Next Article in Journal
Identifying Progression-Specific Alzheimer’s Subtypes Using Multimodal Transformer
Next Article in Special Issue
Navigating HER2-Low Testing in Invasive Breast Cancer: Update Recommendations for Mexican Pathologists
Previous Article in Journal
The Effect of Probiotics on the Prognostication of the Neutrophil-to-Lymphocyte Ratio in Severe Multi-Trauma Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Application of Different Liquid Biopsy Components in Hepatocellular Carcinoma

by
Jing Xu
,
Yuanyuan Zhao
,
Zhishui Chen
and
Lai Wei
*
Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2024, 14(4), 420; https://doi.org/10.3390/jpm14040420
Submission received: 23 February 2024 / Revised: 26 March 2024 / Accepted: 4 April 2024 / Published: 15 April 2024
(This article belongs to the Special Issue Cancer Biomarkers: Promises and Challenges)

Abstract

:
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, usually occurring in the background of chronic liver disease. HCC lethality rate is in the third highest place in the world. Patients with HCC have concealed early symptoms and possess a high-level of heterogeneity. Once diagnosed, most of the tumors are in advanced stages and have a poor prognosis. The sensitivity and specificity of existing detection modalities and protocols are suboptimal. HCC calls for more sophisticated and individualized therapeutic regimens. Liquid biopsy is non-invasive, repeatable, unaffected by location, and can be monitored dynamically. It has emerged as a useable aid in achieving precision malignant tumor treatment. Circulating tumor cells (CTCs), circulating nucleic acids, exosomes and tumor-educated platelets are the commonest components of a liquid biopsy. It possesses the theoretical ability to conquer the high heterogeneity and the difficulty of early detection for HCC patients. In this review, we summarize the common enrichment techniques and the clinical applications in HCC for different liquid biopsy components. Tumor recurrence after HCC-related liver transplantation is more insidious and difficult to treat. The clinical use of liquid biopsy in HCC-related liver transplantation is also summarized in this review.

1. Introduction

Primary liver cancer is the sixth most common cancer [1], and hepatocellular carcinoma (HCC) is the most common histological category, accounting for 70–85% of the morbidity in primary liver cancer patients [2]. Statistically, HCC is the second most fatal malignancy after pancreatic cancer, its five-year survival rate is approximately 18% [3]. The morbidity and mortality of HCC has continued to increase worldwide in recent years compared to other common cancers (e.g., breast, lung and prostate cancer). Frustratingly, the holistic prognosis of HCC continues to be very poor, owing to the concealment of symptoms in the early stage, the lack of validated early detection instruments, restricted therapeutic alternatives and the high recurrence rate [4].
With the perspective of the individuation and precision therapy of oncology taking root, liquid biopsy has gained the limelight. Blood is the most broadly used type of liquid biopsy specimen in medical research. In addition to blood, body fluids such as saliva [5], sputum [6], cerebrospinal fluid [7,8,9,10,11], bile [12], urine [13,14,15,16,17,18,19,20,21], stool [22,23,24], seminal fluid [25], and uterine lavage fluid [26] have also been explored in a variety of malignancies (shown in Figure 1). There are many substances that can be used as liquid biopsy targets, including circulating tumor DNA (ctDNA), circulating miRNA, exosomes, and tumor-educated platelets (TEP), in addition to circulating tumor cells (CTCs) and circulating cell-free DNA (cfDNA), which are more widely used. Liquid biopsy has been clinically approved for the diagnosis, treatment efficacy evaluation, and prognosis assessment of a variety of malignancies [27]. For example, CTCs are approved by the US Food and Drug Administration (FDA) as a prognostic assessment for breast, prostate, and colon carcinomas. Whereas, cfDNA is licensed for the adjuvant detection of non-small cell lung cancer (NSCLC), breast, and ovarian carcinomas.
This review aims to summarize the available enrichment techniques and clinical research on liquid biopsy from blood samples. With the growing number of HCC-associated liver transplant patients, tumor recurrence surveillance for this subset of patients is also an important factor affecting the patients’ overall survival. We specifically summarize clinical studies related to liquid biopsy in HCC-related liver transplant patients.

2. Application of Liquid Biopsy in HCC

Since 1869, when the first detection of circulating tumor cells in breast cancer patients’ blood was made by Thomas Ashworth, the exploration of liquid biopsy has been ongoing for years (shown in Figure 2). Theoretically, liquid biopsy is non-invasive, repeatable, has good patient adherence, without the limitation of the biopsy site and allows for dynamic observation. Moreover, the spatial and temporal heterogeneity of tumor development can be surmounted by repeated follow-ups [28]. Thus, it is straightforward to understand the patient’s tumor progression and even therapeutic effects. Liquid biopsy holds great promise for screening, diagnosis, therapeutics and the monitoring of HCC patients.

2.1. CTCs

Carcinoma cells shedding from primary or metastatic tumors and circulating in body fluids are called CTCs. They are the earliest detectable markers that can be used for biopsy. CTCs are widespread among malignant tumor patients but extremely rare in healthy individuals or benign tumor patients [29]. In accordance with the previous findings, most CTCs are cleared upon entering the peripheral circulation, and only a small percentage of CTCs are capable of surviving in the peripheral circulatory system [30]. The surviving CTCs may come from various locations within the tumor and have different physiologies, and their capabilities of survival, invasion and metastasis vary [31]. In the seed soil hypothesis, the circulatory CTCs are considered the seeds of tumor metastasis [32].
The CTC enrichment approaches are numerous. Physical property-based assays [33,34,35,36] are convenient and fast, but CTCs have high variability in size. Their physical properties overlap significantly with leukocytes. So, this class of assays is less specific. The widest application currently is using immunoaffinity separation techniques for CTC enrichment. In Table 1, we exemplify the current CTC enrichment techniques and the respective advantages and disadvantages of each of them. Among these assays, the Cell Search System represents the only FDA-approved assay available in malignant tumor patients. Epithelial mesenchymal transition (EMT) is a phenotypic change characterized by epithelial property loss and mesenchymal property gain. Due to the role of EMT in tumor progression and metastasis [37], CTCs in malignant patients can be classified into three sub-groups: epithelial CTCs (E-CTCs), mesenchymal CTCs (M-CTCs) and intermediate subtypes undergoing transformation CTCs (E/M-CTCs) [38]. Hence, circulating CTCs in Malignant tumor patients who are undergoing transformation or have completed EMT do not represent the epithelium-derived specific marker, Epithelial cell adhesion molecule (Ep CAM) [39]. The CTCs in HCC patients have also been demonstrated to undergo EMT while in circulation [40]. This leads to a decline in Cell Search System monitoring sensitivity [41].
CTCs are currently widely used in therapeutic modality selection, treatment evaluation and recurrence monitoring of HCC. Jun Yan et al. demonstrated a significant relationship between tumor size and CTC number in an in situ mouse model of HCC, and the number of CTCs was also strongly connected with the treatment response [63]. Consistent results were also reported in HCC patients. CTC counts were significantly correlated with higher serum AFP levels, multiple lesions, more advanced TNM and BCLC stages, and embolisms or micro-embolisms [64]. The research by Lu-Nan Qi et al., which evaluated the role of CTCs in the diagnosis and prognosis of HCC patients, confirmed that CTC counts were substantially higher in advanced HCC patients with Barcelona Clinic Liver Cancer (BCLC)B–C stage than in early stage (BCLC0~A stage). Unexpectedly, two patients with HBV who were included in the same study as controls displayed low levels of CTC positivity and had a confirmed diagnosis of HCC at follow-up [65]. Malignant tumors metastasize in parallel with the development of the primary tumor [66]. CTC counts could serve as a promising indicator for HCC metastasis early diagnosis and disease progression assessment. This conclusion was evidenced in a retrospective study involving 195 patients with HCC [67]. Although there are no trustworthy prospective trials applying CTC for HCC screening, it is theoretically expected. CTCs may also have some guidance on how to maximize patient benefit by selecting from anatomical resection (AR) and non-anatomical resection (NAR). The retrospective study by Lu-Nan Qi et al. on 136 HCC patients treated with R0 resection pointed out that pre-operative CTC analysis could be used to improve the choice of resection procedure for HCC patients, and that AR only improved patient survival in those with low CTC counts and negative M–& E/M-CTC phenotypes [68]. In addition to the surgical choice of liver resection, CTC can also provide pre-operative guidance on the best surgical margins before liver resection. Pre-operative CTC status can also predict the magnitude of microvascular infiltration (MVI) in HCC patients before obtaining laboratory pathology results, thus guiding therapeutic and evaluating prognosis [69]. The amount of CTCs changes over time with the treatment process; therefore, theoretically, the dynamic observation of CTC levels can assess the treatment effect and monitor recurrence. CTC load decreases shortly after resection, and a higher predisposition to tumor recurrence is demonstrated by patients with persistently high post-operative CTC load [70]. Yun-Fan Sun et al. discovered that post-operative CTC count was a predictor of extrahepatic metastasis after curative surgical resection in a retrospective training cohort of 144 HCC patients and this was confirmed in a prospective validation cohort of 53 patients [71].
CTCs possess an intact cellular structure, can fulfill cellular morphology and function research under in vitro conditions, and can even be cultured or developed in vitro. By using CTCs, it is possible to forecast the reaction or resistance of tumor cells to given treatments in vitro and direct individualized programs. The nucleic acids and proteins contained within them can also be analyzed for corresponding histology [72]. Nevertheless, the CTC expression is extremely low in the blood [73] and existing enrichment strategies are not able to fulfill the clinical requirements. No site-specific markers have been found to be stably expressed in HCC patients CTCs [74]. Two systems, the Cell Search System and Can Patrol System [65] still need trustworthy prospective studies to compare their effects on HCC patients. Additionally, there are a few studies related to the use of CTC testing in patients undergoing nonradical therapies, and the future of the use of this liquid biopsy type in such patients is unknown. Existing studies in HCC patient cohorts are scattered and lack consistency, making it impossible to compare the outcomes. Despite the theoretical feasibility of overcoming tumor size and heterogeneity, CTC detection remains only an indirect method in the management process of HCC patients. Large clinical trials are needed to demonstrate this in the future.

2.2. cfDNA & ctDNA

Mandel and Metais identified fragmented DNA in the cell-free component of blood circulation in cancer patients in 1948, giving rise to the concept of cell-free DNA (cfDNA). Thereafter, cfDNA did not receive adequate focus until 1975. Leon et al. concluded that cfDNA could be applied to assess the prognosis of malignancy patients. CfDNA is a typically double-stranded fragment of approximately 150–200 base pairs in length. Free DNA fragments in circulation have a relatively complex origin. In healthy individuals, circulating cfDNA is predominantly originated from hematopoietic cells, especially leukocytes, and it is typically at low steady-state levels [75,76,77]. In the circulation of malignancy patients, the fluctuating intensity of cfDNA ranges widely [78]. The abnormalities of cfDNA in malignant tumor patients include both quantitative and qualitative changes. Quantitative changes refer to elevated levels of cfDNA in malignancy patients compared to healthy individuals or benign disease patients. When patients with malignant tumors receive treatment, the serum levels will be further increased [79]. Qualitative changes include altered DNA integrity and tumor-associated genetic variants such as specific point mutations and DNA methylation. Circulating tumor DNA (ctDNA) is a portion of cfDNA that expresses specific somatic mutations associated with the development of malignant tumors, accounting for approximately 1% of the total [80]. The overwhelming proportion of ctDNA is not generated from CTCs [81].
There are two assays currently in use. One is using PCR techniques to detect some mutations already known, including real-time fluorescence quantitative PCR (qPCR), droplet digital PCR (dd PCR), etc. The other is using non-targeted techniques to classify the detected DNA fragments (e.g., Whole Genome Sequencing (WGS), Sanger sequencing and Next-generation sequencing (NGS)). The former can not only magnify DNA fragments but also dynamically filter aberrations in DNA fragments with high sensitivity and low cost; however, only mutations in a pre-established panel of genes can be detected. The latter one can demonstrate the entire genomic map more thoroughly, but is relatively time-consuming and costly [82].
Ken Chen et al. compared serum cfDNA levels between HCC patients and healthy persons and found that serum cfDNA levels were positively linked to the HCC differentiation degree and metastasis but negatively correlated with patient prognosis. Also, serum cfDNA levels were relevant to survival after hepatectomy. To some extent, it has been indicated that cfDNA could be beneficial for HCC monitoring and could be used as a potential marker for decision-making on surgical or non-surgical treatment [83]. Korean research on post-radiotherapy monitoring of HCC patients also stated that the cfDNA levels of patients with pre- and post-therapy were significantly different, and the cfDNA levels measured immediately after radiotherapy could earlier forecast the partial therapeutic response [84]. Also, serum cfDNA levels have been demonstrated to be a prognostic indicator for extrahepatic recurrence after curative surgery in HCC patients [85]. In addition to variations in content, HCC patients showed abnormalities in the integrity of circulating cfDNA compared to healthy individuals or benign disease patients. A study by Ao Huang et al., using qPCR to detect cfDNA integrity in 69 HCC patients who underwent hepatectomy, stated that patients with HCC had a lower integrity of circulating cfDNA compared to healthy individuals or benign liver disorders, and the cfDNA integrity sharply increased after hepatectomy [86]. Research on circulating cfDNA integrity in HCC patients using parallel sequencing methods, nevertheless, denoted that free DNA fragments in HCC patients’ plasma existed at different lengths, both shorter and longer [87]. There is no controversy that the size of plasma DNA fragments varies depending on their origins. Atsushi Ono et al. investigated that the presence of ctDNA could indicate neoplastic progression and that ctDNA could be tested to anticipate portal vein invasion and neoplasm recurrence, especially extrahepatic metastases within 2 years. In addition, the results of this study also suggested that ctDNA showed a better and more sensitive outcome than traditional tumor markers in some patients [88]. Research on ctDNA, after all, has mainly focused on the analysis of its molecular characteristics. By analyzing whole-exome sequencing data from 11 HCC patients’ tumor samples, Geng Chen et al. proved that different phenotypes of clonal evolution do exist in the HCC patient population, and these phenotypes are significantly connected with patients’ clinical disease duration. Simultaneously, dynamic monitoring of ctDNA levels can mirror HCC load in real time and can be used to pursue tumor clonal evolution in HCC [89]. CtDNA has a short-term middle life and its content depends on tumor load, resulting in broader applications in detecting HCC recurrence and predicting prognosis. Prior to hepatectomy, the status of HCC patients’ ctDNA can be a predictor of tumor recurrence possibility, and the genetic analysis of ctDNA at multiple time points combined with tumor tissue genome sequencing can provide pinpoint information for tumor recurrence [90]. The mutations detected in circulating free DNA fragments can, to some extent, reveal the pool of tumor-associated gene mutations in patients, and therefore, the analysis of ctDNA can deliver a high degree of specificity [91,92,93,94]. In parallel, its quantity correlates with tumor load, the ctDNA is abundant in advanced HCC patients [95]. Johann von Felden et al. utilized mutational signatures of ctDNA to characterize the mutational landscape of advanced HCC patients in detail, filling the vacancy of lacking adequate specimens for biomarker investigations in advanced HCC patients and, to a certain extent, demonstrating again the clinical usefulness and application prospects of ctDNA molecular analysis [96]. But in fact, the amount of ctDNA is very minimal, so there are still missing credible clinical studies in the early diagnosis of HCC. However, some investigators have established a diagnostic prediction model including ctDNA by comparing tumor tissue and normal hematopoietic cell-derived cfDNA gene profiles [97]. This model achieves high diagnostic specificity and sensitivity for HCC tumor load, treatment response and staging, which, to a certain extent, reflects the potential of ctDNA in early diagnosis of HCC. Though free DNA fragment detection is represented in some studies as more sensitive than traditional tumor markers for HCC, more investigations suggest combining the two assays for better sensitivity and accuracy [83,98].
CfDNA/CtDNA correlation analysis yields a comprehensive tumor-associated mutation spectrum. It is more convenient for evaluating tumor heterogeneity, overseeing tumor kinetics, tracing genomic evolution, and monitoring the emergence of acquired drug resistance [95]. However, the tumor histological information carried by DNA fragments is incomplete [99]. This approach fails to recognize the abnormal expression of non-DNA-based alterations in patients. Certain biological elements may also affect the results of the measurements. It is still a perplexing puzzle as to how cfDNA/ctDNA is released into the bloodstream. The contribution of different cellular origins to their content during different physio-pathological processes, such as tumor progression, therapy response, and drug resistance is also not clear [100]. DNA fragments have a short circulating half-life [101], and the appropriate monitoring time needs to be further determined. Existing studies of cfDNA/tDNA analysis are not standardized, and the steps involved prior to analysis may affect the results of the measurements [94], thus the implementation of cfDNA/ctDNA in the clinical milieu remains limited.

2.3. Circulating microRNA

MicroRNAs are a category of homologous, evolutionary conserved, non-coding small molecules with a length of about 20~24 nucleotides. They mainly interact directly with the corresponding target messenger RNA (mRNA) at the post-transcriptional level to exert gene regulation [102,103]. The first report on the connection between miRNAs and malignancies can be dated back to 2002 [104]. As investigations intensified, miRNA signatures displayed remarkable differences according to tumor categories, thus establishing a great potential of miRNAs in the diagnosis, prognosis and therapeutic evaluation of malignant neoplasm patients [105]. For most malignancy patients, miRNAs have a two-fold function: being carcinogenic and tumor-suppressive. The expression of miRNAs in HCC patients’ neoplastic tissues also varied remarkably depending on their roles. In Table 2, abnormal miRNA expression changes in HCC tissues and their roles in the development of HCC are listed. MiRNAs in tissue specimens, despite demonstrating great promise for application are invasive. Circulating miRNAs appear to be a much safer and more effective option. There are two major origins of the extracellular milieu miRNAs, selectively exported by active cells or excreted as a byproduct of cellular activity and cell death [106]. Using integrated serum samples from prostate cancer patients and xenograft models, Patrick S. Mitchell et al. demonstrated that circulating miRNAs are present in the circulation with a high degree of stability. In addition to this, their study indicated that miRNAs have anti-plasma RNase activity in the circulatory system, the levels of miRNAs released into the circulatory system by malignant tumors are adequate for detection. Moreover, it is feasible to apply miRNAs as biomarkers regardless of both serum and plasma sources [107]. Based on this conclusion, circulating miRNAs are well-equipped to perform as circulating biomarkers for the detection of common human cancer species.
The methodologies employed to test circulating miRNAs include PCR, in situ hybridization, microarrays and RNA sequencing, etc. [130]. Quantitative PCR is still the most widely used one.
Serum miRNA-107 levels were significantly elevated in HCC patients compared to healthy volunteers and may be a viable marker for early HCC screening and diagnosis [131]. Circulating miRNA levels were analyzed in a group of hepatitis B virus (HBV) patients with pathologically confirmed dysplastic nodule (DN) and early HCC. miR-122 and let—7b levels exhibited significant differences in this cohort, indicating that miRNA can be used to identify early HCC from DN in patients with chronic hepatitis B [132]. Likewise, plasma miR-148a has demonstrated its ability to recognize HCC patients, cirrhotic patients and healthy individuals. It showed similar susceptibility and specificity to AFP in the diagnosis of HCC. The same applies to AFP-negative HCC patients. Circulating miR-148a levels progressively decreased during the process of cirrhosis to HCC and recovered after hepatectomy, expressing the clinically required sensitivity and specificity of the biomarker for neoplasia [133]. Previous studies have noted that different causes of HCC manifest different miRNA abnormalities [134]. This was reconfirmed in a study that correlated the premature prediction of hepatitis C virus with (HCV)-associated HCC patients. Nine miRNAs (miR-142, miR-150, miR-183, miR-199a, miR-215, miR-217, miR-224, miR-424 and miR-3607) were discovered to be differentially expressed in HCC and HCV patients compared to healthy volunteer serum specimens. The investigators combined miR-150, miR-199a, miR-224, miR-424 and miR-3607 together as an early detection instrument and it did show better sensitivity and specificity [135]. miR-497 and miR-1246 levels not only related to TNM stages and metastasis in HCC but also to some extent, could act as a prognostic predictor [118]. Besides its great promising application in the early diagnosis of HCC, miRNA has also been tried to appraise therapeutic efficacy and prognosis. In two cohorts comprised of HCC patients who underwent radiofrequency ablation (RFA), the Let—7c expression level proved to be a predictor of post-treatment tumor recurrence. Unfortunately, no clear predictive value for recurrence was found in the small group of HCC patients who underwent surgery in the same study [136]. Changes in circulating miRNA levels are equally relevant for surveillance in HCC patients receiving thermal ablation (TA) and transcatheter arterial chemoembolization (TACE) [137]. In the systemic therapeutic treatment of HCC, FDA-approved drugs are targeted at a single point. miRNAs can engage in key pathways of HCC development and progression by binding to multiple mRNAs, which means that miRNAs have the potential to be valuable therapeutically [138]. Douglas D. Young et al. first reported microRNA-related drugs in 2010. miR-122 inhibitors can reduce HCV virus replication in hepatocytes, while their activators can selectively cause apoptosis in hepatocellular carcinoma cells [139]. Since then, many miRNA regulators have been shown to have anti-tumor efficacy [140,141,142].
Circulating microRNA can comprehensively reflect changes in the post-transcriptional level of tumor cells. Its potential therapeutic value is more worthy of attention and in-depth study. There are numerous categories of miRNAs, and the same miRNA may correspond to several different targets [104]. The current studies are mostly retrospective in the laboratory, focusing on specific cases rather than broadly. Meanwhile, miRNA effects in the molecular pathogenesis of HCC at different stages have not been sufficiently investigated in depth. Further investigations are necessary to pinpoint the exact time when circulating miRNAs are detectable in the circulation during the genesis and progression of HCC.

2.4. Exosomes

The concept of the extracellular vesicle (EV) was first used to describe vesicles shed from the plasma membrane of reticulocytes in 1983 [143]. In 1985, Poutsiaka et al. demonstrated that malignant cells can down-regulate tumor–host immune function by releasing EVs [144]. It was later utilized to describe granules that are naturally released by cells into the extracellular space, these granules have a lipid bilayer structure similar to that of human cells but do not contain a nucleus hence they do not have the ability to self-replicate [145]. EVs are regarded as important and acids, proteins, lipids, and many other components are isolated within the bilayer membrane structure of extracellular vesicles, as well as organelles; all of those components play vital functions in multiple physiologic and/or pathologic pathways. It has many designations in published studies, including micro-vesicles, exosomes, nanoparticles, particles, etc. The classification was previously dominated by the biological origin of extracellular vesicles: micro-vesicles (100–1000 nm in diameter, derived from the plasma membrane), exosomes (30–100 nm in diameter, endosomal origin) and apoptotic vesicles (50–200 nm in diameter, released by apoptotic cells) [146]. The guidelines suggest that extracellular vesicles should be designated according to size, density, biochemical composition and cellularity in a detailed classification, on which there is no consensus [145]. For the sake of consistency, this review uses exosomes for description. Under the umbrella of the phospho-bilayer, the materials contained within the exosomes are in a separate environment, and therefore, have better stability than proteins, nucleic acids, and other substances that are individually present in the circulation [147]. Exosomes play a facilitating role in the development of liver disease, cirrhosis and HCC from various causes. Many proteins and nucleic acids contained within exosomes can serve as potential markers of HCC [148]. In Table 3, the components of exosomes associated with the process of HCC development, metastasis, and treatment are presented.
Isolation and enrichment techniques for exosomes include ultracentrifugation, ultrafiltration, immunoaffinity, polymer-based precipitation, size exclusion chromatography, ion exchange chromatography, microfluidic-based technologies and membrane-based isolation techniques [171]. Ultracentrifugation is still considered the gold standard for isolating exosomes till now [172].
By comparing the exosome mRNA in HCC patients with cirrhosis, chronic hepatitis B, and healthy individuals, the investigators found that mRNA detection within the serum exosomes has early HCC diagnostic value [173]. Similarly, the detection of exosomes lncRNA can be applied to HCV-related HCC early diagnosis and screening [174]. Other components share the same role, such as exosome miRNA-10b-5p [175]. Compared to circulating miRNAs, exosome miRNAs performed better. A prospective study in an HCC patient cohort undergoing curative surgery denoted that exosome miRNA-125b had a predictive function in post-operative HCC recurrence and long-term survival. Its low expression levels were associated with tumor numbers, cellular differentiation and TNM stage; the participants with lower levels exhibited shorter recurrence times and total survival [176]. Tomoyuki Suehiro et al. first reported that decreased levels of exosome miRNA-122 expression were associated with poor prognosis after TACE, and they also remarked that serum exosome miRNA-122 levels could reflect liver injury and residual liver function [177]. A similar finding was observed in the study of serum exosome circular RNAs (circ RNAs) in HCC patients. Circ CCAR1 expression was significantly correlated with vascular invasion, tumor size as well as TNM stage and differentiation grade, and HCC cells with high expression levels were resistant to anti-PD1 therapy [178]. Indeed, circulating exosome non-coding RNAs (ncRNAs) have a certain prognostic value and clinical application in HCC patients regardless of the treatment protocols adopted [179].
Compared to other new liquid biopsies, the stability of the exosome’s bilayer membrane structure and the unique capacity to be a “messenger” enable exosomes to participate in the HCC therapeutic schedule. Most chemotherapeutic drugs are toxic to both cancer cells and healthy cells, and their low specificity often causes undesirable toxicity and side effects. To avoid the normal cell or organ toxicity caused by the free diffusion of drugs, specific drug delivery systems have attracted the eyes of researchers. Compared with artificial synthetic nanodrug delivery materials, exosomes possess the merits of small size, easy accessibility, non-toxicity, organ- or cell-specific tropism, higher stability and bioavailability [180,181,182]. Furthermore, human cell-derived exosomes have many unique advantages that are hard to replace with synthetic materials. Their biocompatibility and low immunogenicity, coupled with the presence of bioactive membrane proteins on their surface, permit the specific delivery of goods to target cells without activating or attenuating the immunogenic response of the host’s immune system [183]. Even more importantly, exosome delivery enables their goods to penetrate the biological/physical barriers of the body (e.g., blood-brain barrier) [184,185]. Exosomes can participate in anti-tumor therapy in two manners: first, by using their capacity of material transportation, injecting exosomes that contain anti-tumor drugs or have oncogenic function in patients; second, by using the immunological properties of exosomes and triggering specific immune responses in tumor patients to achieve anti-tumor effects. Guohua Lou et al. in a nude mouse transplantation tumor model demonstrated that miRNA-122-transfected bone marrow mesenchymal stem cell exosomes could alter the expression of miRNA-122-target genes in HCC cells, thus sensitizing cancer cells to sorafenib by the communication with HCC cells and remarkably increasing the in vivo anti-neoplastic efficacy of sorafenib in HCC [186]. In a similar vein, bone marrow mesenchymal stem cell exosomes can serve as an efficient delivery vehicle for miRNA-199a. By targeting the mTOR pathway, it increases the susceptibility of hepatocellular carcinoma cells to chemotherapeutic agents effectively in an orthotopic HCC mouse model [187]. Apart from the relevant genes being loaded to improve the chemotherapy sensitivity of hepatocellular carcinoma, the chemotherapeutic drugs themselves can also be used as exosome “cargo”. Doxorubicin-loaded exosomes were developed. HCC cells and tumor stem cells (CSCs) showed significant cell uptake and cytotoxicity to the Doxorubicin-loaded exosomes. This conclusion has been confirmed in both subcutaneous transplantation tumor models, orthotopic tumor models and advanced metastatic tumor models [188]. This suggests that exosomes have the potential to act as drug carriers to enhance anticancer efficacy. Even more surprisingly, we can use exosomes to deliver multiple drugs simultaneously and achieve synergistic antitumor effects. Dongdong Wang et al. successfully combined photothermal therapy with low-dose chemotherapy by delivering radioactive particles (Bi2Se3) and chemotherapeutic drugs (doxorubicin) via exosomes. The therapeutic regimen produced significant synergistic anti-tumor effects. This study showed that synergistic treatment through an exosome drug delivery system can reduce the dose of anticancer drugs, thus effectively reducing the side effects and improving economic efficiency [189]. Using mesenchymal stem cell-derived exosomes to block the relevant pathways [190], as well as altering the expression levels of tumor-associated macrophage-derived exosomes miRNA-125a and miRNA-125b [191] can inhibit the malignant behavior of HCC cells in cellular studies. In an in situ HCC mouse model, the vaccine formed by modified exosomes generated dramatic tumor inhibition and even complete tumor eradication by inducing oncology-specific immune responses [192]. It does provide a scalable approach for personalized immunotherapy of HCC.
The exosomes are enriched in contents and stabilized in nature. Existing mature enrichment protocols are still inadequate to completely separate extracellular vesicles from circulation. Exosome isolation and purification technologies are still in the process of development, and there is currently no harmonized methodology or platform for clinical trials to be conducted [193]. Further exploration of whether there are interactions between exosome-rich contents is also needed. It has a very huge development potential as a targeted drug delivery carrier [194]. However, the drug delivery system involved in exosomes is still far from being ready for clinical use. In addition to isolation and purification, how to integrate the desired miRNAs into exosomes, and how to ensure that the exosomes are targeted to the correct organ or tissue. These issues need to be addressed urgently [195].

2.5. Platelets & TEPs

Platelets originate from mature megakaryocytes. They are alive for about seven to ten days in peripheral blood and present in abundant quantities [196]. There is no nucleus inside the platelets, but mitochondrial DNA exists in the cytoplasm; various RNA molecules and proteins can be inherited from megakaryocytes, either generated by themselves or obtained externally [197]. Beyond their essential role in physiological processes such as hemostasis and thrombosis, platelets have been found to be important participants in all steps of oncogenesis and neoplastic progression [198]. Increased platelet content associated with solid neoplasms was originally discovered by Leopold Riess in 1872 [199]. This phenomenon was subsequently noted in patients with a variety of cancers to correlate with poor prognosis [200,201,202,203]. A prospective cohort study of 40,000 patients in the United Kingdom showed that about 5% of cancer victims had increased platelet counts prior to diagnosis [204].
The majority of hepatocellular carcinomas develop from cirrhosis; splenomegaly and hypersplenism caused by hepatic fibrosis are often manifested in HCC patients as well; therefore, platelet count increase is hardly noticeable in HCC patients. The degree of thrombocytopenia is tightly correlated with liver functions. Raised platelet counts are connected with HCC progression. Platelet counts may have opposite predictive roles on HCC patients with different hepatic basal circumstances. In HCC patients with combined hepatocirrhosis, thrombocytopenia may indicate a poor prognosis. In contrast, in patients without cirrhosis, thrombocytosis is more frequently associated with a worrisome outcome [205]. A retrospective cohort study found a well-signed positive correlation between pre-therapeutic platelet count and extrahepatic metastasis of HCC [206]. Simultaneously, the study argued that the comparatively lower incidence of extrahepatic metastases in HCC patients could be partly facilitated by thrombocytopenia. Some researchers are skeptical about this conclusion [207]. A study performed in two large HCC cohorts, on the other hand, found that thrombocytosis was independently associated with high tumor load and poor prognosis [208]. Higher pre-treatment platelet levels may be associated with the more aggressive tumor type. On a similar note, contrary findings have been reported. Research by Sangbin Han et al. concluded that there was no correlation between platelet count and tumor aggressiveness. However, they also expressed that the risk of postoperative mortality after HCC-associated living donor liver transplantation increased with an elevated platelet count [209]. Pretreatment measurement of platelet counts in TACE-treated HCC patients may predict, to some extent, both the tumor response to treatment and the long-term prospects of the patient [210]. The relationship between platelet count and HCC patient prognosis is controversial [211].
When malignant tumors develop in the organism, platelets are also “educated” by tumor cells. During this dual interaction, the RNA and protein expression profiles in the platelet cytoplasm are altered accordingly. Therefore, they are also called “tumor-educated platelets” (TEPs) [212]. TEPs possess the potential for early cancer diagnosis [213], disease progression surveillance [214] and treatment response supervision [215]. TEPs are more specific than simple platelet count changes. The previously addressed novel biomarkers are low in early cancer stages. TEPs are abundant in circulation and with standardized isolation procedures, may be a superior candidate [197]. TGF-β, NF-κB, VEGF, AKT, and PI3K were selected as alternative biomarkers based on previous investigations, and blood platelets were tested in 20 confirmed HCC patients and 10 control samples [216]. All selected RNA biomarkers were sufficient to successfully separate HCC patients from controls. Amongst them, AKT and PI3K have good detection potential for early HCC. Likewise, mRNAs such as CTNNB1, Rho A, SPINK1, IFITM3, and SERPIND1 contained in TEPs could permit earlier diagnosis of HCC from cirrhotic nodules. This result was evidenced in a 50-person cohort comprised of healthy subjects, patients with HCC, and cirrhosis [217]. By the bioinformatic analysis of platelet miRNAs, 250 miRNAs with differential expression were identified in blood samples from HCC patients and controls. These miRNAs, such as miR-495-3p and miR-1293, may serve a role in the growth, development, and metastasis of HCC [218].
Simple platelet count changes are convenient to measure, but their significance in HCC patients is unclear. TEPs are characterized by high stability and richness in blood. Due to their “education” by tumor cells, RNA expression and proteomic analysis of TEPs are of special significance. TEPs, although highly feasible in theoretical terms, have too few studies available. Most of the studies focus on the application of HCC screening. In fact, the splicing mechanism in platelets may be distinct from that in cells [219]. The same miRNAs may have entirely divergent effects in platelets and cells. Thus, relevant research needs to be further refined.

3. HCC-Associated Liver Transplantation and Liquid Biopsy

Favored by the simultaneous resection of the primary tumor and malignant-prone cirrhotic milieu, liver transplantation (LT) has been the preferred therapeutic option for early-stage HCC. With the progress of HCC therapeutic protocols and satisfactory preoperative conversion therapy, the criteria for HCC patients’ transplantation selection are being worked on to expand them [220,221]. HCC-associated LT is increasing year by year, and the number of HCC recurrences after liver transplantation is increasing as well. Incidentally, in the post-transplant condition, the recipient’s normative and acquired immunity are suppressed, making HCC recurrence more insidious, aggressive, and difficult to control [222]. There are no consensus or best practice guidelines for cancer surveillance and recurrence management after HCC transplantation. Non-invasive and reproducible liquid biopsies may have enormous potential for post-transplant recurrence monitoring.
As mentioned earlier, more sensitive assays are needed for HCC-related liver graft recipients, due to earlier tumor staging, more complete treatment and insidious recurrence. Feng Xue et al. created an integrated subtractive enrichment and immunostaining-fluorescence in situ hybridization (I FISH) platform to detect circulating CTCs. Superior sensitivity was demonstrated in a cohort of HCC-related liver transplant patients and healthy volunteers. It also indicated that the level of CTCs in the peripheral circulation is a potential prognostic marker for HCC-related liver transplant patients [223]. In a prospective study conducted on 47 HCC-associated LT patients, CTC was tested in subjects’ blood samples from pre-operative, a month post-operative, and three months post-operative [224]. They argued that pre- and post-operative CTC assessment for LT was not statistically significant as a predictor of recurrence. However, CTC subtypes did change throughout the follow-up process. In the same vein, Yun-Liang Xie et al. argued that changes in the total number of CTCs pre- and post-liver transplantation could not be used to evaluate the postoperative prognosis in HCC patients. Yet, the positive M-CTCs or the elevated proportion of M-CTCs after transplantation were associated with postoperative recurrence. M-CTC positivity could be an independent risk factor. This subset of patients has a higher recurrence rate and shorter survival after LT [225]. Through miRNA analysis of pre-operative liver transplantation serum samples from four relapsed and two non-relapsed patients, exosome miRNA-718 and miRNA-1246 were found to be differentially expressed. Subsequently, in 59 preoperative serum samples from patients with HCC-related liver transplantation, the investigators discovered that exosome miRNA-718 could be employed as a biomarker for tumor recurrence monitoring [226]. Toshiaki Nakano et al. proved that circulating exosomes have a well-defined significance in HCC occurrence and development within an animal model. This is ground-breaking in the field of HCC. In parallel, they analyzed blood samples from living donor liver transplantation (LDLT) patients and demonstrated that the HCC patients with a higher risk of early post-transplant recurrence (<2 years) were accompanied by a high level of exosome miR-92b before LDLT. After transplantation, patient exosome miR-92b has favorable forecasting accuracy for early HCC recurrence. This biomarker does not have adequate power to predict late HCC recurrence [169].
The use of liquid biopsy in liver transplant patients goes beyond tumor recurrence monitoring. It aids in assessing graft function [227], ameliorating ischemia-reperfusion injury [228,229,230], and monitoring acute rejection [231,232,233].

4. Discussion

In comparison with lung, breast, and prostate cancers, HCC is highly heterogeneous. As a result, HCC is not only difficult to diagnose at an early stage, but its treatment modalities are more varied and its outcome is more unpredictable. This has led researchers to expect more on liquid biopsy. After all, liquid biopsy is full of theoretical feasibility.
Table 4 summarizes the use of different liquid biopsy components in HCC diagnosis and treatment. While it is far more well-studied in other cancers, detection accuracy also limits the clinical use of liquid biopsies. Only a few components have been approved by the FDA for clinical usage in other cancers. In this regard, the development of liquid biopsy relies on more sensitive and specific enrichment protocols. The clinical significance of this technique in HCC patients is still in an advanced stage of exploration. The majority of existing studies on HCC are cellular or animal studies in the laboratory. Existing clinical studies have not standardized the inclusion criteria for liquid biopsy candidates, and whether or not to participate in the studies is largely dependent on clinical practice and patients’ wishes. How to select the most suitable candidates for liquid biopsy needs to be explored in robust studies. Most of the clinical trials in existence, on the other hand, have the shortcomings of small patient enrollment, short follow-up time, and single-center origin. Conditions such as sample provenance, enrichment techniques, sampling duration, etc., used in the different studies lacked uniformity. Therefore, relevant comparisons cannot be made. Large, prospective, independent, multicenter cohort studies are needed to validate the current results.
In summary, although the development of liquid biopsy lagged behind other types of malignant tumors. Liquid biopsy has shown quite an advantage in HCC screening, diagnosis, treatment and prognostic monitoring as well, and it has room for development. We can expect that liquid biopsy will emerge as an essential instrument in HCC patient management one day in the future.

Author Contributions

J.X. wrote the manuscript and drew the figures and tables. Y.Z. revised the manuscript and conceptualized the figures and tables. L.W. and Z.C. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Health Commission of Hubei Province (funder: ZS.C; Grant No. WJ2021C001) and Key Research and Development Plan of Hubei Province (funder: ZS.C; Grant No. 2022BCA015).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ARanatomical resection
BCLCbarcelona clinic liver cancer
CTCcirculating tumor cell
cfDNAcirculating cell-free DNA
CSCtumor stem cell
ctDNAcirculating tumor DNA
dd PCRdroplet digital PCR
DNdysplastic nodule
E-CTCepithelial circulating tumor cell
Ep CAMepithelial cell adhesion molecule
E/M-CTCintermediate subtypes undergoing transformation circulating tumor cell
EMTepithelial mesenchymal transition
EVextracellular vesicle
FDAUS Food and Drug Administration
HBVhepatitis B virus
HCChepatocellular carcinoma
HCVhepatitis C virus
I FISHimmunostaining-fluorescence in situ hybridization
LDLTliving donor liver transplantation
LTliver transplantation
M-CTCmesenchymal circulating tumor cell
mRNAmessenger RNA
MVImicrovascular infiltration
NARnon-anatomical resection
NGSnext-generation sequencing
NSCLCnon-small cell lung cancer
qPCRquantitative PCR
RFAradiofrequency ablation
TAthermal ablation
TACEtranscatheter arterial chemoembolization
TEPtumor-educated platelets
WGSWhole Genome Sequencing

References

  1. Zhang, C.-H.; Cheng, Y.; Zhang, S.; Fan, J.; Gao, Q. Changing epidemiology of hepatocellular carcinoma in Asia. Liver Int. 2022, 42, 2029–2041. [Google Scholar] [CrossRef] [PubMed]
  2. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed]
  3. Jemal, A.; Ward, E.M.; Johnson, C.J.; Cronin, K.A.; Ma, J.; Ryerson, B.; Mariotto, A.; Lake, A.J.; Wilson, R.; Sherman, R.L.; et al. Annual Report to the Nation on the Status of Cancer, 1975–2014, Featuring Survival. J. Natl. Cancer Inst. 2017, 109, djx030. [Google Scholar] [CrossRef] [PubMed]
  4. Kanwal, F.; Singal, A.G. Surveillance for Hepatocellular Carcinoma: Current Best Practice and Future Direction. Gastroenterology 2019, 157, 54–64. [Google Scholar] [CrossRef] [PubMed]
  5. Nonaka, T.; Wong, D.T.W. Saliva Diagnostics. Annu. Rev. Anal. Chem. 2022, 15, 107–121. [Google Scholar] [CrossRef] [PubMed]
  6. Li, P.; Liu, S.; Du, L.; Mohseni, G.; Zhang, Y.; Wang, C. Liquid biopsies based on DNA methylation as biomarkers for the detection and prognosis of lung cancer. Clin. Epigenetics 2022, 14, 118. [Google Scholar] [CrossRef] [PubMed]
  7. Seoane, J.; De Mattos-Arruda, L.; Le Rhun, E.; Bardelli, A.; Weller, M. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann. Oncol. 2019, 30, 211–218. [Google Scholar] [CrossRef] [PubMed]
  8. Simonelli, M.; Dipasquale, A.; Orzan, F.; Lorenzi, E.; Persico, P.; Navarria, P.; Pessina, F.; Nibali, M.C.; Bello, L.; Santoro, A.; et al. Cerebrospinal fluid tumor DNA for liquid biopsy in glioma patients’ management: Close to the clinic? Crit. Rev. Oncol. Hematol. 2020, 146, 102879. [Google Scholar] [CrossRef]
  9. Cheng, H.; Perez-Soler, R. Leptomeningeal metastases in non-small-cell lung cancer. Lancet Oncol. 2018, 19, e43–e55. [Google Scholar] [CrossRef]
  10. Escudero, L.; Martínez-Ricarte, F.; Seoane, J. ctDNA-Based Liquid Biopsy of Cerebrospinal Fluid in Brain Cancer. Cancers 2021, 13, 1989. [Google Scholar] [CrossRef]
  11. Baraniskin, A.; Schroers, R. Liquid Biopsy and Other Non-Invasive Diagnostic Measures in PCNSL. Cancers 2021, 13, 2665. [Google Scholar] [CrossRef] [PubMed]
  12. Li, Y.-C.; Li, K.-S.; Liu, Z.-L.; Tang, Y.-C.; Hu, X.-Q.; Li, X.-Y.; Shi, A.-D.; Zhao, L.-M.; Shu, L.-Z.; Lian, S.; et al. Research progress of bile biomarkers and their immunoregulatory role in biliary tract cancers. Front. Immunol. 2022, 13, 1049812. [Google Scholar] [CrossRef] [PubMed]
  13. Reckamp, K.L.; Melnikova, V.O.; Karlovich, C.; Sequist, L.V.; Camidge, D.R.; Wakelee, H.; Perol, M.; Oxnard, G.R.; Kosco, K.; Croucher, P.; et al. A Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and Plasma. J. Thorac. Oncol. 2016, 11, 1690–1700. [Google Scholar] [CrossRef] [PubMed]
  14. Park, J.-Y.; Kang, C.-S.; Seo, H.-C.; Shin, J.-C.; Kym, S.-M.; Park, Y.-S.; Shin, T.-S.; Kim, J.-G.; Kim, Y.-K. Bacteria-Derived Extracellular Vesicles in Urine as a Novel Biomarker for Gastric Cancer: Integration of Liquid Biopsy and Metagenome Analysis. Cancers 2021, 13, 4687. [Google Scholar] [CrossRef] [PubMed]
  15. Mouliere, F.; Smith, C.G.; Heider, K.; Su, J.; van der Pol, Y.; Thompson, M.; Morris, J.; Wan, J.C.M.; Chandrananda, D.; Hadfield, J.; et al. Fragmentation patterns and personalized sequencing of cell-free DNA in urine and plasma of glioma patients. EMBO Mol. Med. 2021, 13, e12881. [Google Scholar] [CrossRef] [PubMed]
  16. Bach, S.; Paulis, I.; Sluiter, N.R.; Tibbesma, M.; Martin, I.; van de Wiel, M.A.; Tuynman, J.B.; Bahce, I.; Kazemier, G.; Steenbergen, R.D.M. Detection of colorectal cancer in urine using DNA methylation analysis. Sci. Rep. 2021, 11, 2363. [Google Scholar] [CrossRef] [PubMed]
  17. Zuo, Z.; Tang, J.; Cai, X.; Ke, F.; Shi, Z. Probing of breast cancer using a combination of plasma and urinary circulating cell-free DNA. Biosci. Rep. 2020, 40, BSR20194306. [Google Scholar] [CrossRef] [PubMed]
  18. Satyal, U.; Srivastava, A.; Abbosh, P.H. Urine Biopsy—Liquid Gold for Molecular Detection and Surveillance of Bladder Cancer. Front. Oncol. 2019, 9, 1266. [Google Scholar] [CrossRef] [PubMed]
  19. Terasawa, H.; Kinugasa, H.; Ako, S.; Hirai, M.; Matsushita, H.; Uchida, D.; Tomoda, T.; Matsumoto, K.; Horiguchi, S.; Kato, H.; et al. Utility of liquid biopsy using urine in patients with pancreatic ductal adenocarcinoma. Cancer Biol. Ther. 2019, 20, 1348–1353. [Google Scholar] [CrossRef]
  20. Crocetto, F.; Russo, G.; Di Zazzo, E.; Pisapia, P.; Mirto, B.F.; Palmieri, A.; Pepe, F.; Bellevicine, C.; Russo, A.; La Civita, E.; et al. Liquid Biopsy in Prostate Cancer Management—Current Challenges and Future Perspectives. Cancers 2022, 14, 3272. [Google Scholar] [CrossRef]
  21. Crocetto, F.; Barone, B.; Ferro, M.; Busetto, G.M.; La Civita, E.; Buonerba, C.; Di Lorenzo, G.; Terracciano, D.; Schalken, J.A. Liquid biopsy in bladder cancer: State of the art and future perspectives. Crit. Rev. Oncol. Hematol. 2022, 170, 103577. [Google Scholar] [CrossRef] [PubMed]
  22. Caldas, C.; Hahn, S.A.; Hruban, R.H.; Redston, M.S.; Yeo, C.J.; Kern, S.E. Detection of K-ras mutations in the stool of patients with pancreatic adenocarcinoma and pancreatic ductal hyperplasia. Cancer Res. 1994, 54, 3568–3573. [Google Scholar] [PubMed]
  23. Azuara, D.; Rodriguez-Moranta, F.; de Oca, J.; Soriano-Izquierdo, A.; Mora, J.; Guardiola, J.; Biondo, S.; Blanco, I.; Peinado, M.A.; Moreno, V.; et al. Novel methylation panel for the early detection of colorectal tumors in stool DNA. Clin. Color. Cancer 2010, 9, 168–176. [Google Scholar] [CrossRef] [PubMed]
  24. Youssef, O.; Sarhadi, V.; Ehsan, H.; Bohling, T.; Carpelan-Holmström, M.; Koskensalo, S.; Puolakkainen, P.; Kokkola, A.; Knuutila, S. Gene mutations in stool from gastric and colorectal neoplasia patients by next-generation sequencing. World J. Gastroenterol. 2017, 23, 8291–8299. [Google Scholar] [CrossRef] [PubMed]
  25. Ponti, G.; Maccaferri, M.; Manfredini, M.; Micali, S.; Torricelli, F.; Milandri, R.; Del Prete, C.; Ciarrocchi, A.; Ruini, C.; Benassi, L.; et al. Quick assessment of cell-free DNA in seminal fluid and fragment size for early non-invasive prostate cancer diagnosis. Clin. Chim. Acta Int. J. Clin. Chem. 2019, 497, 76–80. [Google Scholar] [CrossRef] [PubMed]
  26. Nair, N.; Camacho-Vanegas, O.; Rykunov, D.; Dashkoff, M.; Camacho, S.C.; Schumacher, C.A.; Irish, J.C.; Harkins, T.T.; Freeman, E.; Garcia, I.; et al. Genomic Analysis of Uterine Lavage Fluid Detects Early Endometrial Cancers and Reveals a Prevalent Landscape of Driver Mutations in Women without Histopathologic Evidence of Cancer: A Prospective Cross-Sectional Study. PLoS Med. 2016, 13, e1002206. [Google Scholar] [CrossRef] [PubMed]
  27. Nikanjam, M.; Kato, S.; Kurzrock, R. Liquid biopsy: Current technology and clinical applications. J. Hematol. Oncol. 2022, 15, 131. [Google Scholar] [CrossRef]
  28. Keller, L.; Pantel, K. Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Rev. Cancer 2019, 19, 553–567. [Google Scholar] [CrossRef]
  29. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.J.; Uhr, J.W.; Terstappen, L.W.M.M. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef]
  30. Micalizzi, D.S.; Maheswaran, S.; Haber, D.A. A conduit to metastasis: Circulating tumor cell biology. Genes Dev. 2017, 31, 1827–1840. [Google Scholar] [CrossRef]
  31. Plaks, V.; Koopman, C.D.; Werb, Z. Circulating tumor cells. Science 2013, 341, 1186–1188. [Google Scholar] [CrossRef]
  32. Fidler, I.J.; Poste, G. The “seed and soil” hypothesis revisited. Lancet. Oncol. 2008, 9, 808. [Google Scholar] [PubMed]
  33. Liu, H.-Y.; Qian, H.-H.; Zhang, X.-F.; Li, J.; Yang, X.; Sun, B.; Ma, J.-Y.; Chen, L.; Yin, Z.-F. Improved method increases sensitivity for circulating hepatocellular carcinoma cells. World J. Gastroenterol. 2015, 21, 2918–2925. [Google Scholar] [CrossRef]
  34. Hou, H.W.; Warkiani, M.E.; Khoo, B.L.; Li, Z.R.; Soo, R.A.; Tan, D.S.-W.; Lim, W.-T.; Han, J.; Bhagat, A.A.S.; Lim, C.T. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 2013, 3, 1259. [Google Scholar] [CrossRef]
  35. Mitchell, M.J.; Castellanos, C.A.; King, M.R. Surfactant functionalization induces robust, differential adhesion of tumor cells and blood cells to charged nanotube-coated biomaterials under flow. Biomaterials 2015, 56, 179–186. [Google Scholar] [CrossRef] [PubMed]
  36. Shaw Bagnall, J.; Byun, S.; Begum, S.; Miyamoto, D.T.; Hecht, V.C.; Maheswaran, S.; Stott, S.L.; Toner, M.; Hynes, R.O.; Manalis, S.R. Deformability of Tumor Cells versus Blood Cells. Sci. Rep. 2015, 5, 18542. [Google Scholar] [CrossRef] [PubMed]
  37. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. 2018, 13, 395–412. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, X.; Ni, H.; Lu, Z.; Zhang, J.; Zhang, Q.; Ning, S.; Qi, L.; Xiang, B. Mesenchymal circulating tumor cells and Ki67: Their mutual correlation and prognostic implications in hepatocellular carcinoma. BMC Cancer 2023, 23, 10. [Google Scholar] [CrossRef] [PubMed]
  39. Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating Breast Tumor Cells Exhibit Dynamic Changes in Epithelial and Mesenchymal Composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef]
  40. Sun, Y.-F.; Guo, W.; Xu, Y.; Shi, Y.-H.; Gong, Z.-J.; Ji, Y.; Du, M.; Zhang, X.; Hu, B.; Huang, A.; 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. [Google Scholar] [CrossRef]
  41. Deng, G.; Herrler, M.; Burgess, D.; Manna, E.; Krag, D.; Burke, J.F. Enrichment with anti-cytokeratin alone or combined with anti-EpCAM antibodies significantly increases the sensitivity for circulating tumor cell detection in metastatic breast cancer patients. Breast Cancer Res. BCR 2008, 10, R69. [Google Scholar] [CrossRef]
  42. Gerges, N.; Rak, J.; Jabado, N. New technologies for the detection of circulating tumour cells. Br. Med. Bull. 2010, 94, 49–64. [Google Scholar] [CrossRef] [PubMed]
  43. Grover, P.K.; Cummins, A.G.; Price, T.J.; Roberts-Thomson, I.C.; Hardingham, J.E. Circulating tumour cells: The evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer research. Ann. Oncol. 2014, 25, 1506–1516. [Google Scholar] [CrossRef] [PubMed]
  44. Hong, B.; Zu, Y. Detecting circulating tumor cells: Current challenges and new trends. Theranostics 2013, 3, 377–394. [Google Scholar] [CrossRef] [PubMed]
  45. Müller, V.; Riethdorf, S.; Rack, B.; Janni, W.; Fasching, P.A.; Solomayer, E.; Aktas, B.; Kasimir-Bauer, S.; Pantel, K.; Fehm, T.; et al. Prognostic impact of circulating tumor cells assessed with the CellSearch SystemTM and AdnaTest BreastTM in metastatic breast cancer patients: The DETECT study. Breast Cancer Res. BCR 2012, 14, R118. [Google Scholar] [CrossRef] [PubMed]
  46. Danila, D.C.; Samoila, A.; Patel, C.; Schreiber, N.; Herkal, A.; Anand, A.; Bastos, D.; Heller, G.; Fleisher, M.; Scher, H.I. Clinical Validity of Detecting Circulating Tumor Cells by AdnaTest Assay Compared with Direct Detection of Tumor mRNA in Stabilized Whole Blood, as a Biomarker Predicting Overall Survival for Metastatic Castration-Resistant Prostate Cancer Patients. Cancer J. 2016, 22, 315–320. [Google Scholar] [CrossRef] [PubMed]
  47. Scherag, F.D.; Niestroj-Pahl, R.; Krusekopf, S.; Lücke, K.; Brandstetter, T.; Rühe, J. Highly Selective Capture Surfaces on Medical Wires for Fishing Tumor Cells in Whole Blood. Anal. Chem. 2017, 89, 1846–1854. [Google Scholar] [CrossRef] [PubMed]
  48. Wu, S.; Liu, Z.; Liu, S.; Lin, L.; Yang, W.; Xu, J. Enrichment and enumeration of circulating tumor cells by efficient depletion of leukocyte fractions. Clin. Chem. Lab. Med. 2014, 52, 243–251. [Google Scholar] [CrossRef] [PubMed]
  49. Buscail, E.; Alix-Panabières, C.; Quincy, P.; Cauvin, T.; Chauvet, A.; Degrandi, O.; Caumont, C.; Verdon, S.; Lamrissi, I.; Moranvillier, I.; et al. High Clinical Value of Liquid Biopsy to Detect Circulating Tumor Cells and Tumor Exosomes in Pancreatic Ductal Adenocarcinoma Patients Eligible for Up-Front Surgery. Cancers 2019, 11, 1656. [Google Scholar] [CrossRef]
  50. Habli, Z.; AlChamaa, W.; Saab, R.; Kadara, H.; Khraiche, M.L. Circulating Tumor Cell Detection Technologies and Clinical Utility: Challenges and Opportunities. Cancers 2020, 12, 1930. [Google Scholar] [CrossRef]
  51. Yu, H.; Ma, L.; Zhu, Y.; Li, W.; Ding, L.; Gao, H. Significant diagnostic value of circulating tumour cells in colorectal cancer. Oncol. Lett. 2020, 20, 317–325. [Google Scholar] [CrossRef] [PubMed]
  52. Li, Y.; Ma, G.; Zhao, P.; Fu, R.; Gao, L.; Jiang, X.; Hu, P.; Ren, T.; Wu, Y.; Wang, Z.; et al. Improvement of sensitive and specific detection of circulating tumor cells using negative enrichment and immunostaining-FISH. Clin. Chim. Acta 2018, 485, 95–102. [Google Scholar] [CrossRef] [PubMed]
  53. Ozkumur, E.; Shah, A.M.; Ciciliano, J.C.; Emmink, B.L.; Miyamoto, D.T.; Brachtel, E.; Yu, M.; Chen, P.-I.; Morgan, B.; Trautwein, J.; et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci. Transl. Med. 2013, 5, 179ra47. [Google Scholar] [CrossRef] [PubMed]
  54. Karabacak, N.M.; Spuhler, P.S.; Fachin, F.; Lim, E.J.; Pai, V.; Ozkumur, E.; Martel, J.M.; Kojic, N.; Smith, K.; Chen, P.-I.; et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat. Protoc. 2014, 9, 694–710. [Google Scholar] [CrossRef] [PubMed]
  55. Heitzer, E.; Speicher, M.R. Digital Circulating Tumor Cell Analyses for Prostate Cancer Precision Oncology. Cancer Discov. 2018, 8, 269–271. [Google Scholar] [CrossRef] [PubMed]
  56. Effenberger, K.E.; Schroeder, C.; Hanssen, A.; Wolter, S.; Eulenburg, C.; Tachezy, M.; Gebauer, F.; Izbicki, J.R.; Pantel, K.; Bockhorn, M. Improved Risk Stratification by Circulating Tumor Cell Counts in Pancreatic Cancer. Clin. Cancer Res. 2018, 24, 2844–2850. [Google Scholar] [CrossRef] [PubMed]
  57. Woestemeier, A.; Harms-Effenberger, K.; Karstens, K.-F.; Konczalla, L.; Ghadban, T.; Uzunoglu, F.G.; Izbicki, J.R.; Bockhorn, M.; Pantel, K.; Reeh, M. Clinical Relevance of Circulating Tumor Cells in Esophageal Cancer Detected by a Combined MACS Enrichment Method. Cancers 2020, 12, 718. [Google Scholar] [CrossRef] [PubMed]
  58. Feng, Z.; Wu, J.; Lu, Y.; Chan, Y.-T.; Zhang, C.; Wang, D.; Luo, D.; Huang, Y.; Feng, Y.; Wang, N. Circulating tumor cells in the early detection of human cancers. Int. J. Biol. Sci. 2022, 18, 3251–3265. [Google Scholar] [CrossRef]
  59. Li, Y.; Wu, G.; Yang, W.; Wang, X.; Duan, L.; Niu, L.; Zhang, Y.; Liu, J.; Hong, L.; Fan, D. Prognostic value of circulating tumor cells detected with the CellSearch system in esophageal cancer patients: A systematic review and meta-analysis. BMC Cancer 2020, 20, 581. [Google Scholar] [CrossRef]
  60. Hugenschmidt, H.; Labori, K.J.; Borgen, E.; Brunborg, C.; Schirmer, C.B.; Seeberg, L.T.; Naume, B.; Wiedswang, G. Preoperative CTC-Detection by CellSearch® Is Associated with Early Distant Metastasis and Impaired Survival in Resected Pancreatic Cancer. Cancers 2021, 13, 485. [Google Scholar] [CrossRef]
  61. Riethdorf, S.; O’Flaherty, L.; Hille, C.; Pantel, K. Clinical applications of the CellSearch platform in cancer patients. Adv. Drug Deliv. Rev. 2018, 125, 102–121. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, L.; Zhu, Y. The EpCAM overexpression is associated with clinicopathological significance and prognosis in hepatocellular carcinoma patients: A systematic review and meta-analysis. Int. J. Surg. 2018, 56, 274–280. [Google Scholar] [CrossRef]
  63. Yan, J.; Fan, Z.; Wu, X.; Xu, M.; Jiang, J.; Tan, C.; Wu, W.; Wei, X.; Zhou, J. Circulating tumor cells are correlated with disease progression and treatment response in an orthotopic hepatocellular carcinoma model. Cytom. Part A J. Int. Soc. Anal. Cytol. 2015, 87, 1020–1028. [Google Scholar] [CrossRef] [PubMed]
  64. Ou, H.; Huang, Y.; Xiang, L.; Chen, Z.; Fang, Y.; Lin, Y.; Cui, Z.; Yu, S.; Li, X.; Yang, D. Circulating Tumor Cell Phenotype Indicates Poor Survival and Recurrence After Surgery for Hepatocellular Carcinoma. Dig. Dis. Sci. 2018, 63, 2373–2380. [Google Scholar] [CrossRef] [PubMed]
  65. Qi, L.-N.; Xiang, B.-D.; Wu, F.-X.; Ye, J.-Z.; Zhong, J.-H.; Wang, Y.-Y.; Chen, Y.-Y.; Chen, Z.-S.; Ma, L.; Chen, J.; 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. [Google Scholar] [CrossRef] [PubMed]
  66. Klein, C.A. Parallel progression of primary tumours and metastases. Nat. Rev. Cancer 2009, 9, 302–312. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, J.; Cao, S.-W.; 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. Sect. A Dis. Markers 2017, 20, 487–498. [Google Scholar] [CrossRef] [PubMed]
  68. Qi, L.-N.; Ma, L.; Chen, Y.-Y.; Chen, Z.-S.; Zhong, J.-H.; Gong, W.-F.; Lu, Y.; Xiang, B.-D.; Li, L.-Q. Outcomes of anatomical versus non-anatomical resection for hepatocellular carcinoma according to circulating tumour-cell status. Ann. Med. 2020, 52, 21–31. [Google Scholar] [CrossRef] [PubMed]
  69. Zhou, K.-Q.; Sun, Y.-F.; Cheng, J.-W.; Du, M.; Ji, Y.; Wang, P.-X.; Hu, B.; Guo, W.; Gao, Y.; Yin, Y.; et al. Effect of surgical margin on recurrence based on preoperative circulating tumor cell status in hepatocellular carcinoma. EBioMedicine 2020, 62, 103107. [Google Scholar] [CrossRef]
  70. Guo, W.; Sun, Y.-F.; Shen, M.-N.; Ma, X.-L.; Wu, J.; Zhang, C.-Y.; Zhou, Y.; Xu, Y.; Hu, B.; Zhang, M.; 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. [Google Scholar] [CrossRef]
  71. Sun, Y.-F.; Wang, P.-X.; Cheng, J.-W.; Gong, Z.; Huang, A.; Zhou, K.; Hu, B.; Gao, P.; Cao, Y.; Qiu, S.; et al. Postoperative circulating tumor cells: An early predictor of extrahepatic metastases in patients with hepatocellular carcinoma undergoing curative surgical resection. Cancer Cytopathol. 2020, 128, 733–745. [Google Scholar] [CrossRef] [PubMed]
  72. Ahn, J.C.; Teng, P.-C.; Chen, P.-J.; Posadas, E.; Tseng, H.R.; Lu, S.C.; Yang, J.D. 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. [Google Scholar] [CrossRef] [PubMed]
  73. Sun, Y.-F.; Wu, L.; Liu, S.-P.; Jiang, M.-M.; Hu, B.; Zhou, K.-Q.; Guo, W.; Xu, Y.; Zhong, Y.; Zhou, X.-R.; 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. [Google Scholar] [CrossRef] [PubMed]
  74. Chan, L.-K.; Tsui, Y.-M.; Ho, D.W.-H.; Ng, I.O.-L. Cellular heterogeneity and plasticity in liver cancer. Semin. Cancer Biol. 2022, 82, 134–149. [Google Scholar] [CrossRef] [PubMed]
  75. Lui, Y.Y.N.; Chik, K.-W.; Chiu, R.W.K.; Ho, C.-Y.; Lam, C.W.K.; Lo, Y.M.D. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin. Chem. 2002, 48, 421–427. [Google Scholar] [CrossRef] [PubMed]
  76. Sun, K.; Jiang, P.; Chan, K.C.A.; Wong, J.; Cheng, Y.K.Y.; Liang, R.H.S.; Chan, W.-K.; Ma, E.S.K.; Chan, S.L.; Cheng, S.H.; et al. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc. Natl. Acad. Sci. USA 2015, 112, E5503–E5512. [Google Scholar] [CrossRef] [PubMed]
  77. Thierry, A.R.; El Messaoudi, S.; Gahan, P.B.; Anker, P.; Stroun, M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef] [PubMed]
  78. Luo, H.; Wei, W.; Ye, Z.; Zheng, J.; Xu, R.-H. Liquid Biopsy of Methylation Biomarkers in Cell-Free DNA. Trends Mol. Med. 2021, 27, 482–500. [Google Scholar] [CrossRef] [PubMed]
  79. Szilágyi, M.; Pös, O.; Márton, É.; Buglyó, G.; Soltész, B.; Keserű, J.; Penyige, A.; Szemes, T.; Nagy, B. Circulating Cell-Free Nucleic Acids: Main Characteristics and Clinical Application. Int. J. Mol. Sci. 2020, 21, 6827. [Google Scholar] [CrossRef]
  80. Lo, Y.M.D.; Han, D.S.C.; Jiang, P.; Chiu, R.W.K. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science 2021, 372, eaaw3616. [Google Scholar] [CrossRef]
  81. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra24. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, Y.; Liu, Z.; Ji, K.; Li, X.; Wang, C.; Ren, Z.; Liu, Y.; Chen, X.; Han, X.; Meng, L.; et al. Clinical Application Value of Circulating Cell-free DNA in Hepatocellular Carcinoma. Front. Mol. Biosci. 2021, 8, 736330. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, K.; Zhang, H.; Zhang, L.-N.; Ju, S.-Q.; Qi, J.; Huang, D.-F.; Li, F.; Wei, Q.; Zhang, J. Value of circulating cell-free DNA in diagnosis of hepatocelluar carcinoma. World J. Gastroenterol. 2013, 19, 3143–3149. [Google Scholar] [CrossRef]
  84. Park, S.; Lee, E.J.; Rim, C.H.; Seong, J. Plasma Cell-Free DNA as a Predictive Marker after Radiotherapy for Hepatocellular Carcinoma. Yonsei Med. J. 2018, 59, 470–479. [Google Scholar] [CrossRef]
  85. Tokuhisa, Y.; Iizuka, N.; Sakaida, I.; Moribe, T.; Fujita, N.; Miura, T.; Tamatsukuri, S.; Ishitsuka, H.; Uchida, K.; Terai, S.; 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. [Google Scholar] [CrossRef]
  86. Huang, A.; Zhang, X.; Zhou, S.-L.; Cao, Y.; Huang, X.-W.; Fan, J.; Yang, X.-R.; Zhou, J. Plasma Circulating Cell-free DNA Integrity as a Promising Biomarker for Diagnosis and Surveillance in Patients with Hepatocellular Carcinoma. J. Cancer 2016, 7, 1798–1803. [Google Scholar] [CrossRef] [PubMed]
  87. Jiang, P.; Chan, C.W.M.; Chan, K.C.A.; Cheng, S.H.; Wong, J.; Wong, V.W.-S.; Wong, G.L.H.; Chan, S.L.; Mok, T.S.K.; Chan, H.L.Y.; et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, E1317–E1325. [Google Scholar] [CrossRef]
  88. Ono, A.; Fujimoto, A.; Yamamoto, Y.; Akamatsu, S.; Hiraga, N.; Imamura, M.; Kawaoka, T.; Tsuge, M.; Abe, H.; Hayes, C.N.; et al. Circulating Tumor DNA Analysis for Liver Cancers and Its Usefulness as a Liquid Biopsy. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 516–534. [Google Scholar] [CrossRef]
  89. Chen, G.; Cai, Z.; Li, Z.; Dong, X.; Xu, H.; Lin, J.; Chen, L.; Zhang, H.; Liu, X.; Liu, J. Clonal evolution in long-term follow-up patients with hepatocellular carcinoma. Int. J. Cancer 2018, 143, 2862–2870. [Google Scholar] [CrossRef]
  90. Zhu, G.-Q.; Liu, W.-R.; Tang, Z.; Qu, W.-F.; Fang, Y.; Jiang, X.-F.; Song, S.-S.; Wang, H.; Tao, C.-Y.; Zhou, P.-Y.; et al. Serial circulating tumor DNA to predict early recurrence in patients with hepatocellular carcinoma: A prospective study. Mol. Oncol. 2022, 16, 549–561. [Google Scholar] [CrossRef]
  91. Huang, A.; Zhao, X.; Yang, X.-R.; Li, F.-Q.; Zhou, X.-L.; Wu, K.; Zhang, X.; Sun, Q.-M.; Cao, Y.; Zhu, H.-M.; et al. Circumventing intratumoral heterogeneity to identify potential therapeutic targets in hepatocellular carcinoma. J. Hepatol. 2017, 67, 293–301. [Google Scholar] [CrossRef]
  92. Ng, C.K.Y.; Di Costanzo, G.G.; Tosti, N.; Paradiso, V.; Coto-Llerena, M.; Roscigno, G.; Perrina, V.; Quintavalle, C.; Boldanova, T.; Wieland, S.; et al. Genetic profiling using plasma-derived cell-free DNA in therapy-naïve hepatocellular carcinoma patients: A pilot study. Ann. Oncol. 2018, 29, 1286–1291. [Google Scholar] [CrossRef] [PubMed]
  93. Labgaa, I.; Villacorta-Martin, C.; D’Avola, D.; Craig, A.J.; von Felden, J.; Martins-Filho, S.N.; Sia, D.; Stueck, A.; Ward, S.C.; Fiel, M.I.; et al. A pilot study of ultra-deep targeted sequencing of plasma DNA identifies driver mutations in hepatocellular carcinoma. Oncogene 2018, 37, 3740–3752. [Google Scholar] [CrossRef]
  94. Howell, J.; Atkinson, S.R.; Pinato, D.J.; Knapp, S.; Ward, C.; Minisini, R.; Burlone, M.E.; Leutner, M.; Pirisi, M.; Büttner, R.; et al. Identification of mutations in circulating cell-free tumour DNA as a biomarker in hepatocellular carcinoma. Eur. J. Cancer 2019, 116, 56–66. [Google Scholar] [CrossRef] [PubMed]
  95. Diaz, L.A.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef] [PubMed]
  96. von Felden, J.; Craig, A.J.; Garcia-Lezana, T.; Labgaa, I.; Haber, P.K.; D’Avola, D.; Asgharpour, A.; Dieterich, D.; Bonaccorso, A.; Torres-Martin, M.; et al. Mutations in circulating tumor DNA predict primary resistance to systemic therapies in advanced hepatocellular carcinoma. Oncogene 2021, 40, 140–151. [Google Scholar] [CrossRef] [PubMed]
  97. Xu, R.-H.; Wei, W.; Krawczyk, M.; Wang, W.; Luo, H.; Flagg, K.; Yi, S.; Shi, W.; Quan, Q.; Li, K.; et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat. Mater. 2017, 16, 1155–1161. [Google Scholar] [CrossRef]
  98. Qu, C.; Wang, Y.; Wang, P.; Chen, K.; Wang, M.; Zeng, H.; Lu, J.; Song, Q.; Diplas, B.H.; Tan, D.; et al. Detection of early-stage hepatocellular carcinoma in asymptomatic HBsAg-seropositive individuals by liquid biopsy. Proc. Natl. Acad. Sci. USA 2019, 116, 6308–6312. [Google Scholar] [CrossRef]
  99. Li, Y.; Zheng, Y.; Wu, L.; Li, J.; Ji, J.; Yu, Q.; Dai, W.; Feng, J.; Wu, J.; Guo, C. Current status of ctDNA in precision oncology for hepatocellular carcinoma. J. Exp. Clin. Cancer Res. CR 2021, 40, 140. [Google Scholar] [CrossRef]
  100. Lyu, X.; Tsui, Y.-M.; Ho, D.W.-H.; Ng, I.O.-L. Liquid Biopsy Using Cell-Free or Circulating Tumor DNA in the Management of Hepatocellular Carcinoma. Cell. Mol. Gastroenterol. Hepatol. 2022, 13, 1611–1624. [Google Scholar] [CrossRef]
  101. Labgaa, I.; Villanueva, A.; Dormond, O.; Demartines, N.; Melloul, E. The Role of Liquid Biopsy in Hepatocellular Carcinoma Prognostication. Cancers 2021, 13, 659. [Google Scholar] [CrossRef]
  102. Chen, K.; Rajewsky, N. The evolution of gene regulation by transcription factors and microRNAs. Nat. Rev. Genet. 2007, 8, 93–103. [Google Scholar] [CrossRef] [PubMed]
  103. Correia de Sousa, M.; Gjorgjieva, M.; Dolicka, D.; Sobolewski, C.; Foti, M. Deciphering miRNAs’ Action through miRNA Editing. Int. J. Mol. Sci. 2019, 20, 6249. [Google Scholar] [CrossRef]
  104. Yang, N.; Ekanem, N.R.; Sakyi, C.A.; Ray, S.D. Hepatocellular carcinoma and microRNA: New perspectives on therapeutics and diagnostics. Adv. Drug Deliv. Rev. 2015, 81, 62–74. [Google Scholar] [CrossRef]
  105. Calin, G.A.; Croce, C.M. MicroRNA signatures in human cancers. Nat. Rev. Cancer 2006, 6, 857–866. [Google Scholar] [CrossRef]
  106. Turchinovich, A.; Weiz, L.; Burwinkel, B. Extracellular miRNAs: The mystery of their origin and function. Trends Biochem. Sci. 2012, 37, 460–465. [Google Scholar] [CrossRef] [PubMed]
  107. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, J.; Li, J.; Shen, J.; Wang, C.; Yang, L.; Zhang, X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer 2012, 12, 227. [Google Scholar] [CrossRef]
  109. Qin, J.; Luo, M.; Qian, H.; Chen, W. Upregulated miR-182 increases drug resistance in cisplatin-treated HCC cell by regulating TP53INP1. Gene 2014, 538, 342–347. [Google Scholar] [CrossRef]
  110. Cao, M.-Q.; You, A.-B.; Zhu, X.-D.; Zhang, W.; Zhang, Y.-Y.; Zhang, S.-Z.; Zhang, K.-W.; Cai, H.; Shi, W.-K.; Li, X.-L.; et al. miR-182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J. Hematol. Oncol. 2018, 11, 12. [Google Scholar] [CrossRef]
  111. Lai, C.-Y.; Yeh, K.-Y.; Lin, C.-Y.; Hsieh, Y.-W.; Lai, H.-H.; Chen, J.-R.; Hsu, C.-C.; Her, G.M. MicroRNA-21 Plays Multiple Oncometabolic Roles in the Process of NAFLD-Related Hepatocellular Carcinoma via PI3K/AKT, TGF-β, and STAT3 Signaling. Cancers 2021, 13, 940. [Google Scholar] [CrossRef] [PubMed]
  112. Lei, Z.; Tang, X.; Si, A.; Yang, P.; Wang, L.; Luo, T.; Guo, G.; Zhang, Q.; Cheng, Z. microRNA-454 promotes liver tumor-initiating cell expansion by regulating SOCS6. Exp. Cell Res. 2020, 390, 111955. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Y.; Zhang, C.; Zhao, Q.; Wei, W.; Dong, Z.; Shao, L.; Li, J.; Wu, W.; Zhang, H.; Huang, H.; et al. The miR-873/NDFIP1 axis promotes hepatocellular carcinoma growth and metastasis through the AKT/mTOR-mediated Warburg effect. Am. J. Cancer Res. 2019, 9, 927–944. [Google Scholar]
  114. Wang, X.; Lu, J.; Cao, J.; Ma, B.; Gao, C.; Qi, F. MicroRNA-18a promotes hepatocellular carcinoma proliferation, migration, and invasion by targeting Bcl2L10. OncoTargets Ther. 2018, 11, 7919–7934. [Google Scholar] [CrossRef] [PubMed]
  115. Liao, Y.; Wang, C.; Yang, Z.; Liu, W.; Yuan, Y.; Li, K.; Zhang, Y.; Wang, Y.; Shi, Y.; Qiu, Y.; et al. Dysregulated Sp1/miR-130b-3p/HOXA5 axis contributes to tumor angiogenesis and progression of hepatocellular carcinoma. Theranostics 2020, 10, 5209–5224. [Google Scholar] [CrossRef] [PubMed]
  116. Li, D.; Wang, T.; Sun, F.-F.; Feng, J.-Q.; Peng, J.-J.; Li, H.; Wang, C.; Wang, D.; Liu, Y.; Bai, Y.-D.; et al. MicroRNA-375 represses tumor angiogenesis and reverses resistance to sorafenib in hepatocarcinoma. Cancer Gene Ther. 2021, 28, 126–140. [Google Scholar] [CrossRef] [PubMed]
  117. Feng, X.; Zou, B.; Nan, T.; Zheng, X.; Zheng, L.; Lan, J.; Chen, W.; Yu, J. MiR-25 enhances autophagy and promotes sorafenib resistance of hepatocellular carcinoma via targeting FBXW7. Int. J. Med. Sci. 2022, 19, 257–266. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, S.; Fu, Z.; Wen, S.; Yang, X.; Yu, C.; Zhou, W.; Lin, Y.; Lv, Y. Expression and Diagnostic Value of miR-497 and miR-1246 in Hepatocellular Carcinoma. Front. Genet. 2021, 12, 666306. [Google Scholar] [CrossRef] [PubMed]
  119. Shi, D.-M.; Li, L.-X.; Bian, X.-Y.; Shi, X.-J.; Lu, L.-L.; Zhou, H.-X.; Pan, T.-J.; Zhou, J.; Fan, J.; Wu, W.-Z. miR-296-5p suppresses EMT of hepatocellular carcinoma via attenuating NRG1/ERBB2/ERBB3 signaling. J. Exp. Clin. Cancer Res. CR 2018, 37, 294. [Google Scholar] [CrossRef]
  120. Shi, D.-M.; Shi, X.-L.; Xing, K.-L.; Zhou, H.-X.; Lu, L.-L.; Wu, W.-Z. miR-296-5p suppresses stem cell potency of hepatocellular carcinoma cells via regulating Brg1/Sall4 axis. Cell. Signal. 2020, 72, 109650. [Google Scholar] [CrossRef]
  121. Wang, Y.; Tai, Q.; Zhang, J.; Kang, J.; Gao, F.; Zhong, F.; Cai, L.; Fang, F.; Gao, Y. MiRNA-206 inhibits hepatocellular carcinoma cell proliferation and migration but promotes apoptosis by modulating cMET expression. Acta Biochim. Biophys. Sin. 2019, 51, 243–253. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, X.; Chen, Y.; Dong, K.; Ma, Y.; Jin, Q.; Yin, S.; Zhu, X.; Wang, S. Effects of FER1L4-miR-106a-5p/miR-372-5p-E2F1 regulatory axis on drug resistance in liver cancer chemotherapy. Mol. Ther. Nucleic Acids 2021, 24, 449–461. [Google Scholar] [CrossRef] [PubMed]
  123. Du, H.; Xu, Q.; Xiao, S.; Wu, Z.; Gong, J.; Liu, C.; Ren, G.; Wu, H. MicroRNA-424-5p acts as a potential biomarker and inhibits proliferation and invasion in hepatocellular carcinoma by targeting TRIM29. Life Sci. 2019, 224, 1–11. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, P.; Ha, M.; Li, L.; Huang, X.; Liu, C. MicroRNA-3064-5p sponged by MALAT1 suppresses angiogenesis in human hepatocellular carcinoma by targeting the FOXA1/CD24/Src pathway. FASEB J. 2020, 34, 66–81. [Google Scholar] [CrossRef] [PubMed]
  125. Shao, Y.; Zhang, D.; Li, X.; Yang, J.; Chen, L.; Ning, Z.; Xu, Y.; Deng, G.; Tao, M.; Zhu, Y.; et al. MicroRNA-203 Increases Cell Radiosensitivity via Directly Targeting Bmi-1 in Hepatocellular Carcinoma. Mol. Pharm. 2018, 15, 3205–3215. [Google Scholar] [CrossRef] [PubMed]
  126. Luo, J.; Si, Z.-Z.; Li, T.; Li, J.-Q.; Zhang, Z.-Q.; Chen, G.-S.; Qi, H.-Z.; Yao, H.-L. MicroRNA-146a-5p enhances radiosensitivity in hepatocellular carcinoma through replication protein A3-induced activation of the DNA repair pathway. Am. J. Physiol. Cell Physiol. 2019, 316, C299–C311. [Google Scholar] [CrossRef]
  127. Shao, Y.; Song, X.; Jiang, W.; Chen, Y.; Ning, Z.; Gu, W.; Jiang, J. MicroRNA-621 Acts as a Tumor Radiosensitizer by Directly Targeting SETDB1 in Hepatocellular Carcinoma. Mol. Ther. J. Am. Soc. Gene Ther. 2019, 27, 355–364. [Google Scholar] [CrossRef]
  128. Ke, M.; Zhang, Z.; Cong, L.; Zhao, S.; Li, Y.; Wang, X.; Lv, Y.; Zhu, Y.; Dong, J. MicroRNA-148b-colony-stimulating factor-1 signaling-induced tumor-associated macrophage infiltration promotes hepatocellular carcinoma metastasis. Biomed. Pharmacother. 2019, 120, 109523. [Google Scholar] [CrossRef]
  129. Zhao, J.; Li, H.; Zhao, S.; Wang, E.; Zhu, J.; Feng, D.; Zhu, Y.; Dou, W.; Fan, Q.; Hu, J.; et al. Epigenetic silencing of miR-144/451a cluster contributes to HCC progression via paracrine HGF/MIF-mediated TAM remodeling. Mol. Cancer 2021, 20, 46. [Google Scholar] [CrossRef]
  130. Lu, T.X.; Rothenberg, M.E. MicroRNA. J. Allergy Clin. Immunol. 2018, 141, 1202–1207. [Google Scholar] [CrossRef]
  131. Loosen, S.H.; Castoldi, M.; Jördens, M.S.; Roy, S.; Vucur, M.; Kandler, J.; Hammerich, L.; Mohr, R.; Tacke, F.; Ulmer, T.F.; et al. Serum levels of circulating microRNA-107 are elevated in patients with early-stage HCC. PLoS ONE 2021, 16, e0247917. [Google Scholar] [CrossRef]
  132. Hung, C.-H.; Hu, T.-H.; Lu, S.-N.; Kuo, F.-Y.; Chen, C.-H.; Wang, J.-H.; Huang, C.-M.; Lee, C.-M.; Lin, C.-Y.; Yen, Y.-H.; et al. Circulating microRNAs as biomarkers for diagnosis of early hepatocellular carcinoma associated with hepatitis B virus. Int. J. Cancer 2016, 138, 714–720. [Google Scholar] [CrossRef] [PubMed]
  133. Han, J.; Li, J.; Qian, Y.; Liu, W.; Liang, J.; Huang, Z.; Wang, S.; Zhao, C. Identification of plasma miR-148a as a noninvasive biomarker for hepatocellular carcinoma. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 585–593. [Google Scholar] [CrossRef]
  134. Ladeiro, Y.; Couchy, G.; Balabaud, C.; Bioulac-Sage, P.; Pelletier, L.; Rebouissou, S.; Zucman-Rossi, J. MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology 2008, 47, 1955–1963. [Google Scholar] [CrossRef] [PubMed]
  135. Dabbish, A.M.; Abdelzaher, H.M.; Abohawya, M.; Shamma, S.; Mahmoud, Y.H.; Maged, A.; Manaa, M.; Hassany, M.; Kobeissy, F.; Bazgir, O.; et al. Prognostic MicroRNA Panel for HCV-Associated HCC: Integrating Computational Biology and Clinical Validation. Cancers 2022, 14, 3036. [Google Scholar] [CrossRef]
  136. Canale, M.; Foschi, F.G.; Andreone, P.; Ercolani, G.; Marisi, G.; Conti, F.; Vukotic, R.; Guarneri, V.; Burgio, V.; Ratti, F.; et al. Role of circulating microRNAs to predict hepatocellular carcinoma recurrence in patients treated with radiofrequency ablation or surgery. HPB 2022, 24, 244–254. [Google Scholar] [CrossRef] [PubMed]
  137. Andrasina, T.; Juracek, J.; Zavadil, J.; Cechova, B.; Rohan, T.; Vesela, P.; Paldor, M.; Slaby, O.; Goldberg, S.N. Thermal Ablation and Transarterial Chemoembolization are Characterized by Changing Dynamics of Circulating MicroRNAs. J. Vasc. Interv. Radiol. 2021, 32, 403–411. [Google Scholar] [CrossRef]
  138. Zhang, S.; Zhou, Y.; Wang, Y.; Wang, Z.; Xiao, Q.; Zhang, Y.; Lou, Y.; Qiu, Y.; Zhu, F. The mechanistic, diagnostic and therapeutic novel nucleic acids for hepatocellular carcinoma emerging in past score years. Brief. Bioinform. 2021, 22, 1860–1883. [Google Scholar] [CrossRef]
  139. Young, D.D.; Connelly, C.M.; Grohmann, C.; Deiters, A. Small molecule modifiers of microRNA miR-122 function for the treatment of hepatitis C virus infection and hepatocellular carcinoma. J. Am. Chem. Soc. 2010, 132, 7976–7981. [Google Scholar] [CrossRef]
  140. Wang, W.; Zhao, L.-J.; Tan, Y.-X.; Ren, H.; Qi, Z.-T. MiR-138 induces cell cycle arrest by targeting cyclin D3 in hepatocellular carcinoma. Carcinogenesis 2012, 33, 1113–1120. [Google Scholar] [CrossRef]
  141. Xiao, Z.; Li, C.H.; Chan, S.L.; Xu, F.; Feng, L.; Wang, Y.; Jiang, J.-D.; Sung, J.J.Y.; Cheng, C.H.K.; Chen, Y. A small-molecule modulator of the tumor-suppressor miR34a inhibits the growth of hepatocellular carcinoma. Cancer Res. 2014, 74, 6236–6247. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, B.; Wang, X.; Deng, J.; Zheng, H.; Liu, W.; Chen, S.; Tian, J.; Wang, F. p53-dependent upregulation of miR-16-2 by sanguinarine induces cell cycle arrest and apoptosis in hepatocellular carcinoma. Cancer Lett. 2019, 459, 50–58. [Google Scholar] [CrossRef] [PubMed]
  143. Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
  144. Poutsiaka, D.D.; Schroder, E.W.; Taylor, D.D.; Levy, E.M.; Black, P.H. Membrane vesicles shed by murine melanoma cells selectively inhibit the expression of Ia antigen by macrophages. J. Immunol. 1985, 134, 138–144. [Google Scholar] [CrossRef] [PubMed]
  145. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. [Google Scholar] [CrossRef]
  146. Thietart, S.; Rautou, P.-E. Extracellular vesicles as biomarkers in liver diseases: A clinician’s point of view. J. Hepatol. 2020, 73, 1507–1525. [Google Scholar] [CrossRef] [PubMed]
  147. Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
  148. Wang, H.; Lu, Z.; Zhao, X. Tumorigenesis, diagnosis, and therapeutic potential of exosomes in liver cancer. J. Hematol. Oncol. 2019, 12, 133. [Google Scholar] [CrossRef]
  149. Li, R.; Wang, Y.; Zhang, X.; Feng, M.; Ma, J.; Li, J.; Yang, X.; Fang, F.; Xia, Q.; Zhang, Z.; et al. Exosome-mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. Mol. Cancer 2019, 18, 18. [Google Scholar] [CrossRef]
  150. He, M.; Qin, H.; Poon, T.C.W.; Sze, S.-C.; Ding, X.; Co, N.N.; Ngai, S.-M.; Chan, T.-F.; Wong, N. Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis 2015, 36, 1008–1018. [Google Scholar] [CrossRef]
  151. Liu, B.H.M.; Tey, S.K.; Mao, X.; Ma, A.P.Y.; Yeung, C.L.S.; Wong, S.W.K.; Ng, T.H.; Xu, Y.; Yao, Y.; Fung, E.Y.M.; et al. TPI1-reduced extracellular vesicles mediated by Rab20 downregulation promotes aerobic glycolysis to drive hepatocarcinogenesis. J. Extracell. Vesicles 2021, 10, e12135. [Google Scholar] [CrossRef] [PubMed]
  152. Cokakli, M.; Erdal, E.; Nart, D.; Yilmaz, F.; Sagol, O.; Kilic, M.; Karademir, S.; Atabey, N. Differential expression of Caveolin-1 in hepatocellular carcinoma: Correlation with differentiation state, motility and invasion. BMC Cancer 2009, 9, 65. [Google Scholar] [CrossRef] [PubMed]
  153. Jiang, X.; Wu, S.; Hu, C. A narrative review of the role of exosomes and caveolin-1 in liver diseases and cancer. Int. Immunopharmacol. 2023, 120, 110284. [Google Scholar] [CrossRef] [PubMed]
  154. Hou, P.-P.; Luo, L.-J.; Chen, H.-Z.; Chen, Q.-T.; Bian, X.-L.; Wu, S.-F.; Zhou, J.-X.; Zhao, W.-X.; Liu, J.-M.; Wang, X.-M.; et al. Ectosomal PKM2 Promotes HCC by Inducing Macrophage Differentiation and Remodeling the Tumor Microenvironment. Mol. Cell 2020, 78, 1192–1206.e10. [Google Scholar] [CrossRef] [PubMed]
  155. Jiang, K.; Dong, C.; Yin, Z.; Li, R.; Mao, J.; Wang, C.; Zhang, J.; Gao, Z.; Liang, R.; Wang, Q.; et al. Exosome-derived ENO1 regulates integrin α6β4 expression and promotes hepatocellular carcinoma growth and metastasis. Cell Death Dis. 2020, 11, 972. [Google Scholar] [CrossRef]
  156. Cheng, Z.; Lei, Z.; Yang, P.; Si, A.; Xiang, D.; Tang, X.; Guo, G.; Zhou, J.; Hüser, N. Exosome-transmitted p120-catenin suppresses hepatocellular carcinoma progression via STAT3 pathways. Mol. Carcinog. 2019, 58, 1389–1399. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, X.; Shen, H.; Zhangyuan, G.; Huang, R.; Zhang, W.; He, Q.; Jin, K.; Zhuo, H.; Zhang, Z.; Wang, J.; et al. 14-3-3ζ delivered by hepatocellular carcinoma-derived exosomes impaired anti-tumor function of tumor-infiltrating T lymphocytes. Cell Death Dis. 2018, 9, 159. [Google Scholar] [CrossRef] [PubMed]
  158. Mao, X.; Zhou, L.; Tey, S.K.; Ma, A.P.Y.; Yeung, C.L.S.; Ng, T.H.; Wong, S.W.K.; Liu, B.H.M.; Fung, Y.M.E.; Patz, E.F.; et al. Tumour extracellular vesicle-derived Complement Factor H promotes tumorigenesis and metastasis by inhibiting complement-dependent cytotoxicity of tumour cells. J. Extracell. Vesicles 2020, 10, e12031. [Google Scholar] [CrossRef]
  159. Wang, G.; Liu, W.; Zou, Y.; Wang, G.; Deng, Y.; Luo, J.; Zhang, Y.; Li, H.; Zhang, Q.; Yang, Y.; et al. Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine 2019, 40, 432–445. [Google Scholar] [CrossRef]
  160. Huang, X.-Y.; Huang, Z.-L.; Huang, J.; Xu, B.; Huang, X.-Y.; Xu, Y.-H.; Zhou, J.; Tang, Z.-Y. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. J. Exp. Clin. Cancer Res. CR 2020, 39, 20. [Google Scholar] [CrossRef]
  161. Li, B.; Mao, R.; Liu, C.; Zhang, W.; Tang, Y.; Guo, Z. LncRNA FAL1 promotes cell proliferation and migration by acting as a CeRNA of miR-1236 in hepatocellular carcinoma cells. Life Sci. 2018, 197, 122–129. [Google Scholar] [CrossRef]
  162. Li, J.; Xue, J.; Ling, M.; Sun, J.; Xiao, T.; Dai, X.; Sun, Q.; Cheng, C.; Xia, H.; Wei, Y.; et al. MicroRNA-15b in extracellular vesicles from arsenite-treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1-mediated Hippo pathway. Cancer Lett. 2021, 497, 137–153. [Google Scholar] [CrossRef] [PubMed]
  163. Fang, J.-H.; Zhang, Z.-J.; Shang, L.-R.; Luo, Y.W.; Lin, Y.F.; Yuan, Y.; Zhuang, S.M. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018, 68, 1459–1475. [Google Scholar] [CrossRef] [PubMed]
  164. Shao, Z.; Pan, Q.; Zhang, Y. Hepatocellular carcinoma cell-derived extracellular vesicles encapsulated microRNA-584-5p facilitates angiogenesis through PCK1-mediated nuclear factor E2-related factor 2 signaling pathway. Int. J. Biochem. Cell Biol. 2020, 125, 105789. [Google Scholar] [CrossRef] [PubMed]
  165. Lin, X.-J.; Fang, J.-H.; Yang, X.-J.; Zhang, C.; Yuan, Y.; Zheng, L.; Zhuang, S.M. Hepatocellular Carcinoma Cell-Secreted Exosomal MicroRNA-210 Promotes Angiogenesis In Vitro and In Vivo. Mol. Ther. Nucleic Acids 2018, 11, 243–252. [Google Scholar] [CrossRef] [PubMed]
  166. Tian, X.-P.; Wang, C.-Y.; Jin, X.-H.; Li, M.; Wang, F.-W.; Huang, W.-J.; Yun, J.-P.; Xu, R.-H.; Cai, Q.-Q.; Xie, D. Acidic Microenvironment Up-Regulates Exosomal miR-21 and miR-10b in Early-Stage Hepatocellular Carcinoma to Promote Cancer Cell Proliferation and Metastasis. Theranostics 2019, 9, 1965–1979. [Google Scholar] [CrossRef] [PubMed]
  167. Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. CR 2018, 37, 324. [Google Scholar] [CrossRef] [PubMed]
  168. Fang, T.; Lv, H.; Lv, G.; Li, T.; Wang, C.; Han, Q.; Yu, L.; Su, B.; Guo, L.; Huang, S.; et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat. Commun. 2018, 9, 191. [Google Scholar] [CrossRef] [PubMed]
  169. Nakano, T.; Chen, I.-H.; Wang, C.-C.; Chen, P.-J.; Tseng, H.-P.; Huang, K.-T.; Hu, T.-H.; Li, L.-C.; Goto, S.; Cheng, Y.-F.; 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. [Google Scholar] [CrossRef]
  170. Liu, J.; Fan, L.; Yu, H.; Zhang, J.; He, Y.; Feng, D.; Wang, F.; Li, X.; Liu, Q.; Li, Y.; et al. Endoplasmic Reticulum Stress Causes Liver Cancer Cells to Release Exosomal miR-23a-3p and Up-regulate Programmed Death Ligand 1 Expression in Macrophages. J. Hepatol. 2019, 70, 241–258. [Google Scholar] [CrossRef]
  171. Kimiz-Gebologlu, I.; Oncel, S.S. Exosomes: Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J. Control. Release 2022, 347, 533–543. [Google Scholar] [CrossRef] [PubMed]
  172. Li, X.; Li, C.; Zhang, L.; Wu, M.; Cao, K.; Jiang, F.; Chen, D.; Li, N.; Li, W. The significance of exosomes in the development and treatment of hepatocellular carcinoma. Mol. Cancer 2020, 19, 1. [Google Scholar] [CrossRef] [PubMed]
  173. Xu, H.; Dong, X.; Chen, Y.; Wang, X. Serum exosomal hnRNPH1 mRNA as a novel marker for hepatocellular carcinoma. Clin. Chem. Lab. Med. 2018, 56, 479–484. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, C.; Yang, X.; Qi, Q.; Gao, Y.; Wei, Q.; Han, S. lncRNA-HEIH in serum and exosomes as a potential biomarker in the HCV-related hepatocellular carcinoma. Cancer Biomark. Sect. A Dis. Markers 2018, 21, 651–659. [Google Scholar] [CrossRef] [PubMed]
  175. Cho, H.J.; Eun, J.W.; Baek, G.O.; Seo, C.W.; Ahn, H.R.; Kim, S.S.; Cho, S.W.; Cheong, J.Y. Serum Exosomal MicroRNA, miR-10b-5p, as a Potential Diagnostic Biomarker for Early-Stage Hepatocellular Carcinoma. J. Clin. Med. 2020, 9, 281. [Google Scholar] [CrossRef]
  176. Liu, W.; Hu, J.; Zhou, K.; Chen, F.; Wang, Z.; Liao, B.; Dai, Z.; Cao, Y.; Fan, J.; Zhou, J. Serum exosomal miR-125b is a novel prognostic marker for hepatocellular carcinoma. OncoTargets Ther. 2017, 10, 3843–3851. [Google Scholar] [CrossRef] [PubMed]
  177. Suehiro, T.; Miyaaki, H.; Kanda, Y.; Shibata, H.; Honda, T.; Ozawa, E.; Miuma, S.; Taura, N.; Nakao, K. Serum exosomal microRNA-122 and microRNA-21 as predictive biomarkers in transarterial chemoembolization-treated hepatocellular carcinoma patients. Oncol. Lett. 2018, 16, 3267–3273. [Google Scholar] [CrossRef]
  178. Hu, Z.; Chen, G.; Zhao, Y.; Gao, H.; Li, L.; Yin, Y.; Jiang, J.; Wang, L.; Mang, Y.; Gao, Y.; et al. Exosome-derived circCCAR1 promotes CD8+ T-cell dysfunction and anti-PD1 resistance in hepatocellular carcinoma. Mol. Cancer 2023, 22, 55. [Google Scholar] [CrossRef] [PubMed]
  179. Lee, Y.R.; Kim, G.; Tak, W.Y.; Jang, S.Y.; Kweon, Y.O.; Gil Park, J.; Lee, H.W.; Han, Y.S.; Chun, J.M.; Park, S.Y.; et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int. J. Cancer 2019, 144, 1444–1452. [Google Scholar] [CrossRef]
  180. Wan, Y.; Wang, L.; Zhu, C.; Zheng, Q.; Wang, G.; Tong, J.; Fang, Y.; Xia, Y.; Cheng, G.; He, X.; et al. Aptamer-Conjugated Extracellular Nanovesicles for Targeted Drug Delivery. Cancer Res. 2018, 78, 798–808. [Google Scholar] [CrossRef]
  181. Bell, B.M.; Kirk, I.D.; Hiltbrunner, S.; Gabrielsson, S.; Bultema, J.J. Designer exosomes as next-generation cancer immunotherapy. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 163–169. [Google Scholar] [CrossRef] [PubMed]
  182. Burgio, S.; Noori, L.; Marino Gammazza, A.; Campanella, C.; Logozzi, M.; Fais, S.; Bucchieri, F.; Cappello, F.; Bavisotto, C.C. Extracellular Vesicles-Based Drug Delivery Systems: A New Challenge and the Exemplum of Malignant Pleural Mesothelioma. Int. J. Mol. Sci. 2020, 21, 5432. [Google Scholar] [CrossRef] [PubMed]
  183. Armstrong, J.P.K.; Stevens, M.M. Strategic design of extracellular vesicle drug delivery systems. Adv. Drug Deliv. Rev. 2018, 130, 12–16. [Google Scholar] [CrossRef] [PubMed]
  184. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [PubMed]
  185. Yuan, D.; Zhao, Y.; Banks, W.A.; Bullock, K.M.; Haney, M.; Batrakova, E.; Kabanov, A.V. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials 2017, 142, 1–12. [Google Scholar] [CrossRef]
  186. Lou, G.; Song, X.; Yang, F.; Wu, S.; Wang, J.; Chen, Z.; Liu, Y. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J. Hematol. Oncol. 2015, 8, 122. [Google Scholar] [CrossRef] [PubMed]
  187. Lou, G.; Chen, L.; Xia, C.; Wang, W.; Qi, J.; Li, A.; Zhao, L.; Chen, Z.; Zheng, M.; Liu, Y. MiR-199a-modified exosomes from adipose tissue-derived mesenchymal stem cells improve hepatocellular carcinoma chemosensitivity through mTOR pathway. J. Exp. Clin. Cancer Res. 2020, 39, 4. [Google Scholar] [CrossRef] [PubMed]
  188. Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, D.; Yao, Y.; He, J.; Zhong, X.; Li, B.; Rao, S.; Yu, H.; He, S.; Feng, X.; Xu, T.; et al. Engineered Cell-Derived Microparticles Bi2Se3/DOX@MPs for Imaging Guided Synergistic Photothermal/Low-Dose Chemotherapy of Cancer. Adv. Sci. 2020, 7, 1901293. [Google Scholar] [CrossRef]
  190. Gu, H.; Yan, C.; Wan, H.; Wu, L.; Liu, J.; Zhu, Z.; Gao, D. Mesenchymal stem cell-derived exosomes block malignant behaviors of hepatocellular carcinoma stem cells through a lncRNA C5orf66-AS1/microRNA-127-3p/DUSP1/ERK axis. Hum. Cell 2021, 34, 1812–1829. [Google Scholar] [CrossRef]
  191. Wang, Y.; Wang, B.; Xiao, S.; Li, Y.; Chen, Q. miR-125a/b inhibits tumor-associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J. Cell. Biochem. 2019, 120, 3046–3055. [Google Scholar] [CrossRef] [PubMed]
  192. Zuo, B.; Zhang, Y.; Zhao, K.; Wu, L.; Qi, H.; Yang, R.; Gao, X.; Geng, M.; Wu, Y.; Jing, R.; et al. Universal immunotherapeutic strategy for hepatocellular carcinoma with exosome vaccines that engage adaptive and innate immune responses. J. Hematol. Oncol. 2022, 15, 46. [Google Scholar] [CrossRef] [PubMed]
  193. Sorop, A.; Constantinescu, D.; Cojocaru, F.; Dinischiotu, A.; Cucu, D.; Dima, S.O. Exosomal microRNAs as Biomarkers and Therapeutic Targets for Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4997. [Google Scholar] [CrossRef]
  194. Słomka, A.; Wang, B.; Mocan, T.; Horhat, A.; Willms, A.G.; Schmidt-Wolf, I.G.H.; Strassburg, C.P.; Gonzalez-Carmona, M.A.; Lukacs-Kornek, V.; Kornek, M.T. Extracellular Vesicles and Circulating Tumour Cells-complementary liquid biopsies or standalone concepts? Theranostics 2022, 12, 5836–5855. [Google Scholar] [CrossRef]
  195. Li, S.; Yao, J.; Xie, M.; Liu, Y.; Zheng, M. Exosomal miRNAs in hepatocellular carcinoma development and clinical responses. J. Hematol. Oncol. 2018, 11, 54. [Google Scholar] [CrossRef]
  196. Koupenova, M.; Clancy, L.; Corkrey, H.A.; Freedman, J.E. Circulating Platelets as Mediators of Immunity, Inflammation, and Thrombosis. Circ. Res. 2018, 122, 337–351. [Google Scholar] [CrossRef]
  197. Nilsson, R.J.A.; Balaj, L.; Hulleman, E.; van Rijn, S.; Pegtel, D.M.; Walraven, M.; Widmark, A.; Gerritsen, W.R.; Verheul, H.M.; Vandertop, W.P.; et al. Blood platelets contain tumor-derived RNA biomarkers. Blood 2011, 118, 3680–3683. [Google Scholar] [CrossRef] [PubMed]
  198. Schlesinger, M. Role of platelets and platelet receptors in cancer metastasis. J. Hematol. Oncol. 2018, 11, 125. [Google Scholar] [CrossRef] [PubMed]
  199. Haemmerle, M.; Stone, R.L.; Menter, D.G.; Afshar-Kharghan, V.; Sood, A.K. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018, 33, 965–983. [Google Scholar] [CrossRef]
  200. Stone, R.L.; Nick, A.M.; McNeish, I.A.; Balkwill, F.; Han, H.D.; Bottsford-Miller, J.; Rupaimoole, R.; Armaiz-Pena, G.N.; Pecot, C.V.; Coward, J.; et al. Paraneoplastic thrombocytosis in ovarian cancer. N. Engl. J. Med. 2012, 366, 610–618. [Google Scholar] [CrossRef]
  201. Kim, M.-S.; Baek, S.H.; Noh, J.J.; Shim, J.I.; Kang, J.H.; Jeong, S.Y.; Choi, C.H.; Kim, T.-J.; Lee, J.-W.; Lee, Y.-Y. Role of reactive thrombocytosis after primary cytoreductive surgery in advanced ovarian cancer. Front. Oncol. 2022, 12, 926878. [Google Scholar] [CrossRef]
  202. Josa, V.; Krzystanek, M.; Eklund, A.C.; Salamon, F.; Zarand, A.; Szallasi, Z.; Baranyai, Z. Relationship of postoperative thrombocytosis and survival of patients with colorectal cancer. Int. J. Surg. 2015, 18, 1–6. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, Y.-H.; Kang, J.-K.; Zhi, Y.-F.; Zhang, Y.; Wang, Z.-Q.; Zhou, Q.; Niu, W.-Y.; Ma, M.-J. The pretreatment thrombocytosis as one of prognostic factors for gastric cancer: A systematic review and meta-analysis. Int. J. Surg. 2018, 53, 304–311. [Google Scholar] [CrossRef]
  204. Bailey, S.E.; Ukoumunne, O.C.; Shephard, E.A.; Hamilton, W. Clinical relevance of thrombocytosis in primary care: A prospective cohort study of cancer incidence using English electronic medical records and cancer registry data. Br. J. Gen. Pract. J. R. Coll. Gen. Pract. 2017, 67, e405–e413. [Google Scholar] [CrossRef]
  205. Midorikawa, Y.; Takayama, T.; Higaki, T.; Aramaki, O.; Teramoto, K.; Yoshida, N.; Tsuji, S.; Kanda, T.; Moriyama, M. High platelet count as a poor prognostic factor for liver cancer patients without cirrhosis. Biosci. Trends 2020, 14, 368–375. [Google Scholar] [CrossRef]
  206. Lee, C.-H.; Lin, Y.-J.; Lin, C.-C.; Yen, C.; Shen, C.; Chang, C.; Hsieh, S. Pretreatment platelet count early predicts extrahepatic metastasis of human hepatoma. Liver Int. 2015, 35, 2327–2336. [Google Scholar] [CrossRef] [PubMed]
  207. Ozcelik, F.; Yiginer, O.; Özgün, A. Relationship between thrombocytopenia and extrahepatic metastasis of hepatocellular carcinoma: A different perspective. Liver Int. 2016, 36, 614. [Google Scholar] [CrossRef]
  208. Liu, P.-H.; Hsu, C.-Y.; Su, C.-W.; Huang, Y.; Hou, M.; Rich, N.E.; Fujiwara, N.; Hoshida, Y.; Singal, A.G.; Huo, T. Thrombocytosis is associated with worse survival in patients with hepatocellular carcinoma. Liver Int. 2020, 40, 2522–2534. [Google Scholar] [CrossRef] [PubMed]
  209. Han, S.; Lee, S.; Yang, J.D.; Leise, M.D.; Ahn, J.H.; Kim, S.; Jung, K.; Gwak, M.S.; Kim, G.S.; Ko, J.S. Risk of posttransplant hepatocellular carcinoma recurrence is greater in recipients with higher platelet counts in living donor liver transplantation. Liver Transpl. 2018, 24, 44–55. [Google Scholar] [CrossRef]
  210. Wang, B.; Li, F.; Cheng, L.; Zhan, Y.; Wu, B.; Chen, P.; Shen, J.; Wu, W.; Ma, X.; Zhu, J.; et al. The pretreatment platelet count is an independent predictor of tumor progression in patients undergoing transcatheter arterial chemoembolization with hepatitis B virus-related hepatocellular carcinoma. Future Oncol. 2019, 15, 827–839. [Google Scholar] [CrossRef]
  211. Pavlovic, N.; Rani, B.; Gerwins, P.; Heindryckx, F. Platelets as Key Factors in Hepatocellular Carcinoma. Cancers 2019, 11, 1022. [Google Scholar] [CrossRef] [PubMed]
  212. In’t Veld, S.G.J.G.; Wurdinger, T. Tumor-educated platelets. Blood 2019, 133, 2359–2364. [Google Scholar] [CrossRef] [PubMed]
  213. Sabrkhany, S.; Kuijpers, M.J.E.; Oude Egbrink, M.G.A.; Griffioen, A.W. Platelets as messengers of early-stage cancer. Cancer Metastasis Rev. 2021, 40, 563–573. [Google Scholar] [CrossRef] [PubMed]
  214. Best, M.G.; Wesseling, P.; Wurdinger, T. Tumor-Educated Platelets as a Noninvasive Biomarker Source for Cancer Detection and Progression Monitoring. Cancer Res. 2018, 78, 3407–3412. [Google Scholar] [CrossRef] [PubMed]
  215. Hinterleitner, C.; Strähle, J.; Malenke, E.; Hinterleitner, M.; Henning, M.; Seehawer, M.; Bilich, T.; Heitmann, J.; Lutz, M.; Mattern, S.; et al. Platelet PD-L1 reflects collective intratumoral PD-L1 expression and predicts immunotherapy response in non-small cell lung cancer. Nat. Commun. 2021, 12, 7005. [Google Scholar] [CrossRef] [PubMed]
  216. Asghar, S.; Waqar, W.; Umar, M.; Manzoor, S. Tumor educated platelets, a promising source for early detection of hepatocellular carcinoma: Liquid biopsy an alternative approach to tissue biopsy. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 836–844. [Google Scholar] [CrossRef] [PubMed]
  217. Waqar, W.; Asghar, S.; Manzoor, S. Platelets’ RNA as biomarker trove for differentiation of early-stage hepatocellular carcinoma from underlying cirrhotic nodules. PLoS ONE 2021, 16, e0256739. [Google Scholar] [CrossRef] [PubMed]
  218. Zhu, B.; Gu, S.; Wu, X.; He, W.; Zhou, H. Bioinformatics analysis of tumor-educated platelet microRNAs in patients with hepatocellular carcinoma. Biosci. Rep. 2021, 41, BSR20211420. [Google Scholar] [CrossRef] [PubMed]
  219. Ding, S.; Dong, X.; Song, X. Tumor educated platelet: The novel BioSource for cancer detection. Cancer Cell Int. 2023, 23, 91. [Google Scholar] [CrossRef] [PubMed]
  220. Lingiah, V.A.; Niazi, M.; Olivo, R.; Paterno, F.; Guarrera, J.V.; Pyrsopoulos, N.T. Liver Transplantation Beyond Milan Criteria. J. Clin. Transl. Hepatol. 2020, 8, 69–75. [Google Scholar] [CrossRef]
  221. Mazzaferro, V.; Citterio, D.; Bhoori, S.; Bongini, M.; Miceli, R.; De Carlis, L.; Colledan, M.; Salizzoni, M.; Romagnoli, R.; Antonelli, B.; et al. Liver transplantation in hepatocellular carcinoma after tumour downstaging (XXL): A randomised, controlled, phase 2b/3 trial. Lancet Oncol. 2020, 21, 947–956. [Google Scholar] [CrossRef]
  222. Rajendran, L.; Ivanics, T.; Claasen, M.P.; Muaddi, H.; Sapisochin, G. The management of post-transplantation recurrence of hepatocellular carcinoma. Clin. Mol. Hepatol. 2022, 28, 1–16. [Google Scholar] [CrossRef]
  223. Xue, F.; Shi, S.; Zhang, Z.; Xu, C.; Zheng, J.; Qin, T.; Qian, Z.; Zhao, X.; Tong, Y.; Xia, L.; et al. Application of a novel liquid biopsy in patients with hepatocellular carcinoma undergoing liver transplantation. Oncol. Lett. 2018, 15, 5481–5488. [Google Scholar] [CrossRef]
  224. Wang, S.; Zheng, Y.; Liu, J.; Huo, F.; Zhou, J. Analysis of circulating tumor cells in patients with hepatocellular carcinoma recurrence following liver transplantation. J. Investig. Med. 2018, 66, 1–6. [Google Scholar] [CrossRef] [PubMed]
  225. Xie, Y.-L.; Yang, Z.; Feng, X.; Yang, Q.; Ye, L.-S.; Li, X.-B.; Tang, H.; Zhang, Y.-C.; Liu, W.; Zhang, T.; et al. Association of phenotypic transformation of circulating tumor cells and early recurrence in patients with hepatocellular carcinoma following liver transplantation. Asian J. Surg. 2022, 45, 435–440. [Google Scholar] [CrossRef] [PubMed]
  226. Sugimachi, K.; Matsumura, T.; Hirata, H.; Uchi, R.; Ueda, M.; Ueo, H.; Shinden, Y.; Iguchi, T.; Eguchi, H.; Shirabe, K.; et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 2015, 112, 532–538. [Google Scholar] [CrossRef] [PubMed]
  227. Morsiani, C.; Collura, S.; Sevini, F.; Ciurca, E.; Bertuzzo, V.R.; Franceschi, C.; Grazi, G.L.; Cescon, M.; Capri, M. Circulating miR-122-5p, miR-92a-3p, and miR-18a-5p as Potential Biomarkers in Human Liver Transplantation Follow-Up. Int. J. Mol. Sci. 2023, 24, 3457. [Google Scholar] [CrossRef]
  228. Tian, X.; Wu, L.; Li, X.; Zheng, W.; Zuo, H.; Song, H. Exosomes derived from bone marrow mesenchymal stem cells alleviate biliary ischemia reperfusion injury in fatty liver transplantation by inhibiting ferroptosis. Mol. Cell. Biochem. 2023. [Google Scholar] [CrossRef] [PubMed]
  229. Wu, L.; Tian, X.; Zuo, H.; Zheng, W.; Li, X.; Yuan, M.; Tian, X.; Song, H. miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J. Nanobiotechnol. 2022, 20, 196. [Google Scholar] [CrossRef]
  230. Carlson, K.; Kink, J.; Hematti, P.; Al-Adra, D.P. Extracellular Vesicles as a Novel Therapeutic Option in Liver Transplantation. Liver Transpl. 2020, 26, 1522–1531. [Google Scholar] [CrossRef]
  231. Zhang, A.-B.; Peng, Y.-F.; Jia, J.-J.; Nie, Y.; Zhang, S.-Y.; Xie, H.-Y.; Zhou, L.; Zheng, S.-S. Exosome-derived galectin-9 may be a novel predictor of rejection and prognosis after liver transplantation. J. Zhejiang Univ. Sci. B 2019, 20, 605–612. [Google Scholar] [CrossRef] [PubMed]
  232. Muthukumar, T.; Akat, K.M.; Yang, H.; Schwartz, J.E.; Li, C.B.; Bang, H.; Ben-Dov, I.Z.; Lee, J.R.; Ikle, D.; Demetris, A.J.; et al. Serum MicroRNA Transcriptomics and Acute Rejection or Recurrent Hepatitis C Virus in Human Liver Allograft Recipients: A Pilot Study. Transplantation 2022, 106, 806–820. [Google Scholar] [CrossRef] [PubMed]
  233. Ruiz, P.; Millán, O.; Ríos, J.; Díaz, A.; Sastre, L.; Colmenero, J.; Crespo, G.; Brunet, M.; Navasa, M. MicroRNAs 155-5p, 122–125p, and 181a-5p Identify Patients With Graft Dysfunction Due to T Cell-Mediated Rejection After Liver Transplantation. Liver Transpl. 2020, 26, 1275–1286. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Liquid biopsies of non-blood origin in different malignancies.
Figure 1. Liquid biopsies of non-blood origin in different malignancies.
Jpm 14 00420 g001
Figure 2. Historical time-line of the development of liquid biopsy components.
Figure 2. Historical time-line of the development of liquid biopsy components.
Jpm 14 00420 g002
Table 1. Existing detection technologies for CTCs and the pros and cons of each.
Table 1. Existing detection technologies for CTCs and the pros and cons of each.
MethodologiesCharacterizationDrawbacksMeritsReference
physical isolation assay
Deformability; density;
cell size;
cell surface electric load
Based on the physical property differences between CTCs and blood cells
  • Low capture rates
  • Cannot distinguish the source of CTCs
  • High processing efficiency.
  • Simple and easy to implement
[33,34,35,36]
immunoaffinity isolation assay 1
Positive selection
Cell Search system 2Magnetic beads coupled with Ep CAM 3 bound to ferrofluid-based system
  • False-negative possibility- CTCs undergoing EMT probably do not express Ep CAM.
  • low purity of captured cells
  • CTCs can be visualized and quantified under fluorescence microscopy.
  • CTCs isolated can be analyzed for mRNA expression and DNA mutations
  • High sensitivity, detecting approximately 5 CTCs per 7.5 mL whole blood
[42,43,44]
Adna TestMUC1 and Ep CAM antibody-labeled immunomagnetic beads combined with RT-PCR for detection of tumor-associated transcripts
  • False-positive possibility—activated leukocytes can express MUC1
  • Harsh blood sample handling and storage conditions
  • Promising to detect tumor-specific transcripts not typically detected in nucleated blood cells
  • Highly specific and sensitive (detecting approximately 22 CTCs per 5 mL whole blood)
[45,46]
GILUPI CellCollector™Invasive enrichment modality—Ep CAM-coated wire placed into patient’s vein for CTCs capture
  • Invasive
  • Long intravascular presence
  • Single processing of large volumes of blood.
  • High isolation purity
  • Genetic analysis can be continued
[47]
Negative selection
Can Patrol™Negative Enrichment 4 combined with filtration based on cell size
  • Low purity—low levels of leukocyte interference
  • CTCs without classical epithelial antigen expression were captured
[48]
Rosette Sep™Recognition and removal WBCs and RBCs from blood samples by antibody complexes (CD45, CD66b and glycoproteins) in combination with flow cytometry
  • Slow processing speeds
  • Excessive shear forces may affect cell viability and cell ligand attachment.
  • Higher capture specificity and selectivity
  • The captured CTCs can proceed to phenotyping and molecular analysis
[49,50]
Cyttel methodNegative enrichment modality 5 combined with immunofluorescence and fluorescence in situ hybridization.
  • False positive results
  • Lack of harmonized protocols for clinical application
  • High recovery rate
  • Enriched cells with good activity
  • High sensitivity and specificity
[51,52]
Combined positive and negative selection
CTC-iChipMicrofluidic platform using lateral displacement, inertial focusing and magnetophoresis coupled with tumor antigen-dependent and/or non-dependent enrichment of CTCs
  • Low purity
  • Complexity of production
  • High separation efficiency and
  • yield
  • Cytopathological and molecular characterization of both epithelial and non-epithelial cancers
[53,54,55]
MACS systemImmune-magnetic CTC enrichment using cell surface marker antibodies or intracellular anti-pan CK antibodies
  • Identifies Ep CAM-negative cells but not CK-negative ones
  • High efficiency
  • Can be combined with leukocyte depletion
[56,57]
1 Immunoaffinity isolation assay has two main selection methods. Positive selection utilizes tumor-specific markers to select CTCs from the sample. Negative selection achieves CTCs enrichment by removing other cellular components in the sample, and it is not affected by tumor cell surface marker expression variations [58]. 2 The only FDA-approved assay to screen CTCs in metastatic breast, colon and prostate carcinoma [45,59,60,61]. 3 Epithelial cell adhesion molecule (Ep CAM) has been identified abnormal in a spectrum of malignancies, including HCC [62]. 4 The Can Patrol protocol removes erythrocytes by erythrocyte lysis and eliminates CD45+ leukocytes using magnetic bead separation for negative enrichment. 5 The Cyttel method utilizes hypotonic hemolysis to clear erythrocytes and anti-CD45 antibody co-coupled magnetic beads to clear leukocytes for negative enrichment.
Table 2. Expression of anomalous miRNAs in HCC tissues and the roles of each.
Table 2. Expression of anomalous miRNAs in HCC tissues and the roles of each.
miRNAsExpression LevelFunctionRolesReference
miR-182Upward
  • Promote HCC proliferation and metastasis
  • Increase HCC resistance to cisplatin
Prediction of recurrence[108,109,110]
miR-21Upward
  • Promote hepatocyte steatosis, inflammation and fibrosis
  • Increase HCC cells proliferation and invasion, inhibit apoptosis
Potential therapeutic targets[111]
miR-454Upward
  • Participate in HCC genesis and tumor stem cell self-renewal
  • Participate in HCC resistant to sorafenib
Prognostic prediction and therapeutic targets related to HCC resistance[112]
miR-873Upward
  • involve in hepatocyte metabolism and HCC occurrence
Prognostic prediction[113]
miR-18aUpward
  • Enhance HCC invasion, migration, and proliferation
Prognostic prediction[114]
miR-130b-3pUpward
  • Promotes HCC angiogenesis
Potential therapeutic target and prognostic markers[115]
miR-375Upward
  • Inhibit HCC angiogenesis
  • Participate in HCC resistant to sorafenib
Potential therapeutic targets related to HCC resistance[116]
miR-25Upward
  • Induce HCC resistance to sorafenib
  • Regulate HCC apoptosis
Potential therapeutic targets related to HCC resistance[117]
miR-1246Upward
  • Involved in HCC development
Potential early detection and prognostic prediction[118]
miR-296-5pDownward
  • Inhibit HCC invasion, migration, and proliferation
Prognostic prediction[119,120]
miR-206Downward
  • Regulate hepatocyte viability, migration and apoptosis Inhibit HCC development
Multifunctional anti-tumor effects[121]
miR-497Downward
  • Involved in HCC development
Potential early detection and prognostic prediction[118]
miR-106-5p
miR-372-5p
Downward
  • Inhibit HCC proliferation and invasion and promote HCC apoptosis
Potential therapeutic targets related to HCC resistance and prognostic markers[122]
miR-424-5pDownward
  • Inhibit HCC cell proliferation and invasion
Potential therapeutic target and prognostic markers[123]
miR-3064-5pDownward
  • Inhibit HCC angiogenesis
Potential therapeutic target and prognostic markers[124]
miR-203Downward
  • Enhance HCC radiosensitivity
Prognostic prediction and Potential therapeutic target[125]
miR-146aDownward
  • Inhibit HCC proliferation and induce HCC apoptosis
  • Enhance HCC radiosensitivity
Potential therapeutic target[126]
miR-621Downward
  • Enhance HCC radiosensitivity
Potential therapeutic target[127]
miR-148bDownward
  • Promote HCC metastasis
Prognostic prediction and Potential therapeutic target[128]
miR-144
miR-451a
Downward
  • Enhance anti-tumor immunity
Prognostic prediction and Potential therapeutic target[129]
Table 3. Materials with HCC-related aberrant expression in exosomes and the roles of each.
Table 3. Materials with HCC-related aberrant expression in exosomes and the roles of each.
MaterialsBiomarkerSourceExpression LevelsFunctionReference
Protein
LOXL4HCC cellsUpward
  • Involved in HCC metastasis
[149]
METHCC cellsUpward
  • Involved in tumor proliferation, angiogenesis metastasis
[150]
Triosephosphate isomerase 1 (TPI1)HCC cellsDownward
  • Inhibit the development of HCC
[151]
CAV1HCC cellsUpward
  • Involved in HCC formation and metastasis
[150,152,153]
Pyruvate kinase M2 isoform (PKM2)HCC cellsUpward
  • Reshape the Tumor Microenvironment
  • Promote HCC development
[154]
Alpha-enolase (ENO1)HCC cellsUpward
  • Promote HCC growth, metastasis
[155]
p120-cateninHCC cellsDownward
  • Inhibit HCC proliferation and metastasis
[156]
14-3-3 protein zetaHCC cellsUpward
  • Promotes HCC invasion and metastasis
  • Involved in T-lymphocyte depletion
[157]
Complement Factor H (CFH)HCC cellsUpward
  • Promote HCC growth, migration, and invasiveness
[158]
Non-coding RNAs
circRNA-PTGR1HCC cellsUpward
  • Enhances migration and invasion of low-transfer-potential HCC cells
[159]
circRNA-100338HCC cellsUpward
  • Enhance HCC metastasis
  • promote HCC angiogenesis
  • Prognostic prediction
[160]
lncRNA FAL1HCC cellsUpward
  • Enhance HCC proliferation and migration capacity.
[161]
miRNA-15bmacrophagesUpward
  • Promote HCC proliferation, migration and invasion
[162]
miR-103HCC cellsUpward
  • Increase vascular permeability
  • promote HCC metastasis
[163]
miR-584-5pHCC cellsUpward
  • Promote HCC angiogenesis
[164]
Non-coding RNAs
miR-210HCC cellsUpward
  • Promote HCC angiogenesis
[165]
miR-21
miR-10b
HCC cellsUpward
  • Promote HCC proliferation, migration and invasion
  • Prognostic prediction
[166,167]
miR-1247-3pHCC cellsUpward
  • Related to HCC lung metastasis
[168]
miR-92bHCC cellsUpward
  • Enhance HCC migration
  • Down-regulated natural killer (NK) cell-mediated cytotoxicity
  • Prognostic prediction
[169]
miR-23a-3pHCC cellsUpward
  • Upregulate PD-L1 expression in macrophages to help HCC evade anti-tumor immunity.
[170]
Table 4. Application of liquid biopsy in the management of HCC.
Table 4. Application of liquid biopsy in the management of HCC.
CTCscfDNActDNACirculating microRNAExosomesTEPs
ScreeningNeed for researchNeed for research
DiagnosisNeed for researchNeed for researchNeed for research
TreatmentNeed for research
Prognostic monitoringNeed for research
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, J.; Zhao, Y.; Chen, Z.; Wei, L. Clinical Application of Different Liquid Biopsy Components in Hepatocellular Carcinoma. J. Pers. Med. 2024, 14, 420. https://doi.org/10.3390/jpm14040420

AMA Style

Xu J, Zhao Y, Chen Z, Wei L. Clinical Application of Different Liquid Biopsy Components in Hepatocellular Carcinoma. Journal of Personalized Medicine. 2024; 14(4):420. https://doi.org/10.3390/jpm14040420

Chicago/Turabian Style

Xu, Jing, Yuanyuan Zhao, Zhishui Chen, and Lai Wei. 2024. "Clinical Application of Different Liquid Biopsy Components in Hepatocellular Carcinoma" Journal of Personalized Medicine 14, no. 4: 420. https://doi.org/10.3390/jpm14040420

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

Article Metrics

Back to TopTop