Angiogenesis and anti-angiogenesis in hepatocellular carcinoma

Angiogenesis and anti-angiogenesis in hepatocellular carcinoma

Cancer Treatment Reviews (2006) 32, 437– 444 available at www.sciencedirect.com journal homepage: www.elsevierhealth.com/journals/ctrv TUMOUR REVIE...

198KB Sizes 0 Downloads 92 Views

Cancer Treatment Reviews (2006) 32, 437– 444

available at www.sciencedirect.com

journal homepage: www.elsevierhealth.com/journals/ctrv

TUMOUR REVIEW

Angiogenesis and anti-angiogenesis in hepatocellular carcinoma Domenico Ribatti a,*, Angelo Vacca b, Beatrice Nico a, Domenico Sansonno b, Franco Dammacco b a

Department of Human Anatomy and Histology, University of Bari Medical School, Piazza Giulio Cesare, 11 Policlinico, 70124 Bari, Italy b Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy Received 11 March 2006; revised 1 June 2006; accepted 2 June 2006

KEYWORDS

Summary Experimental and clinical data indicate that in human hepatocellular carcinoma (HCC) tumor progression is associated with angiogenesis and that an increase in microvascular density is associated with a poor prognosis. This review summarizes the literature concerning the relationship between angiogenesis and progression in HCC. It is becoming increasingly evident that agents which interfere with blood vessel formation also block tumor progression. Accordingly, anti-angiogenic tumor therapy has gained much interest in preclinical and clinical assessments. The recent applications of anti-angiogenic agents which interfere or block HCC progression are reviewed. c 2006 Elsevier Ltd. All rights reserved.

Angiogenesis; Anti-angiogenesis; Hepatocellular carcinoma; Tumor progression



Tumor angiogenesis Angiogenesis and the production of angiogenic factors are fundamental for tumor growth, invasion and metastasis.1 New vessels promote growth by conveying oxygen and nutrients and removing catabolites, while endothelial cells secrete growth factors for tumor cells1 and a variety of matrix-degrading proteinases that facilitate tumor inva* Corresponding author. Tel.: + 39 80 5478240; fax: +39 80 5478310. E-mail address: [email protected] (D. Ribatti).



sion.2 An expanding endothelial surface also gives tumor cells more opportunities to enter the circulation and metastasize,3 while their release of anti-angiogenic factors explains the control exerted by primary tumors over metastasis. These observations suggest that tumor angiogenesis is linked to a switch in the equilibrium between positive and negative regulators.4 In normal tissues, vascular quiescence is maintained by the dominant influence of endogenous angiogenesis inhibitors over angiogenic stimuli. Tumor angiogenesis, on the other hand, is induced by increased secretion of angiogenic factors and/or down-regulation of angiogenesis inhibitors.

0305-7372/$ - see front matter c 2006 Elsevier Ltd. All rights reserved. doi:10.1016/j.ctrv.2006.06.002

438

Angiogenesis in hepatocellular carcinoma There are two types of microvascular structures in the liver: large vessels, such as portal and central venules and hepatic arterioles, lined with continuous endothelial cells, and fenestrated sinusoids lined with discontinuous sinusoidal endothelial cells (SEC).5 The adult liver is highly vascularized. Its cells are flanked by SEC-lined sinusoids and separated from them by the spaces of Disse.6 In liver tumors capillaries are formed and sinusoid fenestration is lost. Stellate cells are regarded as liver-resident pericytes. They may contribute to angiogenesis through mechanisms different from those attributed to pericytes and become activated. Fibrillar extracellular matrix accumulates and hepatocytes lose microvilli. The result is a distorted sinusoidal structure characterized by capillarization and formation of neovessels. A unique proangiogenic factor, namely the recently described AN-GPTL3, a liver-specific secreted factor showing angiogenic properties by binding to avb3 integrin, has been identified.7 Unlike the dual supply of the normal hepatic parenchyma provided by vessels arising from the systemic arterial circulation and the portal venous circulation, an advanced hepatocellular carcinoma (HCC) is abundantly supplied by systemic arteries alone.8 This abundancy facilitates the angiographic diagnosis of malignancy and differentiates non-neoplastic or preneoplastic conditions, such as liver cirrhosis, hepatocellular adenoma, focal nodular hyperplasia and adenomatous hyperplasia. HCC progresses from a small, well-differentiated tumor with no developed blood vessels to a larger and moderately or poorly differentiated form with a characteristic hypervascularity during dedifferentiation process.9–11 Many immunohistochemical and ultrastructural studies have investigated the immunophenotypical and morphological changes in liver sinusoids and SEC during liver carcinogenesis. These changes, known as sinusoidal capillarization, seem to be concomitant with the malignant transformation of precancerous lesions.12 Modifications of the immunophenotype vascular profile consist of abnormal distribution of endothelial markers (factor VIII-related antigen, CD31, CD34 and Ulex europaeus agglutinin-1) and changes in the basement membrane components (laminin and collagen type IV). Angiogenesis in HCC is usually unremarkable, whereas advanced forms are richly supplied with blood vessels.

