Molecular alterations in the carcinogenesis and progression of hepatocellular carcinoma: Tumor factors and background liver factors (Review)

  • Authors:
    • Yoshikuni Inokawa
    • Kenichi Inaoka
    • Fuminori Sonohara
    • Masamichi Hayashi
    • Mitsuro Kanda
    • Shuji Nomoto
  • View Affiliations

  • Published online on: September 15, 2016     https://doi.org/10.3892/ol.2016.5141
  • Pages: 3662-3668
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although hepatocellular carcinoma (HCC) is associated with poor prognosis worldwide, the molecular mechanisms underlying the carcinogenesis and progression of this disease remain unclear. Several tumor characteristics have previously been demonstrated to be prognostic factors of survival following hepatic resection, or the recurrence of HCC or other types of cancer. Comparisons of normal tissues and HCC tumor tissues have revealed the presence of numerous molecular alterations in HCC, including genetic and epigenetic mechanisms, particularly mutations in certain genes and DNA methylation in the promoter regions of tumor‑suppressor genes. A number of studies have previously used array analysis to detect variations in the expression levels of cancer‑associated genes and microRNAs, and in DNA methylation. However, an investigation of HCC tumor tissues may not determine the effect of noncancerous liver tissues (background liver) in patients with HCC. As HCC may recur multicentrically following resection, a damaged or chronically diseased HCC background liver may be considered as a pre‑cancerous organ. Therefore, the influence of the background liver on HCC requires further study. Detailed studies regarding the background liver may be essential for the improved understanding of the carcinogenesis and progression of this malignancy; however only a few studies have investigated the microenvironment of the HCC background liver. The present review discusses prior molecular studies of hepatocarcinogenesis that focus on HCC and background liver tissues.

Introduction

Hepatocellular carcinoma (HCC) is the sixth most common neoplasm and the third most frequent cause of cancer-associated mortality worldwide (1). Despite advances in the diagnosis (24) and treatment (511) of this disease, its prognosis remains poor (12,13). Therefore, clarification of the mechanisms underlying its development is critically important (14). Numerous studies have investigated gene alterations in tumor tissues of HCC and their functions in hepatocarcinogenesis (1416). Various oncogenic and tumor-suppressor genes have been detected in HCC cell lines and resection specimens. However, HCC usually develops from an established background of chronic liver disease (13). Therefore, the elucidation of the mechanisms underlying HCC carcinogenesis requires the study of the background liver parenchyma as the potential foundation of this malignancy, in addition to the biology of the tumor. The present study reviews previous studies of molecular alterations identified in HCC, with a focus on the tumor factors and background factors.

Prognostic effect of tumor factors

HCC tumor specimens offer important information regarding certain prognostic factors, including vascular invasion (1721), tumor size (2125) and pathological grade (26,27), which are robust predictors of clinical outcomes following curative resection surgeries for HCC. α-fetoprotein levels are also a prognostic indicator (21,25). These factors may affect postoperative prognosis and, thus, HCC tumor tissues require further studies and molecular examinations.

Molecular alterations in HCC tumors

Molecular alterations in HCC tumor tissues have been widely studied (14,28). The Wnt signaling pathway is considered to affect the carcinogenesis of various malignancies, including HCC (29,30). A previous study indicated that the aberrant activation of Wnt/β-catenin signaling is involved in the initiation and progression of HCC (31). Mutation of the β-catenin gene at its exon 3, which is responsible for β-catenin phosphorylation and subsequent proteasome-dependent degradation, may lead to over-activation of the Wnt/β-catenin signaling pathway; this mutation has been reported in patients with HCC (3234). Another frequent mutation in HCC is in tumor protein 53 (p53) (3537). The tumor suppressive effects of p53 are mediated by a variety of mechanisms, including cell-cycle arrest, apoptosis and cellular senescence (38); p53 mutations have been identified in cell lines and tissue samples from patients with HCC (3941). Previous studies have observed that deregulation of the phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B signaling pathway by genetic alteration is another mechanism underlying HCC (4244), which promotes cellular growth and proliferation (42,45).

Epigenetic alterations have also been identified to promote hepatocarcinogenesis; changes in microRNAs (miRNAs) and the expression of their target genes may provide tools and opportunities to detect and treat HCC (4648). Numerous studies have identified specific miRNAs and their target genes, and their possible roles in hepatocarcinogenesis (4954). Previous studies observed that miRNA-101 (49), −122 (50) and −195 (51) levels are downregulated, whereas miRNA-21 (52), −221 (53) and −224 (54) levels are upregulated in HCC. The epigenetic inactivation of tumor-suppressor genes by promoter hypermethylation has been identified as an important mechanism underlying tumorigenesis (55). Hypermethylation has been detected in numerous HCC-associated genes, including cyclin dependent kinase inhibitor 2A (p16INK4a) (56,57), runt related transcription factor 3 (RUNX3) (58,59), suppressor of cytokine signaling 1 (SOCS1) (60,61), secreted frizzled related protein 1 (SFRP1) (62,63), O-6-methylguanine-DNA methyltransferase (MGMT) (6466), Ras association domain family member 1 (RASSF1A) (6769) and glutathione S-transferase pi 1 (GSTP1) (66,70) by investigations using various kinds of HCC cell lines and various stage of clinical HCC samples.

Expression array analysis is frequently used to detect novel cancer-associated genes (7175) by comparing gene expression levels between cancerous and non-cancerous tissues. Okabe et al (72) and Shirota et al (73) extracted mRNA from the cancerous and noncancerous tissues of a number of patients with HCC, in order to evaluate the variations in gene expression levels, and detected certain genes that characterize HCC. Nomoto et al (74) performed a double-combination array analysis consisting of an expression array and single nucleotide polymorphism (SNP) array, which facilitated the identification of novel genes associated with hepatocarcinogenesis using expression profiling and chromosomal copy number analysis (74,7680). Microarray analysis was also used to reveal cancer-associated miRNAs (52,81). Wong et al (81) detected the downregulation of miRNA-223 expression levels in HCC tissues and investigated stathmin 1 (STMN1) as a downstream target. Methylation array analysis has been used to detect cancer-specific epigenetic alterations in HCC (8291). Shen et al (82) analyzed tumor and adjacent non-tumor tissue samples from 62 Taiwanese patients with HCC, using Illumina® methylation arrays, and identified several genes that were hyper-or hypo-methylated in tumor tissues, as compared with adjacent non-tumor tissues (82). The candidate genes were pyrosequenced and the array data was validated; analysis of plasma DNA suggested those genes may be used as biomarkers for early-stage HCC. Okamura et al (83) created a triple-combination array, consisting of an SNP array, gene expression microarray and methylation array analysis (8389). The tumor-suppressor gene candidates were efficiently identified by detecting downregulation and methylation in tumor tissues, and without chromosomal deletion of the loci of the targeted genes. Revill et al (90) performed a genome-wide methylation array analysis of HCC samples and a microarray analysis of gene re-expression in HCC cell lines, and then combined the data to locate an epigenetically-silenced tumor-suppressor gene. These studies demonstrate that the majority of hepatocarcinogenesis studies focus on the changes in tumor tissues and the comparison of HCC cell lines or tumor tissues with adjacent normal liver tissues.

Prognostic impact of background liver factors

Typically, HCC develops within an established background of chronic liver disease (13,92,93). Oncogenic agents, including aflatoxin, are common (94) and the majority of patients with HCC already have background liver disorders, including infections with hepatitis B (HBV) or C (HBC) virus (9598), alcohol intake (99) or non-alcoholic fatty liver disease (100,101). Following hepatic resection, the principal HCC recurrence site is the residual liver (102). Senthilnathan et al (103) demonstrated that the majority of patients treated with loco-regional therapies for HCC succumbed to the disease prior to developing extra-hepatic metastasis, regardless of age; this is unique to HCC, as compared with other types of malignancies. Therefore, previous studies reported that background liver status was an effective prognostic factor following tumor resection in patients with HCC (102,104). Zhou et al (104) performed a multivariate analysis of significant survival predictors in 1,000 patients with HCC (tumor diameter <5 cm) and determined liver cirrhosis to be an independent risk factor.

Multinodular HCCs are classified as multicentric (MC) occurrence (MO) or intrahepatic metastasis (IM) (105). Wang et al (106) investigated 15 high-frequency loss-of-heterozygosity DNA microsatellites in 100 tumor nodules in 60 matched pairs of recurrent HCC tissue samples from 40 patients who underwent liver tumor re-resections. The data demonstrated that IM-type recurrences were more common and had a poorer prognosis, compared with MO-type recurrences. By contrast, Nomoto et al (107) reported clonal analyses of tumor cells that demonstrated recurrent HCC genotypes, mitochondrial gene mutations (107) or patterns of promoter hypermethylation in various tumor-suppressor genes (108), and identified MO to be a more common type of recurrence, compared with IM, in Japanese patients. HCC patients with an HBV infection, which is the principal underlying factor of this malignancy in Eastern Asia and sub-Saharan Africa, were compared with patients with an HCV infection (primarily endemic in North America, Europe and Japan) (13). The patients with HCV-based HCC exhibited poorer liver function and had shorter overall survival and disease-free survival times (109). Therefore, background liver status is crucial in predicting prognosis following HCC surgical treatment, particularly in patients who are likely to have MO-type recurrences. Detailed studies regarding the background liver may be essential for the improved understanding of the carcinogenesis and progression of this malignancy.

Studies of molecular changes in HCC background liver

A number of previous studies have reported the detection of molecular changes in the HCC background liver (110116). Okamoto et al (112) compared gene-expression patterns in noncancerous liver tissue specimens from HCV-positive patients with HCC, between those patients with single-node HCC and those with MC HCC. The study selected 36 genes commonly associated with MC HCC and MC recurrence, and created a scoring system to determine the risk for MC hepatocarcinogenesis; the prediction score was higher in multicentric recurrence group than in that without (112). Utsunomiya et al (113) performed an miRNA microarray to examine the variations in miRNA expression patterns in non-cancerous liver tissue samples between the MC recurrence group and non-MC recurrence group, in order to identify miRNAs associated with MC recurrence. The study detected 20 differentially expressed miRNAs, of which 18 were downregulated and 2 were upregulated in the MC group. The same research group reviewed the concept of field cancerization, in which tumorigenesis may be viewed as cancer initiated by numerous cumulative epigenetic and genetic changes that transform a cell, or a group of cells, in a particular organ (110113). Hoshida et al (114) revealed that the gene expression profiles in non-tumor liver tissue, but not in primary HCC tissues, were highly associated with late recurrence (114). Nomoto et al (115) detected novel candidates for cancer-associated genes from array analyses, which compared supernormal liver tissue samples from resected secondary metastatic liver malignancies without HCC and corresponding normal tissue samples of HCC with chronic HCV (115,116).

Certain studies have investigated the epigenetic status of the HCC background liver and reported that molecular alterations, including DNA methylation, may change gradually during the transition from normal liver tissues to non-tumor liver tissue, and then to malignant HCC tissues (110,117119). Ammerpohl et al (117) performed a methylation array analysis using 17 HCC, 17 cirrhosis and 12 normal control tissues; they detected certain genes with significantly differences in hyper-or hypo-methylation between normal liver controls and cirrhosis as well as between cirrhosis and HCC (117). Um et al (118) evaluated the methylation status in CpG islands of the promoter regions of nine genes in normal liver tissues, cirrhosis, low-grade dysplastic nodules, high-grade dysplastic nodules, early-stage HCC tissues and late-stage HCC tissues (118). The study identified an increased methylation frequency along the sequence of multi-step hepatocarcinogenesis in some of these genes. Arai et al (119) investigated genome-wide DNA methylation profiles in normal liver tissue samples obtained from patients without HCC and cancerous and noncancerous liver tissue samples from patients with HCC; DNA methylation status was revealed to be correlated with the cancer-free and overall survival rates of patients with HCC (119).

Previous studies have also investigated the background tissues of numerous other malignancies (120122). Yoshida et al (120) examined the methylation status of CpG islands in the promoter regions of six genes and two repetitive DNA elements in the gastric mucosae, and revealed that the methylation levels were consistently increased in a stepwise manner with activity of gastric inflammation, among patients who were H. pylori–negative, or had low-or high-titers (120), and concluded that evaluating DNA methylation levels in the gastric mucosae may predict the risk of gastric cancer. Lee et al (121) evaluated promoter region methylation of the cadherin 1 (CDH1) gene in the non-neoplastic mucosae of diffuse gastric cancer and revealed there was a significantly higher methylation ratio, compared with normal non-cancerous gastric mucosae. Kadara et al (122) compared the gene expression profiles in non-small cell lung cancer airway tissues at various distances from the tumor tissues and normal lung tissues (122); significantly higher levels of lysosomal protein transmembrane 4β (LAPTM4B) mRNA transcripts were identified in the airway tissues near tumors. These attempts to clarify the molecular changes in non-cancerous tissues in several kinds of cancers are similar in concept to the evaluation of molecules in background liver of HCC.

Although investigations of non-cancerous tissues adjacent to HCC are required to clarify the mechanisms underlying hepatocarcinogenesis, few studies have previously evaluated the microenvironment of the HCC background liver. The concept of molecular changes in hepatocarcinogenesis is presented in Fig. 1. Although molecular alterations in the background liver tissues and in HCC tissues require further elucidation, the importance of background liver is particularly characteristic of HCC.

Future aspects

Identification of genes associated with hepatocarcinogenesis and their roles in transforming the background liver of HCC may facilitate the early detection of HCC in patients with liver damage, and liver biopsy specimens may provide information regarding early-stage HCC detection for certain patients. Furthermore, analysis of the adjacent liver parenchymal tissues in resected tumor samples from patients with HCC may assist the prediction of recurrence in the remaining liver tissues. Sorafenib is a molecular-targeted agent for HCC, which has been approved as a standard treatment for patients with advanced HCC that may not be treated with surgical resection (123); however, its therapeutic value has yet to be clinically established in patients with HCC who have undergone tumor resection surgery (124). The efficacies of adjuvant chemotherapies are currently limited, although certain postoperative antiviral treatments, including nucleotide analogs administered following surgical resection for HBV-associated HCC, or interferon for HCV-associated HCC, have been previously reported to indirectly reduce the recurrence of HCC following surgery in some studies (125). The elucidation of the mechanisms underlying hepatocarcinogenesis in the background liver may facilitate the development of novel adjuvant chemotherapies, which may be administered to patients following the resection of primary lesions.

In summary, although the mechanisms underlying hepatocarcinogenesis require further study, the status of the background liver tissues in HCC may have a crucial role in the development and progression of this type of malignancy.

Glossary

Abbreviations

Abbreviations:

HCC

hepatocellular carcinoma

miRNA

microRNA

MO

multicentric occurrence

IM

intrahepatic metastasis

HBV

hepatitis B virus

HCV

hepatitis C virus

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Forner A, Vilana R, Ayuso C, Bianchi L, Solé M, Ayuso JR, Boix L, Sala M, Varela M, Llovet JM, et al: Diagnosis of hepatic nodules 20 mm or smaller in cirrhosis: Prospective validation of the noninvasive diagnostic criteria for hepatocellular carcinoma. Hepatology. 47:97–104. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Bolondi L, Sofia S, Siringo S, Gaiani S, Casali A, Zironi G, Piscaglia F, Gramantieri L, Zanetti M and Sherman M: Surveillance programme of cirrhotic patients for early diagnosis and treatment of hepatocellular carcinoma: A cost effectiveness analysis. Gut. 48:251–259. 2001. View Article : Google Scholar : PubMed/NCBI

4 

van Meer S, de Man RA, Siersema PD and van Erpecum KJ: Surveillance for hepatocellular carcinoma in chronic liver disease: Evidence and controversies. World J Gastroenterol. 19:6744–6756. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Llovet JM, Burroughs A and Bruix J: Hepatocellular carcinoma. Lancet. 362:1907–1917. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Tateishi R, Shiina S, Teratani T, Obi S, Sato S, Koike Y, Fujishima T, Yoshida H, Kawabe T and Omata M: Percutaneous radiofrequency ablation for hepatocellular carcinoma-An analysis of 1000 cases. Cancer. 103:1201–1209. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Llovet JM and Bruix J: Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology. 37:429–442. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Abou-Alfa GK, Schwartz L, Ricci S, Amadori D, Santoro A, Figer A, De Greve J, Douillard JY, Lathia C, Schwartz B, et al: Phase II study of sorafenib in patients with advanced hepatocellular carcinoma. J Clin Oncol. 24:4293–4300. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Takayasu K, Arii S, Ikai I, Omata M, Okita K, Ichida T, Matsuyama Y, Nakanuma Y, Kojiro M, Makuuchi M, et al: Prospective cohort study of transarterial chemoembolization for unresectable hepatocellular carcinoma in 8510 patients. Gastroenterology. 131:461–469. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Llovet JM and Bruix J: Novel advancements in the management of hepatocellular carcinoma in 2008. J Hepatol. 48(Suppl 1): S20–S37. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Morise Z, Kawabe N, Tomishige H, Nagata H, Kawase J, Arakawa S, Yoshida R and Isetani M: Recent advances in the surgical treatment of hepatocellular carcinoma. World J Gastroenterol. 20:14381–14392. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Chen X, Liu HP, Li M and Qiao L: Advances in non-surgical management of primary liver cancer. World J Gastroenterol. 20:16630–16638. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI

14 

El-Serag HB and Rudolph L: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PW, Lam CT, Poon RT and Fan ST: Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell. 13:153–166. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S and Zucman-Rossi J: MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology. 47:1955–1963. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Eguchi S, Takatsuki M, Hidaka M, Soyama A, Tomonaga T, Muraoka I and Kanematsu T: Predictor for histological microvascular invasion of hepatocellular carcinoma: A lesson from 229 consecutive cases of curative liver resection. World J Surg. 34:1034–1038. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Roayaie S, Jibara G, Taouli B and Schwartz M: Resection of hepatocellular carcinoma with macroscopic vascular invasion. Ann Surg Oncol. 20:3754–3760. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Faber W, Stockmann M, Kruschke JE, Denecke T, Bahra M and Seehofer D: Implication of microscopic and macroscopic vascular invasion for liver resection in patients with hepatocellular carcinoma. Dig Surg. 31:204–209. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Sumie S, Nakashima O, Okuda K, Kuromatsu R, Kawaguchi A, Nakano M, Satani M, Yamada S, Okamura S, Hori M, et al: The significance of classifying microvascular invasion in patients with hepatocellular carcinoma. Ann Surg Oncol. 21:1002–1009. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Tandon P and Garcia-Tsao G: Prognostic indicators in hepatocellular carcinoma: A systematic review of 72 studies. Liver Int. 29:502–510. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Poon RT and Fan ST: Hepatectomy for hepatocellular carcinoma: Patient selection and postoperative outcome. Liver Transpl. 10(2): Suppl 1. S39–S45. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Pandey D, Lee KH, Wai CT, Wagholikar G and Tan KC: Long term outcome and prognostic factors for large hepatocellular carcinoma (10 cm or more) after surgical resection. Ann Surg Oncol. 14:2817–2823. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Ishii T, Hatano E, Yasuchika K, Taura K, Seo S and Uemoto S: High risk of lung metastasis after resection of hepatocellular carcinoma more than 7 cm in diameter. Surg Today. 44:1900–1905. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Han JH, Kim DG, Na GH, Kim EY, Lee SH, Hong TH and You YK: Evaluation of prognostic factors on recurrence after curative resections for hepatocellular carcinoma. World J Gastroenterol. 20:17132–17140. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Jonas S, Bechstein WO, Steinmüller T, Herrmann M, Radke C, Berg T, Settmacher U and Neuhaus P: Vascular invasion and histopathologic grading determine outcome after liver transplantation for hepatocellular carcinoma in cirrhosis. Hepatology. 33:1080–1086. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Han DH, Choi GH, Kim KS, Choi JS, Park YN, Kim SU, Park JY, Ahn SH and Han KH: Prognostic significance of the worst grade in hepatocellular carcinoma with heterogeneous histologic grades of differentiation. J Gastroenterol Hepatol. 28:1384–1390. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Scaggiante B, Kazemi M, Pozzato G, Dapas B, Farra R, Grassi M, Zanconati F and Grassi G: Novel hepatocellular carcinoma molecules with prognostic and therapeutic potentials. World J Gastroenterol. 20:1268–1288. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Giles RH, van Es JH and Clevers H: Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 1653:1–24. 2003.PubMed/NCBI

30 

Tsao CM, Yan MD, Shih YL, Yu PN, Kuo CC, Lin WC, Li HJ and Lin YW: SOX1 functions as a tumor suppressor by antagonizing the WNT/β-catenin signaling pathway in hepatocellular carcinoma. Hepatology. 56:2277–2287. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Kim M, Lee HC, Tsedensodnom O, Hartley R, Lim YS, Yu E, Merle P and Wands JR: Functional interaction between Wnt3 and Frizzled-7 leads to activation of the Wnt/beta-catenin signaling pathway in hepatocellular carcinoma cells. J Hepatol. 48:780–791. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Wong CM, Fan ST and Ng IO: Beta-Catenin mutation and overexpression in hepatocellular carcinoma: Clinicopathologic and prognostic significance. Cancer. 92:136–145. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, Aoki M, Hosono N, Kubo M, Miya F, et al: Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 44:760–764. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F, et al: Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 44:694–698. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Guan YS, La Z, Yang L, He Q and Li P: p53 gene in treatment of hepatic carcinoma: Status quo. World J Gastroenterol. 13:985–992. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Staib F, Hussain SP, Hofseth LJ, Wang XW and Harris CC: TP53 and liver carcinogenesis. Hum Mutat. 21:201–216. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Hussain SP, Schwank J, Staib F, Wang XW and Harris CC: TP53 mutations and hepatocellular carcinoma: Insights into the etiology and pathogenesis of liver cancer. Oncogene. 26:2166–2176. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Vogelstein B, Lane D and Levine AJ: Surfing the p53 network. Nature. 408:307–310. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Bressac B, Kew M, Wands J and Ozturk M: Selective G to T Mutation Of p53 gene in hepatocellular carcinoma from southern Africa. Nature. 350:429–431. 1991. View Article : Google Scholar : PubMed/NCBI

40 

Hsu IC, Metcalf RA, Sun T, Welsh JA, Wang NJ and Harris CC: Mutational hotspot in the p53 gene in human hepatocellular carcinomas. Nature. 350:427–428. 1991. View Article : Google Scholar : PubMed/NCBI

41 

Oda T, Tsuda H, Scarpa A, Sakamoto M and Hirohashi S: p53 gene mutation spectrum in hepatocellular carcinoma. Cancer Res. 52:6358–6364. 1992.PubMed/NCBI

42 

Zhou JD, Shen F, Ji JS, Zheng K, Huang M and Wu JC: FAM9C plays an anti-apoptotic role through activation of the PI3K/Akt pathway in human hepatocellular carcinoma. Oncol Rep. 30:1275–1284. 2013.PubMed/NCBI

43 

Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X, Zhao Y, Young KH, Hu W and Feng Z: Glutaminase 2 negatively regulates the PI3K/AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget. 5:2635–2647. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Pellegrino R, Calvisi DF, Neumann O, Kolluru V, Wesely J, Chen X, Wang C, Wuestefeld T, Ladu S, Elgohary N, et al: EEF1A2 inactivates p53 by way of PI3K/AKT/mTOR-dependent stabilization of MDM4 in hepatocellular carcinoma. Hepatology. 59:1886–1899. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Zhou Q, Wong CH, Lau CP, Hui CW, Lui VW, Chan SL and Yeo W: enhanced antitumor activity with combining effect of mTOR inhibition and microtubule stabilization in hepatocellular carcinoma. Int J Hepatol. 2013:1038302013. View Article : Google Scholar : PubMed/NCBI

46 

Khare S, Zhang Q and Ibdah JA: Epigenetics of hepatocellular carcinoma: Role of microRNA. World J Gastroenterol. 19:5439–5445. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Law PT and Wong N: Emerging roles of microRNA in the intracellular signaling networks of hepatocellular carcinoma. J Gastroenterol Hepatol. 26:437–449. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Zhu Z, Zhang X, Wang G and Zheng H: Role of MicroRNAs in Hepatocellular Carcinoma. Hepat Mon. 14:e186722014. View Article : Google Scholar : PubMed/NCBI

49 

Xu L, Beckebaum S, Iacob S, Wu G, Kaiser GM, Radtke A, Liu C, Kabar I, Schmidt HH, Zhang X, et al: MicroRNA-101 inhibits human hepatocellular carcinoma progression through EZH2 downregulation and increased cytostatic drug sensitivity. J Hepatol. 60:590–598. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Tsai WC, Hsu PW, Lai TC, Chau GY, Lin CW, Chen CM, Lin CD, Liao YL, Wang JL, Chau YP, et al: MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology. 49:1571–1582. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP and Zhuang SM: MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology. 50:113–121. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Gramantieri L, Fornari F, Ferracin M, Veronese A, Sabbioni S, Calin GA, Grazi GL, Croce CM, Bolondi L and Negrini M: MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality. Clin Cancer Res. 15:5073–5081. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H and Chayama K: Involvement of microRNA-224 in cell proliferation, migration, invasion and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol Hepatol. 28:565–575. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Tischoff I and Tannapfe A: DNA methylation in hepatocellular carcinoma. World J Gastroenterol. 14:1741–1748. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Liew CT, Li HM, Lo KW, Leow CK, Chan JY, Hin LY, Lau WY, Lai PB, Lim BK, Huang J, et al: High frequency of p16INK4A gene alterations in hepatocellular carcinoma. Oncogene. 18:789–795. 1999. View Article : Google Scholar : PubMed/NCBI

57 

Qin Y, Liu JY, Li B, Sun ZL and Sun ZF: Association of low p16INK4a and p15INK4b mRNAs expression with their CpG islands methylation with human hepatocellular carcinogenesis. World J Gastroenterol. 10:1276–1280. 2004.PubMed/NCBI

58 

Mori T, Nomoto S, Koshikawa K, Fujii T, Sakai M, Nishikawa Y, Inoue S, Takeda S, Kaneko T and Nakao A: Decreased expression and frequent allelic inactivation of the RUNX3 gene at 1p36 in human hepatocellular carcinoma. Liver Int. 25:380–388. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Park WS, Cho YG, Kim CJ, Song JH, Lee YS, Kim SY, Nam SW, Lee SH, Yoo NJ and Lee JY: Hypermethylation of the RUNX3 gene in hepatocellular carcinoma. Exp Mol Med. 37:276–281. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Yoshikawa H, Matsubara K, Qian GS, Jackson P, Groopman JD, Manning JE, Harris CC and Herman JG: SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity. Nat Genet. 28:29–35. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Okochi O, Hibi K, Sakai M, Inoue S, Takeda S, Kaneko T and Nakao A: Methylation-mediated silencing of SOCS-1 gene in hepatocellular carcinoma derived from cirrhosis. Clin Cancer Res. 9:5295–5298. 2003.PubMed/NCBI

62 

Shih YL, Shyu RY, Hsieh CB, Lai HC, Liu KY, Chu TY and Lin YW: Promoter methylation of the secreted frizzled-related protein 1 gene SFRP1 is frequent in hepatocellular carcinoma. Cancer. 107:579–590. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Huang J, Zhang YL, Teng XM, Lin Y, Zheng DL, Yang PY and Han ZG: Down-regulation of SFRP1 as a putative tumor suppressor gene can contribute to human hepatocellular carcinoma. BMC cancer. 7:1262007. View Article : Google Scholar : PubMed/NCBI

64 

Zhang YJ, Chen Y, Ahsan H, Lunn RM, Lee PH, Chen CJ and Santella RM: Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation and its relationship to aflatoxin B1-DNA adducts and p53 mutation in hepatocellular carcinoma. Int J Cancer. 103:440–444. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Matsukura S, Soejima H, Nakagawachi T, Yakushiji H, Ogawa A, Fukuhara M, Miyazaki K, Nakabeppu Y, Sekiguchi M and Mukai T: CpG methylation of MGMT and hMLH1 promoter in hepatocellular carcinoma associated with hepatitis viral infection. Br J Cancer. 88:521–529. 2003. View Article : Google Scholar : PubMed/NCBI

66 

Li Z, Zhang H, Yang J, Hao T and Li S: Promoter hypermethylation of DNA damage response genes in hepatocellular carcinoma. Cell Biol Int. 36:427–432. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Schagdarsurengin U, Wilkens L, Steinemann D, Flemming P, Kreipe HH, Pfeifer GP, Schlegelberger B and Dammann R: Frequent epigenetic inactivation of the RASSF1A gene in hepatocellular carcinoma. Oncogene. 22:1866–1871. 2003. View Article : Google Scholar : PubMed/NCBI

68 

Hu L, Chen G, Yu H and Qiu X: Clinicopathological significance of RASSF1A reduced expression and hypermethylation in hepatocellular carcinoma. Hepatol Int. 4:423–432. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Xu B, Di J, Wang Z, Han X, Li Z, Luo X and Zheng Q: Quantitative analysis of RASSF1A promoter methylation in hepatocellular carcinoma and its prognostic implications. Biochem Biophys Res Commun. 438:324–328. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Zhong S, Tang MW, Yeo W, Liu C, Lo YM and Johnson PJ: Silencing of GSTP1 gene by CpG island DNA hypermethylation in HBV-associated hepatocellular carcinomas. Clin Cancer Res. 8:1087–1092. 2002.PubMed/NCBI

71 

Midorikawa Y, Makuuchi M, Tang W and Aburatani H: Microarray-based analysis for hepatocellular carcinoma: From gene expression profiling to new challenges. World J Gastroenterol. 13:1487–1492. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Okabe H, Satoh S, Kato T, Kitahara O, Yanagawa R, Yamaoka Y, Tsunoda T, Furukawa Y and Nakamura Y: Genome-wide analysis of gene expression in human hepatocellular carcinomas using cDNA microarray: Identification of genes involved in viral carcinogenesis and tumor progression. Cancer Res. 61:2129–2137. 2001.PubMed/NCBI

73 

Shirota Y, Kaneko S, Honda M, Kawai HF and Kobayashi K: Identification of differentially expressed genes in hepatocellular carcinoma with cDNA microarrays. Hepatology. 33:832–840. 2001. View Article : Google Scholar : PubMed/NCBI

74 

Nomoto S, Kanda M, Okamura Y, Nishikawa Y, Qiyong L, Fujii T, Sugimoto H, Takeda S and Nakao A: Epidermal growth factor-containing fibulin-like extracellular matrix protein 1, EFEMP1, a novel tumor-suppressor gene detected in hepatocellular carcinoma using double combination array analysis. Ann Surg Oncol. 17:923–932. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Zhang LH and Ji JF: Molecular profiling of hepatocellular carcinomas by cDNA microarray. World J Gastroenterol. 11:463–468. 2005. View Article : Google Scholar : PubMed/NCBI

76 

Kanda M, Nomoto S, Okamura Y, Nishikawa Y, Sugimoto H, Kanazumi N, Takeda S and Nakao A: Detection of metallothionein 1G as a methylated tumor suppressor gene in human hepatocellular carcinoma using a novel method of double combination array analysis. Int J Oncol. 35:477–483. 2009.PubMed/NCBI

77 

Okamura Y, Nomoto S, Kanda M, Li Q, Nishikawa Y, Sugimoto H, Kanazumi N, Takeda S and Nakao A: Leukemia inhibitory factor receptor (LIFR) is detected as a novel suppressor gene of hepatocellular carcinoma using double-combination array. Cancer Lett. 289:170–177. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Kanda M, Nomoto S, Okamura Y, Hayashi M, Hishida M, Fujii T, Nishikawa Y, Sugimoto H, Takeda S and Nakao A: Promoter hypermethylation of fibulin 1 gene is associated with tumor progression in hepatocellular carcinoma. Mol Carcinog. 50:571–579. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Okamura Y, Nomoto S, Kanda M, Hayashi M, Nishikawa Y, Fujii T, Sugimoto H, Takeda S and Nakao A: Reduced expression of reelin (RELN) gene is associated with high recurrence rate of hepatocellular carcinoma. Ann Surg Oncol. 18:572–579. 2011. View Article : Google Scholar : PubMed/NCBI

80 

Hayashi M, Nomoto S, Kanda M, Okamura Y, Nishikawa Y, Yamada S, Fujii T, Sugimoto H, Takeda S and Kodera Y: Identification of the A kinase anchor protein 12 (AKAP12) gene as a candidate tumor suppressor of hepatocellular carcinoma. J Surg Oncol. 105:381–386. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Wong QW, Lung RW, Law PT, Lai PB, Chan KY, To KF and Wong N: MicroRNA-223 is commonly repressed in hepatocellular carcinoma and potentiates expression of Stathmin1. Gastroenterology. 135:257–269. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Shen J, Wang S, Zhang YJ, Kappil M, Wu HC, Kibriya MG, Wang Q, Jasmine F, Ahsan H, Lee PH, et al: Genome-wide DNA methylation profiles in hepatocellular carcinoma. Hepatology. 55:1799–1808. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Okamura Y, Nomoto S, Hayashi M, Hishida M, Nishikawa Y, Yamada S, Fujii T, Sugimoto H, Takeda S, Kodera Y and Nakao A: Identification of the bleomycin hydrolase gene as a methylated tumor suppressor gene in hepatocellular carcinoma using a novel triple-combination array method. Cancer Lett. 312:150–157. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Hishida M, Nomoto S, Inokawa Y, Hayashi M, Kanda M, Okamura Y, Nishikawa Y, Tanaka C, Kobayashi D, Yamada S, et al: Estrogen receptor 1 gene as a tumor suppressor gene in hepatocellular carcinoma detected by triple-combination array analysis. Int J Oncol. 43:88–94. 2013.PubMed/NCBI

85 

Inokawa Y, Nomoto S, Hishida M, Hayashi M, Kanda M, Nishikawa Y, Takeda S, Sugimoto H, Fujii T, Yamada S and Kodera Y: Detection of doublecortin domain-containing 2 (DCDC2), a new candidate tumor suppressor gene of hepatocellular carcinoma, by triple combination array analysis. J Exp Clin Cancer Res. 32:652013. View Article : Google Scholar : PubMed/NCBI

86 

Inokawa Y, Nomoto S, Hishida M, Hayashi M, Kanda M, Nishikawa Y, Takeda S, Fujiwara M, Koike M, Sugimoto H, et al: Dynamin 3: A new candidate tumor suppressor gene in hepatocellular carcinoma detected by triple combination array analysis. Onco Targets Ther. 6:1417–1424. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Hayashi M, Nomoto S, Hishida M, Inokawa Y, Kanda M, Okamura Y, Nishikawa Y, Tanaka C, Kobayashi D, Yamada S, et al: Identification of the collagen type 1 α 1 gene (COL1A1) as a candidate survival-related factor associated with hepatocellular carcinoma. BMC Cancer. 14:1082014. View Article : Google Scholar : PubMed/NCBI

88 

Takano N, Hishida M, Inokawa Y, Hayashi M, Kanda M, Nishikawa Y, Iwata N, Kobayashi D, Tanaka C, Yamada S, et al: CCNJ detected by triple combination array analysis as a tumor-related gene of hepatocellular carcinoma. Int J Oncol. 46:1963–70. 2015.PubMed/NCBI

89 

Hishida M, Inokawa Y, Takano N, Nishikawa Y, Iwata N, Kanda M, Tanaka C, Kobayashi D, Yamada S, Nakayama G, et al: Protein tyrosine kinase 7: A hepatocellular carcinoma-related gene detected by triple-combination array. J Surg Res. 195:444–453. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Revill K, Wang T, Lachenmayer A, Kojima K, Harrington A, Li J, Hoshida Y, Llovet JM and Powers S: Genome-wide methylation analysis and epigenetic unmasking identify tumor suppressor genes in hepatocellular carcinoma. Gastroenterology. 145:1424–1435. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Nishida N, Nishimura T, Nakai T, Chishina H, Arizumi T, Takita M, Kitai S, Yada N, Hagiwara S, Inoue T, et al: Genome-wide profiling of DNA methylation and tumor progression in human hepatocellular carcinoma. Dig Dis. 32:658–663. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Sherman M: Hepatocellular carcinoma: Epidemiology, surveillance, and diagnosis. Semin Liver Dis. 30:3–16. 2010. View Article : Google Scholar : PubMed/NCBI

93 

Fattovich G, Stroffolini T, Zagni I and Donato F: Hepatocellular carcinoma in cirrhosis: Incidence and risk factors. Gastroenterology. 127:5:Suppl 1. S35–S50. 2004. View Article : Google Scholar : PubMed/NCBI

94 

Ladep NG, Lesi OA, Mark P, Lemoine M, Onyekwere C, Afihene M, Crossey MM and Taylor-Robinson SD: Problem of hepatocellular carcinoma in West Africa. World J Hepatol. 6:783–792. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Perz JF, Armstrong GL, Farrington LA, Hutin YJ and Bell BP: The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol. 45:529–538. 2006. View Article : Google Scholar : PubMed/NCBI

96 

Kao JH and Chen DS: Global control of hepatitis B virus infection. Lancet Infect Dis. 2:395–403. 2002. View Article : Google Scholar : PubMed/NCBI

97 

Lavanchy D: The global burden of hepatitis C. Liver Int. 29(Suppl 1): S74–S81. 2009. View Article : Google Scholar

98 

El-Serag HB: Hepatocellular carcinoma and hepatitis C in the United States. Hepatology. 36(5): Suppl 1. S74–S83. 2002. View Article : Google Scholar : PubMed/NCBI

99 

Donato F, Tagger A, Gelatti U, Parrinello G, Boffetta P, Albertini A, Decarli A, Trevisi P, Ribero ML, Martelli C, et al: Alcohol and hepatocellular carcinoma: The effect of lifetime intake and hepatitis virus infections in men and women. Am J Epidemiol. 155:323–331. 2002. View Article : Google Scholar : PubMed/NCBI

100 

Baffy G, Brunt EM and Caldwell SH: Hepatocellular carcinoma in non-alcoholic fatty liver disease: An emerging menace. J Hepatol. 56:1384–1391. 2012. View Article : Google Scholar : PubMed/NCBI

101 

White DL, Kanwal F and El-Serag HB: Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin Gastroenterol Hepatol. 10:1342–1359. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Poon RT, Fan ST, Lo CM, Liu CL and Wong J: Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: Implications for a strategy of salvage transplantation. Ann Surg. 235:373–382. 2002. View Article : Google Scholar : PubMed/NCBI

103 

Senthilnathan S, Memon K, Lewandowski RJ, et al: Extrahepatic metastases occur in a minority of hepatocellular carcinoma patients treated with locoregional therapies: Analyzing patterns of progression in 285 patients. Hepatology. 55:1432–1442. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Zhou XD, Tang ZY, Yang BH, Lin ZY, Ma ZC, Ye SL, Wu ZQ, Fan J, Qin LX and Zheng BH: Experience of 1000 patients who underwent hepatectomy for small hepatocellular carcinoma. Cancer. 91:1479–1486. 2001. View Article : Google Scholar : PubMed/NCBI

105 

Shimada M, Hamatsu T, Yamashita Y, Rikimaru T, Taguchi K, Utsunomiya T, Shirabe K and Sugimachi K: Characteristics of multicentric hepatocellular carcinomas: Comparison with intrahepatic metastasis. World J Surg. 25:991–995. 2001. View Article : Google Scholar : PubMed/NCBI

106 

Wang B, Xia CY, Lau WY, Lu XY, Dong H, Yu WL, Jin GZ, Cong WM and Wu MC: Determination of clonal origin of recurrent hepatocellular carcinoma for personalized therapy and outcomes evaluation: A new strategy for hepatic surgery. J Am Coll Surg. 217:1054–1062. 2013. View Article : Google Scholar : PubMed/NCBI

107 

Nomoto S, Yamashita K, Koshikawa K, Nakao A and Sidransky D: Mitochondrial D-loop mutations as clonal markers in multicentric hepatocellular carcinoma and plasma. Clin Cancer Res. 8:481–487. 2002.PubMed/NCBI

108 

Nomoto S, Kinoshita T, Kato K, Otani S, Kasuya H, Takeda S, Kanazumi N, Sugimoto H and Nakao A: Hypermethylation of multiple genes as clonal markers in multicentric hepatocellular carcinoma. Br J Cancer. 97:1260–1265. 2007. View Article : Google Scholar : PubMed/NCBI

109 

Nomoto S, Hishida M, Inokawa Y, Sugimoto H and Kodera Y: Management of hepatocellular carcinoma should consider both tumor factors and background liver factors. Hepatobiliary Surg Nutr. 3:82–85. 2014.PubMed/NCBI

110 

Utsunomiya T, Shimada M, Morine Y, Tajima A and Imoto I: Specific molecular signatures of non-tumor liver tissue may predict a risk of hepatocarcinogenesis. Cancer Sci. 105:749–754. 2014. View Article : Google Scholar : PubMed/NCBI

111 

Utsunomiya T, Shimada M, Imura S, Morine Y, Ikemoto T and Mori M: Molecular signatures of noncancerous liver tissue can predict the risk for late recurrence of hepatocellular carcinoma. J Gastroenterol. 45:146–152. 2010. View Article : Google Scholar : PubMed/NCBI

112 

Okamoto M, Utsunomiya T, Wakiyama S, Hashimoto M, Fukuzawa K, Ezaki T, Hanai T, Inoue H and Mori M: Specific gene-expression profiles of noncancerous liver tissue predict the risk for multicentric occurrence of hepatocellular carcinoma in hepatitis C virus-positive patients. Ann Surg Oncol. 13:947–954. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Utsunomiya T, Ishikawa D, Asanoma M, Yamada S, Iwahashi S, Kanamoto M, Arakawa Y, Ikemoto T, Morine Y, Imura S, et al: Specific miRNA expression profiles of non-tumor liver tissue predict a risk for recurrence of hepatocellular carcinoma. Hepatol Res. 44:631–638. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Hoshida Y, Villanueva A, Kobayashi M, Peix J, Chiang DY, Camargo A, Gupta S, Moore J, Wrobel MJ, Lerner J, et al: Gene expression in fixed tissues and outcome in hepatocellular carcinoma. N Engl J Med. 359:1995–2004. 2008. View Article : Google Scholar : PubMed/NCBI

115 

Nomoto S, Hishida M, Inokawa Y, Takano N, Kanda M, Nishikawa Y, Fujii T, Koike M, Sugimoto H and Kodera Y: Expression analysis of THOP1 in background liver, a prognostic predictive factor in hepatocellular carcinoma, extracted by multiarray analysis. Ann Surg Oncol. 21(Suppl 3): S443–S450. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Sonohara F, Nomoto S, Inokawa Y, Hishida M, Takano N, Kanda M, Nishikawa Y, Fujii T, Koike M, Sugimoto H and Kodera Y: High expression of Janus kinase 2 in background normal liver tissue of resected hepatocellular carcinoma is associated with worse prognosis. Oncol Rep. 33:767–773. 2015.PubMed/NCBI

117 

Ammerpohl O, Pratschke J, Schafmayer C, Haake A, Faber W, von Kampen O, Brosch M, Sipos B, von Schönfels W, Balschun K, et al: Distinct DNA methylation patterns in cirrhotic liver and hepatocellular carcinoma. Int J Cancer. 130:1319–1328. 2012. View Article : Google Scholar : PubMed/NCBI

118 

Um TH, Kim H, Oh BK, Kim MS, Kim KS, Jung G and Park YN: Aberrant CpG island hypermethylation in dysplastic nodules and early HCC of hepatitis B virus-related human multistep hepatocarcinogenesis. J Hepatol. 54:939–947. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Arai E, Ushijima S, Gotoh M, Ojima H, Kosuge T, Hosoda F, Shibata T, Kondo T, Yokoi S, Imoto I, et al: Genome-wide DNA methylation profiles in liver tissue at the precancerous stage and in hepatocellular carcinoma. Int J Cancer. 125:2854–2862. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Yoshida T, Kato J, Maekita T, Yamashita S, Enomoto S, Ando T, Niwa T, Deguchi H, Ueda K, Inoue I, et al: Altered mucosal DNA methylation in parallel with highly active Helicobacter pylori-related gastritis. Gastric Cancer. 16:488–497. 2013. View Article : Google Scholar : PubMed/NCBI

121 

Lee KH, Hwang D, Kang KY, Lee S, Kim DY, Joo YE and Lee JH: Frequent promoter methylation of CDH1 in non-neoplastic mucosa of sporadic diffuse gastric cancer. Anticancer Res. 33:3765–3774. 2013.PubMed/NCBI

122 

Kadara H, Fujimoto J, Yoo SY, Maki Y, Gower AC, Kabbout M, Garcia MM, Chow CW, Chu Z, Mendoza G, et al: Transcriptomic architecture of the adjacent airway field cancerization in non-small cell lung cancer. J Natl Cancer Inst. 106:dju0042014. View Article : Google Scholar : PubMed/NCBI

123 

Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008. View Article : Google Scholar : PubMed/NCBI

124 

Zheng Z, Liang W, Wang D, Schroder PM, Ju W, Wu L, Zheng Z, Shang Y, Guo Z and He X: Adjuvant chemotherapy for patients with primary hepatocellular carcinoma: A meta-analysis. Int J Cancer. 136:E751–E759. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Kubo S, Takemura S, Sakata C, Urata Y and Uenishi T: Adjuvant therapy after curative resection for hepatocellular carcinoma associated with hepatitis virus. Liver Cancer. 2:40–46. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Inokawa Y, Inaoka K, Sonohara F, Hayashi M, Kanda M and Nomoto S: Molecular alterations in the carcinogenesis and progression of hepatocellular carcinoma: Tumor factors and background liver factors (Review). Oncol Lett 12: 3662-3668, 2016
APA
Inokawa, Y., Inaoka, K., Sonohara, F., Hayashi, M., Kanda, M., & Nomoto, S. (2016). Molecular alterations in the carcinogenesis and progression of hepatocellular carcinoma: Tumor factors and background liver factors (Review). Oncology Letters, 12, 3662-3668. https://doi.org/10.3892/ol.2016.5141
MLA
Inokawa, Y., Inaoka, K., Sonohara, F., Hayashi, M., Kanda, M., Nomoto, S."Molecular alterations in the carcinogenesis and progression of hepatocellular carcinoma: Tumor factors and background liver factors (Review)". Oncology Letters 12.5 (2016): 3662-3668.
Chicago
Inokawa, Y., Inaoka, K., Sonohara, F., Hayashi, M., Kanda, M., Nomoto, S."Molecular alterations in the carcinogenesis and progression of hepatocellular carcinoma: Tumor factors and background liver factors (Review)". Oncology Letters 12, no. 5 (2016): 3662-3668. https://doi.org/10.3892/ol.2016.5141