. Inaccurate localization of
hepatic or extra-hepatic lesions positioned The preoperative work-up of presumed
in close proximity to the liver edge may resectable liver metastases generally
further lead to false-positive or false nega- includes CT of the abdomen and the chest,
tive results, e.g., FDG-PET is unable to associated in case of symptoms with guided
discriminate between lesions close to each investigations such as bone scintigraphy,
other, an overlapping loop of bowel can brain CT, transrectal endoscopic ultra-
occasionally resemble a focal liver lesion. sonography and/or pelvic MRI. Besides
On the other hand, false-positive findings a high accuracy in the detection of liver
of FDG-PET in the liver may rarely occur, and lung metastases, CT usually fails to
for liver lesions that were found in the detect abdominal extrahepatic metastases
published series to represent granuloma, that may be a contraindication to hepate-
liver cyst, adenoma, fibronodular hyper- ctomy and are the main cause of unneces-
plasia or even normal hepatic parenchyma. sary laparotomy in this patient population.
For the latter circumstance, multiple fac- Another important issue is the diagnos-
tors have been involved, such as focal fat tic of locoregional recurrence in patients
infiltration, regionally altered blood flow, undergoing anterior or abdominoperineal
intrinsic heterogeneity of hepatic paren- resection who develop a fibrotic mass in
chyma or image artifact. These benign the presacral operative bed aggravated
hepatic lesions have generally poor FDG by preoperative radiation therapy. These
uptake, except for abscesses. The combi- changes may appear on CT images for
nation of the FDG-PET system with a CT many years and be indistinguishable from
(integrated PET/CT system) was assumed tumor recurrence. With conventional imag-
to avoid these limitations by better local- ing, serial examinations are frequently
izing FDG-PET abnormalities. Actually, required, and the diagnosis of disease
in 76 patients evaluated for resection of recurrence may take 3–6 months or more
liver metastases, PET/CT was equiva- before slowly developing changes become
lent to contrast-enhanced CT for detect- apparent.
ing intrahepatic metastases, as expected, Regarding these extrahepatic sites of
as CT imaging without intravenous con- recurrence, FDG-PET has the advantage
trast does not allow visualization of many over anatomic imaging by providing a
hepatic metastases (Selzner et al., 2004). functional screening of the whole-body
However, PET/CT was superior in estab- and by detecting metabolic changes irre-
lishing the diagnosis of intrahepatic recur- spective of the morphological changes.
rences in patients with prior hepatectomy In the preoperative staging of colorectal
(specificity 50% vs. 100%). In conclusion, liver metastases, FDG-PET accuracy for
although the ability of FDG-PET to detect the detection of extrahepatic recurrences
hepatic lesions has been established, its was difficult to assess. This was due to the
superiority to CT or MRI has not been variability of lesion incidence among the
shown. Due to its limited anatomic reso- studied population, of lesion(s) size and
lution, FDG-PET is unlikely to be a sub- localization and of methodology to define
stitute for conventional imaging but may the true lesion status from one study to
rather complement it. another. Nevertheless, it is now established
392 S. Truant et al.

that FDG-PET is a valuable tool for the of the abundance of the lymphocyte cells
detection of extrahepatic metastases. in the region, which are very glucose avid.
Benign causes of FDG accumulation also
Abdominal Cavity exist, such as fistulas, abscesses, diver-
In overall abdomen, FDG-PET has shown ticulitis, inflammatory bowel disease, and
high rates of sensitivity (63–100%), specifi- adenoma. As a rule, diffuse colonic FDG
city (98–100%), and accuracy (93–100%) uptake pattern, regardless of degree, is
(Flamen et al., 1999; Truant et al., 2005). predictive of normal findings at colonos-
FDG-PET was more sensitive than CT copy, whereas a segmental high uptake
(sensitivity 25–57%), while specificity may imply an inflammatory condition.
rates were comparable (CT specificity of In contrast, focal intense activity in the
91–99%). These results were conditioned colon is predictive of pathologic find-
by the prior knowledge of the specific ings and should be further evaluated with
pitfalls and artifacts encountered in the colonoscopy to exclude a neoplastic proc-
abdominal cavity owing to the many sources ess. Moderate focal FDG uptake can also
of physiologic and benign increased activ- be seen at stomas sites or in laparotomy
ity at FDG-PET, such as uptake in urinary scars. Such increased FDG uptake associ-
and gastrointestinal tract. ated with healing may persist for several
months. Because FDG is excreted in the
Physiologic and Benign Increased Activity of urine, intense FDG activity is encountered
FDG in the Abdominal Cavity
in the intrarenal collecting systems, ure-
Increased activity can be seen in most ters, and bladder. Other benign uptakes
segments of the digestive tract. The are possible, such as increased activity of
mechanism underlying this phenomenon FDG in the uterus during menstruation,
is unclear, partly due to smooth muscle inflammatory changes within the ovary
activity associated with peristalsis, meta- during ovulation, hyperventilation-induced
bolically active mucosa, and bacterial uptake in the diaphragm, or aortic FDG
uptake. Gastric activity can be seen and accumulation due to atherosclerosis. All of
has a characteristic J-shape. Uptake along these sites have potential for tumor recur-
the esophagus is also common, especially rence, and in the case of abdominal 18F-
in the distal portion and at the gastro- FDG uptake, correlation with clinical and
esophageal junction; the esophagus is best CT findings, as well as consideration for
identified on frontal and sagittal views. A biopsy, are mandatory. In case of inflam-
physiologic uptake can also occur in the matory or infectious processes, repeated
small bowel and is usually identified by FDG-PET, in two to four months or after
its pattern that has often a linear “string of a course of appropriate therapy (e.g., anti-
beads” appearance. However, more limited biotics), will often show an absence of or
bowel uptake, which forms discrete foci substantial decrease in uptake intensity at
of increased activity, can be impossible the site. The use of laparoscopy to assess
to distinguish from tumor deposits. The intra-abdominal burden before laparotomy
colonic activity is also variable. Uptake should also be recommended but is not
in the caecum and right colon is usually always possible, given existing adhesions
higher than in the rest of the colon because and the extent of previous surgery.
30. Colorectal Liver Metastases 393

Contribution of FDG-PET imaging findings mainly profiting (Flamen


In the abdominal cavity, FDG-PET has et al., 1999). Importantly especially for this
proven to be especially useful in areas in particular setting, FDG-PET has also shown
which CT has failed to demonstrate high good positive and negative predictive val-
accuracy due to limited anatomic resolu- ues (76% and 92%, respectively) (Moore
tions, such as locoregional recurrence of et al., 2003).
rectal cancer, peritoneal metastases or dis- One must nevertheless keep in mind the
tant lymph nodes metastases. specific pitfalls and limitations of FDG-
PET for diagnosis of pelvic recurrences.
FDG can not differentiate malignant tis-
Pelvic Recurrence of Rectal Cancer
sues from inflammatory process, such as
In spite of radiation therapy and total postoperative chronic fistula, suggesting
mesorectal excision, locoregional recur- to delay data of acquisition to aid differ-
rence occurs in 10–30% of patients and entiation of inflammatory from malignant
is asymptomatic in up to 40% of patients. lesions. Increased FDG uptake following
Locoregional recurrences are a contraindi- radiation may also be due to inflamma-
cation to surgical resection of liver metas- tory changes and is not always associ-
tases except when resectable, although ated with residual tumor. The time course
attempted curative resection often causes of postirradiation FDG activity has not
extreme morbidity with a low chance of been studied systematically; however, it
long-term survival. In contrast to CT that is generally accepted that FDG activity
has low specificity, FDG-PET has proven present six months after completion of
to be particularly valuable in distinguish- radiation therapy most likely represents
ing between local recurrence and scar tis- tumor recurrence, while positive predic-
sue after rectal excision (Figure 30.1). tive value and accuracy have proven to
Most series in recurrent cancer reported improve in FDG-PET imaging performed
high rates of sensitivity and specificity of more than 12 months after completion of
FDG-PET for the diagnosis of locoregional radiation (Moore et al., 2003). Similar to
recurrence, ranging from 84% to 100% anatomic imaging modalities, the altered
and from 80% to 100%, respectively pelvic anatomy associated with surgical
(Moore et al., 2003). The sensitivity of removal of primary rectal tumors may
FDG-PET in detecting locoregional recur- have an effect on the interpretation of
18
rence compared favorably with that of CT F-FDG-PET images, especially in the
and was even higher than that for CT plus differentiation between tumoral 18F-FDG
colonoscopy (90% vs. 71%, respectively) uptake and physiologic 18F-FDG uptake in
(Whiteford et al., 2000). Additional diag- the gastrointestinal or genitourinary tract.
nostic value of FDG-PET in the detection For optimal evaluation of the pelvis, the
of pelvic recurrence was found in 56% bladder should be empty. The use of furo-
of patients if only CT findings were con- semide or even bladder catheterization can
sidered and in 20% of patients if both CT occasionally be helpful in avoiding focal
and transrectal endoscopic ultrasonography ureteral activity. The integrated PET/CT
findings were considered with the subgroup system may overcome the difficulties of
of patients with inconclusive conventional FDG-PET image interpretation associated
394 S. Truant et al.

Figure 30.1. A 68-year-old man with prior colectomy for carcinoma presented with two liver metastases.
Preoperative FDG-PET (b, c) showed not only the two already-known liver metastases but also a small
left pelvic uptake, suggesting local recurrence. In contrast, conventional workup by pelvic CT (a) and
transrectal endoscopic ultrasonography was negative. The patient underwent an exploratory laparotomy
that provided a histological confirmation of local recurrence

with the distorted pelvic anatomy. In 62 were 98%, 96%, 90%, 97%, and 93% for
patients, sensitivity, specificity, positive PET/CT and 82%, 65%, 73%, 75%, and
predictive value, negative predictive value 74% for PET, respectively (Even-Sapir
and accuracy for differentiating malignant et al., 2004). In that study, the most common
from benign 18F-FDG uptake in the pelvis cause for false-positive interpretation of
30. Colorectal Liver Metastases 395

PET findings was physiologic 18F-FDG value of FDG-PET in the preoperative


uptake in displaced pelvic organs. Thus, staging of liver metastases, avoiding futile
although FDG-PET is useful in distin- laparotomy, though there is no specific
guishing viable tumor from necrosis or study on FDG-PET accuracy in this set-
fibrosis in residual masses, the decision ting. As metastatic lymph nodes are fre-
to administer salvage therapy should be quently small and often contain small
made after the positive FDG-PET finding quantities of tumor cells, it is likely that
has been confirmed by biopsy. results may depend on their localization
and their size. False-negative results may
Peritoneal Metastases occur but are difficult to identify given the
Peritoneal dissemination is a major cause impossibility of a systematic histological
of unresectability of liver metastases, but verification. Nevertheless, studies on over-
is not easily detected at an early stage by all recurrent colorectal cancer have shown
anatomic imaging modalities. Few studies sensitivity rates of FDG-PET ranging from
have focused on FDG-PET contribution 65% to 100% for the detection of lymph
in the detection of peritoneal metastases. nodes metastatic to the groin, retroperi-
The largest one which included 23 patients toneum or portal triad (Lai et al., 1996).
with recurrent colorectal cancer was carried FDG-PET sensitivity was two to fourfold
out by Tanaka et al. (2002). In this study, that of CT in association with specificity
sensitivity and diagnostic accuracy of FDG- as high as 90–100%. The sensitivity of
PET (88% and 78%, respectively) were PET/CT (77%) was also superior to CT
about twofold those of CT (38% and 44%, alone (46%) in 76 patients with metastatic
respectively). However, FDG-PET failed colorectal cancer of the liver (Selzner
to show peritoneal lesions < 10–15 mm in et al., 2004). Importantly, as benign or
diameter and detected only some parts of post-therapy inflammatory processes may
the peritoneal deposits, resulting in unusu- occur in lymphoid tissue and cause false-
ally patchy FDG uptake that was sometimes positive results, histological confirmation
difficult to differentiate from benign uptake should be obtained before depriving a
in normal small bowel tissue (Tanaka et al., patient of potentially curative surgery.
2002). In 40 patients being considered Although it is likely that FDG-PET accu-
for hepatic resection but at high risk for racy regarding distant lymph node metas-
unresectable disease, 10% had peritoneal tases has been overestimated, its additional
disease that was not detected by FDG-PET diagnostic value has been established.
(Fong et al., 1999). Depending on the size
of the peritoneal deposits, sensitivity of
FDG-PET for detection of carcinosis varied Extra-Abdominal Organs
greatly from one study to another (range, Colorectal cancer may disseminate in lung
15–80%) but was higher than that obtained (25–30%), which is the second metastastic
by CT (Flamen et al., 1999) (Figure 30.2). site after the liver, and at later stages in
mediastinal lymph nodes, bone, and brain.
Distant Lymph Nodes Metastases
Except for brain, which shows physio-
Metastatic lymph nodes have contributed logic FDG uptake, whole-body FDG-PET
to some extent to the additional diagnostic has proven to be valuable in identifying
396 S. Truant et al.

Figure 30.2. A 63-year-old man, 6 months after colectomy for advanced adenocarcinoma of the colon
(pT3N2M1) was referred for FDG-PET (b, c, d) for preoperative staging of synchronous liver metastases
after receiving neoadjuvant chemotherapy. In contrast to abdominal CT (a), FDG-PET showed carcinosis
that was confirmed by surgical biopsies and contraindicated hepatectomy
30. Colorectal Liver Metastases 397

extraabdominal metastatic spread. As for in the proliferative glandular breast tissue,


abdominal cavity, there are possibilities resulting in generally diffuse FDG uptake.
of normal FDG uptake in extra-abdominal
organs that must be considered in FDG- Contribution of FDG-PET
PET imaging interpretation. Some studies report a significant additional
diagnostic value of FDG-PET for the diag-
Physiologic and Benign Increased Activity of
nosis of unsuspected extra-abdominal sites,
FDG in the Extra-abdominal Organs
mainly in the lung. For this latter site
To avoid problems with image display, the of recurrence, sensitivity rates of FDG-
brain is excluded from the reconstructed PET (94–100%) were equivalent or slightly
whole-body images because of significant higher than those reported for CT (sensitiv-
accumulation of FDG and the fact that ity 50–100%). FDG-PET and CT showed
cerebral metastases may not be detected. similar specificity rates of 90–100%.
There can be an increased diffuse 18F-FDG Nevertheless, in a few of the prior studies
uptake in the bone marrow in patients of FDG-PET scanning, chest CT was rou-
undergoing hemopoietic stimulation ther- tinely used for preoperative staging. Some
apy or showing acute and severe bleeding. authors have further considered as a posi-
Conversely, focal activity within bone tive impact lung metastases detected by
marrow is always suspicious for an abnor- FDG-PET but not by chest X-rays a posi-
mality, although a healing bone, e.g., after tive impact, even though secondarily con-
rib fractures, or a degenerative or inflam- firmed by subsequent chest CT, resulting in
matory joint disease, can also give rise to additional disease in up to 31% of patients.
elevated FDG uptake. Vigorous exercise Other studies have shown that FDG-PET
in the days just before a scan should be was not superior to chest CT for detection
avoided as it can cause intense muscular of lung metastases (Lai et al., 1996; Truant
uptake in the muscle that usually has the et al., 2005). Detection of lung nodules is
distinctive pattern of a symmetric uptake. size dependent and small lung metastases
Stress-induced muscle tension often seen may be overlooked by FDG-PET that does
in the trapezius and paraspinal muscles can not match the spatial resolution of CT in the
be prevented by pretreatment with muscle chest. On the other hand, FDG-PET may
relaxants. Asymmetrical FDG uptake in help to differentiate benign from malignant
muscles and metabolically active fatty tis- lesions. Occasionally, other sites of extra-
sue (brown fat) is more difficult to interpret.abdominal recurrences have been reported
FDG uptake in residual thymic tissue has in patients being considered for hepatic
to be distinguished from mediastinal aden- resection (Figure 30.3), but additional diag-
opathy. Increased FDG uptake which most nostic value of FDG-PET may be difficult to
likely attributable to inflammation may assess due to the small number of patients.
also be seen in noninvolved lymph nodes
in patients with a medical history of silicosis,
bronchitis or granulomatous diseases such Impact of FDG-PET Findings on Patient’s
as tuberculosis or sarcoidosis. Benign Management
distribution of FDG occurs frequently in FDG-PET may have various types of
normal or goitrous thyroid, in heart and impact, mainly avoidance of laparotomy
398 S. Truant et al.

Figure 30.3. A 46-year-old man underwent FDG-PET as preoperative staging of colorectal liver metas-
tases. FDG-PET showed multiple liver metastases in the right lobe in association with a right supracla-
vicular uptake that was related to a metastatic lymph node at sonography

by upstaging disease from resectable to while FDG-PET did not confer any advan-
unresectable, reducing futile morbidity or tage over conventional diagnostic methods
seldom performance of more extensive sur- in the assessment of hepatic metastases.
gery by disproving a positive CT finding. Consequently, FDG-PET improved
It may lead to commencement of palliative resectability rates. In two series (Strasberg
chemotherapy, resulting in improved out- et al., 2001; Truant et al., 2005), 95% of
come or much better palliation. Integration patients who underwent laparotomy actu-
of FDG-PET in the presurgical evaluation ally underwent hepatic resection, similar
of patients with liver metastases from to that achieved by the use of staging
colorectal cancer has also been shown to laparoscopy to detect unresectable tumors
substantially reduce overall costs. Due to before laparotomy. In comparison, 20% of
the lack of randomized controlled clinical patients who underwent exploration on the
trial to assess FDG-PET impact, only stud- basis of conventional imaging were found
ies that focused on preoperative staging of to have unresectable disease at the time
liver metastases have been considered for of surgery. On the other hand, in these
this part of the analysis. series, FDG-PET also falsely upstaged and
understaged up to 7% and 8% of patients,
respectively (Topal et al., 2001; Truant
Additional Disease and Change of
et al., 2005) and showed very poor yield
Management
in patients who were on chemotherapy
When FDG-PET was added to a complete at the time of FDG-PET (Akhurst et al.,
conventional work-up in preoperative 2005). In the preoperative staging of liver
planning for patients with hepatic metas- metastases judged resectable on the basis
tases, additional sites of disease were of conventional imaging findings, FDG-
identified in 9.5–25% of patients (Truant PET resulted in a change of management
et al., 2005). FDG-PET contribution came in 10–25% (Ruers et al., 2002). Selzner
essentially from the discovery of extra- et al. (2004) studied whether the novel
hepatic sites, mainly in the abdomen, PET/CT imaging modality impacted the
30. Colorectal Liver Metastases 399

treatment of patients with metastatic color- before surgical exploration because these
ectal cancer of the liver. As for FDG-PET patients have a low rate of detection of
alone, the main impact of PET/CT came extrahepatic disease and even a non-
from the discovery of extrahepatic disease. negligible rate of false positive findings
New findings in the PET/CT resulted in a (Schussler-Fiorenza et al., 2004; Truant
change in the therapeutic strategy in 21% et al., 2005). As FDG-PET is costly and
of the patients. Therefore, the impact of not widely available, FDG-PET should be
FDG-PET in the particular setting of pre- restricted to high risk patients in centers
sumed resectable liver metastases seems with low availability of FDG-PET in order
to be less than the mean impact of 29% to delay a surgery.
reported by Huebner et al. (2000) in their
meta-analysis on FDG-PET contribution
in overall recurrent colorectal cancer. Survival Impact
The major part of the additional diag- The addition of FDG-PET imaging to pre-
nostic value of FDG-PET resided in surgical evaluation may improve survival
the subgroup of patients at high risk for by appropriately eliminating ineffective
unresectable disease, as predicted by the surgery in patients with inoperable dis-
Memorial Sloan Kettering Cancer Centre ease. Results are, however, highly contro-
clinical risk score based on five criteria: versial regarding the survival benefit of
(1) node positive primary, (2) disease-free FDG-PET. It has been reported that the
interval from primary to liver metastases use of preoperative FDG-PET lessened
< 12 months, (3) largest hepatic tumour the recurrence rate in patients undergo-
> 5 cm, (4) number of hepatic tumours > 1 ing hepatic resection for colorectal liver
(5) CEA level >200 ng/ml (Fong et al., metastases by detection of disease not
1999). Among patients with a low clinical found on conventional imaging. This had a
risk score, 0–3% of patients had additional significant impact on the survival of these
disease detected by FDG-PET, compared patients. In 100 patients having hepatic
with 14–17% for patients with a high clini- resection between March 1995 and June
cal risk score (Schussler-Fiorenza et al., 2002, the 5-year disease-free and actu-
2004; Truant et al., 2005). Thus, patients arial overall survival in patients screened
with large (> 5 cm), multiple and synchro- with FDG-PET was 34.8% and 58%,
nous metastases were more likely to have respectively, which was higher than previ-
extrahepatic disease detected by FDG-PET. ously reported (Fernandez et al., 2004).
In 40 patients being considered for hepatic However, this series included 63% patients
resection but with a high clinical risk score, with a single tumor while in other series,
FDG-PET altered or influenced the clinical resections of solitary liver metastases rep-
management in 16 patients (40%) (Fong et resented the minority of cases and the
al., 1999). This FDG-PET impact is higher resectioning of ten or more tumors was not
than that reported in the aforementioned uncommon. Another study did not confirm
series without consideration of the clini- this survival benefit: 3-year overall and
cal risk score. Some authors now consider disease-free survival rates after exclud-
that patients at low risk of metastatic ing the patients who did not undergo liver
disease should not undergo FDG-PET resection were comparable with those for
400 S. Truant et al.

all patients (51% vs. 47% and 27% vs. However, rising CEA often occurs in asso-
24%, respectively) (Topal et al., 2001). ciation with negative or equivocal results
Most patients selected for surgery by of conventional imaging studies, leading
FDG-PET indeed still had recurrence of to the diagnosis of tumor recurrences at
disease because of the presence of dis- a non-resectable stage. The advantage
ease that was not detected by FDG-PET. of FDG-PET over anatomic imaging to
In the study by Fong et al. (1999), after screen the whole-body in one examination
a relatively short median follow-up (7 session was assumed to be of particular
months), 40% patients had a recurrence value in this setting. Due to the lack of
after liver resection, whereas other series specific study, FDG-PET contribution in
have reported recurrences as early as 3 the postoperative follow-up of resected
months after resection (Strasberg et al., liver metastases can only be extrapolated
2001). Although a fraction of these find- from studies on patients with suspected
ings are likely due to disease that is below recurrent colorectal cancer on the basis of
the level of detection by FDG-PET, at least rising CEA.
some of these early recurrences are due to In asymptomatic patients with a rising
true–false negative findings on FDG-PET. CEA and nondiagnostic radiologic workup,
On the other hand, patients in whom FDG- the sensitivity of FDG-PET for detection of
PET provided additional information had a recurrence has been reported as 79–100%,
dismal prognosis with a 3-year overall sur- specificity as 50–83% and accuracy as
vival of 0% that could be explained by the 74–93% on a patient-by-patient analysis
extent of their disease (Topal et al., 2001). (Flanagan et al., 1998). FDG-PET was
This may be the true survival impact of more sensitive than both CT and serum
FDG-PET: identifying the small number CEA for detection of disease recurrence,
of patients that have no chance for cure except for the liver, and the specificity of
despite reassuring conventional imaging. FDG-PET was equivalent to that of CEA.
FDG-PET showed high positive predictive
value (89–97%) and negative predictive
Postoperative Follow-Up After Resection
value (71–100%) (Flanagan et al., 1998).
of Colorectal Liver Metastases and
FDG-PET was also more accurate than
Rising Carcinoembryonic Antigen
conventional diagnostic methods in pre-
(CEA)
dicting the resectability or non-resectabil-
After hepatic resection, recurrences occur ity of the recurrence, though it did show
in up to 60% of patients, mostly within 2 a low sensitivity on a lesion-by-lesion
years of surgery and with an equal distri- analysis (57%) in 28 patients who under-
bution between hepatic and extrahepatic went second-look laparotomy (Libutti
sites. It is assumed that recurrences diag- et al., 2001). By contrast, in patients with
nosed by systematic follow-up of asymp- normal CEA levels and symptoms, FDG-
tomatic patients are more often resectable PET showed a high rate of false-positive
with a curative intent. Serial determina- results and was inaccurate in predicting
tions of the plasma CEA concentration is recurrence. In the postoperative follow-up
the most frequently used method for the of liver metastases, specific pitfalls have
detection of such asymptomatic recurrences. to be known such as FDG uptake in the
30. Colorectal Liver Metastases 401

scar of previous partial hepatectomy in apy outcome earlier than 2 months after
the case of important inflammatory reac- onset to therapy because resolution of
tion, e.g., postoperative biloma. Studies therapy-induced anatomic changes lags
are now needed to address the impact of behind tumor cell mortality. As metabolic
early diagnosis by FDG-PET on survival changes induced by chemotherapy precede
and its cost-effectiveness in the postopera- the morphological changes, 18F-FDG-PET
tive follow-up of resected liver metastases was assumed to predict response to therapy
before large-scale introduction in clinical at an earlier stage than conventional diag-
practice can be advocated. nostic methods. Evaluation of response
to therapy has involved careful compari-
son of pretreatment and post-treatment
FDG-PET for Monitoring the Response FDG uptake, as assessed by qualitative
to Systemic or Regional Therapy of visual interpretation, standardized uptake
Colorectal Liver Metastases value (SUV) or extensive kinetic analysis.
Hepatic metastases can be treated with Studies performed in patients with hepatic
a curative or palliative intent with sys- metastases from colorectal cancer demon-
temic therapy (chemotherapy, new tar- strated a relatively rapid decline (within
geted therapies) or regional therapy to 72 h) in the SUV in responding tumors after
the liver, including local ablative therapy a single infusion of 5-fluorouracil and foli-
(radiofrequency ablation, cryoablation), nic acid, whereas nonresponding tumors
chemotherapy administered through the showed an increase, no change, or only a
hepatic artery using infusion pomps, selec- small decline in 18F-FDG uptake. Using
tive chemoembolization or radiolabeled SUV in tumors 4–5 weeks after the start
90
Y-microspheres. of treatment with 5-fluorouracil (Findlay
et al., 1996) or after FOLFOX therapy
Anticancer Systemic Therapy (Dimitrakopoulou-Strauss et al., 2003) fur-
ther predicted the nonresponse with a high
Systemic Chemotherapy
accuracy. Quantitative, dynamic FDG-PET
Most patients with colorectal cancer meta- also identified patients not responsive to
static to the liver will receive chemotherapy, treatment with the FOLFOX regimen,
using mostly 5-fluorouracil in combina- although the aggressiveness of the method
tion chemotherapy regimen including (multiple arterial samplings), its sensitiv-
oxaliplatin (FOLFOX) and/or irinotecan ity to the noise and the long time of the
(FOLFIRI). When liver metastases are dynamic acquisition (1 h on a limited field)
non-resectable, chemotherapy alone may are limiting for its use in carcinologic
supply a significant survival benefit pro- domain (Dimitrakopoulou-Strauss et al.,
viding that patient is a responder. Accurate 2003).
prediction of treatment failure may allow FDG-PET negativity following exten-
the oncologist to apply an alternative treat- sive chemotherapy does not, however,
ment regimen without subjecting the patient necessarily mean complete tumor destruc-
to the toxicity of the full treatment scheme. tion because injured but alive tumor cells
Monitoring with radiologic imaging meth- may not accumulate FDG to the same
ods like CT and MRI cannot assess ther- extent as untreated cells. Conversely,
402 S. Truant et al.

a marked increase in 18F-FDG metabolism monitoring the response during the course
has been described in lesions responding of therapy, further clinical trials are needed
after initiation of chemotherapy, because to assess the beneficial effect of early
of major inflammatory response and tran- FDG-PET scanning on the treatment of
sient increase in stromal reaction, and patients and the ultimate outcome. The
may result in overestimation of the frac- method of quantifying tumor 18F-FDG
tion of viable cells. In a tumor mouse uptake, through visual or semiquantitative
model treated by chemotherapy, the FDG methods such as SUV or kinetic analysis,
uptake was stabilized by day 8–10 despite and remain challenging, as well as the
a further reduction in viable tumor cell exact timing of FDG-PET.
fraction (Spaepen et al., 2003). In 18
patients with colorectal liver metastases,
New Targeted Therapies (Monoclonal
the reduction in FDG uptake 1–2 weeks
Antibodies)
after onset of chemotherapy was not sig-
nificantly different between the respond- Systemic chemotherapy is now used in
ing and non-responding groups and was association with new targeted therapies,
false-positive for response in 20% of the such as the monoclonal antibodies bev-
patients (Findlay et al., 1996). Optimal acizumab and cetuximab that specifi-
timing of FDG-PET is thus crucial and cally block the receptors of the vascular
imaging 1–2 weeks after completion of endothelial growth factor (VEGF) and
chemotherapy is recommended to avoid of the epidermal growth factor (EGF),
transient fluctuations in tumor FDG respectively. The VEGF stimulates the
uptake confounding response assessment. proliferation and migration of vascularly
These data suggest that a late 18F-FDG derived endothelial cells and is overex-
examination, namely at 4–5 weeks after pressed in colon cancer. Several studies
the start of treatment, may be better for have suggested that FDG-PET could allow
therapy outcome. FDG-PET may further follow-up of patients treated by an angio-
be of prognostic value in patients under- genesis inhibitor. Endothelial cells have
going chemotherapy. The first FDG-PET indeed proven to contribute to 18F-FDG
study prior to onset of chemotherapy was uptake, with a positive correlation between
predictive for the therapy outcome, with SUV and microvessel density. Recently,
pretreated tumors displaying a low FDG FDG-PET showed an increased metabolic
uptake more likely to have a worse prog- activity in colorectal liver metastases fol-
nosis after chemotherapy than those with lowing removal of the primary tumor, as
a high SUV (Dimitrakopoulou-Strauss et reflected by a rise in FDG uptake. As this
al., 2003). An early response to treatment phenomenon coincided with a decrease in
with 5-fluorouracil or FOLFOX regimen, serum anti-angiogenic factor levels, the
as shown on FDG-PET, was also well cor- authors came to the conclusion that the
related with the ultimate response seen in primary tumor suppressed angiogenesis in
clinical, radiographic or pathologic find- its distant metastases and that its removal
ings a few weeks or months later. resulted in an increase in metabolic activity
Despite interesting findings of sev- in liver metastases, emphasizing the inter-
eral studies investigating FDG-PET for est of angiogenesis inhibitors in colorectal
30. Colorectal Liver Metastases 403

cancer (Peeters et al., 2005). In the only of recurrence with follow-up FDG-PET,
series of seven patients treated with beva- whereas MRI and CT showed only 43%
cizumab and irinotecan for colorectal liver and 50% of these recurrences, respectively
metastases, FDG-PET findings correlated (Anderson et al., 2003). In lesions with
better than those of CT with pathology a positive FDG-PET after treatment, the
(Goshen et al., 2006). Further studies are positive predictive value for the detection
needed to confirm these results, as well of local recurrence was as high as 80%;
as studies on FDG-PET in the evaluation the negative predictive value was 100%
of treatment of colorectal liver metastases (Langenhoff et al., 2002). The authors
with cetuximab. advised to administer a second ablative
treatment on any lesions that remain posi-
Regional Therapies tive. The dual-modality PET/CT, that was
comparable to FDG-PET alone in terms of
Local Ablative Therapy (Cryosurgery sensitivity (65%) in a series of 11 patients
Ablation and Radiofrequency Ablation)
(Veit et al., 2006), might be of great value
Local ablative techniques are used as for guidance of reinterventions. However,
adjuncts to chemotherapy or hepatic resec- detection of residual tumor remains chal-
tion in a curative or palliative intent when lenging, as increased glucose metabolism
complete resection of tumor in the liver due to tissue regeneration surrounding
cannot be achieved. These techniques can the ablative necrosis or abscess formation
be performed laparoscopically or during in the treated area may superimpose on
open surgery, or for radiofrequency abla- small areas of residual tumor, resulting
tion alone by a percutaneous procedure in false-negative FDG-PET results. Some
guided by CT or sonography. In absence authors recommended follow-up FDG-
of evolving extrahepatic lesions, as con- PET directly after radiofrequency abla-
firmed by pretherapy FDG-PET, these tion or at least within 24 h to avoid these
local ablative therapies have been associ- artefacts (Veit et al., 2006). Further studies
ated with improved median survival rates. are needed to confirm these results and to
However, the frequency of incomplete determine the best timing of FDG-PET.
treatment, and thus local recurrence, is still
relatively high due to limitations in ade-
quately monitoring the destruction process Hepatic Arterial Chemotherapy
at the time of treatment. Moreover, after Patients with unresectable liver metastases
local ablative therapy, local recurrences can be proposed hepatic arterial infusional
in the liver are not easily identified with chemotherapy through an hepatic artery
CT, except at advanced stage of relapse, pump placed during a short laparotomy.
because of difficulties in differentiating This treatment provides at least equivalent
viable tumor tissue from adjacent necrosis or even increased response rate for color-
or therapy-induced hyperperfusion. ectal liver metastases when compared to
The time of detection of recurrence by intravenous chemotherapy. It is, however
FDG-PET has proven to be earlier than contraindicated in the presence of extrahe-
the detection by CT alone. Comparative patic disease due to limited systemic effi-
studies reported a fully accurate prediction ciency. Because patients with unresectable
404 S. Truant et al.

liver metastases have a high risk of occult benefit of resected patients needs to be
extrahepatic disease, FDG-PET may spare confirmed. In the postoperative follow-up
patients an unnecessary laparotomy in this of resected liver metastases, FDG-PET
particular setting. may be useful in locating recurrences in
asymptomatic patients with a rising CEA
Other Regional Therapies and nondiagnostic radiologic workup,
although a specific study is still needed in
Some authors have studied the effects of
this setting, as well as an analysis of the
regional chemoembolization therapy using
impact of early diagnosis by FDG-PET
FDG uptake as a criterion and found that
on survival. Results of preliminary studies
decreased FDG uptake correlated with
have also suggested the relevance of
response, while presence of residual uptake
FDG-PET in monitoring the response to
was used to guide further regional therapy
systemic or regional therapy of colorectal
(Vitola et al., 1996a, b). A few studies have
liver metastases. The improvement of ther-
also suggested the effectiveness of FDG-
apeutic management and the cost savings
PET in monitoring the therapeutic response
associated with the use of FDG-PET justify
of hepatic metastases to 90Y-glass micro-
its generalization in clinical practice.
spheres, that are introduced via the hepatic
artery that supplies the majority of blood
flow to the metastases and exerts a local REFERENCES
radiotherapeutic effect (Wong et al., 2004). Akhurst, T., Kates, T. J., Mazumdar, M., Yeung,
H., Riedel, E. R., Burt, B. M., Blumgart, L.,
Jarnagin, W., Larson, S. M., and Fong, Y. 2005.
CONCLUSION Recent chemotherapy reduces the sensitivity
of [18F]fluorodeoxyglucose positron emission
In the preoperative staging of presumed tomography in the detection of colorectal metas-
resectable colorectal liver metastases, the tases. J. Clin. Oncol. 23: 8713–8716.
Anderson, G. S., Brinkmann, F., Soulen, M. C.,
best results of FDG-PET are obtained in the Alavi, A., and Zhuang, H. 2003. FDG positron
detection of extra-hepatic disease, mainly emission tomography in the surveillance of
in the abdomen, while high-quality CT may hepatic tumors treated with radiofrequency abla-
be as accurate as FDG-PET in depicting tion. Clin. Nucl. Med. 28: 192–197.
liver or lung metastases. FDG-PET may be Bipat, S., van Leeuwen, M. S., Comans, E. F.,
especially valuable in patients with high Pijl, M. E., Bossuyt, P. M., Zwinderman, A. H.,
and Stoker, J. 2005. Colorectal liver metastases:
clinical risk scores or synchronous metas- CT, MR imaging, and PET for diagnosis meta-
tases, which represent a greater proportion analysis. Radiology 237: 123–131.
of patients referred for surgery. Conversely, Dimitrakopoulou-Strauss, A., Strauss, L. G., and
in those instances in which FDG-PET Rudi, J. 2003. PET-FDG as predictor of therapy
findings result in the patient being denied response in patients with colorectal carcinoma.
potentially curative surgery, further inves- Q. J. Nucl. Med. 47: 8–13.
Even-Sapir, E., Parag, Y., Lerman, H., Gutman, M.,
tigations are needed, particularly in low Levine, C., Rabau, M., Figer, A., and Metser, U.
risk patients. The main impact involves 2004. Detection of recurrence in patients with
patients that are contraindicated before rectal cancer: PET/CT after abdominoperineal or
undergoing futile surgery, whereas survival anterior resection. Radiology 232: 815–822.
30. Colorectal Liver Metastases 405

Fernandez, F. G., Drebin, J. A., Linehan, D. C., MR imaging, PET): a meta-analysis. Radiology
Dehdashti, F., Siegel, B. A., and Strasberg, S. 224: 748–756.
M. 2004. Five-year survival after resection of Lai, D. T., Fulham, M., Stephen, M. S., Chu, K. M.,
hepatic metastases from colorectal cancer in Solomon, M., Thompson, J. F., Sheldon, D. M.,
patients screened by positron emission tomogra- and Storey, D. W. 1996. The role of whole-body
phy with F-18 fluorodeoxyglucose (FDG-PET). positron emission tomography with [18F]fluoro-
Ann. Surg. 240: 438–447; discussion 447–450. deoxyglucose in identifying operable colorectal
Findlay, M., Young, H., Cunningham, D., Iveson, cancer metastases to the liver. Arch. Surg. 131:
A., Cronin, B., Hickish, T., Pratt, B., Husband, 703–707.
J., Flower, M., and Ott, R. 1996. Noninvasive Langenhoff, B. S., Oyen, W. J., Jager, G. J., Strijk,
monitoring of tumor metabolism using fluorode- S. P., Wobbes, T., Corstens, F. H., and Ruers,
oxyglucose and positron emission tomography T. J. 2002. Efficacy of fluorine-18-deoxyglu-
in colorectal cancer liver metastases: correlation cose positron emission tomography in detecting
with tumor response to fluorouracil. J. Clin. tumor recurrence after local ablative therapy for
Oncol. 14: 700–708. liver metastases: a prospective study. J. Clin.
Flamen, P., Stroobants, S., Van Cutsem, E., Dupont, Oncol. 20: 4453–4458.
P., Bormans, G., De Vadder, N., Penninckx, Libutti, S. K., Alexander, H. R., Jr., Choyke, P.,
F., Van Hoe, L., and Mortelmans, L. 1999. Bartlett, D. L., Bacharach, S. L., Whatley,
Additional value of whole-body positron emis- M., Jousse, F., Eckelman, W. C., Kranda, K.,
sion tomography with fluorine-18–2-fluoro-2- Neumann, R. D., and Carrasquillo, J. A. 2001.
deoxy-D-glucose in recurrent colorectal cancer. A prospective study of 2-[18F] fluoro-2-de-
J. Clin. Oncol. 17: 894–901. oxy-D-glucose/positron emission tomography
Flanagan, F. L., Dehdashti, F., Ogunbiyi, O. A., scan, 99mTc-labeled arcitumomab (CEA-scan),
Kodner, I. J., and Siegel, B. A. 1998. Utility of and blind second-look laparotomy for detecting
FDG-PET for investigating unexplained plasma colon cancer recurrence in patients with increas-
CEA elevation in patients with colorectal cancer. ing carcinoembryonic antigen levels. Ann. Surg.
Ann. Surg. 227: 319–323. Oncol. 8: 779–786.
Fong, Y., Saldinger, P. F., Akhurst, T., Macapinlac, Moore, H. G., Akhurst, T., Larson, S. M., Minsky,
H., Yeung, H., Finn, R. D., Cohen, A., Kemeny, B. D., Mazumdar, M., and Guillem, J. G. 2003.
N., Blumgart, L. H., and Larson, S. M. 1999. A case-controlled study of 18-fluorodeoxyglu-
Utility of 18F-FDG positron emission tom- cose positron emission tomography in the detec-
ography scanning on selection of patients for tion of pelvic recurrence in previously irradiated
resection of hepatic colorectal metastases. Am. rectal cancer patients. J. Am. Coll. Surg. 197:
J. Surg. 178: 282–287. 22–28.
Goshen, E., Davidson, T., Zwas, S. T., and Aderka, Peeters, C. F., de Geus, L. F., Westphal, J. R., de
D. 2006. PET/CT in the Evaluation of Response Waal, R. M., Ruiter, D. J., Wobbes, T., Oyen,
to Treatment of Liver Metastases from Colorectal W. J., and Ruers, T. J. 2005. Decrease in circu-
Cancer with Bevacizumab and Irinotecan. lating anti-angiogenic factors (angiostatin and
Technol. Cancer. Res. Treat. 5: 37–44. endostatin) after surgical removal of primary
Huebner, R. H., Park, K. C., Shepherd, J. E., colorectal carcinoma coincides with increased
Schwimmer, J., Czernin, J., Phelps, M. E., and metabolic activity of liver metastases. Surgery
Gambhir, S. S. 2000. A meta-analysis of the 137: 246–249.
literature for whole-body FDG PET detection Rohren, E. M., Paulson, E. K., Hagge, R., Wong, T.
of recurrent colorectal cancer. J. Nucl. Med. 41: Z., Killius, J., Clavien, P. A., and Nelson, R. C.
1177–1189. 2002. The role of F-18 FDG positron emission
Kinkel, K., Lu, Y., Both, M., Warren, R. S., and tomography in preoperative assessment of the
Thoeni, R. F. 2002. Detection of hepatic metas- liver in patients being considered for curative
tases from cancers of the gastrointestinal tract resection of hepatic metastases from colorectal
by using noninvasive imaging methods (US, CT, cancer. Clin. Nucl. Med. 27: 550–555.
406 S. Truant et al.

