Next Article in Journal
Exploration of the Effect on Genome-Wide DNA Methylation by miR-143 Knock-Out in Mice Liver
Next Article in Special Issue
Multigene Profiling of Circulating Tumor Cells (CTCs) for Prognostic Assessment in Treatment-Naïve Metastatic Hormone-Sensitive Prostate Cancer (mHSPC)
Previous Article in Journal
The Secretome of Human Neonatal Mesenchymal Stem Cells Modulates Doxorubicin-Induced Cytotoxicity: Impact in Non-Tumor Cells
Previous Article in Special Issue
Liquid Biopsy in Melanoma: Significance in Diagnostics, Prediction and Treatment Monitoring
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circulating Tumor Cells in Hepatocellular Carcinoma: A Comprehensive Review and Critical Appraisal

by
María Lola Espejo-Cruz
1,†,
Sandra González-Rubio
1,†,
Javier Zamora-Olaya
1,2,
Víctor Amado-Torres
1,2,
Rafael Alejandre
1,2,
Marina Sánchez-Frías
3,
Rubén Ciria
1,4,
Manuel De la Mata
1,2,5,
Manuel Rodríguez-Perálvarez
1,2,5,* and
Gustavo Ferrín
1,5
1
Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
2
Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
3
Department of Pathology, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
4
Department of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, 14004 Córdoba, Spain
5
Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
These two authors have contributed equally and may be considered co-first authors in the present manuscript.
Int. J. Mol. Sci. 2021, 22(23), 13073; https://doi.org/10.3390/ijms222313073
Submission received: 29 October 2021 / Revised: 27 November 2021 / Accepted: 30 November 2021 / Published: 3 December 2021
(This article belongs to the Special Issue Circulating Tumor Cells: The Next Generation Liquid Biopsy)

Abstract

:
Hepatocellular carcinoma (HCC) is the fifth most common neoplasm and a major cause of cancer-related death worldwide. There is no ideal biomarker allowing early diagnosis of HCC and tumor surveillance in patients receiving therapy. Liquid biopsy, and particularly circulating tumor cells (CTCs), have emerged as a useful tool for diagnosis and monitoring therapeutic responses in different tumors. In the present manuscript, we evaluate the current evidence supporting the quantitative and qualitative assessment of CTCs as potential biomarkers of HCC, as well as technical aspects related to isolation, identification, and classification of CTCs. Although the dynamic assessment of CTCs in patients with HCC may aid the decision-making process, there are still many uncertainties and technical caveats to be solved before this methodology has a true impact on clinical practice guidelines. More studies are needed to identify the optimal combination of surface markers, to increase the efficiency of ex-vivo expansion of CTCs, or even to target CTCs as a potential therapeutic strategy to prevent HCC recurrence after surgery or to hamper tumor progression and extrahepatic spreading.

1. Introduction

Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer, representing 90% of primary liver malignancies in patients with cirrhosis. According to the World Health Organization, HCC is the fifth most common neoplasm worldwide and the third leading cause of cancer-related death [1,2]. The geographic distribution of HCC is very heterogeneous and closely associated with the prevalence of the different etiologies of chronic liver disease [3,4,5]. In recent decades, different healthcare strategies such as vaccination campaigns, development, and liberal prescription of direct-acting antivirals against hepatitis C, and promotion of healthy lifestyle, have been implemented to reduce the incidence of HCC [5,6], but liver cirrhosis is still prevalent and forms a major driver of HCC [7]. The Barcelona Clinic Liver Cancer (BCLC) classification is nowadays the most widely used staging system of HCC [8,9], which also provides therapeutic guidance, and it has been recommended by international clinical practice guidelines [5,10]. Patients with early-stage HCC (BCLC 0-A) may be candidates for potentially curative therapies such as radiofrequency ablation, surgical liver resection (LR), or liver transplantation (LT), with 5-year survival rates of 50–80%. On the other hand, patients with advanced HCC (BCLC C) are candidates for systemic therapies, mainly (multi) tyrosine kinase inhibitors such as sorafenib or regorafenib, and the more recently approved combination of atezolizumab (anti-programmed death-ligand 1 -PD-L1- monoclonal antibody) and bevacizumab (anti-vascular endothelial growth factor monoclonal antibody), which may provide median survival rates shorter than 2 years [4].
Therefore, early diagnosis of HCC is paramount in patients with liver cirrhosis to maximize survival [11,12]. Nowadays, patients with advanced liver fibrosis or cirrhosis may undergo screening for HCC, which consists of an abdominal ultrasound every six months. Serum biomarkers of HCC such as alpha-fetoprotein (AFP), AFP-L3 (a glycoform of AFP), des-gamma-carboxyprothrombin, glypican-3 (GPC3), or osteopontin [13] may provide information about the biological aggressiveness of HCC but, unfortunately, they are not sufficiently accurate to form part of screening strategies. There is a need for new biomarkers of HCC to refine screening and diagnosis, to better predict tumor recurrence after surgical or ablative therapies, and to monitor response in patients receiving systemic therapies. In the present review, we have comprehensively evaluated the current evidence supporting the quantitative and qualitative assessment of circulating tumor cells (CTCs) as potential biomarkers of HCC.

2. Liquid Biopsy and CTCs

The term liquid biopsy refers to the detection of cancer byproducts mainly in the bloodstream which could provide first-hand information about the features of the primary tumor from which they belong. Liquid biopsy offers promising benefits in terms of refined diagnosis, prognostic stratification, and therapeutic guidance in different types of cancers such as metastatic breast, colorectal, or prostate cancer [13]. There are several modalities of liquid biopsy, including the detection of CTCs, extracellular vesicles, or circulating tumor nucleic acids (ribonucleic acid -RNA-, microRNA -miRNA-, tumor deoxyribonucleic acid -DNA-). The most evident advantage of liquid biopsy is the broadly accessible study specimens, which usually consist of peripheral blood samples [14]. CTCs were first described in 1869 as malignant cells derived from the primary or metastatic tumor, able to access the systemic circulation [15,16]. They are very scarce in peripheral blood, with a very short circulating half-life (1 h to 2.4 h) [17]. Despite this, CTCs can be detected even at earlier tumor stages, being associated with a more aggressive biological behavior. Unlike other liquid biopsy markers such as cell-free DNA, CTCs are unequivocally associated with the presence of viable tumors (even if not detected by conventional imaging techniques) and the progressive increase of CTCs indicates tumor spreading recurrence and metastases.
Prior to the identification of CTCs, blood samples require an enrichment method to purify such a minority cell population. Methods for CTC enrichment may be classified into positive and negative. These are not mutually exclusive and can therefore be combined to optimize the capture of CTCs. Negative-enrichment methods are based on the removal of white blood cells by antibody-based depletion strategies usually based on anti-CD45-coated magnetic beads or CD45 depletion cocktails. Enriched CTCs can be detected afterward by several different methodologies such as immunocytochemistry (ICC) [18,19,20,21,22,23], quantitative real-time polymerase chain reaction (PCR) [24,25,26], flow cytometry, or immunofluorescence in situ hybridization (iFISH) [27,28,29,30,31,32], which may combine several probes or antibodies to identify different CTC subpopulations. In contrast, positive-enrichment methods take advantage of specific biological and physical properties of CTCs to distinguish them from non-tumoral blood cells. Physical enrichment strategies are frequently based on centrifugation and microfiltration [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50] to selectively isolate CTCs according to their differential density and size, respectively [51]. Among all the positive-enrichment methods, immunoaffinity is the most widely used but the optimal biomarker matching all CTC subpopulations is unknown. Different combinations of antibodies can be immobilized on the surface of magnetic particles or microdevices with increased capture efficiency [51,52]. As with negative-enrichment methods, isolated CTCs can be subsequently identified by common methodologies (Figure 1). The sensitivity for the detection of CTCs varies greatly between the different methods used, and even between different studies that use the same methodology.
The only CTC detection system approved by the Food-and-Drug Administration is the CellSearch® system (Menarini-Silicon Biosystems, Bologna, Italy), which is based on immunocapture (positive enrichment) of CTCs expressing epithelial-to-cell adhesion molecule (EpCAM+ CTCs). Although the CellSearch® system has been widely used in HCC research with promising results [26,53,54,55,56,57,58,59,60,61,62], novel methodologies are in the pipeline claiming for increased CTC detection rate, sensitivity, and specificity [30,35,63,64]. Thus, CTC analysis by the IsoFlux® system [64] or by negative enrichment and iFISH [30] showed an increased sensitivity in HCC candidates for LT compared to the CellSearch® system (90.5% vs. 4.7% and 70% vs. 26.67%, respectively).

3. Surface Markers to Identify CTCs in HCC and Clinical Significance

The key to ensuring adequate sensitivity and specificity is to identify a surface marker to be targeted for optimal positive cell identification. Recent advances have shown that there is no single optimal marker, and probably the best approach is to combine different surface markers with additional selection filters according to physical properties. In this section, we provide an insight into the most widely used CTC markers in HCC with their benefits and limitations.

3.1. Liver and HCC Specific Markers

A logical source of candidate markers for the detection of CTCs in HCC is the liver-specific or HCC-specific proteome. Since hepatocytes do not circulate under physiological conditions, any cell expressing a liver or HCC-specific marker detected in the bloodstream may be considered a potential CTC. GPC3 is a cell membrane-anchored protein currently used in clinical practice for HCC pathological analysis and characterization [65]. Since GPC3 is more frequently observed in moderately and poorly differentiated HCC tumor cells, which are more prone for extrahepatic spreading, the presence of GPC3+ CTCs may inform about a de-differentiated metastatic HCC. Thus, the immunomagnetic enrichment of GPC3+ CTCs and subsequent analysis by fluorescent activated cell sorter with anti-cytokeratin (CK, an epithelial marker) antibodies was postulated as a valid methodology to identify poor prognostic HCC patients. Indeed, an increased number of CTCs obtained by this approach was associated with microvascular invasion (a well-known poor prognostic histological feature [66]), increased tumor recurrence rates, and shorter overall survival [67]. Asialoglycoprotein receptor (ASGPR) is a transmembrane protein exclusively expressed on the surface of hepatocytes [68], but unlike GPC3, it is highly expressed in well-differentiated HCC. Xu et al. developed a system based on magnetic beads to capture ASGPR+ CTCs, which were subsequently identified by ICC with antibodies anti-hepatocyte-specific hepatocyte paraffin 1 (HepPar 1; a marker for differential diagnosis of HCC [69]) or anti-CK alone. The positivity rate and the number of CTCs isolated by this technique were associated with increased tumor burden, macrovascular invasion, and poor histological differentiation [70]. This methodology was modified afterward using a new anti-ASGPR monoclonal antibody for the isolation of CTCs and a combination of anti-CK and anti-CPS1 (an antigen for HepPar 1) antibodies, thus resulting in higher sensitivity for CTC detection (89% vs. 81%) [71]. The authors designed a platform to capture and release living HCC CTCs for their ulterior expansion in a 3D cell culture assay [72]. The combination of EpCAM, ASGPR, and GPC3 antibodies in the so-called NanoVelcro CTC Assay resulted in HCC CTCs detection in up to 97.6% of patients with HCC at various stages, being the number of CTCs associated with clinical outcomes. In addition, the NanoVelcro CTC Assay identified a vimentin-positive subpopulation of CTCs that indicated a particularly aggressive biological tumor behavior [52]. Using a labyrinth device that exploits the physical characteristics of CTCs and a combination of HCC-specific antibodies against GPC3, glutamine synthetase, and HepPar-1 for ICC analysis, Wan et al. detected CTCs in 88.1% of patients with HCC and correlated the positivity rate of CTCs with the tumor stage [73]. Liver-specific or HCC-specific markers could also be useful for the identification of HCC CTCs after removal of CD45+ cells and/or red blood cells. Immunofluorescence analysis of negative-enriched CTCs from HCC patients with antibodies against ASGPR and CPS1 or GPC3 showed high sensitivity and specificity for detecting CTCs and could provide relevant prognostic information [18,74]. Thus, the incorporation of several specific markers into a single platform for the isolation and/or detection of HCC CTCs across all tumor stages may be more efficient than a single marker.

