You are on page 1of 14

THE ANATOMICAL RECORD 291:721–734 (2008)

Vascular Changes in Hepatocellular


Carcinoma
ZHEN FAN YANG AND RONNIE T.P. POON*
Centre for Cancer Research, Department of Surgery, Queen Mary Hospital,
The University of Hong Kong, Hong Kong, China

ABSTRACT
Hepatocellular carcinoma (HCC) is one of the most vascular solid
tumors, in which angiogenesis plays an important role. The status of
angiogenesis in HCC correlates with the disease progression and progno-
sis, and thus provides a potential therapeutic target. This review summa-
rizes the vascular changes and molecular and cellular basis of angiogene-
sis in HCC. Development of HCC is characterized by arterialization of its
blood supply and sinusoidal capillarization. Vascular endothelial growth
factor (VEGF) is a potent angiogenic factor that plays a critical role in
mediating angiogenesis in HCC. The VEGF can function on various types
of cells, such as endothelial cells, hepatic stellate cells, endothelial pro-
genitor cells and hemangiocytes, to induce vascular changes in HCC.
Therefore, blockade of VEGF-mediated pathways, either by anti-VEGF
neutralizing antibody or tyrosine kinase inhibitors that target VEGF
receptors, suppresses carcinogenesis and angiogenesis in HCC. In addi-
tion to VEGF, several other angiogenic factors in HCC have recently been
identified. These factors can also regulate angiogenic processes through
interaction with VEGF or VEGF-independent pathways. Despite the fact
that treatment of HCC remains a tough task due to lack of effective sys-
temic therapy, antiangiogenic therapy has already entered clinical trials
in HCC patients and sheds light on a promising novel treatment for this
disease. Anat Rec, 291:721–734, 2008. Ó 2008 Wiley-Liss, Inc.

Key words: neoangiogenesis; vascular endothelial growth fac-


tor; hepatocellular carcinoma

Hepatocellular carcinoma (HCC) is one of the five lular mechanisms that modulate the development and
most common malignancies worldwide, with an increas- progression of HCC is crucial for the design of therapeu-
ing incidence due to hepatitis B and C viral infection tic strategies.
(Llovet et al., 2003; McMahon, 2005; El-Serag, 2004). Hepatocellular carcinoma is one of the most vascular
HCC is characterized as a highly vascularized solid tu- solid tumors, in which angiogenesis plays an important
mor with rapid growth rate and poor prognosis. Hepatic role in its development, progression, and metastasis.
resection and liver transplantation are the two main- Vascular changes in HCC are characterized by arteriali-
stays that may cure HCC, but the long-term patient sur-
vival is dependent on the stage of tumor at the time of
diagnosis (Llovet et al., 1999; Arii et al., 2000). Most Grant sponsor: The University of Hong Kong.
patients with HCC present with advanced stage tumors *Correspondence to: Ronnie T.P. Poon, Department of Sur-
beyond surgical treatment, and chemotherapy and radio- gery, The University of Hong Kong, Queen Mary Hospital, 102
therapy have limited efficacy in these cases (Groupe Pokfulam Road, Hong Kong, China. Fax: 852-2817-5475.
d’Etude et de Traitement du Carcinome Hépatocellu- E-mail: poontp@hkucc.hku.hk
laire, 1995; Lo et al., 2002; Raoul et al., 1997). Cur- Received 29 April 2007; Accepted 19 December 2007
rently, there is no proven effective systemic chemother- DOI 10.1002/ar.20668
apy for HCC patients with metastasis (Bruix et al., Published online in Wiley InterScience (www.interscience.wiley.
2005). Therefore, understanding the molecular and cel- com).

Ó 2008 WILEY-LISS, INC.


722 YANG AND POON

Fig. 1. Determination of microvessel density (MVD) in hepatocellular carcinoma (HCC) by CD34 immu-
nohistochemical staining. A significant higher level of MVD was detected in the tumor tissue than the
adjacent nontumorous tissue. T, tumor tissue; NT, nontumorous tissue. Arrows point to microvessels
formed in the tumor tissue. Copyright from Poon et al. (2002).

zation and sinusoidal capillarization. Recently, accumu- substantial amount of nutrient and oxygen from the cir-
lating data have provided insights into the molecular culation. Hence, neoangiogenesis is required to provide
mechanisms that are related to angiogenesis in HCC the nutrient and oxygen for tumor cell survival. Another
(Zhao et al., 2003; Cervello and Montalto, 2006; Lee consequence of neoangiogenesis is that tumor cells may
et al., 2006). Antiangiogenic strategies that target angio- enter into the circulation and develop distal metastasis
genic molecules and inhibit activation of various types of (Gwak et al., 2005; Tanaka et al., 2006). In fact, the
cells that participate in the angiogenic processes become expression of markers for microvessel density (MVD)
potential therapies for HCC. In this review, we summa- has been reported to be associated with the development
rize the recent findings in molecular and cellular mecha- and progression of HCC (Fig. 1). A study has demon-
nisms of angiogenesis in HCC, and the potential implica- strated that the expression of CD31, CD34, and BNH9
tions on the design of therapeutic strategies. in endothelial cells was absent in cirrhotic liver and dys-
plastic nodules, but became detectable in human HCC
tumor tissues (Frachon et al., 2001). However, in an ex-
SIGNIFICANCE OF ANGIOGENESIS IN HCC perimental cirrhotic model induced by bile duct ligation,
Angiogenesis is switched on when precancerous up-regulation of CD31 was detected, suggesting that
lesions, also named dysplastic nodules, develop into angiogenesis might also be present in the premalignant
HCC, and the process of angiogenesis carries on with lesions (Geerts et al., 2006). In established HCC, MVD
the progression of the tumor (Poon et al., 2001). Due to has been shown to impact on prognosis of patients. In a
the rapid growth pattern of HCC, tumor cells require a previous study from our group, high tumor MVD was a
VASCULAR CHANGES IN HCC 723

Fig. 2. Contrast computed tomography scans showing the typical enhancement pattern of HCC. A: A
hypodense lesion in the right lobe of the liver in noncontrast scan. B: The lesion is densely enhanced in
arterial phase, indicating its predominant arterial blood supply. C: The lesion becomes hypoattenuated in
the portal venous phase. An arrow points to a tumor nodule in the right lobe of the liver.

significant prognostic factor for tumor recurrence after showed that well-differentiated HCC with a diameter
resection of early HCC < 5 cm in diameter (Poon et al., less than 3 cm often received preferential portal venous
2002). blood, whereas moderately and poorly differentiated
ones were all supplied with arterial blood. When tumors
grow in size beyond 3 cm, most HCCs including well-dif-
VASCULAR CHANGES ASSOCIATED WITH
ferentiated ones generally develop a predominant arte-
DEVELOPMENT OF HCC rial blood supply.
The liver has dual blood supply from the portal vein Sinusoidal capillarization, another neoangiogenic
and the hepatic artery. HCC usually develops blood sup- process, has also been found to be common in HCC and
ply predominantly from the arterial system due to its occurs even in early small HCC (Kin et al., 1994).
high requirement of oxygen during progression. The Sinusoidal capillarization involves transformation of
presence of new unpaired arteries not accompanied by fenestrated hepatic sinusoids into continuous capillaries,
bile duct has been shown to be able to differentiate coupled with collagenization of the extravascular spaces
neoplastic nodules from regenerative cirrhotic nodules of Disse and deposition of laminin and basement mem-
(Himeno et al., 1994; Ueda et al., 1992; Matsui, 2001). branes near the endothelial cells and hepatocytes
The number of unpaired arteries gradually increases (Schaffner and Poper, 1963; Kin et al., 1994). This vas-
from cirrhotic nodules (lowest) to low- and high-grade cular change also accompanies the development of cir-
dysplastic nodules and with the greatest numbers in rhosis. The sinusoidal capillarization was characterized
HCC, particularly in HCC smaller than 3 cm in diame- by the expression of CD34, a marker which is absent in
ter. Using smooth muscle actin as a marker to label the normal sinusoidal endothelium (Couvelard et al.,
arteries in the liver parenchyma, a study demonstrated 1996; Frachon et al., 2001). While arterialization and si-
that unpaired arteries were histologically useful for dis- nusoidal capillarization both occur in HCC, no correla-
tinguishing dysplastic nodules from large cirrhotic nod- tion has been identified between the location of arteriali-
ules (Park et al., 1998). The arterialization of HCC pro- zation and sinusoidal capillarization (Park et al., 1998).
vides a characteristic vascular enhancement pattern in The process of sinusoidal capillarization also appears to
contrast computed tomography (CT) scan, which is use- be related to tumor differentiation, with mixed sinusoi-
ful as a diagnostic feature of HCC to differentiate it dal and capillary types of microvessels present in well-
from other liver tumors such as secondary metastatic differentiated HCC, whereas capillary type is the only
tumors. Typically, HCC manifests as a hyperattenuated type observed in poorly differentiated HCC (Yamamoto
lesion against a background of minimally enhanced liver et al., 2001). A recent study evaluated the correlation of
parenchyma in the arterial phase because the tumor is arterialization and sinusoidal capillarization by immuno-
supplied by the hepatic artery and is hypervascular, staining histologically with the degree of arterial con-
whereas on delayed phase images, the HCC becomes low trast enhancement in dynamic CT scan in HCC (Kim
attenuating compared with the surrounding liver paren- et al., 2005). The study identified a positive correlation
chyma owing to early washout of contrast (Figures 2A, between the number of unpaired arteries in nodular
B, and C). However, not all HCCs develop the typical ar- HCC and the degree of contrast enhancement in the ar-
terial blood supply. Some small HCCs can remain ‘‘hypo- terial phase by CT imaging. In contrast, there was no
vascular’’ on CT scan. A previous study has shown that correlation between the degree of sinusoidal capillariza-
the degree of arterialization correlated with tumor dif- tion and the degree of contrast enhancement in the arte-
ferentiation of HCC (Yamamoto et al., 2001). The study rial phase.
724 YANG AND POON