Ruers, T. J., Langenhoff, B. S., Neeleman, N., Jager, curable colorectal liver metastases. Eur. J. Surg.
G. J., Strijk, S., Wobbes, T., Corstens, F. H., and Oncol. 27: 175–179.
Oyen, W. J. 2002. Value of positron emission Truant, S., Huglo, D., Hebbar, M., Ernst, O.,
tomography with [F-18]fluorodeoxyglucose in Steinling, M., and Pruvot, F. R. 2005. Prospective
patients with colorectal liver metastases: a pro- evaluation of the impact of [18F]fluoro-2-deoxy-
spective study. J. Clin. Oncol. 20: 388–395. D-glucose positron emission tomography of
Schussler-Fiorenza, C. M., Mahvi, D. M., resectable colorectal liver metastases. Br. J.
Niederhuber, J., Rikkers, L. F., and Weber, S. M. Surg. 92: 362–369.
2004. Clinical risk score correlates with yield Veit, P., Antoch, G., Stergar, H., Bockisch, A.,
of PET scan in patients with colorectal hepatic Forsting, M., and Kuehl, H. 2006. Detection of
metastases. J. Gastrointest. Surg. 8: 150–157; residual tumor after radiofrequency ablation of
discussion 157–158. liver metastasis with dual-modality PET/CT:
Selzner, M., Hany, T. F., Wildbrett, P., McCormack, initial results. Eur. Radiol. 16: 80–87.
L., Kadry, Z., and Clavien, P. A. 2004. Does the Vitola, J. V., Delbeke, D., Meranze, S. G., Mazer,
novel PET/CT imaging modality impact on the M. J., and Pinson, C. W. 1996a. Positron emis-
treatment of patients with metastatic colorectal sion tomography with F-18-fluorodeoxyglucose
cancer of the liver? Ann. Surg. 240: 1027–1034; to evaluate the results of hepatic chemoemboli-
discussion 1035–1036. zation. Cancer 78: 2216–2222.
Spaepen, K., Stroobants, S., Dupont, P., Bormans, Vitola, J. V., Delbeke, D., Sandler, M. P., Campbell,
G., Balzarini, J., Verhoef, G., Mortelmans, L., M. G., Powers, T. A., Wright, J. K., Chapman,
Vandenberghe, P., and De Wolf-Peeters, C. 2003. W. C., and Pinson, C. W. 1996b. Positron emis-
[(18)F]FDG PET monitoring of tumour response sion tomography to stage suspected metastatic
to chemotherapy: does [(18)F]FDG uptake cor- colorectal carcinoma to the liver. Am. J. Surg.
relate with the viable tumour cell fraction? Eur. 171: 21–26.
J. Nucl. Med. Mol. Imaging 30: 682–688. Whiteford, M. H., Whiteford, H. M., Yee, L. F.,
Strasberg, S. M., Dehdashti, F., Siegel, B. A., Ogunbiyi, O. A., Dehdashti, F., Siegel, B. A.,
Drebin, J. A., and Linehan, D. 2001. Survival of Birnbaum, E. H., Fleshman, J. W., Kodner, I. J.,
patients evaluated by FDG-PET before hepatic and Read, T. E. 2000. Usefulness of FDG-PET
resection for metastatic colorectal carcinoma: scan in the assessment of suspected metastatic
a prospective database study. Ann. Surg. 233: or recurrent adenocarcinoma of the colon and
293–299. rectum. Dis. Colon Rectum 43: 759–767; discus-
Tanaka, T., Kawai, Y., Kanai, M., Taki, Y., sion 767–770.
Nakamoto, Y., and Takabayashi, A. 2002. Wong, C. Y., Salem, R., Qing, F., Wong, K. T.,
Usefulness of FDG-positron emission tomogra- Barker, D., Gates, V., Lewandowski, R., Hill,
phy in diagnosing peritoneal recurrence of color- E. A., Dworkin, H. J., and Nagle, C. 2004.
ectal cancer. Am. J. Surg. 184: 433–436. Metabolic response after intraarterial 90Y-glass
Topal, B., Flamen, P., Aerts, R., D’Hoore, A., microsphere treatment for colorectal liver metas-
Filez, L., Van Cutsem, E., Mortelmans, L., tases: comparison of quantitative and visual
and Penninckx, F. 2001. Clinical value of analyses by 18F-FDG PET. J. Nucl. Med. 45:
whole-body emission tomography in potentially 1892–1897.
A. Diagnosis
31
Biliary Cystic Tumors:
Clinicopathological Features
Yasuni Nakanuma, Hiroko Ikeda, Yasunori Sato, Kenichi Harada,
Koichi Nakamura, and Yoh Zen

INTRODUCTION such as their walls being lined by biliary


or other epithelial cells, are reviewed.
Several types of biliary cystic tumors The tumors can be divided into non-
or tumorous lesions have been reported neoplastic and neoplastic categories. The
(Nakanuma et al., 2000; Portman and former includes hamartomatous lesions
Nakanuma, 2006). Currently, with or developmental anomalies. Recently,
advanced imaging techniques, increasing diagnostic criteria for intraductal papil-
numbers of cystic diseases including rare lary mucinous neoplasm (IPMN) and
types are being found, and their clinico- mucinous cystadenoma and cystadeno-
pathological features are now being char- carcinoma, which are also called muci-
acterized (Okuda, 2001). Hepatobiliary nous cystic neoplasm of the pancreas
cystadenoma and cystadenocarcinoma are (pancreatic MCN), have been established
a prototype of biliary cystic tumors, and with reference to the presence of com-
intraductal papillary neoplasm of the bile munication with the duct system and
duct, particularly with cystic dilatation, is also the presence of ovarian-like stroma
a recently proposed disease entity (Chen in the cystic walls (Longnecker et al.,
et al., 2001). However, the exact defini- 2000; Zamboni et al., 2000). Recent
tion and clinicopathological features of studies showed that two similar types of
these biliary cystic tumors, particularly neoplastic diseases are also present in the
for differential diagnosis, remain to be hepatobiliary system (Chen et al., 2001;
established because the number of cases is Zen et al., 2006b; Abraham et al., 2002,
small. While the cystic lesions themselves 2003). In this review, these biliary cystic
are rather easily detectable by imaging tumors are discussed and compared with
modalities, an exact diagnosis based on pancreatic cystic tumors from the stand-
pathological characteristics is needed at point of their clinicopathological simi-
present. larities. Solitary hepatic cyst, polycystic
In this chapter, the clinicopathologi- liver, and cystic dilatation of the bile duct
cal features of biliary cystic tumors, will not be discussed here.

411
412 Y. Nakanuma et al.

ANATOMICAL complex (VMC) and multicystic biliary


CLASSIFICATION hamartoma, are included in this category.
OF THE BILIARY TREE A majority of them may be ectopic lesions
or malformation of the biliary tree, and
The biliary tree is divided into the intrahe- some are acquired lesions.
patic, hilar (right and left hepatic duct) and
extrahepatic bile duct (EHBD) (Nakanuma Peribiliary Cysts
et al., 1997). The intrahepatic bile duct is Peribiliary Cysts in the Large
a branch of the right or left hepatic bile Portal Tracts and Hepatic Hilum
duct, and the first to third branches of both
hepatic ducts are known as the intrahepatic Peribiliary cysts (the latest discovery in
large bile duct. The extrahepatic bile duct, the field of hepatic cystic diseases or
hilar bile duct, and intrahepatic large bile lesions) are grossly visible and multi-
ducts are referred to collectively as the large ple serous cysts with thin walls are
bile duct in this chapter. Around the large found along the fourth-order bile duct
bile ducts, there are physiologically peri- branch up to the hepatic hilum (Figure
biliary glands with serous and mucinous 31.1B) (Nakanuma et al., 1984; Terada
acini in various combinations in the peri- and Nakanuma, 1990a). Peribiliary cysts
ductal connective tissue shown by Terada are usually tiny, < 10–15 mm in diameter,
et al. (1987), which are different from and do not communicate with the lumen
biliary hamarotoma (see below). These of the adjoining large bile ducts. When
glands are connected with bile duct lumen these cysts are clustered, they appear as a
via their own conduits. Occasionally, pan- cystic neoplasm, particularly resembling
creatic exocrine acini are admixed within a multilocular hepatobiliary cystadenoma.
these glandular lobules. These peribiliary Histologically, these cysts are located in
glands are regularly distributed along the the fibrous connective tissue of the portal
intrahepatic large bile ducts, while their tract and hepatic hilum. The lining of the
distribution is rather irregular or sparse at the cysts is a single layer of columnar or cuboi-
extrahepatic bile ducts. Intrahepatic small dal biliary epithelium, and the cysts do not
bile ducts are classified into septal and have their own fibrous wall (Figure 31.1C).
interlobular bile ducts, and bile ductules The latter is a differential histological point
are located at the periphery of the portal distinguishing from the adjoining, original
tracts. The biliary tree is covered by a layer bile ducts with its own fibrous wall. Some
of biliary epithelial cells or cholangiocytes cysts show a stratification of lining epithe-
and the large bile duct and septal bile ducts lial cells reflecting proliferation. The cysts
have their own dense fibrous wall. are admixed with lobules and conduits
of the peribiliary glands, some of which
show varying degrees of luminal dilatation
NON-NEOPLASTIC BILIARY (microcystic to macrocystic). These find-
CYSTIC LESIONS ings strongly suggest that peribiliary cysts
are derived from the cystic dilatation of
Peribiliary cysts, hepatic foregut cysts, bil- pre-existing peribiliary glands of the large
iary hamartoma such as von Meyenburg’s intrahepatic bile ducts.
31. Biliary Cystic Tumors: Clinicopathological Features 413

a b c

Figure 31.1. Radiological and pathological features of peribiliary cysts. (a) An image of computed
tomography (CT) reveals cystic lesions along the hilar bile ducts. (b) A macroscopic image shows small
cysts in periductal connective tissue. (c) Microscopically, cysts are lined by a single-layered columnar
epithelium (H&E staining, ×40)

As for the underlying disease, peri- which arise from the dilatation of VMC
biliary cysts were originally found in or biliary microhamartoma (Kida et al.,
cases of advanced liver disease such as 1992; Qian et al., 2003). Parenchymal
liver cirrhosis and other portal hyper- cysts are predominant in some cases,
tensive diseases, and in patients with while peribiliary cysts are predominant
other hepatobiliary diseases (Nakanuma, in others. Peribiliary cysts are reportedly
2004) (see below). Peribiliary cysts are identified by computed tomography (CT)
clinically suggested by computed tomog- in 73% of adult polycystic liver disease.
raphy (CT) in ~ 10% of cirrhotic livers That is, in adult polycystic liver disease,
and more in autopsied cirrhotic livers intrahepatic peribiliary glands show fre-
by histopathological studies. In these quent and variable cystic dilatation. Such
diseases, peribiliary cysts may develop peribiliary cysts may comprise a propor-
secondarily, possibly related to disturbed tion of numerous cysts of adult polycystic
circulatory changes. Peribiliary cysts in liver. The genetic events responsible for the
cases of these chronic liver diseases are development of liver cysts in the paren-
not infrequently associated with inflam- chyma may also be a factor in the develop-
matory and edematous changes (Terada ment of peribiliary cysts from peribiliary
and Nakanuma, 1990a). glands. Peribiliary cysts also develop to a
Peribiliary cysts are frequently found lesser degree and frequency in livers with
in adult polycystic liver disease with solitary nonparasitic cysts. In this context,
autosomal-dominant polycystic kidney peribiliary cysts could be a component of
disease or alone, in addition to the cysts the hepatobiliary fibropolycystic disease
in the parenchyma (parenchymal cysts) family. In contrast, peribiliary glands of
414 Y. Nakanuma et al.

the extrahepatic bile ducts show no cystic defects in cholangiography, though their
dilatation in most cases of liver cirrhosis etiopathogenesis, particularly their relation
or adult polycystic liver disease. to the peribiliary cysts in the hepatic hilus
Clinically, peribiliary cysts do not usu- and the portal tracts described above,
ally show any symptom. However, other remains speculative.
hepatobiliary diseases such as ascending
cholangitis or obstructive jaundice may
Hepatic Foregut Cyst
develop directly or indirectly due to com-
pression by multiple enlarged peribiliary Ciliated hepatic foregut cysts (CHFCs)
are rare cystic lesions, usually located
cysts on the adjoining bile ducts. Follow-up
radiological studies have disclosed that in the subcapsular region (Portman and
Nakanuma, 2006; Terada et al., 1990)
peribiliary cysts gradually increase in size
and number during the course of liver cir- immediately beneath the hepatic capsule,
rhosis, and the increase can be used as a mostly in Segment IV. They are solitary
marker of progression of liver cirrhosis. and unilocular, usually < 3 cm in diameter,
Peribiliary cysts can be demonstrated byand filled with viscous mucinous fluid
CT, ultrasonography (US), and magnetic surrounded by fibrous tissue. Histologically,
resonance imaging (MRI) in the vicin- the cyst wall consists of four layers: pseu-
ity of the hepatic hilum (Figure 31.1A). dostratified ciliated columnar epithelia with
Radiologically, these peribiliary cysts aremucous cells, subepithelial connective tis-
seen as tiny cysts anywhere in the large sue, smooth-muscle bundles immunoreac-
portal tracts and/or hepatic hilum. Drip- tive to actin and desmin, and an outermost
infusion cholangiography CT clearly dem- fibrous capsule. The lining epithelial cells
onstrates the exact location of the cysts. contain neutral, carboxylated, and sulfated
Magnetic resonance (MR) cholangiography mucus, and are also positive for keratin and
epithelial membrane antigen, immunohis-
easily clarifies the distribution of tiny cysts
tochemically. Cilia are immunoreactive to
just along the large intrahepatic bile ducts,
actin and tubulin. Ultrastructurally, definite
hepatic bile ducts or both. While the adjoin-
ing bile ducts are occasionally compressed cilia arranged in a 9 + 2 pattern as well as
and locally deformed by the cysts, there ismucous cells are observed in these cells.
no communication between the cysts and These morphologic features are similar to
those of normal bronchi or bronchogenic
the bile ducts. The differential diagnosis is
cysts, suggesting that CHFC arises from
based on the diffuse or local dilation of int-
rahepatic bile ducts, VMC, and cholangitis.the embryonic foregut and differentiates
Ultrasonography shows round or tubular toward bronchial structures in the liver,
anechoic lesions around the large portal analogous to a bronchogenic cyst of the
mediastinum (Terada et al., 1990).
tracts mimicking dilatation of the bile ducts.
Clinically, CHFCs are symptomless and
Peribiliary Cysts Around the Extrahepatic incidentally detected radiologically or at
Bile Ducts
autopsy. CHFCs are a cystic lesion. With
Peribiliary glands around the extrahepatic US, CHFCs appear anechoic or hypoechoic.
bile ducts also occasionally show grossly These cystic lesions include high-density
visible cystic changes and exhibit filling contents, due to thick mucus (Okuda,
31. Biliary Cystic Tumors: Clinicopathological Features 415

2001). Therefore, unenhanced CT demon- tally in 1–6% of reported autopsy series. Von
strates the lesion to be a high-density or Meyenburg complex (VMC) is a develop-
iso-density lesion relative to the surround- mental malformation that is thought to rep-
ing liver parenchyma. Magnetic resonance resent persistence of the embryonic ductal
imaging (MRI) demonstrated the lesion to plate. The histological appearance is quite
be hyperintense on T1-weighted image. characteristic. Individual lesions are usually
These cystic lesions do not show a contrast < 5 mm in diameter, but sometimes up to
enhancement. When a subcapsular nodular 10 mm. Von Meyenburg complexes (VMC)
lesion shows the above-mentioned find- consist of a variable number of irregular bile
ings, a diagnosis of CHFC should be con- ductular structures embedded in a fibrocol-
sidered (Kadoya et al., 1990). This type of lagenous stroma and are located along the
cyst may be mistaken for a hypovascular portal tracts. They are typically adjacent to
solid tumor. a portal area and consist of a fibrous stroma
that contains irregularly shaped duct-like
Variants: Recently, a histologic variant of
structures lined by a low cuboidal epithe-
CHFC without a smooth muscle layer or
lium. The ducts are usually somewhat dilated
with inconspicuous muscle elements has
and often U-shaped or branching. They may
also been reported. It was < 2.0 cm in diam-
contain proteinous or bile-stained secretions.
eter, and was located subcapusularly in the
In some lesions, one of the ducts may dilate
medial segment (S4) of the liver. It had an
sufficiently to form a macroscopic cyst.
epithelial lining of ciliated pseudostratified
Multiple VMCs are considered part of the
cells with occasional goblet cells, and lacked
spectrum of hepatic fibropolycystic disease.
a smooth muscle layer. The lining epithe-
It is generally believed that both solitary bile
lium contained cells positive for immunohis-
duct cyst and cysts of polycystic disease
tochemical staining of surfactant apoprotein
arise from the dilatation of VMC.
A, suggesting an embryonic foregut origin
Clinically, VMCs are symptomless
of the cysts and differentiation toward bron-
and occasionally misdiagnosed as malig-
chiolar structures (Sato et al., 2006).
nant liver disease including metastasis.
Biliary Hamartoma Imaging findings of VMC are nonspecific.
Ultrasonography (US), CT, and MRI show
The following two types are representative multiple periportal small nodular or dot-like
of biliary hamartoma. lesions. Von Meyenburg complex (VMC)
shows multiple, or numerous tiny hypoat-
Von Meyenburg Complexes
tenuating lesons, which are small hypoe-
Von Meyenburg complexes (VMCs), also choic structures occasionally associated
known as biliary microhamartoma or mul- with comet-tail echoes on US, and hyperin-
tiple bile duct hamartoma, are benign liver tesne lesions on T2-weighted MRI (Okuda,
malformations (Okuda, 2001). These com- 2001).
plexes are usually multiple, though their
Multicystic Biliary Hamartoma
number varies on the individual basis. They
are present in ~ 90% of livers with adult This lesion is a solitary and localized
polycystic liver disease, in ~ 20% of livers cystic lesion ~ 2–5 cm in diameter, which
with solitary nonparasitic cysts, and inciden- when located around the hepatic capsule
416 Y. Nakanuma et al.

of the hepatic lobe, characteristically pro- lesions). These lesions might be related to
trudes from the liver (Figure 31.2a). Some developmental abnormalities of the biliary
cases not associated with inflammatory tract or embryonal foregut.
changes have a honeycomb appearance. Ultrasonography (US) and CT reveal rel-
Histologically, these nodular lesions consist atively well-circumscribed nodules in the
of ductal structures with ductal epithelium medial segment of the liver, some of which
resembling biliary epithelium and periduc- are protruding from the liver. This nodule
tal glands resembling peribiliary glands, is enhanced on CT by contrast medium
and fibrous connective tissues containing and sustained until the delayed phase.
blood vessels (Zen et al., 2006c). Biliary Computed tomography (CT) reveales a
epithelial cells and glandular epithelial relatively well-circumscribed nodule just
cells express biliary-type cytokeratins such beneath the hepatic capsule. Angiographies
as cytokeratins 7 and 19. Smooth muscle revealed that this lesion is supplied by the
bundles focally surround ductal structures hepatic artery. This nodule shows a multi-
in some cases (Figure 31.2B). Bile-like locular cystic lesion containing many small
materials are observed within some ducts. cystic spaces on radiologic images.
Two cases are associated with xanthogran-
ulomatous inflammation around bile-like
materials. This cystic lesion shares several BILIARY CYSTIC NEOPLASM
pathologic characteristics of CHFCs, such
as their location (around the falciform Biliary cystic neoplasms are heterogene-
ligament) and periductal smooth muscle ous and can be divided into several cat-
bundles, but does not fulfill the diagnostic egories. Hepatobiliary cystadenoma and
criteria (no ciliated cells and multilocular adenocarcinoma are a prototype of neo-

a b

Figure 31.2. Pathological features of multicystic biliary hamartoma. (a) A nodular lesion is protruding
from the liver. This lesion shows a honeycomb appearance consisting of many ductal structures and sur-
rounding connective tissue. (b) Microscopically, this cystic lesion is covered by a biliary type epithelium.
Periductal glands are observed in fibrous connective tissue (H&E staining, ×100)
31. Biliary Cystic Tumors: Clinicopathological Features 417

plastic biliary cystic disease, and intraductal


ring in the liver and along the extrahepatic
papillary neoplasm of the bile duct (IPNB),
bile ducts or gallbladder. They account
particularly with cystic dilatation of the
for < 5% of reported cystic lesions of the
affected bile ducts. While recent clinico-
liver (Devaney et al., 1994; Wheeler and
pathological studies suggested that these
Edmondson, 1985). They are solitary, usu-
two are different from each other, the simi-
ally multilocular and occasionally uniloc-
larities and differences between these two
ular, and the cyst wall is composed of
categories of biliary cystic neoplasms have
irregularly thickened fibrous tissue (Figure
not been well characterized or discussed
31.3B). In cystadenocarcinoma, tumor
in detail, and therefore, there may be some
infiltration into the adjacent parenchyma
confusion, especially among clinicians and
is found. Tumor sizes are variable and
pathologists who are not familiar with hepa-
may be as large as 28.0 cm in diameter.
tobiliary diseases. In addition, there could
Histologically, hepatobiliary cystadenoma/
be other uncategorized hepatobiliary cystic
cystadenocarcinoma are nearly always
neoplasms sharing some features of these
mucinous, and these tumors are composed
two categories of cystic neoplasms. of two components: an inner epithelial
layer and an outer fibrous layer. The
Hepatobiliary Cystadenoma most common type is mucinous cystad-
and Cystadenocarcinoma enoma with ovarian-like stroma occur-
ring in women (Devaney et al., 1994).
Clinicopathological Features
The cysts are lined by a mucin-secreting
Hepatobiliary cystadenoma and cystad- epithelium which may be flat or may
enocarcinoma are rare neoplasms occur- form papillary or polypoid projections,

a b c

Figure 31.3. Radiological and pathological features of hepatobiliary cystadenoma/cystadenocarcinoma.


(a) A multilocular cystic lesion with focal calcification is observed in the liver. (b) Macroscopically, the
inner surface of this lesion is rough, and associated with hemorrhage and fibrin exudates. (c) The epithe-
lium covering this lesion is made up of columnar cells containing intracytoplasmic mucin. A ovarian-like
stroma is observed beneath the epithelium (H&E staining, ×200)
418 Y. Nakanuma et al.

pseudostratification and crypt-like invagi- et al., 2003). Such communication might


nation (Figure 31.3C). The epithelial cells have developed secondarily after the inva-
are usually columnar, resembling a biliary sion of biliary cystadenocarcinoma.
or gastric foveolar epithelium with muci- Clinically, these tumors preferentially
carmine and alcian blue-positive cyto- affect women, especially middle-aged
plasmic mucin. Intestinal metaplasia with women. The patients complain of an
goblet cells and neuroendocrine cells is enlarging abdominal mass. Occasionally,
not infrequently present. According to the a spontaneous rupture of cystic neoplasms
grade of intraepithelial neoplasia, tumors is reported. The background or preced-
may be classified as adenoma, or border- ing conditions leading to hepatobiliary
line (low-grade malignant), non-invasive, MCNs are not known. Differentiating these
or invasive adenocarcinoma, suggesting tumors from benign hepatic cysts may be
a multi-step progression of hepatobiliary difficult. Because of their rarity, a diag-
cystadenoma. Distinguishing benign cys- nosis is often delayed and may result in
tadenoma from cystadenocarcinoma is inaccurate treatment, resulting in unnec-
often difficult in tumors without distinct essary morbidity and mortality. Surgical
invasion. resectability is high, and the post-operative
As for the outer layer, highly cellular progress is favorable, especially in MCNs
mesenchymal tissue resembling ovarian without invasion. Aspirated fluid of hepa-
stroma is characteristically seen in almost tobiliary MCNs frequently shows high
all cases (Figure 31.3C). For this feature, levels of tumor markers such as CEA and
the name “cystadenoma with mesenchymal CA19-9, but it does not allow a diagnosis
stroma” has been used. The ovarian-like of malignancy.
stroma is found only in females, and the Radiologically, these tumors appear
stroma cells in the ovarian-like stroma are as large multilocular or unilocular cysts
positive for the estrogen receptor and pro- with internal septa and sometimes calci-
gesterone receptor. fication (Okuda, 2001) (Figure 31.3A).
As secondary changes, the epithelium Ultrasonography (US) shows a multilocu-
may become ulcerated with extravasation lar cyst with papillary projections that
of cystic fluid into the stroma or wall, or are enhanced by dynamic CT. Hepatic
the lesions may be traumatized secondary angiography demonstrates tumor stains
to their large size, so that inflammation, corresponding to the projected papillary
xanthogranulomatous reaction, scarring, portions and displacement of vessels.
and sometimes calcification are frequently T1-weighted images show multilocules
present in the tumors. While most reported with low signal intensity, unless hemor-
cases of hepatobiliary cystadenoma/ade- rhage or concentrated mucous fluid is
nocarcinoma, particularly those with a present. Most T2-weighted images dem-
ovarian-like stroma, had no luminal com- onstrate hyperintensity within the cystic
munication between the cystic tumor and areas, but the signal intensity sometimes
the adjacent bile duct, imaging modalities varies, depending on the fluid contents.
revealed occasionally direct luminal com- These above-mentioned findings are often
munication (Hamazaki et al., 1996; Sato diagnostic for hepatobiliary MCNs.
31. Biliary Cystic Tumors: Clinicopathological Features 419

Comparison with Mucinous Cystic Neoplasm cystic disease is also often mistaken for
of the Pancreas hepatobiliary cystadenocarcinoma.
Pancreatic MCN to which mucinous cys-
tadenoma and cystadenocarcinoma of the Intraductal Papillary Neoplasm
pancreas belong in addition to MCN of of the Bile Duct (IPNB)
the retroperitoneum are cystic epithelial Clinicopathological Features
neoplasms occurring almost exclusively in
women. They are composed of a colum- Biliary papillary tumors differing in malig-
nar, mucin-producing epithelium, sup- nant potential and biological behavior from
ported by ovarian-type cellular stroma. ordinary tubular cholangiocarcinomas are
Tumors may be classified as adenoma, known to develop in the intrahepatic and
borderline (low grade malignant), and extrahepatic bile ducts (Adbores-Saavedra
non-invasive or invasive adenocarcinoma. et al., 2000), though their clinicopatho-
Pancreatic MCN shows no communica- logic features remain to be clarified in
tion with the pancreatic ductal system. detail. As described below, these biliary
Interestingly, hepatobiliary cystadenoma/ papillary tumors share clinicopathologi-
cystadenocarcinoma also presents these cal features of pancreatic IPMN. We pro-
clinicopathological features of pancreatic posed referring to these papillary biliary
MCN. In this context, hepatobiliary cysta- tumors collectively as intraductal papil-
denoma/adenocarcinoma are a counterpart lary neoplasm of the bile duct (IPNB) as a
of pancreatic MCN and could be called biliary counterpart of pancreatic IPMN
hepatobiliary mucinous cystic neoplasm (Chen et al., 2001; Zen et al., 2006b).
(hepatobiliary MCN) (Zen et al., 2006a). Both IPNBs and pancreatic IPMN
As for stromal components of these present with dilatation or multilocular
MCNs, they might be derived from the cystic changes of the affected ducts and
ovarian primodium. The ovarian-like the pancreatic ducts, respectively.
stroma in mucinous cystadenomas can
(1) Classification into three subgroups
originate from gonadal epithelium during
based on clinicopathological features
early embryonic development. In human
embryos, preceding their ‘descent’, the Our recent studies showed that IPNB is
gonads are situated directly under the characterized by three key pathologic
diaphragm, dorsal to the liver, the tail of lesions in various components: (1) papillary
the pancreas and the spleen, but separated proliferation of neoplastic biliary epithelial
from these organs by the peritoneal cavity. cells (dysplastic or adenocarcinomatous)
In contrast to the peritoneal epithelium with delicate fibrovascular stalks within the
elsewhere, the cells covering the gonads bile ducts, (2) mucin hypersecretion (mac-
show an activated morphology. The ovar- roscopic mucin is recognizable in some
ian stroma in mucinous cystadenomas of cases), and (3) variable dilatation or mul-
the liver and pancreas suggests a com- tilocular cystic changes of the affected bile
mon origin in epithelial cells that cover ducts (Figure 31.4B). Some IPNB cases
the embryonic gonads in early fetal life. have been referred to as biliary papilloma
Cholangiocarcinoma arising in liver with and papillary cholangiocarcinoma or the
420 Y. Nakanuma et al.

a b c

Figure 31.4. Radiological and pathological features of intraductal papillary mucinous neoplasm of
the bile duct (IPNB). A multilocular cystic lesion with a mural nodule is observed in the liver. (b)
Macroscopically, the multilocular cystic space is occupied by papillary structures. (c) Tumor cells show
prominent papillary proliferation with distinct fibrovascular cores (H&E staining, ×40)

intraductal growth-type of cholangiocarci- bile ducts. All cystic tumors contained


noma. Multiple and recurring biliary papil- mucinous fluid. Except for mural nodules,
lomas are known as biliary papillomatosis the internal surface of cystic tumors was
(BP). Biliary papilloma (papillomatosis) is generally fine granular. Papillary mural
thought to be a pre-malignant or borderline nodules, ranging from 0.5 to 3.2 cm, were
malignant lesion followed by progression observed in all cases.
to papillary cholangiocarcinoma (Lee et al., Clinically, IPNB affects both genders
2004). Other subgroups of IPNB are and most cases arise in adults. IPNB
characterized by tumors with clinically develops in an apparently normal liver
recognizable mucin overproduction (IPNB and biliary tree, and also occasionally in
with mucin overproduction), and “mucin- cases of hepatolithiasis, primary scleros-
producing bile duct tumors” is an alterna- ing cholangitis, Caroli’s disease, choledo-
tive term for this subgroup. The cystic, chal cysts, parasite infection, and liver
aneurismal, or fusiform dilatation of the cirrhosis related to viral hepatitis. The left
affected bile ducts is prominent in another hepatic lobe is more commonly affected.
subgroup of IPNB. These tumors appear All cystic tumors are located within the
multilocular or unilocular. The average size liver, and rarely multilocular cystic tumors
of such a dilated IPNB was 6.1 cm (range: protrude into hepatic hilum. Symptoms
2.5–20 cm) in our series. Multilocular cystic result from biliary obstruction and its
tumors might have been formed secondar- complication. Mucinous discharge from
ily by twisting or strangulation or dilatation Vater’s ampulla, which is frequent in pan-
due to mucin overproduction, disturbed bile creatic IPMN, is occasionally observed in
flow, and biliary obstruction, in the affected IPNB on endoscopic examination during
31. Biliary Cystic Tumors: Clinicopathological Features 421

endoscopic retrograde cholangiography tumors or saccular or aneurysmal bile


(ERC). We have recently experienced one duct dilatation in the liver (Figure 31.4A).
autopsy case of IPNB in which bile duct In the cystic tumors, septation and lobula-
necrosis due to mucin overproduction by tion are identifiable. Intraductal papillary
IPNB showing biliary papillomatosis was neoplasm of the bile duct (IPNB) itself
the first clinical sign. In many cases, sur- may appear as a small and flat or polypoid
gical resections are performed based on a mass or nodule in the dilated bile ducts or
radiological diagnosis of biliary MCNs, cysts (Figure 31.4B). Large tumors may
and a pathological diagnosis of IPNB be depicted by US, CT, endoscopic ret-
is eventually made postoperatively. They rograde cholangiography (ERC) or MRI
show a rather favorable prognosis after (Okuda, 2001). However, tumors that are
total surgical excision, when compared relatively small or sessile or that spread
to ordinary tubular cholangiocarcinoma. along the mucosal surface, are difficult to
Curative resection in cases with multifo- visualize with imaging modalities. Mural
cality of the disease is difficult because nodules are also detectable on radiological
of frequent recurrence, and invasive carci- images. Endoscopic retrograde cholangi-
noma frequently develops in IPNB. ography (ERC) and intraoperative cholan-
giography from cystic tumors (cystgraphy)
(2) Communication to bile duct lumen
frequently reveal luminal communication
A majority of IPNBs show direct luminal between cystic tumors and the adjacent
communication between cystic spaces of the bile ducts. With US, mucin is usually ane-
tumors and adjoining bile ducts on preop- choic, like bile. In the future, advances in
erative or intraoperative cholangiographies, diagnostic techniques, particularly imag-
and also such communication is occasion- ing modalities and widespread recognition
ally demonstrable in surgically resected of this disease entity, may allow easy and
specimens, macroscopically. Intraepithelial correct recognition and diagnosis of IPNB,
neoplasm is also observed within adjacent and may lead to increased numbers of
non-dilated bile ducts, suggesting a direct IPNB patients.
in-situ like spread of neoplastic epithelial
cells from the affected bile ducts to the (4) Histologic grade of the tumor and
adjacent bile ducts or multiple occurrences progression to invasive carcinoma
of such neoplastic processes along the bil-
Commonly in these subgroups of IPNB, the
iary tree. More importantly, the walls of
affected bile ducts are lined by a papillary
these dilated lesions are microscopically
epithelium (micropapillary to grossly vis-
bile duct fibrous walls themselves, andible papillary) with dysplastic epithelium,
ovarian-like stroma is not observed, and
borderline malignancy to well-differenti-
immunoreactivity for estrogen receptor or
ated papillary adenocarcinoma (Zen et al.,
progesterone receptor are not detected in
2006b) (Figure 31.4C). Interestingly, many
the walls at all. of these cases are of borderline malignancy,
or invasive or non-invasive well-differenti-
(3) Radiological features
ated papillary adenocarcinoma, and only a
Computed tomography (CT) and MRI few are categorized as dysplasia. Intraductal
show multilocular or unilocular cystic papillary neoplasm of the bile duct (IPNB)
422 Y. Nakanuma et al.

of either of these subgroups could progress gastrointestinal metaplasia with aberrant


to two types of invasive carcinoma; tubular expression of MUC2 and MUC5AC, while
adenocarcinoma or mucinous carcinoma. the expression of MUC2 is not seen in con-
These two histological types of invasive ventional ICC (Ishikawa et al., 2004a). The
lesions are closely related to the post- nuclear expression of CDX2, a homeobox
operative survival status of patients. That gene involved in the regulation of intestinal
is, the latter shows a favorable prognosis in development and differentiation, is closely
comparison with the former. Interestingly, associated with the expression of MUC2
phenotypic changes of MUC occurring in IPNB, suggesting its role in intestinal
during tumor progression also relate to metaplasia and its possible association
the histological types of invasive lesions. with carcinogenesis in IPNBs. Such fea-
Mucus core protein 2 (MUC2) expression tures are also reported in pancreatic IPMN.
is frequently observed in IPNB, including As mentioned above, mucus hypersecre-
non-invasive tumors and invasive lesions, tion is a key feature of IPNBs. Mucin is
whereas MUC1 is expressed in almost macroscopically identifiable (macroscopic
all cases of tubular adenocarcinoma, and mucin) on the tumor surface and mac-
rarely in non-invasive IPNB and mucinous roscopic oversecretion of mucin into the
carcinoma. ductal lumen occurred in one fourth of
IPNB cases examined in our study, whereas
(5) Phenotypes
abundant mucin-production is observed in
The neoplastic cells of IPNB are categorized almost all cases of pancreatic IPMN (Zen
into three phenotypes (Zen et al., 2006b): et al., 2006d). Intraductal papillary neo-
(1) the pancreaticobiliary type, composed plasm of the bile duct (IPNB) of the intes-
of columnar cells with an eosinophilic tinal phenotype has many goblet cells (one
cytoplasm and round nucleus; (2) the intes- of the mucin producing cells) and IPNB
tinal type, characterized by stratified tall commonly expresses the secretory type of
columnar cells with some goblet cells, mucin core proteins, such as MUC2 and
resembling intestinal adenoma or adenocar- MUC5AC, as seen in pancreatic IPMN.
cinoma; and (3) the gastric type, composed The difference in the frequency of mac-
of columnar cells with an abundant intracy- roscopic mucin hypersecretion between
toplasmic mucin. In addition, the oncocytic IPNB and pancreatic IPMN might be
type, which is characterized by abundant related to the extent of mucin-production.
eosinophilic cytoplasm and round nucleus, Approximately, one half of IPNB cases is
is also reported as a variant of the pancreati- of the pancreatobiliary type and the other
cobiliary type (Sudo et al., 2001; Zen et al., half is of the intestinal type. The gastric
2006b). These three phenotypes including type is rare in IPNB.
the oncocytic type are also well known in
Molecular and Genetic Aspects of IPNB
pancreatic IPMN.
Like pancreatic IPMN, IPNB can con-
(6) Mucin phenotypes and mucin sists of purely intraductal lesions with
hypersecretion step-wise histologic grades or can give
Intraductal papillary neoplasm of the rise to invasive adenocarcinomas. Unlike
bile duct (IPNB) frequently presents as pancreatic IPMNs, little is known about
31. Biliary Cystic Tumors: Clinicopathological Features 423

the molecular and genetic pathogenesis as MMP-2, -7 and MT1-MMP in IPNB are
and progression processes of IPNB. As low or weakly positive in comparison with
for microsatellite instability, high-level conventional cholangiocarcinoma (CC) and
instability was present in 12 % of IPNB a balance of expression of MMP-2/tissue
cases, and low-level instability was present inhibitor of matrix metalloproteinase-2
in 35 % of IPNB cases (Abraham et al., (TIMP-2) is also significantly decreased in
2002). No IPNB cases showed methyla- conventional CC compared to IPNB. The
tion of hMLH1. These results indicate participation of MMP/TIMP in the devel-
that microsatellite instability is a relatively opment and progression of IPNB remains
frequent event in IPNB but is not associated to be clarified.
with hMLH1 promoter hypermethylation. As for K-ras gene, activating muta-
Interestingly, the pattern of allelic shifts tions in codon 12 of the K-ras oncogene
was frequently not identical between the are present in 30 % of IPNB cases and
intraductal and invasive cholangiocarci- identical K-ras mutations are present in
noma components of IPNB, suggesting that both the intraductal and invasive com-
there is significant genetic heterogeneity ponents, suggesting that these mutations
within these neoplasms. arise early in the tumorigenesis of IPNB
E-cadherin is one subtype of trans- (Abraham et al., 2003). This frequency
membrane glycoprotein, whose cytoplas- of K-ras gene mutations is lower than
mic domain is bound to α- and β-catenin that previously reported for pancreatic
(Ashida et al., 1998), and β-catenin acts IPMNs. The expression of cyclin D1,
as a key structural molecule in E-cadherin- p21WAF1/CIP, pRb, p53, and MDM2 is
mediated cell-to-cell adhesion. Membranous gradually increased with the progression
β-catenin and E-cadherin expression is well of IPNB. p53 expression is found in 40 %
preserved in non-neoplastic epithelium. of dysplastic IPNB and the expression
Expression levels of both molecules are rate reaches a plateau in borderline malig-
significantly decreased in parallel in inva- nant or non-invasive IPNB and invasive
sive IPNB as compared with non-invasive IPNB. Cyclin D1 and c-myc, a target of
counterparts, respectively, suggesting that Wnt signaling, are frequently present in
a loss or reduction of the membranous IPNB, and interestingly, nuclear β-catenin
expression of β-catenin and E-cadherin is staining, reflecting activation of Wnt sign-
involved in the progression of IPNB. A aling, is observed in 25 % of non-invasive
reduction of β-catenin in the cell membrane IPNB and also in invasive IPNB without
is presumed to disrupt cell to cell adhesive a significant difference. This suggests
stability, and therefore, such a reduction that nuclear staining of β-catenin, prob-
may be followed by invasion and then ably reflecting stimulation of the Wnt
metastasis of carcinoma cells. Matrix met- signaling pathway, is probably involved
alloproteinase (MMP) and tissue inhibitor at an early stage in the progression of
of metalloproteinases (TIMP)-2 also play IPNB. However, no β-catenin gene muta-
important roles in the degradation of the tions have been demonstrated in IPNB.
basement membrane during tumor inva- Interestingly, p53 expression, K-ras muta-
sion. It is well known that the expression or tion, and the nuclear accumulation of
activities of matrix metalloproteinases such β-catenin are greater in conventional CC
424 Y. Nakanuma et al.

than IPNB, suggesting that the carcino- cytoplasm. Our results of Dpc4 expression
genesis is different in these two types of are not in line with the results of previous
biliary neoplasm. studies. This discrepancy might be depend-
Inactivation of p16INK4A, a cancer ent on the cases examined. Our study con-
suppressor gene, is reportedly involved sisted of Asian cases, some of which were
in the carcinogenesis in several organs. associated with hepatolithiasis.
Our recent studies showed that while
p16INK4A is frequently expressed in IPNB Corresponds to IPMN of Main
non-neoplastic, stone-containing bile Pancreatic Duct Type
ducts, its expression is markedly reduced The disease entity of pancreatic IPMN is
in low grade IPNB to invasive IPNB in well established, and it arises in the main
hepatolithiasis (Ishikawa et al., 2004b). pancreatic duct (main-duct type) and also
Methylation-specific PCR revealed that in its major branches (branch-duct type).
54.6 % of 44 IPNB foci harbored p16INK4a The histologic grade and invasiveness, and
promoter hypermethylation, and such foci phenotypes of papillary epithelium compo-
are significantly correlated with decreased nent, and the degree of mucin secretion and
expression of p16INK4, suggesting that mucin phenotypes are different depending
p16INK4a inactivation mainly due to its on these two types of pancreatic IPMN. For
promoter hypermethylation, is a frequent example, while pancreatic IPMN is divided
and early event of IPNB. DeHaan et al. into benign, borderline, and malignant non-
(2007) recently reported that a homozygous invasive or invasive lesions. Benign lesions
9p21 deletion is frequently detected in are common in the branch-duct IPMN and
cholangiocarcinoma with complete loss of borderline and malignant lesions are com-
p16INK4a. And this may be the case in mon in the main-duct type.
IPNB. This inactivation may be partially Interestingly, the gross and microscopic
responsible for the step-wise increase in the characteristics and corresponding radio-
cell proliferative activities of biliary epithe- logic features of IPNB are similar to those
lial cells during the progression of INPB, of pancreatic IPMN, particularly those
followed by further genetic and epigenetic arising in the main pancreatic duct. The
alterations of other cell cycle regulators affected ducts of IPNB and pancreatic
shown in IPNB. DPC4, the gene encod- IPMN are cystically or cylindrically dilated
ing Smad4/Dpc4, is a tumor-suppressor and some appear to be multilocular. They
gene located on chromosome 18q. Dpc4 are characterized by the prominent intra-
protein is almost universally expressed in ductal papillary proliferation of biliary
pancreatic IPMN and pancreatic cancer lining cells. Phenotypically, IPNB and pan-
derived from pancreatic IPMN. Abraham creatic IPMN show one of three epithelial
et al. (2003) revealed that Dpc4 expression phenotypes; pancreaticobiliary, intestinal
was intact in all cases of IPNB examined and gastric types. In addition, an oncocytic
by immunohistochemistry. However, in our variant belonging to the pancreatobiliary
studies, Dpc4 expression was lost in 21.4 % phenotype is also reported in IPNB and
of invasive IPNB cases, while Dpc4 expres- pancreatic IPMN. Like pancreatic IPMN,
sion was diffusely observed in all cases of IPNB could progress to tubular adenocar-
non-neoplastic biliary epithelium in their cinoma or mucinous carcinoma. These
31. Biliary Cystic Tumors: Clinicopathological Features 425

clinicopathological characteristics and the were biliary cystadenoma, and 18 (26 %)


survival status of IPNB closely resemble were cystadenocarcinoma. Ovarian-like
those of pancreatic IPMN, suggesting that stroma was observed in 85 % (44 of 52
IPNB may be the biliary counterpart of cases) of hepatobiliary cystadenoma and
pancreatic IPMN. The simultaneous devel- 28 % (5 of 18 cases) of cystadenocarci-
opment of IPNB and IPMN is occasionally noma. So far, IPNBs and hepatobiliary
reported, suggesting a common pathogene- MCNs have been confused because they
sis of these neoplasms and field canceriza- are both cystic and have a similar epithelial
tion of the bile ducts and pancreatic ducts. component. As for the lining epithelial cells
There are, however, several pathological and their histologic grade, IPNB and hepa-
differences between IPNB and pancreatic tobiliary MCN range pathobiologically
IPMN. As for the histological grade of from adenoma or dysplasia, borderline
IPNB and pancreatic IPMN, almost all malignancy and non-invasive malignancy
cases of IPNB belong to non-invasive to invasive carcinoma. Intestinal or gas-
or invasive well-differentiated papillary tric metaplasias are found in both cystic
adenocarcinoma or borderline malignancy, lesions, while oncocytic changes are found
while dysplasia or adenoma is infrequent. in IPNB.
Pancreatic IPMN of the main-duct type IPNB, particularly with a multilocu-
also shows such features, while pancreatic lar appearance or cystic dilatation of the
IPMN of the branch-duct type is usu- affected bile ducts (a cystic variant of
ally an adenoma. Phenotypically, a great IPNB), may resemble hepatobiliary MCNs.
majority of IPNBs are of pancreatobiliary Prevalence of tumor locations (intrahepatic)
or intestinal type, and the gastric type is and the type of cyst (multilocular cyst) are
rare. This proportion is similar to the main similar in both tumor groups. It is hard
duct type of IPMN. In contrast, the major- to differentiate IPNB from MCN radio-
ity of the branch-duct type is gastric type. logically. As described above, it is becom-
While IPNB and pancreatic IPMN of the ing evident that IPNBs and hepatobiliary
main duct type, particularly those of intes- MCNs are distinct entities, and they can
tinal phenotype, are characterized by the be separated based on clinicopathological
common expression of MUC2, CDX2 and features. Most biliary cystic tumors with an
cytokeratin 20 (CK20), pancreatic IPMN ovarian-like stroma are hepatobiliary cysta-
of the branch type shows lower frequencies denoma and some are cystadenocarcinoma.
of CK20 expression and a higher percent- It is likely that many of the hepatobiliary
age of gastric-type phenotype and mucin, MCNs reported in men or those without
thus differing from IPNB. Taken together, mesenchymal ovarian like stroma in the
it is likely that IPNB is a counterpart of early literature could actually belong to
pancreatic IPMN of the main pancreatic IPNB, as demonstrated in pancreatic MCNs
duct-type (Zen et al., 2006b). and IPMN. In addition, the presence of
luminal communication with the bile duct
Differentiation from Hepatobiliary MCNs
favors the diagnosis of IPNB.
According to Devaney’s report (1994), However, luminal communication
which is the largest study of “hepatobiliary is occasionally reported in hepatobil-
MCN”, 52 out of 70 tumors (74 %) iary MCNs with ovarian-like stroma. It
426 Y. Nakanuma et al.

remains to be clarified whether all hepato- nosis of and distinguishing between these
biliary “cystadenoma and cystadenocarci- cystic lesions are clinically important in
noma” with luminal communication to the the choice of treatment, and genetic and
bile ducts and those without ovarian-like molecular approaches are needed to clarify
stroma are IPNB, and also whether uncate- the etiology and pathogenesis of these
gorized biliary neoplasm(s) may constitute cystic diseases.
these unusual cystic tumors.
Other Types of Neoplastic Cystic REFERENCES
Diseases Abraham, S.C., Lee, J.H., Hruban, R.H., Argani, P.,
Furth, E.E., and Wu, T.T. 2003. Molecular and
There may be other types or categories of immunohistochemical analysis of intraductal
biliary cystic tumors which are different papillary neoplasms of the biliary tract. Hum.
from hepatobiliary MCN or IPNB with Pathol. 34: 902–910.
cystic dilatation or multilocular changes Abraham, S.C., Lee, J.H., Boitnott, J.K., Argani,
of the affected bile ducts. For example, P., Furth, E.E., and Wu, T.T. 2002. Microsatellite
instability in intraductal papillary neoplasms of
cholangiocarcinoma is known to develop the biliary tract. Mod. Pathol. 15: 1309–1317.
in biliary diseases showing cystic dilata- Adbores-Saavedra, J., Scoazec, J.C., Wittekind,
tion such as congenital choledochal cyst C., Sripa, B., Menck, H.R., Soehendra, N., and
and Caroli’s disease, and also in hepatic Sriram, P.V.J. 2000. Carcinoma of the gallbladder
cystic diseases such as adult polycystic and extrahepatic bile ducts. In: Hamilton, S.R.,
liver disease or solitary liver cysts, and Aaltonen, L.A., eds. World Health Organization
Classification of Tumours. Pathology and
rarely in the peribiliary glands or cysts Genetics of Tumours of the Digestive System.
(Nakanuma et al., 2000; Terada and Lyon: IARC Press, 207–214.
Nakanuma, 1990b). Cholangiocarcinoma Ashida, K., Terada, T., Kitamura,Y., and Kaibara, N.
arising in these diseases may present cystic 1998. Expression of E-cadherin, alpha-catenin,
changes within or adjacent to the tumor beta-catenin, and CD44 (standard and variant iso-
tissue as suggested by Fujii et al. (2005). forms) in human cholangiocarcinoma: an immu-
nohistochemical study. Hepatology 27:974–82.
In conclusion, biliary cystic tumors are Chen, T.C., Nakanuma, Y., Zen, Y., Chen, M.F.,
largely divided into non-neoplastic, largely Jan, Y.Y., Yeh, T.S., Chiu, C.T., Kuo, T.T.,
hamartomatous, and neoplastic etiologies. Kamiya, J., Oda, K., Hamaguchi, M., Ohno, Y.,
The former includes peribiliary cysts and Hsieh, L.L., and Nimura, Y. 2001. Intraductal
multicystic biliary hamartoma. The lat- papillary neoplasia of the liver associated with
ter includes hepatobiliary cystadenoma/ hepatolithiasis. Hepatology 34: 651–658.
DeHaan, R.D., Kipp, B.R., Smyrk, T.C., Abraham,
adenocarcinoma and IPNB, particularly S.C., Roberts, L.R., and Halling, K.C. 2007. An
those with cystic dilatation or multilocular assessment of chromosomal alterations detected
cystic changes of the affected bile ducts by fluorescence in situ hybridization and p16
(cystic variant of IPNB). Intraductal pap- expression in sporadic and primary scleros-
illary neoplasm of the bile duct (IPNB) ing cholangitis-associated cholangiocarcinomas.
may be a biliary equivalent of pancreatic Hum. Pathol. 38: 491–499.
Devaney, K., Goodman, Z.D., and Ishak, K.G.
IPMN, and hepatobiliary cystadenoma/cys- 1994. Hepatobiliary cystadenoma and cystade-
tadenocarcinoma share clinicopathological nocarcinoma. A light microscopic and immuno-
features of pancreatic MCT, and therefore histochemical study of 70 patients. Am. J. Surg.
can be called biliary MCT. An exact diag- Pathol. 18: 1078–1091.
31. Biliary Cystic Tumors: Clinicopathological Features 427

Fujii, T., Harada, K., Katayanagi, K., Kurumaya, H., normal and pathologic biliary tract in humans,
and Nakanuma, Y. 2005. Intrahepatic cholangi- including blood supply. Microsc. Res. Tech. 38:
ocarcinoma with multicystic, mucinous appearance 552–570.
and oncocytic change. Pathol. Int. 55: 206–209. Nakanuma, Y., Leong, A.S.Y., Sripa, B.,
Hamazaki, K., Gochi, A., Shimamura, H., Kaihara, Ponchon, T., Vatanasapt, V., and Ishak, K.G.
A., Maruo, Y., Doi, Y., Orita, K., Lygidakis, N.J. 2000. Intrahepatic cholangiocarcinoma. In:
1996. Serum levels of circulating intercellular Hamilton, S.R., Aaltonen, L.A., eds. World
adhesion molecule 1 in hepatocellular carci- Health Organization Classification of Tumours.
noma. Hepatogastroenterology 43: 229–234. Pathology and Genetics of Tumours of the
Ishikawa, A., Sasaki, M., Ohira, S., Ohta, T., Oda, Digestive System. Lyon: IARC Press, 173–180.
K., Nimura, Y., Chen, M.F., Jan, Y.Y., Yeh, T.S., Nakanuma, Y. 2004. Peribiliary cysts have at least
and Nakanuma, Y. 2004a. Aberrant expression two different pathogeneses. J. Gastroenterol. 39:
of CDX2 is closely related to the intestinal 407–408.
metaplasia and MUC2 expression in intraductal Okuda, K. 2001. Okuda, K., Mitchell, D.G., Itai,
papillary neoplasm of the liver in hepatolithiasis. Y., Ariyama, J., eds. Hepatobiliary Diseases.
Lab. Invest. 84: 629–638. Oxford: Blackwell Science.
Ishikawa, A., Sasaki, M., Sato, Y., Ohira, S., Chen, Portman, B.C., and Nakanuma, Y. 2006. Diseases
M.F., Huang, S.F., Oda, K., Nimura, Y., and of bile ducts. Burt, A.D., Portman, B.C. Ferrell,
Nakanuma, Y. 2004b. Frequent p16ink4a inac- L.D., eds. MacSween’s Pathology of the Liver.
tivation is an early and frequent event of intra- Churchill Livingstone 5th ed., pp. 517–581.
ductal papillary neoplasm of the liver arising in Qian, Q., Li, A., King, B.F., Kamath, P.S., Lager,
hepatolithiasis. Hum. Pathol. 35: 1505–1514. D.J., Huston, J., 3rd, Shub, C., Davila, S.,
Kadoya, M., Matsui, O., Nakanuma, Y., Yoshikawa, Somlo, S., and Torres, V.E. 2003. Clinical profile
J., Arai, K., Takashima, T., Amano, M., and of autosomal dominant polycystic liver disease.
Kimura, M. 1990. Ciliated hepatic foregut cyst: Hepatology 37: 164–71.
radiologic features. Radiology 175: 475–477. Sato, Y., Kitagawa, S., Zen, Y., Minato, H., and
Kida, T., Nakanuma, Y., and Terada, T. 1992. Nakanuma, Y. 2006. Ciliated hepatic cyst with-
Cystic dilatation of peribiliary glands in livers out smooth muscle layer: a variant of ciliated
with adult polycystic disease and livers with hepatic foregut cyst? Pathol. Int. 56: 340–344.
solitary nonparasitic cysts: an autopsy study. Sato, M., Watanabe, Y., Tokui, K., Kohtani, T.,
Hepatology 16: 334–340. Nakata, Y., Chen, Y., and Kawachi, K. 2003.
Lee, S.S., Kim, M.H., Lee, S.K., Jang, S.J., Song, Hepatobiliary cystadenocarcinoma connected to
M.H., Kim, K.P., Kim, H.J., Seo, D.W., Song, the hepatic duct: a case report and review of the lit-
D.E., Yu, E., Lee, S.G., and Miin, Y.I. 2004. erature. Hepatogastroenterology 50: 1621–1624.
Clinicopathologic review of 58 patients with bil- Sudo, Y., Harada, K., Tsuneyama, K., Katayanagi,
iary papillomatosis. Cancer 100: 783–793. K., Zen, Y., and Nakanuma, Y. 2001. Oncocytic
Longnecker, D.S., Hruban, R.H., Adler, G., and biliary cystadenocarcinoma is a form of intra-
Kloppel, G. 2000. Intraductal papillary-mucinous ductal oncocytic papillary neoplasm of the liver.
neoplasms of the pancreas. In: Hamilton, S.R., Mod. Pathol. 14: 1304–1309.
Aaltonen, L.A., eds. World Health Organization Terada, T., and Nakanuma, Y. 1990a. Pathological
Classification of Tumours. Pathology and observations of intrahepatic peribiliary glands
Genetics of Tumours of the Digestive System. in 1,000 consecutive autopsy livers. III. Survey
Lyon: IARC Press, 237–240. of necroinflammation and cystic dilatation.
Nakanuma, Y., Kurumaya, H., and Ohta, G. Hepatology 12: 1229–1233.
1984. Multiple cysts in the hepatic hilum and Terada, T., Nakanuma, Y., and Ohta, G. 1987.
their pathogenesis. A suggestion of periductal Glandular elements around the intrahepatic bile
gland origin. Virchows. Arch. A. Pathol. Anat. ducts in man; their morphology and distribution
Histopathol. 404: 341–350. in normal livers. Liver 7: 1–8.
Nakanuma, Y., Hoso, M., Sanzen, T., and Sasaki, Terada, T., Nakanuma, Y., Kono, N., Ueda, K.,
M. 1997. Microstructure and development of the Kadoya, M., and Matsui, O. 1990. Ciliated
428 Y. Nakanuma et al.

hepatic foregut cyst. A mucus histochemical, cystic tumors with bile duct communication: a
immunohistochemical, and ultrastructural study cystic variant of intraductal papillary neoplasm
in three cases in comparison with normal bronchi of the bile duct. Mod. Pathol. 19: 1243–1254.
and intrahepatic bile ducts. Am. J. Surg. Pathol. Zen, Y., Fujii, T., Itatsu, K., Nakamura, K., Minato,
14: 356–363. H., Kasashima, S., Kurumaya, H., Katayanagi,
Terada, T., and Nakanuma, Y. 1990b. Pathological K., Kawashima, A., Masuda, S., Niwa, H.,
observations of intrahepatic peribiliary glands in Mitsui, T., Asada, Y., Miura, S., Ohta, T., and
1,000 consecutive autopsy livers. II. A possible Nakanuma, Y. 2006b. Biliary papillary tumors
source of cholangiocarcinoma. Hepatology 12: share pathological features with intraductal
92–97. papillary mucinous neoplasm of the pancreas.
Wheeler, D.A., and Edmondson, H.A. 1985. Hepatology 44: 1333–1343.
Cystadenoma with mesenchymal stroma (CMS) Zen, Y., Terahata, S., Miyayama, S., Mitsui, T.,
in the liver and bile ducts. A clinicopathologic Takehara, A., Miura, S., Nobata, K., Kitao, A.,
study of 17 cases, 4 with malignant change. Kakuda, K., Kiyohara, K., and Nakanuma, Y.
Cancer 56: 1434–1445. 2006c. Multicystic biliary hamartoma: a hitherto
Zamboni, G., Longnecker, D.S., Kloppel, G., Adler, undescribed lesion. Hum. Pathol. 37: 339–344.
G., and Hruban, R.H. 2000. Mucinous cystic neo- Zen, Y., Sasaki, M., Fujii, T., Chen, T.C., Chen,
plasm of the pancreas. In: Hamilton SR, Aaltonen M.F., Yeh, T.S., Jan, Y.Y., Huang, S.F., Nimura,
LA, eds. World Health Organization Classification Y., and Nakanuma, Y. 2006d. Different expres-
of Tumours. Pathology and Genetics of Tumours sion patterns of mucin core proteins and
of the Digestive System. Lyon: IARC Press, pp. cytokeratins during intrahepatic cholangiocar-
234–236. cinogenesis from biliary intraepithelial neo-
Zen, Y., Fujii, T., Itatsu, K., Nakamura, K., Konishi, plasia and intraductal papillary neoplasm of
F., Masuda, S., Mitsui, T., Asada, Y., Miura, S., the bile duct – an immunohistochemical study
Miyayama, S., Uehara, T., Katsuyama, T., Ohta, of 110 cases of hepatolithiasis. J. Hepatol. 44:
T., Minato, H., and Nakanuma, Y. 2006a. Biliary 350–358.
32
Cholangiocarcinoma: Intraductal
Sonography
Kazuo Inui, Hironao Miyoshi, and Junji Yoshino

INTRODUCTION
METHODS
The recent development of magnetic reso-
Instruments
nance cholangiopancreatography (MRCP)
and multiple-detector-row computed tom- Several types of ultrasonic probes, a 2.5 mm
ography (MD-CT) facilitate diagnosis of diameter miniature ultrasonic probe (UM-
the obstruction site in patients with cholan- 2R or UM-3R, Olympus Medical Systems,
giocarcinoma. However, the extension of Tokyo, Japan) and a 2.1 mm diameter ultra-
cholangiocarcinoma is still difficult to diag- sonic probe (UM-4R, Olympus Medical
nose precisely. The correct diagnosis of Systems) have been developed. The UM-2R
cancer extension, especially invasion to ultrasonic probe incorporates a radial scan-
the portal vein and pancreas, is necessary ning system with a frequency of 12 MHz, and
to decide the operative procedures for a the UM-3R and UM-4R with 20 MHz. These
complete resection. Intraductal ultrasonog- are connected to an endoscopic ultrasonic
raphy (IDUS) has been reported by several observation unit (EU-M30 or EU-M2000,
authors, (Kuroiwa et al., 1998; Fujita et al., Olympus Medical Systems) and incorpo-
1998; and Tamada et al., 2001a), as a rated by a probe-driving unit (MAJ 335 or
reliable method for the evaluation of extra- MAJ 2000, Olympus Medical Systems). The
hepatic cholangiocarcinoma and aiding in probes are easily passed through the 2.8 mm
its accurate staging. We, Kanemaki et al. diameter biopsy channel of a duodenoscope
(1997) and Inui et al. (1998), reported on the (JF-200, Olympus Medical Systems) or the
usefulness of three-dimensional intraductal 3.6 mm diameter biopsy channel of a new
ultrasonography (3D-IDUS) as a compre- electroduodenoscope (JF-V260, Olympus
hensive image display, and the instruments Medical Systems).
and processor systems of 3D-IDUS have We use the 3D-IDUS system developed
been developed. In this chapter, we introduce by Olympus Medical Systems (Figure
the methodology of IDUS and 3D-IDUS for 32.1); a 3D ultrasonic probe, UM-3D-2R
the diagnosis of cholangiocarcinoma. (12 MHz) or UM-3D-3R (20 MHz), with

429
430 K. Inui et al.

Ultrasonography is performed while the


ultrasonic probe in the outer sheath is with-
drawn automatically. During scanning, the
outer sheath does not move. Linear recon-
struction (longitudinal) images are pro-
duced from integrating 40 serial radial
images. We can obtain from 40 to 118
serial radial images. The length of the
longitudinal images can be set at 10, 20,
30, or 40 mm, and in pitches of 0.25, 0.5,
0.75, or 1.0 mm. We routinely obtain 118
serial images at the length of 40 mm and
in pitches of 0.5 mm. For 3D-IDUS, we
use an ultrasound image-processing unit
(EU-IP2, Olympus Medical Systems) to
produce the reconstruction images such as
the dual plane reconstruction images.

Insertion Methods
Transpapillary Approach
The patient is placed on the fluoroscopy
table in the prone position. Premedication
consists of 0.5 mg atropine sulfate, 15 mg
pentazocine, 2 mg scopolamine butylbro-
Figure 32.1. Photograph of a 3D-IDUS system, mide intramuscularly, and 10 mg diazepam
which includes a 3D ultrasonic probe and a probe intravenously. After insertion of the elec-
driving unit, the MAJ 2000. The ultrasonic probe troduodenoscope into the second portion
(arrow), a UM-DG20–35R, is a new type of ultra- of the duodenum, endoscopic retrograde
sonic probe with a ropeway system cholangiopancreatography (ERCP) is
performed. After ERCP, we insert the
a diameter of 3.4 mm; and a probe driving ultrasonic probe into the stricture of the
unit, the MAJ 355 beginning in 1995 and bile duct through the biopsy channel of
the MAJ 2000 since 2002. Recently, we the electroduodenoscope (Figure 32.2).
usually use a new type of ultrasonic probe Ultrasonography is performed, and the
with a ropeway system, UM-DG20-35R area observed by the probe is confirmed
(Figure 32.1). These probes are connected by fluoroscopy.
to an endoscopic ultrasonic observation
Percutaneous Transhepatic Approach
unit (EU-M30 or EU-M2000). The 3D
ultrasonic probe consists of an external In patients with obstructive jaundice, per-
tube as an outer sheath and the probe itself cutaneous transhepatic biliary drainage
with a diameter of 2.4 mm and a 12 or (PTBD) is performed immediately after
20 MHz radial scan transducer at its tip. admission to our hospital. We dilate the
32. Cholangiocarcinoma: Intraductal Sonography 431

16 F-PTBD drainage catheter is inserted


into the sinus tract along with a guide-
wire. The ultrasonic probe is inserted into
the stricture of the bile duct through the
PTBD drainage catheter (Figure 32.2).
Intraductal ultrasonography or 3D-IDUS
is performed, and the area observed by the
probe is confirmed by fluoroscopy. For
better images, 2 mg scopolamine butyl-
bromide is given intramuscularly before
scanning to stop the peristalsis of the
duodenum.

Indications
We have treated patients with several
types of pancreatobiliary disease since
1989. The diseases include extrahepatic
cholangiocarcinoma, gallbladder carci-
noma, carcinoma of the duodenal Papilla
Vater, pancreatic carcinoma, intraductal
papillary mucinous neoplasia of the pan-
creas, benign biliary strictures, chronic
pancreatitis, autoimmune pancreatitis
and other rare conditions. Four patients
Figure 32.2. An ultrasonic probe is inserted into with cholangiocarcinoma had 3D-IDUS
the bile duct through the biopsy channel of a duo- after treatment by Nd-YAG laser irradia-
denoscope along with a guide-wire tion. In only four patients, the ultrasonic
probes could not be inserted into the bile
duct and the stricture. In about one third
of patients, we used the percutaneous
sinus tract of the PTBD to 16 F and per- approach.
form percutaneous transhepatic cholangi- Intraductal ultrasonography and
oscopy (PTCS), IDUS, and/or 3D-IDUS 1 3D-IDUS may be indicated for the diag-
week after PTBD. nosis of remnant stones after endoscopic
The patient is placed on the fluoroscopy lithotripsy of common bile duct stones.
table in the supine position. Premedication They also may be indicated for the
consists of 0.5 mg atropine sulfate, 15 mg diagnosis of biliary strictures and the
pentazocine intramuscularly, and 10 mg precise extension of cancer in patients
diazepam intravenously. We observe the with cholangiocarcinoma and intraduc-
biliary stricture with PTCS and take biop- tal papillary mucinous neoplasia of the
sies from the stricture. After PTCS, a pancreas.
432 K. Inui et al.

IMAGES AND CLINICAL without intraductal spread were diagnosed


UTILITY correctly and three of five patients with intra-
ductal spreading were diagnosed correctly
Intraductal Sonography with IDUS. The overall accuracy of IDUS
for intraductal tumor spreading was 84.6 %.
Findings of Cholangiocarcinoma
The normal bile duct wall is observed as Three-Dimensional Intraductal
two to three layers. On the other hand, Sonography
IDUS reveals a carcinoma lesion as an
image of low echoic, irregular thickness. One of the advantages of 3D-IDUS is that
When the tumor echogram extends into the time required for the examinations is
the high echoic layer of the bile duct, we reduced compared with that required for
diagnose the tumor as invading the subse- conventional IDUS, because conventional
rosa. When the tumor echogram extends to IDUS needs to clarify the relationship
the parenchyma of the pancreas, we diag- between the lesions and the surrounding
nose the tumor as invading the pancreas. organs and vessels. Three types of images
The important demarcation of tumor inva- can be produced using the functions of the
sion into the pancreas is the disappearance 3D-IDUS system: dual plane reconstruc-
of the high echoic layer between the bile tion (DPR) images; oblique reconstruction
duct and the pancreas. When the tumor images; and surface rendering reconstruc-
echogram extends to the high echoic wall tion images.
of the portal vein, we diagnose the tumor
Dual Plane Reconstruction Images
as invading the portal vein. Longitudinal
tumor spread is known to be one of Dual plane reconstruction images, includ-
the characteristics of tumor extension of ing radial and longitudinal images, are
cholangiocarcinoma. Its diagnosis is an useful in assessing the tumor extension
important issue for a curable resection. and the tumor’s relationship with the sur-
If an irregular thickness of the bile duct rounding organs (Figure 32.3). The lon-
is observed consecutively and away from gitudinal images can be rotated without
the main lesion, we diagnose longitudinal changing the radial images, and the radial
spread of the tumor. images can also be changed without alter-
ing the linear images. Because the longi-
Precise Diagnosis of Cancer Extension tudinal reconstruction images are similar
The results of IDUS and pathological stud- to an X-ray cholangiogram, it is easy to
ies for tumor extension in 13 patients with recognize the relationship between the
bile duct carcinoma were investigated (Inui lesions and the surrounding organs. When
et al., 2002). The overall accuracy for the rotated in an anti-clockwise direction,
depth of tumor invasion was 84.6 %, for tumor invasion into the pancreas can be
tumor invasion of the pancreas was 88.9 %, clearly observed.
and invasion of the portal vein was 92.3 %.
Oblique Reconstruction Images
The results of IDUS and the pathological
diagnosis for longitudinal tumor spread The oblique reconstruction images (Figure
were studied in 13 patients. Eight patients 32.3b) and surface rendering reconstruction
32. Cholangiocarcinoma: Intraductal Sonography 433

accuracy rate of 3D-IDUS was 90 %, and


the accuracy rate of conventional IDUS was
80 %. On the other hand, the accuracy rates
for CT scans and angiography were 62.5 %
and 50%, respectively. Three-dimensional
IDUS is useful for the precise diagnosis of
cancer extension in cholangiocarcinoma,
especially for assessing invasion into the
portal vein and pancreas (Figure 32.4).
Volume Measurement of Tumors
Tumor volume could be calculated within
a few minutes using the trace areas of the
serial radial images. With EU-IP2, a new
type of image-processing, it is easier to
calculate tumor volume than with EU-IP.
In 11 of the 15 patients (73.3 %), the whole
outline of the tumor could be visualized,
and we could measure the tumor volumes.
We treated high-risk patients with bile
duct carcinoma using Nd-YAG laser irra-
Figure 32.3. An ultrasonic probe is inserted into diation under PTCS. Cholangiography
the stenosis of the bile duct through the PTBD
drainage catheter
revealed complete recanalization of the
biliary stenosis. We performed 3D-IDUS
before and after laser therapy. The tumor
volume ranged from 913 to 16,960 mm3
images can be produced automatically as ste- (mean ± SD, 5,922 ± 7,593) before treat-
reographic views in real time. It is useful to ment, while it had decreased to 484–
understand the relationship between tumors 10,060 mm3 (mean ± SD, 3,148 ± 4,621)
and the surrounding organs or vessels as after treatment. The tumor echos were
a comprehensive image display. However, decreased significantly after laser therapy
the 3D-images are not yet satisfactory when compared with those before therapy. The
compared with those of a CT scan or MRI. results of cholangiography also differed
The technology will likely be improved for after laser therapy. The tumor reduction
clinical use in the near future. rates were 20–73 % (mean ± SD, 45.3
± 21.7). The survival time of the four
Diagnositc Accuracy for Cancer Extension
patients was from 105 to 920 days (mean
To determine the diagnostic ability of ± SD, 463 ± 352) after laser therapy. The
3D-IDUS with regard to tumor infiltration calculation of tumor volume may help
into the pancreas, the 3D-IDUS findings facilitate the evaluation of the efficacy
in ten surgical cases were compared with of laser therapy for patients with bile
the findings of other imaging modalities duct cancer (Inui and Miyoshi, 2004)
as well as the histological findings. The (Figure 32.5).
434 K. Inui et al.

Figure 32.4. 3D-IDUS of cholangiocarcinoma. The DPR images, including the radial and linear images,
easily reveal the relationship between the tumor (T) and the portal vein (PV)

including us reported that it is a reliable


method for the evaluation of extrahepatic
cholangiocarcinoma. On the other hand,
the first report of 3D-IDUS was published
by Kallimanis et al. (1995). We also
reported the usefulness of 3D-IDUS as
a comprehensive image display in 1997
(Kanemaki et al., 1997).
The instruments for IDUS, including a
miniature ultrasonic probe, an observation
unit, the processing system, etc., have been
under development. At first, the minimum
diameter of a miniature probe was 3.6 mm,
while an ultrasonic probe 2.1 mm in diam-
eter is now available. Stavropoulos et al.
Figure 32.5. Oblique reconstruction images reveal
the tumor echo (arrow) which has not invaded the (2005) reported that the applicability of
portal vein (PV) IDUS is limited in tight stenosis compared
with endoscopic ultrasonography, although
IDUS using a high-frequency miniature
DISCUSSION ultrasound probe is accurate for the stag-
ing of bile duct cancers. For improved
Intraductal ultrasonography was reported resolution in cases where there is a limita-
by Silverstein et al. (1989) in an experi- tion in insertion, an ultrasonic probe with
mental study in canines. Many authors a ropeway system has been developed, and
32. Cholangiocarcinoma: Intraductal Sonography 435

an increased success rate using a trans- of IDUS in assessing tumor invasion to the
papillary approach was reported by Fujita pancreas was reported to be 88.9–100 %
et al. (2000). Using the ropeway system (Inui et al., 2002). When the high-echoic
with a guide-wire, scanning of the bile layer between the tumor and a vessel dis-
duct was achieved in 98.2–99.2 % of the appears, it is diagnosed as positive vessel
patients, regardless of the site and length invasion. The accuracy of IDUS in assess-
of the biliary strictures. Once the guide- ing tumor invasion to the right hepatic
wire is passed through the biliary stenosis, artery and portal vein was reported to be
no further technique is required to intro- 92.3–100 % (Inui et al., 2002). IDUS is
duce the ultrasonic probe. useful for the staging of cholangiocarci-
The clinical indications for IDUS of noma, especially invasion to the portal
the biliary tract include the diagnosis of vein and pancreas. Menzel and Dosmschke
choledocholithiasis (Palazzo, 1997), the (1999) reported on the usefulness of IDUS
differential diagnosis of biliary strictures for diagnosis of cholangiocarcinoma, and
(Menzel et al., 2000) and the local stag- showed that tumor extent was diagnosed
ing of cholangiocarcinoma. Wada et al. correctly in 76.8 % of cases when using
(2000) reported that the diagnosis of bil- IDUS and in 53.6 % cases in EUS.
iary metallic stent obstruction and evalu- Tamada et al. (2001b) reported that an
ation for microwave coagulation therapy accurate distinction of T1 and T2 tumor
were new clinical applications of IDUS. was difficult. In these cases, even if the
Menzel et al. (2000) proposed that tumor was limited to the inside low-echoic
malignancy should be suspected for hyp- layer, it could be a T2 tumor (the tumor
oechoic masses with irregular margins and invaded the peri-muscular connective tis-
inhomogeneous echo-poor areas invading sue) or a T1 tumor (the tumor was con-
the surrounding tissue. Penetration was fined to the fibromuscular layer).
defined as a continuation of the main The diagnosis of the longitudinal spread
hypo-echoic tumor mass into adjacent of a tumor is the most important issue
structures. On the other hand, homogene- for a curable resection, but this is diffi-
ous echo patterns and smoothly defined cult for IDUS. When the irregular thick-
margins were most likely not malignant. ness of the bile duct can be observed
They reported that the sensitivity, specifi- consecutively and away from the main
city, and accuracy of IDUS were 91.1 %, lesion, the longitudinal spread of tumor
80 %, and 89.1 %, respectively. Tamada can be diagnosed. The overall accuracy
et al. (1998) reported that if the inter- of IDUS for intraductal tumor spread was
ruption of the bile duct wall structure or 84.6%, and the result was not satisfactory.
a lesion with a diameter of greater than However, Tamada et al. (2001b) reported
8.0 mm were diagnosed as malignant, the that transpapillary IDUS prior to biliary
accuracy of IDUS was 76%. drainage is a tactic that can minimize the
When a tumor protrudes into the pan- influence of the biliary drainage catheter.
creatic parenchyma, or if the outer echoic Nevertheless, their results were the same
layer of the intra-pancreatic bile duct is as our prior study (Inui et al., 2002) that
interrupted by a tumor, invasion to the used a percutaneous approach, with an
pancreas can be diagnosed. The accuracy accuracy of 85 %.
436 K. Inui et al.

Three dimensional intraductal Newly developed ultrasonic probe with ropeway


Ultrasonography is more useful for the system for transpapillary Intraductal ultrasonog-
raphy of the bilio-pancreatic ductal system. Dig.
precise diagnosis of cancer extension in
Endosc. 12:250–254.
cholangiocarcinoma than conventional Inui, K., Nakazawa, S., Yoshino, J., Wakabayashi,
IDUS. Visualization of the left hepatic T., Okushima, K., Nakamura, Y., Hattori, T.,
artery and proper hepatic artery was poor Miyoshi, H. 1998. Ultrasound probes for biliary
(14 %/18 %) due to echo attenuation in lesions. Endoscopy 30(S1):A 120–123.
conventional IDUS as reported by Tamada Inui, K., Yoshino, J., Okushima, K., Miyoshi,
H., Nakamura, Y. 2002. Intraductal EUS.
et al. (2001a). On the other hand, Kanemaki
Gastrointest. Endosc. 4:S58–62.
et al. (1997) reported that 3D-IDUS was Inui, K. Miyoshi, H. 2004. Cholangiocarcinoma
useful to estimate tumor invasion to the and intraductgal sonography. Gastrointest.
portal vein with an accuracy of 100 %. Endosc. N. Am. 15:143–155.
However, Tamada et al. (2001b) described Kallimanis, G., Garra, B.S., Tio, T.L., Krasner, B.,
same limitations of 3D-IDUS in the assess- al-Kawas, F.H., Fleischer, D.E., Zeman, R.K.,
Nguyen, C.C., Benjamin, S.B. 1995. The fea-
ment of the longitudinal spread of cancer.
sibility of three-dimensional endoscopic ultra-
Another limitation of both IDUS and sonography: a preliminary report. Gastrointest.
3D-IDUS in evaluating cholangiocarci- Endosc. 41:235–239.
noma is its difficulty in diagnosing lymph Kanemaki, N., Nakazawa, S., Inui, K., Yoshino,
node metastasis. J., Yamao, K., Okushima, K. 1997. Three-
dimensional intraductal ultrasonography: pre-
liminary results of a new technique for the
diagnosis of diseases of the pancreatobiliary
CONCLUSIONS system. Endoscopy 29:726–731.
Kuroiwa, M., Goto, H., Hirooka, Y., Furukawa,
IDUS is a reliable method for the evalu- T., Hayakawa, T., Naitoh, Y. 1998. Intraductal
ation and staging of cholangiocarcinoma. ultrasonography for the diagnosis of proximal
DPR images produced by 3D-IDUS are invasion in extrahepatic bile duct cancer. J.
Gastroenterol. Hematol. 13:715–719.
useful for assessing the tumor extension
Menzel, J. and Dosmschke, W. 1999. Intraductal
and its relationship with the surrounding ultrasonography (IDUS) of the pancreato-biliary
organs. 3D-IDUS is useful for the precise duct system. Personal experience and review of
diagnosis of cancer extension in cholan- literature. Euro. J. Ultrasound 10:105–109.
giocarcinoma, especially for invasion into Menzel, J., Poremba, C., Dietl, K.H., Domschke,
the portal vein and pancreas. Volume cal- W. 2000. Preoperative diagnosis of bile duct
strictures- Comparison of intraductal ultra-
culation should facilitate the evaluation of
sonography with conventional endosonography.
the efficacy of non-surgical treatments. Scand. J. Gastroenterol. 35:77–82.
Palazzo, L. 1997. Which test for common bile duct
REFERENCES stones? Endoscopic and intraductal ultrasonog-
raphy. Endoscopy 29:655–665.
Fujita, N., Noda, Y., Kobayashi, G., Kimura, Silverstein, F.E., Martin, R.W., Kimmey, M.B.,
K., Yago, A. 1998. Staging of bile duct carci- Jiranek, G.C., Franklin, D.W., Proctor, A. 1989.
noma by EUS and IDUS. Endoscopy. 30(S1): Experimental evaluation of an endoscopic ultra-
A132–134. sound probe: in vitro and in vivo canine studies.
Fujita, N., Noda, Y., Yokohata, K., Tanaka, M., Gastroenterology 96:1058–1062.
Maguchi, H., Komatsu, Y., Omata, M., Inui, K., Stavropoulos, S., Larghi, A., Verna, E., Battezzati,
Nakazawa, S., Mukai, H., Yasuda, K. 2000. P., Stevens, P. 2005. Intraductal ultrasound for
32. Cholangiocarcinoma: Intraductal Sonography 437

the evaluation of patients with biliary strictures Tamada, K., Nagai, H., Yasuda, Y., Tomiyama, T.,
and no abdominal mass on computed tomogra- Ohashi. A., Wada, S., Kanai, N., Satoh, Y., Ido,
phy. Endoscopy 37:715–721. K., Sugano, K. 2001b. Transpapillary intraductal
Tamada, K., Ueno, N., Tomiyama, T., Oohashi, ultrasonography prior to biliary drainage in the
A., Wada, S., Nishizono, T., Tano, S., assessment of longitudinal spread of extrahe-
Aizawa, T., Ido, K., and Kimura, K. 1998. patic bile duct carcinoma. Gastrointest. Endosc.
Characterization of biliary structures using 53:300–307.
intraductal Ultrasonography: comparison Wada, S., Tamada, K., Tomiyama, T., Ohashi, A.,
with percutaneous cholangioscopic biopsy. Utsunomiya, K., Higashisawa, T., Satoh, Y.,
Gastrointest. Endosc. 47:341–349. Miyata, T., Sugano, K. 2000. Endoscopic micro-
Tamada, K., Inui, K., Menzel, J. 2001a. Intraductal wave coagulation therapy for bile duct cancer with
ultrasonography of the bile duct system. intraductal ultrasonographic monitoring: brief case
Endoscopy 33:878–885. report. Am. J. Gastroenterol. 95:1104–1105.
B. Prognosis
33
Extrahepatic Bile Duct Carcinoma:
Role of the p53 Protein Family
Alexander I. Zaika and Seung-Mo Hong

INTRODUCTION the bile ducts. Based on the location of


the primary tumor, this disease is classi-
Bile duct carcinomas (cholangiocarci- fied into intrahepatic bile duct carcinoma
noma) are devastating tumors. Surgical (peripheral cholangiocarcinoma), perihilar
resection is the mainstay of current treat- cholangiocarcinoma (Klatskin tumor), and
ment; however, it is only effective for a distal bile duct carcinoma. Extrahepatic
limited number of patients diagnosed at bile duct (EBD) carcinoma (including
early stages of the disease. Unfortunately, perihilar and distal cholangiocarcinomas)
the vast majority of patients seek treat- arises from the hepatic hilum to distal
ment of the advanced disease making bile duct, and constitutes approximately
them poor candidates for curative surgery. 80–90 % of all cholangiocarcinomas
Conventional chemotherapy and radiation (Malhi and Gores, 2006). It usually occurs
therapy have not been shown to be effec- in patients over 60 years of age and is
tive in prolonging long-term survival of more frequently observed in males than
patients with cholangiocarcinoma. In addi- females (Hong et al., 2005). The EBD
tion, there are no sensitive and specific carcinoma is a relatively uncommon neo-
tests to diagnose and exclude this disease plasm in the United States and European
in high-risk populations. Thus, there is countries, while the prevalence in Asia,
an urgent need for novel biomarkers and including Korea, Japan, and Vietnam, is
therapies based on a better understanding higher (Hong et al., 2005; Malhi and
of biliary tumorigenesis that would signifi- Gores, 2006; Nagakawa et al., 2002). In
cantly impact the disease outcome. This general, the worldwide incidence of EBD
chapter reviews current clinical data on the carcinoma is slightly decreasing, while
p53 protein family and its role in biliary that of intrahepatic cholangio-carcinoma
tumorigenesis. is increasing.

Epidemiology Etiology
Cholangiocarcinoma is a malignant neo- Several different risk factors may give rise
plasm arising from the epithelium lining to the injury and proliferation of the bile

441
442 A.I. Zaika and S.-M. Hong

duct epithelial cells. These factors are asso- and undifferentiated carcinomas (Hong
ciated with the development of EBD carci- et al., 2005). By tumor growth feature,
noma, which included primary sclerosing EBD carcinoma can be classified as poly-
cholangitis associated with inflammatory poid, nodular, and infiltrating types. One
bowel disease, hepatolithiasis, infection of the often seen histologic characteristics
with liver flukes (Clonorchis sinensis and of EBD is marked desmoplastic stro-
Opisthorchis viverrini), familial adeno- mal reaction, which is extensive stromal
matous polyposis coli, von Meyenberg fibrosis surrounding tumor cells caused
complexes, anomalous pancreaticobiliary by cancer cell infiltration. A few malig-
junction, and choledochal cysts (Choi nant tumors from other organs can show
et al., 2004). desmoplastic stromal reaction. Other than
desmoplastic stromal reaction, micro-
Diagnosis scopic findings of EBD carcinoma are
similar to adenocarcinomas from other
The patients’ symptoms, which include
organs. Usually tumor cells form tubular
abdominal pain, jaundice, and weight
glands. Based on the degree of glandular
loss, are non specific. Therefore, the
formation (histologic grade), EBD carci-
evaluation of patients in regard to the
noma can be divided into well, moderate,
extent of EBD carcinoma is determined
or poorly differentiated adenocarcinomas.
by several radiologic examinations, such
For example, when more than 75 % of all
as ultrasonography, helical-CT, direct
tumor cells make glandular structures, it
cholangiography, and MRCP (magnetic
can be classified as a well-differentiated
resonance cholangiopancreatography)
carcinoma. On the other hand, when less
(Seyama and Makuuchi, 2007). In some
than 25 % of cancer cells form tubular
situations, it is difficult to make a definite
structures, EBD carcinoma can be called
diagnosis with these imaging tools. Bile
poorly differentiated carcinoma. Most
cytology or brushing biopsies are the
EBD carcinomas are classified as well to
tools of choice in this case (Seyama and
moderately differentiated adenocarcinomas
Makuuchi, 2007).
(Albores-Saavedra et al., 2000). Nuclei of
tumor cells show significant difference
Pathology
in size (pleomorphism), are stained more
EBD carcinoma usually appears as an ill- darkly by hematoxylin and eosin staining
demarcated, firm, whitish tan mass with (hyperchromasia), and demonstrate atypi-
induration or ulceration of the bile duct cal, asymmetric mitotic features. In about
wall. Adenocarcinoma is the most com- 10 % of EBD carcinoma cases, dysplasia is
mon histologic type, and consists of about observed in the peritumoral noncancerous
80 % of EBD carcinomas. Other histo- epithelial cells (Albores-Saavedra et al.,
logic variants that compose the remain- 2000). At the time of diagnosis, many EBD
ing 20 % include: papillary carcinomas, carcinoma cases show tumor cell invasion
intestinal-type adenocarcinomas, adenos- into lymphatic system, blood vessels, nerve
quamous carcinomas, mucinous carcino- fibers, lymph nodes, and other adjacent
mas, clear cell carcinomas, signet ring organs such as pancreas, duodenum, and
cell carcinomas, small cell carcinomas, liver.
33. Extrahepatic Bile Duct Carcinoma: Role of the p53 Protein Family 443

Prognosis cellular senescence, autophagy, differen-


The prognosis of EBD carcinoma is very tiation, and repair from genotoxic damage.
poor. For patients who undergo surgi- Inactivation of p53 is strongly associated
cal resection, the 5-year survival rate is with an increased susceptibility to tumors
approximately 20 % (Nathan et al., 2007). in laboratory animals and humans. In
Several factors are related to the prognosis fact, chronic bile duct injury provokes
of patients with EBD carcinomas. The cholangiocarcinoma in p53-deficient mice
most important factors are the extent of (Farazi et al., 2006).
tumor cell invasion (T classification of On average, one-half of cholangiocar-
TNM staging system) and metastasis to cinoma patients carry mutations in the
lymph nodes (N classification). In addi- p53 gene. Depending on the study, its
tion, several other factors, including his- range is from 19–86 % which may reflect
tologic type, histologic grade, presence of differences in etiology between studied
tumor cells at resection margin, vascular, populations, site of the disease along the
lymphatic, and perineural invasion, are biliary tree and detection techniques. A
also reported to show significant survival comprehensive analysis of p53 by Khan
difference in patients with EBD carcino- et al. (2005) concluded that the presence
mas (Green et al., 2002). and type of p53 alterations are dependent
on environmental factors and might be
caused by changes in the composition and
ROLE OF P53 IN MALIGNANT metabolism of cytotoxic biliary constitu-
TUMORS OF THE BILE DUCT ents. Tumor location may also play a role
as the rate of p53 mutations is higher in
Extensive molecular studies of cholan- EBD carcinomas compared with proximal
giocarcinoma significantly improved our tumors (Argani et al., 2001; Diamantis
understanding of the tumorigenic proc- et al., 1995). p53 inactivation occurs rela-
esses in the bile duct. Aberrant activation tively late in neoplastic progression in the
of several key oncogenes, such as recep- biliary tract. Murata et al. (2002) and Tan
tor tyrosine kinases Erb-b2, IL6R/gp130, et al. (2004) have suggested that immu-
c-Met, EGFR, signaling molecules Ki-ras, nohistochemical staining for p53 is use-
BRAF; anti-apoptotic proteins Mcl-1, Bcl- ful for distinguishing cholangiocarcinoma
2, Bcl-xL, and potent inducers of cellular from benign lesions and determination of
proliferation, COX-2 and β-catenin, have tumor spread using preoperative biopsies.
been found in these tumors. Inactivation of The role of p53 mutations in predicting
tumor suppressor genes TP53, APC, p16/ patients’ survival has been examined in
INK4a, DPC4 by genetic and epigenetic several studies of cholangiocarcinoma.
mechanisms have also been reported. p53 Some studies have reported favorable
deserves special attention, as this protein prognosis for patients lacking p53 muta-
plays a pivotal role in tumor suppression. tions (Ahrendt et al., 2000; Diamantis
p53 is a transcription factor that at the et al., 1995). However, other investigators
cellular level, is involved in the regula- have not found such association (Argani
tion of multiple intracellular processes et al., 2001; Lee et al., 1996; Suto et al.,
including apoptosis, cell cycle progression, 1997).
444 A.I. Zaika and S.-M. Hong

Analysis of p53 Mutations discovered p63 and p73 proteins. These


For the study of p53 in tumors, the scien- proteins share significant structural and
tific community employs two broadly used functional similarities with p53. The high-
techniques: immunohistochemical staining est similarity is found in the DNA-binding
(IHC) and various methods of mutational domain, in which p53, p63 and p73 share
analysis (sequencing, single strand confor- approximately 60 % of amino acid identity.
mation polymorphism (SSCP) and others). p73 and p63 proteins are expressed as
Immunochistochemical detection of p53 a complex variety of protein isoforms
is based on the assumption that mutant that originate from two p73 and p63 gene
p53 is stabilized in cancer cells and can be promoters (P1 and P2) and extensive gene
detected by staining with a p53-specific splicing at the NH2- and COOH-termini.
antibody, typically DO-7 or CM1. IHC is These mechanisms lead to the generation
a useful and cost-effective approach for of multiple isoforms with and without
detection of p53 mutations. However, the transactivation domain. The latter isoforms
IHC results should be interpreted with are termed ΔN (or ΔTA) isoforms (transac-
caution as positive IHC does not neces- tivation-deficient) while the former are TA
sarily imply that p53 is fully inactive, nor isoforms (Figure 33.1). Recently, it has
does the absence of staining mean that been confirmed that the principal organi-
p53 is functionally active (Munro et al., zation of the p53 gene is similar to that of
2005). More accurate information about the TP73 and TP63 genes, and multiple
p53 mutations can be achieved by direct p53 isoforms are expressed in human
sequencing alone or combined with SSCP. tumors.
Typically, mutations are determined in A number of activities attributed to p53
exons 5–8 of the p53 gene, since the are shared by TAp73 or TAp63 proteins.
majority of cancer-related mutations are These include induction of apoptosis, cell
located in this region. Exons 5–8 encode cycle arrest and cellular senescence. p73
a highly conserved DNA binding domain and p63 can bind to the p53 consensus
that is critical for the biological functions binding site and activate transcription of
of p53. However, 10–25 % of all muta- most of the p53-target genes that are
tions occur outside exons 5–8 and include involved in cell cycle regulation and apop-
a prevalence of frameshift, nonsense and tosis. It has been demonstrated that the
splice site mutations. Therefore, when it is blockade of TAp73 or TAp63 with a domi-
feasible, complete sequencing of the p53 nant-negative mutant or a small-inhibitory
gene is the preferable strategy. RNA (siRNA) suppresses apoptosis in
cancer cells, irrespective of p53’s status
(Vilgelm et al., 2008). The combined loss
of p63 and p73 results in the failure of
ROLES P73 AND P63 mouse embryonic fibroblasts containing
IN MALIGNANT TUMORS p53 to undergo apoptosis in response to
OF THE BILE DUCT DNA damage.
A recent study of p73- and p63-deficient
p53 is the founding member of the protein mice found that p73+/− (and p63+/−) hetero-
family, which also includes the recently zygous animals developed both malignant
33. Extrahepatic Bile Duct Carcinoma: Role of the p53 Protein Family 445

Figure 33.1. Architectures of human p53, p63 and p73 genes and proteins. TP53, TP63 and TP73 genes
encode a transactivation domain (TAD), DNA-binding domain (DBD), oligomerization domain (OD),
and have two promoters (P1 and P2). The P1 promoter drives expression of transactivation-competent
full-length proteins (TA isoforms). Conversely, the P2 promoter produces TAD-deficient proteins (ΔN
isoforms) with dominant-negative functions. p73 and p63 proteins have also additional domain, termed
SAM (sterile alpha motif) that is not found in p53. Different isoforms are designated with Greek letters

and benign lesions. Similar to the p53+/− However, analyses of ΔN isoforms


mice, tumors from these animals undergo show that these proteins can function as
a loss of the remaining wild-type allele. transcriptional inhibitors of p53, TAp63,
Moreover, loss of p73 and p63 can coop- and TAp73 and bona fide oncoproteins.
erate with the loss of p53 function. These Overexpression of ΔNp63 leads to a sig-
data are consistent with the idea that p73 nificant increase in colony growth in soft
and p63 function as tumor suppressors agar and xenograft tumor formation in
(Flores et al., 2005). nude mice. ΔNp63 has been identified as a
446 A.I. Zaika and S.-M. Hong

downstream target of the phosphoinositide levels in the normal biliary epithelium. In


3-kinase (PI3K) pathway, a cell survival contrast, approximately 20 % of biliary EBD
and proliferation pathway in cancer. displasias are positive for p63 and p73. The
Similarly, ΔNp73 facilitates cell immor- immunoreactivity for these proteins is pri-
talization and cooperates with oncogenic marily confined to the cell nuclei (Figure
Ras in transformation of mouse embryonic 33.2). The staining is further increased
fibroblasts. Implantation of these cells in adenocarcinoma with 26 % and 41 %
into immunocompromised mice results in p63- and p73-positive tumors, respectively.
tumor development. Surrounding normal tissue and fibrovascular
Data from multiple tumors show that stroma within the tumor are uniformly nega-
loss-of-function mutations in p73 and p63 tive (Hong et al., 2007).
open reading frames are rare. Moreover, p63 is a good epithelial basal cell marker
p73 and p63 are frequently overexpressed that is frequently expressed in squamous cell
in human tumors. These data are consistent carcinomas. In rare cases of adenosquamous
with an increased titer of p73 antibodies in carcinoma of the bile duct, p63 staining
patients with various types of cancers. In was only noticed in the areas with squa-
a number of human tumors high-global mous carcinoma and not in adenocarcinoma.
p73 and p63 protein expression correlates Expression of p63 was also found in the areas
with reduced patient survival suggesting of squamous metaplasia. p63 staining was
that these proteins play an important role significantly higher in EBD tumors located
in tumorigenesis and may serve as useful in the distal bile duct and cases with vascular
tumor markers. invasion (Hong et al., 2007). Increased p63
Currently, a limited number of studies expression was also found in intrahepatic
have investigated p63 and p73 expression cholangiocarcinoma accompanied by liver
in cholangiocarcinoma. Immunohistoche- cirrhosis (Nomoto et al., 2006).
mical analyses revealed that both p73 and A study conducted by Tannapfel et al.
p63 are present at low or undetectable (1999) found that p73 expression associated

a b

Figure 33.2. Immunohistochemical staining for p63 and p73 proteins in extrahepatic bile duct carcinoma.
(a). Moderately differentiated EBD adenocarcinoma shows nuclear staining of p63 (X200) (b). Strong
staining for p73 in papillary carcinoma of the bile duct (X200)
33. Extrahepatic Bile Duct Carcinoma: Role of the p53 Protein Family 447

with patients’ survival in intrahepaticp63 transcripts. However, two important


cholangiocarcinoma. The median survivalaspects need to be taken into considera-
time for patients with p73-positive tumors
tion before designing the experiment. p73
was 132 days, whereas patients without and p63 are expressed as a complex set of
detectable p73 protein had a survival time
isoforms. Past studies that have searched
of 320 days. Because the antibody used in
for transcripts have thus far identified
this study can potentially recognize multi-
nine TAp73, three TAp63 transcripts, at
ple p73 isoforms, it remains unclear which
least six p73 mRNAs, and three p63 iso-
p73 isoform, TA or ΔN, is associated with
forms that lack the entire N-terminus (ΔN
the patients’ prognosis. isoforms) or its part (Figure 33.1). As a
In a large study that encompassed 175
result, RT-PCR primers should be carefully
cases of EBD cholangiocarcinoma, neither
designed to avoid detection of unrelated
p73 nor p63 was correlated with 1-, 3- and
isoforms. Detailed information about p73
5-year survival rates (Hong et al., 2007).
and p63 isoforms are provided in Vilgelm
However, analysis of tumors co-express-et al. (2008), primer sequences and PCR
ing p63 and p73 has found that combinedconditions in Concin et al. (2004) and
p63 and p73 expression is a significantHong et al. (2007). It is also important to
prognostic factor (Hong et al., 2007). note that p73 and p63 proteins are control-
This interesting phenomenon may find a led at post-translation levels and for that
mechanistic explanation in studies of non-
reason mRNA might not directly reflect
biliary tumors. Ellisen’s group at Harvard
protein expression (Vilgelm et al., 2008).
Medical School found that co-expression Proteins: Routine immunohistochemical
of p63 isoform, ΔNp63, with TAp73 leds techniques are applicable to the detection
to the inhibition of anti-proliferative and
of p73 and p63 proteins in formalin-fixed,
pro-apoptotic potential of TAp73 (Leongparaffin-embedded tumor tissues. Detailed
et al., 2007). It is plausible to suggest that
immunostaining protocol for p73 and p63
TAp73 is inhibited in cholangiocarcinoma
is described by Hong et al. (2007). Of note,
and ΔNp63 expression is one of the mul-current immunohistochemical applications
tiple mechanisms of p73 inactivation. This
are limited by availability of isoform-
is consistent with data showing that the
specific antibodies.
promoter responsible for expression of Western blotting has also been shown to
TAp73 (P1) is hypermethylated in a subset
be useful for the detection of p73 and p63
of extrahepatic and intrahepatic cholangi-
proteins in tumor tissues. Although this
ocarcinomas (Yang et al., 2005). Loss of
approach permits evaluation of all known
heterozygosity on the chromosome 1p36 isoforms, it requires high quality clinical
region that includes the TP73 gene was materials.
also noted in some intrahepatic cholangi- In summary, current analysis suggests
ocarcinoma (Momoi et al., 2001). that expression of p63 and p73 proteins
may serve as useful prognostic markers in
cholangiocarcinoma. These proteins may
Analyses of p63 and p73 in Tumors
also play an important role in tumorigenic
mRNA: RT-PCR is a robust and effi- processes in the bile duct. However, cur-
cient approach for analyses of p73 and rent studies need to be expanded to more
448 A.I. Zaika and S.-M. Hong

patients and interpreted based on isoform- mice mutant for p63 and p73: evidence for
specific analysis. broader tumor suppressor functions for the p53
family. Cancer Cell 7: 363–373.
Green, F.L., Page, D.L., Fleming, I.D., Fritz, A.G.,
REFERENCES Balch, C.M., Haller, D.G., and Morrow, M.
2002. AJCC Cancer Staging Manual, Springer,
Ahrendt, S.A., Rashid, A., Chow, J.T., Eisenberger, New York.
C.F., Pitt, H.A., and Sidransky, D. 2000. p53 Hong, S.M., Cho, H., Moskaluk, C.A., Yu, E., and
overexpression and K-ras gene mutations in Zaika, A.I. 2007. p63 and p73 expression in ext-
primary sclerosing cholangitis-associated biliary rahepatic bile duct carcinoma and their clinical
tract cancer. J. HepatoBiliary-Pancreat. Surg. 7: significance. J. Mol. Histol. 38: 167–175.
426–431. Hong, S.M., Kim, M.J., Pi, D.Y., Jo, D., Cho, H.J.,
Albores-Saavedra, J., Henson, D.E., and Yu, E., and Ro, J.Y. 2005. Analysis of extrahe-
Klimstra, D.S. 2000. Tumors of the Gall blad- patic bile duct carcinomas according to the New
der, Extrahepatic Bile Ducts and Ampulla of American Joint Committee on Cancer staging
Vater. Atlas of Tumor Pathology, Armed Forces system focused on tumor classification problems
Institute of Pathology, Washington, DC. in 222 patients. Cancer 104: 802–810.
Argani, P., Shaukat, A., Kaushal, M., Wilentz, Khan, S.A., Thomas, H.C., Toledano, M.B., Cox,
R.E., Su, G.H., Sohn, T.A., Yeo, C.J., Cameron, I.J., and Taylor-Robinson, S.D. 2005. p53
J.L., Kern, S.E., and Hruban, R.H. 2001. Mutations in human cholangiocarcinoma: a
Differing rates of loss of DPC4 expression review. Liver Int. 25: 704–716.
and of p53 overexpression among carcinomas Lee, Y.C., Song, S.Y., Chung, J.B., Kang, J.K.,
of the proximal and distal bile ducts. Cancer and Park, I.S. 1996. p53 protein expression in
91: 1332–1341. extrahepatic bile duct cancer. Yonsei Med. J. 37:
Choi, B.I., Han, J.K., Hong, S.T., and Lee, K.H. 112–117.
2004. Clonorchiasis and cholangiocarcinoma: Leong, C.O., Vidnovic, N., DeYoung, M.P., Sgroi,
etiologic relationship and imaging diagnosis. D., and Ellisen, L.W. 2007. The p63/p73 net-
Clin. Microbiol. Rev. 17: 540–552. work mediates chemosensitivity to cisplatin in
Concin, N., Becker, K., Slade, N., Erster, S., a biologically defined subset of primary breast
Mueller-Holzner, E., Ulmer, H., Daxenbichler, cancers. J. Clin. Invest. 117: 1370–1380.
G., Zeimet, A., Zeillinger, R., Marth, C., and Malhi, H., and Gores, G.J. 2006.
Moll, U.M. 2004. Transdominant DeltaTAp73 Cholangiocarcinoma: modern advances in
isoforms are frequently up-regulated in ovarian understanding a deadly old disease. J. Hepatol.
cancer. Evidence for their role as epigenetic p53 45: 856–867.
inhibitors in vivo. Cancer Res. 64: 2449–2460. Momoi, H., Okabe, H., Kamikawa, T., Satoh,
Diamantis, I., Karamitopoulou, E., Perentes, S., Ikai, I., Yamamoto, M., Nakagawara, A.,
E., and Zimmermann, A. 1995. p53 protein Shimahara, Y., Yamaoka, Y., and Fukumoto,
immunoreactivity in extrahepatic bile duct M. 2001. Comprehensive allelotyping of human
and gallbladder cancer: correlation with tumor intrahepatic cholangiocarcinoma. Clin. Cancer
grade and survival, Hepatology 22: 774–779, Res. 7: 2648–2655.
Baltimore, MD. Munro, A.J., Lain, S., and Lane, D.P. 2005.
Farazi, P.A., Zeisberg, M., Glickman, J., Zhang, Y., P53 abnormalities and outcomes in colorectal
Kalluri, R., and DePinho, R.A. 2006. Chronic cancer: a systematic review. Br. J. Cancer 92:
bile duct injury associated with fibrotic matrix 434–444.
microenvironment provokes cholangiocarci- Murata, T., Nagasaka, T., Kamiya, J., Nimura,
noma in p53-deficient mice. Cancer Res. 66: Y., Wakai, K., Yoshida, K., and Nakashima, N.
6622–6627. 2002. P53 labeling index in cholangioscopic
Flores, E.R., Sengupta, S., Miller, J.B., Newman, biopsies is useful for determining spread of
J.J., Bronson, R., Crowley, D., Yang, A., McKeon, bile duct carcinomas. Gastrointest. Endos. 56:
F., and Jacks, T. 2005. Tumor predisposition in 688–695.
33. Extrahepatic Bile Duct Carcinoma: Role of the p53 Protein Family 449

Nagakawa, T., Kayahara, M., Ikeda, S., Futakawa, Immunohistochemical detection of proliferating
S., Kakita, A., Kawarada, H., Matsuno, M., cell nuclear antigen and p53 expression in car-
Takada, T., Takasaki, K., Tanimura, H., Tashiro, cinoma of the extrahepatic bile duct. Oncology
S., and Yamaoka, Y. 2002. Biliary tract cancer 54: 407–413.
treatment: results from the Biliary Tract Cancer Tan, G., Yilmaz, A., De Young, B.R., Behling,
Statistics Registry in Japan. J. HepatoBiliary- C., Lehman, A., and Frankel, W.L. 2004.
Pancreat. Surg. 9: 569–575. Immunohistochemical analysis of biliary tract
Nathan, H., Pawlik, T.M., Wolfgang, C.L., Choti, lesions. Appl. Immunohistochem. Mol. Morphol.
M.A., Cameron, J.L., and Schulick, R.D. 2007. 12: 193–197.
Trends in survival after surgery for cholangi- Tannapfel, A., Engeland, K., Weinans, L., Katalinic,
ocarcinoma: a 30-year population-based SEER A., Hauss, J., Mossner, J., and Wittekind, C.
database analysis. J. Gastrointest. Surg. 11: 1999. Expression of p73, a novel protein related
1488–1496; discussion: 1496–1487. to the p53 tumour suppressor p53, and apoptosis
Nomoto, K., Tsuneyama, K., Cheng, C., Takahashi, in cholangiocellular carcinoma of the liver. Br. J.
H., Hori, R., Murai, Y., and Takano, Y. 2006. Cancer 80: 1069–1074.
Intrahepatic cholangiocarcinoma arising in cir- Vilgelm, A., El-Rifai, W., and Zaika, A. 2008.
rhotic liver frequently expressed p63-positive Therapeutic prospects for p73 and p63: Rising
basal/stem-cell phenotype. Pathol. Res. Pract. from the shadow of p53. Drug Resist. Updat.
202: 71–76. 11: 152–163.
Seyama, Y., and Makuuchi, M. 2007. Current Yang, B., House, M.G., Guo, M., Herman, J.G., and
surgical treatment for bile duct cancer. World J. Clark, D.P. 2005. Promoter methylation profiles
Gastroenterol. 13: 1505–1515. of tumor suppressor genes in intrahepatic and
Suto, T., Sugai, T., Nakamura, S., Uesugi, N., extrahepatic cholangiocarcinoma. Mod. Pathol.
Sasaki, R., Kanno, S., and Saito, K. 1997. 18: 412–420.
34
Extrahepatic Bile Duct Carcinoma:
Mucin 4, a Poor Prognostic Factor
Michiyo Higashi, Shugo Tamada, Kohji Nagata, Masamichi Goto,
and Suguru Yonezawa

INTRODUCTION MUCIN CHARACTERISTICS


Malignant tumors of the extrahepatic bile In general, mucins remain at the api-
ducts are relatively rare, although they cal surface of epithelial cells and are
are one of the most common causes of capable of providing a lubricant func-
extrahepatic bile duct obstruction. Most tion by their abilities to retain water ions
tumors of extrahepatic bile ducts are car- and their entanglement through intra- or
cinomas, and they can develop at any intermolecular disulfide linkages. Their
level of bile duct tree. Extrahepatic bile functions are not only protection and
duct carcinoma (EHBDC) is difficult to lubrication of epithelial surfaces, but
treat because most patients with this car- also signaling modulators and affect-
cinoma are not surgically resectable at the ing tumor cell phenotype. Mucins are
time of diagnosis despite its small size. high molecular weight glycoproteins with
A gradual improvement with better prog- oligosaccharides attached to serine or
nosis has been obtained with a combined threonine residues of the mucin core
modality approach consisting of surgery, protein backbone by O-glycosidic link-
radiation therapy, and chemotherapy, but ages, which are produced by various
survival rate is still low. A complete surgi- epithelial cells. In the past few years,
cal resection offers the only possibility of 20 core proteins for human mucin genes
cure; however, many patients treated with (MUC) have been identified and des-
curative resection have a poor outcome. ignated as MUC1, MUC2, MUC3A,
Our previous immunohistochemical stud- MUC3B, MUC4, MUC5AC, MUC5B,
ies of EHBDC showed that MUC4 mucin MUC6-13, MUC15-17, MUC19, and
expression is a new independent factor for MUC20 (Hollingsworth and Swanson,
poor prognosis and predicts the outcome 2004). Mucins are categorized into mem-
of patients with EHBDC (Tamada et al., brane-associated mucins (MUC1, MUC3
2006), in addition to MUC1 mucin expres- MUC4, MUC12, MUC16 and MUC17),
sion (Tamada et al., 2002). gel forming mucins (MUC2, MUC5AC,

451
452 M. Higashi et al.

