Detection of Circulating Tumor Cells and Their Implications as a Biomarker for Diagnosis, Prognostication, and Therapeutic Monitoring in Hepatocellular Carcinoma
Abstract
Hepatocellular carcinoma (HCC) is among the leading causes of worldwide cancer-related morbidity and mortality. Poor prognosis of HCC is attributed primarily to tumor presentation at an advanced stage when there is no effective treatment to achieve the long term survival of patients. Currently available tests such as alpha-fetoprotein have limited accuracy as a diagnostic or prognostic biomarker for HCC. Liver biopsy provides tissue that can reveal tumor biology but it is not used routinely due to its invasiveness and risk of tumor seeding, especially in early-stage patients. Liver biopsy is also limited in revealing comprehensive tumor biology due to intratumoral heterogeneity. There is a clear need for new biomarkers to improve HCC detection, prognostication, prediction of treatment response, and disease monitoring with treatment. Liquid biopsy could be an effective method of early detection and management of HCC. Circulating tumor cells (CTCs) are cancer cells in circulation derived from the original tumor or metastatic foci, and their measurement by liquid biopsy represents a great potential in facilitating the implementation of precision medicine in patients with HCC. CTCs can be detected by a simple peripheral blood draw and potentially show global features of tumor characteristics. Various CTC detection platforms using immunoaffinity and biophysical properties have been developed to identify and capture CTCs with high efficiency. Quantitative abundance of CTCs, as well as biological characteristics and genomic heterogeneity among the CTCs, can predict disease prognosis and response to therapy in patients with HCC. This review article will discuss the currently available technologies for CTC detection and isolation, their utility in the clinical management of HCC patients, their limitations, and future directions of research.
Abbreviations
-
- AFP
-
- alpha-fetoprotein
-
- ASGPR
-
- asialoglycoprotein receptor
-
- BCLC
-
- Barcelona Clinic Liver Cancer
-
- CK
-
- cytokeratin
-
- CTC
-
- circulating tumor cell
-
- ctDNA
-
- circulating tumor DNA
-
- DEP
-
- dielectrophoresis
-
- EMT
-
- epithelial to mesenchymal transition
-
- EpCAM
-
- epithelial cell adhesion molecule
-
- FDA
-
- Food and Drug Administration
-
- GPC3
-
- glypican 3
-
- HCC
-
- hepatocellular carcinoma
-
- HKR
-
- higher karyoplasmic ratio
-
- IFC
-
- imaging flow cytometry
-
- ISET
-
- isolation by size of epithelial tumor cells
-
- MACS
-
- magnetic activated cell sorting
-
- PD-L1
-
- programmed death ligand 1
-
- RT-PCR
-
- reverse-transcription PCR
-
- SE-iFISH
-
- subtraction enrichment and immunostaining fluorescence in situ hybridization
-
- Treg
-
- regulatory T cells
Hepatocellular carcinoma (HCC) is an aggressive primary liver cancer that typically occurs in the setting of chronic liver disease and cirrhosis, and it is the sixth leading cause of cancer incidence and the fourth cause of cancer death globally.(1) Although a select group of patients with small, localized HCC may undergo curative therapies, those with large tumor burden, vascular invasion, or metastasis have a poor prognosis and are managed with systemic treatment and supportive care. There exists an unmet need for HCC biomarkers for early detection and prognostication, as well as prediction and monitoring for treatment response.
Currently, alpha-fetoprotein (AFP) is the most widely used biomarker for HCC. Biannual liver ultrasound with or without serum AFP is the main HCC screening strategy recommended by major societies.(2-4) AFP is used as a prognostic and predictive biomarker in patients with HCC. Elevated levels of AFP have been associated with increased tumor size and portal vein thrombosis, as well as increased risks of liver transplant wait-list dropout and posttransplant recurrence.(5, 6) Serum AFP is also a predictor of treatment response in patients with HCC after liver transplant and ramucirumab treatment.(7, 8) However, AFP’s role as a biomarker for early detection of HCC is limited by its poor sensitivity. Alternative protein-based serum tumor markers such as AFP lectin fraction (AFP-L3) and des-y-carboxy prothrombin (DCP) have been shown to improve the diagnostic performances when used in combination with AFP.(9) Glypican 3 (GPC3),(10) cytokeratin 19 (CK19),(11) golgi protein 73 (GP73),(12) midkine,(13) osteopontin,(14) squamous cell carcinoma antigen (SCCA),(15) and annexin A2(16) have all been shown to have diagnostic and prognostic roles in HCC as well, but they have not been adopted for widespread clinical practice.
Liver biopsy allows direct sampling of the tumor tissue and molecular characterization of the tumor. However, it is an invasive test with a risk of bleeding and concern for possible tumor seeding. Moreover, as HCCs exhibit significant intertumoral or intratumoral heterogeneity from genetic aberrations, transcriptional and epigenetic dysregulation, a single biopsy specimen containing a small amount of tumor tissue may not be representative of the whole HCC tumor.(17)
Over recent years, a variety of “liquid biopsy” techniques have shown significant promise as biomarkers for HCC. In liquid biopsy, samples of body fluids are collected to obtain vital pieces of phenotypic, genetic, and transcriptomic information about the primary tumor.(18) The primary forms of liquid biopsy include circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNA, and extracellular vesicles. First described in 1869, CTCs are malignant cells derived from either the primary tumor or metastases that migrate into the systemic circulation.(19) CTCs represent a unique biomarker different from any of the existing cancer biomarkers, as they represent a sampling of the patient’s live tumor cells.(20) Analysis of CTCs can help guide treatment plans by identifying specific mutations in target genes and predicting response or resistance to specific treatments. This review article will discuss the CTCs and provide an overview of their biology, current and emerging techniques, and various studies investigating their roles as potential diagnostic and prognostic biomarkers for HCC.
