Liver Cancer
Liver Cancer
Edited by
Consolato M. Sergi, MD, PhD, MPH, FRCPC, FCAP, Departments of Pediatrics,
Laboratory Medicine and Pathology, Stollery Children’s Hospital, University of
Alberta, Edmonton, AB, Canada
Published by
Exon Publications, Brisbane, Australia
CONTENTS
Foreword vii
Preface ix
List of Contributors xi
4 Gross Dissection of Liver for Hepatocellular Carcinoma
Using AJCC Cancer Staging Manual 8th Edition:
Anatomical and Practical Considerations 77
Marla Beach, Laura Henao Caviedes, Consolato M. Sergi
6
Achieving a Cure: The Next Frontier in Hepatitis B
Treatment 109
Tina Boortalary, Brianna Shinn, Dina Halegoua-DeMarzio,
Hie-Won Hann
vi Contents
7 Locoregional Therapies for Bridging and Downstaging
Hepatocellular Carcinoma Prior to Liver Transplant 127
Sandeep A. Ponniah, Andreas G. Zori, Roniel Cabrera
8 Hepatoblastoma 145
Josef Hager, Consolato M. Sergi
Index 179
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.frontmatter
Foreword vii
FOREWORD
Investigating liver tumors in children and adults is a task for the intrepid,
undaunted, and naturally curious scientist. As I count myself among those who
wade in these treacherous and thoroughly stimulating waters, I understand the
value and rare nature of a book that successfully explores both widely established
general concepts and new discoveries with ease. Liver Cancer achieves these goals,
and I am delighted to be part of the effort to bring it to all of you.
Liver tumors are a heterogeneous and complex mix of benign and malignant
neoplasms that may arise in the setting of chronic liver injury or due to no prior
insult. In children, hepatoblastoma is the most common malignant primary liver
tumor and hepatocellular carcinoma is rare. In adults, however, hepatocellular
carcinoma is most common and undifferentiated embryonal sarcoma is vanish-
ingly rare. Liver Cancer explores these, and the myriad of other entities in between,
with a depth and precision that is highly informative and practical to the modern
physician scientist. Descriptions of grossing techniques, histopathologic features,
ancillary testing modalities, molecular/genetic abnormalities, imaging characteris-
tics, treatment options, clinical signs/symptoms and surgical approaches are con-
temporary contributions to this exciting field.
Liver Cancer represents the latest knowledge of primary liver tumors.
Refreshingly, it focuses on tumors and underlying processes that affect both chil-
dren and adults. This is uncommon in recent books and, therefore, places Liver
Cancer among the best in its field. Written by and for practicing Pathologists,
Oncologists, Surgeons, Gastroenterologists and Radiologists, its comprehensive
content makes it THE go to reference for primary liver tumors. It is easy to read
format also makes it a valuable tool for teaching clinical trainees, medical students
and graduate students.
It has been my pleasure to preview this important book and help bring it forth
to readers. The authors and editors should be commended for their tremendous
work. It is my hope readers will include this book as an important reference on
their bookshelves and consult it often. I know I will.
Florette K. Hazard, MD
Medical Director and Chief of Pathology,
Lucile Packard Children’s Hospital Stanford
Director of Pediatric Pathology,
Stanford University School of Medicine
Stanford, CA, USA
March 2021
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.fr
Preface ix
PREFACE
Liver cancers are complex neoplastic diseases with varied etiologies. Despite
numerous research programs and clinical trials targeting several molecules in the
last 50 years, hepatocellular carcinoma, the most common form of liver cancer, is
still difficult to cure, and has become the second most common cause of cancer-
related deaths in adults. This, at least in part, is due to the rising incidence of
obesity, non-alcoholic fatty liver diseases, and excessive alcohol consumption. An
adequate knowledge of the disease, including its etiology and pathology, is essen-
tial to develop effective therapies. Contributed by some of the leading hepatolo-
gists and pathologists in the field, this book is an effort to provide students, basic
scientists, clinicians, and pathologists with a comprehensive understanding of
the pathology, diagnosis, treatment, and management of various types of liver
cancers. There are nine chapters in the book.
Chapter 1 provides a comprehensive review of malignant epithelial tumors of
the liver in children and adolescents. Hepatocellular carcinoma, lipid-rich hepa-
tocellular carcinoma, fibrolamellar carcinoma, and cholangiocellular carcinoma
are discussed. This is followed by an in-depth discussion of hepatocellular carci-
noma in adults in Chapter 2, covering the etiology and pathogenesis of hepatocel-
lular carcinoma, various diagnostics tests, and treatment options.
While the etiology of liver cancers is multifactorial, obesity and non-alcoholic
fatty liver disease are emerging as significant risk factors for liver cancer. Chapter 3
highlights our current knowledge on the progression of non-alcoholic fatty liver
disease to malignancy. It gives insight into the diagnosis, treatment options, and
future directions for non-alcoholic steatohepatitis-related tumorigenesis. A com-
prehensive understanding of the functional liver segments and anatomy of resec-
tion samples is essential to generate clinically relevant pathology reports. Based on
the College of American Pathologists Cancer Protocols, Chapter 4 guides the gross
dissection procedure for producing a valuable pathology report. Radiology plays
a vital role in the screening, diagnosis, and treatment of liver lesions. Pathology
provides further diagnostic insight into material obtained through biopsy or surgi-
cal resection. Gross appearances guide sampling approaches, and microscopic
examination can lead to a final diagnosis. Chapter 5 introduces the reader to the
radiologic and histopathologic features of common tumor or tumor-like lesions
within the liver, emphasizing hepatocellular lesions, biliary lesions, vascular
mesenchymal lesions, and metastatic malignancies.
Chapters 6 and 7 focus on therapy and management. Although antiviral thera-
pies have reduced the incidence of hepatitis B virus-related hepatocellular carci-
noma, patients are still at risk of developing hepatocellular carcinoma. Chapter 6
discusses the immunopathogenesis of the virus, the effectiveness of nucleos(t)ide
analogues, and recent therapeutic developments. Hepatocellular carcinoma is a
common indication for liver transplantation. While awaiting liver transplantation,
locoregional therapy can be used as bridging therapy to maintain the tumor
burden. Chapter 7 provides a comprehensive review of various locoregional
therapies, including trans-arterial chemoembolization, transcatheter arterial
radioembolization, thermal ablation, and stereotactic body radiation therapy.
x Preface
Chapter 8 and 9 deal with two relatively rare entities: hepatoblastoma and
undifferentiated embryonal sarcoma of the liver. Hepatoblastoma is the most
common liver cancer in children aged three years and younger. Chapter 8 pro-
vides an overview of the etiology, pathogenesis, epidemiology, incidence, symp-
toms, and therapeutic considerations of hepatoblastoma. The diagnostic measures
necessary from a surgical point of view and the essential operational and technical
considerations for the various hepatoblastoma stages are discussed. Undifferentiated
embryonal sarcoma of the liver is an aggressive malignancy that most commonly
affects the pediatric age group. This tumor very rarely occurs in adults. Chapter 9
outlines the clinical, radiological, and pathological features of hepatic undifferen-
tiated embryonal sarcoma in adults. An in-depth discussion is undertaken to
describe the diagnostic approach and the differential diagnosis for this rare and
challenging tumor.
I thank the authors for their dedication and professionalism in contributing to
this book. I believe this book will encourage readers to delve deeper into this field
and take up the critical challenge of working toward effective treatments for liver
cancers.
Consolato M. Sergi, MD, PhD, MPH, FRCPC, FCAP
University of Alberta
Edmonton, Canada
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.pr
List of Contributors xi
LIST OF CONTRIBUTORS
ANDREAS G. ZORI, MD
Division of Gastroenterology, Hepatology, and Nutrition, University of Florida,
Gainesville, FL, USA
BRIANNA SHINN, MD
Division of Gastroenterology and Hepatology, Department of Medicine,
Thomas Jefferson University Hospital, Philadelphia, PA, USA
DINA HALEGOUA-DEMARZIO, MD
Division of Gastroenterology and Hepatology, Department of Medicine,
Thomas Jefferson University Hospital, Philadelphia, PA, USA
ERIC LACHANCE, MD
Department of Radiology and Diagnostic Imaging, University of Alberta,
Edmonton, Alberta, Canada
JAKE MANDZIUK, MD
Department of Laboratory Medicine and Pathology, University of Alberta,
Edmonton, Alberta, Canada
JINGYANG HUANG, MD
Department of Pathology and Laboratory Medicine, University of Alberta,
Edmonton, Canada
JOSEPH I. SHAPIRO, MD
Marshall University Joan Edwards School of Medicine,
Huntington WV, USA
KOMAL SODHI, MD
Department of Surgery, Marshall University Joan Edwards School of Medicine,
Huntington WV, USA
List of Contributors xiii
SANDEEP A. PONNIAH, MD
Division of Gastroenterology, Hepatology, and Nutrition, University of Florida,
Gainesville, FL, USA
TINA BOORTALARY, MD
Department of Medicine, Thomas Jefferson University Hospital,
Philadelphia, PA, USA
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.cont
1
Carcinoma of the Liver in Children
and Adolescents
Consolato M. Sergi
Departments of Pediatrics, Laboratory Medicine and Pathology, Stollery Children’s
Hospital, University of Alberta, Edmonton, AB, Canada.
Author for Correspondence: Consolato M. Sergi, Departments of Pediatrics,
Laboratory Medicine and Pathology, Stollery Children’s Hospital, University of Alberta,
Edmonton, AB, Canada. Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch1
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
1
2 Sergi C M
INTRODUCTION
Liver tumors in children and adolescents are catastrophic events. While most
occur in otherwise healthy liver, they may be associated with congenital anomalies,
genetic syndromes or metabolic conditions. Two significant entities are hepato-
blastoma (HBL) and liver carcinoma/hepatocellular carcinoma (HCC). Both
neoplasms account for 0.5–1.5% of all childhood neoplasms (1) and 4% of all
pediatric orthotopic liver transplantations (OLT). HBL accounts for 67–80% of
all pediatric liver cancers worldwide, while 20–33% are HCC. Prevalence of HCC
is geographically and demographically dependent and is associated with the
prevalence of Hepatitis B in the population (1). Differential diagnosis can be
genuinely challenging considering the number of variants and subtypes that have
been delineated over the last decade. Known predisposing factors for HCC in
childhood and youth are alpha-1-antitrypsin deficiency, hereditary tyrosinemia,
genetic hemochromatosis, Wilson’s disease, acute intermittent porphyria,
progressive familial intrahepatic cholestasis type 2, mitochondrial ETC disorders,
glycogen storages diseases, and transaldolase deficiency. Several neoplastic
conditions must also be considered in the differential diagnosis of HCC, including
epithelioid hemangioendothelioma of the liver, hemangiosarcoma, malignant
rhabdoid tumor of the liver, undifferentiated embryonal sarcoma of the liver,
biliary tract rhabdomyosarcoma, and the unique variant of neuroblastoma IVS.
Some note should be given about fibrolamellar carcinomas of the liver, which are
tumors of adolescence with a predilection for the left liver and have a tendency to
be challenging to resect (2–6).
HEPATOCELLULAR CARCINOMA
HCC is the second most common primary liver malignancy in a pediatric setting
(27%) following HBL. Vascular tumors and sarcomas constitute the remaining
(1, 7). HCC is becoming a common indication for liver transplantation in child-
hood with an incidence rate of 0.3 to 0.45 individuals per million per year (23%).
HCC is diagnosed more commonly in adolescents (10–14 years) but has been
described in children age 5 and younger. Male children have a greater predisposi-
tion to HCC (3:1). HCC typically presents at a more advanced stage in children
than in adults. Overall, only 0.5–1% of all HCCs occur in childhood. HCC has a
broad etiologic spectrum including alcoholic cirrhosis, hepatitis B virus (HBV)
infection, hepatitis C virus (HCV) infection and ingestion of aflatoxins from
Aspergillus flavum in contaminated food, but these factors show a geographic vari-
ation. HCC is most common in central Africa (sub-Saharan regions) and southeast
Asia, with an incidence rate of up to 1 per 1,000, most commonly as a result of
HBV infection. HCC remains a common diagnosis in areas with high endemic
HBV infection rates. There are rates of up to 100% positivity for HBV infection in
Taiwan, and 64% positivity for HBV infection in Hong Kong SAR (Special
Administrative Region of the People’s Republic of China) (8, 9). The integration of
the HBV genome into the neoplasm genome can be detected by immunohisto-
chemistry and demonstrated at the molecular level, but this event is not necessar-
ily oncogenic; alone it does not result in liver carcinogenesis. A secondary
Liver Cancer in Children and Adolescents 3
Biliary atresia
Biliary atresia (BA) is characterized by a necro-inflammatory process with destruc-
tive cholangitis (15, 16). The prevalence rate of HCC in BA is 1.3% (five out of
387 patients) with all but one patient below five years of age. This figure is par-
ticularly alarming in the West where rates of BA are up to 30%, although it has
been argued that the said figure may be confounded by surveillance bias (1).
tumor masses, but resection and shunt occlusion, as well as OLT, are necessary for
malignant tumors (20).
Budd-Chiari syndrome
Budd-Chiari syndrome (BCS) is characterized by abdominal pain, ascites, and
liver enlargement caused by blockage of the hepatic veins that drain the liver. BCS
can be primary (75%) (polycythemia rubra vera, pregnancy, postpartum state, use
of oral contraceptives, paroxysmal nocturnal hemoglobinuria, and lupus antico-
agulants) or secondary (25%) (compression of the hepatic vein by an outside
structure [for example, a neoplasm]). HCC is a known sequela of BCS in adults,
with a 5-year cumulative incidence of 4% (1). Nodular regenerative hyperplasia is
more commonly associated with the onset of BCS, and is more often associated
with male gender, FVL mutation, and inferior vena cava (IVC) obstruction,
but can also be seen in tuberculosis, congenital venous webs, and IVC stenosis
(25, 26).
Alagille syndrome
Alagille is a multisystem genetic disorder characterized by interlobular bile ductal
paucity and chronic hepatocellular cholestasis (27). Alagille syndrome is a result
of mutations on one of the two genes: JAG-1 in 90% of cases, and NOTCH2 in
about 1% of cases. Liver carcinogenesis in Alagille syndrome has been occasion-
ally reported (28–32). In the first two decades of life, patients can develop HCC
in conjunction with Alagille syndrome and cirrhosis (32). Upregulation of Notch2
in hepatocytes results in proliferation of hepatocyte and biliary epithelial cells; it
also triggers diethylnitrosamine induced HCC formation in mice (33). Neoplastic
cells commonly lose contact inhibition to undergo anchorage-independent prolif-
eration. They become resistant to programmed cell death by inactivating the
Hippo signaling pathway. It results in activation of the transcriptional co-activator
yes-associated protein (YAP). The transcriptional regulator YAP upregulates
JAGGED-1 and activates NOTCH pathways in mouse models and humans with
HCC and colorectal and pancreatic carcinomas (34). YAP is a transcriptional co-
activator and is responsible for the rapid proliferation of neoplastic cells becoming
apoptosis-resistant.
Autoimmune hepatitis
Autoimmune hepatitis (AIH) is a non-metabolic disorder that predisposes to
HCC (35). The risk of HCC in adults with AIH is 3.06 per 1,000. Absolute risk of
HCC as a sequela of AIH is slightly lower than the risk of HCC attributable to
other causes of cirrhosis in adults like hepatitis B, C, or primary biliary cholangitis
(PBC) (36). The risk of HCC is related to the extent of cirrhosis at the time of clini-
cal diagnosis of AIH (OR = 4.08). Abnormal liver enzymes (transaminases) are
seen on final observation (OR = 3.66) (36, 37). Although the incidence of HCC in
children is rare, AIH remains a common pediatric liver condition. Non-alcoholic
fatty liver disease (NAFLD), type 2 diabetes mellitus, genetic hemochromatosis,
and porphyria (acute intermittent and porphyria cutanea tarda) carry a high risk
Liver Cancer in Children and Adolescents 5
of development of HCC in adults, but not in children (38). About 33–50% of all
HCC cases in childhood are sporadic (1).
Symptoms
Symptoms of HCC in noncirrhotic individuals are similar to those seen in patients
harboring other liver tumors. The “tumor quia tumet” or mass-effect includes an
abdominal mass, pain, and jaundice. Abnormal liver biochemistry alongside spe-
cific clinical signs and symptoms may be present if the tumor originates as a result
of liver disease. Clinical signs may include splenomegaly due to portal hyperten-
sion and spider nevi, among others. There are no specific findings on full blood
count associated with HCC. Liver function tests frequently give abnormal results
particularly in children in whom HCC has occurred with frank cirrhosis. Alpha-
fetoprotein (AFP) is increased in 50–70% of patients with HCC, although it
should be kept in mind that persistent elevations of AFP are found in hepatic
regeneration. On the other hand, AFP levels greater than 400 to 500 ng/mL in a
patient with liver cirrhosis is strongly suggestive of HCC. HCC should be sus-
pected when AFP levels fall between 200 and 300 ng/mL. AFP has prognostic
value and has been used for years as a prognostic (relapse) indicator after chemo-
therapy. Elevations of the vitamin-B12-binding protein transcobalamin I is a useful
indicator for the fibrolamellar variant of HCC or fibrolamellar carcinoma (FLC)
(vide infra). Finally, the exclusion of known risk factors, such as HBV/HCV
serology, plasma and urine amino acid, urinary succinylacetone for hereditary
tyrosinemia, biochemical serum level, and isoelectricfocusing-based phenotype
for AAT should be routinely performed. If there is a suspicion of a genetic disease,
a genetic confirmation of PFIC 2, hereditary tyrosinemia, and Alagille syndrome
should be carried out as soon as possible.
Imaging
Imaging is useful to determine the site and characteristics of the tumor, to estab-
lish the presence of tumor metastases, and to help the surgeon to assess the suit-
ability for prompt resection. HCC is difficult to distinguish from HBL on imagery.
Both HCC and HBL are typically large and multifocal. In both tumors, there may
be evidence of venous invasion, tissue calcification, and lung metastases.
Ultrasound of HCC shows a large, heterogeneous, generally hyperechoic vascular
mass. Computed tomography (CT) provides detailed information on the anatomic
boundaries of liver tumors, which is essential for the pediatric surgeon. MRI scan-
ning gives information about tumor infiltration into surrounding tissue, enabling
precise evaluation of segmental involvement. MRI findings of HCC show a hetero-
geneous, mostly hypointense neoplasm on T1-weighted images. MRI findings on
T2-weighted images of HCC is mildly hyperintense in comparison with healthy
liver. Contrast-enhanced T1-weighted images identify a pattern like CT. There is
early arterial enhancement and reduced signal intensity in the portal venous
phase. Imaging data is essential for clinical staging and surgical intervention. PET
scans show areas of high metabolism, which is useful in localizing early metasta-
ses or recurrence of the neoplastic disease. In all children without cirrhosis, unless
primary surgery is feasible, a biopsy is necessary. Image-guided needle biopsy is
6 Sergi C M
usually favored to open biopsy due to the multifocal nature of the hepatocellular
neoplasm. To avoid tumor seeding in children, the tumor should be approached
along a short route through unaffected liver, avoiding Coinaud liver segments that
will not be removed at subsequent surgery. Moreover, a coaxial biopsy should be
used to obtain several cores of tissue with a single shot. The needle track should
be plugged, either with gelatin foam or with a slurry of collagen. HCC in adults
has been studied in greater detail than pediatric HCC in terms of cytogenetic and
molecular characteristics. Future studies aim to outline molecular pathology of
HCC in children (39).
Morphology
Morphology of adult and pediatric HCC are similar. There are several growth
patterns including trabecular (plate-like), solid/compact, pseudoglandular/acinar,
and scirrhous (scleroting architectural pattern). Cytologic variants include
pleomorphic cells, clear cells, and sarcomatous cells (spindle cells or bizarre giant
cells). The cytoplasm of the tumor cells is stained with Hep Par 1, a monoclonal
antibody (clone OCH1E5.2.10) in a granular manner, without canalicular or
zonal accentuation. The antigen detected by Hep Par 1 appears specifically associ-
ated with the mitochondrial membrane of hepatocytes. The use of antigen retrieval
(high pH buffer) and a detection kit which eliminates non-specific biotin reactiv-
ity allows the pathologist to use Hep Par 1, which is highly effective on formalin-
fixed, paraffin embedded tissues. Cautious use of Hep Par 1 antibody in a panel
with other positive (AFP, carcinoembryonic antigen with a polyclonal antibody,
CD10) as well as negative (epithelial membrane antigen, carcinoembryonic anti-
gen with a monoclonal antibody, CD15) markers of hepatocellular differentiation
may assist in accurate diagnosis of hepatocellular carcinoma (Figure 1). Epithelial
cell adhesion molecule (EPCAM) expression seems to be far more robust in
pediatric than adult HCC (7, 40). In the future, transcriptomics studies may reveal
additional data comparing adult and pediatric HCC.
Differential diagnosis
Distinguishing HBL from HCC may be straightforward in a few cases. HBL may
be easily recognized if both epithelial and mesenchymal components are present,
but when it shows a macrotrabecular architecture with well-differentiated fetal
epithelial cells and no mesenchymal elements, distinction may be practically
impossible histologically (7, 41). In this scenario, liaison with the clinician is
critical. The pathologist should consider that HBL occurs more frequently than
HCC in patients aged 5 or younger, very-low-birth-weight infants and premature
infants, and with a multiple congenital anomaly syndrome, such as Beckwith-
Wiedemann syndrome. On the other hand, the presence of underlying hepatic
disease, especially with a known risk factor, such as hereditary tyrosinemia,
favors a diagnosis of HCC over HBL. In the most recent pediatric liver tumor
consensus classification, the category of “hepatocellular neoplasm not otherwise
specified” has been introduced to acknowledge the challenge of differentiating
sporadic cases which show cells that are “transitional” between hepatoblasts and
hepatocytes (39, 42).
Liver Cancer in Children and Adolescents 7
Staging
Staging pediatric liver neoplasms is crucial, but the reality is that there are
no uniformly and broadly accepted staging systems for the pediatric HCC (7).
In adulthood, the Barcelona Clinic Liver Cancer score is probably the most widely
accepted system, because it assesses the state of the tumor, the state of the patient,
and the condition of the liver. Use of the tumor node metastasis (TNM) staging
system is controversial. TNM does not seem to be well-suited for predicting prog-
nosis in either pediatric HCC or adult HCC (43). Other staging systems include
the Cancer of the Liver Italian Program (CLIP) scale and the Okuda system, among
others (43).
Once a diagnosis is reached, in the United States of America and Canada, the
Children’s Oncology Group (COG) provides vital information to children and
their families. This support continues throughout treatment and carries on
after cure. The COG’s adoption of a system based mostly on surgical findings in
the late 1980s has been particularly important for the treatment of HCC. In the
late 80’s, the PRETEXT (PRETreatment EXTent) system was developed by the
International Society of Pediatric Oncology (SIOP) Liver Tumor Study (SIOPEL)
Group. The PRETEXT was revised in 2005, and again in 2017 (7, 44). This sys-
tem divides children with liver neoplasms into four categories consistent with the
amount of liver segments not involved by the tumor (7, 45). A separate classifica-
tion includes the involvement of the portal and hepatic veins and IVC as well as
metastatic disease. The interobserver reliability of the system remains neverthe-
less quite high. The significant advantage of PRETEXT staging is that it allows
stratifying of patients to determine the most effective treatment. Involvement of
hepatoduodenal lymph nodes has a role in staging. It is important therefore to
take samples from lymph nodes associated with the hepatoduodenal ligament at
the time of surgical resection. After resection of the tumor, the 5-year survival rate
(5-YSR) ranges between 35and 50%, while the 5-YRR (5-years recurrence rate) is
20–30% (7).
ultrasound examination, which must correlate with the volume of the liver that
needs to be removed using CT- or MRI-based calculations (46). In pediatric HCC,
the usual limit of resection is calculated by dividing the remnant liver volume
(milliliters) by the patient’s body weight (kilograms). This value can safely exceed
the usual 0.6–0.8 mL/kg. Orthotopic liver transplantations (OLT) as a treatment
for HCC is still controversial, although the criteria for OLT in adults has been
modified over time from unrestricted tumor limits to precise tumor burden which
meets the conventional Milan criteria (CMC). The CMC-based safe limits for OLT
are either a single tumor smaller than 5 cm in size, or in case of multiple tumors,
no more than 3, and each not exceeding 3 cm in diameter. Satisfying this criterion
allows a 5-YSR of about 70%. Unfortunately, there are no randomized prospective
studies comparing liver resection with OLT as treatments for childhood HCC,
although we must acknowledge that experience with OLT in pediatric HCC is
limited (47). A low 5-YSR has been observed in earlier reports (48, 49) with val-
ues ranging between 29–35% in children with unresectable neoplasms. More
recent reports from Pittsburgh and other centers show an improvement in 5-YSR
rates, with values of up to 60% (50). Contraindications for OLT include extrahe-
patic disease and FLC. It has been proposed that decisions on which treatment is
most appropriate should be made on a case-by-case basis.
Ablative therapies include radiofrequency ablation and percutaneous ethanol
injection (51), although these techniques have been explored mostly for adult
HCC. Ablative therapy is comparable to surgery for tumor size equal to 3 cm or
below, but its effect on HCC up to 5 cm or more is not confirmed to be better than
tumor removal. Usefulness of ablative therapy for treatment of HCC of a size
greater than 5 cm seems to be uncertain.
Transfemoral hepatic artery chemoembolization (TACE) is an option for the
management of adult HCC with improved 5-YSR in randomized controlled trials
(52). TACE indications for pediatric HCC is similar to adult HCC; it may be con-
sidered a bridge to OLT or resection with attempted conversion of tumors from
nonresectable to resectable. Advantages of TACE include the delivery of high con-
centrations of cytotoxic drugs to the tumor center, prolonged intratumoral dwell
time of drug, and reduced systemic toxicity. TACE appears to be a safe and effec-
tive method in children with unresectable hepatocellular neoplasm and is useful
for handling hepatic tumor burden, downstaging, and bridging to OLT (53–55).
Occasionally, pulmonary embolism and thrombosis of some branches of the
hepatic artery have been recorded as severe complications. Despite widespread
use of neoadjuvant and adjuvant chemotherapy, there is little evidence to suggest
that this translates to a benefit in survival as shown in the North American
Intergroup Hepatoma study (INT-0098) and the International and in the
Childhood Liver Tumor Study Group (SIOPEL) study (7, 44). Nevertheless, adju-
vant chemotherapy may be of benefit for children with completely resected HCC.
The outcome was uniformly poor for patients with advanced-stage disease. These
patients showed a 5-year event-free survival (5-YEFS) with stage III and IV HCC
of 23% and 10% respectively. The resection of the neoplasm after neoadjuvant
chemotherapy was practicable in two patients only. Similarly, the SIOPEL identi-
fied a 5-YEFS of 17% (1). In the three cooperative studies of the German Society
for Pediatric Oncology and Hematology (GPOH), HCC has been targeted with the
same regimens as HBL, with surgical resection highly recommended as the pri-
mary treatment in all patients (7, 56).
10 Sergi C M
Treatment-response follow-up
Repeat CT scans or ultrasonography are useful to observe response to treatment.
Imaging can document shrinkage of the liver cancer and chest radiography or CT
scans can monitor potential progress of pulmonary metastases. Also, the presence
of AFP levels alongside the lack of radiological disease is highly suspicious of
minimal residual disease and more sophisticated investigation is mandatory. An
increase in AFP levels after initiation of chemotherapy is usually a sensitive marker
of treatment failure or relapse. Transcobalamin I has been used as a biomarker of
fibrolamellar carcinoma of the liver or fibrolamellar variant of HCC.
Long-term outcomes are dependent on demographics and treatment type.
Survival of children aged 0–4 years is better than that of children aged five years
and older, with a 5-YSR of 53% (unadjusted) compared with 32% for children
aged 5–19 years. Prognosis is better in male children when compared with female
children. Some ethnic evaluation studies identified that Asian children have rela-
tively low survival (13%), compared with white and black children (survival of
33% and 46% respectively) (7). On the other hand, results of Surveillance,
Epidemiology and End Results (SEER) database are puzzling. A large series of 218
pediatric patients with HCC seem to show no significant difference in the overall
survival by age at diagnosis, gender, race/ethnicity, or use of radiation (79–81).
Pediatric excisable HCC seems to bear a better prognosis irrespective of histologic
subtype. These patients seem to do worse than those with HBL (48). Analysis of
post-OLT showed that 1-, 5-, and 10-YSR were 86%, 63%, and 58%, respectively.
The primary cause of death of these patients was metastatic or recurrent disease,
responsible for 86% of deaths in children with HCC (49).
The most common histological patterns of HCC are solid, trabecular, pseudoglan-
dular, and scirrhous, with three recognized cytologic variants: pleomorphic, clear
cell, and sarcomatoid. In the clear cell variant, cytoplasmic changes are related to
glycogen and/or lipid accumulation (lipid-rich HCC, LR-HCC) within the tumor
cells. In cytologic samples of low-grade lipid-containing hepatic neoplasms, dif-
ferentiation of neoplastic cells from benign steatotic cells can be extremely chal-
lenging (92–94). Fine-needle aspirates of benign lesions contain rigid cores and
large tissue fragments, while LR-HCC shows a breakdown of the reticulin frame-
work (visualized with a routine special stain) with scattered cells and a fine gran-
ular-appearing background. LR-HCC is an uncommon HCC variant and needs to
be taken into consideration, particularly now that non-alcoholic steatohepatitis
and fatty cell change are more common in adolescence and youth (95–98).
FIBROLAMELLAR CARCINOMA
FLC or fibrolamellar variant of HCC is a rare variant of HCC, and recent investiga-
tions have identified a specific molecular signature (99–103). This neoplasm
affects adolescents and youth with no significant gender prevalence primarily.
FLC chiefly affects Caucasians. Unlike HCC, FLC is never associated with a
14 Sergi C M
Presentation
Clinically, abdominal pain, hepatomegaly, obstructive jaundice, ascites, systemic
manifestations (for example, fever or sense of “fullness”), and rarely gynecomastia
and BCS have been observed in patients harboring an FLC (100). Laboratory val-
ues that need to be taken into consideration are neurotensin, vitamin B12 binding
capacity (transcobalamin), and des-gamma-carboxyprothrombin (104). Serum
AFP is increased in just 10% of FLC cases, unlike with HCC where AFP is elevated
in 60% of cases. X-ray imaging shows a central scar which resembles the central
scar observed in FNH, a benign lesion with no correlation with FLC. The central
scar of FLC is often calcified, which is unusual with FNH.
Grossly, FLC is typically larger than conventional HCC. FLC has an odd
propensity to metastasize to regional lymph nodes. Thus, the pediatric surgeon
should attempt to sample the regional nodes. FLC generally appears as well-
circumscribed, single white-brown hard mass that shows fibrous bands, similar to
FNH. It usually involves the left lobe of the liver, but it has been reported in both
lobes.
Pathology
Microscopically, the tumor is made of large polygonal cells with copious eosino-
philic cytoplasm (about 50 microns, that is, approximately double the size of a
normal human hepatocyte, which is 20–30 microns) with granular character and
large nuclei containing prominent nucleoli. At low magnification, thick fibrous
collagen bands are seen transecting the tumor (Figure 2A). They may partially
encircle or surround the tumor cells. On histology, pale cytoplasmic bodies (pale
bodies) or ground glass cells and PAS-positive hyaline globules and copper depo-
sitions are also seen (Figure 2B). Bile is observed using negative Prussian Perl’s
blue iron stain. In the differential diagnosis of FLC, sclerotic variant of HCC
should be mentioned, which rarely harbors the parallel (or almost parallel) run-
ning collagen bands. Vascular invasion and necrosis may be seen. Radiological
calcification may also be confirmed on histology.
Liver Cancer in Children and Adolescents 15
Figure 2. Fibrolamellar carcinoma. A. Tumor tissue showing extensive intratumoral lamellar
bands of collagen (blue) separating large polygonal cells (Masson’s trichromic stain, scale bar:
50 µm). B. Tumor tissue showing large polygonal cells with vesiculated nuclei, large nucleoli
and intracytoplasmic pale bodies (Hematoxylin and Eosin staining, scale bar: 100 µm).
16 Sergi C M
A controversial issue is the “mixed” FLC and HCC, but molecular pathology
may resolve this dilemma in the future. Additional features that can be encoun-
tered are trabecular, adenoid or pelioid patterns that need to be taken into
consideration when evaluating needle or open biopsy. Metastases to the regional
lymph nodes, peritoneum, and lungs are commonly seen. Immunohistochemistry
is an important ancillary tool. FLC shows HepPar-1, keratin 7, epithelial mem-
brane antigen (EMA), CD68 and carcinoembryonic antigen (polyclonal anti-
body, p-CEA).
Immunohistochemically, there is usually a lack of expression of AFP, synapto-
physin, and chromogranin. The p-CEA is shown in a canalicular pattern due to
cross-reactivity to biliary glycoprotein I existing in bile canaliculi of healthy liver
and HCC. Of note, the monoclonal CEA is usually negative. These findings have
suggested to some authors that FLC may be a hybrid neoplasm of hepatobiliary
type. FLC may also be positive for antibodies against CAM5.2 (a low molecular
weight cytokeratin panel against keratin 7 and keratin 8 present on secretory epi-
thelia but not on stratified squamous epithelia), AE1–3 (a pan-cytokeratin mark-
ers grouping low and high molecular weight antibodies), and neurotensin. AAT is
usually inconsistently expressed. CK19 is usually negative (105). There may be an
expression of epithelial growth factor receptor (EGFR), and human epidermal
growth factor receptor (Her-2) (106). Electron microscopy reveals copious mito-
chondria (107). Pale bodies contain fibrinogen. There is an abundance of rough
endoplasmic reticulum (ER) in the neoplastic cells, which may also show dense
core neuroendocrine-like granules. However, the neoplastic cells are not positive
for neuroendocrine markers by immunohistochemistry (vide supra) or immuno-
gold-marked electron microscopy. Differential diagnoses include sclerosis-
accompanied adenosquamous carcinoma, cholangiocellular carcinoma, which is
markedly glandular and mucin-positive (108), and FNH. FNH is smaller in size
than FLC. FNH contains bile ductular proliferating associated stroma (fibrous)
and inflammatory cells. Microscopically, FNH has neither gross bile staining nor
hepatocyte atypia. Also, FLC must be differentiated from the sclerosing variant of
HCC, which shows no oncocytic change of the neoplastic cells. This tumor is
smaller in size than the tumor cells of FLC and shows a typical pseudoglandular
pattern. Other differential diagnoses include metastatic carcinoma with sclerotic
stroma (a full history is invaluable), neuroendocrine tumors (neuroendocrine
markers proof by immunohistochemistry is useful), and paraganglioma, which
harbors a Zellballen growth pattern, round/ovoid nuclei without atypia, vascular
stroma but no dense fibrosis and is positive for neuroendocrine markers (109).
A clear cell variant of FLC has also been described. (110). In addition to classic
FLC, the neoplastic clear cells with empty cytoplasm have ballooning and rarefac-
tive changes to their mitochondria (100).
A fusion gene between DNAJB1 and PRKACA characteristically distinguishes
FLC from conventional HCC (103). This genetic abnormality is found with loss
of genetic material on chromosome 19. It leads to the unusual combination of
two genes, DNAJB1 and PRKACA. DNAJB1 is a DnaJ (Hsp40) homolog, subfam-
ily B, member 1 (DNAJB1) or a transcriptional target of forkhead box protein E3
(FOXE3) in a pathway apparently crucial for the development of the eye.
PRKACA encodes one of the catalytic subunits of protein kinase A. The “chimeric”
gene, which in turn produces an abnormal protein product, is an unusual com-
bination of the two normal proteins. Common pathways that are altered in HCC,
Liver Cancer in Children and Adolescents 17
such as β-catenin and p53, are not differentially controlled in FLC. Occasionally,
there is also an upregulation of other pathways including EGFR, MAPK, PI3K,
and RAS (111). In 2015, Riehle et al. highlighted some of these pathways, show-
ing that mTORC1 is activated in FLC and has been found in association with
fibroblast growth factor receptor 1 (FGFR1) overexpression. This data supports
the use of FGFR1 inhibitors in patients with FLC (112). Recently, data from the
Fibrolamellar Carcinoma Consortium has identified and updated overall survival
of FLC (104). About 3/4 of patients with FLC undergo surgical resection and/or
OLT and 1/3 receive perioperative therapy. Preoperative chemotherapy, includ-
ing external beam radiation, and TACE with doxorubicin are the milestones of
the perioperative therapy. Chemotherapy agents used in FLC include cisplatin,
doxorubicin, fluoropyrimidines, gemcitabine, irinotecan, and oxaliplatin.
Postoperative adjuvant chemotherapy and/or radiation should also be consid-
ered as relevant options (104).
This section lists metabolic conditions that may predispose to the development of
HCC. Liver cirrhosis has been considered a precursor of HCC irrespective of etiol-
ogy because dysplasia can occur in the setting of a regenerating nodule, but this
argument is mainly relevant for adult HCC. In childhood, pediatric metabolic
conditions play a major role. About one-third of pediatric cases of HCC are associ-
ated with cirrhosis, while liver cirrhosis is present in two-thirds to 90% of adult-
onset HCC. The list of pediatric metabolic conditions may be not complete but
illustrates most of these conditions. This list includes alpha-1-antitrypsin (AAT)
deficiency, hereditary tyrosinemia 1, glycogen storage diseases types I, III, and IV,
genetic hemochromatosis, Wilson’s disease, acute intermittent porphyria, progres-
sive familial intrahepatic cholestasis type 2, mitochondrial electron transport
chain (ETC) disorders, and transaldolase deficiency. However, in some of the
above conditions, HCC does not occur at the pediatric age. The most prominent
disease in this category of predisposing conditions to HCC is AATD.
Alpha-1-antitrypsin deficiency
AATD (Laurell-Eriksson-Syndrome) is the result of a gene mutation, commonly
found in Caucasians. AAT expression is regulated by the SERPINA1 (SERine
Proteinase INhibitor) gene which expresses codominant alleles. The SERPINA1
gene is situated on the protease inhibitor (Pi) locus. It is on the long arm of chro-
mosome 14 (14q31–32). The three most common alleles are the normal M allele,
the S allele and the Z allele. In the setting of the S allele, AAT plasma levels are
about 60% of normal, while in the setting of the Z allele AAT plasma levels are
about 10–15% (113, 114). The nomenclature of SERPINA1 alleles is based on
isoelectric focusing-determined migration of the protein variants that were identi-
fied long before the gene was known. The coding of the alleles was based on
migration in gel electrophoresis using the prefix PI (protease inhibitor) serving as
18 Sergi C M
an alias for the gene (115). The normal AAT protein (the M protein) migrates to
the middle of the isoelectric field, while the abnormal AATD protein (the Z vari-
ant) migrates slowly. Null alleles are pathogenic alleles that result in either no
mRNA product or no protein product. Homozygosity for the Z mutant allele is
accompanied by abnormal retention of altered AAT molecules in the liver, which
may lead to neonatal hepatitis, liver disease in children, and liver disease with the
development of HCC in young adults (116). AAT is an acute-phase protein with
strong inhibitory activity towards proteolytic enzymes, mainly elastase but also
trypsin, chymotrypsin, and thrombin. Lack of AAT is also associated with higher
risk of chronic obstructive pulmonary disease (COPD), which is characterized
clinically by lung emphysema, chronic bronchitis and/or persistent airflow
obstruction in young and middle-aged adults, especially smokers. Adult-onset
AATD-determined liver disease manifests with fibrosis and cirrhosis. It may occur
in the absence of a history of childhood (or neonatal) liver disease. The risk for
HCC is increased in individuals with AATD. The hepatocytes are the main source
of AAT.
The SERPINA1 gene is closely controlled by different cytokines, including
interleukin-1 (IL-1), tumor necrosis factor α (TNFα), and the interleukin-6 family
of cytokines (interleukin-6, leukemia inhibitory factor, oncostatin M). Genetic
mutations of SERPINA1 cause disturbance in protein structure, resulting in
polymerization and intracellular accumulation of the protein. The point muta-
tions accountable for the molecular instability of the protein occur in the axes and
sliding regions of the protein. They are located in the areas involved in the move-
ment of the reactive loop (117). These mutations allow for the spontaneous open-
ing of the main β-pleated sheet of the molecule. It leads to a swift insertion into
the sheet of the reactive loop of the subsequent AAT molecule. As a result, loop-
sheet polymers are formed. Individuals who are either homozygous for most
common mutated variant (– Z), or two other rare variants (Siiyama and M-Malton/
M-Cagliari), show protein aggregation (113, 114, 118). The histological hallmark
of liver disease in AATD is the identification of AAT-containing globules which are
positive to diastase-resistant PAS-D staining in periportal hepatocytes. The link
between AAT and liver malignancy has been explored from several aspects, for
example, AAT polymorphisms, elevated serum levels and changes in the content
of oligosaccharides of AAT. The impossibility to extrude AAT from the ER leads to
the accumulation of polymers in the ER of hepatocytes, leading to damage of the
hepatocytes by the gain-of-function mechanism. This mechanism, by which accu-
mulation of protein damages the cell, is crucial. There is evidence from investiga-
tions using mice transgenic for the Z mutant of the human AAT (AAT-Z) gene in
which hepatitis, and then HCC, developed (119). Marcus et al. recognized an
AAT-Z altered regulation of several genes, including cyclin D1 and MCAM, both
of which led to cell proliferation and tumorigenesis (120). Genetically engineered
fibroblast cell lines arising from the skin of individuals with PiZZ phenotype with
or without liver disease indicate that there is a hold-up in ER degradation of
A1AT-Z (121). The key to liver carcinogenesis seems to reside in inefficient degra-
dation of A1AT-Z in the ER. Variation in the function of mechanisms of intracel-
lular protein degradation and/or variation in the pathways of signal transduction
activated to protect the cell from protein mis-localization and/or aggregation seem
to result in abnormal cell metabolism of hepatocytes, predisposing to liver carci-
nogenesis (122). It is known that terminally misfolded proteins are transported in
Liver Cancer in Children and Adolescents 19
a selective manner from the ER into the cytosol. These molecules are subsequently
ubiquitinated and degraded by the proteasome in a process called ER-associated
degradation (ERAD). Lack of efficient protein degradation or accumulation of
misfolded proteins in the ER overwhelms the degradation machinery of the cell,
and several ER response pathways are activated. These pathways include the pro-
teasomal degradation pathway and autophagy (118, 123, 124). While the protea-
some is accountable for degrading soluble forms of the Z variant protein (125),
autophagy is specific for the removal of insoluble polymers and aggregates.
Teckmann et al. found that retention of AAT-Z in the ER is associated with a
marked autophagic response (126, 127). The diagnosis of AATD relies on the
serum concentration of AAT by radial immunodiffusion or nephelometry with
molecular genetic confirmation of the variants by screening for the SERPINA1
gene. In conjunction with the periodic pulmonary function tests, children and
young adults with AATD and established liver disease should undergo periodic
(annually or half-annually) ultrasound examination of the liver to monitor for
fibrotic changes and HCC occurrence.
Hereditary tyrosinemia I
Hereditary tyrosinemia I (HT1, OMIM 276700) is the result of an abnormality of
the tyrosine catabolic pathway due to deficiency of the enzyme fumarylacetoace-
tate hydrolase. It is an autosomal recessive disorder. In the liver, there is an accu-
mulation of toxic metabolites (maleylacetoacetate, fumarylacetoacetate, and
succinylacetone) which lead to hepatic and renal manifestations of the disease and
carcinogenesis. HCC occurs commonly in HT1, with a prevalence of 14–75%
(128, 129). Clinically, the spectrum is widely heterogeneous and includes acute or
chronic liver disease, glomerulosclerosis, hypophosphatemic rickets due to renal
tubular dysfunction, neurological porphyria-like crisis, hypertrophic cardiomy-
opathy, hypoglycemia due to hyperinsulinism, and failure to thrive. Currently,
treatment of HT1 is inhibition of the formation of toxic metabolites by nitisinone
[2-(2-nitro-4-trifluoromethyl-benzoyl)-1,3-cyclohexanedione; NTBC] and the
reduction of tyrosine levels by dietary treatment. Heat shock proteins and anti-
apoptotic proteins have been found to be altered in HT1. Presumably, there is a
mechanism whereby transformed cells have a survival advantage in murine mod-
els of HT1 (130, 131). There is a small risk of liver cancer in NTBC treated HT1
patients because this compound blocks the enzyme parahydroxyphenyl-pyruvic
acid dioxygenase, which is involved in the tyrosine degradation pathway, thus
preventing the accumulation of fumarylacetoacetate and succinylacetone. On the
other hand, animal models have revealed that NTBC treatment is unsuccessful in
normalizing tyrosinemia-induced changes in the expression of transcripts encod-
ing proteins involved in signal transduction, protein turnover, cell growth, and
cell proliferation (132). In one study on pediatric patients who underwent OLT,
HCC was found in 12 (75%) liver explants, but in another Iranian study, HCC
was present in just 5 (23%) of the 22 liver explants (133). Screening is recom-
mended in HT1 patients, using regular measures of AFP and imaging. Surveillance
typically includes ultrasound, but in the event a suspicious lesion is found, MRI
could be used for characterization and staging of lesions, as it is more sensitive
compared to ultrasound.
20 Sergi C M
of HCA to HCC may occur on rare occasions (153–161). In GSD-Ia HCAs, chro-
mosomal peculiarities are present in 60% of tumors, which is similar to the rate
observed in HCA seen in the general population. However, it seems that a simul-
taneous gain of chromosome 6p and loss of 6q are present in GSD-Ia HCA only.
Moreover, GSD-Ia HCAs show reduced expression of insulin-like growth factor-2
receptor (IGF2R) and large tumor suppressor kinase-1 (LATS1), which are candi-
date tumor suppressor genes at 6q in more than 50% of the tumors. This data
suggests a potential but still inexplicable role of 6q in oncogenic transformation of
such GSD-Ia HCAs (162). Type III GSD (debrancher enzyme deficiency, or Forbes
disease, or limit dextrinosis) also presents with HCA, but liver enzymes are
increased due to hepatocellular injury, and there is evidence of ballooning cell
change and fibrosis. GSD-III HCAs occur in 4–25% of children. Malignant onco-
genic transformation of HCA occurs almost always after liver cirrhosis (156, 163).
Rarely, HCC can occur in GSD-IV (brancher enzyme deficiency also known as
Andersen’s disease or amylopectinosis) (164).
Wilson’s disease
Wilson’s disease (WD) is characterized by copper accumulation and seems to be
protective against tumorigenesis, but HCC has been described (165). A Dutch
study targeting 140 adults with WD for over 15 years (166) reported an estimated
annual risk of HCC of 9 in 10,000 (0.09%). However, another multicenter
European study of 1,186 WD patients showed a prevalence rate of 0.67% (167).
Other predisposing conditions to HCC include genetic hemochromatosis, acute
intermittent porphyria, progressive familial intrahepatic cholestasis type 2, mito-
chondrial electron transport chain (ETC) disorders, glycogen storage diseases
types I, III, and IV, and transaldolase deficiency (168). Transaldolase deficiency is
a genetic disease caused by mutations in the transaldolase gene (TALDO1), which
leads to abnormally low levels of the transaldolase enzyme. Transaldolase is a
metabolic enzyme involved in the pentose phosphate pathway.
CHOLANGIOCELLULAR CARCINOMA
cancer antigen (CA) 19–9 was reported, and it was increased (>129 U/mL) in
four of them (50%). The SEER database program analysis and the scientific lit-
erature evidence showed that after a median follow-up time of 8 months (range
0–60 months), 12 (54.6%) patients had died, 9 (40.9%) patients were alive, and
1 (4.6%) had no reported survival data. Most pediatric cases had underlying risk
factors for CCA, such as PSC, while in adults only an estimated 30% of CCAs
occurred in patients with an underlying risk factor (108, 190–194). In adults,
underlying risk factors include bile duct cysts, primary sclerosing cholangitis
(specifically with inflammatory bowel disease), Caroli Disease, and hepatolithia-
sis. Choledocholithiasis, cholangitis, hepatitis C (HCV), hepatitis B (HBV) and
cirrhosis have been considered potential risk factors as well (108, 169). There is
a potential association of these tumors with viral infections (hepatitis B virus,
hepatitis C virus) and alcohol use, which has been seen increasingly when com-
pared with classic textbook associations of liver flukes (Opisthorchis viverrinii and
Clonorchis sinensis) and primary sclerosing cholangitis. Other chronic inflamma-
tory processes of the biliary tract have also been associated with CCA. In addi-
tion, congenital disorders such as BA and PIFC have been identified as
comorbidities. Congenital malformations of the biliary tree, such as choledochal
cysts and Caroli disease, may present with obstructive cholestasis; however,
patients may be asymptomatic and diagnosed later in life.
DPM may have several overlapping entities, which are mostly grouped into:
(i) Von Meyenburg complexes (VMCs), small nonhereditary nodular cystic
lesions; (ii) isolated polycystic liver disease (PCLD); and (iii) autosomal dominant
polycystic kidney disease (ADPKD), which shows multiple cysts in both kidney
and liver (218, 219). An arrest in the development of the intrahepatic biliary sys-
tem and cystic changes of the bile ducts result from disrupted TGF-β, Notch, and
Wnt signaling or ciliopathy (220). Both PCLD and ADPKD show multiple large
cysts replacing normal hepatic parenchyma. Hepatic failure, and complications
associated with cysts, such as infection, hemorrhage or rupture, are frequent, but
PCLD and ADPKD are not risk factors for CCA in general. VMC is a sporadic,
small (diameter less than 0.5 cm), usually singular DPM lesion, also called biliary
micro-hamartoma. Although it is often an incidental finding in the adult liver, it
is rarely encountered in childhood. Multiple VMCs may occur with multiple
cysts, which are grossly visible and diffusely located along the biliary tree.
Histologically, VMCs consist of irregularly dilated small bile ducts embedded in
the fibrous stroma with intraluminal inspissated bile secretion. VMCs are consid-
ered a part of the spectrum of adult PCLD, but there is no known genetic back-
ground such as ADPKD or PCLD (221). There is a large body of knowledge that
VMC has more than a potential role as a preneoplastic lesion for CCA. This
knowledge is based on several studies that have reported CCA arising from VMCs,
suggesting malignant transformation from VMC to CCA, CCA having histologic
similarities with VMC and DPM, and histologic similarity of CCA and VMC in
K-ras and p53 mutated animal models inducing CCA (222–232). Most recently,
the CCA with DPM pattern seems to have well-defined ARID1A genetic altera-
tions (233).
present in normal adult hepatocytes. Patients are almost always negative for keratin
20 (CK20) of the intermediate filaments of the cytoskeleton. In our past investiga-
tions on liver development (209) and ductal plate malformation of the liver (208,
210), we used three monoclonal antibodies against CK-7 (OV-TL 12/ 30, 1:50,
Dako Corporation, Hamburg, Germany), CK-19 (RCK 108, 1:100, Dako
Corporation, Hamburg, Germany), and a mixture of cytokeratin epitopes (AE 1/3,
1:2, Linaris, Camon, Wiesbaden, Germany). AE 1 reacts with specific group A
acidic keratins with a molecular weight of 40,000–50,000 (CK 10, 14, 15, 16, 19),
while AE 3 recognizes all eight group B basic keratins with a molecular weight of
58,000 – 67,000 (CK 1–8). A mixture of monoclonal antibodies AE 1 and AE 3
should theoretically stain both biliary epithelial cells and hepatocytes because AE 1
reacts with CK 19, and AE 3 reacts with both CK 7 and CK 8. However, we found
that in formalin-fixed tissue, the AE 113 cocktail reacts only with biliary cells. This
is probably due to formaldehyde fixation of the tissue, as previously shown
(235–237). The epitope recognized by AE 3 is altered by formalin fixation, making
it less available for binding. Furthermore, it differs from other cytokeratin-related
epitopes because it can only partially be ‘restored’ by antigen-unmasking tech-
niques. In CCA, there is no immunohistochemical detection for CDX2 or Caudal
Type Homeobox 2, whose gene is a component of the caudal-related homeobox
transcription factor family. CDX2 is expressed in the nuclei of intestinal epithelial
cells, and diseases associated with CDX2 include Ampulla of Vater adenocarci-
noma and mucinous adenocarcinoma of the ovary. Among CDX2 related pathways
are transcriptional regulatory networks in embryonic stem cell and incretin synthe-
sis, secretion, and inactivation. Ultrastructurally, CCC tumor cells show cytokera-
tin filaments, an intracytoplasmic lumen, tight junctions at the apices, and
desmosomes at the lateral surfaces of neighboring cells. All these features are char-
acteristics of adenocarcinoma cell origin.
The 8th edition of the Staging Manual of the American Joint Committee on
Cancer (AJCC) has different staging systems for intrahepatic, perihilar, distal
bile duct, and HCC (238). AJCC staging is based on vascular invasion, number
of tumor nodules, perforation of the visceral peritoneum, the involvement of
local extrahepatic tissues and the occurrence of periductal infiltration. In the
differential diagnoses in childhood and youth, it is rare to see metastatic colorec-
tal carcinoma, although I came across one such case in my practice; the tumor
suppressor gene, TP53, was deleted. The major differential diagnosis is with
HCC; particularly with unusual growth patterns such as trabecular and nested
type, careful consideration is necessary. The lack of nucleoli, the lack of bile
production, and the presence of prominent (central) fibrosis is suggestive of
CCA. A nested growth pattern would also suggest a neuroendocrine carcinoma,
but in this case, I would expect to see granular or “salt-and-pepper” (“stippled”)
chromatin, although nucleoli can also be seen in NET of high grade. In this set-
ting, the pathologist should consider abundant apoptosis, mitotic figures and
tumor necrosis, which may be particularly prominent in neuroendocrine carci-
nomas. The distinction of CCA from metastatic gall bladder, pancreatic or upper
gastrointestinal adenocarcinomas requires clinical correlation, although such
tumors are p ractically non-existent in childhood and youth. Neuroendocrine
carcinomas and neuroendocrine tumors are often positive for neuroendocrine
Liver Cancer in Children and Adolescents 25
CONCLUSION
Copyright and permission statement: The author confirms that the materials
included in this chapter do not violate copyright laws. Where relevant, a ppropriate
permissions have been obtained from the original copyright holder(s). All original
sources have been appropriately acknowledged and/or referenced.
26 Sergi C M
REFERENCES
39. Lopez-Terrada D, Alaggio R, de Davila MT, Czauderna P, Hiyama E, Katzenstein H, et al. Towards an
international pediatric liver tumor consensus classification: proceedings of the Los Angeles COG liver
tumors symposium. Mod Pathol. 2014;27(3):472–91. https://doi.org/10.1038/modpathol.2013.80
40. Zen Y, Vara R, Portmann B, Hadzic N. Childhood hepatocellular carcinoma: a clinicopathological
study of 12 cases with special reference to EpCAM. Histopathology. 2014;64(5):671–82. https://doi.
org/10.1111/his.12312
41. Tanaka Y, Inoue T, Horie H. International pediatric liver cancer pathological classification: current
trend. Int J Clin Oncol. 2013;18(6):946–54. https://doi.org/10.1007/s10147-013-0624-8
42. Prokurat A, Kluge P, Kosciesza A, Perek D, Kappeler A, Zimmermann A. Transitional liver cell tumors
(TLCT) in older children and adolescents: a novel group of aggressive hepatic tumors expressing beta-
catenin. Med Pediatr Oncol. 2002;39(5):510–8. https://doi.org/10.1002/mpo.10177
43. Faria SC, Szklaruk J, Kaseb AO, Hassabo HM, Elsayes KM. TNM/Okuda/Barcelona/UNOS/CLIP
International Multidisciplinary Classification of Hepatocellular Carcinoma: concepts, perspec-
tives, and radiologic implications. Abdom Imaging. 2014;39(5):1070–87. https://doi.org/10.1007/
s00261-014-0130-0
44. Towbin AJ, Meyers RL, Woodley H, Miyazaki O, Weldon CB, Morland B, et al. 2017 PRETEXT: radio-
logic staging system for primary hepatic malignancies of childhood revised for the Paediatric Hepatic
International Tumour Trial (PHITT). Pediatr Radiol. 2018;48(4):536–54. https://doi.org/10.1007/
s00247-018-4078-z
45. Aronson DC, Schnater JM, Staalman CR, Weverling GJ, Plaschkes J, Perilongo G, et al. Predictive value
of the pretreatment extent of disease system in hepatoblastoma: results from the International Society
of Pediatric Oncology Liver Tumor Study Group SIOPEL-1 study. J Clin Oncol. 2005;23(6):1245–52.
https://doi.org/10.1200/JCO.2005.07.145
46. Silberhumer GR, Steininger R, Laengle F, Muehlbacher F, Zacherl J, Pokieser P. Intraoperative ultra-
sonography in patients who undergo liver resection or transplantation for hepatocellular carcinoma.
Surg Technol Int. 2004;12:145–51.
47. Achilleos OA, Buist LJ, Kelly DA, Raafat F, McMaster P, Mayer AD, et al. Unresectable hepatic tumors
in childhood and the role of liver transplantation. J Pediatr Surg. 1996;31(11):1563–7. https://doi.
org/10.1016/S0022-3468(96)90179-7
48. Tagge EP, Tagge DU, Reyes J, Tzakis A, Iwatsuki S, Starzl TE, et al. Resection, including trans-
plantation, for hepatoblastoma and hepatocellular carcinoma: impact on survival. J Pediatr Surg.
1992;27(3):292–6; discussion 7. https://doi.org/10.1016/0022-3468(92)90849-3
49. Iwatsuki S, Starzl TE, Sheahan DG, Yokoyama I, Demetris AJ, Todo S, et al. Hepatic resection versus
transplantation for hepatocellular carcinoma. Ann Surg. 1991;214(3):221–8; discussion 8–9. https://
doi.org/10.1097/00000658-199109000-00005
50. Reyes JD, Carr B, Dvorchik I, Kocoshis S, Jaffe R, Gerber D, et al. Liver transplantation and che-
motherapy for hepatoblastoma and hepatocellular cancer in childhood and adolescence. J Pediatr.
2000;136(6):795–804. https://doi.org/10.1016/S0022-3476(00)44469-0
51. Ikeda H, Hirato J, Suzuki N, Kuroiwa M, Maruyama K, Tsuchida Y. Detection of hepatic oxidative
DNA damage in patients with hepatoblastoma and children with non-neoplastic disease. Med Pediatr
Oncol. 2001;37(6):505–10. https://doi.org/10.1002/mpo.1243
52. Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, et al. Randomized controlled trial of tran-
sarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology.
2002;35(5):1164–71. https://doi.org/10.1053/jhep.2002.33156
53. Li Z, Xiao D, Li X, Zhan P, Wang J, Zhang H. Early recurrence detected in hepatocellular carcinoma
patients after transcatheter arterial chemoembolization treatment with plasma cell-free DNA. Eur J
Gastroenterol Hepatol. 2019; 31(7):885–92. https://doi.org/10.1097/MEG.0000000000001373
54. Weiss KE, Sze DY, Rangaswami AA, Esquivel CO, Concepcion W, Lebowitz EA, et al. Transarterial
chemoembolization in children to treat unresectable hepatocellular carcinoma. Pediatr Transplant.
2018;22(4):e13187. https://doi.org/10.1111/petr.13187
55. Kohorst MA, Warad DM, Matsumoto JM, Heimbach JK, El-Youssef M, Arndt CAS, et al. Management
of pediatric hepatocellular carcinoma: A multimodal approach. Pediatr Transplant. 2017;21(6).
https://doi.org/10.1111/petr.13007
Liver Cancer in Children and Adolescents 29
56. Hiyama E. Current therapeutic strategies for childhood hepatic malignant tumors. Int J Clin Oncol.
2013;18(6):943–5. https://doi.org/10.1007/s10147-013-0607-9
57. von Schweinitz D, Burger D, Bode U, Weinel P, Erttmann R, Hecker H, et al. [Results of the HB-89
Study in treatment of malignant epithelial liver tumors in childhood and concept of a new HB-94
protocol]. Klin Padiatr. 1994;206(4):282–8. https://doi.org/10.1055/s-2008-1046615
58. Keating GM. Sorafenib: A Review in Hepatocellular Carcinoma. Target Oncol. 2017;12(2):243–53.
https://doi.org/10.1007/s11523-017-0484-7
59. Schmid I, von Schweinitz D. Pediatric hepatocellular carcinoma: challenges and solutions. J Hepatocell
Carcinoma. 2017;4:15–21. https://doi.org/10.2147/JHC.S94008
60. Schmid I, Haberle B, Albert MH, Corbacioglu S, Frohlich B, Graf N, et al. Sorafenib and cisplatin/doxo-
rubicin (PLADO) in pediatric hepatocellular carcinoma. Pediatr Blood Cancer. 2012;58(4):539–44.
https://doi.org/10.1002/pbc.23295
61. Sun W, Cabrera R. Systemic Treatment of Patients with Advanced, Unresectable Hepatocellular
Carcinoma: Emergence of Therapies. J Gastrointest Cancer. 2018;49(2):107–15. https://doi.org/
10.1007/s12029-018-0065-8
62. Kelley RK, Venook AP. Novel therapeutics in hepatocellular carcinoma: how can we make progress?
Am Soc Clin Oncol Educ Book. 2013. https://doi.org/10.14694/EdBook_AM.2013.33.e137
63. Finn RS. Emerging targeted strategies in advanced hepatocellular carcinoma. Semin Liver Dis.
2013;33 Suppl 1:S11–9. https://doi.org/10.1055/s-0033-1333632
64. Zhu AX. Systemic treatment of hepatocellular carcinoma: dawn of a new era? Ann Surg Oncol.
2010;17(5):1247–56. https://doi.org/10.1245/s10434-010-0975-6
65. Cholongitas E, Mamou C, Rodriguez-Castro KI, Burra P. Mammalian target of rapamycin inhibitors
are associated with lower rates of hepatocellular carcinoma recurrence after liver transplantation: a
systematic review. Transpl Int. 2014;27(10):1039–49. https://doi.org/10.1111/tri.12372
66. Waidmann O, Trojan J. Novel drugs in clinical development for hepatocellular carcinoma. Expert
Opin Investig Drugs. 2015;24(8):1075–82. https://doi.org/10.1517/13543784.2015.1058776
67. Bouattour M, Raymond E, Qin S, Cheng AL, Stammberger U, Locatelli G, et al. Recent develop-
ments of c-Met as a therapeutic target in hepatocellular carcinoma. Hepatology. 2018;67(3):1132–49.
https://doi.org/10.1002/hep.29496
68. Gnoni A, Santini D, Scartozzi M, Russo A, Licchetta A, Palmieri V, et al. Hepatocellular carcinoma
treatment over sorafenib: epigenetics, microRNAs and microenvironment. Is there a light at the end
of the tunnel? Expert Opin Ther Targets. 2015;19(12):1623–35. https://doi.org/10.1517/14728222.
2015.1071354
69. Porta C, Giglione P, Ferrari A, Reversi F, Liguigli W, Imarisio I, et al. Tivantinib (ARQ197) in hepato-
cellular carcinoma. Expert Rev Anticancer Ther. 2015;15(6):615–22. https://doi.org/10.1586/14737
140.2015.1050383
70. Qi XS, Guo XZ, Han GH, Li HY, Chen J. MET inhibitors for treatment of advanced hepatocellular
carcinoma: A review. World J Gastroenterol. 2015;21(18):5445–53. https://doi.org/10.3748/wjg.v21.
i18.5445
71. Rota Caremoli E, Labianca R. Tivantinib: critical review with a focus on hepatocellular carcinoma.
Expert Opin Investig Drugs. 2014;23(11):1563–74. https://doi.org/10.1517/13543784.2014.949339
72. Bertino G, Di Carlo I, Ardiri A, Calvagno GS, Demma S, Malaguarnera G, et al. Systemic therapies
in hepatocellular carcinoma: present and future. Future Oncol. 2013;9(10):1533–48. https://doi.
org/10.2217/fon.13.171
73. Santoro A, Rimassa L, Borbath I, Daniele B, Salvagni S, Van Laethem JL, et al. Tivantinib for second-
line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase 2 study.
Lancet Oncol. 2013;14(1):55–63. https://doi.org/10.1016/S1470-2045(12)70490-4
74. Liu PH, Huo TI, Miksad RA. Hepatocellular Carcinoma with Portal Vein Tumor Involvement:
Best Management Strategies. Semin Liver Dis. 2018;38(3):242–51. https://doi.org/10.1055/
s-0038-1666805
75. Ikeda M, Morizane C, Ueno M, Okusaka T, Ishii H, Furuse J. Chemotherapy for hepatocellular car-
cinoma: current status and future perspectives. Jpn J Clin Oncol. 2018;48(2):103–14. https://doi.
org/10.1093/jjco/hyx180
30 Sergi C M
76. Tovoli F, Lorenzo S, Barbera MA, Garajova I, Frega G, Palloni A, et al. Postsorafenib systemic treat-
ments for hepatocellular carcinoma: questions and opportunities after the regorafenib trial. Future
Oncol. 2017;13(21):1893–905. https://doi.org/10.2217/fon-2017-0166
77. Brizzi MP, Pignataro D, Tampellini M, Scagliotti GV, Di Maio M. Systemic treatment of hepatocellular
carcinoma: why so many failures in the development of new drugs? Expert Rev Anticancer Ther.
2016;16(10):1053–62. https://doi.org/10.1080/14737140.2016.1227706
78. Harding JJ, Abou-Alfa GK. Treating advanced hepatocellular carcinoma: How to get out of first gear.
Cancer. 2014;120(20):3122–30. https://doi.org/10.1002/cncr.28850
79. Nautsch F, Ludwig JM, Xing M, Johnson KM, Kim HS. Racial Disparities and Sociodemographic
Differences in Incidence and Survival Among Pediatric Patients in the United States With Primary
Liver Cancer: A Surveillance, Epidemiology, and End Results (SEER) Population Study. J Clin
Gastroenterol. 2018;52(3):262–7. https://doi.org/10.1097/MCG.0000000000000833
80. Lau CS, Mahendraraj K, Chamberlain RS. Hepatocellular Carcinoma in the Pediatric Population:
A Population Based Clinical Outcomes Study Involving 257 Patients from the Surveillance,
Epidemiology, and End Result (SEER) Database (1973–2011). HPB Surg. 2015;2015:670728. https://
doi.org/10.1155/2015/670728
81. Allan BJ, Wang B, Davis JS, Parikh PP, Perez EA, Neville HL, et al. A review of 218 pediatric
cases of hepatocellular carcinoma. J Pediatr Surg. 2014;49(1):166–71. https://doi.org/10.1016/j.
jpedsurg.2013.09.050
82. Bioulac-Sage P, Rebouissou S, Thomas C, Blanc JF, Saric J, Sa Cunha A, et al. Hepatocellular ade-
noma subtype classification using molecular markers and immunohistochemistry. Hepatology. 2007;
46(3):740–8. https://doi.org/10.1002/hep.21743
83. Zucman-Rossi J, Jeannot E, Nhieu JT, Scoazec JY, Guettier C, Rebouissou S, et al. Genotype-phenotype
correlation in hepatocellular adenoma: new classification and relationship with HCC. Hepatology.
2006;43(3):515–24. https://doi.org/10.1002/hep.21068
84. Wanless IR, Mawdsley C, Adams R. On the pathogenesis of focal nodular hyperplasia of the liver.
Hepatology. 1985;5(6):1194–200. https://doi.org/10.1002/hep.1840050622
85. Mortele KJ, Praet M, Van Vlierberghe H, Kunnen M, Ros PR. CT and MR imaging findings in
focal nodular hyperplasia of the liver: radiologic-pathologic correlation. AJR Am J Roentgenol.
2000;175(3):687–92. https://doi.org/10.2214/ajr.175.3.1750687
86. Nahm CB, Ng K, Lockie P, Samra JS, Hugh TJ. Focal nodular hyperplasia--a review of myths and
truths. J Gastrointest Surg. 2011;15(12):2275–83. https://doi.org/10.1007/s11605-011-1680-x
87. Shafizadeh N, Kakar S. Diagnosis of well-differentiated hepatocellular lesions: role of immuno-
histochemistry and other ancillary techniques. Adv Anat Pathol. 2011;18(6):438–45. https://doi.
org/10.1097/PAP.0b013e318234abb4
88. Liu Q, Teh M, Ito K, Shah N, Ito Y, Yeoh KG. CDX2 expression is progressively decreased in human
gastric intestinal metaplasia, dysplasia and cancer. Mod Pathol. 2007;20(12):1286–97. https://doi.
org/10.1038/modpathol.3800968
89. Bahitham W, Liao X, Peng F, Bamforth F, Chan A, Mason A, et al. Mitochondriome and cholangiocel-
lular carcinoma. PLoS One. 2014;9(8):e104694. https://doi.org/10.1371/journal.pone.0104694
90. Johnston J, Al-Bahrani R, Abuetabh Y, Chiu B, Forsman CL, Nagamori S, et al. Twisted gastrula-
tion expression in cholangiocellular and hepatocellular carcinoma. J Clin Pathol. 2012;65(10):945–8.
https://doi.org/10.1136/jclinpath-2011-200577
91. Dorn L, Menezes LF, Mikuz G, Otto HF, Onuchic LF, Sergi C. Immunohistochemical detection of
polyductin and co-localization with liver progenitor cell markers during normal and abnormal devel-
opment of the intrahepatic biliary system and in adult hepatobiliary carcinomas. J Cell Mol Med.
2009;13(7):1279–90. https://doi.org/10.1111/j.1582-4934.2008.00519.x
92. Yang GC, Yang GY, Tao LC. Distinguishing well-differentiated hepatocellular carcinoma from benign
liver by the physical features of fine-needle aspirates. Mod Pathol. 2004;17(7):798–802. https://doi.
org/10.1038/modpathol.3800121
93. Yang GC, Yang GY, Tao LC. Cytologic features and histologic correlations of microacinar and microtra-
becular types of well-differentiated hepatocellular carcinoma in fine-needle aspiration biopsy. Cancer.
2004;102(1):27–33. https://doi.org/10.1002/cncr.20000
Liver Cancer in Children and Adolescents 31
94. Mitchell CM, Sturgis CD. Lipid-rich hepatocellular carcinoma in fine-needle aspiration biopsy. Diagn
Cytopathol. 2009;37(1):36–7. https://doi.org/10.1002/dc.20891
95. Nakagawa H, Hayata Y, Kawamura S, Yamada T, Fujiwara N, Koike K. Lipid Metabolic Reprogram-
ming in Hepatocellular Carcinoma. Cancers (Basel). 2018;10(11):447. https://doi.org/10.3390/
cancers10110447
96. Yang Y, Shen F, Huang W, Qin S, Huang JT, Sergi C, et al. Glucose Is Involved in the Dynamic
Regulation of m6A in Patients With Type 2 Diabetes. J Clin Endocrinol Metab. 2019;104(3):665–73.
https://doi.org/10.1210/jc.2018-00619
97. Aspichueta P. Lipid-rich environment: a key role promoting carcinogenesis in obesity-related non-
alcoholic fatty liver disease. Gut. 2018;67(8):1376–7. https://doi.org/10.1136/gutjnl-2018-316047
98. Orikasa H, Ohyama R, Tsuka N, Eyden BP, Yamazaki K. Lipid-rich clear-cell hepatocellular carcinoma
arising in non-alcoholic steatohepatitis in a patient with diabetes mellitus. J Submicrosc Cytol Pathol.
2001;33(1–2):195–200.
99. Reid LM, Sethupathy P. The DNAJB1-PRKACA chimera: Candidate biomarker and therapeutic target
for fibrolamellar carcinomas. Hepatology. 2016;63(2):662–4. https://doi.org/10.1002/hep.28307
100. Sergi CM. Hepatocellular Carcinoma, Fibrolamellar Variant: Diagnostic Pathologic Criteria and
Molecular Pathology Update. A Primer. Diagnostics (Basel). 2015;6(1):3. https://doi.org/10.3390/
diagnostics6010003
101. Graham RP, Jin L, Knutson DL, Kloft-Nelson SM, Greipp PT, Waldburger N, et al. DNAJB1-PRKACA
is specific for fibrolamellar carcinoma. Mod Pathol. 2015;28(6):822–9. https://doi.org/10.1038/
modpathol.2015.4
102. Xu L, Hazard FK, Zmoos AF, Jahchan N, Chaib H, Garfin PM, et al. Genomic analysis of fi brolamellar
hepatocellular carcinoma. Hum Mol Genet. 2015;24(1):50–63. https://doi.org/10.1093/hmg/ddu418
103. Honeyman JN, Simon EP, Robine N, Chiaroni-Clarke R, Darcy DG, Lim, II, et al. Detection of a
recurrent DNAJB1-PRKACA chimeric transcript in fibrolamellar hepatocellular carcinoma. Science.
2014;343(6174):1010–4. https://doi.org/10.1126/science.1249484
104. Ang CS, Kelley RK, Choti MA, Cosgrove DP, Chou JF, Klimstra D, et al. Clinicopathologic
characteristics and survival outcomes of patients with fibrolamellar carcinoma: data from the fibrola-
mellar carcinoma consortium. Gastrointest Cancer Res. 2013;6(1):3–9.
105. Klein WM, Molmenti EP, Colombani PM, Grover DS, Schwarz KB, Boitnott J, et al. Primary liver
carcinoma arising in people younger than 30 years. Am J Clin Pathol. 2005;124(4):512–8. https://doi.
org/10.1309/TT0R7KAL32228E99
106. Buckley AF, Burgart LJ, Kakar S. Epidermal growth factor receptor expression and gene copy number
in fibrolamellar hepatocellular carcinoma. Hum Pathol. 2006;37(4):410–4. https://doi.org/10.1016/j.
humpath.2005.12.009
107. Torbenson M. Review of the clinicopathologic features of fibrolamellar carcinoma. Adv Anat Pathol.
2007;14(3):217–23. https://doi.org/10.1097/PAP.0b013e3180504913
108. Al-Bahrani R, Abuetabh Y, Zeitouni N, Sergi C. Cholangiocarcinoma: risk factors, environmental
influences and oncogenesis. Ann Clin Lab Sci. 2013;43(2):195–210.
109. Corti B, D’Errico A, Pierangeli F, Fiorentino M, Altimari A, Grigioni WF. Primary paraganglioma
strictly confined to the liver and mimicking hepatocellular carcinoma: an immunohistochemical
and in situ hybridization study. Am J Surg Pathol. 2002;26(7):945–9. https://doi.org/10.1097/
00000478-200207000-00015
110. Cheuk W, Chan JK. Clear cell variant of fibrolamellar carcinoma of the liver. Arch Pathol Lab Med.
2001;125(9):1235–8. https://doi.org/10.5858/2001-125-1235-CCVOFC
111. Kannangai R, Vivekanandan P, Martinez-Murillo F, Choti M, Torbenson M. Fibrolamellar carcinomas
show overexpression of genes in the RAS, MAPK, PIK3, and xenobiotic degradation pathways. Hum
Pathol. 2007;38(4):639–44. https://doi.org/10.1016/j.humpath.2006.07.019
112. Riehle KJ, Yeh MM, Yu JJ, Kenerson HL, Harris WP, Park JO, et al. mTORC1 and FGFR1 signaling
in fibrolamellar hepatocellular carcinoma. Mod Pathol. 2015;28(1):103–10. https://doi.org/10.1038/
modpathol.2014.78
113. Fabbretti G, Sergi C, Consalez G, Faa G, Brisigotti M, Romeo G, et al. Genetic variants of alpha-1-anti-
trypsin (AAT). Liver. 1992;12(4 Pt 2):296–301. https://doi.org/10.1111/j.1600-0676.1992.tb01064.x
32 Sergi C M
114. Sergi C, Consalez GG, Fabbretti G, Brisigotti M, Faa G, Costa V, et al. Immunohistochemical and
genetic characterization of the M Cagliari alpha-1-antitrypsin molecule (M-like alpha-1-antitrypsin
deficiency). Lab Invest. 1994;70(1):130–3.
115. Spada F, Candiano G, Sergi C, Ghiggeri GM, Callea F, Gusmano R. Purification of alpha-1-antitrypsin
monomer by preparative electrophoresis. J Clin Pathol. 1994;47(7):661–3. https://doi.org/10.1136/
jcp.47.7.661
116. Sergi C, Benstz J, Feist D, Nutzenadel W, Otto HF, Hofmann WJ. Bile duct to portal space ratio and
ductal plate remnants in liver disease of infants aged less than 1 year. Pathology. 2008;40(3):260–7.
https://doi.org/10.1080/00313020801911538
117. Carrell RW, Lomas DA. Alpha1-antitrypsin deficiency--a model for conformational diseases. N Engl J
Med. 2002;346(1):45–53. https://doi.org/10.1056/NEJMra010772
118. Topic A, Ljujic M, Radojkovic D. Alpha-1-antitrypsin in pathogenesis of hepatocellular carcinoma.
Hepat Mon. 2012;12(10 HCC):e7042. https://doi.org/10.5812/hepatmon.7042
119. Carlson JA, Rogers BB, Sifers RN, Finegold MJ, Clift SM, DeMayo FJ, et al. Accumulation of PiZ
alpha 1-antitrypsin causes liver damage in transgenic mice. J Clin Invest. 1989;83(4):1183–90.
https://doi.org/10.1172/JCI113999
120. Marcus NY, Brunt EM, Blomenkamp K, Ali F, Rudnick DA, Ahmad M, et al. Characteristics of
hepatocellular carcinoma in a murine model of alpha-1-antitrypsin deficiency. Hepatol Res.
2010;40(6):641–53. https://doi.org/10.1111/j.1872-034X.2010.00663.x
121. Wu Y, Whitman I, Molmenti E, Moore K, Hippenmeyer P, Perlmutter DH. A lag in intracellular
degradation of mutant alpha 1-antitrypsin correlates with the liver disease phenotype in homozy-
gous PiZZ alpha 1-antitrypsin deficiency. Proc Natl Acad Sci U S A. 1994;91(19):9014–8. https://doi.
org/10.1073/pnas.91.19.9014
122. Hidvegi T, Ewing M, Hale P, Dippold C, Beckett C, Kemp C, et al. An autophagy-enhancing
drug promotes degradation of mutant alpha1-antitrypsin Z and reduces hepatic fibrosis. Science.
2010;329(5988):229–32. https://doi.org/10.1126/science.1190354
123. Chiu B, Jantuan E, Shen F, Chiu B, Sergi C. Autophagy-Inflammasome Interplay in Heart Failure:
A Systematic Review on Basics, Pathways, and Therapeutic Perspectives. Ann Clin Lab Sci. 2017;
47(3):243–52.
124. Perlmutter DH. Pathogenesis of chronic liver injury and hepatocellular carcinoma in alpha-1-antitrypsin
deficiency. Pediatr Res. 2006;60(2):233–8. https://doi.org/10.1203/01.pdr.0000228350.61496.90
125. Qu D, Teckman JH, Omura S, Perlmutter DH. Degradation of a mutant secretory protein,
alpha1-antitrypsin Z, in the endoplasmic reticulum requires proteasome activity. J Biol Chem.
1996;271(37):22791–5. https://doi.org/10.1074/jbc.271.37.22791
126. Teckman JH, Gilmore R, Perlmutter DH. Role of ubiquitin in proteasomal degradation of mutant
alpha(1)-antitrypsin Z in the endoplasmic reticulum. Am J Physiol Gastrointest Liver Physiol.
2000;278(1):G39–48. https://doi.org/10.1152/ajpgi.2000.278.1.G39
127. Teckman JH, An JK, Loethen S, Perlmutter DH. Fasting in alpha1-antitrypsin deficient liver: consti-
tutive [correction of consultative] activation of autophagy. Am J Physiol Gastrointest Liver Physiol.
2002;283(5):G1156–65. https://doi.org/10.1152/ajpgi.00041.2002
128. Mayorandan S, Meyer U, Gokcay G, Segarra NG, de Baulny HO, van Spronsen F, et al. Cross-sectional
study of 168 patients with hepatorenal tyrosinaemia and implications for clinical practice. Orphanet
J Rare Dis. 2014;9:107. https://doi.org/10.1186/s13023-014-0107-7
129. Zeybek AC, Kiykim E, Soyucen E, Cansever S, Altay S, Zubarioglu T, et al. Hereditary tyrosinemia
type 1 in Turkey: twenty year single-center experience. Pediatr Int. 2015;57(2):281–9. https://doi.
org/10.1111/ped.12503
130. Bergeron A, Jorquera R, Orejuela D, Tanguay RM. Involvement of endoplasmic reticulum stress
in hereditary tyrosinemia type I. J Biol Chem. 2006;281(9):5329–34. https://doi.org/10.1074/jbc.
M506804200
131. Vogel A, van Den Berg IE, Al-Dhalimy M, Groopman J, Ou CN, Ryabinina O, et al. Chronic liver
disease in murine hereditary tyrosinemia type 1 induces resistance to cell death. Hepatology.
2004;39(2):433–43. https://doi.org/10.1002/hep.20077
Liver Cancer in Children and Adolescents 33
132. Luijerink MC, Jacobs SM, van Beurden EA, Koornneef LP, Klomp LW, Berger R, et al. Extensive
changes in liver gene expression induced by hereditary tyrosinemia type I are not normalized by
treatment with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC). J Hepatol. 2003;
39(6):901–9. https://doi.org/10.1016/S0168-8278(03)00433-1
133. Bahador A, Dehghani SM, Geramizadeh B, Nikeghbalian S, Bahador M, Malekhosseini SA, et al. Liver
Transplant for Children With Hepatocellular Carcinoma and Hereditary Tyrosinemia Type 1. Exp Clin
Transplant. 2015;13(4):329–32.
134. Al-Bahrani R, Tuertcher D, Zailaie S, Abuetabh Y, Nagamori S, Zetouni N, et al. Differential SIRT1
expression in hepatocellular carcinomas and cholangiocarcinoma of the liver. Ann Clin Lab Sci.
2015;45(1):3–9.
135. Wu Y, Ren F, Chai Y, Xue Z, Shen C, Zhang X, et al. Prognostic value of inflammation-based indexes
for intrahepatic cholangiocarcinoma following curative resection. Oncol Lett. 2019;17(1):165–74.
136. Seo S, Yoh T, Morino K, Fuji H, Taura K, Fukumitsu K, et al. The Relationship Between (18)F-FDG
Uptake on PET/CT and Markers of Systemic Inflammatory Response in Patients Undergoing Surgery
for Intrahepatic Cholangiocarcinoma. Anticancer Res. 2019;39(1):341–6. https://doi.org/10.21873/
anticanres.13117
137. Nakagawa H, Suzuki N, Koike K. Mouse Model for Cholangiocarcinoma from Peribiliary Glands.
Methods Mol Biol. 2019;1905:237–45. https://doi.org/10.1007/978-1-4939-8961-4_21
138. Sripa B, Tangkawattana S, Brindley PJ. Update on Pathogenesis of Opisthorchiasis and
Cholangiocarcinoma. Adv Parasitol. 2018;102:97–113. https://doi.org/10.1016/bs.apar.2018.10.001
139. Edwards SW, Spofford EM, Price C, Wright HL, Salao K, Suttiprapa S, et al. Opisthorchiasis-Induced
Cholangiocarcinoma: How Innate Immunity May Cause Cancer. Adv Parasitol. 2018;101:149–76.
https://doi.org/10.1016/bs.apar.2018.05.006
140. Taghavi SA, Eshraghian A, Niknam R, Sivandzadeh GR, Bagheri Lankarani K. Diagnosis of cholangio-
carcinoma in primary sclerosing cholangitis. Expert Rev Gastroenterol Hepatol. 2018;12(6):575–84.
https://doi.org/10.1080/17474124.2018.1473761
141. Chung BK, Karlsen TH, Folseraas T. Cholangiocytes in the pathogenesis of primary sclerosing
cholangitis and development of cholangiocarcinoma. Biochim Biophys Acta Mol Basis Dis. 2018;
1864(4 Pt B):1390–400. https://doi.org/10.1016/j.bbadis.2017.08.020
142. Surapaitoon A, Suttiprapa S, Mairiang E, Khuntikeo N, Pairojkul C, Bethony J, et al. Subsets of
Inflammatory Cytokine Gene Polymorphisms are Associated with Risk of Carcinogenic Liver Fluke
Opisthorchis viverrini-Associated Advanced Periductal Fibrosis and Cholangiocarcinoma. Korean J
Parasitol. 2017;55(3):295–304. https://doi.org/10.3347/kjp.2017.55.3.295
143. Knisely AS, Strautnieks SS, Meier Y, Stieger B, Byrne JA, Portmann BC, et al. Hepatocellular car-
cinoma in ten children under five years of age with bile salt export pump deficiency. Hepatology.
2006;44(2):478–86. https://doi.org/10.1002/hep.21287
144. Davit-Spraul A, Fabre M, Branchereau S, Baussan C, Gonzales E, Stieger B, et al. ATP8B1 and ABCB11
analysis in 62 children with normal gamma-glutamyl transferase progressive familial intrahepatic
cholestasis (PFIC): phenotypic differences between PFIC1 and PFIC2 and natural history. Hepatology.
2010;51(5):1645–55. https://doi.org/10.1002/hep.23539
145. Pawlikowska L, Strautnieks S, Jankowska I, Czubkowski P, Emerick K, Antoniou A, et al. Differences
in presentation and progression between severe FIC1 and BSEP deficiencies. J Hepatol. 2010;53(1):
170–8. https://doi.org/10.1016/j.jhep.2010.01.034
146. Strautnieks SS, Byrne JA, Pawlikowska L, Cebecauerova D, Rayner A, Dutton L, et al. Severe bile
salt export pump deficiency: 82 different ABCB11 mutations in 109 families. Gastroenterology.
2008;134(4):1203–14. https://doi.org/10.1053/j.gastro.2008.01.038
147. Iannelli F, Collino A, Sinha S, Radaelli E, Nicoli P, D’Antiga L, et al. Corrigendum: Massive gene ampli-
fication drives paediatric hepatocellular carcinoma caused by bile salt export pump deficiency. Nat
Commun. 2015;6:7456. https://doi.org/10.1038/ncomms8456
148. Iannelli F, Collino A, Sinha S, Radaelli E, Nicoli P, D’Antiga L, et al. Massive gene amplification drives
paediatric hepatocellular carcinoma caused by bile salt export pump deficiency. Nat Commun.
2014;5:3850. https://doi.org/10.1038/ncomms4850
34 Sergi C M
149. Chen Y, Song X, Valanejad L, Vasilenko A, More V, Qiu X, et al. Bile salt export pump is dysregu-
lated with altered farnesoid X receptor isoform expression in patients with hepatocellular carcinoma.
Hepatology. 2013;57(4):1530–41. https://doi.org/10.1002/hep.26187
150. Zhou S, Hertel PM, Finegold MJ, Wang L, Kerkar N, Wang J, et al. Hepatocellular carcinoma associated
with tight-junction protein 2 deficiency. Hepatology. 2015;62(6):1914–6. https://doi.org/10.1002/
hep.27872
151. Sambrotta M, Strautnieks S, Papouli E, Rushton P, Clark BE, Parry DA, et al. Mutations in TJP2
cause progressive cholestatic liver disease. Nat Genet. 2014;46(4):326–8. https://doi.org/10.1038/
ng.2918
152. Vij M, Shanmugam NP, Reddy MS, Govil S, Rela M. Hepatocarcinogenesis in multidrug-resistant
P-glycoprotein 3 deficiency. Pediatr Transplant. 2017;21(3). https://doi.org/10.1111/petr.12889
153. Reddy SK, Austin SL, Spencer-Manzon M, Koeberl DD, Clary BM, Desai DM, et al. Liver transplanta-
tion for glycogen storage disease type Ia. J Hepatol. 2009;51(3):483–90. https://doi.org/10.1016/j.
jhep.2009.05.026
154. Sentner CP, Hoogeveen IJ, Weinstein DA, Santer R, Murphy E, McKiernan PJ, et al. Glycogen storage
disease type III: diagnosis, genotype, management, clinical course and outcome. J Inherit Metab Dis.
2016;39(5):697–704. https://doi.org/10.1007/s10545-016-9932-2
155. Kishnani PS, Austin SL, Abdenur JE, Arn P, Bali DS, Boney A, et al. Diagnosis and management of
glycogen storage disease type I: a practice guideline of the American College of Medical Genetics and
Genomics. Genet Med. 2014;16(11):e1. https://doi.org/10.1038/gim.2014.128
156. Kishnani PS, Austin SL, Arn P, Bali DS, Boney A, Case LE, et al. Glycogen storage disease type III
diagnosis and management guidelines. Genet Med. 2010;12(7):446–63. https://doi.org/10.1097/
GIM.0b013e3181e655b6
157. Franco LM, Krishnamurthy V, Bali D, Weinstein DA, Arn P, Clary B, et al. Hepatocellular carcinoma
in glycogen storage disease type Ia: a case series. J Inherit Metab Dis. 2005;28(2):153–62. https://doi.
org/10.1007/s10545-005-7500-2
158. Rake JP, Visser G, Labrune P, Leonard JV, Ullrich K, Smit GP. Glycogen storage disease type I: diag-
nosis, management, clinical course and outcome. Results of the European Study on Glycogen
Storage Disease Type I (ESGSD I). Eur J Pediatr. 2002;161 Suppl 1:S20–34. https://doi.org/10.1007/
BF02679990
159. Bianchi L. Glycogen storage disease I and hepatocellular tumours. Eur J Pediatr. 1993;152
Suppl 1:S63–70. https://doi.org/10.1007/BF02072092
160. Labrune P, Trioche P, Duvaltier I, Chevalier P, Odievre M. Hepatocellular adenomas in glycogen stor-
age disease type I and III: a series of 43 patients and review of the literature. J Pediatr Gastroenterol
Nutr. 1997;24(3):276–9. https://doi.org/10.1097/00005176-199703000-00008
161. Oterdoom LH, Verweij KE, Biermann K, Langeveld M, van Buuren HR. Hepatocellular Adenomas and
Carcinoma in Asymptomatic, Non-Cirrhotic Type III Glycogen Storage Disease. J Gastrointestin Liver
Dis. 2015;24(4):515–8. https://doi.org/10.15403/jgld.2014.1121.244.had
162. Kishnani PS, Chuang TP, Bali D, Koeberl D, Austin S, Weinstein DA, et al. Chromosomal and genetic
alterations in human hepatocellular adenomas associated with type Ia glycogen storage disease. Hum
Mol Genet. 2009;18(24):4781–90. https://doi.org/10.1093/hmg/ddp441
163. Demo E, Frush D, Gottfried M, Koepke J, Boney A, Bali D, et al. Glycogen storage disease type
III-hepatocellular carcinoma a long-term complication? J Hepatol. 2007;46(3):492–8. https://doi.
org/10.1016/j.jhep.2006.09.022
164. Romano F, Stroppa P, Bravi M, Casotti V, Lucianetti A, Guizzetti M, et al. Favorable outcome of pri-
mary liver transplantation in children with cirrhosis and hepatocellular carcinoma. Pediatr Transplant.
2011;15(6):573–9. https://doi.org/10.1111/j.1399-3046.2011.01528.x
165. Rosencrantz RA, LeCompte L, Yusuf Y. Beneath the Copper-Pediatric Wilson’s Disease Cirrhosis
and Hepatocellular Carcinoma: A Case Report with Literature Review. Semin Liver Dis. 2015;35(4):
434–8. https://doi.org/10.1055/s-0035-1567828
166. van Meer S, de Man RA, van den Berg AP, Houwen RH, Linn FH, van Oijen MG, et al. No increased
risk of hepatocellular carcinoma in cirrhosis due to Wilson disease during long-term follow-up.
J Gastroenterol Hepatol. 2015;30(3):535–9. https://doi.org/10.1111/jgh.12716
Liver Cancer in Children and Adolescents 35
167. Pfeiffenberger J, Mogler C, Gotthardt DN, Schulze-Bergkamen H, Litwin T, Reuner U, et al. Hepatobiliary
malignancies in Wilson disease. Liver Int. 2015;35(5):1615–22. https://doi.org/10.1111/liv.12727
168. Sergi C, Jundt K, Seipp S, Goeser T, Theilmann L, Otto G, et al. The distribution of HBV, HCV and
HGV among livers with fulminant hepatic failure of different aetiology. J Hepatol. 1998;29(6):861–71.
https://doi.org/10.1016/S0168-8278(98)80112-8
169. Newsome JR, Venkatramani R, Heczey A, Danysh HE, Fishman DS, Miloh T. Cholangiocarcinoma
Among Children and Adolescents: A Review of the Literature and Surveillance, Epidemiology, and
End Results Program Database Analysis. J Pediatr Gastroenterol Nutr. 2018;66(1):e12-e8. https://doi.
org/10.1097/MPG.0000000000001749
170. Armanino LP. Idiopathic dilation of the common bile duct with coexistent primary hepatic carcinoma:
report of a case. Ann Intern Med. 1946;24:714–26. https://doi.org/10.7326/0003-4819-24-4-714
171. Fujiwara Y, Ohizumi T, Kakizaki G, Ishidate T. A case of congenital choledochal cyst associated with
carcinoma. J Pediatr Surg. 1976;11(4):587–8. https://doi.org/10.1016/S0022-3468(76)80021-8
172. Kulkarni PB, Beatty E Jr. Cholangiocarcinoma associated with biliary cirrhosis due to con-
genital biliary atresia. Am J Dis Child. 1977;131(4):442–4. https://doi.org/10.1001/archpedi.
1977.02120170068013
173. Iwai N, Deguchi E, Yanagihara J, Iwai M, Matsuo H, Todo S, et al. Cancer arising in a chole-
dochal cyst in a 12-year-old girl. J Pediatr Surg. 1990;25(12):1261–3. https://doi.org/10.1016/
0022-3468(90)90525-E
174. Tanaka S, Kubota M, Yagi M, Okuyama N, Ohtaki M, Yamazaki S, et al. An 11-year-old male patient
demonstrating cholangiocarcinoma associated with congenital biliary dilatation. J Pediatr Surg.
2006;41(1):e15–9. https://doi.org/10.1016/j.jpedsurg.2005.10.066
175. Scheimann AO, Strautnieks SS, Knisely AS, Byrne JA, Thompson RJ, Finegold MJ. Mutations in bile
salt export pump (ABCB11) in two children with progressive familial intrahepatic cholestasis and
cholangiocarcinoma. J Pediatr. 2007;150(5):556–9. https://doi.org/10.1016/j.jpeds.2007.02.030
176. Mangeya N, Mafukidze AT, Pascoe M, Mbuwayesango B, Madziva D, Ndlovu N, et al.
Cholangiocarcinoma presenting in an adolescent with vertically acquired HIV infection. Int J STD
AIDS. 2008;19(10):717–8. https://doi.org/10.1258/ijsa.2008.008078
177. Nakamura H, Katayose Y, Rikiyama T, Onogawa T, Yamamoto K, Yoshida H, et al. Advanced bile
duct carcinoma in a 15-year-old patient with pancreaticobiliary maljunction and congenital bili-
ary cystic disease. J Hepatobiliary Pancreat Surg. 2008;15(5):554–9. https://doi.org/10.1007/
s00534-007-1310-x
178. Saikusa N, Naito S, Iinuma Y, Ohtani T, Yokoyama N, Nitta K. Invasive cholangiocarcinoma identified
in congenital biliary dilatation in a 3-year-old boy. J Pediatr Surg. 2009;44(11):2202–5. https://doi.
org/10.1016/j.jpedsurg.2009.06.037
179. Channabasappa N, Johnson-Welch S, Mittal N. De novo cholangiocarcinoma after liver trans-
plantation in a pediatric patient. Pediatr Transplant. 2010;14(8):E110–4. https://doi.org/10.1111/
j.1399-3046.2009.01220.x
180. Deneau M, Adler DG, Schwartz JJ, Hutson W, Sorensen J, Book L, et al. Cholangiocarcinoma in
a 17-year-old boy with primary sclerosing cholangitis and inflammatory bowel disease. J Pediatr
Gastroenterol Nutr. 2011;52(5):617–20. https://doi.org/10.1097/MPG.0b013e3181f9a5d2
181. Huddleston BJ, Lamb RD, Gopez EV, Adler DG, Collins BT. Cholangiocarcinoma in a 17-year-old
boy with primary sclerosing cholangitis and UroVysion fluorescent in situ hybridization. Diagn
Cytopathol. 2012;40(4):337–41. https://doi.org/10.1002/dc.21629
182. Vera A, Villaveces D, Lopez R. Orthotopic liver transplantation for biliary atresia complicated by
incidental cholangiocarcinoma. J Pediatr Gastroenterol Nutr. 2012;55(3):336–7. https://doi.org/
10.1097/MPG.0b013e318233ff17
183. Lai J, Taouli B, Iyer KR, Arnon R, Thung SN, Magid MS, et al. Cholangiolocellular carcinoma
in a pediatric patient with small duct sclerosing cholangitis: a case report. Semin Liver Dis.
2012;32(4):360–6. https://doi.org/10.1055/s-0032-1329904
184. Deneau M, Jensen MK, Holmen J, Williams MS, Book LS, Guthery SL. Primary sclerosing cholangitis,
autoimmune hepatitis, and overlap in Utah children: epidemiology and natural history. Hepatology.
2013;58(4):1392–400. https://doi.org/10.1002/hep.26454
36 Sergi C M
185. Peneau A, Savoye G, Turck D, Dauchet L, Fumery M, Salleron J, et al. Mortality and cancer
in pediatric-onset inflammatory bowel disease: a population-based study. Am J Gastroenterol.
2013;108(10):1647–53. https://doi.org/10.1038/ajg.2013.242
186. Liu R, Cox K, Guthery SL, Book L, Witt B, Chadwick B, et al. Cholangiocarcinoma and high-grade
dysplasia in young patients with primary sclerosing cholangitis. Dig Dis Sci. 2014;59(9):2320–4.
https://doi.org/10.1007/s10620-014-3152-0
187. Yoon HJ, Jeon TY, Yoo SY, Kim JH, Eo H, Lee SK, et al. Hepatic tumours in children with biliary atre-
sia: single-centre experience in 13 cases and review of the literature. Clin Radiol. 2014;69(3):e113–9.
https://doi.org/10.1016/j.crad.2013.10.017
188. Hall C, Mamlok V, Al-Khalil I. A Sporadic Case of Advanced Metastatic Cholangiocarcinoma in a
Child: A Case Report and Review of Literature. J Pediatr Hematol Oncol. 2015;37(5):e333–5. https://
doi.org/10.1097/MPH.0000000000000335
189. Izumi H, Yazawa N, Furukawa D, Masuoka Y, Yamada M, Mashiko T, et al. Bile duct carcinoma associ-
ated with congenital biliary dilatation in a 16-year-old female: a case report and literature review. Surg
Case Rep. 2016;2(1):5. https://doi.org/10.1186/s40792-016-0132-y
190. Tyson GL, El-Serag HB. Risk factors for cholangiocarcinoma. Hepatology. 2011;54(1):173–84. https://
doi.org/10.1002/hep.24351
191. Adenugba A, Khan SA, Taylor-Robinson SD, Cox IJ, Toledano MB, Thillainayagam AV, et al.
Polychlorinated biphenyls in bile of patients with biliary tract cancer. Chemosphere. 2009;76(6):841–6.
https://doi.org/10.1016/j.chemosphere.2009.04.003
192. Burak K, Angulo P, Pasha TM, Egan K, Petz J, Lindor KD. Incidence and risk factors for cholan-
giocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol. 2004;99(3):523–6. https://doi.
org/10.1111/j.1572-0241.2004.04067.x
193. Boberg KM, Bergquist A, Mitchell S, Pares A, Rosina F, Broome U, et al. Cholangiocarcinoma in
primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol.
2002;37(10):1205–11. https://doi.org/10.1080/003655202760373434
194. Khan SA, Davidson BR, Goldin RD, Heaton N, Karani J, Pereira SP, et al. Guidelines for the diag-
nosis and treatment of cholangiocarcinoma: an update. Gut. 2012;61(12):1657–69. https://doi.
org/10.1136/gutjnl-2011-301748
195. Irwin ST, Morison JE. Congenital cyst of common bile-duct containing stones and undergoing cancer-
ous change. Br J Surg. 1944;32:319–21. https://doi.org/10.1002/bjs.18003212614
196. Ferraris LV, Navarro A, Malbran JE. Dilatation congéniata del hepato-colédoco y adenocarcinoma. Bol
Soc Cirug Cordoba. 1944;5:21.
197. Chapman RW. Risk factors for biliary tract carcinogenesis. Ann Oncol. 1999;10 Suppl 4:308–11.
https://doi.org/10.1093/annonc/10.suppl_4.S308
198. Blechacz B. Cholangiocarcinoma: Current Knowledge and New Developments. Gut Liver.
2017;11(1):13–26. https://doi.org/10.5009/gnl15568
199. Blechacz B, Komuta M, Roskams T, Gores GJ. Clinical diagnosis and staging of cholangiocarcinoma.
Nat Rev Gastroenterol Hepatol. 2011;8(9):512–22. https://doi.org/10.1038/nrgastro.2011.131
200. Blechacz BR, Gores GJ. Cholangiocarcinoma. Clin Liver Dis. 2008;12(1):131–50, ix. https://doi.
org/10.1016/j.cld.2007.11.003
201. Todani T, Watanabe Y, Toki A, Morotomi Y. Classification of congenital biliary cystic disease: spe-
cial reference to type Ic and IVA cysts with primary ductal stricture. J Hepatobiliary Pancreat Surg.
2003;10(5):340–4. https://doi.org/10.1007/s00534-002-0733-7
202. Todani T, Watanabe Y, Narusue M, Tabuchi K, Okajima K. Congenital bile duct cysts: Classification,
operative procedures, and review of thirty-seven cases including cancer arising from choledochal cyst.
Am J Surg. 1977;134(2):263–9. https://doi.org/10.1016/0002-9610(77)90359-2
203. Bloustein PA. Association of carcinoma with congenital cystic conditions of the liver and bile ducts.
Am J Gastroenterol. 1977;67(1):40–6.
204. Nagorney DM, McIlrath DC, Adson MA. Choledochal cysts in adults: clinical management. Surgery.
1984;96(4):656–63.
205. Rossi RL, Silverman ML, Braasch JW, Munson JL, ReMine SG. Carcinomas arising in cystic condi-
tions of the bile ducts. A clinical and pathologic study. Ann Surg. 1987;205(4):377–84. https://doi.
org/10.1097/00000658-198704000-00006
Liver Cancer in Children and Adolescents 37
206. Masuhara S, Kasuya K, Aoki T, Yoshimatsu A, Tsuchida A, Koyanagi Y. Relation between K-ras codon
12 mutation and p53 protein overexpression in gallbladder cancer and biliary ductal epithelia in
patients with pancreaticobiliary maljunction. J Hepatobiliary Pancreat Surg. 2000;7(2):198–205.
https://doi.org/10.1007/s005340050176
207. Callea F, Sergi C, Fabbretti G, Brisigotti M, Cozzutto C, Medicina D. Precancerous lesions of the biliary
tree. J Surg Oncol Suppl. 1993;3:131–3. https://doi.org/10.1002/jso.2930530535
208. Sergi C, Adam S, Kahl P, Otto HF. Study of the malformation of ductal plate of the liver in Meckel
syndrome and review of other syndromes presenting with this anomaly. Pediatr Dev Pathol.
2000;3(6):568–83. https://doi.org/10.1007/s100240010104
209. Sergi C, Adam S, Kahl P, Otto HF. The remodeling of the primitive human biliary system. Early Hum
Dev. 2000;58(3):167–78. https://doi.org/10.1016/S0378-3782(00)00065-7
210. Sergi C, Kahl P, Otto HF. Contribution of apoptosis and apoptosis-related proteins to the malformation
of the primitive intrahepatic biliary system in Meckel syndrome. Am J Pathol. 2000;156(5):1589–98.
https://doi.org/10.1016/S0002-9440(10)65031-6
211. Desmet VJ. Congenital diseases of intrahepatic bile ducts: variations on the theme “ductal plate mal-
formation”. Hepatology. 1992;16(4):1069–83. https://doi.org/10.1002/hep.1840160434
212. Desmet VJ. What is congenital hepatic fibrosis? Histopathology. 1992;20(6):465–77. https://doi.
org/10.1111/j.1365-2559.1992.tb01031.x
213. Desmet VJ. Ductal plates in hepatic ductular reactions. Hypothesis and implications. III. Implications
for liver pathology. Virchows Arch. 2011;458(3):271–9. https://doi.org/10.1007/s00428-011-1050-9
214. Roskams T, Desmet V. Embryology of extra- and intrahepatic bile ducts, the ductal plate. Anat Rec
(Hoboken). 2008;291(6):628–35. https://doi.org/10.1002/ar.20710
215. Van Eyken P, Sciot R, Callea F, Van der Steen K, Moerman P, Desmet VJ. The development of the intra-
hepatic bile ducts in man: a keratin-immunohistochemical study. Hepatology. 1988;8(6):1586–95.
https://doi.org/10.1002/hep.1840080619
216. Hammar JA. Uber die erste Entstehung der nicht kapillaren intrahepatischen Gallengange beim
Menschen. Z Mikrosk Anat Forsch. 1926;5:59–89.
217. Lee KB. Histopathology of a benign bile duct lesion in the liver: Morphologic mimicker or precursor
of intrahepatic cholangiocarcinoma. Clin Mol Hepatol. 2016;22(3):400–5. https://doi.org/10.3350/
cmh.2016.0105
218. Zheng W, Shen F, Hu R, Roy B, Yang J, Wang Q, et al. Far Upstream Element-Binding Protein 1
Binds the 3’ Untranslated Region of PKD2 and Suppresses Its Translation. J Am Soc Nephrol.
2016;27(9):2645–57. https://doi.org/10.1681/ASN.2015070836
219. Johnson CA, Gissen P, Sergi C. Molecular pathology and genetics of congenital hepatorenal fibrocystic
syndromes. J Med Genet. 2003;40(5):311–9. https://doi.org/10.1136/jmg.40.5.311
220. Wills ES, Roepman R, Drenth JP. Polycystic liver disease: ductal plate malformation and the primary
cilium. Trends Mol Med. 2014;20(5):261–70. https://doi.org/10.1016/j.molmed.2014.01.003
221. Kida T, Nakanuma Y, Terada T. Cystic dilatation of peribiliary glands in livers with adult polycystic
disease and livers with solitary nonparasitic cysts: an autopsy study. Hepatology. 1992;16(2):334–40.
https://doi.org/10.1002/hep.1840160209
222. Sugawara T, Shindoh J, Hoshi D, Hashimoto M. Intrahepatic cholangiocarcinoma and portal
hypertension developing in a patient with multicystic biliary microhamartomas. Malays J Pathol.
2018;40(3):331–5.
223. Lendvai G, Szekerczes T, Illyes I, Dora R, Kontsek E, Gogl A, et al. Cholangiocarcinoma: Classification,
Histopathology and Molecular Carcinogenesis. Pathol Oncol Res. 2018. https://doi.org/10.1007/
s12253-018-0491-8
224. Bhalla A, Mann SA, Chen S, Cummings OW, Lin J. Histopathological evidence of neoplastic progres-
sion of von Meyenburg complex to intrahepatic cholangiocarcinoma. Hum Pathol. 2017;67:217–24.
https://doi.org/10.1016/j.humpath.2017.08.004
225. Tsokos CG, Krings G, Yilmaz F, Ferrell LD, Gill RM. Proliferative index facilitates distinction between
benign biliary lesions and intrahepatic cholangiocarcinoma. Hum Pathol. 2016;57:61–7. https://doi.
org/10.1016/j.humpath.2016.06.019
226. Ettel M, Eze O, Xu R. Clinical and biological significance of precursor lesions of intrahepatic cholan-
giocarcinoma. World J Hepatol. 2015;7(25):2563–70. https://doi.org/10.4254/wjh.v7.i25.2563
38 Sergi C M
227. Nakanuma Y, Tsutsui A, Ren XS, Harada K, Sato Y, Sasaki M. What are the precursor and early
lesions of peripheral intrahepatic cholangiocarcinoma? Int J Hepatol. 2014;2014:805973. https://doi.
org/10.1155/2014/805973
228. Kim HK, Jin SY. Cholangiocarcinoma arising in von Meyenburg complexes. Korean J Hepatol.
2011;17(2):161–4. https://doi.org/10.3350/kjhep.2011.17.2.161
229. Jain D, Nayak NC, Saigal S. Hepatocellular carcinoma arising in association with von-Meyenburg’s
complexes: an incidental finding or precursor lesions? A clinicopatholigic study of 4 cases. Ann Diagn
Pathol. 2010;14(5):317–20. https://doi.org/10.1016/j.anndiagpath.2010.04.003
230. Heinke T, Pellacani LB, Costa Hde O, Fuziy RA, Franco M. Hepatocellular carcinoma in associa-
tion with bile duct hamartomas: report on 2 cases and review of the literature. Ann Diagn Pathol.
2008;12(3):208–11. https://doi.org/10.1016/j.anndiagpath.2006.12.003
231. Jain D, Sarode VR, Abdul-Karim FW, Homer R, Robert ME. Evidence for the neoplastic transfor-
mation of Von-Meyenburg complexes. Am J Surg Pathol. 2000;24(8):1131–9. https://doi.org/
10.1097/00000478-200008000-00011
232. Blanc JF, Bernard PH, Carles J, Le Bail B, Balabaud C, Bioulac-Sage P. Cholangiocarcinoma arising in
Von Meyenburg complex associated with hepatocellular carcinoma in genetic haemochromatosis. Eur
J Gastroenterol Hepatol. 2000;12(2):233–7. https://doi.org/10.1097/00042737-200012020-00016
233. Sasaki M, Sato Y, Nakanuma Y. Cholangiolocellular Carcinoma With “Ductal Plate Malformation”
Pattern May Be Characterized by ARID1A Genetic Alterations. Am J Surg Pathol. 2019;43(3):352–60.
https://doi.org/10.1097/PAS.0000000000001201
234. Nakanuma Y, Kakuda Y. Pathologic classification of cholangiocarcinoma: New concepts. Best Pract
Res Clin Gastroenterol. 2015;29(2):277–93. https://doi.org/10.1016/j.bpg.2015.02.006
235. Cooper D, Schermer A, Sun TT. Classification of human epithelia and their neoplasms using mono-
clonal antibodies to keratins: strategies, applications, and limitations. Lab Invest. 1985;52(3):243–56.
236. Sun TT, Tseng SC, Huang AJ, Cooper D, Schermer A, Lynch MH, et al. Monoclonal antibody studies
of mammalian epithelial keratins: a review. Ann N Y Acad Sci. 1985;455:307–29. https://doi.org/
10.1111/j.1749-6632.1985.tb50419.x
237. Treem WR, Krzymowski GA, Cartun RW, Pedersen CA, Hyams JS, Berman M. Cytokeratin immunohis-
tochemical examination of liver biopsies in infants with Alagille syndrome and biliary atresia. J Pediatr
Gastroenterol Nutr. 1992;15(1):73–80. https://doi.org/10.1097/00005176-199207000-00011
238. AJCC. AJCC Cancer Staging Manual. 8 ed. New York: Springer; 2016.
239. Sempoux C, Jibara G, Ward SC, Fan C, Qin L, Roayaie S, et al. Intrahepatic cholangiocarcinoma: new
insights in pathology. Semin Liver Dis. 2011;31(1):49–60. https://doi.org/10.1055/s-0031-1272839
240. Nakanuma Y, Sasaki M, Sato Y, Ren X, Ikeda H, Harada K. Multistep carcinogenesis of perihilar
cholangiocarcinoma arising in the intrahepatic large bile ducts. World J Hepatol. 2009;1(1):35–42.
https://doi.org/10.4254/wjh.v1.i1.35
2
Hepatocellular Carcinoma in Adults
Aducio L. Thiesen
Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB,
Canada
Author for correspondence: Aducio L. Thiesen, Department of Laboratry Medicine and
Pathology, University of Alberta Hospital, Edmonton, Alberta, Canada.
Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch2
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
39
40 Thiesen A L
INTRODUCTION
The hepatocytes are active metabolic cells involved in many cell functions and are
subject to a large number of insults that may result in abnormal proliferation (8).
Exhaustion of their regenerative capacity is typically exemplified by cirrhosis.
Thus, the etiologic factors involved in the cirrhotic process are also involved in
tumorigenesis of hepatocellular carcinoma. Chronic hepatitis B and C are by far
the most common causes of hepatocellular carcinoma, followed by alcoholic liver
disease and nonalcoholic steatohepatitis. (9–11). Hepatitis B DNA levels in excess
of 200,000 IU/mL (1,000,000 copies/mL) have been reported to increase the inci-
dence of hepatocellular carcinoma to 1,152 per 100,000 individuals (12, 13).
Other etiologic factors in adults include aflatoxin-contaminated food, diabetes,
obesity, and hemochromatosis (14, 15). In some instances, hepatocellular carci-
noma is detected incidentally during routine image examination. These tumors
are considered sporadic and are result of gene mutations that occur during a per-
son’s lifetime.
Genetic susceptibility to hepatocellular carcinoma has been demonstrated
in animal models, but not established in humans. Family clustering has been
Hepatocellular Carcinoma in Adults 41
reported in some Chinese families and Alaskan natives, but infectious hepatitis B
is a requirement for HCC development in this particular population (16, 17).
Other etiologic factors include conditions associated with congenital, genetic,
and metabolic conditions and are directly related to the capacity of these patholo-
gies to develop cirrhosis. Such examples include hemochromatosis, Wilson’s dis-
ease, alpha-1-antitrypsin deficiency and Budd-Chiari syndrome.
Alcoholic liver disease is a risk factor that illustrates regeneration as the key
factor in tumorigenesis. Active intake of alcohol causes hepatocyte injury with
minimal regeneration, which clinically is a low risk factor for hepatocellular car-
cinoma. In contrast, alcohol abstinence results in significant regeneration and a
higher risk of hepatocellular carcinoma (18).
Once the hepatitis B virus is integrated into the hepatocytes, transactivation of
proto-oncogenes, activation of growth factors, and inactivation of tumor suppres-
sor genes may result in abnormal proliferation (19). The hepatitis C virus damages
double-stranded DNA, increasing the frequency of mutations in genes such as
immunoglobulin genes, BCL-6, TP53, and β-catenin, causing abnormal prolifera-
tion (20). Other common mutations encountered in hepatocellular carcinoma
include: telomerase promoter mutations (30 to 60% of HCC) (21); TP53 muta-
tions (18 to 50%) (22) classically associated to exposure to aflatoxin; beta-catenin
(18 to 40%); AXIN1 and AXIN2 genes; chromatin remodeling pathway (ARID1A
and ARID2); Janus kinase (JAK)/signal transducers and activators of transcription
(STAT) pathway (JAK1, IL6R, and IL6ST); genes involved in ubiquitination
(KEAP1); genes involved in RAS/MAPK signaling (RPS6KA3); and genes involved
the oxidative stress pathway (NFE2L2) (23).
At the cellular level, hepatocellular carcinomas are thought to arise from a dys-
plastic focus (less than 1 mm) that develops into either a low-grade or a high-
grade dysplastic nodule. The high-grade dysplastic nodule, more frequently than
the low-grade dysplastic nodule, progresses to hepatocellular carcinoma (24).
Since the primordial lesion (dysplastic focus) is less than 1 mm, it is not identifi-
able by radiologic studies and usually observed in hepatectomy specimens as focal
cytological atypia within the hepatocytes. Morphologically, the atypia within the
hepatocytes is described as small and large cell changes. The small cell change is
characterized by increased nuclear to cytoplasmic ratio, hyperchromasia and
cytoplasmic basophilia, giving the impression of crowding. Small change atypia is
associated with high-grade dysplastic nodules and a higher risk of developing of
hepatocellular carcinoma. The large cell change is characterized by enlarged cells
with larger nuclei and cytoplasm, but the nuclear to cytoplasmic ratio is pre-
served; frequent multinucleation, nuclear polymorphism and hyperchromasia are
common. Large cell changes are the predominant alteration in low grade dysplas-
tic nodules and indicate a benign feature (25). The biggest diagnostic challenge is
to differentiate a high-grade dysplastic nodule from a well-differentiated hepato-
cellular carcinoma. Reticulin stain with delineation of the hepatic plates is a reli-
able ancillary technique in this context. Preservation of the hepatic plate is typical
42 Thiesen A L
Figure 1. Hematoxylin and Eosin stain 200X. On the left, normal hepatocytes with normal
nuclear cytoplasmic ratio as compared to hepatocellular carcinoma (on the right) with
increased nuclear cytoplasmic ratio, organized into thick trabeculae and few
pesudoglandular structures with bile.
Hepatocellular Carcinoma in Adults 43
Figure 2. Reticulin stain 200X. On the left, normal hepatocytes with one to two cell thick
hepatic plates as compared to hepatocellular carcinoma (on the right) with distended
hepatic plates.
and danazol (synthetic androgen used for treatment of endometriosis) (31). At the
molecular level, hepatocellular adenomas are subdivided based on HNF1a muta-
tions (steatotic adenomas), IL-6 ⁄ STAT3 mutations (inflammatory adenomas), and
β-catenin mutations (subgroup of inflammatory adenomas and unclassified adeno-
mas with cytological atypia) (32). Prognostic implications justify such subtyping
with inflammatory adenomas being frequently associated with metabolic syn-
drome, liver steatosis and alcohol exposure (33), and the β-catenin mutated adeno-
mas immunophenotypically represented by nuclear staining associated with a
higher risk of malignancy. Extensive literature and classification of hepatocellular
adenomas is available, but the key is the identification of the lesions with great risk
of progression to hepatocellular carcinoma.
However, symptoms related to predisposing risk factors (cirrhosis and viral infec-
tions) are frequent, such as ascites and esophageal varices. Paraneoplastic syn-
dromes may be occasionally associated with hepatocellular carcinoma, such as
hypoglycemia (35), erythrocytosis (36), hypercalcemia (37) diarrhea (38) and
cutaneous lesions (39). Other less common presentations include fever, infec-
tions, obstructive jaundice, and hemorrhage due to tumor rupture. Lymph node
involvement is not common, and 10 to 15% of the cases present with advanced
disease and metastases to lung, bone, and adrenal gland (40, 41).
Serological markers of the disease include alpha fetoprotein (AFP), heat shock
protein, human cervical cancer oncogene, human telomerase reverse transcriptase
mRNA, certain cytokines and microRNAs. AFP is by far the most used due to its
availability, but screening limitations are significant due to its low sensitivity (41
to 65%). Pregnancy and primary liver and gastrointestinal disease also raise the
levels of AFP, giving false positive results (42). The most accepted cut-off value is
a serum concentration of 20 ng/mL (43). However, the most valuable indication
of AFP is in the follow-up of previously treated hepatocellular carcinoma to detect
the risk of recurrent disease (44). Three different glycoforms of AFP have been
identified: AFP-L1, AFP-L2 and AFP-L3. The latter has been reported to be associ-
ated with more aggressive liver disease and consequently worse prognosis (45, 46).
Glypican-3, a plasma membrane protein involved in the interaction with growth
factors, has been suggested as an adjuvant serologic marker in addition to AFP to
increase the sensitivity of detecting hepatocellular carcinoma (47, 48). Other liver
enzymes have also been proposed in association with AFP to promote screening
for hepatocellular carcinoma such as gamma-glutamyl transferase, Alpha-l-
fucosidase and Des-gamma-carboxyprothrombin. Alternatively, instead of protein
levels, mRNA levels of AFP and GGT may also be helpful prognostic factor after
initial therapy for HCC (49, 50).
Additional serological markers in conjunction with AFP have been proposed
to increase the sensitivity of detection such as alpha-L-fucosidase (AFU) and
transforming growth factors alpha and beta (TGF-α and TGF-β) (51)
Radiologic diagnosis of hepatocellular carcinoma has significant implications
for treatment options, with dysplastic nodules conservatively followed up based
on the fact that one third of these lesions may progress to hepatocellular carci-
noma; and early hepatocellular carcinoma should be treated with possible curative
options such as ablation, resection and transplant (52). The image modality typi-
cally used is ultrasonography, likely due to the availability, but the sensitivity of
65% and specificity of 90% justify the use of more advanced image studies (53).
In referral centers, the image protocol includes four-phase multidetector com-
puted tomography (CT) or dynamic contrast enhanced magnetic resonance imag-
ing (MRI), complementing each other, when suspicious features are noted (54).
The distinction and detection of hepatocellular carcinoma is based on changes of
vascularization including the presence of unpaired arteries and capillarization of
the sinusoids that can be observed by image studies (55, 56). The definite radio-
logic criteria for the diagnosis of hepatocellular carcinoma are defined by contrast
hyperenhancement at the arterial phase and hypoenhancement at the venous
phase for lesions larger than 2 cm (57, 58). This criterion is highly specific and has
replaced biopsy since 2000 (59). For lesions less than 2 cm, there is intense debate
regarding the need of a biopsy, or not to define the lesion. Lesions less than 1 cm
tend to be biopsied but there is variation in practice among centers (60).
Hepatocellular Carcinoma in Adults 45
Figure 3. Immunohistochemistry for Glypican- 3 200X. On the left, normal hepatocytes with
no staining (brown pigmentation) as compared to hepatocellular carcinoma (on the right)
with cytoplasmic and few nuclei staining positive (brown discoloration).
Figure 4. Immunohistochemistry for CD34 200X. On the left, normal hepatocytes with no
staining (brown discoloration) of the sinusoids as compared to hepatocellular carcinoma
(on the right) with positivity within the sinusoids indicating capillarization.
Hepatocellular Carcinoma in Adults 47
STAGING
Tumor
T0 No tumor
T1 Solitary tumor smaller or larger than 2 cm without vascular invasion
T1a Solitary tumor smaller or equal to 2 cm
T1b Solitary tumor larger than 2 cm without vascular invasion
T2 Solitary tumor larger than 2 cm with vascular invasion; or multiple tumors, none
larger than 5 cm
T3 Multiple tumors, at least one of which is larger than 5 cm
T4 Single tumor or multiple tumors of any size involving a major branch of the portal
vein or hepatic vein; Tumor(s) with direct invasion of adjacent organs other than
the gallbladder; Perforation of visceral peritoneum
Node
N0 No nodal metastasis
N1 Regional lymph node metastasis
Metastasis
M0 No distant metastasis
M1 Distant metastasis
Stage
IA T1AN0M0
Stage IB T1BN0M0
Stage II T2N0M0
Stage IIIA T3N0M0
Stage IIIB T4N0M0
Stage IVA Any T, N1, M0
Stage IVB Any T, Any N, M1
48 Thiesen A L
Okuda
Parameters Criteria Pointsa
Tumor size by imaging >50% 1
Cross-sectional area <50% 0
Ascites Present 1
Absent 0
Serum Albumin (mg/dl) >3 0
<3 1
Serum total bilirubin (mg/dl) <3 0
>3 1
CLIP
Child-Pugh class A 0
B 1
C 2
Tumor morphology Single nodule <50% area 0
Multiple nodules <50% area 1
Massive >50% area 2
AFP (ng/mL) <400 0
≥400 1
Portal vein thrombosis No 0
Yes 1
aTotal
points = Stage.
Performance
Stage Status Tumor Stage Liver Function
A= Early A1 0 Single, <5 cm No portal HTN and normal
bilirubin
A2 0 Single, <5 cm Portal HTN and normal bilirubin
A3 0 Single, <5 cm Portal HTN and elevated bilirubin
A4 0 3 tumors <3 cm Child-Pugh class A-B
B = Intermediate 0 Large multinodular Child-Pugh class A-B
C = Advanced 1 and 2 vascular invasion Child-Pugh class A-B
extrahepatic
spread
D = End-stage 3 and 4 Any Child-Pugh class C
TREATMENT
The therapeutic options for HCC consist of liver transplantation, hepatic resec-
tion, ablation, chemoembolization and chemotherapy. The best available treat-
ment is transplantation removing the entire tumor and replacing the diseased liver
(usually cirrhotic) by a new functional healthy organ. However, due to limitations
regarding availability of donor organs other alternatives should be considered.
Living donor transplantation is an alternative option that has been reported to
have similar results in terms of survival rate (78). Contraindications for liver
transplant include extrahepatic spread, multiple tumors with one of the lesions
larger than 3 cm and a single tumor larger than 5 cm emphasizing the need for
proper staging (79–82). Up to 75% of the patients that received a transplant, pre-
viously had received transarterial chemoembolization (TACE) and one third of
this same population had previous radiofrequency ablation (RFA) preventing pro-
gression of disease and winning time until a donor is available (83). If no evidence
of medical liver disease and/or cirrhosis is documented, the patient may be offered
partial hepatectomy as curative therapy (84). The three-year disease-free survival
in cirrhotic patients treated with partial hepatectomy is 18%, whereas it is 83% in
patients treated by transplantation (85).
CONCLUSION
serologic markers, mostly AFP and radiologic studies. Staging systems attempt
to subdivide the cases of hepatocellular carcinoma in subgroups to predict
behavior and prognosis as well as orientate therapy. Curative therapy may be
achieved with either resection and/or transplant. Adjuvant therapy such and
TACE and RFA are offered as palliative care in attempt to win some time until a
donor liver is available.
Copyright and permission statement: The author confirms that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Katyal S, Oliver III JH, Peterson MS, Ferris JV, Carr BS, Baron RL. Extrahepatic Metastases of Hepatocellular
Carcinoma. Radiol. 2000; Sep 216(3). https://doi.org/10.1148/radiology.216.3.r00se24698
2. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018:
GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA
Cancer J Clin. 2018;68(6):394–424. https://doi.org/10.3322/caac.21492
3. El-Serag HB, Kanwal F. Epidemiology of hepatocellular carcinoma in the United States: where are we?
Where do we go? Hepatology. 2014;60(5):1767–75. https://doi.org/10.1002/hep.27222
4. Patti AM, Santi AL, Pompa MG, Giustini C, Vescia N, Mastroeni I, et al. Viral hepatitis and drugs: a
continuing problem. Int J Epidemiol. 1993;22(1):135–9. https://doi.org/10.1093/ije/22.1.135
5. Bertuccio P, Turati F, Carioli G, Rodriguez T, La Vecchia C, Malvezzi M, et al. Global trends and
predictions in hepatocellular carcinoma mortality. J Hepatol. 2017;67(2):302–9. https://doi.
org/10.1016/j.jhep.2017.03.011
6. Tang A, Hallouch O, Chernyak V, Kamaya A, Sirlin CB. Epidemiology of hepatocellular carci-
noma: target population for surveillance and diagnosis. Abdom Radiol. 2018;43:13–25. https://doi.
org/10.1007/s00261-017-1209-1
7. Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000; Int J
Cancer. 2001;94:153–6. https://doi.org/10.1002/ijc.1440
8. Zhang W, Shi J, Li R, Han Z, Li L, Li G, et al. Effectiveness of Olaparib Treatment in a Patient with
Gallbladder Cancer with an ATM -Inactivating Mutation. Oncologist. 2020;25:375–9. https://doi.
org/10.1634/theoncologist.2019-0498
9. Brar TS, Hilgenfeldt E, Soldevila-Pico C. Etiology and Pathogenesis of Hepatocellular Carcinoma. In:
Liu C, editor. Precision Molecular Pathology of Liver Cancer. Molecular Pathology Library. Springer,
Cham; 2018.p 1–15. https://doi.org/10.1007/978-3-319-68082-8_1
10. Poon D, Anderson BO, Chen LT, Tanaka K, Lau WY, Cutsem EV et al. Management of hepatocel-
lular carcinoma in Asia: consensus statement from the Asian Oncology Summit 2009. Lancet Oncol.
2009;10(11):1111–8. https://doi.org/10.1016/S1470-2045(09)70241-4
11. Shen T, Liu Y, Shang J, Xie Q, Li J, Yan M et al. Incidence and Etiology of Drug-Induced Liver Injury
in Mainland China. Gastroenterology. 2019;156(8):2230–41.
12. Chen C, Yang H, Su J, Jen CL, You SL, Lu SN, et al. Risk of Hepatocellular Carcinoma Across a
Biological Gradient of Serum Hepatitis B Virus DNA Level. JAMA. 2006;295(1):65–73. https://doi.
org/10.1001/jama.295.1.65
13. Chen CJ, Yang HI, Iloeje UH. Hepatitis B virus DNA levels and outcomes in chronic hepatitis B.
Hepatology. 2009;49:S72-S84. https://doi.org/10.1002/hep.22884
Hepatocellular Carcinoma in Adults 51
14. Gomaa AI, Khan SA, Toledano MB, Waked I, Taylor-Robinson SD. Hepatocellular carcinoma: epi-
demiology, risk factors and pathogenesis. World J Gastroenterol. 2008;14(27):4300–8. https://doi.
org/10.3748/wjg.14.4300
15. Montalto G, Cervello M, Giannitrapani L, Dantona F, Teraanova A, Castagnetta LAM. Epidemiology,
Risk Factors, and Natural History of Hepatocellular Carcinoma. Ann NY Acad Sci. 2002;963:13–20.
https://doi.org/10.1111/j.1749-6632.2002.tb04090.x
16. Alberts SR, Lanier AP, McMahon BJ. Clustering of hepatocellular carcinoma in Alaska native families.
Genet Epidemiol. 1991;8:127–39. https://doi.org/10.1002/gepi.1370080206
17. Shen FM, Lee MK, Gong HM. Complex segregation analysis of primary hepatocellular carcinoma
in Chinese families: interaction of inherited susceptibility and hepatitis B viral infection. Am J Hum
Genet. 1991;49:88. 18. Leong TY, Leong AS. Epidemiology and carcinogenesis of hepatocellular car-
cinoma. HPB (Oxford) 2005;7(1):5–15. https://doi.org/10.1080/13651820410024021
19. Blum HE, Offensperger WB, Walter E, Offensperger S, Wahl A, Zeschnigk C, et al. Hepatocellular
carcinoma and hepatitis B virus infection: molecular evidence for monoclonal origin and expansion
of malignantly transformed hepatocytes. J Cancer Res Clin Oncol. 1987;113:466–72. https://doi.
org/10.1007/BF00390041
20. Machida K, Cheng KT, Sung VM, Shimodaira S, Lindsay KL, Levine AM, et al. Hepatitis C virus
induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proc
Natl Acad Sci. U S A 2004;101(12):4262–7. https://doi.org/10.1073/pnas.0303971101
21. Nault J, Mallet M, Pilati C, Calderaro J, Bioulac-Sage P, Laurent C, et al. High frequency of telomer-
ase reverse-transcriptase promoter somatic mutations in hepatocellular carcinoma and preneoplastic
lesions. Nat Commun. 2013;4:2218. https://doi.org/10.1038/ncomms3218
22. Hussain S, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma:
insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26(15):2166–76. https://
doi.org/10.1038/sj.onc.1210279
23. Villanueva A, Llovet JM. Liver cancer in 2013: Mutational landscape of HCC-the end of the beginning.
Nat Rev Clin Oncol. 2014;11(2):73–4. https://doi.org/10.1038/nrclinonc.2013.243
24. Dhanasekaran R, Bandoh S, Roberts LR. Molecular pathogenesis of hepatocellular carcinoma and
impact of therapeutic advances. F1000Research 2016, 5(F1000 Faculty Rev):879. https://doi.
org/10.12688/f1000research.6946.1
25. Giannini A, Zampi G, Bartoloni F, Omer S. Morphological precursors of hepatocellular carcinoma: a
morphometrical analysis. Hepatogastroenterology. 1987;34(3):95–7.
26. Chou CT, Chou JM, Chang TA, Huang SF, Chen CB, Chen YL et al. Differentiation between dysplastic
nodule and early-stage hepatocellular carcinoma: the utility of conventional MR imaging. World J
Gastroenterol. 2013;19(42):7433–9. https://doi.org/10.3748/wjg.v19.i42.7433
27. Roncalli M. Hepatocellular nodules in cirrhosis: Focus on diagnostic criteria on liver biopsy. A Western
experience. Liver Transpl. 2004;10:S9-S15. https://doi.org/10.1002/lt.20047
28. Barthelmes L,Tait IS. Liver cell adenoma and liver cell adenomatosis. HPB. 2005;7:186–96. https://
doi.org/10.1080/13651820510028954
29. Carrasco D, Barrachina M, Prieto M, Berenguer J. Clomiphene citrate and liver-cell adenoma. N Engl
J Med. 1984;310(7):1120–1. https://doi.org/10.1056/NEJM198404263101716
30. Coombes GB, Reiser J, Paradinas FJ, Burn I. An androgen-associated hepatic adenoma in a trans-
sexual. Brit J Surg. 1978;65(2):869–70. https://doi.org/10.1002/bjs.1800651212
31. Fermand JP, Levy Y, Bouscary D, D’Agay MF, Clot P, Frija J, Brouet JC. Danazol-induced hepatocellular
adenoma. Am J Med. 1990;88(5):529–30. https://doi.org/10.1016/0002-9343(90)90434-F
32. Bellamy COC, Maxwell RS, Prost S, Azodo IA, Powell JJ, Manning JR. The value of immunophenotyping
hepatocellular adenomas: consecutive resections at one UK centre. Histopathology. 2013;62:431–45.
https://doi.org/10.1111/his.12011
33. Bioulac-Sage P, Laumonier H, Couchy G, Le Bail B, Sa Cunha A, Rullier A et al. Hepatocellular adenoma
management and phenotypic classification: the Bordeaux experience. Hepatology. 2009;50:481–9.
https://doi.org/10.1002/hep.22995
34. Honda T, Miyaaki H, Ichikawa T, Taura N, Miuma S, Shibata H et al. Clinical characteristics of
hepatocellular carcinoma in elderly patients. Onco Lett. 2011;2(5):851–4. https://doi.org/10.3892/
ol.2011.359
52 Thiesen A L
35. Juturi JV, Hopkins T, Farhangi M. Severe leukocytosis with neutrophilia (leukemoid reaction)
in alcoholic steatohepatitis. Am J Gastroenterol. 1998;93(6):1013. https://doi.org/ 10.1111/j.
1572-0241.1998.1013a.x
36. Kew MC, Fisher JW. Serum erythropoietin concentrations in patients with hepatocellular
carcinoma. Cancer. 1986;58(11):2485–88. https://doi.org/10.1002/1097-0142(19861201)58:11
<2485::AID-CNCR2820581122>3.0.CO;2-N
37. Yen TC, Wang CC, Lee SD, Yen SH. Hypercalcemia and parathyroid hormone-related protein in
hepatocellular carcinoma. Liver. 1993;13(6):311. https://doi.org/10.1111/j.1600-0676.1993.
tb00651.x
38. Solinas A, Biscarini L, Morrelli A, Favero AD. Hepatocellular Carcinoma and the Watery Diarrhea
Syndrome. Arch Surg. 1988;123(1):124. https://doi.org/10.1001/archsurg.1988.01400250134033
39. Gregory B, Ho VC. Cutaneous manifestations of gastrointestinal disorders. Part II. J Am Acad
Dermatol. 1992;26(3 Pt 2):371–383. https://doi.org/10.1016/0190-9622(92)70059-O
40. Jiang X, Ke C, Zhang G, Zhang X, Sai K, Chen Z. Brain metastases from hepatocellular car-
cinoma: clinical features and prognostic factors. BMC Cancer. 2012;12:49. https://doi.org/
10.1186/1471-2407-12-49
41. Harding JJ, Abu-Zeinah G, Chou JF, Owen DH, Ly M, Lowery MA et al. Frequency, Morbidity, and
Mortality of Bone Metastases in Advanced Hepatocellular Carcinoma. J Natl Compr Canc Netw.
2018;16(1):50–8. https://doi.org/10.6004/jnccn.2017.7024
42. Zhao YJ et al. Tumor markers for hepatocellular carcinoma. Mol Clin Oncol. 2013;1(4):593–8. https://
doi.org/10.3892/mco.2013.119
43. Zhou L, Liu J, Luo F. Serum tumor markers for detection of hepatocellular carcinoma. World J
Gastroenterol. 2006;12(8):1175–81. https://doi.org/10.3748/wjg.v12.i8.1175
44. Tan CK, Law NM, Ng HS, Machin D. Simple clinical prognostic model for hepatocellular carcinoma
in developing countries and its validation. J Clin Oncol. 2003;21:2294–8. https://doi.org/10.1200/
JCO.2003.03.151
45. Khien VV, Mao HV, Chinh TT, Ha PT, Bang MH, Lac BV et al. Clinical evaluation of lentil lectin-
reactive alpha-fetoprotein-L3 in histology-proven hepatocellular carcinoma. Int J Biol Markers.
2001;16:105–111. https://doi.org/10.1177/172460080101600204
46. Oka H, Saito A, Ito K, Kumada T, Satomura S, Kasugai H et al. Multicenter prospective analy-
sis of newly diagnosed hepatocellular carcinoma with respect to the percentage of Lens culinaris
agglutinin-reactive alpha-fetoprotein. J Gastroenterol Hepatol. 2001;16:1378–83. https://doi.
org/10.1046/j.1440-1746.2001.02643.x
47. Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E et al. Glypican-3: a novel serum and
histochemical marker for hepatocellular carcinoma. Gastroenterology. 2003;125:89–97. https://doi.
org/10.1016/S0016-5085(03)00689-9
48. Nakatsura T, Yoshitake Y, Senju S, Monji M, Komori H, Motomura Y et al. Glypican-3, overexpressed
specifically in human hepatocellular carcinoma, is a novel tumor marker. Biochem Biophys Res
Commun. 2003;306:16–25. https://doi.org/10.1016/S0006-291X(03)00908-2
49. Minata M, Nishida N, Komeda T, Azechi H, Katsuma H, Nishimura T et al. Postoperative detection
of alpha-fetoprotein mRNA in blood as a predictor for metastatic recurrence of hepatocellular carci-
noma. J Gastroenterol Hepatol. 2001;16:445–51. https://doi.org/10.1046/j.1440-1746.2001.02461.x
50. Ijichi M, Takayama T, Matsumura M, Shiratori Y, Omata M, Makuuchi M. alpha-Fetoprotein mRNA
in the circulation as a predictor of postsurgical recurrence of hepatocellular carcinoma: a prospective
study. Hepatology. 2002;35:853–60. https://doi.org/10.1053/jhep.2002.32100
51. El-Tayeh SF, Hussein TD, El-Houseini ME, Amer MA, El-Sherbini M, Elshemey WM. Serological bio-
markers of hepatocellular carcinoma in Egyptian patients. Dis Markers. 2012;32(4):255–63. https://
doi.org/10.1155/2012/962320
52. Bargellini I, Battaglia V, Bozzi E, Lauretti DL, Lorenzoni G, Bartolozzi C. Radiological diagnosis of
hepatocellular carcinoma. J Hepatocell Carcinoma. 2014;1:137–48. https://doi.org/10.2147/JHC.
S44379
53. Singal A, Volk ML, Waljee A, Salgia R, Higgins P, Rogers MA et al. Meta-analysis: surveillance with
ultrasound for early-stage hepatocellular carcinoma in patients with cirrhosis. Aliment Pharmacol
Ther. 2009;30:37–47. https://doi.org/10.1111/j.1365-2036.2009.04014.x
Hepatocellular Carcinoma in Adults 53
54. Waghray A, Murali AR, Menon KN. Hepatocellular carcinoma: From diagnosis to treatment. World J
Hepatol. 2015;7(8):1020–9. https://doi.org/10.4254/wjh.v7.i8.1020
55. Himeno H, Enzan H, Saibara T, Onishi S, Yamamoto Y. Hitherto unrecognized arterioles within hepa-
tocellular carcinoma. J Pathol. 1994;174:217–22. https://doi.org/10.1002/path.1711740311
56. Kin M, Torimura T, Ueno T, Inuzuka S, Tanikawa K. Sinusoidal capillarization in small hepatocellular
carcinoma. Pathol Int 1994;44:771–8. https://doi.org/10.1111/j.1440-1827.1994.tb02925.x
57. Forner A, Vilana R, Ayuso C, Bianchi L, Solé M, Ayuso JR et al. Diagnosis of hepatic nodules 20 mm
or smaller in cirrhosis: prospective validation of the noninvasive diagnostic criteria for hepatocellular
carcinoma. Hepatology. 2008;47:97–104. https://doi.org/10.1002/hep.21966
58. Leoni S, Piscaglia F, Golfieri R, Camaggi V, Vidili G, Pini P et al. The impact of vascular and non-
vascular findings on the noninvasive diagnosis of small hepatocellular carcinoma based on the
EASL and AASLD criteria. Am J Gastroenterol. 2010;105(3):599–609. https://doi.org/10.1038/
ajg.2009.654
59. International Consensus Group for Hepatocellular Neoplasia Pathologic diagnosis of early hepa-
tocellular carcinoma: a report of the International Consensus Group for Hepatocellular Neoplasia.
Hepatology. 2009;49:658–64. https://doi.org/10.1002/hep.22709
60. Bargellini I, Battaglia V, Bozzi E, Lauretti DL, Lorenzoni G, Bartolozzi C. Radiological diagnosis of hepa-
tocellular carcinoma. J Hepatocell Carcinoma. 2014;1:137–148. https://doi.org/10.2147/JHC.S44379
61. Cochand-Priollet B, Chagnon S, Ferrand J, Blery M, Hoang C, Galian A. Comparison of cytologic
examination of smears and histologic examination of tissue cores obtained by fine needle aspiration
biopsy of the liver. Acta Cytol. 1987;31(4):476–80.
62. Silva MA, Hegab B, Hyde C, Guo B, Buckels JA, Mirza DF. Needle track seeding following biopsy of
liver lesions in the diagnosis of hepatocellular cancer: a systematic review and meta-analysis. Gut.
2008;57:1592–6. https://doi.org/10.1136/gut.2008.149062
63. Brunt EM. Histopathologic features of hepatocellular carcinoma. Clin Liver Dis. 2012;1:194–9.
https://doi.org/10.1002/cld.98
64. Jain D. Tissue diagnosis of hepatocellular carcinoma. J Clin Exp Hepatol. 2014;4(Suppl 3):S67–S73.
https://doi.org/10.1016/j.jceh.2014.03.047
65. Timek DT, Shi J, Liu H, Lin F. Arginase-1, HepPar-1, and Glypican-3 are the most effective
panel of markers in distinguishing hepatocellular carcinoma from metastatic tumor on fine-
needle aspiration specimens. Am J Clin Pathol. 2012;138(2):203–210. https://doi.org/10.1309/
AJCPK1ZC9WNHCCMU
66. Ishak KG, Goodman ZD, Stocker JT. Tumors of the Liver and Intrahepatic Bile Ducts. In; Rosai J, edi-
tor. Atlas of Tumor Pathology. Armed Forces Institute of Pathology Washington, DC. 2001 3rd Series.
Fascicle 31.
67. Hirohashi S, Ishak KG, Kojiro M. Hepatocellular carcinoma. In: Hamilton S.R., Aaltonen L.A., edi-
tors. Tumors of the Digestive System. World Health Organization Classification of Tumors.IARC Press;
Lyon. 2000. pp. 159–172.
68. Jakate S, Yabes A, Giusto D. Diffuse cirrhosis-like hepatocellular carcinoma: a clinically and radio-
graphically undetected variant mimicking cirrhosis. Am J Surg Pathol. 2010;34:935–41. https://doi.
org/10.1097/PAS.0b013e3181ddf52f
69. Goodman ZD, Terraciano LM. Tumours and tumour-like lesions of the liver. In: Burt AD, Portmann
BC, Ferrell LD, editors. MacSween’s Pathology of the Liver. 5th ed. Churchill Livingstone; Philadelphia,
PA 2007. pp. 761–814.
70. Shiina S. Image-guided percutaneous ablation therapies for hepatocellular carcinoma. J Gastroenterol.
2009;44(suppl 19):122–31. https://doi.org/10.1007/s00535-008-2263-9
71. Befeler AS, Di Bisceglie AM. Hepatocellular carcinoma: diagnosis and treatment. Gastroenterology.
2002;122(6):1609–19. https://doi.org/10.1053/gast.2002.33411
72. Izumi R, Shimizu K, Ii T, Yagi M, Matsui O, Nonomura A et al. Prognostic factors of hepatocellular
carcinoma in patients undergoing hepatic resection. Gastroenterology. 1994;106:720–7. https://doi.
org/10.1016/0016-5085(94)90707-2
73. Calvet X, Bruix J, Brú C, Gines P, Vilana R, Sole M. Natural history of hepatocellular carci-
noma in Spain. Five year’s experience in 249 cases. J Hepatol. 1990;10(3):311–7. https://doi.
org/10.1016/0168-8278(90)90138-H
54 Thiesen A L
74. Farinati F, Rinaldi M, Gianni S, Naccarato R. How should patients with hepatocellular carcinoma
be staged? Validation of a new prognostic system. Cancer. 2000;89(11):2266–73. https://doi.
org/10.1002/1097-0142(20001201)89:11<2266::AID-CNCR15>3.0.CO;2-0
75. Llovet JM, Bruix J. Prospective validation of the Cancer of the Liver Italian Program (CLIP) score:
a new prognostic system for patients with cirrhosis and hepatocellular carcinoma. Hepatology.
2000;32(3):679–80. https://doi.org/10.1053/jhep.2000.16475
76. Current Surgical Therapy: Expert Consult - Online and Print, 11e (Current Therapy). Saunders 2019.
77. Oh’s Intensive Care Manual: Expert Consult: Online and Print, 7e. Butterworth-Heinemann. 2013
78. Schiano TD, Kim-Schluger L, Gondolesi G, Miller CM. Adult living donor liver transplantation: The
hepatologist’s perspective. Hepatology. 2001;33:3–9. https://doi.org/10.1053/jhep.2001.21489
79. Iwatsuki S, Starzl TE. Role of liver transplantation in the treatment of hepatocellular carcinoma.
Semin Surg Oncol. 1993;9:337–40. https://doi.org/10.1002/ssu.2980090411
80. Bismuth H, Chiche L, Adam R, Castaing D, Diamond T, Dennison A. Liver resection versus transplan-
tation for hepatocellular carcinoma in cirrhotic patients. Ann Surg. 1993;218:145–51. https://doi.
org/10.1097/00000658-199308000-00005
81. Pichlmayr R, Weimann A, Steinhoff G, Ringe B. Liver transplantation for hepatocellular carcinoma:
clinical results and future aspects. Cancer Chemother Pharmacol. 1992;31:S157-S161. https://doi.
org/10.1007/BF00687127
82. Ismail T, Angrisani L, Gunson BK, Hubscher SG, Buckels JA,Neuberger JM et al. Primary hepatic
malignancy:the role of liver transplantation. Br J Surg. 1990;77:983–87. https://doi.org/10.1002/
bjs.1800770908
83. Rahimi RS, Trotter JF. Liver transplantation for hepatocellular carcinoma: outcomes and treatment
options for recurrence. Ann Gastroenterol. 2015;28:323–30.
84. Loss GE, Nair S, Blazek JL, Farr GH, Eason JD. Liver Transplantation for Hepatocellular Carcinoma:
The Ochsner Experience. Ochsner J. 2002;4(4):215–20.
85. Bismuth H, Chiche L, Adam R. Liver resection versus transplantation for hepatocellular carcinoma in cir-
rhotic patients. Ann Surg. 1993;218:145–51. https://doi.org/10.1097/00000658-199308000-00005
3
Non-Alcoholic Fatty Liver Disease
Progression to Non-Alcoholic
Steatohepatitis-Related Primary
Liver Cancer
Utibe-Abasi Udoh1-2 • Juan D Sanabria1-2 • Pradeep K Rajan1-2 •
Moumita Banerjee1-2 • Mathew Schade1-2 • Jacqueline A Sanabria1-2 •
Gary Smith1-2 • Gideon Udoh1-2 • Komal Sodhi1-2 • Sandrine Pierre2 •
Joseph I Shapiro1-2 • Juan R Sanabria1-3
1Department of Surgery, Marshall University Joan Edwards School of Medicine,
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
55
56 Utibe-Abasi U et al.
INTRODUCTION
Cancer is the second leading cause of death worldwide, accounting for one in
every six deaths (1). Hepatocellular carcinoma (HCC) is the second and fast-
est-growing cause of malignancy-related mortality with an estimate of 840,000
new cases every year, contributing to 9.1% of all cancer deaths (2–4). HCC is
a highly lethal malignancy that primarily originates from a background of
end-stage liver disease (ESLD) or cirrhosis, the most significant risk factor for
HCC. ESLD develops from multiple etiologies, such as infectious (hepatitis B
and C virus infection), genetically inherited (α-1 antitrypsin deficiency, hemo-
chromatosis, primarily biliary cirrhosis, Wilson’s disease), toxic (alcohol,
drugs, aflatoxins), and metabolic (non-alcoholic steatohepatitis [NASH]) (2).
The only treatment currently available to intervene in the concomitant mala-
dies from parenchymal dysfunction, portal hypertension, and early-stage
malignancy, is liver transplantation. Although transplantation is a highly suc-
cessful form of therapy with overall survival rates of >65% at five years, this
form of therapy is limited due to scarcity of graft donors. Moreover, most
patients with HCC at presentation are staged with advanced disease, thereby
precluding resection or transplantation, and locoregional therapies such as
trans-arterial chemoembolization (TACE), Y-90, and ablation, are performed
as a bridge for liver transplantation. Systemic therapy for advanced disease
only prolongs survival in terms of weeks. Nevertheless, new forms of immu-
notherapy may increase further overall survival. In brief, HCC still carries a
poor prognosis, evolving as a prominent and increasing global health chal-
lenge (2, 5).
Metabolic disturbances like dyslipidemia from obesity are increasing in the
Western world, with one out of three or four adults being either overweight or
obese. It is estimated that 2.2 billion will be overweight and 1.1 billion obese
by 2030 (6–8). The epidemic of obesity and its metabolic consequences
(hypertension [HTN], hypertriglyceridemia, and insulin resistance) have led
to a sharp rise in non-alcoholic fatty liver disease (NAFLD) and its progressed
inflammatory form NASH, and its sequelae ESLD and HCC (2, 5, 9–11). In
the United States (US), NASH-related HCC is predicted to rise in the next ten
years with an estimated increase of 21% for NAFLD, 63% for NASH, and
137% for HCC by 2030 (2, 12). We aim in this chapter to discuss, at least one
NAFLD Progression to NASH and HCC 57
EPIDEMIOLOGY
VLDL
FFA
Glucose ROS
β-oxidation
DNL
syndrome, as well as in aging and embryonic development (46, 47). For instance,
genetic deletion of a caveolin binding motif (CBM) at the α1-NKA resulted in a
lethal embryonic phenotype in homozygous mice, despite normal NKA protein
expression and ion pumping function (47). This observation indicates that
α1-NKA-CAV-1 interaction is necessary for the proper execution of developmen-
tal signaling pathways. Conversely, chronic activation of the NKA-CAV-1-Src sig-
naling complex promotes pro-inflammatory pathways and tissue fibrosis through
the amplification of reactive oxygen intermediates. This pathway embraces a
vicious feed-forward mechanism, as evident in several disease phenotypes,
including renal fibrosis, uremic cardiomyopathy, and metabolic disorders such as
obesity (46, 48, 49). Although a balanced signaling mechanism through
NKA-CAV-1-Src is essential for normal physiological function, chronic activation
of this signaling mechanism under pathophysiological conditions can further
promote or aggravate disease conditions such as cancer. Recent in vivo and in vitro
data from our lab have shown that dysregulation of this signaling pathway
resulted in an imbalance in Smac/DIABLO-Survivin apoptotic signaling in the
cell via the upregulation of Survivin (anti-apoptotic protein) and a downregula-
tion of Smac/ DIABLO (pro-apoptotic protein), leading to an “oncogenic apop-
totic switch” that favors the development and progression of NASH to HCC (45).
Additionally, inhibition of this pathway by a novel peptide, known as pNaKtide
(developed from N domain of Na/K-ATPase), resulted in the reversal of the
“oncogenic apoptotic switch”, leading to HCC prevention, tumor regression and
reduction in fibrosis (45). Furthermore, evidence from different backgrounds
enforces the fact that the central pathway controlling energy homeostasis, the
phosphoinositide 3-kinase (PI3K)-AKT-mTOR pathway is involved and mutated
in over 50% of all HCC cases, thus placing cell energy disturbances at the center
of liver tumorigenesis (50).
As mentioned earlier, an excess in the production of ROI in NASH, leads to
an imbalance in the cell’s redox status with progressive respiratory chain disrup-
tion and energy depletion followed by mitochondrial membrane pores opening
and the subsequent leakage of cytochrome C and SMAC, concluding in the acti-
vation of the cell apoptotic cascade (20). Although metabolic stress peaks at the
mitochondria, both insulin resistance and lipotoxicity are linked to several
other cellular mechanisms, such as oxidative and endoplasmic reticulum (ER)
stress, which may also contribute to cell injury and progression of NASH to
HCC (20, 51). Another emerging mechanism that may play a key role in the
progression of NASH to HCC is autophagy. Intracellular organelle/protein
autophagy is critical in cell survival by recycling metabolic components and is
increased during cellular stress. The cell, by this process, removes cytosolic
non-functional organelles or macromolecules by transporting them into double-
membrane vesicles and delivering them to lysosomes for degradation (20). In
the liver, autophagy suppresses protein aggregation, lipid accumulation, oxida-
tive stress, chronic cell death, and inflammation. In addition, autophagy has
been shown to control adipogenesis and adipose tissue differentiation (20, 52).
Nevertheless, studies have also revealed that autophagy enables the parenchy-
mal cells to tolerate more stress, promoting tumorigenesis (20, 53, 54). Despite
the controversial role of autophagy in promoting or inhibiting NASH progres-
sion to HCC, its cellular role in an energy inefficient metabolism via PI3K/mTOR
pathway remains to be determined (20).
NAFLD Progression to NASH and HCC 61
Figure 3. Sensitivity of ultrasound as a surveillance tool for the detection of hepatocellular
carcinoma. Ultrasound is recommended every 6 months in high-risk populations for
monitoring the development of HCC, as well as in cirrhotic patients. Its sensitivity is around
77% and it can be complemented, if needed, with other imaging modalities, such as
computed tomography or magnetic resonance imaging (59).
62 Utibe-Abasi U et al.
HCC
PS 0, Child-Pugh A Okuda 3, PS > 2,
Child-Pugh C
Very early stage (0) Early stage (A) Intermediate stage Advanced stage (C)
Single < 2 cm Single or 3 nodules (B) Portal invasion, Terminal
Carcinoma in situ < 3 cm, PS 0 Multinodular, PS 0 N1, M1, PS 1–2 stage (D)
Single nodules ≤ 3 cm
Portal pressure/bilirubin
Increased Associated
Normal No Yes
Figure 4. Staging system for hepatocellular carcinoma. The Barcelona group developed a
staging system for HCC that includes five stages, given recommended therapy for each stage
and change of cure and overall survival (61).
and Treatment of Cancer (EORTC) and the American Association for the Study of
Liver Diseases (AASLD) (60, 62). This system stages an individual as having very
early, early, intermediate, advanced, and very advanced (terminal) HCC based
on tumor burden, severity of liver disease and his/her performance status
matching the recommended evidence-based treatment by the stage of liver tumor
(55, 60).
The management and treatment of HCC is complex, and still carries a poor prog-
nosis since an advanced stage is diagnosed in over 50% of the cases (63, 64).
NAFLD Progression to NASH and HCC 63
Albeit great advances have been made in the management and treatment of HCC
patients at various stages of the disease (65). Patients in very early or early stage
of HCC may benefit from curative procedures, mainly surgical resection and liver
transplantation, as well as locoregional therapies such as thermal-related ablative
procedures. In the intermediate stage of the disease, locoregional procedures such
as TACE and Y-90 embolization are performed as a bridge for transplantation, as
a method to downstage tumor burden, or as to a limited time local disease control
(63, 65, 66). Image guided ablative therapy by microwave, radiofrequency or
cryotherapy procedures are used in tumors <4 cm in size with the Intent to cure.
They can be delivered in a percutaneous, laparoscopic, or open manner as the
only procedure, or as a complement to surgery (60, 64).
Liver resection
Surgical resection of the liver is a curative therapy and preferred treatment method
for patients with noncirrhotic or compensated cirrhosis (Child A, Low MELD,
Figure 5), solitary nodules (tumor size <5 cm) and adequate liver function with
no microvascular invasion or disease dissemination (60, 66). The preferred treat-
ment option for patients with very early and early stage of HCC with cirrhosis is
surgical resection if their hepatic function is intact and bilirubin level is (<1 mg/dl
or <17.1 μmol/l) with limited or no portal hypertension (60, 62). Such patients
have a survival rate of about 70% at 5 years, limited by a degree of patients who
undergo liver decompensation after surgery (60, 67, 68). Thus, selection of
patients for liver resection therapy is critical as there is a probability of liver failure
after resection with high mortality rates (60, 68).
Liver transplantation
Liver transplantation is a curative modality for the patient with HCC and border-
line to decompensated cirrhosis (Figure 6) (66). It offers a high survival rate and
the lowest probability of tumor recurrence due to the removal of the fibrotic envi-
ronment (68). Nonetheless, due to the scarcity of graft donors, there is a signifi-
cant patient mortality from dropping off the liver waiting list due to tumor
progression. To overcome longer cadaveric organs offer waiting times, living
non- or related-liver transplantation has been presented as an option (66, 68, 69).
Figure 5. Liver imaging and pathology specimen after liver resection for hepatocellular
carcinoma. A. A right tri-segmentectomy was performed. B. Tumor was removed, note the
clear margins. C. MRI, showing the large HCC with multiple satellite lesions before resection.
The patient did well after surgery, and she was discharged home 5 days after procedure, with
no evidence for recurrences at 2 years follow up.
64 Utibe-Abasi U et al.
Figure 6. Liver transplant procedure in a patient with end stage liver disease. A. Liver with
advanced fibrosis is removed. B. Replaced with a liver graft form a deceased donor.
C. Reperfusion showing satisfactory appeaance. D. Classic vascular and biliary
reconstruction performed at the proximal and distal end:end caval anastomosis, porto:portal
and common hepatic to common hepatic arterial reconstruction. In this case, no blood
products were given to the patient during the procedure. The patient was discharged 6 days
after surgery. The patient is alive and doing well 5 years after graft implantation.
h
g
Nodules
ur
na
Up to 3 tumors, none > 3 cm
c
tts
tri
el
6
ul n
en
Pi
rc
M apa
tic
Ba
J
5
on co
Pa Fra se
pl is
4
s
Sa rou
m nc
a
B
i
no
na
ul
3
n
ila
a .Si
Pa
M
rc rk o
on Mt
Ba Yo sc
or
ris
ry m
Re lla gh
i
n na
ew c
Pa
st u
N ran
ur
gi s T
Sa plo
no
el
b
F
ila
tts
Pa
a
1 M
Pi
D
0 Tumor Size
Absence of macroscopic vascular invasion, (cm)
0 1 2 3 4 5 6 7 8 9 10
absence of extrahepatic spread
75–80% 50–75% 35–50%
Expected 5-year survival
Figure 7. The Milan criteria for liver transplantation in patients with end-stage liver disease and
hepatocellular carcinoma. Patients with end-stage liver disease and hepatocellular carcinoma
are entered into the liver transplant waiting list if they fulfil the Milan criteria, which includes
one tumor <5 cm in major diameter, or a maximum of 3 tumors with the largest <3 cm in
major diameter. In addition, the patient should have no vascular invasion or evidence of
extrahepatic disease. Other groups have explored other criteria with the development of the
metro-ticket concept that evaluates the probability of overall survival after liver
transplantation in patients with tumor burden over the Milan criteria (70).
The ideal candidate for liver transplantation (cadaveric or living-related) are those
who satisfy the Milan criteria (solitary tumor up to 5 cm in size, or 3 tumors
where the largest tumor is up to 3 cm in size, Figure 7) (66, 70). The Milan criteria
showed a low probability of tumor recurrence after transplantation, with over
75% survival in 5 years and over 90% tumor reoccurrence-free survival rate
(66, 68). Nevertheless, this concept has been challenged by several groups, and
many transplant centers offer liver transplantation for patients with HCC outside
of the Milan criteria with acceptable outcomes (66, 71). One other criterion is that
of the University of California at San Francisco, which include single tumors
≤6.5 cm or 2 to 3 tumors ≤4.5 cm, with a total tumor diameter ≤8 cm (66, 71).
Radiofrequency ablation
Image-guided RFA is indicated in patients with tumor lesions ≤3 cm (single or up
to 3 lesions) or single lesions ≤4 cm, not in proximity to major vascular or biliary
conduits, with intact liver function belonging to Child-Pugh A or B group and
ECOG status 1-2 (72, 73). The heat emanated from high frequency oscillating
electrical currents at the needle tip of the probe transforms dripping NS0.9% into
vapor, resulting in tissue necrosis (Figure 9) (66, 72, 74). However, the thermal
effect is dissipated by the “sink effect” of a vessel in proximity and by the size of
the tumor (72).
Microwave ablation
Microwave ablation is the term that is used to describe tumor destruction by elec-
tromagnetic waves at frequency ≥900 kHz (74). It is one of the treatments of
choice for HCC patients with tumors that are less than 4 cm in size without the
sink effect limitation of RFA (66). The image-guided probe placement, as in RFA
can be achieved percutaneously, laparoscopically, or during open surgery (66, 75).
Contraindications include macrovascular invasion of tumor, main portal vein
destruction, decompensated cirrhosis (Child-Pugh C), biliary obstruction and
proximity to vital structures mitigated by open or laparoscopic techniques (66).
Cryoablation
Cryoablation, unlike RFA or microwave ablation, uses very low temperatures
from a liquid nitrogen source to destroy tissue by alternating freezing and thawing
based on the Joule-Thompson effect (76). As in other ablative methods, the probe
66 Utibe-Abasi U et al.
can be placed under image guidance to induce “ice ball” formation (76, 77).
Indications for percutaneous cryoablation include patients that satisfy the Milan
criteria; that is, those with tumor size <5 cm in diameter or up to 3 tumors <3 cm,
absence of venous thrombosis, Child- Pugh A and B group without significant
coagulopathy (76). The advantages of cryoablation include the ability to visualize
the ice ball, activate cryo-immunology in cancerous cells, absence of severe dam-
age to blood vessels and less severe pain (77). Cryoablation can be used as an
isolated treatment for patients or in combination with other therapies. Cryotherapy
of liver tumors has become unpopular due to major bleeding after probe removal
and the induction of coagulopathy aggravated by thrombocytopenia, liver decom-
pensation and death (66).
Trans-arterial chemoembolization
TACE is currently the standard of care for patients with intermediate–stage HCC
with preserved liver function (78–80). TACE is useful for patients that have a
Child-Pugh score A or B with tumor diameter of >4 cm or four or more tumors as
well as those with single tumor in which it is challenging to carry out liver resec-
tion or locoregional therapies as a result of systemic co-morbidities or anatomical
limitations (81). TACE takes advantages of the dual arterial and portal venous
liver parenchymal blood supply with preferential arterialization not only in cir-
rhotic liver but of HCC. It involves the selective arterial embolization with a gela-
tin mixed with lipiodol (a radiopaque contrast agent) with or without chemotherapy
(doxorubicin, cisplatin or mitomycin C), into the tumor’s feeding blood vessel (66).
The blockage of the arteries supplying the tumor results in tissue necrosis
(Figure 10) (68, 79). In practice, TACE is a recommended therapy for patients
NAFLD Progression to NASH and HCC 67
Figure 10. Trans-arterial chemoembolization of the liver for hepatocellular carcinoma. A and
B. Selective arterial embolization of the vessels feeding the tumor in the liver. C. Tumor
visualized by CT scan. The procedure is performed with or without chemotherapy. D and
E. After embolization, angiography showed obliteration of feeding vessels with shrinkage of
the tumor. F. Follow up CT scan after embolization. Note an increase in patient’s ascites.
with unresectable HCC, nonvascular invasion or disease outside the liver (64).
TACE can also be used with drug-eluting beads (DEB-TACE) and evidence exists
that patients who are on DEB-TACE treatments for unresectable HCC have better
performance in comparison to those on conventional TACE (66). In addition,
TACE is being used for the downstaging of tumors in association with systemic
therapy or as bridge for transplantation.
Y-90 radioembolization
Y-90 radioembolization is a locoregional technique that involves a catheter-based
administration of Y-90 microspheres into the hepatic artery, leading to the delivery
of high radiation doses up to 50 to 150 Gy to tumors without affecting the paren-
chymal cells (Figure 11) (82, 83) . This exploits the principle; intrahepatic tumors
derive their major blood supply from the hepatic artery rather than the portal
vein. Y-90 emits beta radiation with an average energy of 0.9 MeV and a mean
penetration range of 2.5 mm (approximately 1,000 cell diameters). The physical
half-life of Y-90 is 64.2 hours, and it decays to stable Zirconium-90 (82, 83).
Microspheres (embedded with Y-90) are of varying sizes, ranging from 20 to 60
microns. In the US, the common available forms of Y-90 include the Y-90 tagged
glass (TheraSphere) and resin (SIR-Spheres) microspheres (82). The main differ-
ence between Y-90 radioembolization and TACE is that Y-90 microspheres are
smaller than TACE particles (20-30 microns compared to 200-500 microns).
Therefore, TACE gives a more significant embolic effect in comparison to Y-90
radioembolization. However, the main mechanism of action of Y-90 is related to
68 Utibe-Abasi U et al.
Figure 11. Y-90 embolization for hepatocellular carcinoma. A. Selective angiography of the
liver and detection with obliteration of systemic shunts is performed. B, C and
D. Micro-spheres then are embolized to the liver with no vessel occlusion.
radiation effect and not the embolic occlusion of the blood supply to the cancer
cells, thereby making it a more effective therapy (82, 83).
Irreversible electroporation
Irreversible electroporation (IRE) is a nonthermal type of tumor ablation tech-
nique that is not affected by heat sink, which is a common limitation of other
forms of ablation procedures such RFA ablation. IRE procedure involves the
delivery of short pulses of high-frequency energy to create pores in the lipid
bilayer of cancer cells, which leads to cell death through apoptosis. On the other
hand, acellular elements within the treatment region are not affected, resulting
in the preservation of hepatic parenchymal architecture (88, 89). IRE procedure
gives the best results with tumors that are less than 3 cm (88). Currently the
only commercially available system for IRE is NanoKnife® (AngioDynamics,
NAFLD Progression to NASH and HCC 69
A) B)
1500
1250
750
500
250
0
Initial volume Last FU
C) D)
1.0 1.0
0.8 0.8
0.7 0.7
0.6 0.6
0.5 0.5
0.4 0.4
0.3 0.3
0.2 0.2
HCC HCC
0.1 ICC 0.1 ICC
0.0 0.0
0 10 20 30 40 50 0 12 24 36 48 60
Time (months) Time (months)
Figure 12. Stereotactic body radiosurgery therapy for hepatocellular carcinoma. A. High doses
of focused radiotherapy are applied to liver tumor sparing normal liver parenchyma using
stereotactic body radiosurgery therapy. B. Shows high rates of tumor response (reduction in
tumor size) by RECIST criteria. C and D. The estimated probability of local control and overall
survival for hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are
displayed respectively (87).
Molecular targeted signaling pathways that are considered to play key roles in HCC
tumor formation, growth and progression include: mitogen-activated protein
kinases (MAPK) pathway (Ras/Raf/MEK/ERK pathway), the Phosphatidylinositol-
4,5-bisphosphate 3-kinase (PI3K), mammalian target of rapamycin (mTOR) path-
way and angiogenic pathways (68, 93).
Immunotherapy
Currently there is a dynamic expansion in cancer immunotherapy which focuses
mainly on agents that give a prognostic benefit to cancer patients by stimulating
the immune system to mount a response against developing cancers including
HCC. As earlier stated, there is a paucity of therapeutic options for patients with
advanced stage of HCC, therefore it appears that immunotherapy may hold the
key to effective systemic therapy for these patients (94). Immunotherapy strate-
gies for HCC are based on two main principles, namely: (i) the ability to unmask
ongoing immune responses in the liver during the onset or progression of carci-
nogenesis and (ii) the need to elicit new or different immunological responses.
The first strategy is based on pre-existing immune reactivity to cancer develop-
ment and progression held in check by micro-environmental factors. Such factors
include inhibitory receptors on T cells, especially programmed cell death protein 1
(PD-1) and cytotoxic T-lymphocyte associated antigen 4 (CTLA-4); other factors
are immunosuppressive cytokines such as Transforming growth factor beta
(TGF-β) (94). In this strategy, the therapies are not directed towards a specific
biological molecule produced by the cancer cells, but there is a general heighten-
ing/unmasking of immune responses to destroy tumor formation and progression.
On the other hand, antibodies that directly target molecules expressed on HCC
tumor cells, such as alpha-fetoprotein (AFP) or glypican-3 (GPC-3) fall within the
second strategy. These therapies can be enhanced by coupling these antibodies to
effector cells, such as T cells or natural killer cells (94). Additionally, administra-
tion of vaccines and the use of oncolytic viruses can operate through these two
mechanisms, that is, by unmasking already existing immune responses or prompt-
ing de novo T cell responses to substances expressed by HCC tumor cells (94). It
is worth noting that in line with the first strategy, the US Food and Drug
Administration (FDA) has approved the use of nivolumab (a PD-1 inhibitor) as an
immunotherapy for HCC (94, 95).
CONCLUSION
Over the last two decades the incidence of NASH-related HCC has risen exponen-
tially, mainly due to metabolic disturbances promoted by the epidemic of obesity.
HCC has become a major and steadily increasing global health challenge due to a
paucity of biomarkers for its early detection coupled with few treatment options
and a 50–70% recurrence rate after resection or locoregional therapy. Treatment
options that are available for patients in very early or early stage of HCC include
surgical resection, liver transplant and ablation procedures with the intend to
cure. Those patients in the intermediate-stage are often treated with TACE or Y-90
radioembolization. Immuno-strategies are becoming the first line of therapy, fol-
lowed by sorafenib as systemic treatment for patients with advanced stage of the
disease. There is need for basic, translational, and clinical research targeting cel-
lular and molecular pathways that play key roles in cancer development and pro-
gression, aiming for novel and more effective therapies for NASH-related HCC.
One of such pathways is the α1-Na/K-ATPase-CAV-1-Src signaling pathway at the
cell membrane that plays a role in the regulation of embryonic development and
cell growth. Disturbances in this pathway has been shown to splinter the fragile
balance of apoptosis regulator proteins promoting an oncogenic “apoptotic
switch” that favors hepatic cell tumorigenesis. Furthermore, the inhibition of this
pathway may be a putative target for the treatment of HCC.
Copyright and permission statement: The authors confirm that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, and the Global Burden
of Disease Cancer Collaborators. Global, Regional, and National Cancer Incidence, Mortality,
Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-Years for 29 Cancer
Groups, 1990 to 2016: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol.
2018;4(11):1553–68. https://doi.org/10.1001/jamaoncol.2018.2706
2. Hayes CN, Zhang P, Chayama K. The Role of Lipids in Hepatocellular Carcinoma. In: Tirnitz-Parker
JEE, editor. Hepatocellular Carcinoma. Brisbane (AU)2019. https://doi.org/10.15586/hepatocellular-
carcinoma.2019.ch5
3. Rawla P, Sunkara T, Muralidharan P, Raj JP. Update in global trends and aetiology of hepatocellular
carcinoma. Contemp Oncol (Pozn). 2018;22(3):141–50. https://doi.org/10.5114/wo.2018.78941
72 Utibe-Abasi U et al.
4. Liebig M, Dannenberger D, Vollmar B, Abshagen K. n-3 PUFAs reduce tumor load and improve sur-
vival in a NASH-tumor mouse model. Ther Adv Chronic Dis. 2019;10:2040622319872118. https://
doi.org/10.1177/2040622319872118
5. Carlessi R, Kohn-Gaone J, Olynyk JK, Tirnitz-Parker JEE. Mouse Models of Hepatocellular
Carcinoma. In: Tirnitz-Parker JEE, editor. Hepatocellular Carcinoma. Brisbane (AU)2019. https://doi.
org/10.15586/hepatocellularcarcinoma.2019.ch4
6. Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, and GBDO collaborators. Health Effects of
Overweight and Obesity in 195 Countries over 25 Years. N Engl J Med. 2017;377(1):13–27. https://
doi.org/10.1056/NEJMoa1614362
7. Forouzanfar MH, Alexander L, Anderson HR, Bachman VF, Biryukov S, and GBDRF Collaborators.
Global, regional, and national comparative risk assessment of 79 behavioural, environmental and
occupational, and metabolic risks or clusters of risks in 188 countries, 1990–2013: a systematic
analysis for the Global Burden of Disease Study 2013. Lancet. 2015;386(10010):2287–323. https://
doi.org/10.1016/S0140-6736(15)00128-2
8. Mokdad AH, Ballestros K, Echko M, Glenn S, Olsen HE, and USGBD Collaborators. The State of
US Health, 1990–2016: Burden of Diseases, Injuries, and Risk Factors Among US States. JAMA.
2018;319(14):1444–72. https://doi.org/10.1001/jama.2018.0158
9. Cholankeril G, Patel R, Khurana S, Satapathy SK. Hepatocellular carcinoma in non-alcoholic steato-
hepatitis: Current knowledge and implications for management. World J Hepatol. 2017;9(11):533–43.
https://doi.org/10.4254/wjh.v9.i11.533
10. Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of non-
alcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–21.
https://doi.org/10.1053/j.gastro.2005.04.014
11. Nagaoki Y, Hyogo H, Aikata H, Tanaka M, Naeshiro N, Nakahara T, et al. Recent trend of clini-
cal features in patients with hepatocellular carcinoma. Hepatol Res. 2012;42(4):368–75. https://doi.
org/10.1111/j.1872-034X.2011.00929.x
12. Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the epidemic of nonalcoholic fatty liver
disease demonstrates an exponential increase in burden of disease. Hepatology. 2018;67(1):123–33.
https://doi.org/10.1002/hep.29466
13. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology.
2012;142(6):1264–73 e1. https://doi.org/10.1053/j.gastro.2011.12.061
14. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, and GBD Liver Cancer Collaborators.
The Burden of Primary Liver Cancer and Underlying Etiologies From 1990 to 2015 at the Global,
Regional, and National Level: Results From the Global Burden of Disease Study 2015. JAMA Oncol.
2017;3(12):1683–91.
15. El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011;365(12):1118–27. https://doi.
org/10.1056/NEJMra1001683
16. Noureddin M, Rinella ME. Nonalcoholic Fatty liver disease, diabetes, obesity, and hepatocellular car-
cinoma. Clin Liver Dis. 2015;19(2):361–79. https://doi.org/10.1016/j.cld.2015.01.012
17. Altekruse SF, McGlynn KA, Reichman ME. Hepatocellular carcinoma incidence, mortality, and sur-
vival trends in the United States from 1975 to 2005. J Clin Oncol. 2009;27(9):1485–91. https://doi.
org/10.1200/JCO.2008.20.7753
18. Cholankeril G, Perumpail RB, Pham EA, Ahmed A, Harrison SA. Nonalcoholic Fatty Liver Disease:
Epidemiology, Natural History, and Diagnostic Challenges. Hepatology. 2016;64(3):954. https://doi.
org/10.1002/hep.28719
19. Fleischman MW, Budoff M, Zeb I, Li D, Foster T. NAFLD prevalence differs among hispanic sub-
groups: the Multi-Ethnic Study of Atherosclerosis. World J Gastroenterol. 2014;20(17):4987–93.
https://doi.org/10.3748/wjg.v20.i17.4987
20. Kutlu O, Kaleli HN, Ozer E. Molecular Pathogenesis of Nonalcoholic Steatohepatitis- (NASH-)
Related Hepatocellular Carcinoma. Can J Gastroenterol Hepatol. 2018;2018:8543763. https://doi.
org/10.1155/2018/8543763
21. Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH and liver cancer. Nat Rev Gastroenterol
Hepatol. 2013;10(11):656–65. https://doi.org/10.1038/nrgastro.2013.183
NAFLD Progression to NASH and HCC 73
22. Margini C, Dufour JF. The story of HCC in NAFLD: from epidemiology, across pathogenesis, to pre-
vention and treatment. Liver Int. 2016;36(3):317–24. https://doi.org/10.1111/liv.13031
23. Marra F, Gastaldelli A, Svegliati Baroni G, Tell G, Tiribelli C. Molecular basis and mechanisms of
progression of non-alcoholic steatohepatitis. Trends Mol Med. 2008;14(2):72–81. https://doi.
org/10.1016/j.molmed.2007.12.003
24. Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998;114(4):842–5. https://
doi.org/10.1016/S0016-5085(98)70599-2
25. Gentile CL, Pagliassotti MJ. The role of fatty acids in the development and progression of nonalcoholic
fatty liver disease. J Nutr Biochem. 2008;19(9):567–76. https://doi.org/10.1016/j.jnutbio.2007.10.001
26. Day CP. NASH-related liver failure: one hit too many? Am J Gastroenterol. 2002;97(8):1872–4.
https://doi.org/10.1111/j.1572–0241.2002.05952.x
27. Day CP. Non-alcoholic steatohepatitis (NASH): where are we now and where are we going? Gut.
2002;50(5):585–8. https://doi.org/10.1136/gut.50.5.585
28. Day CP. Clinical spectrum and therapy of non-alcoholic steatohepatitis. Dig Dis. 2012;30
Suppl 1:69–73. https://doi.org/10.1159/000341128
29. Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ. Cytokeratin-18 frag-
ment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: a multicenter validation study.
Hepatology. 2009;50(4):1072–8. https://doi.org/10.1002/hep.23050
30. Wieckowska A, McCullough AJ, Feldstein AE. Noninvasive diagnosis and monitoring of nonalco-
holic steatohepatitis: present and future. Hepatology. 2007;46(2):582–9. https://doi.org/10.1002/
hep.21768
31. Wieckowska A, Zein NN, Yerian LM, Lopez AR, McCullough AJ, Feldstein AE. In vivo assessment
of liver cell apoptosis as a novel biomarker of disease severity in nonalcoholic fatty liver disease.
Hepatology. 2006;44(1):27–33. https://doi.org/10.1002/hep.21223
32. Sumida Y, Niki E, Naito Y, Yoshikawa T. Involvement of free radicals and oxidative stress in NAFLD/
NASH. Free Radic Res. 2013;47(11):869–80. https://doi.org/10.3109/10715762.2013.837577
33. Kawano Y, Cohen DE. Mechanisms of hepatic triglyceride accumulation in non-alcoholic fatty liver
disease. J Gastroenterol. 2013;48(4):434–41. https://doi.org/10.1007/s00535-013-0758-5
34. Al-Busafi SA, Bhat M, Wong P, Ghali P, Deschenes M. Antioxidant therapy in nonalcoholic steatohepa-
titis. Hepat Res Treat. 2012;2012:947575. https://doi.org/10.1155/2012/947575
35. Browning JD, Horton JD. Molecular mediators of hepatic steatosis and liver injury. J Clin Invest.
2004;114(2):147–52. https://doi.org/10.1172/JCI200422422
36. Brunt EM, Kleiner DE, Wilson LA, Belt P, Neuschwander-Tetri BA, Network NCR. Nonalcoholic fatty
liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: distinct clinico-
pathologic meanings. Hepatology. 2011;53(3):810–20. https://doi.org/10.1002/hep.24127
37. Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple paral-
lel hits hypothesis. Hepatology. 2010;52(5):1836–46. https://doi.org/10.1002/hep.24001
38. Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treat-
ment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci. 2013;14(10):20704–28. https://
doi.org/10.3390/ijms141020704
39. De Minicis S, Agostinelli L, Rychlicki C, Sorice GP, Saccomanno S, Candelaresi C, et al. HCC
development is associated to peripheral insulin resistance in a mouse model of NASH. PLoS One.
2014;9(5):e97136. https://doi.org/10.1371/journal.pone.0097136
40. Janku F, Kaseb AO, Tsimberidou AM, Wolff RA, Kurzrock R. Identification of novel therapeutic targets
in the PI3K/AKT/mTOR pathway in hepatocellular carcinoma using targeted next generation sequenc-
ing. Oncotarget. 2014;5(10):3012–22. https://doi.org/10.18632/oncotarget.1687
41. Yang S, Liu G. Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma. Oncol Lett.
2017;13(3):1041–7. https://doi.org/10.3892/ol.2017.5557
42. Kudo Y, Tanaka Y, Tateishi K, Yamamoto K, Yamamoto S, Mohri D, et al. Altered composition of fatty
acids exacerbates hepatotumorigenesis during activation of the phosphatidylinositol 3-kinase path-
way. J Hepatol. 2011;55(6):1400–8. https://doi.org/10.1016/j.jhep.2011.03.025
43. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev
Cancer. 2002;2(7):489–501. https://doi.org/10.1038/nrc839
74 Utibe-Abasi U et al.
66. Gosalia AJ, Martin P, Jones PD. Advances and Future Directions in the Treatment of Hepatocellular
Carcinoma. Gastroenterol Hepatol (N Y). 2017;13(7):398–410.
67. Roayaie S, Obeidat K, Sposito C, Mariani L, Bhoori S, Pellegrinelli A, et al. Resection of hepatocellu-
lar cancer </=2 cm: results from two Western centers. Hepatology. 2013;57(4):1426–35. https://doi.
org/10.1002/hep.25832
68. Chacko S, Samanta S. “Hepatocellular carcinoma: A life-threatening disease”. Biomed Pharmacother.
2016;84:1679–88. https://doi.org/10.1016/j.biopha.2016.10.078
69. Bruix J, Llovet JM. Prognostic prediction and treatment strategy in hepatocellular carcinoma.
Hepatology. 2002;35(3):519–24. https://doi.org/10.1053/jhep.2002.32089
70. Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, et al. Liver transplantation
for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med.
1996;334(11):693–9. https://doi.org/10.1056/NEJM199603143341104
71. Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A, et al. Liver transplantation for hepatocel-
lular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology.
2001;33(6):1394–403. https://doi.org/10.1053/jhep.2001.24563
72. Grandhi MS, Kim AK, Ronnekleiv-Kelly SM, Kamel IR, Ghasebeh MA, Pawlik TM. Hepatocellular
carcinoma: From diagnosis to treatment. Surg Oncol. 2016;25(2):74–85. https://doi.org/10.1016/j.
suronc.2016.03.002
73. Meza-Junco J, Montano-Loza AJ, Liu DM, Sawyer MB, Bain VG, Ma M, et al. Locoregional radiological
treatment for hepatocellular carcinoma; Which, when and how? Cancer Treat Rev. 2012;38(1):54–62.
https://doi.org/10.1016/j.ctrv.2011.05.002
74. Crocetti L, Lencioni R. Thermal ablation of hepatocellular carcinoma. Cancer Imaging. 2008;8:19–26.
https://doi.org/10.1102/1470-7330.2008.0004
75. Sato M, Watanabe Y, Ueda S, Iseki S, Abe Y, Sato N, et al. Microwave coagulation therapy for hepa-
tocellular carcinoma. Gastroenterology. 1996;110(5):1507–14. https://doi.org/10.1053/gast.1996.
v110.pm8613057
76. Song KD. Percutaneous cryoablation for hepatocellular carcinoma. Clin Mol Hepatol. 2016;
22(4):509–15. https://doi.org/10.3350/cmh.2016.0079
77. Niu LZ, Li JL, Xu KC. Percutaneous Cryoablation for Liver Cancer. J Clin Transl Hepatol. 2014;
2(3):182–8.
78. Lencioni R, Petruzzi P, Crocetti L. Chemoembolization of hepatocellular carcinoma. Semin Intervent
Radiol. 2013;30(1):3–11. https://doi.org/10.1055/s-0033-1333648
79. Boulin M, Delhom E, Pierredon-Foulongne MA, Cercueil JP, Guiu B. Transarterial chemoemboli-
zation for hepatocellular carcinoma: An old method, now flavor of the day. Diagn Interv Imaging.
2015;96(6):607–15. https://doi.org/10.1016/j.diii.2015.04.005
80. Sieghart W, Hucke F, Peck-Radosavljevic M. Transarterial chemoembolization: modalities, indi-
cation, and patient selection. J Hepatol. 2015;62(5):1187–95. https://doi.org/10.1016/j.jhep.
2015.02.010
81. Imai N, Ishigami M, Ishizu Y, Kuzuya T, Honda T, Hayashi K, et al. Transarterial chemoembolization
for hepatocellular carcinoma: A review of techniques. World J Hepatol. 2014;6(12):844–50. https://
doi.org/10.4254/wjh.v6.i12.844
82. Bhangoo MS, Karnani DR, Hein PN, Giap H, Knowles H, Issa C, et al. Radioembolization with
Yttrium-90 microspheres for patients with unresectable hepatocellular carcinoma. J Gastrointest
Oncol. 2015;6(5):469–78. https://doi.org/10.3978/j.issn.2078-6891.2015.056
83. Molvar C, Lewandowski R. Yttrium-90 Radioembolization of Hepatocellular Carcinoma-Performance,
Technical Advances, and Future Concepts. Semin Intervent Radiol. 2015;32(4):388–97. https://doi.
org/10.1055/s-0035-1564704
84. Lee HL, Tsai JT, Chen CY, Lin YC, Ho CB, Ting LL, et al. Effectiveness of stereotactic ablative
radiotherapy in patients with advanced hepatocellular carcinoma unsuitable for transarte-
rial chemoembolization. Ther Adv Med Oncol. 2019;11:1758835919889002. https://doi.org/
10.1177/1758835919889002
85. McPartlin AJ, Dawson LA. Stereotactic Body Radiotherapy for Hepatocellular Carcinoma. Cancer J.
2016;22(4):296–301. https://doi.org/10.1097/PPO.0000000000000201
76 Utibe-Abasi U et al.
86. Robbins JR, Schmid RK, Hammad AY, Gamblin TC, Erickson BA. Stereotactic body radiation therapy
for hepatocellular carcinoma: Practice patterns, dose selection and factors impacting survival. Cancer
Med. 2019;8(3):928–38. https://doi.org/10.1002/cam4.1948
87. Ibarra RA, Rojas D, Snyder L, Yao M, Fabien J, Milano M, et al. Multicenter results of stereotactic
body radiotherapy (SBRT) for non-resectable primary liver tumors. Acta Oncol. 2012;51(5):575–83.
https://doi.org/10.3109/0284186X.2011.652736
88. Zimmerman A, Grand D, Charpentier KP. Irreversible electroporation of hepatocellular carcinoma:
patient selection and perspectives. J Hepatocell Carcinoma. 2017;4:49–58. https://doi.org/10.2147/
JHC.S129063
89. Golberg A, Bruinsma BG, Jaramillo M, Yarmush ML, Uygun BE. Rat liver regeneration following abla-
tion with irreversible electroporation. PeerJ. 2016;4:e1571. https://doi.org/10.7717/peerj.1571
90. Nielsen K, Scheffer HJ, Vieveen JM, van Tilborg AA, Meijer S, van Kuijk C, et al. Anaesthetic manage-
ment during open and percutaneous irreversible electroporation. Br J Anaesth. 2014;113(6):985–92.
https://doi.org/10.1093/bja/aeu256
91. Kim HY, Park JW. Current immunotherapeutic strategies in hepatocellular carcinoma: recent
advances and future directions. Therap Adv Gastroenterol. 2017;10(10):805–14. https://doi.org/
10.1177/1756283X17722061
92. Dutta R, Mahato RI. Recent advances in hepatocellular carcinoma therapy. Pharmacol Ther.
2017;173:106–17. https://doi.org/10.1016/j.pharmthera.2017.02.010
93. Ohri N, Kaubisch A, Garg M, Guha C. Targeted Therapy for Hepatocellular Carcinoma. Semin Radiat
Oncol. 2016;26(4):338–43. https://doi.org/10.1016/j.semradonc.2016.06.004
94. Johnston MP, Khakoo SI. Immunotherapy for hepatocellular carcinoma: Current and future. World J
Gastroenterol. 2019;25(24):2977–89. https://doi.org/10.3748/wjg.v25.i24.2977
95. Marrero JA, Kulik LM, Sirlin CB, Zhu AX, Finn RS, Abecassis MM, et al. Diagnosis, Staging, and
Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for
the Study of Liver Diseases. Hepatology. 2018;68(2):723–50. https://doi.org/10.1002/hep.29913
96. Mak LY, Cruz-Ramon V, Chinchilla-Lopez P, Torres HA, LoConte NK, Rice JP, et al. Global
Epidemiology, Prevention, and Management of Hepatocellular Carcinoma. Am Soc Clin Oncol Educ
Book. 2018;38:262–79. https://doi.org/10.1200/EDBK_200939
97. Saran U, Humar B, Kolly P, Dufour JF. Hepatocellular carcinoma and lifestyles. J Hepatol.
2016;64(1):203–14. https://doi.org/10.1016/j.jhep.2015.08.028
4
Gross Dissection of Liver for
Hepatocellular Carcinoma Using
AJCC Cancer Staging Manual
8th Edition: Anatomical and
Practical Considerations
Marla Beach1 • Laura Henao Caviedes1 • Consolato M. Sergi2
1
Department of Laboratory Medicine and Pathology, University of Alberta Hospital and
Stollery Children’s Hospital, Edmonton, AB, Canada; 2Departments of Pediatrics,
Laboratory Medicine and Pathology, Stollery Children’s Hospital, University of Alberta,
Edmonton, AB, Canada.
Author for Correspondence: Marla Beach, Department of Laboratory Medicine and
Pathology, University of Alberta Hospital and Stollery Children’s Hospital, Edmonton, AB,
Canada. Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch4
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
77
78 Beach M et al.
INTRODUCTION
The liver is a solid, asymmetrical organ normally located in the right upper quad-
rant of the abdomen and is found exclusively in vertebrates (1). An exception to
this rule occurs in the case of heterotaxis or Ivemark Syndrome, in which the liver
is located centrally or to the left side of the body (2). The liver synthesizes proteins
and produces biochemicals, which are specifically important for digestion and
growth. The gallbladder is a small sac, which is found on the visceral side of
the liver. The gallbladder is adherent to the posterior liver along one side (the
gallbladder ‘bed’), and in rare cases, is not visible as it can be entirely intrahepatic (3).
A thorough inspection should be given in cases of biliary atresia with Kasai
porto-enterostomy, because the gallbladder can be very small or absent (4). The
gallbladder stores and delivers bile produced by the liver into the digestive tube.
The commonly used College of American Pathologists (CAP) Cancer Protocols
detail protocol for reporting, designed to produce a standardized, comprehensive
reporting of hepatocellular carcinoma (5). These synoptics are derived from the
AJCC Cancer Staging Manual (8th ed) (6). The main categories of a complete
pathological report, using the CAP synoptic, employs three main categories:
(i) primary tumor (T stage), (ii) regional lymph nodes (N stage), and (iii) distant
metastasis (M stage). As with most pathological specimens, unless a metastasec-
tomy is performed, gross bench considerations are primarily T and N staging.
Therefore, the M stage is not discussed here.
Surface inspection
Surface inspection of the liver reveals many anatomical landmarks. These include
the falciform ligament, the ligamentum teres (round ligament), porta hepatis, gall-
bladder, and the inferior vena cava (or its groove). Porta hepatis, or the hilum,
contains the veins entering the liver (portal vein), the hepatic artery, and the bile
ducts exiting the liver. The falciform ligament is thin, membranous, and attaches
the liver to the anterior abdominal wall. At the liver’s inferior edge, the falciform
ligament merges with the round ligament (the umbilical vein’s remanent). The
round ligament sits in a grove called the umbilical fissure. Macroscopically, the
Falciform ligament has commonly been used as the border between the right and
left lobes. It grossly sits at a junction where the liver is larger (right lobe) and
smaller (left lobe). This is the anatomical right and left lobes. If inspected from
below (surgeon’s point of view) or posteriorly on the dissection table, four addi-
tional ‘lobes’ are appreciated. At the top, just medial to the IVC groove is a square
to rectangular protrusion designated the caudate lobe (caudate is Latin for ‘tail’,
Gross Dissection of Liver 79
as if it were the tail of the liver). Below the caudate lobe is the gallbladder (centre
right), and the hilum (centre left). The gallbladder is a soft sac, commonly green
or purple, adjacent to the posterior and inferior liver. The hilum in the resected
liver will likely have multiple sutures, clips, and staples on its structures: portal
vein and hepatic artery entering the liver, and the bile ducts exiting the liver. The
round ligament sits to the left of the hilum and delineates the left anatomical lobe
on the posterior liver surface. The quadrate lobe is not a distinct lobar structure,
rather, it is a four-sided area (hence ‘quad’) bounded by the porta hepatis superi-
orly, the gallbladder laterally, the round ligament medially, and the free edge of the
liver inferiorly.
Three main hepatic veins drain from the liver superiorly; the right, middle and
left hepatic veins. In the resected total liver, careful inspection of the IVC or its
groove will reveal the left and central hepatic veins. Commonly, adjacent to the
IVC to the right, the right hepatic vein is found.
Couinaud segments
Further understanding of blood and bile flow in the liver informs the functional
description of liver segments described by Couinaud (8). This system separates
the liver, based on studies done with corrosion casting into eight independent
segments. The middle hepatic vein level, as it extends from inferior to drain into
the IVC superiorly, divides the functional right and left liver in this system. At the
gross level, an estimation of this border is Cantlie’s line. Cantlie’s line is an imagi-
nary line through the fossa of the inferior vena cava to the gallbladder’s fossa,
extending through to the front of the liver (9). Each segment has its own biliary
drainage and vascular inflow and outflow. In the centre of each segment, there are
branches of the portal vein, hepatic artery, and bile duct. In the periphery of each
liver subsegment, there is a vascular outflow through the hepatic veins. The main
dividing lines of liver segments in the transverse plane are the right and left portal
veins. Dividing lines into the vertical and oblique planes are the three main hepatic
veins (right, middle, and left). The segments are numbered clockwise from 1–8
(or I–VIII) (Figure 1). In this system, Segment 1 corresponds to the caudate lobe.
Segments 2–4 comprise the functional right lobe (to the right of Cantlie’s line), and
segments 5–8 comprise the functional left lobe (to the left of Cantlie’s line). The
only segment that is often subdivided is segment 4, into segments 4a (superior)
and 4b (inferior). The quadrate lobe does not have a corresponding functional
segment, rather is part of segment 4b. The Falciform ligament does not divide the
functional lobes, but the anatomical lobes; Cantlie’s line better approximates func-
tional division of right and left lobes and is the line between the right lobe
segments 5, 6, 7 and 8, and the left lobe segments 2, 3 and 4.
Figure 1. Functional (Couinaud) segments of the liver. A. Anterior surface. B. Posterior surface.
The segments 1-8 are delineated by hashed lines in relation to the anatomical landmarks.
the right of the gallbladder for some, it is essential to recognize two impressions
which are one behind the other and separated by a ridge. The first is the colic
impression, a shallow liver parenchyma compression that is formed by the hepatic
flexure of the large bowel. The second is the renal impression, which lies behind
the colic impression. It accommodates part of the right kidney and part of the
suprarenal gland. Other important impressions include the duodenal and gastric
impressions. The duodenal impression has a distinct, characteristic shape and can
be found medial to the renal impression and lies between the segment 8 and the
Gross Dissection of Liver 81
neck of the gallbladder. Visible on the left lobe of the liver, on the inferior surface
is the gastric impression. Lastly, while not an impression, rather a point of contact,
is the superior, rounded surface of the right and mid liver, where tumour involve-
ment of the diaphragm is possible.
Grossing
When a surgical procedure is performed to resect a liver mass, the specific proce-
dure needs to be correctly identified. It is important to separate total hepatectomy
from partial hepatectomy and wedge resection. Total hepatectomies are received
in the setting of orthotopic liver transplantation (10). Thorough review of clinical
history will enlighten the dissector as to the surgeon’s intraoperative findings, the
lobes, segments, any structures removed, as well has any history of previous liver
surgeries, diagnoses, or therapies. Particular attention should be paid to the surgi-
cal report, number, location, and size of previously identified tumors, as well as
previous therapies such as resection, and oncological or ablative procedures (11).
The partial hepatectomy is subdivided into: (i) major hepatectomy, when three
segments or more are involved, and (ii) minor hepatectomy, when the resection
involves only one or two segments. Review the clinical information regarding the
type of resection, anatomical lobe, and the segments that were resected. Wedge
resections are small and may have one, two, or three sides with the identifiable
liver capsule.
identify the size, shape, landmarks, and attached structures to determine what
was resected. It is imperative to liaise with the surgeon under challenging cases to
ensure the specimen’s proper handling.
Tumor
With regard to the tumor, it is crucial to identify all tumors. The largest five masses
must be described in detail. When multiple tumors are present, care must be
taken not to confuse satellitosis with multiple primary tumors. A satellite nodule
is smaller than the main tumor and will sit within two centimeters from it, sepa-
rated by background liver tissue. All tumors must be described with regards to
their size, location (and structures involved), distance to resection margins, cut
surface/consistency, and presence of hemorrhage or necrosis. For tumor size,
ensure the size is described, three dimensionally, for the largest 5 tumors, in
centimeters.
Regarding the tumor location, it is important to specify which functional
segment the tumor is found. The functional segments cannot be discerned in the
solid organ with the naked eye, hence the use of anatomical landmarks to help
approximate segments. Figures 1A and 1B demonstrate the approximate func-
tional segments overlaid with the gross appearance and anatomical landmarks on
a total hepatectomy. With lesions that are at, near, or straddle the areas where two
segments meet, it is not uncommon to include both segment numbers in the
description. For example, if a mass is identified on the anterior surface, just to the
left of the Falciform ligament may be arising from segment 2 or 3 and can be
described as located in segment 2/3. Carefully inspect the edges of the tumor(s)
for the involvement of vascular structures and select sections to confirm vascular
invasion. With regards to the tumor extension, it is important to select all events
that apply, including no evidence of primary tumor, tumor confined to the liver,
tumor involving a major branch of the portal vein or the involvement of the
hepatic veins, the involvement of the visceral peritoneum with obvious perfora-
tion, the invasion of the gallbladder, and invasion of the diaphragm, that is, the
direct extension of the tumor into other neighboring organs. If the tumor catego-
ries cannot be assessed, this also needs to be specified in the protocol.
Margins are those areas of the specimen where the surgeon has dissected,
either sharp or blunt, to sever it from the patient. It is essential to recognize the
parenchymal margin, which can be involved by the neoplasm. If the neoplasm
does not involve the parenchymal margin, the invasive carcinoma distance from
the margin in centimeters needs to be stated in the gross description. Additional
margins need to be specified if they are required; for example, if there is an
attached organ involved by the mass, the distance to the margin(s) of that tissue
needs to be recorded.
The cut surface of the mass should be described. This includes color, consis-
tency, and description of the tumor border. Clinical history of ablative, chemo-
therapeutic or similar treatments is important to note. In these cases, the tumor
may be completely or partially necrotic. Inspect the entire tumor and estimate the
percent necrosis. Necrosis in treated liver tumors often appears as homogenous,
crumbly, soft to firm but friable cut surface mottled green/brown. The surround-
ing parenchyma is often white, rubbery and fibrous. If residual tumor is present,
give the size of residual tumor in three dimensions, in centimeters.
Gross Dissection of Liver 83
Lymph nodes
If resected with part of the hilum, the specimen may include soft tissue, including
a few lymph nodes. Ensure all soft tissue at the hilum is carefully inspected, and
all potential nodes found are submitted for histology. If the gallbladder is present,
also inspect the soft tissue around the cystic duct for any lymph nodes. Other
regional lymph nodes of liver tumors (hepatoduodenal ligament nodes, inferior
phrenic, and caval nodes) may be received separately. The number of submitted
lymph nodes need to be adequately identified. The number of lymph nodes
involved, and the number of lymph nodes examined need to be specified in the
gross description report.
Pathologic staging
With regards to the pathologic stage classification according to the pTNM, AJCC
8th edition (6), three descriptors are considered, including the letter “m” as mul-
tiple primary tumors, the letter “r” as recurrent, and the letter “y” as posttreat-
ment. In the TNM, a solitary tumour of 2 cm or less is classified as pT1, while a
tumor with 2 cm or more without vascular invasion is considered pT1b. A solitary
tumor of 2 cm or less is pT1a. The labeling of pT1 is also used if the tumor size is
more than 2 cm without vascular invasion. The labeling of pT2 regards a solitary
tumor of a diameter of more than 2 cm with vascular invasion or multiple tumors,
but none more than 5 cm. In the case of multiple tumors in which at least one is
more than 5 cm, the labeling of pT3 is used. Finally, pT4 is used when a single
tumor or multiple tumors of any diameter involve a major branch of the portal
vein or hepatic vein, or when tumor(s) with specific direct invasion of adjacent
organs other than the gallbladder or with perforation of visceral peritoneum are
found. The labeling of pN1 is used in case of regional lymph node metastasis,
while pN0 when no regional lymph node metastases are seen. The labeling of
pM1 is used when distant metastasis is seen and is required only if confirmed
pathologically. Additional pathologic findings include fibrosis, cirrhosis, low-
grade dysplastic nodule, high-grade dysplastic nodule, steatohepatitis, iron over-
load or chronic hepatitis. Also report on fibrosis present and indicate the
assessment scale used. The protocol described above applies only to hepatic resec-
tion specimens containing hepatocellular carcinoma, including fibrolamellar
carcinoma. Carcinoma of the intrahepatic bile ducts are staged using a separate
TNM system. This protocol should not be used for combined hepatocellular
carcinoma - cholangiocellular carcinoma, sarcomas, and metastatic tumors.
CONCLUSION
Copyright and Permission Statement: The authors confirm that the materials
included in this chapter do not violate copyright laws. Where relevant, a ppropriate
permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
7. Bismuth, H. Surgical anatomy and anatomical surgery of the liver. World J. Surg. 1982;6:3–9. https://
doi.org/10.1007/BF01656368
8. Strasberg SM. Nomenclature of hepatic anatomy and resections: a review of the Brisbane 2000 system.
J Hepatobiliary Pancreat Surg. 2005;12:351–5. https://doi.org/10.1007/s00534-005-0999-7
9. Cantlie, J. On a new arrangement of the right and left lobes of the liver. Proceedings - Anatomical
Society of Great Britain and Ireland. 1897;32:4–9.
10. Makowka L, Stieber AC, Sher L, et al. Surgical technique of orthotopic liver transplantation.
Gastroenterol Clin North Am. 1988;17(1):33–51.
11. Weledji EP, Ngounou E. The Impact of Segmental Anatomy on Hepatic Oncologic Resections. Curr
Surg Rep. 2016;4(4). https://doi.org/10.1007/s40137-015-0122-1
12. Dayton AS, Ro JY, Schwartz MR, Ayala AG, Raymond AK. Raymond’s Paragraph System: an alternative
format for the organization of gross pathology reports and its implementation in an academic teaching
hospital. Arch Pathol Lab Med. 2009;133(2):298–302. https://doi.org/10.5858/133.2.298
5
Radiologic-Pathologic Correlation of
Liver Tumors
Eric Lachance1 • Jake Mandziuk2 • Consolato M. Sergi3 • Justin Bateman2 •
Gavin Low1
1
Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton,
Alberta, Canada; 2Department of Laboratory Medicine and Pathology, University of Alberta,
Edmonton, Alberta, Canada; 3Departments of Pediatrics, Laboratory Medicine and
Pathology, Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada.
Author for correspondence: Jake Mandziuk, Department of Laboratory Medicine and
Pathology, University of Alberta, Edmonton, AB, Canada. Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch5
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
87
88 Lachance E et al.
INTRODUCTION
This chapter is designed to introduce the reader to the radiologic and pathologic
features of common and/or critical tumor or tumor-like diagnoses (lesions) within
the liver. It is divided into hepatocellular lesions, biliary lesions, vascular mesen-
chymal lesions, and metastatic malignancies. However, a broad differential is
important when approaching liver lesions, and a more comprehensive list of
lesions is provided in Table 1.
Hepatocellular
Benign Precursors Malignant
Focal Nodular Hyperplasia Low-Grade Dysplastic Nodule Hepatocellular Carcinoma
Macroregenerative Nodule High-Grade Dysplastic Nodule Hepatoblastoma
Hepatocellular Adenoma
Biliary
Benign Precursors Malignant
Bile Duct Hamartoma Intraductal Papillary Intraductal Papillary Neoplasm with
Neoplasma Associated Invasive Carcinomaa
Bile Duct Adenoma Mucinous Cystic Neoplasma Mucinous Cystic Neoplasm with
Associated Invasive Carcinomaa
Biliary Adenofibroma Cholangiocarcinoma
Solitary Biliary Cyst
Mesenchymal
Benign Intermediate Malignant
Cavernous Hemangioma Inflammatory Myofibroblastic Epithelioid Hemangioendothelioma
Tumor
Infantile Hemangioma Solitary Fibrous Tumor Angiosarcoma
Angiomyolipoma Kaposi Sarcoma Embryonal Rhabdomyosarcoma
Mesenchymal Hamartoma Undifferentiated Embryonal Sarcoma
Other
Metastatic Malignancy
Infection/Abscess
Confluent Hepatic Fibrosis
Hydatid Cyst
Neuroendocrine Neoplasm
Lymphoma
aThe 4th edition of the WHO Classification of Tumors of the Digestive System in 2010 removed the terms biliary
cystadenoma and biliary cystadenocarcinoma, refining into mucinous cystic neoplasms (MCN) and intraductal papillary
neoplasm (IPN), “with associated invasive carcinoma” added, if present. Literature in the interval using the term biliary
cystadenoma and/or cystadenocarcinoma may include MCN and/or IPN, with or without associated invasive carcinoma.
Radiologic-Pathologic Correlation of Liver Tumors 89
HEPATOCELLULAR LESIONS
Hepatocytes are the main functional epithelial cells of the liver and are arranged
within hepatic plates supported by a fine reticulin fiber meshwork. The following
sections discuss lesions involving hepatocytes and includes focal nodular hyper-
plasia, hepatocellular adenoma, hepatocellular carcinoma, and hepatoblastoma.
Figure 1. Representative images of focal nodular hyperplasia. A. On portal phase CT in liver
windows the isoenhancing lesion with hypoenhancing central scar in segment 2/3 is subtly
appreciated (white arrow to the edge of lesion), subtly bulging the inner contour of the left
lobe, a so-called “stealth” lesion. B and C. On MRI in a different patient, a similarly located
segment 2/3 lesion is isointense with the background liver on the T1 weighted image (B) and
T2 weighted image (C). D and E. Following intravenous gadoxetic acid administration, the
lesion shows homogenous hyperenhancement during the arterial (D) and portal (E) phases.
F. On the hepatobiliary phase at 20 minutes, the lesion shows uniformly higher signal than
the background liver. G and H. A partial hepatectomy from a separate patient shows a
well-circumscribed lesion with a central stellate scar (white arrow) and radiating fibrous
septae, seen grossly (G) and microscopically (H). I. Ductular reaction (black arrow) is present
at the interface of fibrous regions with nodules. Slides are stained with Hematoxylin and
Eosin. Total image magnification: H - 25X; I - 100X.
scar, and a ductular reaction is seen at the interface between the fibrous septae and
hyperplastic nodules. Normal bile ducts and portal tracts are absent. Glutamine
synthetase demonstrates a characteristic ‘map-like’ pattern of staining by immu-
nohistochemistry (6–7).
FNH can be subtle on unenhanced imaging, due to the benign hyperplastic
tissue blending in with adjacent liver parenchyma, classically described as a
“stealth lesion” (8). On ultrasound, the central scar may be indistinctly visualized;
otherwise, FNH echogenicity is varied and nonspecific. However, contrast-
enhanced ultrasound, CT, or MRI is usually diagnostic. The central feeding artery
with centrifugal flow may be seen as the characteristic “spoke-wheel” pattern
on color Doppler, CEUS, and sometimes CT arterial phase, most commonly in
larger lesions (9). FNH is typically a subtle hypoattenuating or isoattenuating
Radiologic-Pathologic Correlation of Liver Tumors 91
Hepatocellular adenoma
Hepatocellular adenoma (HCA) (Figure 2) is a benign hepatocellular neoplasm,
which may be solitary or multiple (2). Liver adenomatosis is defined by 10 or
more HCAs and is diagnosed most frequently in patients with nodules greater
than 5 cm (12). Hemorrhage is the first presentation in 15% of patients and is
associated with lesions greater than 5 cm (13). Grossly, HCAs are non-encapsulated
and soft tan or red-brown masses that occur within a non-cirrhotic liver.
Microscopically, the tumor is composed of benign hepatocytes arranged in plates
up to two cells thick with unpaired arteries and an absence of portal tracts.
Currently, there are four recognized genotypic-phenotypic subtypes: (i) HNF1α-
inactivated HCA with marked steatosis and loss of liver fatty acid-binding protein
expression by IHC, (ii) inflammatory HCA (JAK/STAT3 pathway activation) with
inflammation, sinusoidal dilatation, ductular reaction, and diffuse expression of
C-reactive protein and serum amyloid A by IHC, (iii) beta-catenin activated
inflammatory HCA, and (iv) beta-catenin activated HCA. Beta-catenin activated
HCAs may show atypical cytological and/or architectural features, suggesting
transformation to hepatocellular carcinoma (HCC) (14). Evaluation of IHC stains
may show nuclear beta-catenin expression along with over-expression of gluta-
mine synthetase within these beta-catenin activated HCAs. Careful evaluation of a
reticulin stain is essential to help exclude malignant transformation.
Imaging features of HCA are often varied, with slightly different characteristics
depending on the subtype. Hemorrhage with possible subsequent calcification,
fat, and encapsulation may be seen. Of note, HCAs 5 cm or larger are at higher
risk for both hemorrhage and malignant transformation into HCC, and require
close imaging follow up. Males with HCAs are often treated by prophylactic surgi-
cal resection, particularly for solitary or large lesions, as a result of the increased
incidence of the beta-catenin subtype.
Ultrasound is nonspecific, demonstrating a well-circumscribed lesion with
variable echogenicity, more often hyperechoic. CEUS may increase specificity,
particularly showing centripetal arterial flow, differentiating HCA from FNH,
which typically illustrate a centrifugal arterial flow. Multiphasic contrast-enhanced
92 Lachance E et al.
Figure 2. Representative images of hepatocellular adenoma. A and B. The large lesion (>5 cm)
in segments 5 and 6 shows signal loss on the opposed phase T1 weighted image (B)
compared to the in-phase T1 weighted image (A) denoting microscopic fat content. C. The
lesion shows heterogeneous high signal on the T2 weighted image. D, E, and F. Following
gadolinium administration, the lesion shows heterogeneous arterial (D) and portal phase (E)
hyperenhancement and delayed phase washout (F). An ancillary finding of an enhancing
capsule is depicted in (E) and (F). G. A partial hepatectomy from a separate patient shows a
tan and poorly defined lesion grossly, with areas of hemorrhage and necrosis. H.
Microscopically the neoplastic lesion (white arrow) shows an absence of portal tracts and is
notably different from the background parenchyma (black arrow). I. Benign hepatocytes are
arranged in cell plates up to two cells thick. Slides are stained with Hematoxylin and Eosin.
Total image magnification: H - 25X; I - 400X.
Hepatocellular carcinoma
Hepatocellular carcinoma (HCC) (Figure 3) is a malignant hepatocellular neo-
plasm, commonly occurring in the setting of underlying chronic liver disease (2).
Traditionally, macroscopic patterns of HCC are described as single or multiple
liver nodules, massive nodules that may involve multiple liver segments, and dif-
fuse forms with multiple small nodules throughout the liver mimicking cirrhosis
(16). Tumor nodules smaller than the main tumor and less than 2 cm away (sepa-
rated by non-neoplastic parenchyma) are classified as satellite nodules and
usually represent intrahepatic metastases. Nodules that are far from the primary
tumor may represent either synchronous tumors or intrahepatic metastases (16).
Involvement and spread through intrahepatic portal vein branches are common.
Grossly, HCC is soft with a variegated appearance ranging from green to light
tan depending on bile and fat content respectively, and foci of hemorrhage or
necrosis. Tumors may have a capsule of fibrotic and inflamed tissue and can be
seen invading large vascular structures. Fibrolamellar and scirrhous subtypes may
appear firmer and more fibrotic compared to conventional HCCs, and these fea-
tures may mimic the central scar of FNH or even cholangiocarcinoma. Fibrolamellar
carcinoma is a distinct lesion affecting younger adults without pre-existing cir-
rhosis or elevated serum AFP. It is typically a large mass with calcification, hemor-
rhage, or necrosis, can mimic FNH with a central scar, commonly metastasizes to
lymph nodes and distant sites, and shows a DNAJB1-PRKACA fusion transcript
(17–18).
Microscopically, HCC shows variable degrees of cytological and architectural
atypia with thickened hepatic plates and disruption/loss of the reticulin frame-
work. Hepatocellular differentiation is often apparent on hematoxylin and eosin
(H&E); however, IHC stains (Hep Par-1 and Arginase-1) are sometimes necessary
for poorly differentiated tumors to establish hepatocellular origin. Furthermore,
careful examination of reticulin stain and identification of abnormal expression of
Glypican-3 IHC may be required in well-differentiated tumors to establish a diag-
nosis of carcinoma. Increased arterial blood flow leads to sinusoidal capillariza-
tion, which can be demonstrated by CD34 IHC. There are four predominant
growth patterns including trabecular (70%), solid (20%), pseudoglandular (10%),
and macrotrabecular with cell plates greater than ten cells in thickness (1%),
although mixed patterns are common (19). Approximately 35% of HCCs show
distinct clinicopathologic subtypes, including steatohepatitic (most commonly in
patients with underlying fatty liver disease), clear cell (cytoplasmic clearing sec-
ondary to glycogen accumulation), scirrhous (fibrosis in the majority of the tumor,
separate small nests of tumor cells in thin trabeculae), and fibrolamellar (large
polygonal hepatocytes with abundant cytoplasm and prominent nucleoli, pale
bodies, thick collagenous bands in parallel orientations, and non-cirrhotic back-
ground liver) (20). Combined hepatocellular cholangiocarcinoma is a rare diag-
nosis with convincing HCC and cholangiocarcinoma morphologies present within
the same tumor.
Precursor lesions of HCC are commonly identified in hepatectomy specimens
and occasionally in biopsy specimens in patients with advanced chronic liver
disease. These precursor lesions include dysplastic foci (<1 mm) and dysplastic
nodules (approximately 5–15 mm) (21). Dysplastic foci are incidental findings
and include small cell and large cell cytological change or small groups of hepato-
cytes with an otherwise clonal appearance (for example, iron free foci). Dysplastic
nodules can be low- or high-grade, and the high-grade dysplastic nodules can
mimic small (<2 cm) early HCCs. Features that favor the designation of HCC over
a dysplastic nodule include a nodule-in-nodule growth pattern, stromal invasion,
loss of reticulin, and expression of Glypican-3 by IHC. Low-grade dysplastic nod-
ules require distinction from large regenerative nodules, although this distinction
can be challenging and of limited significance in explanted livers (2).
Across imaging modalities, differentiation of small HCCs from regenerative or
dysplastic nodules can be challenging in the setting of cirrhosis. Smaller lesions
Radiologic-Pathologic Correlation of Liver Tumors 95
Hepatoblastoma
Hepatoblastoma (HB) is a primary malignant liver neoplasm that recapitulates the
developing fetal/embryonal liver with variable proportions of epithelial and mes-
enchymal elements and is the most common malignant liver tumor in children
(30). HB is frequently a solitary mass; however, poor prognostic features include
multifocality (20%), the involvement of the vena cava or all three hepatic veins
(10%), the involvement of the portal bifurcation or both right and left portal veins
(10%), extrahepatic tumor extension (5%), and tumor rupture (5%) (31). Grossly,
tumors are nodular or bosselated, and cut surfaces depend on tumor components,
96 Lachance E et al.
ranging from soft tan-brown in fetal patterns to gritty whitish speckles of osteoid
in mesenchymal patterns. A variegated appearance can result from cystic degen-
eration, necrosis, or hemorrhage.
Microscopically, HBs are classified according to the International Pediatric
Liver Tumors Consensus Classification of Liver Hepatoblastoma (32). Tumors
can be either purely epithelial or contain a mixture of epithelial and mesenchymal
components (spindle cells, osteoid, or cartilage) (32). Epithelial tumors may
show a single or combination of histologic patterns including: fetal (characterized
by trabecular growth of fetal-type hepatocytes, clear or finely granular cytoplasm
depending on cytoplasmic glycogen content, variable degrees of mitotic activity,
and nuclear pleomorphism), embryonal (primitive tubule formation, angulated
nuclei, and high nuclear-to-cytoplasmic ratios), macrotrabecular (fetal or
embryonal patterns in trabeculae greater than 5 cells thick), small cell
undifferentiated (diffuse sheets or nests of cells with minimal cytoplasm, frequent
apoptosis, mitotic activity, and necrosis), and cholangioblastic (formation of
small ducts). The fetal pattern can be further subclassified into those with
low-mitotic activity (well-differentiated), mitotically active (crowded fetal), and
pleomorphic types. The small cell undifferentiated pattern can be further
subdivided into SMARCB1 (INI1) positive and negative types with the help of
IHC (2). Mixed epithelial and mesenchymal tumors are further subclassified
into those with and without teratoid features. Given the wide variation in
histological patterns, biopsies may not be representative of the tumor overall.
Neoadjuvant chemotherapy can markedly alter the histologic appearance with
changes including fibrosis, necrosis/hemorrhage, and the presence of osteoid-like
tissue (30).
Ultrasound is commonly the first modality used when detecting HB, with
further characterization by CT or MRI. MRI is advantageous in children, given
the lack of radiation, and likely has increased sensitivity and specificity with
gadoxetic acid MRI. Ultrasound appearances are dependent on the underlying
histologic pattern. Generally, these lesions are lobular, well-circumscribed
hypoechoic or heterogeneous lesions. Calcification, necrosis and vascular inva-
sion may be seen. HB is typically heterogeneous and hypoattenuating to the liver
parenchyma on CT. Speckled or amorphous calcification is seen in greater
than 50% of lesions (33). Enhancement is typically less than that of the liver;
however, peripheral arterial enhancement may be observed (33). On MRI,
hepatoblastomas are typically heterogeneously T2WI hyperintense and T1WI
hypointense. On contrast-enhanced MRI, HB is typically hypointense on the
arterial, portal-venous, and delayed phases (34–35). The presence of a liver mass
and elevated serum AFP in a child less than 4 years should alert the radiologist
to the possibility of HB.
BILIARY LESIONS
A network of bile canaliculi and a system of bile ducts located within portal tracts
transports bile into larger intrahepatic ducts, exiting at the liver hilum. The fol-
lowing sections discuss lesions involving bile ducts and includes mucinous cystic
neoplasm and intrahepatic cholangiocarcinoma.
Radiologic-Pathologic Correlation of Liver Tumors 97
Intrahepatic cholangiocarcinoma
Intrahepatic cholangiocarcinoma (ICCA) (Figure 4) is a primary intrahepatic
malignant neoplasm of epithelial cells with biliary differentiation. Two subtypes
with unique clinicopathological features have been described: (i) large duct ICCA,
arising from large perihilar intrahepatic bile ducts proximal to the right and left
hepatic ducts, and (ii) small duct ICCA arising from small bile ducts or ductules
in a peripheral location. Large duct and small duct subtypes account for 45% and
55% of all ICCAs, respectively (40), with 60% of all ICCAs being greater than
5 cm in size (41).
Grossly, large duct ICCAs often present as mass lesions around the larger cali-
ber bile duct branches with invasion into the surrounding hepatic parenchyma.
Some large duct ICCAs will not form a mass lesion but rather spread along bile
ducts in a diffuse and longitudinal manner (referred to as a “periductal infiltrat-
ing” pattern of growth) (42). They may be found in association with macroscopi-
cally visible intraductal papillary neoplasms of the bile ducts, not discussed within
98 Lachance E et al.
Figure 4. Representative images of intrahepatic cholangiocarcinoma. A and B. The lesion shows
low signal on the T1 weighted image (A) and intermediate to high signal on the T2 weighted
image (B) with surrounding mild peripheral intrahepatic biliary dilatation. C. The lesion
shows intense diffusion restriction on the b=150 s/mm2 image. D, E, and F. Following
gadolinium administration, the lesion shows heterogeneous arterial (D) and portal phase (E)
enhancement (predominantly in the periphery), with progressive centripetal filling-in,
completely hyperenhancing on the delayed phase (F) at 5 minutes. G. A partial hepatectomy
from a separate patient shows a tan-white sclerosing lesion grossly, with extensive
involvement of a probed large duct. H. Microscopically an invasive adenocarcinoma (white
arrow) is seen arising from a large duct (black arrow). I. Well-formed glands are seen
invading a nerve. Slides are stained with Hematoxylin and Eosin. Total image magnification:
H - 25X; I - 200X.
cases may lack apparent glandular differentiation and require IHC to exclude
other primary neoplasms (for example, poorly differentiated HCC). Combined
hepatocellular cholangiocarcinoma is a rare malignancy composed of both
unequivocal HCC and ICCA components within the same tumor and may occa-
sionally be encountered in biopsy specimens. ICCA does not show a specific mor-
phology or IHC profile. Therefore, careful clinical and radiological correlation is
essential to rule out a metastatic adenocarcinoma from another site.
On imaging, the mass-forming pattern of ICCA demonstrates a
well-circumscribed, often large, lobulated mass frequently with satellite nodules.
The periductal-infiltrating pattern demonstrates growth along bile ducts with an
elongated, branching, spiculated appearance. Capsular retraction, more typical of
the mass-forming pattern, is characteristic but not pathognomonic for ICCA.
Upstream biliary ductal dilatation is typical but not specific.
Mass-forming ICCAs are typically hyperechoic on ultrasound if larger than
3 cm and isoechoic or hypoechoic if smaller (43). A peripheral hypoechoic rim is
seen in approximately 35% of cases (43). On unenhanced CT, they are typically
hypoattenuating (43), and calcification may be present. They are typically hetero-
geneously T1WI hypointense and demonstrate a T2WI hyperintense peripheral
rim with central hypodensity, which may be hyperintense in the setting of internal
necrosis (43). The enhancement pattern on MRI and CT is typically arterial phase
continuous rim-like enhancement (targetoid) with progressive, concentric cen-
tripetal fill-in on the portal-venous phase and persistent enhancement on delayed
imaging (43).
Periductal-infiltrating ICCAs are typically enhancing on CT and MRI (43). Bile
duct imaging, including endoscopic retrograde cholangiopancreatography (ERCP)
and magnetic resonance cholangiopancreatography (MRCP), are highly sensitive
and specific for bile duct narrowing and stricturing, which may be otherwise subtle.
MRCP is 97% sensitive and 98% specific for the presence of obstruction (44).
Differentiating HCC from ICCA is important as locoregional treatments for
HCC (for example, ablative or transcatheter therapy) are not effective for ICCA,
and liver transplantation is a relative contraindication for ICCA due to the high
risk of recurrence (45). A helpful finding is the presence of tumor thrombus, more
commonly associated with HCC, while ICCA more commonly causes venous
occlusion from extrinsic compression.
Cavernous hemangioma
Cavernous hemangioma (CH) of the liver is a benign venous malformation with
no malignant potential. CHs are most often solitary and are categorized according
100 Lachance E et al.
to size: small (less than 5 cm), large (5–10 cm), and giant (>10 cm). Frequencies
are approximately 30%, 35%, and 35%, respectively (46). Grossly, CHs are well-
circumscribed, soft, spongy purple-red lesions located subcapsular or deep in the
liver parenchyma. Microscopically, they consist of variably sized large vascular
spaces lined by a bland epithelium and are embedded in a fibrous stroma. Thrombi
may be present, and when extensive, can result in significant sclerosis of the
lesion. Although well-circumscribed grossly, the tumor interface microscopically
is irregular, with separate hemangioma-like vessels located 0.1–2 cm away from
the lesion in 80% of cases (47).
As the most common benign liver tumor, CHs are frequently detected inciden-
tally on imaging and can show a wide variety of atypical appearances. CHs are
typically well-circumscribed and hyperechoic on ultrasound, with a well margin-
ated lobulated contour that is better appreciated in larger CHs. Heterogeneity is
more common with giant CHs, although the lesion periphery typically remains
echogenic (48). A central scar may be seen in a minority of giant CHs, mimicking
FNH, and calcification is rare, sometimes seen in the central scar. No central flow
is demonstrated using color Doppler; however, peripheral vascularity can be
present. A “reverse target” morphology with hyperechoic rim may be seen in a
minority, with metastasis or HCC as the diagnosis of exclusion on a background
of normal or cirrhotic liver parenchyma, respectively (49). Where CEUS is
available, enhancement patterns parallel CT and MRI.
On CT, attenuation and enhancement across phases mirror blood pool (for
example, the aorta), due to the underlying disorganized venous histology, with
more rapid (“flash”) filling and delayed/centripetal filling observed in small and
giant CHs, respectively. The peripheral nodular contour with discontinuous
enhancement is more conspicuous with large and giant CHs. The central scar, if
present, is characteristically non-enhancing. MRI has the greatest sensitivity and
specificity for diagnosing CH. On MRI, CHs are T1WI isointense or hypointense,
with hypointensity typically observed in giant CHs. If present, the central scar is
markedly T1WI hypointense. CHs are typically T2WI hyperintense due to the
long T2 relaxation time in these lesions. MRI contrast enhancement patterns are
similar to those described for CT; however, some CHs show slow enhancement
with fill-in on delayed phase images, and rarely a centrifugal (inside-out) enhance-
ment pattern (50).
Typically reserved for diagnostic clarification in patients unable to undergo
MRI or contrast-enhanced studies, Tc-99m labelled RBC scans demonstrate pho-
topenia or decreased activity relative to liver parenchyma on early dynamic scans
with persistent filling of large and giant CHs on delayed imaging. This differenti-
ates CHs from vascular tumors such as HCC, HCA, and FNH. When combined
with multi-headed SPECT the positive predictive value approaches 100%, with
sensitivity markedly improving in larger lesions (20% in lesions 0.5–0.9 cm;
100% in lesions >1.4 cm) (51).
Epithelioid hemangioendothelioma
Epithelioid hemangioendothelioma (EHE) (Figure 5) of the liver is a malignant
endothelial neoplasm. EHE is frequently multifocal (87%), bilobar (81%), and
arises in the absence of chronic liver disease (52). Grossly, there is nodular or
Radiologic-Pathologic Correlation of Liver Tumors 101
Hepatic angiosarcoma
Hepatic angiosarcoma (HAS) is a primary malignant endothelial neoplasm. HAS
is predominantly bilobar (80%) and can show variable patterns of growth, includ-
ing multinodular (40%), solitary (20%), dominant mass with satellite nodules
(20%), and diffusely infiltrating (20%) (59). Grossly, tumors are poorly defined
and heterogenous, with solid gray-white areas and hemorrhagic areas showing
large blood-filled spaces. Microscopically, tumors also display heterogeneity. Well-
formed anastomosing vascular channels alternate with solid sheets of tumor lack-
ing vascular formation. Tumor cells can show high-grade features, including
epithelioid or spindled cells with marked nuclear pleomorphism, hyperchromatic
nuclei, and abundant mitoses. Infarct, necrosis, and fibrosis are common. At the
periphery of the tumor, malignant cells characteristically grow along and replace
endothelial cells present in pre-existing sinusoids. A high index of suspicion is
required in some cases, along with a panel of IHC stains, to make a correct
diagnosis. Epithelioid HAS in particular can mimic carcinomas, melanomas, and
lymphomas by morphology alone, and therefore a panel of IHC stains is generally
recommended, including markers of vascular differentiation such as CD31 and
ERG (keratins and EMA may be positive) (2).
HAS demonstrates a variable appearance on radiologic imaging, reflecting the
underlying pleomorphic histology. HAS is commonly heterogeneous and hyper-
vascular, with multiplicity, multiorgan involvement, rapidly progressive growth,
and a history of exposure to particular carcinogens (for example, vinyl chloride
polymers) pointing toward the underlying diagnosis, which ultimately relies on
histology. Hemorrhage, necrosis, and calcification may be seen. A background of
liver cirrhosis or fibrosis is reported in approximately 40% of patients (60). On
ultrasound, lesions are generally heterogeneous. On unenhanced CT, lesions are
typically hypoattenuating, although heterogeneity may be seen, particularly in the
setting of hemorrhage. On MRI, lesions are heterogeneous and may have an ill-
defined outline. They may be T1WI hypointense but can show high signal with
Radiologic-Pathologic Correlation of Liver Tumors 103
METASTATIC MALIGNANCIES
Metastatic malignancies are common in the liver (more common than primary
malignancies) and frequently multifocal; however, solitary nodules and diffuse
parenchymal involvement without discrete nodule formation is possible. Tumors
are primarily carcinomas (92%), of which adenocarcinomas account for the
majority (75%), but melanomas (2.4%) and sarcomas (1%) are also found (62).
The most frequent primary sites of origin for adenocarcinomas are colorectal
(46%), pancreas (10%), breast (8%), and lung (4%) (62). Gross and microscopic
appearances differ based on the primary site of origin. Many cases of metastases
involving the liver can be quickly sorted out based on a combination of tumor
morphology/immunophenotype, clinical history, and radiology. Some cases, how-
ever, present a much greater challenge for pathologists, radiologists, and clinicians
alike, and a multidisciplinary team-based approach can be tremendously helpful
in determining the site of origin and/or appropriate management.
The radiologic appearance of metastatic malignancies also varies, with
appearances mimicking those seen at the respective sites of primary malignancies.
Common examples include hypovascular epithelial metastases (for example,
colon, lung, and gastric), hypervascular metastases (for example, renal cell
carcinoma, melanoma, thyroid, and neuroendocrine tumors), and cystic liver
metastases (for example, mucinous or serous primary malignancies from the
ovary, colon, and pancreas).
In comparison to the gold standard of resection or intraoperative ultrasound,
conventional ultrasound has modest sensitivity for metastases (50–76%), improv-
ing with contrast-enhancement (82–86%) (63), showing similar sensitivities to
contrast-enhanced CT and MRI (64–65). Ultrasound appearances are varied with
the following typical appearances: hypoechoic in hypovascular metastases, cen-
trally hyperechoic and peripherally hypoechoic (targetoid) in aggressive metasta-
ses, and hyperechoic in hypervascular metastases.
Unenhanced CT has poor sensitivity for most metastases, with high sensitiv-
ity for calcification, a nonspecific but common finding in mucinous lesions.
Metastases are typically T1WI hypointense and T2WI hyperintense, heteroge-
neous in morphology, and may be associated with an ill-defined outline.
Enhancement characteristics are reflective of the predominant hepatic
arterial supply, appearing similar on CT and conventional extracellular
104 Lachance E et al.
CONCLUSION
Copyright and permission statement: The authors confirm that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Barr RG, Wilson SR, Lyshchik A, McCarville B, Darge K, Grant E, et al. Contrast-enhanced Ultrasound-
State of the Art in North America: Society of Radiologists in Ultrasound White Paper. Ultrasound Q.
2020 Sep;36(4S):S1–S39. https://doi.org/10.1097/RUQ.0000000000000515
2. WHO Classification of Tumours Editorial Board. Digestive System Tumours, WHO Classification of
Tumours. 5th ed. Lyon, France: International Agency for Research on Cancer; 2019. p. 221–244,
250–3, 466–7, 471–2.
3. Wanless IR, Mawdsley C, Adams R. On the pathogenesis of focal nodular hyperplasia of the liver.
Hepatology. 1985;5(6):1194–200. https://doi.org/10.1002/hep.1840050622
Radiologic-Pathologic Correlation of Liver Tumors 105
4. Nguyen BN, Fléjou JF, Terris B, Belghiti J, Degott C. Focal nodular hyperplasia of the liver: a compre-
hensive pathologic study of 305 lesions and recognition of new histologic forms. Am J Surg Pathol.
1999;23(12):1441–54. https://doi.org/10.1097/00000478-199912000-00001
5. Shen YH, Fan J, Wu ZQ, Ma ZC, Zhou XD, Zhou J, et al. Focal nodular hyperplasia of the liver in 86
patients. Hepatobiliary Pancreat Dis Int. 2007;6(1)52–7.
6. Bioulac-Sage P, Laumonier H, Rullier A, Cubel G, Laurent C, Zucman-Rossi J, et al. Over-expression of
glutamine synthetase in focal nodular hyperplasia: a novel easy diagnostic tool in surgical pathology.
Liver Int. 2009;29(3):459–65. https://doi.org/10.1111/j.1478-3231.2008.01849.x
7. Bioulac-Sage P, Cubel G, Taouji S, Scoazec JY, Leteurtre E, Paradis V, et al. Immunohistochemical
markers on needle biopsies are helpful for the diagnosis of focal nodular hyperplasia and hepa-
tocellular adenoma subtypes. Am J Surg Pathol. 2012;36(11):1691–9. https://doi.org/10.1097/
PAS.0b013e3182653ece
8. Buetow PC, Pantongrag-Brown L, Buck JL, Ros PR, Goodman ZD. Focal nodular hyperplasia of the
liver: radiologic-pathologic correlation. Radiographics. 1996;16(2):369–88. https://doi.org/10.1148/
radiographics.16.2.8966294
9. Ungermann L, Eliás P, Zizka J, Ryska P, Klzo L. Focal nodular hyperplasia: Spoke-wheel arterial
pattern and other signs on dynamic contrast-enhanced ultrasonography. Eur J Radiol. 2007;63(2):
290–4. https://doi.org/10.1016/j.ejrad.2007.01.026
10. Ronot M, Vilgrain V. Imaging of benign hepatocellular lesions: current concepts and recent updates.
Clin Res Hepatol Gastroenterol. 2014;38(6):681–8. https://doi.org/10.1016/j.clinre.2014.01.014
11. Grazioli L, Bondioni MP, Haradome H, Motosugi U, Tinti R, Frittoli B, et al. Hepatocellular adenoma
and focal nodular hyperplasia: value of gadoxetic acid-enhance MR imaging in differential diagnosis.
Radiology. 2012;262(2):520–9. https://doi.org/10.1148/radiol.11101742
12. Barbier L, Nault JC, Dujardin F, Scotto B, Besson M, de Muret A, et al. Natural history of liver adeno-
matosis: A long-term observational study. J Hepatol. 2019;71(6):1184–92. https://doi.org/10.1016/j.
jhep.2019.08.004
13. Laurent A, Dokmak S, Nault JC, Pruvot, FR, Fabre JM, Letoublon C, et al. European experience of
573 liver resections for hepatocellular adenoma: a cross-sectional study by the AFC-HCA-2013 study
group. HPB (Oxford). 2016;18(9):748–55. https://doi.org/10.1016/j.hpb.2016.06.011
14. Gonzalez RS, Raza A, Propst R, Adeyi O, Bateman J, Sopha SC, et al. Recent Advances in Digestive
Tract Tumors: Updates From the 5th Edition of the World Health Organization “Blue Book”. Arch
Pathol Lab Med. 2020. https://doi.org/10.5858/arpa.2020-0047-RA
15. Dharmana H, Saravana-Bawan S, Girgis S, Low G. Hepatocellular adenoma: imaging review of
the various molecular subtypes. Clin Radiol. 2017;72(4):276–85. https://doi.org/10.1016/j.crad.
2016.12.020
16. Martins-Filho SN, Alves VAF. The strengths and weaknesses of gross and histopathological evaluation
in hepatocellular carcinoma: a brief review. Surg Exp Pathol. 2019;2(1):23. https://doi.org/10.1186/
s42047-019-0047-6
17. Kim DH, Choi SH, Kim SY, Kim MJ, Lee SS, Byun JH. Gadoxetic Acid-enhanced MRI of Hepatocellular
Carcinoma: Value of Washout in Transitional and Hepatobiliary Phases. Radiology. 2019;291(3):651–7.
https://doi.org/10.1148/radiol.2019182587
18. Graham RP, Jin L, Knutson DL, Kloft-Nelson SM, Greipp PT, Waldburger N, et al. DNAJB1-PRKACA
is specific for fibrolamellar carcinoma. Mod Pathol. 2015;28(6):822–9. https://doi.org/10.1038/
modpathol.2015.4
19. Rastogi A. Changing role of histopathology in the diagnosis and management of hepatocellular carci-
noma. World J Gastroenterol. 2018;24(35):4000–13. https://doi.org/10.3748/wjg.v24.i35.4000
20. El Jabbour T, Lagana SM, Lee H. Update on hepatocellular carcinoma: Pathologists’ review. World J
Gastroenterol. 2019;25(14):1653–65. https://doi.org/10.3748/wjg.v25.i14.1653
21. International Working Party. Terminology of nodular hepatocellular lesions. Hepatology.
1995;22(3):983–93. https://doi.org/10.1016/0270-9139(95)90324-0
22. Elsayes KM, Kielar AZ, Elmohr MM, Chernyak V, Masch WR, Furlan A, et al. White paper of the
Society of Abdominal Radiology hepatocellular carcinoma diagnosis disease-focused panel on
106 Lachance E et al.
39. Mavilia MG, Pakala T, Molina M, Wu GY. Differentiating Cystic Liver Lesions: A Review of Imaging
Modalities, Diagnosis and Management. J Clin Transl Hepatol. 2018;6(2):208–16. https://doi.
org/10.14218/JCTH.2017.00069
40. Akita M, Fujikura K, Ajiki T, Fukumoto T, Otani K, Azuma T, et al. Dichotomy in intrahepatic
cholangiocarcinomas based on histologic similarities to hilar cholangiocarcinomas. Mod Pathol.
2017;30(7):986–97. https://doi.org/10.1038/modpathol.2017.22
41. Bagante F, Spolverato G, Merath K, Weiss M, Alexandrescu S, Marques HP, et al. Intrahepatic cholan-
giocarcinoma tumor burden: A classification and regression tree model to define prognostic groups
after resection. Surgery. 2019;166(6):983–90. https://doi.org/10.1016/j.surg.2019.06.005
42. Uno M, Shimada K, Yamamoto Y, Nara S, Esaki M, Sakamoto Y, et al. Periductal infiltrating type of
intrahepatic cholangiocarcinoma: a rare macroscopic type without any apparent mass. Surg Today.
2012;42(12):1189–94. https://doi.org/10.1007/s00595-012-0145-5
43. Chung YE, Kim MJ, Park YN, Choi JY, Pyo JY, Kim YC, et al. Varying appearances of cholangio-
carcinoma: radiologic-pathologic correlation. Radiographics. 2009;29(3):683–700. https://doi.org/
10.1148/rg.293085729
44. Romagnuolo J, Bardou M, Rahme E, Joseph L, Reinhold C, Barkun AN. Magnetic resonance cholan-
giopancreatography: a meta-analysis of test performance in suspected biliary disease. Ann Intern Med.
2003;139(7):547–57. https://doi.org/10.7326/0003-4819-139-7-200310070-00006
45. Ho AK, Girgis S, Low G. Uncommon liver lesions with multimodality imaging and pathology correla-
tion. Clin Radiol. 2018;73(2):191–204. https://doi.org/10.1016/j.crad.2017.07.016
46. Di Carlo I, Koshy R, Al Mudares S, Ardiri A, Bertino G, Toro A. Giant cavernous liver hemangiomas:
is it the time to change the size categories? Hepatobiliary Pancreat Dis Int. 2016;15(1):21–9. https://
doi.org/10.1016/S1499-3872(15)60035-2
47. Kim GE, Thung SN, Tsui WM, Ferrell LD. Hepatic cavernous hemangioma: underrecognized associated
histologic features. Liver Int. 2006;26(3):334–8. https://doi.org/10.1111/j.1478-3231.2005.01226.x
48. Moody AR, Wilson SR. Atypical hepatic hemangioma: a suggestive sonographic morphology.
Radiology. 1993;188(2):413–7. https://doi.org/10.1148/radiology.188.2.8327687
49. Wu S, Zuo D, Hong Y. The Prevalence of Hyperechoic Rim in Solid Focal Liver Lesions and Its
Implication. J Diagn Med Sonogr. 2018;35(1):3–8. https://doi.org/10.1177/8756479318798365
50. Mathew RP, Sam M, Raubenheimer M, Patel V, Low G. Hepatic hemangiomas: the various imag-
ing avatars and its mimickers. Radiol Med. 2020;125(9):801–15. https://doi.org/10.1007/
s11547-020-01185-z
51. Ziessman HA, Silverman PM, Patterson J, Harkness B, Fahey FH, Zeman RK, et al. Improved detection
of small cavernous hemangiomas of the liver with high-resolution three-headed SPECT. J Nucl Med.
1991;32(11):2086–91.
52. Mehrabi A, Kashfi A, Fonouni H, Schemmer P, Schmied BM, Hallscheidt P, et al. Primary malignant
hepatic epithelioid hemangioendothelioma: a comprehensive review of the literature with emphasis
on the surgical therapy. Cancer. 2006;107(9):2108–21. https://doi.org/10.1002/cncr.22225
53. Patel NR, Salim AA, Sayeed H, Sarabia SF, Hollingsworth F, Warren M, et al. Molecular character-
ization of epithelioid haemangioendotheliomas identifies novel WWTR1-CAMTA1 fusion variants.
Histopathology. 2015;67(5):699–708. https://doi.org/10.1111/his.12697
54. Mehrabi A, Kashfi A, Fonouni H, Schemmer P, Schmied BM, Hallscheidt P, et al. Primary malignant
hepatic epithelioid hemangioendothelioma. Cancer. 2006;107(9):2108–21. https://doi.org/10.1002/
cncr.22225
55. Mamone G, Miraglia R. The “Target sign” and the “Lollipop sign” in hepatic epithelioid hemangioen-
dothelioma. Abdom Radiol (NY). 2019;44(4):1617–20. https://doi.org/10.1007/s00261-018-1820-9
56. Paolantonio P, Laghi A, Vanzulli A, Grazioli L, Morana G, Ragozzino A, et al. MRI of hepatic epithelioid
hemangioendothelioma (HEH). J Magn Reson Imaging. 2014;40(3):552–8. https://doi.org/10.1002/
jmri.24391
57. Makhlouf HR, Ishak KG, Goodman ZD. Epithelioid hemangioendothelioma of the liver: a
clincicopathologic study of 137 cases. Cancer. 1999;85(3):562–82. https://doi.org/10.1002/
(SICI)1097-0142(19990201)85:3<562::AID-CNCR7>3.0.CO;2-T
108 Lachance E et al.
58. Alomari AI. The lollipop sign: A new cross-sectional sign of hepatic epithelioid hemangioendothe-
lioma. Eur J Radiol. 2006;59(3):460–4. https://doi.org/10.1016/j.ejrad.2006.03.022
59. Wilson GC, Lluis N, Nalesnik MA, Nassar A, Serrano T, Ramos E, et al. Hepatic Angiosarcoma: A
Multi-institutional, International Experience with 44 Cases. Ann Surg Oncol. 2019;26(2):576–82.
https://doi.org/10.1245/s10434-018-7062-9
60. Pickhardt PJ, Kitchin D, Lubner MG, Ganeshan DM, Bhalla S, Covey AM. Primary hepatic angiosar-
coma: multi-institutional comprehensive cancer centre review of multiphasic CT and MR imaging in
35 patients. Eur Radiol. 2015;25(2):315–22. https://doi.org/10.1007/s00330-014-3442-0
61. Koyama T, Fletcher JG, Johnson CD, Kuo MS, Notohara K, Burgart LJ. Primary hepatic angiosar-
coma: findings at CT and MR imaging. Radiology. 2002;222(3):667–73. https://doi.org/10.1148/
radiol.2223010877
62. de Ridder J, de Wilt JHW, Simmer F, Overbeek L, Lemmens V, Nagetegaal I. Incidence and origin of
histologically confirmed liver metastases: an explorative case-study of 23,154 patients. Oncotarget.
2016;7(34):55368-76. https://doi.org/10.18632/oncotarget.10552
63. Albrecht T. Detection and Characterisation of Liver Metastases. In: Lencioni R, editor. Enhancing
the Role of Ultrasound with Contrast Agents. Milano: Springer; 2006. p. 53–67. https://doi.
org/10.1007/88-470-0476-4_4
64. Dietrich CF, Nolsøe CP, Barr RG, Berzigotti A, Burns PN, Cantisani V, et al. Guidelines and Good
Clinical Practice Recommendations for Contrast-Enhanced Ultrasound (CEUS) in the Liver-Update
2020 WFUMB in Cooperation with EFSUMB, AFSUMB, AIUM, and FLAUS. Ultrasound Med Biol.
2020;46(10):2579–2604. https://doi.org/10.1016/j.ultrasmedbio.2020.08.015
65. Dietrich CF, Kratzer W, Strobe D, Danse E, Fessl R, Bunk A, et al. Assessment of metastatic liver dis-
ease in patients with primary extrahepatic tumors by contrast-enhanced sonography versus CT and
MRI. World J Gastroenterol. 2006;12(11):1699–705. https://doi.org/10.3748/wjg.v12.i11.1699
66. Mahfouz AE, Hamm B, Wolf KJ. Peripheral washout: a sign of malignancy on dynamic gadolinium-
enhanced MR images of focal liver lesions. Radiology. 1994;190(1):49–52. https://doi.org/10.1148/
radiology.190.1.8259426
67. Vreugdenburg TD, Ma N, Duncan JK, Riitano D, Cameron AL, Maddern GJ. Comparative diagnos-
tic accuracy of hepatocyte-specific gadoxetic acid (Gd-EOB-DTPA) enhanced MR imaging and con-
trast enhanced CT for the detection of liver metastases: a systematic review and meta-analysis. Int J
Colorectal Dis. 2016;31(11):1739–49. https://doi.org/10.1007/s00384-016-2664-9
68. Sivesgaard K, Larsen LP, Sørensen M, Kramer S, Schlander S, Amanavicius N, et al. Diagnostic accu-
racy of CE-CT, MRI and FDG PET/CT for detecting colorectal cancer liver metastases in patients con-
sidered eligible for hepatic resection and/or local ablation. Eur Radiol. 2018;28(11):4735–47. https://
doi.org/10.1007/s00330-018-5469-0
6
Achieving a Cure: The Next Frontier
in Hepatitis B Treatment
Tina Boortalary1 • Brianna Shinn1, 2 • Dina Halegoua-DeMarzio1,2 •
Hie-Won Hann1,2
1
Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA;
2
Division of Gastroenterology and Hepatology, Department of Medicine, Thomas Jefferson
University Hospital, Philadelphia, PA, USA
Author for correspondence: Hie-Won Hann, Division of Gastroenterology and
Hepatology, Department of Medicine, Thomas Jefferson University Hospital, Philadelphia,
PA, USA. Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch6
Abstract: Since the discovery of the hepatitis B virus (HBV) by Blumberg et al. in
1965, significant progress has been made in understanding the pathogenesis of
the virus and creating an effective vaccine. In the past two decades, several a ntiviral
therapies have reduced the incidence of HBV-related hepatocellular carcinoma.
The nucleos(t)ide analogues have succeeded in decreasing the viral load to
undetectable levels but have been unable to eradicate the virus due to the persis-
tence of covalently closed circular DNA in the hepatocyte nucleus. Despite being
on successful antiviral therapy for multiple years, patients are still at risk of devel-
oping hepatocellular carcinoma. Recently, a number of different drug targets have
been identified that intervene on the viral replication cycle or host immune
system. This chapter discusses the immunopathogenesis of the virus, the
effectiveness of nucleos(t)ide analogues, and recent therapeutic developments. In
light of robust progress achieved in antiviral therapy, the cure for hepatitis B is
likely on the horizon.
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
109
110 Boortalary T et al.
INTRODUCTION
HBV- associated HCC, the hepatitis vaccine has been one of the greatest
achievements in public health.
Entry inhibitors
• Lipopeptides, RNA interference,
ed
Targeting rat Arrowhead, Tekmira,
e.g. Myrcludex-B eg NA
Int V D
• Cyclosporin derivatives cccDNA HB Alnylam, GSK
enveloped pgRNA
RC-DNA cap pA
virion
mature
core protein
RC-DNA
+ P protein
nucleo- immature
capsid RNA
Caspsid inhibitors
nucleocapsid
Novira, Janssen, Abbvie
Inhibitors of HBsAg AssemblyPHARMA, Gilead
release
Replicor plasma
membrane
Polymerase inhibitors
• Nucleoside analogues Immune modulation
• Non-nucleosides • Toll-like receptors agonists,
PD-1
Y Gilead, Roche
“exaustion” • Anti-PD-1 mAb,
“high antigen load” CD8+
BMS, Merck
T cell
B cell • Vaccine therapy:
Transgene, Gilead, Roche
Dysfunctional
Insufficient Innovio, Medimmune, ITS
T-cell response
B-cell response
Current Opinion in Virology
Figure 1. Hepatitis B replication cycle and potential drug targets. Adapted from Levrero et al.
(20). Used with permission from original copyright holder.
112 Boortalary T et al.
uncoated and the nucleocapsid is taken to the location of the host cell’s nucleus
where the rcDNA is imported through the nuclear pore into the nucleus.
The first step in replication involves the transformation of the rcDNA into a
covalently, closed, circular DNA (cccDNA) mini-chromosome by host cell repair
mechanisms (22). The cccDNA serves as the transcriptional template by utilizing
host RNA polymerase II for the production of a four viral mRNA intermediates:
a “larger-than-genome” pregenomic RNA and shorter, subgenomic transcripts.
The pregenomic RNA (pgRNA) is essential for viral replication given that it
serves as the template for DNA synthesis and for the translation of capsid pro-
teins (HBcAg) and HBV DNA polymerase. Once the viral DNA polymerase is
formed, it attaches to its own transcript forming the pgRNA-polymerase complex.
At the same time, the core particles construct a capsid around the pgRNA-
polymerase complex. Within this complex, viral DNA synthesis takes place
through reverse transcription and the rcDNA is formed. The shorter mRNA is
translated and undergoes additional processing to form proteins that become
HBV surface antigens in the viral envelope (23). In addition to forming new
virions, the capsid containing rcDNA is shuttled back to the nucleus to replenish
cccDNA. Therefore, sources for cccDNA include both the new entry of viral par-
ticles as well as the translocation of newly synthesized rcDNA from the cyto-
plasm to the nucleus (24). Even in the case of HBsAg seroconversion, the
persistence of cccDNA in the nucleus is believed to cause infectivity and poses
as an obstacle for cure (25).
HBV is not directly cytotoxic to liver cells. Liver disease is mediated through inter-
play of HBV replication and the host immune response. During the replication
process, HBsAg is taken up and degraded by antigen presenting cells and dis-
played on MHC class I and class II molecules. A T-lymphocyte mediated response
leads to apoptosis or release of cytokines that can down-regulate viral replication.
The persistence of HBV DNA activates the host immune system leading to the
chronic inflammation implicated in the development of HCC.
The connection between HBV and hepatocarcinogenesis has been firmly estab-
lished since the 1980’s. Although the molecular mechanisms of HBV carcinogen-
esis are not fully understood, it is believed to be due to a multifactorial process
involving chronic liver inflammation, chromosomal instability and disruptions of
cell signaling (26). Studies have shown that HBV DNA integration can lead to
duplications, deletions, and chromosomal translocations of important genes that
code for proteins (P53, Ras, TGF-B, Wnt/β-Catenin, cyclins A and D1) involved
in cell signaling, proliferation and apoptosis (27, 28). The viral genotype is a risk
factor for malignancy, with genotype C having a higher risk of causing HCC
(29, 30). Specific mutations in pre-core regions and open reading frames coding
PreS1/PreS2/S have been established as enhancers of the carcinogenic potential of
HBV (29). HBV protein X is thought to play an important role in regulating cel-
lular transcription, apoptosis pathways, and cellular proliferation (31).
Treatment for Hepatitis B 113
TREATMENT
The goal of HBV treatment has been to reduce viral load and halt progression of
liver disease leading to HCC. To date, these goals have been achieved by the use
of pegylated interferon (PegIFN) and nucleos(t)ide analogues (NAs) including
lamivudine, entecavir, telbivudine, adefovir, tenofovir disoproxil fumarate and
tenofovir alafenamide. The exact mechanism of PegIFN is not known, however
they are believed to act through immunomodulation and, to a lesser extent, by
exerting a weak antiviral effect by intervening in several steps of the viral replica-
tion cycle. Unlike the NAs, PegIFN has a finite course of treatment. Its use is
limited by a significant adverse effect profile, a requirement for weekly injections,
and a large portion of non-responders. Lamivudine is an oral antiviral drug and
was the first NA approved for the treatment of hepatitis B in 1998. Subsequently,
more NAs including adefovir, entecavir, telbivudine, tenofovir disoproxil fuma-
rate and tenofovir alafenamide became available. The best outcome expected of
current antivirals is the seroconversion of HBsAg, but this clearance is only
achieved in 10% of patients (32, 33).
who were treated with lamivudine. The study demonstrated that compared to an
untreated, matched control group, patients with HCC who were treated with
lamivudine achieved significant improvements in Child-Pugh scores at 24 months
and lower mortality rates from liver failure. However, there was no change in HCC
recurrence or overall survival between the two groups (40). Kuzuya et al. (41),
compared outcomes in patients with HBV-related HCC who received lamivudine
(n = 16) and those who did not (n = 33) following treatment for HCC (resection
or radiofrequency ablation). The lamivudine group had better Child-Pugh scores
at the time of HCC recurrence. Although there was no difference in rates of HCC
recurrence or survival between the groups, patients treated with lamivudine had
better remnant liver function at the time of HCC recurrence and were more likely
to receive curative treatment (41). Kubo et. al. demonstrated that patients treated
with lamivudine after curative resection for HBV-related HCC were more likely to
have improved tumor-free survival compared to those who did not receive lami-
vudine treatment (43). Prolonged disease-free survival and overall survival rates
were reported in another study in which patients received antiviral treatment after
surgery for HBV-related HCC (44). In a large Taiwanese cohort, patients receiving
NAs after liver resection were found to have lower risk of HCC recurrence and
improved survival rates (42). In a two-stage longitudinal study that included a
randomized clinical trial, Yin et al. also demonstrated that patients receiving NAs
after surgery for HBV-related HCC had significantly decreased risk of HCC recur-
rence and HCC-related deaths (47). In studies from the US, Hann et al. demon-
strated an increased median survival time of 80 months in patients receiving NAs
for HBV-related HCC (after local tumor ablation) compared to a median survival
time of 16 months in those without NA treatment (45, 46). Many of these studies
were pooled in a meta-analysis performed by Yuan et al. and showed that the one
and three-year recurrence rates of HCC and disease-free survival were signifi-
cantly better in patients treated with NAs after curative treatment (liver transplant,
hepatectomy, radiofrequency ablation) for HBV-related HCC (48). More recently,
a study with 487 subjects demonstrated that HCC recurrence and HCC-related
death were significantly decreased in patients who received antiviral treatment
after hepatectomy for HCC compared to those who did not (p = 0.28, p = 0.004,
respectively) (49).
133 patients with HBV who achieved undetectable viral levels developed HCC.
They identified cirrhosis as an associated risk factor (53). It is important to recog-
nize that “undetectable” is a relative term given the variation in measurements
conducted by different laboratory assays. At the Liver Disease Prevention Center,
Division of Gastroenterology and Hepatology of Thomas Jefferson University
Hospital, long-term follow-up data has demonstrated HCC recurrence in patients
even after a decade of successful viral suppression (54, 55) In our observational
cohort, as of October 2020, 17 patients developed HCC despite having been
treated for 9–19 years (median 13 years) with undetectable HBV DNA for 3–12
years (median 8 years) (Table 1). This is among the longest known follow up stud-
ies on the development of HCC in patients taking antiviral therapy. Recurrence of
HCC has also been noted after the treatment of the initial tumor despite successful
viral suppression. We have observed six patients who, after first HCC treatment,
started antiviral therapy which led to successful HBV suppression for years
(5–11 years) and yet developed subsequent new or recurrent HCC (Figure 2).
Li et al. compared patients who received NA treatment (lamivudine with or with-
out adefovir, n = 43) after hepatectomy for advanced HCC and those who did not
receive NAs (n = 36) (56). There was no difference in the recurrence rate of HCC
or disease-free survival between treatment group and control. Kuzuya et al. also
found that the recurrence rates of HCC were not decreased after the administra-
tion of nucleotide analogues, but more favorable Child-Pugh scores were observed
at the time of HCC reoccurrence (41).
The currently available NAs target the reverse transcription of HBV RNA and have
been proven to delay progression to cirrhosis. However, despite their success in
halting inflammation in the liver, the risk of HCC has persisted. As demonstrated
in Figure 1, NAs suppress HBV replication at one point in the cycle, but do not
eliminate the cccDNA, which remains in the hepatocytes. There are currently a
number of clinical trials, summarized in Table 2, that strategize different
approaches to eradicating HBV (57). Each drug targets a specific point of the viral
replication cycle or acts indirectly on the host immune functions. Targets can
interfere with replication through the following mechanisms: (i) inhibition of viral
entry; (ii) blocking rcDNA entry into the hepatic nucleus; (iii) prevention of
cccDNA formation; (iv) prevention of mRNA transcription; (v) inhibition of cap-
sid formation; (vi) inhibition of reverse transcription; and (vii) inhibition of HBV
release into circulation.
Entry inhibitors
Blocking HBV entry into the hepatocyte can prevent the earliest step of infection.
This is the target of Bulevirtide or formally known as Myrcludex B. The drug
works by inhibiting attachment of viral pre-S1 protein to NTCP, therefore block-
ing the mechanism for entry (58). It has been approved by the European
Commission as the first drug effective for hepatitis D and hepatitis B co-infection.
116
TABLE 1 Development of HCC in patients with cirrhosis on long-term antiviral therapy
B→A
2 1/1998 B→A 3/2008 10 8 68 2.8 Rt UD LAM + TDF Dead
3 5/1998 A→A 2/2008 10 7 76 1.8 Lt UD LAM + TDF Alive
4 7/2001 B→B 9/2010 9 4 54 2.8 Rt UD LAM + TDF Dead
5 8/2004 B→B 11/2010 16 4 53 3.9 Rt UD LAM + TDF Alive
6 7/2001 B→B 1/2011 10 5 55 2.8 Rt UD LAM + TDF Dead
7 2/2004 A→A 6/2013 9 8 57 2.5 Lt med UD TDF Dead
8 2/1996 A→A 7/2013 17 10 73 1.6 Rt UD TDF Dead
9 8/1997 A→A 6/2014 17 6 54 2.2 Lt lat UD ETV Alive
10 3/2004 B→B 6/2013 9 7 57 2.5 Lt UD TDF Dead
11 7/2001 A→A 6/2014 13 7 54 2.2 Lt UD TDF Alive
12 5/1996 A→A 10/2014 18 10 74 3.4 Rt UD LAM + TDF Dead
13 2/2000 A→A 10/2014 14 12 62 3.8 Rt UD ETV + TDF Alive
14 2/2000 A→A 4/2015 15 12 62 3.4 Rt UD TDF Alive
15 2/2000 B→A 5/2015 15 12 65 3.8 Rt UD TDF Alive
16 12/1998 A→A 8/2017 19 8 64 2.0 Rt UD LAM + TDF Alive
17 2/2008 A→A 6/2019 11 10 57 2.2 Rt UD ETV + TDF Alive
Dx: Diagnosis, ETV: Entecavir, LAM: Lamivudine, Lt: Left, Pt: Patient, Rt: Right, TDF: Tenofovir Disoproxil Fumarate, Tx: Treatment, UD: Undetectable,
*Years with HBV DNA (-) at diagnosis of HCC
Treatment for Hepatitis B 117
70 3 years Patient 1
5 years Patient 2
65
10 years Patient 3
60 7 years Patient 4
Patient 5
55 5 years Patient 6
50
45
First HCC Second HCC Third HCC
Event
Figure 2. Development of subsequent new and recurrent HCC while on successful antiviral
treatment. Graph of 6 patients demonstrates long duration time between initial and
subsequent HCC diagnoses.
Phase 2 data has shown that there is a reduction of viral HDV RNA after b
ulevirtide
when combined with tenofovir or peg-interferon-α (59, 60).
Capsid inhibitors
HBV core protein is necessary for capsid formation, genome packaging and deliv-
ery of rcDNA into the nucleus. Therefore, core inhibitors or core modulators dis-
rupt the viral lifecycle by inducing the assembly of defective capsids or capsids
that lack genetic material. By this mechanism, they are thought to intervene on
multiple points of the lifecycle, even targeting cccDNA by causing its destabiliza-
tion and preventing its replenishment. There are several different classes of
core protein assembly modulators (CpAMs) including phenylpropenamide which
have normal morphologies but lack genetic material (empty capsids) and
hetero-aryl-dihydropyrimidines (HAP) derivatives, which have aberrant mor-
phologies (67). The first compound studied in humans in phase 1 b study was
NVR3-778. It showed a 1–2 log reduction in HBV DNA levels but no change in
HBsAg levels until the drug was combined with peg-IFN (68).
Core inhibitors are the first viral specific compounds that are capable of target-
ing cccDNA. A recent phase 2 study (69) demonstrated that ABI-H0731, a core
protein inhibitor, not only interfered with viral capsid formation and entry of
rcDNA into the nucleus, but also decreased levels of pgRNA. In that study, the
combination of the ABI-H0731 and entecavir showed greater reductions of viral
DNA load and pgRNA levels by week 24 in treatment naïve, HBeAg+, individuals
compared to entecavir alone (p = 0.0452). When patients were switched from
entecavir to the combination of both, they saw immediate decline in HBV DNA
and pgRNA levels (Figure 3). By week 48, the mean HBV DNA and pgRNA levels
declined 6.3 logs and 3.0 logs from baseline, respectively. The accelerated decline
after the addition of ABI-H0731 was attributed to the reductions in the cccDNA
pool, the only known source of HBV pgRNA (70).
–1 ETV+731 ETV+731
–1
ETV to Combo
Baseline (log10 IU/mL)
Figure 3. Study 202 and 211 results of Entecavir alone, Entecavir+ABI-H0731 (Core Protein
Inhibitor), and switch from entecavir to combo on HBV DNA levels and pgRNA levels. Adapted
from Sulkowski et al. (70). Used with permission. The combination of the ABI-H0731 and
entecavir showed greater reductions of viral DNA load and pgRNA levels by week 24 in
treatment naïve, HBeAg+, individuals compared to entecavir alone (p = 0.0452). When
patients were switched from entecavir to the combination of both, they saw immediate
decline in HBV DNA and pgRNA levels.
HBsAg inhibitors
HBsAg release inhibitors interfere with the production of HBsAg, which is
needed for the virus to enter and leave the liver cell. Most of viral antigen in the
blood is HBsAg and circulating levels are almost entirely made of HBV subviral
particles (SVP). These small particles are produced independently of viral repli-
cation and are non-infectious. However, it has been suggested that SVPs can
neutralize antibodies to the virus and suppress other aspects of host immune
function (75, 76). The nucleic acid polymer (NAP), REP 2139, is a synthetic
RNA that is taken up by hepatocytes and suppresses the production of SVPs
derived from integrated HBV DNA or cccDNA. A phase 2 randomized control
trial showed that the addition of NAPs to TDF and pegIFN allowed significantly
more patients to achieve HBsAg levels below 1 IU/mL and HBsAg seroconver-
sion (77). Another method by which HBsAg is inhibited is through interruption
in the intracellular re-localization of the protein to the Golgi apparatus. This is
the mechanism utilized by Benzimidazole, BM601, which decreases HBsAg and
HBV release (78).
cccDNA inhibitors
Directly inhibiting cccDNA would be the optimal HBV therapy, but aside from
the capsid inhibitor (ABI-H0731) that has shown promising results, no drugs
have shown to target cccDNA in clinical trials (79). Potential mechanisms to tar-
get cccDNA may involve inhibiting its formation, transfer into the nucleus, con-
version to cccDNA, or transcription after cccDNA is produced. Several
gene-editing technologies utilize site-specific nucleases to cleave HBV DNA. The
zinc finger nucleases (ZFNs), transcription activator-like effector nucleases
120 Boortalary T et al.
CONCLUSION
Copyright and permission statement: The authors confirm that the materi-
als included in this chapter do not violate copyright laws. Where relevant,
appropriate permissions have been obtained from the original copyright
holder(s), and all original sources have been appropriately acknowledged or
referenced.
Treatment for Hepatitis B 121
REFERENCES
1. Tsai K-N, Kuo C-F, Ou J-HJ. Mechanisms of hepatitis B virus persistence. Trends Microbiol.
2018;26(1):33–42. https://doi.org/10.1016/j.tim.2017.07.006
2. Blumberg BS, Alter HJ. A new antigen in leukemia sera. Jama. 1965;191(7):541–6. https://doi.
org/10.1001/jama.1965.03080070025007
3. Blumberg BS. Australia antigen and the biology of hepatitis B. Science. 1977;197(4298):17–25.
https://doi.org/10.1126/science.325649
4. Dane DS, Cameron CH, Briggs M. Virus-like particles in serum of patients with Australia-antigen-
associated hepatitis. Lancet. 1970;1(7649):695–8. https://doi.org/10.1016/S0140-6736(70)90926-8
5. Yuen MF, Chen DS, Dusheiko GM, Janssen HLA, Lau DTY, Locarnini SA, et al. Hepatitis B virus infec-
tion. Nat Rev Dis Primers. 2018;4:18035. https://doi.org/10.1038/nrdp.2018.35
6. Magnius LO, Espmark JÅ. New specificities in Australia antigen positive sera distinct from the
Le Bouvier determinants. J Immunol. 1972;109(5):1017–21.
7. Szmuness W, Stevens CE, Harley EJ, Zang EA, Oleszko WR, William DC, et al. Hepatitis B vaccine:
demonstration of efficacy in a controlled clinical trial in a high-risk population in the United States.
NEJM. 1980;303(15):833–41. https://doi.org/10.1056/NEJM198010093031501
8. Valenzuela P, Medina A, Rutter WJ, Ammerer G, Hall BD. Synthesis and assembly of hepatitis B virus
surface antigen particles in yeast. Nature. 1982;298(5872):347–50. https://doi.org/10.1038/298347a0
9. Chen D-S. Hepatitis B vaccination: the key towards elimination and eradication of hepatitis B.
J Hepatol. 2009;50(4):805–16. https://doi.org/10.1016/j.jhep.2009.01.002
10. Ni YH, Huang LM, Chang MH, Yen CJ, Lu CY, You SL, et al. Two decades of universal
hepatitis B v accination in Taiwan: impact and implication for future strategies. Gastroenterology.
2007;132(4):1287–93. https://doi.org/10.1053/j.gastro.2007.02.055
11. Goh KT, Oon C, Heng B, Lim G. Long-term immunogenicity and efficacy of a reduced dose of
plasma-based hepatitis B vaccine in young adults. Bulletin of the World Health Organization.
1995;73(4):523–7.
12. Wainwright RB, Bulkow LR, Parkinson AJ, Zanis C, McMahon BJ. Protection provided by hepatitis
B vaccine in a Yupik Eskimo population-results of a 10-year study. J Infect Dis. 1997;175(3):674–7.
https://doi.org/10.1093/infdis/175.3.674
13. Gold Y, Somech R, Mandel D, Peled Y, Reif S. Decreased immune response to hepatitis B eight
years after routine vaccination in Israel. Acta Paediatrica. 2003;92(10):1158–62. https://doi.
org/10.1111/j.1651–2227.2003.tb02477.x
14. Mahawal B, Bhai N, Kataria V, Gulati N, Chandola I. Estimation of Anti Hbs antibody titer in adults
during 5–10 years period following three doses of vaccine. IOSR J Pharm Biol Sci. 2013;7(1):20–3.
https://doi.org/10.9790/3008-0712023
15. Poovorawan Y, Chongsrisawat V, Theamboonlers A, Bock HL, Leyssen M, Jacquet J-M. Persistence of
antibodies and immune memory to hepatitis B vaccine 20 years after infant vaccination in Thailand.
Vaccine. 2010;28(3):730–6. https://doi.org/10.1016/j.vaccine.2009.10.074
16. Mendy M, Peterson I, Hossin S, Peto T, Jobarteh ML, Jeng-Barry A, et al. Observational study of vac-
cine efficacy 24 years after the start of hepatitis B vaccination in two Gambian villages: no need for a
booster dose. Plos One. 2013;8(3):e58029. https://doi.org/10.1371/journal.pone.0058029
17. Shepard CW, Simard EP, Finelli L, Fiore AE, Bell BP. Hepatitis B virus infection: epidemiology and
vaccination. Epidemiol Rev. 2006;28(1):112–25. https://doi.org/10.1093/epirev/mxj009
18. Kato H, Sugiyama M, Mizokami M. Hepatitis B Virus Genotypes. In: Liaw Y-F, Zoulim F, editors.
Hepatitis B Virus in Human Diseases. Cham: Springer International Publishing; 2016. p. 63–78.
https://doi.org/10.1007/978-3-319-22330-8_3
19. Zeisel MB, Lucifora J, Mason WS, Sureau C, Beck J, Levrero M, et al. Towards an HBV cure: state-of-the-
art and unresolved questions-report of the ANRS workshop on HBV cure. Gut. 2015;64(8):1314–26.
https://doi.org/10.1136/gutjnl-2014-308943
20. Levrero M, Testoni B, Zoulim F. HBV cure: why, how, when? Curr Opin Virol. 2016;18:135–43.
https://doi.org/10.1016/j.coviro.2016.06.003
122 Boortalary T et al.
21. Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z, et al. Sodium taurocholate cotransporting polypep-
tide is a functional receptor for human hepatitis B and D virus. Elife. 2012;1:e00049. https://doi.
org/10.7554/eLife.00049
22. Wong DJ, Locarnini SA. Molecular Virology and Life Cycle. In: Kao J-H, Chen D-S, editors.
Hepatitis B Virus and Liver Disease. Singapore: Springer Singapore; 2018. p. 1–23. https://doi.
org/10.1007/978-981-10-4843-2_1
23. Trépo C, Chan HL, Lok A. Hepatitis B virus infection. Lancet. 2014;384(9959):2053–63. https://doi.
org/10.1016/S0140-6736(14)60220-8
24. Mahtab M-A. Liver: A Complete Book on Hepato-Pancreato-Biliary Diseases-E-Book: Elsevier Health
Sciences; 2012.
25. Morikawa K, Suda G, Sakamoto N. Viral life cycle of hepatitis B virus: Host factors and druggable
targets. Hepatol Res. 2016;46(9):871–7. https://doi.org/10.1111/hepr.12650
26. Fernández-Rodríguez CM, Gutiérrez-García ML. Prevention of hepatocellular carcinoma in patients
with chronic hepatitis B. World J Gastrointest Pharmacol Ther. 2014;5(3):175–82. https://doi.
org/10.4292/wjgpt.v5.i3.175
27. Boyault S, Rickman DS, de Reynies A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome
classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology.
2007;45(1):42–52. https://doi.org/10.1002/hep.21467
28. Tan YJ. Hepatitis B virus infection and the risk of hepatocellular carcinoma. World J Gastroenterol.
2011;17(44):4853–7. https://doi.org/10.3748/wjg.v17.i44.4853
29. Yang HI, Yeh SH, Chen PJ, Iloeje UH, Jen CL, Su J, et al. Associations between hepatitis B virus geno-
type and mutants and the risk of hepatocellular carcinoma. J Natl Cancer Inst. 2008;100(16):1134–43.
https://doi.org/10.1093/jnci/djn243
30. Chan HL, Hui AY, Wong ML, Tse AM, Hung LC, Wong VW, et al. Genotype C hepatitis B virus
infection is associated with an increased risk of hepatocellular carcinoma. Gut. 2004;53(10):1494–8.
https://doi.org/10.1136/gut.2003.033324
31. Bouchard MJ, Schneider RJ. The enigmatic X gene of hepatitis B virus. J Virol. 2004;78(23):12725–34.
https://doi.org/10.1128/JVI.78.23.12725-12734.2004
32. Gish RG, Given BD, Lai C-L, Locarnini SA, Lau JY, Lewis DL, et al. Chronic hepatitis B: virology, natu-
ral history, current management and a glimpse at future opportunities. Antiviral Res. 2015;121:47–58.
https://doi.org/10.1016/j.antiviral.2015.06.008
33. Zoulim F, Durantel D. Antiviral therapies and prospects for a cure of chronic hepatitis B. Cold Spring
Harb Perspect Med 5: a021501. 2015. https://doi.org/10.1101/cshperspect.a021501
34. Liaw Y-F, Sung JJ, Chow WC, Farrell G, Lee C-Z, Yuen H, et al. Lamivudine for patients with
chronic hepatitis B and advanced liver disease. N Engl J Med. 2004;351(15):1521–31. https://doi.
org/10.1056/NEJMoa033364
35. Hosaka T, Suzuki F, Kobayashi M, Seko Y, Kawamura Y, Sezaki H, et al. Long-term entecavir treatment
reduces hepatocellular carcinoma incidence in patients with hepatitis B virus infection. Hepatology.
2013;58(1):98–107. https://doi.org/10.1002/hep.26180
36. Kim W, Berg T, Loomba R, Schall RA, Dinh P, Yee L, et al. 43 long term tenofovir disoproxil fuma-
rate (tdf) therapy and the risk of hepatocellular carcinoma. J Hepatol. 2013;58:S19. https://doi.
org/10.1016/S0168-8278(13)60045-8
37. Wu JC, Huang YH, Chau GY, Su CW, Lai CR, Lee PC, et al. Risk factors for early and late recurrence in
hepatitis B-related hepatocellular carcinoma. J Hepatol. 2009;51(5):890–7. https://doi.org/10.1016/j.
jhep.2009.07.009
38. Hung IF, Poon RT, Lai CL, Fung J, Fan ST, Yuen MF. Recurrence of hepatitis B-related hepatocel-
lular carcinoma is associated with high viral load at the time of resection. Am J Gastroenterol.
2008;103(7):1663–73. https://doi.org/10.1111/j.1572-0241.2008.01872.x
39. Kim BK, Park JY, Kim DY, Kim JK, Kim KS, Choi JS, et al. Persistent hepatitis B viral replication affects
recurrence of hepatocellular carcinoma after curative resection. Liver Int. 2008;28(3):393–401.
https://doi.org/10.1111/j.1478-3231.2007.01625.x
40. Piao CY, Fujioka S, Iwasaki Y, Fujio K, Kaneyoshi T, Araki Y, et al. Lamivudine treatment in patients
with HBV-related hepatocellular carcinoma--using an untreated, matched control cohort. Acta Med
Okayama. 2005;59(5):217–24.
Treatment for Hepatitis B 123
41. Kuzuya T, Katano Y, Kumada T, Toyoda H, Nakano I, Hirooka Y, et al. Efficacy of antiviral ther-
apy with lamivudine after initial treatment for hepatitis B virus-related hepatocellular carcinoma.
J Gastroenterol Hepatol. 2007;22(11):1929–35. https://doi.org/10.1111/j.1440-1746.2006.04707.x
42. Wu CY, Chen YJ, Ho HJ, Hsu YC, Kuo KN, Wu MS, et al. Association between nucleoside analogues
and risk of hepatitis B virus-related hepatocellular carcinoma recurrence following liver resection.
JAMA. 2012;308(18):1906–14. https://doi.org/10.1001/2012.jama.11975
43. Kubo S, Tanaka H, Takemura S, Yamamoto S, Hai S, Ichikawa T, et al. Effects of lamivudine on out-
come after liver resection for hepatocellular carcinoma in patients with active replication of hepatitis
B virus. Hepatol Res. 2007;37(2):94–100. https://doi.org/10.1111/j.1872-034X.2007.00013.x
44. Chan AC, Chok KS, Yuen WK, Chan SC, Poon RT, Lo CM, et al. Impact of antiviral therapy on the
survival of patients after major hepatectomy for hepatitis B virus-related hepatocellular carcinoma.
Arch Surg. 2011;146(6):675–81. https://doi.org/10.1001/archsurg.2011.125
45. Hann HW, Bergin D, Coben R, DiMarino AJ. Prevention of new hepatocellular carcinoma with con-
comitant antiviral therapy in chronic hepatitis B patients whose initial tumor was successfully ablated.
Int J Cancer. 2011;128(3):739–42. https://doi.org/10.1002/ijc.25382
46. Hann HW, Coben R, Brown D, Needleman L, Rosato E, Min A, et al. A long-term study of the
effects of antiviral therapy on survival of patients with HBV-associated hepatocellular carcinoma
(HCC) following local tumor ablation. Cancer Med. 2014;3(2):390–6. https://doi.org/10.1002/
cam4.197
47. Yin J, Li N, Han Y, Xue J, Deng Y, Shi J, et al. Effect of antiviral treatment with nucleotide/nucleo-
side analogs on postoperative prognosis of hepatitis B virus-related hepatocellular carcinoma: a
two-stage longitudinal clinical study. J Clin Oncol. 2013;31(29):3647–55. https://doi.org/10.1200/
JCO.2012.48.5896
48. Yuan P, Chen P, Qian Y. Evaluation of Antiviral Therapy Performed after Curative Therapy in Patients
with HBV-Related Hepatocellular Carcinoma: An Updated Meta-Analysis. Can J Gastroenterol
Hepatol. 2016;2016:5234969. https://doi.org/10.1155/2016/5234969
49. Zhang X, Li C, Wen T, Yan L, Yang J, Tang H, et al. The Different Effects of Nucleotide and Nucleoside
Analogues on the Prognosis of HBV-Related HCC After Curative Resection. J Gastrointest Surg. 2020.
https://doi.org/10.1007/s11605-020-04633-3
50. Arends P, Sonneveld MJ, Zoutendijk R, Carey I, Brown A, Fasano M, et al. Entecavir treatment does
not eliminate the risk of hepatocellular carcinoma in chronic hepatitis B: limited role for risk scores in
Caucasians. Gut. 2015;64(8):1289–95. https://doi.org/10.1136/gutjnl-2014-307023
51. Papatheodoridis GV, Manolakopoulos S, Touloumi G, Vourli G, Raptopoulou-Gigi M, Vafiadis-
Zoumbouli I, et al. Virological suppression does not prevent the development of hepatocellular
carcinoma in HBeAg-negative chronic hepatitis B patients with cirrhosis receiving oral antiviral(s)
starting with lamivudine monotherapy: results of the nationwide HEPNET. Greece cohort study. Gut.
2011;60(8):1109–16. https://doi.org/10.1136/gut.2010.221846
52. Yoo J, Hann HW, Coben R, Conn M, DiMarino AJ. Update Treatment for HBV Infection and Persistent
Risk for Hepatocellular Carcinoma: Prospect for an HBV Cure. Diseases. 2018;6(2).27 https://doi.
org/10.3390/diseases6020027
53. Ando Y, Ishigami M, Ishizu Y, Kuzuya T, Honda T, Hayashi K, et al. Cumulative incidence and
risk factors for the development of hepatocellular carcinoma in patients with chronic hepatitis B
who achieved sustained disappearance of viremia by nucleos(t)ide analog treatment. Hepatol Res.
2018;48(3):E240-E51. https://doi.org/10.1111/hepr.12976
54. Shinn BJ, Kristler C, Roth C, Hann HW. Need For HBV Cure: Persistent Risk For Subsequent New And
Recurrent HCC Even After A Decade Of Successful Anti-HBV Therapy And Initial Tumor Ablation.
Arch Cancer Res. 2018;6:1–6. https://doi.org/10.21767/2254-6081.100172
55. Shinn BJ, Martin A, Coben RM, Conn MI, Prieto J, Kroop H, et al. Persistent risk for new, subsequent
new and recurrent hepatocellular carcinoma despite successful anti-hepatitis B virus therapy and
tumor ablation: The need for hepatitis B virus cure. World J Hepatol. 2019;11(1):65–73. https://doi.
org/10.4254/wjh.v11.i1.65
56. Li N, Lai EC, Shi J, Guo W-X, Xue J, Huang B, et al. A comparative study of antiviral therapy after
resection of hepatocellular carcinoma in the immune-active phase of hepatitis B virus infection. Ann
Surg Oncol. 2010;17(1):179–85. https://doi.org/10.1245/s10434-009-0694-z
124 Boortalary T et al.
76. Rydell GE, Prakash K, Norder H, Lindh M. Hepatitis B surface antigen on subviral particles
reduces the neutralizing effect of anti-HBs antibodies on hepatitis B viral particles in vitro. Virology.
2017;509:67–70. https://doi.org/10.1016/j.virol.2017.05.017
77. Bazinet M, Pântea V, Placinta G, Moscalu I, Cebotarescu V, Cojuhari L, et al. Safety and efficacy of
48 weeks REP 2139 or REP 2165, tenofovir disoproxil, and pegylated interferon alfa-2a in patients
with chronic HBV infection naive to nucleos(t)ide therapy. Gastroenterology. 2020;158(8):2180–94
https://doi.org/10.1053/j.gastro.2020.02.058
78. Xu Y-B, Yang L, Wang G-F, Tong X-K, Wang Y-J, Yu Y, et al. Benzimidazole derivative, BM601, a
novel inhibitor of hepatitis B virus and HBsAg secretion. Antiviral Res. 2014;107:6–15. https://doi.
org/10.1016/j.antiviral.2014.04.002
79. Yang Y-C, Chen Y-H, Kao J-H, Ching C, Liu I-J, Wang C-C, et al. Permanent inactivation of HBV
genomes by CRISPR/Cas9-mediated non-cleavage base editing. Mol Ther Nucleic Acids. 2020;
20:480–90 https://doi.org/10.1016/j.omtn.2020.03.005
80. Zhu A, Liao X, Li S, Zhao H, Chen L, Xu M, et al. HBV cccDNA and its potential as a therapeutic
target. J Clin Transl Hepatol. 2019;7(3):258–62. https://doi.org/10.14218/JCTH.2018.00054
81. Ma Z, Cao Q, Xiong Y, Zhang E, Lu M. Interaction between Hepatitis B Virus and Toll-Like Receptors:
Current Status and Potential Therapeutic Use for Chronic Hepatitis B. Vaccines (Basel). 2018;6(1).6.
https://doi.org/10.3390/vaccines6010006
82. Feray C, Lopez-Labrador FX. Is PD-1 blockade a potential therapy for HBV? JHEP Rep.
2019;1(3):142–4. https://doi.org/10.1016/j.jhepr.2019.07.007
83. Liu H, Hou J, Zhang X. Targeting cIAPs, a New Option for Functional Cure of Chronic Hepatitis B
Infection? Virologica Sinica. 2018;33(5):459–61. https://doi.org/10.1007/s12250-018-0062-x
7
Locoregional Therapies for Bridging
and Downstaging Hepatocellular
Carcinoma Prior to Liver Transplant
Sandeep A. Ponniah • Andreas G. Zori • Roniel Cabrera
Division of Gastroenterology, Hepatology, and Nutrition, University of Florida,
Gainesville, FL, USA
Author for correspondence: Andreas G. Zori, Division of Gastroenterology,
Hepatology, and Nutrition, University of Florida, Gainesville, FL, USA.
Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch7
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
127
128 Ponniah S A et al.
INTRODUCTION
Therapies for hepatocellular carcinoma (HCC) have evolved over the last several
decades, but liver transplantation remains the only curative option in patients
who are not candidates for resection. Liver transplantation allows for the cure of
both the tumor and any underlying chronic liver disease. However, more than
70% of patients present with advanced disease that does not meet the criteria for
transplantation (1). The first and most widely accepted criteria for liver transplan-
tation in patients with HCC are the “Milan criteria”, developed by Mazzaferro
et al. (2). The Milan criteria include stage T1 (1 lesion <2 cm) and stage T2
(1 lesion 2–5 cm or up to 3 lesions ≤3 cm), tumors without vascular invasion,
lymph node involvement, or extrahepatic metastases (3). When these criteria are
met, four-year survival after transplant is >80% with recurrence rates below 15%
(2). In the United States, the United Network for Organ Sharing (UNOS) affords
patients who meet stage T2 disease Model for End-Stage Liver Disease (MELD)
“exception points” due to poor survival associated with HCC that is not accounted
for in standard MELD scoring system. Although the amount of HCC exception
points has varied over time and by country, the current standard exception criteria
adopted by UNOS assigns points equal to the mean MELD at transplant minus 3
of all liver transplants recipients (except status 1A/1B, living donor, donation after
cardiac death donor and donors more than 500 miles from recipient hospital) in
the last 180 days within 250 nautical miles of the listing center. Patients who do
not meet the Milan criteria (greater than stage T2) can be eligible for liver trans-
plantation if they receive locoregional therapies that reduce their tumor burden
and maintain it within Milan criteria for 6 months. Lesions that are eligible for
downstaging beyond Milan criteria are discussed in the following sections.
A growing body of literature supports that the Milan criteria can be safely expanded
to allow patients with more advanced disease access to liver transplantation, and
under the right circumstances, they have comparable outcomes to patients that
meet the Milan criteria. As such, modifications to the Milan criteria have been col-
lectively termed “expanded criteria.” In 2001, Yao et al. (4) from the University of
California, San Francisco (UCSF), published a study with expanded criteria
termed the “UCSF criteria”: 1 lesion ≤6.5 cm or up to 3 lesions with the largest
lesion ≤4.5 cm, with a total diameter of 8 cm (Table 1). In this study, patients had
1- and 5-year survival rates that were 90% and 75.2% respectively. In 2007, this
Locoregional Therapy Prior to Liver Transplant 129
Milan criteria
• Most common eligibility criteria for LT among patients with HCC
• Single lesion ≤5 cm, or up to 3 lesions each ≤3 cm
• No evidence of extra-hepatic metastases or vascular invasion
Expanded criteria
• UCSF: Single lesion ≤6.5 cm or ≤3 lesions with the largest being ≤4.5 cm and a total diameter
≤8 cm
• Up-to-7: 7 cm as the sum of the size of the largest tumor and the number of tumors. No vascular
invasion.
• Toronto criteria: Any tumor size or number. All lesions require a liver biopsy and must NOT
show poor differentiation. No extra-hepatic metastasis, venous/biliary thrombosis OR cancer
related symptoms.
same group from UCSF published a prospective study that showed a 5 year sur-
vival rate of 81%, which was similar to Milan criteria but with the added benefit
of being able to transplant an additional 5–20% of patients initially not included
by Milan criteria (5). Several studies have validated UCSF criteria having similar
rates of survival compared to the Milan criteria (6).
In 2009, Mazzaferro et al. (7) described the Up-to-Seven criteria, where the
sum of the total number of lesions and size in centimeters (cm) of the largest
lesion can be up to 7 if vascular invasion and metastases are absent. In the initial
study of over 1,500 patients, 5-year survival rates in patients that met Up-to-
Seven criteria were similar (71.2%) to those that met the Milan criteria (73.3%) (7).
This lead to the creation of the “Metro Ticket Calculator”, which provides 3- and
5-year overall survival probabilities based on characteristics of HCC lesions (8).
In 2016, the Toronto criteria was proposed, which do not include size or tumor
number restrictions, but based candidacy on absence of extrahepatic disease,
venous thrombi, cancer-related symptoms, and high-risk features on biopsy. In
validating this criteria, patients transplanted outside MC (with the majority being
beyond UCSF criteria but within Toronto criteria), had 5-year survival rates that
were not statistically different to those within MC (68% vs 78%) (9). Other trans-
plant centers outside the USA and Canada such as in Italy, England, Japan,
Australia, New Zealand, and China have similar “expanded” criteria, and have
shown comparable outcomes to the Milan criteria. Although individual centers
may elect to transplant patients outside of Milan criteria, in the United States,
tumor burden must be brought within the Milan criteria with locoregional ther-
apy to qualify for standard MELD points, which is discussed in more detail in the
downstaging section.
130 Ponniah S A et al.
Bridging therapy
Once listed for transplant, HCC patients may experience long waiting times and
growth of tumor beyond the Milan criteria, putting them at risk for dropout from
the waiting list. Waiting times often exceed 1 year and can be as long as 2 years
(10). Dropout rates while awaiting liver transplantation have been noted to be
25% at 6 months, 38% at 12 months and up to 55.1% at 18 months (11). Bridging
therapy is locoregional therapy administered to patients on the liver transplanta-
tion waitlist to prevent tumor progression and decrease dropout rates, acting as a
“bridge” until a suitable donor is obtained. Use of bridging therapy has steadily
increased and is now used for most patients. Patients derive more benefit from
locoregional therapy as the expected liver transplant list increases (12).
Recommendations from the 2010 International Consensus Conference and EASL/
EORTC Clinical Practice Guidelines suggest bridging therapy for all patients with
HCC within the Milan criteria, with wait times >6 months for liver transplantation
(13). In those meeting criteria for the Milan criteria, bridging therapy has decreased
waitlist dropout to 0–10% (12). Risk factors responsible for an increased risk of
dropout include tumors >3 cm or multifocal disease, serum AFP >200 ng/ml,
waitlist time >6 months, and poor response to bridging therapy (12). The data on
survival benefit is discussed later in this chapter.
Downstaging
“Downstaging” involves treating tumors outside of the Milan criteria with locore-
gional therapy to decrease tumor burden, allowing for application of standard
MELD exceptions and ultimately liver transplantation. UCSF first proposed a
downstaging protocol in 2005 where lesions outside the Milan criteria were eligible
for downstaging. Lesions included were: 1 lesion >5 cm and ≤8 cm, 2–3 lesions
each ≤5 cm or 4–5 lesions each ≤3 cm with a total diameter ≤8 cm. In a study from
this group between 2002 and 2012 (14), survival at 5 years post liver transplanta-
tion in patients outside the Milan criteria but who were successfully down staged
was identical to the Milan criteria control group (78%). Similarly in 2018, in a more
recent multi-center study from this same group, 5-year post liver transplant sur-
vival in down staged patients was excellent at 80% (14). Based on these results,
UNOS adopted the UCSF inclusion criteria for downstaging (UNOS-DS) above and
in 2019 finalized the policy verbiage to clearly outline this criterion. Consequently,
patients that are initially outside the Milan criteria but meet UNOS-DS receive auto-
matic approval for MELD exception if they remain within the Milan criteria after
locoregional therapy. Candidates that are outside UNOS-DS who receive downstag-
ing locoregional therapies to the Milan criteria must be referred to the National
Liver Review Board (NLRB) for consideration for MELD exception.
The options for locoregional therapies in downstaging are discussed in detail
below. The decision on optimal locoregional therapy is contingent on multiple
factors, but the Barcelona Clinic Liver Cancer (BCLC) staging system is often used
to help make this decision (8). BCLC staging factors tumor burden, liver function
and patient performance status to stratify patients by risk. Downstaging patients
often have advanced stage disease and based on BCLS, trans-arterial chemoembo-
lization (TACE) is most utilized followed by trans-arterial radioembolization and
ablation (15).
Locoregional Therapy Prior to Liver Transplant 131
Tumor biology
Over time, the focus of liver transplantation guidelines has shifted beyond tumor
size and number to tumor biology and behavior (16). As a result, UNOS currently
requires a 6-month waiting period prior to granting MELD exception points. This
allows for observation of the tumor to ensure that it does not have aggressive biol-
ogy which rapidly progresses to metastasis and can significantly increase the
chances of post-transplant recurrence. Downstaging is similarly helpful in assess-
ing tumor biology, in that tumors which cannot be brought within transplant
criteria with locoregional therapy confer a poor prognosis. Conversely, tumors
outside of the Milan criteria that respond to downstaging protocols have been
noted with favorable histological changes, including a lack of microvascular inva-
sion, low tumor grading, and lack of satellite lesions (17) which are similarly seen
in patients within the Milan criteria. While histological data, such as degree of
differentiation and microvascular invasion, is helpful in predicting tumor behav-
ior and stratifying risk, it is often not available in pre-transplant settings since in
most cases biopsy is not necessary to make the diagnosis of HCC. Therefore, there
is significant interest in identifying serum markers which can classify tumor biol-
ogy non-invasively.
Serum alpha-fetoprotein (AFP) has been studied extensively as a serum marker
for HCC, and combined with tumor burden prognosticates HCC better than
tumor burden alone (18–20). Absolute AFP >1,000 ng/ml is a strong predictor of
vascular invasion and tumor recurrence. Patients with AFP >1,000 ng/ml had
higher 5-year recurrence rates (53%) compared to patients with AFP 100–1,000
ng/ml (26.8%) and <100 ng/ml (16.2%) (21).
AFP levels have now been adopted by UNOS as a marker for exclusion for liver
transplantation. Patients within the Milan criteria applying for MELD exception
must have an AFP ≤1,000 ng/ml. Patients >1,000 ng/ml can still be granted stan-
dard MELD exception points provided they undergo LRT with drop of AFP <500
ng/ml and remain within this range. If AFP >500 ng/ml after LRT, a review must
be filed with the NLRB.
LOCOREGIONAL THERAPIES
Trans-arterial chemoembolization
TACE is one of the most commonly used bridging therapies and involves
intra-arterial administration of a chemotherapeutic agent, usually doxorubicin,
mitomycin and/or cisplatin followed by an embolizing agent. Lipiodol, an
oily radiopaque marker used as an emulsifying agent, is coupled to the
132
TABLE 2 Overview of Locoregional Therapies
chemotherapeutic agent and used as a carrier to deliver the drug to the tumor.
Induction of ischemia and tumor necrosis through embolization of the tumor’s
microcirculation is the primary mode of tumor killing, which is then augmented
by the chemotherapeutic agent (23). Trans-arterial embolization can also be done
without a chemotherapeutic component, termed bland embolization, however
this is performed less commonly. Currently, TACE using drug-eluting beads (DEB-
TACE) has become more commonly used than conventional TACE, and as such
in some literature the term TACE is used interchangeably with DEB-TACE. DEB-
TACE involves the injection of embolization beads that are loaded with cytotoxic
chemotherapeutic agents to target a tumor. Contrary to conventional TACE where
there is concern of systemic exposure, drug-eluting beads release the chemothera-
peutic in a controlled and sustained manner, minimizing systemic toxicity and
improving drug delivery to the tumor (24). Both forms of TACE are used for treat-
ing larger (>3 cm) HCC without vascular invasion or extrahepatic spread where
the patient has preserved liver function. Damage to normal liver parenchyma from
arterial embolization is usually mitigated by the liver’s dual blood supply from the
portal vein and the hepatic artery. However, recurrent treatment with TACE
increases risk of complications and some centers recommend no more than 3–4
treatments times a year (1).
The most common complication of TACE, occurring in approximately 50% of
patients, is post-embolization syndrome (25). This occurs less frequently in
patients treated with DEB-TACE than TACE. Post-embolization syndrome con-
sists of a constellation of right upper quadrant pain, nausea, fever, fatigue, ele-
vated liver enzymes, bilirubin and mild to moderate ileus. Typically, symptoms
last 3–4 days and self-resolve in 7–10 days. Because this occurs frequently, in
most centers patients are observed as inpatients following TACE. Despite most
chemotherapy being localized to liver, other systemic complications include nau-
sea, vomiting and very rarely bone marrow suppression.
Although serious complications of TACE remain uncommon, liver failure can
occur as a result of ischemic damage. In a meta-analysis (26), liver failure was seen
in 7.5% of patients with HCC treated with TACE. However, liver decompensation
occurs most often in patients who had impaired liver function prior to TACE
treatment; therefore, patient selection is critical to reduce the risk of decompensa-
tion. Other complications of TACE include gastroduodenal ulcers (3–5%), hepatic
abscesses (2%), bile duct injury including strictures (0.5–2%) and renal dysfunc-
tion (2%). Rare but fatal complications include pulmonary and cerebral lipiodol
embolization. Overall, mortality rates from TACE are low at less than 1%, with
rates between 2–3% in patients with large tumors that develop tumor lysis syn-
drome (26). Small prospective studies and a meta-analysis have compared tradi-
tional TACE to DEB-TACE in the past and have suggested lower rates of
complications with similar tumor control in the latter (27–30). In the PRECISION-V
study (31), the largest comparative randomized controlled trial between both,
DEB-TACE showed less hepatotoxicity, better tumor response, and lower doxoru-
bicin related adverse effects compared to conventional TACE (in patients with
Child-Pugh B, ECOG1, bilobar disease and recurrent disease). In the same study,
less post procedural pain was noted with DEB-TACE. By contrast, two retrospec-
tive studies (32, 33) showed a higher risk of liver and biliary injuries with DEB-
TACE than conventional TACE. While the results are favorable, there still exists
some heterogeneity when comparing conventional TACE to DEB-TACE.
134 Ponniah S A et al.
Overall TACE therapies are safe in most patients. Absolute contraindications for
TACE therapies include decompensated cirrhosis (Child-Turcorrte-Pugh B
score >8), compromised portal vein flow or thrombus, extensive tumor in both
lobes of the liver, and renal failure. Several other relative contraindications exist
and include but are not limited to: serum bilirubin >2mg/dl, lactate dehydrogenase
>425 U/L, AST >100 units/L, tumor burden >50% of the liver, severe comorbidi-
ties, and untreated esophageal varices with bleeding risk. The ideal population for
TACE therapies is asymptomatic patients with solitary or few HCC tumors, well
compensated liver disease, and without metastatic disease or vascular invasion.
The use of TACE is not standardized and varies in protocol and type of embolic
and chemotherapeutic agent use. Few randomized studies compare TACE to sup-
portive care alone, however two landmark studies by Lo et al. (34) and Llovet
et al. (35) show a clear survival benefit in TACE compared with supportive treat-
ment. One-year survival rates were 57% (34) and 82% (35) compared to 32% and
63% in the control groups respectively. Mean survival was significantly longer
with TACE (28.6 months) compared to supportive care (17.9 months; P = 0.009)
(35). Additionally, in a recent systemic review (25) including 10,000 patients,
TACE showed response rates in >50% of patients, with 1 year survival rates >70%.
Current European Association for the Study of the Liver (EASL) guidelines sup-
port TACE with strong recommendation, citing high quality evidence. When
comparing TACE to transcatheter arterial radioembolization (TARE [discussed
below]), TACE is supported by a much greater number of trials, while TARE is
largely supported by single-center studies with variable patient cohorts (36).
While studies comparing TACE with TARE report no difference in overall sur-
vival, a single treatment with TACE is significantly less expensive than TARE (36).
Several favorable studies have advocated the use of TACE as effective bridging
therapy. Lower dropout rates (3–13%) have been noted by several studies com-
pared to historical data (3). In one of the most favorable studies by Graziadei et al.
(37), no patient experienced tumor progression following locoregional therapy
with TACE. Five-year survival rates after liver transplantation were high at 93%
and tumor recurrence rates were notably low (2%) despite long waiting times
prior to liver transplantation (mean of 178 days). While these studies show posi-
tive results, other studies (38–40) are less favorable and show no benefit in post
liver transplantation survival or tumor recurrence.
Downstaging using TACE has been shown to be effective, however success
rates have been variable in the literature. In the first ever study on locoregional
therapy in 1997, 62% of patients outside of MC were down staged with improved
5-year survival when compared to patients who could not be down staged with
TACE or did not receive TACE (1). Other studies have shown a similar favorable
role of TACE in downstaging patients to MC, however results remain inconsistent
with success rates ranging from 24% (41) to 90% (42).
Despite continued improvement in technique and patient selection, there con-
tinues to be debate regarding the ideal patient population, tumor burden and
tumor biology for TACE in the treatment of HCC.
attached to a bead and injected into the hepatic artery, designed to emit radiation
over a very small distance within the liver (2.5 mm). Y-90 undergoes beta decay
and irradiates the nearby tumor causing cell death. In comparison to TACE, this
procedure provides therapeutic effect through radiation instead of embolization.
This results in reduced toxicity and damage to the liver parenchyma and can be
used safely in patients with portal vein thrombosis (43, 44). The half-life of Y-90
is 2.67 days, and almost all radiation is delivered to the tumor within 2 weeks
after treatment. TARE has been used for bridging, downstaging, as a palliative
treatment for advanced disease, and as adjuvant therapy for surgically resected
HCC (45, 46).
Before the use of TARE, visceral angiography mapping with technetium-99
and SPECT-CT is required to detect shunts to the GI tract or lung. If shunts can-
not be reduced to less than 20% of the hepatic artery blood flow or less than 30
Gy radiation dose absorbed to the lungs via embolization or other means, there is
a high risk of toxicity and TARE should not be performed. Contraindications are
similar to TACE, however, unlike TACE, main portal vein thrombosis or obstruc-
tion is not a contraindication (47). TARE is also safe in patients with prior tran-
sjugular intrahepatic portosystemic shunt (48). Overall tolerance and safety are
comparable to TACE, despite fewer published studies involving TARE. However,
post-embolization syndrome is less common and tends to be less severe when it
does occur (49). Complications unique to TARE include radiation-induced liver
disease (RILD), radiation-induced biliary stricture, radiation-induced pneumoni-
tis, and radiation induced enteritis. The frequency and risk factors for each have
not been well studied and are listed in only a few studies. RILD is considered to
be a serious event, with two studies (50, 51) suggesting rates between 4% and
20%, and presents as jaundice, ascites, and manifestations of liver decompensa-
tion 2–8 weeks after treatment. The risk of RILD increases with repeated Y-90
administrations (51). Radiation-induced biliary stricturing is a less common
adverse effect of TARE occurring in less than 10% of patients (52). The frequency
of these complications has decreased with improvement in technique and
increased experience. As with TACE, proceduralists must be cautioned against
inadvertent embolization of the cystic artery to prevent gallbladder necrosis.
Radiation-induced pneumonitis and enteritis may occur if shunts to the gastroin-
testinal tract or lung are detected on pre-procedural evaluation, but rarely occur
if standard precautions are followed. Lastly, a theoretical concern with Y-90 used
as a bridge includes the risk of radiation exposure to the surgical or pathology
team handling the explanted liver, but considering its short half-life, this only
seems to be a consideration if liver transplantation is performed within 4 weeks
of TARE (3).
Because there are limited head-to-head comparisons between TACE and TARE,
there is no consensus regarding when/if TARE should be chosen over TACE.
However, TARE has been used increasingly due to a number of favorable studies.
Lewandowski et al. (53) compared TARE to TACE in T3 staged HCC down staged
to T2 which showed a significantly better response (61% vs 37%) and slower time
to progression (33.3 months vs 18.2 months). TARE-based bridging protocols
have also been associated with fewer pre-transplant locoregional therapy and had
lower rates of microvascular invasion in the explanted liver, which is associated
with risk of post-transplant HCC recurrence. Salem et al. (54) performed random-
ized controlled trials (RCTs) comparing TARE to TACE (PREMIERE trial) in 179
136 Ponniah S A et al.
patients and reported a significantly longer time to progression in TARE than with
TACE. The complication rate was also lower in the TARE group. Ettore et al. (55)
performed TARE in a small cohort of patients (22) with the majority being outside
the Milan criteria, and successfully bridged all patients and down staged 79% of
patients outside Milan criteria. More recently, the TRACE trial (56) compared Y-90
to DEB-TACE and included patients with BCLC A/B, ECOG 1 and segmental por-
tal vein thrombosis. This study showed superior results in the Y-90 group with
slower time to progression, improved survival, and comparable safety profiles
between Y-90 and DEB-TACE. While these results are favorable, few current stud-
ies overall have noted a difference in survival between TARE and TACE. Other
limitations to several favorable TARE trials include single centered studies, small
cohorts of patients, inherent physician biases and heterogeneity in results and
reporting. Lastly, TARE is more costly than TACE and may preclude its use at
some institutions (36).
Overall, TARE is likely to be used preferentially where there is higher level of
expertise, familiarity with the procedure, in those with infiltrative tumors, portal
vein invasion, larger tumors (>2 segments) and those with progression of tumor
despite use of TACE.
Thermal ablation
Multiple ablative techniques including radiofrequency ablation (RFA), microwave
ablation (MWA), and cryoablation use directly applied thermal energy to induce
tumor necrosis. RFA is the most commonly used and most widely studied of these
interventions and therefore the focus of discussion for this section. RFA involves
the insertion of narrow probes under imaging guidance, either ultrasound or
computed tomography, into a targeted liver lesion. The probes can be inserted
percutaneously, usually by an interventional radiologist or laparoscopically by a
surgeon. High-frequency alternating currents move from the electrode to the
lesion, creating a frictional pattern of ion movement which induces heating and
necrosis to the target lesion (57). Tumor cells die as the tissue becomes heated
above 60 degrees Celsius. The ablated area consists of the tumor plus 5–10 mm
boundary of nearby liver parenchyma (58). While there is no absolute tumor size
for RFA, most reported favorable outcomes to suggest smaller tumors <3 cm
(59, 60), less than or equal to 3 nodules, and without major vascular or biliary
structures near the target lesions.
Limitations of RFA involve the duration of treatment and the risk of thermal
injury to nearby anatomical structures. RFA typically takes 16–18 mins to ablate
a 3–4 cm lesion and there is a risk of dissipating heat energy to nearby blood
vessels (>3 mm) termed the “heat sink effect”; the “cooler” vascular flow near
the lesion may absorb heat resulting in incomplete tumor necrosis in the target
lesion.
Additionally, RFA is not suitable for lesions near certain anatomical structures,
namely the dome of the liver, the gallbladder and the biliary tree (60). There is
also a risk of liver capsule rupture (~2%) and resulting peritoneal seeding when
treating peripheral tumors (3). Overall, the major complication rate ranges from
2.4–13.1%, but RFA is still superior to surgical resection (13). Livraghi et al. (61),
in 218 patients with small and early HCC, showed no perioperative mortality in
RFA with lower than previously suggested rates of major complications (1.8%).
Locoregional Therapy Prior to Liver Transplant 137
RFA as a bridging therapy in 379 patients meeting Toronto extended criteria. They
found similar 5-year survival and dropout rates across all three modalities. A pro-
spective study by Lee et al. (68) evaluated SBRT in decompensated cirrhosis with
Child-Pugh B or C cirrhosis. Patients had either already completed TACE prior to
SBRT or had TACE in combination with SBRT. This study showed favorable results
as the Child-Pugh class remained stable in 30%, and even improved in 22% of
patients, and was associated with an overall improvement in survival. Other stud-
ies (69–71), mostly retrospective in nature, all show comparable outcomes in
local control of disease. These studies suggest that SBRT can be used safely as
bridging therapy when other modalities have failed or are not applicable.
Additionally, SBRT may offer advantages in patients with liver dysfunction who
may not tolerate TACE or RFA (67). Overall, SBRT has now emerged as an effec-
tive LRT for patients with advanced disease with minimal toxicity.
CONCLUSION
Bridging therapy is now the standard of care to prevent waitlist dropout and
decrease HCC recurrence post-transplant. There are no prospective RCTs compar-
ing locoregional therapy modalities before liver transplantation, therefore no sin-
gle bridging modality is recommended over another (13). The literature remains
difficult to analyze due to non-standardized treatment protocols and heteroge-
neous patient populations. Recently, Kulik et al. compared 18 studies (72) and
found that bridging locoregional therapy had no significant impact on survival
rate or HCC recurrence post-transplant. Several studies were found to have impre-
cision regarding inclusion criteria, as some patients were within the Milan criteria,
outside the Milan criteria, or undefined (11). The lack of randomized studies and
potentially biased patient selection in these studies should also be considered;
patients who received locoregional therapy, when compared to those who did not,
likely had more risk factors such as advanced tumors, aggressive tumor biology,
and longer wait times (72). Overall, bridging locoregional therapy is safe and
effective. The type of locoregional therapy selected is based on liver function, size,
the number of tumors, and institutional experience. Ablation is the preferred
modality for smaller tumors with a size ≤3 cm; the type of ablation is expertise-
dependent, but RFA is frequently used. Larger lesions are typically treated with
TACE or Y-90 TARE based on institutional experience and/or preference. SBRT is
a novel approach and has shown promising outcomes for those with liver dys-
function, failed other locoregional therapies, or for those who are no longer liver
transplant candidates.
Like bridging therapy, there remains no consensus on optimal downstaging
strategy. Several factors result in largely varying success rates (~25–90%) (11) as
numerous inconsistencies in the current literature make data interpretation diffi-
cult. Firstly, the definition of downstaging is poorly defined amongst studies.
None of the present studies were randomized controlled trials (11). Some studies
define downstaging as the reduction of tumors to the Milan criteria, others base
downstaging on the complete absence of tumors, and few use explant pathology
to defined success (11). Secondly, there are inconsistencies in locoregional ther-
apy selection, varying tumor burden before locoregional therapy, and differing
Locoregional Therapy Prior to Liver Transplant 139
Copyright and permission statement: The authors confirm that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Bryce K, Tsochatzis EA. Downstaging for hepatocellular cancer: harm or benefit. Transl Gastroenterol
Hepatol. 2017;2:106. https://doi.org/10.21037/tgh.2017.11.18
2. Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, et al. Liver transplantation for
the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996 Mar
14;334(11):693–9. https://doi.org/10.1056/NEJM199603143341104
3. Byrne TJ, Rakela J. Loco-regional therapies for patients with hepatocellular carcinoma awaiting liver
transplantation: Selecting an optimal therapy. World J Transplant. 2016;6(2):306–13. https://doi.
org/10.5500/wjt.v6.i2.306
4. Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A, et al. Liver transplantation for hepatocel-
lular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology.
2001;33(6):1394–403. https://doi.org/10.1053/jhep.2001.24563
5. Yao FY, Xiao L, Bass NM, Kerlan R, Ascher NL, Roberts JP. Liver transplantation for hepatocellular
carcinoma: validation of the UCSF-expanded criteria based on preoperative imaging. Am J Transplant.
2007;7(11):2587–96. https://doi.org/10.1111/j.1600-6143.2007.01965.x
6. Abdelfattah MR, Elsiesy H, Al-Manea H, Broering DC. Liver transplantation for hepatocellu-
lar carcinoma within the Milan criteria versus the University of California San Francisco criteria:
a comparative study. Eur J Gastroenterol Hepatol. 2018;30(4):398–403. https://doi.org/10.1097/
MEG.0000000000001044
7. Mazzaferro V, Llovet JM, Miceli R, Bhoori S, Schiavo M, Mariani L, et al. Predicting survival after
liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a ret-
rospective, exploratory analysis. Lancet Oncol. 2009;10(1):35–43. https://doi.org/10.1016/
S1470-2045(08)70284-5
8. Lingiah VA, Niazi M, Olivo R, Paterno F, Guarrera JV, Pyrsopoulos NT. Liver Transplantation
Beyond Milan Criteria. J Clin Transl Hepatol. 2020;8(1):69–75. https://doi.org/10.14218/
JCTH.2019.00050
140 Ponniah S A et al.
9. Sapisochin G, Goldaracena N, Laurence JM, Dib M, Barbas A, Ghanekar A, et al. The extended
Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: A prospective
validation study. Hepatology. 2016 12;64(6):2077–88. https://doi.org/10.1002/hep.28643
10. Trieu JA, Bilal M, Hmoud B. Factors associated with waiting time on the liver transplant list: an analy-
sis of the United Network for Organ Sharing (UNOS) database. Ann Gastroenterol. 2018;31(1):84–9.
https://doi.org/10.20524/aog.2017.0217
11. Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD guidelines
for the treatment of hepatocellular carcinoma. Hepatology. 2018 01;67(1):358–80. https://doi.
org/10.1002/hep.29086
12. Coletta M, Nicolini D, Benedetti Cacciaguerra A, Mazzocato S, Rossi R, Vivarelli M. Bridging
patients with hepatocellular cancer waiting for liver transplant: all the patients are the same. Transl
Gastroenterol Hepatol. 2017;2:78. https://doi.org/10.21037/tgh.2017.09.01
13. Oligane HC, Close ON, Xing M, Kim HS. Bridging locoregional therapy: Longitudinal trends and
outcomes in patients with hepatocellular carcinoma. Transplant Rev (Orlando). 2017;31(2):136–43.
https://doi.org/10.1016/j.trre.2017.01.004
14. Mehta N, Dodge JL, Grab JD, Yao FY. National Experience on Down-Staging of Hepatocellular
Carcinoma Before Liver Transplant: Influence of Tumor Burden, Alpha-Fetoprotein, and Wait Time.
Hepatology. 2020;71(3):943–54. https://doi.org/10.1002/hep.30879
15. Marrero JA, Kulik LM, Sirlin CB, Zhu AX, Finn RS, Abecassis MM, et al. Diagnosis, Staging, and
Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for
the Study of Liver Diseases. Hepatology. 2018;68(2):723–50. https://doi.org/10.1002/hep.29913
16. Mehta N, Yao FY. What Are the Optimal Liver Transplantation Criteria for Hepatocellular Carcinoma.
Clin Liver Dis (Hoboken). 2019;13(1):20–5. https://doi.org/10.1002/cld.793
17. Galle PR, Forner A, Llovet JM, Mazzaferro V, Piscaglia F, Raoul JL, et al. EASL Clinical Practice
Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236.
18. Halazun KJ, Tabrizian P, Najjar M, Florman S, Schwartz M, Michelassi F, et al. Is it Time to Abandon
the Milan Criteria?: Results of a Bicoastal US Collaboration to Redefine Hepatocellular Carcinoma
Liver Transplantation Selection Policies. Ann Surg. 2018;268(4):690–9. https://doi.org/10.1097/
SLA.0000000000002964
19. Giard JM, Mehta N, Dodge JL, Roberts JP, Yao FY. Alpha-Fetoprotein Slope >7.5 ng/mL per Month
Predicts Microvascular Invasion and Tumor Recurrence After Liver Transplantation for Hepatocellular
Carcinoma. Transplantation. 2018;102(5):816–22. https://doi.org/10.1097/TP.0000000000002094
20. Mehta N, Dodge JL, Roberts JP, Hirose R, Yao FY. Alpha-Fetoprotein Decrease from >1,000 to
<500 ng/mL in Patients with Hepatocellular Carcinoma Leads to Improved Posttransplant Outcomes.
Hepatology. 2019;69(3):1193–205. https://doi.org/10.1002/hep.30413
21. Hameed B, Mehta N, Sapisochin G, Roberts JP, Yao FY. Alpha-fetoprotein level >1000 ng/mL as an
exclusion criterion for liver transplantation in patients with hepatocellular carcinoma meeting the
Milan criteria. Liver Transpl. 2014;20(8):945–51. https://doi.org/10.1002/lt.23904
22. Mehta N. Hepatocellular Carcinoma-How to Determine Therapeutic Options. Hepatol Commun.
2020;4(3):342–54. https://doi.org/10.1002/hep4.1481
23. Raoul JL, Forner A, Bolondi L, Cheung TT, Kloeckner R, de Baere T. Updated use of TACE for hepa-
tocellular carcinoma treatment: How and when to use it based on clinical evidence. Cancer Treat Rev.
2019;72:28–36. https://doi.org/10.1016/j.ctrv.2018.11.002
24. Thuluvath PJ, To C, Amjad W. Role of Locoregional Therapies in Patients With Hepatocellular Cancer
Awaiting Liver Transplantation. Am J Gastroenterol. 2021;116(1):57–67. https://doi.org/10.14309/
ajg.0000000000000999
25. Lencioni R, de Baere T, Soulen MC, Rilling WS, Geschwind JF. Lipiodol transarterial chemoemboliza-
tion for hepatocellular carcinoma: A systematic review of efficacy and safety data. Hepatology. 2016
07;64(1):106–16. https://doi.org/10.1002/hep.28453
26. Marelli L, Stigliano R, Triantos C, Senzolo M, Cholongitas E, Davies N, et al. Transarterial therapy
for hepatocellular carcinoma: which technique is more effective? A systematic review of cohort
and randomized studies. Cardiovasc Intervent Radiol. 2007;30(1):6–25. https://doi.org/10.1007/
s00270-006-0062-3
Locoregional Therapy Prior to Liver Transplant 141
27. van Malenstein H, Maleux G, Vandecaveye V, Heye S, Laleman W, van Pelt J, et al. A randomized
phase II study of drug-eluting beads versus transarterial chemoembolization for unresectable hepato-
cellular carcinoma. Onkologie. 2011;34(7):368–76. https://doi.org/10.1159/000329602
28. Gao S, Yang Z, Zheng Z, Yao J, Deng M, Xie H, et al. Doxorubicin-eluting bead versus conven-
tional TACE for unresectable hepatocellular carcinoma: a meta-analysis. Hepatogastroenterology.
2013;60(124):813–20.
29. Prajapati HJ, Dhanasekaran R, El-Rayes BF, Kauh JS, Maithel SK, Chen Z, et al. Safety and efficacy of
doxorubicin drug-eluting bead transarterial chemoembolization in patients with advanced hepatocel-
lular carcinoma. J Vasc Interv Radiol. 2013;24(3):307–15. https://doi.org/10.1016/j.jvir.2012.11.026
30. Golfieri R, Giampalma E, Renzulli M, Cioni R, Bargellini I, Bartolozzi C, et al. Randomised controlled
trial of doxorubicin-eluting beads vs conventional chemoembolisation for hepatocellular carcinoma.
Br J Cancer. 2014;111(2):255–64. https://doi.org/10.1038/bjc.2014.199
31. Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, et al. Prospective randomized study
of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of
the PRECISION V study. Cardiovasc Intervent Radiol. 2010;33(1):41–52. https://doi.org/10.1007/
s00270-009-9711-7
32. Guiu B, Deschamps F, Aho S, Munck F, Dromain C, Boige V, et al. Liver/biliary injuries following che-
moembolisation of endocrine tumours and hepatocellular carcinoma: lipiodol vs. drug-eluting beads.
J Hepatol. 2012;56(3):609–17. https://doi.org/10.1016/j.jhep.2011.09.012
33. Monier A, Guiu B, Duran R, Aho S, Bize P, Deltenre P, et al. Liver and biliary damages following trans-
arterial chemoembolization of hepatocellular carcinoma: comparison between drug-eluting beads and
lipiodol emulsion. Eur Radiol. 2017;27(4):1431–9. https://doi.org/10.1007/s00330-016-4488-y
34. Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, et al. Randomized controlled trial of tran-
sarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology.
2002;35(5):1164–71. https://doi.org/10.1053/jhep.2002.33156
35. Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, et al. Arterial embolisation or chemo-
embolisation versus symptomatic treatment in patients with unresectable hepatocellular carci-
noma: a randomised controlled trial. Lancet. 2002;359(9319):1734–9. https://doi.org/10.1016/
S0140-6736(02)08649-X
36. Skef W, Agarwal M, Mikolajczyk AE. Position 1: Transarterial Chemoembolization Should Be the
Primary Locoregional Therapy for Unrespectable Hepatocelluar Carcinoma. Clin Liver Dis (Hoboken).
2020;15(2):71–3. https://doi.org/10.1002/cld.909
37. Graziadei IW, Sandmueller H, Waldenberger P, Koenigsrainer A, Nachbaur K, Jaschke W, et al.
Chemoembolization followed by liver transplantation for hepatocellular carcinoma impedes tumor
progression while on the waiting list and leads to excellent outcome. Liver Transpl. 2003;9(6):
557–63. https://doi.org/10.1053/jlts.2003.50106
38. Cabrera R, Dhanasekaran R, Caridi J, Clark V, Morelli G, Soldevila-Pico C, et al. Impact of transarterial
therapy in hepatitis C-related hepatocellular carcinoma on long-term outcomes after liver transplanta-
tion. Am J Clin Oncol. 2012;35(4):345–50. https://doi.org/10.1097/COC.0b013e31821631f6
39. Decaens T, Roudot-Thoraval F, Bresson-Hadni S, Meyer C, Gugenheim J, Durand F, et al. Impact
of pretransplantation transarterial chemoembolization on survival and recurrence after liver trans-
plantation for hepatocellular carcinoma. Liver Transpl. 2005;11(7):767–75. https://doi.org/10.1002/
lt.20418
40. Porrett PM, Peterman H, Rosen M, Sonnad S, Soulen M, Markmann JF, et al. Lack of benefit of
pre-transplant locoregional hepatic therapy for hepatocellular cancer in the current MELD era. Liver
Transpl. 2006;12(4):665–73. https://doi.org/10.1002/lt.20636
41. Chapman WC, Majella Doyle MB, Stuart JE, Vachharajani N, Crippin JS, Anderson CD, et al.
Outcomes of neoadjuvant transarterial chemoembolization to downstage hepatocellular car-
cinoma before liver transplantation. Ann Surg. 2008;248(4):617–25. https://doi.org/10.1097/
SLA.0b013e31818a07d4
42. Ravaioli M, Grazi GL, Piscaglia F, Trevisani F, Cescon M, Ercolani G, et al. Liver transplantation for
hepatocellular carcinoma: results of down-staging in patients initially outside the Milan selection
criteria. Am J Transplant. 2008;8(12):2547–57. https://doi.org/10.1111/j.1600-6143.2008.02409.x
142 Ponniah S A et al.
43. Sato K, Lewandowski RJ, Bui JT, Omary R, Hunter RD, Kulik L, et al. Treatment of unresectable pri-
mary and metastatic liver cancer with yttrium-90 microspheres (TheraSphere): assessment of hepatic
arterial embolization. Cardiovasc Intervent Radiol. 2006;29(4):522–9. https://doi.org/10.1007/
s00270-005-0171-4
44. Salem R, Thurston KG. Radioembolization with 90Yttrium microspheres: a state-of-the-art brachy-
therapy treatment for primary and secondary liver malignancies. Part 1: Technical and meth-
odologic considerations. J Vasc Interv Radiol. 2006;17(8):1251–78. https://doi.org/10.1097/01.
RVI.0000233785.75257.9A
45. Tohme S, Sukato D, Chen HW, Amesur N, Zajko AB, Humar A, et al. Yttrium-90 radioemboliza-
tion as a bridge to liver transplantation: a single-institution experience. J Vasc Interv Radiol.
2013;24(11):1632–8. https://doi.org/10.1016/j.jvir.2013.07.026
46. Kulik LM, Carr BI, Mulcahy MF, Lewandowski RJ, Atassi B, Ryu RK, et al. Safety and efficacy of
90Y radiotherapy for hepatocellular carcinoma with and without portal vein thrombosis. Hepatology.
2008;47(1):71–81. https://doi.org/10.1002/hep.21980
47. Kennedy A, Nag S, Salem R, Murthy R, McEwan AJ, Nutting C, et al. Recommendations for radioem-
bolization of hepatic malignancies using yttrium-90 microsphere brachytherapy: a consensus panel
report from the radioembolization brachytherapy oncology consortium. Int J Radiat Oncol Biol Phys.
2007;68(1):13–23. https://doi.org/10.1016/j.ijrobp.2006.11.060
48. Donahue LA, Kulik L, Baker T, Ganger DR, Gupta R, Memon K, et al. Yttrium-90 radioemboliza-
tion for the treatment of unresectable hepatocellular carcinoma in patients with transjugular intra-
hepatic portosystemic shunts. J Vasc Interv Radiol. 2013;24(1):74–80. https://doi.org/10.1016/j.
jvir.2012.09.030
49. Riaz A, Awais R, Salem R. Side effects of yttrium-90 radioembolization. Front Oncol. 2014;4:198.
https://doi.org/10.3389/fonc.2014.00198
50. Sangro B, Gil-Alzugaray B, Rodriguez J, Sola I, Martinez-Cuesta A, Viudez A, et al. Liver dis-
ease induced by radioembolization of liver tumors: description and possible risk factors. Cancer.
2008;112(7):1538–46. https://doi.org/10.1002/cncr.23339
51. Lam MG, Louie JD, Iagaru AH, Goris ML, Sze DY. Safety of repeated yttrium-90 radioembolization.
Cardiovasc Intervent Radiol. 2013;36(5):1320–8. https://doi.org/10.1007/s00270-013-0547-9
52. Ng SS, Yu SC, Lai PB, Lau WY. Biliary complications associated with selective internal radiation
(SIR) therapy for unresectable liver malignancies. Dig Dis Sci. 2008;53(10):2813–7. https://doi.
org/10.1007/s10620-008-0222-1
53. Lewandowski RJ, Kulik LM, Riaz A, Senthilnathan S, Mulcahy MF, Ryu RK, et al. A comparative analy-
sis of transarterial downstaging for hepatocellular carcinoma: chemoembolization versus radioembo-
lization. Am J Transplant. 2009;9(8):1920–8. https://doi.org/10.1111/j.1600-6143.2009.02695.x
54. Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, Gabr A, et al. Y90 Radioembolization Significantly
Prolongs Time to Progression Compared With Chemoembolization in Patients With Hepatocellular
Carcinoma. Gastroenterology. 2016;151(6):1155–63. https://doi.org/10.1053/j.gastro.2016.08.029
55. Ettorre GM, Levi Sandri GB, Laurenzi A, Colasanti M, Meniconi RL, Lionetti R, et al. Yttrium-90
Radioembolization for Hepatocellular Carcinoma Prior to Liver Transplantation. World J Surg.
2017;41(1):241–9. https://doi.org/10.1007/s00268-016-3682-z
56. Seinstra BA, Defreyne L, Lambert B, Lam MG, Verkooijen HM, van Erpecum KJ, et al. Transarterial
radioembolization versus chemoembolization for the treatment of hepatocellular carcinoma
(TRACE): study protocol for a randomized controlled trial. Trials. 2012;13:144. https://doi.
org/10.1186/1745-6215-13-144
57. McGahan JP, Brock JM, Tesluk H, Gu WZ, Schneider P, Browning PD. Hepatic ablation with use of
radio-frequency electrocautery in the animal model. J Vasc Interv Radiol. 1992;3(2):291–7. https://
doi.org/10.1016/S1051-0443(92)72028-4
58. Makary MS, Khandpur U, Cloyd JM, Mumtaz K, Dowell JD. Locoregional Therapy Approaches
for Hepatocellular Carcinoma: Recent Advances and Management Strategies. Cancers (Basel).
2020;12(7):E1914. https://doi.org/10.3390/cancers12071914
59. Pompili M, Francica G, Ponziani FR, Iezzi R, Avolio AW. Bridging and downstaging treatments for
hepatocellular carcinoma in patients on the waiting list for liver transplantation. World J Gastroenterol.
2013;19(43):7515–30.
Locoregional Therapy Prior to Liver Transplant 143
60. Head HW, Dodd GD, Dalrymple NC, Prasad SR, El-Merhi FM, Freckleton MW, et al. Percutaneous
radiofrequency ablation of hepatic tumors against the diaphragm: frequency of diaphragmatic injury.
Radiology. 2007;243(3):877–84. https://doi.org/10.1148/radiol.2433060157
61. Livraghi T, Meloni F, Di Stasi M, Rolle E, Solbiati L, Tinelli C, et al. Sustained complete response and
complications rates after radiofrequency ablation of very early hepatocellular carcinoma in cirrho-
sis: Is resection still the treatment of choice. Hepatology. 2008;47(1):82–9. https://doi.org/10.1002/
hep.21933
62. Lu Z, Wen F, Guo Q, Liang H, Mao X, Sun H. Radiofrequency ablation plus chemoembolization
versus radiofrequency ablation alone for hepatocellular carcinoma: a meta-analysis of randomized-
controlled trials. Eur J Gastroenterol Hepatol. 2013;25(2):187–94. https://doi.org/10.1097/
MEG.0b013e32835a0a07
63. Mazzaferro V, Battiston C, Perrone S, Pulvirenti A, Regalia E, Romito R, et al. Radiofrequency ablation
of small hepatocellular carcinoma in cirrhotic patients awaiting liver transplantation: a prospective
study. Ann Surg. 2004;240(5):900–9. https://doi.org/10.1097/01.sla.0000143301.56154.95
64. Chu HH, Kim JH, Yoon HK, Ko HK, Gwon DI, Kim PN, et al. Chemoembolization Combined with
Radiofrequency Ablation for Medium-Sized Hepatocellular Carcinoma: A Propensity-Score Analysis.
J Vasc Interv Radiol. 2019;30(10):1533–43. https://doi.org/10.1016/j.jvir.2019.06.006
65. Andolino DL, Johnson CS, Maluccio M, Kwo P, Tector AJ, Zook J, et al. Stereotactic body radiotherapy
for primary hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2011;81(4):e447–53. https://doi.
org/10.1016/j.ijrobp.2011.04.011
66. Sandroussi C, Dawson LA, Lee M, Guindi M, Fischer S, Ghanekar A, et al. Radiotherapy as a bridge
to liver transplantation for hepatocellular carcinoma. Transpl Int. 2010;23(3):299–306. https://doi.
org/10.1111/j.1432-2277.2009.00980.x
67. Sapisochin G, Barry A, Doherty M, Fischer S, Goldaracena N, Rosales R, et al. Stereotactic body
radiotherapy vs. TACE or RFA as a bridge to transplant in patients with hepatocellular carcinoma. An
intention-to-treat analysis. J Hepatol. 2017;67(1):92–9. https://doi.org/10.1016/j.jhep.2017.02.022
68. Lee P, Ma Y, Zacharias I, Bozorgzadeh A, Wilson S, Foley K, et al. Stereotactic Body Radiation Therapy
for Hepatocellular Carcinoma in Patients With Child-Pugh B or C Cirrhosis. Adv Radiat Oncol.
2020;5(5):889–96. https://doi.org/10.1016/j.adro.2020.01.009
69. Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes After Stereotactic
Body Radiotherapy or Radiofrequency Ablation for Hepatocellular Carcinoma. J Clin Oncol. 2016 Feb
10;34(5):452–9. https://doi.org/10.1200/JCO.2015.61.4925
70. Kim N, Kim HJ, Won JY, Kim DY, Han KH, Jung I, et al. Retrospective analysis of stereotactic body
radiation therapy efficacy over radiofrequency ablation for hepatocellular carcinoma. Radiother
Oncol. 2019;131:81–7. https://doi.org/10.1016/j.radonc.2018.12.013
71. Sapir E, Tao Y, Schipper MJ, Bazzi L, Novelli PM, Devlin P, et al. Stereotactic Body Radiation Therapy
as an Alternative to Transarterial Chemoembolization for Hepatocellular Carcinoma. Int J Radiat
Oncol Biol Phys. 2018;100(1):122–30. https://doi.org/10.1016/j.ijrobp.2017.09.001
72. Kulik L, Heimbach JK, Zaiem F, Almasri J, Prokop LJ, Wang Z, et al. Therapies for patients with
hepatocellular carcinoma awaiting liver transplantation: A systematic review and meta-analysis.
Hepatology. 2018;67(1):381–400. https://doi.org/10.1002/hep.29485
8
Hepatoblastoma
Josef Hager1 • Consolato M. Sergi2
1
Department of Pediatric Surgery, University Hospital of Surgery, Medical University of
Innsbruck, Innsbruck, Austria; 2Departments of Pediatrics, Laboratory Medicine and
Pathology, Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada
Author for Correspondence: Josef Hager, Department of Pediatric Surgery, University
Hospital of Surgery, Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck,
Austria. Email: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch8
INTRODUCTION
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
145
146 Hager J and Sergi C M
ETIOLOGY
The etiology of hepatoblastoma, which usually occurs sporadically, has not yet
been clarified. Somatic gene mutations in hepatoblasts and other observations
suggest tumor development is spontaneous (6). It is believed that premature chil-
dren with a very low birth weight (<1,500 g) have an increased risk of developing
malignant tumors, including hepatoblastoma. This fact, which was first reported
in 1997 (7), was confirmed by a worldwide scientific study in 2019. Still, an
explanation for this observation - accidental or causal connection - could not be
established (8). The fact that relatively common conditions such as pre-eclampsia,
fetal distress before or during childbirth, or congenital malformations could play
roles as possible tumor inducers could allegedly be statistically determined in
some studies, but an explanation for these observations is still missing (9).
Regardless of these observations, over the last few decades, various genetic dis-
eases have been found to be risk factors for developing hepatoblastoma. Some of
them include familial adenomatous polyposis (10), Beckwith-Wiedemann syn-
drome (11), and trisomy 18 (Edwards syndrome) (12). A connection between the
occurrence of hepatoblastoma with other genetic diseases such as Li-Fraumeni
syndrome or Prader-Willi syndrome has been discussed, but the link has not yet
been proven satisfactorily (13). Finally, various external and epigenetic influences
have been debated as possible causes for neoplastic development. Smoking before
and/or during pregnancy is an example of this; opinions on this, however, are
equivocal (6, 14).
PATHOGENESIS
classified based on the original histological classification of Ishak and Glunz (15).
Histologically, hepatoblastoma are broadly classified into two types: epithelial and
mixed. Depending on the degree of differentiation, hepatoblastoma cells can be
distinguished into two subtypes: embryonic and fetal. In some cases, both cell
types are present. Embryonic tumor cells are less differentiated whereas the fetal
cells are well-differentiated. Small-cell anaplastic hepatoblastoma is a unique sub-
type; it mainly infiltrates the bile ducts and is considered prognostically unfavor-
able (16). In addition to epithelial components, the mixed hepatoblastoma type
contains mesenchymal stroma such as osteoid, collagen fibers, and rarely, carti-
lage and skeletal muscle cells (17). Liver progenitor cells harbor the ability to
express of keratin 19 (CK19) and/or the epithelial cell adhesion molecule (EpCAM)
(18–21). EpCAM is a transmembrane glycoprotein mediating calcium-indepen-
dent homotypic cell-cell adhesion in the epithelium. This molecule is also involved
in cellular signaling, migration, proliferation, and differentiation. It plays a role in
the event-free survival outcome of patients harboring hepatoblastoma (18, 20–23).
CK19 expression has been correlated with aggressive behavior in hepatoblastoma
[20] and hepatocellular carcinoma (24). Of tremendous importance is that
EpCAM expression is independent of previous cisplatin-based chemotherapy and
can be utilized as a tumor marker and potential target for immunotherapy (18, 19).
Kiruthiga et al. found that more than 90% of tumors with strong expression of
EpCAM showed viable tumor after chemotherapy (19).
EPIDEMIOLOGY
SYMPTOMS
always when the disease has reached an advanced stage. Overall, the complaints
are nonspecific, including nausea, vomiting, weight loss and increasing general
weakness, which can delay development. In this context, osteopenia can develop.
Children with hepatoblastoma can become conspicuous due to osteopenia, and
resultant pathological fractures (34). Very rarely, obstructive jaundice can occur
when the tumor occludes the intrahepatic biliary tract (2). Spontaneous tumor
rupture with extensive intra- or extra-tumor bleeding is extremely rare (35).
Precocious puberty can occur in boys due to the increased formation of human
chorionic gonadotropin (β-hCG) caused by hepatoblastoma (36). Further symp-
toms can occur depending on the site of metastasis. The lungs are most com-
monly affected, with breathing difficulties, coughing fits, and occasional
hemoptysis (2, 3).
DIAGNOSTICS
The diagnosis of hepatoblastoma involves tumor detection and staging. The fol-
lowing diagnostic measures are initially recommended in addition to a clinical
examination of the patient. During the clinical examination, the primary focus
should be on signs of a genetic diseases (for example, macroglossia and hemihy-
pertrophy, among others, which are characteristic features suggesting Beckwith-
Wiedemann syndrome). Most patients affected by this syndrome will require
surgery (37).
Laboratory tests
Laboratory diagnostics for hepatoblastoma include a blood count as standard
(mild anemia, leukocytosis, and thrombocytosis are possible), a liver function test
(SGOT, LDH, AP can be slightly increased) including bilirubin (raised in the case
of bile duct obstruction) and the tumor markers ferritin, CEA (carcinoembryonic
antigen test) and the NSE (neuron-specific enolase), and, if necessary, urinary
catecholamines (to rule out neuroblastoma) and an evaluation of the titer regard-
ing hepatotropic viruses. It is essential to determine the malignancy-associated
tumor markers, namely alpha-1-fetoprotein (AFP) and beta-human chorionic
gonadotropin (β-HCG). AFP is elevated in 80–90% of patients. The typical AFP
values of the respective age group must be observed. Hepatoblastomas with low
AFP (<100ng /ml) are considered aggressive and have an abysmal prognosis (38).
β-HCG is increased in about 20% of patients. However, this does not seem not to
have prognostic significance (39).
Imaging
If a liver tumor is suspected, first contrast-enhanced sonography of the liver is
performed. If the tumor shows increased echogenicity on contrast-enhanced
sonography and pronounced vascular supply on Doppler ultrasound, possible
tumor invasion into one or more hepatic vessels is suspected. This indicates a
malignant process; however, these are not confirmatory evidence of malignancy
(40). Other imaging options, namely magnetic resonance imaging (MRI) or
Hepatoblastoma 149
computed tomography (CT) of the abdomen with contrast agent, can not only
provide evidence of a malignant tumor of the liver, but also allow for the assess-
ment of the extent of malignancy, and even the relationship of the neoplasm to the
hepatic vessels and the liver segments. Nevertheless, a reliable diagnosis of hepa-
toblastoma cannot be achieved with these examinations either (41). But thanks to
these techniques, apart from particular indications, angiography or liver scintigra-
phy can now be used. A lung CT scan to determine or exclude lung metastases
and skeletal scintigraphy with 99-technetium phosphonate to realize or exclude
possible bone metastases is recommended as a precautionary measure. It remains
to be seen whether FDG-PET/CT performed for the initial diagnosis of a possible
hepatoblastoma is sensible, especially since only a possible correlation between
uptake and tumor-related increased AFP values can be established. It is well
known that FDG-PET/CT is vital during treatment or as part of the follow-up of a
malignant tumor. For example, for hepatoblastoma, the detection of metabolically
active metastases indicates an unfavorable prognosis (42).
Histology
To confirm the diagnosis of hepatoblastoma, histological examination of the
tumor biopsy is the gold standard. In most suspected cases, the biopsy material
can be removed using a percutaneous punch (approximately 3–6 liver cores). If
the tumor is difficult to access percutaneously or is heavily vascularized, a biopsy
is indicated either laparoscopically or via laparotomy. Fine-needle aspiration of a
possible hepatoblastoma for the aspirate’s cytological examination is not regarded
by most oncologists as sufficient for a reliable diagnosis, even if there have been
isolated experiences to the contrary (43). This also applies to a percutaneous
punch biopsy by an interventional radiologist (44). The biopsy material should be
examined both conventionally histologically (as a paraffin preparation) and
immunohistochemically. The diagnosis of hepatoblastoma should always be con-
firmed by a reference pathologist, that is, a pathologist with experience working
for either the Children’s Oncology Group (COG) or the International Society of
Pediatric Oncology (SIOP) or the United Kingdom Children Cancer Study Group
(UKCCSG). According to the guidelines of the (German) Society for Pediatric
Oncology and Hematology (GPOH), children between the ages of 6 months and
3 years of age who have a liver tumor suspected of being hepatoblastoma by imag-
ing and an AFP value of over 1,000 ng/ml or an AFP value that is at least three
times higher than the age norm, biopsy confirming the diagnosis of hepatoblas-
toma is not necessary, especially since in these cases the incriminated liver tumor
is always hepatoblastoma (3). However, this view is not generally approved; on
the contrary, most oncologists require a tumor biopsy to confirm the diagnosis
(19) (Figure 1).
In the case of resected specimens, size, the exterior (solid/cystic), and tumor
necrosis are noted (19). Histology is key in reporting hepatoblastoma, and the
report should include the histological subtype, mitotic activity in 10 high power
fields (HPF) (low mitotic activity when ≤5/10 HPF and high mitotic activity when
>5/10 HPF, presence of extramedullary hematopoiesis, and intratumoral fatty
change [steatosis]). Six major subtypes are recognized, including: pure fetal
epithelial, mixed embryonal and fetal epithelial, macro trabecular, small cell
150 Hager J and Sergi C M
Figure 1. Histology of hepatoblastoma. A. Slice following partial resection of the liver showing
a hepatoblastoma with grey cut surface and small areas of hemorrhages. B. Microphotograph
showing hepatoblastoma with pure fetal histology and minimal mitotic activity (hematoxylin
and eosin staining, x200 original magnification). C. Microphotograph showing
hepatoblastoma of embryonal type (hematoxylin and eosin staining, x200 original
magnification). D. Microphotograph showing a teratoid hepatoblastoma with ribbons and
nephroblastoma-like tubules and acini (hematoxylin and eosin staining, x50 original
magnification, scale bar: 100 micrometers). E. Microphotograph showing lung metastasis of a
hepatoblastoma with two nodules depicting hemosiderin accumulation (blue) both inside
and at the edges of the tumor clusters. The lung tissue in the middle of the microphotograph
shows the characteristic alveolar pattern. Perls’ Prussian Blue (PPB) has its name from the
19th century German pathologist Max Perls, who introduced this technique in histopathology
to stain iron in the ferric state (e.g., ferritin and hemosiderin (Perls’ Prussian Blue, x50 original
magnification, scale bar: 100 micrometers). F. Microphotograph showing a hepatoblastoma
post chemotherapy exhibiting some cell maturation (right) and some hemorrhage (center).
Some fibrosis is encountered on the left side of the microphotograph (hematoxylin and
eosin staining, x200 original magnification).
Hepatoblastoma 151
THERAPEUTIC CONSIDERATIONS
The current therapeutic approach involves three treatment options: (i) pre- and/
or post-operative chemotherapy, (ii) tumorectomy with possible partial liver
resection, and (iii) liver transplantation (29). The use of chemotherapies, includ-
ing platinum compounds for neoadjuvant and adjuvant treatment of hepatoblas-
tomas resulted in a significant improvement in outcome. It has been shown that
the results concerning patient survival are relatively similar between the three
treatment options, even though the appropriate regimens of the various pediatric
oncological groups around the world are not identical (5). Regardless of this fact,
surgical treatment of hepatoblastoma is of great importance. The aim of the surgi-
cal procedure, which aims to be curative, is complete tumor resection. Thanks to
neoadjuvant chemotherapy and various improvements in surgical techniques and
equipment (vascular exclusion, ultrasound knife, etc.), this goal has been achieved
more and more frequently in recent years; unfortunately, in about 10% of
PRETEXT (“PRETreatment EXTension of disease”) IV children, despite aggressive
neoadjuvant chemotherapy, hepatoblastoma is not completely resectable.
Orthotopic liver transplant must be considered in this setting (45).
Staging
According to the generally accepted view, precise clinical staging is a prerequisite
for accurate risk stratification and adequate therapy planning. The so-called
PRETEXT system of the liver tumor study group of the International Society for
Pediatric Oncology (SIOPEL) is used internationally for this purpose. The
PRETEXT system categorizes tumors according to their extent in pre-therapeutic
imaging and has high prognostic relevance (46). Hepatoblastomas are divided
into four different PRETEXT groups (I-IV), depending on how many of the liver’s
four surgical sectors are affected (Table 1).
Also, other characteristics of the extent of the tumor are recorded in capital
letters: C, infiltration of the caudate lobe; V, invasion of the hepatic veins; P, inva-
sion of the portal vein; F, multifocality; E, extrahepatic tumor extension; N, lymph
node involvement; and M, hematogenous metastasis. To describe the extent of the
152 Hager J and Sergi C M
PRETEXT Definition
I Three contiguous hepatic sections are free of tumor
II One or two sections have tumor involvement, but two adjoining sections are tumor-free
III Two or three sections have tumor involvement, but no two adjoining sections are
tumor-free
IV All four sections showing tumor involvement
Stage Definition
I Hepatoblastoma completely resected
II Evidence of microscopic residual neoplastic disease only
III Evidence of gross residual neoplastic disease or positive lymph nodes or “tumor spillage”
IV Evidence of metastatic disease
Source: https://www.childrensoncologygroup.org/index.php/newly-diagnosed-with-hepatoblastoma-or-hepatocellular-
carcinoma-[accessed on 04 February 2021]
tumor after two cycles of neoadjuvant chemotherapy, the same system is then
used for the new staging as the so-called POSTTEXT system (“post-treatment
extension”) (47). It should be noted that the PRETEXT system is accused of being
an almost “overly precise” stage assessment, which is why stages III and IV should
be checked particularly carefully, taking into account the PHITT criteria (48).
An additional classification about the risk assessment of hepatoblastoma was
proposed by the Children’s Hepatic Tumors International Collaboration -
Hepatoblastoma Stratification (CHIC-HS), taking into account the PHITT
consideration. This proposal, which is based on the PRETEXT system and includes
the patient’s age and AFP level, classifies hepatoblastomas into groups as very
low-, low-, medium-, and high-risk (4). Alternatively, hepatoblastomas can be
classified according to the Children’s Cancer Study Group of the Children’s
Oncology Group (Table 2). Hepatoblastomas are not staged using the American
Joint Committee on Cancer criteria (49).
Surgical approach
In the conventional surgical approach, a slightly arched, transverse upper abdom-
inal laparotomy is usually chosen, which can be extended in the linea alba in a
T-shape to the xiphoid if necessary. The liver is fully mobilized, the hepatoduode-
nal ligament and the inferior vena cava located. They are looped under and above
the liver. After the associated supplying and draining vessels have been removed,
the incriminated liver segment is resected, partly blunt, partly with an ultrasound
knife or LigaSure™. All crossing vessels are ligated. It should be noted that ana-
tomical resections such as segment resection, lobectomy or extended lobectomy
(tri-segment resection) are to be preferred to atypical (“wedge”) resections or enu-
cleations, as they usually allow more radical resections and fewer complications.
If the distance to the tumor is large enough, a bleeding-free resection can be made
possible with thorough mattress sutures. There are various discussions about
whether the liver should be pinched out during such an operation. It has been
suggested that some surgical techniques are also critical for postoperative liver
function (64). Avoiding clamping the liver for resection has the advantage that
postoperative function regenerates faster (64). In difficult cases, preoperative
imaging of the arteries supplying the tumor is recommended to evaluate the
tumor supply better. As a primary therapeutic consideration, it is possible in this
context to apply particles loaded with a chemotherapeutic directly into the tumor
(65, 66). As a result, the tumor blood flow can be vastly reduced, and tumor
tissue destruction can be achieved. However, this procedure can lead to severe
complications (67). It is possible to remove up to 80% of the liver tissue through the
operation since the liver can regenerate from the remaining tissue. Because the liver
plays a vital role in producing various proteins that are important for the body,
multiple disorders can temporarily occur after the operation due to tissue loss, for
example, blood clotting disorders, disorders of blood sugar regulation, or a lack
of plasma proteins. In the last few years, liver surgery has seen an incredible
boom, including in children. On one hand this is because of surgical-technical
innovations (for example, minimally invasive partial liver resection) and on the
other hand, because of advances in imaging technologies such as image-guided
three-dimensional reconstructions, intraoperative ultrasound (IOUS), and
indocyanine green (ICG) angiography to detect metastasis (68).
A laparoscopic partial liver resection, even of larger sections, is now a wide-
spread method in adults, especially since various technical aids (for example,
B-mode and Doppler ultrasonography) have been developed for this. According
to the literature, hepatoblastomas can be removed in children in a minimally
Hepatoblastoma 155
invasive manner, but the problem is the abdominal cavity’s size (65, 66). Although
neoadjuvant chemotherapy makes most hepatoblastomas significantly smaller
and thus safely operable, there are no clear international guidelines for laparo-
scopic approach (69). A method that has been known for a long time and is also
suitable for making hepatoblastoma visible is the three-dimensional tumor recon-
struction based on CT data (68, 69). It is crucial because it enables the tumor and
relevant surrounding anatomical structures to be displayed selectively, thanks to
new software and a virtual operation simulation. The three-dimensional recon-
struction provides information about the neoplasm, its topography, whether a
blood vessel has been infiltrated, and the extent of the infiltration (70, 71).
With IOUS, up to 20% of the patients could have morphologically different
results compared to the preoperative MRI examination results, which make it
necessary to change the surgical procedure in individual cases. These changes in
results mainly concern the relationship between hepatoblastomas and hepatic
veins, which is problematic, concerning imaging (72). Apart from the IOUS, an
operating microscope and fluorescent dyes, which accumulate in the tumor and
make it visible under the operating microscope with special filters, are usually
used for tumor and tumor border imaging. ICG has achieved particular impor-
tance for this. It is a fluorescent, colored, water-soluble compound suitable for
various human medical examinations. It has a high binding affinity for all plasma
proteins. ICG absorbs and fluoresces in the visible and near-infrared light spec-
trum (73). ICG allows the monitoring of liver perfusion. Healthy liver tissue
excretes the preparation via the bile within a few hours. In contrast, ICG is retained
in the tumor tissue (29) and therefore ideal for detecting metastases. ICG is usu-
ally administered intravenously 48–72 hours preoperatively to achieve its visual-
ization in the liver. This procedure is also used in hepatoblastoma patients as it
allows the resection margins to be assessed clearly and enable the identification of
residual tumors (74). An essential criterion in the surgical removal of hepatoblas-
toma is, as in all operations, the avoidance of complications (secondary bleeding
and biliary leaks [bilioma formation or occurrence of bilious peritonitis]). During
this phase, growth factors that are increasingly formed in connection with the
surgical trauma can develop a tumor-promoting effect (75). Postoperative chemo-
therapy usually consists of 1–2 courses after liver resection, and twice after liver
transplantation. There are new considerations for reducing ototoxic preparations
as much as possible (76).
Liver transplantation
Liver transplantation is provided for cases of hepatoblastomas that are nonresect-
able: (i) multifocal hepatoblastomas across all four sectors (PRETEXT IV), since
chemotherapy is unlikely to completely eradicate all intrahepatic metastases;
(ii) Central PRETEXT IV hepatoblastomas with vascular invasion, in which the
neoadjuvant chemotherapy cannot downstage the tumor to a PRETEXT III;
(iii) Hepatoblastomas (PRETEXT III), which tightly surround or enclose large
vessels (vena cava inferior, hepatic veins), and (iv) hepatoblastomas that do not
respond to chemotherapy. Also, critical tumor resections may be performed using
the heart-lung machine or ex-situ resection. In some settings, liver transplantation
may produce better long-term results than resection alone from an oncological
point of view (77, 78). If tumor resection cannot be carried out, a so-called
156 Hager J and Sergi C M
“rescue” transplant can be performed, but the prognosis is worse than a primary
liver transplant (79). Otte et al. found that orthotopic (split) liver transplant to
treat hepatoblastomas achieved a 6-year survival rate of 82% in 106 patients,
whereas of the 41 patients who underwent “rescue” liver transplantation, it was
only 30% (80). It should be noted that the prognosis of children who have had a
hepatectomy with orthotopic liver transplantation for hepatoblastoma is as good
as that of children who had conventional resection of smaller tumors (81).
However, it must be noted that liver transplantation is not free from comorbidities
and requires lifelong immunosuppression, which in turn implies side effects.
Against a transplant, it is argued that microscopic residues after a tumorectomy do
not reduce the survival rate of those affected (79). It is expected that the PHITT
study will provide further information (82). Opinions differ on the importance of
post-transplant chemotherapy. Otte et al. compared the relevant results in 147
patients in 2004: 65 received post-transplant chemotherapy, 82 did not. The sur-
vival rates of 77% versus 70% were not statistically significant (80). This means
that post-transplant chemotherapy’s benefit must be weighed against the toxic
risks of the treatment, even though a transplanted liver can withstand adjuvant
chemotherapy (79).
For a liver transplant to treat hepatoblastoma, a living donation is best, from
example from a parent. Until the first year of life, due to the immaturity of the
immune system and immunosuppression, such a transplant can be manageable
without risk for the affected child and during the second year of life with a man-
ageable risk, unlike, for example, blood group compatibility (83). After four
weeks, patients with liver transplants can be switched from tacrolimus to siroli-
mus after completion of chemotherapy, or due to the wound healing disorders
associated with these drugs.
FOLLOW-UP CARE
All children with a treated malignant liver tumor require a follow-up period of at
least 5 years after remission. Regular, initially monthly, later 3-monthly, and then
6-monthly check-ups including liver sonography, chest X-ray, and if necessary, CT
and/or MRI (in the event of an increase in AFP) and laboratory values to rule out
tumor recurrence and to assess the long-term effects of therapy are necessary. In
this context, AFP is of great importance as a tumor marker and thus as an indirect
indicator of therapeutic effectiveness. Normalization of the AFP values can be
expected during the neoadjuvant chemotherapy, and after the removal of hepato-
blastoma. If the AFP values do not normalize, a residual tumor can be assumed. If
they have been regular and then rise, the tumor is likely relapsed (92, 93); how-
ever, some observations show that relapse does not have to be accompanied by an
increase in AFP (94). Chemotherapy-induced changes to cardiac and renal func-
tions, changes in blood parameters, and hearing should be monitored. Attention
must also be paid to the development of a second malignancy.
Treatment of relapses
According to the literature, about 12% of hepatoblastoma patients who have
achieved a complete remission are likely to relapse in the liver and/or lungs. To
achieve remission, chemotherapy (95) and surgical removal of the local recur-
rence or the newly occurring lung metastases is necessary (96). Matsunaga et al.
stated in 2003 that, of the 90 patients (without metastases) in whom the hepato-
blastoma had been fully resected, four had a liver recurrence and eight had lung
metastases. Except for one case with multiple lung metastases, all achieved remis-
sion via medicinal or surgical treatment (97). In liver transplant patients it is less
favorable if additional metastases had initially existed. For example, in 2014,
Yamada et al. reported that about 30% of such the cases relapse (98).
158 Hager J and Sergi C M
CONCLUSION
Hepatoblastoma typically occurs in infants and toddlers and its etiology has not
yet been clarified. Hepatoblastoma is a challenging diagnosis for clinicians and
pathologists. About 20% of children with hepatoblastoma have lung metastases at
the time of tumor diagnosis. Several histological patterns have been associated
with different prognosis. The current therapeutic approach involves chemother-
apy, tumorectomy with possible partial liver dissection, and liver transplantation.
Chemotherapy-induced changes to vital functions should be monitored. About
12% of hepatoblastoma patients who have achieved a complete remission are
likely to relapse. Regular, follow-up is necessary to monitor long-term effects of
therapy.
Acknowledgment: This research has been funded by the generosity of the Stollery
Children’s Hospital Foundation and supporters of the Lois Hole Hospital for
Women through the Women and Children’s Health Research Institute (WCHRI,
Grant ID #: 2096), Hubei Province Natural Science Funding for Hubei University
of Technology (100-Talent Grant for Recruitment Program of Foreign Experts
Total Funding: Digital PCR and NGS-based diagnosis for infection and oncology,
2017–2022), Österreichische Krebshilfe Tyrol (Krebsgesellschaft Tirol, Tyrolean
Cancer Research Institute, Austria, 2007 and 2009 - “DMBTI and cholangiocel-
lular carcinomas” and “Hsp70 and HSPBP1 in carcinomas of the pancreas”),
Austrian Research Fund (Fonds zur Förderung der wissenschaftlichen Forschung,
FWF, Grant ID L313-B13), Canadian Foundation for Women’s Health (“Early
Fetal Heart-RES0000928”), Cancer Research Society (von Willebrand factor gene
expression in cancer cells), Canadian Institutes of Health Research (Omega-3
Hepatoblastoma 159
Copyright and Permission Statement: The authors confirm that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Ng K, Mogul DB. Pediatric Liver Tumors. Clin Liver Dis. 2018;22(4):753–72. https://doi.org/10.1016/j.
cld.2018.06.008
2. Devi LP, Kumar R, Handique A, Kumar M. Hepatoblastoma--a rare liver tumor with review of litera-
ture. J Gastrointest Cancer. 2014;45 Suppl 1:261–4. https://doi.org/10.1007/s12029-014-9659-y
3. von Schweinitz D. Management of liver tumors in childhood. Semin Pediatr Surg. 2006;15(1):17–24.
https://doi.org/10.1053/j.sempedsurg.2005.11.004
4. Meyers RL, Maibach R, Hiyama E, Haberle B, Krailo M, Rangaswami A, et al. Risk-stratified staging
in paediatric hepatoblastoma: a unified analysis from the Children’s Hepatic tumors International
Collaboration. Lancet Oncol. 2017;18(1):122–31. https://doi.org/10.1016/S1470-2045(16)30598-8
5. Aronson DC, Meyers RL. Malignant tumors of the liver in children. Semin Pediatr Surg.
2016;25(5):265–75. https://doi.org/10.1053/j.sempedsurg.2016.09.002
6. Turcotte LM, Spector LG. What do we know about the etiology of hepatoblastoma? Hepat Oncol.
2014;1(1):7–10. https://doi.org/10.2217/hep.13.2
7. Ikeda H, Matsuyama S, Tanimura M. Association between hepatoblastoma and very low birth weight:
a trend or a chance? J Pediatr. 1997;130(4):557–60. https://doi.org/10.1016/S0022-3476(97)70239-7
8. Hubbard AK, Spector LG, Fortuna G, Marcotte EL, Poynter JN. Trends in International Incidence
of Pediatric Cancers in Children Under 5 Years of Age: 1988–2012. JNCI Cancer Spectr.
2019;3(1):pkz007, https://doi.org/10.1093/jncics/pkz007
9. Schraw JM, Desrosiers TA, Nembhard WN, Langlois PH, Meyer RE, Canfield MA, et al. Cancer diag-
nostic profile in children with structural birth defects: An assessment in 15,000 childhood cancer
cases. Cancer. 2020;126(15):3483–92. https://doi.org/10.1002/cncr.32982
10. Heaton TE, Walsh MF. Hepatoblastoma in infants born to parents with familial adenomatous pol-
yposis. In: Guillem J, Friedman G, editors. Management of hereditary colorectal cancer A multidisci-
plinary approach. Springer; 2020. p. 123–36. https://doi.org/10.1007/978-3-030-26234-1_7
11. Cohen MM, Jr. Beckwith-Wiedemann syndrome: historical, clinicopathological, and etiopathogenetic
perspectives. Pediatr Dev Pathol. 2005;8(3):287–304. https://doi.org/10.1007/s10024-005-1154-9
12. Cereda A, Carey JC. The trisomy 18 syndrome. Orphanet J Rare Dis. 2012;7:81. https://doi.
org/10.1186/1750-1172-7-81
13. Meyers RL, Aronson DC, von Schweinitz D, Zimmermann A, Malogolowkin MH. Pediatric liver
tumors. In: Pizzo PA, Poplack DG, editors. Principles and practice of pediatric oncology. 6th ed.
Philadelphia: Lippincott Williams and Wilkins; 2011. p. 838–60.
14. Johnson KJ, Williams KS, Ross JA, Krailo MD, Tomlinson GE, Malogolowkin MH, et al. Parental
tobacco and alcohol use and risk of hepatoblastoma in offspring: a report from the children’s oncol-
ogy group. Cancer Epidemiol Biomarkers Prev. 2013;22(10):1837–43. https://doi.org/10.1158/1055-
9965.EPI-13-0432
160 Hager J and Sergi C M
15. Ishak KG, Glunz PR. Hepatoblastoma and hepatocarcinoma in infancy and childhood.
Report of 47 cases. Cancer. 1967;20(3):396–422. https://doi.org/10.1002/1097-0142(1967)20:
3<396::AID-CNCR2820200308>3.0.CO;2-O
16. Haas JE, Feusner JH, Finegold MJ. Small cell undifferentiated histology in hepatoblastoma may
be unfavorable. Cancer. 2001;92(12):3130–4. https://doi.org/10.1002/1097-0142(20011215)92:
12<3130::AID-CNCR10115>3.0.CO;2-#
17. Weinberg AG, Finegold MJ. Primary hepatic tumors of childhood. Hum Pathol. 1983;14(6):512–37.
https://doi.org/10.1016/S0046-8177(83)80005-7
18. Armeanu-Ebinger S, Hoh A, Wenz J, Fuchs J. Targeting EpCAM (CD326) for immunotherapy in
hepatoblastoma. Oncoimmunology. 2013;2(1):e22620. https://doi.org/10.4161/onci.22620
19. Kiruthiga KG, Ramakrishna B, Saha S, Sen S. Histological and immunohistochemical study of hepato-
blastoma: correlation with tumour behaviour and survival. J Gastrointest Oncol. 2018;9(2):326–37.
https://doi.org/10.21037/jgo.2018.01.08
20. Yun WJ, Shin E, Lee K, Jung HY, Kim SH, Park YN, et al. Clinicopathologic implication of hepatic
progenitor cell marker expression in hepatoblastoma. Pathol Res Pract. 2013;209(9):568–73. https://
doi.org/10.1016/j.prp.2013.06.015
21. Zen Y, Vara R, Portmann B, Hadzic N. Childhood hepatocellular carcinoma: a clinicopathological
study of 12 cases with special reference to EpCAM. Histopathology. 2014;64(5):671–82. https://doi.
org/10.1111/his.12312
22. Ward SC, Thung SN, Lim KH, Tran TT, Hong TK, Hoang PL, et al. Hepatic progeni-
tor cells in liver cancers from Asian children. Liver Int. 2010;30(1):102–11. https://doi.org/
10.1111/j.1478-3231.2009.02126.x
23. Wu JF, Ho MC, Ni YH, Hsu HY, Lee PH, Chang MH. Dysregulation of liver developmental microRNA
contribute to hepatic carcinogenesis. J Formos Med Assoc. 2019;119(6):1041–51. https://doi.
org/10.1016/j.jfma.2019.09.018
24. Ward SC, Waxman S. Fibrolamellar carcinoma: a review with focus on genetics and compari-
son to other malignant primary liver tumors. Semin Liver Dis. 2011;31(1):61–70. https://doi.
org/10.1055/s-0031-1272835
25. Spector LG, Birch J. The epidemiology of hepatoblastoma. Pediatr Blood Cancer. 2012;59(5):776–9.
https://doi.org/10.1002/pbc.24215
26. Pateva IB, Egler RA, Stearns DS. Hepatoblastoma in an 11-year-old: Case report and a review of the lit-
erature. Medicine (Baltimore). 2017;96(2):e5858. https://doi.org/10.1097/MD.0000000000005858
27. Vishnoi JR, Sasidhar A, Misra S, Pareek P, Khera S, Kumar S, et al. Hepatoblastoma in a Young Adult:
a Rare Case Report and Review of the Literature. J Gastrointest Cancer. 2020;51(1):319–24. https://
doi.org/10.1007/s12029-019-00256-3
28. Tulla M, Berthold F, Graf N, Rutkowski S, von Schweinitz D, Spix C, et al. Incidence, Trends, and
Survival of Children With Embryonal Tumors. Pediatrics. 2015;136(3):e623–32. https://doi.
org/10.1542/peds.2015-0224
29. Yang T, Whitlock RS, Vasudevan SA. Surgical Management of Hepatoblastoma and Recent Advances.
Cancers (Basel). 2019;11(12).1944. https://doi.org/10.3390/cancers11121944
30. Dasgupta P, Henshaw C, Youlden DR, Aitken JF, Sullivan A, Irving H, et al. Global trends in incidence
rates of childhood liver cancers: A systematic review and meta-analysis. Paediatr Perinat Epidemiol.
2020;34(5):609–17. https://doi.org/10.1111/ppe.12671
31. Feng J, Polychronidis G, Heger U, Frongia G, Mehrabi A, Hoffmann K. Incidence trends and sur-
vival prediction of hepatoblastoma in children: a population-based study. Cancer Commun (Lond).
2019;39(1):62. https://doi.org/10.1186/s40880-019-0411-7
32. von Schweinitz D. Hepatoblastoma: recent developments in research and treatment. Semin Pediatr
Surg. 2012;21(1):21–30. https://doi.org/10.1053/j.sempedsurg.2011.10.011
33. Gatta G, Ferrari A, Stiller CA, Pastore G, Bisogno G, Trama A, et al. Embryonal cancers in Europe. Eur
J Cancer. 2012;48(10):1425–33. https://doi.org/10.1016/j.ejca.2011.12.027
34. Towbin AJ, Braojos Braga FDC, Zhang B, Geller JI, Tiao GM, Podberesky DJ. Fractures in children
with newly diagnosed hepatoblastoma. Pediatr Radiol. 2018;48(4):581–5. https://doi.org/10.1007/
s00247-017-4050-3
Hepatoblastoma 161
35. Lai M, Burjonrappa S. Perinatal hemorrhage complicating neonatal hepatoblastoma: case report.
J Pediatr Surg. 2012;47(10):e29–32. https://doi.org/10.1016/j.jpedsurg.2012.05.030
36. Yhoshu E, Lone YA, Mahajan JK, Singh UB. Hepatoblastoma with Precocious Puberty. J Indian Assoc
Pediatr Surg. 2019;24(1):68–71. https://doi.org/10.4103/jiaps.JIAPS_102_18
37. Style CC, Cruz SM, Lau PE, Lee TC, Wesson DE, Olutoye OO. Surgical Outcomes of Patients with
Beckwith-Wiedemann Syndrome. J Pediatr Surg. 2018;53(5):1042–5. https://doi.org/10.1016/j.
jpedsurg.2018.02.041
38. De Ioris M, Brugieres L, Zimmermann A, Keeling J, Brock P, Maibach R, et al. Hepatoblastoma
with a low serum alpha-fetoprotein level at diagnosis: the SIOPEL group experience. Eur J Cancer.
2008;44(4):545–50. https://doi.org/10.1016/j.ejca.2007.11.022
39. Hiyama E. Pediatric hepatoblastoma: diagnosis and treatment. Transl Pediatr. 2014;3(4):293–9.
https://doi.org/10.3978/j.issn.2224-4336.2014.09.01
40. Barr RG. Contrast enhanced ultrasound for focal liver lesions: how accurate is it? Abdom Radiol (NY).
2018;43(5):1128–33. https://doi.org/10.1007/s00261-017-1257-6
41. Pugmire BS, Towbin AJ. Magnetic resonance imaging of primary pediatric liver tumors. Pediatr
Radiol. 2016;46(6):764–77. https://doi.org/10.1007/s00247-016-3612-0
42. Derlin T, Hartung D, Hueper K. 18F-FDG PET/CT for Molecular Imaging of Hepatoblastoma in
Beckwith-Wiedemann Syndrome. Clin Nucl Med. 2018;43(5):e164-e5. https://doi.org/10.1097/
RLU.0000000000002040
43. Subramanian KS, Chikhale M, Barwad A, Gochhait D, Toi PC, Siddaraju N. Fine-needle aspira-
tion cytology and cell block in diagnosis and subtyping of hepatoblastoma. Diagn Cytopathol.
2019;47(7):706–10. https://doi.org/10.1002/dc.24167
44. Khanna G. Pediatric hepatoblastoma and hepatocellular carcinoma: lessons learned in the last decade.
Pediatr Radiol. 2018;48(4):531–2. https://doi.org/10.1007/s00247-018-4085-0
45. Lim IIP, Bondoc AJ, Geller JI, Tiao GM. Hepatoblastoma-The Evolution of Biology, Surgery, and
Transplantation. Children (Basel). 2018;6(1).1. https://doi.org/10.3390/children6010001
46. Aronson DC, Schnater JM, Staalman CR, Weverling GJ, Plaschkes J, Perilongo G, et al. Predictive value
of the pretreatment extent of disease system in hepatoblastoma: results from the International Society
of Pediatric Oncology Liver Tumor Study Group SIOPEL-1 study. J Clin Oncol. 2005;23(6):1245–52.
https://doi.org/10.1200/JCO.2005.07.145
47. Meyers RL, Tiao G, de Ville de Goyet J, Superina R, Aronson DC. Hepatoblastoma state of the art:
pre-treatment extent of disease, surgical resection guidelines and the role of liver transplantation. Curr
Opin Pediatr. 2014;26(1):29–36. https://doi.org/10.1097/MOP.0000000000000042
48. Towbin AJ, Meyers RL, Woodley H, Miyazaki O, Weldon CB, Morland B, et al. 2017 PRETEXT: radio-
logic staging system for primary hepatic malignancies of childhood revised for the Paediatric Hepatic
International Tumour Trial (PHITT). Pediatr Radiol. 2018;48(4):536–54. https://doi.org/10.1007/
s00247-018-4078-z
49. AJCC. AJCC Cancer Staging Manual. 8th ed: Springer; 2018.
50. Roebuck DJ, Olsen O, Pariente D. Radiological staging in children with hepatoblastoma. Pediatr
Radiol. 2006;36(3):176–82. https://doi.org/10.1007/s00247-005-0029-6
51. McCarville MB, Roebuck DJ. Diagnosis and staging of hepatoblastoma: imaging aspects. Pediatr Blood
Cancer. 2012;59(5):793–9. https://doi.org/10.1002/pbc.24221
52. El-Ali AM, Davis JC, Cickelli JM, Squires JH. Contrast-enhanced ultrasound of liver lesions in chil-
dren. Pediatr Radiol. 2019;49(11):1422–32. https://doi.org/10.1007/s00247-019-04361-0
53. Chavhan GB, Shelmerdine S, Jhaveri K, Babyn PS. Liver MR Imaging in Children: Current Concepts
and Technique. Radiographics. 2016;36(5):1517–32. https://doi.org/10.1148/rg.2016160017
54. Baheti AD, Chapman T, Rudzinski E, Albert CM, Stanescu AL. Diagnosis, histopathologic cor-
relation and management of hepatoblastoma: What the radiologist needs to know. Clin Imaging.
2018;52:273–9. https://doi.org/10.1016/j.clinimag.2018.08.009
55. Bottcher J, Hansch A, Pfeil A, Schmidt P, Malich A, Schneeweiss A, et al. Detection and classification
of different liver lesions: comparison of Gd-EOB-DTPA-enhanced MRI versus multiphasic spiral CT
in a clinical single centre investigation. Eur J Radiol. 2013;82(11):1860–9. https://doi.org/10.1016/j.
ejrad.2013.06.013
162 Hager J and Sergi C M
56. Warmann SW, Schenk A, Schaefer JF, Ebinger M, Blumenstock G, Tsiflikas I, et al. Computer-assisted
surgery planning in children with complex liver tumors identifies variability of the classical Couinaud
classification. J Pediatr Surg. 2016;51(11):1801–6. https://doi.org/10.1016/j.jpedsurg.2016.05.018
57. Malogolowkin MH, Katzenstein HM, Meyers RL, Krailo MD, Rowland JM, Haas J, et al. Complete
surgical resection is curative for children with hepatoblastoma with pure fetal histology: a report
from the Children’s Oncology Group. J Clin Oncol. 2011;29(24):3301–6. https://doi.org/10.1200/
JCO.2010.29.3837
58. Murphy AJ, Ayers GD, Hilmes MA, Mukherjee K, Wilson KJ, Allen WM, et al. Imaging analysis of
hepatoblastoma resectability across neoadjuvant chemotherapy. J Pediatr Surg. 2013;48(6):1239–48.
https://doi.org/10.1016/j.jpedsurg.2013.03.019
59. Venkatramani R, Stein JE, Sapra A, Genyk Y, Jhaveri V, Malogolowkin M, et al. Effect of neoadjuvant
chemotherapy on resectability of stage III and IV hepatoblastoma. Br J Surg. 2015;102(1):108–13.
https://doi.org/10.1002/bjs.9681
60. Tiao GM, Bobey N, Allen S, Nieves N, Alonso M, Bucuvalas J, Wells R, et al. The current management
of hepatoblastoma: a combination of chemotherapy, conventional resection, and liver transplantation.
J Pediatr. 2005;146(2):204–11. https://doi.org/10.1016/j.jpeds.2004.09.011
61. Fuchs J, Cavdar S, Blumenstock G, Ebinger M, Schafer JF, Sipos B, et al. POST-TEXT III and IV
Hepatoblastoma: Extended Hepatic Resection Avoids Liver Transplantation in Selected Cases. Ann
Surg. 2017;266(2):318–23. https://doi.org/10.1097/SLA.0000000000001936
62. de Freitas Paganoti G, Tannuri ACA, Dantas Marques AC, Torres RR, Mendes Gibelli NE, Tannuri U.
Extensive Hepatectomy as an Alternative to Liver Transplant in Advanced Hepatoblastoma: A New
Protocol Used in a Pediatric Liver Transplantation Center. Transplant Proc. 2019;51(5):1605–10.
https://doi.org/10.1016/j.transproceed.2019.03.004
63. Gentile LF, Islam S. Ruptured hepatoblastoma treated with primary surgical resection. Ann Pediatr
Surg. 2014;10:54–6. https://doi.org/10.1097/01.XPS.0000445134.67605.19
64. Fagenson AM, Gleeson EM, Nabi F, Lau KN, Pitt HA. When does a Pringle Maneuver cause harm?
HPB (Oxford). 2020. https://doi.org/10.1016/j.hpb.2020.07.014
65. Vogl TJ, Naguib NN, Nour-Eldin NE, Bechstein WO, Zeuzem S, Trojan J, et al. Transarterial chemo-
embolization in the treatment of patients with unresectable cholangiocarcinoma: Results and prognos-
tic factors governing treatment success. Int J Cancer. 2012;131(3):733–40. https://doi.org/10.1002/
ijc.26407
66. Vogl TJ, Scheller A, Jakob U, Zangos S, Ahmed M, Nabil M. Transarterial chemoembolization in the
treatment of hepatoblastoma in children. Eur Radiol. 2006;16(6):1393–6. https://doi.org/10.1007/
s00330-005-2827-5
67. Czauderna P, Zbrzezniak G, Narozanski W, Korzon M, Wyszomirska M, Stoba C. Preliminary experi-
ence with arterial chemoembolization for hepatoblastoma and hepatocellular carcinoma in children.
Pediatr Blood Cancer. 2006;46(7):825–8. https://doi.org/10.1002/pbc.20422
68. Schmidt A, Warmann SW, Urla C, Fuchs J. [Innovations in surgical treatment of pediatric solid
tumors]. Chirurg. 2018;89(3):205–11. https://doi.org/10.1007/s00104-017-0568-z
69. Fuchs J. The role of minimally invasive surgery in pediatric solid tumors. Pediatr Surg Int.
2015;31(3):213–28. https://doi.org/10.1007/s00383-015-3660-9
70. Souzaki R, Kinoshita Y, Ieiri S, Hayashida M, Koga Y, Shirabe K, et al. Three-dimensional liver model
based on preoperative CT images as a tool to assist in surgical planning for hepatoblastoma in a child.
Pediatr Surg Int. 2015;31(6):593–6. https://doi.org/10.1007/s00383-015-3709-9
71. Zhang G, Zhou XJ, Zhu CZ, Dong Q, Su L. Usefulness of three-dimensional(3D) simulation soft-
ware in hepatectomy for pediatric hepatoblastoma. Surg Oncol. 2016;25(3):236–43. https://doi.
org/10.1016/j.suronc.2016.05.023
72. Felsted AE, Shi Y, Masand PM, Nuchtern JG, Goss JA, Vasudevan SA. Intraoperative ultrasound
for liver tumor resection in children. J Surg Res. 2015;198(2):418–23. https://doi.org/10.1016/j.
jss.2015.03.087
73. Landsman ML, Kwant G, Mook GA, Zijlstra WG. Light-absorbing properties, stability, and spec-
tral stabilization of indocyanine green. J Appl Physiol. 1976;40(4):575–83. https://doi.org/10.1152/
jappl.1976.40.4.575
Hepatoblastoma 163
74. Yamada Y, Ohno M, Fujino A, Kanamori Y, Irie R, Yoshioka T, et al. Fluorescence-Guided Surgery
for Hepatoblastoma with Indocyanine Green. Cancers (Basel). 2019;11(8). https://doi.org/10.3390/
cancers11081215
75. Becker K, Furch C, Schmid I, von Schweinitz D, Haberle B. Impact of postoperative complications on
overall survival of patients with hepatoblastoma. Pediatr Blood Cancer. 2015;62(1):24–8. https://doi.
org/10.1002/pbc.25240
76. Katzenstein HM, Langham MR, Malogolowkin MH, Krailo MD, Towbin AJ, McCarville MB, et al.
Minimal adjuvant chemotherapy for children with hepatoblastoma resected at diagnosis (AHEP0731):
a Children’s Oncology Group, multicentre, phase 3 trial. Lancet Oncol. 2019;20(5):719–27. https://
doi.org/10.1016/S1470-2045(18)30895-7
77. Khaderi S, Guiteau J, Cotton RT, O’Mahony C, Rana A, Goss JA. Role of liver transplantation in the
management of hepatoblastoma in the pediatric population. World J Transplant. 2014;4(4):294–8.
https://doi.org/10.5500/wjt.v4.i4.294
78. Feng J, He Y, Wei L, Chen D, Yang H, Tan R, et al. Assessment of Survival of Pediatric Patients With
Hepatoblastoma Who Received Chemotherapy Following Liver Transplant or Liver Resection. JAMA
Netw Open. 2019;2(10):e1912676. https://doi.org/10.1001/jamanetworkopen.2019.12676
79. Stringer MD. The role of liver transplantation in the management of paediatric liver tumours. Ann R
Coll Surg Engl. 2007;89(1):12–21. https://doi.org/10.1308/003588407X155527
80. Otte JB, Pritchard J, Aronson DC, Brown J, Czauderna P, Maibach R, et al. International Society
of Pediatric O. Liver transplantation for hepatoblastoma: results from the International Society of
Pediatric Oncology (SIOP) study SIOPEL-1 and review of the world experience. Pediatr Blood Cancer.
2004;42(1):74–83. https://doi.org/10.1002/pbc.10376
81. Sindhi R, Rohan V, Bukowinski A, Tadros S, de Ville de Goyet J, Rapkin L, Ranganathan S. Liver
Transplantation for Pediatric Liver Cancer. Cancers (Basel). 2020;12(3).720. https://doi.org/10.3390/
cancers12030720
82. Czauderna P, Garnier H. Hepatoblastoma: current understanding, recent advances, and controversies.
F1000Res. 2018;7:53. https://doi.org/10.12688/f1000research.12239.1
83. Sakamoto S, Kasahara M, Mizuta K, Kuroda T, Yagi T, Taguchi T, et al. Japanese Liver Transplantation
S. Nationwide survey of the outcomes of living donor liver transplantation for hepatoblastoma in
Japan. Liver Transpl. 2014;20(3):333–46. https://doi.org/10.1002/lt.23803
84. Angelico R, Grimaldi C, Gazia C, Saffioti MC, Manzia TM, et al. How Do Synchronous Lung
Metastases Influence the Surgical Management of Children with Hepatoblastoma? An Update and
Systematic Review of the Literature. Cancers (Basel). 2019;11(11).1693. https://doi.org/10.3390/
cancers11111693
85. Wanaguru D, Shun A, Price N, Karpelowsky J. Outcomes of pulmonary metastases in hepatoblas-
toma--is the prognosis always poor? J Pediatr Surg. 2013;48(12):2474–8. https://doi.org/10.1016/j.
jpedsurg.2013.08.023
86. Lake CM, Tiao GM, Bondoc AJ. Surgical management of locally-advanced and metastatic hepatoblas-
toma. Semin Pediatr Surg. 2019;28(6):150856. https://doi.org/10.1016/j.sempedsurg.2019.150856
87. Meyers RL, Katzenstein HM, Krailo M, McGahren ED 3rd, Malogolowkin MH. Surgical resection of
pulmonary metastatic lesions in children with hepatoblastoma. J Pediatr Surg. 2007;42(12):2050–6.
https://doi.org/10.1016/j.jpedsurg.2007.08.030
88. Kitagawa N, Shinkai M, Mochizuki K, Usui H, Miyagi H, Nakamura K, et al. Navigation using indo-
cyanine green fluorescence imaging for hepatoblastoma pulmonary metastases surgery. Pediatr Surg
Int. 2015;31(4):407–11. https://doi.org/10.1007/s00383-015-3679-y
89. Souzaki R, Kawakubo N, Matsuura T, Yoshimaru K, Koga Y, Takemoto J, et al. Navigation sur-
gery using indocyanine green fluorescent imaging for hepatoblastoma patients. Pediatr Surg Int.
2019;(35):551–7. https://doi.org/10.1007/s00383-019-04458-5
90. Fuchs J, Seitz G, Ellerkamp V, Dietz K, Bosk A, Muller I, et al. Analysis of sternotomy as treatment
option for the resection of bilateral pulmonary metastases in pediatric solid tumors. Surg Oncol.
2008;17(4):323–30. https://doi.org/10.1016/j.suronc.2008.05.004
91. Heaton TE, Davidoff AM. Surgical treatment of pulmonary metastases in pediatric solid tumors.
Semin Pediatr Surg. 2016;25(5):311–7. https://doi.org/10.1053/j.sempedsurg.2016.09.001
164 Hager J and Sergi C M
92. Fuchs J, Rydzynski J, Von Schweinitz D, Bode U, Hecker H, Weinel P, et al. Study Committee of the
Cooperative Pediatric Liver Tumor Study Hb 94 for the German Society for Pediatric O, Hematology.
Pretreatment prognostic factors and treatment results in children with hepatoblastoma: a report from
the German Cooperative Pediatric Liver Tumor Study HB 94. Cancer. 2002;95(1):172–82. https://doi.
org/10.1002/cncr.10632
93. Yang W, Chen Y, Huang Y, Wang H. Analysis of factors related to recurrence of paediatric hepatoblas-
toma - a single Centre retrospective study. BMC Pediatr. 2019;19(1):485. https://doi.org/10.1186/
s12887-019-1870-3
94. Kachanov DY, Aliev TZ, Moiseenko RA, Roschin VY, Metelin AV, Uskova NG, et al. Hepatoblastoma
relapses with a normal level of alpha-fetoprotein: report of two cases. Pediatric Hematology/Oncology
and Immunopathology. 2019;18(4):58–65. https://doi.org/10.24287/1726-1708-2019-18-4-58-65
95. Natarajan E, Auerbach C, Cheron R, Pashankar F. Sustained Remission After Maintenance Irinotecan
in Patient With Multiply Relapsed Hepatoblastoma. J Pediatr Hematol Oncol. 2020;42(7):e659-e61.
https://doi.org/10.1097/MPH.0000000000001544
96. Semeraro M, Branchereau S, Maibach R, Zsiros J, Casanova M, Brock P, et al. Relapses in hepato-
blastoma patients: clinical characteristics and outcome--experience of the International Childhood
Liver Tumour Strategy Group (SIOPEL). Eur J Cancer. 2013;49(4):915–22. https://doi.org/10.1016/j.
ejca.2012.10.003
97. Matsunaga T, Sasaki F, Ohira M, Hashizume K, Hayashi A, Hayashi Y, et al. Japanese Study Group for
Pediatric Liver T. Analysis of treatment outcome for children with recurrent or metastatic hepatoblas-
toma. Pediatr Surg Int. 2003;19(3):142–6. https://doi.org/10.1007/s00383-002-0906-0
98. Yamada Y, Hoshino K, Mori T, Kawaida M, Abe K, Takahashi N, et al. Metastasectomy of
Hepatoblastoma Utilizing a Novel Overlay Fluorescence Imaging System. J Laparoendosc Adv Surg
Tech A. 2018;28(9):1152–5. https://doi.org/10.1089/lap.2017.0735
99. Takahashi N, Yamada Y, Hoshino K, Kawaida M, Mori T, Abe K, et al. Living Donor Liver
Re-Transplantation for Recurrent Hepatoblastoma in the Liver Graft following Complete Eradication
of Peritoneal Metastases under Indocyanine Green Fluorescence Imaging. Cancers (Basel).
2019;11(5).730. https://doi.org/10.3390/cancers11050730
100. Shi Y, Geller JI, Ma IT, Chavan RS, Masand PM, Towbin AJ, et al. Relapsed hepatoblastoma confined
to the lung is effectively treated with pulmonary metastasectomy. J Pediatr Surg. 2016;51(4):525–9.
https://doi.org/10.1016/j.jpedsurg.2015.10.053
101. Passmore SJ, Noblett HR, Wisheart JD, Mott MG. Prolonged survival following multiple thoracoto-
mies for metastatic hepatoblastoma. Med Pediatr Oncol. 1995;24(1):58–60. https://doi.org/10.1002/
mpo.2950240113
102. Yevich S, Calandri M, Gravel G, Fresneau B, Brugieres L, Valteau-Couanet D, et al. Reiterative
Radiofrequency Ablation in the Management of Pediatric Patients with Hepatoblastoma Metastases
to the Lung, Liver, or Bone. Cardiovasc Intervent Radiol. 2019;42(1):41–7. https://doi.org/10.1007/
s00270-018-2097-7
103. Zhang YT, Chang J, Yao YM, Li YN, Zhong XD, Liu ZL. Novel treatment of refractory / recurrent
pulmonary hepatoblastoma. Pediatr Int. 2020;62(3):324–9. https://doi.org/10.1111/ped.14134
9
Undifferentiated Embryonal Sarcoma
of the Liver in Adults
Jingyang Huang
Department of Pathology and Laboratory Medicine, University of Alberta,
Edmonton, Canada
Author for Correspondence: Jingyang Huang, Department of Pathology and
Laboratory Medicine, University of Alberta, Edmonton, Canada.
E-mail: [email protected]
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.ch9
Keywords: diagnosis of exclusion; liver tumor; pediatric liver sarcoma; rare adult
hepatic sarcoma; undifferentiated embryonal sarcoma of the liver
In: Liver Cancer. Sergi CM. (Editor). Exon Publications, Brisbane, Australia.
ISBN: 978-0-6450017-2-3; Doi: https://doi.org/10.36255/exonpublications.livercancer.2021
Copyright: The Authors.
License: This open access article is licenced under Creative Commons Attribution-NonCommercial
4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/
165
166 Huang J
INTRODUCTION
CLINICAL PRESENTATION
UESL is a rare liver mesenchymal neoplasm that most often occurs in children
between 6 and 10 years of age (1). UESL is the third most common primary liver
malignancy in children after hepatoblastoma and hepatocellular carcinoma (1, 7).
This tumor has been reported in adults, but these cases are exceptionally rare (3).
UESL most commonly occurs in the right lobe of the liver with many patients
being asymptomatic for considerable periods of time (3). Those patients who are
symptomatic may present with non-specific features such as nausea, vomiting,
weight loss, fever, and abdominal pain and distention (1, 2). Serology in most
patients is usually negative but in a minority of cases, elevated liver enzymes, alpha
fetoprotein, and erythrocyte sedimentation rates have been encountered (1, 2).
RADIOLOGY
Imaging will demonstrate a heterogeneous liver mass with solid and cystic
areas (8). There are usually no specific imaging features that can help suggest an
undifferentiated embryonal sarcoma (2). On ultrasound, the tumor may appear
more solid than cystic because of its high water content (7). On CT, UESL will be
a hypodense solid and cystic mass (Figure 1A) (2). On MRI, the cysts within the
mass may show multiple fluid-fluid levels (Figure1B) (9). Due to their cystic char-
acter radiologically, UESLs have been known to be mistaken for benign cystic
entities such as biliary cystadenomas, hydatid cyst disease, or venolymphatic mal-
formations (2, 8, 9). In fact, the radiological appearance of USEL can be quite
misleading to a point where delays in diagnosis or even the initiation of the wrong
treatment have occurred (4, 10).
Undifferentiated Embryonal Sarcoma of the Liver 167
Figure 1. Radiology of undifferentiated embryonal sarcoma of the liver (UESL). A. An arterial
phase contrast-enhanced computed tomography from a young adult male with UESL. It
demonstrates a large hypodense mass in the right lobe of the liver. B. Magnetic resonance
image showing a large mass in right lobe of the liver with heterogenous and cystic features
along with fluid-fluid levels.
GROSS FEATURES
Hepatic resection specimens show masses most commonly located in the right
lobe of the liver. Many of these tumors are quite large by the time surgical interven-
tion is undertaken. The tumor commonly has a pseudocapsule and due to its large
size, compresses the adjacent normal liver parenchyma. The tumor itself has a
variegated appearance showing cystic and solid areas that have gelatinous, hemor-
rhagic, and/or necrotic features (Figure 2) (2, 3, 7). Tumors that have been large
enough to rupture through the liver capsule have also been encountered (11–13).
HISTOLOGY
Figure 2. Gross features of undifferentiated embryonal sarcoma of the liver (USEL). This gross
photograph of a UESL shows a variegated gelatinous liver mass with a lobulated architecture
and areas of necrosis and hemorrhage. This case also shows the tumor rupturing through
the capsule of the liver.
IMMUNOHISTOCHEMISTRY
There is no immunohistochemistry that is specific for UESL. In fact, the key role
of immunohistochemistry is to rule out other possible tumors in the differential
(2–4). Variable expression of actin, vimentin, CD68, BCL-2, desmin, and CD10 is
commonly seen in UESLs (Figures 4A and B). Alpha-1-antitrypsin and
alpha-1-antichymotrypsin positivity within the eosinophilic globules is com-
monly encountered (Figure 4C). (2–4, 7). Weak cytokeratin staining has also been
reported, and a minority of UESLs have positivity for glypican 3 and nuclear posi-
tivity for CD117. As a result, glypican 3 should not be used on its own to rule out
a hepatoblastoma and hepatocellular carcinoma. Rather glypican 3 should be
deployed in a panel of stains and interpreted in conjunction with more reliable
markers such as HepPAR1 and β-catenin. UESLs are usually negative for S100,
MART1, myogenin, ALK-1, Beta-catenin, and HepPar1 (2–4, 7).
ELECTRON MICROSCOPY
Figure 3. Histological features of undifferentiated embryonal sarcoma of the liver. A. High-grade
myxomatous areas with considerable pleomorphism and an atypical mitotic figure.
B. Hemorrhagic area with abundant tumor necrosis. C. Hemorrhagic area with cystic change.
D. Benign bile duct along with cords and nests of normal hepatocytes entrapped at the
periphery of the tumor. Tumor pseudocapsule at the right bottom corner of the image.
E. High-grade myxomatous area with PASD-resistant eosinophilic hyaline globules. F. Tumor
cells with PASD-resistant eosinophilic hyaline globules.
the PASD-resistant eosinophilic globules seen histologically (2, 6, 15, 16). Some
tumors in adults demonstrated myofilaments and intermediate filaments, suggest-
ing smooth muscle differentiation (17). Lipoblastic differentiation has also been
reported due to the presence of fat droplets (2, 15).
170 Huang J
MOLECULAR FINDINGS
UESL is predominately a pediatric tumor, and thus the literature about prognosis
is predominantly based on data from pediatric patients. Historically, this tumor
Undifferentiated Embryonal Sarcoma of the Liver 171
DIFFERENTIAL DIAGNOSIS
melanoma (3, 40, 41). Additional follow-up stains such as SOX10, MART-1,
MITF, and HMB45 are often used in conjunction with S100 in cases that need
further workup. It should be noted that before diagnosing melanoma in the liver,
one should always consider the possibility of a perivascular epithelioid cell tumor
(PECOMA), because PECOMAs can also be positive for S100, MART1, MITF, and
HMB45. As a result, appropriate myoid markers such as smooth muscle actin and
desmin along with detailed examination of the histology should allow for proper
distinction between a PECOMA and a melanoma (42, 43).
OTHER SARCOMAS
Higher-grade tumors can be more difficult to identify and, in such cases, immu-
nohistochemistry will be necessary. Positivity for actin, desmin, and caldesmon is
usually sufficient to rule out other possibilities in the differential (3, 47).
CONCLUSION
UESL is an aggressive malignancy that does occur in adults but is very rare. It
presents clinically most often with non-specific symptoms of an abdominal mass
with imaging features of a solid and cystic liver tumor. UESL is a diagnosis of
exclusion in pathology, meaning that all other differentials must be reasonably
excluded before a diagnosis of UESL can be confidently made. The pathogenesis
and origin of UESL is unclear at this time, but molecular aberrations in p53 and
translocation t(11;19) have been found. In terms of treatment, considerable prog-
ress has been made by implementing a multimodality approach which has led to
much more favorable outcomes for this high-grade liver tumor.
Copyright and permission statement: The author confirms that the materials
included in this chapter do not violate copyright laws. Where relevant, appropri-
ate permissions have been obtained from the original copyright holder(s), and all
original sources have been appropriately acknowledged or referenced.
REFERENCES
1. Shi Y, Rojas Y, Zhang W, Beierle EA, Doski JJ, Goldfarb M, et al. Characteristics and outcomes in
children with undifferentiated embryonal sarcoma of the liver: A report from the National Cancer
Database. Pediatr Blood Cancer. 2017;64(4): e26272. https://doi.org/10.1002/pbc.26272
2. Putra J, Ornvold K. Undifferentiated embryonal sarcoma of the liver: a concise review. Arch Pathol
Lab Med. 2015;139(2):269–73.
3. Pinamonti M, Vittone F, Ghiglione F, Borasi A, Silvestri S, Coverlizza S. Unexpected Liver Embryonal
Sarcoma in the Adult: Diagnosis and Treatment. Case Rep Surg. 2018;2018:8362012. https://doi.
org/10.1155/2018/8362012
4. Zhang H, Lei L, Zuppan CW, Raza AS. Undifferentiated embryonal sarcoma of the liver with an
unusual presentation: case report and review of the literature. J Gastrointest Oncol. 2016;7(Suppl 1):
S100-S6.
5. Gerson G, Valença JT, Cavalcante JM, Coêlho RDAB. Undifferentiated embryonal sarcoma of the liver
in elderly: Case report and review of the literature. Indian J Pathol Microbiol.2019;62(1):129–31.
https://doi.org/10.4103/IJPM.IJPM_508_16
6. Lee KH, Maratovich MN, Lee KB. Undifferentiated embryonal sarcoma of the liver in an adult patient.
Clin Mol Hepatol. 2016;22(2):292–95.
Undifferentiated Embryonal Sarcoma of the Liver 177
7. Silverberg, SG, Delellis RA, Sobin LH. Mesenchymal Tumors and Tumor-like Lesions. AFIP Atlas of
Tumor Pathology Series 4: Tumors of the Liver. American Registry of Pathology, 2018. P. 313–28
8. Kallam A, Krishnamurthy J, Kozel J, Shonka N. Undifferentiated Embryonal Sarcoma of Liver. Rare
Tumors. 2015;7(4):6009.
9. Kamrani K, Patel A, Guerrieri C, Bethel CAI, Phatak T. Undifferentiated embryonal sarcoma of the
liver mimicking venolymphatic malformation. Radiol Case Rep. 2019;14(7):795–99.
10. Gao J, Fei L, Li S, Cui K, Zhang J, Yu F, et al. Undifferentiated embryonal sarcoma of the liver in a child:
A case report and review of the literature. Oncol Lett. 2013;5(3):739–42. https://doi.org/10.3892/
ol.2012.1087
11. Hu MH, Zhang WL, Li J, Wen Y, Li F, Zhi T, Huang DS. Report of seven children with undifferenti-
ated embryonal sarcoma of the liver. Chin Med J. 2019;132(18):2244–5. https://doi.org/10.1097/
CM9.0000000000000429
12. Te-Yu Hung 1, David Lu, Min-Chang Liu. Undifferentiated (embryonal) sarcoma of the liver compli-
cated with rupture in a child. J Pediatr Hematol Oncol. 2007;29(1):63–65. https://doi.org/10.1097/
MPH.0b013e318030ac25
13. Chen JH, Lee CH, Wei CK, Chang SM, Yin, WY. Undifferentiated embryonal sarcoma of the liver
with focal osteoid picture-A case report. Asian J Surg. 2013;36(4):174–8. https://doi.org/10.1016/j.
asjsur.2012.06.012
14. Ishak KG, Goodman ZD, Stocker JT. Malignant Mesenchymal Tumors. In: Rosai J, Sobin LH. Atlas of
Tumor Pathology: Tumors of the Liver and Intrahepatic Bile Ducts. Washington DC: Armed Forces
Institute of Pathology, 2001. p. 313–230.
15. Agaram NP, Baren A, Antonescu CR. Pediatric and adult hepatic embryonal sarcoma: a comparative
ultrastructural study with morphologic correlations. Ultrastruct Pathol. 2006;30(6):403–8. https://
doi.org/10.1080/01913120600854699
16. Keating S, Taylor GP. Undifferentiated (embryonal) sarcoma of the liver: Ultrastructural and immu-
nohistochemical similarities with malignant fibrous histiocytoma. Hum Pathol. 1985;16(7):693–9.
https://doi.org/10.1016/S0046-8177(85)80154-4
17. Nishio J, Iwasaki H, Sakashita N, Haraoka S, Isayama T, Naito M, et al. Undifferentiated (embryonal)
sarcoma of the liver in middle-aged adults: smooth muscle differentiation determined by immunohis-
tochemistry and electron microscopy. Hum Pathol. 2003;34(3):246–52.
18. Mori A, Fukase K, Masuda K, Sakata N, Mizuma M, Ohtsuka H, et al. A case of adult undifferentiated
embryonal sarcoma of the liver successfully treated with right trisectionectomy: a case report. Surg
Case Rep. 2017;3:19. https://doi.org/10.1186/s40792-017-0295-1
19. Hu X, Chen H, Jin M, Wang X, Lee J, Xu W, et al. Molecular cytogenetic characterization of undifferen-
tiated embryonal sarcoma of the liver: a case report and literature review. Mol Cytogenet. 2012;5:26.
https://doi.org/10.1186/1755-8166-5-26
20. Khan ZH, Ilyas K, Khan KH, Ghazanfar H, Hussain A, Inayat G, et al. Unresectable Undifferentiated
Embryonal Sarcoma of the Liver in an Adult Male Treated with Chemotherapy and Orthotopic Liver
Transplantation. Cureus. 2017;9(10): e1759. https://doi.org/10.7759/cureus.1759
21. Siddiqui MA, McKenna BJ. Hepatic mesenchymal hamartoma: a short review. Arch Pathol Lab Med.
2006;130(10):1567–1569.
22. Patel RS, Misra V, Verma K, Gupta P, and Dhingra V. Benign Hepatic Mesenchymal Hamartoma
(HMH) - A Case Report. J Clin Diagn Res. 2014;8(3):119–20. https://doi.org/10.7860/
JCDR/2014/6522.4129
23. Gupta R, Parelkar SV, and Sanghvi B. Mesenchymal hamartoma of the liver. Indian J Med Paediatr
Oncol. 2009;30(4):141–3.
24. Ranganthan S. Liver pathology. New York: Demos Medical; c2011. Chapter 28, Hepatoblastoma;
p.411–56.
25. Khanna R, Verma SK. Pediatric hepatocellular carcinoma. World J Gastroenterol. 2018;24(35):3980–99.
26. Lin CC, Yang HM. Fibrolamellar Carcinoma: A Concise Review. Arch Pathol Lab Med.
2018;142(9):1141–5.
27. Lee KB. Sarcomatoid hepatocellular carcinoma with mixed osteoclast-like giant cells and chondroid
differentiation. Clin Mol Hepatol. 2014;20(3):313–6.
178 Huang J
28. Egas-Bejar D, Huh WW. Rhabdomyosarcoma in adolescent and young adult patients: current perspec-
tives. Adolesc Health Med Ther. 2014;5:115–25.
29. Ognjanovic S, Linabery AM, Charbonneau B, Ross JA. Trends in Childhood Rhabdomyosarcoma
Incidence and Survival in the United States (1975–2005). Cancer. 2009;115(18):4218–26.
30. Parham DM, Barr FG. Classification of Rhabdomyosarcoma and Its Molecular Basis. Adv Anat Pathol.
2013;20(6):387–97. https://doi.org/10.1097/PAP.0b013e3182a92d0d
31. Parham DM, Ellison DA. Rhabdomyosarcomas in Adults and Children: An Update. Arch Pathol Lab
Med. 2006;130(10):1454–65.
32. Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology, molecular pathology,
prognosis, and differential diagnosis. Arch Pathol Lab Med. 2006t;130(10):1466–78.
33. Nagai T, Ueda K, Hakoda H, Okata S, Nakata S, Taira T, et al. Primary gastrointestinal stromal tumor
of the liver: a case report and review of the literature. Surg Case Rep. 2016;2(1):87. https://doi.
org/10.1186/s40792-016-0218-6
34. Laurini JA, Carter JE. Gastrointestinal Stromal Tumors: A Review of the Literature. Arch Pathol Lab
Med. 2010;134(1):134–41. https://doi.org/10.1038/labinvest.2010.17
35. Shi YN, Li Y, Wang LP, Wang ZH, Liang XB, Liang H, et al. Gastrointestinal stromal tumor (GIST)
with liver metastases. Medicine (Baltimore). 2017;96(46): e8240. https://doi.org/10.1097/
MD.0000000000008240
36. Zhao X, Yue C. Gastrointestinal stromal tumor. J Gastrointest Oncol. 2012;3(3):189–208.
37. Lech G, Korcz W, Kowalczyk E, Guzel T, Radoch M, Krasnodębski IW. Giant gastrointestinal stro-
mal tumour of rare sarcomatoid epithelioid subtype: Case study and literature review. World J
Gastroenterol. 2015;21(11):3388–93.
38. Parab TM, DeRogatis MJ, Boaz AM, Grasso SA, Issack PS, Duarte DA, et al. Gastrointestinal stromal
tumors: a comprehensive review. J Gastrointest Oncol. 2019;10(1):144–54.
39. Zhang Y, Hu Z, Wu W, Liu J, Hong D, Zhang C. Partial hepatectomy for primary hepatic melanoma:
a report of two cases and review of the literature. World J Surg Oncol. 2014;12:362. https://doi.
org/10.1186/1477-7819-12-362
40. Husain N, Verma N. Current Concepts in Pathology of Soft Tissue Sarcoma. Indian J Surg Oncol.
2011;2(4):302–8. https://doi.org/10.1007/s13193-012-0134-6
41. Goldblum JR. An approach to pleomorphic sarcomas: can we subclassify, and does it matter? Mod
Pathol. 2014;27 Suppl 1:S39–46. https://doi.org/10.1038/modpathol.2013.174
42. Armah HB, Parwani AV. Perivascular epithelioid cell tumor. Arch Pathol Lab Med.
2009;133(4):648–654.
43. Uddin Z, Qureshi A, Fatima S, Kayani N. PEComa in a 12-year-old boy. BMJ Case Rep. 2010;2010:
bcr04.2009.1735.
44. Matthyssens LE, Creytens D, Ceelen WP. Retroperitoneal Liposarcoma: Current Insights in Diagnosis
and Treatment. Front Surg. 2015;2:4. https://doi.org/10.3389/fsurg.2015.00004
45. Huang IH, Wu YY, Huang TC, Chang WK, Chen JH. Statistics and outlook of primary hepatic angio-
sarcoma based on clinical stage. Oncol Lett. 2016;11(5):3218–22.
46. Sardaro A, Bardoscia L, Petruzzelli MF, Portaluri M. Epithelioid Hemangioendothelioma: An
Overview and Update on a Rare Vascular Tumor. Oncol Rev. 2014;8(2):259. https://doi.org/10.4081/
oncol.2014.259
47. Shivathirthan N, Kita J, Iso Y, Hachiya H, KyungHwa P, Sawada T, et al. Primary hepatic leiomyosar-
coma: Case report and literature review. World J Gastrointest Oncol. 2011;3(10):148–52. https://doi.
org/10.4251/wjgo.v3.i10.148
Index 179
Index
Erlotinib, 10 I
Etiology, 40, 146
Ifosfamide, 10
Everolimus, 10
Imaging, 5, 148
Expanded criteria, 127, 129
Immunohistochemistry, 168
Immunopathogenesis, 111
F Immunotherapy, 70
Fibrolamellar carcinoma, 13 International Society of Pediatric
Fluorouracil (5FU), 10 Oncology, 8
Focal nodular hyperplasia, 11, 89 Irinotecan, 10
Follow-up, 11, 157 Irreversible electroporation, 68
Fresh tissue, 81
J
G JAG-1, 4
Gemcitabine, 10 JAGGED-1, 4
Global estimates, 57 JNJ-3989, 118
Glycogen storage disorders, 20
Glypican-3, 46 K
Gross anatomy, 78
Keratin, 13, 16
Gross dissection, 77
Kidney, 7, 23, 80
Gross features, 167
Gross inspection, 81
L
H Laboratory tests, 148
Lamivudine, 113
HBsAg inhibitors, 119
Lifestyle, 70
HBV life cycle, 111
Lipid conjugates, 118
Hematoxylin and eosin stain, 42
Lipid-rich hepatocellular
Hepatic angiosarcoma, 102
carcinoma, 13
Hepatitis B, 109
Liver lesions, 88
Hepatoblastoma, 145
Liver resection, 63
Hepatocellular adenoma, 11, 91
Liver transplantation, 63, 129, 155
Hepatocellular carcinoma, 2, 39, 93
Liver tumors, 2, 87
Hepatocellular lesions, 89
Locoregional therapies, 127, 131
Hereditary tyrosinemia I, 19
Lung metastases, 156
Histology, 149, 167
Lymph nodes, 83
Host immunity, 120
Index 181
M Q
Management, 62 Quadrant, 78
Metastatic malignancies, 103 Quadrate lobe, 79
Micrographs, 7
Microwave ablation, 65 R
Milan criteria, 64, 129
Radiofrequency ablation, 64
Mitomycin, 10
Radiologic-pathologic correlation, 87
Molecular findings, 170
Radiology, 89, 166
Morphology, 6
Reticulin stain, 43
Mucinous cystic neoplasm, 97
Reverse transcriptase inhibitors, 118
Risk assessment, 152
N
Natural history, 112 S
Non-alcoholic fatty liver disease, 55
Sampling, 81, 83
Non-alcoholic steatohepatitis, 55
Sarcoma of the liver, 165
Nucleos(t)ide analogues, 109
Sarcomas, 175
Selection criteria for liver
O
transplantation, 129
Okuda system, 8, 48 Small interfering RNAs, 117
Oxaliplatin, 10 Sorafenib, 10
Staging, 8, 47, 61, 83, 151
P Stereotactic body radiation therapy, 137
Stereotactic body radiotherapy, 68
Pathogenesis, 40, 146
Surface inspection, 78
Pediatric metabolic conditions, 17
Surgical anatomy of the liver, 78
Pegylated interferon, 113
Surgical approach, 154
Percutaneous ethanol injection, 64
Symptoms, 5, 147
Pregenomic RNA (pgRNA), 112
Systemic therapies, 69
Pre-neoplastic lesions, 41
PRETEXT, 8, 152
T
Prevention, 113, 114
Primary liver cancer, 55 Targeted therapies, 69
Prognosis, 170 Telbivudine, 113
Progressive familial intrahepatic cholestasis Temozolomide, 10
type, 2, 20 Tenofovir alafenamide, 113
182 Index
Doi: https://doi.org/10.36255/exonpublications.livercancer.2021.index
Liver Cancer
Consolato M. Sergi, MD, PhD, MPH, FRCPC, FCAP
Editor