MUC5B and MUC6), and soluble mucin central large tandem repeat domain con-
(MUC7) (Hollingsworth and Swanson, taining 16-amino acid repetitive units,
2004). regions rich in potential N-glycosylation
sites, two cystein-rich domains, a putative
GDPH proteolytic cleavage site, three epi-
MUC4 MUCIN dermal growth factor (EGF)-like domains,
a hydrophobic transmembrane domain,
MUC4, a transmembrane mucin, has and a short cytoplasmic tail (Singh
gained significant biological concern in et al., 2007). Approximately two thirds of
the past few years. Like MUC1, MUC4 the MUC4 protein sequence consists of
is a membrane mucin; however, MUC4 identical 16-amino-acid tandem repeats
functions through different mechanisms (Moniaux et al., 2004).
than those of MUC1. As a signaling mol- MUC4/Sialomucin complex (SMC) is
ecule, MUC1 acts as a docking protein, a rat homologue of human mucin gene
whereas MUC4 acts as a receptor ligand MUC4 and its transmembrane subunit acts
(Ramsauer et al., 2003). The difference as an intramembrane ligand for the recep-
of the expression patterns suggests the tor tyrosine kinase ErbB2 to induce the
possibility of different mechanisms in phosphorylation of Tyr-1248 of the ErbB2
the expression of MUC1 and MUC4, (DiGiovanna and Stern, 1995; Jepson
both of which are membrane mucins pos- et al., 2002; Ramsauer et al., 2003). The
sessing cell signaling functions. MUC4 heterodimeric glycoprotein complex con-
extends at least 1.12–2.12 μm above the sists of ASGP-1 (ascites sialoglycopro-
cell membrane, far above all other mem- tein-1), an O-glycosylated mucin subunit,
brane-associated proteins, such as adhe- and ASGP-2, an N-glycosylated trans-
sion molecules (Moniaux et al., 1999). membrane subunit (Singh et al., 2007).
MUC4 with its rigid and extended struc- MUC4/SMC leads to the expression of
ture is considered a modulator for cell–cell the cell-cycle inhibitor p27 (Ramsauer
and cell-extracellular matrix interactions et al., 2003). On the other hand, MUC4/
(Komatsu et al., 1999). SMC acts with neuregulin synergistically
The gene encoding the MUC4 mucin to enhance phosphorylation of both ErbB2
was first identified in 1991 from a tracheo- and ErbB3, resulting in the downregula-
bronchial cDNA library (Porchet et al., tion of p27 and activation of protein kinase
1991). However, due to its large size and B/Akt. It is proposed that complex forma-
complex structure, its complete genomic tion of MUC4/SMC and ErbB2 has an effect
organization could not be established until on epithelial cell behavior, as a switch in
2000 (Moniaux et al., 1999; Moniaux epithelial differentiation and proliferation
et al., 2000). MUC4 gene is located on (Jepson et al., 2002). Furthermore, Singh
chromosome 3 in the q29 region that et al. (2004) proposed that MUC4 partici-
codes a family of 25 alternatively spliced pates in tumor growth and metastasis by
transcripts. The deduced full-length amino directly altering the tumor cell properties,
acid sequence of MUC4 apoprotein shows and/or via modulating ErbB2 expression.
the presence of a leader peptide, serine and Komatsu et al. (1999) showed that SMC
threonin rich non-tandem repeat region, disrupts integrin-mediated cell adhesion
34. Extrahepatic Bile Duct Carcinoma: Mucin 4, a Poor Prognostic Factor 453

to extracellular matrix proteins. In addi- ANTIBODIES AGAINST MUC4


tion, overexpression of SMC masks the
surface antigens on target tumor cells We use two monoclonal antibodies against
and effectively suppresses tumor cell kill- MUC4. One of the antibodies, clone 8G7,
ing by cytotoxic lymphocytes (Komatsu was generated and provided by Surinder
et al., 1999). These phenomena may be K. Batra, University of Nebraska Medical
related with the poor outcome of patients Center, Omaha, NE. The 8G7 shows high
with MUC4 overexpression, although specificity and avidity in ELISA, Western
MUC4 expression is not related to the blotting, immunoprecipitation, and in
morphological invasive parameters, such immunohistochemical examination. This
as lymphatic invasion, venous invasion, antibody recognizes an epitope on the
perineural invasion, and distant lymph 16-amino-acid tandem repeats, which are
node metastasis. flanked by unique sequences (Moniaux
However, in EHBDCs examined in et al., 2004). The other antibody is
our study, ErbB2 expression was not a purchased from Invitrogen, South San
significant prognostic factor (Tamada Francisco, U.S. (previously distributed by
et al., 2006). The combined evaluation Zymed). This antibody, clone 1G8, recog-
of MUC4 and ErbB2 expression did not nizes an epitope on the ASGP-2 subunit,
show significant results. Low expression which is one of the main subunits that
of p27 is reported to be a poor prognos- comprise the MUC4 glycoprotein complex
tic factor in gastric cancer (Sgambato (data sheet). We have shown the represent-
et al., 2000), colorectal carcinoma (Chen ative positive immunohistochemical stain-
et al., 2002), and intrahepatic cholan- ing of MUC4 (Figures 34.1 and 34.2). The
giocarcinoma (Taguchi et al., 2001). In difference between these two antibodies
EHBDCs examined in our study, how- consists of antibodies’ reactivity with the
ever, the p27 expression itself was not an endothelial cells of small blood vessels and
independent prognostic factor (Tamada duodenal mucosal epithelia. The stained
et al., 2006). In the combined evaluation, by the clone 1G8 is seen in the blood ves-
the patients with MUC4 overexpression sels and duodenal mucosal epithelia, in
and p27 overexpression did not show addition to the cancer cells, whereas it by
significantly worse outcome than those the clone 8G7 is not observed in them.
with MUC4 low expression and p27 low
expression. There may be a difference
between the cascade reaction in vitro and MUC1 MUCIN
the immunohistochemical localization in
vivo tumor tissues. In conclusion, impor- MUC1 is a polymorphic, highly glycosy-
tant information on the degree of MUC4 lated, type I transmembrane protein
expression is a useful indicator to predict expressed by ductal epithelial cells of many
the outcome of patients with EHBDCs, organs including gastrointestinal tract, pan-
who had surgical resection of the tumor. creas, breast, and airway. MUC1 and MUC4
MUC4 could potentially be a useful present significantly different structures
marker for the clinical management of although they both belong to the same mucin
patients with EHBDCs. subfamily (membrane-bound mucins).
454 M. Higashi et al.

a a

b b

Figure 34.1. Expression of MUC4 in EHBDC. Figure 34.2. Expression of MUC4 in duodenal
(a). MUC4, clone 8G7 (b) MUC4, clone 1G8. The mucosa. a. MUC4, clone 8G7 The clone 8G7 is
clone 8G7 antibody is expressed in the cytoplasm not expressed in the duodenal mucosa. b. MUC4,
and/or membrane of the carcinoma cells, and is clone 1G8 The clone 1G8 stains the luminal sur-
negative for normal peribilliary glands (arrow- face of the duodenal epithelia
heads). The clone 1G8 is weakly expressed in the
membrane of the carcinoma cells, and also stains amino acids (Hollingsworth and Swanson,
the endothelial cells of the small blood vessels. This is 2004). In normal individuals, there is a
negative for normal peribilliary glands (arrowheads)
variation in the number of repeats per allele
(from 25 to > 125 repeats), implying that
MUC1 has a relatively simple architec- the length of the repetitive array may be
ture. The MUC1 gene codes a membrane- critical for the normal function of MUC1.
associated protein with distinct domains; The large extracellular central domain is
an amino-terminal domain consisting of a heavily O-glycosylated. Glycosylation
20-amino acid signal peptide and degener- varies among different tissues, and tumor-
ate tandem repeats; a large central domain associated MUC1 has been reported to be
(two-thirds of the core protein sequence) either less glycosylated or more glyco-
made up of variable numbers of a 20-amino sylated than the forms expressed by normal
acid repeat; a carboxyl terminus made of tissue (Higashi et al., 1999; Horinouchi
degenerate tandem repeats; a hydrophobic et al., 2003). Another important domain of
membrane-spanning domain of 31 amino MUC1 is the cytoplasmic domain, which is
acids, and a cytoplasmic domain of 69 highly conserved across mammalian species
34. Extrahepatic Bile Duct Carcinoma: Mucin 4, a Poor Prognostic Factor 455

and hypothesized to play a role in its post- sialylated-MUC1 mucin (Yamamoto


translational processing, subcellular locali- et al., 1996). The binding of MUC1/
zation, signal transduction, and intracellular HMFG-1 (provided by Dr. Joyce Taylor-
localization (Hollingsworth and Swanson, Papadimitriou, Imperial Cancer Research
2004). Fund laboratories, London, U.K.) to core
protein epitopes is influenced by the car-
bohydrate chains, and MUC1/HMFG-1
ANTIBODIES AGAINST MUC1 detects fully glycosylated MUC1 mucin,
and its binding is particularly affected by
The most extensively studied mucin is sialic acid (Burchell et al., 1983).
MUC1. As a potential tool in the immuno- The expression of various glycoforms
detection and immunotherapy of cancer, of MUC1 mucins was also recognized in
a large number of monoclonal antibodies other human carcinomas of the stomach
have been raised against the protein core (Utsunomiya et al., 1998), intrahepatic bile
of MUC1. Fifty six antibodies were clas- duct (Higashi et al., 1999), and pancreas
sified at a workshop of the International (Horinouchi et al., 2003). Our study of
Society for Oncodevelopmental Biology EHBDC demonstrated that MUC1 expres-
and Medicine in San Diego in 1996, and sion of poorly glycosylated MUC1 (MUC1/
the results have been summarized else- CORE and MUC1/DF3) and glycosylated
where (1998). The majority of antibod- MUC1 (MUC1/MY.1E12) showed a sig-
ies against MUC1 defined epitopes are nificant relationship with tumor progre-
located within the 20-amino acid tandem sion factors such as poor differentiation,
repeat sequence of the MUC1 protein deep invasion, lymph node metastasis,
core. Of the remaining antibodies, there lymphatic invasion, or perineural inva-
was evidence for the involvement of car- sion. In contrast, the expression of MUC1/
bohydrate residues in the epitopes. HMFG-1 (fully glycosylated MUC1) did
Several monoclonal antibodies against not show a significant relationship with
MUC1 are available, which are used for tumor progression factors (Tamada et al.,
detecting different glycoforms of MUC1. 2002). Figure 34.3 shows representative
In the following section, we focus on four positive immunohistochemical stainings
monoclonal antibodies, NCL-MUC-1- of MUC1 in EHBDC tissues.
CORE (MUC1/CORE), DF3(MUC1/DF3),
MY. 1E12(MUC1/MY.1E12), and HMFG-
1(MUC1/HMFG-1). MUC1/CORE recog-
nizes MUC1 mucin core peptide (TRPAPG) METHODS
(data sheet from Novocastra Laboratories
Tissue Sample Management
Ltd., New Castle, U.K.). The MUC1/ DF3
(Toray-Fuji Bionics, Tokyo, Japan) identi- Appropriate fixation is very important for
fies MUC1 core peptide (TRPAPGS), but handling tissue samples. Less than opti-
MUC1/DF3 binding to protein is thought mal fixation may result in worse results
to be enhanced by the presence of carbo- not only in the histological examina-
hydrates. MUC1/ MY. 1E12 (developed in tion, but also in the immunohistochemi-
the laboratory of Dr. T. Irimura, University cal examination. The resected specimens
of Tokyo, Tokyo, Japan) is specific for should be fixed as soon as possible. Both
456 M. Higashi et al.

b c

d e

Figure 34.3. Expression of MUC1 in EHBDC. a. Hematoxylin and eosin, b. MUC1/CORE, c. MUC1/
DF3, d. MUC1/MY1E12, e. MUC1/HMFG-2. MUC1/DF3 (c) and MUC1/MY1E12 (d) are more widely
expressed in the carcinoma cells than in the MUC1/CORE (b) and MUC1/HMFG-2 (e). MUC1/HMFG-2
is partially glycosylated MUC1 (Burchell et al., 1983)

bile duct and pancreatic duct epithelia are the pancreaticoduodenectomy. A suitable
especially prone to undergo degenera- opening procedure is necessary because
tion (autolytic change), thus much atten- the anatomy of pancreas head is com-
tion should be paid to the specimens of plex. These guidelines are very useful not
34. Extrahepatic Bile Duct Carcinoma: Mucin 4, a Poor Prognostic Factor 457

only for histological diagnosis, but also We should avoid opening both common
for immunohistochemical and genomic bile duct and main pancreatic duct, because
examination. the shape of the resected specimens may
Here, we describe the proper handling be distorted and the orientation will be
procedure of pancreaticoduodenectomy lost. Only common bile duct thus should
specimens. First of all, the orientation of be opened. It is better to stop opening the
duodenum is recognized. There are two bile duct before the papilla of Vater for the
points to distinguish the proximal and dis- preservation of its structure of the papilla
tal sides of the duodenum. One of the dis- of Vater. Photographs with a scale bar are
tinguishing features is at the proximal free taken, and fixation is completed.
end, which is shorter than that at the distal The orientation and the shape should be
free end. The other is that the stomach adequately kept when chemical fixation is
(pyloric region), is sometimes attached at performed. A corkboard, and pins or nee-
the proximal margin. Subsequently, bile dles are useful tools, because the mucosa
duct should be identified. When the gall- of the duodenum and bile duct is kept in
bladder is attached to the resected speci- the correct position when the mucosa is
mens, it is relatively easy to identify the immobilized with these tools on the cork-
bile duct. We follow the gallbladder, and board. The corkboard may be replaced
the cystic duct to common bile duct ante- by a board made of Styrofoam or rubber.
gradely. When the gallbladder has been Injection of fixative solution into the pan-
removed, we have to find the suture thread creas parenchyma before attaching it on
of bile duct ligation. Usually, the distal the board is also recommended in order
side of the bile duct is dilated; and these to avoid autolysis. An 18 gauge is suitable
procedures are easy. But in some cases it is for this purpose. Injection of too much
difficult to find the bile duct. In such cases, fixative should be avoided especially for
we follow the bile duct from the papilla of adipose tissue because the adipose tissue
Vater retrogradely after opening the duo- shows edematous change.
denum. We have to identify the anterior The cases of simple extrahepatic bile
and posterior sides before opening the duct excision are relatively simply handled
duodenum. The superior mesenteric vessel when compared to those of pancreati-
sometimes indents the uncinate process to coduodenectomy. After the identification
form a groove called the pancreatic notch. of the anatomical orientation, the bile
The opening of the duodenum should be duct is opened in a longitudinal direction.
performed after the correct orientation of Injection fixation is useful if the dissection is
the above sites has been noted. It is better not done. Pinning is necessary in all cases.
to open the duodenal wall near the pancreas All specimens are fixed in 10 % buffered
at the posterior side than opening it at the formalin or 10 % formalin. Sampling for
opposite side (lateral side), because with histological examination should be done
the former method it is easy to observe the within 48 h after fixation. Prolonged fixta-
papilla of Vater. The common bile duct is tion may produce formalin pigments and
also opened at the posterior side after open- loss of the antigenecity. Further fixation is
ing the duodenum. A long metal probe of expected by cutting out thin blocks from
small diameter can be used as a guide wire. the large and thick specimens. Sections are
458 M. Higashi et al.

embedded in paraffin, and cut into 4 μm PBS. The sections then receive Elite ABC
thick serial sections for usual hematoxylin(Vector Laboratories, Burlingame, CA)
and eosin staining, and further immunohis- for 30 min. After washing with PBS three
tochemistry. Silan-coated glass slides are times, the sections are finally reacted
suitable for immunohistochemistry. with diaminobenzidine substrate (0.2 mg/
ml) for 10 min for visualization, rinsed
with tap water, counterstained with hema-
IMMUNOHISTOCHEMISTRY toxylin and mounted. Reaction products
are not present when nonimmune serum
Immunohistochemical staining is per- or PBS is used instead of the primary
formed by the immunoperoxidase method antibodies.
using the avidin–biotinylated horserad-
ish peroxidase complex (ABC complex),
REFERENCES
(Vector Laboratories, Burlingame, CA).
The sections are deparaffinized with Burchell, J., Durbin, H., and Taylor-Papadimitriou,
J. 1983. Complexity of expression of antigenic
xylene for 30 min followed by 100 %
determinants recognized by monoclonal anti-
ethanol three times. After hydration in bodies HMFG-1 and HMFG-2 in normal and
decreasing concentrations of ethanol in malignant human mammary epithelial cells. J.
water (95 %, 70 %, 50 %, 30 % and 20 %, Immunol. 131: 508–513.
5 min for each), the sections are washed Chen, A. J., Meng, Q. H., Long, B., and Yang,
in 0.01 mol/l phosphate buffered saline, G. L. 2002. Relationship between p27 expres-
sion and prognosis of colorectal carcinoma. Ai
pH 7.4 (PBS). Endogenous peroxidase
Zheng. 21: 1075–1077.
is blocked by incubating the sections in DiGiovanna, M. P., and Stern, D. F. 1995. Activation
0.3 % hydrogen peroxidase in absolute state-specific monoclonal antibody detects tyro-
methanol at room temperature for 30 min. sine phosphorylated p185neu/erbB-2 in a subset
For the epitope retrieval of MUC4, the of human breast tumors overexpressing this
slides are immersed in a water bath pre- receptor. Cancer Res. 55: 1946–1955.
Higashi, M., Yonezawa, S., Ho, J. J., Tanaka, S.,
treatment at 80°C for 20 min in 0.01 M
Irimura, T., Kim, Y. S., and Sato, E. 1999.
citrate buffer (pH6.0). After heating, the Expression of MUC1 and MUC2 mucin antigens
samples are allowed to cool for 15–20 min in intrahepatic bile duct tumors: its relation-
at room temperature. ship with a new morphological classification of
The sections are washed twice with PBS cholangiocarcinoma. Hepatology 30: 1347–1355.
after cooling. Then, 2 % horse serum in Hollingsworth, M. A., and Swanson, B. J. 2004.
Mucins in cancer: protection and control of the
PBS is applied for 30 min at room tem-
cell surface. Nat. Rev. Cancer 4: 45–60.
perature to prevent nonspecific staining. Horinouchi, M., Nagata, K., Nakamura A., Goto
The sections are washed twice with PBS, M., Takao S., Sakamoto M., Fukushima N., Miwa
and then incubated with diluted anti- A., Irimura T., Imai K., Sato E. and Yonezawa
MUC4 monoclonal antibody (clone 8G7 S. 2003. Expression of different glycoforms
1:3000, clone 1G8 1:100) in PBS with 1 % of membrane mucin (MUC1) and secretory
mucin (MUC2, MUC5AC and MUC6) in pan-
bovine serum albumin for 16 h at 4°C. The
creatic neoplasms. Acta Histochem. Cytochem.
sections are washed three times with PBS 36: 443–453.
and incubated with the biotinylated anti- Jepson, S., Komatsu, M., Haq, B., Arango, M.
mouse IgG, and washed three times with E., Huang, D., Carraway, C. A., and Carraway,
34. Extrahepatic Bile Duct Carcinoma: Mucin 4, a Poor Prognostic Factor 459

K. L. 2002. Muc4/sialomucin complex, the Ramsauer, V. P., Carraway, C. A., Salas, P. J.,
intra-membrane ErbB2 ligand, induces spe- and Carraway, K. L. 2003. Muc4/sialomucin
cific phosphorylation of ErbB2 and enhances complex, the intramembrane ErbB2 ligand,
expression of p27(kip), but does not activate translocates ErbB2 to the apical surface in
mitogen-activated kinase or protein kinaseB/Akt polarized epithelial cells. J. Biol. Chem. 278:
pathways. Oncogene 21: 7524–7532. 30142–30147.
Komatsu, M., Yee, L., and Carraway, K. L. 1999. Sgambato, A., Migaldi, M., Leocata, P., Ventura,
Overexpression of sialomucin complex, a rat L., Criscuolo, M., Di Giacomo, C., Capelli, G.,
homologue of MUC4, inhibits tumor killing by Cittadini, A., and De Gaetani, C. 2000. Loss
lymphokine-activated killer cells. Cancer Res. of p27Kip1 expression is a strong independent
59: 2229–2236. prognostic factor of reduced survival in N0 gas-
Moniaux, N., Escande, F., Batra, S. K., Porchet, tric carcinomas. Cancer 89: 2247–2257.
N., Laine, A., and Aubert, J. P. 2000. Alternative Singh, A. P., Chaturvedi, P., and Batra, S. K. 2007.
splicing generates a family of putative secreted Emerging roles of MUC4 in cancer: a novel
and membrane-associated MUC4 mucins. Eur. J. target for diagnosis and therapy. Cancer Res. 67:
Biochem. 267: 4536–4544. 433–436.
Moniaux, N., Nollet, S., Porchet, N., Degand, P., Singh, A. P., Moniaux, N., Chauhan, S. C., Meza,
Laine, A., and Aubert, J. P. 1999. Complete J. L., and Batra, S. K. 2004. Inhibition of MUC4
sequence of the human mucin MUC4: a putative expression suppresses pancreatic tumor cell growth
cell membrane-associated mucin. Biochem. J. and metastasis. Cancer Res. 64: 622–630.
338: 325–333. Taguchi, K., Aishima, S., Asayama, Y., Kajiyama,
Moniaux, N., Varshney, G. C., Chauhan, S. K., Kinukawa, N., Shimada, M., Sugimachi, K.,
C., Copin, M. C., Jain, M., Wittel, U. A., and Tsuneyoshi, M. 2001. The role of p27kip1
Andrianifahanana, M., Aubert, J. P., and Batra, protein expression on the biological behavior of
S. K. 2004. Generation and characterization intrahepatic cholangiocarcinoma. Hepatology
of anti-MUC4 monoclonal antibodies reactive 33: 1118–1123.
with normal and cancer cells in humans. J. Tamada, S., Goto, M., Nomoto, M., Nagata, K.,
Histochem. Cytochem. 52: 253–261. Shimizu, T., Tanaka, S., Sakoda, K., Imai, K.,
Porchet, N., Nguyen, V. C., Dufosse, J., Audie, J. and Yonezawa, S. 2002. Expression of MUC1
P., Guyonnet-Duperat, V., Gross, M. S., Denis, and MUC2 mucins in extrahepatic bile duct car-
C., Degand, P., Bernheim, A., and Aubert, J. cinomas: its relationship with tumor progression
P. 1991. Molecular cloning and chromosomal and prognosis. Pathol. Int. 52: 713–723.
localization of a novel human tracheo-bronchial Tamada, S., Shibahara, H., Higashi, M., Goto, M.,
mucin cDNA containing tandemly repeated Batra, S. K., Imai, K., and Yonezawa, S. 2006.
sequences of 48 base pairs. Biochem. Biophys. MUC4 is a novel prognostic factor of extrahe-
Res. Commun. 175: 414–422. patic bile duct carcinoma. Clin. Cancer Res. 12:
Price, M. R., Rye, P. D., Petrakou, E., Murray, 4257–4264.
A., Brady, K., Imai, S., Haga, S., Kiyozuka, Y., Utsunomiya, T., Yonezawa, S., Sakamoto, H.,
Schol, D., Meulenbroek, M. F., Snijdewint, F. Kitamura, H., Hokita, S., Aiko, T., Tanaka, S.,
G., von Mensdorff-Pouilly, S., Verstraeten, R. A., Irimura, T., Kim, Y. S., and Sato, E. 1998.
Kenemans, P., Brockzjil, A., Nilsson, K., Nilsson, Expression of MUC1 and MUC2 mucins in gas-
O., Reddish, M., Suresh, M. R., Koganty, R. R., tric carcinomas: its relationship with the prognosis
Fortier, S., Baronic, L., Berg, A., Longenecker, of the patients. Clin. Cancer Res. 4: 2605–2614.
M. B., Hilgers, J. et al. 1998. Summary report Yamamoto, M., Bhavanandan, V. P., Nakamori, S.,
on the ISOBM TD-4 Workshop: Analysis of 56 and Irimura, T. 1996. A novel monoclonal anti-
monoclonal antibodies against the MUC1 mucin. body specific for sialylated MUC1 mucin. Jpn.
Tumour Biol. 19(Suppl 1): 1–20. J. Cancer Res. 87: 488–496.
C. Treatment
35
Hilar Cholangiocarcinoma: Photodynamic
Therapy and Stenting
Marcus Wiedmann, Joachim Mössner, and Helmut Witzigmann

SUMMARY plications can be managed endoscopically


and the bile duct remains accessible for
Carcinomas of the biliary tree are rare tumors endoluminal treatment. Palliative photo-
of the gastrointestinal tract with rising inci- dynamic therapy (PDT) for non-resectable
dence during the last years. Endoscopic hilar CC is effective in prolonging survival,
therapy plays a central role in the preopera- improving biliary drainage and quality of
tive and palliative treatment of extrahepatic life, and represents a promising adjunct to
cholangiocarcinoma (CC). In obstructive palliative therapy. Early referral for PDT
jaundice, biliary drainage may cause only may provide maximum survival benefit.
few complications and relieves symptoms In contrast, neoadjuvant and adjuvant PDT
reliably. It can prevent further complica- for hilar CC is a new approach that needs
tions and is indispensable in the treatment further evaluation.
of cholangitis. The principal drawbacks
of biliary stents are stent occlusion and
cholangitis. Prophylactic application of INTRODUCTION
antibiotics and/or ursodeoxycholic acid has
not been shown to be effective. Technical According to de Groen et al. (1999),
details concerning material, number and 67 % of biliary tract tumors are gallbladder
location of stents await further clarification. carcinomas, 22 % are hilar carcinomas
There are no prospective randomized tri- (Klatskin tumors), 8 % occur in the dis-
als investigating the effect of preoperative tal bile duct, and 3 % are intrahepatic
biliary drainage on morbidity and mortality cholangiocarcinomas (ICC). Since 1970
in jaundized patients before liver resection. the incidence of ICC is increasing in North
Palliative surgical cholangio-jejunostomy America and Western Europe (Welzel
with the bile duct of liver segment III for et al., 2006; West et al., 2006). The cause
Klatskin-tumors is rarely performed due of this rise is unknown and does not appear
to the high success rate of stent therapy. to be explained simply by improvements
Self-expandable metal stents (SEMS) are a in diagnosis or changes in coding practice.
safe, efficacious and less invasive treatment Besides regional etiological factors such as
option for palliation of duodenal obstruc- liver fluke infection and hepatolithiasis, it
tion in biliary tract cancer. Technical com- was hypothesized by Khan et al. (2002) that

463
464 M. Wiedmann et al.

this increase may be the result of a rise in one 1999; Miyazaki et al., 1998; Seyama et al.,
or several genotoxic environmental agents, 2003; Todoroki et al., 2000). We performed
causing cholangiocyte DNA damage. preoperative biliary drainage according to
At present, complete resection is the these criteria in our patients and obtained
only potentially curative therapy, but most a median preoperative bilirubin level of
of the patients with intra- and extrahepatic 2.1 mg/dl. Recovery of hepatic function
CC present with already advanced disease. after biliary decompression takes 4 weeks,
Given the small size of available studies so surgery should not be done too early.
and lack of randomized trials, there is no
established role for neoadjuvant and adju-
vant therapy. In the palliative setting, sys- PALLIATIVE BILE DUCT
temic chemotherapy (chemoradiation) has STENTING
not been clearly proven to prolong survival
significantly as discussed by Lazaridis and The goal of palliative treatment is to achieve
Gores (2005) in a review article. stable long-term biliary drainage in order to
improve quality of life and prolong survival.
Refractory obstructive cholestasis leads to
PREOPERATIVE BILIARY death owing to severe bacterial cholangitis
DRAINAGE or liver failure within 3–6 months. Because
surgical drainage procedures do not reveal
Liver dysfunction caused by obstructive any advantage to nonsurgical palliation
jaundice is a relevant risk factor in major according to studies such as by Kosuge
hepatic resections. There are no prospec- et al. (1999), biliary stenting is regarded as
tive randomized trials investigating the the palliative method of choice. Endoscopic
effect of preoperative biliary drainage approach for biliary stent insertion is pre-
on morbidity and mortality in jaundized ferred over an ultrasound-guided percuta-
patients before liver resection. In non- neous approach due to a lower complication
randomized studies such as by Cherqui rate. Furthermore, cholestasis was more
et al. (2000) no difference in mortality efficiently reduced and 30-day and overall
was observed between patients with or mortality rates were lower in a randomized
without biliary drainage. No difference study (Speer et al., 1987). Percutaneous
with respect to morbidity was seen in three transhepatic cholangiodrainage (PTCD)
studies and in our own series of 60 patients (Figure 35.1) should be used after failure
with hilar CC who underwent resection. of endoscopy and for anatomically dif-
On the other hand, higher complication ficult situations, i.e., after partial stomach
rates after biliary stenting were reported resection or gastrectomies, especially with
by other authors. Despite these inconclu- Y-Roux-anastomosis (Doctor et al., 1999).
sive data, the authors of the majority of Often in these cases the papilla Vateri can-
recent studies dealing with hilar CC recom- not be reached endoscopically. In selected
mend biliary drainage in order to decrease cases both methods can be combined as a
the serum bilirubin level below 2–5 mg/ “rendezvous-approach”. A new technique
dl before resection (Ebata et al., 2003; of endoscopic biliary-enteric bypass was
Hemming et al., 2005; Kawasaki et al., introduced by Burmester et al. (2003),
2003; Kondo et al., 2004; Kosuge et al., which comprises endosonographically
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 465

ursodeoxycholic acid were useful in several


studies.
Technical questions regarding stent mate-
rial, number, and location of stents have
not been answered definitely. Polyurethane
and polyethylene seemed to be slightly
better in comparison to Teflon because of
a four-times lower occlusion rate accord-
ing to preclinical studies.
In a prospective randomized trial of Teflon
versus polyethylene stents for distal malig-
nant biliary obstruction by van Berkel et
al. (1998) Teflon material did not improve
patency in biliary stents with an Amsterdam-
type design. In this study, 42 patients received
Figure 35.1. Klatskin-tumor Bismuth-Corlette a Teflon stent and 42 patients a polyeth-
stage IV. Right-sided external–internal percuta- ylene stent between September 1995 and
neous transhepatic cholangiodrainage (PTCD; November 1996. All patients had a distal
Yamakawa-endoprosthesis; thick white arrows) and malignant biliary stricture without a previ-
left-sided pigtail endoprosthesis (thin white arrows) ous drainage procedure. Diagnoses included
which was inserted endoscopically (* gallbladder)
carcinoma of the pancreas (n = 76), papilla
(n = 1), bile duct (n = 5) and metastases
guided bile duct puncture followed by bil- (n = 2). The internal and external diam-
iary drainage into the duodenum. eter (10 French (F) ), length (9 cm) and stent
The major complication of endoprosthesis design (a straight stent with two side flaps
implantation is stent occlusion with consec- and one side hole at each end) were similar
utive cholangitis. Light and electron micro- for both stents. As a result, a reduction in
scopy studies of blocked stents revealed that bilirubin of more than 20 % within one week
the material blocking the lumina was com- was seen in 91 % of the patients. Early com-
posed of a matrix of bacteria and their fibril- plication rates were similar in both groups
lar anionic extracellular products. Crystals (10 %). The median follow-up was 142 days.
of calcium bilirubinate, calcium palmitate, Stent dysfunction occurred in 28 Teflon and
and cholesterol were embedded within this 29 polyethylene stents. The 30-day mortality
matrix. Bacteria were attached to the stent was 14 % in both groups. Patient survival did
surface by a fibrillar matrix, suggesting not differ significantly between the groups
that the initial event in stent clogging is (median survival: Teflon 165 days, polyeth-
the development of an adherent bacterial ylene 140 days). The median stent patency
biofilm. Bacterial enzyme activity (beta- was 83 days for Teflon and 80 days for
glucuronidase and phospholipase) leads to polyethylene stents, and was not significantly
the deposition of crystals. Therefore, it was different.
assumed that the use of antibiotics and/or To assess whether the use of large stents
ursodeoxycholic acid may reduce bacte- is justified, Speer et al. (1988) retrospectively
rial adhesion and stent clogging. However, reviewed the results of 8 F stents with pig-
prophylactic long-term antibiosis and/or tails and 10 F straight stents in the palliation
466 M. Wiedmann et al.

of biliary obstruction due to malignancy. (2003) concluded that these results showed
The incidence of cholangitis following that DoubleLayer™ stents have a longer
stent insertion was significantly lower with patency period than polyethylene stents.
10 F stents compared with 8 F stents (5 % Another new development, as described
vs. 34 %, p < 0.001). Stent survival until by Raju et al. (2006), is a 10 F “winged”
blockage was significantly longer for 10 F stent without a lumen to prevent stent occlu-
compared with 8 F (median 32 weeks vs. sion. Finally, our own group, Wiedmann
median 12 weeks, p < 0.001). et al. (2004b), studied in a small case
In addition, Kadakia and Starnes (1992) series, whether bacterial colonization and
retrospectively compared the efficacy and incrustration of stents can be avoided
complications of 10 F biliary stents with by using an external-internal Yamakawa-
11.5 F stents in the management of malig- endoprosthesis in combination with a sub-
nant and benign biliary tract diseases. cutaneously implanted port system. A total
They concluded that 10 F stents have the of three patients with bile duct obstruc-
same success rate and complication rate as tion (1 × recurrence of gastric cancer,
11.5 F stents in the management of biliary 1 × invasive gallbladder cancer, 1 × recur-
tract diseases; thus, a stent size larger than rence of bile duct cancer) were included.
10 F offers no significant advantage. In all patients open biliary drainage and
A new approach may be the use of prevention of cholangitis were achieved.
an Olympus DoubleLayer™ stent (DLS; Therefore, this method is currently being
perfluoro alkoxy, without sideholes). In a evaluated in a larger number of patients.
prospective randomized comparison of this Cost analyses according to studies by Prat
stent with the standard polyethylene stent et al. (1998) and Wagner et al. (1993) in
(with sideholes), including a total of 120 patients with middle and distal bile duct or
patients with jaundice due to malignant ampullary cancer showed that metallic stents
strictures of the middle to distal third of were advantageous in patients surviving
the common bile duct, 28 DLS patients more than 6 months, whereas a plastic stent
(47 %) and 17 polyethylene stent patients was advantageous in patients surviving 6
(29 %) died without clinical evidence of months or less (Figure 35.2). Self-expanding
stent occlusion after a mean of 114 and metal stents (SEMS) which can be inserted
105 days, respectively (p < 0.05). Twenty- endoscopically and percutaneously, have
six DLS patients (43 %) and 38 polyeth- the advantage of a higher patency rate with
ylene stent patients (63 %) had symptoms consecutive lower rates of reinterventions
of stent clogging after a mean of 144 and rehospitalizations in comparison to less
and 99 days, respectively (p < 0.05). Stent costly plastic stents (Deviere et al., 1988;
dysfunction (stent orifice impacted in the Wagner et al., 1993). The higher patency
bile duct or duodenal wall, stent migra- rate of metallic stents is related to the wider
tion) was recorded in 6 DLS patients lumen and better drainage of segmental bile
(10 %) and 5 polyethylene patients (8 %) duct branches (Figure 35.3). These data were
(n.s.). Kaplan-Meier analysis of DLS and confirmed by a single-center prospective
PE stent clogging-free survival showed a randomized controlled trial by Soderlund
significantly longer patency period with and Linder (2006) comparing the patency of
the DLS stents (p = 0.0005). Tringali et al. 10 F polyethylene stents and covered 30 F
a b c

Figure 35.2. Endoscopic view of a pigtail plastic endoprosthesis (a), a straight plastic stent (b) and a
metal stent (c), which jut out from the papilla Vateri into the duodenum (black arrows)

Figure 35.3. Different models of uncovered [Wallstent™, Ultraflex™, EndoCoil™ Endoflex™, and
Z-Stent™ (from left to right)] and covered [ePTFE-FEP (expanded polytretrafluoroethylene-fluorinated
ethylene propylene) Silicone, Polycaprolactone, Polyurethane (below)] metal stents
468 M. Wiedmann et al.

steel Wallstents™ in patients with nonresect- days in the covered group and after 319
able malignant common bile duct strictures. days in the uncovered Wallstent group (p >
As a result, median patency times were 1.8 0.05). Stent patency rates were 83, 78, 67,
and 3.6 months in the polyethylene stents and 54 % at 100, 200, 300, and 400 days,
and Wallstents™ groups, respectively (p = respectively, in the covered group and 83,
0.002). Interestingly, when overall costs of 66, 54, and 36 % in the uncovered group,
therapy, determined by adding the cost of which was not significantly different.
hospitalization to the costs of the stent, were However, it must be mentioned that this
compared, there was no difference between study was not a prospective randomized
both groups. The authors recommended study and the sample size was not large.
the more-effective SEMS in unresectable A prospective randomized study by
patients with malignant common bile duct De Palma et al. (2001) revealed a lower
strictures, who survive a median of 4.5 cholangitis rate after unilateral stent
months and less costly plastic stents for insertion in comparison to bilateral stent
patients who have distant metastases. insertion. Howewer, thus far general con-
Despite the higher patency rate of SEMS, sensus regarding unilateral versus bilateral
stent dysfuncion can be induced by tumor bile duct drainage has not been obtained
ingrowth, overgrowth or stent disloca- (Table 35.1). In general, at least 25 % of
tion. Stent dysfunction can be solved by liver tissue should be drained and con-
implantation of additional plastic or metal trast medium should be injected only
stents, laser treatment or balloon dilatation. in those bile duct segments that will be
Whether stent occlusion may be prevented finally drained. There is no need to drain
by covered SEMS or brachytherapy, is a certain liver lobe. It is better to drain the
currently unknown. A prospective ran- easiest accessable one. In case of concur-
domized study by Isayama et al. (2004) rent single-sided liver atrophy as a result
evaluating patients with distal malignant of tumor induced blood vessel occlusion,
bilary duct obstruction showed that a the primarily intact liver lobe has to be
polyurethane covered Diamond™-stent is drained. A recent study by Freeman and
superior to a non-covered metallic stent in Overby (2003) showed that MRCP-guided
terms of patency rate. However, cholecys- drainage of the widest dilated bile ducts
titis and pancreatitis rates were higher in can improve results of stent therapy.
the covered metallic stent group. Overall Despite controversial data with respect
survival was equivalent in both groups. to the benefit of unilateral or bilateral
In contrast, a recent study by Yoon biliary drainage, in studies by Liu et al.
et al. (2006) comparing polyurethane- (1998), Ducreux et al. (1992), Polydorou
covered Wallstents™ (that were used in et al. (1989), and Polydorou et al. (1991)
36 patients) with uncovered Wallstents™ successful endoscopic drainage rate with
(that were used in 41 patients) found no plastic stents was 41 % (20 % reduc-
significant difference between stent paten- tion of bilirubin within the first week),
cies. Cholecystitis occurred in 1 patient of 53 % (30 % reduction of bilirubin within
the covered Wallstent™ group but in none the first week), 74 % (30 % reduction of
of the uncovered Wallstent™ group. Stent bilirubin within the first month), and 80 %
occlusion occurred after a mean of 398 (30 % reduction of bilirubin within the
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 469

Table 35.1. Uni-versus bilateral biliary drainage.


Success Cholangitis 30 day Median
Number of patients rate rate mortality survival Reference
48 (retrospective: unilateral 96% 38% vs. 17% 29% vs. 8% 119 vs. 176 days Deviere et al.
vs. bilateral; attn: bilateral (p < 0.01) (p < 0.01) (p < 0.01) (1988)
contrast injection)
141 (retrospective: group A ? 6% vs. 0% ? Bismuth types II and Chang et al.
(one lobe opacified with vs. 32% III: 145 vs. 225 vs. (1998)
same lobe drained), 46 days (p < 0.001
B (both lobes opacified for A vs. C and
with both lobes drained), B vs. C; p < 0.001
and C (both lobes for C vs. A plus
opacified with one lobe B and for B vs.
drained)) A plus C)
157 (prospective: unilateral 89% vs. 77% 9% vs. 17% 11% vs. 14% 140 vs. 142 days De Palma
vs. bilateral) (p = 0.041) (p = 0.013) (p = 0.638) et al. (2001)

first month), respectively. In contrast, the


ally represents a terminal event, but also
randomized study by Ortner et al. (2003), grossly impairs patients’ quality of life.
which contained 79 % Bismuth type IV We found an incidence of 25 % in our
tumors, showed a decrease of cholestasis own series of 80 biliary tumors. Relief
by at least 50 % within the first week in of obstruction to allow enteral feeding,
only 21 % with stenting alone. In our own thereby improving weight and possibly
study, Witzigmann et al. (2006), hyper- increasing life expectancy, is the primary
bilirubinemia was decreased by at least aim in these patients. Surgical bypass is the
50 % in 39 % of patients three months standard treatment with a reported success
after stenting despite a high percentage rate up to 90 %. The procedure carries a
of Bismuth-Corlette type IV tumors. Very high complication rate of 25–35 % and a
important for long-term success of stent perioperative mortality of 2 % determined
therapy is a close monitoring of patients in studies such as that by Lillemoe et al.
for early detection of latent biliary sepsis.
(1999). A procedure that prolongs hospital
As part of this monitoring plastic endo- stay and increases costs dramatically may
prostheses should be routinely exchanged not always be appropriate, especially in
every 3 months (Prat et al., 1998). terminally ill patients with a poor clinical
condition. Previous endoscopic interven-
tions like balloon dilatation or laser reca-
PALLIATIVE STENTING OF nalization provide only transient success,
MALIGNANT DUODENAL resulting in high reocclusion rates.
OBSTRUCTION Based on the experience in palliation of
obstructive esophageal and biliary tract can-
Malignant gastric outlet obstruction or cer, SEMS also have been used for malig-
duodenal obstruction is a complication of nant gastric outlet and duodenal obstructions
advanced hepatobiliary carcinomas. It usu- since the early 1990s. In contrast to surgical
470 M. Wiedmann et al.

bypass surgery, endoscopic placement of mostly tumor tissue. The mechanisms by


duodenal SEMS is a less invasive procedurewhich this localization occurs are complex
possibly resulting in shorter hospitalization
and not fully understood. High vascular
time and lower costs. However, some stud- permeability of the agents, their affin-
ies reported complications that may increase
ity for proliferating endothelium, and the
the length of hospital stay, especially tumor
lack of lymphatic drainage in tumors may
ingrowth, tumor overgrowth, and stent dis-contribute to an accumulation in tumors.
location. In our own prospective study, com-
Moreover, tumors might have increased
prising 20 patients with biliary-tract-cancer
lipid content, elevated numbers of low-
(7 × gallbladder, 13 × Klatskin tumors) density lipoprotein receptors, abnormal
successful stent placement was achieved invasculature, and decreased pH. In a second
all patients (Schiefke et al., 2003). An addi-
step, non-thermal laser light of a specific
tional stent was required in six cases (four
wavelength is applied, and adapted to
occlusions, two dislocations). Median sur-the absorption spectrum of the sensitizer
vival was 20.5 weeks, no treatment relatedto be excited. Following the absorption
deaths occurred. Twenty-eight biliary stent
of light, the compound is transformed
exchanges were performed in 13/20 (65 %) into a relatively long-lived electronically
patients. Erosive reflux esophagitis improved
excited state via a short-lived excited sin-
in 11/12 (92 %) cases. After 4 weeks, all 17
glet state.
surviving patients tolerated soft-solid or This so-called triplet state can undergo
solid food, while 13/17 (77 %) tolerated atwo kinds of reaction. In the type I reac-
more solid diet (p < 0.001, gastric-outlet-
tion, it can react directly with a substrate,
obstruction-scoring-system). 12/17 (71 %) such as the cell membrane or a molecule,
patients gained a median of 1.5 kg weight and transfer a hydrogen atom or electron
(p = 0.001). The median Karnofsky-Index to form radicals. The radicals interact with
increased from 50 % to 60 % in 13/17 (77 %)
oxygen to produce oxygenated products.
patients. In conclusion, SEMS is a safe, effi-
Alternatively, in the type II reaction (which
cacious and low invasive treatment option is more common), the triplet can transfer its
for palliation of duodenal obstruction in energy directly to oxygen to generate singlet
biliary tract cancer. Technical complications
oxygen (Figure 35.4). Therefore, the effects
can be managed endoscopically and the bileof both PDT reaction types are oxygen-
duct remains accessible for endoluminal dependent. Tumor destruction, mediated
treatment. by oxygenated products and singlet oxy-
gen, can be explained by several mecha-
nisms causing necrosis and apoptosis. First,
PDT kills tumor cells directly; secondly, it
PHOTODYNAMIC THERAPY damages the tumor-associated vasculature,
(PDT) leading to tumor infarction, and finally can
activate an immune response against tumor
Mechanism of Action
cells. In detail, PDT has been demon-
Photodynamic therapy is based on the strated to induce (1) direct cytotoxicity by
administration of a photosensitizer which degeneration of lipids in cell membranes,
localizes selectively within the target, (2) microvascular damage from platelet
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 471

Figure 35.4. Mechanism of action of photodynamic therapy (PDT)

activation and thromboxane-induced vessel sure dose and light fluence rate, the oxygen
constriction and thrombus formation, prob- availability in the target, and finally the
ably mediated by nitric oxide, (3) cytosolic time interval between the administration of
and mitochondrial membrane damage with the photosensitizer and the light treatment.
cytochrome C release, bcl-2 depletion and Because PDT is a cold photochemical proc-
induction of apoptosis, and (4) release ess, there is no tissue heating, and connec-
of inflammatory mediators that induce tive tissues such as collagen and elastin are
T-lymphocyte mediated cellular immune largely unaffected.
response. The extent of photodamage and
cytotoxicity is multifactorial and depends Contraindications for Photodynamic
on the type of photosensitizer used, its Therapy
localization and administered dose, the Photodynamic therapy should not be per-
light source used, generating the light expo- formed in patients with acute porphyria,
472 M. Wiedmann et al.

poor kidney or liver function (creatinine > ative of pyropheophorbide (HPPH), benzo-
3 mg/dl, international normalized ratio of porphyrin derivatives (BPD), ring A-reduced
prothrombin time [INR] > 2.2), encasement monoacid benzoporphyrin derivative
or thrombosis of the main blood vessels, (BPDMA, Vertiporfin, Visudyne™), Sn eti-
leucopenia (leucocytes < 2000/cmm), throm- opurpurin (SnEt2, Rostaporphin, Purlytin™),
bocytopenia (< 50,000/cmm), and terminal meso-tetra (hydroxyphenyl) chlorins
tumor stage. [H2(THPC)] (Temoporfin, Foscan™), bac-
teriochlorin, and 5-aminolevulinic acid
Photosensitizers (ALA, Levulan™) are their representatives.
The second group consists of photosensi-
The ideal photosensitizer should be chemi- tizers based on isomeric porphyrins, like
cally pure and of known specific compo- porphycene, corrphycene, isoporphycene,
sition, should have a high quantum yield hemiporphycene, N-confused porphyrin,
for singlet oxygen production, a strong doubly N-confused porphyrin, and por-
absorption with high extinction coefficient phycene ATMPn (9-acetoxy-2,7,12,17-
ε at longer wavelength (red) region prefer- tetrakis-(β-methoxy-ethyl)-porphycene).
ably between 700–800 nm, and an excellent The third group consists of phthalocyanine
photochemical reactivity. It should also and naphthalocyanine based photosensitiz-
possess minimal dark toxicity and only be ers, like zinc phthalocyanine CGP 55847,
toxic in the presence of light, have prefer- sulphonated aluminium phthalocyanine
able retention by target tissue (tumor cells),(Photosense) and naphthalocyanines. The
be rapidly excreted from the body, be syn- fourth group consists of cationic photo-
thesizable from easy available precursors sensitizers, like Pt (II) complex of bis
and stable and easy to dissolve in the body’s (N,N-dimethylaminomethyl) deutero-
tissue fluids and be capable of formulation. porphyrin IX dimethyl ester, copper imi-
Starting from the development of the first nium salt of octaethylbenzochlorin, and
generation porphyrin based photosensitiz- unsymmetrically substituted benzonaph-
ers, hematoporphyrin derivative (HPD) and thopyrazines. Other potential candidates
the purified hematoporphyrin derivatives are expanded porphyrin based photosensi-
Photofrin™, Photosan™, and Photoheme™, tizers. Their representatives are sapphyrin,
a second generation of photosentizers,
vinylogous porphyrins, texaphyrins (e.g.,
have been developed and are now under
gadolinium (III) texaphyrin (XCYTRIN™)
trial. The first group consists of porphy- or Lutetium (III) texaphyrin (LUTRIN™) ),
rin, chlorin and bacteriochlorin derivatives. and core-modified expanded porphyrins
Boronated protoporphyrin, isohematopor- (e.g., ammonium salt of 5,10,15,20-tetrakis
phyrin, tetrasulfonated meso-tetraphenyl (meso-p-sulfonato phenyl)-25,27, 29-trithia
porphyrin (H2TPPS4), 5,10,15,20-tetrakis sapphyrin) (Table 35.2).
(4-sulphonato-phenyl)-21,23-dichalcogena-
porphyrin, o-,m- and p-isomers of tetra
(hydroxyphenyl)-porphyrin, picket fence Photodynamic Therapy for Palliation
porphyrins, monoaspartyl chlorin e6 (MACE, of Hilar Cholangiocarcinoma
Npe6), diaspartyl chlorin e6 (DACE), Interest in using PDT for the palliative
chlorin p6 lysyl derivative, hexylether deriv- treatment of advanced non-resectable CC
Table 35.2. Photosensitizers, potential indications and activation wavelengths.
Activation
wavelength
Sensitizer Trade name Potential indications (nm)
First Generation Photosensitizers
Purified hematoporphyrin Photofrin, Photosan, Head-, neck-, tracheobronchial, gynecological, esophageal, bladder, brain, gastric, 630
derivatives (HPD) and Photoheme bile duct cancer, skin basalioma, Barrett’s esophagus, psoriasis, Papilloma virus
Second Generation Photosensitizers
A. Porphyrin, Chlorin and Bacteriochlorin Derivatives
Boronated protoporphyrin BOPP Brain tumors 630
Monoaspartyl chlorin e6, (Npe6) MACE Skin cancer 654
Ring A-reduced monoacid benzoporphyrin Visudyne Non-melanoma skin cancer, age-related macular degeneration, rheumatoid arthritis, 689
derivative (BPDMA, Vertiporfin) bone marrow purging, Barett’s esophagus, psoriasis
Sn etiopurpurin (SnEt2, Rostaporfin) Purlytin Metastatic breast cancer, Kaposi’s sarcoma, prostatic cancer, brain, lung, 663
skin, head and neck cancer, age-related macular degeneration
Meso-tetra (hydroxyphenyl) chlorins Foscan Head and neck, upper aero-digestive tract cancers, gastric and prostate 652
[H2(THPC)] (Temoporfin) cancer
5-aminolevulinic acid (ALA) Levulan Actinic keratosis, basal cell carcinoma, squamous cell carcinoma, head and neck cancer, 635
gynecological tumors, Barrett’s esophagus, gastrointestinal tumors
Photodetection of bladder cancer, esophageal cancer, gastrointestinal cancer 410
5-ALA-methylesther Metvix Basal cell carcinoma, actinic keratosis 635
5-ALA-benzylesther Benzvix Gastrointestinal cancer 635
Hexylether derivative of Photochlor Basal cell carcinoma 665
pyropheophorbide (HPPH)
B. Phthalocyanine and Naphthalocyanine Based Photosensitizers
Zinc phthalocyanine CGP 55847 Squamous cell carcinoma 670
Sulphonated aluminium Photosense Skin, breast, lung, bladder, pancreatic, brain, gastrointestinal cancers 670
phthalocyanine
C. Expanded Porphyrin Based Photosensitizers
Gadolinium (III) texaphyrin XCYTRIN Brain metastases 700–780
Lutetium (III) texaphyrin LUTRIN, ANTRIN, Breast, cervical, prostate and brain tumors, angioplasty, age-related 732
OPTRIN macular degeneration
474 M. Wiedmann et al.

was given rise to by a case report, docu- the two monthly follow-ups. Quality of
menting the success of PDT performed via life indices improved dramatically and
percutaneous cholangioscopy in a single remained stable. Thirty-day mortality was
patient with incompletely resected bile 0 %, and median survival 439 days. Side
duct carcinoma (McCaughan et al., 1991). effects were limited to skin hyperpigmen-
This type of treatment caused a prolonged tation, but no acute phototoxicity in all
survival time of > 4 years in a tumor patients. One patient also reported fever
entity with a median survival of only 4–6 and pain, self-limited, for 2 weeks after
months. Ortner et al. (1998) performed the PDT (Table 35.1). Ortner’s work (Ortner,
first prospective non-randomized single- 2000) was subsequently expanded to a
arm study including nine patients with larger study of 21 patients (Table 35.3).
advanced Bismuth type III and IV hilar We performed our own study (Berr
CC, who showed no sufficient drainage et al., 2000b), including 23 patients with
after endoscopic stent insertion. Two days hilar CC (Bismuth type III, n = 2; type IV;
after intravenous application of porfimer n = 21) who were treated with a combina-
sodium (Photofrin™) at 2 mg/kg body tion of bile duct stenting and endocopi-
weight, intraluminal photoactivation at cally performed Photofrin™-PDT (Figure
630 nm was performed cholangioscopi- 35.5). In detail, we intubated the tumor
cally with laser diffusing fibers. After PDT, stenosis with a translucent ERC-cannula
bilirubin serum levels declined signifi- and inserted a 400 μm thin quartz fiber
cantly (p = 0.004) with no increase during with either a 2, 3, or 4 cm long cylindrical

Table 35.3. Palliative Photodynamic Therapy (PDT) in Hilar Cholangiocarcinoma.


Protocol n Photosensitizer Drainage Survival# Reference
Phase I 1 Dihemato- T-Drain 48 McCaughan et al. (1991)
porphyrin 2
Phase II 9 Photofrin II plastic stent 14.6 (3–19.2) Ortner et al. (1998)
Phase II 21 Photofrin II plastic stent 15 Ortner (2000)
Phase II 23 Photofrin II plastic stent 12.0 (10.4–13.6) Berr et al. (2000);
Wiedmann et al. (2004)
Phase II 8 Photosan-3 plastic stent 4.0 (1.7–14.8) Zoepf et al. (2001a)
Phase II 4 5-ALA and PTCD NA Zoepf et al. (2001b)
Phase II 6 Photofrin II plastic stent NA Rumalla et al. (2001)
Phase II 24 Photofrin II metallic stent 9.9 (6.4–13.4)+ Dumoulin et al. (2003)
Phase III 20 Photofrin II plastic stent 16.4** Ortner et al. (2003)
19 plastic stent 3.3
Phase II 8 Photofrin II plastic stent 9.2+ Harewood et al. (2005)
Phase II 36 Photofrin II plastic stent 11.2 (2.5–17.3) Pereira et al. (2005b)
Phase II 24 Hematoporphy- PTCD 18.6 (2.1–27) Shim et al. (2005)
rine derivative
Phase II 25 Photofrin II plastic stent 8.1 Prasad et al. (2005)
Phase III 16 Photosan-3 plastic stent 21* (3–30) Zoepf et al. (2005)
16 plastic stent 7 (1–24)
Phase II 68 Photofrin II plastic stent 12** (8.8–15.3) Witzigmann et al. (2006)
56 plastic stent 6.4 (4.2–8.5)
NA = non-applicable; #median (range) in months from diagnosis; *p < 0.05; **p < 0.01; +median (range) in months from therapy
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 475

grating sphere power meter. The tumor


stenoses were exposed to laser light for
10 min with a light dose of 242 J/cm2.
The patients showed a 6 month survival
rate of 91 % after diagnosis and 74 %
after start of PDT with a median local
tumor response of 74 %, 54 %, 29 %, and
67 % after the first, second, third, and
fourth PDT session, respectively (Table
35.3). Cholestasis, performance, and qual-
ity of life of the patients improved clearly.
Cholangitis rate with PDT was not higher
than in historical control patients with
Bismuth type III tumors and bile duct
drainage, only. After a 5 year follow-up,
median survival time after diagnosis was
18 months for patients without perito-
neal carcinosis (n = 19) and 12 months
for all patients (n = 23) (Wiedmann et al.,
2004a). Survival was 63 %, 26 %, 16 %,
and 5 % after one, two, three, and four
years, respectively. Of the patients who
died, 73.9 % (n = 17) died because of
tumor progression; 26.1 % died as a result
of cholangitis (n = 4), septic shock (n = 1),
or appendicitis/peritonitis (n = 1). For all
patients, except one with diffuse liver
metastases, there was improvement in
cholestasis, performance, and quality of
life, which was maintained for an extended
Figure 35.5. Hilar cholangiocarcinoma Bismuth period of time. These data were confirmed
type IV, (a) tumor imaging with endoscopic retro- by a similar recent phase II study by Pereira
grade cholangiography (ERC; white arrows), (b) et al. (2005b) including 36 patients.
reopened bile ducts 3 months after photodynamic Zoepf et al. (2001a) described the use
therapy (PDT) and stent insertion (white arrows)
of a novel laser diode PDT system in
the treatment of 8 patients with Bismuth
II-IV non-resectable CC using Photosan-3
light diffuser and radioopaque markers (2 mg/kg; dihematoporphin ether) and a
(Lambda Plaus PDL-2, Coherent, Dieburg, light dose of 200 J/cm2 at a wavelength of
Germany). The power emmited by the 633 nm (Table 35.3). Four weeks after ini-
diffuser tip through the ERC cannula tial PDT, which was performed transpapil-
had been calibrated to 0.400 W/cm before lary (n = 4) or percutaneously (n = 4), all
PDT and checked thereafter using an inte- patients showed a marked reduction of bile
476 M. Wiedmann et al.

duct stenosis. The median serum bilirubin sodium porfimer, and 48 h later a com-
value declined from 5.8 to 1.0 mg/dl. The mercially available cylindrical diffusing
median survival time was 119 days (52– laser fiber was inserted into an 8 F biliary
443 days). catheter equipped with a 0.038 in. side-
The therapeutic value of percutane- hole at its distal tip. After positioning
ous transhepatic PDT in patients with the 0.035 inch guidewire proximal to
advanced bile duct cancer was the subject the biliary stricture, the preloaded cath-
of a prospective trial by Shim et al. (2005). eter was advanced over the guidewire
The utility of intraductal ultrasonography by using the monorail technique. Laser
(IDUS) for the assessment of responses light was applied at a power of 400 mW/
and for regular follow-up after PDT was cm fiber for a total energy of 180 J/cm2.
also examined. PTCD was initiated before By using the preloaded biliary catheter,
PDT. Following dilation and maturation adequate positioning of the laser fiber
of the PTCD tract, percutaneous PDT was was achieved in all patients. A fracture of
performed. Intraluminal photoactivation the diffuser tip occurred during one of the
was carried out using percutaneous cholan- treatments. Two patients developed acute
gioscopy 2 days after intravenous applica- cholangitis and two patients experienced
tion of a hematoporphyrin derivative. All skin phototoxicity. The applicability of
patients were provided additionally with this technique was confirmed in recent
percutaneous bile duct drainage catheters studies by Harewood et al. (2005) and
after PDT. IDUS was conducted monthly Prasad et al. (2005) (Table 35.3). It was
to measure the thickness of the tumor concluded from the first study that (1)
mass before and after PDT. Twenty-four PDT treatment every 3 months is applica-
patients with advanced cholangiocarcino- ble, (2) patient selection for PDT need not
mas (Bismuth IIIa, n = 4; IIIb, n = 10; IV, to be based on response to primary biliary
n = 10) were treated with PDT. At 3 months stenting, and (3) the average cost of treat-
after PDT, the mean thickness of the tumor ment per patient would be US$10,337,
mass had decreased from 8.7 ± 3.7 to 5.8 ± which is based on direct medical costs
2.0 mm (p < 0.01). At 4 months after PDT, estimated from USA Medicare ambula-
the thickness of the mass had increased tory patient classification (ABC, 2003)
to 7.0 ± 3.7 mm. Quality of life indices plus professional fees for hospital-based
improved dramatically and remained sta- outpatient procedures, which can be com-
ble 1 month after PDT; the Karnofsky pared with the costs of management with
index increased from 39 to 58 points (p = biliary stenting alone. The conclusion
0.003). The 30-day mortality rate was 0 %, from the second study was that early
and the median survival time was 558 days treatment with PDT may lead to greater
(range 62–810 days). preservation of liver function based on
Another single-arm study by Rumalla a univariate and multivariate analysis,
et al. (2001) described a new method of showing a longer interval from diagnosis
applying photodynamic therapy in the to PDT treatment, which was significantly
biliary tract by using accessories avail- associated with shorter survival following
able in the United States (Table 35.3). PDT (HR 1.23, 1.02–1.47; p = 0.029 and
Patients were injected with 2 mg/kg of HR 2.38, 1.38–4.85; p = 0.0001).
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 477

Another prospective phase II study by fulfilling inclusion criteria (non-resectable


Dumoulin et al. (2003) investigated PDT tumor, tumor diameter > 3 cm, tumor
and consecutive metal stent insertion for clearly visible on CT and ERCP, unequiv-
palliation of hilar CC. It was feasible but ocal positive histology, no evidence of
there was only a modest benefit in overall cancer of another organ) were randomized
survival in comparison to a historical con- to group A (stenting and subsequent PDT)
trol group (Table 35.3). In addition, a small and group B (stenting alone). For PDT,
study by Zoepf et al. (2001b) investigated Photofrin™ at 2 mg/kg body weight was
the use of 5-aminolevulinic acid (5-ALA) injected intravenously 2 days before intra-
for the palliative treatment of hilar CC. luminal photoactivation (wavelength,
Light activation was performed 5 to 7 630 nm; light dose, 180 J/cm2). Further
hours after oral administration of 5-ALA. treatments were performed in cases of
All patients had an endoprosthesis placed residual tumor in the bile duct. Oral cipro-
in the bile duct after PDT. However, 4 floxacin therapy with 250 mg twice daily
weeks after PDT, 5-ALA had failed to was started before ERCP and continued
significantly reduce malignant bile duct for 14 days. The primary outcome param-
obstruction, and thus cannot be recom- eter was survival time. Secondary outcome
mended for the palliative treatment of bile parameters were cholestasis and quality
duct cancer (Table 35.3). Finally, a pro- of life. PDT resulted in prolongation of
spective analysis of resectional and pallia- survival (group A: n = 20, median 493
tive treatment of 184 patients with hilar CC days; group B: n = 19, median 98 days;
was reported from our hospital (60 patients p < 0.0001). It also improved biliary
underwent resection, 8 after neoadjuvant drainage and quality of life. The study
PDT, 68 had PDT in addition to stenting was terminated prematurely because PDT
and 56 had stenting alone) (Witzigmann proved to be so superior to simple stent-
et al., 2006). During a 10-year period it was ing treatment that further randomization
revealed that PDT and stenting resulted in was deemed unnecessary. However, the
longer median survival (12 vs., 6.4 months, study was criticized for the short duration
p < 0.01), lower serum bilirubin levels of survival in the control group, mainly
(p < 0.05), and higher Karnofsky perform- caused by insufficient biliary drainage
ance status (p < 0.01) as compared to by stenting alone. Nevertheless, it was
stenting alone. Median survival after PDT remarkable that an additional group of 31
and stenting, but not after stenting alone, patients who were excluded from rand-
did not differ from that after both R1 and omization because of a statistically signifi-
R2 resection. However, these improve- cant lower Karnovsky performance status,
ments in palliative treatment by PDT will but received PDT treatment voluntarily,
not change the concept of an aggressive performed as well as the randomized PDT
resectional approach. group (Table 35.3). In summary, pallia-
The first prospective, randomized multi- tive PDT for non-resectable hilar CC was
center trial by Ortner et al. (2003), includ- effective in restoring biliary drainage and
ing our hospital, confirmed preliminary improving quality of life, survival was
data from the above mentioned non-ran- prolonged; thus, PDT represents a promis-
domized studies. Patients with hilar CC ing adjunct to palliative therapy.
478 M. Wiedmann et al.

The second prospective randomized trial Similar results have also been reported
by Zoepf et al. (2005) investigated the by Nanashima et al. (2004) who treated
effect of Photosan-3 based PDT plus stent- 8 patients with porfimer sodium PDT in
ing on survival time in 32 patients with an adjuvant setting. Five patients had ext-
advanced hilar CC. In the control group, rahepatic bile duct cancer, two had intra-
patients were treated with endoprostheses hepatic cholangiocarcinoma, and one had
but no PDT. PDT group and the control ampullary carcinoma. Cancer cells were
group were comparable in age, gender, microscopically detected in the stump of
performance status, bilirubin level, and the hepatic duct in six patients, and biliary
Bismuth stage. The median survival time stenosis caused by remnant tumor was
after randomization was 7 months for the observed in one patient. One patient had
control group and 21 months for the PDT tumor recurrence with occlusion of the bile
group (p = 0.01). In half of the initially duct. At 48 h prior to PDT, porfimer sodium
percutaneously treated patients, a change was injected intravenously. A pulse laser by
from percutaneous to transpapillary drain- an eximer dye laser (50–100 J/cm2) with a
age after PDT was possible. Four patients wavelength of 630 nm was applied through
showed infectious complications after PDT an endoscope to the hepatic stump or tumor
versus one patient in the control group. lesion. Marked destruction of the tumor and
ductal epithelium was observed on day 1
Photodynamic Therapy for Neoadjuvant after PDT. After PDT, four patients devel-
and Adjuvant Treatment of Hilar oped mild dermatitis, but no severe morbid-
Cholangiocarcinoma ity or mortality was noted. In patients who
underwent PDT for the stump, one patient
A new approach is the use of PDT for the showed distant metastasis at 31 months, and
treatment of hilar CC in a neoadjuvant four patients did not show tumor recurrence
setting because of the high tumor recur- at 17, 12, 12, and 6 months, respectively.
rence rate of up to 76 % after curative (R0) However, one of the eight patients died at
resection. Having accomplished a success- 2 months due to an unrelated cause. In two
ful treatment in a single case (Berr et al., patients with occlusion caused by tumor
2000a), we investigated the use of PDT growth, resolution of bile duct stenosis
prior to tumor resection in six patients and was noted on day 7. These patients showed
in one patient prior to liver transplantation in
reocclusion by tumor at 20 and 8 months.
a small pilot study (Wiedmann et al., 2003). In summary, neoadjuvant and adjuvant
In all patients, R0 resection was achieved. PDT in these two small studies were safe
Four patients developed minor surgical and useful options for a better survival
complications, even though the bilioenteric benefit in patients with bile duct cancer
anastomoses were sewn to PDT-treated bile undergoing surgical resection; however,
ducts. No viable tumor cells were found in further studies are required.
the inner 4 mm layer of the surgical speci-
mens. The PDT-pretreated epithelium of
Future Directions of Photodynamic
the tumor-free proximal resection margins
Therapy for Hilar Cholangiocarcinoma
exhibited only minimal inflammatory infil-
tration. The 1-year recurrence free survival The penetration depth of Photofrin™ and
rate was 83 %. Photosan™ is 4–4.5 mm, and exceeds the
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 479

penetration depth of 2 mm of 5-aminole- The median survival after diagnosis and


vulinic acid (5-ALA, Levulan™), a prodrug PDT administration was 21 (10–90) and
of heme biosynthesis and a photosensitizer 9 (1–27) months, respectively. PDT was
of the second generation. In the future, generally well-tolerated but two patients
further progress may be made by using developed significant hemobilia, one of
photosensitizers with a higher penetration whom died with a gallbladder empyema 2
depth, such as Temoporfin (Foscan™) with weeks after treatment.
a strong absorption at 652 nm or tetrahy- Further modifications of Foscan™, like
droporphyrin tetratosylat (THPTS) with a a liposomal formulation (Foslip™) or
strong absorption at 760 nm, in combina- pegylation (Fospeg™), aim to reduce pho-
tion with cheaper laser systems and better tosensitizer dose, side effects, and costs. In
light applicators. addition to the development of new pho-
A first preliminary study by Pereira et tosensitizers, other innovative ideas are
al. (2005a) tried to determine the safety currently under investigation. For instance,
and efficacy of Foscan™-PDT in the treat- fractionated drug-dose PDT regimens were
ment of patients with irresectable malignant reported to result in a superior therapeutic
biliary strictures and recurrent stent occlu- effect, compared to single-dose regimens,
sion. Thirteen patients with malignant and were able to induce long-term tumor
biliary obstruction due to carcinoma of growth control. Moreover, the use of spe-
the biliary tract (n = 5), ampulla (n = 5), cific targeting carriers, such as conjugated
pancreas (n = 2) or stomach (n = 1), were antibodies directed to tumor-associated
studied. All had been initially palliated with antigens or vascular antigens are supposed
metal (n = 10) or polyethylene (n = 3) to direct the photosensitizer to a certain cell
biliary stents, and presented with recurrent type or compartment. Others suggest the
obstructive jaundice. Patients received use of different advanced delivery systems,
0.15 mg/kg Foscan™ intravenously 72 h such as ligand-based targeting with insu-
before endoluminal light activation with lin, epidermal growth factor or adenoviral
an endoscopically-placed optical fiber proteins, protease-mediated drug delivery,
(4–5 cm diffusing tip, 652 nm light at photosensitizing adenoviruses, water-sol-
10–50 J, 100 mW/cm diffuser fiber), fol- uble polymer carriers, and pH-responsive
lowed by polyethylene stent insertion in polymeric micelles. A completely different
all but one patient. Before PDT, patients approach is the protection of normal tissue
had a median of 3 (0–5) stent occlusions with drugs like WR-2721 and WR-77913
over 13 (2–27) months, with a median from possible PDT side effects if the pho-
patency of plastic stents placed inside tosensitizer is used in a high dose.
metal stents for recurrent stent occlusion
of 3.5 (0.5–13) months. After PDT, tumor REFERENCES
necrosis and/or metal stent recanalization
was seen in all patients, with a median of Berr, F., Tannapfel, A., Lamesch, P., Pahernik,
S., Wiedmann, M., Halm, U., Goetz, A. E.,
0 (0–3) stent occlusions during 17 (1–78) Mossner, J., and Hauss, J., 2000a. Neoadjuvant
months follow-up. The median patency photodynamic therapy before curative resection
of plastic stents inside metal bile duct of proximal bile duct carcinoma. J. Hepatol. 32:
stents after PDT was 5 (1–52) months. 352–357.
480 M. Wiedmann et al.

Berr, F., Wiedmann, M., Tannapfel, A., Halm, U., 2003. Phase II study of photodynamic therapy
Kohlhaw, K. R., Schmidt, F., Wittekind, C., and metal stent as palliative treatment for nonre-
Hauss, J., and Mossner, J., 2000b. Photodynamic sectable hilar cholangiocarcinoma. Gastrointest.
therapy for advanced bile duct cancer: evidence Endosc. 57: 860–867.
for improved palliation and extended survival. Ebata, T., Nagino, M., Kamiya, J., Uesaka, K.,
Hepatology 31: 291–2988. Nagasaka, T., and Nimura, Y., 2003. Hepatectomy
Burmester, E., Niehaus, J., Leineweber, T., and with portal vein resection for hilar cholangiocar-
Huetteroth, T., 2003. EUS-cholangio-drainage cinoma: audit of 52 consecutive cases. Ann.
of the bile duct: report of 4 cases. Gastrointest. Surg. 238: 720–727.
Endosc. 57: 246–251. Freeman, M. L. and Overby, C., 2003. Selective
Chang, W. H., Kortan, P., and Haber, G. B., MRCP and CT-targeted drainage of malignant
1998. Outcome in patients with bifurcation hilar biliary obstruction with self-expanding
tumors who undergo unilateral versus bilateral metallic stents. Gastrointest. Endosc. 58:
hepatic duct drainage. Gastrointest. Endosc. 47: 41–49.
354–362. Harewood, G. C., Baron, T. H., Rumalla, A., Wang,
Cherqui, D., Benoist, S., Malassagne, B., Humeres, K. K., Gores, G. J., Stadheim, L. M., and de
R., Rodriguez, V., and Fagniez, P. L., 2000. Groen, P. C., 2005. Pilot study to assess patient
Major liver resection for carcinoma in jaundiced outcomes following endoscopic application of
patients without preoperative biliary drainage. photodynamic therapy for advanced cholan-
Arch. Surg. 135: 302–308. giocarcinoma. J. Gastroenterol. Hepatol. 20:
de Groen, P. C., Gores, G. J., LaRusso, N. F., 415–420.
Gunderson, L. L., and Nagorney, D. M., 1999. Hemming, A. W., Reed, A. I., Fujita, S., Foley, D.
Biliary tract cancers. N. Engl. J. Med. 341: P., and Howard, R. J., 2005. Surgical manage-
1368–1378. ment of hilar cholangiocarcinoma. Ann. Surg.
De Palma, G. D., Galloro, G., Siciliano, S., Iovino, 241: 693–699; discussion 699–702.
P., and Catanzano, C., 2001. Unilateral versus Isayama, H., Komatsu, Y., Tsujino, T., Sasahira, N.,
bilateral endoscopic hepatic duct drainage in Hirano, K., Toda, N., Nakai, Y., Yamamoto, N.,
patients with malignant hilar biliary obstruction: Tada, M., Yoshida, H., Shiratori, Y., Kawabe, T.,
results of a prospective, randomized, and control- and Omata, M., 2004. A prospective randomised
led study. Gastrointest. Endosc. 53: 547–553. study of “covered” versus “uncovered” diamond
Deviere, J., Baize, M., de Toeuf, J., and Cremer, stents for the management of distal malignant
M., 1988. Long-term follow-up of patients with biliary obstruction. Gut 53: 729–734.
hilar malignant stricture treated by endoscopic Kadakia, S. C. and Starnes, E., 1992. Comparison
internal biliary drainage. Gastrointest. Endosc. of 10 French gauge stent with 11.5 French gauge
34: 95–101. stent in patients with biliary tract diseases.
Doctor, N., Dick, R., Rai, R., Dafnios, N., Salamat, Gastrointest. Endosc. 38: 454–459.
A., Whiteway, H., Dooley, J., and Davidson, B. Kawasaki, S., Imamura, H., Kobayashi, A., Noike,
R., 1999. Results of percutaneous plastic stents T., Miwa, S., and Miyagawa, S., 2003. Results of
for malignant distal biliary obstruction following surgical resection for patients with hilar bile duct
failed endoscopic stent insertion and comparison cancer: application of extended hepatectomy
with current literature on expandable metal- after biliary drainage and hemihepatic portal
lic stents. Eur. J. Gastroenterol. Hepatol. 11: vein embolization. Ann. Surg. 238: 84–92.
775–780. Khan, S. A., Taylor-Robinson, S. D., Toledano,
Ducreux, M., Liguory, C., Lefebvre, J. F., Ink, O., M. B., Beck, A., Elliott, P., and Thomas, H. C.,
Choury, A., Fritsch, J., Bonnel, D., Derhy, S., 2002. Changing international trends in mortality
and Etienne, J. P., 1992. Management of malig- rates for liver, biliary and pancreatic tumours. J.
nant hilar biliary obstruction by endoscopy. Hepatol. 37: 806–183.
Results and prognostic factors. Dig. Dis. Sci. 37: Kondo, S., Hirano, S., Ambo, Y., Tanaka, E.,
778–783. Okushiba, S., Morikawa, T., and Katoh, H., 2004.
Dumoulin, F. L., Gerhardt, T., Fuchs, S., Scheurlen, Forty consecutive resections of hilar cholangio-
C., Neubrand, M., Layer, G., and Sauerbruch, T., carcinoma with no postoperative mortality and no
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 481

positive ductal margins: results of a prospective domized prospective study. Gastroenterology


study. Ann. Surg. 240: 95–101. 125: 1355–1363.
Kosuge, T., Yamamoto, J., Shimada, K., Yamasaki, Pereira, S. P., Ayaru, L., Hatfield, A. R., Rogowska,
S., and Makuuchi, M., 1999. Improved surgical A., and Bown, S., 2005a. Photodynamic ther-
results for hilar cholangiocarcinoma with pro- apy (PDT) of malignant biliary strictures using
cedures including major hepatic resection. Ann. meso-tetrahydroxyphenylchlorin (mTHPC). J.
Surg. 230: 663–671. Clin. Oncol. 23: abstract 4176 (ASCO).
Lazaridis, K. N. and Gores, G. J., 2005. Pereira, S. P., Ragunath, K., Devlin, J., and Meadows,
Cholangiocarcinoma. Gastroenterology 128: H. M., 2005b. Preliminary results of a phase II
1655–1667. trial to examine the safety and efficacy of por-
Lillemoe, K. D., Cameron, J. L., Hardacre, J. M., fimer sodium photodynamic therapy (PDT) in
Sohn, T. A., Sauter, P. K., Coleman, J., Pitt, H. locally advanced biliary tract carcinoma (BTC). J.
A., and Yeo, C. J., 1999. Is prophylactic gastro- Clin. Oncol. 23: abstract 4180 (ASCO).
jejunostomy indicated for unresectable periam- Polydorou, A. A., Cairns, S. R., Dowsett, J. F.,
pullary cancer? A prospective randomized trial. Hatfield, A. R., Salmon, P. R., Cotton, P. B.,
Ann. Surg. 230: 322–328; discussion 328–330. and Russell, R. C., 1991. Palliation of proximal
Liu, C. L., Lo, C. M., Lai, E. C., and Fan, S. T., malignant biliary obstruction by endoscopic
1998. Endoscopic retrograde cholangiopancrea- endoprosthesis insertion. Gut 32: 685–689.
tography and endoscopic endoprosthesis inser- Polydorou, A. A., Chisholm, E. M., Romanos, A.
tion in patients with Klatskin tumors. Arch. Surg. A., Dowsett, J. F., Cotton, P. B., Hatfield, A. R.,
133: 293–296. and Russell, R. C., 1989. A comparison of right
McCaughan, J. S., Jr., Mertens, B. F., Cho, C., versus left hepatic duct endoprosthesis insertion
Barabash, R. D., and Payton, H. W., 1991. in malignant hilar biliary obstruction. Endoscopy
Photodynamic therapy to treat tumors of the 21: 266–271.
extrahepatic biliary ducts. A case report. Arch. Prasad, G., Wang, K. K., Baron, T. H., Buttar, N. S.,
Surg. 126: 111–113. WongKeeSong, M., Borkenhagen, L., Lutzke, L.,
Miyazaki, M., Ito, H., Nakagawa, K., Ambiru, S., Papanfuss, S., Gores, G., and Roberts, L., 2005.
Shimizu, H., Shimizu, Y., Kato, A., Nakamura, S., Factors predicting Survival in patients with cholan-
Omoto, H., Nakajima, N., Kimura, F., and Suwa, giocarcinoma treated with photodynamic therapy.
T., 1998. Aggressive surgical approaches to hilar Gastroenterology: abstract T1090 (DDW).
cholangiocarcinoma: hepatic or local resection? Prat, F., Chapat, O., Ducot, B., Ponchon, T., Pelletier,
Surgery 123: 131–136. G., Fritsch, J., Choury, A. D., and Buffet, C.,
Nanashima, A., Yamaguchi, H., Shibasaki, S., Ide, 1998. A randomized trial of endoscopic drain-
N., Sawai, T., Tsuji, T., Hidaka, S., Sumida, age methods for inoperable malignant strictures
Y., Nakagoe, T., and Nagayasu, T., 2004. of the common bile duct. Gastrointest. Endosc.
Adjuvant photodynamic therapy for bile duct 47: 1–7.
carcinoma after surgery: a preliminary study. J. Raju, G. S., Sud, R., Elfert, A. A., Enaba, M.,
Gastroenterol. 39: 1095–1101. Kalloo, A., and Pasricha, P. J., 2006. Biliary
Ortner, M. A., 2000. Photodynamic therapy of drainage by using stents without a central lumen:
cholangiocarcinoma cancer. Gastrointest. a pilot study. Gastrointest. Endosc. 63: 317–320.
Endosc. Clin. N. Am. 10: 481–486. Rumalla, A., Baron, T. H., Wang, K. K., Gores,
Ortner, M. A., Liebetruth, J., Schreiber, S., Hanft, G. J., Stadheim, L. M., and de Groen, P. C.,
M., Wruck, U., Fusco, V., Muller, J. M., Hortnagl, 2001. Endoscopic application of photodynamic
H., and Lochs, H., 1998. Photodynamic ther- therapy for cholangiocarcinoma. Gastrointest.
apy of nonresectable cholangiocarcinoma. Endosc. 53: 500–504.
Gastroenterology 114: 536–542. Schiefke, I., Zabel-Langhennig, A., Wiedmann,
Ortner, M. E. J., Caca, K., Berr, F., Liebetruth, M., Huster, D., Witzigmann, H., Mossner, J.,
J., Mansmann, U., Huster, D., Voderholzer, Berr, F., and Caca, K., 2003. Self-expandable
W., Schachschal, G., Mössner, J., and Lochs, metallic stents for malignant duodenal obstruc-
H., 2003. Successful photodynamic therapy tion caused by biliary tract cancer. Gastrointest.
for nonresectable cholangiocarcinoma: a ran- Endosc. 58: 213–219.
482 M. Wiedmann et al.

Seyama, Y., Kubota, K., Sano, K., Noie, T., Wagner, H. J., Knyrim, K., Vakil, N., and Klose,
Takayama, T., Kosuge, T., and Makuuchi, M., K. J., 1993. Plastic endoprostheses versus metal
2003. Long-term outcome of extended hemi- stents in the palliative treatment of malignant
hepatectomy for hilar bile duct cancer with no hilar biliary obstruction. A prospective and ran-
mortality and high survival rate. Ann. Surg. 238: domized trial. Endoscopy 25: 213–218.
73–83. Welzel, T. M., McGlynn, K. A., Hsing, A. W.,
Shim, C. S., Cheon, Y. K., Cha, S. W., Bhandari, O’Brien, T. R., and Pfeiffer, R. M., 2006. Impact
S., Moon, J. H., Cho, Y. D., Kim, Y. S., Lee, L. of classification of hilar cholangiocarcinomas
S., Lee, M. S., and Kim, B. S., 2005. Prospective (Klatskin tumors) on the incidence of intra- and
study of the effectiveness of percutaneous tran- extrahepatic cholangiocarcinoma in the United
shepatic photodynamic therapy for advanced States. J. Natl. Cancer Inst. 98: 873–875.
bile duct cancer and the role of intraductal ultra- West, J., Wood, H., Logan, R. F., Quinn, M., and
sonography in response assessment. Endoscopy Aithal, G. P., 2006. Trends in the incidence
37: 425–433. of primary liver and biliary tract cancers in
Soderlund, C. and Linder, S., 2006. Covered England and Wales 1971–2001. Br. J. Cancer.
metal versus plastic stents for malignant com- 94: 1751–1758.
mon bile duct stenosis: a prospective, rand- Wiedmann, M., Berr, F., Schiefke, I., Witzigmann,
omized, controlled trial. Gastrointest. Endosc. H., Kohlhaw, K., Mossner, J., and Caca, K.,
63: 986–995. 2004a. Photodynamic therapy in patients with
Speer, A. G., Cotton, P. B. and MacRae, K. D., non-resectable hilar cholangiocarcinoma: 5-year
1988. Endoscopic management of malignant follow-up of a prospective phase II study.
biliary obstruction: stents of 10 French gauge Gastrointest. Endosc. 60: 68–75.
are preferable to stents of 8 French gauge. Wiedmann, M., Caca, K., Berr, F., Schiefke, I.,
Gastrointest. Endosc. 34: 412–417. Tannapfel, A., Wittekind, C., Mossner, J., Hauss,
Speer, A. G., Cotton, P. B., Russell, R. C., Mason, J., and Witzigmann, H., 2003. Neoadjuvant pho-
R. R., Hatfield, A. R., Leung, J. W., MacRae, todynamic therapy as a new approach to treating
K. D., Houghton, J., and Lennon, C. A., 1987. hilar cholangiocarcinoma: a phase II pilot study.
Randomised trial of endoscopic versus percu- Cancer 97: 2783–2790.
taneous stent insertion in malignant obstructive Wiedmann, M., Dietrich, A., Mossner, J.,
jaundice. Lancet 2: 57–62. Witzigmann, H., and Caca, K., 2004b. Combined
Todoroki, T., Kawamoto, T., Koike, N., Takahashi, percutaneous transhepatic biliary drainage with
H., Yoshida, S., Kashiwagi, H., Takada, Y., port implantation for management of patients
Otsuka, M., and Fukao, K., 2000. Radical resec- with malignant biliary obstruction. Gastrointest.
tion of hilar bile duct carcinoma and predictors Endosc. 60: 117–120.
of survival. Br. J. Surg. 87: 306–313. Witzigmann, H., Berr, F., Ringel, U., Caca, K.,
Tringali, A., Mutignani, M., Perri, V., Zuccala, G., Uhlmann, D., Schoppmeyer, K., Tannapfel, A.,
Cipolletta, L., Bianco, M. A., Rotondano, G., Wittekind, C., Mossner, J., Hauss, J., and Wiedmann,
Philipper, M., Schumacher, B., Neuhaus, H., M., 2006. Surgical and Palliative Management
Schmit, A., Deviere, J., and Costamagna, G., and Outcome in 184 Patients With Hilar
2003. A prospective, randomized multicenter Cholangiocarcinoma: Palliative Photodynamic
trial comparing DoubleLayer and polyethylene Therapy Plus Stenting is Comparable to R1/R2
stents for malignant distal common bile duct Resection. Ann. Surg. 244: 230–239.
strictures. Endoscopy 35: 992–997. Yoon, W. J., Lee, J. K., Lee, K. H., Lee, W. J.,
van Berkel, A. M., Boland, C., Redekop, W. K., Ryu, J. K., Kim, Y. T., and Yoon, Y. B., 2006. A
Bergman, J. J., Groen, A. K., Tytgat, G. N., and comparison of covered and uncovered Wallstents
Huibregtse, K., 1998. A prospective randomized for the management of distal malignant biliary
trial of Teflon versus polyethylene stents for obstruction. Gastrointest. Endosc. 63: 996–1000.
distal malignant biliary obstruction. Endoscopy Zoepf, T., Jakobs, R., Arnold, J. C., Apel, D., and
30: 681–686. Riemann, J. F., 2005. Palliation of nonresectable
35. Hilar Cholangiocarcinoma: Photodynamic Therapy and Stenting 483

bile duct cancer: improved survival after pho- a new diode laser system. Am. J. Gastroenterol.
todynamic therapy. Am. J. Gastroenterol. 100: 96: 2093–2097.
2426–2430. Zoepf, T., Jakobs, R., Rosenbaum, A., Apel,
Zoepf, T., Jakobs, R., Arnold, J. C., Apel, D., D., Arnold, J. C., and Riemann, J. F., 2001b.
Rosenbaum, A., and Riemann, J. F., 2001a. Photodynamic therapy with 5-aminolevulinic
Photodynamic therapy for palliation of nonresect- acid is not effective in bile duct cancer.
able bile duct cancer – preliminary results with Gastrointest. Endosc. 54: 763–766.
A. Diagnosis
36
Splenic Metastases: Diagnostic Methods
Eva Compérat and Frédéric Charlotte

INTRODUCTION obtained mainly from autopsy series pub-


lished before 1990 and ranged between
Metastases from solid tumors to the spleen due 2.3 % and 7.1 % (Berge, 1974). The larger
to hematogenous dissemination are confined autopsy series was reported by Berge
to the splenic parenchyma, and should not be (1974) who found 312 splenic metastases
confused with small superficial subcapsular for 7,165 autopsies performed for cancer.
foci associated with peritoneal dissemination In this study, microscopic splenic metas-
observed in ovarian cancers (Tserkezoglou tases were identified in 50 % of subjects
et al., 2005). According to this definition, who had metastases in at least five organs.
splenic metastases were thought to be excep- After 1990, studies from different parts
tional but the incidence of reported cases has of the world brought indications regard-
been increasing due to the improvement of ing the prevalence of splenic metastases
medical imaging and the long-term follow- in living patients. In a study from the
up of patients with cancer. Metastases to the U.S.A., (Kraus et al., 2001), 1.3 % of
spleen generally occur in the context of mul- 1,280 sequential tumors in splenectomy
tivisceral metastatic cancer at terminal stage. specimens were metastatic. In the same
Solitary metastases have also been reported study, 9.8 % of 122 splenectomies per-
(Compérat et al., 2007), and are becoming a formed for diagnosis contained metastasis.
diagnostic dilemma with primary tumors of In a Japanese study (Ishida et al., 1997),
the spleen. After a brief description of epi- 0.15 % of 24,761 patients examined by
demiology, pathologic features, pathogenesis ultrasonography had splenic metastasis.
and clinical aspects of splenic metastases, A Chinese report (Lam and Tang, 2000)
this review will outline diagnostic procedures found metastasis in 1.1 % of 1,743 sequen-
and the differential diagnoses. tial splenectomy specimens.

EPIDEMIOLOGY PATHOLOGIC FEATURES


The prevalence of splenic metastases Splenic metastases can present as three
in large populations with cancer was main macroscopic patterns: macronodular,

489
490 E. Compérat and F. Charlotte

micronodular, and diffuse (Berge, 1974). In


the macronodular pattern, the splenic paren-
chyma is partially replaced by macroscopi-
cally discernible, solitary or multiple large
nodules of varying sizes (Figure 36.1). The
micronodular pattern is characterized by a
relatively uniform miliary aspect (Figure
36.2). The tumor nodules can be located in
the white pulp or the red pulp. In the diffuse
pattern, the splenic parenchyma is com-
pletely replaced by the tumor cells. In the
microscopic pattern, no macroscopic lesion
can be seen. In this pattern, the tumor cells
can be confined to the venous sinuses, the
red pulp, the white pulp, or the trabecular
vessels, or can be found in several of these
compartments (Berge, 1974). Whatever the

Figure 36.2. Macroscopic findings of micronodu-


lar aspects with uniform miliary pattern

pattern of splenic infiltration, the splenic


metastasis and primary tumor are generally
similar in terms of cytological and architec-
tural aspect.

PATHOGENESIS
The development of metastases in spe-
cific organs according to the type of pri-
mary source depends on anatomic factors,
the microenvironment of host tissue, and
the intrinsic characteristics of tumor cells
(Chambers et al., 2002). The pathogenesis
of splenic metastases is largely specula-
tive. The relative rarity of splenic metas-
tases might be explained by two main
causes: (1) mechanical factors impeding
Figure 36.1. Macroscopic findings: multiple large the splenic implantation of blood-borne
nodules of varying sizes in spleen parenchyma cancer cells, that is, the constant flow of
36. Splenic Metastases: Diagnostic Methods 491

blood through the spleen and the rhythmic various types of metastatic cells, are found
contraction of splenic capsule; the sharp not only in the preferential sites of metas-
angle of splenic artery branching from the tases such as the bone marrow, lung and
celiac artery preventing large clumps of lymph nodes, but also in the spleen where
tumor cells from passing through; the lack it plays a crucial role in lymphocyte hom-
of afferent lymphatic vessels limiting lym- ing (Chambers et al., 2002). Therefore,
phogenic metastases, and (2) inhibitory implantation of cancer cells in the splenic
effect of splenic microenvironment on the parenchyma may occur, but clinically
growth of metastatic cells (Berge, 1974). detectable metastases are rare because
The respective contribution of these factors the splenic microenvironment may not
to the resistance of the spleen to metastatic facilitate the growth of micrometastatic
involvement is still debated. The mechani- foci. This probably explains the contrast
cal theory was proposed at the time the between the high prevalence of splenic
dissemination from the primary tumor and micrometastases at autopsy in patients
the implantation of tumor cells in distant with multivisceral cancer and the rarity
organs were regarded as a late event in of clinically detectable metastases. This
the multistep process of cancer. However, suggests that mechanical factor seems to
the recent development of sensitive immu- play a minor role in explaining the rarity
nologic and molecular methods has per- of clinical splenic metastases.
mitted the detection of individual tumor
cells in regional lymph nodes, blood, or
distant organs at the time of primary tumor CLINICAL FEATURES
diagnosis for various histological types
of cancer (Pantel and Brakenhoff, 2004). Most splenic metastases are a part of
This supports the concept that micrometa- multivisceral metastatic disease. In this
static dissemination occurs, in fact, early context, breast, lung, ovarian, colorectal, and
in the course of malignant disease and is gastric carcinomas, and skin melanoma are
not affected by mechanical factors. This the most common primary sources (Berge,
is also in accordance with the results of 1974; Lam and Tang, 2000). Skin melanoma
experimental metastasis models using neo- has the highest rate of splenic metastases
plastic cells labeled with green fluorescent per primary tumor since more than 30 % of
protein (Goodison et al., 2003). patients with skin melanoma have splenic
From this recent advance in the knowl- metastasis at autopsy (Berge, 1974; Lam and
edge of metastatic process, it may be Tang, 2000). Splenic metastases also occur
assumed that the presence of disseminated as a solitary splenic mass, synchronous or
cancer cells in the spleen may not be metachronous to the primary tumor. To our
detected at the time of primary diagnosis knowledge, 93 well-documented cases of
by conventional methods. It is noteworthy solitary splenic metastases are known which
that the expression of chemokines in the are listed in Table 36.1. From diagnosis of
spleen does not constitute an obstacle to primary tumor to the discovery of solitary
cancer cell homing because stromal-cell- splenic metastasis it might take between
derived factor 1 (SDF-1) and the ligand 0 to 264 months with a median of 28
for CXC chemoreceptor-4 expressed by months. Colorectal and ovarian carcinomas
492 E. Compérat and F. Charlotte

Table 36.1. Summary of 93 case reports of solitary splenic metastases.


No. of
asymp- No. of cases
Interval (mo) tomatic with positive
Primary site No. Histological type (No.) range (median) cases serum marker References
Colon/rectum 20 Adenocarcinoma 0–132 (27) 18/20 13/20 Ishida et al., 1997
Kim et al., 2000
Agha-Mohammadi and Calne,
2001
Avesani et al., 2001
Okuyama T et al., 2001
Genna et al., 2003
Cavallaro et al., 2004
Ovary 18 Serous cystadeno- 18–183 (51) 15/17* 10/17* Tserkezoglou et al., 2005
carcinoma (17)
Mucinous cystaden- 1 1 1 Koh et al., 2004
ocarcinoma (1)
Lung 10 Adenocarcinoma (4) 0–25(12) 2/4 nd Agha-Mohammadi and Calne,
2001
Large cell carcinoma 0 1/3 nd Schmidt and Smith, 2004
(3)
Bronchioalveolar 20 0 nd
carcinoma (1)
Carcinoid (1) 96 0 nd
Squamous cell 14 1 nd
carcinoma (1)
Endometrium 9 Adenocarcinoma 11–120 (31) 3/9 2/9 Agha-Mohammadi and Calne,
2001
Giuliani et al., 1999
Hadjileontis et al., 2004
Kidney 9 Clear cell carcinoma 0–264 (48) 3/6* nd Ishida et al., 1997
Kugel et al., 2003
McGregor et al., 2003
Stomach 7 Adenocarcinoma 16–102 (48) 3/3* 3/3* Opocher et al., 2000;
Yamanouchi et al., 2002
Cervix 6 Squamous cell 49–69 (56) 1/4* 1/1* Agha-Mohammadi and Calne,
carcinoma (4) 2001
Adenocarcinoma (2) 14–60 (37) 1/2 nd Carvalho et al., 1997
Pang, 2004
Breast 3 Adenocarcinoma 0–108 (24) 0 nd Barreca et al., 2001
Iype S et al., 2002
Hoar et al., 2003
Prostate 3 Adenocarcinoma 0–60 (18) 2/3 2/3 Sharpe et al., 1993
Naseem et al., 1998
Compérat et al., 2007
Esophagus 3 Squamous cell 0–96 (6) 1/3 nd Agha-Mohammadi and Calne,
carcinoma 2001
Vyas et al., 2002;
Kimura et al., 2003
Fallopian tube 1 Adenocarcinoma 48 1 1 Dubecq-Princeteau et al., 1997
Skin 1 Melanoma 30 0 nd Agha-Mohammadi and Calne,
2001
Uveal tract 1 Melanoma 10 0 nd Tas et al., 2004
Thyroid 1 Undifferentiated 6 0 nd Mayayo et al., 2003
carcinoma
Unknown 1 Adenocarcinoma 0 0 1 Alici et al., 2003
Total 93 0–264 (28) 55/86 34/56 (61%)
(64%)
*
data not available for all cases; mo: months; nd: not described; ref: references.
36. Splenic Metastases: Diagnostic Methods 493

are the most common sources. On the FDG-PET is to discriminate benign from
other hand, solitary splenic metastases malignant splenic masses, the latter is char-
from breast carcinoma (Barreca et al., 2001; acterized by an increase in FDG uptake.
Iype et al., 2002; Hoar et al., 2003) or skin Moreover, FDG-PET coupled with CT can
melanoma are rare. Exceptionally, a splenic also identify other tumor sites expressing
metastasis reveals a clinically occult primary as hypermetabolic foci which are unsus-
tumor (McGregor et al., 2003). Only one pected on conventional imaging but may
report described a solitary splenic metas- be more accessible for biopsy.
tasis from unknown primary cancer (Alici Serum level of tumor markers utilized
et al., 2003). in the follow-up of patient with cancer can
Splenic metastases are most often predict the appearance of solitary splenic
incidentally detected by medical imag- metastases before their clinical expression
ing techniques. When isolated, more than or radiological detection and decreases
60 % of splenic metastases are asympto- within normal limits after splenectomy
matic (Table 36.1). Splenic metastasis can (Dubecq-Princeteau et al., 1997). For
also be revealed by fatigue (Ishida et al., example, Compérat et al. (2007) have
1997), weight loss (Agha-Mohammadi and recently seen a solitary splenic metastasis
Calne, 2001), fever (Kugel et al., 2003), from prostate carcinoma occurring 5 years
abdominal pain (Carvalho et al., 1997; after a radical prostatectomy (Figure 36.3).
Agha-Mohammadi and Calne, 2001; Iype A progressive increase in the serum PSA
et al., 2002; Vyas, 2002; Mayayo et al.,
2003; Schmidt and Smith, 2004; Tas et al.,
2004;), splenomegaly (Sharpe et al., 1993;
Agha-Mohammadi and Calne, 2001; Alici
et al., 2003), anemia or thrombocytopenia
due to hypersplenism (Kugel et al., 2003;
Hadjileontis et al., 2004), and more rarely
by splenic rupture (Hoar et al., 2003;
Schmidt and Smith, 2004).

DIAGNOSTIC PROCEDURES
Splenic metastases can be detected by
ultrasonography or computed tomography
(CT) in the regular follow-up of patients
with cancer or in the work-up performed
at the time of any event related to cancer.
The increasing use of 18-fluorodeoxyglu-
cose (FDG)-positron emission tomography
(PET) has resulted in more patients with
asymptomatic metastases being identified
than previously found using conventional Figure 36.3. Solitary splenic metastasis from pros-
radiological techniques. The advantage of tate carcinoma, H&E × 4
494 E. Compérat and F. Charlotte

level was observed before the tumor could 0 % and 2 % and mainly affect patients
be detected by imaging technique. Serum with thrombocytopenia or vascular splenic
PSA level decreased within normal limits neoplasms.
after splenectomy and the patient remained
asymptomatic 13 months thereafter. This
case history along with other published DIFFERENTIAL DIAGNOSIS
similar cases suggests that splenic metas-
tases might result from the growth of early Despite improvement of medical imag-
blood-borne disseminated cancer cells ing techniques such as PET, the clini-
within the spleen after a period of clinical cal diagnosis of splenic metastases is
latency. This could explain why isolated largely dependent on patient history.
splenic metastases are not necessarily the Radiologically, metastatic disease fre-
precursor sign of active metastatic can- quently affects the spleen in a manner
cer at terminal stage because long term that is indistinguishable from primary
remission has been achieved in patients splenic lesions. A splenic mass without
treated with splenectomy alone (Agha- context of active cancerous disease is
Mohammadi and Calne, 2001). Further more suggestive of a primary splenic
follow-up studies of patients with solitary lesion such as lymphomas, vascular
splenic metastasis detected by imaging tumors, or infectious lesions. Conversely,
techniques such as PET, are needed to a previous history of malignant disease
better understand the significance of this constitutes an indication for the diagno-
clinical manifestation. sis of splenic metastasis. The search for
The finding of a splenic nodule is additional clinical findings such as lym-
regarded as metastatic in case of dissemi- phadenopathy or disease in other organs
nated cancerous disease, and splenectomy may aid in the diagnosis of lymphoma or
is generally not performed, as the diagno- metastatic disease. Rarer splenic lesions
sis relies on biopsy of a more accessible such as hamartoma and inflammatory
tumor site. Isolated splenic metastasis can pseudotumor can clinically and radiologi-
be diagnosed by fine-needle aspiration or cally mimic a metastatic tumor but there
percutaneous biopsy in place of splenec- is generally no confusion on histological
tomy. The use of these imaging-guided grounds.
techniques to diagnose splenic metastases Primary malignant lymphoma of the
has rarely been reported in the medical lit- spleen is defined as a disease limited to
erature. This may be attributed to the fear the spleen and splenic hilum. The main
of hemorrhagic complication because the histological types include large B-cell
spleen is highly vascular. However, these lymphoma, marginal zone B-cell lymphoma,
imaging-guided techniques are less aggres- mantle cell lymphoma, hairy-cell leuke-
sive than the surgical approach and permit mia, and gamma-delta T-cell lymphoma
the avoidance of unnecessary splenectomy (Mollejo et al., 2003). Large B-cell lym-
for the diagnosis of benign splenic lesions phoma can be mistaken for metastasis from
such as granulomatous disease. A success- undifferentiated carcinoma or melanoma.
ful diagnosis can be achieved in ∼ 90 % of Immunostaining with cytokeratins for car-
the cases. Complications range between cinoma, common leukocyte antigen for
36. Splenic Metastases: Diagnostic Methods 495

lymphoma, and S-100 protein, HMB45, or


melan A for melanoma is useful in estab-
lishing a correct diagnosis (Figure 36.4).
Interestingly, two case reports described
the association between carcinoma and
lymphoma in the same spleen (Sharpe
et al., 1993; Hoar et al., 2003).
Vascular tumors of the spleen can pose
a diagnostic problem radiologically, but
they are easily distinguished from met-
astatic tumors on histological grounds.
Hemangioma is the most common primary
tumor of the spleen. Littoral cell angioma
is composed of vascular channels lined
by tall endothelial cells expressing both
endothelial (factor VIIIr) and histiocytic
markers (CD68). It is noteworthy that this
vascular tumor is frequently associated
with visceral malignancy such as colorec-
tal, lung, renal, and pancreatic carcinoma.
Primary angiosarcoma of the spleen, par- Figure 36.4. Metastasis of melanoma in spleen
ticularly in its pleomorphic or epithelioid parenchyma, H&E × 20
variant, can be a problem of differential
diagnosis with a metastasis from undif-
ferentiated carcinoma or melanoma. The
right diagnosis is based upon the expres- disorder. Fine needle aspiration is very
sion of vascular makers CD31, factor VIII, useful in the differential diagnosis between
or CD34 by the tumor cells. these infectious causes and neoplasm.
Nonneoplastic disorders of the spleen In conclusion, although classically
include infections such as disseminated regarded as very low, the prevalence of
fungal disease, or septic emboli, and gran- splenic metastases seems to increase with
ulomatous diseases such as sarcoidosis the improvement of medical imaging tech-
or tuberculosis. Findings of associated niques. Therefore, splenic metastases are
lymphadenopathies, pulmonary, and medi- becoming a diagnostic dilemma with pri-
astinal diseases should aid in the diagnosis mary lesions of the spleen, particularly for
of sarcoidosis. In immunocompromised solitary lesions in patients with a history
individuals, tuberculosis, Mycobacterium of malignant disease. The most common
avium intracellulare, Pneumocystis cari- sources of splenic metastases are breast,
nii, and disseminated Kaposi sarcoma may lung, colorectal and ovarian carcinoma,
cause multiple splenic masses. In these and melanoma. Fine-needle aspiration and
cases, the patient’s clinical condition and percutaneous biopsy of splenic lesions are
immune status will help in distinguishing useful in establishing the right diagnosis in
an infectious entity from a neoplastic most cases.
496 E. Compérat and F. Charlotte

REFERENCES metastasis from endometrial carcinoma. J. Exp.


Clin. Cancer Res. 18:93–96.
Agha-Mohammadi, S., and Calne, R.Y. 2001. Goodison, S., Kawai, K., Hihara, J., Jiang, P.,
Solitary splenic metastasis. Case report and Yang, M., Urquidi, V., Hoffman, R.M., and
review of the literature. Am. J. Clin. Oncol. Tarin, D. 2003. Prolonged dormancy and site-
24:306–310. specific growth potential of cancer cells spon-
Alici, S., Kosem, M., and Kotan, C. 2003. Isolated taneously disseminated from non metastatic
splenic metastases occurring as an unknown pri- breast tumors as revealed by labeling with
mary lesion. J. Postgrad. Med. 49:83–84. green Fluorescent Protein. Clin. Cancer Res.
Avesani, E.C., Cioffi, U., de Simone, M., Botti, F., 9:3808–3814.
Carrara, A., and Ferrero, S. 2001. Synchronous Hadjileontis, C., Amplianitis, I., Valsamides, C.,
isolated splenic metastasis from colon carci- Harisis, G., Nepka, H., and Kafanas, A. 2004.
noma. Am. J. Clin. Oncol. 24:311–312. Solitary splenic metastasis of endometrial carci-
Barreca, M., Angelini, D., Gallo, A., Puntillo, F., noma ten years after hysterectomy. Case report
Amodio, P.M., and Fernandes, E. 2001. Single and review of the literature. Eur. J. Gynaecol.
asymptomatic splenic metastasis of breast car- Oncol. 25:233–235.
cinoma: report of a clinical case. G. Chir. Hoar, F.J., Chan, S.Y., Stonelake, P.S., Wolverson,
22:227–228. R.W., and Bareford, D. 2003. Splenic rupture as
Berge T. 1974. Splenic metastases. Frequencies a consequence of dual malignant pathology: a
and patterns. Acta Pathol. Microbiol. Scand. case report. J. Clin. Pathol. 56:709–710.
82:499–506. Ishida, H., Konno, K., Ishida, J., Shirayama, K.,
Carvalho, L., Azevedo, I., Salgado, L., Ferreira, Naganuma, H., Komatsuda, T., Hamashima, Y.,
E.S., Henrique, R., de Carvalho, R.G., and and Masamune, O. 1997. Isolated splenic metas-
Vieira, E. 1997. Squamous cell carcinoma of tases. J. Ultrasound Med. 16:743–749.
the cervix metastatic to the spleen – case report. Iype, S., Akbar, M.A., and Krishna, G. 2002.
Gynecol. Oncol. 67:107–110. Isolated splenic metastasis from carcinoma of
Cavallaro, A., Modugno, P., Specchia, M., Pontenza, the breast. Postgrad. Med. J. 78:173–174.
A.E., Loschiavo, V., Colli, R., Lauriola, L., and Kim, J.C., Jeong, C.S., Kim, H.C., Yu, C.S., Kang,
Barone, C. 2004. Isolated splenic metastasis G.H., and Lee, M.G. 2000. Isolated splenic
from colon cancer. J. Exp. Clin. Cancer Res. metastasis from colorectal carcinoma: a case
23:143–146. report. J. Korean Med. Sci. 15:355–358.
Chambers, A.F., Groom, A.C., and MacDonald, I.C. Kimura, Y., Miyazaki, M., Saeki, H., Ohga, T.,
2002. Dissemination and growth of cancer cells Nozoe, T., and Sugimachi, K. 2003. Solitary
in metastatic sites. Nat. Rev. Cancer 2:563–572. splenic metastasis derived from esophageal can-
Compérat, E., Azzouzi, A.R., Chartier-Kastler cer. Hepatogastroenterology 50:1336–1337.
E., Ménégaux F., Capron, F., Richard, F., and Koh, Y.S., Kim, J.C., and Cho, C.K. 2004.
Charlotte, F. 2007. Late recurrence of a prostatic Splenectomy for solitary splenic metastasis of
adenocarcinoma as a solitary splenic metastasis. ovarian cancer. BMC. Cancer 4:96
Urol. Int. 78:86–88. Kraus, M.D., Fleming, M.D., and Vonderheide,
Dubecq-Princeteau, F., Lefranc, J.P., Touboul, E., R.H. 2001. The spleen as a diagnostic specimen:
Chauvel, A., and Le Charpentier, Y. 1997. a review of 10 years’ experience at two tertiary
Adenocarcinoma of the fallopian tube: natural care institutions. Cancer 91:2001–2009.
history of an isolated splenic metastasis. Acta Kugel, V., Dekel, Y., Konichezky, M., Baniel,
Oncol. 36:221–223. J., Livne, P.M., and Koren, R. 2003. Unusual
Genna, M., Leopardi, F., Valloncini, E., Molfetta, splenic metastasis from renal cell carcinoma. A
M., De Manzoni, G., and Castelli, A. 2003. case report and review of the literature. Pathol.
Metachronous splenic metastasis of colon can- Res. Pract. 199:739–743.
cer. A case report. Minerva. Chir. 58:811–814. Lam, K.Y., and Tang, V. 2000.Metastatic tumors
Giuliani, A., Caporale, A., Di Bari, M., Demoro, to the spleen. A 25-year clinicopathologic study.
M., and Mingazzini, P. 1999. Isolated splenic Arch. Pathol. Lab. Med. 124:526–530.
36. Splenic Metastases: Diagnostic Methods 497

Mayayo, E., Blazquez, S., Gomez-Aracil, V., Sauri, vix in the spleen: case report. South Med. J.
A., and Martinez, S. 2003. Spleen metastasis 97:301–304.
from thyroid carcinoma. Report of a case with Pantel, K., and Brakenhoff, R.H. 2004. Dissecting
diagnosis by fine needle aspiration cytology. the metastatic cascade. Nat. Rev. Cancer 4:
Acta Cytol. 47:1116–1118. 448–456.
McGregor, D.H., Wu, Y., Weston, A.P., Mcanaw, Schmidt, B.J., and Smith, S.L. 2004. Isolated splenic
M.P., Bromfield, C., and Bhattatiry, M.M. 2003. metastasis from primary lung Adenocarcinoma.
Metastatic renal cell carcinoma of spleen diag- South. Med. J. 97:298–300.
nosed by fine-needle aspiration Spleen. Am. J. Sharpe, R.W., Rector, J.T., Rushin, J.M., Garvin,
Med. Sci. 326:51–54. D.F., and Cotelingam, J.D. 1993. Splenic metas-
Mollejo, M., Algara, P., Mateo, M., Menarguez, tasis in hairy cell leukemia. Cancer 71:2222–
J., Pascual, E., Fresno, M.F., Camacho, F., and 2226.
Piris, A. 2003. Large B-cell lymphoma present- Tas, F., Ustuner, Z., Buyukbabani, N., Tenekeci, N.,
ing in the spleen: Identification of different clin- and Topuz, E. 2004. Massive and isolated metas-
icopathologic conditions. Am. J. Surg. Pathol. tases to spleen of uveal malignant melanoma.
27: 895–902. Retina 24:170–172.
Naseem, M.S., Jan, H.A., Britton, K.E., and Tserkezoglou, A., Kontou, S., Hatjieleftheriou,
Nargund, V.H. 1998. Splenic metastasis from G., Nikolaidou, M.E., Plataniotis, G.,
adenocarcinoma of the prostate. Br. J. Urol. Apostolikas, N., and Magiakos, G. 2005.
82:597–598. Solitary parenchymal splenic recurrence
Okuyama, T., Oya, M., and Ishikawa, H. 2001. of ovarian adenocarcinoma: a case report
Isolated splenic metastasis of sigmoid colon and review of the literature. Anticancer Res.
cancer: a case report. Jpn. J. Clin. Oncol. 25:1471–1476.
31:341–345. Vyas, S.J., Chitaleb, A.R., and Deshpande, R.K.
Opocher, E., Santambrogio, R., Bianchi, P., Cioffi, 2002. Late splenic metastasis after curative
U., De Simone, M., Vellini, S., and Montorsi, resection for oesophageal carcinoma. Eur. J.
M. 2000. Isolated splenic metastasis from gastric Cardiothorac. Surg. 22:1011–1013.
carcinoma. value of CEA and CA 19–9 in early Yamanouchi, K., Ikematsu, Y., Waki, S., Kida, H.,
diagnosis: report of two case. Am. J. Clin. Oncol. Nishiwaki, Y., Gotoh, K., Ozawa, T., and
23:579–580. Uchimura, M. 2002. Solitary splenic metastasis
Pang, L.C. 2004. Solitary recurrent metastasis of from gastric cancer: report of a case. Surg. Today
squamous cell carcinoma of the uterine cer- 32:1081–1084.
Index

AASLD. See American Association for the Study Alcohol. See Fatty liver
of Liver Diseases guidelines Aldolase B (liver type), hepatitis C virus-
Abdomen related human hepatocellular
magnetic resonance imaging of, 21 carcinoma and, 348
T2-weighted images of, 22 Alpha-enolase, hepatitis C virus-related
2DT-FSE, 22 human hepatocellular carcinoma
lesion characterization with, 22–23 and, 347
single-shot echo planar, 22, 23 Alpha-fetoprotein (AFP), for hepatocellular
single-shot fast spin-echo, 22, 23 carcinoma screening, 138–139, 174
steady-state free precession, 22 Alternative-free response receiver operating
Abscess, characterization of, 167 characteristic (AFROC), 231
arterial phase, 167 American Association for the Study of Liver
parenchymal phase, 167 Diseases (AASLD) guidelines, 177,
portal vein phase, 167 262–263
typical features, 167 Angiography-assisted computed tomography,
ActiTest, 139, 140 36–37, 253
Adenoma, characterization of, 166–167 Angiosarcoma, characterization of, 171
arterial phase, 167 Antiangiogenesis therapy, evaluation of, future
atypical features, 167 perspectives of, 282–284
parenchymal phase, 167 APRI. See AST to platelet ratio index
portal vein phase, 167 Arginase 1, hepatitis C virus-related
typical features, 167 human hepatocellular carcinoma
Adenomatous hyperplasia, computed and, 348
tomography of, 255 Arterial portal microfistulas, contrast-enhanced
Adenosine triphosphate (ATP) synthase ultrasound and, 164–165
beta chain, hepatitis C virus-related Aspartate transaminase (AST)/alanine
human hepatocellular carcinoma transaminase (ALT) ratio, 139, 140
and, 347 AST/ALT. See Aspartate transaminase/alanine
ADI. See Agent detection imaging transaminase ratio
AFROC. See Alternative-free response receiver AST to platelet ratio index (APRI), 139, 140
operating characteristic ATP. See Adenosine triphosphate synthase beta
Agent detection imaging (ADI), 163 chain
contrast-enhanced, 151–153 Atypical adenomatous hyperplasia, computed
AI-700, 161 tomography of, 255
Albunex, 161 AXOR12, 325

499
500 Index

Bile cyst, characterization of, 166 CE-IOUS. See Contrast-enhanced intraoperative


Bile duct, intraductal papillary neoplasm of, ultrasonography
biliary cystic neoplasm, 419–426 Cervix, splenic metastases to, 492
bile duct lumen communication, 421 CEUS. See Contrast-enhanced ultrasound
classification, 419–421 CHA. See Coded harmonic angio
genetic aspects of, 422–424 cHCC-CC. See Combined hepatocellular and
histologic grade of tumor, 421–422 cholangiocarcinoma
invasive carcinoma progression, 421–422 Chemotherapy
molecular aspects of, 422–424 intraarterial, 287–289
mucin hypersecretion, 422 monitoring of, 2-fluoro-2-deoxy-D-glucose-
mucin phenotypes, 422 positron emission tomography in, 14–15
phenotypes, 422 CHFCs. See Ciliated hepatic foregut cysts
radiological features, 421 Cholangiocarcinoma
Biliary cystic neoplasm, 416–426 combined hepatocellular and
cystadenocarcinoma, 416–419 differential diagnosis of, 246–247
clinicopathologic features of, 416–419 enhanced computed tomographic findings,
pancreas mucinous cystic neoplasm 243–247
comparison, 419 pathogenesis of, 242–243
hepatobiliary cystadenoma endoscopic retrograde
clinicopathologic features of, 416–419 cholangiopancreatography for, 10
pancreas mucinous cystic neoplasm 2-fluoro-2-deoxy-D-glucose-positron emission
comparison, 419 tomography for, 10–11
intraductal papillary neoplasm of bile duct, intraductal ultrasonography for
419–426 cancer extension precise diagnosis, 432
bile duct lumen communication, 421 findings of, 432
classification, 419–421 indications, 431
genetic aspects of, 422–424 insertion methods, 430–431
histologic grade of tumor, 421–422 instruments, 429–430
invasive carcinoma progression, 421–422 methods, 429–431
molecular aspects of, 422–424 percutaneous transhepatic approach, 430–431
mucin hypersecretion, 422 three-dimensional, 432–433
mucin phenotypes, 422 transpapillary approach, 430
phenotypes, 422 magnetic resonance cholangiopancreatography
radiological features, 421 for, 10
Biliary hamartoma, non-neoplastic biliary cystic magnetic resonance imaging for, 10
lesions, 415–416 percutaneous transhepatic cholangiography
multicystic biliary hamartoma, 415–416 for, 10
Von Meyenburg complexes, 415 three-dimensional intraductal ultrasonography
Biliary tree, anatomical classification of, 412 diagnostic accuracy for extension, 433
Biopsy dual plane reconstruction images, 432
fine needle aspiration, of combined hepatocellular oblique reconstruction images, 432–433
and cholangiocarcinoma, 242 tumor volume measurement, 433
of liver, 139, 174 Ciliated hepatic foregut cysts (CHFCs), 414–415
needle, 252–253 Cirrhosis discriminant score (CDS), 139, 140
Biosphere, 161 Cirrhosis, of liver
Breast, splenic metastases to, 492 epidemiology of, 137–138
surrogate tests for, 139–141
Carcinoembryonic antigen (CEA), positron tumor arising in, 35–36
emission tomography and, 57–58 Coagulative necrosis, 87
CDS. See Cirrhosis discriminant score Coded harmonic angio (CHA), 150–151
Index 501

Colon/rectum, splenic metastases to, 492 enhanced findings, combined hepatocellular and
Colorectal cancer cholangiocarcinoma, 243–247
liver metastases and, 66–67 of localized fibrous tumors of liver, 17–19
detection of, 358–359 multidetector, methods of, 253
diagnostic hepatic ultrasound advantages, 357 of nodule-in-nodule hepatocellular
Doppler flow pattern, 359–363 carcinoma, 255
future advancements in, 363–364 spiral, for hepatocellular carcinoma, 265–266
grayscale echo pattern, 357–358 vascular endothelial growth factor, Western
postoperative follow-up ultrasound, 363 blotting measurement of, 202–209
ultrasound scanning technique for, Computed tomography during arterial portography
356–357 (CTAP), 37–39, 214, 253
liver metastases and, 18F-fluorodeoxyglucose Computed tomography during hepatic
positron emission tomography, 389–404 arteriography (CTHA), 37, 39, 253
abdominal cavity, 392–395 Contrast-enhanced agent detection imaging,
additional disease and, 398–399 151–153
anticancer systemic therapy, 401–402 Contrast-enhanced coded harmonic sonography,
benign increased activity of, 392, 397 150–151
change of management, 398–399 Contrast-enhanced color Doppler sonography,
contribution of, 393, 397 146–147
cryosurgery ablation, 403 Contrast-enhanced harmonic power Doppler
detection of, 389–391 sonography, 147–148
distant lymph nodes metastases, 395 Contrast-enhanced intraoperative ultrasonography
extra-abdominal organs, 395–397 (CE-IOUS), 63–64, 65–66, 176
extrahepatic metastases detection, 391–392 classification by, 66
hepatic arterial chemotherapy, 403–404 colorectal cancer, liver metastases and, 66–67
on patient’s management, 397–398 liver metastases, colorectal cancer and, 66–67
pelvic recurrence of rectal cancer, 393–395 tumor location by, 67–69
peritoneal metastases, 395 Contrast-enhanced power Doppler
physiologic activity of, 392, 397 sonography, 147
preoperative staging, 389 Contrast-enhanced pulse-inversion harmonic
radiofrequency ablation, 403 sonography, 148–150
regional therapy for, 401, 403 Contrast-enhanced sonography, hepatocellular
rising carcinoembryonic antigen, 400–401 carcinoma and, 153–156
survival impact, 399–400 Contrast-enhanced ultrasound (CEUS). See also
systemic therapy for, 401 Harmonic imaging; Nonlinear contrast
Combined hepatocellular and cholangiocarcinoma enhancement
(cHCC-CC) arterial portal microfistulas and, 164–165
differential diagnosis of, 246–247 for hepatocellular carcinoma, 264–265
enhanced computed tomographic findings, physical background of, 162–163
243–247 technological background of, 162–163
pathogenesis of, 242–243 Contrast Pulse Sequencing, 163
Computed tomography (CT) Cryoablation, 85
of adenomatous hyperplasia, 255 Cryosurgery ablation, colorectal cancer, liver
of advanced hepatocellular carcinoma, 253–254 metastases and, 18F-fluorodeoxyglucose
angiography, 36–37, 228–229 positron emission tomography, 403
angiography combined, 253 CT. See Computed tomography
of atypical adenomatous hyperplasia, 255 CTAP. See Computed tomography during arterial
contrast material fixed injection duration, 239 portography
contrast material fixed injection rate, 237–239 CTHA. See Computed tomography during hepatic
of early hepatocellular carcinoma, 254–255 arteriography
502 Index

Cystadenocarcinoma, biliary cystic neoplasm, Endoscopic retrograde cholangiopancreatography


416–419 (ERCP), for cholangiocarcinoma, 10
clinicopathologic features of, 416–419 Enoyl-CoA hydratase, hepatitis C virus-related
pancreas mucinous cystic neoplasm human hepatocellular carcinoma and, 348
comparison, 419 ERCP. See Endoscopic retrograde
Cyst, characterization of, 166 cholangiopancreatography
arterial phase, 166 Esophagus, splenic metastases to, 492
bile, 166 Ethanol injection, 85
hydatid, 166 percutaneous, 153
nonvital parasitic, 166 European Association for the Study of Liver
parenchymal phase, 166 Disease (EASLD), 177
portal vein phase, 166 European Association for the Study of the Liver
typical features, 166 (EASL) consensus, 262–263
DCE-MRI. See Dynamic contrast-enhanced Extrahepatic bile duct carcinoma (EHBDC)
magnetic resonance imaging diagnosis of, 442
epidemiology of, 441
Definity, 161 etiology of, 441–442
Diabetes. See Fatty liver mucin characteristics
Diffuse-weighted magnetic resonance imaging, immunohistochemistry, 458
189–190 methods, 455–458
Diffusion-weighted imaging (DWI), hepatic vessel MUC1, 453–455
signal suppression, 214–215 MUC4, 452–453
Doppler flow pattern, liver metastases, colorectal pathology of, 442
cancer and, 359–363 prognosis of, 443
Doppler sonography role of p53 in malignant tumors, 443–444
contrast-enhanced color, 146–147 mutation analysis, 444
contrast-enhanced harmonic power, 147–148 role of p63 in malignant tumors, 444–448
contrast-enhanced power, 147 analysis of, 447–448
Doppler-ultrasonography, for hepatocellular role of p73 in malignant tumors, 444–448
carcinoma, 264–265 analysis of, 447–448
Drugs. See Fatty liver Fallopian tube, splenic metastases to, 492
Dual plane reconstruction images,
cholangiocarcinoma, three-dimensional Fast spin echo (FSE), 215–216
intraductal ultrasonography, 432 Fatty liver, characterization of, 165–166
Dynamic contrast-enhanced magnetic resonance arterial phase, 166
imaging (DCE-MRI), 283 focal fatty change, 166
Dynamic magnetic resonance imaging, 39–40 focal fatty sparing, 165–166
Dysplastic nodule, characterization of parenchymal phase, 166
arterial phase, 168 portal vein phase, 166
parenchymal phase, 168 typical features, 166
portal vein phase, 168 Fatty Liver Shionogi (FLS) mouse, 334
typical features, 168 FDG-PET. See 18F-fluorodeoxyglucose positron
EASL. See European Association for the Study emission tomography; 2-fluoro-2-
of the Liver consensus deoxy-D-glucose-positron emission
tomography
EASLD. See European Association for the Study Ferritin light chain, hepatitis C virus-related human
of Liver Disease hepatocellular carcinoma and, 348
18
Echovist, 161 F-fluorodeoxyglucose positron emission
EHBDC. See Extrahepatic bile duct carcinoma tomography (FDG-PET)
Endometrium, splenic metastases to, 492 colorectal liver metastases and, 389–404
Index 503

abdominal cavity, 392–395 portal vein phase, 167


additional disease and, 398–399 typical features, 167
anticancer systemic therapy, 401–402 adenoma, characterization of, 166–167
benign increased activity of, 392, 397 arterial phase, 167
change of management, 398–399 atypical features, 167
contribution of, 393, 397 parenchymal phase, 167
cryosurgery ablation, 403 portal vein phase, 167
detection of, 389–391 typical features, 167
distant lymph nodes metastases, 395 angiosarcoma, characterization of, 171
extra-abdominal organs, 395–397 cyst, characterization of, 166
extrahepatic metastases detection, 391–392 arterial phase, 166
hepatic arterial chemotherapy, 403–404 bile, 166
on patient’s management, 397–398 hydatid, 166
pelvic recurrence of rectal cancer, 393–395 nonvital parasitic, 166
peritoneal metastases, 395 parenchymal phase, 166
physiologic activity of, 392, 397 portal vein phase, 166
preoperative staging, 389 typical features, 166
radiofrequency ablation, 403 detection of, 171–174
regional therapy for, 401, 403 hepatic, 172
rising carcinoembryonic antigen, 400–401 hepatocellular carcinoma surveillance,
survival impact, 399–400 173–174
systemic therapy for, 401 oncologic patient follow-up, 172–173
patient outcome of, 54–55 staging, 172–173
pitfalls of, 388 dysplastic nodule, characterization of
principle of, 388 arterial phase, 168
Fibroscan, 139 parenchymal phase, 168
Fibrosis, of liver, surrogate tests for, 139–141 portal vein phase, 168
Fibrotest, 139, 140 typical features, 168
Fine needle aspiration biopsy (FNAB), fatty liver, characterization of, 165–166
of combined hepatocellular and arterial phase, 166
cholangiocarcinoma, 242 focal fatty change, 166
FLLs. See Focal liver lesions focal fatty sparing, 165–166
FLS. See Fatty Liver Shionogi mouse parenchymal phase, 166
2-fluoro-2-deoxy-D-glucose-positron emission portal vein phase, 166
tomography (FDG-PET) typical features, 166
for chemotherapy monitoring, 14–15 hemangioma, characterization of, 164–165
for cholangiocarcinoma, 10–11 arterial phase, 165
for gallbladder carcinoma, 11–12 atypical features, 165
for hepatocellular carcinoma, 12–14 capillary, 164
for metastatic liver disease, 7–10 cavernous, 164
false-negative findings, 8 focal liver lesions, nonlinear contrast
false-positive findings, 8 enhancement of, 164–165
FNAB. See Fine needle aspiration biopsy parenchymal phase, 165
(FNAB) portal vein phase, 165
Focal liver lesions (FLLs), 66 typical features, 165
Focal liver lesions (FLLs), nonlinear contrast hepatocellular carcinoma, characterization of
enhancement of arterial phase, 168
abscess, characterization of, 167 parenchymal phase, 168
arterial phase, 167 portal vein phase, 168
parenchymal phase, 167 typical features, 168
504 Index

Focal liver lesions (FLLs), nonlinear contrast Glucose-regulated 78 kDa protein (GRP78),
enhancement of (Continued) hepatitis C virus-related human
histiocytoma, characterization of, 171 hepatocellular carcinoma and, 347
inflammatory pseudo tumors, characterization Glutamine synthetase, hepatitis C virus-related
of, 167–168 human hepatocellular carcinoma and, 347
leiomyoma, characterization of, 171 Gradient-recalled echo (GRE), 184
liver metastases, characterization of, 168–171 Grayscale echo pattern, liver metastases,
arterial phase, 170, 171 colorectal cancer and, 357–358
hyper-vascular, 171 GRE. See Gradient-recalled echo
hypo-vascular, 170 GRP75. See Glucose-regulated 75 kDa protein
parenchymal phase, 170, 171 GRP78. See Glucose-regulated 78 kDa protein
portal vein phase, 170, 171
typical features, 170, 171 HAIC. See Hepatic arterial infusion
lymphoma, characterization of, 171 chemotherapy
percutaneous ablative treatment monitoring, HAP. See Hepatic arterial-dominant phase
174–176 scanning
regenerative nodule, characterization of, 166 Harmonic imaging, 160–161
arterial phase, 166 advantage of, 160
parenchymal phase, 166 disadvantage of, 160
portal vein phase, 166 HBV. See Hepatitis B virus
typical features, 166 HCC. See Hepatocellular carcinoma
sarcomas, characterization of, 171 HCV. See Hepatitis C virus
Focal nodular hyperplasia, characterization of, 165 HCV-HCC. See Hepatitis C virus-related human
arterial phase, 165 hepatocellular carcinoma
atypical features, 165 HDGF. See Hepatoma-derived growth factor
parenchymal phase, 165 Heat shock 60 kDa protein (HSP60), hepatitis
portal vein phase, 165 C virus-related human hepatocellular
typical features, 165 carcinoma and, 347
Forns index, 139, 140 Heat shock 70 kDa protein 1 (HSP70.1), hepatitis
FSE. See Fast spin echo C virus-related human hepatocellular
carcinoma and, 347
Gadobenate dimeglumine (Gd-BOPTA), 89, 227 Heat shock cognate 71 kDa protein (HSC70),
Gadolinium hepatitis C virus-related human
hepatocellular carcinoma, enhancement of, hepatocellular carcinoma and, 347
185–187 Hemangioma, characterization of
superparamagnetic iron oxide and, 187 arterial phase, 165
Gadolinium chelates, 39–40, 88 atypical features, 165
Gadoxetic acid (Gd-ethoxybenzyldiethylenetriami capillary, 164
nepentaacetic acid; Gd-EOB-DTPA), 89, cavernous, 164
94, 227 focal liver lesions, nonlinear contrast
Gallbladder carcinoma, 2-fluoro-2-deoxy-D- enhancement of, 164–165
glucose-positron emission tomography parenchymal phase, 165
for, 11–12 portal vein phase, 165
Gd-BOPTA. See Gadobenate dimeglumine typical features, 165
Gd-EOB-DTPA. See Gadoxetic acid Hepatic arterial-dominant phase (HAP)
Gd-ethoxybenzyldiethylenetriaminepentaacetic scanning, 237
acid. See Gadoxetic acid Hepatic arterial infusion chemotherapy (HAIC),
Glucose-regulated 75 kDa protein (GRP75), 287, 289–290
hepatitis C virus-related human Hepatic foregut cyst, non-neoplastic biliary cystic
hepatocellular carcinoma and, 347 lesions, 414–415
Index 505

Hepatic intraarterial catheters, perfusion territory characterization of


visualization, 291–298 arterial phase, 168
image analysis, 293–297 parenchymal phase, 168
imaging techniques, 291 portal vein phase, 168
single-photon emission computed tomography typical features, 168
with integrated computed tomography contrast-enhanced sonography and, 153–156
for port perfusion scintigraphy, 291–293 development of, 196–199
therapeutic consequences, 297–298 diagnosis accuracy of, 231–232
Hepatic steatosis. See Fatty liver diagnosis of, magnetic resonance imaging for,
Hepatitis B virus (HBV), 137–138, 183 267–268
Hepatitis C virus (HCV), 137–138, 183 early, computed tomography images of, 254–255
Hepatitis C virus-related human hepatocellular epidemiology of, 137–138
carcinoma (HCV-HCC), 343 2-fluoro-2-deoxy-D-glucose-positron emission
glucose-regulated 75 kDa protein, 347 tomography for, 12–14
glucose-regulated 78 kDa protein, 347 gadolinium enhancement of, 185–187
glutamine synthetase, 347 hepatitis C virus-related human, 343
heat shock 60 kDa protein, 347 hepatoma-derived growth factor
heat shock 70 kDa protein 1, 347 evaluation of, 337–338
heat shock cognate 71 kDa protein, 347 prognostic significance, 338–340
proteomics for, 345–348 homeoprotein six1 overexpression
amino acid sequencing by liquid cell lines in, 314, 315–317
chromatography-tandem mass clinical samples of, 314
spectrometry, 346 materials in, 314–315
gel image analysis, 346 methods in, 314–315
in-gel digestion, 346 results of, 315–319
isoelectric focusing, 345 reverse transcription-polymerase chain
mass spectrometry analysis, 346 reaction, 314–315
proteins expression decreased in, 347–348 statistical analysis, 315
proteins expression increased in, 346–347 Western blot, 315
sodium dodecyl sulfate-polyacrylamide gel immunohistochemistry, research on, 200–202
electrophoresis, 345–346 KiSS-1 overexpression
tissue sample preparation, 345 immunohistochemistry, 328
two-dimensional gel electrophoresis, 345 materials, 327
Hepatobiliary cystadenoma, biliary cystic methods, 327–328
neoplasm results of, 328–330
clinicopathologic features of, 416–419 tissue preparation, 327
pancreas mucinous cystic neoplasm magnetic resonance imaging
comparison, 419 appearance of, 184–185
Hepatocarcinogenesis, multistep progression features of, 229–230
of, 250–251 nodule-in-nodule, computed tomography of, 255
Hepatocellular carcinoma (HCC) postoperative interferon alpha treatment of
after percutaneous radiofrequency ablation clinicopathological data in, 305–306
therapy, 232–233 disease-free survival of, 306–307
after transcatheter arterial immunohistochemistry, 304
chemoembolization, 232 materials in, 304–305
alpha-fetoprotein and, 138–139, 174 methods in, 304–305
atypical appearances of, 188–189 overall survival of, 306–307
diffuse, 189 results of, 305–307
large, 188–189 scoring of p48 immunohistochemistry, 304–305
small, 188 statistical analysis, 305
506 Index

Hepatocellular carcinoma (HCC) (Continued) Hilar cholangiocarcinoma, palliation of,


survival of, 306 photodynamic therapy for, 472–478
tissue microarray, 304 Histiocytoma, characterization of, 171
proteomicss for, 344 Hodgkin’s lymphoma, 171
antibodies and, 348–349 Homeoprotein six1 overexpression, in
radiologic diagnosis history of, 194–196 hepatocellular carcinoma, 314, 315–317
resection of, 69–73 cell lines in, 314, 315–317
limited, 70–71 clinical samples of, 314
parenchymal dissection, 71–72 materials in, 314–315
portal branch, compression of, 70 methods in, 314–315
portal branch, hooking of, 70 results of, 315–319
portal branch, puncture of, 69–70 reverse transcription-polymerase chain reaction,
post-resectional control, 72–73 314–315
systematic segmentectomy, 69 statistical analysis, 315
screening benefit of, 138–139 Western blot, 315
cost-effectiveness analyses of, 139 HSC70. See Heat shock cognate 71 kDa protein
ultrasonographic, 141–142 HSP60. See Heat shock 60 kDa protein
small nodules and, 268–270 HSP70.1. See Heat shock 70 kDa protein 1
staging of, 231 Hydatid cyst, characterization of, 166
surveillance for Hyper-vascular metastases, characterization of, 171
contrast-enhanced ultrasonography, 264–265 Hypo-attenuating nodular lesions
Doppler-ultrasonography, 264–265 natural outcome of, 256
power Doppler-ultrasonography, 264–265 treatment of, 257
radiologic techniques in, 263–264 Hypo-vascular metastases, characterization of, 171
spiral computed tomography, 265–266
ultrasonography, 264–265 IEF. See Isoelectric focusing
thalidomide and, vascular response evaluation, Imagent, 161
279–282 iMRI. See Intraoperative magnetic resonance
tumor vascularity evaluation, with power imaging
Doppler ultrasound, 279 Inflammatory pseudo tumors, characterization
ultrasonography and, 138–139 of, 167–168
vascular endothelial growth factor expression, Interferon alpha treatment, postoperative, of
196–199 hepatocellular carcinoma
vasculature of, 196–199 clinicopathological data in, 305–306
Hepatoma-derived growth factor (HDGF), disease-free survival of, 306–307
333–336 immunohistochemistry, 304
analytical methods, 336–337 materials in, 304–305
angiogenesis, role in, 335–336 methods in, 304–305
cancer progression, role in, 335–336 overall survival of, 306–307
developmentally regulated expression of, 334 results of, 305–307
hepatocarcinogenesis, role in, 334–335 scoring of p48 immunohistochemistry, 304–305
in hepatocellular carcinoma, evaluation of, statistical analysis, 305
337–338 survival of, 306
in hepatocellular carcinoma, prognostic tissue microarray, 304
significance, 338–340 Intraarterial chemotherapy, 287–289
immunohistochemical methods, 336–337 Intraductal papillary neoplasm of bile duct (IPNB),
materials in, 336–337 biliary cystic neoplasm, 419–426
High-frequency ultrasound bile duct lumen communication, 421
imaging of, preclinical liver metastases, 376–379 classification, 419–421
preclinical liver metastases, 373–374 genetic aspects of, 422–424
Index 507

histologic grade of tumor, 421–422 Laser-induced thermotherapy (LITT), 85


invasive carcinoma progression, 421–422 LC-MS/MS. See Liquid chromatography-tandem
molecular aspects of, 422–424 mass spectrometry
mucin hypersecretion, 422 Leiomyoma, characterization of, 171
mucin phenotypes, 422 LFTs. See Localized fibrous tumors
phenotypes, 422 Liquid chromatography-tandem mass
radiological features, 421 spectrometry (LC-MS/MS), amino acid
Intraductal ultrasonography (IDUS), sequencing by, for hepatitis C
cholangiocarcinoma virus-related human hepatocellular
cancer extension precise diagnosis, 432 carcinoma, 346
findings of, 432 LITT. See Laser-induced thermotherapy
indications, 431 Liver. See also Hepatocellular carcinoma
insertion methods, 430–431 biopsy of, 139, 174
instruments, 429–430 cirrhosis of
methods, 429–431 epidemiology of, 137–138
percutaneous transhepatic approach, 430–431 surrogate tests for, 139–141
three-dimensional, 432–433 tumor arising in, 35–36
transpapillary approach, 430 exploration of, intraoperative ultrasonography
Intraoperative magnetic resonance imaging for, 64–69
(iMRI), 81, 88 fibrosis of, surrogate tests for, 139–141
developments in, 96–97 localized fibrous tumors of, 17–19
radiofrequency ablation and, 90–98 noncirrhotic, tumor arising in, 36
conduct of, 92–96 radiofrequency ablation of, 14
future developments of, 96–98 transplant of, 83
potential indications for, 92 Liver metastases
rationale of, 90 characterization of, 168–171
technical developments in, 90–92 arterial phase, 170, 171
Intraoperative ultrasonography (IOUS) hyper-vascular, 171
contrast-enhanced, 63–64 hypo-vascular, 170
in hepatic surgery parenchymal phase, 170, 171
liver exploration in, 64–69 portal vein phase, 170, 171
technical aspects of, 63–64 typical features, 170, 171
tumor vessel classification, 68–69 colorectal cancer and, 66–67
IOUS. See Intraoperative ultrasonography detection of, 358–359
IPNB. See Intraductal papillary neoplasm diagnostic hepatic ultrasound advantages, 357
of bile duct Doppler flow pattern, 359–363
Isoelectric focusing (IEF), for hepatitis C virus-related future advancements in, 363–364
human hepatocellular carcinoma, 345 grayscale echo pattern, 357–358
postoperative follow-up ultrasound, 363
Ketohexokinase, hepatitis C virus-related human ultrasound scanning technique for,
hepatocellular carcinoma and, 348 356–357
Kidney, splenic metastases to, 492 colorectal cancer and, 18F-fluorodeoxyglucose
KiSS-1 overexpression, in hepatocellular positron emission tomography, 389–404
carcinoma abdominal cavity, 392–395
immunohistochemistry, 328 additional disease and, 398–399
materials, 327 anticancer systemic therapy, 401–402
methods, 327–328 benign increased activity of, 392, 397
results of, 328–330 change of management, 398–399
tissue preparation, 327 contribution of, 393, 397
Kupffer cells, of liver, 40–41 cryosurgery ablation, 403
508 Index

Liver metastases (Continued) Lung, splenic metastases to, 492


detection of, 389–391 Lymphoma, characterization of, 171
distant lymph nodes metastases, 395
extra-abdominal organs, 395–397 MAA. See Macroaggregated albumin
extrahepatic metastases detection, 391–392 Macroaggregated albumin (MAA), 105
hepatic arterial chemotherapy, 403–404 Magnetic resonance cholangiopancreatography
on patient’s management, 397–398 (MRCP), 429
pelvic recurrence of rectal cancer, 393–395 for cholangiocarcinoma, 10
peritoneal metastases, 395 Magnetic resonance imaging (MRI), 39, 225
physiologic activity of, 392, 397 3T, 190
preoperative staging, 389 of abdomen, 21
radiofrequency ablation, 403 for cholangiocarcinoma, 10
regional therapy for, 401, 403 contrast-enhanced dynamic, 226
rising carcinoembryonic antigen, 400–401 diffuse-weighted, 189–190
survival impact, 399–400 dynamic, 39–40
systemic therapy for, 401 hepatocellular carcinoma, appearance
2-fluoro-2-deoxy-D-glucose-positron emission of, 184–185
tomography for, 7–10 for hepatocellular carcinoma diagnosis,
false-negative findings, 9 267–268
false-positive findings, 8 hepatocellular carcinoma, features of, 229–230
liver dissection and, 73–74 of localized fibrous tumors of liver, 17–19
magnetic resonance imaging for, 7–8 for metastatic liver disease, 7–8
positron emission tomography assessment perfusion-weighted, 190
of, 104–105 preclinical liver metastases and, 371
post-resectional control of, 73–74 scanning technique, 225–226
preclinical subtraction images and, 88
bioluminescent imaging, 372 superparamagnetic iron oxide-enhanced,
fluorescent imaging, 372 213–214
high-frequency ultrasound, 373–374 of hepatocellular carcinoma, 230–231
high-frequency ultrasound imaging of, technique of, 183–184
376–379 thermal injury and, 86–90
magnetic resonance imaging and, 371 characteristics of, 86–88
positron emission tomography, 372 extent of, 88–90
three-dimensional imaging, 374–376 functional potential utility, 89–90
volume calculation, 374–376 tissue-specific contrast agents, 187–188
X-ray computed tomography, 371–372 tissue-specific contrast-enhanced, 226–227
resection of, 73–74 vascular endothelial growth factor, Western
area definition in, 73 blotting measurement of, 202–209
Liver tumors Malignant duodenal obstruction, palliative stenting
cryoablation for, 85 of, 469–470
ethanol injection for, 85 Mangafodipir trisodium (Mn-DPDP), 227
radiofrequency ablation for, 81–85 MCT. See Microwave coagulation therapy
alternatives of, 85 MDCT. See Multi-detector computed tomography
indications for, 83–84 Metastases, splenic
outcomes of, 84–85 clinical features, 491–493
principles of, 81–83 diagnostic procedures, 493–494
Localized fibrous mesothelioma. See Localized differential diagnosis, 494–495
fibrous tumors epidemiology of, 489
Localized fibrous tumors (LFTs), 17–19 pathogenesis of, 490–491
Low mechanical index imaging, 153 pathologic features, 489–490
Index 509

Metastatic liver disease duration of, 223


characterization of, 168–171 injection flow rate, 223
arterial phase, 170, 171 iodine concentration, 223
hyper-vascular, 171 image processing, 224–225
hypo-vascular, 170 optimal scanning delay, 223–224
parenchymal phase, 170, 171 scanning technique, 222
portal vein phase, 170, 171 scanning timing, 224
typical features, 170, 171 Multi-shot 2DFT-FSE, 22
colorectal cancer and, 66–67
2-fluoro-2-deoxy-D-glucose-positron emission Needle biopsy, 252–253
tomography for, 7–10 Nodular hepatocellular lesions, classification
liver dissection and, 73–74 of, 249–250
magnetic resonance imaging for, 7–8 Nodule-in-nodule hepatocellular carcinoma,
positron emission tomography assessment computed tomography of, 255
of, 104–105 Noncirrhotic liver, tumor arising in, 36
post-resectional control of, 73–74 Non Hodgkin’s lymphoma, 171
resection area definition in, 73 Nonlinear contrast enhancement, of focal liver
resection of, 73–74 lesions, 163–176
Microbubbles, 161 abscess, characterization of, 167
physics of, 146 arterial phase, 167
Microwave coagulation therapy (MCT), 85 parenchymal phase, 167
Mn-DPDP. See Mangafodipir trisodium portal vein phase, 167
Motion-probing gradient (MPG) pulses, 214 typical features, 167
MPG. See Motion-probing gradient pulses adenoma, characterization of, 166–167
MRCP. See Magnetic resonance arterial phase, 167
cholangiopancreatography atypical features, 167
MRI. See Magnetic resonance imaging parenchymal phase, 167
MUC1, 453–455 portal vein phase, 167
antibodies against, 455 typical features, 167
MUC4, 452–453 angiosarcoma, characterization of, 171
antibodies against, 453 cyst, characterization of, 166
Mucin characteristics, extrahepatic bile duct arterial phase, 166
carcinoma bile, 166
immunohistochemistry, 458 hydatid, 166
methods, 455–458 nonvital parasitic, 166
MUC1, 453–455 parenchymal phase, 166
MUC4, 452–453 portal vein phase, 166
Mucin hypersecretion, intraductal papillary typical features, 166
neoplasm of bile duct, 422 detection of, 171–174
Mucin phenotypes, intraductal papillary neoplasm hepatic, 172
of bile duct, 422 hepatocellular carcinoma surveillance,
Multicystic biliary hamartoma, 415–416 173–174
Multi-detector computed tomography (MDCT), oncologic patient follow-up, 172–173
33–34, 42, 429 staging, 172–173
limitations of, 233 dysplastic nodule, characterization of
prospects of, 233 arterial phase, 168
Multidetector-row helical computed tomography parenchymal phase, 168
(MDCT), 221 portal vein phase, 168
contrast administration for, 222–223 typical features, 168
dose, 222–223 fatty liver, characterization of, 165–166
510 Index

Nonlinear contrast enhancement, of focal liver Non-neoplastic biliary cystic lesions, 412–416
lesions (Continued) biliary hamartoma, 415–416
arterial phase, 166 multicystic biliary hamartoma, 415–416
focal fatty change, 166 Von Meyenburg complexes, 415
focal fatty sparing, 165–166 hepatic foregut cyst, 414–415
parenchymal phase, 166 prebiliary cysts, 412–414
portal vein phase, 166 around extrahepatic bile ducts, 414
typical features, 166 in hepatic hilum, 412–414
focal nodular hyperplasia, characterization in large portal tracts, 412–414
of, 165 Nonvital parasitic cyst, characterization of, 166
arterial phase, 165 Nyquist criteria, 27
atypical features, 165
parenchymal phase, 165 Obesity. See Fatty liver
portal vein phase, 165 Oblique reconstruction images,
typical features, 165 cholangiocarcinoma, three-dimensional
hemangioma, characterization of, 164–165 intraductal ultrasonography, 432–433
arterial phase, 165 Oncologic patients, tumor arising in, 36
atypical features, 165 Optison, 161
capillary, 164 Ovary, splenic metastases to, 492
cavernous, 164
focal liver lesions, nonlinear contrast P53, role of, in malignant tumors of bile duct,
enhancement of, 164–165 443–444
parenchymal phase, 165 analysis of, 444
portal vein phase, 165 P63, role of, in malignant tumors of bile duct,
typical features, 165 444–448
hepatocellular carcinoma, characterization of analysis of, 447–448
arterial phase, 168 P73, role of, in malignant tumors of bile duct,
parenchymal phase, 168 444–448
portal vein phase, 168 analysis of, 447–448
typical features, 168 Palliative bile duct stenting, 464–469
histiocytoma, characterization of, 171 Pancreas mucinous cystic neoplasm
inflammatory pseudo tumors, characterization cystadenocarcinoma comparison, 419
of, 167–168 hepatobiliary cystadenoma comparison, 419
leiomyoma, characterization of, 171 Parenchymal dissection, in hepatocellular
liver metastases, characterization of, 168–171 carcinoma, 71–72
arterial phase, 170, 171 PEI. See Percutaneous ethanol injection
hyper-vascular, 171 Percutaneous ablative treatment monitoring,
hypo-vascular, 170 nonlinear contrast enhancement, of focal
parenchymal phase, 170, 171 liver lesions, 174–176
portal vein phase, 170, 171 Percutaneous ethanol injection (PEI), 153, 174
typical features, 170, 171 Percutaneous radiofrequency ablation therapy,
lymphoma, characterization of, 171 hepatocellular carcinoma after, 232–233
percutaneous ablative treatment monitoring, Percutaneous transhepatic cholangiography (PTC),
174–176 for cholangiocarcinoma, 10
regenerative nodule, characterization of, 166 Perfusion-weighted magnetic resonance
arterial phase, 166 imaging, 190
parenchymal phase, 166 Periodically Rotated Overlapping ParallEL
portal vein phase, 166 Lines with Enhanced Reconstruction
typical features, 166 (PROPELLER), 215–216
sarcomas, characterization of, 171 superparamagnetic iron oxide-enhanced DWI
Index 511

T2 fast spin echo imaging using, extra-abdominal organs, 395–397


216–218 extrahepatic metastases detection, 391–392
PET. See Positron emission tomography hepatic arterial chemotherapy, 403–404
PGAA index, 139, 140 on patient’s management, 397–398
PGA index, 139, 140 pelvic recurrence of rectal cancer, 393–395
Photodynamic therapy (PDT), 470–479 peritoneal metastases, 395
contraindications for, 471–472 physiologic activity of, 392, 397
for hilar cholangiocarcinoma palliation, preoperative staging, 389
472–478 radiofrequency ablation, 403
mechanism of action, 470–471 regional therapy for, 401, 403
photosensitizers and, 472 rising carcinoembryonic antigen, 400–401
Photosensitizers, photodynamic therapy and, 472 survival impact, 399–400
PIHI. See Pulse-inversion harmonic sonography systemic therapy for, 401
(PIHI) patient outcome of, 54–55
Platelet counts, 139, 140 pitfalls of, 388
Portal branch principle of, 388
compression of, in hepatocellular carcinoma, 70 Postoperative interferon alpha treatment,
hooking of, in hepatocellular carcinoma, 70 of hepatocellular carcinoma
puncture of, in hepatocellular carcinoma, 69–70 clinicopathological data in, 305–306
Positron emission tomography (PET) disease-free survival of, 306–307
for detection of extrahepatic disease, 50–51 immunohistochemistry, 304
for detection of intrahepatic disease, 51–52 materials in, 304–305
in elevated carcinoembryonic antigen, 57–58 methods in, 304–305
for follow-up, 57 overall survival of, 306–307
in follow-up post hepatic resection, 58 results of, 305–307
liver metastases and scoring of p48 immunohistochemistry, 304–305
negative impact of, 54 statistical analysis, 305
positive impact of, 53–54 survival of, 306
metastatic liver disease, assessment of, 104–105 tissue microarray, 304
preclinical liver metastases and, 372 Power-Doppler ultrasound, for hepatocellular
as staging modality, to complement carcinoma, 264–265
conventional imaging, 50 for tumor vascularity evaluation of, 279
for surveillance, 57 Prebiliary cysts, non-neoplastic biliary cystic
value of, prognostic clinical risk score, 52–53 lesions, 412–414
in work-up of hepatic colorectal metastases, around extrahepatic bile ducts, 414
58–59 in hepatic hilum, 412–414
Yttrium-90 microsphere radioembolization and, in large portal tracts, 412–414
109–112 Preclinical liver metastases
Positron emission tomography (PET), bioluminescent imaging, 372
18
F-fluorodeoxyglucose (FDG) fluorescent imaging, 372
colorectal liver metastases and, 389–404 high-frequency ultrasound, 373–374
abdominal cavity, 392–395 high-frequency ultrasound imaging of, 376–379
additional disease and, 398–399 magnetic resonance imaging and, 371
anticancer systemic therapy, 401–402 positron emission tomography and, 372
benign increased activity of, 392, 397 three-dimensional imaging and, 374–376
change of management, 398–399 volume calculation, 374–376
contribution of, 393, 397 Preoperative biliary drainage, 464
cryosurgery ablation, 403 PROPELLER. See Periodically Rotated
detection of, 389–391 Overlapping ParallEL Lines with
distant lymph nodes metastases, 395 Enhanced Reconstruction
512 Index

Prostate, splenic metastases to, 492 laparoscopic disadvantages of, 119


Proteomics, 343–344 open surgical advantages of, 119
for hepatitis C virus-related human open surgical disadvantages of, 119
hepatocellular carcinoma, 345–348 percutaneous advantages of, 118
amino acid sequencing by liquid percutaneous disadvantages of, 118
chromatography-tandem mass operative (open), 83
spectrometry, 346 percutaneous, 83
gel image analysis, 346 prognosis using, 120–124
in-gel digestion, 346 intraoperative evaluation, 121
isoelectric focusing, 345 long-term results, 123–124
mass spectrometry analysis, 346 methods and, 120, 121–122
proteins expression decreased in, 347–348 patients and, 120
proteins expression increased in, 346–347 postoperative follow-up, 122
sodium dodecyl sulfate-polyacrylamide gel preoperative evaluation, 121
electrophoresis, 345–346 short-term results, 123
tissue sample preparation, 345 statistical analyses, 122
two-dimensional gel electrophoresis, 345 tumor unresectability, 120–121
for hepatocellular carcinoma tissues, 344 role of, 124–128
Prothrombin time, 139, 140 Radiologic diagnosis history, of hepatocellular
PTC. See Percutaneous transhepatic carcinoma, 194–196
cholangiography vascular endothelial growth factor expression
PTK787/ZK 222584 (PTK/ZK), 193 and, 199
Pulse-inversion harmonic sonography (PIHI), Receiver operating characteristic (ROC)
contrast-enhanced, 148–150 analyses, 231
Regenerative nodule, characterization of, 166
Quantison, 161 arterial phase, 166
parenchymal phase, 166
Radial fast spin-echo, 27–30 portal vein phase, 166
Radioembolization, 290–291 typical features, 166
Radiofrequency ablation (RFA) Resection
endpoint monitoring of, 86 of hepatocellular carcinoma (HCC), 69–73
impedance, 86 limited, 70–71
temperature, 86 parenchymal dissection, 71–72
ultrasound and, 86 portal branch, compression of, 70
future perspective of, 128–129 portal branch, hooking of, 70
intraoperative magnetic resonance imaging portal branch, puncture of, 69–70
and, 90–98 post-resectional control, 72–73
conduct of, 92–96 systematic segmentectomy, 69
future developments of, 96–98 of liver metastases, 73–74
potential indications for, 92 liver dissection, 73–74
rationale of, 90 post-resectional control, 73–74
technical developments in, 90–92 resection area definition, 73
of liver, 14 RFA. See Radiofrequency ablation
for liver tumor treatment, 81–85 ROC. See Receiver operating characteristic analyses
alternatives of, 85
indications for, 83–84 Sarcomas, characterization of, 171
outcomes of, 84–85 SDS-PAGE. See Sodium dodecyl sulfate-
principles of, 81–83 polyacrylamide gel electrophoresis
methodology of, 118–119 Serum albumin, hepatitis C virus-related human
laparoscopic advantages of, 118 hepatocellular carcinoma and, 348
Index 513

Single-photon emission computed tomography single-shot fast spin-echo, 22, 23


(SPECT), 106 steady-state free precession, 22
with integrated computed tomography for TACE. See Transcatheter arterial
port perfusion scintigraphy, hepatic chemoembolization
intraarterial catheters, perfusion territory Thalidomide, hepatocellular carcinoma and,
visualization, 291–293 vascular response evaluation, 279–282
Single-shot echo planar (SSEPI), 22, 23 Thermal injury, magnetic resonance imaging and
Single-shot fast spin-echo (SSFSE), 22, 23 characteristics of, 86–88
image quality improvement in, 24–27 extent of, 88–90
motion artifact reduction in, 24–27 functional potential utility, 89–90
Skin, splenic metastases to, 492 Thorotrast vinyl chloride, 171
Smoothelin, hepatitis C virus-related human 3D-IDUS. See Three-dimensional intraductal
hepatocellular carcinoma and, 348 ultrasonography
Sodium dodecyl sulfate-polyacrylamide gel Three-dimensional intraductal ultrasonography
electrophoresis (SDS-PAGE), for (3D-IDUS)
hepatitis C virus-related human cholangiocarcinoma, diagnostic accuracy for
hepatocellular carcinoma, 345–346 extension, 433
Solitary fibrous tumors. See Localized fibrous tumors cholangiocsarcoma
Sonazoid, 161 dual plane reconstruction images, 432
SonoVue®, 64, 161 oblique reconstruction images, 432–433
SPECT. See Single photon emission computed tumor volume measurement, 433
tomography 3T magnetic resonance imaging, 190
SPIO. See Superparamagnetic iron oxide Thyroid, splenic metastases to, 492
Spiral computed tomography, for hepatocellular Transarterial radioembolization, 289
carcinoma, 265–266 Transcatheter arterial chemoembolization (TACE),
Splenic metastases hepatocellular carcinoma after, 232
clinical features, 491–493 Triosephosphate isomerase, hepatitis C virus-
diagnostic procedures, 493–494 related human hepatocellular carcinoma
differential diagnosis, 494–495 and, 347
epidemiology of, 489 Tropomyosin beta chain, hepatitis C virus-related
pathogenesis of, 490–491 human hepatocellular carcinoma and, 348
pathologic features, 489–490 Tuberculosis, 167
SSEPI. See Single-shot echo planar TUF. See Tumor uptake fraction
SSFP. See Steady-state free precession Tumor(s)
SSFSE. See Single-shot fast spin-echo cryoablation for, 85
Steady-state free precession (SSFP), 22 ethanol injection for, 85
Stomach, splenic metastases to, 492 in liver cirrhosis, 35–36
Subtraction images, magnetic resonance imaging in noncirrhotic liver, 36
and, 88 in oncologic patients, 36
Superparamagnetic iron oxide (SPIO), 39, 40–42 radiofrequency ablation for, 81–85
enhanced magnetic resonance imaging, 213–214 alternatives of, 85
of hepatocellular carcinoma, 230–231 indications for, 83–84
gadolinium and, 187 outcomes of, 84–85
Systematic segmentectomy, in hepatocellular principles of, 81–83
carcinoma, 69 Tumor uptake fraction (TUF), 108
Tumor volume measurement, cholangiocsarcoma,
T2-weighted images, of abdomen, 22 three-dimensional intraductal
2DT-FSE, 22 ultrasonography, 433
lesion characterization with, 22–23 2DFT-FSE. See Two-dimensional Fourier
single-shot echo planar, 22, 23 transform-fast spin-echo
514 Index

Two-dimensional Fourier transform (2DFT) fast three-dimensional intraductal


spin-echo (FSE), 22 for cholangiocarcinoma, diagnostic accuracy
image quality improvement in, 24–27 for extension, 433
motion artifact reduction in, 24–27 for cholangiocarcinoma, dual plane
Two-dimensional gel electrophoresis (2-DE), reconstruction images, 432
for hepatitis C virus-related human for cholangiocarcinoma, oblique
hepatocellular carcinoma, 345 reconstruction images, 432–433
2-DE. See Two-dimensional gel electrophoresis for cholangiocarcinoma, tumor volume
measurement, 433
Ultrasound Uveal tract, splenic metastases to, 492
contrast-enhanced
arterial portal microfistulas, 164–165 Vascular endothelial growth factor
arterial portal microfistulas and, 164–165 (VEGF)
physical background of, 162–163 expression, hepatocellular carcinoma,
technological background of, 162–163 196–199
for hepatocellular carcinoma, 264–265 radiologic diagnosis history, 199
screening, 138–139 Western blotting measurement of, 202–209
high-frequency, preclinical liver metastases, VCMs. See Von Meyenburg complexes
373–374 VEGF. See Vascular endothelial growth factor
intraductal, cholangiocarcioma VIBE. See Volumetric Interpolated Breath-hold
cancer extension precise diagnosis, 432 Examination
findings of, 432 Volumetric Interpolated Breath-hold Examination
indications, 431 (VIBE), 226
insertion methods, 430–431 Von Meyenburg complexes (VMCs), 415
instruments, 429–430
methods, 429–431 Western blotting measurement
percutaneous transhepatic approach, 430–431 computed tomography, vascular endothelial
three-dimensional, 432–433 growth factor, 202–209
transpapillary approach, 430 hepatocellular carcinoma, homeoprotein six1
intraoperative overexpression, 315
contrast-enhanced, 63–64
tumor vessel classification, 68–69 X-ray computed tomography, preclinical liver
postoperative follow-up, colorectal cancer, liver metastases and, 371–372
metastases and, 363
power-Doppler Y-90. See Yttrium-90 microsphere
for hepatocellular carcinoma, 264–265 radioembolization
for tumor vascularity evaluation of, 279 Yttrium-90 (Y-90) microsphere radioembolization,
hepatocellular carcinoma, tumor vascularity 105–112
evaluation, 279 administration of, 105–106
radiofrequency ablation and, endpoint clinical results of, 109–111
monitoring of, 86 patient evaluation in, 106–108
radiofrequency ablation endpoint monitoring positron emission tomography and, 109–112
and, 86 treatment planning of, 105–106

Potrebbero piacerti anche