Clin Mol Hepatol > Volume 31(Suppl); 2025 > Article
Park, Lee, Agopian, Liu, Koltsova, You, Zhu, Tseng, and Yang: Liquid biopsy in hepatocellular carcinoma: Challenges, advances, and clinical implications

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive primary liver malignancy often diagnosed at an advanced stage, resulting in a poor prognosis. Accurate risk stratification and early detection of HCC are critical unmet needs for improving outcomes. Several blood-based biomarkers and imaging tests are available for early detection, prediction, and monitoring of HCC. However, serum protein biomarkers such as alpha-fetoprotein have shown relatively low sensitivity, leading to inaccurate performance. Imaging studies also face limitations related to suboptimal accuracy, high cost, and limited implementation. Recently, liquid biopsy techniques have gained attention for addressing these unmet needs. Liquid biopsy is non-invasive and provides more objective readouts, requiring less reliance on healthcare professional’s skills compared to imaging. Circulating tumor cells, cell-free DNA, and extracellular vesicles are targeted in liquid biopsies as novel biomarkers for HCC. Despite their potential, there are debates regarding the role of these novel biomarkers in the HCC care continuum. This review article aims to discuss the technical challenges, recent technical advancements, advantages and disadvantages of these liquid biopsies, as well as their current clinical application and future directions of liquid biopsy in HCC.

INTRODUCTION

Hepatocellular carcinoma (HCC) is the most common histological type of primary liver cancer with poor long-term prognosis [1]. Moreover, its prognosis did not improve significantly despite the development of diagnostic and therapeutic tools compared to other common malignancies such as breast, prostate, and colon cancer. This poor outcome is attributable to absence of symptoms in the early stage, the unsatisfied sensitivity of the current surveillance method, limited therapeutic strategies, and a high recurrence rate after curative treatment [2]. Accurate and convenient cancer surveillance is the first step toward improving the overall prognosis of HCC.
Currently, several biomarkers are available for early detection of HCC. Alpha-fetoprotein (AFP) is the most widely used biomarker in combination with ultrasound for HCC surveillance among high-risk patients. It can also be used for the prognostication and prediction of treatment response. Despite its wide use in clinical practice, AFP alone is not endorsed by current practice guidelines for detecting HCC due to its limited sensitivity [3]. Given these unmet needs, developing highly accurate and non-invasive tools for early HCC detection has been the main interest of healthcare professionals and researchers for the past decades [4]. Alternative protein-based serum tumor markers such as AFP lectin fraction (AFP-L3), des-y-carboxyprothrombin (DCP), and the derived algorithm GALAD score (Gender, Age, AFP-L3, AFP, DCP) are promising biomarkers that can potentially improve HCC detection rate [3,4]. Recently, liquid biopsy technology has also emerged as a solution to address this growing need.
Liquid biopsy refers to the detection of byproducts that are shed from or produced during the proliferation of cancer cells [5]. Common targets of liquid biopsy include circulating tumor cells (CTCs), cell-free DNA (cfDNA), and extracellular vesicles (EVs). As a non-invasive procedure, liquid biopsy allows obtaining molecular information from cancer cells, making it an attractive option as a novel biomarker for early detection, prognostication, and disease monitoring in HCC.
This review article aims to provide an overview of CTCs, cfDNA, and EVs, discuss the technical challenges in applying these liquid biopsies, and address recent advancements in technology (Fig. 1), current clinical applications, and future directions in this field (Fig. 2).

LIQUID BIOPSY – CTC, cfDNA, AND EV IN HCC

The term “liquid biopsy” refers to a test that detects circulating cancer cells or DNA, RNA, and other molecules released by tumor cells through all types of body fluids [5]. Through this process, genetic, epigenetic, transcriptomic, and proteomic information representing cancer could be collected in a non-invasive manner. In this section, we focus on three common types of liquid biopsy in HCC: CTCs, cfDNA, and EVs. We briefly introduce their biology and characteristics, outline the technical challenges of applying these liquid biopsies, provide updates on recent technological advancements, and discuss their advantages and disadvantages (Table 1).

CIRCULATING TUMOR CELLS (CTCs)

Biology and characteristics

CTCs refer to cells that have detached from the tumor mass and circulated in the bloodstream for distant metastases. As malignant tumors grow, matrix metalloproteinases are secreted, breaking down the basement membrane and allowing the tumor cells to enter blood vessels or lymphatic vessels [6]. Once entering the blood vessels, tumor cells become CTCs, which undergo epithelial-to-mesenchymal transition (EMT) process. This process equips CTCs with mesenchymal properties to survive immune attacks, shear stress, and anoikis - a form of programmed cell death that occurs in anchorage-dependent cells when they detach from the surrounding extracellular matrix [7]. Only a few CTCs escape immune system surveillance and extravasate to metastasize to other organs. Given the nature of CTCs, their presence is regarded as a biomarker for both cancer detection and prognostication.

Technical challenges and technological advancements

CTCs are extremely rare in circulation, with a prevalence of one CTC per 106–107 white blood cells (WBCs) [8,9]. As such, efficient enrichment of CTCs from blood is crucial for downstream analysis. There are two main categories of technologies to enrich CTCs: immunoaffinity-based and physical property-based CTC enrichment techniques. The immunoaffinity-based techniques use antibodies to target antigens on the cell membrane, which can be employed for either positive selection to detect CTCs or negative selection to filter out WBCs [10]. For positive selection of CTCs, the most used marker is epithelial cell adhesion molecule (EpCAM), as many cancers originate from epithelium. The CellSearch® System is the first and only EpCAM-based method approved by Food and Drug Administration (FDA) for enrichment and enumeration of CTCs in metastatic breast, colorectal, and prostate cancers [11-13]. This system uses ferromagnetic beads coated with anti-EpCAM antibodies to capture CTCs, followed by magnetic-activated cell sorting for isolation. However, cancer cells often undergo EMT, and HCC CTCs are derived from hepatocytes rather than epithelial cells. Therefore, using anti-EpCAM antibodies alone may not be sufficient for effective enrichment of HCC CTCs [14]. To address this limitation, cocktails of different antibodies targeting asialoglycoprotein receptor (ASGPR), glypican‐3 (GPC3), and vimentin have been developed, demonstrating increased capture rates of HCC CTCs [15]. Microfluidic devices, such as CTC-ChipTM [16-19] and NanoVelcro ChipTM [20-23], which can also be used with different antibodies, have been developed to improve the capture efficiency and the purity of HCC CTCs. For example, nanosubstrates that mimic Velcro® and the overlying chaotic mixer of NanoVelcro ChipTM significantly increase the contact frequency and affinity between CTCs and the chip surfaces, achieving >80% capture efficiency in HCC patients [15]. Compared to the positive selection strategy mentioned above, negative selection utilizes anti-CD45 antibodies to target and deplete hematopoietic cells from the background [24], thereby enriching CTCs. However, negative selection is typically used in conjunction with positive selection, physical property-based isolation, or microfluidic devices, as the purity of enriched CTCs from negative selection alone is usually low [10]. The third generation of CTC-ChipTM, CTC-iChip [17,19,25], and its updated version combine either positive selection or negative selection with an arrayformat, inertial-enhanced design to fulfill high-throughput (107 cells/s) and enrich all CTC populations with high purity. A 10-gene signature in HCC CTCs enriched by CTC-iChip was quantified as a surrogate for the presence of CTCs and demonstrated as a promising biomarker for early detection of HCC [19].
The second CTC enrichment method, physical property-based isolation, leverages the differences in size, mechanical plasticity and dielectric mobility of CTCs [26]. Techniques in this category include microfiltration, density-based gradient centrifugation, dielectrophoresis, and microfluidic devices. As these methods are antigen-independent, they can address the issue of EpCAM loss during EMT and the heterogeneous antigen expression of CTCs. They also offer the advantages of relatively low cost and ease of downstream analysis, as there is no need to separate CTCs from coated antibodies. The Parsortix® PC1 system is an example of microfluidic device recently cleared by FDA to enrich CTCs in metastatic breast cancer patients based on cell size and deformability [27].
Despite enrichment using the aforementioned technologies, more than 1,000 WBCs per CTC still inevitably remain in the background [8]. Therefore, immunofluorescence staining is typically applied to distinguish CTCs from the background WBCs. CTCs are usually defined as cells with DAPI+ oval nuclei, CK+ cytoplasma, CD45- cytoplasma, and positive staining of targeted surface markers such as EpCAM, vimentin, or ASGPR, particularly for HCC [15,28]. The identified CTCs could either be enumerated or collected for single-cell analysis [28,29]. In addition to manual enumeration and identification of CTCs, bioinformatics analysis was proposed to select a panel of genes that are highly expressed in CTCs but lowly expressed in background WBCs [19,22,30]. This approach enables “bulk” genetic analysis of the samples after CTC enrichment, streamlining the entire analysis process for further clinical applications like HCC detection and prognostication.

Advantages and disadvantages of CTCs

Compared to other types of liquid biopsy, CTCs preserve the most complete molecular information due to the protection of their cell membranes. With the advancement of CTC isolation technologies and single-cell analysis, CTCs can be released from the captured substrates with high cell viability [28]. This enables downstream analyses of DNA, RNA and proteins to investigate the mechanisms of tumor progression, EMT, and metastasis, as well as to surrogate clonal evolution of the cancer. In addition, in vitro proliferation of the CTCs allows for functional tests to assess drug resistance and to understand the interactions between CTCs and epithelial cells, platelets, and immune cells during EMT, providing insights into the blood microenvironment [28].
However, CTCs also have several disadvantages. First, no standardized pre-analytical protocols exist for CTC studies.9 While the two FDA-cleared devices, CellSearch® and Parsortix® PC1 Systems, each have their own protocols and corresponding blood collection tubes (BCTs), they are not approved for use in HCC patients. Additionally, the choice of BCT varies based on the downstream CTC analysis. 9 For example, the CellSave BCT is used for CTC enumeration, 31 while acid citrate dextrose solution A (ACD-A) and ethylenediaminetetraacetic acid (EDTA) BCTs are preferred for CTC mRNA analysis [32,33]. With appropriate BCTs and storage at 4°C, CTCs and their molecular contents are generally stable for up to 3 days [34,35]. However, for molecular analyses, it is recommended to verify the stability of the target molecular panels over the processing interval to ensure study reliability. Second, despite advancements in automated reading system and artificial intelligence/deep learning that assist in the identification of CTCs [36,37], enumeration still heavily relies on manpower and requires significant experience, which is inevitably subjective to some degree. Third, CTCs are heterogenous as their originated tumor cells [38,39]. While continuously profiling these heterogeneous CTCs could help unravel the dynamic changes in the tumor and its evolving biology [24], this poses challenges for the CTC enrichment process. This is particularly true for immunoaffinity-based methods, as different CTC populations may express varying surface antigens. Lastly, due to their nature, CTCs are relatively rare in patients with early-stage cancer, which makes their utility as a biomarker for early detection of HCC challenging.

CELL-FREE DNA (cfDNA)

Biology and characteristics

CfDNA are small fragments of degraded DNA (<200 bp) that originate from disrupted or apoptotic cells and circulate in the bloodstream [40]. CfDNA derived from malignant cells is also known as circulating tumor DNA (ctDNA). In normal conditions, the concentration of cfDNA is typically low, but it increases in certain conditions such as exercise, inflammation, diabetes, and malignancy [41,42]. In HCC patients, cfDNA exhibits both quantitative changes, such as elevated levels of cfDNA, and qualitative changes, including copy number alterations (CNVs), single-nucleotide variations (SNVs), methylation patterns, 5-hydroxymethylcytosine (5hmC), and fragment features [40]. For CNVs in HCC cfDNA, arm-level amplifications of 1q and 8q, and deletions of 1p, 4q, and 8p, are frequently observed [43]. SNVs in TP53, CTNNB1, and the promoter region of TERT are common in cfDNA from HCC patients, offering potential for cancer detection [44,45]. DNA methylation is known to be involved in DNA regulation and carcinogenesis, notably appearing even in the early stage of tumor development. Therefore, cfDNA methylation has been regarded as a promising biomarker for early-stage detection of HCC [46]. Recent studies suggested that at least 10 to 50 cfDNA methylation markers are required to robustly distinguish HCC from controls [47,48], while around 200 cfDNA methylation markers are needed to differentiate between different cancers [49]. Alternatively, incorporating relevant clinical variables such as age, sex, AFP, AFP-L3%, and DCP along with a few cfDNA methylation markers is a common approach to enhance the sensitivity for HCC detection [47,48]. In addition to mutation and methylation patterns, cfDNA fragmentation has attracted the attention of many researchers. Genome-wide fragmentation profiles, fragmentation size, nucleosome footprint, DNA 5’ end motif, jagged ends, and endpoint locations were shown to provide valuable information to differentiate HCC patients [50].

Technical challenges and technological advancements

It Is estimated that ctDNA could be <1% of the total amount of cfDNA, with levels even lower at around 0.01% in early-stage cancer [51]. Unlike the various methods available for isolating CTCs, there are currently no reliable methods for enriching ctDNA from cfDNA. As the size of cfDNA in HCC patients tends to be shorter, approximately 144 bps [52], a study suggested applying fragment-size selection of <150 bp to increase the proportion of ctDNA [53]. However, this approach is not commonly used in most cfDNA-based biomarker studies. Instead, researchers are focusing on developing PCR and sequencing technologies to either increase sensitivity or broaden the genomic coverage to detect subtle changes in ctDNA. In general, PCR techniques are used to detect known SNVs and methylation markers, offering high sensitivity and quick results [54]. Among the promising commercial cfDNA platforms for HCC detection [55], Oncoguard® Liver by Exact Science has developed a PCR-based technology, the Target Enrichment Long probe Quantitative Amplified Signal (TELQAS) assay [56], to measure selected methylation markers. The TELQAS assay combines three key features: 1) a multiplexed pre-amplification process, 2) longer and high-melting-temperature probes, and 3) a second round of amplification of individual target loci, which enables sensitive and specific detection of low-abundance methylation targets in cfDNA without requiring whole-genome pre-amplification.
The other methods, besides PCR-based technologies, are next-generation sequencing (NGS), including whole-genome sequencing, whole-exome sequencing, and target-panel sequencing. While NGS provides a comprehensive view of the entire genomic map, those methods are more expensive and time-consuming [54]. For the detection of low-frequency SNVs in HCC ctDNA, Lee et al. [45] designed an algorithm that condenses hopped indexes for barcode-based targeted sequencing to eliminate PCR and sequencing errors, thereby achieving ultra-high depth and enhanced sensitivity. Similarly, HCCscreenTM by GENETRON uses barcode-based targeted sequencing with rapid amplification of cDNA ends and multiple primers to detect the SNVs in TP53, CTNNB1, and AXIN1, the promoter region of TERT, and the HBV integration breakpoint, which are included in a panel for HCC detection. The researchers further developed the Mutation Capsule Plus (MCP) technology [57], which enables 1) parallel profiling of mutation and methylation changes in the same reaction, 2) repeated profiling of a single cfDNA sample without sacrificing sensitivity, and 3) genome-wide screening for unidentified methylation markers. For methylation sequencing of cfDNA, HelioLiverTM’s ECLIPSE platform employs a gentler enzymatic conversion approach, yielding a greater quantity and quality of converted DNA compared to traditional bisulfite conversion, which is well-known for causing substantial DNA and signal damage [58]. Additionally, the ECLIPSE platform incorporates a bioinformatics pipeline named Cell Heterogeneity-Adjusted cLonal Methylation (CHALM) [59], which is able to assess the heterogeneity of sequenced bulk cfDNA, thereby enhancing the accuracy of quantifying differentially methylated markers. More recently, a research team led by Zhou developed a cost-effective assay named cell-free DNA Methylome Sequencing (cfMethyl-Seq) to profile CpG-rich regions in cfDNA [49]. Since these regions comprise only 3% of the whole genome, the cost of cfMethyl-Seq is significantly reduced while still covering over 90% of CpG islands and achieving more than 12× enrichment compared to whole-genome bisulfite sequencing. Leveraging cfMethyl-Seq, cancer-specific and tissue-specific hypermethylation and hypomethylation cfDNA markers were identified and demonstrated excellent performance in detecting and locating cancers, including HCC.

