Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 14, 2014; 20(30): 10249-10261
Published online Aug 14, 2014. doi: 10.3748/wjg.v20.i30.10249
Implications of biomarkers in human hepatocellular carcinoma pathogenesis and therapy
Li-Li Han, Hui Guo, Zhi-Ping Ruan, Ke-Jun Nan, Department of Medical Oncology, The First Affiliated Hospital of Medical College, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
Yi Lv, Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
Author contributions: Han LL conducted the literature review and wrote the manuscript; Guo H and Ruan ZP revised the manuscript; Lv Y and Nan KJ provided guidance in the preparation of the manuscript; all authors have read and approved the final version to be published.
Supported by National Natural Science Foundation of China, No. 81172361
Correspondence to: Ke-Jun Nan, PhD, Department of Medical Oncology, The First Affiliated Hospital of Medical College, Xi’an Jiaotong University, No. 58 Chang’an North Road, Beilin District, Xi’an 710061, Shaanxi Province, China. nankj@163.com
Telephone: +86-29-85324086 Fax: +86-29-85324086
Received: September 27, 2013
Revised: March 21, 2014
Accepted: April 27, 2014
Published online: August 14, 2014

Abstract

Hepatocellular carcinoma (HCC) is one of the most frequent tumors worldwide and accounts for approximately one-third of all malignancies. In the past decade, advances have been made to improve the prognosis of HCC, including improvement in the clinical diagnosis of early-stage HCC using molecular biomarkers and molecular-targeted therapy to treat advanced HCC. However, the diagnosis, pathogenesis and targeted therapy of HCC are not completely independent, and should be comprehensively studied. For example, a number of tumor markers provide useful clinical information not only for prognosis, but also in pathogenesis and treatment efficacy. Therefore, this review will focus on the role of several specific biomarkers implicated in the pathogenesis of HCC and several promising molecular-targeted drugs that target the biomarkers of HCC.

Key Words: Biomarkers, Pathogenesis, Hepatocellular carcinoma, Targeted therapy, MicroRNAs

Core tip: Advances made in the prognosis of hepatocellular carcinoma (HCC) in recent years include improvements in clinical diagnosis using biomarkers and the potential of molecular-targeted therapy. The diagnosis, pathogenesis and targeted therapy of HCC should be comprehensively studied. Several biomarkers, including both traditional biomarkers and novel biomarkers, such as microRNAs, are also essential in the pathogenesis of HCC, and represent important new targets for HCC therapy. Ongoing studies and clinical trials suggest that molecular-targeted drugs that target biomarkers and their pathways will be used to treat HCC.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death in men, the sixth leading cause of cancer-related death in women and is the most frequently diagnosed cancer worldwide[1]. In addition, HCC is one of the deadliest primary cancers, with a 5-year survival rate of 10% or less[2].

Chronic infections with either hepatitis B virus (HBV) or hepatitis C virus (HCV) greatly increase the relative risk of HCC. These chronic viral infections are present in more than 70% of HCC cases, and iatrogenic interventions for these viruses significantly reduce the risk of developing HCC[3]. In addition to HBV and HCV infection, other major pathogenetic factors include alcoholic cirrhosis, non-alcoholic steatohepatitis, metabolic syndrome and fatty liver. These diseases are all associated with impaired liver function in HCC patients, making treatment of HCC very difficult. Furthermore, the early stages of HCC are generally silent, with rapid growth of the tumor[4].

Despite the possibility of early detection using such methods as serum biomarkers, alpha-fetoprotein (AFP) and ultrasound examination, surgical resection or liver transplantation are not performed in the majority of HCC cases, because surgery can only be carried out at the early and confined stages of HCC. Chemotherapy has a response rate of less than 20%, thus is not a good treatment option. Limited treatment options and late diagnosis may explain the low survival rate of patients with HCC[5]. Only early diagnosis of HCC can improve the survival rate; therefore, studies on specific biomarkers, particularly novel biomarkers, such as microRNAs, over the last two decades are of profound importance. Both traditional tumor markers including AFP, glypican-3 and transforming growth factor (TGF)-β and novel biomarkers including microRNAs provide useful clinical data, not only on prognosis, but also on pathogenesis and treatment efficacy. Furthermore, specific biomarkers may be potential therapeutic targets.

This review will focus on the role of several specific biomarkers in the pathogenesis and treatment of HCC.

TRADITIONAL MOLECULAR BIOMARKERS AND THEIR IMPORTANCE IN PATHOGENESIS AND THERAPY

Traditional molecular markers used for HCC diagnosis can be classified into three major types: (1) serological markers; (2) cancer stem cell markers; and (3) tumor tissue markers. Of these, serological markers are most commonly used in the clinic, and are closely related to the pathogenesis and targeted therapies of HCC. Therefore, we will discuss several traditional serological molecular biomarkers involved in the pathogenesis and targeted therapies of HCC.

AFP

AFP is abundantly expressed in fetal liver cells, but not in normal adult liver cells, and is the most commonly used HCC serum biomarker. However, its sensitivity and predictive accuracy often depend on the AFP cut-off value, and the results are less than satisfactory. Sensitivity of HCC diagnosis is 60%-80% with a 20 ng/mL AFP cut-off value; however, the sensitivity decreases to 20%-40% for the detection of small tumors[6]. High serum AFP levels can also be found in several hepatitis infections and chronic liver conditions. Therefore, AFP is not considered an ideal diagnostic biomarker for HCC[5].

Further research has shown that the role of AFP in HCC targeted therapies is a concern. AFP computational secreted network construction and analysis of HCC is very useful to identify novel markers and potential targets for prognosis and therapy[7,8]. The specific expression level of AFP led to the AFP promoter being used as an HCC targeting promoter to drive the adenovirus E1A gene[9] or suicide genes, such as herpes simplex virus thymidine kinase.

Recent studies showed that AD55-Apoptin, in which the E1A gene was driven by the AFP promoter along with a 55 kDa deletion in the E1B gene to form AD55-Apoptin, may be a potential anti-hepatoma agent and has shown marked antitumor efficacy and safety in a cancer targeting gene-viro-therapy system[10]. Therefore, AFP may contribute to the therapy of HCC.

Glypican-3

Glypican-3 (GP3) is a member of the heat-shock protein family and plays a pivotal role in cell growth, differentiation and migration[11]. GP3 is expressed by most HCCs. Initially, only GP3 mRNA was reported to be significantly elevated in HCC compared with normal liver and was used as a biomarker[12]. A study by Capurro et al[12] in 2003 confirmed these results at the protein level. The authors found that 72% of HCC patients expressed GP3 in serum using immunoblotting and ELISA. Several independent studies confirmed these results[13-15]. The serological sensitivity and specificity of GP3 as a HCC biomarker were 77% and 96%, respectively, as reported by the International Consensus Group. Consequently, GP3 is used in the clinic to confirm a diagnosis of HCC.

In addition to being a biomarker of HCC, GP3 plays a pivotal role in the pathogenesis and progression of the disease[11]. GP3 promotes the growth of HCC by regulating the signaling activity of several growth factors, including the Wnt/β-catenin pathway, which is crucial for the progression of HCC[16,17]. This is based on the ability of GP3 to increase the binding of Wnt to its signaling receptor[18,19]. Several reports have confirmed GP3-induced activation of canonical Wnt signaling in HCC cells[19]. Furthermore, GP3 can stimulate fibroblast growth factor (FGF), which is activated in a significant proportion of HCC[20].

A humanized anti-GP3 monoclonal antibody has been produced and was safely administered intravenously up to 20 mg/kg per week in a phase I clinical trial of patients with advanced HCC[21]. In addition, investigators have attempted to inhibit HCC growth by blocking GP3 function using targeted GP3 in immunotherapeutic approaches[22,23]. Consequently, GP3 not only has a significant role in the diagnosis of HCC, but also an established role in the future therapy of HCC.

Serum Fas/FasL

Fas is a type I membrane protein and belongs to the tumor necrosis factor (TNF) receptor family, whereas FasL is a member of the TNF family. The Fas/FasL (Fas-ligand) system, which plays a major and unique role in HCC growth and metastasis, is upregulated in various chronic liver diseases and accelerates their progression. Serum Fas (sFas) expression is upregulated in chronic hepatitis B, chronic hepatitis C[24] and acute liver failure[25]. Furthermore, sFas is significantly higher in HCC patients compared with patients suffering from chronic hepatitis C or liver cirrhosis. In addition, the level of sFas and FasL increase from chronic hepatitis to cirrhosis[26]. The linear relationship between expression of Fas in liver tissue and its serum levels suggests that these parameters could be considered predictive markers of tumorigenesis in HCC[27].

Several independent studies have shown that overexpression of FasL is associated with many tumors, including the advanced stages of HCC[28]. SFas is believed to act as a decoy receptor that prevents Fas/FasL binding and inhibits Fas-mediated apoptosis[29]. The sFas protein inhibits the activity of cytotoxic T lymphocytes in a dose-dependent manner, and is capable of inhibiting hepatic apoptosis by binding to FasL or anti-Fas antibodies, and triggering a cascade of intracellular signaling events that end in cell death by apoptosis. Fas/FasL signaling contributes to phenylalanine-induced apoptosis of HCC cells[30].

Apoptosis and the Fas system are significantly involved in the conversion of liver cirrhosis to HCC. Downregulation of Fas expression and upregulation of FasL expression in hepatocytes, and elevation of serum sFas levels are important in tumor evasion from immune surveillance and in hepatic carcinogenesis. Therefore, attention has been focused on the use of these components of the Fas system as targets for anticancer therapy.

TGF-β

The TGF-β family plays a pivotal role in physiology during embryonic development, as well as in the control of tissue homeostasis in adults, such as regeneration via cytokines, which regulates the growth and differentiation of both normal and transformed liver cells[31].

Although the presence of TGF may lead to some false positive results because of its high expression in liver cirrhosis[32], TGF is still a significant HCC biomarker, especially in patients with HCC caused by chronic hepatitis B; high serum TGF levels generally indicate a poor prognosis[33]. As a serological biomarker of HCC, TGF has a sensitivity of 68% at a cut-off value of 800 pg/mL, which is much higher than AFP at the same cut-off value, especially in the diagnosis of early stage HCC.

The TGF-β family regulates growth inhibition and induces apoptosis in hepatocytes[34]. Several independent studies have shown that TGF-β acts as an important tumor suppressor during the early stages of tumor development and as a proto-oncogene during the late phase of carcinogenesis. TGF-β acts as a tumor suppressor by inhibiting cell proliferation, and as a tumor promoter, TGF-β induces an epithelial-mesenchymal transition (EMT), cell motility and invasion[35] by regulation of vascular endothelial growth factor (VEGF), extracellular regulated protein kinases (ERK) and hypoxia inducible factor (HIF-1) and over-activation of cyclin and cyclin-dependent kinases[36].

Carmona-Cuenca et al[37] reported that TGF-β upregulates the Rac-independent NADPH oxidase, NOX4, which is inhibited by PI3K or mitogen-activated protein kinase (MAPK)/ERK anti-apoptotic signal pathways in both rat and human hepatocytes. This correlates with its proapoptotic activity, leading to mitochondrial-dependent apoptosis[38]. Furthermore, TGF reduces migration and invasion of HCC cells by upregulating E-cadherin[39].

In conclusion, any advance in the understanding of the molecular mechanisms that allow HCC cells to escape from TGF-β-induced apoptosis may have potential for future targeted therapy of HCC.

VEGF/VEGF receptor

VEGF was initially identified in 1983 as a protein secreted by tumor cells[40]. The expression of VEGF and its receptors, which include VEGF receptor (VEGFR)1, VEGFR2, and VEGFR3, is elevated in HCC cell lines and tissues, as well as in the blood circulation of patients with HCC. The combination of a serum AFP value > 19.8 ng/mL and a serum VEGF value > 355.2 pg/mL increased HCC screening sensitivity to 95.5% compared with the individual sensitivities of 68.2% and 86.4%, respectively[41]. Moreover, VEGF plays an important role in screening patients suitable for liver transplantation, because of its close relationship with microscopic venous invasion and intrahepatic metastasis. Higher VEGF expression in the serum of patients with small HCC (< 5 cm) predicts a poor prognosis after liver transplantation, radiofrequency ablation or transcatheter arterial chemoembolization. Furthermore, other studies have suggested that the expressions of VEGF-C, VEGFR1, and VEGFR3 in peritumoral liver tissue are associated with a unique type of HCC that had a poorer outcome after hepatectomy[42]. In addition, patients with remotely metastasized tumors showed much higher levels of serum VEGF compared with HCC patients without metastasis.

Numerous studies have shown that angiogenesis is pivotal in the pathogenesis of human HCC. Angiogenesis is mediated by the activation of different pathways in the tumor and endothelium, and the most critical player is VEGFR. As a permeability factor, VEGF promotes extravasation of plasma fibrinogen, leading to the formation of fibrin scaffolding, which facilitates cell migration during invasion. In addition, as an endothelial growth factor, the receptors of VEGF, which induce cell proliferation in an autocrine fashion, activate intracellular signals, such as the RAF/MEK/ERK pathway and the PI3K/AKT/mTOR pathway.

HCC is a vascular tumor, in which increased levels of VEGF and microvessel density have been observed. There are many anti-angiogenic agents in clinical trials of HCC, and most target VEGF and VEGFR[43]. There is no doubt that anti-angiogenesis therapies have shown promise in the treatment of HCC[44]. Therefore, future studies should identify and characterize these pathways, with the goal of targeting anti-VEGF therapies. Sorafenib, an oral multi-kinase inhibitor, targets VEGF-mediated angiogenesis and is the first drug to prolong the survival of patients with advanced HCC[45]. This has opened a new era for anti-angiogenic therapies in HCC[46]. Several important molecular-targeted therapies that inhibit the VEGF/VEGFR signaling pathways involved in HCC are summarized in Table 1.

Table 1 Several important molecular-targeted therapies that inhibit the vascular endothelial growth factor/vascular endothelial growth factor receptor signaling pathways involved in hepatocellular carcinoma.
Names of drugTargeted signaling pathwaysPhase/number of patientsMain side effects and prevalenceEfficiency (mo) (PFS/OS)
SorafenibRaf/MAPK/ERK, VEGFR-2, -3, PDGFRII/137Diarrhea (8%), hand-foot skin reaction (5%)5.5/9.2[49]
BevacizumabVEGF/VEGFRII/46Hypertension (15%), thrombosis (6%), and major bleeding (11%)6.9/12.4[52]
SunitinibVEGFR1, VEGFR2, PDGFR, c-KIT, FLT3, RET kinasesII/45Fatigue (62%), diarrhea (47%), nausea (44%)1.5/9.3[54]
BrivanibVEGFR, FGFRII/55Hypertension (33.8%), proteinuria (14.7%), hemorrhage (11.8%)2.7/10[56]
LinifanibVEGFR and PDGFRII/44Hypertension (25.0%) and fatigue (13.6%)3.7/9.7[58]
RamucirumabVEGFR-2II/40Hypertension (14%), gastrointestinal hemorrhage and infusion-related reactions (7% each), and fatigue (5%)4.0/12[59]

Sorafenib inhibits Raf/MAPK/ERK signaling, VEGFR-2, -3, and PDGFR, increases apoptosis, decreases angiogenesis and cell proliferation, and inhibits overall tumor cell signaling[47]. Based on the results of the recent large randomized phase III studies, Sorafenib has been approved by the United States Food and Drug Administration for the treatment of patients with advanced HCC. In the sorafenib HCC assessment randomized protocol (SHARP) trials, the median overall survival (OS) increased from 7.9 mo in the placebo group to 10.7 mo in the sorafenib group. Sorafenib also showed significant benefit in terms of time to progression (TTP), with a median of 5.5 mo in the sorafenib group and 2.8 mo in the placebo group[48]. It is worth noting that the impact of viral etiology in HCC on the survival of patients is still controversial. Survival benefits of sorafenib are different according to etiologies of HCC, with or without extrahepatic spreading and vascular invasion, as well as other risk factors by subgroup analyses. An Italian study suggested a worse survival for patients with HBV-related HCC, particularly in advanced-stage disease. A meta-analysis of 14 randomized clinical trials of systemic therapy indicated that HBV-related HCC is an independent predictor of better survival[49].

As the SHARP trial reported, the overall incidence of treatment-related adverse events was 80% in the sorafenib group and 52% in the placebo group. The rate of discontinuation of the study drug due to adverse events (38% vs 37%), grade 3-4 drug-related adverse events and the overall incidence of serious adverse events from any cause(52% vs 54%) were similar in the two study groups. The most common serious adverse events of any cause (aside from death) were liver dysfunction (7% and 5%, respectively), diarrhea (5% and 2%) and ascites (5% and 4%).

Sorafenib is currently undergoing investigation in a phase III study (the STORM trial) as an adjuvant therapy for the prevention of recurrence following surgery or local ablation. Moreover, a phase II trial is currently recruiting patients to determine the progression-free survival of patients with advanced or metastatic HCC treated with sorafenib plus tegafur/uracil.

Bevacizumab

Bevacizumab is a recombinant humanized monoclonal antibody that targets the VEGF signaling pathway and blocks tumor vascularization[50]. A phase II study of 46 patients with unresectable HCC, but no overt extrahepatic metastases or invasion of major blood vessels, treated with bevacizumab, found that six patients (13%) had objective responses (95%CI: 3-23), 65% were progression-free at 6 mo, median PFS was 6.9 mo (95%CI: 6.5-9.1), and median OS was 12.4 mo (95%CI: 9.4-19.9). Grade 3-4 adverse events included hypertension (15%) and thrombosis (6%, including 4% with arterial thrombosis)[51].

Sunitinib

Sunitinib, a multi-kinase inhibitor, blocks a number of angiogenesis-related signaling pathways, including VEGFR1, VEGFR2, PDGFR, c-KIT, FLT3, and RET kinases[52]. Sunitinib is a very promising multi-kinase inhibitor, as a study by Koeberle and colleagues showed that in 45 patients treated with a daily dose of 37.5 mg sunitinib, median PFS and OS were 1.5 and 9.3 mo, respectively[53].

In addition, a randomized phase III study comparing sunitinib with sorafenib in patients with advanced HCC was conducted. In this large study of 1073 patients, although sunitinib failed to demonstrate superiority in OS compared with sorafenib[54], both PFS and TTP in sunitinib-treated patients were superior to those in sorafenib-treated patients. Unfortunately, the use of sunitinib was discontinued because of adverse effects during phase II and phase III clinical trials.

Brivanib

Brivanib is a dual inhibitor of VEGFR and fibroblast growth factor receptor (FGFR). A phase II study was undertaken to assess the efficacy and safety of brivanib in patients with advanced HCC, and the antitumor activity of brivanib was preliminarily demonstrated. Median PFS and OS were 2.7 mo (95%CI: 1.4-3.0) and 10 mo (95%CI: 6.8-15.2) in the first line-study[55]. Median OS and TTP as assessed by investigators following second-line treatment with brivanib were 9.79 and 2.7 mo, respectively[56].

Linifanib (ABT-869)

Linifanib (ABT-869) is a potent inhibitor of VEGFR and PDGFR. Preliminary results from an open label, multicenter phase II study of linifanib in advanced HCC have been reported[57]. The median TTP/PFS was 112 d and median OS was 295 d (95%CI: 182-333). A phase III study comparing linifanib with sorafenib in advanced HCC is ongoing and the results are pending.

Ramucirumab (IMC-1121B)

Ramucirumab is a recombinant human monoclonal antibody against VEGFR-2. A phase II study demonstrated that the response rate (RR) was 10%, PFS was 4.0 mo, and OS was 12.0 mo in patients who had not received prior systemic therapy[58]. Furthermore, a phase III study of ramucirumab compared with placebo is ongoing in patients with advanced HCC who have failed or could not tolerate sorafenib.

Cediranib (AZD2171)

Cediranib has potent activity against pan-VEGFR inhibitor, PDGFR and c-Kit. In a phase II study, the median OS was 5.8 mo (95%CI: 3.4-7.3) and TTP was 2.8 mo (95%CI: 2.3-4.4) in patients with advanced HCC[59].

Other oral tyrosine-kinase inhibitors Pazopanib (GW786034), TSU-68, Lenvatinib (E7080) and Lenalidomide block a number of angiogenesis-related signaling pathways. Pazopanib targets VEGFR, PDGFR, and c-Kit, TSU-68 targets EGFR-2, PDGFR, and FGFR-2, Lenalidomide inhibits VEGF and FGF, and Lenvatinib (E7080) targets VEGFR1-3, FGFR1-4, RET, KIT, and PDGFR-beta. These agents showed promising results in phase II clinical trials of patients with advanced HCC[60-63].

NOVEL MOLECULAR BIOMARKERS AND THEIR IMPORTANCE IN PATHOGENESIS AND THERAPY
MicroRNAs

MicroRNAs (miRNAs), a class of noncoding RNAs of 15-25 nucleotides in length found in both plants and animals, have emerged as key posttranscriptional regulators of gene expression by interacting with the 3’ UTRs of protein-coding mRNAs[64]. Clinically, because of their tumor-specific expression and stability both in tissues and in the circulation, miRNAs have been proposed as novel biomarkers in the diagnostic and prognostic stratification of HCC. Numerous studies have reported the key role of miRNAs in tumor cell proliferation, apoptosis, metastasis and drug resistance[65]. By targeting different genes in tumor development, there is accumulating evidence to indicate the role of miRNAs as tumor suppressors or oncogenes in hepatic malignancies[66]. Furthermore, the encouraging therapeutic potential of miRNAs has been demonstrated in various studies in recent years.

miRNAs as biomarkers in HCC

The differential expression of miRNAs in HCC cells indicates the potential value of miRNA detection in the prediction of HCC diagnosis and prognosis. Table 2 summarizes studies on the clinical value of miRNA detection in HCC.

Table 2 Clinical relevance of deregulated microRNAs in hepatocellular carcinoma.
Molecular alterationmiRNAsClinical significance
Down regulationmiR-139Poor survival[70]
miR-26Shorter overall survival[82-87]
miR-124Gain of metastatic properties early recurrence[69]
miR-145Advanced tumor progression, poor prognosis[71]
miR-199a-3pReduced time to recurrence[72]
miR-199b-5pPoor overall survival and Progression-free survival rates[72]
miR-122Poor prognosis[82-87]
Up regulationmiR-222Shorter disease-free survival[73]
miR--135Poor prognosis worse Edmondson-Steiner grade, vein invasion, shortened overall survival and disease-free survival[74]
miR-221Multinodularity, reduced time to recurrence, gain of metastatic properties high tumor capsular infiltration[79]
miR--155Poorer recurrence-free survival and overall survival[75]
miR--182Intrahepatic metastasis and poor prognosis[76]
miR- 10bPoor prognosis[77]
miR-17-5pPoor prognosis[78]
miR-21Poor prognosis[79]

Downregulation of miR-99a[67], -124[68], -139[69], -145[70] and -199b[71] was significantly associated with poor prognosis, shorter disease-free survival and features of metastatic tumors including venous invasion, microsatellite formation, absence of tumor encapsulation and reduced differentiation. Conversely, high levels of miR-222[72], -135a[73], -155[74], -182[75], -10b[76], -17-5p[77], -221 and -21[78], correlated with poor prognosis, such as increased risk of tumor recurrence and shorter overall survival.

In addition, for classification purposes, miR-200c, miR-141 and miR-126, alone or in combination, could be used to distinguish primary HCC compared to tumor metastases to the liver with very high accuracy; moreover, the ratio of miR-205 to miR-194 expression could be used to distinguish between gastrointestinal tumors and metastases outside the gastrointestinal system[79].

Extracellular miRNAs in the circulation are stable because RNAse in the blood protects them from enzymatic cleavage, suggesting that the expression profile of miRNAs in serum or plasma may also serve as novel diagnostic markers[80]. To date, more than 20 circulating miRNAs have been associated with HCC detection, of these miRNAs, the levels of miR-122, -192, -21, -223, -26a, -27a and -801a were significantly higher in patients with HCC and could help detect early-stage HCC with high diagnostic accuracy[81-86]. However, because increased levels are also detected in chronic hepatitis, their usefulness as clinical tumor markers needs to be further validated. Several independent studies showed that the amount of miR-500 in the serum of HCC patients increased and then returned to normal after surgical treatment. Moreover, the relative amount of miR-92a in the plasma of HCC patients was reduced compared with that in healthy persons[87]. In addition, the expression profile of miR-25, miR-375, and let-7f in serum could identify HCC cases[88]. These studies confirmed the potential use of miRNAs as sensitive markers for the detection of underlying HCC and for prognostic stratification of the disease.

Involvement of miRNAs in HCC pathogenesis

In addition to being biomarkers of HCC, miRNAs play a pivotal role in the pathogenesis and progression of this disease.

MiRNAs and HCC-associated virus infection: miRNAs regulate HBV and HCV infection, which significantly increase the relative risk of HCC, at the transcription level either by targeting crucial cellular transcription factors required for HBV gene expression or by directly binding to HBV transcripts[89]. For example miR-1, miR-152 and miR-148a play protective roles in HBV-induced HCC[66]. Similarly, miR-196 inhibits HCV transcription by upregulating heme oxygenase (decycling) 1 (HMOX1) expression[90]. In addition, miR-217 promotes hepatocyte ethanol-induced fat accumulation, which is another risk factor for HCC[91].

MiRNAs and HCC-associated molecular pathways: Numerous studies have demonstrated that HCC develops via deregulation of various molecular pathways, such as PTEN/PI3K/AKT/mTOR, p53, RAS/MAPK, WNT/β-catenin, MET, CDKN1B/p27/Kip1, CDKN1C/p57/Kip2 and transforming growth factor beta. MiRNAs play key roles in regulating these diverse molecular pathways, similar to transcription factors. It has been reported that miRNAs exert their functions either as oncogenes or tumor suppressor genes in HCC. Deregulated miRNAs in cancer cells contribute to diverse molecular pathways, including sustained proliferative signaling, survival, angiogenesis, invasion, and metastasis[92] (Table 3).

Table 3 MicroRNAs deregulated in hepatocellular carcinoma.
miRNAsExpression in HCCTargetInvolvement in cellular processes
miR-199DownMet, mTOR, PAK4, DDR1, caveolin-2Proliferation, autophagy, metastasis, invasion; autophagy, drug resistance; cell growth[95]
miR-449Downc-MetProliferation; apoptosis[97]
miR-122DownBcl-w, ADAM-1, Wnt-1Apoptosis; metastasis; angiogenesis[100-102]
miR-34aDownc-Met; CCL22Metastasis[103]
miR-200aDownHDAC4Proliferation; metastasis[104]
miRs-let-7a, -7b, -7c, -7d, -7f-1DownCaspase-3, HMGA2, C-myc, Bcl-xlProliferation, apoptosis[112]
miR-125a, -125bDownMMP11, SIRT7, VEGF-A, LIN28B2, Bcl-2, Mcl-1, Bcl-wAngiogenesis, apoptosis, metastasis, proliferation[115,116]
miR-29DownSPARCProliferation[114]
miR-224UpRKIP; CDC42; CDH1; PAK2; BCL-2; MAPK1; API-5Metastasis; proliferation; apoptosis[113]
miR-519dUpCDKN1A/p21; PTEN; AKT3; TIMP2Proliferation; invasion; apoptosis[109]
miR-216aUpTSLC1,PTENTumorigenesis[107]
miR-148aUpPTENTumorigenesis[108]
miR-21UpPTEN, RhoB, PDCD4Drug resistance, metastasis[105,106]
miR-221UpBmf, DDIT4, Arnt, CDKN1B/p27, CDKN1C/p57Angiogenesis, apoptosis, proliferation[98,99]

MiR-199 regulates several targets, including target genes, such as MET, mTOR, HIF-1α48-51 and transmembrane glycoprotein CD44. Restoring the expression of miRNA-199 in HCC cells led to reduced invasive capability, enhanced susceptibility to hypoxia, and increased sensitivity to doxorubicin-induced apoptosis[93]. Similarly, downregulation of miR-199a in patients with HCC is associated with a higher recurrence rate and shorter time to recurrence after surgery. Therefore, miR-199 was suggested to act as a tumor suppressor gene in HCC. C-Met can also be suppressed by other miRNAs, including miR-198[94] and miR-449[95].

MiR-221 affects several cancer pathways, such as cyclin-dependent kinase inhibitors CDKN1B/p27 and CDKN1C/p57, and the PTEN-PI3K-AKT-mTOR pathway. Other important targets include the BH3-only protein, Bcl-2-modifying factor (Bmf) and DNA damage-inducible transcript TIMP3, a tissue inhibitor of metalloproteases[96]. Overexpression of miR-221 increased growth, proliferation, migration, and invasion in HCC cells. Similarly, overexpression of miR-221 promoted tumor progression and shortened the survival of animals with cancer. Recently, a study using a transgenic mouse model with overexpression of miR-221 demonstrated that cancer in these animals was partly inhibited by anti-miR-221 oligonucleotides. Taken together, miR-199 is thought to be a tumor oncogene in HCC[97].

MiR-122, a negative regulator of p53, is downregulated in approximately 70% of HCCs, suggesting that it has tumor suppressor function. Studies using miR-122 knockout mice demonstrated a direct role for miR-122 in liver cancer[98,99]. It was shown that high expression of miR-122 induced apoptosis, arrested the cancer cell cycle, inhibited tumorigenicity and sensitized HCC cells to sorafenib or doxorubicin[100]. On the other hand, loss of miR-122 expression in patients with HCC correlated with metastasis and recurrence.

MiR-34a, another downstream target of tumor suppressor p53, functions as a link between p53 signaling and cell cycle regulation by targeting cyclin D1, cyclin-dependent kinase 4 (CDK4) and CDK2 in HCC[101]. In addition, p53 upregulates the miR-200 and miR-192 families of miRNAs to inhibit ZEB1/2-mediated EMT[102].

MiR-21, which downregulates the expression of tumor suppressor phosphatase and tensin homolog (PTEN)[103], is also a potent oncogene in HCC. Overexpression of miR-21 in HCC cells can increase tumor cell proliferation and migration. In addition, miR-21 plays a role in the inhibition of cell proliferation and increased apoptosis in vivo[104].

PTEN is downregulated by many other miRNAs in HCC, such as miR-216a[105], miR-148a[106], miR-519d[107], and miR-29a[108], leading to activation of the PI3K/AKT/mTOR pathway. Moreover, miR-7 regulates the PTEN/PI3K/Akt pathway by targeting phosphoinositide3-kinase (PIK3CD), mTOR, and p70S6K[109].

Let-7 miRNAs negatively regulate B-cell lymphoma-extra large (Bcl-xL) expression and enhance the sensitivity of HCC cells to apoptosis induced by Mcl-1-targeting anticancer drugs[110]. On the other hand, B-cell lymphoma 2 (Bcl-2), induced myeloid leukemia cell differentiation protein (Mcl-1) and Bcl-2-like protein 2 (Bcl-w) are targets of miR-224[111], miR-29[112] and miR-125b, respectively[113,114].

Potential roles of miRNAs in HCC therapy

Recent studies have suggested that strategies based on the modulation of miRNA activity may provide a novel approach to treating HCC.

MiRNA inhibition: several independent studies have demonstrated that inhibition of miR-122 by administering anti-miRNA oligonucleotides in nonhuman primates resulted in a reduction in miRNA activity in the adult liver without any evidence of toxicity, and led to prolonged survival or a reduction in the number and size of tumor nodules[115,116]. It was confirmed that AAV-mediated delivery of miR-122 inhibited tumorigenesis in a myc mouse HCC model[100]. Treatment with an LNA-modified anti-miR-122 was well tolerated and led to stable suppression of HCV infection in cynomolgus monkey models[117]. In addition, an antisense inhibitor of miR-122 could produce a dose-dependent and prolonged decrease in HCV RNA levels in patients with chronic HCV genotype 1 infection, which is a major cause of hepatocellular carcinoma in many Western countries[118].

Anti-miR-221 was previously shown to have antitumor activity in prostate carcinoma, melanoma and multiple myeloma[119]. Two recent independent studies using mouse models of HCC showed that anti-miR-221 molecules reduced the proliferation of tumor cells, promoted survival and achieved a significant reduction in the number and size of tumors compared with untreated mice[116,117].

In addition, separate restoration of miR-143 and miR-124 significantly inhibited tumorigenesis and metastasis in vivo[120].

Miravirsen SPC3649, the first miRNA-targeted drug, has been used in phase I investigations and is currently in a phase II clinical trial for the treatment of HCV infection[121].

MiRNA replacement: further studies demonstrating the antitumor effects of miRNAs have been reported in other types of tumors and experimental settings[122]. Thus, in addition to the inhibition of oncomirs, another approach to treating cancer is based on restoration of tumor suppressor miRNAs.

MiR-31 regulates the invasive properties of tumor cells, and elicits metastatic regression in already-established metastases[123].

AAV8 miR-199, which effectively restored miR-199a/b-3p, was reported to reduce tumor size in HCC[124].

Cholesterol-conjugated 2’-O-methyl, which effectively restored miR-375, significantly suppressed the growth of HCC in a mouse model[125].

MiR-34a is the first microRNA replacement therapy scheduled for use in the clinic[126]. In earlier studies, miR-34a was observed to prevent tumor growth and progression in mouse models of lung adenocarcinoma and multiple myeloma xenografts induced by K-ras and p53[127,128]. Recently, miR-34 combined with the cytokine, interleukin-24, showed synergistic antitumor activity in a mouse model of HCC, indicating its possible use in HCC therapy[129].

Such miR replacement therapies have been demonstrated in the case of miRs-26a[130] and -124[124] in mouse models of HCC.

MiRNAs influencing the chemosensitivity of HCC cells: miRNAs regulate the chemosensitivity of cancer cells. Tumors with high expression of oncomiR-21[131] and -181b[132] were resistant to IFN-5FU combination therapy and doxorubicin treatment, respectively. Therefore, antagomirs targeting miR-21 or miR-181b may be useful to increase drug efficacy. In contrast, restoration of miR-122 in HCC cells resulted in HCC cells being more sensitive to sorafenib treatment via downregulation of the expression of multidrug resistance (MDR) proteins[133]. In addition, recent studies showed that microRNA-200a/b influenced the therapeutic effects of curcumin in HCC cells, and miR-23a potentiated HCC cell response to etoposide via the inhibition of topoisomerase 1 expression.

Anti-miRNA oligonucleotides and miRNA mimics have antitumor activities. By confirming the feasibility as well as the safety and efficacy of these molecules, these studies established the basis for the use of anti-miRNAs as efficient therapeutic targets of HCC in clinical trials. Further studies are expected to assess the clinical value of miRNA-based approaches in HCC.

Potential mRNA biomarkers in HCC

Recently, molecularly-detected peripheral blood mRNA transcripts have been considered as new cancer biomarkers[134], especially with the application of new generation qPCR and sequencing platforms in mRNA biomarker analysis[135]. AFP and GPC3 mRNA in peripheral blood had clinical value as predictors of HCC[136]. Furthermore, some RNA-binding proteins regulated the expression of target spots through mRNA stability[137]; thus, mRNA biomarkers may represent potential therapeutic targets in HCC.

It is true that among the new molecules, miRNAs have a more clear pathophysiological association with HCC and a better diagnostic accuracy for HCC; however, it is also true that new biomarkers/molecules may not be easily available in all clinical settings (especially primary and secondary clinical settings) and accessibility may not be straightforward everywhere. Therefore, traditional molecular markers such as AFP, widely available and a relatively cheap biomarker for screening, may remain the main clinical marker for the foreseeable future.

CONCLUSION

Exhaustive research on the molecular biology of HCC in recent years has resulted in two major steps to improve the management of HCC: (1) a wide variety of molecular markers have been shown to be excellent diagnostic tools for HCC; and (2) improvements in the identification of several key molecular pathways involved in the pathogenesis of HCC have led to the development of new targeted therapies for HCC.

However, the diagnosis, pathogenesis and targeted therapy of HCC are not completely independent, and should be comprehensively studied. For example, a number of molecular markers, which have a significant impact on the molecular pathogenesis of HCC, were shown to be promising therapeutic targets for this disease, providing the rationale to develop new effective treatments.

There is new hope for improving the survival of patients with advanced HCC. However, additional clinical trials are required to improve the treatment of HCC patients.

Footnotes

P- Reviewer: Di Lorenzo G, Muengtaweepongsa S, Jamil K, Sebastiani G, Tanaka T, Yao DF S- Editor: Gou SX L- Editor: Stewart G E- Editor: Wang CH

References
1.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  He G, Karin M. NF-κB and STAT3 - key players in liver inflammation and cancer. Cell Res. 2011;21:159-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 749]  [Cited by in F6Publishing: 867]  [Article Influence: 61.9]  [Reference Citation Analysis (0)]
3.  El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264-1273.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Lin CL, Kao JH. The clinical implications of hepatitis B virus genotype: Recent advances. J Gastroenterol Hepatol. 2011;26 Suppl 1:123-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 259]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
5.  Lok AS, Sterling RK, Everhart JE, Wright EC, Hoefs JC, Di Bisceglie AM, Morgan TR, Kim HY, Lee WM, Bonkovsky HL. Des-gamma-carboxy prothrombin and alpha-fetoprotein as biomarkers for the early detection of hepatocellular carcinoma. Gastroenterology. 2010;138:493-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 412]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
6.  Chen L, Ho DW, Lee NP, Sun S, Lam B, Wong KF, Yi X, Lau GK, Ng EW, Poon TC. Enhanced detection of early hepatocellular carcinoma by serum SELDI-TOF proteomic signature combined with alpha-fetoprotein marker. Ann Surg Oncol. 2010;17:2518-2525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
7.  Wang L, Huang J, Jiang M, Zheng X. AFP computational secreted network construction and analysis between human hepatocellular carcinoma (HCC) and no-tumor hepatitis/cirrhotic liver tissues. Tumour Biol. 2010;31:417-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
8.  Kaleta EJ, Tolan NV, Ness KA, O’Kane D, Algeciras-Schimnich A. CEA, AFP and CA 19-9 analysis in peritoneal fluid to differentiate causes of ascites formation. Clin Biochem. 2013;46:814-818.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Ducher A, Armelle A, Renaud A, Chloe O. Protein of human adenovirus inhibits the overall activation of the target of type I interferons by preventing mono-ubiquitantion of histone 2B. Irologie. 2013;17:300-301.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Zhang KJ, Qian J, Wang SB, Yang Y. Targeting Gene-Viro-Therapy with AFP driving Apoptin gene shows potent antitumor effect in hepatocarcinoma. J Biomed Sci. 2012;19:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
11.  Capurro MI, Xiang YY, Lobe C, Filmus J. Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling. Cancer Res. 2005;65:6245-6254.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E, Filmus J. Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology. 2003;125:89-97.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Zhang L, Liu H, Sun L, Li N, Ding H, Zheng J. Glypican-3 as a potential differential diagnosis marker for hepatocellular carcinoma: a tissue microarray-based study. Acta Histochem. 2012;114:547-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
14.  Wang F, Jing X, Wang T, Li G, Li T, Zhang Q, Huang Y, Li J, Wang Y, Gao Y. Differential diagnostic value of GPC3-CD34 combined staining in small liver nodules with diameter less than 3 cm. Am J Clin Pathol. 2012;137:937-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
15.  Wang FH, Yip YC, Zhang M, Vong HT, Chan KI, Wai KC, Wen JM. Diagnostic utility of glypican-3 for hepatocellular carcinoma on liver needle biopsy. J Clin Pathol. 2010;63:599-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
16.  Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell. 2012;149:1192-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3740]  [Cited by in F6Publishing: 4054]  [Article Influence: 337.8]  [Reference Citation Analysis (0)]
17.  White BD, Chien AJ, Dawson DW. Dysregulation of Wnt/β-catenin signaling in gastrointestinal cancers. Gastroenterology. 2012;142:219-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 364]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
18.  Liu H, Li P, Zhai Y, Qu CF, Zhang LJ, Tan YF, Li N, Ding HG. Diagnostic value of glypican-3 in serum and liver for primary hepatocellular carcinoma. World J Gastroenterol. 2010;16:4410-4415.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Li L, Jin R, Zhang X, Lv F, Liu L, Liu D, Liu K, Li N, Chen D. Oncogenic activation of glypican-3 by c-Myc in human hepatocellular carcinoma. Hepatology. 2012;56:1380-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
20.  Gauglhofer C, Sagmeister S, Schrottmaier W, Fischer C, Rodgarkia-Dara C, Mohr T, Stättner S, Bichler C, Kandioler D, Wrba F. Up-regulation of the fibroblast growth factor 8 subfamily in human hepatocellular carcinoma for cell survival and neoangiogenesis. Hepatology. 2011;53:854-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 99]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
21.  Zhu AX, Gold PJ, El-Khoueiry AB, Abrams TA, Morikawa H, Ohishi N, Ohtomo T, Philip PA. First-in-man phase I study of GC33, a novel recombinant humanized antibody against glypican-3, in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2013;19:920-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 141]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
22.  Zittermann SI, Capurro MI, Shi W, Filmus J. Soluble glypican 3 inhibits the growth of hepatocellular carcinoma in vitro and in vivo. Int J Cancer. 2010;126:1291-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
23.  Feng M, Kim H, Phung Y, Ho M. Recombinant soluble glypican 3 protein inhibits the growth of hepatocellular carcinoma in vitro. Int J Cancer. 2011;128:2246-2247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
24.  El Bassiouny AE, El-Bassiouni NE, Nosseir MM, Zoheiry MM, El-Ahwany EG, Salah F, Omran ZS, Ibrahim RA. Circulating and hepatic Fas expression in HCV-induced chronic liver disease and hepatocellular carcinoma. Medscape J Med. 2008;10:130.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Galle PR, Hofmann WJ, Walczak H, Schaller H, Otto G, Stremmel W, Krammer PH, Runkel L. Involvement of the CD95 (APO-1/Fas) receptor and ligand in liver damage. J Exp Med. 1995;182:1223-1230.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Bortolami M, Kotsafti A, Cardin R, Farinati F. Fas / FasL system, IL-1beta expression and apoptosis in chronic HBV and HCV liver disease. J Viral Hepat. 2008;15:515-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
27.  Hammam O, Mahmoud O, Zahran M, Aly S, Hosny K, Helmy A, Anas A. The role of fas/fas ligand system in the pathogenesis of liver cirrhosis and hepatocellular carcinoma. Hepat Mon. 2012;12:e6132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
28.  Pryczynicz A, Guzińska-Ustymowicz K, Kemona A. Fas/FasL expression in colorectal cancer. An immunohistochemical study. Folia Histochem Cytobiol. 2010;48:425-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
29.  García-Santos G, Martin V, Rodríguez-Blanco J, Herrera F, Casado-Zapico S, Sánchez-Sánchez AM, Antolín I, Rodríguez C. Fas/Fas ligand regulation mediates cell death in human Ewing’s sarcoma cells treated with melatonin. Br J Cancer. 2012;106:1288-1296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
30.  Huang X, Lu Z, Lv Z, Yu T, Yang P, Shen Y, Ding Y, Fu D, Zhang X, Fu Q. The Fas/Fas ligand death receptor pathway contributes to phenylalanine-induced apoptosis in cortical neurons. PLoS One. 2013;8:e71553.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Heldin CH, Landström M, Moustakas A. Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr Opin Cell Biol. 2009;21:166-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
32.  Wright LM, Kreikemeier JT, Fimmel CJ. A concise review of serum markers for hepatocellular cancer. Cancer Detect Prev. 2007;31:35-44.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Fabregat I. Dysregulation of apoptosis in hepatocellular carcinoma cells. World J Gastroenterol. 2009;15:513-520.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sánchez A, Fabregat I. Growth factor- and cytokine-driven pathways governing liver stemness and differentiation. World J Gastroenterol. 2010;16:5148-5161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
35.  Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 843]  [Cited by in F6Publishing: 854]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
36.  Hough C, Radu M, Doré JJ. Tgf-beta induced Erk phosphorylation of smad linker region regulates smad signaling. PLoS One. 2012;7:e42513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 112]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
37.  Carmona-Cuenca I, Roncero C, Sancho P, Caja L, Fausto N, Fernández M, Fabregat I. Upregulation of the NADPH oxidase NOX4 by TGF-beta in hepatocytes is required for its pro-apoptotic activity. J Hepatol. 2008;49:965-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 180]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
38.  Caja L, Sancho P, Bertran E, Iglesias-Serret D, Gil J, Fabregat I. Overactivation of the MEK/ERK pathway in liver tumor cells confers resistance to TGF-{beta}-induced cell death through impairing up-regulation of the NADPH oxidase NOX4. Cancer Res. 2009;69:7595-7602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
39.  Fransvea E, Angelotti U, Antonaci S, Giannelli G. Blocking transforming growth factor-beta up-regulates E-cadherin and reduces migration and invasion of hepatocellular carcinoma cells. Hepatology. 2008;47:1557-1566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 209]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
40.  Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983-985.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  el-Houseini ME, Mohammed MS, Elshemey WM, Hussein TD, Desouky OS, Elsayed AA. Enhanced detection of hepatocellular carcinoma. Cancer Control. 2005;12:248-253.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Zhuang PY, Shen J, Zhu XD, Lu L, Wang L, Tang ZY, Sun HC. Prognostic roles of cross-talk between peritumoral hepatocytes and stromal cells in hepatocellular carcinoma involving peritumoral VEGF-C, VEGFR-1 and VEGFR-3. PLoS One. 2013;8:e64598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
43.  Llovet JM, Peña CE, Lathia CD, Shan M, Meinhardt G, Bruix J. Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2290-2300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
44.  Zhu AX, Duda DG, Sahani DV, Jain RK. HCC and angiogenesis: possible targets and future directions. Nat Rev Clin Oncol. 2011;8:292-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 16]  [Reference Citation Analysis (0)]
45.  Zhao Y, Wang WJ, Guan S, Li HL, Xu RC, Wu JB, Liu JS, Li HP, Bai W, Yin ZX. Sorafenib combined with transarterial chemoembolization for the treatment of advanced hepatocellular carcinoma: a large-scale multicenter study of 222 patients. Ann Oncol. 2013;24:1786-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
46.  Dufour JF, Hoppe H, Heim MH, Helbling B, Maurhofer O, Szucs-Farkas Z, Kickuth R, Borner M, Candinas D, Saar B. Continuous administration of sorafenib in combination with transarterial chemoembolization in patients with hepatocellular carcinoma: results of a phase I study. Oncologist. 2010;15:1198-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 18]  [Reference Citation Analysis (0)]
47.  Samant RS, Shevde LA. Recent advances in anti-angiogenic therapy of cancer. Oncotarget. 2011;2:122-134.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9016]  [Cited by in F6Publishing: 9515]  [Article Influence: 594.7]  [Reference Citation Analysis (1)]
49.  Hsu C, Shen YC, Cheng CC, Hu FC, Cheng AL. Geographic difference in survival outcome for advanced hepatocellular carcinoma: implications on future clinical trial design. Contemp Clin Trials. 2010;31:55-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
50.  Hsu CH, Yang TS, Hsu C, Toh HC, Epstein RJ, Hsiao LT, Chen PJ, Lin ZZ, Chao TY, Cheng AL. Efficacy and tolerability of bevacizumab plus capecitabine as first-line therapy in patients with advanced hepatocellular carcinoma. Br J Cancer. 2010;102:981-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
51.  Siegel AB, Cohen EI, Ocean A, Lehrer D, Goldenberg A, Knox JJ, Chen H, Clark-Garvey S, Weinberg A, Mandeli J. Phase II trial evaluating the clinical and biologic effects of bevacizumab in unresectable hepatocellular carcinoma. J Clin Oncol. 2008;26:2992-2998.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Iyer R, Fetterly G, Lugade A, Thanavala Y. Sorafenib: a clinical and pharmacologic review. Expert Opin Pharmacother. 2010;11:1943-1955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
53.  Koeberle D, Montemurro M, Samaras P, Majno P, Simcock M, Limacher A, Lerch S, Kovàcs K, Inauen R, Hess V. Continuous Sunitinib treatment in patients with advanced hepatocellular carcinoma: a Swiss Group for Clinical Cancer Research (SAKK) and Swiss Association for the Study of the Liver (SASL) multicenter phase II trial (SAKK 77/06). Oncologist. 2010;15:285-292.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, Chung HC, Song X, Xu J, Poggi G. Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase III trial. J Clin Oncol. 2013;31:4067-4075.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Park JW, Finn RS, Kim JS, Karwal M, Li RK, Ismail F, Thomas M, Harris R, Baudelet C, Walters I. Phase II, open-label study of brivanib as first-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2011;17:1973-1983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 132]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
56.  Finn RS, Kang YK, Mulcahy M, Polite BN, Lim HY, Walters I, Baudelet C, Manekas D, Park JW. Phase II, open-label study of brivanib as second-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2090-2098.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Toh HC, Chen PJ, Carr BI, Knox JJ, Gill S, Ansell P, McKeegan EM, Dowell B, Pedersen M, Qin Q. Phase 2 trial of linifanib (ABT-869) in patients with unresectable or metastatic hepatocellular carcinoma. Cancer. 2013;119:380-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 87]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
58.  Zhu AX, Finn RS, Mulcahy M, Gurtler J, Sun W, Schwartz JD, Dalal RP, Joshi A, Hozak RR, Xu Y. A phase II and biomarker study of ramucirumab, a human monoclonal antibody targeting the VEGF receptor-2, as first-line monotherapy in patients with advanced hepatocellular cancer. Clin Cancer Res. 2013;19:6614-6623.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Alberts SR, Fitch TR, Kim GP, Morlan BW, Dakhil SR, Gross HM, Nair S. Cediranib (AZD2171) in patients with advanced hepatocellular carcinoma: a phase II North Central Cancer Treatment Group Clinical Trial. Am J Clin Oncol. 2012;35:329-333.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Yau T, Chen PJ, Chan P, Curtis CM, Murphy PS, Suttle AB, Gauvin J, Hodge JP, Dar MM, Poon RT. Phase I dose-finding study of pazopanib in hepatocellular carcinoma: evaluation of early efficacy, pharmacokinetics, and pharmacodynamics. Clin Cancer Res. 2011;17:6914-6923.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Kanai F, Yoshida H, Tateishi R, Sato S, Kawabe T, Obi S, Kondo Y, Taniguchi M, Tagawa K, Ikeda M. A phase I/II trial of the oral antiangiogenic agent TSU-68 in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol. 2011;67:315-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 75]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
62.  Safran H, Charpentier KP, Kaubisch A, Mantripragada K, Dubel G, Perez K, Faricy-Anderson K, Miner T, Eng Y, Victor J. Lenalidomide for Second-line Treatment of Advanced Hepatocellular Cancer: A Brown University Oncology Group Phase II Study. Am J Clin Oncol. 2013;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Yamada K, Yamamoto N, Yamada Y, Nokihara H, Fujiwara Y, Hirata T, Koizumi F, Nishio K, Koyama N, Tamura T. Phase I dose-escalation study and biomarker analysis of E7080 in patients with advanced solid tumors. Clin Cancer Res. 2011;17:2528-2537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
64.  Zhang LY, Liu M, Li X, Tang H. miR-490-3p modulates cell growth and epithelial to mesenchymal transition of hepatocellular carcinoma cells by targeting endoplasmic reticulum-Golgi intermediate compartment protein 3 (ERGIC3). J Biol Chem. 2013;288:4035-4047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
65.  Zhang X, Zhang E, Ma Z, Pei R, Jiang M, Schlaak JF, Roggendorf M, Lu M. Modulation of hepatitis B virus replication and hepatocyte differentiation by MicroRNA-1. Hepatology. 2011;53:1476-1485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 154]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
66.  Au SL, Wong CC, Lee JM, Fan DN, Tsang FH, Ng IO, Wong CM. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatology. 2012;56:622-631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
67.  Li D, Liu X, Lin L, Hou J, Li N, Wang C, Wang P, Zhang Q, Zhang P, Zhou W. MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with prognosis of patients with hepatocellular carcinoma. J Biol Chem. 2011;286:36677-36685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 184]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
68.  Zheng F, Liao YJ, Cai MY, Liu YH, Liu TH, Chen SP, Bian XW, Guan XY, Lin MC, Zeng YX. The putative tumour suppressor microRNA-124 modulates hepatocellular carcinoma cell aggressiveness by repressing ROCK2 and EZH2. Gut. 2012;61:278-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 311]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
69.  Wong CC, Wong CM, Tung EK, Au SL, Lee JM, Poon RT, Man K, Ng IO. The microRNA miR-139 suppresses metastasis and progression of hepatocellular carcinoma by down-regulating Rho-kinase 2. Gastroenterology. 2011;140:322-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 245]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
70.  Jia Y, Liu H, Zhuang Q, Xu S, Yang Z, Li J, Lou J, Zhang W. Tumorigenicity of cancer stem-like cells derived from hepatocarcinoma is regulated by microRNA-145. Oncol Rep. 2012;27:1865-1872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
71.  Wang C, Song B, Song W, Liu J, Sun A, Wu D, Yu H, Lian J, Chen L, Han J. Underexpressed microRNA-199b-5p targets hypoxia-inducible factor-1α in hepatocellular carcinoma and predicts prognosis of hepatocellular carcinoma patients. J Gastroenterol Hepatol. 2011;26:1630-1637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
72.  Augello C, Vaira V, Caruso L, Destro A, Maggioni M, Park YN, Montorsi M, Santambrogio R, Roncalli M, Bosari S. MicroRNA profiling of hepatocarcinogenesis identifies C19MC cluster as a novel prognostic biomarker in hepatocellular carcinoma. Liver Int. 2012;32:772-782.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Liu S, Guo W, Shi J, Li N, Yu X, Xue J, Fu X, Chu K, Lu C, Zhao J. MicroRNA-135a contributes to the development of portal vein tumor thrombus by promoting metastasis in hepatocellular carcinoma. J Hepatol. 2012;56:389-396.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Han ZB, Chen HY, Fan JW, Wu JY, Tang HM, Peng ZH. Up-regulation of microRNA-155 promotes cancer cell invasion and predicts poor survival of hepatocellular carcinoma following liver transplantation. J Cancer Res Clin Oncol. 2012;138:153-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
75.  Wang J, Li J, Shen J, Wang C, Yang L, Zhang X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer. 2012;12:227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
76.  Li QJ, Zhou L, Yang F, Wang GX, Zheng H, Wang DS, He Y, Dou KF. MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells. Tumour Biol. 2012;33:1455-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 82]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
77.  Chen L, Jiang M, Yuan W, Tang H. miR-17-5p as a novel prognostic marker for hepatocellular carcinoma. J Invest Surg. 2012;25:156-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
78.  Karakatsanis A, Papaconstantinou I, Gazouli M, Lyberopoulou A, Polymeneas G, Voros D. Expression of microRNAs, miR-21, miR-31, miR-122, miR-145, miR-146a, miR-200c, miR-221, miR-222, and miR-223 in patients with hepatocellular carcinoma or intrahepatic cholangiocarcinoma and its prognostic significance. Mol Carcinog. 2013;52:297-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 278]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
79.  Huang YH, Lin YH, Chi HC, Liao CH, Liao CJ, Wu SM, Chen CY, Tseng YH, Tsai CY, Lin SY. Thyroid hormone regulation of miR-21 enhances migration and invasion of hepatoma. Cancer Res. 2013;73:2505-2517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 655]  [Reference Citation Analysis (0)]
80.  Qi J, Wang J, Katayama H, Sen S, Liu SM. Circulating microRNAs (cmiRNAs) as novel potential biomarkers for hepatocellular carcinoma. Neoplasma. 2013;60:135-142.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Zhou J, Yu L, Gao X, Hu J, Wang J, Dai Z, Wang JF, Zhang Z, Lu S, Huang X. Plasma microRNA panel to diagnose hepatitis B virus-related hepatocellular carcinoma. J Clin Oncol. 2011;29:4781-4788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 469]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
82.  Xu J, Wu C, Che X, Wang L, Yu D, Zhang T, Huang L, Li H, Tan W, Wang C. Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol Carcinog. 2011;50:136-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 410]  [Cited by in F6Publishing: 448]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
83.  Cermelli S, Ruggieri A, Marrero JA, Ioannou GN, Beretta L. Circulating microRNAs in patients with chronic hepatitis C and non-alcoholic fatty liver disease. PLoS One. 2011;6:e23937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 436]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
84.  Tomimaru Y, Eguchi H, Nagano H, Wada H, Kobayashi S, Marubashi S, Tanemura M, Tomokuni A, Takemasa I, Umeshita K. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J Hepatol. 2012;56:167-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 271]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
85.  Trebicka J, Anadol E, Elfimova N, Strack I, Roggendorf M, Viazov S, Wedemeyer I, Drebber U, Rockstroh J, Sauerbruch T. Hepatic and serum levels of miR-122 after chronic HCV-induced fibrosis. J Hepatol. 2013;58:234-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 139]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
86.  van der Meer AJ, Farid WR, Sonneveld MJ, de Ruiter PE, Boonstra A, van Vuuren AJ, Verheij J, Hansen BE, de Knegt RJ, van der Laan LJ. Sensitive detection of hepatocellular injury in chronic hepatitis C patients with circulating hepatocyte-derived microRNA-122. J Viral Hepat. 2013;20:158-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
87.  Shigoka M, Tsuchida A, Matsudo T, Nagakawa Y, Saito H, Suzuki Y, Aoki T, Murakami Y, Toyoda H, Kumada T. Deregulation of miR-92a expression is implicated in hepatocellular carcinoma development. Pathol Int. 2010;60:351-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 138]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
88.  Li LM, Hu ZB, Zhou ZX, Chen X, Liu FY, Zhang JF, Shen HB, Zhang CY, Zen K. Serum microRNA profiles serve as novel biomarkers for HBV infection and diagnosis of HBV-positive hepatocarcinoma. Cancer Res. 2010;70:9798-9807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 365]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
89.  Liu WH, Yeh SH, Chen PJ. Role of microRNAs in hepatitis B virus replication and pathogenesis. Biochim Biophys Acta. 2011;1809:678-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
90.  Hou W, Tian Q, Zheng J, Bonkovsky HL. MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expression in human hepatoma cells expressing hepatitis C viral proteins. Hepatology. 2010;51:1494-1504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 163]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
91.  Yin H, Hu M, Zhang R, Shen Z, Flatow L, You M. MicroRNA-217 promotes ethanol-induced fat accumulation in hepatocytes by down-regulating SIRT1. J Biol Chem. 2012;287:9817-9826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 125]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
92.  Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39812]  [Cited by in F6Publishing: 43011]  [Article Influence: 3308.5]  [Reference Citation Analysis (4)]
93.  Fornari F, Milazzo M, Chieco P, Negrini M, Calin GA, Grazi GL, Pollutri D, Croce CM, Bolondi L, Gramantieri L. MiR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2010;70:5184-5193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 328]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
94.  Tan S, Li R, Ding K, Lobie PE, Zhu T. miR-198 inhibits migration and invasion of hepatocellular carcinoma cells by targeting the HGF/c-MET pathway. FEBS Lett. 2011;585:2229-2234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
95.  Buurman R, Gürlevik E, Schäffer V, Eilers M, Sandbothe M, Kreipe H, Wilkens L, Schlegelberger B, Kühnel F, Skawran B. Histone deacetylases activate hepatocyte growth factor signaling by repressing microRNA-449 in hepatocellular carcinoma cells. Gastroenterology. 2012;143:811-20.e1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 156]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
96.  Sharma AD, Narain N, Händel EM, Iken M, Singhal N, Cathomen T, Manns MP, Schöler HR, Ott M, Cantz T. MicroRNA-221 regulates FAS-induced fulminant liver failure. Hepatology. 2011;53:1651-1661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
97.  Santhekadur PK, Das SK, Gredler R, Chen D, Srivastava J, Robertson C, Baldwin AS, Fisher PB, Sarkar D. Multifunction protein staphylococcal nuclease domain containing 1 (SND1) promotes tumor angiogenesis in human hepatocellular carcinoma through novel pathway that involves nuclear factor κB and miR-221. J Biol Chem. 2012;287:13952-13958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
98.  Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, Yu L, Bai S, La Perle K, Chivukula RR. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest. 2012;122:2871-2883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 591]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
99.  Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest. 2012;122:2884-2897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 612]  [Cited by in F6Publishing: 632]  [Article Influence: 52.7]  [Reference Citation Analysis (0)]
100.  Ma L, Liu J, Shen J, Liu L, Wu J, Li W, Luo J, Chen Q, Qian C. Expression of miR-122 mediated by adenoviral vector induces apoptosis and cell cycle arrest of cancer cells. Cancer Biol Ther. 2010;9:554-561.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Cheng J, Zhou L, Xie QF, Xie HY, Wei XY, Gao F, Xing CY, Xu X, Li LJ, Zheng SS. The impact of miR-34a on protein output in hepatocellular carcinoma HepG2 cells. Proteomics. 2010;10:1557-1572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
102.  Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon YJ, Volinia S, Pineau P, Marchio A, Palatini J, Suh SS. p53 regulates epithelial-mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J Exp Med. 2011;208:875-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 404]  [Cited by in F6Publishing: 425]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
103.  Buscaglia LE, Li Y. Apoptosis and the target genes of microRNA-21. Chin J Cancer. 2011;30:371-380.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Medina PP, Nolde M, Slack FJ. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature. 2010;467:86-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 726]  [Cited by in F6Publishing: 736]  [Article Influence: 52.6]  [Reference Citation Analysis (0)]
105.  Wu K, Ding J, Chen C, Sun W, Ning BF, Wen W, Huang L, Han T, Yang W, Wang C. Hepatic transforming growth factor beta gives rise to tumor-initiating cells and promotes liver cancer development. Hepatology. 2012;56:2255-2267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
106.  Yuan K, Lian Z, Sun B, Clayton MM, Ng IO, Feitelson MA. Role of miR-148a in hepatitis B associated hepatocellular carcinoma. PLoS One. 2012;7:e35331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
107.  Fornari F, Milazzo M, Chieco P, Negrini M, Marasco E, Capranico G, Mantovani V, Marinello J, Sabbioni S, Callegari E. In hepatocellular carcinoma miR-519d is up-regulated by p53 and DNA hypomethylation and targets CDKN1A/p21, PTEN, AKT3 and TIMP2. J Pathol. 2012;227:275-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 159]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
108.  Kong G, Zhang J, Zhang S, Shan C, Ye L, Zhang X. Upregulated microRNA-29a by hepatitis B virus X protein enhances hepatoma cell migration by targeting PTEN in cell culture model. PLoS One. 2011;6:e19518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 146]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
109.  Fang Y, Xue JL, Shen Q, Chen J, Tian L. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 2012;55:1852-1862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 329]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
110.  Shimizu S, Takehara T, Hikita H, Kodama T, Miyagi T, Hosui A, Tatsumi T, Ishida H, Noda T, Nagano H. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. J Hepatol. 2010;52:698-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 263]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
111.  Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H, Chayama K. Involvement of microRNA-224 in cell proliferation, migration, invasion, and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol Hepatol. 2013;28:565-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
112.  Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH, Zhuang SM. Effects of microRNA-29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology. 2010;51:836-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 286]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
113.  Zhao A, Zeng Q, Xie X, Zhou J, Yue W, Li Y, Pei X. MicroRNA-125b induces cancer cell apoptosis through suppression of Bcl-2 expression. J Genet Genomics. 2012;39:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
114.  Gong J, Zhang JP, Li B, Zeng C, You K, Chen MX, Yuan Y, Zhuang SM. MicroRNA-125b promotes apoptosis by regulating the expression of Mcl-1, Bcl-w and IL-6R. Oncogene. 2013;32:3071-3079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 161]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
115.  Park JK, Kogure T, Nuovo GJ, Jiang J, He L, Kim JH, Phelps MA, Papenfuss TL, Croce CM, Patel T. miR-221 silencing blocks hepatocellular carcinoma and promotes survival. Cancer Res. 2011;71:7608-7616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 181]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
116.  Callegari E, Elamin BK, Giannone F, Milazzo M, Altavilla G, Fornari F, Giacomelli L, D’Abundo L, Ferracin M, Bassi C. Liver tumorigenicity promoted by microRNA-221 in a mouse transgenic model. Hepatology. 2012;56:1025-1033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 134]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
117.  Hildebrandt-Eriksen ES, Aarup V, Persson R, Hansen HF, Munk ME, Ørum H. A locked nucleic acid oligonucleotide targeting microRNA 122 is well-tolerated in cynomolgus monkeys. Nucleic Acid Ther. 2012;22:152-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
118.  Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685-1694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1665]  [Cited by in F6Publishing: 1627]  [Article Influence: 147.9]  [Reference Citation Analysis (0)]
119.  Di Martino MT, Gullà A, Cantafio ME, Lionetti M, Leone E, Amodio N, Guzzi PH, Foresta U, Conforti F, Cannataro M. In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget. 2013;4:242-255.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Lang Q, Ling C. MiR-124 suppresses cell proliferation in hepatocellular carcinoma by targeting PIK3CA. Biochem Biophys Res Commun. 2012;426:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 108]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
121.  Lindow M, Kauppinen S. Discovering the first microRNA-targeted drug. J Cell Biol. 2012;199:407-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 207]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
122.  Ibrahim AF, Weirauch U, Thomas M, Grünweller A, Hartmann RK, Aigner A. MicroRNA replacement therapy for miR-145 and miR-33a is efficacious in a model of colon carcinoma. Cancer Res. 2011;71:5214-5224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 304]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
123.  Valastyan S, Chang A, Benaich N, Reinhardt F, Weinberg RA. Activation of miR-31 function in already-established metastases elicits metastatic regression. Genes Dev. 2011;25:646-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
124.  Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q, Qin L, Wu X, Zheng Y, Yang Y. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell. 2011;19:232-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 542]  [Cited by in F6Publishing: 570]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
125.  He XX, Chang Y, Meng FY, Wang MY, Xie QH, Tang F, Li PY, Song YH, Lin JS. MicroRNA-375 targets AEG-1 in hepatocellular carcinoma and suppresses liver cancer cell growth in vitro and in vivo. Oncogene. 2012;31:3357-3369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 277]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
126.  Bader AG. miR-34 - a microRNA replacement therapy is headed to the clinic. Front Genet. 2012;3:120.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Kasinski AL, Slack FJ. miRNA-34 prevents cancer initiation and progression in a therapeutically resistant K-ras and p53-induced mouse model of lung adenocarcinoma. Cancer Res. 2012;72:5576-5587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 197]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
128.  Di Martino MT, Leone E, Amodio N, Foresta U, Lionetti M, Pitari MR, Cantafio ME, Gullà A, Conforti F, Morelli E. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: in vitro and in vivo evidence. Clin Cancer Res. 2012;18:6260-6270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 190]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
129.  Lou W, Chen Q, Ma L, Liu J, Yang Z, Shen J, Cui Y, Bian XW, Qian C. Oncolytic adenovirus co-expressing miRNA-34a and IL-24 induces superior antitumor activity in experimental tumor model. J Mol Med (Berl). 2013;91:715-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
130.  Zhu Y, Lu Y, Zhang Q, Liu JJ, Li TJ, Yang JR, Zeng C, Zhuang SM. MicroRNA-26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb protein. Nucleic Acids Res. 2012;40:4615-4625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 169]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
131.  Tomimaru Y, Eguchi H, Nagano H, Wada H, Tomokuni A, Kobayashi S, Marubashi S, Takeda Y, Tanemura M, Umeshita K. MicroRNA-21 induces resistance to the anti-tumour effect of interferon-α/5-fluorouracil in hepatocellular carcinoma cells. Br J Cancer. 2010;103:1617-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
132.  Wang B, Hsu SH, Majumder S, Kutay H, Huang W, Jacob ST, Ghoshal K. TGFbeta-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene. 2010;29:1787-1797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 303]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
133.  Bai S, Nasser MW, Wang B, Hsu SH, Datta J, Kutay H, Yadav A, Nuovo G, Kumar P, Ghoshal K. MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J Biol Chem. 2009;284:32015-32027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in F6Publishing: 409]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
134.  Lasa A, Garcia A, Alonso C, Millet P, Cornet M, Ramón y Cajal T, Baiget M, Barnadas A. Molecular detection of peripheral blood breast cancer mRNA transcripts as a surrogate biomarker for circulating tumor cells. PLoS One. 2013;8:e74079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
135.  Devonshire AS, Sanders R, Wilkes TM, Taylor MS, Foy CA, Huggett JF. Application of next generation qPCR and sequencing platforms to mRNA biomarker analysis. Methods. 2013;59:89-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
136.  Wang Y, Shen Z, Zhu Z, Han R, Huai M. Clinical values of AFP, GPC3 mRNA in peripheral blood for prediction of hepatocellular carcinoma recurrence following OLT: AFP, GPC3 mRNA for prediction of HCC. Hepat Mon. 2011;11:195-199.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Yin T, Cho SJ, Chen X. RNPC1, an RNA-binding protein and a p53 target, regulates macrophage inhibitory cytokine-1 (MIC-1) expression through mRNA stability. J Biol Chem. 2013;288:23680-23686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]