3.2. Epithelial Markers

EpCAM is one of the most used membrane-associated proteins for capturing CTCs in peripheral blood samples as it is not expressed by normal blood cells [75]. EpCAM+ CTCs have been associated with different clinicopathological features of HCC, including vascular invasion, high serum AFP (≥400 ng/mL), and more advanced BCLC stage [58,59]. In patients undergoing LT, LR, transarterial chemoembolization (TACE), or radiotherapy for HCC, EpCAM+ CTC count at baseline has been associated with increased tumor recurrence, shorter overall survival, and shorter progression-free survival [26,54,55,60]. EpCAM+ CTCs could also aid to assess the optimal surgical margin size during LR [62], optimizing the management of HCC patients on the waiting list for LT [76], and for a better selection of candidates to certain therapies [61]. In other words, EpCAM+ CTCs are strongly correlated with tumor aggressiveness and could assist in the adequate stratification of HCC patients for optimal therapy and in predicting their response to treatment [56]. Unfortunately, EpCAM-based CTC enrichment may overlook CTCs with low expression of the epithelial marker [77,78] or those which expression has been lost during the dynamic process of epithelial-to-mesenchymal transition (EMT) [75,79]. Thus, the use of additional criteria such as size and brightfield image could enhance CTC detection sensitivity in HCC [22]. In addition, since EpCAM has been identified as a surface marker of HCC cells displaying stem cell features [80,81] and EpCAM+ CTCs from HCC patients are highly tumorigenic in vivo [60], it is possible that EpCAM+ CTCs account for both tumorigenic and non-tumorigenic cells.

3.3. Epithelial-to-Mesenchymal Transition Markers

The EMT is the process by which a CTC with an epithelial phenotype acquires mesenchymal characteristics that confer increased migratory ability, invasiveness and/or drug resistance, thus contributing to HCC spreading [82]. The activation of the EMT process in CTCs occurs primarily in the bloodstream and involves dynamic adaptive mechanisms that are associated with stress response, cell cycle, or immune evasion [21,40,83,84]. This process may not be considered dichotomous: CTCs do not necessarily exist in ‘pure’ epithelial or mesenchymal states. CTCs can be found in intermediate states showing characteristics of both epithelial and mesenchymal phenotypes [85]. Therefore, the use of a combination of epithelial and mesenchymal markers would improve the sensitivity and the overall accuracy of a CTC enrichment method. The expression of EMT markers such as vimentin, twist, Zinc finger E-box-binding (ZEB)1, ZEB2, snail, slug, and E-cadherin has been studied in liver-derived CTCs from HCC patients, being the co-expression of twist and vimentin significantly correlated with tumor burden, macrovascular invasion, and more advanced stages [86]. The CanPatrolTM system, which combines a positive-enrichment filter-based method and an RNA-in situ hybridization (RNA-ISH) technique, was able to stratify CTCs into three phenotypic subgroups accordingly to the predominantly expressed markers: epithelial phenotype (EpCAM, CK 8/18/19), mesenchymal phenotype (vimentin, twist), and mixed/hybrid phenotype. Overall CTC count (which comprises all phenotypes) performed better than serum AFP to discriminate between patients diagnosed with HCC and those with non-malignant liver diseases [44]. Mesenchymal CTCs (mCTC) and mixed CTCs were associated with clinical features indicating poor prognoses, such as high serum AFP levels, advanced tumor stages, and microvascular invasion. Thus, EMT-based classification of CTCs could predict early recurrence of HCC, metastases, and shorter overall survival [33,36,40,43,45,48,50].
Epithelial CTCs and mCTC are occasionally found in the bloodstream bound to immune cells, platelets, and fibroblast as circulating tumor microemboli or CTC clusters, which may arise from the detachment of multicellular aggregates from the primary tumor mass or the reaggregation of CTCs in the bloodstream [84]. These clusters contribute to the survival and adoption of invasive advantages of CTCs and highlight the relevance of the tumor environment in the metastatic process [87,88]. The microfluidic isolation and subsequent immunofluorescence identification of CTC clusters in HCC patients with more advanced tumor stages indicated their association with tumor progression [73]. By using the CanPatrolTM system, Luo et al. showed that the presence of CTC clusters consisting of CTCs (epithelial and/or mesenchymal) and white blood cells was associated with tumor burden and vascular invasion, being an independent predictor of poor prognosis in HCC [41]. In addition, the authors correlated the number of mCTC and CTC clusters with a specific mutational profile of EMT-related genes involved in the metastatic process of HCC [42].

3.4. Stem-Cells Markers

Stem cell markers are expressed in a small subpopulation of CTCs which may be considered tumor-initiating cells related to increased aggressiveness and poor prognosis. These circulating cancer stem cells (cCSC) may derive directly from the tumor or an adaptive process during the EMT. Due to its scarcity in peripheral blood, CTC-derived xenograft models and CTC-derived ex vivo cultures have been used for the study of cCSC [89]. CSCs may express surface markers of regular stem cells such as EpCAM, CD133, CD44, CD90, or ICAM-1. CD44 is an adhesion molecule that facilitates tumor cell invasion and migration [90] and it is concomitantly expressed with other stem-cell markers. In HCC cells and tumor xenograft models, CD133+CD44+ cells (but not CD133+CD44-) display stem cell properties, including extensive proliferation, self-renewal capacity, tumorigenic potential, resistance to chemotherapy, and expression of stem cell-associated genes [91]. In this sense, CD133+CD44+ cCSC has been correlated with increased serum transaminases, serum AFP and poorer outcomes in HCC patients [92]. CD90+CD44+ cells represent a more aggressive cell subpopulation within the CD90+ group, with increased in vivo tumorigenic capacity than CD90+CD44- cells. Interestingly, CD90+ CTCs express higher levels of CD44 than those cells located within the primary tumor, suggesting increased tumor aggressiveness. Indeed, the number of CD90+ cCSC was related to disease progression in HCC patients [93]. The concomitant expression of CD90 with other surface markers such as CXCR4 (receptor for stromal cell-derived factor-1 (SDF-1/CXCL12)) may be required by cCSC to motivate HCC progression [94]. EpCAM+ CTCs from HCC patients exhibit high tumorigenic activity in vivo and were associated with the expression of others CSC markers such as CD133 and ABCG2 [60]. Based on a strategy combining negative cell enrichment and quantitative real-time PCR, the expression of nine CSC markers was analyzed in the blood of HCC patients. Aligning with the above-referred studies, EpCAM, CD133, CD90, and CK19 (another hepatic stem marker [95]) were significantly overexpressed in the HCC group and associated with the presence of a corresponding subpopulation of HCC CTCs and with a higher tumor recurrence rate [25]. Similarly, EpCAM+CD90+ CTCs have been associated with HCC recurrence after LT [96]. Although expression levels of ICAM-1 did not differ significantly from the control group in that study, hepatic ICAM-1+ CTCs from HCC patients show high tumorigenic activity in vivo and their frequency is an independent risk factor of portal vein tumor thrombus and ascites [97].

4. Dynamic Changes of CTC Counts after HCC Therapy

Preoperative CTCs have been widely associated with HCC recurrence after LR [24,36,55,56,60], LT [29,30,96] or local ablation [28]. Theoretically, a complete tumor removal by LR or LT may motivate a rapid decline in CTC count in the post-operative period. Similarly, local ablation therapies could release CTCs to the bloodstream initially, followed by a progressive decline until complete clearance. In patients receiving systemic therapies, the dynamic of CTC counts may vary depending on the therapeutic response. In any of these situations, trends of CTC count after therapy could provide valuable information to predict tumor recurrence or to anticipate resistance to systemic therapies (Figure 2).

4.1. CTC Dynamics after Liver Resection

Studies evaluating intraoperative CTC monitoring have shown that surgical manipulation during LR is not associated with a significant CTC release into the bloodstream [49]. LR appeared to have little effect on CTC count in the immediate postoperative period [53]. The decline of CTC count after surgical resection may be more evident between postoperative days 7 to 10 [40,49] and could continue declining up to the first post-operative month [26,37,56,60]. The postoperative increase in CTCs and the persistence of a high CTC count after LR may increase the risk of tumor recurrence and extrahepatic metastases, as well as shorter overall survival. These results were consistent using the CellSearch® [26,53,56,60] and the CanPatrolTM [37,40] systems, as well as the isolation by size of epithelial tumor cells method [49]. It may well be that these patients with persistence of CTCs had an incomplete tumor removal or additional HCC microscopic foci within the liver, which in the last term would motivate local recurrence or distant metastases. The percentage of mCTC (and total CTC count) could anticipate tumor recurrence or metastases up to two months before it is evident in imaging techniques [40].

4.2. CTC Dynamics after Liver Transplantation

LT is considered a more radical therapy than LR for patients with HCC since the whole cirrhotic liver is removed, including the known HCC nodules and the possible nascent tumors in the remaining liver parenchyma. A more rapid decline of CTCs would therefore be expected. A single study using negative-enrichment and iFISH platform showed a significant decrease in CTC count 3 months after surgery in most patients [30]. However, CTC dynamics assessed by the CanPatrolTM system did not find a significant association with HCC recurrence [34]. More recently, Wang et al. reported that those patients who did not have CTCs pre-LT and showed positive counts 2–4 weeks after LT had an increased risk of tumor recurrence [20]. This study used a negative-enrichment method and ICC detection of CTCs and, unlike the results obtained by Xue et al. by using iFISH [30], no significant difference in preoperative CTC count or CTC-positive rate was observed between patients who experienced tumor recurrence and those who did not. Very recently, Hwang et al. have shown that CTCs with cancer stemness could predict HCC recurrence when they are detected preoperatively and 1 day after LT using fluorescent activated cell sorter [96]. We have evaluated the clearance kinetics of CTCs in HCC patients undergoing LT and found that the presence of CTC clusters before surgery was associated with an incomplete clearance of CTCs at postoperative day 30, which in turn predicted mortality due to extrahepatic recurrence of HCC [98]. The unpredictable evolution of CTCs after LT could be in part explained using immunosuppressive drugs, which are required to prevent graft rejection. Since evading the immune system is one of the hallmarks of cancer [99], the use of immunosuppressive agents such as calcineurin inhibitors may increase the risk of tumor recurrence in a dose-dependent manner [100]. A reduced CTC count after LT in a patient deeply immunosuppressed may be sufficient to persist in the bloodstream leading to tumor recurrence. Different protocols including CTCs monitoring have been proposed to decrease the risk of tumor recurrence after LT including the following aspects: (1) an adequate selection of recipients with limited tumor burden and reduced baseline CTC count, (2) minimizing the handling of large HCCs during transplantation to decrease the risk of CTC release, (3) decreasing liver graft ischemia-reperfusion injury to prevent the engraftment of CTCs in the liver and (4) consider using anticancer drugs and (5) minimization of immunosuppression [101].

4.3. CTC Dynamics after Local Ablative Therapies and TACE

It has been suggested that radiofrequency ablation [46] and TACE [31,102] could motivate the release of CTCs. Conversely, microwave ablation has been associated with a significant decrease of CTCs and a higher tumoricidal effect [103]. Dynamic changes of CTCs after radiofrequency ablation [39], chemoembolization [31,103,104], or microwave ablation [39,103] seemed to have no impact on tumor recurrence rates when CTCs were identified by using the CanPatrolTM system, negative-enrichment, and flow cytometry or iFISH, and immunomagnetic enrichment. However, these studies were limited by the reduced sample size. In a study including 155 HCC patients assessing CTCs by negative-enrichment and iFISH, patients who responded to TACE (complete response and partial response) had a significant CTC count decline whereas patients without radiological response did not [32]. Similarly, when CTCs were assessed by the CellSearch® system, patients with stable or decreasing CTC count one month after TACE or radiotherapy showed radiological response or stable disease, while most of the patients with persistently high CTC count showed disease progression [26].

4.4. CTC Dynamics during Systemic Treatment

There is no evidence regarding the dynamics of CTCs in patients with HCC receiving multikinase inhibitors or immunotherapy. In a highly metastatic orthotopic nude mouse model of green fluorescent protein-labeled HCC, Yan et al. used a non-invasive in vivo flow cytometry method and showed that sorafenib could reduce the number of CTCs by inhibiting tumor proliferation and angiogenesis [105]. A study evaluating cell-free DNA has identified a mutational profile that indicates resistance to sorafenib [106]. More reliable information could be obtained by analyzing the dynamic of CTC counts and by characterizing the mutational profile of CTCs longitudinally to anticipate therapeutic resistance and radiological progression, thus allowing patients to access a second-line therapy in a timely manner.

5. Cell Culture of HCC CTCs

Ex vivo expansion of viable CTCs can be useful to obtain prognostic information and to investigate the biochemical, gene expressional, and behavioral properties of the primary cancer cells [107]. CTC culture would allow identifying specific targets for cancer therapy, monitoring the acquisition of mutations conferring drug resistance, and even testing the vulnerability of the tumor against multiple possible therapeutic agents in a non-invasive manner [108]. Compared to preclinical CTC-derived xenograft models, the ex vivo culture of CTCs is less time-consuming, which can be paramount when making individualized clinical decisions [89]. Researchers frequently use 3D culture conditions and the high glucose DMEM/F12 medium with epidermal growth factor and/or basic fibroblast growth factor for the enrichment of cancer cell lines-derived CSC spheroids in vitro [109,110]. Optional supplements are N2, B27, inhibitors Y27632 and SB431542, heparin, insulin-transferrin-selenium, or fetal bovine serum. Even though, CTC culture is challenging. Stable ex vivo culture of cCSC has been successfully achieved mainly in patients with metastatic cancer and increased CTC count [111,112], but CSC spheroids often survive for only a few days or weeks [113]. Therefore, ex vivo CTC expansion is inefficient even in patients with advanced cancer [114,115].
Ex vivo culture of cCSC in patients with HCC has been reported even more anecdotally. The few successful studies included intermediate/advanced cancer patients and reported similar culture conditions including normoxia, MatrigelTM solution, and DMEM (with or without 10% fetal bovine serum). cCSC isolation methods varied widely, including negative selection with magnetic beads [23,116] and positive selection by microfluidic chip [72] or fluorescent activated cell sorter [117]. A success rate of 86% to 100% has been reported with a median cell survival time of 7–14 days, although a publication bias is highly probable.
The main caveat for CTC culture is again the reduced number of these cells in the bloodstream. Different approaches are under investigation to increase the isolation rate of CTCs [118]. New methodologies for stable ex vivo culture of CTCs will need to mirror the tumor microenvironment as accurately as possible. Stress conditions naturally occurring in solid tumors such as glucose depletion and hypoxia have been tested. Glucose deprivation slows down cell proliferation but increases the expression of stem cell markers and other stem cell phenotypic characteristics [119]. Regarding the oxygenation conditions, the culture of cCSC has been successfully achieved both in normoxia and hypoxia. Hypoxia is a critical microenvironmental factor promoting the self-renewal of CSCs and a more aggressive phenotype in tumor cells [83,110,111,120], which in turn is associated with resistance to therapy [121,122]. Hypoxic conditions have been successfully used for long-term CTC culture from patients with different types of cancer including colorectal [123], lung [124], breast [89,108], or gastroesophageal [125] tumors. In an ongoing research project analyzing the clearance of CTCs after local ablation of HCC, we tried to obtain cCSC spheroids by using different strategies. First, we used anti-EpCAM-coated immunomagnetic beads and the IsoFlux® system (Fluxion Biosciences, Alameda, CA, USA), which displays great sensitivity in the identification of HCC-derived CTCs [64]. Although we were able to obtain CSC spheres from established HCC cell lines using this strategy, we failed to obtain cCSC spheroids in any of the 14 patients analyzed after 2 weeks of cell culture in normoxia, high glucose, and ultra-low adherence. By contrast, in an independent cohort of 12 patients, the negative enrichment and subsequent culture of CD45- CTCs under the same conditions allowed to obtain CSC-like spheroids in 50% of patients. Despite this encouraging result, spheroids could not be expanded for more than 7–10 days and they finally died off after this period (data not shown). In addition to the difficulties of cultivating CTCs, it is worth mentioning that most patients included in this study were intermediate-stage HCC patients, with lower CTC counts than those obtained in advanced HCC.

6. CTCs and Microenvironment as Therapeutic Targets in HCC

Targeting CTCs is an attractive strategy to hamper tumor progression. Indeed, the development of tumor cell populations with metastatic potential is a critical hallmark of cancer. There are several therapeutic strategies directed against the tumor microenvironment to avoid CTC survival and differentiation to more aggressive phenotypes, which are shown in Table 1.

6.1. CTCs as Therapeutic Targets in HCC

The approved multikinase inhibitors for advanced HCC may affect the viability of CTCs. Adjuvant therapy with sorafenib after surgical resection could eliminate residual CTCs to reduce the risk of postoperative recurrence and metastases [145]. pERK+/pAkt CTCs are vulnerable to sorafenib and their clearance is associated with improved progression-free survival [23]. However, the most effective therapy against CTCs would be their physical removal before they can migrate to other organs and produce metastases. The Viatar cancer dialysis system (Viatar CTC Solutions Inc., Lowell, MA, USA), the temporary indwelling intravascular CTC isolation system [126], the GILUPI CellCollector® [127] or the black phosphorus and antibody-functionalized intravenous catheter [128] are different technologies designed for in vivo removal and destruction of EpCAM+ CTCs [128]. Most of them were designed to allow the screening of a large volume of blood and they could be used as adjuvant therapies after surgical resection or locoregional ablation. Although each of these systems claims for inherent technical advantages, additional studies are needed to determine their safety and efficacy in vivo.
Gene therapy and immunotherapy have also been proposed as promising tools to selectively target resistant subpopulations of CTCs. Immune checkpoint inhibitors in combination with epidermal growth factor receptor (EGFR) inhibitors (atezolizumab/bevacizumab) have recently become first-line therapies in patients with advanced HCC, as they have demonstrated improved progression-free and overall survival rates as compared with sorafenib, the previous standard of care [146]. Anti-programmed cell death protein 1 (PD-1) therapy could target PD-L1+ CTCs, which are associated with more aggressive disease [129]. Inhibition of EGFR signaling reduces total CTC count and metastases after local ablation [130,147]. There are other potential therapeutic targets able to interfere with the viability of CTCs which could reshape future therapeutic algorithms in HCC. Major vault protein (MVP) depletion by siRNA technology or by anti-MVP antibodies reduces survival, migratory capacity, and invasiveness of HCC cells [19]. Other strategies to target CTCs, either as a whole population or as subpopulations with particularly aggressive phenotypes are depletion of ubiquitin-specific protease 1 [133], inhibition of Intercellular Adhesion Molecule 1 (ICAM-1) using a plasmid expressing a short hairpin RNA [97], blockade of CD44 by specific antibodies [93,131] and promotion of the expression of androgen receptor, which suppresses CD90 expression and cell migration and increases cell death [132].
Interfering with the EMT activation or the colonization of CTCs could be therapeutically attractive to prevent the extrahepatic spread of HCC [84,134]. Tacrolimus, an immunosuppressant commonly used to prevent allograft rejection after LT, has been associated with the induction of motility and invasiveness of HCC cells in vitro and it is able to increase the risk of tumor recurrence in a dose-dependent manner [100]. One of the main mechanisms underlying this clinical observation is the activation of the Rho/ROCK signaling pathway. The administration of Y-27632, a ROCK-specific inhibitor, prevents tumor recurrence after LT in an animal model of HCC [135,148]. However, this mechanism should be further investigated since ROCK inhibition also promotes survival and expansion of tumor stem cells in vitro [149].

6.2. Tumor Microenvironment as a Therapeutic Target in HCC

Many interactions or communication factors are forming the pre-metastatic niche, which promotes the arrival, survival, and growth of tumor cells. Targeting this crosstalk could prevent extrahepatic spreading of HCC. The SDF-1/CXCR4 axis participates in the acquisition of stemness and migratory potential of HCC cells and could be considered a potential target to prevent metastases [136], with promising results obtained in preclinical studies targeting CXCR4 [137,138] or SDF-1 [139] by specific inhibitors or gene expressing traps respectively. MiRNAs can be released by many cell types into the extracellular space where they regulate different processes. Thus, the tumor-derived exosomal miR-1247-3p transforms fibroblasts into cancer-associated fibroblasts to promote lung metastases of liver cancer [140]. MiR-155 released in exosomes by HCC cells stimulates cell colony formation, migration, and invasion in vitro [141] and promotes HCC proliferation in vivo [150]. Similarly, SMAD Family Member 3-enriched exosomes derived from primary tumors promote the metastatic potential of CTCs [142].
Other interactions between CTCs and the tumor microenvironment involve the immune system [151,152,153], and particularly the adhesive myeloid-derived suppressor cells, tumor-associated macrophages, and regulatory T cells (Tregs). These interactions protect CTCs from immune responses, thus increasing their metastatic potential. Liver sinusoidal endothelial cells are the major liver cell type responsible for TGF-β-dependent hepatic Treg induction and immune tolerance [154]. The immunosuppressive chemokine CCL5 recruits Tregs to the tumor microenvironment to promote immune escape [155]. This may explain the correlation between the number of Tregs and CTC count [24]. Moreover, Treg could in turn induce CCL5 expression in CTCs by secretion of TGF-β1 [21]. Targeting liver sinusoidal endothelial cells [143] or blocking the CCL5 pathway [21,155] could interfere in this feedback, thus inducing a hostile microenvironment [144] which hampers CTC survival and metastatic colonization.

7. Future Perspectives

Liquid biopsy, and particularly quantitative and qualitative assessment of CTCs, will probably reshape the future diagnostic and therapeutic algorithms of HCC. This valuable information may be easily obtained from low-volume peripheral blood samples, either with diagnostic, therapeutic purposes or even to stratify patients according to the biological aggressiveness of the primary tumor. However, current methodologies pose important caveats which need to be addressed before this technology is implemented in routine clinical practice. First, the optimal combination of markers for the enrichment of CTCs, able to detect only viable cells with metastatic potential, is unknown. Most studies have a limited sample size, lack external validation and their results become obsolete shortly after their publication due to the vertiginous technological development. Secondly, isolating CTCs for ulterior culture and evaluation of their mutational profile is challenging and very inefficient under current strategies. The development of optimal culture conditions to expand CTCs is paramount to evaluate their resistance to available therapies and to advance towards truly personalized medicine in HCC. Finally, CTC counting is a time-consuming process that requires highly specialized personnel and expensive technology. Blood samples should be analyzed as soon as possible after the extraction and storing samples longer than 24 h could threaten the viability of CTCs. The whole process should be automatized, and costs reduced so this technology is made available in all institutions taking care of patients with HCC. Only then, liquid biopsy could be incorporated as part of the diagnostic and therapeutic armamentarium of HCC.

Author Contributions

Conceptualization, M.D.l.M., M.R.-P. and G.F.; literature search, M.L.E.-C., S.G.-R., J.Z.-O., V.A.-T., R.A., M.S.-F., R.C., M.R.-P. and G.F.; writing—original draft preparation, M.L.E.-C., S.G.-R., M.R.-P. and G.F.; writing—review and editing, G.F. and M.R.-P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the “Plan Nacional de I+D+I Proyectos de Investigación en Salud of Instituto de Salud Carlos III (ISCIII), Subdirección General de Evaluación and Fondo Europeo de Desarrollo Regional (FEDER)” (grant number PI18/01736). G.F. was supported by the CIBERehd. S.G.-R. is supported by the “Consejería de Salud y Familias de la Junta de Andalucía and Fondo Social Europeo” (grant number RH-0075-2020). M.L.E.-C. was supported by the ISCIII (grant number FI19/00357).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Reig, M.; Forner, A.; Ávila, M.A.; Ayuso, C.; Mínguez, B.; Varela, M.; Bilbao, I.; Bilbao, J.I.; Burrel, M.; Bustamante, J.; et al. Diagnosis and Treatment of Hepatocellular Carcinoma. Update of the Consensus Document of the AEEH, AEC, SEOM, SERAM, SERVEI, and SETH. Medicina Clínica 2021, 156, 463.e1–463.e30. [Google Scholar] [CrossRef]
  4. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 2021, 7, 6. [Google Scholar] [CrossRef]
  5. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef]
  6. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Hernandez–Gea, V.; Toffanin, S.; Friedman, S.L.; Llovet, J.M. Role of the Microenvironment in the Pathogenesis and Treatment of Hepatocellular Carcinoma. Gastroenterology 2013, 144, 512–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Llovet, J.M.; Brú, C.; Bruix, J. Prognosis of Hepatocellular Carcinoma: The BCLC Staging Classification. Semin. Liver Dis. 1999, 19, 329–338. [Google Scholar] [CrossRef] [PubMed]
  9. Reig, M.; Darnell, A.; Forner, A.; Rimola, J.; Ayuso, C.; Bruix, J. Systemic Therapy for Hepatocellular Carcinoma: The Issue of Treatment Stage Migration and Registration of Progression Using the BCLC-Refined RECIST. Semin. Liver Dis. 2014, 34, 444–455. [Google Scholar] [CrossRef]
  10. European Association for the Study of the Liver. European Organisation for Research and Treatment of Cancer. EASL–EORTC clinical practice guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2012, 56, 908–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Yang, J.D.; Kim, W.R. Surveillance for Hepatocellular Carcinoma in Patients with Cirrhosis. Clin. Gastroenterol. Hepatol. 2012, 10, 16–21. [Google Scholar] [CrossRef]
  12. Yang, J.D.; Mannalithara, A.; Piscitello, A.J.; Kisiel, J.B.; Gores, G.J.; Roberts, L.R.; Kim, W.R. Impact of surveillance for hepatocellular carcinoma on survival in patients with compensated cirrhosis. Hepatology 2018, 68, 78–88. [Google Scholar] [CrossRef]
  13. Mocan, T.; Simão, A.L.; Castro, R.E.; Rodrigues, C.M.P.; Słomka, A.; Wang, B.; Strassburg, C.; Wöhler, A.; Willms, A.G.; Kornek, M. Liquid Biopsies in Hepatocellular Carcinoma: Are We Winning? J. Clin. Med. 2020, 9, 1541. [Google Scholar] [CrossRef] [PubMed]
  14. Von Felden, J.; Garcia-Lezana, T.; Schulze, K.; Losic, B.; Villanueva, A. Liquid biopsy in the clinical management of hepatocellular carcinoma. Gut 2020, 69, 2025–2034. [Google Scholar] [CrossRef] [PubMed]
  15. Ashworth, T.R. A Case of Cancer in Which Cells Similar to Those in the Tumours Were Seen in the Blood after Death. Aust. Med. J. 1869, 14, 146. [Google Scholar]
  16. Parkinson, D.R.; Dracopoli, N.; Petty, B.G.; Compton, C.; Cristofanilli, M.; Deisseroth, A.; Hayes, D.F.; Kapke, G.; Kumar, P.; Lee, J.S.; et al. Considerations in the development of circulating tumor cell technology for clinical use. J. Transl. Med. 2012, 10, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Okajima, W.; Komatsu, S.; Ichikawa, D.; Miyamae, M.; Ohashi, T.; Imamura, T.; Kiuchi, J.; Nishibeppu, K.; Arita, T.; Konishi, H.; et al. Liquid biopsy in patients with hepatocellular carcinoma: Circulating tumor cells and cell-free nucleic acids. World J. Gastroenterol. 2017, 23, 5650–5668. [Google Scholar] [CrossRef] [PubMed]
  18. 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]
  19. Lee, H.M.; Joh, J.W.; Seong, C.G.; Kim, W.-T.; Kim, M.K.; Choi, H.S.; Kim, S.Y.; Jang, Y.-J.; Sinn, D.H.; Choi, G.S.; et al. Cell-surface major vault protein promotes cancer progression through harboring mesenchymal and intermediate circulating tumor cells in hepatocellular carcinomas. Sci. Rep. 2017, 7, 13201. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, P.; Xu, Y.; Sun, Y.; Cheng, J.; Zhou, K.; Wu, S.; Hu, B.; Zhang, Z.; Guo, W.; Cao, Y.; et al. Detection of circulating tumour cells enables early recurrence prediction in hepatocellular carcinoma patients undergoing liver transplantation. Liver Int. 2021, 41, 562–573. [Google Scholar] [CrossRef]
  21. 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, 1–14. [Google Scholar] [CrossRef]
  22. Ogle, L.F.; Orr, J.G.; Willoughby, C.; Hutton, C.; McPherson, S.; Plummer, R.; Boddy, A.; Curtin, N.; Jamieson, D.; Reeves, H.L. Imagestream detection and characterisation of circulating tumour cells—A liquid biopsy for hepatocellular carcinoma? J. Hepatol. 2016, 65, 305–313. [Google Scholar] [CrossRef] [Green Version]
  23. Li, J.; Shi, L.; Zhang, X.; Sun, B.; Yang, Y.; Ge, N.; Liu, H.; Yang, X.; Chen, L.; Qian, H.; et al. pERK/pAkt phenotyping in circulating tumor cells as a biomarker for sorafenib efficacy in patients with advanced hepatocellular carcinoma. Oncotarget 2015, 7, 2646–2659. [Google Scholar] [CrossRef] [Green Version]
  24. Zhou, Y.; Wang, B.; Wu, J.; Zhang, C.; Zhou, Y.; Yang, X.; Zhou, J.; Guo, W.; Fan, J. Association of preoperative EpCAM Circulating Tumor Cells and peripheral Treg cell levels with early recurrence of hepatocellular carcinoma following radical hepatic resection. BMC Cancer 2016, 16, 506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. 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] [PubMed] [Green Version]
  26. Guo, W.; Yang, X.-R.; Sun, Y.-F.; Shen, M.-N.; Ma, X.-L.; Wu, J.; Zhang, C.-Y.; Zhou, Y.; Xu, Y.; Hu, B.; et al. Clinical Significance of EpCAM mRNA-Positive Circulating Tumor Cells in Hepatocellular Carcinoma by an Optimized Negative Enrichment and qRT-PCR–Based Platform. Clin. Cancer Res. 2014, 20, 4794–4805. [Google Scholar] [CrossRef] [Green Version]
  27. He, Y.Z.; He, K.; Huang, R.Q.; Wang, Z.L.; Ye, S.W.; Liu, L.W.; Luo, Q.J.; Hu, Z.M. Preoperative evaluation and prediction of clinical scores for hepatocellular carcinoma microvascular invasion: A single-center retrospective analysis. Ann. Hepatol. 2020, 19, 654–661. [Google Scholar] [CrossRef] [PubMed]
  28. He, Y.Z.; He, K.; Huang, R.Q.; Liu, L.W.; Ye, S.W.; Qian, J.L.; Peng, P.; Luo, Q.J.; Wang, Z.L.; Hu, Z.M. A clinical scoring system for predicting tumor recurrence after percutaneous radiofrequency ablation for 3 cm or less hepatocellular carcinoma. Sci. Rep. 2021, 11, 1–14. [Google Scholar] [CrossRef]
  29. Chen, Z.; Lin, X.; Chen, C.; Chen, Y.; Zhao, Q.; Wu, L.; Wang, D.; Ma, Y.; Ju, W.; Chen, M.; et al. Analysis of preoperative circulating tumor cells for recurrence in patients with hepatocellular carcinoma after liver transplantation. Ann. Transl. Med. 2020, 8, 1067. [Google Scholar] [CrossRef]
  30. 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]
  31. Deng, J.; Chen, W.; Wu, X.; Zhou, Y.; Li, J. Evaluating the predictive power of circulating tumor cells for the prognosis of transarterial chemoembolization treatment on patients with advanced hepatocellular carcinoma. Medicine 2021, 100, e24060. [Google Scholar] [CrossRef]
  32. Wu, X.; Yang, C.; Yu, H.; Cao, F.; Shan, Y.; Zhao, W. The predictive values of serum dickkopf-1 and circulating tumor cells in evaluating the efficacy of transcatheter arterial chemoembolization treatment on hepatocellular carcinoma. Medicine 2019, 98, e16579. [Google Scholar] [CrossRef]
  33. Liu, Y.-K.; Hu, B.-S.; Li, Z.-L.; He, X.; Li, Y.; Lu, L.-G. An improved strategy to detect the epithelial-mesenchymal transition process in circulating tumor cells in hepatocellular carcinoma patients. Hepatol. Int. 2016, 10, 640–646. [Google Scholar] [CrossRef]
  34. 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]
  35. Morris, K.L.; Tugwood, J.D.; Khoja, L.; Lancashire, M.; Sloane, R.S.; Burt, D.; Shenjere, P.; Zhou, C.; Hodgson, C.; Ohtomo, T.; et al. Circulating biomarkers in hepatocellular carcinoma. Cancer Chemother. Pharmacol. 2014, 74, 323–332. [Google Scholar] [CrossRef]
  36. 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]
  37. Ye, X.; Li, G.; Han, C.; Han, Q.; Shang, L.; Su, H.; Han, B.; Gong, Y.; Lu, G.; Peng, T. Circulating tumor cells as a potential biomarker for postoperative clinical outcome in HBV-related hepatocellular carcinoma. Cancer Manag. Res. 2018, 10, 5639–5647. [Google Scholar] [CrossRef]
  38. Takahashi, K.; Ofuji, K.; Hiramatsu, K.; Nosaka, T.; Naito, T.; Matsuda, H.; Endo, K.; Higuchi, M.; Ohtani, M.; Nemoto, T.; et al. Circulating tumor cells detected with a microcavity array predict clinical outcome in hepatocellular carcinoma. Cancer Med. 2021, 10, 2300–2309. [Google Scholar] [CrossRef] [PubMed]
  39. Chen, Y.; Li, S.; Li, W.; Yang, R.; Zhang, X.; Ye, Y.; Yu, J.; Ye, L.; Tang, W. Circulating tumor cells undergoing EMT are poorly correlated with clinical stages or predictive of recurrence in hepatocellular carcinoma. Sci. Rep. 2019, 9, 7084. [Google Scholar] [CrossRef] [Green Version]
  40. 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] [Green Version]
  41. Luo, Q.; Wang, C.; Peng, B.; Pu, X.; Cai, L.; Liao, H.; Chen, K.; Zhang, C.; Cheng, Y.; Pan, M. Circulating Tumor-Cell-Associated White Blood Cell Clusters in Peripheral Blood Indicate Poor Prognosis in Patients With Hepatocellular Carcinoma. Front. Oncol. 2020, 10, 1758. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, C.; Luo, Q.; Huang, W.; Zhang, C.; Liao, H.; Chen, K.; Pan, M. Correlation Between Circulating Tumor Cell DNA Genomic Alterations and Mesenchymal CTCs or CTC-Associated White Blood Cell Clusters in Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 686365. [Google Scholar] [CrossRef]
  43. Wang, Z.; Luo, L.; Cheng, Y.; Ze-Sheng, J.; Peng, B.; Gao, Y.; Jiang, Z.-S.; Pan, M. Correlation Between Postoperative Early Recurrence of Hepatocellular Carcinoma and Mesenchymal Circulating Tumor Cells in Peripheral Blood. J. Gastrointest. Surg. 2018, 22, 633–639. [Google Scholar] [CrossRef] [Green Version]
  44. Cheng, Y.; Luo, L.; Zhang, J.; Zhou, M.; Tang, Y.; He, G.; Lu, Y.; Wang, Z.; Pan, M. Diagnostic Value of Different Phenotype Circulating Tumor Cells in Hepatocellular Carcinoma. J. Gastrointest. Surg. 2019, 23, 2354–2361. [Google Scholar] [CrossRef]
  45. 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 Biomarkers 2017, 20, 487–498. [Google Scholar] [CrossRef]
  46. Li, Y.; Huang, N.; Wang, C.; Ma, H.; Zhou, M.; Lin, L.; Huang, Z.; Sun, L.; Shi, M.; Liao, W. Impact of liver tumor percutaneous radiofrequency ablation on circulating tumor cells. Oncol. Lett. 2018, 16, 2839–2850. [Google Scholar] [CrossRef]
  47. Vona, G.; Sabile, A.; Louha, M.; Sitruk, V.; Romana, S.P.; Schütze, K.; Capron, F.; Franco, D.; Pazzagli, M.; Vekemans, M.; et al. Isolation by Size of Epithelial Tumor Cells: A New Method for the Immunomorphological and Molecular Characterization of Circulating Tumor Cells. Am. J. Pathol. 2000, 156, 57–63. [Google Scholar] [CrossRef]
  48. 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]
  49. Zhou, J.; Zhang, Z.; Zhou, H.; Leng, C.; Hou, B.; Zhou, C.; Hu, X.; Wang, J.; Chen, X. Preoperative circulating tumor cells to predict microvascular invasion and dynamical detection indicate the prognosis of hepatocellular carcinoma. BMC Cancer 2020, 20, 1047. [Google Scholar] [CrossRef]
  50. Yin, L.-C.; Luo, Z.-C.; Gao, Y.-X.; Li, Y.; Peng, Q.; Gao, Y. Twist Expression in Circulating Hepatocellular Carcinoma Cells Predicts Metastasis and Prognoses. BioMed Res. Int. 2018, 2018, 3789613. [Google Scholar] [CrossRef]
  51. Bankó, P.; Lee, S.Y.; Nagygyörgy, V.; Zrínyi, M.; Chae, C.H.; Cho, D.H.; Telekes, A. Technologies for circulating tumor cell separation from whole blood. J. Hematol. Oncol. 2019, 12, 1–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Court, C.M.; Hou, S.; Winograd, P.; Segel, N.H.; Li, Q.W.; Zhu, Y.; Sadeghi, S.; Finn, R.S.; Ganapathy, E.; Song, M.; et al. A novel multimarker assay for the phenotypic profiling of circulating tumor cells in hepatocellular carcinoma. Liver Transplant. 2018, 24, 946–960. [Google Scholar] [CrossRef]
  53. Yu, J.-J.; Xiao, W.; Dong, S.-L.; Liang, H.-F.; Zhang, Z.-W.; Zhang, B.-X.; Huang, Z.-Y.; Chen, Y.-F.; Zhang, W.-G.; Luo, H.-P.; et al. Effect of surgical liver resection on circulating tumor cells in patients with hepatocellular carcinoma. BMC Cancer 2018, 18, 835. [Google Scholar] [CrossRef]
  54. Shen, J.; Wang, W.-S.; Zhu, X.-L.; Ni, C.-F. High Epithelial Cell Adhesion Molecule–Positive Circulating Tumor Cell Count Predicts Poor Survival of Patients with Unresectable Hepatocellular Carcinoma Treated with Transcatheter Arterial Chemoembolization. J. Vasc. Interv. Radiol. 2018, 29, 1678–1684. [Google Scholar] [CrossRef]
  55. Von Felden, J.; Schulze, K.; Krech, T.; Ewald, F.; Nashan, B.; Pantel, K.; Lohse, A.W.; Riethdorf, S.; Wege, H. Circulating tumor cells as liquid biomarker for high HCC recurrence risk after curative liver resection. Oncotarget 2017, 8, 89978–89987. [Google Scholar] [CrossRef] [Green Version]
  56. Sun, Y.; Wang, P.; Cheng, J.; 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]
  57. Ge, Z.; Helmijr, J.C.; Jansen, M.P.; Boor, P.P.; Noordam, L.; Peppelenbosch, M.; Kwekkeboom, J.; Kraan, J.; Sprengers, D. Detection of oncogenic mutations in paired circulating tumor DNA and circulating tumor cells in patients with hepatocellular carcinoma. Transl. Oncol. 2021, 14, 101073. [Google Scholar] [CrossRef]
  58. Schulze, K.; Gasch, C.; Staufer, K.; Nashan, B.; Lohse, A.W.; Pantel, K.; Riethdorf, S.; Wege, H. Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma. Int. J. Cancer 2013, 133, 2165–2171. [Google Scholar] [CrossRef]
  59. Kelley, R.K.; Magbanua, M.J.M.; Butler, T.M.; Collisson, E.A.; Hwang, J.; Sidiropoulos, N.; Evason, K.; McWhirter, R.M.; Hameed, B.; Wayne, E.M.; et al. Circulating tumor cells in hepatocellular carcinoma: A pilot study of detection, enumeration, and next-generation sequencing in cases and controls. BMC Cancer 2015, 15, 206. [Google Scholar] [CrossRef]
  60. Sun, Y.-F.; Xu, Y.; Yang, X.-R.; Guo, W.; Zhang, X.; Qiu, S.-J.; Shi, R.-Y.; Hu, B.; Zhou, J.; Fan, J. Circulating stem cell-like epithelial cell adhesion molecule-positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection. Hepatology 2013, 57, 1458–1468. [Google Scholar] [CrossRef]
  61. Wang, P.; Sun, Y.; Zhou, K.; Cheng, J.; Hu, B.; Guo, W.; Yin, Y.; Huang, J.; Zhou, J.; Fan, J.; et al. Circulating tumor cells are an indicator for the administration of adjuvant transarterial chemoembolization in hepatocellular carcinoma: A single-center, retrospective, propensity-matched study. Clin. Transl. Med. 2020, 10, e137. [Google Scholar] [CrossRef] [PubMed]
  62. 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]
  63. Fischer, J.; Niederacher, D.; Topp, S.A.; Honisch, E.; Schumacher, S.; Schmitz, N.; Fohrding, L.Z.; Vay, C.; Hoffmann, I.; Kasprowicz, N.S.; et al. Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. Proc. Natl. Acad. Sci. USA 2013, 110, 16580–16585. [Google Scholar] [CrossRef] [Green Version]
  64. Sánchez-Lorencio, M.; Ramirez, P.; Saenz, L.; Sánchez, M.D.R.G.; De La Orden, V.; Mediero-Valeros, B.; Veganzones, S.; Baroja-Mazo, A.; Nuin, B.C.R.; Gonzalez, M.; et al. Comparison of Two Types of Liquid Biopsies in Patients With Hepatocellular Carcinoma Awaiting Orthotopic Liver Transplantation. Transplant. Proc. 2015, 47, 2639–2642. [Google Scholar] [CrossRef]
  65. Tsuchiya, N. Biomarkers for the early diagnosis of hepatocellular carcinoma. World J. Gastroenterol. 2015, 21, 10573–10583. [Google Scholar] [CrossRef]
  66. Rodríguez-Perálvarez, M.; Luong, T.V.; Andreana, L.; Meyer, T.; Dhillon, A.P.; Burroughs, A.K. A Systematic Review of Microvascular Invasion in Hepatocellular Carcinoma: Diagnostic and Prognostic Variability. Ann. Surg. Oncol. 2012, 20, 325–339. [Google Scholar] [CrossRef]
  67. Hamaoka, M.; Kobayashi, T.; Tanaka, Y.; Mashima, H.; Ohdan, H. Clinical significance of glypican-3-positive circulating tumor cells of hepatocellular carcinoma patients: A prospective study. PLoS ONE 2019, 14, e0217586. [Google Scholar] [CrossRef] [Green Version]
  68. Spiess, M. The asialoglycoprotein receptor: A model for endocytic transport receptors. Biochemistry 1990, 29, 10009–10018. [Google Scholar] [CrossRef]
  69. Fan, Z.; Van De Rijn, M.; Montgomery, K.; Rouse, R.V. Hep Par 1 Antibody Stain for the Differential Diagnosis of Hepatocellular Carcinoma: 676 Tumors Tested Using Tissue Microarrays and Conventional Tissue Sections. Mod. Pathol. 2003, 16, 137–144. [Google Scholar] [CrossRef] [Green Version]
  70. Xu, W.; Cao, L.; Chen, L.; Li, J.; Zhang, X.-F.; Qian, H.-H.; Kang, X.-Y.; Zhang, Y.; Liao, J.; Shi, L.-H.; et al. Isolation of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma Using a Novel Cell Separation Strategy. Clin. Cancer Res. 2011, 17, 3783–3793. [Google Scholar] [CrossRef] [Green Version]
  71. Li, J.; Chen, L.; Zhang, X.; Zhang, Y.; Liu, H.; Sun, B.; Zhao, L.; Ge, N.; Qian, H.; Yang, Y.; et al. Detection of Circulating Tumor Cells in Hepatocellular Carcinoma Using Antibodies against Asialoglycoprotein Receptor, Carbamoyl Phosphate Synthetase 1 and Pan-Cytokeratin. PLoS ONE 2014, 9, e96185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, Y.; Zhang, X.; Zhang, J.; Sun, B.; Zheng, L.; Li, J.; Liu, S.; Sui, G.; Yin, Z. Microfluidic chip for isolation of viable circulating tumor cells of hepatocellular carcinoma for their culture and drug sensitivity assay. Cancer Biol. Ther. 2016, 17, 1177–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wan, S.; Kim, T.H.; Smith, K.J.; Delaney, R.; Park, G.-S.; Guo, H.; Lin, E.; Plegue, T.; Kuo, N.; Steffes, J.; et al. New Labyrinth Microfluidic Device Detects Circulating Tumor Cells Expressing Cancer Stem Cell Marker and Circulating Tumor Microemboli in Hepatocellular Carcinoma. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef] [PubMed]
  74. Mu, H. Identification of biomarkers for hepatocellular carcinoma by semiquantitative immunocytochemistry. World J. Gastroenterol. 2014, 20, 5826–5838. [Google Scholar] [CrossRef] [PubMed]
  75. Gires, O.; Stoecklein, N.H. Dynamic EpCAM expression on circulating and disseminating tumor cells: Causes and consequences. Cell. Mol. Life Sci. 2014, 71, 4393–4402. [Google Scholar] [CrossRef] [PubMed]
  76. Ramirez, P.; Sáenz, L.; Cascales-Campos, P.; Sánchez, M.G.; Llàcer-Millán, E.; Sánchez-Lorencio, M.; Díaz-Rubio, E.; De La Orden, V.; Mediero-Valeros, B.; Navarro, J.; et al. Oncological Evaluation by Positron-emission Tomography, Circulating Tumor Cells and Alpha Fetoprotein in Patients with Hepatocellular Carcinoma on the Waiting List for Liver Transplantation. Transplant. Proc. 2016, 48, 2962–2965. [Google Scholar] [CrossRef] [PubMed]
  77. De Wit, S.; Manicone, M.; Rossi, E.; Lampignano, R.; Yang, L.; Zill, B.; Rengel-Puertas, A.; Ouhlen, M.; Crespo, M.; Berghuis, A.M.S.; et al. EpCAMhigh and EpCAMlow circulating tumor cells in metastatic prostate and breast cancer patients. Oncotarget 2018, 9, 35705–35716. [Google Scholar] [CrossRef] [Green Version]
  78. Nicolazzo, C.; Gradilone, A.; Loreni, F.; Raimondi, C.; Gazzaniga, P. EpCAMlow Circulating Tumor Cells: Gold in the Waste. Dis. Markers 2019, 2019, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Giannelli, G.; Koudelkova, P.; Dituri, F.; Mikulits, W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J. Hepatol. 2016, 65, 798–808. [Google Scholar] [CrossRef] [Green Version]
  80. Terris, B.; Cavard, C.; Perret, C. EpCAM, a new marker for cancer stem cells in hepatocellular carcinoma. J. Hepatol. 2010, 52, 280–281. [Google Scholar] [CrossRef] [Green Version]
  81. Yamashita, T.; Ji, J.; Budhu, A.; Forgues, M.; Yang, W.; Wang, H.; Jia, H.; Ye, Q.; Qin, L.; Wauthier, E.; et al. EpCAM-Positive Hepatocellular Carcinoma Cells Are Tumor-Initiating Cells With Stem/Progenitor Cell Features. Gastroenterology 2009, 136, 1012–1024. [Google Scholar] [CrossRef] [Green Version]
  82. Zhang, Y.; Weinberg, R.A. Epithelial-to-mesenchymal transition in cancer: Complexity and opportunities. Front. Med. 2018, 12, 361–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef] [PubMed]
  84. 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. 2017, 24, 547–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sha, Y.; Haensel, D.; Gutierrez, G.; Du, H.; Dai, X.; Nie, Q. Intermediate cell states in epithelial-to-mesenchymal transition. Phys. Biol. 2019, 16, 021001. [Google Scholar] [CrossRef] [PubMed]
  86. Li, Y.-M.; Xu, S.-C.; Li, J.; Han, K.-Q.; Pi, H.-F.; Zheng, L.; Zuo, G.-H.; Huang, X.-B.; Li, H.-Y.; Zhao, H.-Z.; et al. Epithelial–mesenchymal transition markers expressed in circulating tumor cells in hepatocellular carcinoma patients with different stages of disease. Cell Death Dis. 2013, 4, e831. [Google Scholar] [CrossRef] [PubMed]
  87. Hong, Y.; Fang, F.; Zhang, Q. Circulating tumor cell clusters: What we know and what we expect (Review). Int. J. Oncol. 2016, 49, 2206–2216. [Google Scholar] [CrossRef] [Green Version]
  88. Duda, D.G.; Duyverman, A.M.M.J.; Kohno, M.; Snuderl, M.; Steller, E.J.A.; Fukumura, D.; Jain, R.K. Malignant cells facilitate lung metastasis by bringing their own soil. Proc. Natl. Acad. Sci. USA 2010, 107, 21677–21682. [Google Scholar] [CrossRef] [Green Version]
  89. Tayoun, T.; Faugeroux, V.; Oulhen, M.; Aberlenc, A.; Pawlikowska, P.; Farace, F. CTC-Derived Models: A Window into the Seeding Capacity of Circulating Tumor Cells (CTCs). Cells 2019, 8, 1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Lara-Pezzi, E.; Serrador, J.M.; Montoya, M.C.; Zamora, D.; Yáñez-Mó, M.; Carretero, M.; Furthmayr, H.; Sánchez-Madrid, F.; López-Cabrera, M. The hepatitis B virus X protein (HBx) induces a migratory phenotype in a CD44-dependent manner: Possible role of HBx in invasion and metastasis. Hepatology 2001, 33, 1270–1281. [Google Scholar] [CrossRef]
  91. Zhu, Z.; Hao, X.; Yan, M.; Yao, M.; Ge, C.; Gu, J.; Li, J. Cancer stem/progenitor cells are highly enriched in CD133+CD44+population in hepatocellular carcinoma. Int. J. Cancer 2009, 126, 2067–2078. [Google Scholar] [CrossRef]
  92. Zahran, A.M.; Abdel-Rahim, M.; Refaat, A.; Sayed, M.; Othman, M.M.; Khalak, L.M.R.; Hetta, H.F. Circulating hematopoietic stem cells, endothelial progenitor cells and cancer stem cells in hepatocellular carcinoma patients: Contribution to diagnosis and prognosis. Acta Oncol. 2020, 59, 33–39. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, Z.F.; Ho, D.W.; Ng, M.N.; Lau, C.K.; Yu, W.C.; Ngai, P.; Chu, P.W.; Lam, C.T.; Poon, R.T.; Fan, S.T. Significance of CD90+ Cancer Stem Cells in Human Liver Cancer. Cancer Cell 2008, 13, 153–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhu, L.; Zhang, W.; Wang, J.; Liu, R. Evidence of CD90+CXCR4+ cells as circulating tumor stem cells in hepatocellular carcinoma. Tumor Biol. 2015, 36, 5353–5360. [Google Scholar] [CrossRef] [PubMed]
  95. Govaere, O.; Komuta, M.; Berkers, J.; Spee, B.; Janssen, C.; de Luca, F.; Katoonizadeh, A.; Wouters, J.; van Kempen, L.C.; Durnez, A.; et al. Keratin 19: A key role player in the invasion of human hepatocellular carcinomas. Gut 2014, 63, 674–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Hwang, H.S.; Yoo, J.E.; Han, D.H.; Choi, J.S.; Lee, J.G.; Joo, D.J.; Kim, M.S.; Kim, S.I.; Choi, G.H.; Park, A.Y.N. Circulating Cancer Stem Cells Expressing EpCAM/CD90 in Hepatocellular Carcinoma: A Pilot Study for Predicting Tumor Recurrence after Living Donor Liver Transplantation. Gut Liver 2021. [Google Scholar] [CrossRef]
  97. Liu, S.; Li, N.; Yu, X.; Xiao, X.; Cheng, K.; Hu, J.; Wang, J.; Zhang, D.; Cheng, S.; Liu, S. Expression of Intercellular Adhesion Molecule 1 by Hepatocellular Carcinoma Stem Cells and Circulating Tumor Cells. Gastroenterology 2013, 144, 1031–1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Amado, V.; González-Rubio, S.; Zamora, J.; Alejandre, R.; Espejo-Cruz, M.; Linares, C.; Sánchez-Frías, M.; García-Jurado, G.; Montero, J.; Ciria, R.; et al. Clearance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma Undergoing Surgical Resection or Liver Transplantation. Cancers 2021, 13, 2476. [Google Scholar] [CrossRef]
  99. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  100. Rodríguez-Perálvarez, M.; Tsochatzis, E.; Naveas, M.C.; Pieri, G.; García-Caparrós, C.; O’Beirne, J.; Poyato-González, A.; Ferrín, G.; Montero-Álvarez, J.L.; Patch, D.; et al. Reduced exposure to calcineurin inhibitors early after liver transplantation prevents recurrence of hepatocellular carcinoma. J. Hepatol. 2013, 59, 1193–1199. [Google Scholar] [CrossRef] [PubMed]
  101. Toso, C.; Mentha, G.; Majno, P. Liver Transplantation for Hepatocellular Carcinoma: Five Steps to Prevent Recurrence. Arab. Archaeol. Epigr. 2011, 11, 2031–2035. [Google Scholar] [CrossRef]
  102. Wang, J.-H.; Fang, Z.-T.; Wang, G.Z.; Zhang, W.; Qu, X.-D.; Liu, R.; Qian, S.; Zhu, L.; Zhou, B. Transcatheter arterial embolization promotes liver tumor metastasis by increasing the population of circulating tumor cells. OncoTargets Ther. 2013, 6, 1563–1572. [Google Scholar] [CrossRef] [Green Version]
  103. Vogl, T.J.; Riegelbauer, L.J.; Oppermann, E.; Kostantin, M.; Ackermann, H.; Trzmiel, A.; Stein, S.; Eichler, K.; Zharov, V.P.; Roy, D.; et al. Early dynamic changes in circulating tumor cells and prognostic relevance following interventional radiological treatments in patients with hepatocellular carcinoma. PLoS ONE 2021, 16, e0246527. [Google Scholar] [CrossRef]
  104. Fang, Z.-T.; Wang, J.-H.; Zhang, W.; Wang, G.Z.; Yang, G.-W.; Qu, X.-D.; Liu, R.; Qian, S.; Zhu, L.; Zhou, B.; et al. Circulating tumor cells in the central and peripheral venous compartment—Assessing hematogenous dissemination after transarterial chemoembolization of hepatocellular carcinoma. OncoTargets Ther. 2014, 7, 1311–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. 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 2015, 87, 1020–1028. [Google Scholar] [CrossRef] [PubMed]
  106. 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]
  107. Pizon, M.; Schott, D.; Pachmann, U.; Pachmann, K. The number of tumorspheres cultured from peripheral blood is a predictor for presence of metastasis in patients with breast cancer. Oncotarget 2016, 7, 48143–48154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Yu, M.; Bardia, A.; Aceto, N.; Bersani, F.; Madden, M.W.; Donaldson, M.C.; Desai, R.; Zhu, H.; Comaills, V.; Zheng, Z.; et al. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 2014, 345, 216–220. [Google Scholar] [CrossRef] [Green Version]
  109. Cao, L.; Zhou, Y.; Zhai, B.; Liao, J.; Xu, W.; Zhang, R.; Li, J.; Zhang, Y.; Chen, L.; Qian, H.; et al. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol. 2011, 11, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Huo, Y.; Chen, W.S.; Lee, J.; Feng, G.-S.; Newton, I.G. Stress Conditions Induced by Locoregional Therapies Stimulate Enrichment and Proliferation of Liver Cancer Stem Cells. J. Vasc. Interv. Radiol. 2019, 30, 2016–2025. [Google Scholar] [CrossRef] [PubMed]
  111. Hernández, L.E.C.; Eslami-S, Z.; Alix-Panabières, C. Circulating tumor cell as the functional aspect of liquid biopsy to understand the metastatic cascade in solid cancer. Mol. Asp. Med. 2020, 72, 100816. [Google Scholar] [CrossRef] [PubMed]
  112. Grillet, F.; Bayet, E.; Villeronce, O.; Zappia, L.; Lagerqvist, E.L.; Lunke, S.; Charafe-Jauffret, E.; Pham, K.; Molck, C.; Rolland, N.; et al. Circulating tumour cells from patients with colorectal cancer have cancer stem cell hallmarks inex vivoculture. Gut 2017, 66, 1802–1810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Luo, Y.T.; Cheng, J.; Feng, X.; He, S.J.; Wang, Y.W.; Huang, Q. The viable circulating tumor cells with cancer stem cells feature, where is the way out? J. Exp. Clin. Cancer Res. 2018, 37, 1–13. [Google Scholar] [CrossRef]
  114. Zhao, P.; Zhou, W.; Liu, C.; Zhang, H.; Cheng, Z.; Wu, W.; Liu, K.; Hu, H.; Zhong, C.; Zhang, Y.; et al. Establishment and Characterization of a CTC Cell Line from Peripheral Blood of Breast Cancer Patient. J. Cancer 2019, 10, 6095–6104. [Google Scholar] [CrossRef]
  115. Que, Z.; Luo, B.; Zhou, Z.; Dong, C.; Jiang, Y.; Wang, L.; Shi, Q.; Tian, J. Establishment and characterization of a patient-derived circulating lung tumor cell line in vitro and in vivo. Cancer Cell Int. 2019, 19, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hu, C.-L.; Zhang, Y.-J.; Zhang, X.-F.; Fei, X.; Zhang, H.; Li, C.-G.; Sun, B. 3D Culture of Circulating Tumor Cells for Evaluating Early Recurrence and Metastasis in Patients with Hepatocellular Carcinoma. OncoTargets Ther. 2021, 14, 2673–2688. [Google Scholar] [CrossRef] [PubMed]
  117. Abdel-Salam, I.M.; Awadein, N.E.-S.; Ashour, M. Cytotoxicity of Luffa cylindrica (L.) M. Roem extract against circulating cancer stem cells in hepatocellular carcinoma. J. Ethnopharmacol. 2019, 229, 89–96. [Google Scholar] [CrossRef]
  118. Wu, L.-J.; Pan, Y.-D.; Pei, X.-Y.; Chen, H.; Nguyen, S.; Kashyap, A.; Liu, J.; Wu, J. Capturing circulating tumor cells of hepatocellular carcinoma. Cancer Lett. 2012, 326, 17–22. [Google Scholar] [CrossRef] [Green Version]
  119. Yoshikawa, N.; Saito, Y.; Manabe, H.; Nakaoka, T.; Uchida, R.; Furukawa, R.; Muramatsu, T.; Sugiyama, Y.; Kimura, M.; Saito, H. Glucose Depletion Enhances the Stem Cell Phenotype and Gemcitabine Resistance of Cholangiocarcinoma Organoids through AKT Phosphorylation and Reactive Oxygen Species. Cancers 2019, 11, 1993. [Google Scholar] [CrossRef] [Green Version]
  120. Buffa, F.M.; Carmeliet, P.; Giaccia, A.J.; Harris, A.L.; Imtiyaz, H.Z.; Johnson, R.S.; Johnsson, A.S.; Li, Z. Diverse Effects of Hypoxia on Tumor Progression; Springer: Berlin/Heidelberg, Germany, 2010; Volume 345, ISBN 978-3-642-13328-2. [Google Scholar]
  121. Cosse, J.-P. Tumour Hypoxia Affects the Responsiveness of Cancer Cells to Chemotherapy and Promotes Cancer Progression. Anti-Cancer Agents Med. Chem. 2008, 8, 790–797. [Google Scholar] [CrossRef] [PubMed]
  122. Rhee, H.; Nahm, J.H.; Kim, H.; Choi, G.H.; Yoo, J.E.; Lee, H.S.; Koh, M.J.; Park, Y.N. Poor outcome of hepatocellular carcinoma with stemness marker under hypoxia: Resistance to transarterial chemoembolization. Mod. Pathol. 2016, 29, 1038–1049. [Google Scholar] [CrossRef]
  123. Cayrefourcq, L.; Mazard, T.; Joosse, S.; Solassol, J.; Ramos, J.; Assenat, E.; Schumacher, U.; Costes, V.; Maudelonde, T.; Pantel, K.; et al. Establishment and Characterization of a Cell Line from Human Circulating Colon Cancer Cells. Cancer Res. 2015, 75, 892–901. [Google Scholar] [CrossRef] [Green Version]
  124. Wang, Z.; Wu, W.; Wang, Z.; Tang, Y.; Deng, Y.; Xu, L.; Tian, J.; Shi, Q. Ex vivo expansion of circulating lung tumor cells based on one-step microfluidics-based immunomagnetic isolation. Analyst 2016, 141, 3621–3625. [Google Scholar] [CrossRef]
  125. Brungs, D.; Minaei, E.; Piper, A.-K.; Perry, J.; Splitt, A.; Carolan, M.; Ryan, S.; Wu, X.J.; Corde, S.; Tehei, M.; et al. Establishment of novel long-term cultures from EpCAM positive and negative circulating tumour cells from patients with metastatic gastroesophageal cancer. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef]
  126. Kim, T.H.; Wang, Y.; Oliver, C.R.; Thamm, D.; Cooling, L.; Paoletti, C.; Smith, K.J.; Nagrath, S.; Hayes, D.F. A temporary indwelling intravascular aphaeretic system for in vivo enrichment of circulating tumor cells. Nat. Commun. 2019, 10, 1478. [Google Scholar] [CrossRef] [Green Version]
  127. Saucedo-Zeni, N.; Mewes, S.; Niestroj, R.; Gasiorowski, L.; Murawa, D.; Nowaczyk, P.; Tomasi, T.; Weber, E.; Dworacki, G.; Morgenthaler, N.G.; et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int. J. Oncol. 2012, 41, 1241–1250. [Google Scholar] [CrossRef] [Green Version]
  128. Wang, D.; Ge, C.; Liang, W.; Yang, Q.; Liu, Q.; Ma, W.; Shi, L.; Wu, H.; Zhang, Y.; Wu, Z.; et al. In Vivo Enrichment and Elimination of Circulating Tumor Cells by Using a Black Phosphorus and Antibody Functionalized Intravenous Catheter. Adv. Sci. 2020, 7, 2000940. [Google Scholar] [CrossRef]
  129. Winograd, P.; Hou, S.; Court, C.M.; Lee, Y.-T.; Chen, P.-J.; Zhu, Y.; Sadeghi, S.; Finn, R.S.; Teng, P.-C.; Wang, J.J.; et al. Hepatocellular Carcinoma–Circulating Tumor Cells Expressing PD-L1 Are Prognostic and Potentially Associated With Response to Checkpoint Inhibitors. Hepatol. Commun. 2020, 4, 1527–1540. [Google Scholar] [CrossRef] [PubMed]
  130. Zhu, L.; Liu, R.; Zhang, W.; Qian, S.; Wang, J. Application of EGFR inhibitor reduces circulating tumor cells during transcatheter arterial embolization. Clin. Transl. Oncol. 2017, 20, 639–646. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, Z.F.; Ngai, P.; Ho, D.W.; Yu, W.C.; Ng, M.N.; Lau, C.K.; Li, M.L.Y.; Tam, K.H.; Lam, C.T.; Poon, R.T.P.; et al. Identification of local and circulating cancer stem cells in human liver cancer. Hepatology 2008, 47, 919–928. [Google Scholar] [CrossRef] [PubMed]
  132. Lai, H.-C.; Yeh, C.-C.; Jeng, L.-B.; Huang, S.-F.; Liao, P.-Y.; Lei, F.-J.; Cheng, W.-C.; Hsu, C.-L.; Cai, X.; Chang, C.; et al. Androgen receptor mitigates postoperative disease progression of hepatocellular carcinoma by suppressing CD90+ populations and cell migration and by promoting anoikis in circulating tumor cells. Oncotarget 2016, 7, 46448–46465. [Google Scholar] [CrossRef] [Green Version]
  133. Li, Y.; Xu, Y.; Gao, C.; Sun, Y.; Zhou, K.; Wang, P.; Cheng, J.; Guo, W.; Ya, C.; Fan, J.; et al. USP1 Maintains the Survival of Liver Circulating Tumor Cells by Deubiquitinating and Stabilizing TBLR1. Front. Oncol. 2020, 10, 554809. [Google Scholar] [CrossRef]
  134. Lee, D.; Na, J.; Ryu, J.; Kim, H.-J.; Nam, S.H.; Kang, M.; Jung, J.W.; Lee, M.-S.; Song, H.E.; Choi, J.; et al. Interaction of tetraspan(in) TM4SF5 with CD44 promotes self-renewal and circulating capacities of hepatocarcinoma cells. Hepatology 2015, 61, 1978–1997. [Google Scholar] [CrossRef]
  135. Ogawa, T.; Tashiro, H.; Miyata, Y.; Ushitora, Y.; Fudaba, Y.; Kobayashi, T.; Arihiro, K.; Okajima, M.; Asahara, T. Rho-Associated Kinase Inhibitor Reduces Tumor Recurrence After Liver Transplantation in a Rat Hepatoma Model. Arab. Archaeol. Epigr. 2007, 7, 347–355. [Google Scholar] [CrossRef]
  136. Tang, Y.; Lu, Y.; Chen, Y.; Luo, L.; Cai, L.; Peng, B.; Huang, W.; Liao, H.; Zhao, L.; Pan, M. Pre-metastatic niche triggers SDF-1/CXCR4 axis and promotes organ colonisation by hepatocellular circulating tumour cells via downregulation of Prrx1. J. Exp. Clin. Cancer Res. 2019, 38, 1–13. [Google Scholar] [CrossRef] [Green Version]
  137. Gentilini, A.; Rombouts, K.; Galastri, S.; Caligiuri, A.; Mingarelli, E.; Mello, T.; Marra, F.; Mantero, S.; Roncalli, M.; Invernizzi, P.; et al. Role of the stromal-derived factor-1 (SDF-1)–CXCR4 axis in the interaction between hepatic stellate cells and cholangiocarcinoma. J. Hepatol. 2012, 57, 813–820. [Google Scholar] [CrossRef] [PubMed]
  138. Matsusue, R.; Kubo, H.; Hisamori, S.; Okoshi, K.; Takagi, H.; Hida, K.; Nakano, K.; Itami, A.; Kawada, K.; Nagayama, S.; et al. Hepatic Stellate Cells Promote Liver Metastasis of Colon Cancer Cells by the Action of SDF-1/CXCR4 Axis. Ann. Surg. Oncol. 2009, 16, 2645–2653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Goodwin, T.J.; Zhou, Y.; Musetti, S.N.; Liu, R.; Huang, L. Local and transient gene expression primes the liver to resist cancer metastasis. Sci. Transl. Med. 2016, 8, 364ra153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. 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] [Green Version]
  141. Ji, J.; Zheng, X.; Forgues, M.; Yamashita, T.; Wauthier, E.L.; Reid, L.M.; Wen, X.; Song, Y.; Wei, J.S.; Khan, J.; et al. Identification of microRNAs specific for epithelial cell adhesion molecule-positive tumor cells in hepatocellular carcinoma. Hepatology 2015, 62, 829–840. [Google Scholar] [CrossRef] [PubMed]
  142. Fu, Q.; Zhang, Q.; Lou, Y.; Yang, J.; Nie, G.; Chen, Q.; Chen, Y.; Zhang, J.; Wang, J.; Wei, T.; et al. Primary tumor-derived exosomes facilitate metastasis by regulating adhesion of circulating tumor cells via SMAD3 in liver cancer. Oncogene 2018, 37, 6105–6118. [Google Scholar] [CrossRef] [Green Version]
  143. Yang, M.; Zhang, C. The role of liver sinusoidal endothelial cells in cancer liver metastasis. Am. J. Cancer Res. 2021, 11, 1845–1860. [Google Scholar]
  144. Zhuang, P.-Y.; Shen, J.; Zhu, X.-D.; Zhang, J.-B.; Tang, Z.-Y.; Qin, L.-X.; Sun, H.-C. Direct Transformation of Lung Microenvironment by Interferon-α Treatment Counteracts Growth of Lung Metastasis of Hepatocellular Carcinoma. PLoS ONE 2013, 8, e58913. [Google Scholar] [CrossRef] [Green Version]
  145. Zhang, Y.; Shi, Z.-L.; Yang, X.; Yin, Z.-F. Targeting of circulating hepatocellular carcinoma cells to prevent postoperative recurrence and metastasis. World J. Gastroenterol. 2014, 20, 142–147. [Google Scholar] [CrossRef] [PubMed]
  146. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  147. Chen, Y.; Chen, X.; Ding, X.; Wang, Y. Afatinib, an EGFR inhibitor, decreases EMT and tumorigenesis of Huh-7 cells by regulating the ERK-VEGF/MMP9 signaling pathway. Mol. Med. Rep. 2019, 20, 3317–3325. [Google Scholar] [CrossRef] [Green Version]
  148. Xue, F.; Takahara, T.; Yata, Y.; Xia, Q.; Nonome, K.; Shinno, E.; Kanayama, M.; Takahara, S.; Sugiyama, T. Blockade of RhoRho-associated coiled coil-forming kinase signaling can prevent progression of hepatocellular carcinoma in matrix metalloproteinase-dependent manner. Hepatol. Res. 2008, 38, 810–817. [Google Scholar] [CrossRef]
  149. de Sousa, G.R.; Vieira, G.M.; das Chagas, P.F.; Pezuk, J.A.; Brassesco, M.S. Should we keep rocking? Portraits from targeting Rho kinases in cancer. Pharmacol. Res. 2020, 160, 105093. [CrossRef] [PubMed]
  150. Sun, J.-F.; Zhang, D.; Gao, C.-J.; Zhang, Y.-W.; Dai, Q.-S. Exosome-Mediated MiR-155 Transfer Contributes to Hepatocellular Carcinoma Cell Proliferation by Targeting PTEN. Med. Sci. Monit. Basic Res. 2019, 25, 218–228. [Google Scholar] [CrossRef] [PubMed]
  151. Chen, F.; Zhong, Z.; Tan, H.-Y.; Wang, N.; Feng, Y. The Significance of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy. Cancers 2020, 12, 1734. [Google Scholar] [CrossRef]
  152. Liu, Q.; Liao, Q.; Zhao, Y. Myeloid-derived suppressor cells (MDSC) facilitate distant metastasis of malignancies by shielding circulating tumor cells (CTC) from immune surveillance. Med. Hypotheses 2016, 87, 34–39. [Google Scholar] [CrossRef] [PubMed]
  153. Hamilton, G.; Rath, B.; Klameth, L.; Hochmair, M.J. Small cell lung cancer: Recruitment of macrophages by circulating tumor cells. OncoImmunology 2016, 5, e1093277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Carambia, A.; Freund, B.; Schwinge, D.; Heine, M.; Laschtowitz, A.; Huber, S.; Wraith, D.C.; Korn, T.; Schramm, C.; Lohse, A.W.; et al. TGF-β-dependent induction of CD4+CD25+Foxp3+ Tregs by liver sinusoidal endothelial cells. J. Hepatol. 2014, 61, 594–599. [Google Scholar] [CrossRef] [PubMed]
  155. Serrels, A.; Lund, T.; Serrels, B.; Byron, A.; McPherson, R.C.; von Kriegsheim, A.; Gómez-Cuadrado, L.; Canel, M.; Muir, M.; Ring, J.E.; et al. Nuclear FAK Controls Chemokine Transcription, Tregs, and Evasion of Anti-tumor Immunity. Cell 2015, 163, 160–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Illustration summarizing the most relevant methodologies for circulating tumor cells (CTCs) enrichment and detection.
Figure 1. Illustration summarizing the most relevant methodologies for circulating tumor cells (CTCs) enrichment and detection.
Ijms 22 13073 g001
Figure 2. Illustration depicting the theoretical dynamics of CTCs after different successful therapeutic interventions.
Figure 2. Illustration depicting the theoretical dynamics of CTCs after different successful therapeutic interventions.
Ijms 22 13073 g002
Table 1. Potential strategies to target CTCs or tumor microenvironment to prevent HCC recurrence and metastases. The table includes systemic treatments, physical removal methods, and strategies based on gene therapy and immunotherapy.
Table 1. Potential strategies to target CTCs or tumor microenvironment to prevent HCC recurrence and metastases. The table includes systemic treatments, physical removal methods, and strategies based on gene therapy and immunotherapy.
Methodology /TherapyTargetStudy PopulationKey Findings
SorafenibpERK+/pAkt CTCsHCC patients pERK+/pAkt CTCs are sensible to sorafenib [23].
Viatar SystemEpCAM+ CTCs Physical removing of CTCs. It requires a dialysis system. Proof of concepts.
TIIASIn vitro,
canine model
Miniaturized Viatar System. Portable aphaeretic system. It analyzes 1–2% of whole blood in 2 hours [126].
FSMWCancer patientsPassive CTC-capturing device. Estimated analysis of 1.5–3 liters of blood in 30 min [127].
BPAFIC In vitro,
rabbit model
Passive CTC-capturing device. Captures 2.1% of CTCs in 5 min and kills them with 100% efficiency [128].
Anti-PD-1PD-L1+ CTCsHCC patientsFavorable response to anti-PD-1 therapy is associated with the presence of PD-L1+ CTCs [129].
Anti-EGFRCTCs (angiogenesis, cell migration)Mouse modelEGFR inhibition may reduce CTCs after transarterial chemoembolization [130].
Anti-MVPMVP+ CTCsIn vitroAnti-MVP therapy target MVP+EpCAM CTCs, which are related to metastases [19].
Anti-CD44CD90+CD44+ CSCsIn vitroCD44 blockade could induce the death of CD90+ cells [131].
AR overexpressionCD90 expressionMouse modelEnhancing AR expression in CTCs might reduce the risk of HCC recurrence [132].
Anti-USP1USP1+ CTCsMouse modelUSP1 upregulation in CTCs correlates with metastases and reduced survival. USP1 inhibition is a potential therapy for HCC [133].
Anti-ICAM-1ICAM-1+ CTCsMouse modelInhibition of ICAM-1 reduces tumor initiation and metastases [97].
Anti-TM4SF5TM4SF5+ CTCsMouse modelTargeting TM4SF5 or interaction between TM4SF5 and CD44 may lead to efficient inhibition of TM4SF5-mediated metastases [134].
Y-27632 Rho-associated kinaseRat model Y-27632 inhibits tacrolimus-enhanced invasiveness of cancer cells and could be used to prevent tumor recurrence after LT [135].
CXCR4/SDF-1 axis blockade CXCR4+ CTCsIn vitro, mouse modelSTAT3 inhibition and CXCR4 blockade have clinical therapeutic potential in HCC [136]; Hepatic stellate cells play an important role in liver metastases by releasing SDF-1 [137,138].
HepatocytesMouse modelNon-viral SDF-1 trap gene decreases liver metastases in models of colorectal and breast cancer [139].
miR-1247-3p/IL-6, IL-8 axis blockade miR-1247-3p/ IL-6/ IL-8In vitro, mouse modelTumor-derived exosomal miR-1247-3p converts fibroblasts to cancer-associated fibroblasts which promote tumor stemness, EMT, chemoresistance, and tumorigenicity [140].
miR-155 blockadeTumor-derived miR-155In vitro, mouse modelMiR-155 is highly elevated in EpCAM+HCC cells and could be an actionable target to remove the EpCAM+CSC population [141].
Anti-SMAD3CTCsIn vitro, mouse modelSMAD3-containing exosomes from primary tumors could favor the viability and adhesion of CTCs and the risk of metastases [142].
Function modulationLSECs In vitroCancer-activated LSECs can enhance the proliferation of Tregs and promote cancer cell liver colonization [143].
anti-CCL5/ CCR5,CCR4CCL5/ CCR5+, CCR4+ TregsIn vitro, mouse modelChemokine CCL5 recruits regulatory T cells to facilitate the immune escape of CTCs [21].
IFN-αMicroenvironmentMouse modelIFN-α does not decrease the number of CTCs but could modulate the tissue microenvironment to resist CTCs and metastases [144].
TIIAS, Temporary Indwelling Intravascular Aphaeretic System; FSMW, Structured and Functionali-zed Medical Wire; BPAFIC, Black Phosphorus and Antibody Functionalized Intravenous Catheter; AR, Androgen receptor; CCR, colorec-tal cancer; EMT, Epithelial-Mesenchymal Transition; LSECs, Liver sinusoidal endothelial cells; Tregs, regulatory T cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Espejo-Cruz, M.L.; González-Rubio, S.; Zamora-Olaya, J.; Amado-Torres, V.; Alejandre, R.; Sánchez-Frías, M.; Ciria, R.; De la Mata, M.; Rodríguez-Perálvarez, M.; Ferrín, G. Circulating Tumor Cells in Hepatocellular Carcinoma: A Comprehensive Review and Critical Appraisal. Int. J. Mol. Sci. 2021, 22, 13073. https://doi.org/10.3390/ijms222313073

AMA Style

Espejo-Cruz ML, González-Rubio S, Zamora-Olaya J, Amado-Torres V, Alejandre R, Sánchez-Frías M, Ciria R, De la Mata M, Rodríguez-Perálvarez M, Ferrín G. Circulating Tumor Cells in Hepatocellular Carcinoma: A Comprehensive Review and Critical Appraisal. International Journal of Molecular Sciences. 2021; 22(23):13073. https://doi.org/10.3390/ijms222313073

Chicago/Turabian Style

Espejo-Cruz, María Lola, Sandra González-Rubio, Javier Zamora-Olaya, Víctor Amado-Torres, Rafael Alejandre, Marina Sánchez-Frías, Rubén Ciria, Manuel De la Mata, Manuel Rodríguez-Perálvarez, and Gustavo Ferrín. 2021. "Circulating Tumor Cells in Hepatocellular Carcinoma: A Comprehensive Review and Critical Appraisal" International Journal of Molecular Sciences 22, no. 23: 13073. https://doi.org/10.3390/ijms222313073

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