Advantages and disadvantages of cfDNA

Unlike the absence of standardized guidelines for the collection, processing, and storage of CTCs, National Cancer Institute’s (NCI) Biorepositories and Biospecimen Research Branch (BBRB) has introduced a cfDNA-specific guideline [60] to harmonize the pre-analytical processes and minimize potential cfDNA degradation, analytical artifacts, and contamination. This guideline includes evidence-based recommendations on BCTs, temperature and duration of ambient storage or transport, centrifugation speed and duration for blood processing, as well as cfDNA extraction and storage. Notably, if blood is collected in Streck tubes, processing can be delayed for up to 3 days. In addition, the pre-analytical centrifugation requirement involves only two steps within an hour and does not require specialized centrifugation equipment, making it feasible in standard laboratories. These advantages facilitate multicenter clinical trials investigating cfDNA’s potential for HCC detection and prognostication and simplify the establishment of institutional biorepositories for future studies. Indeed, compared to CTCs and EVs, the genomic landscape of HCC ctDNA is the most extensively profiled, and several cfDNA-based assays are commercially available and validated in large-scale clinical studies.
Despite its advantages, cfDNA-based assays are relatively expensive and time-consuming, especially for NGS-based assays. Since cfDNA is fragmented, some chromosomal analyses like karyotyping and fluorescence in situ hybridization cannot be performed. Additionally, as there are no reliable methods to isolate ctDNA from cfDNA, it is challenging to specifically attribute identified genetic or epigenetic characteristics to HCC. Finally, cfDNA heterogeneity may interfere with quantification results. However, several of the aforementioned studies [49,56,58] are underway to improve cost-effectiveness and minimize the impact of cfDNA heterogeneity.

EXTRACELLULAR VESICLES (EVs)

Biology and characteristics

EVs are a group of lipid-bilayer “cargo” vesicles containing proteins, lipids, RNA, DNA and non-coding RNA released by cells into the extracellular environment [61,62]. Their major functions are intercellular communication, transfer of “cargo” cell-to-cell, and modulating physiologic conditions. In HCC, EVs from various cells influence the tumor microenvironment [63] and drive cancer progression by modulating cancer cell proliferation and migration, angiogenesis, extravasation, immune escape, and metastasis [64,65]. Recent evidence also shows that EVs derived from HCC can promote tumorigenesis and cancer stemness [66,67]. As such, EVs are regarded as promising HCC biomarkers for early detection of cancer, prognostication, and monitoring cancer progression.

Technical challenges and technological advancements

Multiple EV isolation methods are endorsed by The International Society for Extracellular Vesicles (ISEV) and its latest guideline, “Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches.” [62] Among these methods, differential ultracentrifugation, performed at speeds of 10,000–20,000g for 10 to 90 minutes followed by 100,000–200,000g for 45 to 150 minutes, is the most commonly used for EV isolation in research. However, this method is time-consuming, and ultracentrifugation machines are not commonly available, which hinders its application in clinical settings. Precipitation using hydrophilic polymers, such as polyethylene glycol, is also commonly used to isolate EVs due to its low cost, speed, simplicity, and high yield [62,68]. But this method typically results in low-purity EVs, often co-precipitating with vesicle-free miRNA and proteins like albumin and lipoproteins [69]. Filtration leverages membranes with different pore sizes to isolate EVs, allowing not only for the separation of EVs from proteins but also for the harvesting of EVs of specific sizes. Compared to precipitation, EVs enriched through filtration are purer, though the yield is slightly lower. Additional advantages include the lack of requirement for special equipment and simplicity, although filter clogging and the co-retention of proteins of similar size remain inevitable. Size exclusion chromatography separates nanoparticles based on their size as they pass through the column packed with numerous porous polymers and buffers. Because EVs are larger than the pore size, they cannot enter the pores like smaller molecules, allowing them to flow through the column more quickly and be separated effectively. This method can achieve higher purity of isolated EVs compared to filtration. However, it is more time-consuming and may require an additional step to concentrate the diluted EV samples for downstream analysis.
Although the aforementioned methods are effective to isolate EVs and urine and plasma samples of HCC patients, they cannot distinguish between HCC-derived EVs from those of non-HCC origins, making it challenging to detect tumor-specific molecular information within HCC-derived EVs. To overcome this limitation, immunoaffinity-based isolation methods using antibodies to target specific EV surface molecules have been investigated to capture the HCC-associated EVs [70,71]. Julich-Haertel et al. [72] demonstrated that AnnexinV+ EpCAM+ ASPGR1+ EVs are elevated in HCC patient’s sera and decrease after curative treatment, indicating that utilizing these markers might be a potential method to enrich HCC-associated EVs. Assuming that the surface markers of EVs resemble those of the parental tumor cells due to their shared cytomembrane, Sun et al. [71] targeted EpCAM, ASPGR1, CD147, and GPC3 to purify HCC-associated EVs. The research team developed methyltetrazine-modified microbeads, termed Click Beads, which trigger a click chemistry reaction upon encountering EVs coated with antibodies modified with trans-cyclooctene. Compared with conventional streptavidin-biotin-mediated immunoaffinity-based approaches, the click chemistry reaction enables faster and irreversible purification [71,73], thereby improving capture efficiency and reducing nonspecific EV capture. In combination with duplex real-time immuno-PCR, the authors enabled sensitive quantification of EV subpopulations positive for both the aforementioned HCC-associated markers (EpCAM, ASPGR1, CD147, and GPC3) and EV-related markers (CD63 and CD9) [62], which have shown promise for detecting early-stage HCC. Microfluidic devices leveraging immunoaffinity and physical characteristics are also a novel strategy to purify HCC-associated EVs [62,68,74]. For example, the same research team developed EV Click Chips [70,75], which integrates the following features: 1) an antibody cocktail (EpCAM, ASPGR1, and CD147) to enrich HCC-associated EVs, 2) a chaotic mixer and nanostructured substrate to significantly increase the surface area as EVs flow through, thereby enhancing capture efficiency, and 3) click chemistry-mediated EV capture and disulfide cleavage-driven EV release, which further increases the purity of the targeted EVs for downstream analyses. Although immunoaffinity-based isolation methods, including microfluidics technology, can isolate HCC-associated EVs and achieve high EV purity, they have some limitations [62,68,74], including low EV yield, the cost of antibodies, and the need for separation between EVs and antibodies or devices if further functional studies are required.
Currently, there is no universal pre-analytical protocol for blood collection, processing, and storage in blood-based EV studies. Acknowledging that the initial quality of plasma and serum can significantly affect study results, the ISEV Blood Task Force developed the Minimal Information for Blood EV Research (MIBlood-EV) guideline to help researchers transparently report crucial pre-analytical information, thereby improving the reproducibility of results. Most recently, Dhondt et al. [76] conducted the first large‐ scale benchmarking study to comprehensively compare the impact of BCTs and blood processing intervals on blood sample quality and downstream analyses. In brief, plasma is preferred over serum for analyses, as less soluble platelet activation is observed in plasma, reducing potential confounding of the results. ACD-A, citrate, or EDTA BCTs are recommended for plasma collection, with ACD-A and EDTA approved for a blood processing interval of up to 8 hours after collection. Unlike CTC and cfDNA, which remain stable after 72 hours, significantly elevated levels of platelet-derived EVs and erythrocyte-derived EVs due to platelet activation and hemolysis are observed after 72 hours from blood collection, potentially affecting the study results. The Streck BCT can also be used for plasma collection in EV studies, although it offers no clear advantage over ACD-A, citrate, or EDTA BCTs regardless of the blood processing interval.
Urine is the second most commonly used biofluid in EV studies and offers the advantages of easy and noninvasive collection. The ISEV Urine Task Force has developed guidelines [77,78] for collection, processing, and storage to standardize and facilitate urinary EV studies. Similar to blood samples, urine samples should be processed within 8 hours after collection to prevent microbial growth. Further detailed recommendations for each processing step can be found in the quick reference card [78].

Advantages and disadvantages of EVs

Similar to CTCs, EVs are protected by a lipid bilayer and contain not only DNA but also RNA, proteins, and lipids, providing additional molecular information, while being easier to collect and preserve. Given the roles of EVs in HCC progression, functional studies of EVs can be conducted to reveal the underlying mechanisms when new EV-based biomarkers are discovered. Although blood samples for plasma collection in EV studies are recommended to be processed within 8 hours, which can pose challenges for fresh sample transfer, plasma can be obtained through a simple two-step centrifugation process that is feasible in both clinical and research laboratories. The plasma can then be stored and transported at –80°C for subsequent clinical EV studies. However, due to the lack of a standardized pre-analytical protocol for EV studies, factors such as BCTs, processing and storage times, and the number of freeze-thaw cycles may introduce technical variations. Unlike CTCs and cfDNA, research and biomarker exploration in the field of HCC EVs are still in the early stages of development. For example, given the small amounts of the HCC-derived EVs enriched using the novel technologies [70,71], more sensitive methods are required to detect and quantify scarce molecules, such as RNA, within these EV subpopulations. Lastly, larger scale clinical trials are needed to validate the findings of HCC EV biomarker studies.

ROLE OF LIQUID BIOPSY FOR EARLY DETECTION OF HCC

Early detection would be the most effective way to improve the prognosis in HCC patients. This section discusses the clinical utility of early detection according to the target substances of liquid biopsy. The main research findings are summarized in Tables 24 [79-94].

Early detection of HCC using CTCs

Earlier efforts to detect CTCs used immunoaffinity against EpCAM, one of the commonly used membrane-associated proteins, not expressed by normal blood cells. However, EpCAM positive rate was only about 35% in CTC [95]. This result is likely because its expression is low in the early-stage HCC and is often lost during the EMT process. To overcome these issues, research has been conducted using HCC-specific markers such as GPC3, a cell membrane-anchored protein, more frequently observed in moderately or poorly differentiated HCC tumor cells and ASGPR, a transmembrane protein exclusively expressed on the surface of hepatocyte [96-98]. While they did not show excellent detection performance as an individual marker, combination of EpCAM, GPC3 and ASGPR antibodies using microfluidic synergistic chip achieved a detection rate of up to 97% in patients with HCC at various stages [99]. On the other hand, analyzing various types of CTCs, such as epithelial, mesenchymal, and mixed type, can help with enhanced accuracy for early HCC detection [100]. Chen et al. [100] enumerated all the CTCs, including epithelial, mesenchymal, and mixed type, which showed better performance in discriminating between HCC and non-malignant liver disease compared to AFP.
More research has been conducted by combining CTCs with other biological markers to overcome the low sensitivity of CTCs. Combining CTCs with AFP resulted in better diagnostic yield compared to CTCs alone or AFP alone (0.821 vs. 0.774 vs. 0.669) [101]. Guanine nucleotide-binding protein subunit beta-4 (GNB4) and Riplet gene methylation showed high sensitivity and specificity when they were combined with CTCs [102]. Despite advancements in CTC technology to detect HCC, CTCs for HCC surveillance are not endorsed due to high error rates, low detection rates in early stages, and difficulties in developing standardized testing methods [103].

Early detection of HCC using cfDNA

In earlier cfDNA research, the primary focus was on the quantitative changes in cfDNA as a biomarker for early-stage HCC detection. Following the results of the Iizuka group, which showed that cfDNA concentration was higher in HCV-associated HCC patients compared to the HCV carrier group [104], several other studies reported that the concentration of cfDNA in HCC patients was 2 to 4 times higher than in those with chronic hepatitis, and nearly 20 times higher than in healthy individuals [105,106]. Additionally, tumor size, stage, and shorter survival time are correlated to cfDNA concentration. However, since cfDNA concentration can be influenced by liver inflammation, it would not be a reliable indicator for HCC, making it challenging to accurately determine the status of HCC [40].
Despite the clear evidence that analysis of alteration in cfDNA is highly beneficial for HCC detection, individual cfDNA alterations alone may lack sufficient diagnostic value for HCC [107]. On the other hand, the development of NGS technology has improved the understanding of the genetic alteration landscape in HCC. Recent studies using NGS have been able to detect most of the mutations present within tumors, and in some cases, the mutation detection rate was reported to be higher in liquid biopsy than in tissue biopsy. This indicates that intratumoral heterogeneity can be effectively identified, making it a powerful tool for improving early-stage diagnosis [108,109]. Guo et al. analyzed the cfDNA mutation profile using a targeted gene panel. High frequency of HCC-related gene mutations was observed in patients with chronic hepatitis B or liver cirrhosis. The area under a receiver operating characteristic curve (AUROC) of the gene mutation burden calculated from a panel of genes for HCC detection was 0.92 [110]. These results indicate that somatic mutation detected in cfDNA may be an excellent biomarker for the early detection of HCC. Also, genetic alteration of ctDNA provides valuable diagnostic value in combination with conventional serum protein tumor markers. Cohen et al applied CancerSEEK test, which is composed of protein biomarkers (CA-125, CEA, CA 19-9, hepatocyte growth factor, prolactin etc.) and 16 gene mutations of ctDNA (NRAS, CTNNB1, PIK3CA, FBSW7, APC, EGFR, BRAF, CDKN2A, PTEN, FGFR2, HRAS, KRA, AKT1, TP42, PPP2R1A, GNAS) to detect 8 different solid tumors, including liver cancer. Overall sensitivity of liver cancer was about 98% and specificity was over 99% [111]. These remarkable results suggest that cfDNA might play a crucial role in the early detection of HCC.
CfDNA methylation pattern also can provide valuable information to detect HCC [112]. Analyzing the methylation pattern of the SEPT9 promoter, an important tumor suppressor, in cfDNA demonstrated a potential role for HCC detection among cirrhotic patients [113]. HCC-specific circulating DNA methylation profiles identified 283 CpGs with methylation differences, enabling differentiation between HCC and non-HCC samples with AUROC of 0.957 (sensitivity=90%, specificity=97%) [114]. Blinded multicenter validation study using HelioLiverTM test, which is generated using methylated cfDNA markers, serum protein markers and patient demographics, demonstrated better overall detection ability of HelioLiverTM test (AUROC=0.944) compared to AFP (AUROC=0.851) and GALAD (gender, age, AFP-L3, AFP, and des-carboxy-prothrombin) score (AUROC=0.899). HelioLiverTM test also showed better sensitivity (75.7%) for detecting early-stage HCC (I/II) compared to AFP (≥20 ng/mL, 57%) or GALAD score (≥−0.63, 65%) [58]. Another multicenter study used mt-HBT (multitarget HCC blood test), which combined 3 methylation markers (HOXA1, TSPYL5 and B3GALT6), AFP and patient sex, demonstrated superior sensitivity for early-stage HCC (I/II, 82%) detection compared to AFP (≥20 ng/mL, 40%) or GALAD (≥–0.63, 71%) [115]. Another study also demonstrated that combining cfDNA methylation markers with traditional serum protein biomarkers provides excellent accuracy for HCC detection, with an AUROC of 0.87 [116].
CfDNA fragment pattern also provides valuable information for detecting HCC. Non-tumor cfDNA fragments are, on average, 167 bp in size, while ctDNA fragments are typically shorter, around 145 bp [117]. This result is potentially due to the highly active transcriptional state of specific regions. A multi-national study that used whole genome cfDNA fragmentome profile was conducted to detect HCC patients using a machine learning model. The sensitivity for detecting cancer was 88% in an average-risk population at 98% specificity and 85% among high-risk individuals at 80% specificity.118 Another study analyzed cancer-related mutations and fragment length profile classification model using three fragmentomic features of ctDNA through deep sequencing of 13 genes associated with HCC. They demonstrated an AUROC of 0.88, a sensitivity of 89%, and a specificity of 82% for HCC detection in the discovery cohort [119]. Chen et al. [120] applied a combined profile of 5-Hydroxymethylcytosine/Motif/Fragmentation/nucleosome footprint using NGS technology and showed 95% sensitivity and 97% specificity for detecting HCC from cirrhotic patients. A recent meta-analysis, which analyzed 33 papers, reported an overall sensitivity of 72.2% and a specificity of 82.3% for 7 quantitative studies and an overall sensitivity of 56.8% and a specificity of 88.2% for 26 qualitative studies [121].

EARLY DETECTION OF HCC USING EVs

It is well known that the concentration of EVs is higher in HCC patients compared to those with liver cirrhosis [122]. Biomolecular cargo in the EVs is a useful biomarker for HCC, with higher accuracy than EV concentration. EV hnRNPH1 mRNA levels in HCC patients were higher than in the control groups and showed good diagnostic performance to detect HCC [123]. Wang et al. [124] demonstrated that EV miR-122, miR-148a, and miR-1246 had superior performance to detect HCC compared to AFP. EV long noncoding RNAs (IncRNAs) have shown promising results in early-stage HCC detection too. A Korean study utilizing six upregulated EV IncRNAs reported excellent accuracy, with an AUC of 0.96 [125]. Another study showed that combining EV lncRNA and AFP further enhances the accuracy of EV-based biomarkers for HCC detection [126]. EV-derived LDHC mRNA demonstrated better AUC compared with serum-only LDHC mRNA for early HCC detection (EV LDHC 0.945 vs. serum LDHC 0.838) [127].
Our team developed an HCC EV purification system (i.e., EV Click Chips) by synergistically integrating covalent chemistry-mediated EV capture/release, multi-marker antibody cocktails, nanostructured substrates, and microfluidic chaotic mixers. The purified HCC EVs were subjected to reverse-transcription droplet digital PCR for quantification of 10 HCC-specific genes, i.e., alpha-fetoprotein (AFP), glypican 3 (GPC3), albumin (ALB), apolipoprotein H (APOH), fatty acid binding protein 1 (FABP1), fibrinogen beta chain (FGB), fibrinogen gamma chain (FGG), alpha 2-HS glycoprotein (AHSG), retinol binding protein 4 (RBP4), and transferrin (TF). The HCC EV-derived molecular signatures could detect early-stage HCC (BCLC stage 0-A) with high sensitivity (94.4%), specificity (88.5%), and AUROC of 0.93, which was superior to serum AFP (AUROC of 0.69) [70]. More recently, our team developed an HCC EV surface protein assay, composed of covalent chemistry-mediated HCC EV purification and real-time immuno-polymerase chain reaction readouts, quantifying subpopulations of HCC derived EVs. An HCC EV ECG score, calculated from the readouts of three HCC EV subpopulations can detect early HCC with AUROC of 0.94, which was higher compared to AFP (AUROC of 0.79). ECG score also demonstrated excellent early-stage HCC (BCLC 0-A) detectability (sensitivity=91%, specificity=81%) [71].

ROLE OF LIQUID BIOPSY FOR PROGNOSTICATION

In the following section, we discuss examples of clinical applications related to prognostication for each target substance of liquid biopsy. Main research findings are summarized in Tables 57 [128-142].

Prognostication using CTCs

Several studies have shown that the number of CTCs is correlated with tumor size, portal vein tumor thrombosis, tumor stage, differentiation, and shorter survival [15,143]. Our team developed the HCC-CTC mRNA scoring system for the prognosis of HCC. This system is composed of 2 steps: enrichment of CTCs from blood collected from HCC patients using the NanoVelcro CTC assay and lysis of the enriched CTCs to extract mRNA; second, reverse transcription to obtain HCC-CTC-derived cDNA. The extracted cDNA is reacted with the HCC-CTC RS panel, a final set of 10 genes with prognostic value, using the Nanostring nCounter technique. Based on the results, patients were categorized into low-risk and high-risk groups to compare their prognosis. In the validation cohort, the overall survival (OS) for the high-risk group showed significantly worse results (P=0.02), HCC-CTC panel was an independent predictor of survival (HR 5.7; 95% CI 1.5–21.3; P=0.009), along with BCLC stage [30]. A long-term study tracking patients with locally advanced or metastatic HCC reported that a significant increase in CTC count is associated with worsening treatment response [144]. The mesenchymal phenotype of CTCs (M-CTCs) is also associated with poor prognosis, including not only tumor aggressiveness and portal vein tumor thrombosis but also reduced OS [145,146].

Prognostication using cfDNA

CfDNA holds strong prognostic value for HCC due to its short half-life and reflection of tumor burden and biology. One study demonstrated that SNV and CNV of cfDNA correlate with tumor burden and predict prognostic outcomes better for recurrence-free survival (RFS) and OS than traditional protein biomarkers such as AFP, AFP-L3% and DCP [147]. A similar study showed that poor disease-free survival is observed in HCC patients with cfDNA mutations in their post-operative plasma [148]. Oh et al. analyzed the prognosis of HCC patients using cfDNA concentration and the I-score, which indicates genomic instability. cfDNA is an independent prognostic factor for OS (HR 2.51; 95% CI 1.62–3.89; P<0.001), while the I-score is an independent prognostic factor for both time to progression (TTP) (HR 1.71; 95% CI 1.13–2.58; P=0.011) and OS (HR 1.85; 95% CI 1.16–2.96, P=0.010) [149].
Multiple studies revealed the association between epigenetic alteration of cfDNA and HCC prognosis. One study developed a combined prognostic score (cp-score) from 8 methylation markers and revealed that the high-risk group based on cp-score is an independent predictor for survival (HR 1.548; 95% CI 1.246–1.924; P<0.001) [48]. A recent multicenter study validated 20 differentially methylated regions (DMRs), which predict a high recurrence risk (HR 3.33; 95% CI 1.87–5.92; P<0.001) [150]. A systematic review analyzing eight studies (5 studies with pre-treatment samples, 2 studies with pre & post-treatment samples, and 1 study with post-treatment samples) reported that the mutation of ctDNA was independently associated with reduced disease-free survival (HR 3.01; 95% CI 1.23–11.30) [151]. A study using data from participants in the SORAMIC trial showed that cfDNA levels significantly correlated with metastases and survival. In addition, cfDNA kinetics over time revealed a trend with the clinical history of the patients, supporting its use as a biomarker to monitor therapeutic response [152].

Prognostication using EVs

Several studies demonstrated the prognostic value of EV in HCC. High EV miRNA-21 levels positively correlated with the stage of HCC and linked to the PTEN/Akt signaling pathway, which enhances EMT and tumor proliferation. That suggests that EV miRNA-21 could be a potential predictive prognostic marker of HCC [153]. The EV miRNA-665, which is linked to the Mark/ERK anti-apoptotic pathway, also showed a correlation with HCC progression and shorter survival time [154]. Some studies revealed that the prognostic impact of EV is mediated by angiogenesis. EV miRNA-210 suppresses the expression of SMAD4/STAT6, therefore strengthening tumor angiogenesis [155]. Another study has reported that EV miR-103 inhibits VE-cadherin, p120, and zonula occludens, weakening endothelial cell junctions and ultimately leading to tumor metastasis [156]. On the other hand, the downregulation of tumor suppressor EV miRNAs can also indicate poor prognosis. Studies have reported that decreased EV miRNA-125b and miRNA-638 levels are associated with shorter time to recurrence (TTR) and OS [157,158]. EV LncRNA-ATB, which induces tumor invasion, EMT, and metastasis, is positively correlated to advanced HCC stage, portal vein thrombosis, leading to reduced OS and progression-free survival (PFS) [159]. EV circular RNAs also showed prognostic value for HCC. Upregulation of circPTGR1, which acts in decreasing interaction between miRNA-449 and MET, is associated with advanced cancer stage including metastasis and poor prognosis [160]. EV may predict the treatment response to immunotherapy. The high expression of circUHRF1 is associated with a decreased NK cell proportion in blood (P<0.001), which inhibits NK cell-derived IFN-γ and TNF-α secretion in HCC, ultimately leading to immunosuppression. Also, high expression of EV circUHRF1 is an independent prognostic factor for recurrence (HR 1.762; 95% CI 1.172–2.428; P=0.019) [161], suggesting that EV cirUHRF1 may induce poor response to anti-PD1 immunotherapy.

ROLE OF LIQUID BIOPSY FOR MINIMAL RESIDUAL DISEASE (MRD) AND DISEASE MONITORING

MRD in solid tumors refers to a small number of cancer cells remaining after treatment [162]. For surgical resection, a wider resection margin can improve survival by decreasing the chance of MRD [163]. Microvascular invasion is also known to be an important factor in predicting early recurrence from MRD [163]. However, these surrogates need surgical procedures, making continuous monitoring after surgery not feasible in routine clinical practice. With the recent approval of adjuvant treatment for HCC in high-risk patients, the detection of MRD became even more important. Therefore, there is a clinical unmet need in HCC to develop non-invasive, validated methods to detect MRD.
One notable study reported that a CTC count greater than 15 and mesenchymal CTCs greater than 2% serve as predictors for early recurrence and metastasis. Importantly, in the post-operative monitoring of 10 patients, an increase in CTC count was recognized before clinically detectable nodules were found [146]. These results support that CTC could be a surrogate marker for MRD.
Regarding cfDNA, one study reported that patients with detectable cfDNA at 1 and 4 months after hepatic resection demonstrated a significantly shorter RFS [164]. Similarly, overexpressed miR-92b at 1 month after liver transplantation was associated with early recurrence [165]. Xu et al. [162] reported that a positive MRD monitoring gene (TP53, TERT, CTNNB1, APC, RMB10, NTRK3, NOTCH1, NOTCH2, NF1, CREBBP, GLI3, CDKN2A, EZH2) is an independent predictor of shorter RFS (HR 13.00; 95% CI 2.6–69.0). A recent pilot study compared protein surface markers of EVs in patients with recurrent HCC and non-recurrent HCC. When comparing 1 year after hepatic surgery, they revealed significantly lower EV CD31 levels in the recurrent HCC group than in the non-recurrent HCC group (P<0.01) [166], suggesting that EVs may play a role in the early detection of recurrent HCC. Yi et al. measured EV lncRNA (LUCAT-1, EGFR-AS-1) in HCC patients who underwent hepatic resection to assess its prognostic role. Interestingly, a significant number of patients who achieved complete radiologic response experienced recurrence or progression in the context of positive EV dual lncRNA results [167]. This suggests that EV lncRNA may have the potential to be a biomarker for MRD. Sefrioui et al. analyzed ctDNA with TERT mutation and cfDNA in patients who underwent transarterial chemoembolization (TACE). When comparing preprocedural and 1 month post procedural cfDNA and ctDNA levels, the progression rate was significantly higher in the non-responder group compared to the responder group in any type of DNA (responder vs. non-responder=4.3% vs. 80%, P<0.001) [168].
Monitoring for disease recurrence or progression after cancer treatment is a critical component of HCC care continuum. Protein biomarkers such as serum AFP are well known to play a role in monitoring after surgical resection, locoregional therapy, and immune therapies [3,169]. However, due to low sensitivity and applicability, there is an unmet need for more accurate and sensitive biomarkers. Liquid biopsy could be particularly helpful for recurrence and progression monitoring among HCC patients with normal AFP levels.
Winograd et al. observed that in a small set of patients, those with PD-L1 positive CTCs seemed to have a better response to immune checkpoint inhibitors (ICI) compared to those with PD-L1 negative CTCs. This suggests that PD-L1 expression on CTCs could serve as a predictive biomarker, helping to identify which patients are more likely to benefit from immunotherapy [21]. Nosaka et al. [170] assessed serial treatment response before and after Atezolizumab plus Bevacizumab by investigating PD1 RNA expression in CTCs. PD-L1 expression decreased significantly in cases with partial response or stable disease [170]. Rau et al. sequentially measured CTC counts in patients who received various treatments, and in most cases, the CTC counts were consistent with the patient’s radiologic treatment response. Notably, in patients whose AFP levels were normal but did not show a complete response in CT or MRI scan, the CTC count accurately reflected the patient’s actual status [113]. This highlights the potential value of CTC count as a biomarker for monitoring treatment response and recurrence in the future. Regarding ctDNA, Ikeda et al. [171] suggested that cfDNA could be a biomarker for treatment response as most HCC patients had detectable cfDNA alteration, which can reflect the burden of diseases. However, more studies are required to establish the use of CTC counts, cfDNA, or EV for monitoring HCC recurrence, given the lack of robust evidence.

CLINICALLY AVAILABLE TESTS AND ONGOING CLINICAL TRIALS

The following section will show currently applicable technologies. HelioLiverTM test serves as a notable example. As previously described, it demonstrates superior HCC detection rate compared to serum AFP and the GALAD score [58]. Furthermore, in the very recently completed CLiMB trial, the HelioLiverTM test showed outperformed sensitivity and non-inferior specificity compared with ultrasound as well as ultrasound with AFP (≥20 ng/mL) (sensitivity=47.8% vs. 28.3% vs. 39.1%). HelioLiverTM test detected HCC lesion of ≤2 cm with sensitivity of 28.6% (95% CI 11.3–52.2). In contrast, ultrasound failed to detect any HCC lesion of ≤2 cm [172]. The Oncoguard® Liver platform, which combined 3 methylation markers (HOXA1, TSPYL5 and B3GALT6), AFP and patient sex, demonstrated superior sensitivity for early-stage HCC (I/II, 82%) detection compared to AFP (≥20 ng/mL, 40%) or GALAD (≥–0.63, 71%) [115]. A new clinical trial, known as ALTUS is on the way. This observation trial aims to evaluate the sensitivity and specificity of Oncoguard® in adults at increased risk for HCC [173].
HCCscreenTM by GENETRON uses barcode-based targeted sequencing with rapid amplification of cDNA ends and multiple primers to detect the SNVs in TP53, CTNNB1, and AXIN1, the promoter region of TERT, and the HBV integration breakpoint, which are incorporated into a panel for HCC detection. This test is commercially available in China based on Qu et al.’s research findings demonstrating that the HCCscreenTM achieved a sensitivity of 85% and specificity of 93% (AUROC=0.928) in distinguishing 65 HCC patients from 70 non-HCC patients among individuals with hepatic nodules and/or elevated AFP [174]. After that, Wang et al. applied the Mutation Capsule Plus (MCP) technology to the HCCscreenTM that follows for parallel profiling of mutation and methylation changes on a single cfDNA sample. Their validated retrospective cohort with 58 HCC and 198 non-HCC showed 90% sensitivity with 94% specificity [175]. The HCC blood test by Epigenomics, which is PCR assay for the assessment of methylated Septin 9 in bisulfite-converted DNA, is also available based on the case-control study, which showed 76.7% sensitivity and 64.1% specificity from 60 early stage HCC and 103 non-HCC patients, and also showed superior to serum AFP regarding detectability.176 Although neither of these tests has yet received FDA approval, they nonetheless offer considerable value in patient care.
Building on the currently commercially available test discussed above, we now turn to an exploration of ongoing clinical trials. Among the ongoing clinical trials listed in Table 8 [177-183], those conducted by Exact Sciences and Helio Genomics are particularly noteworthy. Additionally, emerging research findings suggest these trials may assist in predicting responses to immune checkpoint inhibitors, highlighting the growing interest in focus on this area.

BARRIERS TO CLINICAL TRANSLATION & CONCLUSION

Liquid biopsy allows non-invasive serial monitoring of HCC and detection of intratumoral heterogeneity through genomic analysis. Figure 2 provides an overview of potential future application of liquid biopsy for early detection, prognostication, and detection of minimal residual disease in HCC. We anticipate that liquid biopsy will enhance the detectability of early-stage HCC, positioning itself as a complementary or alternative test to the current standard imaging study with or without AFP [2]. Considering the low sensitivity of AFP and the operator-dependent nature of ultrasound, liquid biopsy could become a standardized tool for HCC surveillance. Liquid biopsy can enable the early detection of minimal residual disease post-treatment, leading to additional interventions that ultimately improve patient outcomes. Furthermore, it can monitor treatment efficacy following locoregional or systemic treatments, providing accurate assessments. However, several areas still need improvement for successful clinical translation. Despite advancements in various diagnostic technologies, the lack of standardized protocols for CTCs and EVs, background noise, and relatively high cost of NGS remain a critical issue. Furthermore, while technological advancement progresses, improving accuracy for early HCC detection remains a pressing need. The development of artificial intelligence (AI) would be one of the solutions to address these issues [184]. Due to the nature of blood tests, liquid biopsy cannot provide spatial information about HCC. Further refinement of liquid biopsy using multi-omics data, which encompass gene mutations, DNA methylation, fragmentome, proteome, and nucleosomes from CTCs, ctDNA and EVs in a patient’s blood sample all at once, may further enhance the accuracy of these novel biomarkers [185]. Also, research utilizing single-molecule sequencing techniques is now underway. This approach is expected to address limitations associated with conventional NGS, such as the inability to efficiently detect molecules longer than 600 bp. Additionally, it offers a solution to the issue of DNA degradation caused by disulfide treatment during DNA methylation analysis [186,187]. Ultimately, large-scale clinical trials are essential to validate its effectiveness and facilitate its adoption in clinical practice for HCC. Through these efforts, liquid biopsy could significantly aid in the entire spectrum of HCC care continuum.

FOOTNOTES

Authors’ contribution
Ju Dong Yang, Jaeho Park and Yi-Te Lee made contributions to the following: the conception and design of the study, or acquisition of data, or analysis and interpretation of data.
All authors have made contributions to all of the following: (1) drafting the article or revising it critically for important intellectual content, (2) final approval of the version to be submitted. The manuscript, including related data, figures and tables has not been previously published and that the manuscript is not under consideration elsewhere.
All authors approve the final version of the manuscript, including the authorship list and agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Acknowledgements
Figure 1 and Figure 2 are created in BioRender.
This work is supported by National Institutes of Health (R01CA277530, R01CA255727, R01CA253651, R01CA253651-04S1, R01 CA273925, R21CA280444, R01CA246304, U01EB026421, K08CA259534, R44CA288163, and U01CA271887). The funders had no role in the collection of data; the design and conduct of the study; management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Conflicts of Interest
Dr. Ju Dong Yang provides a consulting service for Fuji-Film Medical Sciences, Exact Sciences, AstraZeneca, Eisai, Exelixis, and Merck. Dr. Yazhen Zhu is a co-founder and shareholder in Eximius Diagnostics Corp.
Following the management plan provide by UCLA Conflict of Interest Review Committee, Dr. Hsian-Rong Tseng would like to disclose that (1) he has a financial interest in CytoLumina Technologies Corp. and Pulsar Therapeutics Corp., and (2) the UC Regents have licensed intellectual properties invented by Dr. Tseng to CytoLumina and Pulsar. Dr. Vatche Agopian provides a consulting service for Merck, Eximius Diagnostics Corp, and Early Diagnostics Corp.

Figure 1.
Concept and workflow of liquid biopsy in HCC, including CTCs, EVs, and cfDNA. This schematic figure highlights the key components of the concept and workflow of liquid biopsy in HCC: the pre-analytical process, technical challenges associated with each type of liquid biopsy, technology development to address these challenges and clinical validation of liquid biopsy assays. CfDNA, cellfree DNA; CTC, circulating tumor cell; ctDNA, circulating tumor DNA; EV, extracellular vesicle; HCC, hepatocellular carcinoma; NGS, next-generation sequencing; PCR, polymerase chain reaction; WBC, white blood cell.

cmh-2024-0541f1.jpg
Figure 2.
Future application of liquid biopsy for early detection, prognostication, and detection of minimal residual disease in HCC. (A) Early detection of HCC. Signals from CTCs, cfDNA, and EVs are measured every 6 months in patients with cirrhosis or chronic hepatitis B who require HCC surveillance. Once these signals exceed predefined cut-off values, diagnostic methods such as CT or MRI are used to confirm an HCC diagnosis. (B) Prognostication of HCC. Blood samples are collected from HCC patients before surgery or locoregional therapies. Based on signals from CTCs, cfDNA, and EVs, HCC patients are classified into high-risk and low-risk groups for recurrence or progression. For high-risk patients, adjuvant therapy and close monitoring of early recurrence or progression may be warranted. (C) Detection of MRD in HCC. After curative surgery or locoregional therapies, blood samples are collected from HCC patients. High signals from CTCs, cfDNA, and EVs indicate an increased likelihood of MRD, warranting adjuvant therapy and close monitoring for early recurrence. CfDNA, cell-free DNA; CTC, circulating tumor cell; EV, extracellular vesicle; HCC, hepatocellular carcinoma; MRD, minimal residual disease.

cmh-2024-0541f2.jpg
Table 1.
Comparison of CTCs, cfDNA, and EVs as liquid biopsies for HCC
Comparative features CTCs CfDNA EVs
Molecular contents DNA, RNA, proteins, lipids DNA DNA, RNA, proteins, lipids
Sources Blood (peripheral blood mononuclear cells) Blood, urine Blood, urine
Omics analysis capability Genomic, epigenomic, transcriptomic, proteomic, metabolomic analyses Genomic and epigenomic analyses Genomic, epigenomic, transcriptomic, proteomic, metabolomic analyses
Functional analysis capability Yes No Yes
Single cell analysis Yes No No
Standardized pre-analytical protocols No Yes (NCI Biospecimen Evidence-Based Practices) No
Blood collection tube CellSave BCT is used for CTC enumeration; EDTA BCT, Streck BCT or other cfDNA stabilizing BCT ACD-A, citrate, EDTA, Streck BCTs
ACD-A and EDTA BCTs for CTC mRNA analysis
Blood processing interval Up to 3 days at 4°C; stability of molecular panels over processing intervals needs to be verified EDTA BCT: ideally within 2 hours; 4 hours at 4°C/RT or 24 hours at 4°C is acceptable Within 8 hours
Streck BCT: Up to 3 days at RT
Technical challenges in detection Rarity in blood (1 CTC per 106-107 WBCs) Requirement of high sensitivity and broad genomic coverage to detect subtle changes in ctDNA Purification of HCC-associated EVs from bulk Evs
Other limitations Subjectivity in CTC enumeration; Not an ideal candidate for early HCC detection due to its inherent nature High cost of NGS Challenges in molecular profiling in the small amounts of EV subpopulations
Clinical validation status No ongoing clinical trials for early detection and prognostication of HCC Several ongoing clinicals trials to validate the performance of cfDNA-based assays for early detection of HCC No ongoing clinical trials for early detection and prognostication of HCC

ACD-A, acid citrate dextrose solution A; BCT, blood collection tube; cfDNA, cell-free DNA; CTC, circulating tumor cell; ctDNA, circulating tumor DNA; EDTA, ethylenediaminetetraacetic acid; EV, extracellular vesicle; HCC, hepatocellular carcinoma; NCI, National Cancer Institute; NGS, next-generation sequencing; RT, room temperature; WBC, white blood cell.

Table 2.
CTCs for early detection of HCC
Author Year Cohort Yield (sensitivity, specificity) Isolation technique/biomarker
Bahnassy et al. [79] 2014 Prospective CK19+ CTCs (sen: 87.1%, spe: 82.5%) Flow cytometry, RT-PCR
120 HCC/30 HCV/33 healthy control CD90+ CTCs (sen: 82.5%, spe: 89.6%) CK19, CD 133 and CD90
CD133+ CTCs (sen: 40%, spe: 6.3%)
Armakolas et al. [80] 2022 Prospective Sen: 46% EpCAM, vimentin, AFP, sMVP and qRT-PCR
89 HCC/28 cirrhosis
Cheng et al. [101] 2018 113 HCC/57 CLD Total CTCs >2 (sen: 62%, spe: 90%) CanPatrol: assay to isolate each type of CTCs
Epithelial CTCs >0 (sen: 45%, spe: 79%)
Mesenchymal CTCs >0: (sen: 49.6%, spe: 87.7%)
Liang et al. [102] 2022 17 HCC/11 HBV cirrhosis sen: 70.6%, spe: 90.9% CTCBIOPSY device
Guo et al. [81] 2018 395 HCC/201 HBV/100 benign liver disease/210 healthy control sen: 72.5%, spe: 95% Multimarker qRT-RNA
Detection flatform
Bahn et al. [82] 2018 54 HCC/39 CLD CTC> 4/10 mL: sensitivity: 81% CTC-iChip digital PCR with glypican-3
10 healthy control

CLD, chronic liver disease; CTC, circulating tumor cell; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; qRT-PCR, quantitative reverse transcription polymerase chain reaction; RT-PCR, reverse transcription polymerase chain reaction; sen, sensitivity; spe, specificity; EpCAM, epithelial cell adhesion molecule; AFP, alpha-fetoprotein; sMVP, soluble microvascular permeability; CTCBIOPSY, circulating tumor cell biopsy; CTC-iChip, circulating tumor cell isolation chip.

Table 3.
CfDNA for early detection of HCC
Author Year Cohort Yield (sensitivity, specificity) Isolation technique /biomarker
Chen et al. [83] 2024 Cross-sectional study PreCar Score (sen: 51.3%, spe: 95.5%) PreCar score using cfDNA genomic features. (Nucleosome footprint, 5-hydroxymethylcytosine, fragment size, 5’ end motif, copy number variation)
510 HCC (BCLC 0/A) US (sen: 23.7%, spe: 99.37%)
4367 LC PreCar+US (sen: 60.5%, spe: 95.08%)
Lin et al. [58] 2022 HCC 35 (I/II) HelioLiverTM test (sen: 76%, spe: 91%) NGS with ECLIPSE platform
125 CLD AFP (≥12.1 ng/mL,sen: 57%) 28 target genes (77CpG sites)
GALAD (≥−0.63, sen: 65%)
Chalasani et al. [115] (follow-up study of Kisiel JB et al) 2022 78 HCC (BCLC 0-A) mt-HBT (Oncoguard® by Exact Science) (sen: 82%, spe: 87%) Target Enrichment Long probe Quantitative Amplified Signal (TELQAS) assay
245 CLD AFP (≥20 ng/mL, sen: 40%)
GALAD (≥0.63, sen: 71%) HOXA1, TSPYL5, B3GALT6
Kisiel et al. [56] 2019 95 HCC (BCLC 0-D) AUROC (6 markers vs. AFP) =0.93 vs. 0.74 TELQAS assay/
51 LC/98 HC 6 methylation markers
Cai et al. [84] 2019 1204 HCC (BCLC 0-C) HCC vs. CLD (sen: 82.7%, spe: 76.4%) 5hmC-Seal/
392 LC/958 HC BCLC 0 vs. non-HCC: AUC=0.846 32 gene markers
Oussalah et al. [113] 2018 98 HCC (BCLC 0-D) Pooled AUROC: 0.940 MSP/
191 non-HCC HCC (BCLC A): AUROC=0.863 SEPT9 methylation
Cohen et al. [111] 2018 44 HCC (BCLC 0-D) HCC (sen: 100%, spe>99%) NGS/
8 proteins+1933 ctDNA mutations
Jiang et al. [85] 2015 90 HCC (BCLC A-B) HCC vs. HC (sen: 80%, spe: 94%) NGS/
67 CHB/36 LC/ 32 HC WGS (plasma DNA size measurement)
Han et al. [112] 2014 160 HCC (TNM I-IV) TGR5+AFP (sen: 65%, spe: 85.2%) MSP/
TGR5 promoter hypermethylation
Huang et al. [86] 2014 66 HCC (TNM I-IV) sen: 65.3%, spe: 87.2% Pyrosequencing/
34 CLD INK4A
Piciocchi et al. [87] 2013 66 HCC sen: 91%, spe: 43% qRT-PCR/
35 LC/41 CH (HCV) hTERT
Huang et al. [105] 2012 72 HCC (TNM I-IV) cfDNA (sen: 90.2%, spe: 90.3%) qRT-PCR/
37 LC cfDNA+AFP (sen: 95.1%, spe: 94.4%) NA

AFP, alpha-fetoprotein; AUROC, area under the receiver operating characteristic curve; BCLC, Barcelona clinic liver cancer; cfDNA, cell-free DNA; CLD, chronic liver disease; CHB, chronic hepatitis B; ctDNA, circulating tumor DNA; GALAD, gender; age; AFP-L3; AFP; and des-carboxy-prothrombin; HC, healthy control; HCC, hepatocellular carcinoma; hTERT, human telomerase reverse transcriptase; LC, liver cirrhosis; mt-HBT, multitarget hepatocellular carcinoma blood test; MSP, methylation-specific polymerase chain reaction; NA, nucleic acid; NGS, next generation sequencing; qRT-PCR, quantitative real-time PCR; sen, sensitivity; SEPT9, septin-9; spe, specificity; TELQAS, target enrichment long-probe quantitative amplified signal; TGR5, Takeda G-protein-coupled Receptor 5; TNM, tumor; nodes; and metastasis; US, ultrasound; WGS, whole genome sequencing.

Table 4.
EVs for early detection of HCC
Author Year Cohort Sensitivity, specificity Marker type Expression level Isolation technique/biomarker
Sun et al. [71] 2023 Validation cohort 91%, 81% Amounts of EV subpopulations EV surface protein assay (Click Bead+immune-PCR)/
HCC 35 (BCLC 0-A) ECG score from 3 EV subpopulations
LC 37
Son et al. [88] 2023 External cohort AUROCs N/A qRT-PCR (quantitative reverse)/
28 HCC (TMN I) SF3B4=0.940 EV-SF3B4
49 CH/LC AFP=0.785
Kim et al. [89] 2021 HCC 72 (BCLC 0-D) Positive rate: 86–96% (stage I) lncRNA qRT-PCR/
LC 25, CH 21, MALAT1, DLEU2, HOTTIP, SNHG1
von Felden et al. [90] 2021 HCC 105 (BCLC 0-A) 85%, 94% smRC qRT-PCR/
CLD 85 smRC_135709, smRC_119591, smRC_48615
Ghosh et al. [91] 2020 38 HCC (TMN I-III) 59%, 95% miRNA ExoEnrichTM instant exosome isolation Kit & anti-ASGR2/
35 CH+25 LC miR-10b-5p, miR-221-3p, miR-223-3p & miR-21-5p
Kim et al. [125] 2020 32 HCC (BCLC 0) 94%, 85% lncRNA ExoQuickTM Exosome precipitation solution/
28 CH+35 LC LINC00853
Huang et al. [92] 2020 122 HCC 80%, 76.5% lncRNA RiboTM Exosome isolation reagent/
43 LC Lnc85
Sun et al. [93] 2020 71 HCC (TMN I-IV) 91%, 78% circRNA Ultracentrifugation/
40 HD Circ_0004001, Circ_0004123, Circ_0075792
Cui et al. [127] 2020 50 HCC (TMN I, II) 88%, 93% mRNA exoRNeays Midi Kit/
100 HD LDHC
Sun et al. [70] 2020 36 HCC (BCLC 0-A) 84%, 88% mRNA HCC EV digital scoring assay (EV Click Chip+RT-ddPCR)/mRNA Panel (AFP, GPC3, ALB, APOH, FABP1, FGB, FGG, AHSG, RBP4, TF)
26 LC
Wang et al. [124] 2018 50 HCC (TMN I-III) 86%, 88% miRNA+AFP Ultracentrifugation, filtration & precipitation/
40 LC miR-122, miR-148a & AFP
Arbelaiz et al. [94] 2017 29 HCC 83%, 90% Protein Ultracentrifugation/
12 iCCA FGG
Wang et al. [122] 2013 28 HCC (TMN I) 63%, 89% Amounts of EV Ultracentrifugation/
40 LC Amount of total EVs

AFP, alpha fetoprotein; AHSG, alpha-2-Heremans-Schmid-glycoprotein; ALB, albumin; APOH, apolipoprotein H; AUROC, area under the receiver operating characteristic curve; BCLC, Barcelona Clinic Liver Cancer; CH, chronic hepatitis; circRNA, circular RNA; CLD, chronic liver disease; DLEU2, deleted in lymphocytic leukemia 2; EV, extracellular vesicle; FABP1, fatty acid binding protein 1; FGB, fibrinogen beta chain; FGG, fibrinogen gamma chain; GPC3, glypican-3; HCC, hepatocellular carcinoma; HD, healthy donor; HOTTIP, HOXA transcript at the distal tip; iCCA, intrahepatic cholangiocarcinoma; LC, liver cirrhosis; LDHC, lactate dehydrogenase C; lncRNA, long non-coding RNAs; MALATI, metastasis associated lung adenocarcinoma transcript 1; miR-122, microRNA-122; miR-148a, microRNA 148a; miRNA, microRNA; mRNA, messenger RNA; smRC, small RNA clusters; qRTPCR, quantitative reverse transcription polymerase chain reaction; RBP4, retinol-binding protein 4; RT-ddPCR, reverse transcription droplet digital PCR; SF3B4, splicing factor 3b subunit 4; SNHG1, small nucleolar RNA host gene 1; TF, transcription factor; TNM, Tumor; Nodes and Metastasis.

Table 5.
CTCs for prognostication of HCC
Author Year Cohort Associated findings Isolation technique/biomarker
Chen et al. [128] 2024 124 HCC PD-L1>1 or CTC-NLR>0 circulating tumor cell sorter, CytoNanoChip
 : independent predictor for OS↓ PD-L1+ CTC, CTC-NLR
Zhao et al. [129] 2023 270 HCC Independent risk for recurrence IF staining, RNA ISH, WGA
 : PCP (HR 25.7; 95% CI 5.2–106.6) CAPI, CK, Ki67
 : CTC cluster (HR 9.9; 95% CI 2.5–38.6)
Yang et al. [145] 2023 105 HCC M-CTC >0 per 5 mL CanPatrol
 : AFP ≥400 ng/mL, tumor size≥ 5 cm
 : poorly differentiation, multiplicity
 : advanced stage, microvascular invasion
 : portal vein tumor thrombosis
Lee et al. [30] 2022 Validation cohort 40 Independent predictor of survival NanoVelcro CTC assay
HCC  : HR 5.7; 95% CI 1.5–21.3; P=0.009 mRNA Risk Score panel
Chen et al. [130] 2022 Retrospective analysis CTC-WBC >0 per 5 mL CanPatrol, filtration & multiple mRNA ISH
136 HCC  : distant metastasis↑, recurrence↑, RFS↓
Kelley et al. [131] 2015 20 HCC CTC >0 per 7.5 mL CellSearch
10 NMLD  : AFP ≥400 ng/mL, vascular invasion CTC counts
Liu et al. [132] 2013 60 HCC CD45-ICAM-1+ qRT-PCR
 : DFS↓, OS↓ ICAM-1
Schulze et al. [133] 2013 59 HCC Presence of CTC CellSearch
 : OS↓, advanced BCLC stage CTC counts
 : microscopic vascular invasion
 : AFP ≥400 ng/mL

AFP, alpha-fetoprotein; BCLC, Barcelona Clinic Liver Cancer; CAPI, cytopathological immunofluorescence; CK, cytokeratin; CTC, circulating tumor cells; DFS, disease-free survival; HCC, hepatocellular carcinoma; HR, hazard ratio; ICAM-1, intercellular adhesion molecule 1; IF, immunofluorescence; ISH, in situ hybridization; mRNA, messenger RNA; NLR, neutrophil-to-lymphocyte ratio; NMLD, non-malignant liver disease; OS, overall survival; PCP, proliferative CTC percentage; PD-L1, programmed death-ligand 1; qRT-PCR, quantitative reverse transcription polymerase chain reaction; RFS, recurrence-free survival; WGA, whole genome amplification.

Table 6.
CfDNA for prognostication of HCC
Author Year Cohort Associated finding Isolation technique/biomarker
Guo et al. [150] 2024 293 HCC ctDNA methylation+ qMSP assay
96 CHB/LC, 23 BHL, 147 HC  : post-operative cumulative recurrence (HR 3.33; 95% CI 1.87–5.92) HCC specific DMR
Sogbe et al. [134] 2024 73 HCC Detectable ctDNA ULP-WGS
 : mPFS↓ (4.2 vs. 8.7 months)
Zhao et al. [135] 2022 80 HCC CTCs & ctDNA from PPWES NGS/PPWES, UPTS
 : RFS↓ (HR 11.88, 95% CI 4.06–34.75)
Kim et al. [136] 2020 107 HCC No SNV is associated with OS. ddPCR/
High-ctDNA group has lower OS. MLH1, PTEN, STK11, CTNNB1
Oversoe et al. [137] 2020 95 HCC No TERT in non-HCC ddPCR/
45 LC mortality↑ (HR 2.16; 95% CI 1.20–3.88) TERT
Li et al. [138] 2018 155 HCC Independent prognostic predictor of MSP
 : OS (P<0.01) IGFBP7 promoter methylation
 : early tumor recurrence (P=0.008)
Xu et al. [48] 2017 369 HCC (validation set) Panel score: independent risk for survival↓ (HR 1.54; 95% CI 1.25–1.92) Targeted bisulfite sequencing
8 methylation markers
Ji et al. [139] 2014 72 HCC (TNM I-IV) MT1M & MT1G promoter methylation MSP/
37 LC  : vascular invasion↑ or metastasis↑ MT1M, MT1G
Piciocchi et al. [87] 2013 66 HCC cfDNA level>2 ng/uL qRT-PCR/hTERT
35 LC/ 41 CH (HCV)  : shorter survival (37 vs. 24 months)

BHL, benign hepatic lesions; cfDNA, cell-free DNA; CDKN1A, cyclin dependent kinase inhibitor 1A; CHB, chronic hepatitis B; CI, confidence interval; circRNA, circular RNA; ctDNA, circulating tumor DNA; CTCs, circulating tumor cells; CTNNB, catenin beta; ddPCR, digital-droplet polymerase chain reaction; DMR, differentially methylated region; ERK, extracellular signal-regulated kinase; HC, healthy control; HCC, hepatocellular carcinoma; hnRNPH1, heterogeneous nuclear ribonucleoprotein H1; HR, hazard ratio; IGFBP7, insulinlike growth factor binding protein 7; LC, liver cirrhosis; lncRNA, long non-coding RNA; MARK, mitogen-activated protein kinase; miRNA, microRNA; MLH1, mutL homolog 1; mPFS, median progression-free survival; MSP, methylation-specific polymerase chain reaction; MT1G, metallothionein 1G; MT1M, metallothionein 1M; NGS, next-generation sequencing; OS, overall survival; PPWES, personalized panel targeting mutations from whole-exome sequencing; PTEN, phosphatase and tensin homolog; qMSP, quantitative methylationspecific polymerase chain reaction; qRT-PCR, quantitative real-time polymerase chain reaction; RFS, recurrence-free survival; RT-PCR, reverse transcription polymerase chain reaction; SNV, single nucleotide variant; STK11, serine/threonine kinase 11; TERT, telomerase reverse transcriptase; TIMP2, tissue inhibitor of metalloproteinases 2; TP53INP1, tumor protein 53-induced nuclear protein 1; TTR, time to recurrence; ULP-WGS, ultra-low-pass whole-genome sequencing; UPTS, universal personalized targeted panel.

Table 7.
EVs for prognostication of HCC
Author Year Cohort Associated finding Cargo Cargo level Isolation technique target gene or RNA
Sun et al. [140] 2021 HCC 168 High S100A4: independent risk factor for Worse OS, RFS protein RT-PCR
S100A4
Lee et al. [159] 2019 79 HCC Independent risk factor for shorter OS and faster progress lncRNA-ATB RT-PCR
miR-21
Wang et al. [160] 2019 HCC 71 High circPTGR1 circPTGR1 qRT-PCR
NC 41  : lower survival rate miR449a/MET
Xue et al. [141] 2018 85 HCC Cumulative survival: lower in high miR-93 (P=0.046) miR-93 qRT-PCR
TP53INP1, TIMP2, CDKN1A
Shi et al. [157] 2018 126 HCC 3 yr survival: HR 3.52 (95% CI 1.37–6.02) miR-638 qRT-PCR
21 HC 5 yr survival: HR 2.80 (95% CI 1.24–4.31) miR-638
Xu et al. [142] 2018 60 HCC High level of 2 lncRNA lncRNA RT-PCR
85 LC/ CHB 96  : associated with worse OS (log-rank test) ENSG00000258332.1
60 HC LINC000635
Xu et al. [123] 2018 88 HCC High mRNA-hnRNPH1 mRNA RT-PCR
67 LC, CHB 68  : worse OS mRNA-hnRNPH1
68 HC
Qu et al. [154] 2017 30 HCC High miR-665 miRNA-665 qRT-PCR/
10 NC  : OS↓ MARK/ERK
Liu et al. [158] 2017 HCC 128 Low miR-125 on multivariate analysis miR-125 qRT-PCR
TTR: HR 0.14 (95% CI 0.07–0.29) miR-125
OS: HR 0.36 (95% CI 0.18–0.74)

CDKN1A, cyclin dependent kinase inhibitor 1A; CHB, chronic hepatitis B; CI, confidence interval; circRNA, circular RNA; ERK, extracellular signal-regulated kinase; HC, healthy control; HCC, hepatocellular carcinoma; hnRNPH1, heterogeneous nuclear ribonucleoprotein H1; LC, liver cirrhosis; lncRNA, long non-coding RNA; MARK, mitogen-activated protein kinase; miRNA, microRNA; NC, normal control; OS, overall survival; qRT-PCR, quantitative real-time polymerase chain reaction; RFS, recurrence-free survival; RT-PCR, reverse transcription polymerase chain reaction; TIMP2, tissue inhibitor of metalloproteinases 2; TP53INP1, tumor protein 53-induced nuclear protein 1; TTR, time to recurrence.

Table 8.
Ongoing clinical trials for HCC
Test category Trial identifier Estimated enrollment Subject Study type Primary outcome measures Status Ref.
cfDNA NCT05064553 2,500 Increased risk for HCC Observational Show non-inferior sensitivity compared to ultrasound Recruiting [173]
cfDNA NCT03694600 1,900 LC Observational Compare sensitivity and specificity to ultrasound Active, not recruiting [172]
cfDNA NCT04111029 30 Unresectable HCC Observational Comparison of exome sequencing of tumor tissue and cfDNA after non-curative treatment Recruiting [177]
cfDNA NCT06134973 1,000 Hepatic nodules in CLD patients Observational Establishment of risk assessment model Recruiting [178]
ctDNA NCT04484636 400 Histologic confirmed HCC Interventional Distribution of mutations in solid tumor including HCC Completed [179]
exo-miRNA NCT06342414 400 Confirmed HCC or ICC Observational Ability to differentiate HCC and ICC Recruiting [180]
CTCs NCT04800497 70 HCC (BCLC 0-B) Observational Evaluate the association between variation in CTCs and prognosis Recruiting [181]
cfDNA mutation profile NCT04965454 80 Unresectable HCC Interventional Predict a lack of objective response after 16 weeks of ICI Recruiting [182]
Mutation in ctDNA NCT06028724 782 Histologically proven solid tumor including HCC Observational Real world prevalence of clinical useful mutations in solid tumors Recruiting [183]

BCLC, Barcelona clinic liver cancer; cfDNA, cell-free DNA; ctDNA, circulating tumor DNA; CLD, chronic liver disease; CTC, circulating cell tumor; HCC, hepatocellular carcinoma; ICI, Immune Checkpoint inhibitors; ICC, Intrahepatic cholangiocarcinoma; LC, liver cirrhosis; NCT, national clinical trial.

Abbreviations

ACD-A
acid citrate dextrose solution A
AFP
alpha-fetoprotein
AFP-L3
AFP lectin fraction
AHSG
alpha 2 - HS glycoprotein
AI
artificial intelligence
ALB
albumin
APOH
apolipoprotein
ASGPR
asialoglycoprotein receptor
AUROC
area under the receiver operating characteristic
BBRB
biospecimen research branch
BCLC
barcelona clinic liver cancer
BCT
blood collection tube
cfDNA
cell free DNA
cfMethyl-Seq
cell-free DNA methylome sequencing
CHALM
cell heterogeneity-adjusted cLonal methylation
circUHRF1
circular ubiquitin-like with PHD and ring finger domain 1 RNA
CK
creatinine kinase
CNVs
copy number alterations
cp-score
combined prognostic score
CT
computed tomography
CTC
circulating tumor cell
ctDNA
circulating tumor DNA
DAPI
4’
DCP
des-y-carboxyprothrombin
ddPCR
droplet digital PCR
DMRs
differentially methylated regions
EDTA
ethylenediaminetetraacetic acid
EEMT
epithelial to mesenchymal transition
EpCAM
epithelial cell adhesion molecule
EVs
extracellular vesicles
FABP1
fatty acid binding protein 1
FDA
food and drug administration
FGB
fibrinogen beta chain
FGG
fibrinogen gamma chain
GALAD
gender
GNB4
guanine nucleotide-binding protein subunit beta-4
GPC3
glypican 3
HCC
hepatocellular carcinoma
HCV
hepatitis C virus
ICI
immune checkpoint inhibitors
ISEV
international society for extracellular vesicles
LDHC
lactate dehydrogenase C
lncRNAs
long noncoding RNAs
MCP
mutation capsule plus
M-CTCs
mesenchymal phenotype of CTCs
MIBlood-EV
minimal information for blood extracellular vesicle research
miRNA
microRNA
MRD
minimal residual disease
mRNA
messenger RNA
MRI
magnetic resonance imaging
Mt-HBT
multitarget HCC blood test
NCI
national cancer institute
NGS
next-generation sequencing
NK
natural killer
NSCLC
non-small cell lung cancer
OS
overall survival
PCR
polymerase chain reaction
PFS
progression-free
qRT-PCR
quantitative real-time PCR
RBP4
retinol binding protein 4
RFS
recurrence-free Survival
SNVs
single nucleotide variations
TACE
transarterial chemoembolization
TF
transferrin
TTP
time to progression
WBCs
white blood cells
WGS
whole genome sequencing
5hmC
5-hydroxymethylcytosine

REFERENCES

1. Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, et al. Annual report to the nation on the status of cancer, 1975-2014, featuring survival. J Natl Cancer Inst 2017;109:djx030.
crossref pmid pmc
2. Kanwal F, Singal AG. Surveillance for hepatocellular carcinoma: current best practice and future direction. Gastroenterology 2019;157:54-64.
crossref pmid pmc
3. Yeo YH, Lee YT, Tseng HR, Zhu Y, You S, Agopian VG, et al. Alpha-fetoprotein: past, present, and future. Hepatol Commun 2024;8:e0422.
crossref pmid pmc
4. Lee YT, Fujiwara N, Yang JD, Hoshida Y. Risk stratification and early detection biomarkers for precision HCC screening. Hepatology 2023;78:319-362.
crossref pmid pmc pdf
5. Poulet G, Massias J, Taly V. Liquid biopsy: general concepts. Acta Cytol 2019;63:449-455.
crossref pmid pdf
6. Kleiner DE, Stetler-Stevenson WG. Matrix metalloproteinases and metastasis. Cancer Chemother Pharmacol 1999;43 Suppl:S42-S51.
crossref pmid pdf
7. Miller MC, Doyle GV, Terstappen LW. Significance of Circulating tumor cells detected by the cellsearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol 2010;2010:617421.
crossref pmid pmc pdf
8. Edd JF, Mishra A, Smith KC, Kapur R, Maheswaran S, Haber DA, et al. Isolation of circulating tumor cells. iScience 2022;25:104696.
crossref pmid pmc
9. Grölz D, Hauch S, Schlumpberger M, Guenther K, Voss T, Sprenger-Haussels M, et al. Liquid biopsy preservation solutions for standardized pre-analytical workflows-venous whole blood and plasma. Curr Pathobiol Rep 2018;6:275-286.
crossref pmid pmc pdf
10. Ahn JC, Teng PC, Chen PJ, Posadas E, Tseng HR, Lu SC, et al. Detection of circulating tumor cells and their implications as a biomarker for diagnosis, prognostication, and therapeutic monitoring in hepatocellular carcinoma. Hepatology 2021;73:422-436.
crossref pmid pmc pdf
11. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 2004;351:781-791.
crossref pmid
12. Negin BP, Cohen SJ. Circulating tumor cells in colorectal cancer: past, present, and future challenges. Curr Treat Options Oncol 2010;11:1-13.
crossref pmid pdf
13. Millner LM, Linder MW, Valdes R Jr. Circulating tumor cells: a review of present methods and the need to identify heterogeneous phenotypes. Ann Clin Lab Sci 2013;43:295-304.
pmid pmc
14. Lawrence R, Watters M, Davies CR, Pantel K, Lu YJ. Circulating tumour cells for early detection of clinically relevant cancer. Nat Rev Clin Oncol 2023;20:487-500.
crossref pmid pmc pdf
15. Court CM, Hou S, Winograd P, Segel NH, Li QW, Zhu Y, et al. A novel multimarker assay for the phenotypic profiling of circulating tumor cells in hepatocellular carcinoma. Liver Transpl 2018;24:946-960.
crossref pmid pmc pdf
16. Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007;450:1235-1239.
crossref pmid pmc pdf
17. Ozkumur E, Shah AM, Ciciliano JC, Emmink BL, Miyamoto DT, Brachtel E, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci Transl Med 2013;5:179ra47.
crossref pmid pmc
18. Fachin F, Spuhler P, Martel-Foley JM, Edd JF, Barber TA, Walsh J, et al. Monolithic chip for high-throughput blood cell depletion to sort rare circulating tumor cells. Sci Rep 2017;7:10936.
crossref pmid pmc pdf
19. Kalinich M, Bhan I, Kwan TT, Miyamoto DT, Javaid S, LiCausi JA, et al. An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma. Proc Natl Acad Sci U S A 2017;114:1123-1128.
crossref pmid pmc
20. Jan YJ, Chen JF, Zhu Y, Lu YT, Chen SH, Chung H, et al. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018;125:78-93.
crossref pmid pmc
21. Winograd P, Hou S, Court CM, Lee YT, Chen PJ, Zhu Y, et al. Hepatocellular carcinoma-circulating tumor cells expressing PD-L1 are prognostic and potentially associated with response to checkpoint inhibitors. Hepatol Commun 2020;4:1527-1540.
crossref pmid pmc pdf
22. Sun N, Lee YT, Kim M, Wang JJ, Zhang C, Teng PC, et al. Covalent chemistry-mediated multimarker purification of circulating tumor cells enables noninvasive detection of molecular signatures of hepatocellular carcinoma. Adv Mater Technol 2021;6:2001056.
crossref pmid pmc pdf
23. Jan YJ, Yoon J, Chen JF, Teng PC, Yao N, Cheng S, et al. A circulating tumor cell-RNA assay for assessment of androgen receptor signaling inhibitor sensitivity in metastatic castration-resistant prostate cancer. Theranostics 2019;9:2812-2826.
crossref pmid pmc
24. Ring A, Nguyen-Sträuli BD, Wicki A, Aceto N. Biology, vulnerabilities and clinical applications of circulating tumour cells. Nat Rev Cancer 2023;23:95-111.
crossref pmid pmc pdf
25. Mutlu BR, Smith KC, Edd JF, Nadar P, Dlamini M, Kapur R, et al. Non-equilibrium inertial separation array for high-throughput, large-volume blood fractionation. Sci Rep 2017;7:9915.
crossref pmid pmc pdf
26. Vona G, Estepa L, Béroud C, Damotte D, Capron F, Nalpas B, et al. Impact of cytomorphological detection of circulating tumor cells in patients with liver cancer. Hepatology 2004;39:792-797.
crossref pmid
27. Miller MC, Robinson PS, Wagner C, O’Shannessy DJ. The Parsortix™ cell separation system-a versatile liquid biopsy platform. Cytometry A 2018;93:1234-1239.
crossref pmid pmc pdf
28. Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, et al. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021;6:404.
crossref pmid pmc pdf
29. Visal TH, den Hollander P, Cristofanilli M, Mani SA. Circulating tumour cells in the -omics era: how far are we from achieving the ‘singularity’? Br J Cancer 2022;127:173-184.
crossref pmid pmc pdf
30. Lee YT, Sun N, Kim M, Wang JJ, Tran BV, Zhang RY, et al. Circulating tumor cell-based messenger RNA Scoring system for prognostication of hepatocellular carcinoma: translating tissue-based messenger RNA profiling into a noninvasive setting. Liver Transpl 2022;28:200-214.
crossref pmid pmc pdf
31. Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res 2007;13:920-928.
crossref pmid pdf
32. Luk AWS, Ma Y, Ding PN, Young FP, Chua W, Balakrishnar B, et al. CTC-mRNA (AR-V7) analysis from blood samples-impact of blood collection tube and storage time. Int J Mol Sci 2017;18:1047.
crossref pmid pmc
33. Zavridou M, Mastoraki S, Strati A, Tzanikou E, Chimonidou M, Lianidou E. Evaluation of preanalytical conditions and implementation of quality control steps for reliable gene expression and DNA methylation analyses in liquid biopsies. Clin Chem 2018;64:1522-1533.
crossref pmid pdf
34. Wong KHK, Tessier SN, Miyamoto DT, Miller KL, Bookstaver LD, Carey TR, et al. Whole blood stabilization for the microfluidic isolation and molecular characterization of circulating tumor cells. Nat Commun 2017;8:1733.
crossref pmid pmc pdf
35. Apostolou P, Ntanovasilis DA, Papasotiriou I. Evaluation of a simple method for storage of blood samples that enables isolation of circulating tumor cells 96 h after sample collection. J Biol Res (Thessalon) 2017;24:11.
crossref pmid pmc pdf
36. Deng Z, Wu S, Wang Y, Shi D. Circulating tumor cell isolation for cancer diagnosis and prognosis. EBioMedicine 2022;83:104237.
crossref pmid pmc
37. Guo Z, Lin X, Hui Y, Wang J, Zhang Q, Kong F. Circulating tumor cell identification based on deep learning. Front Oncol 2022;12:843879.
crossref pmid pmc
38. Sun YF, Wu L, Liu SP, Jiang MM, Hu B, Zhou KQ, et al. Dissecting spatial heterogeneity and the immune-evasion mechanism of CTCs by single-cell RNA-seq in hepatocellular carcinoma. Nat Commun 2021;12:4091.
crossref pmid pmc pdf
39. Sun YF, Guo W, Xu Y, Shi YH, Gong ZJ, Ji Y, et al. Circulating tumor cells from different vascular sites exhibit spatial heterogeneity in epithelial and mesenchymal composition and distinct clinical significance in hepatocellular carcinoma. Clin Cancer Res 2018;24:547-559.
crossref pmid pdf
40. Gao Q, Zeng Q, Wang Z, Li C, Xu Y, Cui P, et al. Circulating cell-free DNA for cancer early detection. Innovation (Camb) 2022;3:100259.
crossref pmid pmc
41. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24.
pmid pmc
42. Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther 2019;20:1057-1067.
crossref pmid pmc pdf
43. Lyu X, Tsui YM, Ho DW, Ng IO. Liquid biopsy using cell-free or circulating tumor DNA in the management of hepatocellular carcinoma. Cell Mol Gastroenterol Hepatol 2022;13:1611-1624.
crossref pmid pmc
44. Liao W, Yang H, Xu H, Wang Y, Ge P, Ren J, et al. Noninvasive detection of tumor-associated mutations from circulating cell-free DNA in hepatocellular carcinoma patients by targeted deep sequencing. Oncotarget 2016;7:40481-40490.
crossref pmid pmc
45. Lee HW, Kim E, Cho KJ, Park HJ, Seo J, Lee H, et al. Applications of molecular barcode sequencing for the detection of low-frequency variants in circulating tumour DNA from hepatocellular carcinoma. Liver Int 2022;42:2317-2326.
pmid
46. Villanueva A, Portela A, Sayols S, Battiston C, Hoshida Y, Méndez-González J, et al. DNA methylation-based prognosis and epidrivers in hepatocellular carcinoma. Hepatology 2015;61:1945-1956.
crossref pmid pdf
47. Hao X, Luo H, Krawczyk M, Wei W, Wang W, Wang J, et al. DNA methylation markers for diagnosis and prognosis of common cancers. Proc Natl Acad Sci U S A 2017;114:7414-7419.
crossref pmid pmc
48. Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater 2017;16:1155-1161.
crossref pmid pdf
49. Stackpole ML, Zeng W, Li S, Liu CC, Zhou Y, He S, et al. Cost-effective methylome sequencing of cell-free DNA for accurately detecting and locating cancer. Nat Commun 2022;13:5566.
crossref pmid pmc pdf
50. Jiang P, Sun K, Peng W, Cheng SH, Ni M, Yeung PC, et al. Plasma DNA end-motif profiling as a fragmentomic marker in cancer, pregnancy, and transplantation. Cancer Discov 2020;10:664-673.
crossref pmid pdf
51. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A 2005;102:16368-16373.
crossref pmid pmc
52. Lo YMD, Chan KCA, Sun H, Chen EZ, Jiang P, Lun FMF, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med 2010;2:61ra91.
crossref pmid
53. Jin C, Liu X, Zheng W, Su L, Liu Y, Guo X, et al. Characterization of fragment sizes, copy number aberrations and 4-mer end motifs in cell-free DNA of hepatocellular carcinoma for enhanced liquid biopsy-based cancer detection. Mol Oncol 2021;15:2377-2389.
crossref pmid pmc pdf
54. Yang JD, Liu MC, Kisiel JB. Circulating tumor DNA and hepatocellular carcinoma. Semin Liver Dis 2019;39:452-462.
crossref pmid pmc
55. Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024;80:515-530.
crossref pmid pmc
56. Kisiel JB, Dukek BA, Kanipakam VSR, Ghoz HM, Yab TC, Berger CK, et al. Hepatocellular carcinoma detection by plasma methylated DNA: discovery, phase I pilot, and phase II clinical validation. Hepatology 2019;69:1180-1192.
crossref pmid pmc pdf
57. Wang P, Song Q, Ren J, Zhang W, Wang Y, Zhou L, et al. Simultaneous analysis of mutations and methylations in circulating cell-free DNA for hepatocellular carcinoma detection. Sci Transl Med 2022;14:eabp8704.
crossref pmid
58. Lin N, Lin Y, Xu J, Liu D, Li D, Meng H, et al. A multi-analyte cell-free DNA-based blood test for early detection of hepatocellular carcinoma. Hepatol Commun 2022;6:1753-1763.
crossref pmid pmc pdf
59. Xu J, Shi J, Cui X, Cui Y, Li JJ, Goel A, et al. Cellular heterogeneity-adjusted cLonal Methylation (CHALM) improves prediction of gene expression. Nat Commun 2021;12:400.
crossref pmid pmc pdf
60. Greytak SR, Engel KB, Parpart-Li S, Murtaza M, Bronkhorst AJ, Pertile MD, et al. Harmonizing cell-free DNA collection and processing practices through evidence-based guidance. Clin Cancer Res 2020;26:3104-3109.
crossref pmid pmc pdf
61. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7:1535750.
pmid pmc
62. Welsh JA, Goberdhan DCI, O’Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles 2024;13:e12404.
pmid pmc
63. Cheng L, Zhang L, Wang X, Wang Y, Yu J, Li M, et al. Extracellular vesicles in the HCC microenvironment: implications for therapy and biomarkers. Pharmacol Res 2024;209:107419.
crossref pmid
64. Wang H, Lu Z, Zhao X. Tumorigenesis, diagnosis, and therapeutic potential of exosomes in liver cancer. J Hematol Oncol 2019;12:133.
crossref pmid pmc pdf
65. Rios-Colon L, Arthur E, Niture S, Qi Q, Moore JT, Kumar D. The role of exosomes in the crosstalk between adipocytes and liver cancer cells. Cells 2020;9:1988.
crossref pmid pmc
66. Tey SK, Wong SWK, Chan JYT, Mao X, Ng TH, Yeung CLS, et al. Patient pIgR-enriched extracellular vesicles drive cancer stemness, tumorigenesis and metastasis in hepatocellular carcinoma. J Hepatol 2022;76:883-895.
crossref pmid
67. Tian L, Lu J, Ng IO. Extracellular vesicles and cancer stemness in hepatocellular carcinoma - is there a link? Front Immunol 2024;15:1368898.
crossref
68. Clos-Sansalvador M, Monguió-Tortajada M, Roura S, Franquesa M, Borràs FE. Commonly used methods for extracellular vesicles’ enrichment: Implications in downstream analyses and use. Eur J Cell Biol 2022;101:151227.
crossref
69. Karttunen J, Heiskanen M, Navarro-Ferrandis V, Das Gupta S, Lipponen A, Puhakka N, et al. Precipitation-based extracellular vesicle isolation from rat plasma co-precipitate vesicle-free microRNAs. J Extracell Vesicles 2019;8:1555410.
crossref pmid pmc
70. Sun N, Lee YT, Zhang RY, Kao R, Teng PC, Yang Y, et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat Commun 2020;11:4489.
crossref pdf
71. Sun N, Zhang C, Lee YT, Tran BV, Wang J, Kim H, et al. HCC EV ECG score: an extracellular vesicle-based protein assay for detection of early-stage hepatocellular carcinoma. Hepatology 2023;77:774-788.
pmid
72. Julich-Haertel H, Urban SK, Krawczyk M, Willms A, Jankowski K, Patkowski W, et al. Cancer-associated circulating large extracellular vesicles in cholangiocarcinoma and hepatocellular carcinoma. J Hepatol 2017;67:282-292.
crossref pmid
73. Karver MR, Weissleder R, Hilderbrand SA. Synthesis and evaluation of a series of 1,2,4,5-tetrazines for bioorthogonal conjugation. Bioconjug Chem 2011;22:2263-2270.
crossref pmid pmc
74. Lee YT, Tran BV, Wang JJ, Liang IY, You S, Zhu Y, et al. The role of extracellular vesicles in disease progression and detection of hepatocellular carcinoma. Cancers (Basel) 2021;13:3076.
crossref pmid pmc
75. Wang JJ, Sun N, Lee YT, Kim M, Vagner T, Rohena-Rivera K, et al. Prostate cancer extracellular vesicle digital scoring assay - a rapid noninvasive approach for quantification of disease-relevant mRNAs. Nano Today 2023;48:101746.
crossref pmid pmc
76. Dhondt B, Pinheiro C, Geeurickx E, Tulkens J, Vergauwen G, Van Der Pol E, et al. Benchmarking blood collection tubes and processing intervals for extracellular vesicle performance metrics. J Extracell Vesicles 2023;12:e12315.
crossref pmid pmc
77. Erdbrügger U, Blijdorp CJ, Bijnsdorp IV, Borràs FE, Burger D, Bussolati B, et al. Urinary extracellular vesicles: a position paper by the Urine Task Force of the International Society for Extracellular Vesicles. J Extracell Vesicles 2021;10:e12093.
pmid pmc
78. van Royen ME, Soekmadji C, Grange C, Webber JP, Tertel T, Droste M, et al. The quick reference card “Storage of urinary EVs” - a practical guideline tool for research and clinical laboratories. J Extracell Vesicles 2023;12:e12286.
pmid
79. Bahnassy AA, Zekri AR, El-Bastawisy A, Fawzy A, Shetta M, Hussein N, et al. Circulating tumor and cancer stem cells in hepatitis C virus-associated liver disease. World J Gastroenterol 2014;20:18240-18248.
crossref pmid pmc
80. Armakolas A, Dimopoulou V, Nezos A, Stamatakis G, Samiotaki M, Panayotou G, et al. Cellular, molecular and proteomic characteristics of early hepatocellular carcinoma. Curr Issues Mol Biol 2022;44:4714-4734.
crossref pmid pmc
81. Guo W, Sun YF, Shen MN, Ma XL, Wu J, Zhang CY, et al. Circulating tumor cells with stem-like phenotypes for diagnosis, prognosis, and therapeutic response evaluation in hepatocellular carcinoma. Clin Cancer Res 2018;24:2203-2213.
crossref pmid pdf
82. Bhan I, Mosesso K, Goyal L, Philipp J, Kalinich M, Franses JW, et al. Detection and analysis of circulating epithelial cells in liquid biopsies from patients with liver disease. Gastroenterology 2018;155:2016-2018.e11.
crossref pmid pmc
83. Chen L, Wu T, Fan R, Qian YS, Liu JF, Bai J, et al. Cell-free DNA testing for early hepatocellular carcinoma surveillance. EBioMedicine 2024;100:104962.
crossref pmid pmc
84. Cai J, Chen L, Zhang Z, Zhang X, Lu X, Liu W, et al. Genome-wide mapping of 5-hydroxymethylcytosines in circulating cell-free DNA as a non-invasive approach for early detection of hepatocellular carcinoma. Gut 2019;68:2195-2205.
crossref pmid pmc
85. Jiang P, Chan CW, Chan KC, Cheng SH, Wong J, Wong VW, et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc Natl Acad Sci U S A 2015;112:E1317-E1325.
crossref pmid pmc
86. Huang G, Krocker JD, Kirk JL, Merwat SN, Ju H, Soloway RD, et al. Evaluation of INK4A promoter methylation using pyrosequencing and circulating cell-free DNA from patients with hepatocellular carcinoma. Clin Chem Lab Med 2014;52:899-909.
crossref pmid pmc
87. Piciocchi M, Cardin R, Vitale A, Vanin V, Giacomin A, Pozzan C, et al. Circulating free DNA in the progression of liver damage to hepatocellular carcinoma. Hepatol Int 2013;7:1050-1057.
crossref pmid pdf
88. Son JA, Weon JH, Baek GO, Ahn HR, Choi JY, Yoon MG, et al. Circulating small extracellular vesicle-derived splicing factor 3b subunit 4 as a non-invasive diagnostic biomarker of early hepatocellular carcinoma. J Exp Clin Cancer Res 2023;42:288.
crossref pmid pmc pdf
89. Kim SS, Baek GO, Son JA, Ahn HR, Yoon MK, Cho HJ, et al. Early detection of hepatocellular carcinoma via liquid biopsy: panel of small extracellular vesicle-derived long noncoding RNAs identified as markers. Mol Oncol 2021;15:2715-2731.
pmid pmc
90. von Felden J, Garcia-Lezana T, Dogra N, Gonzalez-Kozlova E, Ahsen ME, Craig A, et al. Unannotated small RNA clusters associated with circulating extracellular vesicles detect early stage liver cancer. Gut 2022;71:2069-2080.
crossref pmid pmc
91. Ghosh S, Bhowmik S, Majumdar S, Goswami A, Chakraborty J, Gupta S, et al. The exosome encapsulated microRNAs as circulating diagnostic marker for hepatocellular carcinoma with low alpha-fetoprotein. Int J Cancer 2020;147:2934-2947.
pmid
92. Huang X, Sun L, Wen S, Deng D, Wan F, He X, et al. RNA sequencing of plasma exosomes revealed novel functional long noncoding RNAs in hepatocellular carcinoma. Cancer Sci 2020;111:3338-3349.
crossref pmid pmc pdf
93. Sun XH, Wang YT, Li GF, Zhang N, Fan L. Serum-derived three-circRNA signature as a diagnostic biomarker for hepatocellular carcinoma. Cancer Cell Int 2020;20:226.
crossref pmid pmc pdf
94. Arbelaiz A, Azkargorta M, Krawczyk M, Santos-Laso A, Lapitz A, Perugorria MJ, et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2017;66:1125-1143.
crossref pmid pdf
95. Went PT, Lugli A, Meier S, Bundi M, Mirlacher M, Sauter G, et al. Frequent EpCam protein expression in human carcinomas. Hum Pathol 2004;35:122-128.
crossref pmid
96. Espejo-Cruz ML, González-Rubio S, Zamora-Olaya J, Amado-Torres V, Alejandre R, Sánchez-Frías M, et al. Circulating tumor cells in hepatocellular carcinoma: a comprehensive review and critical appraisal. Int J Mol Sci 2021;22:13073.
crossref pmid pmc
97. Hamaoka M, Kobayashi T, Tanaka Y, Mashima H, Ohdan H. Clinical significance of glypican-3-positive circulating tumor cells of hepatocellular carcinoma patients: a prospective study. PLoS One 2019;14:e0217586.
crossref pmid pmc
98. Li YM, Xu SC, Li J, Han KQ, Pi HF, Zheng L, et al. Epithelial-mesenchymal transition markers expressed in circulating tumor cells in hepatocellular carcinoma patients with different stages of disease. Cell Death Dis 2013;4:e831.
crossref pmid pmc pdf
99. Zhu L, Lin H, Wan S, Chen X, Wu L, Zhu Z, et al. Efficient isolation and phenotypic profiling of circulating hepatocellular carcinoma cells via a combinatorial-antibody-functionalized microfluidic synergetic-chip. Anal Chem 2020;92:15229-15235.
crossref pmid
100. Chen J, Cao SW, Cai Z, Zheng L, Wang Q. Epithelial-mesenchymal transition phenotypes of circulating tumor cells correlate with the clinical stages and cancer metastasis in hepatocellular carcinoma patients. Cancer Biomark 2017;20:487-498.
crossref pmid
101. Cheng Y, Luo L, Zhang J, Zhou M, Tang Y, He G, et al. Diagnostic value of different phenotype circulating tumor cells in hepatocellular carcinoma. J Gastrointest Surg 2019;23:2354-2361.
crossref pmid pdf
102. Liang W, Xu Z, Kong F, Huang X, Xiao Y, Zhou W, et al. Circulating tumour cell combined with DNA methylation for early detection of hepatocellular carcinoma. Front Genet 2022;13:1065693.
crossref pmid pmc
103. Cui K, Ou Y, Shen Y, Li S, Sun Z. Clinical value of circulating tumor cells for the diagnosis and prognosis of hepatocellular carcinoma (HCC): a systematic review and meta-analysis. Medicine (Baltimore) 2020;99:e22242.
pmid pmc
104. Iizuka N, Sakaida I, Moribe T, Fujita N, Miura T, Stark M, et al. Elevated levels of circulating cell-free DNA in the blood of patients with hepatitis C virus-associated hepatocellular carcinoma. Anticancer Res 2006;26:4713-4719.
pmid
105. Huang Z, Hua D, Hu Y, Cheng Z, Zhou X, Xie Q, et al. Quantitation of plasma circulating DNA using quantitative PCR for the detection of hepatocellular carcinoma. Pathol Oncol Res 2012;18:271-276.
crossref pmid pdf
106. Tokuhisa Y, Iizuka N, Sakaida I, Moribe T, Fujita N, Miura T, et al. Circulating cell-free DNA as a predictive marker for distant metastasis of hepatitis C virus-related hepatocellular carcinoma. Br J Cancer 2007;97:1399-1403.
crossref pmid pmc pdf
107. Yan L, Chen Y, Zhou J, Zhao H, Zhang H, Wang G. Diagnostic value of circulating cell-free DNA levels for hepatocellular carcinoma. Int J Infect Dis 2018;67:92-97.
crossref pmid
108. Coto-Llerena M, Benjak A, Gallon J, Meier MA, Boldanova T, Terracciano LM, et al. Circulating cell-free DNA captures the intratumor heterogeneity in multinodular hepatocellular carcinoma. JCO Precis Oncol 2022;6:e2100335.
crossref pmid pmc
109. Huang A, Zhang X, Zhou SL, Cao Y, Huang XW, Fan J, et al. Detecting circulating tumor DNA in hepatocellular carcinoma patients using droplet digital PCR is feasible and reflects intratumoral heterogeneity. J Cancer 2016;7:1907-1914.
crossref pmid pmc
110. Guo W, Lu J, Yan L, Sun D, Gong L, Shi W. Molecular alterations of circulating cell-free DNA in the pathological progression of hepatocellular carcinoma. J Oncol 2021;2021:3637436.
crossref pmid pmc pdf
111. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018;359:926-930.
crossref pmid pmc
112. Han LY, Fan YC, Mu NN, Gao S, Li F, Ji XF, et al. Aberrant DNA methylation of G-protein-coupled bile acid receptor Gpbar1 (TGR5) is a potential biomarker for hepatitis B Virus associated hepatocellular carcinoma. Int J Med Sci 2014;11:164-171.
crossref pmid pmc
113. Oussalah A, Rischer S, Bensenane M, Conroy G, Filhine-Tresarrieu P, Debard R, et al. Plasma mSEPT9: a novel circulating cell-free DNA-based epigenetic biomarker to diagnose hepatocellular carcinoma. EBioMedicine 2018;30:138-147.
crossref pmid pmc
114. Guo P, Zheng H, Li Y, Li Y, Xiao Y, Zheng J, et al. Hepatocellular carcinoma detection via targeted enzymatic methyl sequencing of plasma cell-free DNA. Clin Epigenetics 2023;15:2.
crossref pmid pmc pdf
115. Chalasani NP, Porter K, Bhattacharya A, Book AJ, Neis BM, Xiong KM, et al. Validation of a novel multitarget blood test shows high sensitivity to detect early stage hepatocellular carcinoma. Clin Gastroenterol Hepatol 2022;20:173-182.e7.
crossref pmid
116. Phan TH, Chi Nguyen VT, Thi Pham TT, Nguyen VC, Ho TD, Quynh Pham TM, et al. Circulating DNA methylation profile improves the accuracy of serum biomarkers for the detection of nonmetastatic hepatocellular carcinoma. Future Oncol 2022;18:4399-4413.
crossref pmid
117. Udomruk S, Orrapin S, Pruksakorn D, Chaiyawat P. Size distribution of cell-free DNA in oncology. Crit Rev Oncol Hematol 2021;166:103455.
crossref pmid
118. Foda ZH, Annapragada AV, Boyapati K, Bruhm DC, Vulpescu NA, Medina JE, et al. Detecting liver cancer using cell-free DNA fragmentomes. Cancer Discov 2023;13:616-631.
crossref pmid pmc pdf
119. Nguyen VC, Nguyen TH, Phan TH, Tran TT, Pham TTT, Ho TD, et al. Fragment length profiles of cancer mutations enhance detection of circulating tumor DNA in patients with early-stage hepatocellular carcinoma. BMC Cancer 2023;23:233.
crossref pmid pmc pdf
120. Chen L, Abou-Alfa GK, Zheng B, Liu JF, Bai J, Du LT, et al. Genome-scale profiling of circulating cell-free DNA signatures for early detection of hepatocellular carcinoma in cirrhotic patients. Cell Res 2021;31:589-592.
crossref pmid pmc pdf
121. Zhang Z, Chen P, Xie H, Cao P. Using circulating tumor DNA as a novel biomarker to screen and diagnose hepatocellular carcinoma: a systematic review and meta-analysis. Cancer Med 2020;9:1349-1364.
crossref pmid pmc pdf
122. Wang W, Li H, Zhou Y, Jie S. Peripheral blood microvesicles are potential biomarkers for hepatocellular carcinoma. Cancer Biomark 2013;13:351-357.
crossref pmid
123. 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.
crossref pmid
124. Wang Y, Zhang C, Zhang P, Guo G, Jiang T, Zhao X, et al. Serum exosomal microRNAs combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer Med 2018;7:1670-1679.
pmid pmc
125. Kim SS, Baek GO, Ahn HR, Sung S, Seo CW, Cho HJ, et al. Serum small extracellular vesicle-derived LINC00853 as a novel diagnostic marker for early hepatocellular carcinoma. Mol Oncol 2020;14:2646-2659.
crossref pmid pmc pdf
126. Lu Y, Duan Y, Xu Q, Zhang L, Chen W, Qu Z, et al. Circulating exosome-derived bona fide long non-coding RNAs predicting the occurrence and metastasis of hepatocellular carcinoma. J Cell Mol Med 2020;24:1311-1318.
crossref pmid pmc pdf
127. Cui Z, Li Y, Gao Y, Kong L, Lin Y, Chen Y. Cancer-testis antigen lactate dehydrogenase C4 in hepatocellular carcinoma: a promising biomarker for early diagnosis, efficacy evaluation and prognosis prediction. Aging (Albany NY) 2020;12:19455-19467.
crossref pmid pmc
128. Chen JL, Guo L, Wu ZY, He K, Li H, Yang C, et al. Prognostic value of circulating tumor cells combined with neutrophil-lymphocyte ratio in patients with hepatocellular carcinoma. World J Gastrointest Oncol 2024;16:372-385.
crossref pmid pmc
129. Zhao L, Song J, Sun Y, Ju Q, Mu H, Dong X, et al. Tumor-derived proliferative CTCs and CTC clusters predict aggressiveness and early recurrence in hepatocellular carcinoma patients. Cancer Med 2023;12:13912-13927.
pmid pmc
130. Chen J, Luo Y, Xi X, Li H, Li S, Zheng L, et al. Circulating tumor cell associated white blood cell cluster as a biomarker for metastasis and recurrence in hepatocellular carcinoma. Front Oncol 2022;12:931140.
crossref pmid pmc
131. Kelley RK, Magbanua MJ, Butler TM, Collisson EA, Hwang J, Sidiropoulos N, et al. Circulating tumor cells in hepatocellular carcinoma: a pilot study of detection, enumeration, and next-generation sequencing in cases and controls. BMC Cancer 2015;15:206.
crossref pmid pmc pdf
132. Liu S, Li N, Yu X, Xiao X, Cheng K, Hu J, et al. Expression of intercellular adhesion molecule 1 by hepatocellular carcinoma stem cells and circulating tumor cells. Gastroenterology 2013;144:1031-1041 e10.
crossref pmid
133. Schulze K, Gasch C, Staufer K, Nashan B, Lohse AW, Pantel K, et al. Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma. Int J Cancer 2013;133:2165-2171.
crossref pmid
134. Sogbe M, Bilbao I, Marchese FP, Zazpe J, De Vito A, Pozuelo M, et al. Prognostic value of ultra-low-pass whole-genome sequencing of circulating tumor DNA in hepatocellular carcinoma under systemic treatment. Clin Mol Hepatol 2024;30:177-190.
crossref pmid pmc pdf
135. Zhao L, Jiang L, Liu Y, Wang X, Song J, Sun Y, et al. Integrated analysis of circulating tumour cells and circulating tumour DNA to detect minimal residual disease in hepatocellular carcinoma. Clin Transl Med 2022;12:e793.
crossref pmid pmc pdf
136. Kim SS, Eun JW, Choi JH, Woo HG, Cho HJ, Ahn HR, et al. MLH1 single-nucleotide variant in circulating tumor DNA predicts overall survival of patients with hepatocellular carcinoma. Sci Rep 2020;10:17862.
crossref pmid pmc pdf
137. Oversoe SK, Clement MS, Pedersen MH, Weber B, Aagaard NK, Villadsen GE, et al. TERT promoter mutated circulating tumor DNA as a biomarker for prognosis in hepatocellular carcinoma. Scand J Gastroenterol 2020;55:1433-1440.
crossref pmid
138. Li F, Qiao CY, Gao S, Fan YC, Chen LY, Wang K. Circulating cell-free DNA of methylated insulin-like growth factor-binding protein 7 predicts a poor prognosis in hepatitis B virus-associated hepatocellular carcinoma after hepatectomy. Free Radic Res 2018;52:455-464.
crossref pmid
139. Ji XF, Fan YC, Gao S, Yang Y, Zhang JJ, Wang K. MT1M and MT1G promoter methylation as biomarkers for hepatocellular carcinoma. World J Gastroenterol 2014;20:4723-4729.
crossref pmid pmc
140. Sun H, Wang C, Hu B, Gao X, Zou T, Luo Q, et al. Exosomal S100A4 derived from highly metastatic hepatocellular carcinoma cells promotes metastasis by activating STAT3. Signal Transduct Target Ther 2021;6:187.
crossref pmid pmc pdf
141. 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.
crossref pmid
142. 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 Biomarkers Prev 31 Oct 2024.
crossref pmid pdf
143. Wang S, Zhang C, Wang G, Cheng B, Wang Y, Chen F, et al. Aptamer-mediated transparent-biocompatible nanostructured surfaces for hepotocellular circulating tumor cells enrichment. Theranostics 2016;6:1877-1886.
crossref pmid pmc
144. Rau KM, Liu CT, Hsiao YC, Hsiao KY, Wang TM, Hung WS, et al. Sequential circulating tumor cell counts in patients with locally advanced or metastatic hepatocellular carcinoma: monitoring the treatment response. J Clin Med 2020;9:188.
crossref pmid pmc
145. Yang X, Ni H, Lu Z, Zhang J, Zhang Q, Ning S, et al. Mesenchymal circulating tumor cells and Ki67: their mutual correlation and prognostic implications in hepatocellular carcinoma. BMC Cancer 2023;23:10.
crossref pmid pmc pdf
146. Qi LN, Xiang BD, Wu FX, Ye JZ, Zhong JH, Wang YY, et al. Circulating tumor cells undergoing EMT provide a metric for diagnosis and prognosis of patients with hepatocellular carcinoma. Cancer Res 2018;78:4731-4744.
crossref pmid pdf
147. Cai Z, Chen G, Zeng Y, Dong X, Li Z, Huang Y, et al. Comprehensive liquid profiling of circulating tumor DNA and protein biomarkers in long-term follow-up patients with hepatocellular carcinoma. Clin Cancer Res 2019;25:5284-5294.
crossref pmid pdf
148. An Y, Guan Y, Xu Y, Han Y, Wu C, Bao C, et al. The diagnostic and prognostic usage of circulating tumor DNA in operable hepatocellular carcinoma. Am J Transl Res 2019;11:6462-6474.
pmid pmc
149. Oh CR, Kong SY, Im HS, Kim HJ, Kim MK, Yoon KA, et al. Genome-wide copy number alteration and VEGFA amplification of circulating cell-free DNA as a biomarker in advanced hepatocellular carcinoma patients treated with Sorafenib. BMC Cancer 2019;19:292.
crossref pmid pmc pdf
150. Guo DZ, Huang A, Wang YC, Zhou S, Wang H, Xing XL, et al. Early detection and prognosis evaluation for hepatocellular carcinoma by circulating tumour DNA methylation: a multicentre cohort study. Clin Transl Med 2024;14:e1652.
crossref pmid pmc
151. Liu H, Yang H, Chen X. Prognostic value of circulating tumour DNA in Asian patients with hepatocellular carcinoma: a systematic review and meta-analysis. Evid Based Complement Alternat Med 2022;2022:8019652.
crossref pmid pmc pdf
152. Alunni-Fabbroni M, Rönsch K, Huber T, Cyran CC, Seidensticker M, Mayerle J, et al. Circulating DNA as prognostic biomarker in patients with advanced hepatocellular carcinoma: a translational exploratory study from the SORAMIC trial. J Transl Med 2019;17:328.
crossref pmid pmc pdf
153. Wang H, Hou L, Li A, Duan Y, Gao H, Song X. Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed Res Int 2014;2014:864894.
crossref pmid pmc pdf
154. Qu Z, Wu J, Wu J, Ji A, Qiang G, Jiang Y, et al. Exosomal miR-665 as a novel minimally invasive biomarker for hepatocellular carcinoma diagnosis and prognosis. Oncotarget 2017;8:80666-80678.
crossref pmid pmc
155. Lin XJ, Fang JH, Yang XJ, Zhang C, Yuan Y, Zheng L, et al. Hepatocellular carcinoma cell-secreted exosomal microRNA-210 promotes angiogenesis in vitro and in vivo. Mol Ther Nucleic Acids 2018;11:243-252.
crossref pmid pmc
156. Fang JH, Zhang ZJ, Shang LR, Luo YW, Lin YF, Yuan Y, et al. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018;68:1459-1475.
crossref pmid pdf
157. Shi M, Jiang Y, Yang L, Yan S, Wang YG, Lu XJ. Decreased levels of serum exosomal miR-638 predict poor prognosis in hepatocellular carcinoma. J Cell Biochem 2018;119:4711-4716.
crossref pmid pdf
158. Liu W, Hu J, Zhou K, Chen F, Wang Z, Liao B, et al. Serum exosomal miR-125b is a novel prognostic marker for hepatocellular carcinoma. Onco Targets Ther 2017;10:3843-3851.
crossref pmid pmc pdf
159. Lee YR, Kim G, Tak WY, Jang SY, Kweon YO, Park JG, et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int J Cancer 2019;144:1444-1452.
crossref pmid pdf
160. Wang G, Liu W, Zou Y, Wang G, Deng Y, Luo J, et al. Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine 2019;40:432-445.
crossref pmid pmc
161. Zhang PF, Gao C, Huang XY, Lu JC, Guo XJ, Shi GM, et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol Cancer 2020;19:110.
crossref pmid pmc pdf
162. Xu Y, Cai J, Zhong K, Wen Y, Cai L, He G, et al. Plasma-only circulating tumor DNA analysis detects minimal residual disease and predicts early relapse in hepatocellular carcinoma patients undergoing curative resection. Front Oncol 2023;13:1119744.
crossref pmid pmc
163. Yang P, Si A, Yang J, Cheng Z, Wang K, Li J, et al. A wide-margin liver resection improves long-term outcomes for patients with HBV-related hepatocellular carcinoma with microvascular invasion. Surgery 2019;165:721-730.
crossref pmid
164. Zhu GQ, Liu WR, Tang Z, Qu WF, Fang Y, Jiang XF, et al. Serial circulating tumor DNA to predict early recurrence in patients with hepatocellular carcinoma: a prospective study. Mol Oncol 2022;16:549-561.
pmid
165. Nakano T, Chen IH, Wang CC, Chen PJ, Tseng HP, Huang KT, et al. Circulating exosomal miR-92b: its role for cancer immunoediting and clinical value for prediction of posttransplant hepatocellular carcinoma recurrence. Am J Transplant 2019;19:3250-3262.
crossref pmid pdf
166. Juratli MA, Pollmann NS, Oppermann E, Mohr A, Roy D, Schnitzbauer A, et al. Extracellular vesicles as potential biomarkers for diagnosis and recurrence detection of hepatocellular carcinoma. Sci Rep 2024;14:5322.
crossref pmid pmc pdf
167. Yi K, Wang Y, Rong Y, Bao Y, Liang Y, Chen Y, et al. Transcriptomic signature of 3D hierarchical porous chip enriched exosomes for early detection and progression monitoring of hepatocellular carcinoma. Adv Sci (Weinh) 2024;11:e2305204.
crossref pmid pmc
168. Sefrioui D, Verdier V, Savoye-Collet C, Beaussire L, Ghomadi S, Gangloff A, et al. Circulating DNA changes are predictive of disease progression after transarterial chemoembolization. Int J Cancer 2022;150:532-541.
pmid
169. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894-1905.
crossref pmid
170. Nosaka T, Murata Y, Akazawa Y, Tanaka T, Takahashi K, Naito T, et al. Programmed death ligand 1 expression in circulating tumor cells as a predictor and monitor of response to atezolizumab plus bevacizumab treatment in patients with hepatocellular carcinoma. Cancers (Basel) 2024;16:1785.
crossref pmid pmc
171. Ikeda S, Tsigelny IF, Skjevik ÅA, Kono Y, Mendler M, Kuo A, et al. Next-generation sequencing of circulating tumor DNA reveals frequent alterations in advanced hepatocellular carcinoma. Oncologist 2018;23:586-593.
crossref pmid pmc pdf
172. Clinical Trial Multi-analyte Blood Test (CLiMB). ClinicalTrials. gov web site <https://clinicaltrials.gov/study/NCT03694600>. Accessed 31 Oct 2024.

173. ALTUS: Performance of a Multi- Target Hepatocellular Carcinoma (HCC) Test in Subjects With Increased Risk. ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT05064553>. Accessed 31 Oct 2024.

174. Qu C, Wang Y, Wang P, Chen K, Wang M, Zeng H, et al. Detection of early-stage hepatocellular carcinoma in asymptomatic HBsAg-seropositive individuals by liquid biopsy. Proc Natl Acad Sci U S A 2019;116:6308-6312.
crossref pmid pmc
175. Wang P, Song Q, Ren J, Zhang W, Wang Y, Zhou L, et al. Simultaneous analysis of mutations and methylations in circulating cell-free DNA for hepatocellular carcinoma detection. Sci Transl Med 2022;14:eabp8704.
crossref pmid
176. Lewin J, Kottwitz D, Aoyama J, deVos T, Garces J, Hasinger O, et al. Plasma cell free DNA methylation markers for hepatocellular carcinoma surveillance in patients with cirrhosis: a case control study. BMC Gastroenterol 2021;21:136.
crossref pmid pmc pdf
177. Liquid Biopsy in Hepatocellular Carcinoma (HCCGenePanel). ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT04111029>. Accessed 31 Oct 2024.

178. A Prospective Study on the Application of Liquid Biopsy in the Surveillance of High-risk Population of HCC. ClinicalTrials. gov web site <https://clinicaltrials.gov/study/NCT06134973>. Accessed 31 Oct 2024.

179. PLATON - Platform for Analyzing Targetable Tumor Mutations (Pilot-study) (PLATON). ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT04484636>. Accessed 31 Oct 2024.

180. An Exosome-Based Liquid Biopsy for the Differential Diagnosis of Primary Liver Cancer (ELUCIDATE). ClinicalTrials. gov web site <https://clinicaltrials.gov/study/NCT06342414>. Accessed 31 Oct 2024.

181. The Role Of Circulating Tumor Cells As Markers Of Advanced Disease And Prognosis In HCC. ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT04800497>. Accessed 31 Oct 2024.

182. Evaluation of Treatment Predictors Reflecting Beta-catenin Activation in Hepatocellular Carcinoma (ExTRACT-HCC). ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT04965454>. Accessed 31 Oct 2024.

183. A Study on the Prevalence of Clinically Useful Mutations in Solid Tumor Characterized by Next Generation Sequencing Methods on Liquid Biopsy Analysis (POPCORN) (POPCORN). ClinicalTrials.gov web site <https://clinicaltrials.gov/study/NCT06028724>. Accessed 31 Oct 2024.

184. Aquino IMC, Pascut D. Liquid biopsy: new opportunities for precision medicine in hepatocellular carcinoma care. Ann Hepatol 2024;29:101176.
crossref pmid
185. Chen G, Zhang J, Fu Q, Taly V, Tan F. Integrative analysis of multi-omics data for liquid biopsy. Br J Cancer 2023;128:505-518.
crossref pmid pmc pdf
186. Choy LYL, Peng W, Jiang P, Cheng SH, Yu SCY, Shang H, et al. Single-molecule sequencing enables long cell-free DNA detection and direct methylation analysis for cancer patients. Clin Chem 2022;68:1151-1163.
crossref pmid pdf
187. Long F, Zhong W, Zhao F, Xu Y, Hu X, Jia G, et al. DAB2+ macrophages support FAP+ fibroblasts in shaping tumor barrier and inducing poor clinical outcomes in liver cancer. Theranostics 2024;14:4822-4843.
crossref pmid pmc

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 1525
TOTAL : 2560005
Close layer