The exact significance of arterialization and sinusoidal et al., 2004). The levels of VEGF expression during the
capillarization in the process of angiogenesis of HCC development of HCC have been shown to be associated
remains to be elucidated in further studies. Other vascu- with the increase of unpaired arteries and sinusoidal
lar changes associated with neovascularization include capillarization (Park et al., 2000). Increasing evidence of
increase in MVD and development of heterogeneous up-regulated expression of VEGF in small HCC suggests
immature vessels with increased permeability, all con- the importance of VEGF not only in tumor progression
tributing to the contrast enhancement pattern of HCC but also in hepatocarcinogenesis (Iavarone et al., 2007).
in imaging studies. It is now possible to use perfusion This hypothesis has been proven in a diethylnitros-
magnetic resonance imaging (MRI) to study changes in amine-induced murine hepatocarcinogenesis model
these parameters (Huwart et al., 2007). A dynamic mon- (Yoshiji et al., 2004). The expression of VEGF in HCC
itoring of vascular changes may play an important role has been found to be associated with the clinicpathologi-
in monitoring the response of the tumor to antiangio- cal features of HCC patients in several studies. Tori-
genic therapy. mura et al. (1998) showed a significant association
between higher levels of VEGF protein by immunostain-
ing and poorly differentiated HCC. A higher level of
MAJOR MOLECULAR PATHWAYS THAT VEGF expression in HCC is also associated with a
MEDIATE ANGIOGENESIS IN HCC higher proliferation index, poor encapsulation of tumors,
Tumor cells release angiogenic factors to initiate the and venous tumor emboli or portal vein thrombosis
process of angiogenesis (Sun and Tang, 2004). Endothe- (Chow et al., 1997; Li et al., 1998). In addition, circulat-
lial cells express a variety of receptors that bind to ing VEGF levels before treatment could predict disease
angiogenic molecules secreted by the tumor cells. It has outcome after a variety of treatments, including hepatic
been postulated for many years that neoangiogenesis resection, radiofrequency ablation, transcatheter arterial
originates from migration of endothelial cells in the pre- chemoembolization (TACE; Poon et al., 2003a, 2004a,
existing vessels in tumor-surrounding tissues. Accumu- 2007). Parallel to VEGF, VEGFR expression is also asso-
lated evidence suggests that some of the tumor neoves- ciated with the clinical outcome of HCC patients,
sels may be derived from circulating bone marrow- although only the level of VEGFR1 mRNA significantly
derived endothelial progenitor cells (EPCs; Shi et al., correlated with the VEGF mRNA level in tumor tissues
1998; Asahara et al., 1999), although some other studies and intrahepatic metastasis in HCC (Ng et al., 2001).
propose a limited role of EPCs in tumor neovasculariza- Another study also reported that VEGFR1 expression in
tion (Yung et al., 2004; Peters et al., 2005). tumor tissues correlated with CD31 expression, a
In addition to tumor cells, other types of cells, such as marker for tumor microvessels (Dhar et al., 2002). The
endothelial cells and infiltrating lymphocytes, can also above findings suggest that VEGFR1 may play a more
secrete angiogenic factors. On the other hand, endoge- important role in VEGF/VEGFR pathway in mediating
nous presence of antiangiogenic factors has also been angiogenesis of HCC.
identified. Therefore, tumor angiogenesis is a dynamic In addition to VEGFR1 and VEGFR2, VEGF also
process regulated by the balance between angiogenic interacts with the members of the neuropilin family,
and antiangiogenic pathways. In our previous review, neuropilin-1 and neuropilin-2, to regulate angiogenesis.
we have listed some of the relatively well-characterized Moreover, these neuropilin members can act together
angiogenic and antiangiogenic factors, with a focus on with VEGFR1 and VEGFR2 to mediate signal transduc-
their potential roles in HCC (Pang and Poon, 2006). The tion (Staton et al., 2007). Little evidence has shown how
roles of a few more important factors are briefly neuropilins participate in the angiogenic process of
described as follows, including updated information from HCC. However, some studies on other types of cancers
the recent literature. reported that neuropilins that are expressed on tumor
cells could directly modulate cell survival, proliferation,
and invasion (Hong et al., 2007; Fukasawa et al., 2007).
Vascular Endothelial Growth Factor
In endothelial cells, neuropilin-1 has been identified to
Vascular endothelial growth factor (VEGF) is the most be a coreceptor of VEGFR2 and function on angiogenic
well-studied angiogenic factor in HCC. The angiogenic behavior by means of an enhancement of the VEGFR-2
effects of VEGF are achieved by binding to its receptors, phosphorylation threshold (Favier et al., 2006). Hence,
VEGFR1 (Flt-1) and VEGFR2 (Flk-1), expressed on en- exploring the potential role of the neuropilin family on
dothelial cells. Interaction between VEGF and VEGFR HCC, either alone or combined with other VEGFRs, is
initiate several signaling pathways, such as Akt/PI3K/ warranted, and may shed light on a novel antiangio-
MAPK pathway, resulting in proliferation, migration, genic therapy for this malignancy.
and invasion of endothelial cells (Gerber et al., 1998; The VEGF promoter contains a hypoxia-responsive
Ahmad et al., 2006; Vogel et al., 2007). In addition to en- element, which is regulated by hypoxia inducible factor-
dothelial cells, recent findings have demonstrated the 1a (HIF-1a), a transcription factor. Activated HIF-1a
expression of VEGFR in tumor cells, proposing an auto- under stress, such as hypoxia, can bind to hypoxia-re-
crine loop of VEGF/VEGFR in tumor cells (Masood sponsive elements expressed in several pro-survival and
et al., 2001; Weigand et al., 2005). pro-proliferation genes, leading to up-regulation of HIF-
Mise et al. (1996) first reported the expression of 1a downstream proteins (Harris, 2002; Brown and Wil-
VEGF in HCC and detected significantly higher levels of son, 2004). HIF-1a–mediated up-regulation of VEGF is
VEGF mRNA and protein in the tumor tissues than the pivotal for hypoxia-induced angiogenesis (von Marschall
nontumorous counterpart. Subsequent studies reported et al., 2001; Yang et al., 2004), and a higher level of
similar findings of high VEGF expression at both mRNA HIF-1a is also associated with a poorer prognosis of
and protein levels in HCC (Torimura et al., 2004; Yasuda HCC patients (Huang et al., 2005). Transcriptional acti-
VASCULAR CHANGES IN HCC 725
vation of HIF-1a can also be induced by hepatitis B vi- angiopoietin-2 and VEGF. Based on the above findings,
rus X protein (Yoo et al., 2003), indicating the potential the expression of angiopoietins might be a potent param-
interaction among viral infection, HIF-1a and VEGF up- eter to predict the outcome of HCC patients. Indeed,
regulation, and hepatocarcinogenesis. Mitsuhashi et al. (2003) reported that HCC patients har-
VEGF can directly function on HCC tumor cells, as boring tumors with high angiopoietin-2/1 mRNA ratio
VEGFRs are also expressed by tumor cells. Our previous had a shorter survival than those with tumors having
study demonstrated that stimulation of HCC cell lines low angiopoietin-2/1 mRNA ratio. A study by Wada et al.
by exogenous VEGF recombinant protein could stimu- (2006) also revealed that a higher level of angiopoietin-2
late tumor cell proliferation (Liu Y et al., 2005), suggest- in the tumors was associated with a shorter disease-free
ing the presence of an autocrine loop in HCC tumor survival in HCC patients with hepatic resection.
cells. Moreover, VEGF may also have an angiogenesis- However, the mechanism responsible for the regula-
independent effect on HCC cell invasion. A study by tion of angiopoietin-2 expression appears to be different
Schmitt et al. (2004) showed that VEGF induced a from that of VEGF in HCC. Unlike VEGF, hypoxia does
marked loss of pseudocanaliculi and disruption of occlu- not regulate the expression of angiopoietin-2 in HCC
din-delineated tight junctions in tumor cells. Sections (Sugimachi et al., 2003). In addition, the role of angio-
from surgically removed tumor specimens showed VEGF poietin-1 in HCC is controversial. Sugimachi et al.
expression in the tumor and occludin disassembly in (2003) showed that angiopoietin-1 was more frequently
normal liver parenchyma next to the tumor, suggesting expressed in normal liver, whereas angiopoietin-2 was
that VEGF may enhance invasion of HCC cells into nor- more frequently expressed in HCC. Hence, a balance
mal liver parenchyma. between angiopoietin-1 and angiopoietin-2 may play a
VEGF has been reported to be overexpressed in cirrho- more important role in mediating angiogenesis in HCC.
sis, a well-recognized precancerous lesion of HCC. There-
fore, VEGF is also postulated to play an important role Epidermal Growth Factor
in the development of liver cirrhosis (Deli et al., 2005;
Medina et al., 2005). However, there is no solid evidence The epidermal growth factor (EGF) family contains six
showing the relationship among VEGF expression, cir- ligands, that is, EGF, tumor growth factor (TGF) -a,
rhosis and the development of HCC, although a recent amphiregulin, betacellulin (BTC), heparin-binding EGF-
study demonstrated that a higher level of VEGF165 like growth factor and epiregulin, and four receptors,
mRNA in noncancerous liver tissue correlated signifi- that is, EGFR, ErbB2, ErbB3, and ErbB4 (Salomon
cantly with a higher risk of HCC recurrence (Sheen et al., 1995; Riese and Stern, 1998). All the EGF family
et al., 2005). The significance of angiogenesis in cirrhosis members share common features including an extracellu-
and its relationship with hepatocarcinogenesis in cir- lar ligand binding domain, a transmembrane domain,
rhotic liver are areas that need further investigation. and an intracellular tyrosine kinase domain. The interac-
tion between any of the six ligands to EGFR activates
cell signaling pathways that lead to cell proliferation and
Angiopoietins differentiation. HCC tumor cells secrete EGF, which may
Angiopoietins constitute another group of angiogenic act on multiple EGFRs expressed by tumor cells, and
molecules. Different members in the angiopoietin family thus achieve an autocrine cascade (Zhang et al., 2004).
play different roles in the process of angiogenesis. For Recent evidence has shown the potential role of EGF
instance, angiopoietin-1 provides survival signals to family in angiogenesis of HCC. Moon et al. (2006) dem-
endothelial cells and promotes recruitment of pericytes onstrated that BTC mRNA was prominently expressed
and smooth muscle cells to form mature blood vessels, by tumor cells in human HCC specimens, whereas
whereas angiopoietin-2 functions as a natural antagonist EGFR was mainly expressed by sinusoidal endothelial
of angiopoietin-1 (Tanaka et al., 1999). The presence of cells. In addition, a strong correlation between BTC
angiopoietin-2 enhances the sensitivity of endothelial expression in tumor cells and EGFR expression in tumor
cells to other angiogenic factors, such as VEGF. Higher endothelial cells was identified. Furthermore, BTC was
levels of angiopoietin-2 expression have been detected in secreted by HCC cell lines, suggesting the potential
the tumor tissues than in the nontumorous tissues in interaction between BTC and EGFR in tumor angiogene-
HCC. The level of angiopoietin-2 was found to be associ- sis. Targeting EGFR signaling has been recently demon-
ated with the clinicopathological parameters of HCC strated to be effective in blockade of angiogenesis in ani-
patients, and the ratio between angiopoietin-2 and mal models (Ueda et al., 2006).
angiopoietin-1 indicated the status of tumor angiogene-
sis (Zhang et al., 2006). A study by Torimura et al. Platelet-derived Endothelial Cell
(2004) also showed a positive correlation between angio-
Growth Factor
poietin-1 and angiopoietin-2 expression with tumor de-
differentiation. By using immunohistochemistry, angio- Platelet-derived endothelial cell growth factor (PD-
poietin-1 and angiopoietin-2 were detected in HCC cells, ECGF) is another angiogenic molecule that can promote
hepatic stellate cells, and smooth muscle cells, whereas endothelial cell migration, but it has little effect on cell
their receptor Tie-2 was detected in endothelial cells, he- proliferation. Its angiogenic effect is mediated by the
patic stellate cells and smooth muscle cells, suggesting release of 2-deoxy-d-ribose as a result of breakdown of
that multiple cell types are involved in the angiopoietin/ thymidine by its thymidine phosphorylase activity (Grif-
Tie-2 signaling pathways to mediate tumor angiogenesis. fiths and Stratford, 1997). Up-regulation of PD-ECGF
Direct interaction between angiopoietin-2 and VEGF has has been identified in HCC tumor tissues compared with
been identified using a murine HCC model (Yoshiji the adjacent nontumorous tissues, suggesting the poten-
et al., 2005), suggesting a synergistic effect between tial role of PD-ECGF in tumor angiogenesis. However,
726 YANG AND POON

in a study, the expression of PD-ECGF in tumor tissues and MVD in HCC tumor tissue (Cheng et al., 2004). Our
did not correlate with the outcome of HCC patients data also showed that tumor cytosolic COX-2 levels in
(Morinaga et al., 2003). Interestingly, another study HCC correlated with VEGF levels and features of inva-
revealed that a higher level of thymidine phosphorylase siveness such as venous invasion and microsatellite
activity in nontumorous tissues adjacent to tumor tis- lesions (Tang et al., 2005). These findings suggest an
sues was associated with an earlier tumor recurrence angiogenic effect of COX-2 in HCC.
after hepatic resection, suggesting a prognostic value of Other angiogenic factors, such as macrophage migra-
thymidine phosphorylase activity in nontumorous livers tion inhibitory factor (Ren et al., 2003; Hisai et al.,
(Ezaki et al., 2005). Another two studies have reported a 2003) and thrombospondin-1 (Poon et al., 2004b), might
positive correlation between PD-ECGF expression in the also play important roles in mediating tumor angiogene-
tumor tissue and portal vein tumor thrombosis in HCC sis in HCC, though further studies are needed to clarify
(Zhou et al., 2000; Guo et al., 2001). the molecular pathways of these molecules in angiogene-
sis. With the development of gene and protein expres-
sion profiling technologies, novel angiogenic factors,
Other Angiogenic Factors
such as pituitary tumor transforming gene 1 (PTTG1),
Several other factors secreted by tumor cells have also brain-derived neurotrophic factor (BDNF) and stromal-
been reported to participate in the angiogenic process. derived factor-1 (SDF-1), have been identified. Fujii
Fibroblast growth factors, a family of heparin-binding et al. (2006) reported that PTTG1 was remarkably over-
growth factors, exhibit potent angiogenic properties. Ba- expressed in HCC tumor tissues, as compared with non-
sic fibroblast growth factor (bFGF) could be secreted by tumorous tissues. In addition, PTTG1 mRNA expression
both tumor cells and endothelial cells, suggesting the significantly correlated with MVD. Finally, PTTG1
presence of both autocrine and paracrine cascades in mRNA was an independent prognostic factor for disease-
bFGF-mediated angiogenic activities (Schweigerer et al., free survival of HCC patients. These findings suggest
1987). El-Assal et al. (2001) demonstrated that the that PTTG1 might be involved in the development of
expression of bFGF protein in human HCC specimens HCC through promotion of angiogenesis.
correlated with tumor angiogenesis. HCC cells also Our previous animal study has detected an increasing
secrete platelet-derived growth factor (PDGF), another production of BDNF by tumor cells during HCC tumor
angiogenic factor, to function on endothelial cells which development (Yang et al., 2005). As endothelial cells and
express PDGFR, resulting in an increased metastatic bone marrow-derived progenitor cells express BDNF re-
potential (Zhang et al., 2005). A recent report demon- ceptor, tyrosine kinase receptor B (TrkB), it is reasonable
strated that PDGF links TGF-beta signaling to nuclear to hypothesize that BDNF might exhibit angiogenic
beta-catenin accumulation for HCC progression (Fischer effects through recruitment of endothelial cells and bone
et al., 2007). Inducible nitric oxide synthase (iNOS) is marrow-derived progenitors. In fact, some studies have
also regarded to be an angiogenic factor, supported by shown the presence of autocrine and paracrine loops of
the evidence that its expression was positively correlated BDNF in endothelial cells (Kim et al., 2004), and BDNF
with MVD and tumor recurrence after hepatic resection, could stimulate mobilization of TrkB1 progenitor cells to
and its angiogenic effects seemed to be mediated by ma- the site of hypoxia to promote revascularization (Kermani
trix metalloproteinase related pathways (Sun et al., et al., 2005). In addition, BDNF/TrkB interaction could
2005). However, detailed mechanism studies are still up-regulate HIF-1a and VEGF expression in tumor cells
required to investigate the role of inducible nitric oxide (Nakamura et al., 2006), providing another clue that
synthase in HCC angiogenesis. BDNF might be a tumor angiogenic factor. Therefore, fur-
Our previous study showed that the expression of tis- ther studies are warranted to explore the expression of
sue factor, a hemostatic protein, correlated with tumor BDNF in relation to angiogenic parameters in HCC.
angiogenesis and tumor invasiveness (Poon et al., SDF-1 is a recently identified angiogenic factor that
2003b), suggesting that clotting-dependent induction of could recruit endothelial progenitors to the site where
tumor angiogenesis is mediated by tissue factor-induced neovasculature is formed (Aghi et al., 2006). Because
generation of thrombin and subsequent deposition of a bone marrow-derived progenitors (CXCR41Flt-11
cross-linked fibrin network, which provides a proangio- hemagiocytes) express CXCR4, a receptor of SDF-1, the
genic matrix that facilitates blood vessel infiltration. chemotaxic role of SDF-1 seems to be more prominent in
Interleukin-8 (IL-8) is another potential angiogenic these endothelial progenitors. However, available studies
factor that is expressed by various types of tumor cells about SDF-1 and CXCR4 in HCC mainly focused on
including HCC (Shin et al., 2002; Azenshtein et al., CXCR41 tumor cell migration and metastasis (Schi-
2005). However, controversial findings have been manski et al., 2006; Sutton et al., 2007). The role of
reported about the angiogenic property of IL-8 in HCC. SDF-1/CXCR4 interaction in angiogenesis of HCC
One study demonstrated that increased expression of IL- deserves further studies.
8 in human HCC specimens was associated with venous
and bile duct invasion, and IL-8 produced by HCC cell
Antiangiogenic Factors
lines stimulated proliferation of human umbilical vein
cells (Akiba et al., 2001). In contrast, a more recent It has been postulated that physiological angiogenesis
study reported that the expression of IL-8 in human needs a balanced network between angiogenic and anti-
HCC was related to vascular invasion but not tumor angiogenic cascades, whereas tumor angiogenesis is an
angiogenesis as assessed by MVD (Kubo et al., 2005). imbalance between them (Zhao et al., 2003). However,
Cyclo-oxygenase (COX-2) contributes to several physi- there are few data about antiangiogenic factors in tumor
ological and pathological processes. COX-2 expression angiogenesis in HCC. A study has shown that angiogen-
has been found to be associated with VEGF expression esis in HCC depends on a net balance between VEGF
VASCULAR CHANGES IN HCC 727
and matrix metalloelastase, a matrix metalloproteinase sial whether MVD evaluated by CD34 or vWF staining
responsible for the generation of the natural angiogene- provides better prognostic information in HCC. The vari-
sis inhibitor angiostatin (Gorrin-Rivas et al., 2000). ation in results among the studies may be partly related
Endostatin is another important endogenous angiogene- to the subjective bias in counting microvessels irrespec-
sis inhibitor. Musso et al. (2001) demonstrated that both tive of the endothelial markers used.
long form and short form of endostatin could be detected Recently, CD105 (endoglin), a new endothelial cell
in HCC cells and cancerous stromal cells. Lower mRNA marker which may represent the proliferation status of
level of tissue collagen XVIII (the precursor of endosta- endothelial cells, has been shown to be superior to CD34
tin) in tumor tissues was associated with larger tumor in other cancers as an angiogenesis marker (Tanaka
size and higher MVD, suggesting that low endostatin et al., 2001). However, a study by our group showed that
expression in HCC may be associated with increased CD105 was positively stained only in a subset of micro-
angiogenic activity and tumor progression. vessels in HCC compared with CD34, and MVD by
CD105 staining did not provide significant prognostic in-
formation in HCC (Ho et al., 2005a). In contrast, other
CELLULAR BASIS OF ANGIOGENESIS IN HCC studies reported a significant correlation between CD105
Tumor cells secrete pro-angiogenic factors that attract staining and VEGF expression in HCC tumor tissues,
various types of cells to the site where angiogenesis is and suggested that CD105 could serve as an independ-
needed. Previous studies have suggested that migration ent prognostic marker for patient survival (Yang et al.,
of endothelial cells from adjacent nontumorous tissues to 2006; Yao et al., 2007). There are several drawbacks of
tumor tissues contribute mostly to the formation of tumor using MVD in quantifying angiogenesis in tissue sec-
neovasculature. However, recent studies have identified tions that have hindered its potential for clinical appli-
the potential role of other cell types, such as circulating cation. They include bias in selecting the areas for
endothelial cells, hepatic stellate cells (HSCs), bone mar- counting, subjective errors in counting, conflicting data
row-derived EPCs and hemangiocytes, in neoangiogene- on the predictive value of various MVD markers, and
sis, though molecular mechanisms that mediate the lack of universally accepted criteria of assessment.
recruitment of these types of cells remain largely unclear.
Hepatic Stellate Cells (HSCs)
Tumor Endothelial Cells HSCs play an essential role in the development of
Endothelial cells express a variety of receptors of liver fibrosis. Some studies demonstrated that this type
angiogenic factors, including VEGFRs (Ryschich et al., of cells might also participate in the development of dys-
2006), Tie-2 (angiopoietin receptor; Yu, 2005), EGFR plastic nodules and hepatocarcinogenesis (Park et al.,
(Amin et al., 2006), PDGFR (Rolny et al., 2006) and 1997; Sano et al., 2005). In the study by Park et al.
CXCRs (Ryschich et al., 2006). The interaction between (1997), the expression of a-smooth muscle actin (a
ligands and their corresponding receptors initiates sev- marker for activated HSCs) was found to be significantly
eral signaling pathways that regulate survival, prolifera- lower in dysplastic nodules than that in cirrhotic nod-
tion, mobilization and invasion of endothelial cells. MVD ules or in HCCs, leading to a hypothesis that dysplastic
is a commonly used parameter to assess the number of nodules develop from clonal expansions of neoplastic he-
neovessels formed in the tumor tissues. The level of patocytes in advance of or parallel to the development of
MVD is usually quantified by immunohistochemical cirrhosis. In a rat HCC model, blockade of HSC activa-
staining using endothelial cell markers. The commonly tion significantly prevent the development of fibrogene-
used endothelial cell markers for HCC include CD31, sis and HCC, suggesting a sequential change from liver
CD34 and von Willebrand factor (vWF; Poon et al., injury, fibrosis and HCC (Sano et al., 2005).
2002; Wang et al., 2006; Lau et al., 2006). Previous stud- HSCs share various types of endothelial receptors,
ies by our group (Poon et al., 2002) and Tanigawa et al. such as VEGFRs (Ankoma-Sey et al., 2000), PDGFR
(1997) showed that CD34 stained mainly larger vessels (Borkham-Kamphorst et al., 2007), and Tie-2 (Novo
with wider lumen in the fibrous tissue within the tumor et al., 2007). Therefore, the angiogenic factors secreted
tissues, rather than the capillary-like vessels that are by tumor cells may also function on HSCs. Olaso et al.
stained by CD31, suggesting that CD34 might more (2003) reported that hypoxic induction of VEGF in tu-
accurately represent MVD in HCC. In the study by mor-activated HSCs may create a proangiogenic micro-
Tanigawa et al. (1997), MVD by CD34 staining, but not environment that favors recruitment and survival of en-
vWF staining, was an independent prognostic factor of dothelial cells during hepatic metastasis transition from
patient survival after resection of HCC. However, an avascular to a vascular stage. An in vitro experiment
another study of MVD by CD34 staining in 40 HCC revealed that tumor cells could directly activate HSCs,
patients with hepatic resection did not show any signifi- and the activation process depended on the PDGF/
cant correlation between tumor MVD and pathological PDGFR pathway (Faouzi et al., 1999).
features of the tumor, such as portal vein invasion and
tumor stage (Kimura et al., 1998). Two other studies Bone Marrow-Derived Endothelial
reported that MVD stained by vWF, but not CD34, could
Progenitor Cells
predict HCC patient outcome after hepatic resection
(Yamamoto et al., 1998; Ker et al., 1999). However, our Recent research interest has moved to another poten-
study and others’ showed that CD34-MVD could achieve tial angiogenic cell type, namely EPCs. EPCs express
a significant prognostic value for tumor recurrence after some cell surface markers, such as CD31 and VEGFR2,
resection of small HCC (5 cm in diameter; Sun et al., that are similar to mature endothelial cells, but display
1999; Poon et al., 2002). Therefore, it remains controver- more abundant phenotypes of stem/progenitor cells, such
728 YANG AND POON

Fig. 3. Measurement of circulating endothelial progenitor cells rescein isothiocyanate-labeled Ultex europaeus agglutinin-1. B: Levels
(EPCs) in hepatocellular carcinoma (HCC). A: The level of circulating of circulating EPCs were significantly higher in HCC patients, particu-
EPCs was assessed by colony formation unit (a) and (b), and the con- larly unresectable HCC, than that in normal controls and patients with
firmation of endothelial cell lineage was evaluated by colony’s positive cirrhosis. Copyright from Ho et al. (2006).
uptake of (c) DiI-labled acetylated low-density lipoprotein and (d) fluo-

as CD133 (Bertolini et al., 2006). The first study on the identified in the premetastatic niche (Kaplan et al.,
role of EPCs in tumor angiogenesis was reported by 2005). Jin et al. (2006) further characterized the pheno-
Lyden et al. in 2001. Several other studies have subse- types of bone marrow-derived Flt-11 cells and found
quently demonstrated the contribution of EPCs to tumor that these cells also simultaneously expressed CXCR4, a
vasculature (De Palma and Naldini, 2006; Li et al., chemokine receptor of SDF-1. The mobilization of
2006). However, these studies are mainly performed in CXCR41Flt-11 cells (hemangiocytes) to the site of is-
animal models of tumors. A recent report showed that chemia could be inhibited by CXCR4 blockade. These
by using the fluorescence in situ hybridization technol- studies provide a hint that tumor cell-secreted VEGF
ogy, EPCs were found in approximately 4.9% of vascula- may recruit hemangiocytes for tumor angiogenesis.
ture in human tumor specimens, suggesting a less prom- Because HCC tumor cells secrete a high level of VEGF,
inent role of EPCs in tumor angiogenesis than previ- it is reasonable to postulate that VEGF-mediated pro-
ously suggested (Peters et al., 2005). genitor cell migration might contribute to tumor angio-
Our previous clinical data showed that the level of cir- genesis in HCC, though further experimental and clini-
culating EPCs was significantly higher in HCC patients cal evidence is needed to explore this hypothesis.
than that in patients with cirrhosis and normal subjects
(Ho et al., 2006; Fig. 3). In addition, patients with an Other Types of Cells
advance stage HCC had a higher level of circulating
EPCs. Moreover, circulating EPC level was significantly HCC tumor cells are also one of the key players in the
correlated with serum AFP levels, plasma VEGF and IL- process of angiogenesis by secreting various types of
8 levels, suggesting that VEGF and IL-8 might contrib- angiogenic factors that function on endothelial cells-
ute to the mobilization of circulating EPCs to the tumor expressed corresponding receptors. Some of the major
tissues. However, direct evidence to show the localiza- angiogenic factors secreted by the tumor cells have been
tion of EPCs in the endothelia of tumor tissues is needed discussed in the section Major Molecular Pathways That
to more strongly support the role of EPCs in angiogene- Mediate Angiogenesis in HCC earlier in this review. On
sis of HCC. In addition to natural course of tumor devel- the other hand, a recent work suggested that tumor cells
opment, some therapeutic approaches, such as vascula- might be directly involved in the process of vasculogenic
ture destruction agents, could also induce acute recruit- mimicry (Sun et al., 2006), another type of angiogenesis,
ment of EPCs to the tumor tissues (Shaked et al., 2006), though limited evidence demonstrated its mechanism.
suggesting the potential role of EPC mobilization in tu- Pericytes/smooth muscle cells are another type of cells
mor angiogenesis in response to anti-cancer treatment. that participate in the maturation of tumor neovascula-
However, more clinical investigation is needed to prove ture (Dickson et al., 2007). However, little evidence has
this hypothesis. shown the presence of this type of cells in HCC and
their effects on tumor angiogenesis. The major concern
is that the marker, smooth muscle actin, which is used
Bone Marrow-Derived Hemangiocytes
to label smooth muscle cells, can also be used to recog-
Another type of bone marrow-derived stem/progenitor nize HSCs, leading to the hypothesis that HSCs belong
cells, which express Flt-1 and CXCR4, has recently been to the pericyte/smooth muscle cell family.
VASCULAR CHANGES IN HCC 729
ANTIANGIOGENIC THERAPY FOR HCC intense interest in research on antiangiogenic therapy
for cancers. There are reports of some clinical trials
Targeting endothelial cells as an anticancer strategy
using antiangiogenic agents, such as Thalidomide, for
may have the following advantages: (i) drugs targeted at
the treatment of HCC, but the tumor response rate is
the endothelial cells are less likely than cytotoxic drugs
low (Patt et al., 2005). Preliminary results from clinical
to induce drug resistance due to a relatively stable
trials of single-agent antiangiogenic therapy in advanced
genetic background of the microvascular endothelial
solid cancers showed poor efficacy (Chen et al., 2005).
cells; (ii) low toxicity has been found when targeting tu-
Current evidence suggests that antiangiogenic therapy
mor vasculature, which is characterized by a relatively
works better in combination with cytotoxic chemother-
immature structure compared with normal quiescent apy (Gasparini et al., 2005). In addition, antiangiogenic
vasculature; (iii) endothelial cells are a major obstacle to therapy may also have a synergistic effect with other
conventional anticancer therapy as they may hinder molecular targeted therapies (Feng et al., 2005). Our
drug delivery to tumor cells. Therefore, targeting endo- group is currently conducting a phase I/II trial on combi-
thelial cells may enhance delivery of cytotoxic drugs to nation of systemic doxorubicin and PTK787 for patients
tumor cells. with advanced or metastatic HCC. Antiangiogenic ther-
Several preclinical studies have demonstrated the effi- apy may also be used as an adjunctive therapy with
cacy of antiangiogenesis as a potent anticancer strategy established therapies for HCC. Our animal model has
for HCC. Blockade of VEGF/VEGFR pathway by gene shown that by using the combination of hepatic artery
delivery vehicle encoding antisense VEGF could inhibit ligation with PTK787, significant inhibition of tumor
the growth of HCC xenograft in nude mice (Kang et al., growth and prolongation of animal survival could be
2000). Our group also demonstrated that using a tyro- achieved (Yang et al., 2006), suggesting that addition of
sine kinase inhibitor, PTK 787, targeting VEGFRs could antiangiogenic therapy may enhance the therapeutic ef-
suppress HCC tumor cell growth both in vitro and in ficacy of chemoembolization in clinical setting. Another
vivo, and the significant inhibition of tumor growth in group also demonstrated that VEGF antisense oligodeox-
vivo corresponded to the decreased expression of MVD ynucleotides mixed with lipiodol more significantly
(Liu Y. et al., 2005). Moreover, inhibition of VEGFR ac- inhibited HCC growth in rats than arterial embolization
tivity using PTK787 was also effective in blocking revas- with lipiodol alone (Wu et al., 2004). These pre-clinical
cularization in tumor tissues after hepatic artery liga- data suggest that it is reasonable to conduct a clinical
tion, suggesting that blockade VEGF/VEGFR pathway trial to test whether combined anti-VEGF therapy can
by molecular targeted therapy may enhance the thera- enhance the efficacy of TACE for the treatment of HCC.
peutic efficacy of TACE for the treatment of unresectable
HCC (Yang et al., 2006).
Another approach of antiangiogenic therapy is to use
CONCLUSIONS
drugs that directly inhibit the proliferation of endothe- Angiogenesis plays a crucial role in carcinogenesis,
lial cells. TNP-470, a fumagillin analogue that can in- growth and progression of HCC, which is one of the
hibit endothelial cell proliferation, has been shown to most vascularized human cancers. Morphologically,
suppress the growth and metastasis of human HCC xen- arterialization and sinusoidal capillarization are two
otransplant in an animal model (Xia et al., 1997). Inter- important vascular changes associated with tumor
feron-alpha, an immunomodulatory agent, has been angiogenesis in HCC. VEGF appears to be the most crit-
shown to directly inhibit endothelial cell proliferation ical angiogenic factor regulating angiogenesis in HCC.
and suppress angiogenesis in vivo (Wang et al., 2000). Recently, other cell types in addition to endothelial cells
More recently, our group has shown that a novel immu- have been identified to play important roles in tumor
nomodulatory agent FTY720 could also inhibit the angiogenesis. However, the molecular and cellular mech-
growth of HCC xenotransplant in nude mice by means anisms that regulate angiogenesis in HCC still remain
of an antiangiogenic effect (Ho et al., 2005b). largely unclear. Further studies to elucidate the mecha-
Researchers have also used agents that can prevent nisms involved in HCC angiogenesis are not only crucial
the degradation of extracellular matrix and basement to our understanding of tumor biology of this disease,
membrane, a step essential for angiogenesis, for antian- but may also provide guidance to the use of appropriate
giogenic treatment of cancers (Qin et al., 1999). Gene antiangiogenic agents for HCC.
therapy by delivery of natural antiangiogenic factors,
such as angiostatin and endostatin, can also achieve ACKNOWLEDGMENTS
antiangiogenic effects in HCC xenograft in nude mice The study was supported by the Seed Funding Pro-
(Ishikawa et al., 2003; Liu H. et al., 2005). Analysis of gram for Basic Research 2007 of the University of Hong
the vascular density in the tumor tissues showed that Kong.
inhibition of tumor growth by angiostatin correlated
with suppression of tumor vascularity. Some analogues
LITERATURE CITED
of VEGF receptors, such as soluble Flt-1 recombinant
protein, could competitively bind to Flt-1 expressed on Aghi M, Cohen KS, Klein RJ, Scadden DT, Chiocca EA. 2006. Tu-
endothelial cells, and thus inhibit the formation of neo- mor stromal-derived factor-1 recruits vascular progenitors to
mitotic neovasculature, where microenvironment influences their
vasculature (Graepler et al., 2005).
differentiated phenotypes. Cancer Res 66:9054–9064.
On-going clinical trials have demonstrated the efficacy Ahmad S, Hewett PW, Wang P, Al-Ani B, Cudmore M, Fujisawa T,
of VEGF/VEGFR blockade using anti-VEGF antibody, Haigh JJ, le Noble F, Wang L, Mukhopadhyay D, Ahmed A. 2006.
bevacizumab, in combination with chemotherapy in Direct evidence for endothelial vascular endothelial growth factor
treating various types of human cancers including HCC receptor-1 function in nitric oxide-mediated angiogenesis. Circ
(Zhu et al., 2006; Fanale and Younes, 2007), leading to Res 99:715–722.
730 YANG AND POON

Akiba J, Yano H, Ogasawara S, Higaki K, Kojiro M. 2001. Expres- El-Assal ON, Yamanoi A, Ono T, Kohno H, Nagasue N. 2001. The
sion and function of interleukin-8 in human hepatocellular carci- clinicopathological significance of heparanase and basic fibroblast
noma. Int J Oncol 18:257–264. growth factor expressions in hepatocellular carcinoma. Clin Can-
Amin DN, Hida K, Bielenberg DR, Klagsbrun M. 2006. Tumor endo- cer Res 7:1299–1305.
thelial cells express epidermal growth factor receptor (EGFR) but El-Serag HB, 2004. Hepatocellular carcinoma: recent trend in the
not ErbB3 and are responsive to EGF and to EGFR kinase inhibi- United States. Gastroenterology 127:S27–S34.
tors. Cancer Res 66:2173–2180. Ezaki T, Ikegami T, Maeda T, Yamada T, Ishida T, Hashizume M,
Ankoma-Sey V, Wang Y, Dai Z. 2000. Hypoxic stimulation of vascu- Maehara Y. 2005. Prognostic value of thymidine phosphorylase
lar endothelial growth factor expression in activated rat hepatic activity in liver tissue adjacent to hepatocellular carcinoma. Int J
stellate cells. Hepatology 31:141–148. Clin Oncol 10:171–176.
Arii S, Yamaoka Y, Futagawa S, Inoue K, Kobayashi K, Kojiro M, Fanale MA, Younes A. 2007. Monoclonal antibodies in the treat-
Makuuchi M, Nakamura Y, Okita K, Yamada R. 2000. Results ment of non-Hodgkin’s lymphoma. Drugs 67:333–350.
of surgical and nonsurgical treatment for small-sized hepatocellu- Faouzi S, Lepreux S, Bedin C, Dubuisson L, Balabaud C, Bioulac-
lar carcinomas: a retrospective and nationwide survey in Japan. Sage P, Desmouliere A, Rosenbaum J. 1999. Activation of cul-
The Liver Cancer Study Group of Japan. Hepatology 32:1224– tured rat hepatic stellate cells by tumoral hepatocytes. Lab Invest
1229. 79:485–493.
Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Favier B, Alam A, Barron P, Bonnin J, Laboudie P, Fons P, Man-
Kearne M, Magner M, Isner JM. 1999. Bone marrow origin of en- dron M, Herault JP, Neufeld G, Savi P, Herbert JM, Bono F.
dothelial cell progenitor cells responsible for postnatal vasculo- 2006. Neuropilin-2 interacts with VEGFR-2 and VEGFR-3 and
genesis in physiological and pathological neovascularization. Circ promotes human endothelial cell survival and migration. Blood
Res 85:221–228. 108:1243–1250.
Azenshtein E, Meshel T, Shina S, Barak N, Keydar I, Ben-Baruch Feng KK, Zhao HY, Qiu H, Liu JX, Chen J. 2005. Combined ther-
A. 2005. The angiogenic factors CXCL8 and VEGF in breast can- apy with flk1-based DNA vaccine and interleukin-12 results in
cer: regulation by an array of pro-malignancy factors. Cancer Lett enhanced antiangiogenic and antitumor effects. Cancer Lett
217:73–86. 221:41–47.
Bertolini F, Shaked Y, Mancuso P, Kerbel RS. 2006. The multifac- Fischer AN, Fuchs E, Mikula M, Huber H, Beug H, Mikulits W.
eted circulating endothelial cell in cancer: towards marker and 2007. PDGF essentially links TGF-beta signaling to nuclear beta-
target identification. Nat Rev Cancer 6:835–845. catenin accumulation in hepatocellular carcinoma progression.
Borkham-Kamphorst E, van Roeyen CR, Ostendorf T, Floege J, Oncogene 26:3395–3405.
Gressner AM, Weiskirchen R. 2007. Pro-fibrogenic potential of Frachon S, Gouysse G, Dumortier J, Couvelard A, Nejjari M, Mion
PDGF-D in liver fibrosis. J Hepatol 46:1064–1074. F, Berger F, Paliard P, Boillot O, Scoazec JY. 2001. Endothelial
Brown JM, Wilson WR. 2004. Exploiting tumour hypoxia in cancer cell marker expression in dysplastic lesions of the liver: an immu-
treatment. Nat Rev Cancer 4:437–447. nohistochemical study. J Hepatol 34:850–857.
Bruix J, Sherman M, Practice Guidelines Committee, American Fujii T, Nomoto S, Koshikawa K, Yatabe Y, Teshigawara O, Mori T,
Association for the Study of Liver Diseases. 2005. Management of Inoue S, Takeda S, Nakao A. 2006. Overexpression of pituitary
hepatocellular carcinoma. Hepatology 42:1208–1236. tumor transforming gene 1 in HCC is associated with angiogene-
Cervello M, Montalto G. 2006. Cyclooxygenases in hepatocellular sis and poor prognosis. Hepatology 43:1267–1275.
carcinoma. World J Gastroenterol 12:5113–5121. Fukasawa M, Matsushita A, Korc M. 2007. Neuropilin-1 interacts
Chen LT, Liu TW, Chao Y, Shiah HS, Chang JY, Juang SH, Chen with integrin beta1 and modulates pancreatic cancer cell growth,
SC, Chuang TR, Chin YH, Whang-Peng J. 2005. Alpha-fetopro- survival and invasion. Cancer Biol Ther 6:1173–1180.
tein response predicts survival benefits of thalidomide in Gasparini G, Longo R, Fanelli M, Teicher BA. 2005. Combination of
advanced hepatocellular carcinoma. Aliment Pharmacol Ther antiangiogenic therapy with other anticancer therapies: results,
22:217–226. challenges, and open questions. J Clin Oncol 23:1295–1311.
Cheng AS, Chan HL, To KF, Leung WK, Chan KK, Liew CT, Sung Geerts AM, De Vriese AS, Vanheule E, Van Vlierberghe H, Mortier
JJ. 2004. Cyclooxygenase-2 pathway correlates with vascular S, Cheung KJ, Demetter P, Lameire N, De Vos M, Colle I. 2006.
endothelial growth factor expression and tumor angiogenesis in Increased angiogenesis and permeability in the mesenteric micro-
hepatitis B virus-associated hepatocellular carcinoma. Int J Oncol vasculature of rats with cirrhosis and portal hypertension: an in
24:853–860. vivo study. Liver Int 26:889–898.
Chow NH, Hsu PI, Lin XZ, Yang HB, Chan SH, Cheng KS, Huang Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit V,
SM, Su IJ. 1997. Expression of vascular endothelial growth factor Ferrara N. 1998. Vascular endothelial growth factor regulates en-
in normal liver and hepatocellular carcinoma: an immunohisto- dothelial cell survival through the phosphatidylinositol 30 -kinase/
chemical study. Hum Pathol 28:698–703. Akt signal transduction pathway. Requirement for Flk-1/KDR
Couvelard A, Scoazec JY, Dauge MC, Bringuier AF, Potet F, Feld- activation. J Biol Chem 273:30336–30343.
mann G. 1996. Structural and functional differentiation of sinu- Gorrin-Rivas MJ, Arii S, Mori A, Takeda Y, Takeda Y, Mizumoto M,
soidal endothelial cells during liver organogenesis in humans. Furutani M, Imamura M. 2000. Implications of human macro-
Blood 87:4568–4580. phage metalloelastase and vascular endothelial growth factor
De Palma M, Naldini L. 2006. Role of haematopoietic cells and en- gene expression in angiogenesis of hepatocellular carcinoma. Ann
dothelial progenitors in tumour angiogenesis. Biochim Biophys Surg 231:67–73.
Acta 1766:159–166. Graepler F, Verbeek B, Graeter T, Smirnow I, Kong HL, Schuppan
Deli G, Jin CH, Mu R, Yang S, Liang Y, Chen D, Makuuchi M. D, Bauer M, Vonthein R, Gregor M, Lauer UM. 2005. Combined
2005. Immunohistochemical assessment of angiogenesis in hepa- endostatin/sFlt-1 antiangiogenic gene therapy is highly effective
tocellular carcinoma and surrounding cirrhotic liver tissues. in a rat model of HCC. Hepatology 41:879–886.
World J Gastroenterol 11:960–963. Griffiths L, Stratford IJ. 1997. Platelet-derived endothelial cell
Dhar DK, Naora H, Yamanoi A, Ono T, Kohno H, Otani H, Nagasue growth factor thymidine phosphorylase in tumour growth and
N. 2002. Requisite role of VEGF receptors in angiogenesis of response to therapy. Br J Cancer 76:689–693.
hepatocellular carcinoma: a comparison with angiopoietin/Tie Groupe d0 Etude et de Traitement du Carcinome Hépatocellulaire.
pathway. Anticancer Res 22:379–386. 1995. A comparison of lipiodol chemoembolization and conserva-
Dickson PV, Hamner JB, Streck CJ, Ng CY, McCarville MB, Calabr- tive treatment for unresectable hepatocellular carcinoma. N Engl
ese C, Gilbertson RJ, Stewart CF, Wilson CM, Gaber MW, Pfeffer J Med 332:1256–1261.
LM, Skapek SX, Nathwani AC, Davidoff AM. 2007. Continuous Guo L, Kuroda N, Toi M, Miyazaki E, Hayashi Y, Enzan H, Jin Y.
delivery of IFN-beta promotes sustained maturation of intratu- 2001. Increased expression of platelet-derived endothelial cell
moral vasculature. Mol Cancer Res 5:531–542. growth factor in human hepatocellular carcinomas correlated
VASCULAR CHANGES IN HCC 731
with high Edmondson grades and portal vein tumor thrombosis. noma: correlation between dynamic CT and density of tumor
Oncol Rep 8:871–876. microvessels. Radiology 237:529–534.
Gwak GY, Yoon JH, Kim KM, Lee HS, Chung JW, Gores GJ. 2005. Kimura H, Nakajima T, Kagawa K, Deguchi T, Kakusui M, Kata-
Hypoxia stimulates proliferation of human hepatoma cells gishi T, Okanoue T, Kashima K, Ashihara T. 1998. Angiogenesis
through the induction of hexokinase II expression. J Hepatol in hepatocellular carcinoma as evaluated by CD34 immunohisto-
42:358–364. chemistry. Liver 18:14–19.
Harris AL. 2002. Hypoxia--a key regulatory factor in tumour Kin M, Torimura T, Ueno T, Inuzuka S, Tanikawa K. 1994. Sinusoi-
growth. Nat Rev Cancer 2:38–47. dal capillarization in small hepatocellular carcinoma. Pathol Int
Himeno H, Enzan H, Saibara T, Onishi S, Yamamoto Y. 1994. Hith- 44:771–778.
erto unrecognized arterioles within hepatocellular carcinoma. J Kubo F, Ueno S, Hiwatashi K, Sakoda M, Kawaida K, Nuruki K,
Pathol 174:217–222. Aikou T. 2005. Interleukin 8 in human hepatocellular carcinoma
Hisai H, Kato J, Kobune M, Murakami T, Miyanishi K, Takahashi correlates with cancer cell invasion of vessels but not with tumor
M. 2003. Increased expression of angiogenin in hepatocellular angiogenesis. Ann Surg Oncol 12:800–807.
carcinoma in correlation with tumor vascularity. Clin Cancer Res Lau CP, Poon RT, Cheung ST, Yu WC, Fan ST. 2006. SPARC and
9:4852–4859. Hevin expression correlate with tumour angiogenesis in hepato-
Ho JW, Poon RT, Sun CK, Xue WC, Fan ST. 2005a. Clinicopatholog- cellular carcinoma. J Pathol 210:459–468.
ical and prognostic implications of endoglin (CD105) expression in Lee TK, Poon RT, Yuen AP, Ling MT, Wang XH, Wong YC, Guan
hepatocellular carcinoma and its adjacent non-tumorous liver. XY, Man K, Tang ZY, Fan ST. 2006. Regulation of angiogenesis by
World J Gastroenterol 11:176–181. Id-1 through hypoxia-inducible factor-1alpha-mediated vascular
Ho JW, Man K, Sun CK, Lee TK, Poon RT, Fan ST. 2005b. Effects endothelial growth factor up-regulation in hepatocellular carci-
of a novel immunomodulating agent, FTY720, on tumor growth noma. Clin Cancer Res 12:6910–6919.
and angiogenesis in hepatocellular carcinoma. Mol Cancer Ther Li M, Tang ZY, Zhou G, Lui YK, Ye SL. 1998. Significance of vascu-
4:1430–1438. lar endothelial growth factor mRNA expression in invasion and
Ho JW, Pang RW, Lau C, Sun CK, Yu WC, Fan ST, Poon RT. 2006. metastasis of hepatocellular carcinoma. J Exp Clin Cancer Res
Significance of circulating endothelial progenitor cells in hepato- 17:13–17.
cellular carcinoma. Hepatology 44:836–843. Li B, Sharpe EE, Maupin AB, Teleron AA, Pyle AL, Carmeliet P,
Hong TM, Chen YL, Wu YY, Yuan A, Chao YC, Chung YC, Wu MH, Young PP. 2006. VEGF and PlGF promote adult vasculogenesis
Yang SC, Pan SH, Shih JY, Chan WK, Yang PC. 2007. Targeting by enhancing EPC recruitment and vessel formation at the site of
neuropilin 1 as an antitumor strategy in lung cancer. Clin Cancer tumor neovascularization. FASEB J 20:1495–1497.
Res 13:4759–4768. Liu H, Peng CH, Liu YB, Wu YL, Zhao ZM, Wang Y, Han BS. 2005.
Huang GW, Yang LY, Lu WQ. 2005. Expression of hypoxia-inducible Inhibitory effect of adeno-associated virus-mediated gene transfer
factor 1alpha and vascular endothelial growth factor in hepatocel- of human endostatin on hepatocellular carcinoma. World J Gas-
lular carcinoma: impact on neovascularization and survival. troenterol 11:3331–3334.
World J Gastroenterol 11:1705–1708. Liu Y, Poon RT, Li Q, Kok TW, Lau C, Fan ST. 2005. Both antian-
Huwart L, Michoux N, Van Beers B. 2007. Magnetic resonance giogenesis- and angiogenesis-independent effects are responsible
imaging of angiogenesis in tumors. J Radiol 88(Pt 1):331–338. for hepatocellular carcinoma growth arrest by tyrosine kinase in-
Iavarone M, Lampertico P, Iannuzzi F, Manenti E, Donato MF, Aro- hibitor PTK787/ZK222584. Cancer Res 65:3691–3699.
sio E, Bertolini F, Primignani M, Sangiovanni A, Colombo M. Llovet JM, Fuster J, Bruix J. 1999. Intention-to-treat analysis of
2007. Increased expression of vascular endothelial growth factor surgical treatment for early hepatocellular carcinoma: resection
in small hepatocellular carcinoma. J Viral Hepat 14:133–139. versus transplantation. Hepatology 30:1434–1440.
Ishikawa H, Nakao K, Matsumoto K, Ichikawa T, Hamasaki K, Llovet JM, Burroughs A, Bruix J. 2003. Hepatocellular carcinoma.
Nakata K. 2003. Antiangiogenic gene therapy for hepatocellular Lancet 362:1907–1917.
carcinoma using angiostatin gene. Hepatology 37:696–704. Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, Fan ST,
Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV, Young LM, Hoo- Wong J. 2002. Randomized controlled trial of transarterial lipio-
per AT, Amano H, Avecilla ST, Heissig B, Hattori K, Zhang F, dol chemoembolization for unresectable hepatocellular carcinoma.
Hicklin DJ, Wu Y, Zhu Z, Dunn A, Salari H, Werb Z, Hackett NR, Hepatology 35:1164–1171.
Crystal RG, Lyden D, Rafii S. 2006. Cytokine-mediated deploy- Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn
ment of SDF-1 induces revascularization through recruitment of A, Heissig B, Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett
CXCR41 hemangiocytes. Nat Med 12:557–567. NR, Crystal RG, Moore MA, Hajjar KA, Manova K, Benezra R,
Kang MA, Kim KY, Seol JY, Kim KC, Nam MJ. 2000. The growth Rafii S. 2001. Impaired recruitment of bone-marrow-derived endo-
inhibition of hepatoma by gene transfer of antisense vascular en- thelial and hematopoietic precursor cells blocks tumor angiogene-
dothelial growth factor. J Gene Med 2:289–296. sis and growth. Nat Med 7:1194–1201.
Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa Masood R, Cai J, Zheng T, Smith DL, Hinton DR, Gill PS. 2001.
C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Vascular endothelial growth factor (VEGF) is an autocrine growth
Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jen- factor for VEGF receptor-positive human tumors. Blood 98:1904–
sen KK, Rafii S, Lyden D. 2005. VEGFR1-positive haematopoietic 1913.
bone marrow progenitors initiate the pre-metastatic niche. Na- Matsui O. 2001. Analysis of intranodular hemodynamics of dysplas-
ture 438:820–827. tic nodules in cirrhotic livers. Radiology 218:603–605.
Ker CG, Chen HY, Juan CC, Lo HW, Shen YY, Chen JS, Lee KT, McMahon BJ. 2005. Epidemiology and natural history of hepatitis
Sheen PC. 1999. Role of angiogenesis in hepatitis and hepatocel- B. Semin Liver Dis 25:3–8.
lular carcinoma. Hepatogastroenterology 46:646–650. Medina J, Sanz-Cameno P, Garcia-Buey L, Martin-Vilchez S, Lopez-
Kermani P, Rafii D, Jin DK, Whitlock P, Schaffer W, Chiang A, Vin- Cabrera M, Moreno-Otero R. 2005. Evidence of angiogenesis in
cent L, Friedrich M, Shido K, Hackett NR, Crystal RG, Rafii S, primary biliary cirrhosis: an immunohistochemical descriptive
Hempstead BL. 2005. Neurotrophins promote revascularization by study. J Hepatol 42:124–131.
local recruitment of TrkB1 endothelial cells and systemic mobiliza- Mise M, Arii H, Higashituji M, Furutani M, Niwano M, Harada T,
tion of hematopoietic progenitors. J Clin Invest 115:653–663. Ishigami S, Toda Y, Nakayama H, Fukumoto M, Fujita J, Ima-
Kim H, Li Q, Hempstead BL, Madri JA. 2004. Paracrine and auto- mura M. 1996. Clinical significance of vascular endothelial
crine functions of brain-derived neurotrophic factor (BDNF) and growth factor and basic fibroblast growth factor gene expression
nerve growth factor (NGF) in brain-derived endothelial cells. J in liver tumor. Hepatology 23:455–464.
Biol Chem 279:33538–33546. Mitsuhashi N, Shimizu H, Ohtsuka M, Wakabayashi Y, Ito H,
Kim CK, Lim JH, Park CK, Choi D, Lim HK, Lee WJ. 2005. Neoan- Kimura F, Yoshidome H, Kato A, Nukui Y, Miyazaki M. 2003.
giogenesis and sinusoidal capillarization in hepatocellular carci- Angiopoietins and Tie-2 expression in angiogenesis and prolifera-
732 YANG AND POON

tion of human hepatocellular carcinoma. Hepatology 37:1105– transarterial chemoembolization in hepatocellular carcinoma: a
1113. prospective study. Oncol Rep 11:1077–1084.
Moon WS, Park HS, Yu KH, Park MY, Kim KR, Jang KY, Kim JS, Poon RT, Chung KK, Cheung ST, Lau C, Tong SW, Leung KL, Yu
Cho BH. 2006. Expression of betacellulin and epidermal growth WC, Tuszynski GP, Fan ST. 2004b. Clinical significance of throm-
factor receptor in hepatocellular carcinoma: implications for bospondin 1 expression in hepatocellular carcinoma. Clin Cancer
angiogenesis. Hum Pathol 37:1324–1332. Res 10:4150–4157.
Morinaga S, Yamamoto Y, Noguchi Y, Imada T, Rino Y, Akaike M, Poon RT, Lau C, Pang R, Ng KK, Yuen J, Fan ST. 2007. High se-
Sugimasa Y, Takemiya S, Takanashi Y. 2003. Platelet-derived en- rum vascular endothelial growth factor levels predict poor prog-
dothelial cell growth factor (PD-ECGF) is up-regulated in human nosis after radiofrequency ablation of hepatocellular carcinoma:
hepatocellular carcinoma (HCC) and the corresponding hepatitis importance of tumor biomarker in ablative therapies. Ann Surg
liver. Hepatogastroenterology 50:1521–1526. Oncol 14:1835–1845.
Musso O, Theret N, Heljasvaara R, Rehn M, Turlin B, Campion JP, Qin LX, Tang ZY, Li XM, Bu W, Xia JL. 1999. Effect of antiangio-
Pihlajaniemi T, Clement B. 2001. Tumor hepatocytes and base- genic agents on experimental animal models of hepatocellular
ment membrane-producing cells specifically express two different carcinoma. Ann Acad Med Singapore 28:147–151.
forms of the endostatin precursor, collagen XVIII, in human liver Raoul JL, Guyader D, Bretagne JF, Heautot JF, Duvauferrier R,
cancers. Hepatology 33:868–876. Bourguet P, Bekhechi D, Deugnier YM, Gosselin M. 1997. Pro-
Nakamura K, Martin KC, Jackson JK, Beppu K, Woo CW, Thiele spective randomized trial of chemoembolization versus intra-arte-
CJ. 2006. Brain-derived neurotrophic factor activation of TrkB rial injection of 131I-labeled-iodized oil in the treatment of hepa-
induces vascular endothelial growth factor expression via hy- tocellular carcinoma. Hepatology 26:1156–1161.
poxia-inducible factor-1alpha in neuroblastoma cells. Cancer Res Ren Y, Tsui HT, Poon RT, Ng IO, Li Z, Chen Y, Jiang G, Lau C, Yu
66:4249–4255. WC, Bacher M, Fan ST. 2003. Macrophage migration inhibitory
Ng IO, Poon RT, Lee JM, Fan ST, Ng M, Tso WK. 2001. Microvessel factor: roles in regulating tumor cell migration and expression of
density, vascular endothelial growth factor and its receptors Flt-1 angiogenic factors in hepatocellular carcinoma. Int J Cancer
and Flk-1/KDR in hepatocellular carcinoma. Am J Clin Pathol 107:22–29.
116:838–845. Riese DJ Jr, Stern DF. 1998. Specificity within the EGF family/
Novo E, Cannito S, Zamara E, Valfre di Bonzo L, Caligiuri A, Cra- ErbB receptor family signaling network. Bioessays 20:41–48.
vanzola C, Compagnone A, Colombatto S, Marra F, Pinzani M, Rolny C, Nilsson I, Magnusson P, Armulik A, Jakobsson L, Wentzel
Parola M. 2007. Proangiogenic cytokines as hypoxia-dependent P, Lindblom P, Norlin J, Betsholtz C, Heuchel R, Welsh M, Claes-
factors stimulating migration of human hepatic stellate cells. Am son-Welsh L. 2006. Platelet-derived growth factor receptor-beta
J Pathol 170:1942–1953. promotes early endothelial cell differentiation. Blood 108:1877–
Olaso E, Salado C, Egilegor E, Gutierrez V, Santisteban A, Sancho- 1886.
Bru P, Friedman SL, Vidal-Vanaclocha F. 2003. Proangiogenic Ryschich E, Lizdenis P, Ittrich C, Benner A, Stahl S, Hamann A,
role of tumor-activated hepatic stellate cells in experimental mel- Schmidt J, Knolle P, Arnold B, Hammerling GJ, Ganss R. 2006.
anoma metastasis. Hepatology 37:674–685. Molecular fingerprinting and autocrine growth regulation of endo-
Pang R, Poon RT. 2006. Angiogenesis and antiangiogenic therapy in thelial cells in a murine model of hepatocellular carcinoma. Can-
hepatocellular carcinoma. Cancer Lett 242:151–167. cer Res 66:198–211.
Park YN, Yang CP, Cubukcu O, Thung SN, Theise ND. 1997. He- Salomon DS, Brandt R, Ciardiello F, Normanno N. 1995. Epidermal
patic stellate cell activation in dysplastic nodules: evidence for an growth factor-related peptides and their receptors in human
alternate hypothesis concerning human hepatocarcinogenesis. malignancies. Crit Rev Oncol Hematol 19:183–232.
Liver 17:271–274. Sano T, Kagawa M, Okuno M, Ishibashi N, Hashimoto M, Yama-
Park YN, Yang CP, Fernandez GJ, Cubukcu O, Thung SN, Theise moto M, Suzuki R, Kohno H, Matsushima-Nishiwaki R, Takano
ND. 1998. Neoangiogenesis and sinusoidal ‘‘capillarization’’ in Y, Tsurumi H, Kojima S, Friedman SL, Moriwaki H, Tanaka T.
dysplastic nodules of the liver. Am J Surg Pathol 22:656–662. 2005. Prevention of rat hepatocarcinogenesis by acyclic retinoid is
Park YN, Kim YB, Yang KM, Park C. 2000. Increased expression of accompanied by reduction in emergence of both TGF-alpha-
vascular endothelial growth factor and angiogenesis in the early expressing oval-like cells and activated hepatic stellate cells. Nutr
stage of multistep hepatocarcinogenesis. Arch Pathol Lab Med Cancer 51:197–206.
124:1061–1065. Schaffner F, Poper H. 1963. Capillarization of hepatic sinusoids in
Patt YZ, Hassan MM, Lozano RD, Nooka AK, Schnirer II, Zeldis man. Gastroenterology 44:239–242.
JB, Abbruzzese JL, Brown TD. 2005. Thalidomide in the treat- Schimanski CC, Bahre R, Gockel I, Muller A, Frerichs K, Horner V,
ment of patients with hepatocellular carcinoma: a phase II trial. Teufel A, Simiantonaki N, Biesterfeld S, Wehler T, Schuler M,
Cancer 103:749–755. Achenbach T, Junginger T, Galle PR, Moehler M. 2006. Dissemi-
Peters BA, Diaz LA, Polyak K, Meszler L, Romans K, Guinan EC, nation of hepatocellular carcinoma is mediated via chemokine re-
Antin JH, Myerson D, Hamilton SR, Vogelstein B, Kinzler KW, ceptor CXCR4. Br J Cancer 95:210–217.
Lengauer C. 2005. Contribution of bone marrow-derived Schmitt M, Horbach A, Kubitz R, Frilling A, Haussinger D. 2004.
endothelial cells to human tumor vasculature. Nat Med 11:261– Disruption of hepatocellular tight junctions by vascular endothe-
262. lial growth factor (VEGF): a novel mechanism for tumor invasion.
Poon RT, Fan ST, Wong J. 2001. Clinical implications of circulating J Hepatol 41:274–283.
angiogenic factors in cancer patients. J Clin Oncol 19:1207–1225. Schweigerer L, Neufeld G, Friedman J, Abraham JA, Fiddes JC,
Poon RT, Ng IO, Lau C, Yu WC, Yang ZF, Fan ST, Wong J. 2002. Gospodarowicz D. 1987. Capillary endothelial cells express basic
Tumor microvessel density as a predictor of recurrence after fibroblast growth factor, a mitogen that promotes their own
resection of hepatocellular carcinoma: a prospective study. J Clin growth. Nature 325:257–259.
Oncol 20:1775–1785. Shaked Y, Ciarrocchi A, Franco M, Lee CR, Man S, Cheung AM,
Poon RT, Lau CP, Cheung ST, Yu WC, Fan ST. 2003a. Quantitative Hicklin DJ, Chaplin D, Foster FS, Benezra R, Kerbel RS. 2006.
correlation of serum levels and tumor expression of vascular en- Therapy-induced acute recruitment of circulating endothelial pro-
dothelial growth factor in patients with hepatocellular carcinoma. genitor cells to tumors. Science 313:1785–1787.
Cancer Res 63:3121–3126. Sheen IS, Jeng KS, Shih SC, Kao CR, Chang WH, Wang HY, Wang
Poon RT, Lau CP, Ho J, Yu WC, Fan ST, Wong J. 2003b. Tissue fac- PC, Wang TE, Shyung LR, Chen CZ. 2005. Clinical significance of
tor expression correlates with tumor angiogenesis and invasive- the expression of isoform 165 vascular endothelial growth factor
ness in human hepatocellular carcinoma. Clin Cancer Res mRNA in noncancerous liver remnants of patients with hepatocel-
9:5339–5345. lular carcinoma. World J Gastroenterol 11:187–192.
Poon RT, Lau C, Yu WC, Fan ST, Wong J. 2004a. High serum levels Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y,
of vascular endothelial growth factor predict poor response to Kothari S, Mohle R, Sauvage LR, Moore MA, Storb RF, Hammond
VASCULAR CHANGES IN HCC 733
WP. 1998. Evidence for circulating bone marrow-derived endothe- von Marschall Z, Cramer T, Hocker M, Finkenzeller G, Wieden-
lial cells. Blood 92:362–367. mann B, Rosewicz S. 2001. Dual mechanism of vascular endothe-
Shin EC, Choi YH, Kim JS, Kim SJ, Park JH. 2002. Expression lial growth factor upregulation by hypoxia in human hepatocellu-
patterns of cytokines and chemokines genes in human hepatoma lar carcinoma. Gut 48:87–96.
cells. Yonsei Med J 43:657–664. Wada H, Nagano H, Yamamoto H, Yang Y, Kondo M, Ota H, Naka-
Staton CA, Kumar I, Reed MW, Brown NJ. 2007. Neuropilins in phys- mura M, Yoshioka S, Kato H, Damdinsuren B, Tang D, Maruba-
iological and pathological angiogenesis. J Pathol 212:237–248. shi S, Miyamoto A, Takeda Y, Umeshita K, Nakamori S, Sakon
Sugimachi K, Tanaka S, Taguchi K, Aishima S, Shimada M, Tsu- M, Dono K, Wakasa K, Monden M. 2006. Expression pattern of
neyoshi M. 2003. Angiopoietin switching regulates angiogenesis angiogenic factors and prognosis after hepatic resection in hepato-
and progression of human hepatocellular carcinoma. J Clin cellular carcinoma: importance of angiopoietin-2 and hypoxia-
Pathol 56:854–860. induced factor-1 alpha. Liver Int 26:414–423.
Sun HC, Tang ZY. 2004. Angiogenesis in hepatocellular carcinoma: Wang L, Tang ZY, Qin LX, Wu XF, Sun HC, Xue Q, Ye SL. 2000.
the retrospectives and perspectives. J Cancer Res Clin Oncol High-dose and long-term therapy with interferon-alfa inhibits tu-
130:307–319. mor growth and recurrence in nude mice bearing human hepato-
Sun HC, Tang ZY, Li XM, Zhou YN, Sun BR, Ma ZC. 1999. Micro- cellular carcinoma xenografts with high metastatic potential.
vessel density of hepatocellular carcinoma: its relationship with Hepatology 32:43–48.
prognosis. Cancer Res Clin Oncol 125:419–426. Wang SN, Yeh YT, Yang SF, Chai CY, Lee KT. 2006. Potential role
Sun MH, Han XC, Jia MK, Jiang WD, Wang M, Zhang H, Han G, of leptin expression in hepatocellular carcinoma. J Clin Pathol
Jiang Y. 2005. Expressions of inducible nitric oxide synthase and 59:930–934.
matrix metalloproteinase-9 and their effects on angiogenesis and Weigand M, Hantel P, Kreienberg R, Waltenberger J. 2005. Auto-
progression of hepatocellular carcinoma. World J Gastroenterol crine vascular endothelial growth factor signalling in breast can-
11:5931–5937. cer. Evidence from cell lines and primary breast cancer cultures
Sun B, Zhang S, Zhang D, Du J, Guo H, Zhao X, Zhang W, Hao X. in vitro. Angiogenesis 8:197–204.
2006. Vasculogenic mimicry is associated with high tumor grade, Wu H, Feng G, Liang H, Zheng C, Li X. 2004. Vascular endothelial
invasion and metastasis, and short survival in patients with growth factor antisense oligodeoxynucleotides with lipiodol in ar-
hepatocellular carcinoma. Oncol Rep 16:693–698. terial embolization of liver cancer in rats. World J Gastroenterol
Sutton A, Friand V, Brule-Donneger S, Chaigneau T, Ziol M, 10:813–818.
Sainte-Catherine O, Poire A, Saffar L, Kraemer M, Vassy J, Xia JL, Yang BH, Tang ZY, Sun FX, Xue Q, Gao DM. 1997. Inhibi-
Nahon P, Salzmann JL, Gattegno L, Charnaux N. 2007. Stromal tory effect of the angiogenesis inhibitor TNP-470 on tumor growth
cell-derived factor-1/chemokine (C-X-C motif) ligand 12 stimulates and metastasis in nude mice bearing human hepatocellular carci-
human hepatoma cell growth, migration, and invasion. Mol Can- noma. J Cancer Res Clin Oncol 123:383–387.
cer Res 5:21–33. Yamamoto A, Dhar DK, El-Assal ON, Igarashi M, Tabara H, Naga-
Tanaka S, Mori M, Sakamoto Y, Makuuchi M, Sugimachi K, Wands sue N. 1998. Thymidine phosphorylase (platelet-derived endothe-
JR. 1999. Biologic significance of angiopoietin-2 expression in lial cell growth factor), microvessel density and clinical outcome
human hepatocellular carcinoma. J Clin Invest 103:341–345. in hepatocellular carcinoma. J Hepatol 29:290–299.
Tanaka F, Otake Y, Yanagihara K, Kawano Y, Miyahara R, Li M, Yamamoto T, Hirohashi K, Kaneda K, Ikebe T, Mikami S, Uenishi
Yamada T, Hanaoka N, Inui K, Wada H. 2001. Evaluation of T, Kanazawa A, Takemura S, Shuto T, Tanaka H, Kubo S,
angiogenesis in non-small cell lung cancer: comparison between Sakurai M, Kinoshita H. 2001. Relationship of the microvascular
anti-CD34 antibody and anti-CD105 antibody. Clin Cancer Res type to the tumor size, arterialization and dedifferentiation of
7:3410–3415. human hepatocellular carcinoma. Jpn J Cancer Res 92:1207–
Tanaka H, Yamamoto M, Hashimoto N, Miyakoshi M, Tamakawa S, 1213.
Yoshie M, Tokusashi Y, Yokoyama K, Yaginuma Y, Ogawa K. Yang ZF, Poon RT, To J, Ho DW, Fan ST. 2004. The potential role of
2006. Hypoxia-independent overexpression of hypoxia-inducible hypoxia inducible factor 1alpha in tumor progression after hy-
factor 1alpha as an early change in mouse hepatocarcinogenesis. poxia and chemotherapy in hepatocellular carcinoma. Cancer Res
Cancer Res 66:11263–11270. 64:5496–5503.
Tang TC, Poon RT, Lau CP, Xie D, Fan ST. 2005. Tumor cyclooxy- Yang ZF, Ho DW, Lam CT, Luk JM, Lum CT, Yu WC, Poon RT, Fan
genase-2 levels correlate with tumor invasiveness in human hepa- ST. 2005. Identification of brain-derived neurotrophic factor as a
tocellular carcinoma. World J Gastroenterol 11:1896–1902. novel functional protein in hepatocellular carcinoma. Cancer Res
Tanigawa N, Lu C, Mitsui T, Miura S. 1997. Quantitation of sinu- 65:219–225.
soid-like vessels in hepatocellular carcinoma: its clinical and prog- Yang LY, Lu WQ, Huang GW, Wang W. 2006. Correlation between
nostic significance. Hepatology 26:1216–1223. CD105 expression and postoperative recurrence and metastasis of
Torimura T, Sata M, Ueno T, Kin M, Tsuji R, Suzaku K, Hashimoto hepatocellular carcinoma. BMC Cancer 6:110.
O, Sugawara H, Tanikawa K. 1998. Increased expression of vas- Yang ZF, Poon RT, Liu Y, Lau CK, Tam KH, Lam CT, Fan ST. 2006.
cular endothelial growth factor is associated with tumor progres- High doses of tyrosine kinase inhibitor PTK787 enhance the effi-
sion in hepatocellular carcinoma. Hum Pathol 29:986–991. cacy of ischemic hypoxia for the treatment of HCC: dual effects
Torimura T, Ueno T, Kin M, Harada R, Taniguchi E, Nakamura T, on cancer cell and angiogenesis. Mol Cancer Ther 5:2261–2270.
Sakata R, Hashimoto O, Sakamoto M, Kumashiro R, Sata M, Yao Y, Pan Y, Chen J, Sun X, Qiu Y, Ding Y. 2007. Endoglin
Nakashima O, Yano H, Kojiro M. 2004. Overexpression of angio- (CD105) expression in angiogenesis of primary hepatocellular car-
poietin-1 and angiopoietin-2 in hepatocellular carcinoma. J Hepa- cinomas: analysis using tissue microarrays and comparisons with
tol 40:799–807. CD34 and VEGF. Ann Clin Lab Sci 37:39–48.
Ueda K, Terada T, Nakanuma Y, Matsui O. 1992. Vascular supply Yasuda S, Arii S, Mori A, Isobe N, Yang W, Oe H, Fujimoto A, Yone-
in adenomatous hyperplasia of the liver and hepatocellular carci- naga Y, Sakashita H, Imamura M. 2004. Hexokinase II and
noma: a morphometric study. Hum Pathol 23:619–626. VEGF expression in liver tumors: correlation with hypoxia-induci-
Ueda S, Basaki Y, Yoshie M, Ogawa K, Sakisaka S, Kuwano M, ble factor 1 alpha and its significance. J Hepatol 40:117–123.
Ono M. 2006. PTEN/Akt signaling through epidermal growth fac- Yoo YG, Oh SH, Park ES, Cho H, Lee N, Park H, Kim DK, Yu DY,
tor receptor is prerequisite for angiogenesis by hepatocellular car- Seong JK, Lee MO. 2003. Hepatitis B virus X protein enhances
cinoma cells that is susceptible to inhibition by gefitinib. Cancer transcriptional activity of hypoxia-inducible factor-1alpha through
Res 66:5346–5353. activation of mitogen-activated protein kinase pathway. J Biol
Vogel C, Bauer A, Wiesnet M, Preissner KT, Schaper W, Marti HH, Chem 278:39076–39084.
Fischer S. 2007. Flt-1, but not Flk-1 mediates hyperpermeability Yoshiji H, Kuriyama S, Yoshii J, Ikenaka Y, Noguchi R, Hicklin DJ,
through activation of the PI3-K/Akt pathway. J Cell Physiol 212: Wu Y, Yanase K, Namisaki T, Kitade M, Yamazaki M, Tsujinoue
236–243. H, Masaki T, Fukui H. 2004. Halting the interaction between vas-
734 YANG AND POON

cular endothelial growth factor and its receptors attenuates liver carcinoma associated with high metastatic potential. Clin Cancer
carcinogenesis in mice. Hepatology 39:1517–1524. Res 11(Pt 1):8557–8563.
Yoshiji H, Kuriyama S, Noguchi R, Yoshii J, Ikenaka Y, Yanase K, Zhang ZL, Liu ZS, Sun Q. 2006. Expression of angiopoietins, Tie2
Namisaki T, Kitade M, Uemura M, Masaki T, Fukui H. 2005. and vascular endothelial growth factor in angiogenesis and pro-
Angiopoietin 2 displays a vascular endothelial growth factor de- gression of hepatocellular carcinoma. World J Gastroenterol
pendent synergistic effect in hepatocellular carcinoma develop- 12:4241–4245.
ment in mice. Gut 54:1768–1775. Zhao ZC, Zheng SS, Wan YL, Jia CK, Xie HY. 2003. The molecular
Yu Q. 2005. The dynamic roles of angiopoietins in tumor angiogene- mechanism underlying angiogenesis in hepatocellular carcinoma:
sis. Future Oncol 1:475–484. the imbalance activation of signaling pathways. Hepatobiliary
Yung YC, Cheshier S, Santarelli JG, Huang Z, Wagers A, Weissman Pancreat Dis Int 2:529–536.
I, Tse V. 2004. Incorporation of naive bone marrow derived cells Zhou J, Tang ZY, Fan J, Wu ZQ, Li XM, Liu YK, Liu F, Sun HC, Ye
into the vascular architecture of brain tumor. Microcirculation SL. 2000. Expression of platelet-derived endothelial cell growth
11:699–708. factor and vascular endothelial growth factor in hepatocellular
Zhang J, Wang WL, Li Q, Qiao Q. 2004. Expression of transforming carcinoma and portal vein tumor thrombus. J Cancer Res Clin
growth factor-alpha and hepatitis B surface antigen in human he- Oncol 126:57–61.
patocellular carcinoma tissues and its significance. World J Gas- Zhu AX, Blaszkowsky LS, Ryan DP, Clark JW, Muzikansky A, Hor-
troenterol 10:830–833. gan K, Sheehan S, Hale KE, Enzinger PC, Bhargava P, Stuart K.
Zhang T, Sun HC, Xu Y, Zhang KZ, Wang L, Qin LX, Wu WZ, Liu 2006. Phase II study of gemcitabine and oxaliplatin in combina-
YK, Ye SL, Tang ZY. 2005. Overexpression of platelet-derived tion with bevacizumab in patients with advanced hepatocellular
growth factor receptor alpha in endothelial cells of hepatocellular carcinoma. J Clin Oncol 24:1898–1903.

You might also like