Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Review ArticleReviews

Alternative Biomarkers to Predict Tumor Biology in Hepatocellular Carcinoma

PHILLIPE ABREU, RAPHAELLA FERREIRA, VICTOR MINELI, MAURICIO ALVES RIBEIRO, FABIO GONÇALVES FERREIRA, RODRIGO MARTINEZ DE MELLO VIANNA, FLAVIO DANIEL SAAVEDRA TOMASICH and LUIZ ARNALDO SZUTAN
Anticancer Research December 2020, 40 (12) 6573-6784; DOI: https://doi.org/10.21873/anticanres.14682
PHILLIPE ABREU
1Department of Surgery, University of Miami, Jackson Memorial Hospital, Miami Transplant Institute, Miami, FL, U.S.A.
2Department of Surgery, Division of Liver Surgery, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: Dr.PhillipeAbreu@gmail.com
RAPHAELLA FERREIRA
2Department of Surgery, Division of Liver Surgery, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
VICTOR MINELI
3Faculty of Medicine, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MAURICIO ALVES RIBEIRO
2Department of Surgery, Division of Liver Surgery, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
FABIO GONÇALVES FERREIRA
2Department of Surgery, Division of Liver Surgery, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RODRIGO MARTINEZ DE MELLO VIANNA
1Department of Surgery, University of Miami, Jackson Memorial Hospital, Miami Transplant Institute, Miami, FL, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
FLAVIO DANIEL SAAVEDRA TOMASICH
4Department of Surgery, Federal University of Parana, Erasto Gaertner Cancer Center, Curitiba, PR, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
LUIZ ARNALDO SZUTAN
2Department of Surgery, Division of Liver Surgery, Santa Casa of Sao Paulo School of Medical Sciences, Sao Paulo, SP, Brazil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Hepatocellular carcinoma (HCC) is the most frequent primary malignant liver tumor, with more than 800,000 new cases diagnosed each year and with high mortality, ranking fourth in the world in cancer deaths. The worst prognosis is related to the late diagnosis, in which the tumor is at an advanced stage and curative treatments are not efficient in terms of increasing overall survival. Currently, screening and monitoring tests based on current guidelines have limited accuracy, which points to the need for the development of new biomarkers that improve HCC detection as well as its early diagnosis. This review will discuss the five phases of development of a biomarker, from its discovery to its application in clinical practice, and indicate the main biomarkers per development phase. Potential emerging technologies such as “Radiomics”, “Proteomics” and “Metabolomics” will also be discussed, which should serve as tools for the elucidation of tumor heterogeneity, as well as provide data for future studies on HCC biomarkers.

Key Words:
  • Hepatocellular carcinoma
  • surveillance
  • screening
  • diagnosis
  • biomarkers
  • tumor biology
  • liver tumors
  • review

Hepatocellular carcinoma (HCC) is the most frequent primary tumor of the liver, and its incidence is increasing, currently occupying the sixth position among the incidences of all cancers, and the fourth position among cancer-related deaths (1). The prevalence of HCC is higher in men than in women (1). As for the geographical distribution of the disease, HCC has the highest incidence in developing countries, with more than 80% of cases reported in sub-Saharan Africa and East Asia as a result of the hepatitis B endemic in these regions; while the incidence has been decreasing in some Eastern countries such as China and Japan due to vaccination programs and treatment of viral hepatitis. Cases of HCC have increased in Western countries, such as the United States, related to the aging population of patients with hepatitis C and the emergence of non-alcoholic steatohepatitis (NASH) as a chronic liver disease (2, 3). The pre-existence of cirrhosis is estimated in 80% of patients diagnosed with HCC (4), but currently any etiological agent leading to chronic liver lesions and cirrhosis is considered a risk factor associated with the development of HCC, the most common being hepatitis B, hepatitis C, high alcohol consumption and NASH (5).

Once diagnosed, the prognosis of the HCC patient depends, among other factors, on the intention of the treatment: curative or palliative. Among the curative treatments, surgical resection, local ablative therapies and liver transplantation may be indicated, the latter being the most indicated for early stage patients and the one offering the best overall survival (6, 7). Other factors that directly influence the patient’s prognosis, such as number of lymph nodes involved, presence of metastasis, tumor size and history of hepatitis, the analysis of these factors being important in the definition of survival rates and individualized therapeutic plans (8).

The development of biomarkers as complementary diagnostic methods and in the follow-up after treatment, has been the object of study in the last two decades, but before their widespread adoption, there are validation phases (9, 10). In this review, the development stages of the current biomarkers will be addressed and the most promising ones in the early detection of HCC will be highlighted. Emerging technologies that may assist in the diagnosis of HCC in the coming years, such as artificial intelligence, are also pointed out.

Current Recommendations

The diagnosis of HCC is made by imaging examinations or liver biopsy, but for early detection of HCC, screening of asymptomatic patients by monitoring tests should be performed. Therefore, the American Association for the Study of Liver Diseases (AASLD) recommends monitoring every 6 months by abdominal ultrasonography, with or without alpha-fetoprotein (AFP) dosage, in all adults with cirrhosis, except in cirrhotic patients with Child-Pugh C Score who are not on the transplant list (6). Active surveillance then becomes an early detection tool for HCC, which is directly associated with improved survival rates of patients, as demonstrated by the meta-analysis of Singal et al. (11).

Despite the surveillance recommendations, there are flaws in this process (12). In a meta-analysis, Singal tried to characterize the performance of abdominal ultrasound in the detection of HCC and obtained only a 63% sensitivity in the detection of early-stage tumors, without any statistically significant improvement in sensitivity when associated with the serum AFP examination (13). Lack of screening and follow-up contribute to the detection of HCC in more advanced tumor stages in one-third of the cases; however, the low sensitivity in detecting the tumor in currently available tests represents the most common cause of late diagnosis, suggesting the need for more effective investigation strategies (12).

In Brazil, the Diagnostic and Therapeutic Guidelines for hepatocellular carcinoma were published by the Ministry of Health in 2012, and recommend screening for HCC by abdominal ultrasonography every 6 months, associated or not with AFP dosage, among patients diagnosed with hepatic cirrhosis who may benefit from curative treatments (patients classified as Child-Pugh A-C without comorbidities) (14). The diagnosis can be preferably made by radiological methods such as computed tomography, magnetic resonance imaging or contrast ultrasonography, reserving the anatomopathological diagnosis to non-cirrhotic patients and to cases in which the radiological methods are inconclusive (14). Screening patients in risk groups is the best way to ensure an early diagnosis and the possibility of healing, and increase the overall survival of these patients (15).

Development of Biomarkers

A biomarker can be defined as an objective and quantifiable feature of a biological process (16, 17). In practice, we can define it as a clinical sign, a parameter observed by the physician, which can be precisely measured and which can be reproduced, and can be used in cancer screening, in measuring therapeutic response or in defining prognosis (17). In HCC, biomarkers are widely applied, whether in decision making for a therapeutic choice or in the selection of patients most likely to get the best results from different treatments (18). The development of potential biomarkers for HCC faces challenges, mainly due to the tumor heterogeneity in this cancer, which makes its clinical applicability difficult (16, 18). Recently, gene expression analysis of known potential biomarkers of HCC was used to correlate their expression in tumoral tissues with tumor prognosis (16).

The expansion of studies on biomarkers as monitoring tools led to the establishment of the Early Detection Research Network (EDRN) by the National Cancer Institute (NCI), with the objectives of coordinating research among the biomarker development and validation laboratories, in order to maintain rigor and promote collaboration among them, generating greater efficiency in the studies (10, 19). With the objectives of EDRN as a basis, Pepe et al. categorized the development of biomarkers into five phases, from their discovery to their application, as a monitoring tool in the early detection of cancer (10).

Phase 1. Phase 1 development usually begins with pre-clinical studies, comparing tumor tissue with non-tumoral tissue (10). The objective is to identify unique characteristics of tumor tissues that may lead to a candidate biomarker (10). Techniques such as immunohistochemistry, western blot and more recent technologies such as gene expression profiles based on mass spectrometry have been used to identify genes or proteins in tumor tissues when compared to control tissues (10). Thus, phase 1 aims to identify and prioritize potential biomarkers (establishing the best case selection and control for the development of studies), establish reliable and reproducible assays, determine how well the biomarker in question distinguishes case and control, analyze data obtained and conduct other confirmatory studies with new tissue samples (10).

Phase 2. Phase two consists of clinical trials executed with samples obtained in a non-invasive manner (10). The choice of samples, as well as their sizes, needs to be carefully considered, and even if blood bank samples are used, it is recommended that the final conclusions be obtained from studies with population samples (10). The clinical trials should be performed to show the ability of the biomarker to distinguish between the samples with and without cancer, thus allowing it to be considered promising for screening (10). An ideal biomarker for early detection of cancer should be highly sensitive, which indicates that the main objective of phase 2 is estimate the true-positive and false-positive rates in the trial and the ability of the biomarker to distinguish between samples with and without cancer, in addition to optimizing procedures to ensure the reproducibility of the trial in other laboratories (10). Phase 2 has other important objectives, such as evaluating the variations in the levels of the biomarker in question based on clinical-epidemiological factors of the individuals, such as age, gender and also based on tumor characteristics, such as histology, tumor staging and prognosis (10).

Phase 3. The main objective of phase 3 is to evaluate the ability of the biomarker to identify the disease before clinical diagnosis and thus, define a criterion for a positive test (10). Analyzing the group of cases, a biomarker whose levels change months before the diagnosis of the disease has more potential to become a useful tool in early detection when compared to a biomarker whose levels change days before the diagnosis (10). In this way, this capacity is evaluated and a criterion is defined to classify the test as positive (10). In addition, this phase aims to explore the impact of variables such as demographic factors, characteristics associated with the disease and clinical information of individuals on the discriminatory ability of the biomarker and, in case the biomarker performs better in certain subgroups, assist in the development of prospective studies of phase 4 (10).

Phase 4. Phase 4 consists of a prospective study, in which the objective is to describe the characteristics of the tumor detected by the screening test and these characteristics with possible benefits offered by early detection (10). The samples in this phase should be chosen based on a population that is possibly the target of the screening, that is, a group at risk for development of the disease, which will improve the performance of the biomarker when compared to phase 3 (10). Based on the results obtained, the receiver operating characteristics of the screening test are determined, such as the true-positive rate (proportion of those tested positive versus those that have the disease) and the false-positive rate (proportion of those who tested positive versus those that do not have the disease) (10).

Phase 5. Phase 5 assesses whether screening reduces mortality from the disease (10). Even when there is early detection of the disease, there may be no benefits for the specific population (10). The reasons for this may be related to ineffective treatment of the detected tumors, difficulties in implementing screening programs or overdiagnosis (10). This phase also considers cost-benefit information, assesses compliance with screening and compares the different approach protocols in the treatment of screened diseases in relation to their effects on mortality and cost (10).

Biomarkers for HCC per Development Phase

Table I presents the main biomarkers per study development phase for HCC as well as their applications and initial results.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Biomarkers available in the studies of HCC.

Phase 1. MicroRNAs. MicroRNAs (miRNAs) are posttranscription gene regulators, which silence messenger RNA and thus, play a homeostatic role by adjusting protein translation, while their deregulation is related to tumor progression (20). The potential of 19 circulating microRNAs as biomarkers for HCC has been studied by Huang et al. (21). In this meta-analysis, when the ability of the marker to differentiate between HCC patients and control group individuals was evaluated, the sensitivity and specificity of miRNA-21 was 86.6% and 79.5, respectively, while miRNA-122 obtained a sensitivity of 68% and specificity of 73.3% (21). Huang et al. have studied the ability of miRNA to differentiate between patients with HCC and hepatic cirrhosis (22). They obtained 16 miRNAs with a statistically significant difference between groups, namely miRNA-15a, miRNA-21, miRNA-29a, miRNA-30c, miRNA-486-3p, Let-7g, miRNA-122, miRNA18a, miRNA-338-3p, miRNA-126, miRNA-222, miRNA-223, miRNA-26a, miRNA192, miRNA-27a and miRNA-124 (22). In the recent study by Wu et al., serum levels of miRNA-199a in patients with HCC were significantly lower than those in patients with hepatic cirrhosis, pointing to its potential as a marker for the diagnosis of HCC (23). Thus, the study of microRNAs becomes a useful tool for the development of HCC markers (20).

Metabolomics. Metabolomics is a field of study that aims to provide information about endogenous metabolites in a biological sample, and with these data one can map biochemical changes in a disease and thus develop potential predictive biomarkers (24). Intracellular and extracellular metabolic changes, as well as metabolic pathways, are associated with tumor progression and reprogramming of the microenvironment caused by tumor growth (25).

Recent advances in the Metabolomics field can provide several new types of markers for HCC (26). Wang et al. have proposed an analysis of metabolic profiles based on high efficiency liquid chromatography (UPLC) and mass spectrometry (MS) of samples of HCC patients with cirrhosis of the liver and healthy individuals (27). The metabolic profile showed 100% sensitivity and specificity in differentiating HCC patients from those with hepatic cirrhosis, and 79.3% sensitivity and 100% specificity in the diagnosis of HCC. In addition, the analysis of the profiles indicated 13 potential biomarkers suggestive of metabolic disorders, such as phospholipid and organic acid alterations, in patients with HCC (27). Luo et al. have characterized the serum metabolic profiles of 1448 individuals using UPLC and MS-based methods and a panel was defined after selecting the appropriate biomarkers (28). The ability of the panel to discriminate between HCC and cirrhosis patients was compared with that of the AFP values, and was found to be more efficient (AUC 0.866 vs. 0.682), revealing its role in early stage HCC screening (28). The analysis of serum metabolites values is a promising tool for predicting metabolic changes at tissue levels due to the tumor installation (25).

Proteomics. Proteomics is an area that studies the complete set of proteins expressed in a cell, tissue or biological fluid (29). Proteomics, besides determining protein expression profiles, also identifies structure, location, activity, modifications and interactions of proteins in physiological or pathological conditions (29). In HCC, proteomics has the potential not only to elucidate the mechanisms of installation and progression of HCC, but also to provide a basis for the development of biomarkers for the disease (29). Wang et al. have proposed a useful algorithm to predict HCC that associates AFP with 4 other clinical variables, being age, sex, alkaline phosphatase and alanine aminotransferase (30). More recently, they added to the algorithm the biomarker fucosilated kininogen, to improve HCC detection in early stages and in patients with AFP<20 ng/ml (31). As a result, they obtained AUC greater than that of AFP alone (0.977 vs. 0.828), and 89% HCC detection rate in patients with AFP<20 ng/ml (31).

Gao et al. have conducted a proteogenomic HCC characterization in patients with hepatitis B (32). They found two enzymes, PYCR2 and ADG1A, which can be potential biomarkers for the prognosis of HCC, and observed lower expression of hepatitis B virus proteins and receptors in patients with HCC suggesting that metabolic changes are related to the advanced stage of the disease and worse prognosis, and may serve as a basis for future studies on HCC treatment (29). In general, proteomic data can provide information that helps in the clinical, biological and therapeutic understanding of HCC (29).

Radiomics. Radiomics is an emerging field that is based on the conversion of images from exams such as computed tomography and magnetic resonance imaging into high dimension data, which are analyzed by artificial intelligence to reveal hidden pathophysiological characteristics that, when added to the patient’s characteristics, function as a prognostic or diagnostic tool (33). Regarding HCC, “omics” although at embryonic stage, it has presented promising results (34). Zhout et al. published a study with 215 patients to predict early recurrence of HCC based on the radiomic characteristics of the patients and obtained a predictive AUC of 0.83 in a combined model, showing that the Radiomics signature may be promising in predicting early recurrence (35). Zheng et al. have reported that the combination of factors obtained from the Radiomics model with clinical variables of the patients has good performance in predicting microvascular invasion after HCC resection (36). Radiomics has shown promise in diagnosis, choice of therapeutic treatment, screening evaluation and prognosis of HCC (37).

Polo-like Kinase Proteins

Polo-like kinase proteins (PLK) are characterized by an N-terminal serine/threonine kinase domain, highly conserved at one or two polo boxes in the C-terminal region, which are crucial for subcellular location, specific phospho-peptide binding and centrioles duplication (38). PLK-4 shows cytoplasmic expression in most human tissues. The protein is located in centrioles and microtubules, and regulates the duplication of centrioles during the cell cycle. Also, tissue expression of PLK-4 is very variable among human tissues, and depends on the number of cells with a high concentration of centromeres, which is directly related to the cellular function of tissues during cell division (39). It is considered a prognostic marker in colorectal cancer (favorable), lung cancer (unfavorable) and pancreatic cancer (unfavorable) (39). In HCC, low expression of PLK-4 is possibly adversely associated with global and disease-free survival in preclinical models (38, 40-43).

Phase 2

AFP-L3. AFP-L3 is an AFP isoform and its percentage relative to the total AFP has been used as a marker for HCC detection, with a cut-off value of 10% (44). In the study by Toyoda et al., hs-AFP-L3 (highly sensitive AFP-L3) was tested in HCC patients under surveillance with AFP values below 20 ng/ml (45). As a result, for cut-off value of 5% of hs-AFP-L3 relative to total AFP, sensitivity 41.5% and specificity 85.1% in diagnosis of patients with AFP<20 ng/ml were obtained (45). In that study, the relationship between the percentage of hs-AFP-L3 and prognosis was also evaluated, showing that the measurement of hs-AFP-L3 before treatment can help predict prognosis and define therapies (45). Thus, it has been shown that hs-AFP-L3 may be useful in the early detection of HCC in patients with AFP<20 ng/ml, having improved sensitivity when combined with Des-carboxy-prothrombin, and may be part of pretherapy as a diagnostic predictor (45).

Dickkopf-1 (DKK-1). DKK-1 is a glycoprotein that acts as an inhibitor of the Wnt/beta-catenine signaling pathway (46). Shen et al. have shown significantly higher serum DKK-1 levels in HCC patients when compared to healthy individuals with chronic hepatitis B or liver cirrhosis (47). In the meta-analysis of Zhenije et al., the combination of DKK-1 and AFP showed better accuracy in the diagnosis of HCC when compared to the single use of these biomarkers (48).

Glipican-3 (GPC3). GPC3 is a member of the proteoglycan heparan sulfate family, comprised of 6 subtypes of glypicans (GPC1-6), which have several functions, the main one being to regulate Wnt, Hedgehog, bone morphogenic protein (BMP) and fibroblastic growth factor signaling (49). GPC3 is present in different stages and different tissues during embryonic development, which indicates its involvement in morphogenesis (50). In adult liver, no GPC3 expression is detected (51) while microarray analysis of tissue samples revealed increased GPC3 expression in 63.6% of HCC patients (52). The mechanisms involving GPC3 in the development of HCC have been described by Zhou et al., and include stimulation of Wnt signaling, interaction with growth factors, stimulation of macrophage recruitment and promotion of epithelium-mesenchymal transition (EMT) (53). The literature indicates GPC3 as a highly specific biomarker, and the combination of markers has shown improved sensitivity in the distinction between HCC and non-malignant hepatocellular lesions, as Enan et al. pointed out by combining GPC3 with CD34 and obtaining 82% sensitivity (54). Nault et al. have studied the serum levels of GPC3 in patients with hepatic cirrhosis, obtaining significantly higher values in patients with advanced HCC when compared to patients without HCC or in patients with early stage disease (55).

Alpha-1-fucosidase (AFU). AFU is a lysosomal enzyme that can be detected in the serum of healthy patients, but its activity is increased in patients with HCC, cirrhosis and chronic hepatitis (56). In the study of Fawzy et al., the AFU values of 80 patients (40 with HCC and 40 with chronic liver diseases) and 40 healthy individuals were measured and compared (57). The results showed significantly higher serum AFU values in the HCC group when compared to the other two groups (p<0.001), and for AFU cut-off value of 2.3005 micromol (l–1) (min–1) the sensitivity and specificity delivered were 90% and 97.5%, respectively (57). The study also found a decrease in AFU values in patients who underwent local therapies for HCC [radiofrequency ablation (RFA) or transarterial chemoembolization (TACE)] (57). Thus, AFU has shown promise as a biomarker for the diagnosis of HCC and for the follow-up of therapeutic results (57).

Squamous cell carcinoma antigen (SCCA). SCCA is a serine protease inhibitor, physiologically found in squamous epithelium, but also expressed in epithelial cell neoplasms (58). Caterina et al., have studied the serum levels of the immunocomplex SCCA with IgM (SCCA-IgM) in patients with HCC and found sensitivity and specificity values of 89% and 50%, respectively, regarding the diagnosis of HCC (59). In the meta-analysis of Liu et al., both SCCA and SCCA-IgM showed moderate diagnostic accuracy in HCC, but combining SCCA-IgM with AFP there was an increase in diagnostic accuracy and early detection of HCC (60).

Golgi protein-73 (GP73). The GP73, is a transmembrane protein of the Golgi apparatus expressed in the normal liver, is mostly secreted by cells of the biliary epithelium and has insignificant expression in the hepatocytes (61). It was isolated for the first time in 2000, when its expression was observed in liver tissue cells of patients with cirrhosis and chronic hepatitis but not in normal liver cells in in vitro experiments (61). Later, it was demonstrated that serum levels of GP73 are significantly higher in HCC patients compared to healthy individuals (62). In the study of Jiao et al., 180 patients with HCC, 61 with cirrhosis of the liver, 99 with chronic hepatitis, and 103 healthy individuals, were submitted to blood sampling (63). It was shown, that the serum levels of GP73 were significantly higher in patients with HCC when compared to those in the other three groups (63). From the analysis of clinical and pathological variables, the presence of lymphatic metastasis was associated with high GP73 levels in patients with lymph node metastasis (63). As for HCC diagnosis, GP73 showed, for the optimal cut-off value of 117.53 ng/ml, a sensitivity of 78.3% and a specificity of 85.4%. However, AFP showed sensitivity and specificity values of 51.1% and 99%, respectively, for the optimal cut-off value of 176.91ng/ml, which shows that GP73 is more efficient in the early detection of HCC compared to AFP (63). The combination of GP73 with AFP presented the largest AUC, and obtained better sensitivity and specificity values (63).

Phase 3

Osteopontine (OPN). OPN is a multifunctional protein that is involved in several pathological processes such as inflammation and carcinogenesis in several tissues (64). In HCC, the plasma levels of OPN have been considered as a potential biomarker (65). Zhu et al. have conducted a study with 322 patients (105 with chronic hepatitis, 116 with cirrhosis and 101 with hepatocellular carcinoma) (66). Serum samples from patients were obtained before any invasive procedure, such as surgery or biopsy, and before any nonsurgical oncologic treatment, such as chemotherapy or radiotherapy. The results showed significantly higher OPN values in patients with HCC (median 39.84 ng/ml, IQR=15.55-91.81) when compared with the values of patients with chronic hepatitis (median 10.18 ng/ml) and liver cirrhosis (10.93 ng/ml) (66). In the diagnosis of HCC in the risk group (patients with chronic hepatitis and liver cirrhosis) and AFP-negative individuals (with cut-off value of 20 ng/ml), OPN showed sensitivity of 78.26% and specificity of 80.45% (66). In the diagnosis of HCC, OPN reached higher values of AUC when compared to AFP [AUC: 0.851 (95%CI=0.807-0.888) vs. AUC: 0.68 (95%CI=0.629-0.734)], and in the optimal cut off value of OPN (14.64 ng/ml) the sensitivity was 79.21% and the specificity 79.24% (66). When AFP and OPN were combined, the value of AUC increased to 0.876 (p<0.0001), higher than AFP and OPN alone, and reached 88.12% and 74.21% of sensitivity and specificity, respectively (66). Regarding the early detection of HCC, patients with tumors smaller than 2 cm presented significantly higher OPN values (mean=29.67 ng/ml, IQR=14.69-47.97) when compared to those with chronic hepatitis (p<0.0001) and those with hepatic cirrhosis (p=0.0029), and the performance of OPN was higher than AFP in these patients (66). These results suggest that OPN is promising in the early detection of HCC, although the number of patients in this group was low and other studies need to confirm it (66).

Midkine (MDK). MDK is a growth factor that binds to heparin which is encoded by the MDK gene on chromosome 11. It plays a key role in carcinogenic activities such as proliferation, migration and angiogenesis, its serum values are increased in most cases of HCC, and can play a key role in AFP-negative patients and patients with tumors in the early stages (67, 68). In the most recent meta-analysis of Zhang et al., 10 studies were selected representing a total of 1730 individuals (753 with HCC and 977 without HCC); the sensitivity and specificity of MDK for the diagnosis of HCC was 86% (95%CI=83.3-88.4%) and 75.4% (95%CI=72.6-78.1%), respectively (69). This meta-analysis, however, presented significant heterogeneity as well as limitations that may have affected the results. In a study by Zekri et al., single or combined biomarkers were compared; for optimal MDK cut-off values, sensitivity was 88.5% and specificity 80.6% (p<0.001), and their serum levels were significantly higher in HCC patients (70). Vongsuvanh et al. investigated the role of MDK in AFP-negative patients with HCC (AFP<20 ng/ml), and found that in 59.18% of these patients the levels of MDK (MDK>0.44 ng/ml) were high, which indicated the possible importance of MDK complementary to AFP (68). It is, therefore, necessary to develop more rigorous studies with larger samples to obtain future evidence on the role of MDK in the diagnosis of HCC (70).

Des-carboxyprothrombin (DCP). DCP, also known as vitamin K-induced protein, is a protein produced by hepatocytes in situations of vitamin K deficiency (71). Multiple mechanisms raise the levels of CPD in patients with HCC, including genetic alterations in the hepatocytes, changes in their cytoskeleton or unregulated entry of vitamin K (71). In the systematic review of Ji De et al., the results showed 66% grouped sensitivity and 88% grouped specificity of serum DCP in the diagnosis of HCC (72). The study of Lok. et al. who compared the accuracy of AFP and DCP in the early diagnosis of HCC, showed that these biomarkers were complementary, achieving better sensitivity results when combined (73). However, further studies are needed to determine whether this combination of biomarkers improves early detection of HCC.

GALAD score (Gender, Age, AFP-L3, DCP). The GALAD Score is a tool based on parameters such as gender, age and the serological biomarkers AFP, DCP and AFP-L3 (74). Yang et al. have compared GALAD Score and liver ultrasonography (74). They obtained 91% sensitivity and 85% specificity in HCC detection (74). When the analysis was restricted to initial stage HCC patients, the AUC of the GALAD Score remained high and was higher than the ultrasound (0.92 vs. 0.82, p<0.001) (74). Thus, despite the limitations of the study, the GALAD score showed potential as a complementary parameter compared to ultrasound, being particularly important in the detection of HCC in patients with high liver dysfunction and obesity, conditions that increase the risk of obtaining false-negative abdominal ultrasound (74). Berhane et al. compared the GALAD Score with the AFP, AFP-L3 and DCP single biomarkers and showed 91.6% sensitivity and 89.7% specificity for the GALAD Score in HCC diagnosis, significantly higher than the single biomarkers, indicating the GALAD Score as a tool capable of detecting tumors in which curative treatments are more applicable (75).

Phase 4

To date, there are no phase 4 biomarkers that have completed the tests for early diagnosis of HCC. Progress is hampered by the lack of suitable phase 3 samples for validation (76).

Phase 5

Alpha-fetoprotein (AFP). Although not all HCCs secrete AFP and other liver diseases can show increased levels of this marker, AFP is the most commonly used biomarker in HCC surveillance (77). Tzartzeva et al. have analyzed studies on HCC surveillance performance in patients with hepatic cirrhosis and obtained results indicating best sensitivity in early stage HCC detection when AFP was associated with ultrasound (mean sensitivity of 97%) (78). AFP can correlate with alanine aminotransferase (ALT) and aspartate aminotransferase (AST), especially in patients with hepatitis C, which implies reduced specificity of this biomarker (79). However, Liu et al. have studied whether the relationship between AFP, AST and ALT could result in an index that would be useful in HCC and, despite the limitations of the research, this relationship was able to provide information regarding the diagnosis, therapeutic outcome and prognosis of HCC (80). AFP cut-off values vary according to guidelines around the world (81). The meta-analysis of Zhang et al. examined these values and concluded that the AFP cut off of 400 ng/ml shows the best accuracy in HCC diagnosis, while the cut off of 20 ng/ml can be used for follow-up after treatment, as it presents high sensitivity (81). However, future studies should focus on the dynamics of changing AFP values as HCC progresses. The potential of AFP values in predicting transplant outcomes has been studied by Toso et al. (82). They have shown that AFP values can be used as a predictor of post-transplant global survival and can be included in the selection criteria along with morphological variables (size and number) (82). Duvoux et al. have proposed an HCC recurrence prediction model that incorporates AFP values and compared its accuracy with the Milan criterion. Establishing the relationship between AFP levels and tumor behavior, such as presence of vascular invasion, and consequently with prognosis, they pointed out that the adoption of AFP, a simple and reliable tool, to the selection criteria of patients for transplantation can refine this choice (83). Metha et al. have demonstrated that the serum levels of AFP are an important biomarker in predicting post-transplant recurrence (84). In patients with preoperative serum AFP>1000 ng/ml, the disease-free survival was 53%, while in patients with AFP<1000 ng/ml it was 80% (84). Halazun et al. have published a model that incorporates the AFP response concept, which is the difference between the highest and final value of serum AFP before transplantation while in the waiting list, to the criteria for patient selection. AFP response to treatment is an important tool that can improve patient selection and outcomes after transplantation (85).

Future Directions

Artificial intelligence (AI) can be defined as the study of algorithms that give machines the ability to reason and perform cognitive functions such as problem solving, object and word recognition, and decision making (86). One of the subfields of AI is machine learning (ML). ML allows machines to learn from the data and make predictions through pattern recognition (87). In the case of supervised ML, data are analysed using an ML algorithm with reinforcement learning to potentially identify clinically applicable patterns, and from a set of predictors (such as the clinical characteristics of a patient) to be able to predict outcomes (such as HCC recurrence) (87).

ML algorithms can be static or incorporate new information that allows constant improvement of the forecasting tool, allowing the algorithm to perfect itself (87). Thus, ML can be used to build a forecasting tool for a given population of HCC patients to identify those most likely to benefit from the various treatments and determine the most important variables to distinguish these patients, as presented in Figure 1. ML considers all the variables available in a given database as “Big-data” and incorporates the interrelationships between them in the outcome prediction tool (87).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

“Machine learning” application algorithm.

Conclusion

New biomarkers predictors of tumor biology have been investigated to adequately select patients for each treatment offered. To date, the response to initial treatment and the biological behavior of the tumor are effectively used in clinical practice in HCC patients. Artificial intelligence technologies, capable of taking into account clinical-laboratory and biological tumor data, need to be implemented, so that information from the so-called Big-data, is correctly interpreted for anticipating the chances of tumor recurrence in HCC. Several molecules related to HCC can contribute to the complete understanding of the disease, and indirectly estimate the response to the indicated treatment.

Footnotes

  • Authors’ Contributions

    This manuscript was written under the role assignment specified below. PA as corresponding author, and RF, were primarily in charge of manuscript drafting and contributed equally to it. VM, MAR, FGF conducted literature review and participated in study design. RMMV, FDST, LAS supervised drafting of the manuscript and gave final approval of the version to be published. All Authors read and approved the final manuscript.

  • This article is freely accessible online.

  • Conflicts of Interest

    The Authors have no conflicts of interest to declare regarding this study.

  • Received October 12, 2020.
  • Revision received October 21, 2020.
  • Accepted October 22, 2020.
  • Copyright © 2020 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

References

  1. ↵
    1. Ferlay J,
    2. Soerjomataram I,
    3. Dikshit R,
    4. Eser S,
    5. Mathers C,
    6. Rebelo M,
    7. Parkin DM,
    8. Forman D and
    9. Bray F
    : Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5): E359-86, 2015. PMID: 25220842. DOI: 10.1002/ijc.29210
    OpenUrlCrossRefPubMed
  2. ↵
    1. Setshedi M,
    2. Andersson M,
    3. Kgatle MM and
    4. Roberts L
    : Molecular and cellular oncogenic mechanisms in hepatocellular carcinoma. S Afr Med J 108(8b): 41-46, 2018. PMID: 30182912. DOI: 10.7196/SAMJ.2018.v108i8b.13500
    OpenUrlCrossRef
  3. ↵
    1. White DL,
    2. Thrift AP,
    3. Kanwal F,
    4. Davila J and
    5. El-Serag HB
    : Incidence of hepatocellular carcinoma in all 50 United States, from 2000 through 2012. Gastroenterology 152(4): 812-820.e5, 2017. PMID: 27889576. DOI: 10.1053/j.gastro.2016.11.020
    OpenUrlCrossRefPubMed
  4. ↵
    1. Harris PS,
    2. Hansen RM,
    3. Gray ME,
    4. Massoud OI,
    5. McGuire BM and
    6. Shoreibah MG
    : Hepatocellular carcinoma surveillance: An evidence-based approach. World J Gastroenterol 25(13): 1550-1559, 2019. PMID: 30983815. DOI: 10.3748/wjg.v25.i13.1550
    OpenUrlCrossRef
  5. ↵
    1. Bruix J and
    2. Sherman M
    : Management of hepatocellular carcinoma: An update. Hepatology 53(3): 1020-1022, 2011. PMID: 21374666. DOI: 10.1002/hep.24199
    OpenUrlCrossRefPubMed
  6. ↵
    1. Marrero JA,
    2. Kulik LM,
    3. Sirlin CB,
    4. Zhu AX,
    5. Finn RS,
    6. Abecassis MM,
    7. Roberts LR and
    8. Heimbach JK
    : Diagnosis, staging, and management of hepatocellular carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 68(2): 723-750, 2018. PMID: 29624699. DOI: 10.1002/hep.29913
    OpenUrlCrossRefPubMed
  7. ↵
    1. Llovet JM,
    2. Fuster J and
    3. Bruix J
    : The Barcelona approach: Diagnosis, staging, and treatment of hepatocellular carcinoma. Liver Transplantation 10(S2): S115-20, 2004. PMID: 14762851. DOI: 10.1002/lt.20034
    OpenUrlCrossRef
  8. ↵
    1. Pinna AD,
    2. Yang T,
    3. Mazzaferro V,
    4. de Carlis L,
    5. Zhou J,
    6. Roayaie S,
    7. Shen F,
    8. Sposito C,
    9. Cescon M,
    10. Di-Sandro S,
    11. Yi-Feng H,
    12. Johnson P and
    13. Cucchetti A
    : Liver transplantation and hepatic resection can achieve cure for hepatocellular carcinoma. Ann Surg 268(5): 868-875, 2018. PMID: 30080736. DOI: 10.1097/SLA.0000000000002889
    OpenUrlCrossRef
  9. ↵
    1. Sarveazad A,
    2. Agah S,
    3. Babahajian A,
    4. Amini N and
    5. Bahardoust M
    : Predictors of 5 year survival rate in hepatocellular carcinoma patients. J Res Med Sci 24(1): 86, 2019. PMID: 31741658. DOI: 10.4103/jrms.JRMS_1017_18
    OpenUrlCrossRef
  10. ↵
    1. Pepe MS,
    2. Etzioni R,
    3. Feng Z,
    4. Potter JD,
    5. Thompson ML,
    6. Thornquist M,
    7. Winget M and
    8. Yasui Y
    : Phases of biomarker development for early detection of cancer. J Natl Cancer Inst 93(14): 1054-1061, 2001. PMID: 11459866. DOI: 10.1093/jnci/93.14.1054
    OpenUrlCrossRefPubMed
  11. ↵
    1. Singal AG,
    2. Pillai A and
    3. Tiro J
    : Early detection, curative treatment, and survival rates for hepatocellular carcinoma surveillance in patients with cirrhosis: A meta-analysis. PLoS Med 11(4): e1001624, 2014. PMID: 24691105. DOI: 10.1371/journal.pmed.1001624.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Singal AG,
    2. Nehra M,
    3. Adams-Huet B,
    4. Yopp AC,
    5. Tiro JA,
    6. Marrero JA,
    7. Lok AS and
    8. Lee WM
    : Detection of hepatocellular carcinoma at advanced stages among patients in the HALT-C trial: Where did surveillance fail? Am J Gastroenterol 108(3): 425-432, 2013. PMID: 23337478. DOI: 10.1038/ajg.2012.449
    OpenUrlCrossRefPubMed
  13. ↵
    1. Singal A,
    2. Volk ML,
    3. Waljee A,
    4. Salgia R,
    5. Higgins P,
    6. Rogers MAM and
    7. Marrero JA
    : Meta-analysis: Surveillance with ultrasound for early-stage hepatocellular carcinoma in patients with cirrhosis. Aliment Pharmacol Ther 30(1): 37-47, 2009. PMID: 19392863. DOI: 10.1111/j.1365-2036.2009.04014.x
    OpenUrlCrossRefPubMed
  14. ↵
    1. Chagas AL,
    2. Mattos AA,
    3. Carrilho FJ,
    4. Bittencourt PL; Members of the Panel of the 2nd Consensus of the Brazilian Society of Hepatology on the Diagnosis and Management of Hepatocellular Carcinoma,
    5. Vezozzo DCP,
    6. Horvat N,
    7. Rocha MS,
    8. Alves VAF,
    9. Coral GP,
    10. Alvares-DA-Silva MR,
    11. Barros FMDR,
    12. Menezes MR,
    13. Monsignore LM,
    14. Coelho FF,
    15. Silva RFD,
    16. Silva RCMA,
    17. Boin IFSF,
    18. D Albuquerque LAC,
    19. Garcia JHP,
    20. Felga GEG,
    21. Moreira AM,
    22. Braghiroli MIFM,
    23. Hoff PMG,
    24. Mello VB,
    25. Dottori MF,
    26. Branco TP,
    27. Schiavon LL and
    28. Costa TFA
    : Brazilian society of hepatology updated recommendations for diagnosis and treatment of hepatocellular carcinoma. Arq Gastroenterol 57(suppl 1): 1-20, 2020. PMID: 32294682. DOI: 10.1590/S0004-2803.202000000-20
    OpenUrlCrossRef
  15. ↵
    1. Paranaguá-Vezozzo DC,
    2. Ono SK,
    3. Alvarado-Mora MV,
    4. Farias AQ,
    5. Cunha-Silva M,
    6. França JID,
    7. Alves VAF,
    8. Sherman M and
    9. Carrilho FJ
    : Epidemiology of HCC in Brazil: Incidence and risk factors in a ten-year cohort. Ann Hepatol 13(4): 386-393, 2014. PMID: 24927609.
    OpenUrl
  16. ↵
    1. Hass HG,
    2. Jobst J,
    3. Scheurlen M,
    4. Vogel U and
    5. Nehls O
    : Gene expression analysis for evaluation of potential biomarkers in hepatocellular carcinoma. Anticancer Res 35(4): 2021-2028, 2015. PMID: 25862856.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Strimbu K and
    2. Tavel JA
    : What are biomarkers? Curr Opin HIV AIDS 5(6): 463-466, 2010. PMID: 20978388. DOI: 10.1097/COH.0b013e32833ed177
    OpenUrlCrossRefPubMed
  18. ↵
    1. Sia D,
    2. Villanueva A,
    3. Friedman SL and
    4. Llovet JM
    : Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology 152(4): 745-761, 2017. PMID: 28043904. DOI: 10.1053/j.gastro.2016.11.048
    OpenUrlCrossRefPubMed
  19. ↵
    1. Borges KA,
    2. Dai J,
    3. Parikh ND,
    4. Schwartz M,
    5. Nguyen MH,
    6. Roberts LR,
    7. Befeler AS,
    8. Srivastava S,
    9. Rinaudo JA,
    10. Feng Z,
    11. Marrero JA and
    12. Reddy KR
    : Rationale and design of the hepatocellular carcinoma early detection strategy study: A multicenter longitudinal initiative of the National Cancer Institute’s Early Detection Research Network. Contemp Clin Trials 76: 49-54, 2019. PMID: 30439517. DOI: 10.1016/j.cct.2018.11.008
    OpenUrlCrossRef
  20. ↵
    1. Sartorius K,
    2. Sartorius B,
    3. Winkler C,
    4. Chuturgoon A and
    5. Makarova J
    : The biological and diagnostic role of miRNA’s in hepatocellular carcinoma. Front Biosci (Landmark Ed) 23(9): 1701-1720, 2018. PMID: 29293458. DOI: 10.2741/4668
    OpenUrlCrossRef
  21. ↵
    1. Huang JT,
    2. Liu SM,
    3. Ma H,
    4. Yang Y,
    5. Zhang X,
    6. Sun H,
    7. Zhang X,
    8. Xu J and
    9. Wang J
    : Systematic review and meta-analysis: Circulating miRNAs for diagnosis of hepatocellular carcinoma. J Cell Physiol 231(2): 328-335, 2016. PMID: 26291451. DOI: 10.1002/jcp.25135
    OpenUrlCrossRef
  22. ↵
    1. Huang YH,
    2. Liang KH,
    3. Chien RN,
    4. Hu TH,
    5. Lin KH,
    6. Hsu CW,
    7. Lin CL,
    8. Pan TL,
    9. Ke PY and
    10. Yeh CT
    : A circulating MicroRNA signature capable of assessing the risk of hepatocellular carcinoma in cirrhotic patients. Sci Rep 7(1): 523, 2017. PMID: 28364124. DOI: 10.1038/s41598-017-00631-9
    OpenUrlCrossRef
  23. ↵
    1. Wu J,
    2. Wu Y,
    3. Luo Y,
    4. Li X,
    5. Lin N,
    6. Yang X,
    7. Lin Y and
    8. Chen M
    : Circulating miRNA-199a and miRNA-122 levels as potential diagnostic and prognostic biomarkers for hepatocellular carcinoma. Ann Clin Lab Sci 50(2): 219-227, 2020. PMID: 32366560.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Sugimoto M,
    2. Wong DT,
    3. Hirayama A,
    4. Soga T and
    5. Tomita M
    : Capillary electrophoresis mass spectrometry-based saliva metabolomics identified oral, breast and pancreatic cancer-specific profiles. Metabolomics 6(1): 78-95, 2010. PMID: 20300169. DOI: 10.1007/s11306-009-0178-y
    OpenUrlCrossRefPubMed
  25. ↵
    1. Matteis S de, Ragusa A,
    2. Marisi G,
    3. Domenico S de, Gardini AC,
    4. Bonafè M and
    5. Giudetti AM
    : Aberrant metabolism in hepatocellular carcinoma provides diagnostic and therapeutic opportunities. Oxid Med Cell Longev 2018: 7512159, 2018. PMID: 30524660. DOI: 10.1155/2018/7512159
    OpenUrlCrossRef
  26. ↵
    1. Li ZF,
    2. Wang J,
    3. Huang C,
    4. Zhang S,
    5. Yang J,
    6. Jiang A,
    7. Zhou R and
    8. Pan D
    : Gas chromatography/time-of-flight mass spectrometry-based metabonomics of hepatocarcinoma in rats with lung metastasis: elucidation of the metabolic characteristics of hepatocarcinoma at formation and metastasis. Rapid Commun Mass Spec 24(18): 2765-2775, 2010. PMID: 20814984. DOI: 10.1002/rcm.4703
    OpenUrlCrossRefPubMed
  27. ↵
    1. Wang B,
    2. Chen D,
    3. Chen Y,
    4. Hu Z,
    5. Cao M,
    6. Xie Q,
    7. Chen Y,
    8. Xu J,
    9. Zheng S and
    10. Li L
    : Metabonomic profiles discriminate hepatocellular carcinoma from liver cirrhosis by ultraperformance liquid chromatography-mass spectrometry. J Proteome Res 11(2): 1217-1227, 2012. PMID: 22200553. DOI: 10.1021/pr2009252
    OpenUrlCrossRefPubMed
  28. ↵
    1. Luo P,
    2. Yin P,
    3. Hua R,
    4. Tan Y,
    5. Li Z,
    6. Qiu G,
    7. Yin Z,
    8. Xie X,
    9. Wang X,
    10. Chen W,
    11. Zhou L,
    12. Wang X,
    13. Li Y,
    14. Chen H,
    15. Gao L,
    16. Lu X,
    17. Wu T,
    18. Wang H,
    19. Niu J and
    20. Xu G
    : A Large-scale, multicenter serum metabolite biomarker identification study for the early detection of hepatocellular carcinoma. Hepatology 67(2): 662-675, 2018. PMID: 28960374. DOI: 10.1002/hep.29561
    OpenUrlCrossRef
  29. ↵
    1. Feng JT,
    2. Shang S and
    3. Beretta L
    : Proteomics for the early detection and treatment of hepatocellular carcinoma. Oncogene 25(27): 3810-3817, 2006. PMID: 16799622. DOI: 10.1038/sj.onc.1209551
    OpenUrlCrossRefPubMed
  30. ↵
    1. Wang M,
    2. Devarajan K,
    3. Singal AG,
    4. Marrero JA,
    5. Dai J,
    6. Feng Z,
    7. Rinaudo JA,
    8. Srivastava S,
    9. Evans A,
    10. Hann HW,
    11. Lai Y,
    12. Yang H,
    13. Block TM and
    14. Mehta A
    : The Doylestown Algorithm: A test to improve the performance of AFP in the detection of hepatocellular carcinoma. Cancer Prevent Res 9(2): 172-179, 2016. PMID: 26712941. DOI: 10.1158/1940-6207.CAPR-15-0186
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Wang M,
    2. Sanda M,
    3. Comunale MA,
    4. Herrera H,
    5. Swindell C,
    6. Kono Y,
    7. Singal AG,
    8. Marrero J,
    9. Block T,
    10. Goldman R and
    11. Mehta A
    : Changes in the glycosylation of kininogen and the development of a kininogen-based algorithm for the early detection of HCC. Cancer Epidemiol Biomarkers Prevent 26(5): 795-803, 2017. PMID: 28223431. DOI: 10.1158/1055-9965.EPI-16-0974
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Gao Q,
    2. Zhu H,
    3. Dong L,
    4. Shi W,
    5. Chen R,
    6. Song Z,
    7. Huang C,
    8. Li J,
    9. Dong X,
    10. Zhou Y,
    11. Liu Q,
    12. Ma L,
    13. Wang X,
    14. Zhou J,
    15. Liu Y,
    16. Boja E,
    17. Robles AI,
    18. Ma W,
    19. Wang P,
    20. Li Y,
    21. Ding L,
    22. Wen B,
    23. Zhang B,
    24. Rodriguez H,
    25. Gao D,
    26. Zhou H and
    27. Fan J
    : Integrated proteogenomic characterization of HBV-related hepatocellular carcinoma. Cell 179(5): 1240, 2019. PMID: 31730861. DOI: 10.1016/j.cell.2019.10.038
    OpenUrlCrossRef
  33. ↵
    1. Gillies RJ,
    2. Kinahan PE and
    3. Hricak H
    : Radiomics: Images are more than pictures, they are data. Radiology 278(2): 563-577, 2016. PMID: 26579733. DOI: 10.1148/radiol.2015151169
    OpenUrlCrossRefPubMed
  34. ↵
    1. Hesketh RL,
    2. Zhu AX and
    3. Oklu R
    : Radiomics and circulating tumor cells: Personalized care in hepatocellular carcinoma? Diagn Intervent Radiol 21(1): 78-84, 2015. PMID: 25430530. DOI: 10.5152/dir.2014.14237
    OpenUrlCrossRef
  35. ↵
    1. Zhou Y,
    2. He L,
    3. Huang Y,
    4. Chen S,
    5. Wu P,
    6. Ye W,
    7. Liu Z and
    8. Liang C
    : CT-based radiomics signature: a potential biomarker for preoperative prediction of early recurrence in hepatocellular carcinoma. Abdom Radiol 42(6): 1695-1704, 2017. PMID: 28180924. DOI: 10.1007/s00261-017-1072-0
    OpenUrlCrossRef
  36. ↵
    1. Zheng J,
    2. Chakraborty J,
    3. Chapman WC,
    4. Gerst S,
    5. Gonen M,
    6. Pak LM,
    7. Jarnagin WR,
    8. DeMatteo RP,
    9. Do RKG and
    10. Simpson AL
    : Preoperative prediction of microvascular invasion in hepatocellular carcinoma using quantitative image analysis. J Am Coll Surg 225(6): 778-788.e1, 2017. PMID: 28941728. DOI: 10.1016/j.jamcollsurg.2017.09.003
    OpenUrlCrossRef
  37. ↵
    1. Ma M,
    2. Feng Z,
    3. Peng T,
    4. Yan H,
    5. Rong P and
    6. Jumbe MM
    : Radiomics and its advances in hepatocellular carcinoma. Zhong Nan Da Xue Xue Bao Yi Xue Ban 44(3): 225-232, 2019. PMID: 30971513. DOI: 10.11817/j.issn.1672-7347.2019.03.001
    OpenUrlCrossRef
  38. ↵
    1. Bao J,
    2. Yu Y,
    3. Chen J,
    4. He Y,
    5. Chen X,
    6. Ren Z,
    7. Xue C,
    8. Liu L,
    9. Hu Q,
    10. Li J,
    11. Cui G and
    12. Sun R
    : MiR-126 negatively regulates PLK-4 to impact the development of hepatocellular carcinoma via ATR/CHEK1 pathway. Cell Death Dis 9(10): 1045, 2018. PMID: 30315225. DOI: 10.1038/s41419-018-1020-0
    OpenUrlCrossRef
  39. ↵
    1. Colwill K,
    2. Gräslund S,
    3. Persson H,
    4. Jarvik NE,
    5. Wyrzucki A and
    6. Wojcik J
    : A roadmap to generate renewable protein binders to the human proteome. Nature 8(7): 551-561, 2011. PMID: 21572409. DOI: 10.1038/nmeth.1607
    OpenUrlCrossRefPubMed
  40. ↵
    1. Liao Z,
    2. Zhang H,
    3. Fan P,
    4. Huang Q,
    5. Dong K,
    6. Qi Y,
    7. Song J,
    8. Chen L,
    9. Liang H,
    10. Chen X,
    11. Zhang Z,
    12. Zhang B.
    High PLK4 expression promotes tumor progression and induces epithelial-mesenchymal transition by regulating the Wnt/β-catenin signaling pathway in colorectal cancer. Int J Oncol 54(2): 479-490, 2019. PMID: 30570110. DOI: 10.3892/ijo.2018.4659
    OpenUrlCrossRef
    1. Korzeniewski N,
    2. Hohenfellner M and
    3. Duensing S
    : CAND1 promotes PLK4-mediated centriole overduplication and is frequently disrupted in prostate cancer. Neoplasia 14(9): 799-806, 2012. PMID: 23019411. DOI: 10.1593/neo.12580
    OpenUrlCrossRefPubMed
    1. Coelho PA,
    2. Bury L,
    3. Shahbazi MN,
    4. Liakath-Ali K,
    5. Tate PH,
    6. Wormald S,
    7. Hindley CJ,
    8. Huch M,
    9. Archer J,
    10. Skarnes WC,
    11. Zernicka-Goetz M and
    12. Glover DM
    : Over-expression of Plk4 induces centrosome amplification, loss of primary cilia and associated tissue hyperplasia in the mouse. Open Biol 5(12): 150209, 2015. PMID: 26701933. DOI: 10.1098/rsob.150209
    OpenUrlCrossRefPubMed
  41. ↵
    1. Levine MS,
    2. Bakker B,
    3. Boeckx B,
    4. Moyett J,
    5. Lu J,
    6. Vitre B,
    7. Spierings DC,
    8. Lansdorp PM,
    9. Cleveland DW,
    10. Lambrechts D,
    11. Foijer F and
    12. Holland AJ
    : Centrosome amplification is sufficient to promote spontaneous tumorigenesis in mammals. Dev Cell 40(3): 313-322.e5, 2017. PMID: 28132847. DOI: 10.1016/j.devcel.2016.12.022
    OpenUrlCrossRefPubMed
  42. ↵
    1. Khien VV,
    2. Mao HV,
    3. Chinh TT,
    4. Ha PT,
    5. Bang MH,
    6. Lac BV,
    7. Hop TV,
    8. Tuan NA,
    9. Don LV,
    10. Taketa K and
    11. Satomura S
    : Clinical evaluation of lentil lectin-reactive alpha-fetoprotein-l3 in histology-proven hepatocellular carcinoma. Int J Biol Markers 16(2): 105-111, 2001. PMID: 11471892.
    OpenUrlPubMed
  43. ↵
    1. Toyoda H,
    2. Kumada T,
    3. Tada T,
    4. Kaneoka Y,
    5. Maeda A,
    6. Kanke F and
    7. Satomura S
    : Clinical utility of highly sensitive Lens culinaris agglutinin-reactive alpha-fetoprotein in hepatocellular carcinoma patients with alpha-fetoprotein <20ng/mL. Cancer Sci 102(5): 1025-1031, 2011. PMID: 21244578. DOI: 10.1111/j.1349-7006.2011.01875.x
    OpenUrlCrossRefPubMed
  44. ↵
    1. Lozada ME,
    2. Chaiteerakij R and
    3. Roberts LR
    : Screening for hepatocellular carcinoma and cholangiocarcinoma: Can biomarkers replace imaging? Curr Hep Rep 14(2): 128-138, 2015. PMID: 26328266. DOI: 10.1007/s11901-015-0261-y
    OpenUrlCrossRef
  45. ↵
    1. Shen Q,
    2. Fan J,
    3. Yang XR,
    4. Tan Y,
    5. Zhao W,
    6. Xu Y,
    7. Wang N,
    8. Niu Y,
    9. Wu Z,
    10. Zhou J,
    11. Qiu SJ,
    12. Shi YH,
    13. Yu B,
    14. Tang N,
    15. Chu W,
    16. Wang M,
    17. Wu J,
    18. Zhang Z,
    19. Yang S,
    20. Gu J,
    21. Wang H and
    22. Qin W
    : Serum DKK1 as a protein biomarker for the diagnosis of hepatocellular carcinoma: A large-scale, multicentre study. Lancet Oncol 13(8): 817-826, 2012. PMID: 22738799. DOI: 10.1016/S1470-2045(12)70233-4
    OpenUrlCrossRefPubMed
  46. ↵
    1. Li Z,
    2. Mou L,
    3. Gao H,
    4. Zeng Y,
    5. Tang X,
    6. Deng X,
    7. Pu Z,
    8. Ni Y and
    9. Zhan Y
    : Diagnostic accuracy of serum dickkopf-1 protein in diagnosis hepatocellular carcinoma: An updated meta-analysis. Medicine (Baltimore) 98(32): e16725, 2019. PMID: 31393380. DOI: 10.1097/MD.0000000000016725
    OpenUrlCrossRef
  47. ↵
    1. Filmus J,
    2. Capurro M and
    3. Rast J
    : Glypicans. Genome Biol 9(5): 224, 2008. PMID: 18505598. DOI: 10.1186/gb-2008-9-5-224
    OpenUrlCrossRefPubMed
  48. ↵
    1. Iglesias BV,
    2. Centeno G,
    3. Pascuccelli H,
    4. Ward F,
    5. Peters MG,
    6. Filmus J,
    7. Puricelli L and
    8. Joffé EBK
    : Expression pattern of glypican-3 (GPC3) during human embryonic and fetal development. Histol Histopathol 23(11): 1333-1340, 2008. PMID: 18785116. DOI: 10.14670/HH-23.1333
    OpenUrlCrossRefPubMed
  49. ↵
    1. Haruyama Y and
    2. Kataoka H
    : Glypican-3 is a prognostic factor and an immunotherapeutic target in hepatocellular carcinoma. World J Gastroenterol 22(11): 275-283, 2016. PMID: 26755876. DOI: 10.3748/wjg.v22.i1.275
    OpenUrlCrossRef
  50. ↵
    1. Baumhoer D,
    2. Tornillo L,
    3. Stadlmann S,
    4. Roncalli M,
    5. Diamantis EK and
    6. Terracciano LM
    : Glypican 3 expression in human nonneoplastic, preneoplastic, and neoplastic tissues: A tissue microarray analysis of 4,387 tissue samples. Am J Clin Pathol 129(6): 899-906, 2008. PMID: 18480006. DOI: 10.1309/HCQWPWD50XHD2DW6
    OpenUrlCrossRefPubMed
  51. ↵
    1. Zhou F,
    2. Shang W,
    3. Yu X and
    4. Tian J
    : Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev 38(2): 741-767, 2018. PMID: 28621802. DOI: 10.1002/med.21455
    OpenUrlCrossRef
  52. ↵
    1. Enan ET,
    2. El-Hawary AK,
    3. El-Tantawy DAEA,
    4. Abu-Hashim MM and
    5. Helal NM
    : Diagnostic role of glypican 3 and CD34 for differentiating hepatocellular carcinoma from nonmalignant hepatocellular lesions. Ann Diagn Pathol 17(6): 490-493, 2013. PMID: 24012547. DOI: 10.1016/j.anndiagpath.2013.08.001
    OpenUrlCrossRef
  53. ↵
    1. Nault JC,
    2. Guyot E,
    3. Laguillier C,
    4. Chevret S,
    5. Ganne-Carrie N,
    6. N’Kontchou G,
    7. Beaugrand M,
    8. Seror O,
    9. Trinchet JC,
    10. Coelho J,
    11. Lasalle P,
    12. Charnaux N,
    13. Delehedde M,
    14. Sutton A and
    15. Nahon P
    : Serum proteoglycans as prognostic biomarkers of hepatocellular carcinoma in patients with alcoholic cirrhosis. Cancer Epidemiol Biomarkers Prevent 22(8): 1343-1352, 2013. PMID: 23780836. DOI: 10.1158/1055-9965.EPI-13-0179
    OpenUrlAbstract/FREE Full Text
  54. ↵
    1. Giardina MG,
    2. Matarazzo M,
    3. Varriale A,
    4. Morante R,
    5. Napoli A and
    6. Martino R
    : Serum alpha-L-fucosidase. A useful marker in the diagnosis of hepatocellular carcinoma. Cancer 70(5): 1044-1048, 1992. PMID: 1381268. DOI: 10.1002/1097-0142(19920901)70:5<1044::aid-cncr2820700506>3.0.co;2-u
    OpenUrlCrossRefPubMed
  55. ↵
    1. Fawzy Montaser M,
    2. Amin Sakr M and
    3. Omar Khalifa M
    : Alpha-l-fucosidase as a tumour marker of hepatocellular carcinoma. Arab J Gastroenterol 13(1): 9-13, 2012. PMID: 22560818. DOI: 10.1016/j.ajg.2012.03.006
    OpenUrlCrossRefPubMed
  56. ↵
    1. Kato H
    : Expression and function of squamous cell carcinoma antigen. Anticancer 16(4B): 2149-2153, 1996. PMID: 8694535.
    OpenUrl
  57. ↵
    1. Pozzan C,
    2. Cardin R,
    3. Piciocchi M,
    4. Cazzagon N,
    5. Maddalo G,
    6. Vanin V,
    7. Giacomin A,
    8. Pontisso P,
    9. Cillo U and
    10. Farinati F
    : Diagnostic and prognostic role of SCCA-IgM serum levels in hepatocellular carcinoma (HCC). J Gastroenterol Hepatol 29(8): 1637-1644, 2014. PMID: 24635038. DOI: 10.1111/jgh.12576
    OpenUrlCrossRef
  58. ↵
    1. Liu CH,
    2. Gil-Gómez A,
    3. Ampuero J and
    4. Romero-Gómez M
    : Diagnostic accuracy of SCCA and SCCA-IgM for hepatocellular carcinoma: A meta-analysis. Liver Int 38(10): 1820-1831, 2018. PMID: 29704434. DOI: 10.1111/liv.13867
    OpenUrlCrossRef
  59. ↵
    1. Kladney RD,
    2. Bulla GA,
    3. Guo L,
    4. Mason AL,
    5. Tollefson AE,
    6. Simon DJ,
    7. Koutoubi Z and
    8. Fimmel CJ
    : GP73, a novel Golgilocalized protein upregulated by viral infection. Gene 249(1-2): 53-65, 2000. PMID: 10831838. DOI: 10.1016/s0378-1119(00)00136-0
    OpenUrlCrossRefPubMed
  60. ↵
    1. Tian L,
    2. Wang Y,
    3. Xu D,
    4. Gui J,
    5. Jia X,
    6. Tong H,
    7. Wen X,
    8. Dong Z and
    9. Tian Y
    : Serological AFP/Golgi protein 73 could be a new diagnostic parameter of hepatic diseases. International J Cancer 129(8): 1923-1931, 2011. PMID: 21140449. DOI: 10.1002/ijc.25838
    OpenUrlCrossRefPubMed
  61. ↵
    1. Jiao C,
    2. Cui L,
    3. Piao J,
    4. Qi Y and
    5. Yu Z
    : Clinical significance and expression of serum Golgi protein 73 in primary hepatocellular carcinoma. J Cancer Res Ther 14(6): 1239-1244, 2018. PMID: 30488837. DOI: 10.4103/0973-1482.199784
    OpenUrlCrossRef
  62. ↵
    1. Wen Y,
    2. Jeong S,
    3. Xia Q and
    4. Kong X
    : Role of osteopontin in liver diseases. Int J Biol Sci 12(9): 1121-1128, 2016. PMID: 27570486. DOI: 10.7150/ijbs.16445
    OpenUrlCrossRef
  63. ↵
    1. Shang S,
    2. Plymoth A,
    3. Ge S,
    4. Feng Z,
    5. Rosen HR,
    6. Sangrajrang S,
    7. Hainaut P,
    8. Marrero JA and
    9. Beretta L
    : Identification of osteopontin as a novel marker for early hepatocellular carcinoma. Hepatology 55(2): 483-490, 2012. PMID: 21953299. DOI: 10.1002/hep.24703
    OpenUrlCrossRefPubMed
  64. ↵
    1. Zhu M,
    2. Zheng J,
    3. Wu F,
    4. Kang B,
    5. Liang J,
    6. Heskia F,
    7. Zhang X and
    8. Shan Y
    : OPN is a promising serological biomarker for hepatocellular carcinoma diagnosis. J Med Virol, 2020. PMID: 32043608. DOI: 10.1002/jmv.25704
    OpenUrlCrossRef
  65. ↵
    1. Yan Q,
    2. Huang HL,
    3. Yao X,
    4. Li J,
    5. Li LQ,
    6. Zhong J,
    7. Min LS,
    8. Dai LC and
    9. Zheng SS
    : Novel functional proteins interact with midkine in hepatic cancer cells. Hepatobiliary Pancreat Dis Int 11(3): 272-277, 2012. PMID: 22672821. DOI: 10.1016/s1499-3872(12)60160-x
    OpenUrlCrossRefPubMed
  66. ↵
    1. Vongsuvanh R,
    2. van Poorten D der,
    3. Iseli T,
    4. Strasser SI,
    5. McCaughan GW and
    6. George J
    : Midkine increases diagnostic yield in AFP negative and NASH-related hepatocellular carcinoma. PLoS One 11(5): e0155800, 2016. PMID: 27219517. DOI: 10.1371/journal.pone.0155800
    OpenUrlCrossRef
  67. ↵
    1. Zhang Y,
    2. Tang J,
    3. Zhou X,
    4. Zhu SL and
    5. Li LQ
    : Diagnostic accuracy of midkine for hepatocellular carcinoma: A meta-analysis. Mol Genet Genom Med 8(2): e1071, 2020. PMID: 31777190. DOI: 10.1002/mgg3.1071
    OpenUrlCrossRef
  68. ↵
    1. N Zekri AR,
    2. el Kassas M,
    3. Salam ESTA el, Hassan RM,
    4. Mohanad M,
    5. Gabr RM,
    6. Lotfy MM,
    7. Abdel-Zaher RAT,
    8. Bahnassy AA and
    9. Ahmed OS
    : The possible role of Dickkopf-1, Golgi protein- 73 and Midkine as predictors of hepatocarcinogenesis: a review and an Egyptian study. Sci Rep 10(1): 5156, 2020. PMID: 32198440. DOI: 10.1038/s41598-020-62051-6
    OpenUrlCrossRef
  69. ↵
    1. Bertino G,
    2. Ardiri AM,
    3. Calvagno GS,
    4. Bertino N and
    5. Boemi PM
    : Prognostic and diagnostic value of des-γ-carboxy prothrombin in liver cancer. Drug News Perspect 23(8): 498-508, 2010. PMID: 21031166. DOI: 10.1358/dnp.2010.23.8.1444236
    OpenUrlCrossRefPubMed
  70. ↵
    1. De J,
    2. Shen Y,
    3. Qin J,
    4. Feng L,
    5. Wang Y and
    6. Yang L
    : A systematic review of des-γ-carboxy prothrombin for the diagnosis of primary hepatocellular carcinoma. Medicine (Baltimore) 95(17): e3448, 2016. PMID: 27124038. DOI: 10.1097/MD.0000000000003448
    OpenUrlCrossRef
  71. ↵
    1. Lok AS,
    2. Sterling RK,
    3. Everhart JE,
    4. Wright EC,
    5. Hoefs JC,
    6. di Bisceglie AM,
    7. Morgan TR,
    8. Kim HY,
    9. Lee WM,
    10. Bonkovsky HL and
    11. Dienstag JL
    : Des-γ-carboxy prothrombin and α-fetoprotein as biomarkers for the early detection of hepatocellular carcinoma. Gastroenterology 138(2): 493-502, 2010. PMID: 19852963. DOI: 10.1053/j.gastro.2009.10.031
    OpenUrlCrossRefPubMed
  72. ↵
    1. Yang JD,
    2. Addissie BD,
    3. Mara KC,
    4. Harmsen WS,
    5. Dai J,
    6. Zhang N,
    7. Wongjarupong N,
    8. Ali HM,
    9. Ali HA,
    10. Hassan FA,
    11. Lavu S,
    12. Cvinar JL,
    13. Giama NH,
    14. Moser CD,
    15. Miyabe K,
    16. Allotey LK,
    17. Algeciras-Schimnich A,
    18. Theobald JP,
    19. Ward MM,
    20. Nguyen MH,
    21. Befeler AS,
    22. Reddy KR,
    23. Schwartz M,
    24. Harnois DM,
    25. Yamada H,
    26. Srivastava S,
    27. Rinaudo JA,
    28. Gores GJ,
    29. Feng Z,
    30. Marrero JA and
    31. Roberts LR
    : GALAD score for hepatocellular carcinoma detection in comparison with liver ultrasound and proposal of Galadus score. Cancer Epidemiol Biomarkers Prev 28(3): 531-538, 2019. PMID: 30464023. DOI: 10.1158/1055-9965.EPI-18-0281
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Berhane S,
    2. Toyoda H,
    3. Tada T,
    4. Kumada T,
    5. Kagebayashi C,
    6. Satomura S,
    7. Schweitzer N,
    8. Vogel A,
    9. Manns MP,
    10. Benckert J,
    11. Berg T,
    12. Ebker M,
    13. Best J,
    14. Dechêne A,
    15. Gerken G,
    16. Schlaak JF,
    17. Weinmann A,
    18. Wörns MA,
    19. Galle P,
    20. Yeo W,
    21. Mo F,
    22. Chan SL,
    23. Reeves H,
    24. Cox T and
    25. Johnson P
    : Role of the GALAD and BALAD-2 Serologic models in diagnosis of hepatocellular carcinoma and prediction of survival in patients. Clin Gastroenterol Hepatol 14(6): 875-886.e6, 2016. PMID: 26775025. DOI: 10.1016/j.cgh.2015.12.042
    OpenUrlCrossRefPubMed
  74. ↵
    1. Sengupta S and
    2. Parikh ND
    : Biomarker development for hepatocellular carcinoma early detection: current and future perspectives. Hepat Oncol 4(4): 111-122, 2017. PMID: 30191058. DOI: 10.2217/hep-2017-0019
    OpenUrlCrossRef
  75. ↵
    1. Marrero JA,
    2. Feng Z,
    3. Wang Y,
    4. Nguyen MH,
    5. Befeler AS,
    6. Roberts LR,
    7. Reddy KR,
    8. Harnois D,
    9. Llovet JM,
    10. Normolle D,
    11. Dalhgren J,
    12. Chia D,
    13. Lok AS,
    14. Wagner PD,
    15. Srivastava S and
    16. Schwartz M
    : Alpha-fetoprotein, des-gamma carboxyprothrombin, and lectin-bound alpha-fetoprotein in early hepatocellular carcinoma. Gastroenterology 137(1): 110-118, 2009. PMID: 19362088. DOI: 10.1053/j.gastro.2009.04.005
    OpenUrlCrossRefPubMed
  76. ↵
    1. Tzartzeva K,
    2. Obi J,
    3. Rich NE,
    4. Parikh ND,
    5. Marrero JA,
    6. Yopp A,
    7. Waljee AK and
    8. Singal AG
    : Surveillance imaging and alpha fetoprotein for early detection of hepatocellular carcinoma in patients with cirrhosis: A meta-analysis. Gastroenterology 154(6): 1706-1718.e1, 2018. PMID: 29425931. DOI: 10.1053/j.gastro.2018.01.064
    OpenUrlCrossRefPubMed
  77. ↵
    1. Yang JD,
    2. Dai J,
    3. Singal AG,
    4. Gopal P,
    5. Addissie BD,
    6. Nguyen MH,
    7. Befeler AS,
    8. Reddy KR,
    9. Schwartz M,
    10. Harnois DM,
    11. Yamada H,
    12. Gores GJ,
    13. Feng Z,
    14. Marrero JA and
    15. Roberts LR
    : Improved performance of serum alpha-fetoprotein for hepatocellular carcinoma diagnosis in hcv cirrhosis with normal alanine transaminase. Cancer Epidemiol Biomarkers Prev 26(7): 1085-1092, 2017. PMID: 28258053. DOI: 10.1158/1055-9965.EPI-16-0747
    OpenUrlAbstract/FREE Full Text
  78. ↵
    1. Liu X,
    2. Meng J,
    3. Xu H and
    4. Niu J
    : Alpha-fetoprotein to transaminase ratio is related to higher diagnostic efficacy for hepatocellular carcinoma. Medicine (Baltimore) 98(17): e15414, 2019. PMID: 31027143. DOI: 10.1097/MD.0000000000015414
    OpenUrlCrossRef
  79. ↵
    1. Zhang J,
    2. Chen G,
    3. Zhang P,
    4. Zhang J,
    5. Li X,
    6. Gan D,
    7. Cao X,
    8. Han M,
    9. Du H and
    10. Ye Y
    : The threshold of alpha-fetoprotein (AFP) for the diagnosis of hepatocellular carcinoma: A systematic review and meta-analysis. PLoS One 15(2): e0228857, 2020. PMID: 32053643. DOI: 10.1371/journal.pone.0228857
    OpenUrlCrossRef
  80. ↵
    1. Merani S,
    2. Majno P,
    3. Kneteman NM,
    4. Berney T,
    5. Morel P,
    6. Mentha G and
    7. Toso M
    : The impact of waiting list alpha-fetoprotein changes on the outcome of liver transplant for hepatocellular carcinoma. J Hepatol 55(4): 814-819, 2011. PMID: 21334400. DOI: 10.1016/j.jhep.2010.12.040
    OpenUrlCrossRefPubMed
  81. ↵
    1. Duvoux C,
    2. Roudot-Thoraval F,
    3. Decaens T,
    4. Pessione F,
    5. Badran H,
    6. Piardi T,
    7. Francoz C,
    8. Compagnon P,
    9. Vanlemmens C,
    10. Dumortier J,
    11. Dharancy S,
    12. Gugenheim J,
    13. Bernard PH,
    14. Adam R,
    15. Radenne S,
    16. Muscari F,
    17. Conti F,
    18. Hardwigsen J,
    19. Pageaux GP,
    20. Chazouillères O,
    21. Salame E,
    22. Hilleret MN,
    23. Lebray P,
    24. Abergel A,
    25. Debette-Gratien M,
    26. Kluger MD,
    27. Mallat A,
    28. Azoulay D,
    29. Cherqui D and Liver Transplantation French Study Group
    : Liver transplantation for hepatocellular carcinoma: A model including α-fetoprotein improves the performance of milan criteria. Gastroenterology 143(4): 986-94.e3, 2012. PMID: 22750200. DOI: 10.1053/j.gastro.2012.05.052
    OpenUrlCrossRefPubMed
  82. ↵
    1. Hameed B,
    2. Mehta N,
    3. Sapisochin G,
    4. Roberts JP and
    5. Yao FY
    : Alpha-fetoprotein level > 1000 ng/mL as an exclusion criterion for liver transplantation in patients with hepatocellular carcinoma meeting the Milan criteria. Liver Transpl 20(8): 945-951, 2014. PMID: 24797281. DOI: 10.1002/lt.23904
    OpenUrlCrossRefPubMed
  83. ↵
    1. Halazun KJ,
    2. Tabrizian P,
    3. Najjar M,
    4. Florman S,
    5. Schwartz M,
    6. Michelassi F,
    7. Benjamin Samstein B,
    8. Brown-Jr RS,
    9. Emond JC,
    10. Busuttil RW and
    11. Agopian VA
    : Is it time to abandon the Milan criteria? Results of a bicoastal US collaboration to redefine hepatocellular carcinoma liver transplantation selection policies. Ann Surg 268(4): 690-699, 2018. PMID: 30048307. DOI: 10.1097/SLA.0000000000002964
    OpenUrlCrossRef
  84. ↵
    1. Mintz Y and
    2. Brodie R
    : Introduction to artificial intelligence in medicine. Minim Invasive Ther Allied Technol 28(2): 73-81, 2019. PMID: 30810430. DOI: 10.1080/13645706.2019.1575882
    OpenUrlCrossRef
  85. ↵
    1. Hashimoto DA,
    2. Rosman G,
    3. Rus D and
    4. Meireles OR
    : Artificial intelligence in surgery: Promises and perils. Ann Surg 268(1): 70-76, 2018. PMID: 29389679. DOI: 10.1097/SLA.0000000000002693
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 40 (12)
Anticancer Research
Vol. 40, Issue 12
December 2020
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Alternative Biomarkers to Predict Tumor Biology in Hepatocellular Carcinoma
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
4 + 8 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Alternative Biomarkers to Predict Tumor Biology in Hepatocellular Carcinoma
PHILLIPE ABREU, RAPHAELLA FERREIRA, VICTOR MINELI, MAURICIO ALVES RIBEIRO, FABIO GONÇALVES FERREIRA, RODRIGO MARTINEZ DE MELLO VIANNA, FLAVIO DANIEL SAAVEDRA TOMASICH, LUIZ ARNALDO SZUTAN
Anticancer Research Dec 2020, 40 (12) 6573-6784; DOI: 10.21873/anticanres.14682

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Alternative Biomarkers to Predict Tumor Biology in Hepatocellular Carcinoma
PHILLIPE ABREU, RAPHAELLA FERREIRA, VICTOR MINELI, MAURICIO ALVES RIBEIRO, FABIO GONÇALVES FERREIRA, RODRIGO MARTINEZ DE MELLO VIANNA, FLAVIO DANIEL SAAVEDRA TOMASICH, LUIZ ARNALDO SZUTAN
Anticancer Research Dec 2020, 40 (12) 6573-6784; DOI: 10.21873/anticanres.14682
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Current Recommendations
    • Development of Biomarkers
    • Biomarkers for HCC per Development Phase
    • Polo-like Kinase Proteins
    • Future Directions
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Artificial Intelligence Models Could Enhance the Diagnostic Accuracy (DA) of Fecal Immunochemical Test (FIT) in the Detection of Colorectal Adenoma in a Screening Setting
  • Google Scholar

More in this TOC Section

  • Cytokine-based Cancer Immunotherapy: Challenges and Opportunities for IL-10
  • Proteolytic Enzyme Therapy in Complementary Oncology: A Systematic Review
  • Multimodal Treatment of Primary Advanced Ovarian Cancer
Show more Reviews

Similar Articles

Keywords

  • hepatocellular carcinoma
  • surveillance
  • screening
  • diagnosis
  • Biomarkers
  • tumor biology
  • liver tumors
  • review
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire