You are on page 1of 12

Update on Precursor and Early Lesions of

Hepatocellular Carcinomas
Young Nyun Park, MD, PhD

Nmultistep
Context.—There is increasing evidence to support a
model of the process of human hepatocarcino-
and large cell change is a rather heterogeneous lesion that
may represent both reactive change and true dysplasia.
genesis. Precursor lesions are characterized by the Dysplastic nodules can be categorized as low or high grade
appearance of dysplastic lesions in the form of microscopic according to the degree of atypia. High-grade dysplastic
dysplastic foci and macroscopic dysplastic nodules. There nodules have been reported to show molecular changes
are 2 types of small hepatocellular carcinoma (HCC) similar to HCC and have a high risk of malignant
(#2 cm in diameter): (1) early HCC with an indistinct transformation. Early HCC, which may correspond to
margin and (2) progressed HCC with a distinct margin. microinvasive carcinomas of other organs, is a well-
Pathologic diagnostic criteria for early HCC have recently differentiated HCC, and differential diagnosis between
been set up based on a consensus between Eastern and early HCC and high-grade dysplastic nodule is difficult.
Western pathologists. Identification of stromal invasion and application of a
Objective.—To review the nomenclature, pathology, panel of markers (glypican-3, heat shock protein 70, and
and biomarkers of precursor and early lesions of HCC. glutamine synthetase) is helpful for diagnosis of early HCC.
Data Sources.—Literature review and illustrations from Detection of precursor lesions of HCC is important in
case materials were used. recognizing patients with higher risk of developing HCC,
Conclusions.—Dysplastic foci are composed of large and and diagnosis of early HCC can improve patient survival by
small cell changes. Small cell change is considered to be a allowing for early and adequate treatment.
more advanced precursor lesion than large cell change, (Arch Pathol Lab Med. 2011;135:704–715)

IHepatocellular
n humans, most hepatocellular carcinomas (HCCs) arise
in the background of chronic hepatitis/cirrhosis.
carcinoma usually follows a multistep
This term was introduced by an international consensus
article published in 1995,1 and its arbitrary size criterion is
related to the fact that these microscopic lesions are
sequence, and de novo development of HCC may occur, usually contained within a single hepatic lobule or
although this appears to be rare. Morphologically recog- cirrhotic nodule.2,3 In most cases, dysplastic foci are
nizable lesions during hepatocarcinogenesis include dys- recognized incidentally in liver biopsies or resected liver
plastic lesions (dysplastic foci and dysplastic nodules specimens, as they are not macroscopically detectable.
[DNs]) and small cancerous lesions (#2 cm in diameter) Dysplastic foci are categorized according to the main cell
(early HCC and small progressed HCC) (Figure 1; type as small cell foci, large cell foci, or iron-free foci, and
Table 1). Their pathologic features are discussed with dysplastic hepatocytes showing small cell changes (SCCs)4
emerging biomarkers. or large cell changes (LCCs)5 are also part of the spectrum
of cytologic abnormalities occurring in DNs.
DYSPLASTIC FOCI Small cell change, originally described as small cell
Dysplastic foci are microscopic lesions composed of dysplasia,4 is defined as hepatocytes showing decreased
dysplastic hepatocytes that measure less than 1 mm in size cell volume, increased nuclear to cytoplasmic ratio, mild
and occur in chronic liver disease, particularly in cirrhosis. nuclear pleomorphism and hyperchromasia, and cyto-
plasmic basophilia, giving the impression of nuclear
Accepted for publication January 11, 2011. crowding (Figure 2, A).3 Small cell change is characterized
From the Department of Pathology, Brain Korea 21 Project for by higher proliferative activity than the surrounding
Medical Science, Center for Chronic Metabolic Disease, and the normal-looking hepatocytes, telomere shortening with
Severance Biomedical Science Institute, Yonsei University Health
System, Seoul, South Korea.
p21 checkpoint inactivation, increased formation of
The author has no relevant financial interest in the products or micronuclei suggesting chromosomal instability, chromo-
companies described in this article. somal gains and losses, a morphologic resemblance to
Presented at the 9th Spring Seminar of the Korean Pathologists well-differentiated HCC, and a histologic continuum to
Association of North America; March 18–20, 2010; Washington, DC; in HCC.2,3,6,7 These data support the precancerous nature of
conjunction with the 99th Annual Meeting of the United States and SCC. Small cell change occurring in small foci, with
Canadian Academy of Pathology.
Reprints: Young Nyun Park, MD, PhD, Department of Pathology, expansile rather than widespread growth, was reported to
Yonsei University Health System, 250 Seongsanno, Seodaemun-gu, be associated with HCC development.2 Thus, expansile
Seoul 120-752, South Korea (e-mail: young0608@yuhs.ac). foci of SCC are considered to correspond to dysplastic foci
704 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
Figure 1. Scheme of preneoplastic and neoplastic nodules during hepatocarcinogenesis. Abbreviations: DN, dysplastic nodule; HCC,
hepatocellular carcinoma; LCC, large liver cell change; SCC, small liver cell change.

in the International Working Party classification.1 In alterations present in adjacent HCCs.6 Data showing that
contrast, a diffuse pattern of SCC may represent regener- LCC has higher argyrophilic nucleolar organizer region
ating, rather than dysplastic, hepatocytes.8,9 The difficulty values than other normal-looking hepatocytes (with the
in distinguishing precancerous SCC from regenerative highest argyrophilic nucleolar organizer region values
changes may be one reason for the limited predictive being strongly associated with HCC development during
value of SCC for HCC development.2 Some reports have follow-up),17 abnormal DNA content,18–20 chromosomal
shown a significant correlation between the presence of numerical aberrations, and chromosomal gains and
SCC and HCC,10,11 whereas others have not.12–14 losses21,22 all demonstrate that LCC is a precancerous
Large cell change, originally termed liver cell dysplasia,5 lesion. In addition, the cirrhotic macronodules with LCC
is defined as hepatocytes with both nuclear and cytoplas- were reported to be monoclonal, a prerequisite step of
mic enlargement (therefore, a preserved nuclear to hepatocarcinogenesis.23 A recent study reported that
cytoplasmic ratio), nuclear pleomorphism and hyperchro- telomere shortening, DNA damage, increased prolifera-
masia, and frequent multinucleation (Figure 2, B).3 A tion, increased DNA damage, and inactivation of the cell
definitive consensus has not been reached regarding the cycle check point (p16 and p21) were found in LCC of B
biologic nature of LCC.2 The data supporting LCC as a viral cirrhosis, while these features are not found in LCC
reactive process related to chronic injury, or senescence, of chronic cholestasis.24 These conflicting data suggest that
include low proliferative activity and increased apopto- LCC is a heterogeneous entity with 2 types: one that is not
sis,15 absence of a histologic continuum with HCC,15 totally benign and tumor-related (true dysplasia), and
association with cholestasis,16 and absence of genetic another one that is benign and non–tumor-related

Table 1. Hepatic Precancerous Lesions and Small Hepatocellular Carcinoma (HCC)


Dysplastic focus
N Cluster of hepatocytes, ,1 mm in size, characterized by presence of either small cell change or large cell change
Dysplastic nodule
N A distinctly nodular lesion (usually 1 cm in size) differing from the surrounding liver parenchyma regarding size, color, and texture and that
bulges from the surrounding liver on the cut surface
N Dysplastic nodule, low grade: mild atypia
N Dysplastic nodule, high grade: at least moderate atypia, but insufficient for a diagnosis of HCC
Small HCC (#2 cm in diameter)
N Early HCC (small HCC with indistinct margins, small HCC with vaguely nodular type)
N Small and progressed HCC (small HCC with distinct margins)

Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 705
bulges from the surrounding liver on a cut surface
(Figure 3, A). Dysplastic nodules mostly measure about
1 cm in diameter. They may be single or multiple (less
than 10 in number). According to international consen-
sus,1 DNs are classified as low grade or high grade,
depending on the degree of atypia.
Histologically, low-grade DNs show mild increases in
cell density with a monotonous pattern compared with
that of cirrhotic nodules (Figure 3, B through D). They
have features suggestive of a clonal cell population, such
as diffuse iron or copper accumulation, or uniform
steatosis in livers without fatty change. Cytologic atypia
is mild. Large cell change may be found; however SCC is
not present. Cell density is slightly higher than in the
surrounding liver but less than 1.3-fold. Architectural
changes beyond clearly regenerative features are not
present; these lesions do not contain pseudoglands.
High-grade DNs show at least moderate cytologic or
architectural atypia that approaches but does not quite
reach the levels of HCC (Figure 3, E). These lesions most
often show increased cell density, 1.3 to 2 times greater
than that of the adjacent liver tissue, and SCC is the most
frequently seen form of cytologic atypia in high-grade
DNs.
Dysplastic nodules may derive their blood supply both
from portal vessels and from newly formed arteries
(termed unpaired or nontriadal arteries) (Figure 3, F).2,3,29–31
Portal tracts are commonly seen in DNs but may be few
and scarred. Unpaired arteries, which have no accompa-
nying bile ducts, are not present in cirrhosis. They are least
prominent in low-grade DNs, both in number and in size,
increasing in numbers through high-grade DNs and are
most abundant in HCC. Similarly, sinusoidal capillariza-
tion is minimal in cirrhosis, and it increases from low- to
high-grade DN, with the highest levels in HCC. Neoan-
Figure 2. Dysplastic foci. A, Dysplastic foci showing small liver cell giogenesis is caused by growth factor production includ-
change (arrows). B, Dysplastic foci showing large liver cell change ing vascular endothelial growth factor by lesional cells.32 It
(arrows) (hematoxylin-eosin, original magnifications 3200 [A and B]). is also correlated with radiologic features; DN usually
appears isovascular or hypovascular compared with the
(reactive change).25 Actually, the presence of LCC in B surrounding parenchyma, whereas HCC usually shows a
viral or C viral cirrhosis has been reported to be an hypervascular appearance in the arterial phase of con-
important independent risk factor for subsequent devel- trast-enhanced hepatic imaging.2,3,27
opment of HCC.12–14 Interestingly, in chronic liver disease, The following evidence supports the theory of a
telomere shortening, chromosomal instability, cell cycle premalignant nature of DN:
check point inactivation, and DNA damage were progres- 1. An epidemiologic association of DNs with HCC in
sively increased from LCC to SCC with peak levels at resected and explanted livers, particularly in end-stage
HCC.7,24 These data suggest that SCC may be a more cirrhotic patients.33–37 Their association with HCC takes
advanced precancerous lesion and that LCC might be a 2 forms. First, both low- and high-grade DNs are
very early precursor of HCC in chronic viral hepatitis B markers of an increased risk of cancer development in
or C. the liver. In fact, HCCs do not always develop within
Iron-free foci in genetic hemochromatosis were report- DNs but may be found elsewhere in the liver,
ed to be proliferative lesions associated with a high independent of the latter. Second, subnodules of
incidence of HCC during follow-up.26 From a practical HCC are often seen within DNs (nodule-within-
point of view, identification of SCC, LCC, or iron-free foci nodule pattern) (Figure 4).
in liver biopsies may signify an increased risk of HCC over 2. The natural history of DNs shows an increased risk
time. The presence of such lesions should be noted on of malignant transformation compared with control
pathology reports. patients with cirrhosis. There are several reports
that have evaluated the natural history of large
DYSPLASTIC NODULE hepatic nodules. Borzio and colleagues38 followed 90
Dysplastic nodules are usually detected in cirrhotic cirrhotic patients with histologically proven large
livers but are occasionally found along with chronic liver regenerative nodules (n 5 56) or low-grade (n 5 16)
disease without cirrhosis.1–3,27,28 A DN is a distinctly or high-grade (n 5 19) DNs and found that the
nodular lesion that differs from the surrounding liver presence of high-grade DNs was associated with a
parenchyma regarding size, color, and texture and that higher risk for HCC development (hazard ratio, 2.4
706 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
Figure 3. Dysplastic nodule (DN). A, Macroscopic feature of DN (arrows) in B viral cirrhosis. B, Microscopic feature of low-grade DN (above
dotted line). Portal tracts are present inside DN. Comparison of cell density among cirrhotic nodule (C), low-grade DN (D), and high-grade DN (E) at
the same magnification. Low-grade DN (D) shows a slightly increased cell density compared with extranodular cirrhotic liver and mild cellular
atypia. The intranodular portal tract is marked by arrows. High-grade DN (E) shows high cell density, a thin trabecular pattern, and small cell change
with an increased nuclear to cytoplasmic ratio. F, Immunohistochemical stain for CD34 shows increased sinusoidal capillarization and unpaired
arteries (arrow) in high-grade DN (hematoxylin-eosin, original magnifications 3100 [B] and 3200 [C through E]; original magnification 3200 [F]).

during a mean follow-up period of 33 months). colleagues39 followed 154 patients who had histo-
Large regenerative nodules and low-grade DNs logically proven large regenerative nodules (n 5 99)
were associated with a low risk for malignant or low-grade (n 5 42) or high-grade (n 5 13) DNs
transformation. In another study, Kobayashi and for a median period of 2.8 years. The hazard ratios
Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 707
Figure 4. Hepatocellular carcinoma (HCC) arising in dysplastic nodule. A, A well-defined and dark brown nodule in cirrhosis shows a nodule-in-
nodule pattern. The inner nodule shows a yellowish tan color. B, The outer nodule shows the features of high-grade dysplastic nodule. The
intranodular portal tract is marked by arrows. C, Nodule-in-nodule pattern. The inner nodule (below dotted line) shows a well-differentiated HCC
with fatty change and Mallory bodies. D, Immunohistochemical stain for CD34 shows increased sinusoidal capillarization and unpaired arteries in
the inner nodule of HCC (below dotted line) than in the outer nodules of the dysplastic nodule (hematoxylin-eosin, original magnifications 3200 [B
and C]; original magnification 3200 [D]).

of high-grade DN, low-grade DN, and large regenera- vulnerable to further genetic change for malignant
tive nodules for malignant transformation were 16.8, transformation.
2.96, and 1.0, respectively. In this study, however, SCC Important pathologic features that may be helpful for
was considered to be a feature of low-grade as well as the differential diagnosis of hepatic nodules are summa-
high-grade DNs. Finally, Terasaki and colleagues40 rized in Table 2. The differentiation between high-grade
found that histologic features predictive of malignant DNs and well-differentiated HCC is often difficult and
transformation of DNs included increased cellularity, recognition of stromal invasion of tumor cells supports the
clear cell change, SCC, and fatty change. diagnosis of HCC. This is further discussed in the section
3. Similarities in both genetic and epigenetic changes on early HCC and biomarkers.50 Another diagnostic
between DNs and HCC have been detected. The difficulty is making the distinction between low-grade
incidence and extent of these changes is between those DN and large regenerative nodules, as these lesions share
observed in cirrhotic nodules and those in HCC. In a number of histologic features. The presence of unpaired
some cases, the very same molecular alteration has arteries or evidence of a clonelike cell population in a large
been reported in DNs and the adjacent HCC.41–45 nodule suggest that it is dysplastic rather than regenera-
4. Dysplastic nodules, especially high-grade DNs, have tive. Interestingly, about 15% of low-grade DNs showed
shortened telomeres with increased chromosomal insta- shortened telomere length and 7% showed telomerase
bility, and most high-grade DNs strongly express activity equal to the levels found in high-grade DNs,
hTERT mRNA at levels similar to those of HCCs, and although there was no histologic difference between the
33% of them showed high telomerase activity.46–48 Fur- lesions with and without these changes.46–48 Chromosomal
thermore, the p21 check point is inactivated in DNs in instability, indicated by micronuclei, is higher in low-
contrast to cirrhotic nodules where they are activated.49 grade DN than in cirrhotic nodules, and the p21
Thus, DNs, especially high grade, are considered to be checkpoint is deactivated in low-grade DNs unlike in
708 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
Table 2. Important Pathologic Features of Hepatocellular Nodules
LRN LGDN HGDN WD HCC
Cytologic features
Small cell change 2 2 + +
Large cell change 6 6 6 2
Clonelike foci (clear, fatty) 2 6 + +
Architectural features
Plate thickening $3 cells 2 2 2 +
Increased cell density compared with surroundings 2 2 1.3 to 2 times .2 times
Pseudoglands 2 2 6 +
Nodule-in-nodule 2 2 2 6
Portal tract + + + 6
Unpaired arteries and capillarized sinusoidsa 2 6 6 +
Stromal invasionb 2 2 2 +
Reticulin framework + + + 6
Tumor markers
Glypican-3 2 2 6 (9%) + (69%)
Heat shock protein 70 2 2 6 (5%) + (78%)
Glutamine synthetase 2 2 6 (13%) + (59%)
Positive in at least 2 of above 3 markers 2 2 2 + (72%)
Abbreviations: 2, absent; 6, may be present; +, usually present; ( ), % of positivity in resected specimen; HGDN, high-grade dysplastic nodule;
LGDN, low-grade dysplastic nodule; LRN, large regenerative nodule; WD HCC, well-differentiated hepatocellular carcinoma.
a
Immunohistochemical stain for CD34 is helpful.
b
Immunohistochemical stain for keratin 7/19 is helpful.

cirrhotic nodules, where the p21 is activated.49 Thus, some of well-differentiated HCC mimic nonneoplastic hepato-
low-grade DNs may have more potential to progress cytes, and identification of these lesions as cancer may be
along the multistep hepatocarcinogenesis course, and this difficult microscopically. The cell density of early HCC is
diagnosis may indicate that the patient is at increased risk more than 2 times that of the surrounding hepatic
of developing HCC. In a report from the International parenchyma. Careful examination of areas with increased
Consensus Group for Hepatocellular Neoplasia, diagnos- cell density on low power can be helpful to not miss the
tic agreement for low-grade DN is low, and from a region of a well-differentiated HCC. Fatty change is
practical standpoint, the distinction between low-grade common, reported in about 40% of cases. In areas of fatty
DN and large regenerative nodules of cirrhotic livers is change, the nuclear to cytoplasmic ratio is relatively low
difficult to make by morphology alone.27 Fortunately, this and cytologic atypia may be difficult to detect. Tumor cells
distinction does not have any significant clinical impact at often invade the fibrous tissue of portal tracts within or
present, as low-grade DNs are followed up and urgent outside the nodule, and detection of stromal invasion can
treatment is not required. be aided by the use of Victoria blue staining outlining
residual portal tracts or by a lack of ductular reaction,
EARLY HCC which usually takes place around nonmalignant nodules,
Small HCC is defined arbitrarily as carcinoma that visualized by keratin 7/19 immunostaining (Figure 5, E
measures less than 2.0 cm in diameter. Studies from and F).50,54 However, early HCC does not show vascular
Japan36,51,52 have shown that there are 2 types of small invasion, and it usually does not metastasize. Therefore,
HCCs: early HCC and (small) progressed HCC. Early early HCC can correspond to an entity in between in situ
HCC is a low-grade, early-stage tumor whose definition carcinoma and invasive carcinoma, similar to microinva-
was agreed upon after a prolonged discussion with sive carcinoma of other organs. Varying numbers of portal
circulation of slides between a group of Eastern and tracts are found within the nodule and arterial neovascu-
Western pathologists.27 Early HCC is a vaguely nodular larization, which can be easily detected by immunohisto-
and small-sized nodule (Figure 5, A). In a series of 116 chemistry for CD34, is more developed than in high-grade
small HCCs, the mean size of the early HCCs was 11.9 mm, DNs but incomplete compared with progressed HCC2,3
but that of small progressed HCCs (small HCCs with a (Figure 5, G and H). Therefore, early HCCs usually show
distinct margin) was 16.0 mm.28 The margin of early HCCs isovascular or hypovascular and rarely hypervascular
is usually indistinct as tumor cells grow by replacing the appearance in the arterial phase of contrast-enhanced
nonneoplastic hepatocyte cords without forming a tumor hepatic imaging. The reticulin framework is usually
capsule (Figure 5, B). Therefore, it may be difficult to reduced but not totally lost as in progressed HCCs. Early
recognize at a macroscopic level. On the other hand, in the HCC is considered to be a precursor of distinctly nodular
setting of advanced cirrhosis, early HCC may show a (progressed) HCC, and actually in some cases of pro-
more distinctly nodular feature as it is surrounded by gressed HCCs, a component of early, well-differentiated
cirrhotic septa (not tumor capsule).33,53 HCC is detected at the periphery of tumor, indicating a
Histologically, early HCCs are usually well differenti- multistep progression of hepatocarcinogenesis. It should
ated and consist of small neoplastic cells (cytologically also be noted that the characteristic histologic features of
reminiscent of SCC) with increased nuclear to cytoplasmic early HCC may sometimes be seen in tumors measuring
ratio arranged in irregular, thin trabeculae and pseudo- more than 2 cm in diameter, suggesting that some HCCs
glandular structures (Figure 5, C and D). Thus, tumor cells have a slow biologic evolution.
Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 709
Figure 5. Early hepatocellular carcinoma (HCC). A, Macroscopic features of early HCC showing a vaguely nodular appearance with an ill-defined
margin. B, At low power, early HCC (left side) shows a replacing growth pattern without a tumor capsule at the tumor border. The cell density of the
tumor is more than 2 times higher than the surrounding cirrhotic nodules (right side). Fatty change and some portal tracts are present in the tumor. C,
At medium power, an increased cell density is more obvious in the areas without fatty change. D, At high power, early HCC shows a well-
differentiated histology and small tumor cells with high nuclear to cytoplasmic ratios, thin trabecular pattern with 2- to 3-cell thickness, and acinar
pattern. E, Stromal invasion of tumor cells (arrows) into fibrous stroma of the portal tract. F, Immunohistochemical stain for keratin 19 showing no
ductular reaction around the stromal invasion of tumor cells (arrows). Bile ducts show a positive expression of keratin 19. G, Development of
unpaired arteries (arrows) without accompanying bile ducts in early HCC. H, Immunohistochemical stain for CD34 showing increased sinusoidal
capillarization and unpaired arteries (arrows) in early HCC (hematoxylin-eosin, original magnifications 340 [B], 3100 [C], and 3200 [D, E, and G];
original magnifications 3200 [F and H]).

710 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
DN. In well-differentiated HCC, tumor cells mimic
nontumor hepatocytes; therefore, stromal invasion is
sometimes difficult to distinguish from clusters of
nonneoplastic hepatocytes that are localized around the
portal tracts or fibrous septa of chronic hepatitis/cirrhosis
(pseudoinvasion), close to the infiltrating edge of the
tumor. These can be distinguished by immunostaining for
keratin 7 and keratin 19, which highlight the presence of a
ductular reaction around reactive and regenerating
clusters (pseudoinvasion) as a ductular reaction is absent
around tumor cells of stromal invasion (Figure 5, F).50
Glypican-3
Glypican-3 (GPC3), a cell-surface heparin sulfate pro-
teoglycan, has recently become established as a serum and
tissue marker for HCC.27,56–59 Immunohistochemistry for
GPC3 demonstrates a cytoplasmic and sometimes mem-
branous or canalicular staining pattern (Figure 7, E).
GPC3 immunoreactivity of resected liver specimens was
reported in 69% of well-differentiated HCCs and in 81% of
moderately to poorly differentiated HCCs, while 9% of the
high-grade DNs and none of the low-grade DNs were
positive.60 GPC3 staining must be interpreted in the
context of histologic features because it is also reported
in regenerating hepatocytes of chronic hepatitis61 and in
melanocytic lesions.62
Heat Shock Protein 70
Heat shock protein 70 (HSP70) is a chaperone stress
protein and a potent antiapoptotic survival factor.63,64
Significantly more upregulation of HSP70 was reported
in early HCC than in DNs among a set of 12 600 genes
studied, and it was furthermore significantly overex-
pressed in progressed HCC compared with early HCC.65
HSP70 immunoreactivity is found in the nucleus and
Figure 6. Small and progressed hepatocellular carcinoma (HCC). A, cytoplasm (Figure 7, F). HSP70 immunoreactivity in a
Macroscopic features of small progressed HCC showing a distinctively resected specimen was recently reported in 78% of well-
nodular appearance with tumor capsule in cirrhosis. B, Microscopic
features of small progressed HCC showing moderately differentiated
differentiated HCCs and in 67% of moderately to poorly
histology. Invasion of the tumor capsule is noted (arrows) (hematoxylin- differentiated HCCs, while it was detected in 5% of high-
eosin, original magnification 3200 [B]). grade DNs and in no low-grade DNs.60 The hepatobiliary
cells of ductular reactions also showed positive reactions,
On the other hand, small and progressed HCCs (small which can be used as an internal positive control.
HCCs of the distinctly nodular type) are usually moder-
ately differentiated and often form subnodules with less Glutamine Synthetase
differentiation (Figure 6). A tumor capsule is found in Glutamine synthetase (GS) is a target gene of b-catenin
about 50% of cirrhotic livers. Invasion of portal vein and its overexpression is associated with mutations of b-
branches by tumor cells and minute intrahepatic metas- catenin or with activation of this pathway.27,66,67 Upregula-
tases have been described in 27% and 10% of cases, tion of GS messenger RNA, protein, and activity was
respectively.52 Unpaired (nontriadal) arteries are more shown in human HCC,66,67 and the stepwise increase in GS
developed in distinctly nodular HCCs than in early HCCs, immunoreactivity was reported from precancerous le-
and portal tracts are absent in the nodule55 (Figure 7, A, C, sions to early HCC to progressed HCC.68 Glutamine
and D). Therefore, progressed HCC shows a hypervas- synthetase is expressed in hepatocytes surrounding the
cular appearance in the arterial phase of contrast- terminal hepatic venules in normal livers,69 so to increase
enhanced hepatic imaging. its specificity as a marker of malignancy, GS immuno-
BIOMARKERS: APPLICATION OF IMMUNOCHEMICAL staining should be diffuse and of strong intensity
STAIN FOR DIFFERENTIAL DIAGNOSIS OF (Figure 7, B). This pattern was reported in 14% of high-
HEPATIC NODULES grade DNs, 59% of well-differentiated HCCs, and 86% of
moderately to poorly differentiated HCCs of resected
Keratins 7 and 19 specimens.60
Stromal invasion is defined as the presence of tumor Diffuse immunoreactivity of GS is also reported in the
cells in portal tracts or fibrous septa within or outside the b-catenin–activated subtype of hepatocellular adenoma.
lesion under examination (Figure 5, E). The presence of Although hepatocellular adenomas rarely transform to
stromal invasion is a very helpful feature in the dis- HCC, a subtype of hepatocellular adenoma has been
crimination of well-differentiated HCC from high-grade recently identified that has an increased risk of malignant
Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 711
Figure 7. A panel of immunohistochemical markers for hepatocellular carcinoma (HCC). A well- to moderately differentiated HCC showing a
nodule-in-nodule pattern (A, C). The margin of the tumor is marked by the dotted line. Immunohistochemical stain for glutamine synthetase (B)
shows a positive reaction in the tumor, with the nontumor hepatocytes in zone 3 also showing a positive reaction. Sinusoidal capillarization (CD34
immunostaining, D) is more marked in the moderately differentiated inner HCC nodule than in the well-differentiated outer HCC nodule. Glypican-3
(E) and heat shock protein 70 (F) are positive in HCC, with weak staining in well-differentiated HCC (outer nodule) and strong staining in moderately
differentiated HCC (inner nodule). Bile ducts and ductules are also positive for heat shock protein 70 (F) (hematoxylin-eosin, original magnifications
3100 [A] and 3200 [C]; original magnifications 340 [B] and 3200 [D through F]).

712 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
transformation and is characterized by activating muta- immunohistochemistry findings, which provides confir-
tions in the gene encoding b-catenin.70 matory evidence of malignancy in doubtful cases.
The Combination of GPC3, HSP70, and GS LIVER BIOPSY INTERPRETATION FOR PRECURSOR
The sensitivity and specificity of individual markers for AND EARLY LESIONS OF HCC
the detection of well-differentiated HCC, including early International guidelines recommend that a biopsy
HCC, were reported to be 59% and 86% for GS, 69% and should be performed for nodules less than 2 cm in size if
91% for GPC3, and 78% and 95% for HSP70, respectively, their radiologic findings are not typical of HCC, and a
and the immunoreactivity of these markers may be weak biopsy is not necessary for lesions showing characteristic
or only focal in well-differentiated HCC, including early radiologic findings.73 Moderately or poorly differentiated
HCC. Thus, application of a single marker does not seem HCC can be easily diagnosed based on histology, while
to be very helpful in diagnosing well-differentiated HCC. there is diagnostic difficulty in the distinction of high-
Application of more than 1 marker raises the overall grade DN from well-differentiated HCC. In addition, it is
accuracy. When applying a panel of these 3 markers even more difficult when the diagnosis is made on a small
(GPC3, HSP70, and GS) to resected small lesions, the biopsied sample, as only a limited portion of the lesion is
positivity for any 2 markers had a sensitivity of 72% and a available. The distinction between low-grade DNs and
specificity of 100% for detecting well-differentiated HCC. large regenerative nodules is also very difficult to make,
When applying 2 markers, the combination of HSP70 and and it is not recommended to attempt such a distinction
GPC3 seems to be best with 59% sensitivity and 100% on a biopsy, as it cannot be made confidently based on
specificity for well-differentiated HCC in resected cases.60 morphology alone.27
It is recommended that sampling includes both
CD34 intralesional and extralesional tissues because the archi-
Newly formed arteries have no accompanying bile tectural and cytologic abnormalities are better appreci-
ducts, unlike normal hepatic arteries; thus, they are termed ated by comparing these 2 regions with each other.
unpaired or nontriadal arteries. Unpaired (nontriadal) Microscopically, the identification of stromal invasion is
arteries and capillarized sinusoids can be easily detected important for the diagnosis of HCC, but evaluating
via immunohistochemical stain for CD34 in contrast to stromal invasion is difficult in a biopsy because only a
normal sinusoidal endothelial cells that do not express small portion of the tumor-nontumor interface and
CD34 (Figure 7, D). Sinusoidal capillarization may occur in intralesional portal tracts is sampled, and stromal
relation to increased blood flow and pressure. These invasion may not be included in the biopsied tissue. In
features increase from low-grade DN, high-grade DN, a recent study based on liver biopsies, a negative or focal
and early HCC to progressed HCCs29,30; it can therefore be (,10% of stromal-parenchymal interface) ductular reac-
concluded that the semiquantitative evaluation of CD34 tion in a desmoplastic area adjacent to abnormal-looking
expression can be helpful in supporting diagnosis based on hepatocytes suggests stromal invasion of tumor cells,
histology. However, it is not very helpful to differentiate indicating HCC. In contrast, the presence of a keratin 7/
high-grade DN from well-differentiated HCC because this 19–positive ductular reaction around abnormal-looking
is a gradual rather than stepwise change, and no definite hepatocytes located in portal/periportal areas represents
cutoff values have been validated for each lesion during pseudoinvasion and is evidence against HCC.50 Appli-
multistep hepatocarcinogenesis. cation of a panel of GPC3, HSP70, and GS is reported to
be helpful in detecting well-differentiated HCCs, and the
PCNA or Ki-67
sensitivity and specificity in liver biopsies were 73% and
Proliferative activity gradually increases from low-grade 100%, respectively, when any 2 of the 3 markers were
DNs, high-grade DNs, and early HCCs to HCCs, and positive, although sensitivity was lower than in resected
although activity is low in low-grade DNs, it is slightly specimens.74 These tumor markers are also helpful in
higher than in cirrhosis.71 Application of PCNA or Ki-67 is detecting less-differentiated HCC, when the tumor
helpful in detecting the proliferation rate of the lesions; portion is minimally sampled and the tissue artifact is
however, semiquantitative evaluation of proliferative activ- severe. Before making the final diagnosis based on
ity is not very helpful in differentiating high-grade DN from biopsied liver tissue, clinicopathologic or radiopatholo-
well-differentiated HCC, as no definite cutoff values have gic correlation is recommended as gross morphology is
been established for the various histologic parameters. not available, and only a limited portion of the nodular
lesion is present in the biopsied material.
a-Fetoprotein
Cytology alone is not recommended for the distinction
a-Fetoprotein has long been a well-established marker between well-differentiated HCC and high-grade DN, as
for HCC; however, serum a-fetoprotein is rarely elevated it cannot provide architectural information. It is rather
in early HCCs and is not detected in DNs.27,72 Likewise, it suggested to be used as a confirmatory procedure for
is not useful as a tissue marker for HCC because of its low progressed and less-differentiated HCC or as a method of
sensitivity (about 30%), even in progressed and less- cell sampling for molecular studies.
differentiated HCC.27 In conclusion, morphologically recognizable lesions
during hepatocarcinogenesis include dysplastic foci,
p53 DNs, early HCCs, and progressed HCCs. The pathologic
Mutations of TP53, which seem to play a pivotal role in diagnosis of high-grade DNs or small HCCs, including
hepatocarcinogenesis, occur late in the process, being both early and progressed HCCs, is important as these
detectable in progressed and less-differentiated HCC. lesions should be treated with local ablation, surgical
Mutated TP53 can be detected by nuclear positivity in resection, or liver transplantation.
Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 713
This work was supported by a National Research Foundation 28. Kojiro M. Pathology of Hepatocellular Carcinoma. Malden, MA: Blackwell
grant funded by the Korean government (MEST) (R13-2002-054- Publishing; 2006:31–49.
29. Park YN, Yang CP, Fernandez GJ, Cubukcu O, Thung SN, Theise ND.
03004-0). Neoangiogenesis and sinusoidal ‘‘capillarization’’ in dysplastic nodules of the
References liver. Am J Surg Pathol. 1998;22(6):656–662.
1. International Working Party. Terminology of nodular hepatocellular lesions. 30. Roncalli M, Roz E, Coggi G, et al. The vascular profile of regenerative and
Hepatology. 1995;22(3):983–993. dysplastic nodules of the cirrhotic liver: implications for diagnosis and
2. Hytiroglou P, Park YN, Krinsky G, Theise ND. Hepatic precancerous lesions classification. Hepatology. 1999;30(5):1174–1178.
and small hepatocellular carcinoma. Gastroenterol Clin North Am. 2007;36(4): 31. Ueda K, Terada T, Nakanuma Y, Matsui O. Vascular supply in
867–887, vii. adenomatous hyperplasia of the liver and hepatocellular carcinoma: a
3. Theise ND, Curado MP, Franceschi S, et al. Hepatocellular carcinoma. In: morphometric study. Hum Pathol. 1992;23(6):619–626.
Bosman FT, Carneiro F, Hruban RH, Heise ND, eds. WHO Classification of 32. Park YN, Kim YB, Yang KM, Park C. Increased expression of vascular
Tumours of the Digestive System. 4th ed. Lyon, France: IARC Press; 2010:205– endothelial growth factor and angiogenesis in the early stage of multistep
216. hepatocarcinogenesis. Arch Pathol Lab Med. 2000;124(7):1061–1065.
4. Watanabe S, Okita K, Harada T, et al. Morphologic studies of the liver cell 33. Theise ND, Park YN, Kojiro M. Dysplastic nodules and hepatocarcino-
dysplasia. Cancer. 1983;51(12):2197–2205. genesis. Clin Liver Dis. 2002;6(2):497–512.
5. Anthony PP, Vogel CL, Barker LF. Liver cell dysplasia: a premalignant 34. Terada T, Terasaki S, Nakanuma Y. A clinicopathologic study of
condition. J Clin Pathol. 1973;26(3):217–223. adenomatous hyperplasia of the liver in 209 consecutive cirrhotic livers
6. Marchio A, Terris B, Meddeb M, et al. Chromosomal abnormalities in liver examined by autopsy. Cancer. 1993;72(5):1551–1556.
cell dysplasia detected by comparative genomic hybridisation. Mol Pathol. 2001; 35. Furuya K, Nakamura M, Yamamoto Y, Togei K, Otsuka H. Macroregen-
54(4):270–274. erative nodule of the liver: a clinicopathologic study of 345 autopsy cases of
7. Plentz RR, Park YN, Lechel A, et al. Telomere shortening and inactivation of chronic liver disease. Cancer. 1988;61(1):99–105.
cell cycle checkpoints characterize human hepatocarcinogenesis. Hepatology. 36. Sakamoto M, Hirohashi S, Shimosato Y. Early stages of multistep
2007;45(4):968–976. hepatocarcinogenesis: adenomatous hyperplasia and early hepatocellular carci-
8. Nakanuma Y, Hirata K. Unusual hepatocellular lesions in primary biliary noma. Hum Pathol. 1991;22(2):172–178.
cirrhosis resembling but unrelated to hepatocellular neoplasms. Virchows Arch A 37. Theise ND, Schwartz M, Miller C, Thung SN. Macroregenerative nodules
Pathol Anat Histopathol. 1993;422(1):17–23. and hepatocellular carcinoma in forty-four sequential adult liver explants with
9. Zhao M, Zhang NX, Du ZY, Laissue JA, Zimmermann A. Three types of liver cirrhosis. Hepatology. 1992;16(4):949–955.
cell dysplasia (LCD) in small cirrhotic nodules are distinguishable by karyometry 38. Borzio M, Fargion S, Borzio F, et al. Impact of large regenerative, low
and PCNA labelling, and their features resemble distinct grades of hepatocellular grade and high grade dysplastic nodules in hepatocellular carcinoma develop-
carcinoma. Histol Histopathol. 1994;9(1):73–83. ment. J Hepatol. 2003;39(2):208–214.
10. Makino Y, Shiraki K, Sugimoto K, et al. Histological features of cirrhosis 39. Kobayashi M, Ikeda K, Hosaka T, et al. Dysplastic nodules frequently
with hepatitis C virus for prediction of hepatocellular carcinoma development; a develop into hepatocellular carcinoma in patients with chronic viral hepatitis and
prospective study. Anticancer Res. 2000;20(5C):3709–3715. cirrhosis. Cancer. 2006;106(3):636–647.
11. Libbrecht L, Craninx M, Nevens F, Desmet V, Roskams T. Predictive value 40. Terasaki S, Kaneko S, Kobayashi K, Nonomura A, Nakanuma Y.
of liver cell dysplasia for development of hepatocellular carcinoma in patients Histological features predicting malignant transformation of nonmalignant
with non-cirrhotic and cirrhotic chronic viral hepatitis. Histopathology. 2001; hepatocellular nodules: a prospective study. Gastroenterology. 1998;115(5):
39(1):66–73. 1216–1222.
12. Borzio M, Bruno S, Roncalli M, et al. Liver cell dysplasia is a major risk 41. Maggioni M, Coggi G, Cassani B, et al. Molecular changes in
factor for hepatocellular carcinoma in cirrhosis: a prospective study. Gastroen- hepatocellular dysplastic nodules on microdissected liver biopsies. Hepatology.
terology. 1995;108(3):812–817. 2000;32(5):942–946.
13. Ganne-Carrie N, Chastang C, Chapel F, et al. Predictive score for the 42. Sun M, Eshleman JR, Ferrell LD, et al. An early lesion in hepatic
development of hepatocellular carcinoma and additional value of liver large cell carcinogenesis: loss of heterozygosity in human cirrhotic livers and dysplastic
dysplasia in western patients with cirrhosis. Hepatology. 1996;23(5):1112–1118. nodules at the 1p36-p34 region. Hepatology. 2001;33(6):1415–1424.
14. Koo JS, Kim H, Park BK, et al. Predictive value of liver cell dysplasia for 43. Tornillo L, Carafa V, Sauter G, et al. Chromosomal alterations in
development of hepatocellular carcinoma in patients with chronic hepatitis B. J hepatocellular nodules by comparative genomic hybridization: high-grade
Clin Gastroenterol. 2008;42(6):738–743. dysplastic nodules represent early stages of hepatocellular carcinoma. Lab
15. Lee RG, Tsamandas AC, Demetris AJ. Large cell change (liver cell Invest. 2002;82(5):547–553.
dysplasia) and hepatocellular carcinoma in cirrhosis: matched case-control 44. Wong CM, Ng IO. Molecular pathogenesis of hepatocellular carcinoma.
study, pathological analysis, and pathogenetic hypothesis. Hepatology. 1997; Liver Int. 2008;28(2):160–174.
26(6):1415–1422. 45. Kondo Y, Kanai Y, Sakamoto M, Mizokami M, Ueda R, Hirohashi S.
16. Natarajan S, Theise ND, Thung SN, Antonio L, Paronetto F, Hytiroglou P. Genetic instability and aberrant DNA methylation in chronic hepatitis and
Large-cell change of hepatocytes in cirrhosis may represent a reaction to cirrhosis—a comprehensive study of loss of heterozygosity and microsatellite
prolonged cholestasis. Am J Surg Pathol. 1997;21(3):312–318. instability at 39 loci and DNA hypermethylation on 8 CpG islands in
17. Borzio M, Trere D, Borzio F, et al. Hepatocyte proliferation rate is a microdissected specimens from patients with hepatocellular carcinoma. Hepa-
powerful parameter for predicting hepatocellular carcinoma development in liver tology. 2000;32(5):970–979.
cirrhosis. Mol Pathol. 1998;51(2):96–101. 46. Oh BK, Chae KJ, Park C, et al. Telomere shortening and telomerase
18. Roncalli M, Borzio M, Brando B, Colloredo G, Servida E. Abnormal DNA reactivation in dysplastic nodules of human hepatocarcinogenesis. J Hepatol.
content in liver-cell dysplasia: a flow cytometric study. Int J Cancer. 1989;44(2): 2003;39(5):786–792.
204–207. 47. Oh BK, Kim YJ, Park YN, Choi KJ, Kim KS, Park C. Quantitative assessment
19. Thomas RM, Berman JJ, Yetter RA, Moore GW, Hutchins GM. Liver cell of hTERT mRNA expression in dysplastic nodules of HBV-related hepatocarcin-
dysplasia: a DNA aneuploid lesion with distinct morphologic features. Hum ogenesis. Am J Gastroenterol. 2006;101(4):831–838.
Pathol. 1992;23(5):496–503. 48. Oh BK, Kim YJ, Park C, Park YN. Up-regulation of telomere-binding
20. El-Sayed SS, El-Sadany M, Tabll AA, Soltan A, El-Dosoky I, Attallah AM. proteins, TRF1, TRF2, and TIN2 is related to telomere shortening during human
DNA ploidy and liver cell dysplasia in liver biopsies from patients with liver multistep hepatocarcinogenesis. Am J Pathol. 2005;166(1):73–80.
cirrhosis. Can J Gastroenterol. 2004;18(2):87–91. 49. Lee YH, Oh BK, Yoo JE, et al. Chromosomal instability, telomere
21. Terris B, Ingster O, Rubbia L, et al. Interphase cytogenetic analysis reveals shortening, and inactivation of p21(WAF1/CIP1) in dysplastic nodules of hepatitis
numerical chromosome aberrations in large liver cell dysplasia. J Hepatol. 1997; B virus-associated multistep hepatocarcinogenesis. Mod Pathol. 2009;22(8):
27(2):313–319. 1121–1131.
22. Zondervan PE, Wink J, Alers JC, et al. Molecular cytogenetic evaluation of 50. Park YN, Kojiro M, Di Tommaso L, et al. Ductular reaction is helpful in
virus-associated and non-viral hepatocellular carcinoma: analysis of 26 defining early stromal invasion, small hepatocellular carcinomas, and dysplastic
carcinomas and 12 concurrent dysplasias. J Pathol. 2000;192(2):207–215. nodules. Cancer. 2007;109(5):915–923.
23. Paradis V, Laurendeau I, Vidaud M, Bedossa P. Clonal analysis of 51. Nakashima O, Sugihara S, Kage M, Kojiro M. Pathomorphologic
macronodules in cirrhosis. Hepatology. 1998;28(4):953–958. characteristics of small hepatocellular carcinoma: a special reference to small
24. Kim H, Oh BK, Roncalli M, et al. Large liver cell change in hepatitis B hepatocellular carcinoma with indistinct margins. Hepatology. 1995;22(1):101–
virus-related liver cirrhosis. Hepatology. 2009;50(3):752–762. 105.
25. Park YN, Roncalli M. Large liver cell dysplasia: a controversial entity. J 52. Kojiro M, Nakashima O. Histopathologic evaluation of hepatocellular
Hepatol. 2006;45(5):734–743. carcinoma with special reference to small early stage tumors. Semin Liver Dis.
26. Deugnier YM, Charalambous P, Le Quilleuc D, et al. Preneoplastic 1999;19(3):287–296.
significance of hepatic iron-free foci in genetic hemochromatosis: a study of 185 53. Roncalli M, Park YN, Di Tommaso L. Histopathological classification of
patients. Hepatology. 1993;18(6):1363–1369. hepatocellular carcinoma. Dig Liver Dis. 2010;42(suppl 3):S228–S234.
27. International Consensus Group for Hepatocellular Neoplasia. Pathologic 54. Kondo F, Kondo Y, Nagato Y, Tomizawa M, Wada K. Interstitial tumour
diagnosis of early hepatocellular carcinoma: a report of the international cell invasion in small hepatocellular carcinoma: evaluation in microscopic and
consensus group for hepatocellular neoplasia. Hepatology. 2009;49(2):658–664. low magnification views. J Gastroenterol Hepatol. 1994;9(6):604–612.

714 Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park
55. Nakashima Y, Nakashima O, Hsia CC, Kojiro M, Tabor E. Vascularization 65. Chuma M, Sakamoto M, Yamazaki K, et al. Expression profiling in
of small hepatocellular carcinomas: correlation with differentiation. Liver. 1999; multistage hepatocarcinogenesis: identification of HSP70 as a molecular marker
19(1):12–18. of early hepatocellular carcinoma. Hepatology. 2003;37(1):198–207.
56. Capurro M, Wanless IR, Sherman M, et al. Glypican-3: a novel serum and 66. Christa L, Simon MT, Flinois JP, Gebhardt R, Brechot C, Lasserre C.
histochemical marker for hepatocellular carcinoma. Gastroenterology. 2003; Overexpression of glutamine synthetase in human primary liver cancer.
125(1):89–97. Gastroenterology. 1994;106(5):1312–1320.
57. Midorikawa Y, Ishikawa S, Iwanari H, et al. Glypican-3, overexpressed in 67. Zucman-Rossi J, Benhamouche S, Godard C, et al. Differential effects of
hepatocellular carcinoma, modulates FGF2 and BMP-7 signaling. Int J Cancer. inactivated Axin1 and activated beta-catenin mutations in human hepatocellular
2003;103(4):455–465. carcinomas. Oncogene. 2007;26(5):774–780.
58. Moriguchi H, Sato C. The values and limitations of glypican-3 as a novel 68. Osada T, Sakamoto M, Nagawa H, et al. Acquisition of glutamine
tumor marker for hepatocellular carcinoma from clinical and economic view synthetase expression in human hepatocarcinogenesis: relation to disease
points. Gastroenterology. 2004;127(2):679–680. recurrence and possible regulation by ubiquitin-dependent proteolysis. Cancer.
59. Kandil D, Leiman G, Allegretta M, et al. Glypican-3 immunocytochemistry 1999;85(4):819–831.
in liver fine-needle aspirates: a novel stain to assist in the differentiation of benign 69. Moorman AF, de Boer PA, Geerts WJ, van den Zande L, Lamers WH,
and malignant liver lesions. Cancer. 2007;111(5):316–322. Charles R. Complementary distribution of carbamoylphosphate synthetase
60. Di Tommaso L, Franchi G, Park YN, et al. Diagnostic value of HSP70, (ammonia) and glutamine synthetase in rat liver acinus is regulated at a
glypican 3, and glutamine synthetase in hepatocellular nodules in cirrhosis. pretranslational level. J Histochem Cytochem. 1988;36(7):751–755.
Hepatology. 2007;45(3):725–734. 70. Bioulac-Sage P, Rebouissou S, Thomas C, et al. Hepatocellular adenoma
61. Abdul-Al HM, Makhlouf HR, Wang G, Goodman ZD. Glypican-3 subtype classification using molecular markers and immunohistochemistry.
expression in benign liver tissue with active hepatitis C: implications for the Hepatology. 2007;46(3):740–748.
diagnosis of hepatocellular carcinoma. Hum Pathol. 2008;39(2):209–212. 71. Park YN, Chae KJ, Kim YB, Park C, Theise N. Apoptosis and proliferation
62. Nakatsura T, Kageshita T, Ito S, et al. Identification of glypican-3 as in hepatocarcinogenesis related to cirrhosis. Cancer. 2001;92(11):2733–2738.
a novel tumor marker for melanoma. Clin Cancer Res. 2004;10(19):6612– 72. Theise ND, Fiel IM, Hytiroglou P, et al. Macroregenerative nodules in
6621. cirrhosis are not associated with elevated serum or stainable tissue alpha-
63. Garrido C, Gurbuxani S, Ravagnan L, Kroemer G. Heat shock proteins: fetoprotein. Liver. 1995;15(1):30–34.
endogenous modulators of apoptotic cell death. Biochem Biophys Res Commun. 73. Bruix J, Sherman M. Management of hepatocellular carcinoma. Hepatol-
2001;286(3):433–442. ogy. 2005;42:1208–1236.
64. Jolly C, Morimoto RI. Role of the heat shock response and molecular 74. Di Tommaso L, Destro A, Seok JY, et al. The application of markers
chaperones in oncogenesis and cell death. J Natl Cancer Inst. 2000;92(19):1564– (HSP70 GPC3 and GS) in liver biopsies is useful for detection of hepatocellular
1572. carcinoma. J Hepatol. 2009;50:746–754.

Arch Pathol Lab Med—Vol 135, June 2011 Precursor and Early Lesions of Hepatocellular Carcinoma—Park 715

You might also like