Next Article in Journal
RbfA Is Involved in Two Important Stages of 30S Subunit Assembly: Formation of the Central Pseudoknot and Docking of Helix 44 to the Decoding Center
Next Article in Special Issue
Inflammation, Extracellular Matrix Remodeling, and Proteostasis in Tumor Microenvironment
Previous Article in Journal
Neuroinflammation as a Common Denominator of Complex Diseases (Cancer, Diabetes Type 2, and Neuropsychiatric Disorders)
Previous Article in Special Issue
Syndecan-1 Depletion Has a Differential Impact on Hyaluronic Acid Metabolism and Tumor Cell Behavior in Luminal and Triple-Negative Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunodiagnostic Biomarkers for Hepatocellular Carcinoma (HCC): The First Step in Detection and Treatment

1
Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
2
Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
3
Jiangsu Key Laboratory of Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
*
Authors to whom correspondence should be addressed.
Xinzhi Wang and Mengtao Xing contributed equally to this paper.
Int. J. Mol. Sci. 2021, 22(11), 6139; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22116139
Submission received: 30 April 2021 / Revised: 27 May 2021 / Accepted: 27 May 2021 / Published: 7 June 2021
(This article belongs to the Special Issue Extracellular Matrix in the Tumor Microenvironment)

Abstract

:
Hepatocellular carcinoma (HCC) exerts huge effects on the health burden of the world because of its high mortality and poor prognosis. HCC is often clinically detected late in patients. If HCC could be detected and treated earlier, the survival rate of patients will be greatly improved. Therefore, identifying specific biomarkers is urgent and important for HCC. The liver is also recognized as an immune organ. The occurrence of HCC is related to exacerbation of immune tolerance and/or immunosurveillance escape. The host immune system plays an important role in the recognition and targeting of tumor cells in cancer immunotherapy, as can be seen from the clinical success of immune checkpoint inhibitors and chimeric antigen receptor (CAR) T cells. Thus, there is a pressing medical need to discover immunodiagnostic biomarkers specific to HCC for understanding the pathological mechanisms of HCC, especially for immunotherapy targets. We have reviewed the existing literature to summarize the immunodiagnostic markers of HCC, including autoantibodies against tumor-associated antigens (TAAs) and exosomes, to provide new insights into HCC and early detection of this deadly cancer.

1. Introduction

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, which constitutes around 80% of cases [1]. It poses a huge health risk and is the sixth most diagnosed cancer worldwide and the fourth leading cause in 2018. [2]. Contributing factor to its poor prognosis is the absence of obvious symptoms during the early stages of HCC. Consequently, greater than 60% of patients with HCC are diagnosed in advanced stages [3], leading to an extremely low overall five-year survival rate (less than 16%) [4]. However, there are many effective curable therapies for early HCC, making a good prognosis for early patients. For example, when a patient is diagnosed with HCC in Barcelona-based clinical liver cancer (BCLC) stage 0 and A, the 5-year survival rate is higher than 93% through surgical intervention therapies [5]. Thus, novel biomarkers for early HCC detection significantly impact curative treatment regimens. This exciting development of HCC biomarkers may encourage the use of more effective and novel chemoprevention strategies for people at high risk for HCC, such as HBV-infected individuals.
The ideal HCC biomarker is one that can be widely employed during a rapid and inexpensive screening process, and even asymptomatic patients can be diagnosed by trained clinicians. Generally, clinically useful biomarkers can achieve at least 90% sensitivity and specificity levels, and are non-invasive and cost-effective to be widely used [6]. Therefore, the most desirable biomarkers are tumor-specific and readily detectable in body fluids, for example, serum, plasma, and bile. Over the past decades, serum α-fetoprotein (AFP) has been used for HCC early detection, but elevated levels of AFP has also been shown to have predictive power in other disease, such as acute viral hepatitis A (AHA) [7]. Since AFP is generally less sensitive to HCC detection (20–65%), serum AFP is no longer in the recommended guidelines [8]. The use of ultrasonography has gained some acceptance, however its effectiveness depends on the operator and has limited ability to clearly differentiate between HCC and benign nodules [9]. Dual-phase computed tomography (CT) scan, and magnetic resonance imaging (MRI) are more effective when the nodules are larger than 1–2 cm [10]. Ineffective diagnostic biomarkers result in an inadequate diagnosis of a large number of HCC patients and false positives in patients with non-tumor liver disease, such as hepatitis and cirrhosis. Because of its dual blood supply from both the systemic circulation and the portal vein, the liver is an important immune organ. Under physiologic conditions, it exerts a protective effect by promoting immune tolerance [11]. Since HCC has been shown as an inflammatory tumor [12], it is necessary to review early immunospecific biomarkers to immunologically diagnose HCC. The use of serum autoantibodies to tumor-associated antigens (TAAs) as serological cancer biomarkers is growing in popularity. The source of this interest is based on the cognition that anti-TAAs antibodies are "indicators" of the tumorigenesis associated with molecular events.

2. Tumor-Associated Antigens (TAAs) and Anti-TAAs Autoantibodies

As early as the 1960s, Robert W. Baldwin showed that the human immune system can produce autoantibodies in the early stage of cancer development [13]. The normal proteins of the human body do not have immunogenicity because of self-tolerance. A widely accepted view is that mutations, ectopic or via recombination, can occur early in the development and progression of tumor cells [14]. During tumorigenesis and progression, altered protein expression levels, an environment of chronic inflammation, protein structure changes, and cell death mechanisms will trigger specific proteins, called TAAs, promoting the host’s immune responses. Ultimately, the autoantibodies against TAAs, which can be regarded as "sensors" or "reporters" that indicate abnormalities or dysregulation of cellular mechanisms during tumorigenesis, are detectable in the serum. Therefore, these specific autoantibodies can be used as potential biomarkers in early cancer diagnoses, a detector of treatment results, and even an indicator of cancer prognosis [15,16].
Cancer-specific autoantibodies, usually present at very low titers or absent in non-cancer individuals, are produced by the self-immune system in response to TAAs. Modern biological methods have been able to detect low concentrations of antibodies that cannot be detected by traditional in vitro testing [17]. In addition, antibodies are not only stable in vivo, whose survival time in the circulation of cancer patients is usually up to 30 days, but also more stable in vitro than other biomarkers. Based on these unique advantages, anti-TAAs autoantibodies are ideal biomarkers for the early detection of tumorigenesis [18]. The shortcoming of autoantibodies against single TAA in diagnosis is the low frequency (10–20 %) in HCC sera [15]. For this disadvantage, our group found that this drawback can be overcome by using a carefully selected TAA microarray. But for different types of cancers, may need to design different TAA arrays to achieve the needed sensitivity and specificity to make this method a reliable reference for clinicians’ tumor diagnosis (Figure 1). We designed a microarray for HCC diagnosis that includes various TAAs, such as tumor suppressors p53, Ink4a/p16, NPM1/B23, CAPERα/HCC1.4, and oncoproteins, including IMP1, IMP2/p62, IMP3/Koc, CIP2A/p90, RalA, c-Myc, survivin, cyclin B1, 14-3-3ζ, and MDM2. The sensitivity and specificity of 14 TAAs for immunological diagnosis of HCC were 69.7% and 83.0%, respectively [19]. It is noteworthy that 43.8% of HCC patients identified by this microarray had normal serum AFP levels [19]. In a recent study, a panel of 12 TAAs was used to establish a predictive value based on the sera from 160 HCC patients and 90 normal controls and then successfully used to diagnose HCC in 16 of 17 patients who had no earlier clinical information prior to diagnosis [20]. In a series of studies, it was shown that the study of anti-TAAs panels as biomarkers acts as a potential early detection method for HCC.
The nature of autoantibodies against different types of TAAs is different. For tumor suppressors, the most likely reason is mutation. TP53, as a famous tumor suppressor gene, frequently mutated in HCC patients (~30%), and its autoantibodies can be detected in 20% of HCC patients. Different from this, the autoantibodies against the proteins encoded by oncogenes are usually produced, due to the protein overexpression [21,22]. Oncoproteins IMP1, IMP2/p62, and IMP3/Koc belong to the IGF-II mRNA binding protein (IMPs) family. They are highly conserved and share a similar structure: The identity of the overall amino acid sequences approximately is 60-80%; all three members have six characteristic RNA binding modules, including two N-terminal RNA recognition motifs (RRMs), and four heteronuclear ribonucleoproteins (hnRNP) K-homology (KH) domains in their C-terminal regions [23]. IMPs are mainly expressed during embryonic development and almost disappear completely in adult tissues. Nevertheless, they are again highly expressed in a variety of cancer tissues, so they are considered oncofetal proteins [24,25]. In 1999, we first identified IMP2 as a TAA in the sera of HCC patients. The autoantibodies against IMP2 can be detected in the early stage of HCC and approximately 21% of HCC patients [19,23]. In subsequent work, we found that the expression of IMP2 was elevated in both mRNA and protein levels in HCC. Overexpression of IMP2 can enhance the signal of the Wnt/β-Catenin pathway in HCC, induce the genomic instability and improve the migration ability of HCC cells [26,27]. In addition, IMP1 and IMP3 play important roles in the development of HCC [28,29]. We also found a high frequency of autoantibodies of IMP1 and IMP3 existed in the sera of HCC patients [19]. The IGF2 mRNA is one of the common targets of IMPs, which is a fetal growth factor and has a similar structure and function to IGF1 [30]. IGF2 is not expressed in healthy adults. However, the frequency of various cancers occurred more frequently in IGF2 transgenic mice [31]. In human HCC, overexpression of IGF2 is associated with fetal malformations and a variety of cancers [32]. Under the regulation of IMPs, the expression of IGF2 in HCC was significantly increased [33]. Meanwhile, IGF-2 can also be recognized as a cofactor or a second signal for transformation in SV40 oncogene-induced tumorigenesis [34]. In summary, the TAAs like IMPs, can play an important role in carcinogenesis by modulating its downstream signaling, and their autoantibodies could be regarded as valuable biomarkers in the early diagnosis of HCC.
In Table 1, a comprehensive list of autoantibodies used in HCC diagnosis which have been reported since 1997.

3. HCC-Derived Exosome

Exosomes are membrane-bound extracellular vesicles that are 40–100 nm in size. Under normal and pathological conditions, almost all cell types secrete exosomes. Exosomes existing in various body fluids (blood, urine, ascites, and et al.) cargoes a wide range of biomolecules, including lipids, messenger RNAs (mRNAs), microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and proteins [82]. These features of exosomes could act as potential diagnostic and monitoring biomarkers for cancer [83]. (1) The amount and content of exosomes reflect the actual conditions of their original cells with high sensitivity, because exosomes can be immune-isolated using antibodies against tissue-specific proteins on the cell membrane surface. (2) The stable structure of lipid bilayer protects exosomal cargoes from damage by widespread RNases and proteases in circulation, thus exosomes can keep their cargos intact and have high fidelity and consistency. (3) Release of exosomes into biological fluids provides a non-invasive way to determine patients with tumors. (4) In contrast to the hardly detectable signal in biological fluids, exosomes comprise abundant specific cancer proteins, non-coding nuclear acids, and other markers, which facilitates the recognition of low abundance nucleic acids or proteins while increasing the signal-to-noise ratio during detection [84]. (5) The comparatively uncomplicated structure of exosomes reduces the complexity of the identification. Moreover, they may have circulating biomarkers with more accuracy, which can be a benefit for the early diagnosis of tumors. (6) Exosome analysis can directly generate details on disease lineage, staging, relapse, and drug reaction [85]. Therefore, exosomes might represent a “liquid biopsy” for malignancies.
Extensive researches have investigated serum exosomal miRNAs as potential biomarkers. As shown in Table 2, two exosome miRNAs, miR-21, and miR-122, appear repeatedly. Serum miR-21 is an important indicator used to independently assess relapse and is revealed to have more sensitivity than AFP in detecting HCC [86]. In comparison to normal individuals or patients with chronic hepatitis B (CHB), patients with HCC showed increased exosomal miR-21 levels, which is related to LC and advanced tumor stage. Moreover, compared with whole serum free of exosomes, the exosomal miR-21 level is obviously more elevated, which shows higher diagnostic sensitivity [87]. The above studies suggest that miRNAs in exosomes of circulation, like miR-21, can be used to predict HCC risk and detect HCC early. However, a report has also shown that miR-21 level in serum of HCC patients is decreased in comparison to chronic hepatitis patients [88]. Similarly, there is no increase in miR-21 levels of HBV-related HCC patients. Therefore, circulating miR-21 levels cannot distinguish patients with cirrhosis from those with HCC [89]. Identification techniques of miRNA, patient selection, and shortage of general internal controls of miRNA may contribute to this difference. Liver-specific miR-122 plays a suppressive role in the HCC progress by binding to target genes related to cell proliferation, migration, differentiation, apoptosis, and angiogenesis in HCC [90]. MiR-122 not only has diagnostic value for HCC, but also has been shown to have relation to the fibrosis of the liver [9,91], and viral replication rate [92]. Circulating levels of miR-122 are related to liver damage, as well as high ALT levels [93]. If the levels of exosomal miRNA match those in the parental cells, the result may suggest that exosomal miR-122 reflect residual liver function and capacity [94].
Many studies have been performed to investigate the possibility of exosomes as markers for different stages of HCC. Compared with serum circulating mi-RNA levels, serum exosomal mi-RNA levels can better distinguish HCC from CHB or LC. It has been revealed that the circulating levels of exosomal miR-18a, miR-221, miR-222, and miR-224 in HCC patients are significantly increased compared to patients of CHB or liver cirrhosis (LC) [95]. However, the serum levels of exosomal miR-101, miR-106b, miR-122, and miR-195 of HCC patients are reduced compared to CHB patients [95]. The prognosis for HCC patients remains unsatisfactory because of the great prevalence of postoperative relapse and cancer proliferation. MiR-125b in exosomes of serum is served as a biomarker in predicting relapse and survival in HCC patients after surgery [96]. In addition, exosomal lncRNA-ATB acts as a non-invasive predictor of HCC prognosis, which is independent of age, gender, the existence of LC, or cause [97]. It has been demonstrated that lncRNA-ATB promotes ZEB1 and ZEB2, thereby cause EMT, attack from cancer cells, and spread of cancer [98]. The serum level of exosomal miR-718 of recurrent HCC patients after LT is obviously reduced compared with patients free of recurrence, which has a positive relation to poor prognosis and aggressiveness of HCC patients [99]. Recently, accumulating evidence has identified that exosomes select particular miRNAs into their packages dependent on conditions [100]. The miRNA expression profiling in exosomes of the HCC cell line SMMC-7721, is different from the parental cells, suggesting that exosomes act as regulators of miRNAs in cells [101]. Exosomes can also cargo proteins with significant diagnostic or prognostic potential. Proteomics identification has found differences in serum exosomes of cholangiocarcinoma (CCA), primary sclerosing cholangitis (PSC), HCC, and normal controls. Serum exosomal galectin-3-binding protein (LG3BP) and polymeric immune receptor (PIGR) reveal more sensitivity in the detection of HCC than AFP. Both are oncogenic proteins, promoting the progression of HCC, malignant transformation, aggression, and proliferation [102]. LG3BP is also considered a biomarker for the poor prognosis of HCC [103].
Exosomes are a kind of non-invasive, sensitive, and specific biomarkers that serve as diagnostic, prognostic, and predictive markers for HCC (Table 2). Unlike conventional cancer biomarkers, including carcinoembryonic antigen and AFP, exosomal miRNAs can transfer their functions to target cells and modulate cell signaling [104]. Generally, tumor cells produce an increasing number of exosomes than normal cells [105]. Tumor-derived exosomes contribute to an appropriate microenvironment for tumor cells to proliferate, reduce in effectiveness of the drug, generate blood vessels, invasion, regulate the immune system, and form metastatic niche before metastasis [106]. In particular, exosomes are essential for cell-cell communication by transmitting information from tumor cells to nearby or distant cells. In addition, exosomes can induce immune responses and modulate the immune system [107]. Overcoming the resistance of HCC to chemotherapy is also a challenge. Exosomes display a range of HCC antigens [108]. Cancer-derived exosomes have been shown to trigger a stronger immune response mediated by dendritic cells (DC) rather than cell lysates, and ameliorate the tumor microenvironment (TEM) of HCC [109]. Exosomes secreted by AFP-expressing DCs (DEXAFP) induce intense immune responses based on antigen specificity, which lead to significantly delayed cancer development and increased survival rates in murine models of HCC. Therefore, DEXAFP may provide a promising vaccine without cells for immunotherapy of HCC [110]. Moreover, HCC cell-derived exosomes enhance drug resistance of sorafenib in vitro. However, AMSC-derived exosomes are effective carriers of miRNA-122 and can increase the chemosensitivity of HCC [111]. These results provide potential immunotherapies for the improvement of therapeutic efficacy.

4. Other Immunodiagnostic Biomarkers

Squamous cell carcinoma antigen (SCCA) is a kind of serine protease inhibitor known as serpins [124]. Under physiological conditions, SCCA is expressed by the stratum granulosum of normal squamous epithelium. Under pathological conditions, it is found in carcinoma cells, such as lung tumors, cervical tumors, and head and neck tumors [125]. SCCA is absent in normal liver cells, whereas its expression is elevated when liver inflammation occurs [126]. Immunohistochemical results showed that variants of SCCA (SCCA1 and SCCA2) are overexpressed in biopsies specimens of HCC [124]. IgM antibodies in circulation constitute antigen-antibody complexes with specific cancer biomarkers, offers chances for the prediction and treatment of HCC patients [127]. The emergence of the biomarker IgM immune complex appears to be confirmed by a model of cancer immune editing, which recognizes natural IgMs as participants of great significance in innate immunity [128]. IgM in circulation identifies new epitopes on the surface of cancer cells and promotes the phagocytosis and clearance of transformed cells by dendritic cells and macrophages. The mechanism reveals a possible host defense that attempts to exert selective pressure on newborn neoplastic cells to fight cancer growth [129]. Compared with LC and control groups, elevated levels of SCCA-IgM immune complexes can be detected in the serum of cirrhosis and HCC patient and assessing SCCA-IgM levels have better diagnostic value than determining the corresponding single molecule [126,130,131]. A significantly positive correlation exists between SCCA-IgM and AFP [132]. The diagnostic accurateness of serum SCCA-IgM is greater than that of AFP, due to its higher AUC, sensitivity, and specificity, which distinguish HCC patients from cirrhosis patients [133]. The level of SCCA-IgM decreases after treatment, suggesting that SCCA-IgM plays a role in the prognostic prediction of HCC. Reduced levels of SCCA–IgM may relate to a decrease in the innate immunity and/or reduced release activity of tumor cells in patients tested positive. This data indicates that the reduced concentration or activity of SCCA in the liver may reflect the decreased invasion, proliferation, and anti-apoptotic properties of cancer cells [134]. Elevated serum SCCA-IgM levels of HCC patients have also been reported to have a relation to the decreased survival, suggesting that serum SCCA–IgM levels may make a significant contribution to the prediction of HCC therapy response [130,135].
In addition, an oncofetal protein, Glypican-3 (GPC3), is regarded as a potential biomarker in serum recently. It is a member of the Glypican family and is a heparan sulfate proteoglycan located on the outer surface of cell membranes [136]. GPC3 is expressed in the liver of the fetus, whereas it cannot be detected in the liver of healthy adults [137]. In contrast, GPC3 is overexpressed not only at the gene, but also protein levels in HCC patients. It has been demonstrated in a study that GPC3 mRNA is overexpressed in 55.7-100% of HCC tissues [138]. The same conclusions have also been shown in hepatitis C virus (HCV) infected HCC patients [139]. At the protein level, GPC3 is highly expressed in more than 70% of HCC patients by immunohistochemical staining (21/29), but not expressed in healthy or benign hepatic lesions, cirrhosis, hepatitis, or healthy adult tissues (0/38) [140]. More importantly, research has revealed that GPC3 levels are increased not only in liver cancer tissues, but also in serum. GPC3 can be cleaved and released from the cell surface into the serum. At the same time, GPC3 autoantibodies and GPC3-specific cytotoxic T lymphocytes (CTL) can be detected in the blood of HCC patients [141]. Therefore, the levels of serum GPC3, its autoantibodies, and even specific immune cells can be used as potential diagnostic biomarkers for HCC [142]. In liver cancer diagnosis, AFP is a traditional biomarker, and the expression of GPC3 can distinguish benign nodular liver tumor with AFP negative. Therefore, when diagnosing liver cancer, the combined use of GPC3 and AFP may make the diagnosis more reliable [143]. GPC3 has been the target of molecular imaging for early liver cancer, especially the positron emission tomography imaging technology for GPC3 has matured nowadays [144]. Various immunotherapies targeting GPC3 have also been studied, such as the use of antibodies against anti-GPC3, gene therapy against GPC3, vaccines based on GPC3 peptides, and GPC3-targeted CART therapy, etc. [145].
Chemokines in serum can also be deemed as ideal potential biomarkers for HCC diagnosis. For instance, CXCL13 can cooperate with its receptor CXCR5, attracting T helper cells and to encourage lymphocyte homing of naive B cells [146]. Hence, CXCL13 is also known as B cell attracting chemokine 1 (BCA 1) in humans [147]. Many studies have demonstrated that CXCL13 is deeply involved in the occurrence, development, and metastasis of multiple tumors [148,149]. In HCC, the expression of CXCL13 and CXCR5 are both up-regulated compared to healthy tissues, and even higher in poorly differentiated cancer tissues, which may be related to the harsh tumor microenvironment [150]. Clinical data showed that CXCL13 is elevated in the serum of more than 60% of HCC patients compared to healthy controls. As tumors become larger, the levels of CXCL13 in serum become higher and higher. Correlation analysis showed that serum CXCL13 levels are of high value for the diagnosis and prognosis of HCC [151]. Another clinical research also indicated that the combination of CXCL13 and AFP may potentially increase the sensitivity of AFP to HCC and can be used in the clinical diagnosis of HCC [152]. In the future, we believe that the above-mentioned potential biomarkers will be used in combination with AFP to have a more accurate diagnosis of early HCC.

5. Conclusions

The immune system of cancer patients suggests that it can perceive structure, function, internal location of cells, and other changes in cellular participants during tumorigenesis, which can be the first sign of carcinogenesis. Immunodiagnostic markers have been described as the reporters, internal outposts, and immune surveillance of the immune system. Moreover, immunodiagnostic biomarkers, even ones with low sensitivity, have the potential to serve as an important indicator for a tumor-targeted drug. This unexpected assistance by the immune system provides cancer researchers with powerful research tools to unlock important clues to our understanding of oncogenesis and ultimately lead to better cancer treatment strategies.

Author Contributions

Writing—Original Draft Preparation, X.W. and M.X.; Writing—Review & Editing, L.H., R.A.K. and J.-Y.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grant 5U54MD007592 from the National Institutes on Minority Health and Health Disparities (NIMHD), a component of the National Institutes of Health (NIH). This work was partly supported by the National Natural Science Foundation of China (81703626), the China Scholarship Council and basic research funding of China Pharmaceutical University (2632021PY05).

Conflicts of Interest

The authors declare that they have no financial conflict of interest.

References

  1. Hilmi, M.; Vienot, A.; Rousseau, B.; Neuzillet, C. Immune Therapy for Liver Cancers. Cancers 2019, 12, 77. [Google Scholar] [CrossRef] [Green Version]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  3. Altekruse, S.F.; McGlynn, K.A.; Reichman, M.E. Hepatocellular Carcinoma Incidence, Mortality, and Survival Trends in the United States From 1975 to 2005. J. Clin. Oncol. 2009, 27, 1485–1491. [Google Scholar] [CrossRef] [Green Version]
  4. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics. CA A Cancer J. Clin. 2013, 63, 11–30. [Google Scholar] [CrossRef] [Green Version]
  5. Takayama, T.; Makuuchi, M.; Kojiro, M.; Lauwers, G.Y.; Adams, R.B.; Wilson, S.R.; Jang, H.-J.; Charnsangavej, C.; Taouli, B. Early Hepatocellular Carcinoma: Pathology, Imaging, and Therapy. Ann. Surg. Oncol. 2008, 15, 972–978. [Google Scholar] [CrossRef] [PubMed]
  6. Tsuchiya, N.; Sawada, Y.; Endo, I.; Saito, K.; Uemura, Y.; Nakatsura, T. Biomarkers for the early diagnosis of hepatocellular carci-noma. World J. Gastroenterol. 2015, 21, 10573–10583. [Google Scholar] [CrossRef] [PubMed]
  7. Huo, T.-I.; Hsu, C.-Y.; Liu, P.-H. Magic mirror on the wall: Which is the best biomarker for hepatocellular carcinoma? Hepatology 2018, 67, 2482–2483. [Google Scholar] [CrossRef] [Green Version]
  8. Collier, J.; Sherman, M. Screening for hepatocellular carcinoma. Hepatology 1998, 27, 273–278. [Google Scholar] [CrossRef] [PubMed]
  9. Liu, W.H.; Ren, L.N.; Wang, X.; Wang, T.; Zhang, N.; Gao, Y.; Luo, H.; Navarro-Alvarez, N.; Tang, L.J. Combination of exosomes and circulating microRNAs may serve as a promising tumor marker complementary to alpha-fetoprotein for early-stage hepato-cellular carcinoma diagnosis in rats. J. Cancer Res. Clin. Oncol. 2015, 141, 1767–1778. [Google Scholar] [CrossRef]
  10. Labgaa, A.; Villanueva, I. Liquid biopsy in liver cancer. Discov. Med. 2015, 19, 263–273. [Google Scholar]
  11. Bogdanos, D.P.; Gao, B.; Gershwin, M.E. Liver Immunology. Compr. Physiol. 2013, 3, 567–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Tampaki, M.; Ionas, E.; Hadziyannis, E.; Deutsch, M.; Malagari, K.; Koskinas, J. Association of TIM-3 with BCLC Stage, Serum PD-L1 Detection, and Response to Transarterial Chemoembolization in Patients with Hepatocellular Carcinoma. Cancers 2020, 12, 212. [Google Scholar] [CrossRef] [Green Version]
  13. Baldwin, R.W. Tumour-associated antigens and tumour-host interactions. Proc. R. Soc. Med. 1971, 64, 1039–1042. [Google Scholar] [PubMed]
  14. Zhang, H.-F.; Qin, J.-J.; Ren, P.-F.; Shi, J.-X.; Xia, J.-F.; Ye, H.; Wang, P.; Song, C.-H.; Wang, K.-J.; Zhang, J.-Y. A panel of autoantibodies against multiple tumor-associated antigens in the immunodiagnosis of esophageal squamous cell cancer. Cancer Immunol. Immunother. 2016, 65, 1233–1242. [Google Scholar] [CrossRef]
  15. Zhou, J.-W.; Li, Y.; Yue, L.-X.; Luo, C.-L.; Chen, Y.; Zhang, J.-Y. Autoantibody response to Sui1 and its tissue-specific expression in hepatocellular carcinoma. Tumor Biol. 2015, 37, 2547–2553. [Google Scholar] [CrossRef] [PubMed]
  16. Liu, M.; Varela-Ramirez, A.; Li, J.; Dai, L.; Aguilera, R.J.; Zhang, J.-Y. Humoral autoimmune response to nucleophosmin in the immunodiagnosis of hepatocellular carcinoma. Oncol. Rep. 2015, 33, 2245–2252. [Google Scholar] [CrossRef] [Green Version]
  17. Zhang, J.-Y.; Tan, E.M. Autoantibodies to tumor-associated antigens as diagnostic biomarkers in hepatocellular carcinoma and other solid tumors. Expert Rev. Mol. Diagn. 2010, 10, 321–328. [Google Scholar] [CrossRef] [Green Version]
  18. Anderson, K.S.; LaBaer, J. The Sentinel Within: Exploiting the Immune System for Cancer Biomarkers. J. Proteome Res. 2005, 4, 1123–1133. [Google Scholar] [CrossRef] [Green Version]
  19. Dai, L.; Ren, P.; Liu, M.; Imai, H.; Tan, E.M.; Zhang, J.-Y. Using immunomic approach to enhance tumor-associated autoantibody detection in diagnosis of hepatocellular carcinoma. Clin. Immunol. 2014, 152, 127–139. [Google Scholar] [CrossRef] [Green Version]
  20. Koziol, J.A.; Imai, H.; Dai, L.; Zhang, J.-Y.; Tan, E.M. Early detection of hepatocellular carcinoma using autoantibody profiles from a panel of tumor-associated antigens. Cancer Immunol. Immunother. 2018, 67, 835–841. [Google Scholar] [CrossRef]
  21. Schulze, K.; Imbeaud, S.; Letouzé, E.; Alexandrov, L.B.; Calderaro, J.; Rebouissou, S.; Couchy, G.; Meiller, C.; Shinde, J.; Soysouvanh, F.; et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 2015, 47, 505–511. [Google Scholar] [CrossRef] [PubMed]
  22. Suppiah, A.; Greenman, J. Clinical utility of anti-p53 auto-antibody: Systematic review and focus on colorectal cancer. World J. Gastroenterol. 2013, 19, 4651–4670. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, J.-Y.; Chan, E.K.; Peng, X.-X.; Tan, E.M. A Novel Cytoplasmic Protein with RNA-binding Motifs Is an Autoantigen in Human Hepatocellular Carcinoma. J. Exp. Med. 1999, 189, 1101–1110. [Google Scholar] [CrossRef] [PubMed]
  24. Nielsen, J.; Christiansen, J.; Lykke-Andersen, J.; Johnsen, A.H.; Wewer, U.M.; Nielsen, F.C. A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development. Mol. Cell Biol. 1999, 19, 1262–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mueller-Pillasch, F.; Lacher, U.; Wallrapp, C.; Micha, A.; Zimmerhackl, F.; Hameister, H.; Varga, G.; Friess, H.; Büchler, M.; Beger, H.G.; et al. Cloning of a gene highly overexpressed in cancer coding for a novel KH-domain containing protein. Oncogene 1997, 14, 2729–2733. [Google Scholar] [CrossRef] [Green Version]
  26. Kessler, S.M.; Laggai, S.; Barghash, A.E.M.; Schultheiss, C.S.; Lederer, E.; Artl, M.; Helms, V.; Haybaeck, J.; Kiemer, A.K. IMP2/p62 induces genomic instability and an aggressive hepatocellular carcinoma phenotype. Cell Death Dis. 2015, 6, e1894. [Google Scholar] [CrossRef] [Green Version]
  27. Xing, M.; Li, P.; Wang, X.; Li, J.; Shi, J.; Qin, J.; Zhang, X.; Ma, Y.; Francia, G.; Zhang, J.Y. Overexpression of p62/IMP2 can Promote Cell Migration in Hepatocellular Carcinoma via Activation of the Wnt/beta-Catenin Pathway. Cancers 2019, 12, 7. [Google Scholar] [CrossRef] [Green Version]
  28. Wachter, D.L.; Kristiansen, G.; Soll, C.; Hellerbrand, C.; Breuhahn, K.; Fritzsche, F.; Agaimy, A.; Hartmann, A.; Riener, M.O. Insu-lin-like growth factor II mRNA-binding protein 3 (IMP3) expression in hepatocellular carcinoma. A clinicopathological analysis with emphasis on diagnostic value. Histopathology 2012, 60, 278–286. [Google Scholar] [CrossRef] [PubMed]
  29. Degrauwe, N.; Suvà, M.-L.; Janiszewska, M.; Riggi, N.; Stamenkovic, I. IMPs: An RNA-binding protein family that provides a link between stem cell maintenance in normal development and cancer. Genes Dev. 2016, 30, 2459–2474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Kessler, S.; Haybaeck, J.; Kiemer, A. Insulin-Like Growth Factor 2—The Oncogene and its Accomplices. Curr. Pharm. Des. 2016, 22, 5948–5961. [Google Scholar] [CrossRef] [PubMed]
  31. Rogler, C.E.; Yang, D.; Rossetti, L.; Donohoe, J.; Alt, E.; Chang, C.J.; Rosenfeld, R.; Neely, K.; Hintz, R. Altered body composition and increased frequency of diverse malignancies in insulin-like growth factor-II transgenic mice. J. Biol. Chem. 1994, 269, 13779–13784. [Google Scholar] [CrossRef]
  32. Agrogiannis, G.D.; Sifakis, S.; Patsouris, E.S.; Konstantinidou, A.E. Insulin-like growth factors in embryonic and fetal growth and skeletal development (Review). Mol. Med. Rep. 2014, 10, 579–584. [Google Scholar] [CrossRef] [Green Version]
  33. Cariani, E.; Lasserre, C.; Seurin, D.; Hamelin, B.; Kemeny, F.; Franco, D.; Czech, M.P.; Ullrich, A.; Brechot, C. Differential expression of insulin-like growth factor II mRNA in human primary liver cancers, benign liver tumors, and liver cirrhosis. Cancer Res. 1988, 48, 6844–6849. [Google Scholar] [PubMed]
  34. Christofori, G.; Naik, P.; Hanahan, D. A second signal supplied by insulin-like growth factor II in oncogene-induced tumorigenesis. Nat. Cell Biol. 1994, 369, 414–418. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, H.; Zhang, Y.; Li, S.; Li, N.; Chen, Y.; Zhang, B.; Qu, C.; Ding, H.; Huang, J.; Dai, M. Direct comparison of five serum biomarkers in early diagnosis of hepatocellular carcinoma. Cancer Manag. Res. 2018, 10, 1947–1958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ren, B.; Zou, G.; Xu, F.; Huang, Y.; Xu, G.; He, J.; Li, Y.; Zhu, H.; Yu, P. Serum levels of anti-sperm-associated antigen 9 antibody are elevated in patients with hepatocellular carcinoma. Oncol. Lett. 2017, 14, 7608–7614. [Google Scholar] [CrossRef]
  37. Ren, B.; Luo, S.; Xu, F.; Zou, G.; Xu, G.; He, J.; Huang, Y.; Zhu, H.; Li, Y. The expression of DAMP proteins HSP70 and cancer-testis antigen SPAG9 in peripheral blood of patients with HCC and lung cancer. Cell Stress Chaperon 2016, 22, 237–244. [Google Scholar] [CrossRef]
  38. Liu, M.; Liu, X.; Ren, P.; Li, J.; Chai, Y.; Zheng, S.J.; Chen, Y.; Duan, Z.P.; Li, N.; Zhang, J.Y. A cancer-related protein 14-3-3zeta is a potential tumor-associated antigen in immunodiagnosis of hepatocellular carcinoma. Tumour Biol. 2014, 35, 4247–4256. [Google Scholar] [CrossRef] [Green Version]
  39. Liu, M.; Zheng, S.-J.; Chen, Y.; Li, N.; Ren, P.-F.; Dai, L.-P.; Duan, Z.-P.; Zhang, J.-Y. Autoantibody Response to Murine Double Minute 2 Protein in Immunodiagnosis of Hepatocellular Carcinoma. J. Immunol. Res. 2014, 2014, 1–10. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, H.; Zhang, J.; Wang, S.; Pang, Z.; Wang, Z.; Zhou, W.; Wu, M. Screening of autoantibodies as potential biomarkers for hepa-tocellular carcinoma by using T7 phase display system. Cancer Epidemiol. 2012, 36, 82–88. [Google Scholar] [CrossRef]
  41. Li, L.; Chen, S.-H.; Yu, C.-H.; Li, Y.-M.; Wang, S.-Q. Identification of Hepatocellular-Carcinoma-Associated Antigens and Autoantibodies by Serological Proteome Analysis Combined with Protein Microarray. J. Proteome Res. 2008, 7, 611–620. [Google Scholar] [CrossRef]
  42. Heo, C.-K.; Hwang, H.-M.; Lee, H.-J.; Kwak, S.-S.; Yoo, J.-S.; Yu, D.-Y.; Lim, K.-J.; Lee, S.; Cho, E.-W. Serum anti-EIF3A autoantibody as a potential diagnostic marker for hepatocellular carcinoma. Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  43. Hwang, H.M.; Heo, C.K.; Lee, H.J.; Kwak, S.S.; Lim, W.H.; Yoo, J.S.; Yu, D.Y.; Lim, K.J.; Kim, J.Y.; Cho, E.W. Identification of an-ti-SF3B1 autoantibody as a diagnostic marker in patients with hepatocellular carcinoma. J. Transl. Med. 2018, 16, 177. [Google Scholar] [CrossRef]
  44. Chao, N.X.; Li, L.Z.; Luo, G.R.; Zhong, W.G.; Huang, R.S.; Fan, R.; Zhao, F.L. Cancer-testis antigen GAGE-1 expression and serum immunoreactivity in hepatocellular carcinoma. Niger. J. Clin. Pract. 2018, 21, 1361–1367. [Google Scholar]
  45. Dai, L.; Peng, X.X.; Tan, E.M.; Zhang, J.Y. Tumor-associated antigen CAPERalpha and microvessel density in hepatocellular car-cinoma. Oncotarget 2016, 7, 16985–16995. [Google Scholar] [CrossRef] [Green Version]
  46. Oshima, Y.; Shimada, H.; Yajima, S.; Nanami, T.; Matsushita, K.; Nomura, F.; Kainuma, O.; Takiguchi, N.; Soda, H.; Ueda, T.; et al. NY-ESO-1 autoantibody as a tumor-specific biomarker for esophageal cancer: Screening in 1969 patients with various cancers. J. Gastroenterol. 2016, 51, 30–34. [Google Scholar] [CrossRef] [PubMed]
  47. Middleton, C.H.; Irving, W.; Robertson, J.F.R.; Murray, A.; Parsy-Kowalska, C.B.; Macdonald, I.K.; McElveen, J.; Allen, J.; Healey, G.F.; Thomson, B.J.; et al. Serum Autoantibody Measurement for the Detection of Hepatocellular Carcinoma. PLoS ONE 2014, 9, e103867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Chen, Y.; Zhou, Y.; Qiu, S.; Wang, K.; Liu, S.; Peng, X.-X.; Li, J.; Tan, E.M.; Zhang, J.-Y. Autoantibodies to tumor-associated antigens combined with abnormal alpha-fetoprotein enhance immunodiagnosis of hepatocellular carcinoma. Cancer Lett. 2010, 289, 32–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Heo, C.K.; Woo, M.K.; Yu, D.Y.; Lee, J.Y.; Yoo, J.S.; Yoo, H.S.; Ko, J.H.; Kim, J.M.; Choi, J.Y.; Kim, I.G.; et al. Identifi-cation of autoantibody against fatty acid synthase in hepatocellular carcinoma mouse model and its application to diagnosis of HCC. Int. J. Oncol. 2010, 36, 1453–1459. [Google Scholar]
  50. Wang, K.; Chen, Y.; Liu, S.; Qiu, S.; Gao, S.; Huang, X.; Zhang, J.; Peng, X.; Qiani, W. Immunogenicity of Ra1A and its Tissue-Specific Expression in Hepatocellular Carcinoma. Int. J. Immunopathol. Pharmacol. 2009, 22, 735–743. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, K.; Xu, X.; Nie, Y.; Dai, L.; Wang, P.; Zhang, J. Identification of tumor-associated antigens by using SEREX in hepatocellular carcinoma. Cancer Lett. 2009, 281, 144–150. [Google Scholar] [CrossRef]
  52. Looi, K.S.; Nakayasu, E.S.; de Diaz, R.A.; Tan, E.M.; Almeida, I.C.; Zhang, J.-Y. Using Proteomic Approach to Identify Tumor-Associated Antigens as Markers in Hepatocellular Carcinoma. J. Proteome Res. 2008, 7, 4004–4012. [Google Scholar] [CrossRef] [Green Version]
  53. Chen, X.; Fu, S.; Chen, F.; Chen, H.; Chen, Z. Identification of tumor-associated antigens in human hepatocellular carcinoma by autoantibodies. Oncol. Rep. 2008, 20, 979–985. [Google Scholar] [PubMed]
  54. Zhang, J.-Y.; Megliorino, R.; Peng, X.-X.; Tan, E.M.; Chen, Y.; Chan, E.K. Antibody detection using tumor-associated antigen mini-array in immunodiagnosing human hepatocellular carcinoma. J. Hepatol. 2007, 46, 107–114. [Google Scholar] [CrossRef] [Green Version]
  55. Takashima, M.; Kuramitsu, Y.; Yokoyama, Y.; Iizuka, N.; Harada, T.; Fujimoto, M.; Sakaida, I.; Okita, K.; Oka, M.; Nakamura, K. Proteomic analysis of autoantibodies in patients with hepatocellular carcinoma. Proteomics 2006, 6, 3894–3900. [Google Scholar] [CrossRef] [PubMed]
  56. Himoto, T.; Kuriyama, S.; Zhang, J.Y.; Chan, E.K.; Kimura, Y.; Masaki, T.; Uchida, N.; Nishioka, M.; Tan, E.M. Analyses of autoan-tibodies against tumor-associated antigens in patients with hepatocellular carcinoma. Int. J. Oncol. 2005, 27, 1079–1085. [Google Scholar] [PubMed]
  57. Looi, K.; Megliorino, R.; Shi, F.-D.; Peng, X.-X.; Chen, Y.; Zhang, J.-Y. Humoral immune response to p16, a cyclin-dependent kinase inhibitor in human malignancies. Oncol. Rep. 2006, 16, 1105–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Zhang, J.-Y.; Casiano, C.A.; Peng, X.-X.; Koziol, J.A.; Chan, E.K.L.; Tan, E.M. Enhancement of antibody detection in cancer using panel of recombinant tumor-associated antigens. Cancer Epidemiol. Biomark. Prev. 2003, 12, 136–143. [Google Scholar]
  59. Koziol, J.A.; Zhang, J.-Y.; Casiano, C.A.; Peng, X.-X.; Shi, F.-D.; Feng, A.C.; Chan, E.K.L.; Tan, E.M. Recursive partitioning as an approach to selection of immune markers for tumor diagnosis. Clin. Cancer Res. 2003, 9, 5120–5126. [Google Scholar]
  60. Soo Hoo, L.; Zhang, J.Y.; Chan, E.K. Cloning and characterization of a novel 90 kDa ’companion’ auto-antigen of p62 overex-pressed in cancer. Oncogene 2002, 21, 5006–5015. [Google Scholar] [CrossRef] [Green Version]
  61. Le Naour, F.; Brichory, F.; Misek, D.E.; Bréchot, C.; Hanash, S.M.; Beretta, L. A Distinct Repertoire of Autoantibodies in Hepatocellular Carcinoma Identified by Proteomic Analysis. Mol. Cell. Proteom. 2002, 1, 197–203. [Google Scholar] [CrossRef] [Green Version]
  62. Zhang, W.; Zhu, H.; Imai, K.; Kiyosawa, E.K.; Chan, E.M.; Tan, J.Y. De-novo humoral immune responses to cancer-associated autoantigens during transition from chronic liver disease to hepatocellular carcinoma. Clin. Exp. Immunol. 2001, 125, 3–9. [Google Scholar] [CrossRef] [PubMed]
  63. Raedle, J.; Oremek, G.; Truschnowitsch, M.; Lorenz, M.; Roth, W.; Caspary, W.; Zeuzem, S. Clinical evaluation of autoantibodies to p53 protein in patients with chronic liver disease and hepatocellular carcinoma. Eur. J. Cancer 1998, 34, 1198–1203. [Google Scholar] [CrossRef]
  64. Edis, C.; Kähler, C.; Klotz, W.; Herold, M.; Feichtinger, H.; Königsreiner, A.; Margreiter, R.; Jaschke, W.; Vogel, W. A Comparison Between α-Fetoprotein and P53 Antibodies in the Diagnosis of Hepatocellular Carcinoma. Transplant. Proc. 1998, 30, 780–781. [Google Scholar] [CrossRef]
  65. Covini, E.K.; Chan, M.; Nishioka, S.A.; Morshed, S.I.; Reed, E.M.; Tan, G. Immune response to cyclin B1 in hepatocellular carcinoma. Hepatology 1997, 25, 75–80. [Google Scholar] [CrossRef]
  66. Shao, Q.; Ren, P.; Li, Y.; Peng, B.; Dai, L.; Lei, N.; Yao, W.; Zhao, G.; Li, L.; Zhang, J. Autoantibodies against glucose-regulated protein 78 as serological diagnostic biomarkers in hepatocellular carcinoma. Int. J. Oncol. 2012, 41, 1061–1067. [Google Scholar] [CrossRef] [Green Version]
  67. Ying, S.X.; Han, C.C.; He, C.Y.; Zhou, Y.P.; Dong, M.J.; Cai, X.; Sui, C.X.; Ma, X.; Sun, Y.Y.; Zhang, W.L.; et al. Autoantibodies against glucose-regulated protein 78 as serological biomarkers in metastatic and recurrent hepatocellular car-cinoma. Oncotarget 2017, 8, 24828–24839. [Google Scholar] [CrossRef] [Green Version]
  68. Ying, X.; Zhao, Y.; Wang, J.-L.; Zhou, X.; Zhao, J.; He, C.-C.; Guo, X.-J.; Jin, G.-H.; Wang, L.-J.; Zhu, Q.; et al. Serum anti-osteopontin autoantibody as a novel diagnostic and prognostic biomarker in patients with hepatocellular carcinoma. Oncol. Rep. 2014, 32, 1550–1556. [Google Scholar] [CrossRef] [Green Version]
  69. Takeji, S.; Hirooka, M.; Koizumi, Y.; Tokumoto, Y.; Abe, M.; Ikeda, Y.; Nadano, S.; Hiasa, Y.; Onji, M. Des-gamma-carboxy pro-thrombin identified by P-11 and P-16 antibodies reflects prognosis for patients with hepatocellular carcinoma. J. Gastroenterol. Hepatol. 2013, 28, 671–677. [Google Scholar] [CrossRef] [PubMed]
  70. Su, Y.; Qian, H.; Zhang, J.; Wang, S.; Shi, P.; Peng, X. The diversity expression of p62 in digestive system cancers. Clin. Immunol. 2005, 116, 118–123. [Google Scholar] [CrossRef]
  71. Zhang, X.; Wang, J.; Xu, Y.; Zhao, H. Change of circulating antibodies against CD25-derived peptide antigen in hepatocellular carcinoma. J. Cancer Res. Ther. 2017, 13, 813. [Google Scholar] [CrossRef]
  72. Yu, Y.-Q.; Wang, L.; Jin, Y.; Zhou, J.-L.; Geng, Y.-H.; Jin, X.; Zhang, X.-X.; Yang, J.-J.; Qian, C.-M.; Zhou, D.-E.; et al. Identification of serologic biomarkers for predicting microvascular invasion in hepatocellular carcinoma. Oncotarget 2016, 7, 16362–16371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zheng, S.-X.; Xiang, B.-D.; Long, J.-M.; Qu, C.; Mo, Z.-J.; Li, K.; Zhuang, Y.; Lv, Z.-L.; Zhou, S.-F. Diagnostic Value of Serum SMP30 and Anti-SMP30 Antibody in Hepatocellular Carcinoma. Lab. Med. 2018, 49, 203–210. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, M.; Liu, S.J.; Zheng, D.D.; Bian, J.Y.; Zhang, J.; Yao, Q.F.; Zheng, A.M.; Shi, W.H.; Li, L.; Li, Y.; et al. Tumor-associated autoantibodies are useful biomarkers in immunodiagnosis of alpha-fetoprotein-negative hepato-cellular carcinoma. World J. Gastroenterol. 2017, 23, 3496–3504. [Google Scholar] [CrossRef]
  75. Zhou, S.-F.; Xie, X.-X.; Bin, Y.-H.; Lan, L.; Chen, F.; Luo, G.-R. Identification of HCC-22-5 tumor-associated antigen and antibody response in patients. Clin. Chim. Acta 2006, 366, 274–280. [Google Scholar] [CrossRef] [PubMed]
  76. Himoto, S.; Kuriyama, J.Y.; Zhang, E.K.; Chan, M.; Nishioka, E.M.; Tan, T. Significance of autoantibodies against insulin-like growth factor II mRNA-binding proteins in patients with hepatocellular carcinoma. Int. J. Oncol. 2005, 26, 311–317. [Google Scholar] [CrossRef] [PubMed]
  77. Xu, Y.; Liu, A.J.; Gao, Y.X.; Hu, M.G.; Zhao, G.D.; Zhao, Z.M.; Liu, R. Expression of Ku86 and presence of Ku86 antibody as bi-omarkers of hepatitis B virus related hepatocellular carcinoma. Dig. Dis. Sci. 2014, 59, 614–622. [Google Scholar] [CrossRef]
  78. Yau, W.-Y.; Shih, H.-C.; Tsai, M.-H.; Sheu, J.-C.; Chen, C.-H.; Chow, L.-P. Autoantibody recognition of an N-terminal epitope of hnRNP L marks the risk for developing HBV-related hepatocellular carcinoma. J. Proteom. 2013, 94, 346–358. [Google Scholar] [CrossRef]
  79. Akada, J.; Kamei, S.; Ito, A.; Ito, M.; Kitagawa, T.; Furumoto, H.; Kato, Y.; Tamesa, M.; Takashima, M.; Shirai, M.; et al. A new type of protein chip to detect hepatocellular carcinoma-related autoimmune antibodies in the sera of hepatitis C virus-positive patients. Proteome Sci. 2013, 11, 33. [Google Scholar] [CrossRef] [Green Version]
  80. Nomura, F.; Sogawa, K.; Noda, K.; Seimiya, M.; Matsushita, K.; Miura, T.; Tomonaga, T.; Yoshitomi, H.; Imazeki, F.; Takizawa, H.; et al. Serum anti-Ku86 is a potential biomarker for early detection of hepatitis C virus-related hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2012, 421, 837–843. [Google Scholar] [CrossRef] [PubMed]
  81. Ezzikouri, S.; Kimura, K.; Sunagozaka, H.; Kaneko, S.; Inoue, K.; Nishimura, T.; Hishima, T.; Kohara, M.; Tsukiyama-Kohara, K. Serum DHCR24 Auto-antibody as a new Biomarker for Progression of Hepatitis, C. eBioMedicine 2015, 2, 604–612. [Google Scholar] [CrossRef] [Green Version]
  82. Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 2019, 49, 347–360. [Google Scholar] [CrossRef]
  83. Sun, F.; Wang, J.-Z.; Luo, J.-J.; Wang, Y.-Q.; Pan, Q. Exosomes in the Oncobiology, Diagnosis, and Therapy of Hepatic Carcinoma: A New Player of an Old Game. BioMed Res. Int. 2018, 2018, 1–10. [Google Scholar] [CrossRef]
  84. Bach, D.-H.; Hong, J.-Y.; Park, H.J.; Lee, S.K. The role of exosomes and miRNAs in drug-resistance of cancer cells. Int. J. Cancer 2017, 141, 220–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Liu, H.; Li, B. The functional role of exosome in hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 2018, 144, 2085–2095. [Google Scholar] [CrossRef] [PubMed]
  86. Tomimaru, Y.; Eguchi, H.; Nagano, H.; Wada, H.; Kobayashi, S.; Marubashi, S.; Tanemura, M.; Tomokuni, A.; Takemasa, I.; Umeshita, K.; et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J. Hepatol. 2012, 56, 167–175. [Google Scholar] [CrossRef]
  87. Wang, L.; Hou, A.; Li, Y.; Duan, H.; Gao, X.; Song, H. Expression of serum exosomal microRNA-21 in human hepatocellular car-cinoma. BioMed Res. Int. 2014, 2014, 864894. [Google Scholar]
  88. Xu, J.; Wu, C.; Che, X.; Wang, L.; Yu, D.; Zhang, T.; Huang, L.; Li, H.; Tan, W.; Wang, C.; et al. Circulating MicroRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol. Carcinog. 2011, 50, 136–142. [Google Scholar] [CrossRef]
  89. Qi, P.; Cheng, S.-Q.; Wang, H.; Li, N.; Chen, Y.-F.; Gao, C.-F. Serum MicroRNAs as Biomarkers for Hepatocellular Carcinoma in Chinese Patients with Chronic Hepatitis B Virus Infection. PLoS ONE 2011, 6, e28486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Szabo, G.; Bala, S. MicroRNAs in liver disease. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 542–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Halasz, G.; Horvath, G.; Par, K.; Werling, A.; Kiss, Z.; Schaff, G.; Lendvai, T. miR-122 negatively correlates with liver fibrosis as detected by histology and FibroScan. World J. Gastroenterol. 2015, 21, 7814–7823. [Google Scholar] [CrossRef] [Green Version]
  92. Conrad, K.D.; Giering, F.; Erfurth, C.; Neumann, A.; Fehr, C.; Meister, G.; Niepmann, M. microRNA-122 Dependent Binding of Ago2 Protein to Hepatitis C Virus RNA Is Associated with Enhanced RNA Stability and Translation Stimulation. PLoS ONE 2013, 8, e56272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ding, J.; Ding, X.; Ning, J.; Yi, F.; Chen, J.; Zhao, D.; Zheng, J.; Liang, Z.; Hu, Z.; Du, Q. Circulating microRNA-122 as a potential biomarker for liver injury. Mol. Med. Rep. 2012, 5, 1428–1432. [Google Scholar] [CrossRef] [PubMed]
  94. Suehiro, T.; Miyaaki, H.; Kanda, Y.; Shibata, H.; Honda, T.; Ozawa, E.; Miuma, S.; Taura, N.; Nakao, K. Serum exosomal mi-croRNA-122 and microRNA-21 as predictive biomarkers in transarterial chemoembolization-treated hepatocellular carcinoma patients. Oncol. Lett. 2018, 16, 3267–3273. [Google Scholar] [PubMed]
  95. Sohn, J.; Kim, S.H.; Kang, S.R.; Yang, J.Y.; Cho, H.C.; Cho, S.G.; Shim, Y.H.; Paik, J. Serum exosomal microRNAs as novel bi-omarkers for hepatocellular carcinoma. Exp. Mol. Med. 2015, 47, e184. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, W.; Hu, J.; Zhou, K.; Chen, F.; Wang, Z.; Liao, B.; Dai, Z.; Cao, Y.; Fan, J.; Zhou, J. Serum exosomal miR-125b is a novel prognostic marker for hepatocellular carcinoma. OncoTargets Ther. 2017, 10, 3843–3851. [Google Scholar] [CrossRef] [Green Version]
  97. Lee, Y.R.; Kim, G.; Tak, W.Y.; Jang, S.Y.; Kweon, Y.O.; Gil Park, J.; Lee, H.W.; Han, Y.S.; Chun, J.M.; Park, S.Y.; et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int. J. Cancer 2019, 144, 1444–1452. [Google Scholar] [CrossRef] [Green Version]
  98. Yuan, F.; Yang, F.; Wang, J.Z.; Ma, Y.J.; Guo, Q.F.; Tao, F.; Liu, W.; Pan, T.T.; Wang, C.C.; Zhou, S.B.; et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014, 25, 666–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Sugimachi, T.; Matsumura, H.; Hirata, R.; Uchi, M.; Ueda, H.; Ueo, Y.; Shinden, T.; Iguchi, H.; Eguchi, K.; Shirabe, T.; et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular car-cinoma recurrence after liver transplantation. Br. J. Cancer 2015, 112, 532–538. [Google Scholar] [CrossRef]
  100. Hu, G.; Drescher, K.M.; Chen, X.-M. Exosomal miRNAs: Biological Properties and Therapeutic Potential. Front. Genet. 2012, 3, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wei, J.-X.; Lv, L.-H.; Wan, Y.-L.; Cao, Y.; Li, G.-L.; Lin, H.-M.; Zhou, R.; Shang, C.-Z.; Cao, J.; He, H.; et al. Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology 2015, 61, 1284–1294. [Google Scholar] [CrossRef]
  102. Arbelaiz, A.; Azkargorta, M.; Krawczyk, M.; Santos-Laso, A.; Lapitz, A.; Perugorria, M.J.; Erice, O.; Gonzalez, E.; Jimenez-Agüero, R.; Lacasta, A.; et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2017, 66, 1125–1143. [Google Scholar] [CrossRef] [PubMed]
  103. Jiang, S.-S.; Weng, D.-S.; Wang, Q.-J.; Pan, K.; Zhang, Y.-J.; Li, Y.-Q.; Li, J.-J.; Zhao, J.-J.; He, J.; Lv, L.; et al. Galectin-3 is associated with a poor prognosis in primary hepatocellular carcinoma. J. Transl. Med. 2014, 12, 273. [Google Scholar] [CrossRef] [Green Version]
  104. Kogure, T.; Lin, W.-L.; Yan, I.K.; Braconi, C.; Patel, T. Intercellular nanovesicle-mediated microRNA transfer: A mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology 2011, 54, 1237–1248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Huang, T.; Deng, C.-X. Current Progresses of Exosomes as Cancer Diagnostic and Prognostic Biomarkers. Int. J. Biol. Sci. 2019, 15, 1–11. [Google Scholar] [CrossRef] [PubMed]
  106. Li, X.; Wang, X. The emerging roles and therapeutic potential of exosomes in epithelial ovarian cancer. Mol. Cancer 2017, 16, 1–10. [Google Scholar] [CrossRef] [Green Version]
  107. Wu, Q.; Zhou, L.; Lv, D.; Zhu, X.; Tang, H. Exosome-mediated communication in the tumor microenvironment contributes to hepatocellular carcinoma development and progression. J. Hematol. Oncol. 2019, 12, 53. [Google Scholar] [CrossRef]
  108. Chen, R.; Xu, X.; Tao, Y.; Qian, Z.; Yu, Y. Exosomes in hepatocellular carcinoma: A new horizon. Cell Commun. Signal. 2019, 17, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Rao, Q.; Zuo, B.; Lu, Z.; Gao, X.; You, A.; Wu, C.; Du, Z.; Yin, H. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology 2016, 64, 456–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Lu, Z.; Zuo, B.; Jing, R.; Gao, X.; Rao, Q.; Liu, Z.; Qi, H.; Guo, H.; Yin, H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J. Hepatol. 2017, 67, 739–748. [Google Scholar] [CrossRef] [PubMed]
  111. Li, L.-M.; Liu, Z.-X.; Cheng, Q.-Y. Exosome plays an important role in the development of hepatocellular carcinoma. Pathol. Res. Pract. 2019, 215, 152468. [Google Scholar] [CrossRef]
  112. Xu, H.; Dong, X.; Chen, Y.; Wang, X. Serum exosomal hnRNPH1 mRNA as a novel marker for hepatocellular carcinoma. Clin. Chem. Lab. Med. 2018, 56, 479–484. [Google Scholar] [CrossRef]
  113. Zhang, C.; Yang, X.; Qi, Q.; Gao, Y.; Wei, Q.; Han, S. lncRNA-HEIH in serum and exosomes as a potential biomarker in the HCV-related hepatocellular carcinoma. Cancer Biomark. 2018, 21, 651–659. [Google Scholar] [CrossRef] [PubMed]
  114. Tang, J.; Li, Y.; Liu, K.; Zhu, Q.; Yang, W.-H.; Xiong, L.-K.; Guo, D.-L. Exosomal miR-9-3p suppresses HBGF-5 expression and is a functional biomarker in hepatocellular carcinomaa. Minerva Med. 2017, 109, 15–23. [Google Scholar] [PubMed]
  115. Li, B.; Mao, R.; Liu, C.; Zhang, W.; Tang, Y.; Guo, Z. LncRNA FAL1 promotes cell proliferation and migration by acting as a CeRNA of miR-1236 in hepatocellular carcinoma cells. Life Sci. 2018, 197, 122–129. [Google Scholar] [CrossRef] [PubMed]
  116. Fornari, F.; Ferracin, M.; Trerè, D.; Milazzo, M.; Marinelli, S.; Galassi, M.; Venerandi, L.; Pollutri, D.; Patrizi, C.; Borghi, A.; et al. Circulating microRNAs, miR-939, miR-595, miR-519d and miR-494, Identify Cirrhotic Patients with HCC. PLoS ONE 2015, 10, e0141448. [Google Scholar] [CrossRef] [Green Version]
  117. Xue, X.; Zhao, Y.; Wang, X.; Qin, L.; Hu, R. Development and validation of serum exosomal microRNAs as diagnostic and prognostic biomarkers for hepatocellular carcinoma. J. Cell. Biochem. 2019, 120, 135–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Xue, X.; Wang, X.; Zhao, Y.; Hu, R.; Qin, L. Exosomal miR-93 promotes proliferation and invasion in hepatocellular carcinoma by directly inhibiting TIMP2/TP53INP1/CDKN1A. Biochem. Biophys. Res. Commun. 2018, 502, 515–521. [Google Scholar] [CrossRef]
  119. Qu, Z.; Wu, J.; Wu, J.; Ji, A.; Qiang, G.; Jiang, Y.; Jiang, C.; Ding, Y. Exosomal miR-665 as a novel minimally invasive biomarker for hepatocellular carcinoma diagnosis and prognosis. Oncotarget 2017, 8, 80666–80678. [Google Scholar] [CrossRef] [Green Version]
  120. Xu, H.; Chen, Y.; Dong, X.; Wang, X. Serum Exosomal Long Noncoding RNAs ENSG00000258332.1 and LINC00635 for the Diagnosis and Prognosis of Hepatocellular Carcinoma. Cancer Epidemiol. Biomark. Prev. 2018, 27, 710–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Fang, J.H.; Zhang, Z.J.; Shang, L.R.; Luo, Y.W.; Lin, Y.F.; Yuan, Y.; Zhuang, S.M. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018, 68, 1459–1475. [Google Scholar] [CrossRef] [Green Version]
  122. Gramantieri, L.; Baglioni, M.; Fornari, F.; Laginestra, M.A.; Ferracin, M.; Indio, V.; Ravaioli, M.; Cescon, M.; De Pace, V.; Leoni, S.; et al. LncRNAs as novel players in hepatocellular carcinoma recurrence. Oncotarget 2018, 9, 35085–35099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Shi, M.; Jiang, Y.; Yang, L.; Yan, S.; Wang, Y.; Lu, X. Decreased levels of serum exosomal miR-638 predict poor prognosis in hepatocellular carcinoma. J. Cell. Biochem. 2018, 119, 4711–4716. [Google Scholar] [CrossRef] [PubMed]
  124. Pontisso, P.; Calabrese, F.; Benvegnù, L.; Lise, M.; Belluco, C.; Ruvoletto, M.G.; De Falco, S.; Marino, M.; Valente, M.; Nitti, D.; et al. Overexpression of squamous cell carcinoma antigen variants in hepatocellular carcinoma. Br. J. Cancer 2004, 90, 833–837. [Google Scholar] [CrossRef] [PubMed]
  125. Cataltepe, S.; Gornstein, E.R.; Schick, C.; Kamachi, Y.; Chatson, K.; Fries, J.; Silverman, G.A.; Upton, M.P. Co-expression of the Squamous Cell Carcinoma Antigens 1 and 2 in Normal Adult Human Tissues and Squamous Cell Carcinomas. J. Histochem. Cytochem. 2000, 48, 113–122. [Google Scholar] [CrossRef] [Green Version]
  126. Beneduce, L.; Castaldi, F.; Marino, M.; Quarta, S.; Ruvoletto, M.; Benvegnù, L.; Calabrese, F.; Gatta, A.; Pontisso, P.; Fassina, G. Squamous cell carcinoma antigen-immunoglobulin M complexes as novel biomarkers for hepatocellular carcinoma. Cancer 2005, 103, 2558–2565. [Google Scholar] [CrossRef] [PubMed]
  127. Yuen, M.-F.; Tanaka, Y.; Fong, D.; Fung, J.; Wong, D.K.-H.; Yuen, J.C.-H.; But, D.Y.-K.; Chan, A.O.-O.; Wong, B.C.-Y.; Mizokami, M.; et al. Independent risk factors and predictive score for the development of hepatocellular carcinoma in chronic hepatitis B. J. Hepatol. 2009, 50, 80–88. [Google Scholar] [CrossRef] [PubMed]
  128. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
  129. Silverman, G.J. Regulatory natural autoantibodies to apoptotic cells: Pallbearers and protectors. Arthritis Rheum. 2011, 63, 597–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Pozzan, R.; Cardin, M.; Piciocchi, N.; Cazzagon, G.; Maddalo, V.; Vanin, A.; Giacomin, P.; Pontisso, U.; Cillo, F.; Farinati, C. Di-agnostic and prognostic role of SCCA-IgM serum levels in hepatocellular carcinoma (HCC). J. Gastroenterol. Hepatol. 2014, 29, 1637–1644. [Google Scholar] [CrossRef] [PubMed]
  131. Biasiolo, L.; Chemello, S.; Quarta, L.; Cavalletto, F.; Bortolotti, C.; Caberlotto, L.; Beneduce, E.; Bernardinello, N.; Tono, G.; Fassina, A.; et al. Monitoring SCCA-IgM complexes in serum predicts liver disease progression in patients with chronic hepatitis. J. Viral Hepat. 2008, 15, 246–249. [Google Scholar] [CrossRef] [PubMed]
  132. Mossad, E.H.; Mahmoud, E.A.; Osman, S.H.; Mahmoud, H.I.; Shousha, N.A. Evaluation of squamous cell carcinoma anti-gen-immunoglobulin M complex (SCCA-IGM) and alpha-L-fucosidase (AFU) as novel diagnostic biomarkers for hepatocellular carcinoma. Tumour Biol. 2014, 35, 11559–11564. [Google Scholar] [CrossRef] [PubMed]
  133. Ali, A.M.; Higazi, H.M.; Moness, N.M.; Farag, Z.M.; Saad, H.A.; Moukareb, W.; Soliman, G.; El Sagheer, S.R.; Abd El Hamid, H.; Abdl Hamid, L.H. Clinical significances and diagnostic utilities of both miR-215 and squamous cell carcinoma antigen-IgM versus alpha-fetoprotein in Egyptian patients with hepatitis C virus-induced hepatocellular carcinoma. Clin. Exp. Gastroenterol. 2019, 12, 51–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Suminami, Y.; Nagashima, S.; Murakami, A.; Nawata, S.; Gondo, T.; Hirakawa, H.; Numa, F.; Silverman, G.A.; Kato, H. Suppression of a squamous cell carcinoma (SCC)-related serpin, SCC antigen, inhibits tumor growth with increased intratumor infiltration of natural killer cells. Cancer Res. 2001, 61, 1776–1780. [Google Scholar]
  135. Guarino, G.G.; Di Costanzo, A.; Gallotta, R.; Tortora, L.; Paneghetti, F.; Auriemma, C.; Tuccillo, G.; Fassina, N.; Caporaso, F.; Morisco, M. Circulating SCCA-IgM complex is a useful biomarker to predict the outcome of therapy in hepatocellular carcinoma patients. Scand. J. Clin. Lab. Investig. 2017, 77, 448–453. [Google Scholar] [CrossRef]
  136. Filmus, J. The contribution of in vivo manipulation of gene expression to the understanding of the function of glypicans. Glycoconj. J. 2002, 19, 319–323. [Google Scholar] [CrossRef] [PubMed]
  137. Iglesias, B.V.; Centeno, G.; Pascuccelli, H.; Ward, F.; Peters, M.G.; Filmus, J.; Puricelli, L.; Joffé, E.B.D.K. Expression pattern of glypican-3 (GPC3) during human embryonic and fetal development. Histol. Histopathol. 2008, 23, 1333–1340. [Google Scholar] [PubMed]
  138. Yao, M.; Yao, D.-F.; Bian, Y.-Z.; Zhang, C.-G.; Qiu, L.-W.; Wu, W.; Sai, W.-L.; Yang, J.-L.; Zhang, H.-J. Oncofetal antigen glypican-3 as a promising early diagnostic marker for hepatocellular carcinoma. Hepatobiliary Pancreat. Dis. Int. 2011, 10, 289–294. [Google Scholar] [CrossRef]
  139. Saber, M.A.; Khorshed, S.F.E.; Aboushousha, T.S.; Hamdy, H.E.; Seleem, M.I.; Soliman, A.H. Differential Expression of Glypican-3 and Insulin-Like Growth Factor-II mRNAs and Alpha-Fetoprotein and Ki-67 Markers in HCV Related Hepato-cellular Carcinomas in Egyptian Patients. Asian Pac. J. Cancer Prev. 2017, 18, 121–127. [Google Scholar] [PubMed]
  140. Capurro, M.; Wanless, I.R.; Sherman, M.; Deboer, G.; Shi, W.; Miyoshi, E.; Filmus, J. Glypican-3: A novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology 2003, 125, 89–97. [Google Scholar] [CrossRef]
  141. Hayashi, E.; Motomura, Y.; Shirakawa, H.; Yoshikawa, T.; Oba, N.; Nishinakagawa, S.; Mizuguchi, Y.; Kojima, T.; Nomura, K.; Nakatsura, T. Detection of glypican-3-specific CTLs in chronic hepatitis and liver cirrhosis. Oncol. Rep. 2009, 22, 149–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Nakatsura, T.; Shirakawa, H.; Kuronuma, T.; Nishimura, Y.; Hasebe, T.; Nakano, M.; Gotohda, N.; Takahashi, S.; Nakagohri, T.; Konishi, M.; et al. Glypican-3 is a useful diagnostic marker for a component of hepatocellular carcinoma in human liver cancer. Int. J. Oncol. 2009, 34, 649–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wang, X.Y.; Degos, F.; Dubois, S.; Tessiore, S.; Allegretta, M.; Guttmann, R.D.; Jothy, S.; Belghiti, J.; Bedossa, P.; Paradis, V. Glypican-3 expression in hepatocellular tumors: Diagnostic value for preneoplastic lesions and hepatocellular carcinomas. Hum. Pathol. 2006, 37, 1435–1441. [Google Scholar] [CrossRef] [PubMed]
  144. Qin, Z.; Wang, J.; Wang, Y.; Wang, G.; Wang, X.; Zhou, Z.; Liu, G.; Gao, S.; Zhu, L. Identification of a Glypican-3-Binding Peptide for In Vivo Non-Invasive Human Hepatocellular Carcinoma Detection. Macromol. Biosci. 2017, 17, 17. [Google Scholar] [CrossRef]
  145. Zhou, F.; Shang, W.; Yu, X.; Tian, J. Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med. Res. Rev. 2018, 38, 741–767. [Google Scholar] [CrossRef] [PubMed]
  146. Fischer, L.; Korfel, A.; Pfeiffer, S.; Kiewe, P.; Volk, H.-D.; Cakiroglu, H.; Widmann, T.; Thiel, E. CXCL13 and CXCL12 in Central Nervous System Lymphoma Patients. Clin. Cancer Res. 2009, 15, 5968–5973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Nhan-Chang, R.; Romero, J.P.; Kusanovic, F.; Gotsch, S.S.; Edwin, O.; Erez, P.; Mittal, C.J.; Kim, M.J.; Kim, J.; Espinoza, L.A.; et al. A role for CXCL13 (BCA-1) in pregnancy and intra-amniotic in-fection/inflammation. J. Matern. Fetal Neonatal. Med. 2008, 21, 763–775. [Google Scholar] [CrossRef]
  148. Widney, D.; Gui, L.M.; Popoviciu, J.W.; Said, E.C.; Breen, X.; Huang, C.M.; Kitchen, J.M.; Alcantar, J.B.; Smith, R.; Detels, O.; et al. Expression and Function of the Chemokine, CXCL13, and Its Receptor, CXCR5, in Aids-Associated Non-Hodgkin’s Lymphoma. AIDS Res. Treat. 2010, 2010, 164586. [Google Scholar] [CrossRef] [PubMed]
  149. Kim, S.J.; Ryu, K.J.; Hong, M.; Ko, Y.H.; Kim, W.S. The serum CXCL13 level is associated with the Glasgow Prognostic Score in extranodal NK/T-cell lymphoma patients. J. Hematol. Oncol. 2015, 8, 1–9. [Google Scholar] [CrossRef] [Green Version]
  150. Duan, Z.; Gao, J.; Zhang, L.; Liang, H.; Huang, X.; Xu, Q.; Zhang, Y.; Shen, T.; Lu, F. Phenotype and function of CXCR5+CD45RA−CD4+ T cells were altered in HBV-related hepatocellular carcinoma and elevated serum CXCL13 predicted better prognosis. Oncotarget 2015, 6, 44239–44253. [Google Scholar] [CrossRef] [Green Version]
  151. Li, C.; Kang, D.; Sun, X.; Liu, Y.; Wang, J.; Gao, P. The Effect of C-X-C Motif Chemokine 13 on Hepatocellular Carcinoma Associates with Wnt Signaling. BioMed Res. Int. 2015, 2015, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Li, B.; Su, H.; Cao, J.; Zhang, L. CXCL13 rather than IL-31 is a potential indicator in patients with hepatocellular carcinoma. Cytokine 2017, 89, 91–97. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Using immunoproteomic approach for identification and validation of TAAs. Firstly, the sera from HCC patients with high titer fluorescently stained were selected out with western blotting analysis and indirect immunofluorescence by using cultured tissue cells as antigen. Candidates of TAA were then separated using 2D-SDS-PAGE and analyzed by mass spectrometry. Finally, multiple methods, such as enzyme-linked immunosorbent assay (ELISA), Western blotting and immunohistochemistry (IHC), and tissue arrays, are used to validate identified potential tumor-associated antigen-antibody systems. These validated TAAs can be used to form TAA arrays for the immunodiagnosing early-stage cancers.
Figure 1. Using immunoproteomic approach for identification and validation of TAAs. Firstly, the sera from HCC patients with high titer fluorescently stained were selected out with western blotting analysis and indirect immunofluorescence by using cultured tissue cells as antigen. Candidates of TAA were then separated using 2D-SDS-PAGE and analyzed by mass spectrometry. Finally, multiple methods, such as enzyme-linked immunosorbent assay (ELISA), Western blotting and immunohistochemistry (IHC), and tissue arrays, are used to validate identified potential tumor-associated antigen-antibody systems. These validated TAAs can be used to form TAA arrays for the immunodiagnosing early-stage cancers.
Ijms 22 06139 g001
Table 1. TAA as biomarkers in HCC.
Table 1. TAA as biomarkers in HCC.
FunctionAutoantibodiesControlRefs.
Early diagnosis↑HCC1, P16, P53, P90, survivinHealthy individuals[20]
↑AFP, AFP-L3, DCP, CENPFLC, CH, NHS[35]
↑SPAG9LC, CH, NHS[36,37]
↑NPM1LC, CH, SLE, NHS[16]
↑14-3-3ζLC, CH, NHS[38]
↑MDM2LC, CH, NHS[39]
↑CENPF, DDX3, HSPA4, HSPA5, VIM, LMNB1, p53CH, NHS[40]
↑DDX3, eEF2, AIF, hnRNP A2, PBP, TIMCH, NHS, lung cancer, EC, BC, GC[41]
Diagnose (complementarydiagnosis)↑EIF3ANHS[42]
↑SF3B1NHS[43]
↑GAGE-1LC, HB, NHS[44]
↑CAPERαProstate cancer, breast cancer[45]
↑NY-ESO-1NHS[46]
↑IMP1, IMP2/p62, IMP3/Koc, CIP2A/p90, RalA, c-Myc, survivin, cyclin B1, 14-3-3 ζ, MDM2, p53, CAPERα/HCC1.4, p16, NPM1LC, CH, NHS[19]
↑AFP, Cyclin B1, Gankyrin, p53, NY-ESO-1, RalA, CK8, H-RAS-1, p16, WT1hepatitis C with either cirrhosis or chronic liver disease, NHS[47]
↑ Sui1LC, CH, NHS[15]
↑IMP1, p62, Koc, p53, c-myc, cyclin B1, survivin, p16, RalA, Sui1LC, CH, NHS[48]
↑FASNNHS[49]
↑RalALC, CH, NHS[50]
↑KRT23, AHSG, RPL17, FTL, DDX41CH, NHS[51]
↑ORP150, aconitate dehydratase, HSP70, protein disulfide-isomerase A3, NDRG1, GLUD1, PA2G4, fumarate hydratase, VDAC1, PEBP, peroxiredoxinLC, CH, NHS[52]
↑EIF3SI, LDHA, RFC2, MCART1LC, CH, NHS, GC, PC[53]
↑IMP1, p62, Koc, p53, c-myc, cyclin B1, survivin, p16LC, CH, NHS[54]
↑HSP70, GAPDH, PRX, Mn-SODNHS[55]
↑ IMP1, IMP3, p53LC, CH, NHS[56]
↑p16NHS[57]
↑c-myc, p53, cyclin B1, p62, Koc, IMP1, survivinNHS[58,59]
↑p90CH, AH, HBsAg carriers, NHS[60]
↑calreticulin, CK8, and NDPKA, and ATP5F1B,Other cancers, CH, active SLE, NHS[61]
↑p62, CENPFLC, CH, autoimmune hepatitis[62]
↑p62Asymptomatic HBsAg carrier, AH, CH, NHS[23]
↑p53, AFPchronic liver disease (non-viral/viral liver disease )[63,64]
↑cyclin B1LC, CH, NHS[65]
Diagnosis,
recurrence/metastasis prediction
↑GRP78LC, CH, NHS

SUN449, A549, T24, MOLT-4, KOPN63
[66,67]
Diagnosis/prognostic marker↑OPNLC, CH, NHS[68]
↑NX-PVKA, DCPCompare using Child-Pugh class and TNM classification[69]
↑p62EC, GC, large intestine cancer[70]
Prognostic marker↑anti-CD25 IgGNHS[71]
MVI prediction in HCC↑HSP 70 and Eno-1NHS[72]
Diagnosis of AFP-negative HCC↑SMP30LC, CH, NHS[73]
↑NPM1, 14-3-3 ζ, MDM2chronic liver disease, normal human control, AFP-positive HCC cases[74]
↑HCC-22-5LC, CH, NHS, Nasopharynx cancer, lung cancer, gastric-intestine disease[75]
↑IMPsNHS[76]
Diagnosis of HBV-HCC↑Ku86LC, NHS[77]
↑hnRNP LHBV carrier, HBV LC, NHS[78]
Diagnose of HCV-HCC↑HSP70, SOD2, PRDX6HCV-/HCC-, HCV+/HCC-, NHS[79]
↑Ku86LC, NHS[80]
↑DHCR24HBV+ including LC, CH[81]
AH, acute hepatitis; AIF, apoptosis-inducing factor; AFP, alpha-fetoprotein; AFP-L3, lens culinaris agglutinin-reactive AFP; anti-CENPF, centromere protein F autoantibody; BC, breast cancer; CK8, cytokeratin 8; CH, chronic hepatitis; DCP, des-gamma-carboxyprothrombin; DDX3, DEAD (Asp-Glu-Ala-Asp) box polypeptide 3; DHCR24, 3β-hydroxysterol Δ24-reductase; EC, esophageal cancer; eEf2, eukaryotic translation elongation factor 2; Eno-1, alpha-enolase; FASN, fatty acid synthase; F1-ATP synthase β-subunit; GAGE-1, cancer-testis antigen G antigen 1; GC, gastric carcinoma; GLUD1, glutamate dehydrogenase 1; GRP78, glucose-regulated protein 78; hnRNP A2, heterogeneous nuclear ribonucleoprotein A2; HBV-HCC, hepatitis B virus-related hepatocellular carcinoma; HCV-HCC, hepatitis C virus-related hepatocellular carcinoma; hnRNP L, hnRNP Lheterogeneous nuclear ribonucleoprotein L; LC, liver cirrhosis; MDM2, mouse double minute 2 homolog; MVI, microvascular invasion; NDPKA, nucleoside diphosphate kinase A; NDRG1, N-myc downstream-regulated gene 1; NHS, normal human serum; NPM1, nucleophosmin; OPN, osteopontin; PC, pancreatic carcinoma; PEBP, phosphatidylethanolamine-binding protein; PBP, prostatic binding protein; TIM, triosephosphate isomerase; SLE, systemic lupus erythematosus; SPAG9, sperm-associated antigen 9; VDAC1, voltage-dependent anion-selective channel protein 1. ↑: upregulated biomarkers.
Table 2. Exosomes are used as biomarkers of HCC.
Table 2. Exosomes are used as biomarkers of HCC.
FunctionSpeciesSourceExosome contentsControlRef.
Early predictionRatSerum↑miR-10b, ↑miR-21,
↓miR-122, ↓miR-200a
Normal, degeneration, fibrosis, cirrhosis[9]
HumanSerum↑miR-21Healthy individuals, CHB patients[87]
DiagnoseHumanSerum↑hnRNPH1Healthy individuals, CHB and LC patients[112]
HumanSerum↑LncRNA HEIH

↓LncRNA HEIH
HCV-induced cirrhosis
CHC patients
[113]
HumanSerum↓miR-9-3pHealthy individuals[114]
HumanSerum↑LncRNA-FAL1Healthy individuals[115]
HumanSerum↑LG3BP, ↑PIGRHealthy individuals[102]
HumanSerum↑miR-18a, ↑miR-221, ↑miR-222, ↑miR-224
↓miR-101, ↓miR-106b, ↓miR-122, ↓miR-195
CHB or LC[95]
HumanSerum↑miR-519d, ↑miR-494, ↑miR-21, ↑miR-22Cirrhotic patients without HCC[116]
Diagnosis and prognosis predictionHumanSerum↑miR-122, ↑miR-125b, ↑miR-145, ↑miR-192, ↑miR-194, ↑miR-29a, ↑miR-17-5p, ↑miR-106aHealthy individuals[117]
HepG2, SMMC7721, SKHEP1, Huh7 cells
Human
Cell culture media
Serum
↑miR-93WRL68 cell
Healthy individuals
[118]
MHCC-97H
Human
Cell culture
media
Serum
↑miR-665MHCC-97L and L02
Healthy individuals
[119]
Diagnosis, clinical staging and recurrence predictionHumanSerum↑ENSG00000258332.1, ↑LINC00635Healthy individuals, CHB and LC patients[120]
Recurrence/metastasis predictionHumanSerum↑miR-103Recurrence-free survival groups[121]
HumanSerum↑CASC9Low recurrence survival groups[122]
Prognosis
prediction
HumanSerum↑miRNA-21, ↑lncRNA-ATBTwo different non-human miRNAs[97]
HumanSerum↓miR-638Healthy individuals[123]
HumanSerum↓miR-125bCHB[96]
Prognosis prediction after LTHumanSerum↓miR-718HCC patients without recurrence[99]
Prognosis prediction of TACEHumanSerum↓miR-122LC and CH[94]
CHB, chronic hepatitis B; CHC, chronic hepatitis C; LC, liver cirrhosis; LG3BP, Galectin-3-binding protein; LT, liver transplantation; PIGR, polymeric immune receptor; TACE, transarterial chemoembolization. ↑: upregulated biomarkers ↓: downregulated biomarkers.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xing, M.; Wang, X.; Kirken, R.A.; He, L.; Zhang, J.-Y. Immunodiagnostic Biomarkers for Hepatocellular Carcinoma (HCC): The First Step in Detection and Treatment. Int. J. Mol. Sci. 2021, 22, 6139. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22116139

AMA Style

Xing M, Wang X, Kirken RA, He L, Zhang J-Y. Immunodiagnostic Biomarkers for Hepatocellular Carcinoma (HCC): The First Step in Detection and Treatment. International Journal of Molecular Sciences. 2021; 22(11):6139. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22116139

Chicago/Turabian Style

Xing, Mengtao, Xinzhi Wang, Robert A. Kirken, Ling He, and Jian-Ying Zhang. 2021. "Immunodiagnostic Biomarkers for Hepatocellular Carcinoma (HCC): The First Step in Detection and Treatment" International Journal of Molecular Sciences 22, no. 11: 6139. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22116139

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop