Hepatocellular Carcinoma: Targeted Therapy and Multidisciplinary P5 ppsx

10 389 0
Hepatocellular Carcinoma: Targeted Therapy and Multidisciplinary P5 ppsx

Đang tải... (xem toàn văn)

Thông tin tài liệu

2 Biology of Hepatocellular Carcinoma 25 disease. The liver is different from the skin and gastrointestinal tract with regard to transit-amplifying cells, in that the most highly differentiated cells, the hepato- cytes, are not terminally differentiated and can respond to injury or loss by rapid, highly regulated proliferation. Thus, in the liver, differentiated hepatocytes per se can be viewed as the hepatic version of transit-amplifying cells. Given the postu- lated presence of a hepatic lineage of cells from periductal stem cells, to bipolar ductal progenitor cells, to hepatocytes, with each cell type proliferation competent, it would not be surprising to find that HCCs can arise from the stem cells, the bipo- lar ductal progenitor cells, or the hepatocytes [ 17]. The fact that stem cell activation precedes the development of HCC in almost all models of hepatocarcinogenesis and invariably accompanies chronic liver damage in humans makes it likely that the mature hepatocyte is not the cell of origin of all HCCs [18]. Detailed immunophe- notyping of HCCs indicated that 28–50% of HCCs express markers of progenitor cells such as CK7 and CK19 [19]. But what transforms a stem cell into a cancer stem cell (CSC)? There are at least two proposed mechanisms of CSC origin: onco- genic mutations may inactivate the constraints on normal stem cell expansion or, alternatively, oncogenic mutations in a more differentiated cell generate continual proliferation of cells that no longer enter a postmitotic differentiated state, thereby creating a pool of self-renewing cells in which further mutations can accumulate [20]. Several pathways have been proposed to be implicated in stem cell prolif- eration: TGFβ, Notch, Wnt and Hedgehog are some examples [19]. The detailed mechanisms directing transformation of stem cells to cancer stem cells and hepa- tocellular cancer, however, remain still to be elucidated and definitive markers for these putative cancer stem cells have not yet been established. Angiogenesis HCC is one of the most vascular solid cancers, associated with a high propen- sity for vascular invasion. In fact, its active neovascularization can be visualized in angiography and is used as a diagnostic criterion for HCC. The development of neovasculature in the tumour provides two essential functions for the growth and metastasis of a cancer. First, the vessels provide a route f or supply of nutrient and oxygen to sustain growth and excretion of metabolic waste. Second, the neovessels provide access for tumour cells to enter the circulation and spread as metastases. In fact, HCC is characterized by a high propensity for vascular invasion, and the angiogenic activity of HCC correlates with the risk of vascular invasion [21]. HCC typically develops from dysplastic nodules in a cirrhotic liver and the endothelial cells in these nodules undergo phenotypic changes during malignant transformation as demonstrated by changes in endothelial cell markers. The process of angiogene- sis is a complex multistep process initiated by the release of angiogenic factors from tumour cells. The angiogenic factors bind to specific receptors of endothelial cells of preexisting blood vessels and activate the endothelial cells, which then secrete enzymes to degrade the underlying basement membrane. The activated endothelial 26 M.L. Balmer and J F. Dufour cells then proliferate, migrate and assemble into new capillary tubes, followed by the synthesis of a new basement membrane. However, some recent studies suggested that some of the neovessels in tumours may be derived from circulating endothelial precursor cells that originate from the bone marrow [22, 23]. There is also evidence indicating that some tumours may be vascularized without significant angiogenesis, probably by using existing vessels through a process described as vascular co-option or even by forming vascular channels on their own through a nonendothelial cell process called ‘vascular mimicry’ [24]. Angiogenesis can be triggered by activation of oncogenes like ras or inactivation of tumour suppressor genes like p53 [25, 26]. In addition, a number of