Overview of CTC Biology and Clinical Implications
As a malignant tumor proliferates and invades into the adjacent tissue, the tumor cells secrete matrix metalloproteinase, which breaks the basement membrane, enabling the tumor cells to gain direct access to the nearby blood and lymphatic vessels(21) (Fig. 1). Once in the bloodstream, the tumor cells become CTCs and can remain in circulation until they reach different tissues of the body and invade them. Most of the CTCs introduced into circulation get rapidly killed by processes such as anoikis, immune attacks, or shear stress.(22) Thus, the CTCs undergo a number of adaptations to survive in their hostile new environment. A key process is epithelial to mesenchymal transition (EMT), in which CTCs lose epithelial-type surface markers and gain mesenchymal markers, allowing them to behave like mesenchymal cells. CTCs that have undergone EMT can easily detach themselves from the primary tumor tissue and invade the capillaries and possess significantly improved ability to survive and metastasize. In addition, CTCs may form aggregates with fibroblasts, leukocytes, endothelial cells, or platelets to form CTC clusters, which possess a significantly higher metastatic potential and increased ability to survive, compared with individual CTCs.(23) It should be noted that CTCs are not identical clones of each other, but represent a heterogeneous population of cells from different tumor foci, with abilities to change their phenotypic and molecular characteristics under selective microenvironmental and therapeutic pressures.(24) Studies have shown that profound heterogeneities exist between tumor cells within the primary tumors and those within sites of metastasis. Therefore, liquid biopsy using CTCs should not be regarded merely as a less invasive alternative to an actual biopsy, but a way to obtain a comprehensive understanding of the heterogeneous tumor cells throughout the body. Detection and isolation of CTCs can enable the implementation of precision medicine by revealing the molecular characteristics of the tumor and identify markers for targeted therapy.

CTCs are being extensively studied in a variety of solid organ malignancies. In patients with metastatic prostate cancer, CTC enumeration has been shown to be a reliable predictor of prognosis and treatment response.(25) Detection of HER2+ CTCs in patients with breast cancer can identify candidates for targeted therapy.(26) Although CTCs are highly promising as a form of liquid biopsy, there are technical challenges to be overcome before there can be widespread use of CTCs. Most importantly, CTCs have an extremely rare frequency in the circulation, and the number of CTCs tends to be proportional to tumor volume, which makes their detection in early-stage disease challenging.(27) Therefore, the challenge of CTC research lies in improving the sensitivity and specificity of CTC detection to the levels necessary for accurate and comprehensive molecular characterization.
Another cornerstone of liquid biopsy, ctDNA is the fraction of cell-free DNA (cfDNA), which originates from dying tumor cells or macrophages that have phagocytized tumor cells.(28) As ctDNA contains genetic mutations identical to those of their originating tumor cells, ctDNA can be used to identify heterogeneous tumor-specific mutations and epigenetic changes, and to monitor tumor dynamics in patients who are undergoing therapy. ctDNA is already used for treatment-response monitoring or early detection of relapse, and to identify specific mutations to make therapy decisions.(29-31) Similar to CTCs, the clinical application of ctDNA is challenged by the technical difficulty of identifying ctDNA in the background of a significantly larger amount of cfDNA from other tissues. Moreover, it is unclear whether ctDNA accurately represents the genetic make-up of actively proliferating and metastasizing tumor cells, as ctDNA may disproportionately represent DNA from “weaker” tumor cells that are more prone to dying and releasing their contents into circulation.(32) Compared with ctDNA, the main advantage of CTCs is that intact, viable tumor cells are being captured. As long as their cellular integrity is preserved, the captured CTCs can be used for functional assays and be cultured to evaluate drug resistance.(33)
CTC Detection and Isolation
Several systems have been developed to improve the detection and isolation of CTCs, using their distinct physical and molecular characteristics. The platforms can largely be broken down into three categories of immunoaffinity-based enrichment, biophysical property-based enrichment, and enrichment-free methods (Table 1).
Subcategory | Key Features | Capture Yield* | Refs. | |
---|---|---|---|---|
Immunoaffinity | ||||
CellSearch | Immunomagnetic | Ferrofluid beads functionalized with anti-EpCAM. The only FDA-approved platform for CTC enumeration in metastatic prostate, breast, and colorectal cancer | ≥85% | 35 |
MACS | Immunomagnetic | MNPs conjugated to various antibodies. Large surface area to volume ratio. Can be used for both positive/negative enrichment | 40%-90% | 34 |
SERS | Immunomagnetic | MNPs used as the CTC enrichment platform and SERS signal amplification substrate. Dual-selectivity using anti-ASGPR nanoparticles and anti-GPC3 nanorods for HCC CTC isolation | >90% | 77 |
SE-iFISH | Immunomagnetic, in situ karyotyping | Combines subtraction enrichment followed by in situ phenotypic and karyotypic characterization; especially useful for identifying chromosome aneuploidy | 70%-87% | 94 |
CTC-chip | Microfluidic | Microposts (µpCTC-Chip) with geometric arrangement generate laminar flow, optimizing cell attachment. Herringbone-shaped grooves (HBCTC-chip) and microvortices increase cell contact toward antibody-coated surfaces. High-purity processing of whole blood | >60% | 36 |
CTC-iChip† | Microfluidic, immunomagnetic, inertial focusing | Sequential steps of micropillar array, inertial focusing, and magnetophoresis (isolation of nucleated cells including CTCs and WBCs using deterministic lateral displacement, alignment of nucleated cells in a microfluific channel, and collection of magnetically tagged cells). Combining strengths of microfluidics and magnetic-based cell sorting | 97% | 37 |
NanoVelcro† | Microfluidic | Anti-EpCAM/anti-ASGPR/anti-GPC-3 coated nanosubstrates, with integrated microfluidic chaotic mixers to facilitate CTC-substrate contact to achieve enhanced HCC-CTC capture. Vimentin(+) CTCs identified as a poor prognostic subset in HCC | 80%-94% | 38 |
Biophysical Property | ||||
ISET | Microfiltration | Size-based isolation of CTCs that are usually larger than hematologic cells. Track-etched membranes with nano-sized to micron-sized pores in thin polycarbonate films | N/A | 42 |
ScreenCell | Microfiltration | Size-based isolation of CTCs that are usually larger than hematologic cells. Track-etched membranes with nano-sized to micron-sized pores in thin polycarbonate films | 74%-91% | 43 |
CellSieve | Microfiltration | Precision pores arranged in arrayed patterns to enable CTC capture under low-pressure state and preserve intracellular architecture | 83%-91% | 44 |
FMSA | Microfiltration | Flexible polymer micro springs minimize cell damage. Allows rapid enrichment directly from whole blood | 90% | 45 |
CanPatrol | Microfiltration, immunomagnetic | Microfiltration followed by detection of EMT markers using RNA in situ hybridization | 80%-89% | 85 |
Ficoll-Paque | DGC | Inexpensive, easy-to-use in combination with other techniques. The ratio of CTCs to PBMCs remains unchanged | 84% | 48 |
OncoQuick | DGC | Porous membrane above separation media for additional separation by filtration. Superior CTC capture ratio compared with Ficoll-Paque | 87% | 49 |
Microfiltration | ||||
RosetteSep CTC Enrichment Cocktail | DGC immunomagnetic | Antibody complexes targeting an extensive mixture of CTC antigens. Can be used with centrifugation platforms such as Ficoll-Paque to enhance capture efficiency | 36%-60% | 50 |
DEPArray | DEP | Traps single cells in DEP cages generated through an array of electrodes. Can recover single CTCs | N/A | 55 |
Enrichment-Free | ||||
ImageStream | IFC | Uses size and karyoplasmic ratios to detect CTCs, without requiring antibodies or complex handling of blood samples | N/A | 57 |
PAFC | In vivo direct imaging | Absorption of laser by nanoparticles, which are tagged on targeted cells through antibodies. Enables real-time detection of CTCs in veins using a laser-based technology | N/A | 59 |
EPISPOT | Functional assay | Detects viable CTCs at the single-cell level by identifying proteins that are secreted/released/shed (antibody-based) from single epithelial cancer cells. Theoretically can be combined with any CTC enrichment step with live tumor cells | N/A | 60 |
- * From cell line spiking study (the capture yield may vary in different cancer types and different generations of the technology).
- † In each processed patient sample, background 500 and 200-1,000 white blood cells were nonspecifically captured in the CTC-iChip and NanoVelcro platform, respectively. Other platforms did not report a reliable purity.
- Abbreviations: DGC, density gradient centrifugation; FMSA, flexible micro spring array; MNP, magnetic nanoparticle; N/A, not available; PAFC, photoacoustic flow cytometry; PBMC, peripheral blood mononuclear cell; SERS, surface-enhanced Raman scattering; WBC, white blood cells.
Immunoaffinity
Immunoaffinity-based CTC enrichment techniques use antibodies against cell surface markers tethered to the device surface or a magnetic substance. Positive enrichment refers to capturing CTCs by using antibodies against specific tumor-associated antigens expressed on CTC surfaces, whereas negative enrichment refers to depleting the hematopoietic cells in the background by using antibodies against CD45.(34) Until recent years, CTCs were defined as nucleated epithelial cell adhesion molecule (EpCAM+)/CK+/CD45- cells. The only Food and Drug Administration (FDA)–approved CTC diagnostic technology, the CellSearch system (Menarini Silicon Biosystems Inc., Bologna, Italy) uses an immunomagnetic separation system of ferrofluid beads coated with antibody to EpCAM (anti-EpCAM).(35) Microfluidic devices such as the CTC-chip (developed at the Massachusetts General Hospital, Boston, MA) are composed of antibody-coated microposts with a geometric arrangement to optimize cell attachment.(36) The CTC-iChip combines microfluidic and immunomagnetic technology and has demonstrated a greater sensitivity of CTC detection compared with CellSearch.(37) In parallel, Wang et al. pioneered a unique concept of “NanoVelcro,” which uses antibody-coated nanostructured substrates with integrated microfluidic chaotic mixers to facilitate CTC-substrate contact to enhance CTC capture.(38)
Epithelial markers such as EpCAM are often down-regulated or lost during the EMT.(39) These CTCs with EMT phenotype have highly metastatic properties and can escape positive enrichment systems that target the epithelial markers such as EpCAM and CK. Given such limitations of relying on epithelial markers, positive enrichment strategies targeting stem cell markers (e.g., CD133), mesenchymal markers (e.g., vimentin), and cancer-specific antigens (e.g., HER2, PSMA) have also been developed.(40)
However, even antibody cocktails targeting a wide variety of antigens may not account for the heterogeneity of CTC antigens. Negative enrichment strategies address this by targeting and removing background hematopoietic cells. There are commercialized platforms dedicated to negative enrichment, and some of the positive-enrichment technologies such as magnetic activated cell sorting (MACS) and CTC-iChip can be used for negative enrichment by replacing anti-EpCAM with anti-CD45.(34) Although they allow for higher sensitivity compared with positive-enrichment technologies, negative-enrichment technologies alone typically have a much lower purity.(41)
Biophysical Properties
Compared with blood cells, CTCs are distinguished by their large size, mechanical plasticity, and dielectric mobility properties. This enables them to be isolated using techniques such as membrane filtration, density gradient stratification, inertial focusing, and dielectric mobility. These so-called “label-free” methods are increasingly popular, as they avoid the challenges of targeting numerous specific antigens. They also make downstream processing easier, as the isolated CTCs are not tagged with antibodies.(34)
Microfiltration uses the larger, more rigid phenotype observed by the CTCs. Isolation by ISET (size of epithelial tumor cells; Berlex Laboratories, Inc., Montville, NJ)(42) and ScreenCell (Sarcelles, France)(43) use track-etched membranes, whereas CellSieve (Creatv MicroTech, Inc., Rockville, MD)(44) and flexible micro spring array(45) use photolithography to construct membranes that minimize captured cell damage. The Cluster-Chip (Massachusetts General Hospital) is a unique 3D microfiltration system that is designed specifically to capture CTC clusters.(46) The main advantage of microfiltration is its ability to rapidly process blood for CTC enrichment. However, microfiltration systems are subject to clogging, and size overlap between leukocytes and CTCs makes it challenging to achieve high purity.(34)
Density-based gradient centrifugation uses the differences in specific gravities of leukocytes and CTCs.(47) Although not initially developed for CTC isolation, Ficoll-Paque (GE Healthcare Life Sciences) has been able to detect CTCs in patients with various cancers.(48) OncoQuick (Greiner Bio-One) combines centrifugation and filtration and has superior CTC capture ratio compared with Ficoll-Paque.(49) Finally, the RosetteSep CTC Enrichment Cocktail (STEMCELL Technologies, Inc.) integrates immunoaffinity with density centrifugation, using antibody complexes targeting an extensive mixture of antigens.(50) Centrifugation is used widely used for CTC isolation due to its reliability and inexpensiveness, but it is best used as an initial step before additional enrichment using other strategies.(34)
Inertial focusing uses the differential inertial effect on different sizes of cells to help with the isolation of CTCs. It is applicable to CTCs larger than background hematologic cells. This method has been combined into many microfluidic devices.(51) By establishing a model accounting for wall interaction force, shear gradient lift force, and secondary-flow drag force, as well as other effects including particle properties (i.e., cells in CTC isolation devices), rotation, interparticle spacing, and fluid properties, researchers have recently made significant improvements in predicting the motion of cells according to inertial focusing in microfluidic devices.(52) These devices have shown promising results with a high sensitivity of CTC detection and viability of retrieved CTCs.(53)
Another technology, dielectrophoresis (DEP), separates cells using distinct electrical fingerprints among different cell types.(54) The dielectric characteristics of cancer cells are significantly different from blood cells. By applying an alternating electrical field, cells are electrically polarized, and CTCs can be isolated through differential electric forces. Becker et al. demonstrated differential dielectric parameters in breast cancer cell lines, lymphocytes, and erythrocytes. The dielectric differences between cancer and blood cells may result from the different morphologic features of the cell membrane, including microvilli, membrane folds, and blebbing.(54) DEPArray (Menarini Silicon Biosystems, Inc.) can trap single cells in DEP cages and has been used to recover single CTCs for highly specific genetic analyses.(55)
Enrichment-Free Methods
All enrichment technologies require verification of the captured cells, which can be significantly time-consuming. Advancements in the field of high-speed, fluorescence imaging have led to the development of imaging platforms that enable a direct identification of CTCs from blood samples without the enrichment step.(56) Imaging flow cytometry distinguishes CTCs from leukocytes using parameters such as higher karyoplasmic ratio and size.(57, 58) Another unique, in vivo direct imaging method to detect CTCs is photoacoustic flow cytometry, which enables real-time detection of CTCs in veins using a laser-based technology.(59) Finally, there are functional assays to detect CTC, which exploit aspects of liver cellular activity such as secretion of tumor-associated proteins and preferential adhesion of CTCs to a specialized matrix.(60)
CTCs as a Biomarker in HCC
EpCAM-Based CTC Detection in HCC
The commonly used, immunoaffinity-based CTC enrichment techniques such as CellSearch have been used to detect and capture EpCAM+ CTCs in patients with HCC (Table 2). In 2013, Sun et al. used CellSearch to detect EpCAM+ CTCs in patients with HCC undergoing tumor resection. The authors preoperatively detected EpCAM+ CTCs in 67% of patients with HCC. A preoperative CTC count of two or greater was shown to be a predictor of tumor recurrence after surgery.(61) Additional studies using CellSearch demonstrated that the presence of EpCAM+ CTCs in patients with HCC was associated with vascular invasion,(62, 63) significantly elevated AFP,(63) more advanced Barcelona Clinic Liver Cancer (BCLC) stage,(62) disease progression,(64) higher recurrence rate,(65) and shorter overall survival.(62-66) In 2014, Guo et al. established an optimized platform based on negative enrichment and quantitative reverse-transcription PCR (RT-PCR) for the detection of EpCAM+ CTCs and exhibited 76.7% consistency with the CellSearch system.(67) In 2016, Wang et al. used the CTC-BioTChip to detect EpCAM+ CTCs in 60% of patients with HCC (n = 42) and found significant correlations between both the positive rate and number of CTCs with tumor, node, and metastases staging.(68) In 2016, Zhou et al. studied the prognostic value of EpCAM+ CTCs and regulatory T cells (Treg) in patients with HCC and found that elevated EpCAM+ CTC and Treg levels were associated with early recurrence and poor clinical outcome.(69) Unfortunately, EpCAM-based CTC enrichment platforms are limited by the loss of epithelial markers in CTCs undergoing EMT. Furthermore, a study showed that only a small proportion (30%-40%) of HCC cells express EpCAM.(70)
Study | Patients | Method | CTC Marker | Main Findings |
---|---|---|---|---|
EpCAM-Based CTC Detection in HCC | ||||
Sun et al.,(61) 2013 | 123 HCC; 5 BLD; 10 HV | CellSearch | EpCAM | CTCs identified in 67% of pre-op patients, and 28% 1 month after resection; ≥2 CTCs/7.5 mL predicted recurrence |
Schulze et al.,(62) 2013 | 59 HCC; 19 BLD | CellSearch | EpCAM | CTCs identified in 31% of patients with HCC. Associated with advanced stage, vascular invasion, and shorter OS |
Guo et al.,(67) 2014 | 299 HCC; 24 BHT; 25 CLD; 71 HV | RosetteSep, MACS, quantitative RT-PCR | EpCAM | Negative enrichment and quantitative RT-PCR-based platform had AUROC = 0.70, sensitivity = 42.6%, and specificity = 96.7%. Combined with AFP level, AUROC improved to 0.86 with sensitivity = 73.0% and specificity = 93.4% |
Kelley et al.,(63) 2015 | 20 HCC; 10 BLD | CellSearch | EpCAM | CTCs detected in 40% of patients with metastatic HCC; ≥1/7.5 mL associated with vascular invasion and decreased OS |
Wang et al.,(68) 2016 | 42 HCC | CTC-BioTChip | EpCAM | CTCs detected in 60% of patients with HCC. Positive rate and number of CTCs highly correlated with TNM staging |
Zhou et al.,(69) 2016 | 49 HCC undergoing curative resection; 50 HV | RosetteSep | EpCAM | Patients with high EpCAM mRNA+ CTCs and CD4+CD25+Foxp3+ Treg cells had higher risk of postoperative recurrence (67% vs. 10%) and 1-year recurrence (50% vs. 10%) |
Quantitative RT-PCR | CD4+CD25+Foxp3+ | |||
von Felden et al.,(65) 2017 | 57 HCC undergoing resection | CellSearch | EpCAM | CTCs detected in 15% of patients with HCC. CTC positivity associated with higher recurrence and shorter median RFS |
Shen et al.,(64) 2018 | 89 HCC undergoing TACE | CellSearch | EpCAM | CTCs detected in 56% of patients with HCC. Higher number of CTCs associated with mortality and progression |
Yu et al.,(66) 2018 | 139 HCC; 23 BHT | CellSearch | EpCAM | Patients with CTC ≥ 2 had shorter DFS and OS compared to patients with CTC < 2 |
Combined Markers-Based CTC Detection in HCC | ||||
Xu et al.,(71) 2011 | 85 HCC; 37 BLD; 14 OT; 20 HV | AutoMACS | ASGPR, HER2, TP53 | CTCs identified in 81% of patients with HCC. Positivity and number of CTCs correlated with tumor size, portal vein tumor thrombus, differentiation status, and disease extent |
Li et al.,(73) 2014 | 27 HCC; 12 OT; 13 LC; 11 BLT; 7 CH; 15 HV | AutoMACS | ASGPR; CPS1 | CTCs detected in 89% of patients with HCC. Anti-ASGPR-specific and efficient for HCC-CTC enrichment. Combining anti-P-CK and anti-CPS1 was superior (96.7%) to using one antibody alone (CPS1: 62.1%, P-CK: 85.2%) |
P-CK | ||||
Mu et al.,(74) 2014 | 62 HCC; 7 CH; 15 HV | MidiMACS | ASGPR, GPC3, CK | GPC3, ASGPR, and CK expression increased in patients with HCC. PPV and NPV: 90% and 71% for GPC3; 93% and 75% for ASGPR; 83% and 29% for CK. GPC3 and ASGPR expression associated with decreased OS |
Liu et al.,(75) 2015 | 32 HCC; 17 OT; 12 BLT; 15 LC; 10 CH; 3 AH; 20 HV | Ficoll-Paque, RosetteSep | ASGPR | CD45 depletion of leukocytes recovered more CTCs compared with ASGPR+ selection. Combining anti-ASGPR and anti-CPS1 improved CTC detection vs. either antibody alone, and detected CTCs in 91% of patients with HCC |
CPS1 | ||||
Zhang et al.,(76) 2016 | 36 HCC; 14 BLD | CTC-chip | ASGPR, P-CK, CPS1 | CTCs detected in 100% of patients with HCC. Captured CTCs readily released from CTC-chip and could subsequently be expanded to form a spheroid-like structure in a 3-dimensional cell culture assay |
Pang et al.,(77) 2018 | 8 HCC; 5 OT; 5 HV | SERS | ASGPR, GPC3 | SERS nanoplatform detected CTCs in 100% of patients with HCC, and enabled detection with small volume of blood |
Label-Free (Biophysical and Enrichment-Free) Methods to Detect CTCs in HCC | ||||
Vona et al.,(42) 2000 | 7 HCC undergoing hepatectomy; 8 CH; 8 HV | ISET | AFP | CTCs detected in 43% and 86% of patients with HCC before and after surgery. ISET allowed subsequent analysis of cell morphology, enumeration of CTCs, and demonstration of tumor microemboli during surgery |
RT-PCR | PSA | |||
Vona et al.,(79) 2004 | 44 HCC; 30 CH; 39 LC; 38 HV | ISET | N/A | CTCs found in 52% of patients with HCC. Associated with portal tumor thrombosis and shorter survival |
Morris et al.,(80) 2014 | 52 HCC | ISET | EpCAM | CellSearch detected CTCs in 28%, whereas ISET detected CTCs in 100% of patients with HCC. Presence of GPC3-positive CTCs by ISET was 100% concordant with GPC3-positive cells in the original tumor |
CellSearch | GPC3 | |||
Liu et al.,(57) 2016 | 52 HCC; 12 HV | IFC | HKR | Using HKR, IFC had an AUROC of 0.82. Patients with HCC had significantly higher presence of microvascular thrombosis, as well as higher risk of recurrence and mortality |
Ogle et al.,(58) 2016 | 69 HCC; 16 LC; 15 HV | IFC | Cell size | Size criteria + absence of CD45 led to capture of all positive biomarker CTCs, and 28% of biomarker negative CTCs. Increased CTCs associated with advanced tumor stage, portal vein thrombosis, and poorer survival |
CK, EpCAM, AFP, GPC-3, DNA-K | ||||
Biologic Characterization and Heterogeneity of CTCs in HCC | ||||
Li et al.,(72) 2013 | 60 HCC; 10 BLD; 10 OT; 10 HV | MiniMACS | ASGPR, vimentin, twist, ZEB1/2, snail, slug, cadherin | CTCs detected in 77% of patients with HCC. CTC positivity higher in patients with portal vein thrombus and advanced stages. EMT markers highly correlated with portal vein thrombus, TNM classification, and tumor size |
Flow cytometry | ||||
Li et al.,(82) 2016 | 109 HCC on sorafenib treatment | Ficoll-Paque | pERK | pERK/pAkt expression in CTC and tissue concordant in 90%. pERK+/pAkt- CTCs associated with PFS and predicted good prognosis. In vitro, pERK+/pAkt- HCC cells showed the greatest response to sorafenib |
RosetteSep | pAkt | |||
Shi et al(81) 2016 | 47 HCC undergoing cryoablation | MACS | MAGE-3, survivin | Average CTCs decreased significantly following cryosurgery. Gene expression for tumor markers MAGE-3, surviving, and CEA all significantly decreased following cryosurgery as well |
Quantitative RT-PCR | CEA | |||
Kalinich et al.,(84) 2017 | 63 HCC; 31 CLD; 6 LM; 38 OT; 26 HV | CTC-iChip | AFP, AHSG, ALB, APOH, FABP1, FGB, FGG, GPC3, RBP4, TF | 10 liver-specific transcripts identified CTCs in 56% of untreated patients with HCC vs. 3% of patients with nonmalignant liver disease at risk of developing HCC. CTC positivity declined in patients with HCC receiving therapy |
Digital PCR | ||||
Chen et al.,(86) 2017 | 195 HCC | CanPatrol | CK, EpCAM, twist, cadherin, snail, vimentin, AKT2 | CTCs detected in 95% of patients with HCC. Able to discriminate metastatic HCC with AUROC = 0.86, sensitivity = 86%, specificity = 81%. Mesenchymal CTCs associated with age, BCLC stages, metastasis, AFP levels, recurrence |
Court et al.,(78) 2018 | 61 HCC; 11 BLD; 8 HV | NanoVelcro CTC Assay | EpCAM, ASGPR | CTCs detected in 97% of patients with HCC. Panel identified HCC with sensitivity = 84.2%, specificity = 88.5%, PPV = 69.6%, NPV = 94.7%, and AUROC = 0.92. Vimentin-positive CTCs associated with aggressive disease and metastasis |
GPC3, vimentin | ||||
Qi et al.,(87) 2018 | 112 HCC treated with R0 resection; 12 HBV; 20 HV | CanPatrol | EpCAM, CK, vimentin, twist | CTCs detected in 90% of patients with HCC. CTC count ≥16 and mesenchymal-CTC percentage ≥2% associated with recurrence and metastasis. BCAT1 gene identified as a potential trigger of EMT |
Ou et al.,(88) 2018 | 165 HCC undergoing radical resection | CanPatrol | EpCAM, CK, vimentin, twist | CTCs detected in 71% of patients with HCC. Increased CTCs associated with higher AFP, multiple tumors, advanced staging, and tumor embolus. Mesenchymal CTCs predicted earlier recurrence and shortest RFS |
Yin et al.,(89) 2018 | 80 HCC; 10 HV | CanPatrol | Twist | Twist+ correlated with tumor burden/aggressiveness, staging, as well as post-op recurrence and mortality |
Ye et al.,(90) 2018 | 42 HCC | CanPatrol | TP53 | Post-op CTC counts and changes in CTC counts were independent prognostic indicators for PFS |
Cheng et al.,(91) 2018 | 113 HCC; 57 BLD; 6 LM | CanPatrol | CK, EpCAM | Total number of CTCs had AUROC = 0.774, which improved to 0.821 when combined with serum AFP. Mesenchymal CTCs were increased in late-stage patients with HCC |
Vimentin, twist | ||||
Wang et al.,(85) 2018 | 62 HCC undergoing radical resection | CanPatrol | CK, EpCAM, vimentin, twist | CTCs detected in 84% of patients with HCC. Patients with post-op recurrence had significantly higher number of CTCs, mesenchymal CTCs, and mixed CTCs. Mesenchymal CTCs associated with shortened postoperative DFS |
D’Avola et al.,(92) 2018 | 6 HCC; 1 HV | Single-cell RNA sequencing | CK, EpCAM | Single-cell RNA sequencing of CTCs showed significant transcriptome heterogeneity. Nonhepatic expression of ASGPR1 was detected in a significant proportion of non-CTCs, primarily monocytes |
GPC3, ASGPR1 | ||||
Sun et al.,(93) 2018 | 73 HCC undergoing curative resection | CellSearch | EpCAM, CK, cadherin, slug, vimentin, snail | EMT status of CTCs was heterogeneous across different vascular compartments (peripheral vein, peripheral artery, hepatic vein, portal vein, IVC) |
Quantitative RT-PCR | ||||
Wang et al.,(94) 2018 | 14 HCC; 16 CCA; 4 GBC undergoing resection | SE-iFISH | C8 aneuploidy | Among CTCs detected, 8% were EpCAM+, and 86% were EpCAM−. Small aneuploid HCC CTCs were discovered. Post-op quantity of triploid CTCs, multiploid CTCs, or CTMs were correlated with poor prognosis |
Winograd et al.,(83) 2018 | 73 HCC; 11 BLD; 8 HV | CTC-iChip | PD-L1 | Presence of PD-L1+ CTCs predicted response to anti-PD1 therapy |
- Abbreviations: AH, acute hepatitis; AUROC, area under the receiver operating characteristic curve; BHT, benign hepatic tumor; BLD, benign liver disease; CCA, cholangiocarcinoma; CH, chronic hepatitis; CLD, chronic liver disease; CTM, circulating tumor microemboli; DFS, disease-free survival; GBC, gallbladder cancer; HBV, hepatitis B virus; HD, hepatic disease without HCC; HV, healthy volunteers; IVC, inferior vena cava; LC, liver cirrhosis; LM, liver metastasis; NPV, negative predictive value; OS, overall survival; OT, other malignant tumors; PD1, programmed death receptor 1; PFS, progress-free survival; PPV, positive predictive value; RFS, recurrence-free survival; SERS, surface-enhanced Raman scattering; TNM, tumor, node, and metastases.
Combined Markers-Based CTC Detection in HCC
To overcome the low sensitivity of EpCAM-based CTC detection platforms in HCC, alternative markers have been investigated (Table 2). For example, asialoglycoprotein receptor (ASGPR), a transmembrane protein exclusively expressed on hepatocyte surfaces, has been used in a variety of enrichment methods for CTC detection in patients with HCC.(71-78) In 2011, Xu et al. performed immunomagnetic separation using HCC cells bound by biotinylated asialofetuin, a ligand of ASGPR, and was able to detect CTCs in 81% of patients with HCC.(71) Both the presence of CTCs and the quantity of CTCs significantly correlated with tumor extent, portal vein tumor thrombus, and differentiation status.(71) In 2014, the same group used a synthetic anti-ASGPR antibody instead of ASGPR ligand with successful CTC detection in 89% of patients with HCC.(73) Another study achieved an even higher CTC detection rate of 91% in patients with HCC by using a mixture of antibodies against ASGPR and CPS1, combined with negative enrichment.(75) In 2016, Zhang et al. used a CTC-chip with antibodies to ASGPR, P-CK, and CPS1 and isolated CTCs from 100% of patients with HCC (n = 36).(76) Recently, Court et al. used the NanoVelcro CTC Assay with a multimarker panel of EpCAM, ASGPR, and GPC3, and captured CTCs in 97% of patients with HCC.(78)
Label-Free (Biophysical and Enrichment-Free) Methods of CTC Detection in HCC
“Label-free” methods have also been investigated for CTC detection in HCC patients (Table 2). In 2000, Vona et al. used the ISET, a 2-dimensional microfiltration system, to detect CTCs in patients with HCC undergoing liver resection, and was able to capture tumor microemboli during surgery.(42) Vona et al. also found that the number of CTCs detected by ISET correlated with the presence of a diffuse tumor, portal tumor thrombosis, more advanced liver disease, as well as shorter survival.(79) A direct comparison of CellSearch and ISET in their ability to detect CTCs in patients with HCC revealed a significantly superior performance by ISET (ISET: 100%, CellSearch: 28%).(80) However, one disadvantage of the ISET is the difficulty of releasing CTCs for downstream genetic analysis.(27)
In 2016, Liu et al. used imaging flow cytometry (IFC) to detect CTCs from blood samples of patients with HCC, using a higher karyoplasmic ratio (HKR) seen with CTCs. When compared with the traditional CTC detection method based on CD45- EpCAM+ cells, IFC using HKR cells showed significantly greater area under the receiver operating characteristic curve of 0.82 versus 0.73.(57) Ogle et al. studied IFC focusing on cell size and compared its effectiveness to a multimarker panel consisting of CK, EpCAM, AFP, GPC-3, and DNA-K. IFC with the inclusion of size criteria, combined with the depletion of CD45 cells, led to the capture of all positive biomarker CTCs, as well as an additional 28% of CTCs that did not express any of the biomarkers tested.(58) The advantages of IFC are its ease and cost-effectiveness without requiring antibodies or complex handling of blood samples. However, its specificity could be limited by the presence of immune cells, which also have large size and HKRs.(57)
Biological Characterization and Heterogeneity of CTCs
Multimarker combination panels and downstream molecular analysis can reveal biological characteristics and heterogeneities of HCC CTCs (Table 2). Identification of CTCs expressing specific tumor markers and drug targets allows providers to predict response to therapy. Shi et al. used three CTC markers (MAGE3, survivin, and CEA) to evaluate the efficacy of cryosurgery on unresectable HCC, and found that they were reliable in predicting treatment response.(81) In 2016, Li et al. used the expression status of pERK and pAkt to detect CTCs in 93% of patients with advanced HCC undergoing sorafenib treatment.(82) Among different patterns of pERK/pAkt expression, the presence of pERK+/pAkt- CTCs was significantly associated with longer progression-free survival and higher rate of response to sorafenib.(82) Winograd et al. evaluated for CTCs expressing programmed death ligand 1 (PD-L1) in patients with HCC, and found that the presence of PD-L1+ CTCs discriminated patients with HCC with early-stage and advanced/metastatic disease. Of the 6 patients receiving anti–programmed death protein 1 therapy, all 3 patients demonstrating response had PD-L1+ CTCs, compared with only 1 of 3 nonresponders, suggesting the possibility that PD-L1+ CTCs might be predictive of treatment response to immunotherapy.(83) In 2017, Kalinich et al. also devised a unique strategy of using the CTC-iChip to isolate CTCs, then applying RNA-based digital PCR to detect a panel of liver-specific transcripts, which were combined into a single metric CTC score. The CTC score was highly correlated with BCLC staging and declined in patients receiving therapy.(84)
Expression of EMT markers such as vimentin and twist in CTCs of patients with HCC has been studied as markers for predicting recurrence and prognosis. Most of these studies used the CanPatrol CTC analysis platform (SurExam, Guangzhou, China), a two-step technique including microfiltration and subsequent characterization of CTCs using a variety of epithelial (EpCAM, CK8/9/19) and mesenchymal markers (vimentin and twist).(85) Using this method, Chen et al. detected CTCs in 95% of 195 patients with HCC.(86) The proportion of hybrid and mesenchymal CTCs was associated with increased age, BCLC stages, metastasis, and AFP levels.(86) Wang et al. studied 62 patients with HCC undergoing surgical resection and found that patients with a higher number of mesenchymal CTCs had increased risk of recurrence and shortened disease-free survival.(85) Other studies using CanPatrol showed a consistent association between mesenchymal CTCs and poor clinical outcomes.(87-91)
An emerging body of literature suggests profound heterogeneity among the HCC CTCs. D’Avola et al. developed an analytical technique that combines IFC and single-cell mRNA sequencing.(92) Genome-wide expression profiling of CTCs using this approach demonstrated significant transcriptome heterogeneity, even among CTCs from the same patient with HCC.(92) Such marked heterogeneity among CTCs was also reported by Sun et al., who found significant heterogeneity in the EMT status of CTCs across different vascular compartments, with predominantly epithelial phenotype at release but switching to EMT phenotype during transit.(93) A recent study demonstrated even more heterogeneity among CTCs, both in size and karyotype. Wang et al. used subtraction enrichment and immunostaining fluorescence in situ hybridization (SE-iFISH), which uses in situ characterization of phenotypes and karyotypes to examine both chromosomal aneuploidy and tumor marker expressions. Using this strategy, the authors discovered the existence of small HCC CTCs smaller than white blood cells, with a very high prevalence of chromosome 8 aneuploidy. The postsurgical quantity of triploid or multiploid CTCs significantly correlated with poor prognosis.(94)
Conclusion and Future Directions
Decades of research have led to significant advancements in the clinical utility of CTCs in patients with HCC (Fig. 1). As a form of liquid biopsy, CTCs hold great potential to facilitate the implementation of precision medicine in patients with HCC. CTCs are already FDA-approved for disease monitoring and prognostication in patients with metastatic breast and colorectal cancer, and there is an ongoing National Institutes of Health–sponsored clinical trial (NCT02973204) to investigate CTCs and ctDNA as clinical support tools in HCC. However, significant challenges remain before CTCs can be adopted for widespread clinical use in HCC. In addition to the technical challenges with CTC detection and isolation, there are numerous CTC detection methods, each with its own protocols for sample preparation, enrichment, and analysis. Most studies are small, single-center, case-control studies with widely varying patient demographics such as ethnicity, etiology of liver disease, and stage of HCC, which makes validation studies very difficult. Therefore, there needs to be a standardized assay protocol with high sensitivity and specificity that can capture the full spectrum of CTCs. This can potentially be achieved by multicenter, prospective studies with a larger sample size using a uniform CTC detection platform, which can provide effective validation of the findings.(95)
Author Contributions
J.C.A.: Review of the literature, drafting of the manuscript, and approval of the final version. P.-C.T., P.-J.C.: Review of the literature, critical revision of the manuscript, and approval of the final version. E.P., H.-R.T., S.C.L.: Conceptualization, critical revision of the manuscript, and approval of the final version. J.D.Y.: Conceptualization, supervision, critical revision of the manuscript, and approval of the final version.