Skip to main content
Biomarkers in Cancer logoLink to Biomarkers in Cancer
. 2016 Jul 4;8(Suppl 1):37–55. doi: 10.4137/BIC.S34413

Tissue- and Serum-Associated Biomarkers of Hepatocellular Carcinoma

Ranjit Chauhan 1,2,, Nivedita Lahiri 3
PMCID: PMC4933537  PMID: 27398029

Abstract

Hepatocellular carcinoma (HCC), one of the leading causes of cancer deaths in the world, is offering a challenge to human beings, with the current modes of treatment being a palliative approach. Lack of proper curative or preventive treatment methods encouraged extensive research around the world with an aim to detect a vaccine or therapeutic target biomolecule that could lead to development of a drug or vaccine against HCC. Biomarkers or biological disease markers have emerged as a potential tool as drug/vaccine targets, as they can accurately diagnose, predict, and even prevent the diseases. Biomarker expression in tissue, serum, plasma, or urine can detect tumor in very early stages of its development and monitor the cancer progression and also the effect of therapeutic interventions. Biomarker discoveries are driven by advanced techniques, such as proteomics, transcriptomics, whole genome sequencing, micro- and micro-RNA arrays, and translational clinics. In this review, an overview of the potential of tissue- and serum-associated HCC biomarkers as diagnostic, prognostic, and therapeutic targets for drug development is presented. In addition, we highlight recently developed micro-RNA, long noncoding RNA biomarkers, and single-nucleotide changes, which may be used independently or as complementary biomarkers. These active investigations going on around the world aimed at conquering HCC might show a bright light in the near future.

Keywords: biomarker, HCC, liver, miRNA, lncRNA, prognosis, prevention, therapeutic

Introduction

Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer mortality. HCC is defined as a primary tumorigenesis in the liver, mainly in patients suffering from chronic liver cirrhosis or hepatitis B or C. The tumor gradually spreads to hepatocytes and in advanced stages metastasizes to other organs, such as lungs and brain. HCC has become one of the very common cancers causing death, affecting more than 500,000 people in the world.1 The other main risk factors for HCC are alcohol and aflatoxin.2,3

The Development of HCC

HCC is one of the major liver-related mortalities in cirrhosis. As shown in Figure 1, when the healthy liver is affected by hepatitis B virus (HBV) or hepatitis C virus (HCV), a gradual chronic infection leads to liver cirrhosis. Cirrhosis is characterized by a decrease in the growth of healthy hepatocytes and is due to degeneration and regeneration of cells.3 Regeneration leads to increase in fibrous scar tissue following the destruction of the healthy liver cells, which provides the environment for forming cancerous tumors.4 Telomerase, which plays an important role in maintaining telomere length and chromosomal stability in hepatocytes5 and their shortening, limits the regenerative capacity of organs during chronic disease.6 In a cirrhotic liver, the telomeres of the hepatocytes are significantly shorter than in noncirrhotic tissue.6 Telomere dysfunctions, along with p53 mutation, are associated with the onset of early-stage hepatic neoplasms.7

Figure 1.

Figure 1

Stages of Hepatocellular carcinoma development and induction of biomarkers. Highlighted liver indicates decisive stage during which liver is either dysregulated by insult or it tolerates such insult till certain limits.

Another characteristic of cirrhosis is the activation of stellate cells. This leads to an increase in the production of cytokines, growth factors, and products of oxidative stress, many of which have been shown to affect hepatocyte proliferation and so could play a role in tumor formation.8

The main oncogenic pathways involved in HCC are phosphoinositol-3-kinase (PI3K)/Akt, myc, Wnt/β-catenin, c-Met, and hedgehog (Fig. 14).9,10 Activation of Akt signaling is thought to promote tumor formation by suppressing transforming growth factor (TGF)-β-induced apoptosis, which in turn activates Wnt/β-catenin signaling, so further driving the hepatocarcinogenic process.

Figure 2.

Figure 2

Main factors and signaling involved in hepatocarcinogenesis.

Figure 3.

Figure 3

Mechanism of action of sGPC3. (A) Wild-type GPC3 bound to the cell membrane facilitates/stabilizes the interaction of Wnt and other growth factors (bFGF and HGF) with their signaling receptors. (B) sGPC3 sequesters growth factors and inhibits their interaction with signaling receptors. © 2009 UICC, reused with permission from 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.

Figure 4.

Figure 4

14-3-3–HSF-1/HSP70 cell signaling in HCC cells. Modified from Wu YJ, Jan YJ, Ko BS, Liang SM, Liou JY. Involvement of 14-3-3 proteins in regulating tumor progression of hepatocellular carcinoma. Cancers. 2015;7(2):1022–1036, under the Creative Commons Attribution License.

Current HCC therapeutic and management methods

HCC patients are subjected to multiple treatments, including transcatheter arterial chemoembolization, brachytherapy with radioactive yytrium,1113 and also cytokine/hormonal therapy, such as interleukin (IL)-2.14 A potent drug sorafenib, an oral agent with antiangiogenic, proapoptotic, and Raf-kinase inhibitory properties, has been safely evaluated in clinical phase II and phase III trials.1517 Several drugs modulating different cell signaling mechanisms, mainly PI3K–Akt, mammalian target of rapamycin (mTOR), and RAF/MEK/ERK pathways, are being investigated for their effectiveness to control the progression of HCC. For example, KU-0060648 was found to inhibit HCC cell proliferation by both DNA-activated protein kinase-dependent and -independent me chanisms.18 A study involving VO-OHpic, a phosphatase and tensin homolog (PTEN) inhibitor in HCC cells, demonstrated that VO-OHpic inhibited HCC cell viability, cell proliferation, and colony formation in synergy with PI3K/mTOR and RAF/MEK/ERK pathway inhibitors.19 Antroquinonol, a traditional Chinese liver treatment drug, exhibited anticancer activity by activating 5′-adenosine mono phosphate (AMP) kinase and inhibiting mTOR pathway,20 leading to G1 cycle arrest and cellular apoptosis. In another study, it is shown that overexpression of far upstream element-binding protein in HCC and other cancers,21,22 which is directly regulated by P13K/Akt/mTOR pathway, could be substantially reduced by sorafenib.23,24

Role of biomarkers in HCC

Biomarkers are the molecular indicators of the physiological status detectable in blood, urine, or tissue and can be important for the management of various diseases. Their concentrations and changes in body fluids can provide an estimate about the disease progression. Ideally, biomarkers should have high sensitivity of detection where there is small cancer, should be highly specific, and must not increase in noncancer. Their levels should increase with tumor progression and predict the prognosis. HCC is one such life-threatening condition that could be detected early or, in an optimistic note, hoped to be prevented by biomarkers’ therapeutic and prognostic capabilities. Table 1 lists various categories of biomarkers upregulated in HCC.

Table 1.

Biomarkers, their origin, and the significance.

NAME CATEGORY DESCRIPTION REFERENCE
Alpha Fetoprotein (AFP) Serum glycoprotein antigen AFP >500 ng/mL are indicative of HCC, early diagnosis and monitoring 61, 226, 227
Glypican 3 (GPC3) Tissue biomarker-membrane bound heparin sulfate proteoglycan GPC3 mRNA and protein significantly upregulated in HCC specifically, early diagnosis 29, 228
Des-Gamma Carboxyl Prothrombin (DCP) Cellular enzyme-secreted by the cancerous hepatocyte (serum biomarker) DCP >0.1 AU/mL (100 ng/mL) are highly suggestive of HCC, early diagnoses, metastasis 229
Gamma Glutamyl Transferase (GGT) Serum biomarker GGTII isoform significantly upregulated in HCC 230
Alpha-1-Fucosidase (AFU) Lysosomal enzyme, serum biomarker More than 2-fold increase in HCC patients compared to normal, liver cirrhosis or chronic hepatitis patients, early diagnosis 86
DKK-1 Serum/secreted protein biomarker Enhances HCC progression by accumulation of beta-catenin by Wnt signaling, early HCC biomarker 76, 231
Human carbonyl reductase-2 Secreted by Liver and kidney, serum biomarker Detoxicating enzyme, level falls in HCC, prognosis 80
Golgi Phosphoprotein-2 (GOLPH2)/Golgi protein 73 (GP73) Serum biomarker Increased significantly, higher sensitivity than AFP, tumor aggressiveness 232
Transforming Growth factor beta (TGF-beta) Serum biomarker Tumor invasiveness, mRNA overexpressed in HCC tissue 233235
Epidermal Growth Factor Receptor Family (EGFR) Cellular (transmembrane) biomarker Early recurrence and reduced disease-free survival following resection of hepatocellular carcinoma 236
Hepatocyte Growth Factor (HGF) Serum biomarker (cytokine) Prognosis, invasiveness and metastasis 237
Fibroblast Growth factor (FGF) Heparin binding polypeptide FGF level >10.8 pg/mL predicts HCC and reduced disease free survival 238
Molecular markers
• AFP mRNA
• GGT mRNA, type B
• (Insulin-like Growth factor) IGF-II mRNA
• Albumin mRNA
• miRNA (micro RNAs)
Serum biomarker HCC tissue
Serum biomarker
Serum biomarker
Nucleotide biomarker
Increased postsurgical circulation depicts more chances of recurrence, extrahepatic metastasis, monitoring of postoperative recurrence, tumor spread and survival 184, 239245
Pathological biomarkers
• Heat Shock Protein (HSP70)
• Glutamine synthase
• Ki-67
• Survivin
• E-Cadherin
• Vascular Endothelial Growth Factor (VEGF)
Tissue biomarker Increased transformed cell proliferation, survival, cell-to-cell adhesion, neoangiogenesis 246
Hepatocyte Paraffin 1 (HepPar1) Tissue biomarker Decreased expression in HCC cells 247
Squamous Cell Carcinoma Antigen (SCCA)-IgM immunocomplex Serum biomarker Significant enhancement during progression to HCC 248,249
Tumor Associated Glycoprotein 72 (TAG-72) Tissue biomarker Upregulation indicates tumor progression and metastasis, associated with poor survival 46
Zinc-α2-glycoprotein (ZAG) Serum biomarker (soluble glycoprotein) Overexpressed in HCC 250
DLK-1 (Delta like protein-1) Embryonic, kidney and neural tissue Significantly elevated in HCC 251
Villin 1 (Vil1) HCC tumor tissue Upregulated Vil1 indicates transformation, vascular invasion and cancer progression 252

Tumor Tissue Biomarkers of HCC

The biopsies and extensive research on tumor tissues of HCC provide a wide range of information about the abnormal constituents of tumor cells and what constituent is upregulated in transformed or neoplastic conditions. This information has provided researchers with therapeutic targets for vaccine, drug development, and screening for surveillance or device prognostic measures to prevent HCC in the past couple of decades.2528 Here, we focus on some of the potential tissue biomarkers of HCC that function as appropriate targets for early diagnosis and development of antimetastatic vaccines/drugs.

Glypican-3

Glypican-3 (GPC3), a potent tissue biomarker of HCC, links to the cell membrane by a glycosylphosphatidylinositol anchor. It is a heparan sulfate proteoglycan that is involved in regulating the cell growth. Besides, GPC3 can remove growth factors, such as hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), from the cell surface and inhibit the growth of HCC.29,30 GPC3 is particularly expressed in HCC, but it is not produced by normal or cirrhotic hepatic cells.29,31 Genetic studies in many mammals exhibited that glypicans can regulate the signaling of Wnt, hedgehog, and FGF.3235

The glypicans interact with Wnts and their signaling receptors, the frizzleds.36 The GPC3 stimulates Wnt signaling activity by stabilizing the interaction with of Wnt with frizzled, resulting in the proliferation of the HCC cells. The GPC3-induced stimulatory activity requires the attachment of GPC3 to the cell membrane.36 In a study by Zittermann et al,30 it was demonstrated that a mutant GPC3 could not attach to the cell membrane (sGP3) and is secreted extracellularly (in the serum) and, thus, could remove the autocrine/paracrine Wnt from the cell surface of the HCC. The removal of Wnt could inhibit the proliferation of HCC and block the activity of FGF, VEGF, epidermal growth factor, and HGF.3739 Hence, sGPC3 (mutant GPC3) is a potent tissue biomarker that can be targeted for HCC management.

Heat shock protein 70

Heat shock protein 70 (HSP70) is an essential molecular chaperon upregulated in response to heat, stress, or cell survival protection, tightly controlled by heat shock factor-1 (HSF-1). Studies have demonstrated that HSF-1 and HSP70 are involved in HCC tumor invasion and metastasis.40,41 It has been demonstrated that 14-3-3σ protein induces HSP70 via a β-catenin/HSF-1-dependent pathway, which in its downstream modulates HCC.42 Hence, 14-3-3σ–HSF-1 or HSF-1/HSP70 complex is another important tissue biomarker to be targeted for developing a prognostic tool for HCC.

Tumor-associated glycoprotein 72

Tumor-associated glycoprotein 72 (TAG-72) is a cell membrane mucin-like glycoprotein complex, overexpressed in many of human adenocarcinomas but not in normal tissues.4345 In a study by Zhang et al,46 it was demonstrated that TAG-72 is a new tissue biomarker in HCC that indicates the poor survival of patients (Fig. 5).

Figure 5.

Figure 5

TAG-72 as a potent prognostic marker in various cancers: Curves showing an estimate of overall survivability of cancer patients with absence of TAG-72 expression and presence of TAG-72 expression in cancer tissue after days of treatment.

Ki-67 antigen

Ki-67 is a nuclear protein associated with cellular proliferation.47 The Ki-67 levels were expressed significantly correlating with higher increases from grades I to IV HCC tumors. This proliferative marker, with increased expression, indicated increased severity or spread of HCC (ranging from a mean+/− SD of 4.1+/−4.01 in tumor grade I to 29+/−15.01 in grade IV tumors).48

Hepatocyte paraffin 1

Hepatocyte paraffin 1 (HepPar1) is a monoclonal antibody prepared from a failed liver allograft,49 which recognizes an epitope that is a component of hepatic mitochondria and not found in other normal tissues.49,50 Hep-Par1 is often used as a marker to distinguish between HCC and secondary or metastatic hepatic neoplasms.51,52 A strong HepPar1 expression was detected in majority of the HCC tumors (35 out of 48 HCCs),53 whereas it was also expressed in nonhepatic tumors, such as lung, gallbladder, stomach, pancreas, colon, and malignant melanoma, but to a much lesser extent.5458

HERC5

Homologous to the E6-AP carboxyl terminus (HECT) domain and RCC-1-like domain-containing protein 5 is E3 ligase that conjugates with ISG15 to regulate several proteins. One of the main mechanisms of action of this molecule is by induction of CCL20, which in turn increases T regulatory infiltration, and it is one of the important prognostic biomarkers for tumor recurrence in HCC patients as well as survival in liver transplant patients.59 It is also an important biomarker in the prognosis of lung cancer, and hypermethylation of promoter of HERC5 was associated with poor survival of stage 1 adenocarcinoma.59,60

Serum Biomarkers of HCC

Alpha fetoprotein

Alpha fetoprotein (AFP) is produced by embryonic liver cells during pregnancy. It is in abundance during gestational stages, but its production is minimal after birth.61 AFP is one of the most common serum biomarkers used for the diagnosis of HCC by clinicians; however, the specificity and reliability of AFP biomarker is questioned, and it is of less value in the early stages of HCC when the tumor size is <3 cm.62

AFP-L3

It is a different form of AFP that differs in binding affinity with a lectin Lens culinaris agglutinin. This form of AFP-L is used as an early biomarker of HCC when the size of tumor is <2 cm. As the size of tumor increases, the sensitivity of this marker is increased.63 AFP-L is connected with Ki-67; as a marker of increased nuclear expression of Ki-67, there is a decrease in the expression of β-catenin, which is associated with distant metastasis.64 In case of the AFP-negative HCC, β-catenin positivity is more common.65

Glycoprotein 73

Glycoprotein 73 (GP73) is a type II Golgi-localized phosphoprotein, encoded by Golgi trans-membrane type II (GOLPH2) gene located on chromosome 9q21. It is expressed in epithelial cells in several human tissues and was first identified by serum glycoproteomics.66 GP73 is highly expressed in the tumor part of the liver; however, its expression is comparatively very less in normal part of the same liver.67,68 Similar observations were noted in woodchuck animal model of HCC.67 Interestingly, it can differentiate patients developing cirrhosis with that of high-grade HCC and is also an early biomarker of HCC. During insult to the liver, ie, acute hepatitis and autoimmune hepatitis, GP73 is highly expressed. Viral infections are also found to increase the GP73 secretion.69 Oncostatin M, produced in the adipose tissues and a proinflammatory cytokine IL-6, increases the mRNA levels of GP73 as detected in HepG2 cell lines.70 In addition to HCC, it has been found in other cancers, such as lung adenocarcinoma, testicular seminomas, renal cell carcinoma, and prostate cancer; however, in prostate cancer, it is detected only in urine and not in serum and, therefore, not a specific liver cancer biomarker.71 As given in Figure 6A, there are two major isoforms of GP73: one is complete and another is incomplete. In the incomplete form, first 10 amino acids at the N-terminal region are lacking and may have functional implications.

Figure 6.

Figure 6

(A) Alignment of the amino acid sequence of the two isoforms of GP73 taken from Uniprot protein ID-Q8NBJ4-1 and Uniprot protein ID-Q8NBJ4-2. Three blue box represents the glycosylation sites in two isoforms. Compared to isoform-1, isoform-2 is 10 amino acid shorter in N-terminal region. (B) Increased expression of GP73 in cirrhosis and HCC compared to the healthy controls. Increased expression of GP73 in patients infected with HBV-associated HCC and cirrhosis. Increased expression also correlates with the grading of the liver necroinflammation. Panel (B) is reused from Xu Z, Liu L, Pan X, et al. Serum golgi protein 73 (GP73) is a diagnostic and prognostic marker of chronic HBV liver disease. Tarantino G, ed. Medicine. 2015;94(12):e659, under the Creative Commons Attribution License.

APO-J

APO-J is a glycoprotein with seven glycosylated sites and is also known as clusterin.72 It is more sensitive and specific than AFP.73 APO-J is significantly decreased in HCC patients compared to healthy controls and can be used as an independent marker of HCC.72 One of the recent studies also showed that it can be used as a prognostic marker and can also monitor HCC progression and metastasis.74,75

DKK-1 Dickkopf-p1

DKK biomarker is of high importance in cases where AFP biomarker misses HCC diagnosis. Importantly, it can diagnose HCC in very early stages of cancer.76,77 One of the studies by Zhu et al reported that DKK-1 can be used as a biomarker for HCC patients undergoing liver transplantation (LT) and can predict the prognosis of such patients.78 Studies on this biomarker are ongoing, and more robust studies are required to consider it as a biomarker.

Human carbonyl reductase-2

It is expressed in human liver as well as kidneys. During oxidative stress, the released reactive oxygen species and alpha dicarbonyl are detoxified by this enzyme.79 The expression of human carbonyl reductase-2 (HCR2) is significantly decreased in the tumor part of the liver compared to the normal cells.80 The decreased expression of carbonyl reductase-2 leads to cancer growth as it triggers the cell damage through reactive oxygen species and other carcinogens.

Midkine

Midkine is expressed at the time of early embryogenesis and is a heparin-binding growth factor. It is expressed during wound healing, tumorogenesis, and inflammation.81 Compared to AFP, which is significantly raised during advanced stages of HCC, midkine is marginally raised and cannot be used as the biomarker in advanced stages of HCC. However, midkine expression markedly rises during early development of HCC.82

Des-γ-carboxy prothrombin

It is an important biomarker in large size HCC, and in cases where AFP misses HCC diagnosis, des-γ-carboxy prothrombin is able to detect HCC.83 It is an abnormal prothrombin protein of VEGF family expressed during vitamin K deficiency/antagonist-II (PIVKA-II). During insult to the liver cells, when normal cells convert to fibroid cells or epithelial-to-mesenchymal transition, hypoxic shock leads to the induction of DES.84

α-1-Fucosidase

α-1-Fucosidase is a lysosomal enzyme that hydrolyzes fucose glucosidic bonds of glycolipids and glycoprotein. Its expression increases with liver insult similar to chronic hepatitis, cirrhosis, and HCC patients.85 It is one of the early HCC biomarkers and has a cut-off value of 870 nm/mL/h. Interestingly, it is raised preceding six months of development of HCC, and its value is over 700 nm/mL/h in 85% of HCC patients.86

Hepatocyte growth factor

HGF is a cytokine produced by nonparenchymal Ito cells in the liver.87 It stimulates the expression of immediate early genes in primary cultures of hepatocytes.88 In hepatic regeneration, chronic hepatitis, cirrhosis, and HCC, HGF levels increase, and a level of 1.0 ng/mL indicates poor survival.89 It acts as a prognostic biomarker and can predict the early tumor recurrence and metastasis.90

Nerve growth factor

Nerve growth factor (NGF) is a member of neurotrophin family and is important for differentiation, survival, and preservation of peripheral and central nervous systems.91 NGF levels are associated with tumor growth, invasion, and metastasis. There are two receptors of NGF: high-affinity trkANGF and low-affinity p75NTR. Expression of NGF and trkANGF increases significantly during HCC. It can differentiate between cirrhosis that develops to HCC and cirrhosis that does not lead to HCC.92

Vascular endothelial growth factor

VEGF is a glycosylated cytokine that acts as a mitogen and mediates vascular permeability, angiogenesis, vasculogenesis, and endothelial cell growth-reduced survival.93 Tumor characteristic and environment promotes VEGF expression and initiates VEGF signaling and thus triggers downstream MAPK cascade (Ras/RAF/MEK/ERK) which is involved in angiogenesis, proliferation and metastasis and is shown in Figure 7. VEGF levels envisage HCC recurrence, and it is a substantial biomarker for the survival of HCC patients.94,95 A phase III clinical study conducted on 602 HCC patients receiving sorafenib showed that VEGF was one of the molecules that predicted patients’ survival suggesting its role as a biomarker in the prognosis of HCC.96

Figure 7.

Figure 7

Interaction between VEGF, VEGFR-2, and sVEGFR-2 via MAPK pathway. Modified from Ratnasari N, Yano Y. Do soluble vascular endothelial growth factor and its receptors predict the progression of chronic hepatitis to hepatocellular carcinoma? Hepatitis. 2015;1(1):4, under the Creative Commons Attribution License.

Transforming growth factor-β

TGF-β plays an important role in the control of cellular proliferation and differentiation in HCC cells. Serum TGF-β levels are raised in HCC patients and is a long sought biomarker of HCC.97 One of the recent reports documented the role of the TGF-β-interacting factor as one of the prognostic biomarkers of HCC.98 Moreover, its partner mucin1 mediates TGF-β signaling by activating JNK/AP1 pathway and can also be used as a therapeutic target for the treatment of HCC.99

Epidermal growth factor

Epidermal growth factor receptor (EGFR) signaling is one of the important players in all the phases of hepatic injury from very early stages of inflammation to HCC development, including fibrogenesis and neoplastic transformation.100 One of the forms of EGFR, ErbB3, was detected in the serum of HCC patients during the early stages of HCC development and was associated with portal vein invasion and metasatsis.101 One of the recent studies reported that HBV HBx protein downregulates the EGFR expression by inducing miRNA-7 in HCC cells.102 In addition, it is an important player and has tumor promoting role in non-HCC cells via live resident macrophages.103 Using an in vivo model, a recent study determined the potential role of soluble EGFR in HCC metastasis.104

Wnt

Wnt-1 protein has been described as a prognostic biomarker of HBV-related and HCV-related HCC after surgery.105 GPC3 molecule, which is described in the “Glypican-3” section, promotes the growth of HCC by stimulating Wnt signaling.106

Angiopointin-1/2

Angiopointin-2 levels are raised in HCC and cirrhosis, and it has been documented by several studies that elevated levels of Ang2 could be used as a marker of advanced pathological invasiveness and overall survival of HCC patients.96,107,108 It is also shown that Ang2 contributes to multiple organ failure and sepsis.109

NOTCH

Activation of NOTCH plays a prominent role in HBV-mediated HCC by proliferating hepatic cells and further supporting the growth of HCC, and this is also proved in in vivo mouse models.110,111 NOTCH1 is one of the possible therapeutic targets for the treatment of HBx-associated HCC.112 Recent studies showed that NOTCH1 and NOTCH4 are important biomarkers revealing the poor prognosis of HCC.113,114

Oncostatin M

OSM is a member of cytokine family, which is very early secreted from the hematopoietic cells, and induces the differentiation of hepatocytes by regulating HNF4 alpha.115 It regulates cytokine production, such as IL-6, GM-CSF, and G-CSF. It is also shown to be elevated in HCC and acts in synergy with IL-6.116

Alpha-1 antitrypsin

Alpha-1 antitrypsin is a member of the SERPINA1 family of proteins, which is controlled by IL-6, TNα, and IL-1.117,118 Increased levels of A1AT have been associated with HCC. A recent study revealed changes in the status of A1AT glycosylation during HCC and also documented that the core fucosyalation of HCC is one of the main reasons behind such changes.119 Compared to cirrhosis, it is significantly elevated in HCC and could also be used as a differentiation marker.120

WFA+ M2BP

Kuno et al121 first reported an assay that uses Wisteria floribunda agglutinin-positive human mac2-binding protein in assessing liver fibrosis. A recent study by Yamasaki et al122 showed high value of measuring WFA+ M2BP and highlighted that it can be used as an independent risk factor biomarker for HCC development. Interestingly, WFA+ M2BP can predict HCC in HCV patients who respond well to the treatment and achieve the sustained virological response.123

Lymphotoxin beta receptor

Lymphotoxin beta receptor is a cytokine and a member of tumor necrosis factor family, which is well known for controlling the development of lymphoid organs.124 In HCC as well as in cholangiocarcinoma cells, lymphotoxin beta receptor is vastly expressed and sustains the oncogene activity.125 It correlates with the upregulated Akt/NOTCH1 signaling and is a marker of poor survival in cholangiocarcinoma patients.126

Long Noncoding RNA as Biomarkers of HCC

MALAT1

Long noncoding RNA (lncRNA) is an RNA molecule with a length of 200 bp to 100 kbp and lacks protein-coding capacity. Metastasis-associated lung adenocarcinoma transcript 1 is the lncRNA of >8 kbp transcribed from chromosome 11q13. Recently, Lai et al127 reported that an overexpressed MALAT1 transcript could predict HCC recurrence after LT and importantly in those patients whose survival rate was also reduced. There are five SP-binding motifs upstream of the MALAT1, which lead to its overexpression,128 In addition, an in vitro study suggested an interaction of hn-RNP-C with MALAT1 regulating cell cycle as recently studied in HepG2 cells.129

HOTAIR

Hox antisense intergenic RNA plays a role in chromatin dynamics, cell differentiation, and cancer metastasis, which is encoded by HOXC gene cluster.130 Once transcribed, it acts in trans to control the HOXD genes by recruiting the polycomb repressive complex 2 and silencing the transcriptional machinery.131 Patients with elevated expression of HOTAIR shows poor prognosis compared to those with reduced HOTAIR expression. Yang et al132 documented that HOTAIR can act as an independent prognostic biomarker in predicting the HCC recurrence in patients undergoing LT.

H19

Oncofetal H19 mRNA is abundantly expressed in the fetus.133 It is paternally imprinted, which resides at chromosome 11p15.5 and is significantly expressed during tumorogenesis. Compared to AFP, the expression of H19 mRNA is much higher in HCC cases.134 It also inhibits metastasis by stimulating miRNA-200 and inducing histone acetylation.135

Highly upregulated in liver cancer

Highly upregulated in liver cancer is a 500 nucleotide lncRNA, which is expressed from chromosome 6p24.3136 loci. Compared to normal liver tissue, it is highly expressed in adjacent tumor part of the liver.137 One of the main agents responsible for HCC is HBV infection, particularly HBx. A recent study by Du et al138 documented an important role of highly upregulated in liver cancer in HBx-mediated HCC and further implicated that it is possible due to the downregulation of p18.

Long interspersed nuclear element-1

Retrotransposons are the jumping genes, and long interspersed nuclear element-1 (LINE-1) is one of the autonomously regulated retrotransposons. A recent study by Tangkijvanich et al139 tested the hypomethylation status of LINE-1 in 85 patients and concluded that advanced disease and tumor size are associated with levels of LINE-1 hypomethylation. Another report by Piao et al highlighted the role of LINE-1 hypomethylation in early childhood tumorogensis.140 Gao et al141 also precisely documented that hypomethylation at two of the sites, CpG 7 and 18, is associated with poor prognosis in HCC.

Micro-RNA biomarkers of HCC

Micro-RNA and its role as biomarkers

A micro-RNA (miRNA) is a small noncoding RNA molecule that mainly functions in RNA silencing.142 Transcription of miRNA initiates from intronic region of a host gene leading to long pri-miRNA transcript. This is further processed to pre-miRNA, Pre-miRNA then transport to cytoplasm by Exportin, where it goes through further modification by Dicer developing into complete miRNA molecule as given in Figure 8. miRNAs exhibit their silencing activities by base pairing with complementary sequences within mRNA molecules,143 by cleavage of the mRNA, chopping off its poly(A) tail, or less efficient translation of the mRNA into protein by ribosomes.144 The human genome encodes for over 1000 miRNAs,145 most of which are abundant in many mammalian cell types.146

Figure 8.

Figure 8

Biogenesis of miRNA. Modified from Anwar SL, Lehmann U. MicroRNAs: Emerging novel clinical biomarkers for hepatocellular carcinomas. Journal of Clinical Medicine. 2015;4(8):1631–1650, under the Creative Commons Attribution License.

Several studies indicate that miRNAs are involved in a variety of physiological processes, including cell proliferation, differentiation, metabolism, and apoptosis. As given in Figure 9, miRNAs regulate important cellular angiogenesis, apoptosis and metastasis pathways. The miRNA was recognized as a distinct class of biological regulators during early 2000s.147,148 Ongoing research has identified different miR-NAs expressed in different cells and tissues.146,149 miRNAs are detectable and stable in clinical samples, such as blood, serum, plasma, urine, and feces. Besides, the abnormal expression of miRNAs has been observed under different disease conditions, especially cancers, establishing miRNA as an important biomarker. Hence, new miRNA-based therapies are now under investigation.150153

Figure 9.

Figure 9

Mature miRNAs silenced by aberrant DNA methylation and their affected target genes and pathways that are important in the development and progression of HCC.

Abbreviations: PI3K, phosphatidylinositol-3-kinase; MAPK, mitogen-activated protein kinase; EGF, epidermal growth factor; PDGF, platelet-derived growth factor; HGF, hepatocyte growth factor; mTOR, mammalian target of rapamycin; VEGF, vascular endothelial growth factor; MMP, matrix metalloproteinase; APC, activated protein C; CTNNB1, β-catenin.

Role of miRNA biomarkers in HCC

miRNA dysregulation has been known to be associated with many cancers, the first studied being lymphocytic leukemia,154 and hence sometimes referred to as oncomirs. miRNA levels are also used as prognostic for cancers, as low miR-324 levels could serve as indicator of poor survival in non-small-cell lung carcinoma samples.155 High miR-185 or low miR-133b may correlate with metastasis and poor survival in colorectal cancer.156 miR-10b is implicated in the metastasis of breast cancer cells157 and the development of gastric cancer and pancreatic cancer.158,159

A number of studies have demonstrated extensive miRNA dysregulation in various stages of HCC.160 Furthermore, unique patterns of miRNA expression could be utilized as potential biomarkers for diagnosis, prognosis, staging, and prediction of therapeutic responses in HCC.161163 Expression of particular miRNAs tends to change gradually during the progression of HCC, and many tumor suppressor genes are demonstrated as the targets of the HCC oncomirs (eg, PTEN for miR-21, miR-221, and miR-222). Specific signaling pathways, such as Wnt/β-catenin, RAS, TGF-β, and JAK/STAT, are established targets for miRNA dysregulation in HCC.163

A comprehensive demonstration of the role of miRNA dysregulation and differential miRNA expression in HCC has been done.164,165 The advantage of miRNAs is that they are stable in frozen samples, formalin fixed paraffin embedded tissues, and body fluids, including plasma/serum, urine, and saliva. This property of miRNA makes them an excellent tool for early cancer diagnosis.

Primary tissue specimens. Basal miRNA expression has been studied using deep sequencing in primary HCC specimens as well as in the healthy liver.165,166 The most abundant miRNA expressed in healthy liver is miR-122, which is commonly downregulated in HCC. miR-199a/b is also downregulated in HCC and associated with poor survival.166

Serum. Serum levels of miR-122 are significantly upregulated in HCC patients compared to healthy individuals, and the levels are decreased after therapy.167 A case study reported by Li et al168 involving 500 serum samples from HCC patients showed that a combination of three miRNAs’ profile change, such as miR-25, miR-375, and let-7f, could distinguish between HCC and healthy controls. Serum levels of miR-16, miR-195, and miR-199a, alone or in combination, could distinguish between HCC and chronic hepatitis.169 Overexpression of miR-15b, miR-21, miR-130b, and miR-183 is demonstrated in 96 tumors, which indicate that these circulating miRNAs are derived from tumor cells.168 Lin et al170 reported very recently that the cluster of seven miRNAs (miR-29a, miR-29c, miR-133a, miR-143, miR-145, miR-192, and miR-505) can detect the HCC with better sensitivity and similar specificity than AFP. Another recent study demonstrated that low miR-150 level in HBV-related HCC patients was associated with a significantly decreased survival (P < 0.0001).161

Zhou et al171 reported that a particular miRNA panel was able to specifically distinguish HCC from healthy individuals and HBV and cirrhosis patients, using plasma samples from 934 HBV-associated HCC patients. For example, miR-106 could distinguish between HCC from healthy individuals and chronic hepatitis patients;172 while four miRNA panels (miT-20a-5p, miR-320a, miR-324-3p, and miR-375) have a high sensitivity and specificity to differentiate HCC from benign liver lesions.173 These findings clearly demonstrate that circulating miRNAs could be a potential diagnostic marker in HCC. As given in Figure 10, distinctive miRNAs are up-/down-regulated in hepatitis, fibrosis, steatosis and HCC, however miRNA-21, miRNA-122 and miRNA-223 are involved in Hepatitis as well as HCC.

Figure 10.

Figure 10

Summary of miRNAs and their targets associated with HCC, liver fibrosis, NAFLD, and hepatitis (HBV or HCV infection). miRNAs that are upregulated are indicated by red text, and miRNAs that are downregulated are indicated by green text.

miRNA as a prognostic tool in HCC

Other than their utility as a diagnostic biomarker, miRNAs are also important as prognostic tool, able to determine the tumor size, nodal, and metastasis stage of HCC, invasion, recurrence, and overall survival. miR-25 has been demonstrated to have a significant role in tumor size, nodal, and metastasis as reported by Su et al;174 upregulation of miR-183175 and miR-17-5p176 in primary HCC samples after surgery has been associated with large tumors and higher risk of metastasis. High miR-221 expression, along with downregulation of miR-100 and miR-22, is demonstrated in primary HCC tissues with distant metastasis.177179 miR-222 level is important to determine the advancement of the tumor.180 An overall miRNA profiling study demonstrated that upregulation of miR-25, miR-372, miR-155, and miR-182174,181183 strongly correlates with shorter survival time, whereas downregulation of miR-29a-5p, miR-100, miR-29, miR-101, and miR-148a in HCC tissues depicts the cure of the HCC and an increased survival.177,184186

miRNA as a therapeutic target

Currently, active research is going on to utilize these potential diagnostic and prognostic biomarkers as potential therapeutic targets to manage HCC. For therapy, antagonists are being developed against oncogenic miRNAs or oncomirs, referred to as antagomirs (locked nucleic acids). A very interesting example is of miravirsen (anti-miR-122) to treat chronic HCV infection, which is currently on phase III clinical trials.187 The success of the miRNA-based therapy would definitely lead to the better effectiveness of the current interferon therapy by the downregulation of the cell signaling pathway that generally renders the cells’ interferon resistant. It is reported that sorafenib, the drug used to treat HCC patients, regulates miRNA expression. Fourteen miRNAs are upregulated by sorafenib treatment. miR-122 is an important liver-specific miRNA that maintains liver homeostasis, so the delivery of miR-122 in HCV-infected patients is currently on clinical trials.188 miR-122, which is generally downregulated in HCC patients, is restored by sorafenib.189 Restoration of miR-122 by sorafenib also leads to an increased sensitivity of the tumors to other drugs, such as doxorubicin190 adriamycin, and vincristine.191 miR-26b has been shown to affect NF-κB signaling molecules to mediate chemosensitivity.192 Certain miRNAs when downregulated or upregulated induce drug resistance, hence they are targeted to develop their specific antagomirs to induce drug sensitivity for HCC treatment.

miRNA profiling and expression studies have tremendous potential for the development of new biomarkers, diagnostic and prognostic markers, as well as therapeutic molecules for the management of HCC. Hence, miRNAs are definitely the anticancer medicine for the future.

Tables 2A–C summarize the names and the roles of miR-NAs as diagnostic, prognostic, and therapeutic markers, respectively, in HCC.193

Table 2.

This table summarizes the list of miRNA from different sources, being upregulated or downregulated during HCC, and their roles in HCC progression. A. Daignostic markers. B. Prognostic markers C. Therapeutic markers.

microRNA REGULATION SOURCE INFORMATION
A. Diagnostic biomarkers
miR-106 Up Plasma Differentiate HCC from healthy control and chronic disease
miR-122 Up Serum Differentiate HCC from healthy control
miR-15b, miR-130b Up Serum Differentiate HCC from healthy control
miR-16, miR-199a Down Serum Differentiate HCC from healthy control and Chronic hepatitis
miR-183 Up Tissue Differentiate benign and malignant liver tumor
miR-15b, miR-130b Up Serum Differentiate HCC and healthy patients and reduce after surgery
miR-18a Up Serum Differentiate HCC from healthy control
miR-122, miR-192, miR-21, miR-223, miR-26a, miR-27a, miR801- Signature Plasma Differentiate HCC from healthy control
miR-21 Up Serum, Plasma Differentiate HCC from cirrhosis and healthy controls
miR-375 Up Serum Differentiate HBV- and HCV-related HCC from healthy controls
miR-483 Up Plasma Differentiate HCC from healthy control
miR-618/miR-650 Up Urine Differentiate HCC and control
miR-885 Up Serum Differentiate HCC, cirrhosis and chronic liver patients from healthy controls
miR-92a Down Plasma Differentiate HCC from healthy control
miR-25, miR-375, let-7f Up Serum Differentiate HCC from healthy control
miR-20a-5p, miR-320a, miR-324-3p, miR-375 Up Plasma Differentiate HCC from non-cancerous lesions
miR-29a, miR-29c, miR-133a, miR-143, miR-145, miR-192, miR-505 Signature Serum Detect early stage HCC and AFP-negative HCC
B. Prognostic markers
miR-10b Up Tissue Poor prognosis
miR-122 Down Tissue Poor prognosis
miR-124 Down Tissue Poor prognosis and aggressive type
miR-135a Up Tissue Shorter overall survival and disease free survival
miR-139 Down Tissue Metastasis and poor prognosis
miR-155 Up Tissue Poor prognosis, recurrence, micro-vascular invasion
miR-182 Up Tissue Intrahepatic metastasis and poor prognosis
miR-199b-5p Down Tissue Shorter overall survival
miR-203 Up Tissue Better prognosis, longer survival
miR-21, miR-221 Up Tissue Tumor stage and poor prognosis
miR-22 Down Tissue Poor survival
miR-221 Up Tissue Poor survival
miR-29 Down Tissue Shorter disease free survival
miR-29a-5p Up Tissue Recurrence in early stage of HCC
miR-99a Down Tissue Shorter survival
Let-7g Down Tissue Poor survival
DLK1-DIO3 miRNA cluster Up Tissue Poor prognosis
C19MC microRNA cluster Up Tissue Poor clinico-pathological features, recurrence and shorter overall survival
miR-155, miR-15a, miR-432, miR-486-3p, miR-15b, miR-30b Up Tissue Recurrence-free survival
miR-19a, miR-886, miR-43, miR-486-3p, miR-15b, miR-30b Signature Tissue Overall survival and recurrent free survival
67 miRNA signature
miR signatures in tumor and non-tumor tissues
Signature
Signature
Tissue
Tissue
Differentiate recurrence after liver transplantation
Differentiate early and late recurrence
miR-326, miR-3677, miR-511-1, miR-511-2, miR-9-1, miR-9-2 Signature Tissue Negatively associated with overall survival
C. Therapeutic markers
miR-122 Down Cells, tissue Decreased sensitivity to doxorubicin
miR-122 Down Cells, tissue Decreased sensitivity to adriamycin, vincristin
miR-122 Down Cells, tissue Suppressed sensitivity to sorafenib
miR-146a Up Cells Suppressed sensitivity to interferon-α
miR-193a-3p Down Cells, tissue Resistance to 5-FU
miR-193b Up Cells, tissue Sensitivity to cisplatin
miR-199a-3p Down Cells, tissue Increased sensitivity to doxorubicin
miR-1247a Down Cells Resistance to sorafenib
miR-21 Up Cells, tissue Resistance to interferon-α/5-FU in HCC cells
miR-34a Down Cells, tissue Resistance to sorafenib
13 microRNA signature Signature Cells, tissue Multidrug resistance

Note: Taken from Ref. 193.

Exosomal miRNA as a biomarker of HCC

One of the recent studies by Sohn et al194 isolated exosomes from the serum of chronic hepatitis B (CHB), cirrhosis, and HCC patients and found elevated levels of miRNAs, such as miR-18a, miR-221, miR-222, and miR-224, in HCC patients compared to those with CHB or liver cirrhosis (Fig. 11), whereas serum levels of miR-101, miR-106b, miR-122, and miR-195 were lower in HCC patients compared to CHB patients.

Figure 11.

Figure 11

The distribution of upregulated exosomal miRNAs (miR-18a, miR-221, miR-222, and miR-224) in CHB patients, LC, and HCC. Reprinted by permission from Macmillan Publishers Ltd: Experimental & Molecular Medicine, Sohn W, Kim J, Kang SH. Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Exp Mol Med. 2015;47:e184. Copyright 2015.

Genetic Variants as Biomarkers

P53 A249S

AGG-to-AGT single-nucleotide transversion leads to change of codon from arginine to serine at position 249 of P53 gene and one of the important hotspot genetic variations in HCC.195 It is majorly detected in patients who are exposed to aflatoxin and HBV.196 Some recent investigations deciphered the mechanisms by which it leads to HCC development: (1) inactivation of p16INK4 gene by overmethylation of this gene197 and (2) regulation of genes that play a main role in controlling the cell cycle switch from G to S phases.198

HBV core promoter mutations

HBV mutations in the basal core promoter region are important etiological risk factors for severe liver disease and HCC.199,200 BCP 1762 and 1764 mutations identified as an independent risk factor of HCC.201,202 Using the plasma samples, two studies reported that these mutations have the high predictive power to reveal the predisposition to HCC and its development.203,204

HBV PreS1 mutations/truncations

One of the largest studies, while recruiting 11,582 HBV-infected patients, reported that HBV preS mutants are associated with high risk of HCC.205 T53C, PreS2 initiation codon mutation, PreS1 deletion, C7A, A2962G, C2964A, and C3116T were significantly associated with HCC.201,206 One of the effects of few such mutations/deletions is that it also alters the overlapping polymerase gene reading frame; for example, rtA181T mutation in polymerase gene leads to stop codon mutation sW172 stop in the surface region and is associated with high risk of HCC development in patients’ refractory to the nucleoside therapy.207 Due to PreS envelope protein mutations/deletions, high-level uneven expression of these proteins leads to endoplasmic reticulum stress, causing genomic instability resulting in HCC.208

HBx region mutations and truncations

There are two main mechanisms by which HBV causes HCC, ie, direct and indirect. In direct mechanism, it integrates into the host liver cell genome and modulates several host genes in cis manner,209 and the indirect way is by trans activation of several genes by HBx protein.210 HBx protein in serum has been shown to be one of the markers of the liver cirrhosis and liver cancer in HBV-infected HCC.211 HBx induces AFP and promotes malignant transformation of hepatocytes by activating PI3K/mTOR signaling.212

Genetic Variations Identified by Next-generation Sequencing

Accumulation of genetic alterations is one of the most important mechanisms of causing HCC. Next-generation sequencing (NGS) through whole genome, whole exome, and whole transcriptome approaches has lead to revolutionary studies. A study by Ley et al213 reporting NGS, first described NGS as an unbiased method discovering cancer-initiating mutations. HBV integration into the host liver genome is considered as a direct way through which HBV causes HCC. Through NGS, three recent reports described HBV integration sites in the host liver genome. Using the whole genome sequencing, Jiang et al214 found 255 HBV integration sites in only three patients. In the second study by Sung et al,215 399 integrations sites were identified 75 out of 81 HCC patients. In the third study, Toh et al used 48 patients and, using the deep sequencing-mediated enrichment of HBV genomes, found that HBV integrates significantly in higher frequency in the regulatory regions of the host genes. It was reported that one of the important genes having recurrent HBV integration was TERT gene.216 A recent study also found important TERT promoter mutations in 60% of HCC patients, and other mutated genes identified were P53, CTNNB1, and ARID1 genes.217 Although not by NGS, but by clonal sequencing, our recent in-vitro study demonstrated that hepatitis B virus can integrate in the host genome immediately after infection, and the same was proved in in vivo study by infecting Woodchucks with Woodchuck hepatitis virus.218 Such HBV-host integration junctions possibly have the potential to act as very early molecular biomarkers of HBV related hepatocellular dysregulation. However, the question of whether they can act as a serum biomarker (fused virus-host protein secretion) will need further work and validations. Moreover, it will be interesting to discover, if such virus-host fused, secreted, properly folded proteins indeed play a role in the development of hepatocarcinoma and could be used as a prognostic biomarker or a therapeutic target. Using the whole-exome sequencing, a recent study for the first time identified recurrent alterations in four genes: AR1D1A, RPS6KA3, NFE2L2 and IRF2. In addition in liver tumors G > T transversion were significantly enriched in the non-transcribed DNA strand and highlighted their role in HCC.219 This study demonstrated alterations in genes responsible for activating the PI3K/Akt/mTOR pathway and an important mutation of mTOR at position S2215Y was reported.220

Summary

HCC, the neoplastic transformation of the hepatocytes, is one of the leading causes of cancer deaths around the world. People at risk of developing HCC include chronic liver disease patients with hepatitis B or C, obese or diabetic people, and heavy drinkers. HCC is developed as a consequence of chronic liver cirrhosis, where there is a decrease in hepatocyte growth and proliferation, along with scar tissue formation, that provides the platform for neoplastic tumor growth. Current methods of HCC management and treatment are more tumor oriented (chemotherapy using drugs, radiotherapy, surgical methods, such as liver resection and transplantation, and ethanol injection in the tumor cells). However, these methods are more of a palliative approach to HCC, aiming to extend the life span of patients rather than devising a curative approach. Therefore, due to the lack of a proper curative treatment of HCC, it is very important to prevent the onset of HCC or make attempts to detect the disease at a very early stage. The purpose of this review is to present before the readers, scientists, and medical professionals a detailed report of the ongoing research aiming at the discovery of biological tools for the prevention and prognosis of HCC. There are some biomolecules in the tissues and body fluids of humans whose levels change with the development of many abnormal conditions, various disease states, as well as carcinogenesis. These molecules, called biomarkers, are now regarded as important prognostic tools for early diagnosis of HCC. Active research is being conducted both at the basic and clinical levels to accurately detect the molecular targets, for developing drugs and vaccines to cure or prevent HCC. Many classes of biomarkers have been studied and detected in various stages of HCC (early and late), including peptides, glycoproteins, enzymes (soluble biomarkers), and mRNAs (nucleotide biomarkers), and can be obtained from liver tissue and serum of HCC patients. These biomolecules have also shown strong promise as molecular targets for the development of anti-metastatic vaccines or drugs. For example, some biomarkers, such as TAG-72, Golgi-localized phosphoprotein 73 (GP73), enzymes, such as α-1-fucosidase, and HGF, are overexpressed in HCC tissues and indicate poor survivability in patients. There are other markers, such as HCR2 enzyme, the glycoprotein APO-J/clusterin, that are reduced significantly during HCC progression and metastasis. From the start of the new millenium, miRNAs that are small non-coding RNA silencing molecules were recognized to be a new and distinct class of cancer biomarkers, especially in HCC. miRNAs that are effectively stable and easily extractable from tissues, plasma, serum, urine, and feces have shown great potential as prognostic tools and therapeutic targets in HCC.

This review has been documented to discuss the new research going on with an aim to harness the SOS signals of the human body, the biomarkers, to detect early and prevent HCC. The success of this research would bring a new era where not only HCC but also other cancers would no more be a death sentence.

Acknowledgments

Authors thank all biomarker investigators and scientists who are directly and indirectly associated with the present review. We have made an effort to accommodate most of the references; however, due to space constraints, all references could not be accommodated.

Abbreviations

CHB

chronic hepatitis B

HCC

hepatocellular carcinoma

HBV

hepatitis B virus

HCV

hepatitis C virus

LT

liver transplantation

OLT

orthotopic liver transplantation

TACE

transcatheter arterial chemoembolization

Footnotes

ACADEMIC EDITOR: Barbara Guinn, Editor in Chief

PEER REVIEW: Four peer reviewers contributed to the peer review report. Reviewers’ reports totaled 354 words, excluding any confidential comments to the academic editor.

FUNDING: RC thanks Canadian Institute of Health Research for providing funds for postdoctoral fellowship. The authors confirm that the funder had no influence over the study design, content of the article, or selection of this journal.

COMPETING INTERESTS: NL is a scientist at Pfizer. RC worked as a postdoctoral research fellow in a Canadian Institute of Health Research-funded project (MOP-14818) during inception and part of writing of this review. RC and NL have no roles in the discovery of any of the biomarker/s described in this review. Description of biomarkers given in this review is based on the recent developments in the field. The views are of authors, and there is no association or influence of any public or private company, institute, or person.

Paper subject to independent expert single-blind peer review. All editorial decisions made by independent academic editor. Upon submission manuscript was subject to anti-plagiarism scanning. Prior to publication all authors have given signed confirmation of agreement to article publication and compliance with all applicable ethical and legal requirements, including the accuracy of author and contributor information, disclosure of competing interests and funding sources, compliance with ethical requirements relating to human and animal study participants, and compliance with any copyright requirements of third parties. This journal is a member of the Committee on Publication Ethics (COPE).

Author Contributions

Wrote first draft of the manuscript: RC. Contributed to the writing of manuscript: RC, NL. Agree with manuscript results and conclusions: RC, NL. Jointly developed the structure and arguments for the paper: RC, NL. Both authors reviewed and approved of the final manuscript.

REFERENCES

  • 1.Bugianesi E. Non-alcoholic steatohepatitis and cancer. Clin Liver Dis. 2007;11:191–207. x–xi. doi: 10.1016/j.cld.2007.02.006. [DOI] [PubMed] [Google Scholar]
  • 2.Bosch FX, Ribes J, Diaz M, Cleries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16. doi: 10.1053/j.gastro.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 3.Delhaye M, Louis H, Degraef C, et al. Relationship between hepatocyte proliferative activity and liver functional reserve in human cirrhosis. Hepatology. 1996;23:1003–1011. doi: 10.1053/jhep.1996.v23.pm0008621125. [DOI] [PubMed] [Google Scholar]
  • 4.Caillot F, Derambure C, Bioulac-Sage P, et al. Transient and etiology-related transcription regulation in cirrhosis prior to hepatocellular carcinoma occurrence. World J Gastroenterol. 2009;15:300–309. doi: 10.3748/wjg.15.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wiemann SU, Satyanarayana A, Tsahuridu M, et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 2002;16:935–942. doi: 10.1096/fj.01-0977com. [DOI] [PubMed] [Google Scholar]
  • 6.Farazi PA, Glickman J, Jiang S, Yu A, Rudolph KL, DePinho RA. Differential impact of telomere dysfunction on initiation and progression of hepatocellular carcinoma. Cancer Res. 2003;63:5021–5027. [PubMed] [Google Scholar]
  • 7.Farazi PA, Glickman J, Horner J, Depinho RA. Cooperative interactions of p53 mutation, telomere dysfunction, and chronic liver damage in hepatocellular carcinoma progression. Cancer Res. 2006;66:4766–4773. doi: 10.1158/0008-5472.CAN-05-4608. [DOI] [PubMed] [Google Scholar]
  • 8.Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115:209–218. doi: 10.1172/JCI24282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Monga SP, Pediaditakis P, Mule K, Stolz DB, Michalopoulos GK. Changes in WNT/beta-catenin pathway during regulated growth in rat liver regeneration. Hepatology. 2001;33:1098–1109. doi: 10.1053/jhep.2001.23786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Monga SP. Role of Wnt/beta-catenin signaling in liver metabolism and cancer. Int J Biochem Cell Biol. 2011;43:1021–1029. doi: 10.1016/j.biocel.2009.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cormier JN, Thomas KT, Chari RS, Pinson CW. Management of hepatocellular carcinoma. J Gastrointest Surg. 2006;10:761–780. doi: 10.1016/j.gassur.2005.10.006. [DOI] [PubMed] [Google Scholar]
  • 12.Marelli L, Stigliano R, Triantos C, et al. Treatment outcomes for hepatocellular carcinoma using chemoembolization in combination with other therapies. Cancer Treat Rev. 2006;32:594–606. doi: 10.1016/j.ctrv.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 13.Salem R, Hunter RD. Yttrium-90 microspheres for the treatment of hepatocellular carcinoma: a review. Int J Radiat Oncol Biol Phys. 2006;66:S83–S88. doi: 10.1016/j.ijrobp.2006.02.061. [DOI] [PubMed] [Google Scholar]
  • 14.Zhu AX. Systemic therapy of advanced hepatocellular carcinoma: how hopeful should we be? Oncologist. 2006;11:790–800. doi: 10.1634/theoncologist.11-7-790. [DOI] [PubMed] [Google Scholar]
  • 15.Abou-Alfa GK, Schwartz L, Ricci S, et al. Phase II study of sorafenib in patients with advanced hepatocellular carcinoma. J Clin Oncol. 2006;24:293–4300. doi: 10.1200/JCO.2005.01.3441. [DOI] [PubMed] [Google Scholar]
  • 16.Liu L, Cao Y, Chen C, et al. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006;66:11851–11858. doi: 10.1158/0008-5472.CAN-06-1377. [DOI] [PubMed] [Google Scholar]
  • 17.Forner A, Hessheimer AJ, Isabel Real M, Bruix J. Treatment of hepatocellular carcinoma. Crit Rev Oncol Hematol. 2006;60:89–98. doi: 10.1016/j.critrevonc.2006.06.001. [DOI] [PubMed] [Google Scholar]
  • 18.Chen MB, Zhou ZT, Yang L, et al. KU-0060648 inhibits hepatocellular carconoma cells through DNA-PKcs dependent and DNA-PKcs-independent mechanisms. Oncotarget. 2016 doi: 10.18632/oncotarget.7742. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Augello G, Puleio R, Emma MR, et al. A PTEN inhibitor displays preclinical activity against hepatocarcinoma cells. Cell Cycle. 2016;15:573–583. doi: 10.1080/15384101.2016.1138183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chiang PC, Lin SC, Pan SL, et al. Antroquinonol displays anticancer potential against human hepatocellular carcinoma cells: a crucial role of AMPK and mTOR pathways. Biochem Pharmacol. 2010;79:162–171. doi: 10.1016/j.bcp.2009.08.022. [DOI] [PubMed] [Google Scholar]
  • 21.Malz M, Weber A, Singer S, et al. Overexpression of far upstream element-binding proteins: a mechanism regulating proliferation and migration of liver cancer cells. Hepatology. 2009;50:1130–1139. doi: 10.1002/hep.23051. [DOI] [PubMed] [Google Scholar]
  • 22.Rabenhorst U, Beinoraviciute-Kellner R, Brezniceanu ML, et al. Overexpression of the far upstream element binding protein-1 in hepatocellular carcinoma is required for tumor growth. Hepatology. 2009;50:1121–1129. doi: 10.1002/hep.23098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Samarin J, Laketa V, Malz M, et al. PI3K/AKT/mTOR-dependent stabilization of oncogenic far-upstream element binding proteins in hepatocellular carcinoma cells. Hepatology. 2016;63:813–826. doi: 10.1002/hep.28357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Niederau C, Fischer R, Purschel A, Stremmel W, Haussinger D, Strohmeyer G. Long-term survival in patients with hereditary hemochromatosis. Gastroenterology. 1996;110:1107–1119. doi: 10.1053/gast.1996.v110.pm8613000. [DOI] [PubMed] [Google Scholar]
  • 25.Aravalli RN, Steer CJ, Cressman EN. Molecular mechanisms of hepatocellular carcinoma. Hepatology. 2008;48:2047–2063. doi: 10.1002/hep.22580. [DOI] [PubMed] [Google Scholar]
  • 26.Zhu K, Dai Z, Pan Q, et al. Metadherin promotes hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition. Clin Cancer Res. 2011;17:7294–7302. doi: 10.1158/1078-0432.CCR-11-1327. [DOI] [PubMed] [Google Scholar]
  • 27.Sengupta B, Siddiqi SA. Hepatocellular carcinoma: important biomarkers and their significance in molecular diagnostics and therapy. Curr Med Chem. 2012;19:3722–3729. doi: 10.2174/092986712801661059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Shi GM, Ke AW, Zhou J, et al. CD151 modulates expression of matrix metal-loproteinase 9 and promotes neoangiogenesis and progression of hepatocellular carcinoma. Hepatology. 2010;52:183–196. doi: 10.1002/hep.23661. [DOI] [PubMed] [Google Scholar]
  • 29.Capurro M, Wanless IR, Sherman M, et al. Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology. 2003;125:89–97. doi: 10.1016/s0016-5085(03)00689-9. [DOI] [PubMed] [Google Scholar]
  • 30.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. doi: 10.1002/ijc.24941. [DOI] [PubMed] [Google Scholar]
  • 31.Yamauchi N, Watanabe A, Hishinuma M, et al. The glypican 3 oncofetal protein is a promising diagnostic marker for hepatocellular carcinoma. Mod Pathol. 2005;18:1591–1598. doi: 10.1038/modpathol.3800436. [DOI] [PubMed] [Google Scholar]
  • 32.Baeg GH, Perrimon N. Functional binding of secreted molecules to heparan sulfate proteoglycans in Drosophila. Curr Opin Cell Biol. 2000;12:575–580. doi: 10.1016/s0955-0674(00)00134-4. [DOI] [PubMed] [Google Scholar]
  • 33.Perrimon N, Bernfield M. Specificities of heparan sulphate proteoglycans in developmental processes. Nature. 2000;404:725–728. doi: 10.1038/35008000. [DOI] [PubMed] [Google Scholar]
  • 34.Capurro MI, Xu P, Shi W, Li F, Jia A, Filmus J. Glypican-3 inhibits Hedgehog signaling during development by competing with patched for Hedgehog binding. Dev Cell. 2008;14:700–711. doi: 10.1016/j.devcel.2008.03.006. [DOI] [PubMed] [Google Scholar]
  • 35.Lin X. Functions of heparan sulfate proteoglycans in cell signaling during development. Development. 2004;131:6009–6021. doi: 10.1242/dev.01522. [DOI] [PubMed] [Google Scholar]
  • 36.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. doi: 10.1158/0008-5472.CAN-04-4244. [DOI] [PubMed] [Google Scholar]
  • 37.Ogasawara S, Yano H, Iemura A, Hisaka T, Kojiro M. Expressions of basic fibroblast growth factor and its receptors and their relationship to proliferation of human hepatocellular carcinoma cell lines. Hepatology. 1996;24:198–205. doi: 10.1053/jhep.1996.v24.pm0008707262. [DOI] [PubMed] [Google Scholar]
  • 38.Ito Y, Takeda T, Higashiyama S, et al. Expression of heparin binding epidermal growth factor-like growth factor in hepatocellular carcinoma: an immunohisto-chemical study. Oncol Rep. 2001;8:903–907. doi: 10.3892/or.8.4.903. [DOI] [PubMed] [Google Scholar]
  • 39.Horiguchi N, Takayama H, Toyoda M, et al. Hepatocyte growth factor promotes hepatocarcinogenesis through c-Met autocrine activation and enhanced angio-genesis in transgenic mice treated with diethylnitrosamine. Oncogene. 2002;21:1791–1799. doi: 10.1038/sj.onc.1205248. [DOI] [PubMed] [Google Scholar]
  • 40.Jin X, Moskophidis D, Mivechi NF. Heat shock transcription factor 1 is a key determinant of HCC development by regulating hepatic steatosis and metabolic syndrome. Cell Metab. 2011;14:91–103. doi: 10.1016/j.cmet.2011.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fang F, Chang R, Yang L. Heat shock factor 1 promotes invasion and metastasis of hepatocellular carcinoma in vitro and in vivo. Cancer. 2012;118:1782–1794. doi: 10.1002/cncr.26482. [DOI] [PubMed] [Google Scholar]
  • 42.Liu CC, Jan YJ, Ko BS, et al. 14-3-3sigma induces heat shock protein 70 expression in hepatocellular carcinoma. BMC Cancer. 2014;14:425. doi: 10.1186/1471-2407-14-425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jin B, Wang X, Jin Y, et al. Detection of serum gastric cancer-associated MG7-Ag from gastric cancer patients using a sensitive and convenient ELISA method. Cancer Invest. 2009;27:227–233. doi: 10.1080/07357900802175609. [DOI] [PubMed] [Google Scholar]
  • 44.Chauhan SC, Vinayek N, Maher DM, et al. Combined staining of TAG-72, MUC1, and CA125 improves labeling sensitivity in ovarian cancer: antigens for multi-targeted antibody-guided therapy. J Histochem Cytochem. 2007;55:867–875. doi: 10.1369/jhc.7A7213.2007. [DOI] [PubMed] [Google Scholar]
  • 45.Santos-Juanes J, Bernaldo de Quiros JF, Galache Osuna C, et al. Apocrine carcinoma, adenopathies, and raised TAG-72 serum tumor marker. Dermatol Surg. 2004;30:566–569. doi: 10.1111/j.1524-4725.2004.30180.x. [DOI] [PubMed] [Google Scholar]
  • 46.Zhang Y, Deng ZS, Liao MM, et al. Tumor associated glycoprotein-72 is a novel marker for poor survival in hepatocellular carcinoma. Pathol Oncol Res. 2012;18:911–916. doi: 10.1007/s12253-012-9521-0. [DOI] [PubMed] [Google Scholar]
  • 47.Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182:311–322. doi: 10.1002/(SICI)1097-4652(200003)182:3<311::AID-JCP1>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 48.Mohamed WS, Omar MM, Khayri TM, Fakhr IM. Assessment of the proliferative marker Ki-67 and p53 protein expression in HBV- and HCV-related hepatocellular carcinoma cases in Egypt. Int J Health Sci (Qassim) 2008;2:27–34. [PMC free article] [PubMed] [Google Scholar]
  • 49.Wennerberg AE, Nalesnik MA, Coleman WB. Hepatocyte paraffin 1: a monoclonal antibody that reacts with hepatocytes and can be used for differential diagnosis of hepatic tumors. Am J Pathol. 1993;143:1050–1054. [PMC free article] [PubMed] [Google Scholar]
  • 50.Fasano M, Theise ND, Nalesnik M, et al. Immunohistochemical evaluation of hepatoblastomas with use of the hepatocyte-specific marker, hepatocyte paraffin 1, and the polyclonal anti-carcinoembryonic antigen. Mod Pathol. 1998;11:934–938. [PubMed] [Google Scholar]
  • 51.Maitra A, Murakata LA, Albores-Saavedra J. Immunoreactivity for hepatocyte paraffin 1 antibody in hepatoid adenocarcinomas of the gastrointestinal tract. Am J Clin Pathol. 2001;115:689–694. doi: 10.1309/5C2C-FP3H-GE7Q-2XJ5. [DOI] [PubMed] [Google Scholar]
  • 52.Murakata LA, Ishak KG, Nzeako UC. Clear cell carcinoma of the liver: a comparative immunohistochemical study with renal clear cell carcinoma. Mod Pathol. 2000;13:874–881. doi: 10.1038/modpathol.3880156. [DOI] [PubMed] [Google Scholar]
  • 53.Lugli A, Tornillo L, Mirlacher M, Bundi M, Sauter G, Terracciano LM. Hepatocyte paraffin 1 expression in human normal and neoplastic tissues: tissue microarray analysis on 3,940 tissue samples. Am J Clin Pathol. 2004;122:721–727. doi: 10.1309/KC09-YTF2-M4DL-UYQ6. [DOI] [PubMed] [Google Scholar]
  • 54.Lau SK, Prakash S, Geller SA, Alsabeh R. Comparative immunohistochemical profile of hepatocellular carcinoma, cholangiocarcinoma, and metastatic adenocarcinoma. Hum Pathol. 2002;33:1175–1181. doi: 10.1053/hupa.2002.130104. [DOI] [PubMed] [Google Scholar]
  • 55.Villari D, Caruso R, Grosso M, Vitarelli E, Righi M, Barresi G. Hep Par 1 in gastric and bowel carcinomas: an immunohistochemical study. Pathology. 2002;34:423–426. doi: 10.1080/0031302021000009333. [DOI] [PubMed] [Google Scholar]
  • 56.Zimmerman RL, Burke MA, Young NA, Solomides CC, Bibbo M. Diagnostic value of hepatocyte paraffin 1 antibody to discriminate hepatocellular carcinoma from metastatic carcinoma in fine-needle aspiration biopsies of the liver. Cancer. 2001;93:288–291. doi: 10.1002/cncr.9043. [DOI] [PubMed] [Google Scholar]
  • 57.Kakar S, Muir T, Murphy LM, Lloyd RV, Burgart LJ. Immunoreactivity of Hep Par 1 in hepatic and extrahepatic tumors and its correlation with albumin in situ hybridization in hepatocellular carcinoma. Am J Clin Pathol. 2003;119:361–366. doi: 10.1309/8l872rphejrkf5jj. [DOI] [PubMed] [Google Scholar]
  • 58.Pitman MB, Triratanachat S, Young RH, Oliva E. Hepatocyte paraffin 1 antibody does not distinguish primary ovarian tumors with hepatoid differentiation from metastatic hepatocellular carcinoma. Int J Gynecol Pathol. 2004;23:58–64. doi: 10.1097/01.pgp.0000101141.31270.a1. [DOI] [PubMed] [Google Scholar]
  • 59.Xue F, Higgs BW, Huang J, et al. HERC5 is a prognostic biomarker for post-liver transplant recurrent human hepatocellular carcinoma. J Transl Med. 2015;13:379. doi: 10.1186/s12967-015-0743-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bernassola F, Karin M, Ciechanover A, Melino G. The HECT family of E3 ubiquitin ligases: multiple players in cancer development. Cancer Cell. 2008;14:10–21. doi: 10.1016/j.ccr.2008.06.001. [DOI] [PubMed] [Google Scholar]
  • 61.Chen DS, Sung JL, Sheu JC, et al. Serum alpha-fetoprotein in the early stage of human hepatocellular carcinoma. Gastroenterology. 1984;86:1404–1409. [PubMed] [Google Scholar]
  • 62.Wang CS, Lin CL, Lee HC, et al. Usefulness of serum des-gamma-carboxy pro-thrombin in detection of hepatocellular carcinoma. World J Gastroenterol. 2005;11:6115–6119. doi: 10.3748/wjg.v11.i39.6115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cheng J, Wang W, Zhang Y, et al. Prognostic role of pre-treatment serum AFP-L3% in hepatocellular carcinoma: systematic review and meta-analysis. PLoS One. 2014;9:e87011. doi: 10.1371/journal.pone.0087011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Guzman G, Alagiozian-Angelova V, Layden-Almer JE, et al. p53, Ki-67, and serum alpha feto-protein as predictors of hepatocellular carcinoma recurrence in liver transplant patients. Mod Pathol. 2005;18:1498–1503. doi: 10.1038/modpathol.3800458. [DOI] [PubMed] [Google Scholar]
  • 65.Gorog D, Regoly-Merei J, Paku S, Kopper L, Nagy P. Alpha-fetoprotein expression is a potential prognostic marker in hepatocellular carcinoma. World J Gastroenterol. 2005;11:5015–5018. doi: 10.3748/wjg.v11.i32.5015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Norton PA, Comunale MA, Krakover J, et al. N-linked glycosylation of the liver cancer biomarker GP73. J Cell Biochem. 2008;104:136–149. doi: 10.1002/jcb.21610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Block TM, Comunale MA, Lowman M, et al. Use of targeted glycoproteomics to identify serum glycoproteins that correlate with liver cancer in woodchucks and humans. Proc Natl Acad Sci U S A. 2005;102:779–784. doi: 10.1073/pnas.0408928102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Block T, Mehta AS, London WT. Hepatocellular carcinoma of the liver. Cancer Biomark. 2010;9:375–383. doi: 10.3233/CBM-2011-0165. [DOI] [PubMed] [Google Scholar]
  • 69.Kladney RD, Bulla GA, Guo L, et al. GP73, a novel Golgi-localized protein upregulated by viral infection. Gene. 2000;249:53–65. doi: 10.1016/S0378-1119(00)00136-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Liang H, Block TM, Wang M, et al. Interleukin-6 and oncostatin M are elevated in liver disease in conjunction with candidate hepatocellular carcinoma biomarker GP73. Cancer Biomark. 2012;11:161–171. doi: 10.3233/CBM-2012-00276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kristiansen G, Fritzsche FR, Wassermann K, et al. GOLPH2 protein expression as a novel tissue biomarker for prostate cancer: implications for tissue-based diagnostics. Br J Cancer. 2008;99:939–948. doi: 10.1038/sj.bjc.6604614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Comunale MA, Wang M, Rodemich-Betesh L, et al. Novel changes in glycosylation of serum Apo-J in patients with hepatocellular carcinoma. Cancer Epidemiol Biomarkers Prev. 2011;20:1222–1229. doi: 10.1158/1055-9965.EPI-10-1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang Y, Liu YH, Mai SJ, et al. Evaluation of serum clusterin as a surveillance tool for human hepatocellular carcinoma with hepatitis B virus related cirrhosis. J Gastroenterol Hepatol. 2010;25:1123–1128. doi: 10.1111/j.1440-1746.2009.06205.x. [DOI] [PubMed] [Google Scholar]
  • 74.Nafee AM, Pasha HF, Abd El, Aal SM, Mostafa NA. Clinical significance of serum clusterin as a biomarker for evaluating diagnosis and metastasis potential of viral-related hepatocellular carcinoma. Clin Biochem. 2012;45:1070–1074. doi: 10.1016/j.clinbiochem.2012.04.024. [DOI] [PubMed] [Google Scholar]
  • 75.Zheng W, Yao M, Sai W, et al. Diagnostic and prognostic significance of secretory clusterin expression in patients with hepatocellular carcinoma. Tumour Biol. 2015:1–12. doi: 10.1007/s13277-015-3875-3. [DOI] [PubMed] [Google Scholar]
  • 76.Shen Q, Fan J, Yang XR, et al. Serum DKK1 as a protein biomarker for the diagnosis of hepatocellular carcinoma: a large-scale, multicentre study. Lancet Oncol. 2012;13:817–826. doi: 10.1016/S1470-2045(12)70233-4. [DOI] [PubMed] [Google Scholar]
  • 77.Tsai JF, Jeng JE, Chuang WL. Dickkopf-1 and hepatocellular carcinoma. Lancet Oncol. 2012;13:e410. doi: 10.1016/S1470-2045(12)70416-3. author reply e410–411. [DOI] [PubMed] [Google Scholar]
  • 78.Zhu K, Dai Z, Zhou J. Biomarkers for hepatocellular carcinoma: progression in early diagnosis, prognosis, and personalized therapy. Biomark Res. 2013;1:10. doi: 10.1186/2050-7771-1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wang C, Qi R, Li N, et al. Notch1 signaling sensitizes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in human hepatocellular carcinoma cells by inhibiting Akt/Hdm2-mediated p53 degradation and up-regulating p53-dependent DR5 expression. J Biol Chem. 2009;284:16183–16190. doi: 10.1074/jbc.M109.002105. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 80.Liu S, Ma L, Huang W, et al. Decreased expression of the human carbonyl reductase 2 gene HCR2 in hepatocellular carcinoma. Cell Mol Biol Lett. 2006;11:230–241. doi: 10.2478/s11658-006-0022-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhu WW, Guo JJ, Guo L, et al. Evaluation of midkine as a diagnostic serum biomarker in hepatocellular carcinoma. Clin Cancer Res. 2013;19:3944–3954. doi: 10.1158/1078-0432.CCR-12-3363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nault JC. Molecular determinants of prognosis in hepatocellular carcinoma. J Clin Transl Hepatol. 2014;2:31–36. doi: 10.14218/JCTH.2013.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Marrero JA, Feng Z, Wang Y, et al. Alpha-fetoprotein, des-gamma carboxypro-thrombin, and lectin-bound alpha-fetoprotein in early hepatocellular carcinoma. Gastroenterology. 2009;137:110–118. doi: 10.1053/j.gastro.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Murata K, Suzuki H, Okano H, Oyamada T, Yasuda Y, Sakamoto A. Hypoxia-induced des-gamma-carboxy prothrombin production in hepatocellular carcinoma. Int J Oncol. 2010;36:161–170. [PubMed] [Google Scholar]
  • 85.Wang K, Guo W, Li N, et al. Alpha-1-fucosidase as a prognostic indicator for hepatocellular carcinoma following hepatectomy: a large-scale, long-term study. Br J Cancer. 2014;110:1811–1819. doi: 10.1038/bjc.2014.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tangkijvanich P, Tosukhowong P, Bunyongyod P, et al. Alpha-L-fucosidase as a serum marker of hepatocellular carcinoma in Thailand. Southeast Asian J Trop Med Public Health. 1999;30:110–114. [PubMed] [Google Scholar]
  • 87.Nakamura S, Muro H, Suzuki S, et al. Immunohistochemical studies on endothelial cell phenotype in hepatocellular carcinoma. Hepatology. 1997;26:407–415. doi: 10.1002/hep.510260222. [DOI] [PubMed] [Google Scholar]
  • 88.Tewari M, Mohn KL, Yue FE, Taub R. Sequence of rat RL/IF-1 encoding an IkappaB, and comparison with related proteins containing notch-like repeats [corrected] Nucleic Acids Res. 1992;20:607. doi: 10.1093/nar/20.3.607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yamagamim H, Moriyama M, Matsumura H, et al. Serum concentrations of human hepatocyte growth factor is a useful indicator for predicting the occurrence of hepatocellular carcinomas in C-viral chronic liver diseases. Cancer. 2002;95:824–834. doi: 10.1002/cncr.10732. [DOI] [PubMed] [Google Scholar]
  • 90.Mizuguchi T, Katsuramaki T, Nobuoka T, et al. Serum hyaluronate level for predicting subclinical liver dysfunction after hepatectomy. World J Surg. 2004;28:971–976. doi: 10.1007/s00268-004-7389-1. [DOI] [PubMed] [Google Scholar]
  • 91.Levi-Montalcini R. The nerve growth factor 35 years later. Science. 1987;237:1154–1162. doi: 10.1126/science.3306916. [DOI] [PubMed] [Google Scholar]
  • 92.Rasi G, Serafino A, Bellis L, et al. Nerve growth factor involvement in liver cirrhosis and hepatocellular carcinoma. World J Gastroenterol. 2007;13:4986–4995. doi: 10.3748/wjg.v13.i37.4986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Treiber G, Wex T, Rocken C, Fostitsch P, Malfertheiner P. Impact of biomarkers on disease survival and progression in patients treated with octreotide for advanced hepatocellular carcinoma. J Cancer Res Clin Oncol. 2006;132:699–708. doi: 10.1007/s00432-006-0118-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhu AX, Finn RS, Mulcahy M, et al. 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. doi: 10.1158/1078-0432.CCR-13-1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Poon RT, Ho JW, Tong CS, Lau C, Ng IO, Fan ST. Prognostic significance of serum vascular endothelial growth factor and endostatin in patients with hepatocellular carcinoma. Br J Surg. 2004;91:1354–1360. doi: 10.1002/bjs.4594. [DOI] [PubMed] [Google Scholar]
  • 96.Llovet JM, Pena CE, Lathia CD, et al. Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2290–2300. doi: 10.1158/1078-0432.CCR-11-2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Bedossa P, Peltier E, Terris B, Franco D, Poynard T. Transforming growth factor-beta 1 (TGF-beta 1) and TGF-beta 1 receptors in normal, cirrhotic, and neoplastic human livers. Hepatology. 1995;21:760–766. [PubMed] [Google Scholar]
  • 98.Liu ZM, Tseng HY, Tsai HW, Su FC, Huang HS. Transforming growth factor beta-interacting factor-induced malignant progression of hepatocellular carcinoma cells depends on superoxide production from Nox4. Free Radic Biol Med. 2015;84:54–64. doi: 10.1016/j.freeradbiomed.2015.03.028. [DOI] [PubMed] [Google Scholar]
  • 99.Li Q, Liu G, Shao D, et al. Mucin1 mediates autocrine transforming growth factor beta signaling through activating the c-Jun N-terminal kinase/activator protein 1 pathway in human hepatocellular carcinoma cells. Int J Biochem Cell Biol. 2015;59:116–125. doi: 10.1016/j.biocel.2014.11.012. [DOI] [PubMed] [Google Scholar]
  • 100.Berasain C, Avila MA. The EGFR signalling system in the liver: from hepatoprotection to hepatocarcinogenesis. J Gastroenterol. 2014;49:9–23. doi: 10.1007/s00535-013-0907-x. [DOI] [PubMed] [Google Scholar]
  • 101.Hsieh SY, He JR, Yu MC, et al. Secreted ERBB3 isoforms are serum markers for early hepatoma in patients with chronic hepatitis and cirrhosis. J Proteome Res. 2011;10:4715–4724. doi: 10.1021/pr200519q. [DOI] [PubMed] [Google Scholar]
  • 102.Chen YJ, Chien PH, Chen WS, et al. Hepatitis B virus-encoded X protein downregulates EGFR expression via inducing microRNA-7 in hepatocellular carcinoma cells. Evidence Based Complement Alternat Med. 2013;2013:10. doi: 10.1155/2013/682380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lanaya H, Natarajan A, Komposch K, et al. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nat Cell Biol. 2014;16(972–981):971–977. doi: 10.1038/ncb3031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hu H, Gao L, Wang C, et al. Lower serum soluble-EGFR is a potential biomarker for metastasis of HCC demonstrated by N-glycoproteomic analysis. Discov Med. 2015;19:333–341. [PubMed] [Google Scholar]
  • 105.Lee HH, Uen YH, Tian YF, et al. Wnt-1 protein as a prognostic biomarker for hepatitis B-related and hepatitis C-related hepatocellular carcinoma after surgery. Cancer Epidemiol Biomarkers Prev. 2009;18:1562–1569. doi: 10.1158/1055-9965.EPI-09-0039. [DOI] [PubMed] [Google Scholar]
  • 106.Ho M, Kim H. Glypican-3: a new target for cancer immunotherapy. Eur J Cancer. 2011;47:333–338. doi: 10.1016/j.ejca.2010.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Scholz A, Rehm VA, Rieke S, et al. Angiopoietin-2 serum levels are elevated in patients with liver cirrhosis and hepatocellular carcinoma. Am J Gastroenterol. 2007;102:2471–2481. doi: 10.1111/j.1572-0241.2007.01377.x. [DOI] [PubMed] [Google Scholar]
  • 108.Diaz-Sanchez A, Matilla A, Nunez O, et al. Serum angiopoietin-2 level as a predictor of tumor invasiveness in patients with hepatocellular carcinoma. Scand J Gastroenterol. 2013;48:334–343. doi: 10.3109/00365521.2012.746391. [DOI] [PubMed] [Google Scholar]
  • 109.David S, Mukherjee A, Ghosh CC, et al. Angiopoietin-2 may contribute to multiple organ dysfunction and death in sepsis*. Crit Care Med. 2012;40:3034–3041. doi: 10.1097/CCM.0b013e31825fdc31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wang F, Zhou H, Xia X, Sun Q, Wang Y, Cheng B. Activated Notch signaling is required for hepatitis B virus X protein to promote proliferation and survival of human hepatic cells. Cancer Lett. 2010;298:64–73. doi: 10.1016/j.canlet.2010.06.003. [DOI] [PubMed] [Google Scholar]
  • 111.Villanueva A, Alsinet C, Yanger K, et al. Notch signaling is activated in human hepatocellular carcinoma and induces tumor formation in mice. Gastroenterology. 2012;143(1660–1669):e1667. doi: 10.1053/j.gastro.2012.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sun Q, Wang R, Wang Y, Luo J, Wang P, Cheng B. Notch1 is a potential therapeutic target for the treatment of human hepatitis B virus X protein-associated hepatocellular carcinoma. Oncol Rep. 2014;31:933–939. doi: 10.3892/or.2013.2917. [DOI] [PubMed] [Google Scholar]
  • 113.Ahn S, Hyeon J, Park CK. Notch1 and Notch4 are markers for poor prognosis of hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int. 2013;12:286–294. doi: 10.1016/s1499-3872(13)60046-6. [DOI] [PubMed] [Google Scholar]
  • 114.Wu T, Jiao M, Jing L, et al. Prognostic value of Notch-1 expression in hepatocellular carcinoma: a meta-analysis. Onco Targets Ther. 2015;8:3105–3114. doi: 10.2147/OTT.S92945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rose TM, Bruce AG. Oncostatin M is a member of a cytokine family that includes leukemia-inhibitory factor, granulocyte colony-stimulating factor, and interleukin 6. Proc Natl Acad Sci U S A. 1991;88:8641–8645. doi: 10.1073/pnas.88.19.8641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kinoshita T, Sekiguchi T, Xu MJ, et al. Hepatic differentiation induced by oncostatin M attenuates fetal liver hematopoiesis. Proc Natl Acad Sci U S A. 1999;96:7265–7270. doi: 10.1073/pnas.96.13.7265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Blum HE. Molecular targets for prevention of hepatocellular carcinoma. Dig Dis. 2002;20:81–90. doi: 10.1159/000063163. [DOI] [PubMed] [Google Scholar]
  • 118.Morgan K, Kalsheker NA. Regulation of the serine proteinase inhibitor (SER-PIN) gene alpha 1-antitrypsin: a paradigm for other SERPINs. Int J Biochem Cell Biol. 1997;29:1501–1511. doi: 10.1016/s1357-2725(97)00118-0. [DOI] [PubMed] [Google Scholar]
  • 119.Comunale MA, Rodemich-Betesh L, Hafner J, et al. Linkage specific fucosylation of alpha-1-antitrypsin in liver cirrhosis and cancer patients: implications for a biomarker of hepatocellular carcinoma. PLoS One. 2010;5:e12419. doi: 10.1371/journal.pone.0012419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wang M, Long RE, Comunale MA, et al. Novel fucosylated biomarkers for the early detection of hepatocellular carcinoma. Cancer Epidemiol Biomarkers Prev. 2009;18:1914–1921. doi: 10.1158/1055-9965.EPI-08-0980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kuno A, Ikehara Y, Tanaka Y, et al. A serum “sweet-doughnut” protein facilitates fibrosis evaluation and therapy assessment in patients with viral hepatitis. Sci Rep. 2013;3:1065. doi: 10.1038/srep01065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yamasaki K, Tateyama M, Abiru S, et al. Elevated serum levels of Wisteria floribunda agglutinin-positive human Mac-2 binding protein predict the development of hepatocellular carcinoma in hepatitis C patients. Hepatology. 2014;60:1563–1570. doi: 10.1002/hep.27305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Sasaki R, Yamasaki K, Abiru S, et al. Serum wisteria floribunda agglutinin-positive mac-2 binding protein values predict the development of hepatocellular carcinoma among patients with chronic hepatitis C after sustained virological response. PLoS One. 2015;10:e0129053. doi: 10.1371/journal.pone.0129053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wolf MJ, Seleznik GM, Zeller N, Heikenwalder M. The unexpected role of lymphotoxin beta receptor signaling in carcinogenesis: from lymphoid tissue formation to liver and prostate cancer development. Oncogene. 2010;29:5006–5018. doi: 10.1038/onc.2010.260. [DOI] [PubMed] [Google Scholar]
  • 125.Haybaeck J, Zeller N, Wolf MJ, et al. A lymphotoxin-driven pathway to hepatocellular carcinoma. Cancer Cell. 2009;16:295–308. doi: 10.1016/j.ccr.2009.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Scarzello AJ, Jiang Q, Back T, et al. LT βR signalling preferentially accelerates oncogenic AKT-initiated liver tumours. Gut. 2015 doi: 10.1136/gutjnl-2014-308810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lai MC, Yang Z, Zhou L, et al. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 2012;29:1810–1816. doi: 10.1007/s12032-011-0004-z. [DOI] [PubMed] [Google Scholar]
  • 128.Huang Z, Huang L, Shen S, et al. Sp1 cooperates with Sp3 to upregulate MALAT1 expression in human hepatocellular carcinoma. Oncol Rep. 2015;34:2403–2412. doi: 10.3892/or.2015.4259. [DOI] [PubMed] [Google Scholar]
  • 129.Liu WT, Lu X, Tang GH, et al. LncRNAs expression signatures of hepatocellular carcinoma revealed by microarray. World J Gastroenterol. 2014;20:6314–6321. doi: 10.3748/wjg.v20.i20.6314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Bhan A, Mandal SS. LncRNA HOTAIR: a master regulator of chromatin dynamics and cancer. Biochim Biophys Acta. 2015;1856:151–164. doi: 10.1016/j.bbcan.2015.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Cai B, Song XQ, Cai JP, Zhang S. HOTAIR: a cancer-related long non-coding RNA. Neoplasma. 2014;61:379–391. doi: 10.4149/neo_2014_075. [DOI] [PubMed] [Google Scholar]
  • 132.Yang Z, Zhou L, Wu LM, et al. Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann Surg Oncol. 2011;18:1243–1250. doi: 10.1245/s10434-011-1581-y. [DOI] [PubMed] [Google Scholar]
  • 133.Gabory A, Jammes H, Dandolo L. The H19 locus: role of an imprinted non-coding RNA in growth and development. Bioessays. 2010;32:473–480. doi: 10.1002/bies.200900170. [DOI] [PubMed] [Google Scholar]
  • 134.Shi X, Sun M, Liu H, Yao Y, Song Y. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett. 2013;339:159–166. doi: 10.1016/j.canlet.2013.06.013. [DOI] [PubMed] [Google Scholar]
  • 135.Zhang L, Yang F, Yuan JH, et al. Epigenetic activation of the MiR-200 family contributes to H19-mediated metastasis suppression in hepatocellular carcinoma. Carcinogenesis. 2013;34:577–586. doi: 10.1093/carcin/bgs381. [DOI] [PubMed] [Google Scholar]
  • 136.Zhao Y, Guo Q, Chen J, Hu J, Wang S, Sun Y. Role of long non-coding RNA HULC in cell proliferation, apoptosis and tumor metastasis of gastric cancer: a clinical and in vitro investigation. Oncol Rep. 2014;31:358–364. doi: 10.3892/or.2013.2850. [DOI] [PubMed] [Google Scholar]
  • 137.Panzitt K, Tschernatsch MM, Guelly C, et al. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330–342. doi: 10.1053/j.gastro.2006.08.026. [DOI] [PubMed] [Google Scholar]
  • 138.Du Y, Kong G, You X, et al. Elevation of highly up-regulated in liver cancer (HULC) by hepatitis B virus X protein promotes hepatoma cell proliferation via down-regulating p18. J Biol Chem. 2012;287:26302–26311. doi: 10.1074/jbc.M112.342113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Tangkijvanich P, Hourpai N, Rattanatanyong P, Wisedopas N, Mahachai V, Mutirangura A. Serum LINE-1 hypomethylation as a potential prognostic marker for hepatocellular carcinoma. Clin Chim Acta. 2007;379:127–133. doi: 10.1016/j.cca.2006.12.029. [DOI] [PubMed] [Google Scholar]
  • 140.Piao W, Wang W, Zhang H, et al. Hypomethylation of long interspersed nuclear element-1 is involved in the early tumorigenesis of hepatocellular carcinoma. J Interdiscipl Histopathol. 2014;2(4):191–196. [Google Scholar]
  • 141.Gao XD, Qu JH, Chang XJ, et al. Hypomethylation of long interspersed nuclear element-1 promoter is associated with poor outcomes for curative resected hepatocellular carcinoma. Liver Int. 2014;34:136–146. doi: 10.1111/liv.12264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–355. doi: 10.1038/nature02871. [DOI] [PubMed] [Google Scholar]
  • 143.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–379. doi: 10.1146/annurev-biochem-060308-103103. [DOI] [PubMed] [Google Scholar]
  • 145.Bentwich I, Avniel A, Karov Y, et al. Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet. 2005;37:766–770. doi: 10.1038/ng1590. [DOI] [PubMed] [Google Scholar]
  • 146.Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol. 2002;12:735–739. doi: 10.1016/s0960-9822(02)00809-6. [DOI] [PubMed] [Google Scholar]
  • 147.Reinhart BJ, Slack FJ, Basson M, et al. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000;403:901–906. doi: 10.1038/35002607. [DOI] [PubMed] [Google Scholar]
  • 148.Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294:858–862. doi: 10.1126/science.1065062. [DOI] [PubMed] [Google Scholar]
  • 149.Wienholds E, Kloosterman WP, Miska E, et al. MicroRNA expression in zebrafish embryonic development. Science. 2005;309:310–311. doi: 10.1126/science.1114519. [DOI] [PubMed] [Google Scholar]
  • 150.Li C, Feng Y, Coukos G, Zhang L. Therapeutic microRNA strategies in human cancer. AAPS J. 2009;11:747–757. doi: 10.1208/s12248-009-9145-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Trang P, Weidhaas JB, Slack FJ. MicroRNAs as potential cancer therapeutics. Oncogene. 2008;27(suppl 2):S52–S57. doi: 10.1038/onc.2009.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Hydbring P, Badalian-Very G. Gayane clinical applications of microRNAs. F1000Res. 2013;2:136. doi: 10.12688/f1000research.2-136.v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Fasanaro P, Greco S, Ivan M, Capogrossi MC, Martelli F. microRNA: emerging therapeutic targets in acute ischemic diseases. Pharmacol Ther. 2010;125:92–104. doi: 10.1016/j.pharmthera.2009.10.003. [DOI] [PubMed] [Google Scholar]
  • 154.Musilova K, Mraz M. MicroRNAs in B-cell lymphomas: how a complex biology gets more complex. Leukemia. 2015;29:1004–1017. doi: 10.1038/leu.2014.351. [DOI] [PubMed] [Google Scholar]
  • 155.Vosa U, Vooder T, Kolde R, et al. Identification of miR-374a as a prognostic marker for survival in patients with early-stage nonsmall cell lung cancer. Genes Chromosomes Cancer. 2011;50:812–822. doi: 10.1002/gcc.20902. [DOI] [PubMed] [Google Scholar]
  • 156.Akcakaya P, Ekelund S, Kolosenko I, et al. miR-185 and miR-133b deregulation is associated with overall survival and metastasis in colorectal cancer. Int J Oncol. 2011;39:311–318. doi: 10.3892/ijo.2011.1043. [DOI] [PubMed] [Google Scholar]
  • 157.Ma L. Role of miR-10b in breast cancer metastasis. Breast Cancer Res. 2010;12:210. doi: 10.1186/bcr2720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Liu Z, Zhu J, Cao H, Ren H, Fang X. miR-10b promotes cell invasion through RhoC-AKT signaling pathway by targeting HOXD10 in gastric cancer. Int J Oncol. 2012;40:1553–1560. doi: 10.3892/ijo.2012.1342. [DOI] [PubMed] [Google Scholar]
  • 159.Ouyang H, Gore J, Deitz S, Korc M. microRNA-10b enhances pancreatic cancer cell invasion by suppressing TIP30 expression and promoting EGF and TGF-beta actions. Oncogene. 2014;33:4664–4674. doi: 10.1038/onc.2013.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Borel F, Konstantinova P, Jansen PL. Diagnostic and therapeutic potential of miRNA signatures in patients with hepatocellular carcinoma. J Hepatol. 2012;56:1371–1383. doi: 10.1016/j.jhep.2011.11.026. [DOI] [PubMed] [Google Scholar]
  • 161.Wong CM, Wong CC, Lee JM, Fan DN, Au SL, Ng IO. Sequential alterations of microRNA expression in hepatocellular carcinoma development and venous metastasis. Hepatology. 2012;55:1453–1461. doi: 10.1002/hep.25512. [DOI] [PubMed] [Google Scholar]
  • 162.Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med. 2012;4:143–159. doi: 10.1002/emmm.201100209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Wei R, Huang GL, Zhang MY, et al. Clinical significance and prognostic value of microRNA expression signatures in hepatocellular carcinoma. Clin Cancer Res. 2013;19:4780–4791. doi: 10.1158/1078-0432.CCR-12-2728. [DOI] [PubMed] [Google Scholar]
  • 164.Huang S, He X. The role of microRNAs in liver cancer progression. Br J Cancer. 2011;104:235–240. doi: 10.1038/sj.bjc.6606010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Law PT, Wong N. Emerging roles of microRNA in the intracellular signaling networks of hepatocellular carcinoma. J Gastroenterol Hepatol. 2011;26:437–449. doi: 10.1111/j.1440-1746.2010.06512.x. [DOI] [PubMed] [Google Scholar]
  • 166.Hou J, Lin L, Zhou W, et al. 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. doi: 10.1016/j.ccr.2011.01.001. [DOI] [PubMed] [Google Scholar]
  • 167.Qi P, Cheng SQ, Wang H, Li N, Chen YF, Gao CF. Serum microRNAs as biomarkers for hepatocellular carcinoma in Chinese patients with chronic hepatitis B virus infection. PLoS One. 2011;6:e28486. doi: 10.1371/journal.pone.0028486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Li LM, Hu ZB, Zhou ZX, et al. Serum microRNA profiles serve as novel biomarkers for HBV infection and diagnosis of HBV-positive hepatocarcinoma. Cancer Res. 2010;70:9798–9807. doi: 10.1158/0008-5472.CAN-10-1001. [DOI] [PubMed] [Google Scholar]
  • 169.Jiang L, Li X, Cheng Q, Zhang BH. Plasma microRNA might as a potential biomarker for hepatocellular carcinoma and chronic liver disease screening. Tumour Biol. 2015;36:7167–7174. doi: 10.1007/s13277-015-3446-7. [DOI] [PubMed] [Google Scholar]
  • 170.Lin XJ, Chong Y, Guo ZW, et al. A serum microRNA classifier for early detection of hepatocellular carcinoma: a multicentre, retrospective, longitudinal biomarker identification study with a nested case-control study. Lancet Oncol. 2015;16:804–815. doi: 10.1016/S1470-2045(15)00048-0. [DOI] [PubMed] [Google Scholar]
  • 171.Zhou J, Yu L, Gao X, et al. Plasma microRNA panel to diagnose hepatitis B virus-related hepatocellular carcinoma. J Clin Oncol. 2011;29:4781–4788. doi: 10.1200/JCO.2011.38.2697. [DOI] [PubMed] [Google Scholar]
  • 172.Sun W, Ma J, Wu S, et al. Characterization of the liver tissue interstitial fluid (TIF) proteome indicates potential for application in liver disease biomarker discovery. J Proteome Res. 2010;9:1020–1031. doi: 10.1021/pr9009172. [DOI] [PubMed] [Google Scholar]
  • 173.Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer. 2010;127:118–126. doi: 10.1002/ijc.25007. [DOI] [PubMed] [Google Scholar]
  • 174.Su ZX, Zhao J, Rong ZH, Geng WM, Wu YG, Qin CK. Upregulation of microRNA-25 associates with prognosis in hepatocellular carcinoma. Diagn Pathol. 2014;9:47. doi: 10.1186/1746-1596-9-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Liang Z, Gao Y, Shi W, et al. Expression and significance of microRNA-183 in hepatocellular carcinoma. Sci World J. 2013(2013):381874. doi: 10.1155/2013/381874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Yang F, Yin Y, Wang F, et al. miR-17-5p promotes migration of human hepatocellular carcinoma cells through the p38 mitogen-activated protein kinase-heat shock protein 27 pathway. Hepatology. 2010;51:1614–1623. doi: 10.1002/hep.23566. [DOI] [PubMed] [Google Scholar]
  • 177.Yoon SO, Chun SM, Han EH, et al. Deregulated expression of microRNA-221 with the potential for prognostic biomarkers in surgically resected hepatocellular carcinoma. Hum Pathol. 2011;42:1391–1400. doi: 10.1016/j.humpath.2010.12.010. [DOI] [PubMed] [Google Scholar]
  • 178.Chen P, Zhao X, Ma L. Downregulation of microRNA-100 correlates with tumor progression and poor prognosis in hepatocellular carcinoma. Mol Cell Biochem. 2013;383:49–58. doi: 10.1007/s11010-013-1753-0. [DOI] [PubMed] [Google Scholar]
  • 179.Shi C, Xu X. MicroRNA-22 is down-regulated in hepatitis B virus-related hepatocellular carcinoma. Biomed Pharmacother. 2013;67:375–380. doi: 10.1016/j.biopha.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • 180.Li J, Wang Y, Yu W, Chen J, Luo J. Expression of serum miR-221 in human hepatocellular carcinoma and its prognostic significance. Biochem Biophys Res Commun. 2011;406:70–73. doi: 10.1016/j.bbrc.2011.01.111. [DOI] [PubMed] [Google Scholar]
  • 181.Gu H, Guo X, Zou L, Zhu H, Zhang J. Upregulation of microRNA-372 associates with tumor progression and prognosis in hepatocellular carcinoma. Mol Cell Biochem. 2013;375:23–30. doi: 10.1007/s11010-012-1521-6. [DOI] [PubMed] [Google Scholar]
  • 182.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. doi: 10.1007/s00432-011-1076-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.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. doi: 10.1186/1471-2407-12-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Xiong Y, Fang JH, Yun JP, et al. Effects of microRNA-29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology. 2010;51:836–845. doi: 10.1002/hep.23380. [DOI] [PubMed] [Google Scholar]
  • 185.Zhang Z, Zheng W, Hai J. MicroRNA-148b expression is decreased in hepatocellular carcinoma and associated with prognosis. Med Oncol. 2014;31:984. doi: 10.1007/s12032-014-0984-6. [DOI] [PubMed] [Google Scholar]
  • 186.Zhu HT, Dong QZ, Sheng YY, et al. MicroRNA-29a-5p is a novel predictor for early recurrence of hepatitis B virus-related hepatocellular carcinoma after surgical resection. PLoS One. 2012;7:e52393. doi: 10.1371/journal.pone.0052393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685–1694. doi: 10.1056/NEJMoa1209026. [DOI] [PubMed] [Google Scholar]
  • 188.Hsu SH, Wang B, Kota J, et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest. 2012;122:2871–2883. doi: 10.1172/JCI63539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Bai S, Nasser MW, Wang B, et al. MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J Biol Chem. 2009;284:32015–32027. doi: 10.1074/jbc.M109.016774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Yang F, Zhang L, Wang F, et al. Modulation of the unfolded protein response is the core of microRNA-122-involved sensitivity to chemotherapy in hepatocellular carcinoma. Neoplasia. 2011;13:590–600. doi: 10.1593/neo.11422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Xu Y, Xia F, Ma L, et al. MicroRNA-122 sensitizes HCC cancer cells to adriamycin and vincristine through modulating expression of MDR and inducing cell cycle arrest. Cancer Lett. 2011;310:160–169. doi: 10.1016/j.canlet.2011.06.027. [DOI] [PubMed] [Google Scholar]
  • 192.Zhao N, Wang R, Zhou L, Zhu Y, Gong J, Zhuang SM. MicroRNA-26b suppresses the NF-kappaB signaling and enhances the chemosensitivity of hepatocellular carcinoma cells by targeting TAK1 and TAB3. Mol Cancer. 2014;13:35. doi: 10.1186/1476-4598-13-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Anwar SL, Lehmann U. MicroRNAs: emerging novel clinical biomarkers for hepatocellular carcinomas. J Clin Med. 2015;4:1631–1650. doi: 10.3390/jcm4081631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Sohn W, Kim J, Kang SH, et al. Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Exp Mol Med. 2015;47:e184. doi: 10.1038/emm.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26:2166–2176. doi: 10.1038/sj.onc.1210279. [DOI] [PubMed] [Google Scholar]
  • 196.Gouas D, Shi H, Hainaut P. The aflatoxin-induced TP53 mutation at codon 249 (R249S): biomarker of exposure, early detection and target for therapy. Cancer Lett. 2009;286:29–37. doi: 10.1016/j.canlet.2009.02.057. [DOI] [PubMed] [Google Scholar]
  • 197.Matsuda Y, Ichida T, Matsuzawa J, Sugimura K, Asakura H. p16(INK4) is inactivated by extensive CpG methylation in human hepatocellular carcinoma. Gastroenterology. 1999;116:394–400. doi: 10.1016/s0016-5085(99)70137-x. [DOI] [PubMed] [Google Scholar]
  • 198.Zhang YJ, Jiang W, Chen CJ, et al. Amplification and overexpression of cyclin D1 in human hepatocellular carcinoma. Biochem Biophys Res Commun. 1993;196:1010–1016. doi: 10.1006/bbrc.1993.2350. [DOI] [PubMed] [Google Scholar]
  • 199.Chauhan R, Kazim SN, Bhattacharjee J, Sakhuja P, Sarin SK. Basal core promoter, precore region mutations of HBV and their association with e antigen, genotype, and severity of liver disease in patients with chronic hepatitis B in India. J Med Virol. 2006;78:1047–1054. doi: 10.1002/jmv.20661. [DOI] [PubMed] [Google Scholar]
  • 200.Fang ZL, Sabin CA, Dong BQ, et al. HBV A1762T, G1764A mutations are a valuable biomarker for identifying a subset of male HBsAg carriers at extremely high risk of hepatocellular carcinoma: a prospective study. Am J Gastroenterol. 2008;103:2254–2262. doi: 10.1111/j.1572-0241.2008.01974.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Liu S, Zhang H, Gu C, et al. Associations between hepatitis B virus mutations and the risk of hepatocellular carcinoma: a meta-analysis. J Natl Cancer Inst. 2009;101:1066–1082. doi: 10.1093/jnci/djp180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Yang HI, Yeh SH, Chen PJ, et al. Associations between hepatitis B virus genotype and mutants and the risk of hepatocellular carcinoma. J Natl Cancer Inst. 2008;100:1134–1143. doi: 10.1093/jnci/djn243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Kuang SY, Jackson PE, Wang JB, et al. Specific mutations of hepatitis B virus in plasma predict liver cancer development. Proc Natl Acad Sci U S A. 2004;101:3575–3580. doi: 10.1073/pnas.0308232100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Kuang SY, Lekawanvijit S, Maneekarn N, et al. Hepatitis B 1762T/1764A mutations, hepatitis C infection, and codon 249 p53 mutations in hepatocellular carcinomas from Thailand. Cancer Epidemiol Biomarkers Prev. 2005;14:380–384. doi: 10.1158/1055-9965.EPI-04-0380. [DOI] [PubMed] [Google Scholar]
  • 205.Pollicino T, Cacciola I, Saffioti F, Raimondo G. Hepatitis B virus PreS/S gene variants: pathobiology and clinical implications. J Hepatol. 2014;61:408–417. doi: 10.1016/j.jhep.2014.04.041. [DOI] [PubMed] [Google Scholar]
  • 206.Qu LS, Liu JX, Liu TT, et al. Association of hepatitis B virus pre-S deletions with the development of hepatocellular carcinoma in Qidong, China. PLoS One. 2014;9:e98257. doi: 10.1371/journal.pone.0098257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yeh CT, Chen T, Hsu CW, et al. Emergence of the rtA181T/sW172* mutant increased the risk of hepatoma occurrence in patients with lamivudine-resistant chronic hepatitis B. BMC Cancer. 2011;11:398. doi: 10.1186/1471-2407-11-398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Wang HC, Wu HC, Chen CF, Fausto N, Lei HY, Su IJ. Different types of ground glass hepatocytes in chronic hepatitis B virus infection contain specific pre-S mutants that may induce endoplasmic reticulum stress. Am J Pathol. 2003;163:2441–2449. doi: 10.1016/S0002-9440(10)63599-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Buendia MA, Neuveut C. Hepatocellular carcinoma. Cold Spring Harb Perspect Med. 2015;5:a021444. doi: 10.1101/cshperspect.a021444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Gearhart TL, Bouchard MJ. The hepatitis B virus X protein modulates hepatocyte proliferation pathways to stimulate viral replication. J Virol. 2010;84:2675–2686. doi: 10.1128/JVI.02196-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Zhang H, Wu LY, Zhang S, et al. Anti-hepatitis B virus X protein in sera is one of the markers of development of liver cirrhosis and liver cancer mediated by HBV. J Biomed Biotechnol. 2009;2009:289068. doi: 10.1155/2009/289068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zhu M, Guo J, Li W, et al. Hepatitis B virus X protein induces expression of alpha-fetoprotein and activates PI3K/mTOR signaling pathway in liver cells. Oncotarget. 2015;6:12196–12208. doi: 10.18632/oncotarget.2906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Ley TJ, Mardis ER, Ding L, et al. DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature. 2008;456:66–72. doi: 10.1038/nature07485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Jiang Z, Jhunjhunwala S, Liu J, et al. The effects of hepatitis B virus integration into the genomes of hepatocellular carcinoma patients. Genome Res. 2012;22:593–601. doi: 10.1101/gr.133926.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Sung WK, Zheng H, Li S, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 2012;44:765–769. doi: 10.1038/ng.2295. [DOI] [PubMed] [Google Scholar]
  • 216.Toh ST, Jin Y, Liu L, et al. Deep sequencing of the hepatitis B virus in hepatocellular carcinoma patients reveals enriched integration events, structural alterations and sequence variations. Carcinogenesis. 2013;34:787–798. doi: 10.1093/carcin/bgs406. [DOI] [PubMed] [Google Scholar]
  • 217.Nault JC, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol. 2016;40:9–14. doi: 10.1016/j.clinre.2015.07.006. [DOI] [PubMed] [Google Scholar]
  • 218.Chauhan R, Churchill ND, Michalak TI. Initial sites of hepadnavirus integration with host genome after de novoinfection of human hepatocytes and woodchuck model of hepatitis B. unpublished. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Guichard C, Amaddeo G, Imbeaud S, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nature Genetics. 2012;44(6):694–698. doi: 10.1038/ng.2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Janku F, Kaseb AO, Tsimberidou AM, Wolff RA, Kurzrock R. Identification of novel therapeutic targets in the PI3K/AKT/mTOR pathway in hepatocellular carcinoma using targeted next generation sequencing. Oncotarget. 2014;5:3012–3022. doi: 10.18632/oncotarget.1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Sanyal AJ, Yoon SK, Lencioni R. The etiology of hepatocellular carcinoma and consequences for treatment. Oncologist. 2010;15(suppl 4):14–22. doi: 10.1634/theoncologist.2010-S4-14. [DOI] [PubMed] [Google Scholar]
  • 222.Wu YJ, Jan YJ, Ko BS, Liang SM, Liou JY. Involvement of 14-3-3 proteins in regulating tumor progression of hepatocellular carcinoma. Cancers. 2015;7:1022–1036. doi: 10.3390/cancers7020822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Xu Z, Liu L, Pan X, et al. Serum Golgi protein 73 (GP73) is a diagnostic and prognostic marker of chronic HBV liver disease. Medicine (Baltimore) 2015;94:e659. doi: 10.1097/MD.0000000000000659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ratnasari N, Yano Y. Do soluble vascular endothelial growth factor and its receptors predict the progression of chronic hepatitis to hepatocellular carcinoma? Journal of Hepatitis. 2015;1:4. [Google Scholar]
  • 225.Fu WM, Zhu X, Wang WM, et al. Hotair mediates hepatocarcinogenesis through suppressing miRNA-218 expression and activating P14 and P16 signaling. J Hepatol. 2015;63:886–895. doi: 10.1016/j.jhep.2015.05.016. [DOI] [PubMed] [Google Scholar]
  • 226.Wu JT. Serum alpha-fetoprotein and its lectin reactivity in liver diseases: a review. Ann Clin Lab Sci. 1990;20:98–105. [PubMed] [Google Scholar]
  • 227.Khien VV, Mao HV, Chinh TT, et al. Clinical evaluation of lentil lectin-reactive alpha-fetoprotein-L3 in histology-proven hepatocellular carcinoma. Int J Biol Markers. 2001;16:105–111. doi: 10.1177/172460080101600204. [DOI] [PubMed] [Google Scholar]
  • 228.Nakatsura T, Yoshitake Y, Senju S, et al. Glypican-3, overexpressed specifically in human hepatocellular carcinoma, is a novel tumor marker. Biochem Biophys Res Commun. 2003;306:16–25. doi: 10.1016/s0006-291x(03)00908-2. [DOI] [PubMed] [Google Scholar]
  • 229.Weitz IC, Liebman HA. Des-gamma-carboxy (abnormal) prothrombin and hepatocellular carcinoma: a critical review. Hepatology. 1993;18:990–997. doi: 10.1002/hep.1840180434. [DOI] [PubMed] [Google Scholar]
  • 230.Cui R, He J, Zhang F, et al. Diagnostic value of protein induced by vitamin K absence (PIVKAII) and hepatoma-specific band of serum gamma-glutamyl transferase (GGTII) as hepatocellular carcinoma markers complementary to alpha-fetoprotein. Br J Cancer. 2003;88:1878–1882. doi: 10.1038/sj.bjc.6601018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Yu B, Yang X, Xu Y, et al. Elevated expression of DKK1 is associated with cytoplasmic/nuclear beta-catenin accumulation and poor prognosis in hepatocellular carcinomas. J Hepatol. 2009;50:948–957. doi: 10.1016/j.jhep.2008.11.020. [DOI] [PubMed] [Google Scholar]
  • 232.Marrero JA, Romano PR, Nikolaeva O, et al. GP73, a resident Golgi glycoprotein, is a novel serum marker for hepatocellular carcinoma. J Hepatol. 2005;43:1007–1012. doi: 10.1016/j.jhep.2005.05.028. [DOI] [PubMed] [Google Scholar]
  • 233.Okumoto K, Hattori E, Tamura K, et al. Possible contribution of circulating transforming growth factor-beta1 to immunity and prognosis in unresectable hepatocellular carcinoma. Liver Int. 2004;24:21–28. doi: 10.1111/j.1478-3231.2004.00882.x. [DOI] [PubMed] [Google Scholar]
  • 234.Ikeguchi M, Iwamoto A, Taniguchi K, Katano K, Hirooka Y. The gene expression level of transforming growth factor-beta (TGF-beta) as a biological prognostic marker of hepatocellular carcinoma. J Exp Clin Cancer Res. 2005;24:415–421. [PubMed] [Google Scholar]
  • 235.Song BC, Chung YH, Kim JA, et al. Transforming growth factor-beta1 as a useful serologic marker of small hepatocellular carcinoma. Cancer. 2002;94:175–180. doi: 10.1002/cncr.10170. [DOI] [PubMed] [Google Scholar]
  • 236.Ito Y, Takeda T, Sakon M, et al. Expression and clinical significance of erb-B receptor family in hepatocellular carcinoma. Br J Cancer. 2001;84:1377–1383. doi: 10.1054/bjoc.2000.1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Osada S, Kanematsu M, Imai H, Goshima S. Clinical significance of serum HGF and c-Met expression in tumor tissue for evaluation of properties and treatment of hepatocellular carcinoma. Hepatogastroenterology. 2008;55:544–549. [PubMed] [Google Scholar]
  • 238.Poon RT, Ng IO, Lau C, Yu WC, Fan ST, Wong J. Correlation of serum basic fibroblast growth factor levels with clinicopathologic features and postoperative recurrence in hepatocellular carcinoma. Am J Surg. 2001;182:298–304. doi: 10.1016/s0002-9610(01)00708-5. [DOI] [PubMed] [Google Scholar]
  • 239.Jeng KS, Sheen IS, Tsai YC. Circulating messenger RNA of alpha-fetoprotein: a possible risk factor of recurrence after resection of hepatocellular carcinoma. Arch Surg. 2004;139:1055–1060. doi: 10.1001/archsurg.139.10.1055. [DOI] [PubMed] [Google Scholar]
  • 240.Sheen IS, Jeng KS, Tsai YC. Is the expression of gamma-glutamyl transpeptidase messenger RNA an indicator of biological behavior in recurrent hepatocellular carcinoma? World J Gastroenterol. 2003;9:468–473. doi: 10.3748/wjg.v9.i3.468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Himoto T, Kuriyama S, Zhang JY, et al. Analyses of autoantibodies against tumor-associated antigens in patients with hepatocellular carcinoma. Int J Oncol. 2005;27:1079–1085. [PubMed] [Google Scholar]
  • 242.Cheung ST, Fan ST, Lee YT, et al. Albumin mRNA in plasma predicts post-transplant recurrence of patients with hepatocellular carcinoma. Transplantation. 2008;85:81–87. doi: 10.1097/01.tp.0000298003.88530.11. [DOI] [PubMed] [Google Scholar]
  • 243.Yao J, Liang L, Huang S, et al. MicroRNA-30d promotes tumor invasion and metastasis by targeting Galphai2 in hepatocellular carcinoma. Hepatology. 2010;51:846–856. doi: 10.1002/hep.23443. [DOI] [PubMed] [Google Scholar]
  • 244.Coulouarn C, Factor VM, Andersen JB, Durkin ME, Thorgeirsson SS. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene. 2009;28:3526–3536. doi: 10.1038/onc.2009.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Ura S, Honda M, Yamashita T, et al. Differential microRNA expression between hepatitis B and hepatitis C leading disease progression to hepatocellular carcinoma. Hepatology. 2009;49:1098–1112. doi: 10.1002/hep.22749. [DOI] [PubMed] [Google Scholar]
  • 246.Fartoux L, Decaens T. Contribution of biomarkers and imaging in the management of hepatocellular carcinoma. Clin Res Hepatol Gastroenterol. 2011;35(suppl 1):S21–S30. doi: 10.1016/S2210-7401(11)70004-3. [DOI] [PubMed] [Google Scholar]
  • 247.Minervini MI, Demetris AJ, Lee RG, Carr BI, Madariaga J, Nalesnik MA. Utilization of hepatocyte-specific antibody in the immunocytochemical evaluation of liver tumors. Mod Pathol. 1997;10:686–692. [PubMed] [Google Scholar]
  • 248.Guido M, Roskams T, Pontisso P, et al. Squamous cell carcinoma antigen in human liver carcinogenesis. J Clin Pathol. 2008;61:445–447. doi: 10.1136/jcp.2007.051383. [DOI] [PubMed] [Google Scholar]
  • 249.Beneduce L, Castaldi F, Marino M, et al. Squamous cell carcinoma antigen-immunoglobulin M complexes as novel biomarkers for hepatocellular carcinoma. Cancer. 2005;103:2558–2565. doi: 10.1002/cncr.21106. [DOI] [PubMed] [Google Scholar]
  • 250.Zhao YJ, Ju Q, Li GC. Tumor markers for hepatocellular carcinoma. Mol Clin Oncol. 2013;1:593–598. doi: 10.3892/mco.2013.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Jin ZH, Yang RJ, Dong B, Xing BC. Progenitor gene DLK1 might be an independent prognostic factor of liver cancer. Expert Opin Biol Ther. 2008;8:371–377. doi: 10.1517/14712598.8.4.371. [DOI] [PubMed] [Google Scholar]
  • 252.Xieraili M, Yasen M, Mogushi K, et al. Villin 1 is a predictive factor for the recurrence of high serum alpha-fetoprotein-associated hepatocellular carcinoma after hepatectomy. Cancer Sci. 2012;103:1493–1501. doi: 10.1111/j.1349-7006.2012.02315.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Biomarkers in Cancer are provided here courtesy of SAGE Publications

RESOURCES