Endogenous angiogenic stimulators expressed in HCC Vascular endothelial growth factor (VEGF) Since liver cells express VEGF13,14 and endothelial cells express its tyrosine kinase receptors VEGFR-1 and VEGFR2,15,16 it has been suggested that they communicate by means of VEGF signaling: signaling via VEGFR-1 induces SEC to release cytokines that stimulate hepatocyte proliferation; signaling via VEGFR-2 stimulates SEC proliferation.17 VEGF is expressed in HCC.18,19 Mise et al.19 have shown that most HCC samples display a stronger expression of VEGF mRNA. No apparent correlation was observed between the angiogenic gene expression and tumor size or grade.

D. Ribatti et al. Chow et al.20 found that immunoreactivity for VEGF is present in the extracellular matrix of the portal tracts in the normal liver, whereas reactivity in neoplastic tissue may characterize a progression towards higher proliferation in vivo. Yamaguchi et al.18 reported that VEGF protein expression is related to HCC grade: the highest expression was observed in well-differentiated HCC, followed by moderately and poorly differentiated HCC. VEGF expression increases gradually from low-grade dysplastic nodules to high-grade dysplastic nodules to early HCC.21 Its degree correlates with microvascular density and with CD34 staining as a marker of sinusoidal capillarization.21 Tumor expression of VEGF (mRNA and protein expression) significantly correlates with serum VEGF levels in HCC patients.22 Concentration of circulating VEGF increases with advancing HCC stage and is highest in patients with metastasis.23 In 98 patients with resectable HCC, preoperative serum VEGF was a significant and independent predictor of recurrence, disease-free survival and overall survival.24 In a prospective study of 80 patients undergoing transarterial chemoembolization in HCC, pretreatment serum VEGF levels were significantly lower in patients with stable or responsive disease than in those with progressive disease.25 Overexpression of VEGF has been found in HCC compared with cirrhosis or normal liver and is strongly increased in areas directly adjacent to necrotic/hypoxic regions.26

Fibroblast growth factor-2 (FGF-2) A direct correlation between the levels of FGF-2 expression and microvessel density in HCC has been found.27 Moreover, the serum FGF-2 level correlates with clinicopathological features and increased rate of postoperative recurrence.28

Angiogenin Serum concentration and tumoral mRNA expression of angiogenin correlated with microvascular density in a study of 41 HCC patients.29 Angiogenin serum levels significantly decreased after successful TACE or percutaneous ethanol injection. HCC patients with values higher than the mean level had a lower 5-year survival rate.29

Angiopoietins (Ang) Tanaka et al.30 reported that the expression of the Ang-2 gene is up-regulated gene in the hypervascular type of human HCC and that its ectopic expression in non-expressing HCC cells promotes rapid tumor growth and extensive hemorrhage in mice, suggesting that the Ang-Tie system might be involved in angiogenesis in HCC. Mitsuhashi et al.31 investigated the expression of Ang-1, Ang-2, their ligand Tie-2, and VEGF by RT-PCR and immunohistochemical staining in HCC, including adjacent non-tumor liver tissue. VEGF mRNA was significantly upregulated in HCC compared with normal liver tissue, but no differences were found between HCC and adjacent liver tissue. In contrast, Ang-2 mRNA expression in HCC significantly increased when compared with adjacent liver tissue. Ang-1 and Tie-2 expression was not significantly different. Immunohistochemical studies also showed increased Ang-2

Angiogenesis and anti-angiogenesis in hepatocellular carcinoma protein in HCC. The Ang-2/1 mRNA ratios in HCC were closely associated with tumor portal vein invasion, tumor diameter, and the microvascular density. The high Ang-2/ 1 mRNA ratio group showed a significantly poorer prognosis. Sugimachi et al.32 showed that in resected HCC samples, Ang-2 was immunohistochemically detected in more poorly differentiated HCC cells, and Tie-2 was detected in vascular endothelial cells and pericytes. In contrast, Ang-1 expression was observed in normal hepatocytes. Ang-2 has been detected in hepatoma cells at the mRNA level as well as immunohistochemically, whereas its ligand Tie-2 was detected in endothelial cells.33

How different cells and factors contribute to an enhanced angiogenesis in HCC Secretion by HCC cells, infiltrating inflammatory cells and hepatic stellate cells of angiogenic factors, such as VEGF, FGF-2, angiogenin and Angs, promotes the sprouting of new vessels from pre-existing ones. In fact, tumor angiogenesis does not depend on a single molecule, since many angiogenic inducers and inhibitors are simultaneously expressed. Peng et al.34 demonstrated that the counts of tumorassociated macrophages, mast cells and microvessels were significantly higher in metastatic HCC than that in non-metastatic HCC. The infiltrating inflammatory cells secrete several angiogenic factors and may contribute to tumorassociated angiogenesis and there are many reports of associations between macrophage and mast cell infiltration, vascularity and prognosis.35,36 Hepatitis B virus X protein has been shown to increase the transcriptional activity and protein level of hypoxia inducible factor (HIF), a transcriptional factor inducing the expression of several hypoxia response genes, such as VEGF.37 Accordingly, hepatitis B virus promotes angiogenesis during hepatocarcinogenesis.38 Human HCC cells express high levels of matrix metalloproteinases (MMP)39,40 and this overexpression correlates with invasive phenotype.41 MMPs, in turn, promote the release of extracellular matrix-bound or cell-surface-bound cytokines,42 such as VEGF, and further upregulates proangiogenic factors in hepatocarcinogenesis.

Endogenous angiogenic inhibitors expressed in HCC

439 Ishikawa et al.45 stably transfected a vector expressing the angiostatin gene into an HCC cell line to investigate the mechanism of the anti-angiogenic property of angiostatin gene transduction on the expression of VEGF and the effect of transfection on the proliferation and migration of cultured HUVECs in vitro and on the progression of tumors implanted subcutaneously in nude mice. Results showed that expression of angiostatin in HCC cells did not influence the expression of VEGF. However, conditioned media derived from angiostatin-gene-transfected cells suppressed the proliferation and migration of HUVECs. Lastly, suppression of tumor growth was noted in mice implanted with these cells and analysis of the tumor vessel density showed that growth suppression by angiostatin correlated with suppression of vascularity.

Thrombospondin-1 (TSP-1) Kawahara et al.46 reported that the mRNA level of TSP-1 significantly increased in hypovascular cholangiocarcinoma compared to hypervascular HCC, whereas Poon et al.47 showed that TSP-1 expression is associated with invasion and tumor progression in HCC.

Anti-angiogenesis in HCC (Table 1) In 1971, Folkman48 advanced the view that tumor growth depends on angiogenesis and that targeting such angiogenesis could be a new way of preventing tumor progression. Three strategies block tumor growth in experimental models through regression of angiogenesis: vascular targeting, gene therapy and direct inhibition of proliferating and migrating endothelial cells. Alternatively, indirect antiangiogenic drugs prevent the expression or block the activity of tumor proangiogenic factors by interfering with their endothelial cell receptors. Direct angiogenesis inhibitors target the microvascular endothelial cells recruited to the tumor and prevent them from responding to mitogens and motogens. Indirect angiogenesis inhibitors generally prevent the expression of or block the activity of a tumor protein that activates angiogenesis, such as FGF-2 and VEGF, or block the expression of its receptors on endothelial cells. Approximately 75 anti-angiogenic compounds have been developed and are being tested clinically. An overview of ongoing studies on anti-angiogenic treatment is provided by the National Cancer Institute (http://www.cancer.gov).

Endostatin Tyrosine kinase inhibitors Dhar et al.43 measured serum endostatin, VEGF and FGF-2 levels by ELISA in samples from 33 patients who had received no preoperative therapy. They demonstrated a significant decrease in postoperative compared with preoperative serum endostatin and FGF-2 values, and a very strong direct correlation between VEGF and endostatin. Moreover, they found that the serum endostatin was significantly higher in living patients and those with a high level had tended to survive longer. Similarly, Hu et al.44 reported that poorly differentiated hepatoma cell lines exhibited increased endostatin levels compared with the well-differentiated ones.

PTK 787/ZK22254 is a small molecular weight inhibitor of the tyrosine kinase activity of VEGFR-1 and VEGFR-2 and is given orally.49 Preclinical studies have demonstrated its inhibition of tumor vasculature, alone or in combination with chemotherapy or radiotherapy. Liu et al.50 examined the effects of PTK 787 on tumor growth and angiogenesis of HCC xenografts in nude mice. Oral administration significantly reduced tumor volume and microvessel formation, inhibited tumor cell proliferation in a dose-dependent manner and also induced tumor cell apoptosis both in vivo and in vitro. The proapoptotic

440 Table 1

D. Ribatti et al. Antiangiogenic agents applied for treatment of HCC

Agent

Mechanism of action

Stage

References

Bevacizumab Thalidomide IFN-a IL-12 PTK 787 Gefitinib TNP-470 Batimastat (BB-94) Endostatin

Inhibition Inhibition Inhibition Inhibition Inhibition Inhibition Inhibition Inhibition Inhibition

Phase II Phase II Phase II Murine model Murine model Murine model Murine model Murine model Phase I

Schwartz et al.56,57 Lin et al.65 Patt et al.66 Peron et al.77 Liu et al.50 Matsuo et al.53 Kin et al.81 Bu et al.82 Eder Jr., et al.85

of of of of of of of of of

VEGF endothelial cell proliferation endothelial cell migration angiogenesis tyrosine kinase EGFR angiogenesis MMP tumor cell growth

response was associated with down-regulation of Bcl-2 and Bcl-xL expression and induction of cleavage of caspase 3. In addition, PTK 787 induced growth arrest in HCC cells. This was associated with G1 arrest and partial G2-M block. Raskpof et al.51 used a recombinant adenoviral vector encoding a soluble dominant negative fragment of VEGFR2, Ads Flk-1, to control murine orthotopic and metastatic hepatomas. Systemic treatment of tumor-bearing mice inhibited tumor growth by 84% compared with the corresponding control group. Numerous reports have described overexpression of epidermal growth factor receptor (EGFR) in epithelial tumors and substantiated the view that deregulated EGFR signaling has an important role in human cancer. EGFR inhibitors are currently under clinical development. Many display antiangiogenic activity, at least in preclinical models, and this could partly explain their clinical efficacy.52 In a murine HCC model, oral Gefitinib (ZD 1839 or Iressa), a potent EGFR inhibitor, inhibited the growth of implanted HCC and intrahepatic metastasis by approximately 50% and prevented EGF-induced chemotactic migration of HCC cells and production of active MMP-9 in vitro.53

Thalidomide Thalidomide is a compound with anti-angiogenic, immunomodulatory and antitumor effects. Its most promising clinical results have been obtained in multiple myeloma,59 whereas it has proved poorly effective in recurrent glioma, breast cancer, melanoma, renal and ovarian cancer and hormone-refractory prostate cancer.60–62 Hsu et al.63 reported the results of a trial with 68 patients with unresectable and non-embolizable HCC, treated with thalidomide. One complete and three partial responses were seen, with a response rate of 6.3%. Wang et al.64 treated 99 patients and obtained six partial responses. Lin et al.65 treated 27 patients with unresectable HCC. One patient showed near complete recovery of a-fetoprotein levels and a partial radiographic response; another two patients had stable disease. The overall median survival was 123 days. Finally, Patt et al.66 treated 37 patients with thalidomide and showed that one patient had a partial response, one a minor response and 10 a stable disease. The overall median survival was 6.8 months.

Interferon alpha (IFN-a) Bevacizumab (Avastin) Bevacizumab is a humanized murine anti-VEGF monoclonal antibody, which recognizes all VEGF isoforms without cross-reacting with other growth factors, displaying potent antitumor activity in experimental models.54 First-line use of bevacizumab with irinotecan, 5-fluorouracil and leucovorin produced significantly improvements in response rate, duration of response and survival in a phase III study of patients with metastatic colorectal cancer.55 Bevacizumab has been tested in a phase II study in patients with unresectable non-metastatic HCC and without main portal invasion.56,57 Bevacizumab is safe and potentially efficacious in localized HCC in patients with adequate liver function and results suggest significant disease-modifying effect, including six month stability in patients with prior rapid growth.56,57 More recently, a phase II study undertaken to examine the efficacy and safety profiles of combining bevacizumab with the combination of gemcitabine and oxaplatin (GEMOX) in patients with unresectable or metastatic HCC, has demonstrated a moderate antitumor activity of this combination.58

IFN-a was first shown to inhibit endothelial cell migration by Brouty-Boye and Zetter.67 This led to its subsequent characterization as an angiogenesis inhibitor. Its anti-angiogenic effects in humans are illustrated by its efficacy in the treatment of pediatric hemangiomas,68 which express high levels of FGF-2 as their major angiogenic mediator. Metastasis and recurrence of human HCC after resection were inhibited by IFN-a in a study with nude mice showing decreased VEGF serum levels and decreased tumoral microvascular density, but not FGF-2 levels.69 Shiratori et al.70 treated with IFN-a patients with compensated cirrhosis, three or fewer nodules of HCC and low HCV RNA loads with complete ablation of lesions by percutaneous ethanol injection therapy. Of the 49 patients treated, 21 showed a sustained biochemical response and 14 showed a sustained virologic response. A clinical prospective randomized controlled study found that IFN-a prevented HCC recurrence reduced after medical ablation therapy for primary tumors.71 In a phase II trial, 34 patients with HCC and 9 with fibrolamellar HCC were treated with subcutaneous recombinant IFN-a2b and systemic

Angiogenesis and anti-angiogenesis in hepatocellular carcinoma continuous 5-fluorouracil.72 A partial response was observed in 14% of those with HCC and 62.5% of those with fibrolamellar HCC.

441 Rats with deficient-diet-induced HCC treated with TNP470 developed fewer and smaller tumors than control animals. This was accompanied by a reduction of tumor vascularity.81

Interleukin-12 (IL-12) TIMPs Il-12 blocks corneal angiogenesis in normal and immunodeficient mice, suggesting that the presence of immunocompetent T-cells is not a prerequisite for its anti-angiogenic activity.73 Il-12 acts via the release of IFN-b and consequently interferon-inducible protein 10 (IP-10), a member of the CXC chemokine family, which can itself block the activity of FGF-2.74 In addition, down-regulation of MMP-9 and increase of tissue inhibitor of metalloproteinase (TIMP-1) may also contribute to the angioinhibitory effects of IL-12.75 In a rat model of orthotopic HCC, adenovirus-mediated gene transfer of IL-12 inhibited tumor growth in a dosedependent manner.76 Similar results were observed in a murine model with either tumor cells expressing IL-12 or injection of the tumor with fibroblasts secreting IL-12.77

TNP-470 TNP-470 (or AGM-1470) is a synthetic analog of fumagillin, a natural angiogenesis inhibitor. It acts by inhibiting the phosphorylation of methionine aminopeptidase-2, a cytoplasmic enzyme of endothelial cells, which is overexpressed in tumor cells. TNP-470 has been applied in clinical trials in patients with carcinoma of the cervix metastasized to the lung78 and renal cell carcinoma.79 Combination of TNP470 with conventional cytotoxic agents, such as cisplatin, paclitaxel or cyclophosphamide, significantly improve their antitumor efficacy.80

Endostatin In preclinical studies in mice endostatin has been found to inhibit the growth of metastatic and primary liver tumors without drug resistance or toxicity.83,84 A phase I clinical trial of recombinant human endostatin given in 15 patients with refractory solid tumors showed no significant toxicities and evidence of clinical benefit in three patients.85

The winning strategy The complex relationship between angiogenic cascade and anti-angiogenic agents in the tumor vascular phase, as well as identification and characterization of angiogenesis inhibitors, have indicated that anti-angiogenesis can be considered as a strategy for the adjuvant therapy of HCC (Fig. 1).

Cirrhotic liver

Normal liver

Sinusoid capillaries

MMP inhibitors have been extensively assessed in a broad range of malignant diseases because of their ability to inhibit MMP-2 and MMP-9, which are thought to be important for an early stage of angiogenesis. Because they inhibit MMP, it was believed they could inhibit tumor growth, invasion, angiogenesis and metastasis. Batimastat (BB-94), a synthetic inhibitor of MMP, inhibited primary tumor growth, local invasion and lung metastasis, and also prolonged survival in an orthotopic metastatic human HCC nude mice model.82

Extracellular matrix

HCC

Fibrosis with laminin and collagen type IV

Capillaries stained with FVIII-RA, CD31, CD34

Tumor nodule with neovessels and fibrosis

Inducers VEGF FGF-2 Angiogenin Angiopoietins

Figure 1

Endogenous inhibitors

Endostatin Thrombospondin-1

Major angiogenesis inducers and endogenous angiogenesis inhibitors in hepatocellular carcinoma (HCC).

442 The results of preclinical studies with anti-angiogenic agents had been very promising showing partly or complete tumor regression without any drug resistance. However, many of these results could not be confirmed in clinical trials. In fact, objective tumor responses were achieved only in some cases. Especially, the minimal antitumoral effects of endostatin in doses equivalent to those used in animal models has been disappointing. VEGF receptors tyrosine kinase inhibitors or thalidomide might actually the most promising substances (Table 1). The advantage of thalidomide is that it is administered orally and, in general, is well tolerated, especially in patients without cirrhosis. Conventional cytotoxic chemotherapeutic drugs have been used at low and non-cytotoxic concentrations as anti-angiogenic. Anti-endothelial effects have been demonstrated in vitro and in vivo for several cytostatic agents. The presence of dividing endothelial cells in newly forming tumor blood vessels should render such vessels, in contrast to the mature vessels found in normal adult tissues, sensitive to the cytotoxic effects of such drugs. The use of low doses in ‘‘metronomic’’ chemotherapy (namely, very frequent or continuous low-dose chemotherapy) as anti-angiogenic targeting strategy, seems particularly effective against drug-resistant tumors, especially when it is combined with a second anti-angiogenic drug.86 Although there are no studies of metronomic chemotherapy in patients with HCC, this modality might be useful in patients with diminished hepatic functional reserve and a reduced capacity to metabolize and excrete chemotherapeutic drugs. There is accumulating evidences that a number of antiangiogenic substances can lose their activity over time. This could be caused by several possible mechanisms, e.g. an overexpression of other angiogenic factors that may antagonize the function of the anti-angiogenic agents if only one of such factors is the target for an anti-angiogenic therapy. For example, blockade of the VEGF pathway seems too narrow as an approach to inhibit angiogenesis, because a tumor may use a compensatory pathway. For this reason, the combined application of angiogenesis inhibitors might be a promising strategy. Inhibition of various receptor/ligand systems could be effective due to the different mechanism of action within the angiogenesis process and combination of inhibitors with different mechanisms of action attacking different biochemical pathways should shut down the multifactor stimulated cascade of biochemical angiogenic processes.

Acknowledgement Supported by Associazione Italiana per la Ricerca sul Cancro AIRC (Regional Funds) Milan, Fondazione Italiana per la Lotta al Neuroblastoma, Genoa, MIUR (FIRB2001 and PRIN2005) Rome, Italy.

References 1. Ribatti D, Vacca A, Dammacco F. The role of the vascular phase in solid tumor growth: a historical review. Neoplasia 1999;1: 293–302. 2. Mignatti P, Rifkin DB. Biology and biochemistry of proteinases in tumor invasion. Physiol Rev 1993;73:161–95.

D. Ribatti et al. 3. Aznavoorian S, Murphy AN, Stetler-Stevenson WG, Liotta LA. Molecular aspects of tumor cell invasion. Cancer 1993;71: 1368–83. 4. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86: 353–64. 5. Mc Cuskey RS, Reilly F. Hepatic microvasculature: dynamic structure and its regulation. Sem Liver Dis 1993;13:1–12. 6. Mc Cuskey RS. The hepatic microvascular system. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Schachter DA, Shafritz DA, editors. The liver: biology and pathology. New York: Raven Press; 1994. p. 1089–106. 7. Camenisch G, Pisabarro M, Sherman D, et al. ANGPTL3 stimulates endothelial cell adhesion and migration via integrin alpha v beta 3 and induces blood vessel formation in vivo. J Biol Chem 2002;227:17281–90. 8. Nakashima T, Kojiro M. Angioarchitecture of hepatocellular carcinoma. In: Hepatocellular carcinoma: an atlas of its pathology. Tokyo: Springer Verlag; 1987. p. 105–15. 9. Maeda T, Adachi E, Kajiyama K, et al. CD34 expression in endothelial cells of small hepatocellular carcinoma: its correlation with tumour progression and angiographic findings. J Gastroenterol Hepatol 1995;10:650–4. 10. Sonoda T, Shirabe K, Takenaka K, Kanematsu T, Yasumori K, Sugimachi K. Angiographically undetected small hepatocellular carcinoma: clinicopathological characteristics, follow-up and treatment. Hepatology 1989;10:1003–7. 11. Sakamoto M, Ino Y, Fujii T, Hirohashi S. Phenotype changes in tumor vessels associated with the progression of hepatocellular carcinoma. Jpn Clin Oncol 1993;23:98–104. 12. Haratake J, Scheuer PJ. An immunohistochemical and ultrastructural study of the sinusoids of hepatocellular carcinoma. Cancer 1990;65:1985–93. 13. Mochida S, Ishikawa K, Toshima K, et al. The mechanisms of hepatic sinusoidal endothelial cell regeneration: a possible communication system associated with vascular endothelial growth factor in liver cells. J Gastroenterol Hepatol 1998;13S: S1–5. 14. Yamane A, Seetharam L, Yamaguchi S, et al. A new communication system between hepatocytes and sinusoidal endothelial cells in liver through vascular endothelial growth factor and Flt tyrosine kinase receptor family (Flt-1 and KDR/Flk-1). Oncogene 1994;9:2683–90. 15. De Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT. The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science 1992;255:989–91. 16. Quinn TP, Peters KG, De Vries C, Ferrara N, Williams LT. Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc Natl Acad Sci USA 1993;90:7533–7. 17. Le Couter J, Moritz D, Li B, et al. Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 2003;299:890–3. 18. Yamaguchi R, Yano H, Iemura A, Ogasawara S, Haramaki M, Kojiro M. Expression of vascular endothelial growth factor in human hepatocellular carcinoma. Hepatology 1998;28:68–77. 19. Mise M, Arii S, Higashituji H, et al. Clinical significance of vascular endothelial growth factor in normal liver and hepatocellular carcinoma: an immunohistochemical study. Hepatology 1996;23:455–64. 20. Chow NH, Hsu PI, Lin XZ, et al. Expression of vascular endothelial growth factor in normal liver and hepatocellular carcinoma: an immunohistochemical study. Hum Pathol 1997;28:698–703. 21. Park YN, Kim YB, Yang KM, Park C. Increased expression of vascular endothelial growth factor and angiogenesis in the early stage of multistep hepatocarcinogenesis. Arch Pathol Lab Med 2000;124:1061–5.

Angiogenesis and anti-angiogenesis in hepatocellular carcinoma 22. Poon RT, Lau CP, Cheung ST, Yu WC, Fan ST. Quantitative correlation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Res 2003;63:3121–6. 23. Jinno K, Tanimizu M, Hyodo I, et al. Circulating vascular endothelial growth factor (VEGF) is a possible tumor marker for metastasis in human hepatocellular carcinoma. J Gastroenterol 1998;33:376–82. 24. Chao Y, Li CP, Chau GY, et al. Prognostic significance of vascular endothelial growth factor, basic fibroblast growth factor, and angiogenin in patients with resectable hepatocellular carcinoma after surgery. Ann Surg Oncol 2003;10: 355–62. 25. Poon R, Lau C, Yu W, Fan S, Wong J. High serum level of vascular endothelial growth factor predict poor response to transarterial chemoembolization in hepatocellular carcinoma: a prospective study. Oncol Rep 2004;11:1077–84. 26. von Marschall Z, Cramer T, Hocker M, Finkenzeller G, Wiedenmann B, Rosewicz S. Dual mechanism of vascular endothelial growth factor upregulation by hypoxia in human hepatocellular carcinoma. Gut 2001;48:87–96. 27. El-Assal ON, Yamanoi A, Ono T, Kohno H, Nagasue N. The clinicopathological significance of heparanase and basic fibroblast growth factor expression in hepatocellular carcinoma. Clin Cancer Res 2001;7:1299–305. 28. Poon RT, Hg IO, Lau C, Yu WC, Fan ST, Wong J. Correlation of serum basic fibroblast growth factor levels with clinicopathological features and postoperative recurrence in hepatocellular carcinoma. Am J Surg 2001;182:298–304. 29. Hisai H, Kato J, Kobune M, et al. Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity. Clin Cancer Res 2003;9:4849–52. 30. Tanaka S, Mori M, Sakamoto Y, Makuuchi M, Sugimachi K, Wands JR. Biologic significance of angiopoietin-2 expression in human hepatocellular carcinoma. J Clin Invest 1999;103:341–5. 31. Mitsuhashi N, Shimizu H, Ohtsuka M, et al. Angiopoietin and Tie-2 expression in angiogenesis and proliferation of human hepatocellular carcinoma. Hepatology 2003;37:1105–13. 32. Sugimachi K, Tanaka S, Terashi T, Taguchi K, Rikimaru T. The mechanisms of angiogenesis in hepatocellular carcinoma: angiogenic switch during tumor progression. Surgery 2002;131:S135–41. 33. Chen L, Yang Z, Wang G, Wang C. Expression of angiopoietin-2 gene and its receptor Tie-2 in hepatocellular carcinoma. J Tongji Med Univ 2001;21:228–30. 34. Peng SH, Deng H, Yang JF, et al. Significance and relationship between infiltrating inflammatory cell and tumor angiogenesis in hepatocellular carcinoma tissues. World J Gastroenterol 2005;11:6521–4. 35. Bingle L, Brown NJ, Lewis CE. The role of tumor associated macrophages in tumor progression: implications for new anticancer therapies. J Pathol 2002;196:254–65. 36. Ribatti D, Crivellato E, Roccaro AM, Ria R, Vacca A. Mast cell contribution to angiogenesis related to tumor progression. Clin Exp Allergy 2004;34:1660–4. 37. Semenza GL. Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med 2001;7:345–50. 38. Moon EJ, Jeong CH, Jeong JW, et al. Hepatitis B virus X protein induces angiogenesis by stabilizing hypoxia-inducible factor 1 alpha. FASEB J 2004;18:382–4. 39. Kim JH, Kim TH, Jang JW, et al. Analysis of matrix metalloproteinase mRNAs expressed in hepatocellular carcinoma cell lines. Mol Cells 2001;12:32–40. 40. Monvoisin A, Bisson C, Si-Tayeb K, Balabaud C, Desmouliere A, Rosenbaum J. Involvement of matrix metalloproteinase type-3 in hepatocyte growth factor-induced invasion of human hepatocellular carcinoma cells. Int J Cancer 2002;97:157–62.

443 41. Giannelli G, Bergamini C, Fransvea E, Marinosci F, Quaranta V, Antonaci S. Human hepatocellular carcinoma (HCC) cells require both alpha 3 beta 1 integrin and matrix metalloproteinase activity for migration and invasion. Lab Invest 2001;81: 613–27. 42. McMahon G, Vu TH, Ito HT, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2000;2:737–44. 43. Dhar DK, Ono T, Yamanoi A, et al. Serum endostatin predicts tumor vascularity in hepatocellular carcinoma. Cancer 2002;95:2188–95. 44. Hu TH, Huang CC, Wu CL, et al. Increased endostatin/collagen XVIII expression correlates with elevated VEGF level and poor prognosis in hepatocellular carcinoma. Mod Pathol 2005;18: 663–72. 45. Ishikawa H, Nakao K, Matsumoto K, et al. Antiangiogenic gene therapy for hepatocellular carcinoma using angiostatin gene. Hepatology 2003;37:696–704. 46. Kawahara N, Ono M, Taguchi K, et al. Enhanced expression of thrombospondin-1 and hypovascularity in human choloangiocarcinoma. Hepatology 1998;28:1512–7. 47. Poon RT, Chung KK, Cheung ST, et al. Clinical significance of thrombospondin-1 expression in hepatocellular carcinoma. Clin Cancer Res 2004;10:4150–7. 48. Folkman J. Tumor angiogenesis: therapeutic implications. New Engl J Med 1971;285:1182–6. 49. Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growthfactor-induced responses and tumor growth after oral administration. Cancer Res 2000;60:2178–89. 50. Liu Y, Poon RT, Li Q, Kok TW, Lau C, Fan S. Both antiangiogenesis and angiogenesis-independent effects are responsible for hepatocellular carcinoma growth arrest by tyrosine kinase inhibitor PTK787/ZK22584. Cancer Res 2005;65: 3691–9. 51. Raskopf E, Dzienisowicz C, Hilbert T, et al. Effective angiostatic treatment in a murine metastatic and orthotopic hepatoma model. Hepatology 2005;41:1233–40. 52. Ranson M. Epidermal growth factor receptor tyrosine kinase inhibitors. Br J Cancer 2004;90:2250–5. 53. Matsuo M, Sakurai H, Saiki I. ZD1839, a selective epidermal growth factor receptor tyrosine kinase inhibitor, shows antimetastatic activity using a hepatocellular carcinoma model. Mol Cancer Ther 2003;2:557–61. 54. Presta LG, Chen H, O’Connor SJ, et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res 1997;57:4539–99. 55. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350: 2335–42. 56. Schwartz JD, Schwartz M, Goldman J, et al. Bavacizumab in hepatocellular carcinoma in patients without invasion of the portal vein. J Clin Oncol, 2004 ASCO Annual Meeting Proceedings 2004;22(14S):4088. 57. Schwartz JD, Schwartz M, Lehrer D, et al. Bevacizumab in hepatocellular carcinoma (HCC) in patients without metastasis and without invasion of the portal vein. J Clin Oncol, 2005 ASCO Annual Meeting Proceedings 2005;23(16S):4122. 58. Zhu AX, Blaszkowsky LS, Ryan DP, et al. Phase II study of gemcitabine and oxaliplatin in combination with bevacizumab in patients with advanced hepatocellular carcinoma. J Clin Oncol 2006;24:1898–903. 59. Ribatti D, Vacca A. Therapeutic renaissance of thalidomide in the treatment of haematological malignancies. Leukemia 2005;19:1525–31.

444 60. Figg WD, Raje S, Bauer SK, et al. Pharmacokinetics of thalidomide in AIDS-related Kaposi’s sarcoma. J Pharmacol Sci 1999;88:121–5. 61. Fine HA, Figg WD, Jaeckle K, et al. Phase II trial of the antiangiogenic agent thalidomide in patients with recurrent high-grade gliomas. J Clin Oncol 2000;18:708–15. 62. Eisen T, Boschoff C, Mak I, et al. Continuous low dose thalidomide: phase II study in advanced melanoma, renal cell, ovarian and breast cancer. Br J Cancer 2000;82:812–7. 63. Hsu C, Chen CN, Chen LT, et al. Low-dose thalidomide treatment for advanced hepatocellular carcinoma. Oncology 2003;65:242–9. 64. Wang TE, Kao CR, Lin SC, et al. Salvage therapy for hepatocellular carcinoma with thalidomide. World J Gastroenterol 2004;10:649–53. 65. Lin AY, Brophy N, Fisher GA, et al. Phase II study of thalidomide in patients with unresectable hepatocellular carcinoma. Cancer 2005;103:119–25. 66. Patt YZ, Hassan MM, Lozano RE, et al. Thalidomide in the treatment of patients with hepatocellular carcinoma: a Phase II trial. Cancer 2005;103:749–55. 67. Brouty-Boye D, Zetter BR. Inhibition of cell motility by interferon. Science 1980;208:516–8. 68. Folkman J. Endogenous angiogenesis inhibitors. APMIS 2004;112:496–507. 69. Wang L, Wu WZ, Sun HC, et al. Mechanism of interferon alpha on inhibition of metastasis and angiogenesis of hepatocellular carcinoma after curative resection in nude mice. J Gastroenterol Surg 2003;7:587–94. 70. Shiratori Y, Shiina S, Teratani T, et al. Interferon therapy after tumor ablation improves prognosis in patients with hepatocellular carcinoma associated with hepatitis C virus. Ann Intern Med 2003;138:299–306. 71. Lin SM, Lin CJ, Hsu CW, et al. Prospective randomized controlled study of interferon-alpha in preventing hepatocellular carcinoma recurrence after medical ablation therapy for primary tumors. Cancer 2004;100:376–82. 72. Patt YZ, Hassan MM, Lozano RD, et al. Phase II trial of systemic continuous fluorouracil and subcutaneous recombinant interferon alpha-2b for treatment of hepatocellular carcinoma. J Clin Oncol 2003;21:421–7. 73. Voest EE, Kenyon BM, O’Reilly MS, Truitt G, D’Amato RJ, Folkman J. Inhibition of angiogenesis in vivo by interleukin 12. J Natl Cancer Inst 1995;87:581–6.

D. Ribatti et al. 74. Sgadari C, Angiolillo AL, Tosato G. Inhibition of angiogenesis by interleukin-12 is mediated by the interferon-inducible protein 10. Blood 1996;87:3877–82. 75. Dias S, Boyd R, Balkwill F. IL-12 regulates VEGF and MMPs in a murine breast cancer model. Int J Cancer 1998;78:361–5. 76. Barajas M, Mazzolini G, Genove G, et al. Gene therapy of orthotopic hepatocellular carcinoma in rats using adenovirus coding for interelukin 12. Hepatology 2001;33:52–61. 77. Peron J, Couderc B, Rochaix P, et al. Treatment of murine hepatocellular carcinoma using genetically modified cells to express interleukin-12. J Gastroenterol Hepatol 2004;19: 388–96. 78. Kudelka AP, Verschraegen CF, Loyer E. Complete remission of metastatic cervical cancer with the angiogenesis inhibitor TNP470. New Engl J Med 1998;338:981–92. 79. Stadler WM, Kuzel T, Shapiro C, Sosman J, Clark J, Vogelzang NJ. Multi-institutional study of the angiogenesis inhibitor TNP470 in metastatic renal carcinoma. J Clin Oncol 1999;17: 2541–5. 80. Teicher BA. Potentiation of cytotoxic cancer therapies by antiangiogenic agents. In: Teicher BA, editor. Antiangiogenic Agents in Cancer Therapy. Totowa: Humana Press; 1999. p. 277–316. 81. Kin M, Torimura T, Uen T, et al. Angiogenesis inhibitor TNP470 suppresses the progression of experimentally-induced hepatocellular carcinoma in rats. Int J Oncol 2000;16: 375–82. 82. Bu W, Tang ZY, Sun FX, et al. Effects of matrix metalloproteinase inhibitor BB-94 on liver cancer growth and metastasis in a patient-like orthotopic model LCI-D20. Hepatogastroenterology 1998;45:1956–61. 83. Tai KF, Chen PJ, Chen DS, Hwang LH. Concurrent delivery of GM-CSF and endostatin genes by a single adenoviral vector provides a synergistic effect on the treatment of orthotopic liver tumors. J Gene Med 2003;5:386–98. 84. Chen CT, Lin J, Li Q, et al. Antiangiogenic gene therapy for cancer via systemic administration of adenoviral vector expressing secretable endostatin. Hum Gene Ther 2000;11: 1983–96. 85. Eder Jr JP, Supki JG, Clark JW, et al. Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily. J Clin Oncol 2002;20:3772–84. 86. Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nat Rev Cancer 2002;2:727–39.