cellular stress factors such as hypoxia, nutrient deprivation or inducers of reactive oxygen species are important stimuli of angiogenic signalling [27]. Many angiogenic and antiangiogenic factors have been studied in recent years. Vascular endothelial growth factor (VEGF) is one of the first isolated angiogenic peptides and is the most well-studied angiogenic factor so far. It has a specific mito- genic effect on endothelial cells, and it also increases vascular permeability (hence also known as vascular permeability factor) and promotes extravasation of proteins from tumour vessels, leading to the formation of a fibrin matrix that supports the growth of endothelial cells and allows invasion of stromal cells into the developing tumour [28]. The expression of VEGF protein was found to correlate with clinico- pathological factors such as proliferation, vascular invasion and tumour multiplicity and was reported to associate with not only invasion and metastasis of HCC but also postoperative recurrence [29]. Expression of VEGF is regulated by microenviron- mental and genetic alterations in cancer cells. Hypoxia is a key microenvironmental factor of angiogenesis, and hypoxia-inducible factors (HIF) are known to stimulate VEGF expression [30, 31]. The upregulation of VEGF in HCC is controlled at tran- scriptional levels as well as by the mRNA stability of VEGF [32]. In addition, the p53 tumour suppressor and HBx genes might regulate VEGF expression in HCC [33, 34]. Fibroblast growth factors (FGFs) are a family of heparin-binding growth factors that includes at least 22 structurally related members, of which acidic fibroblast growth factor (aFGF) and basic fibroblast growth factor (bFGF) are the best-known members. FGFs exert their pro-angiogenic activity by interacting with various endothelial cell surface receptors, including tyrosine kinase receptors FGFR1 and FGFR2, heparan sulphate proteoglycans and integrins [35]. bFGF appears to act synergistically with VEGF in the induction of angiogenesis [36]. Aside from its angiogenic effect, bFGF has also been shown to act as a mitogen for HCC cell proliferation via an autocrine mechanism [37]. Angiopoietins play an important role in angiogenesis. Angiopoietin-1 (Ang-1) is a survival signal for endothelial cells and it promotes recruitment of pericytes and smooth muscle cells to form mature blood vessels. In contrast to Ang-1, angiopoi- etin 2 (Ang-2) induces vascular regression in the absence of VEGF but increases vascular sprouting in its presence. It has been shown that the ectopic expression of Ang-2 in HCC cells promotes rapid development of tumour and aggravates its prognosis [38]. 2 Biology of Hepatocellular Carcinoma 27 Other angiogeneic factors have also been shown to be involved in tumour angiogenesis including platelet-derived endothelial cell growth factor [39], tissue factor [40], cyclooxygenase-2 [41] and angiogenin [42]. Telomere Shortening The progressive shortening of telomeres with each cell division serves in most somatic cells as a “mitotic clock,” indicating cell age and cellular senescence. After a certain number of cell doublings, when a threshold level of telomeric length is reached, a signal is initiated to cease cell division and further progression to S-phase is prevented. Loss of telomeres initiates or drives chromosomal instability, which in turn results in chromosomal abnormalities such as end-to-end fusion and rearrangement. In carcinogenesis, certain cells such as those that have undergone viral transformation, irradiation or mutagenesis will continue to divide, have their telomeres further shortened and eventually die. However, prior to death, the result- ing genomic instability causes a small number of these cells to undergo multiple mutations, including the regaining of telomerase activity which thereafter serves to maintain telomere length and genomic stability indefinitely [43]. Telomere shorten- ing is accelerated in chronic liver disease and critically short t elomeres characterize cirrhosis stage [44]. The cancer risk increases in response to telomere shorten- ing during aging and chronic liver disease. Furthermore, telomerase activity has been detected in human HCC while it is absent in adjacent non-tumour tissues [43]. Studies in telomerase knockout mice have provided experimental evidence that telomere shortening influences stem cell function, aging and carcinogene- sis. These mice exhibit an impaired maintenance and function of adult stem cells and reduced regenerative reserve in response to organ damage. Interestingly they show an increase in chromosomal instability and tumour initiation but impaired tumour progression [45]. Telomerase has been shown to be a critical component for in vivo progression of p53 mutant HCC with short telomeres in the chroni- cally damaged liver. In this molecular context, telomerase limits the accumulation of telomere dysfunction, the evolution of excessive aneuploidy and the activation of p53-independent checkpoints suppressing hepatocarcinogenesis [46]. This dual role of telomeres may point to new treatment options in patients with HCC. HCC in the Non-cirrhotic Liver The incidence of HCC arising in the non-cirrhotic liver varies greatly between different studies, ranging from 10 to 50% [47–50]. These tumours have been char- acterized as often uninodular, encapsulated and expansive growing and they seem to be bigger than normal HCC [50]. All factors that can induce a HCC with cirrho- sis can also lead to a non-cirrhotic HCC. Nevertheless, there are several conditions which are known to be associated predominantly with non-cirrhotic liver cancer: sexual steroids induce HCCs through the development of liver adenomas; patients 28 M.L. Balmer and J F. Dufour with Alagille syndrome (arteriohepatic dysplasia) [51], hypercitrullinemia [51], α1-antitrypsin deficiency [52] and glycogenosis type 1 [53] often develop HCC without cirrhosis. Iron overload seems to be a general risk factor for developing a non-cirrhotic HCC [54], as well as clinically unapparent mutations in the HFE gene [55]. It has been estimated that up to 40% of hepatitis B virus (HBV)-related HCC occur in persons who do not have cirrhosis, while almost all cases of hep- atitis C virus (HCV)-related HCC occur in the setting of cirrhosis [56, 57]. Direct oncogenic potential of HBV through chromosomal integration (cis-activation) or trans-activation of cellular genes seems to be an important feature in the pathogene- sis of these cancers [58]. Nevertheless, the two major risk factors for developing an HCC without underlying liver cirrhosis seem to be the metabolic syndrome (MS) with fatty liver disease and liver adenomas. The association between diabetes, obesity, steatosis, non-alcoholic fatty liver disease (NAFLD) and the develop- ment of HCC is not well elucidated so far. Changes in fat metabolism, including expression of adipocyte-like gene pathways, appear to play a role both in hepatic regeneration and sometimes in neoplastic transformation [59, 60]. This relation- ship to fat metabolism appears to be important both in NAFLD-related cancer and in HCV, where steatosis, steatohepatitis and associated oxidative stress are increasingly recognized as significant risk or cofactors in HCC development [61]. Furthermore steatosis is an independent predictor of postoperative HCC recurrence in HCV-associated HCC [62]. Additional epidemiological data indicate a signifi- cantly increased risk of hepatocellular carcinoma among diabetic patients [63]. The pathophysiological components of this disorder, especially when steatohepatitis is present, include lipid peroxidation, stem cell proliferation, and increased growth factors, such as insulin and TGF. Proliferation of cells in the setting of oxidative stress and increased trophic factors associated with the metabolic syndrome such as hyperinsulinemia seems to be the hallmark of non-cirrhotic HCC in context with the metabolic syndrome [61]. Recently it has been shown that most of the tumours in context with the metabolic syndrome develop in nonfibrotic livers [64]. Wnt signalling pathway deregulation did not represent the main carcinogenic process involved in this context [64]. A significant percentage of HCCs that develop in the context of MS without significant fibrosis arises from malignant transformation of liver adenoma, especially the TA (telangiectatic) subtype. It has been proposed that liver adenomas can transform into malignant liver tumours [53, 65–67]. However, the incidence and underlying molecular mechanisms still remain unclear. Moreover, the published data usually derive from patients with- out liver cirrhosis. This is rather due to the conceptual problem that adenomas are only diagnosed in non-cirrhotic livers and are labelled as macroregenerative nod- ules or adenomatous hyperplasias in patients with underlying liver cirrhosis, than due to a fundamentally different pathogenesis of these tumours. However, the con- ceptual adenoma-carcinoma sequence, as observed in other organs like the gut, seems to exist in a similar manner in HCC as well. Among the three genotypi- cally identified subtypes of liver adenoma HNF1α inactivated (35–50% of cases), β-catenin activated (15–18% of cases) and inflammatory (40–55% of cases), the β-catenin-activated adenomas seem to be at higher risk of HCC [68]. 2 Biology of Hepatocellular Carcinoma 29 Metastasis HCC is characterized by early development of intrahepatic metastasis, whereas distant organs are usually late involved in the disease. In the diagnosis of intra- hepatic HCC metastases, one problem is to separate metastatic dissemination from multifocal tumours in the liver. The possibility that more than one nodule may be detected in patients with HCC has been known for more than 50 years [69]. This is not only semantic but of biological importance. ‘Metastatic dissemination or mul- tifocal tumour?’ is not a Hamletic question, because the two possibilities are not mutually exclusive as cirrhotic liver can generate more than one cancer nodule with the same, still unknown mechanisms, during the history of the disease. However, the identification of these two distinct hypotheses seems to be an underestimated problem by clinicians, although a patient with a multifocal tumour has a better prog- nosis than a patient with a metastatic cancer [70, 71]. Multiple HCC nodules are an expression of metastasis rather than of multifocal cancer in more than 60% of cases [72], but more sensitive tests are needed to distinguish metastatic from multifocal HCC in the liver. The process of metastasis involves an intricate interplay between altered cell adhesion, survival, proteolysis, migration, lymph/angiogenesis, immune escape mechanisms and homing on target organs. Not surprisingly, the molecular mech- anisms that propel invasive growth and metastasis are also found in embryonic development and, however, to a less perpetual/chronic/aggressive/quantitatively dif- ferent extent, in adult tissue maintenance (e.g. involving stem cell differentiation) and repair processes [73]. Several molecular examples and pathways are involved and all act in concert to guide the tumour cell to its new home (Table 2.1). All these players are tightly orchestrated and interact through several molecular pathways: The Wnt/β-catenin pathway links cell–cell adhesion and downstream signalling and mutations in β-catenin genes can be detected in 12–26% of human HCC; p53 mutation is involved in determining dedifferentiation, proliferating activity and tumour pro- gression [74]; is strongly related to the invasiveness of HCC and also influences the postoperative course (particularly recurrence within 1 year) [75]. The mitogen- activated protein kinase pathway (MAPK) and the Raf kinase inhibitor protein Table 2.1 Molecular examples and pathways are involved and all act in concert to guide the tumour cell to its new home Biological capability Molecular examples/pathway entities Survival IGF survival factors Adhesion and deadhesion CAMs, cadherins, integrins Migration Met-SF/HGF signalling, FAK Proteolysis/ECM remodelling MMPs, uPA, ADAMs, heparanase Immune escape Downregulation of intrinsic immunogenicity, MHC loss Lymph/angiogenesis VEGF, PDGF, bFGF Homing on target organs Chemokines/chemokine receptors, CD44, osteopontin 30 M.L. Balmer and J F. Dufour (RKIP) as an inhibitor of this pathway revealed prominent roles during human HCC metastasis [76]. Signalling pathways in liver cancer metastasis are highly com- plex and little is known about the importance of every single cascade in metastasis development. A bioinformatics analysis of metastasis-related proteins in hepatocel- lular carcinoma resulted in a gigantic diversity of involved partners in metastasis development (506 proteins, 83 pathways) [77]. From a more macroscopic point of view, there are four proposed models of metastasis development: According to Chambers and coworkers, only a very small population of injected tumour cells in mice form micrometastases, although most of them are arrested in the liver. Furthermore, not all of the micrometastases persist, and the progressively growing metastases arise only from a small subset (0.02%) of cells [78]. Muschel and coworkers recently proposed a new model for pul- monary metastasis in which endothelium-attached tumour cells that survived the initial apoptotic stimuli proliferate intravascularly. Thus, a principal tenet of this new model is that the extravasation of tumour cells is not a prerequisite for metastatic colony formation and that the initial proliferation t akes place within the blood vessels [79]. The unique ability of aggressive tumour cells to generate patterned networks, similar to the patterned networks during embryonic vasculogenesis, and concomitantly to express vascular markers associated with endothelial cells, their precursors and other vascular cells has been termed ‘vasculogenic mimicry’ by Hendrix and coworkers [80]. It has been shown that tumour cells can migrate as tumour emboli that conserve a tissue architecture reminiscent of the primary tumour. Tumour cells are thus protected from anoikis and direct immunological engagement during dissemination [81]. All of these mechanisms are supposed to have an impact on the pathogenesis of HCC metastasis and should be taken into account when planning new treatment strategies. HCC in comparison to other solid tumours is a biologically highly multifaceted tumour in which a variety of different events have an impact on its development and characteristics. The epic voyage from genotype to phenotype includes chromoso- mal instability, altered transcription and translation, deregulated protein synthesis, miRNAs, altered signalling pathways and finally results in a tumour cell perfectly adapted to escape proliferation control and immune surveillance. References 1. Pei Y, Zhang T, Renault V, Zhang X (2009) An overview of hepatocellular carcinoma study by omics-based methods. Acta Biochim Biophys Sin (Shanghai) 41:1–15 2. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100:57–70 3. Weinberg RA (1996) The retinoblastoma protein and cell cycle control. Cell 81:323–330 4. Harris CC (1996) p53 tumor suppressor gene: from the basic research laboratory to the clinic—an abridged historical perspective. Carcinogenesis 17:1187–1198 5. Bryan TM, Cech TR (1999) Telomerase and the maintenance of chromosome ends. Curr Opin Cell Biol 11:318–324 6. Bryan TM, Englezou A, Gupta J, Bacchetti S, Reddel RR (1995) Telomere elongation in immortal human cells without detectable telomerase activity. EMBO J 14:4240–4248 2 Biology of Hepatocellular Carcinoma 31 7. Lee JS, Heo J, Libbrecht L, Chu IS, Kaposi-Novak P, Calvisi DF, Mikaelyan A, et al (2006) A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progen- itor cells. Nat Med 12:410–416 8. Farazi PA, DePinho RA (2006) Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 6:674–687 9. Borzio M, Bruno S, Roncalli M, Mels GC, Ramella G, Borzio F, Leandro G, et al (1995) Liver cell dysplasia is a major risk factor for hepatocellular carcinoma in cirrhosis: a prospective study. Gastroenterology 108:812–817 10. Nakamoto Y, Kaneko S, Fan H, Momoi T, Tsutsui H, Nakanishi K, Kobayashi K, et al (2002) Prevention of hepatocellular carcinoma development associated with chronic hepatitis by anti- fas ligand antibody therapy. J Exp Med 196:1105–1111 11. Huang J, Sheng HH, Shen T, Hu YJ, Xiao HS, Zhang Q, Zhang QH, et al (2006) Correlation between genomic DNA copy number alterations and transcriptional expression in hepatitis B virus-associated hepatocellular carcinoma. FEBS Lett 580:3571–3581 12. Pineau P, Marchio A, Battiston C, Cordina E, Russo A, Terris B, Qin LX, et al (2008) Chromosome instability in human hepatocellular carcinoma depends on p53 status and aflatoxin exposure. Mutat Res 653:6–13 13. Saigo K, Yoshida K, Ikeda R, Sakamoto Y, Murakami Y, Urashima T, Asano T, et al (2008) Integration of hepatitis B virus DNA into the myeloid/lymphoid or mixed-lineage leukemia (MLL4) gene and rearrangements of MLL4 in human hepatocellular carcinoma. Hum Mutat 29:703–708 14. Gramantieri L, Fornari F, Callegari E, Sabbioni S, Lanza G, Croce CM, Bolondi L, et al (2008) MicroRNA involvement in hepatocellular carcinoma. J Cell Mol Med 12:2189–2204 15. Morrison SJ, Kimble J (2006) Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441:1068–1074 16. Roskams T, Yang SQ, Koteish A, Durnez A, DeVos R, Huang X, Achten R, et al (2003) Oxidative stress and oval cell accumulation in mice and humans with alcoholic and nonalco- holic fatty liver disease. Am J Pathol 163:1301–1311 17. Sell S (2001) Heterogeneity and plasticity of hepatocyte lineage cells. Hepatology 33:738–750 18. Alison MR, Islam S, Lim S (2009) Stem cells in liver regeneration, fibrosis and cancer: the good, the bad and the ugly. J Pathol 217:282–298 19. Mishra L, Banker T, Murray J, Byers S, Thenappan A, He AR, Shetty K, et al (2009) Liver stem cells and hepatocellular carcinoma. Hepatology 49:318–329 20. Pardal R, Clarke MF, Morrison SJ (2003) Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 3:895–902 21. Poon RT, Ng IO, Lau C, Zhu LX, Yu WC, Lo CM, Fan ST, et al (2001) Serum vascular endothelial growth factor predicts venous invasion in hepatocellular carcinoma: a prospective study. Ann Surg 233:227–235 22. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M, et al (1999) Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 85:221–228 23. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C , Ishida A, Fujita Y, et al (1998) Evidence for circulating bone marrow-derived endothelial cells. Blood 92:362–367 24. Ribatti D, Vacca A, Dammacco F (2003) New non-angiogenesis dependent pathways for tumour growth. Eur J Cancer 39:1835–1841 25. Kieser A, Weich HA, Brandner G, Marme D, Kolch W (1994) Mutant p53 potentiates protein kinase C induction of vascular endothelial growth factor expression. Oncogene 9:963–969 26. Rak J, Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T, Kerbel RS (1995) Mutant ras oncogenes upregulate VEGF/VPF expression: implications for induction and inhibition of tumor angiogenesis. Cancer Res 55:4575–4580 27. North S, Moenner M, Bikfalvi A (2005) Recent developments in the regulation of the angiogenic switch by cellular stress factors in tumors. Cancer Lett 218:1–14 32 M.L. Balmer and J F. Dufour 28. Dvorak HF, Nagy JA, Berse B, Brown LF, Yeo KT, Yeo TK, Dvorak AM, et al (1992) Vascular permeability factor, fibrin, and the pathogenesis of tumor stroma formation. Ann N Y Acad Sci 667:101–111 29. El-Assal ON, Yamanoi A, Soda Y, Yamaguchi M, Igarashi M, Yamamoto A, Nabika T, et al (1998) Clinical significance of microvessel density and vascular endothelial growth factor expression in hepatocellular carcinoma and surrounding liver: possible involvement of vascular endothelial growth factor in the angiogenesis of cirrhotic liver. Hepatology 27:1554–1562 30. Sugimachi K, Tanaka S, Taguchi K, Aishima S, Shimada M, Tsuneyoshi M (2003) Angiopoietin switching regulates angiogenesis and progression of human hepatocellular carcinoma. J Clin Pathol 56:854–860 31. Yasuda S, Arii S, Mori A, Isobe N, Yang W, Oe H, Fujimoto A, et al (2004) Hexokinase II and VEGF expression in liver tumors: correlation with hypoxia-inducible factor 1 alpha and its significance. J Hepatol 40:117–123 32. von Marschall Z, Cramer T, Hocker M, Finkenzeller G, Wiedenmann B, Rosewicz S (2001) Dual mechanism of vascular endothelial growth factor upregulation by hypoxia in human hepatocellular carcinoma. Gut 48:87–96 33. Lee SW, Lee YM, Bae SK, Murakami S, Yun Y, Kim KW (2000) Human hepatitis B virus X protein is a possible mediator of hypoxia-induced angiogenesis in hepatocarcinogenesis. Biochem Biophys Res Commun 268:456–461 34. Tsukamoto A, Kaneko Y, Yoshida T, Ichinose M, Kimura S (1999) Regulation of angiogenesis in human hepatomas: possible involvement of p53-inducible inhibitor of vascular endothelial cell proliferation. Cancer Lett 141:79–84 35. Pang R, Poon RT (2006) Angiogenesis and antiangiogenic therapy in hepatocellular carci- noma. Cancer Lett 242:151–167 36. Yoshiji H, Kuriyama S, Yoshii J, Ikenaka Y, Noguchi R, Hicklin DJ, Huber J, et al (2002) Synergistic effect of basic fibroblast growth factor and vascular endothelial growth factor in murine hepatocellular carcinoma. Hepatology 35:834–842 37. Kin M, Sata M, Ueno T, Torimura T, Inuzuka S, Tsuji R, Sujaku K, et al (1997) Basic fibroblast growth factor regulates proliferation and motility of human hepatoma cells by an autocrine mechanism. J Hepatol 27:677–687 38. Mitsuhashi N, Shimizu H, Ohtsuka M, Wakabayashi Y, Ito H, Kimura F, Yoshidome H, et al (2003) Angiopoietins and Tie-2 expression in angiogenesis and proliferation of human hepatocellular carcinoma. Hepatology 37:1105–1113 39. Morinaga S, Yamamoto Y, Noguchi Y, Imada T, Rino Y, Akaike M, Sugimasa Y, et al (2003) Platelet-derived endothelial cell growth factor (PD-ECGF) is up-regulated in human hepa- tocellular carcinoma (HCC) and the corresponding hepatitis liver. Hepatogastroenterology 50:1521–1526 40. Poon RT, Lau CP, Ho JW, Yu WC, Fan ST, Wong J (2003) Tissue factor expression correlates with tumor angiogenesis and invasiveness in human hepatocellular carcinoma. Clin Cancer Res 9:5339–5345 41. Tang TC, Poon RT, Lau CP, Xie D, Fan ST (2005) Tumor cyclooxygenase-2 levels corre- late with tumor invasiveness in human hepatocellular carcinoma. World J Gastroenterol 11: 1896–1902 42. Hisai H, Kato J, Kobune M, Murakami T, Miyanishi K, Takahashi M, Yoshizaki N, et al (2003) Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity. Clin Cancer Res 9:4852–4859 43. Erlitzki R, Minuk GY (1999) Telomeres, telomerase and HCC: the long and the short of it. J Hepatol 31:939–945 44. Plentz RR, Park YN, Lechel A, Kim H, Nellessen F, Langkopf BH, Wilkens L, et al (2007) Telomere shortening and inactivation of cell cycle checkpoints characterize human hepatocarcinogenesis. Hepatology 45:968–976 2 Biology of Hepatocellular Carcinoma 33 45. Begus-Nahrmann Y, Lechel A, Obenauf AC, Nalapareddy K, Peit E, Hoffmann E, Schlaudraff F, et al.(2009) p53 deletion impairs clearance of chromosomal-instable stem cells in aging telomere-dysfunctional mice. Nat Genet 41:1138–1143 46. Lechel A, Holstege H, Begus Y, Schienke A, Kamino K, Lehmann U, Kubicka S, et al (2007) Telomerase deletion limits progression of p53-mutant hepatocellular carcinoma with short telomeres in chronic liver disease. Gastroenterology 132:1465–1475 47. Nzeako UC, Goodman ZD, Ishak KG (1996) Hepatocellular carcinoma in cirrhotic and non- cirrhotic livers. A clinico-histopathologic study of 804 North American patients. Am J Clin Pathol 105:65–75 48. Okuda K, Nakashima T, Kojiro M, Kondo Y, Wada K (1989) Hepatocellular carcinoma without cirrhosis in Japanese patients. Gastroenterology 97:140–146 49. Regimbeau JM, Colombat M, Mognol P, Durand F, Abdalla E, Degott C, Degos F, et al (2004) Obesity and diabetes as a risk factor for hepatocellular carcinoma. Liver Transpl 10: S69–S73 50. Trevisani F, D’Intino PE, Caraceni P, Pizzo M, Stefanini GF, Mazziotti A, Grazi GL, et al (1995) Etiologic factors and clinical presentation of hepatocellular carcinoma. Differences between cirrhotic and noncirrhotic Italian patients. Cancer 75:2220–2232 51. Okuda K (1997) Liver cancer. Churchill Livingstone, London 52. Zhou H, Ortiz-Pallardo ME, Ko Y, Fischer HP (2000) Is heterozygous alpha-1-antitrypsin deficiency type PIZ a risk factor for primary liver carcinoma? Cancer 88:2668–2676 53. Bianchi L (1993) Glycogen storage disease I and hepatocellular tumours. Eur J Pediatr 152(Suppl 1):S63–S70 54. Kowdley KV (2004) Iron, hemochromatosis, and hepatocellular carcinoma. Gastroenterology 127:S79–S86 55. Turlin B, Juguet F, Moirand R, Le Quilleuc D, Loreal O, Campion JP, Launois B, et al (1995) Increased liver iron stores in patients with hepatocellular carcinoma developed on a noncirrhotic liver. Hepatology 22:446–450 56. Lok AS (2009) Hepatitis B: liver fibrosis and hepatocellular carcinoma. Gastroenterol Clin Biol 33:911–915 57. But DY, Lai CL, Yuen MF (2008) Natural history of hepatitis-related hepatocellular carci- noma. World J Gastroenterol 14:1652–1656 58. Park NH, Song IH, Chung YH (2006) Chronic hepatitis B in hepatocarcinogenesis. Postgrad Med J 82:507–515 59. Terasaki S, Kaneko S, Kobayashi K, Nonomura A, Nakanuma Y (1998) Histological features predicting malignant transformation of nonmalignant hepatocellular nodules: a prospective study. Gastroenterology 115:1216–1222 60. Watanabe S, Horie Y, Kataoka E, Sato W, Dohmen T, Ohshima S, Goto T, et al (2007) Non-alcoholic steatohepatitis and hepatocellular carcinoma: lessons from hepatocyte-specific phosphatase and tensin homolog (PTEN)-deficient mice. J Gastroenterol Hepatol 22(Suppl 1):S96–S100 61. Caldwell S, Park SH (2009) The epidemiology of hepatocellular cancer: from the perspectives of public health problem to tumor biology. J Gastroenterol 44(Suppl 19):96–101 62. Takuma Y, Nouso K, Makino Y, Saito S, Takayama H, Takahara M, Takahashi H, et al (2007) Hepatic steatosis correlates with the postoperative recurrence of hepatitis C virus-associated hepatocellular carcinoma. Liver Int 27:620–626 63. Lagiou P, Kuper H, Stuver SO, Tzonou A, Trichopoulos D, Adami HO (2000) Role of diabetes mellitus in the etiology of hepatocellular carcinoma. J Natl Cancer Inst 92:1096–1099 64. Paradis V, Zalinski S, Chelbi E, Guedj N, Degos F, Vilgrain V, Bedossa P, et al (2009) Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: a pathological analysis. Hepatology 49:851–859 65. Closset J, Veys I, Peny MO, Braude P, Van Gansbeke D, Lambilliotte JP, Gelin M (2000) Retrospective analysis of 29 patients surgically treated for hepatocellular adenoma or focal nodular hyperplasia. Hepatogastroenterology 47:1382–1384 34 M.L. Balmer and J F. Dufour 66. Ferrell LD (1993) Hepatocellular carcinoma arising in a focus of multilobular adenoma. A case report. Am J Surg Pathol 17:525–529 67. Foster JH, Berman MM (1994) The malignant transformation of liver cell adenomas. Arch Surg 129:712–717 68. Bioulac-Sage P, Laumonier H, Couchy G, Le Bail B, Sa Cunha A, Rullier A, Laurent C, et al (2009) Hepatocellular adenoma management and phenotypic classification: the Bordeaux experience. Hepatology 50:481–489 69. Plopper H, Schaffner F (1957) Primary hepatic carcinomas. McGraw-Hill, New York, pp. 593–612 70. Nakano S, Haratake J, Okamoto K, Takeda S (1994) Investigation of resected multinodular hepatocellular carcinoma: assessment of unicentric or multicentric genesis from histological and prognostic viewpoint. Am J Gastroenterol 89:189–193 71. Yasui M, Harada A, Nonami T, Takeuchi Y, Taniguchi K, Nakao A, Takagi H (1997) Potentially multicentric hepatocellular carcinoma: clinicopathologic characteristics and post- operative prognosis. World J Surg 21:860–865 72. Ng IO, Guan XY, Poon RT, Fan ST, Lee JM (2003) Determination of the molecular relation- ship between multiple tumour nodules in hepatocellular carcinoma differentiates multicentric origin from intrahepatic metastasis. J Pathol 199:345–353 73. Dvorak HF (1986) Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315:1650–1659 74. Itoh T, Shiro T, Seki T, Nakagawa T, Wakabayashi M, Inoue K, Okamura A (2000) Relationship between p53 overexpression and the proliferative activity in hepatocellular carcinoma. Int J Mol Med 6:137–142 75. Jeng KS, Sheen IS, Chen BF, Wu JY (2000) Is the p53 gene mutation of prognostic value in hepatocellular carcinoma after resection? Arch Surg 135:1329–1333 76. Guo K, Liu Y, Zhou H, Dai Z, Zhang J, Sun R, Chen J, et al (2008) Involvement of protein kinase C beta-extracellular signal-regulating kinase 1/2/p38 mitogen-activated protein kinase- heat shock protein 27 activation in hepatocellular carcinoma cell motility and invasion. Cancer Sci 99:486–496 77. Song PM, Zhang Y, He YF, Bao HM, Luo JH, Liu YK, Yang PY, et al (2008) Bioinformatics analysis of metastasis-related proteins in hepatocellular carcinoma. World J Gastroenterol 14:5816–5822 78. Chambers AF, Wilson SM, Kerkvliet N, O’Malley FP, Harris JF, Casson AG (1996) Osteopontin expression in lung cancer. Lung Cancer 15:311–323 79. Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L, Lee A, Muschel RJ (2000) Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis. Nat Med 6:100–102 80. Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, Trent JM, et al (1999) Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155:739–752 81. Sugino T, Yamaguchi T, Hoshi N, Kusakabe T, Ogura G, Goodison S, Suzuki T (2008) Sinusoidal tumor angiogenesis is a key component in hepatocellular carcinoma metastasis. Clin Exp Metastasis 25:835–841 . (2003) Oxidative stress and oval cell accumulation in mice and humans with alcoholic and nonalco- holic fatty liver disease. Am J Pathol 163:1301–1311 17. Sell S (2001) Heterogeneity and plasticity of. regeneration, fibrosis and cancer: the good, the bad and the ugly. J Pathol 217:282–298 19. Mishra L, Banker T, Murray J, Byers S, Thenappan A, He AR, Shetty K, et al (2009) Liver stem cells and hepatocellular. involvement of p53 -inducible inhibitor of vascular endothelial cell proliferation. Cancer Lett 141:79–84 35. Pang R, Poon RT (2006) Angiogenesis and antiangiogenic therapy in hepatocellular carci- noma.

Ngày đăng: 07/07/2014, 09:20

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan