Next Article in Journal
Time Does Matter: The Cellular Response to Resveratrol Varies Depending on the Exposure Duration
Previous Article in Journal
Transcriptome Analysis Reveals Mechanisms of Stripe Rust Response in Wheat Cultivar Anmai1350
Previous Article in Special Issue
Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Precision Oncology Framework Using Circulating Tumor Cells

1
Department of Personalized Cancer Medicine, Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto 606-8507, Japan
2
AFI Corporation, Medical Innovation Center, Building, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto 606-8507, Japan
3
Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto 606-8507, Japan
4
Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto 606-8507, Japan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(12), 5539; https://doi.org/10.3390/ijms26125539
Submission received: 19 April 2025 / Revised: 20 May 2025 / Accepted: 30 May 2025 / Published: 10 June 2025

Abstract

Circulating tumor cells (CTCs) are multifaceted biomarkers with significant potential for precision oncology, offering opportunities to refine diagnoses and personalize treatments across various cancer types, including colorectal and breast cancer. CTC assays serve as reliable prognostic indicators, even during chemotherapy and/or molecularly targeted therapies. Notably, CTCs exhibit heterogeneity that gradually develops during carcinogenesis and becomes more pronounced in advanced disease stages. These intra- and intertumoral heterogeneities pose challenges, particularly when drug-resistant clones emerge following therapy. The dynamic behavior of CTCs provides valuable insights into treatment response and prognosis. Extensive efforts have led to the development of technologies for effective CTC isolation, accelerating their clinical implementation. While both CTC and circulating tumor DNA (ctDNA) tests offer prognostic value, they reflect different aspects of tumor biology: CTC counts indicate tumor progression, while ctDNA levels correlate with tumor burden. The combined analysis is expected to yield complementary insights. CTC tests are feasible in general hospitals and may serve as tumor markers comparable to, or even superior to, conventional markers such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) for colorectal cancer, and CA15-3 for breast cancer. Early incorporation of CTC tests into routine blood panels appears to be a rational and promising approach.

1. Introduction

Precision oncology is an approach to cancer treatment that integrates the molecular and cellular features of tumors, the characteristics of the tumor microenvironment, and patient-specific factors to develop individualized therapeutic strategies [1,2,3]. Recent clinical trials of molecularly targeted therapies have demonstrated significant improvements in survival outcomes among patients with cancers, including colorectal and breast carcinomas, whose tumors harbor specific molecular alterations. Currently, such biomarker-driven therapies are available for an increasing number of cancer types [3].
Circulating tumor cells (CTCs) are multifaceted biomarkers that hold significant promise for the precision oncology, offering the potential to refine diagnoses and tailor treatments. One key advantage of CTCs is their minimally invasive nature, particularly compared to traditional biopsies, as they are obtained solely through blood collection. Once isolated, CTCs can undergo various types of genomic and molecular analyses and are often referred to as a real-time “liquid biopsy” for cancer patients [4,5,6].
CTC-derived data have applications in several clinical settings, including tumor screening and early diagnosis, detection of recurrence, and surveillance of disease progression [5,7]. The genomic profiling of individual CTCs is expected to be a critical component of precision oncology and is predicted to enhance the precision of cancer treatments for approximately 20% of molecularly matched patients [1,8]. Numerous basic and clinical studies have demonstrated that CTC enumeration serves as a predictive and prognostic biomarker [7].
In addition to CTCs, another blood-based biomarker is circulating tumor DNA (ctDNA). ctDNA, released from apoptotic or necrotic tumor cells, serves as a biomarker for detecting somatic variants present in tumor tissue [9]. Like CTCs, ctDNA can be obtained through a simple blood draw, offering a minimally invasive alternative to traditional biopsies. Although ctDNA cannot be used for transcriptional or pathological analyses, it has several clinical applications, including early cancer detection, detection of minimal/molecular residual disease (MRD), and monitoring of treatment response [9].
In this review, we outline the clinical applications of CTCs in relation to metastasis, molecular pathology, prognostic biomarkers, comparison with ctDNA, and detection methods. We place particular emphasis on colorectal cancer, which typically exhibits lower CTC levels, and breast cancer, which tends to show higher CTC levels. Globally, colorectal and breast cancers are the second and fourth leading causes of cancer-related mortality, and the third and second most commonly diagnosed malignancies worldwide, respectively [10]. These cancers have been extensively studied in the context of CTC analysis due to their clinical relevance, distinct patterns of metastasis, and the availability of comprehensive data on CTC isolation. Including both tumor types enables comparative evaluation of CTC characteristics across malignancies with significant clinical impact.

2. CTCs in the Metastatic Process

Carcinomas invade surrounding tissues and enter the bloodstream via nearby veins. Portions of the primary tumor dissociates, circulating as single cells or clusters within blood vessels before being transported to distant organs. These CTCs can adhere to the vessel walls, extravasate, and, albeit at a low frequency, establish metastatic colonies (Figure 1) [11].
In the bloodstream, colon cancer cells are frequently transported to the liver via the mesenteric and portal veins and subsequently reach the lungs (Figure 1a) [12,13]. In contrast, lower rectal cancer cells have a greater tendency to metastasize to the lungs directly via the inferior rectal vein, inferior vena cava, and heart (Figure 1a) [12,13]. Similarly, breast cancer cells are thought to spread hematogenously to the bones, lungs, and liver via the venous circulation and heart (Figure 1b), and colonize distant organs with molecular signature-driven organotropism [14].
Although cancer researchers have studied the metastatic process for over 120 years, it is only recently that molecular insights have emerged clarifying the organotropism of certain tumor types, including colorectal and breast cancer (Figure 1) [15,16]. Namely, cancer cell-secreted factors, including tumor-derived extracellular vesicles and particles (EVPs), circulate and act on distant organs to establish a tumor microenvironment (TME) that is favorable for the initiation of metastasis. These changes lead to the formation of “pre-metastatic niches” (PMNs), which are receptive to the colonization of CTCs [17]. In addition, tumor-associated macrophages (TAMs) are the particularly abundant stromal cells in TME of most solid tumors, and a high TAM density is associated with poor prognosis in cancer patients [16,18].
Nevertheless, cancer metastasis is considered an inefficient process [19]. CTCs are subjected to various forms of cell death due to factors such as detachment-induced apoptosis (anoikis), immune attacks by CD8⁺ T cells and natural killer (NK) cells [16], and fluid shear stress as they traverse capillaries [20]. CD8⁺ T cells and NK cells typically induce apoptosis in CTCs through the secretion of perforin and granzymes, as well as through the expression of death ligands such as Fas ligand (FASL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [21]. On the other hand, neutrophils contribute to both tumoricidal and tumor-promoting phenotypes [16,22,23].
It is well established that CTCs undergoing epithelial–mesenchymal transition (EMT) express mesenchymal markers. Moreover, some CTCs have been reported to maintain a quiescent or dormant state [24]. These observations suggest that the behavior of CTCs reflects not only their ability to adapt to the circulatory environment but also their inherent plasticity.
Several studies suggest that CTC clusters exhibit higher metastatic potential than single CTCs [25,26,27], but due to their rarity, reports on their clinical significance remain limited [7]. CTCs are also believed to reflect the biology of metastasis itself [28]. Therefore, methods that enable early detection of colorectal and breast cancer and real-time monitoring of CTCs may hold great promise for improving clinical outcomes and patient survival [29].

3. Heterogeneity and Molecular Pathology of CTCs

Tumor heterogeneity, a key factor in the emergence of drug-resistant cells following chemotherapy and/or endocrine therapy, can arise during the development and progression of primary tumors [30], or be induced by various treatments within these primary tumors [31,32]. Consequently, multiple tumor cell subtypes appear as CTC subpopulations in the later stages of the disease [33,34].
In primary colorectal tumors, driver mutations in genes such as adenomatous polyposis coli (APC), tumor protein p53 (TP53), Kirsten rat sarcoma viral oncogene homolog (KRAS), and SMAD family member 4 (SMAD4) are associated with multistep tumorigenesis through the adenoma–carcinoma sequence [30,35,36,37]. Mutations in KRAS at codon 12 or 13 (observed in 35% to 45% of cases) or the v-raf murine sarcoma viral oncogene homolog B (BRAF) V600E mutation (approximately 10%) confer resistance to cetuximab-based therapies in metastatic colorectal cancer (mCRC) [38]. Notably, KRAS and BRAF mutations are mutually exclusive in colorectal cancers [39].
Interestingly, several studies have highlighted inconsistencies between the molecular profiles of CTCs and their corresponding primary tumors. Mutation analyses of driver genes in colorectal cancer CTCs have revealed discordance between CTCs and primary tumors [40,41]. Specifically, mutations in KRAS, BRAF, or phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) were detected in CTCs but were absent in the primary tumors [40,41], suggesting a potential role in resistance to molecularly targeted therapies [42,43].Similarly, in luminal-type breast cancer, certain activating mutations are detected specifically in CTCs but are absent in the primary tumor. For instance, mutations in estrogen receptor 1 (ESR1) and phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2 gamma (PIK3C2G) have been identified in metastatic lesions but occur at low frequencies in primary tumors [44,45]. Such discrepancies may reflect tumor evolution under therapeutic pressure and the emergence of resistant clones [46,47]. On the other hand, other reports have shown little or no difference in driver gene mutations between the primary tumors and metastatic lesions [30,48].
The heterogeneity of CTCs can be demonstrated by their expression of key molecular markers. In metastatic colorectal cancer, this heterogeneity can be detected through the distinct expression patterns of caudal type homeobox 1 (CDX1), CDX2, and/or catenin beta 1 (CTNNB1) (Figure 2a). In metastatic breast cancer, it is reflected in the varied expression patterns of ESR1, ESR2, progesterone receptor (PGR), and/or erb-b2 receptor tyrosine kinase 2 (ERBB2), consistent with the heterogeneity observed in the primary tumors and/or metastatic lesions (Figure 2b) [25,49,50,51,52,53,54].
One of the major contributors to gene expression heterogeneity is EMT, a process that facilitates tumor progression and metastasis [31,55]. Since vimentin, a stromal intermediate filament protein is highly expressed during EMT, antibodies against cell-surface vimentin (CSV), a cancer-specific antigen, have been used to detect live CTCs in patients with EMT-associated carcinoma or malignant sarcomas [56,57,58,59]. Notably, approximately 70% of CTCs in both metastatic colorectal and breast cancers express the epithelial cell adhesion molecule (EPCAM), members of the Keratin (KRT) family, and/or vimentin (VIM) (Figure 2a,b).
A subset of CTCs in recurrent or metastatic breast cancer exhibits a KI67-negative state, indicative of quiescence or dormancy, during which the cells undergo cell cycle arrest [60,61] (Figure 2b). The expression levels of proliferation markers such as marker of proliferation Ki-67 (MKI67), telomerase reverse transcriptase (TERT), myelocytomatosis oncogene (MYC), and cyclin B1 (CCNB1) are significantly downregulated in CTCs compared to those in their parental cell lines or in primary tumors (Figure 2b) [62]. Importantly, this quiescent phase contributes to resistance to anticancer drugs and facilitates tumor recurrence.
These genomic, transcriptomic, and phenotypic studies demonstrate that CTCs can exhibit significant heterogeneity, even within the same patient. As a result, identifying reliable biomarkers for CTC detection remains challenging [63]. Consequently, researchers have explored alternative approaches for CTC detection, which are discussed in Section 6.
Figure 2. Heterogeneity in CTC transcriptomes of metastatic colorectal and breast cancers. (a,b) Heatmaps showing CTC transcriptomic profiles in metastatic colorectal (n = 59; panel (a)) and breast cancers (n = 339; panel (b)) [25,49,50,51,52,53,54,64]. In panel (b), ctcRbase integrates seven datasets (GSE111065, GSE51827, GSE55807, GSE67939, GSE75367, GSE86978, and GSE41245) as previously described [64]. Panels (a) and (b) present expression values in transcripts per million (TPM) and fragments per kilobase of transcript per million mapped reads (FPKM), respectively. These heatmaps were reconstructed by the authors based on data reported in previous studies [25,49,50,51,52,53,54,64]. As only limited genomic data on CTCs was available in these datasets, gene mutation data is not presented.
Figure 2. Heterogeneity in CTC transcriptomes of metastatic colorectal and breast cancers. (a,b) Heatmaps showing CTC transcriptomic profiles in metastatic colorectal (n = 59; panel (a)) and breast cancers (n = 339; panel (b)) [25,49,50,51,52,53,54,64]. In panel (b), ctcRbase integrates seven datasets (GSE111065, GSE51827, GSE55807, GSE67939, GSE75367, GSE86978, and GSE41245) as previously described [64]. Panels (a) and (b) present expression values in transcripts per million (TPM) and fragments per kilobase of transcript per million mapped reads (FPKM), respectively. These heatmaps were reconstructed by the authors based on data reported in previous studies [25,49,50,51,52,53,54,64]. As only limited genomic data on CTCs was available in these datasets, gene mutation data is not presented.
Ijms 26 05539 g002

4. CTCs as Prognostic and Predictive Biomarkers in Cancer Therapy

Based on accumulated evidence highlighting the prognostic significance of CTCs in various cancers, researchers have investigated the clinical utility of CTC enumeration. The dynamics of CTCs provide insights into therapeutic response levels. In colorectal cancer patients, the presence of CTCs has been significantly correlated with poor overall survival (OS), poor disease-free survival (DFS), poor progression-free survival (PFS), and advanced tumor, node, and metastasis (TNM) stages [65,66,67,68].
In a meta-analysis of colorectal cancer patients with mixed stages, CTC counts of ≥1, ≥2, and ≥3 were associated with hazard ratios (HRs) for overall mortality (95% CI) of 2.33 (0.44–12.18), 2.12 (1.13–3.98), and 1.72 (1.28–2.32), respectively [29]. A threshold of ≥3 CTCs per 7.5 mL of blood has frequently been used as a marker to define “high CTC” status in metastatic colorectal cancer [4,29,69,70,71,72]. Additionally, CTC enumeration demonstrated the added benefits of bevacizumab plus FOLFOXIRI (folinic acid, fluorouracil, oxaliplatin, and irinotecan) compared to bevacizumab plus FOLFOX (folinic acid, fluorouracil, and oxaliplatin) in colorectal cancer patients [4,72,73].
Interestingly, CTC-based stratification identified two distinct response groups among metastatic colorectal cancer patients treated with bevacizumab and FOLFOX: a favorable response group (complete response [CR], partial response [PR], or stable disease [SD]) and an unfavorable response group (progressive disease [PD]) [74]. Separately, in locally advanced rectal cancer, CTCs expressing thymidylate synthase (TYMS) and RAD23 homolog B (RAD23B) were identified as markers of resistance to chemotherapy and radiotherapy (Figure 2) [75].
In clinical trials involving early- and late-stage breast cancer patients undergoing chemotherapy, CTC positivity was associated with unfavorable OS and DFS [76,77,78,79]. A meta-analysis pooling data from 21 studies, which included 2,239 non-metastatic breast cancer patients who received neoadjuvant chemotherapy, demonstrated that higher CTC counts correlated with poor survival outcomes [80]. Specifically, the hazard ratios (HRs) for death (95% CI) associated with CTC counts of 1, 2, 3–4, and ≥5 were 1.09 (0.65–1.69), 2.63 (1.42–4.54), 3.83 (2.08–6.66), and 6.25 (4.34–9.09), respectively.
Furthermore, CTC counts serve as prognostic markers in metastatic breast cancer, not only in patients receiving chemotherapy but also in those undergoing molecular-targeted therapies, such as letrozole plus bevacizumab and anti-HER2 (human epidermal growth factor receptor 2, also known as ERBB2) monoclonal antibodies [81,82,83,84]. Changes in CTC counts during treatment are significantly associated with PFS and OS, underscoring the value of CTC tracking in improving prognostic accuracy for metastatic breast cancer [7,79,85,86].
Therefore, CTC monitoring can serve as a complementary tool for therapeutic decision-making, clinical evaluation, and treatment efficacy assessment. Enumeration of CTCs provides real-time indicators of tumor response and has been recognized as an effective prognostic biomarker. In patients with early-stage, operable, estrogen receptor-positive (ER+) breast cancer, CTC counts have also been used as indicators for detecting minimal/molecular residual disease (MRD) up to four years earlier [87]. However, combining CTCs with ctDNA may improve the sensitivity of MRD detection [88], as discussed below.

5. Feasibility of the CTC Test: A Comparison with the ctDNA Test

CTCs exhibit a variety of features over time in individual patients, with their numbers typically increasing prior to recurrence [87,89]. While both CTCs and ctDNA offer prognostic information, CTC count reflects cancer progression, whereas ctDNA quantity provides an indication of tumor burden [7,89]. Although few colorectal cancer studies have reported a direct comparison between CTC and ctDNA detection [90], the breast cancer study shows concordance between them was approximately 48% in early-stage breast cancer and 67% in the metastatic setting, with the latter showing a higher level of concordance [91]. These findings suggest that CTC and ctDNA detection provide complementary insight. The CTC test is superior to the ctDNA test for downstream analyses such as cytological tests, chemosensitivity tests, and gene expression profiling (Figure 2) (Table 1). However, the ctDNA shows strengths in sequential quantification of tumor burden [92,93]. Regarding the ctDNA test, it is important to consider that a laboratory testing consortium has highlighted the following limitations: “ctDNA exists in extremely low quantities and is highly fragmented, requiring deep sequencing of a large number of DNA fragments. Additionally, a bioinformatics pipeline capable of detecting variants with low allele frequencies is necessary... [94]” This suggests that the ctDNA test may not be feasible in general hospitals (Table 1). In contrast, CTCs can be analyzed if the appropriate equipment is available, and many detection systems are compatible with pathology laboratories to a certain degree [95,96,97]. Once CTCs are isolated using minimally damaging separation systems such as ClearCell FX1, CROSSORTER, or Parsortix (see Section 7) [98,99,100], they can be processed using standard cytological procedures. Specifically, the cells are attached to glass slides via routine cytocentrifugation, stained using conventional immunocytochemistry, and examined under a standard light microscope. For enhanced sensitivity, quantitative polymerase chain reaction (qPCR) can also be performed using Real-Time PCR instruments that are widely available in clinical laboratories (Table 1) [101].

6. CTC Separation Techniques

CellSearch is a CTC detection system that was approved by The United States Food and Drug Administration (FDA) in 2004 for use in the diagnosis and prognosis prediction of metastatic colorectal, breast, and prostate cancer [97]. In addition, Parsortix was also granted de novo classification by the FDA in 2022 for isolation of CTC in metastatic breast cancer [98]. Furthermore, various researchers and developers have meticulously designed devices for CTC isolation. The following sections and Table 2 describe these techniques in more detail.

6.1. Size-Based Separation, Including Hydrodynamic Methods

Size-based separation utilizes the fact that CTCs are generally larger than normal blood cells. By employing fine filters of specific sizes, CTCs can be selectively separated. For instance, white blood cells and red blood cells that are smaller than 8 µm can pass through the filter, while CTCs, which typically range from 10 to 20 µm, are retained and collected. A representative system using this principle is Isolation by Size of Epithelial Tumor Cells (ISET) [106].
Another approach in this category involves microfluidic constrictions, where microfluidic chips are designed with narrow constriction regions. Owing to their larger size, CTCs are physically unable to pass through these narrow constrictions, enabling their separation and collection. This method has the added advantage of reducing the risk of filter clogging, a common issue in membrane filtration systems.
Hydrodynamic separation techniques address similar challenges by exploiting fluid dynamics for CTC isolation [107]. One such method is inertial microfluidics, which utilizes the inertial forces and dean flow characteristics generated as the fluid passes through microchannels. Larger CTCs are directed toward the center or specific regions of the flow path, allowing for their separation from smaller blood cells. Spiral microfluidics and Dean Flow Fractionation are well-known systems utilizing this approach [108,109].
Another hydrodynamic separation method is deterministic lateral displacement (DLD) [109], which employs microfluidic channels with systematically arranged obstacles. As cells pass through these obstacles, they follow different trajectories based on their sizes, enabling effective separation. This technique is advantageous for high-throughput separation processes.

6.2. Density-Based Separation

Density-based separation relies on the differences in density between CTCs and other blood cells, such as red and white blood cells. Centrifugal force is applied to separate cells based on their density differences. Common techniques include density gradient centrifugation using Ficoll or OncoQuick to concentrate CTCs [110,111]. Additionally, some microfluidic chips integrate centrifugal separation functions to enhance this approach [112].

6.3. Immunoseparation

Immunoseparation methods leverage the expression of specific surface markers on CTCs for selective isolation [106]. Positive selection involves capturing CTCs using antibodies targeting specific markers such as epithelial cell adhesion molecule (EpCAM). Systems such as CTC-iChip [107], which combines immunomagnetic separation with fluidic separation, and CTC-Chip [113], which captures CTCs using antibody-coated micropillars, are based on this principle. Negative selection, on the other hand, removes normal blood cells using antibodies targeting markers like CD45 and CD34. By eliminating these cells, only CTCs remain, including those that may be EpCAM-negative. This method enables the isolation of a broader spectrum of CTC subpopulations.
Immunomagnetic beads, such as those coated with EpCAM antibodies, allow for CTCs to be separated under a magnetic field. Representative systems include CellSearch [97], an FDA-approved CTC isolation platform, and MagSweeper [62], which integrates microfluidic chips with magnetic separation capabilities.

6.4. Acoustic Separation

Acoustic separation employs standing ultrasonic waves to manipulate cells based on their size, density, and compressibility [114]. Since CTCs and normal blood cells exhibit different physical properties, ultrasound waves can be used to selectively direct CTCs into specific regions for collection. Acoustophoresis, also known as acoustic microfluidics, is a well-established system based on this principle.

6.5. Dielectrophoresis (DEP)

DEP is a separation technique that utilizes non-uniform electric fields to induce motion in cells based on their dielectric properties [99,115,116]. Cells respond differently to the electric field, either being attracted to regions of high field intensity (positive DEP) or repelled toward low-intensity regions (negative DEP). This unique behavior allows label-free separation of CTCs without the need for antibodies or chemical markers.
DEP is particularly effective in isolating CTCs, as their dielectric properties differ from those of normal blood cells. By incorporating microelectrodes into microfluidic chips, real-time manipulation and separation of cells can be achieved. A notable DEP-based system is DEPArray, which enables single-cell-level CTC isolation using microelectrode technology [115]. Moreover, the CROSSORTER system combines DEP with size-based separation to achieve low-damage CTC isolation [99].
Table 2. Overview of CTC separation techniques and their characteristics.
Table 2. Overview of CTC separation techniques and their characteristics.
Separation TechniqueSeparation PrincipleAdvantagesRepresentative SystemsReferences
Size-based
(including hydrodynamic)
Filtration,
microchannel constrictions,
inertial microfluidics
Simple,
high-throughput,
label-free
ClearCell FX1, CROSSORTER,
CTC-iChip 1, ISET, Parsortix, VTX-1
[98,99,100,
106,107,109,
117,118]
Density-basedCentrifugation,
gradient
Simple,
scalable
Ficoll, OncoQuick, RosetteSep[110,111,119]
ImmunoseparationPositive/negative selectionHigh selectivity, specificityAdnaTest 2, CellSearch 2, CTC-iChip, GEDI Chip, HB-chip, MagSweeper 2[62,106,107,
113,120,121,122]
Acoustic-basedUltrasonic fieldNon-invasive,
label-free
AcouTrap 3[114]
DEP-basedDielectrophoresisLabel-free,
precise control
Apostream, CROSSORTER 4,
DEPArray, 3DEP
[99,115,116,
122]
1 Uses immunolabeling; 2 Uses magnetic labeling; 3 Separation relies on size, density, and compressibility; 4 Also incorporates size-based separation.

7. Low-Damage CTC Isolation Systems

Systems that are considered to cause low damage should meet the following conditions: (1) minimal physical stress, avoiding strong centrifugal forces, shear forces, or pressure; (2) label-free separation, meaning no need for antibodies or magnetic beads, thereby preserving the integrity of the cell surface; and (3) the ability to maintain cells in a viable state. Several systems fulfill these criteria. The Parsortix PC1 System employs microfluidic technology to capture cells based on size and deformability, without requiring chemical treatments such as antibody labeling, allowing for the captured cells to be recovered alive (Table 2) [98]. It has been FDA-cleared and is well-suited for downstream analysis, including gene expression studies and cell culture. The ClearCell FX1 System utilizes inertial focusing technology to separate CTCs solely based on fluidic properties, minimizing shear stress and ensuring a high cell survival rate [100]. Since it does not require centrifugation, it enables label-free CTC isolation. The CROSSORTER integrates size-based and DEP-based technologies for label-free CTC separation (Table 2) [99]. Although it involves exposure to an electric field, it minimizes the need for physical and biological damage, thereby reducing cellular stress. This system has a high live-cell recovery rate, making it suitable for culture and genetic analysis. The VTX-1 Liquid Biopsy System is another label-free microfluidic platform that isolates CTCs based on size and morphology, without the use of antibodies, thereby reducing the risk of cell membrane damage [118]. These are particularly promising choices for researchers aiming to recover viable CTCs for subsequent culture or genetic analysis.
Further information on CTC isolation devices can be found in recent comprehensive reviews [7,104,123].

8. Current Challenges

While CTC-based technologies hold great promise for cancer diagnosis and treatment, their integration into routine clinical practice poses several clinical and technical challenges. The key points are as follows:
(1)
Definition of CTCs using both gene expression and mutation profiles.
(2)
Standardization of CTC isolation devices and protocols.
(3)
Formulation of guidelines by an international consortium.
To define CTCs, robust evidence is required to confirm that they are indeed cancer cells. Although this can be achieved by identifying driver-gene mutations, a number of studies fail to provide this essential evidence. Furthermore, the genetic heterogeneity of CTCs, which can vary across patients and even within the same patient over time, adds complexity to their definition and characterization.
Once such evidence is obtained, we will be able to establish benchmarks for CTC isolation devices and approve these devices for clinical use. These benchmarks will need to consider not only sensitivity and specificity but also the reproducibility of results across different laboratory settings and patient populations.
To accomplish this, it is crucial to collaborate with international CTC consortia. This collaboration will allow for the development of standardized guidelines, which will provide a unified approach to CTC isolation and clinical application. These guidelines will play a key role in ensuring that CTC-based technologies are implemented safely, effectively, and consistently across healthcare systems worldwide.

9. Precision Oncology Framework Using CTCs

After blood collection, CTCs are isolated from blood using low-damage separation systems such as ClearCell FX1, CROSSORTER, or Parsortix (Section 6 and Section 7). The isolated CTCs can then be processed using standard cytological techniques, including routine cytocentrifugation and immunocytochemistry. These cells can be examined and enumerated microscopically or alternatively detected via signal amplification by qPCR (Section 5). CTC profiling provides valuable insights into the biology of metastasis and contributes to clinical decision-making, particularly in the context of molecularly targeted therapies (Section 3 and Section 4).
CTCs may reflect metastatic potential, risk of recurrence, or prognostic relevance based on parameters such as cell count, the presence of gene mutations, or EMT characteristics (Section 2 and Section 3). Even if CTCs are not initially detectable at the start of treatment, they may emerge during long-term monitoring, sometimes years later (Section 4). Furthermore, various immune cell populations can influence the survival and proliferation of CTCs or disseminated tumor cells (DTCs), which may enter reversible states of dormancy or quiescence (Section 3).
Thus, CTCs represent multifaceted biomarkers with significant promise in precision oncology, offering the potential to enhance diagnostic accuracy and enable individualized treatment strategies. A key advantage of CTC analysis lies in its minimally invasive nature, requiring only blood collection compared to conventional tissue biopsies. CTC-based assays are expected to be integrated into clinical blood panels as tumor markers with greater clinical utility than traditional markers such as CEA, CA19-9, or CA15-3, thereby supporting oncologists in therapeutic decision-making.
Currently, over 3,000 clinical trials are investigating CTC-based diagnostics—approximately twice the number of trials focusing on ctDNA [124]. These ongoing studies have demonstrated the feasibility and clinical relevance of CTC testing in prospective trials involving patients with colorectal and breast cancers [125,126,127,128].

10. Limitations

The role of CTCs as a liquid biopsy has been recognized in many solid tumors; however, the prognostic implications of CTC counts and molecular characteristics are strongly influenced by both disease stage (early vs. metastatic) and tumor type. Therefore, combining distinct tumor types should be approached with caution, as it may lead to misinterpretation of the biological significance and clinical relevance of CTCs.

Author Contributions

Conceptualization, F.K., K.O. (Kyoichi Oshiro), Y.E., K.K., M.K., H.M. (Hiroyuki Miyoshi), and Y.T.; writing—original draft preparation, F.K.; writing—review and editing, F.K., K.O. (Kyoichi Oshiro), Y.E., K.K., N.M., H.M. (Hisatsugu Maekawa), J.M., M.K., H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama); supervision, K.O. (Kyoichi Oshiro), H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama); project administration, F.K., K.O. (Kyoichi Oshiro), Y.E., K.K., N.M., H.M. (Hisatsugu Maekawa), J.M., M.K., H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama); funding acquisition, H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the development of innovative personalized medicine based on patient-derived cancer stem cell culture in Alliance program for innovative Medical/healthcare research by Government–Academia–Industry Collaboration (AIMGAIN) from the Japan Agency for Medical Research and Development (K.Ob.), grant number 23zf0227002h0001, and also by AFI Corporation, Kyo Diagnostics K.K.; and SCREEN Holdings Co., Ltd.

Acknowledgments

We acknowledge the members of Department of Personalized Cancer Medicine and Gastrointestinal Surgery at KUHP for help in manuscript preparation.

Conflicts of Interest

The authors declare the following conflicts of interest: F.K., H.M. (Hisatsugu Maekawa), H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama) are affiliated with an endowed chair established through funding received from AFI Corporation, Kyo Diagnostics K.K., and SCREEN Holdings Co., Ltd. F.K., H.M. (Hisatsugu Maekawa), H.M. (Hiroyuki Miyoshi), Y.T., and K.O. (Kazutaka Obama) received research funds from AFI Corporation, Kyo Diagnostics K.K., and SCREEN Holdings Co., Ltd. K.O. (Kyoichi Oshiro) is an employee of AFI Corporation.

Abbreviations

The following abbreviations are used in this manuscript:
APCAdenomatous polyposis coli
BRAFV-raf murine sarcoma viral oncogene homolog B
CA15-3Carbohydrate antigen 15-3
CA19-9Carbohydrate antigen 19-9
CCNB1Cyclin B1
CD8Cluster of differentiation 8
CEACarcinoembryonic Antigen
CIConfidence interval
CRComplete response
CSVCell-surface vimentin
CTCCirculating tumor cell
CTC-ChipCirculating tumor cell chip
CTC-iChipCirculating tumor cell inertial chip
ctDNACirculating tumor DNA
DEPDielectrophoresis
DFSDisease-free survival
DTCDisseminated tumor cell
EMTEpithelial–mesenchymal transition
EPCAMEpithelial cell adhesion molecule
EREstrogen receptor
ESR1Estrogen receptor 1
FASLFas ligand
FDAThe United States Food and Drug Administration
FOLFOXFolinic acid, fluorouracil, and oxaliplatin
FOLFOXIRIFolinic acid, fluorouracil, oxaliplatin, and irinotecan
HER2human epidermal growth factor receptor 2
HRHazard ratio
ISETIsolation by Size of Epithelial Tumor Cells
MKI67Marker of proliferation Ki-67
MRDMinimal/molecular residual disease
MYCMyelocytomatosis oncogene
NKNatural killer
KRASKirsten rat sarcoma viral oncogene homolog
KRTKeratin
OSOverall survival
PFSProgression-free survival
PIK3CAPhosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
PIK3C2GPhosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2 gamma
PMEPre-metastatic niche
PRPartial response
qPCRquantitative polymerase chain reaction
RAD23BRAD23 homolog B
SDStable disease
SMAD4SMAD family member 4
TAMTumor-associated macrophage
TERTTelomerase reverse transcriptase
TMETumor microenvironment
TNMTumor, node, and metastasis
TP53Tumor protein 53
TYMSThymidylate synthase
VIMVimentin

References

  1. Le Tourneau, C.; Borcoman, E.; Kamal, M. Molecular profiling in precision medicine oncology. Nat. Med. 2019, 25, 711–712. [Google Scholar] [CrossRef]
  2. Yates, L.R.; Seoane, J.; Le Tourneau, C.; Siu, L.L.; Marais, R.; Michiels, S.; Soria, J.C.; Campbell, P.; Normanno, N.; Scarpa, A.; et al. The European Society for Medical Oncology (ESMO) Precision Medicine Glossary. Ann. Oncol. 2018, 29, 30–35. [Google Scholar] [CrossRef]
  3. Mateo, J.; Steuten, L.; Aftimos, P.; André, F.; Davies, M.; Garralda, E.; Geissler, J.; Husereau, D.; Martinez-Lopez, I.; Normanno, N.; et al. Delivering precision oncology to patients with cancer. Nat. Med. 2022, 28, 658–665. [Google Scholar] [CrossRef] [PubMed]
  4. Veyrune, L.; Naumann, D.N.; Christou, N. Circulating Tumour Cells as Prognostic Biomarkers in Colorectal Cancer: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 3437. [Google Scholar] [CrossRef]
  5. Hu, M.; Wang, Z.; Wu, Z.; Ding, P.; Pei, R.; Wang, Q.; Xing, C. Circulating tumor cells in colorectal cancer in the era of precision medicine. J. Mol. Med. 2022, 100, 197–213. [Google Scholar] [CrossRef] [PubMed]
  6. Alix-Panabières, C. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–491. [Google Scholar] [CrossRef]
  7. Fabisiewicz, A.; Szostakowska-Rodzos, M.; Grzybowska, E.A. Improving the Prognostic and Predictive Value of Circulating Tumor Cell Enumeration: Is Longitudinal Monitoring the Answer? Int. J. Mol. Sci. 2024, 25, 10612. [Google Scholar] [CrossRef] [PubMed]
  8. Rodon, J.; Soria, J.-C.; Berger, R.; Miller, W.H.; Rubin, E.; Kugel, A.; Tsimberidou, A.; Saintigny, P.; Ackerstein, A.; Braña, I.; et al. Genomic and transcriptomic profiling expands precision cancer medicine: The WINTHER trial. Nat. Med. 2019, 25, 751–758. [Google Scholar] [CrossRef]
  9. Cohen, S.A.; Liu, M.C.; Aleshin, A. Practical recommendations for using ctDNA in clinical decision making. Nature 2023, 619, 259–268. [Google Scholar] [CrossRef]
  10. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  11. Fidler, I.J. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 2003, 3, 453–458. [Google Scholar] [CrossRef] [PubMed]
  12. Qiu, M.; Hu, J.; Yang, D.; Cosgrove, D.P.; Xu, R. Pattern of distant metastases in colorectal cancer: A SEER based study. Oncotarget 2015, 6, 38658–38666. [Google Scholar] [CrossRef]
  13. Riihimäki, M.; Hemminki, A.; Sundquist, J.; Hemminki, K. Patterns of metastasis in colon and rectal cancer. Sci. Rep. 2016, 6, 29765. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, W.; Hoffmann, A.D.; Liu, H.; Liu, X. Organotropism: New insights into molecular mechanisms of breast cancer metastasis. NPJ Precis. Oncol. 2018, 2, 4. [Google Scholar] [CrossRef]
  15. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
  16. Kitamura, T.; Qian, B.-Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef]
  17. Patras, L.; Shaashua, L.; Matei, I.; Lyden, D. Immune determinants of the pre-metastatic niche. Cancer Cell 2023, 41, 546–572. [Google Scholar] [CrossRef] [PubMed]
  18. Kitamura, T. Tumour-associated macrophages as a potential target to improve natural killer cell-based immunotherapies. Essays Biochem. 2023, 67, 1003–1014. [Google Scholar]
  19. Fidler, I.J. Metastasis: Quantitative analysis of distribution and fate of tumor embolilabeled with 125 I-5-iodo-2’-deoxyuridine. J. Natl. Cancer Inst. 1970, 45, 773–782. [Google Scholar]
  20. Follain, G.; Herrmann, D.; Harlepp, S.; Hyenne, V.; Osmani, N.; Warren, S.C.; Timpson, P.; Goetz, J.G. Fluids and their mechanics in tumour transit: Shaping metastasis. Nat. Rev. Cancer 2020, 20, 107–124. [Google Scholar] [CrossRef]
  21. Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef] [PubMed]
  22. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of Tumor-Associated Neutrophil Phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef]
  23. Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef]
  24. Nasr, M.M.; Lynch, C.C. How circulating tumor cluster biology contributes to the metastatic cascade: From invasion to dissemination and dormancy. Cancer Metastasis Rev. 2023, 42, 1133–1146. [Google Scholar] [CrossRef]
  25. Aceto, N.; Bardia, A.; Miyamoto, T.D.; Donaldson, C.M.; Wittner, S.B.; Spencer, A.J.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [PubMed]
  26. Larsson, A.-M.; Jansson, S.; Bendahl, P.-O.; Levin Tykjaer Jörgensen, C.; Loman, N.; Graffman, C.; Lundgren, L.; Aaltonen, K.; Rydén, L. Longitudinal enumeration and cluster evaluation of circulating tumor cells improve prognostication for patients with newly diagnosed metastatic breast cancer in a prospective observational trial. Breast Cancer Res. 2018, 20, 48. [Google Scholar] [CrossRef] [PubMed]
  27. Kok, S.Y.; Nakayama, M.; Morita, A.; Oshima, H.; Oshima, M. Genetic and nongenetic mechanisms for colorectal cancer evolution. Cancer Sci. 2023, 114, 3478–3486. [Google Scholar] [CrossRef]
  28. Pantel, K.; Alix-panabières, C.; Riethdorf, S. Cancer micrometastases. Nat. Rev. Clin. Oncol. 2009, 6, 339–351. [Google Scholar] [CrossRef]
  29. Tan, Y.; Wu, H. The significant prognostic value of circulating tumor cells in colorectal cancer: A systematic review and meta-analysis. Curr. Probl. Cancer 2018, 42, 95–106. [Google Scholar] [CrossRef]
  30. Reiter, J.G.; Baretti, M.; Gerold, J.M.; Makohon-Moore, A.P.; Daud, A.; Iacobuzio-Donahue, C.A.; Azad, N.S.; Kinzler, K.W.; Nowak, M.A.; Vogelstein, B. An analysis of genetic heterogeneity in untreated cancers. Nat. Rev. Cancer 2019, 19, 639–650. [Google Scholar] [CrossRef]
  31. Menyailo, M.E.; Tretyakova, M.S.; Denisov, E.V. Heterogeneity of Circulating Tumor Cells in Breast Cancer: Identifying Metastatic Seeds. Int. J. Mol. Sci. 2020, 21, 1696. [Google Scholar] [CrossRef] [PubMed]
  32. Crucitta, S.; Crucitta, S.; Cucchiara, F.; Mathijssen, R.; Mateo, J.; Jager, A.; Joosse, A.; Passaro, A.; Attili, I.; Petrini, I.; et al. Treatment-driven tumour heterogeneity and drug resistance: Lessons from solid tumours. Cancer Treat. Rev. 2022, 104, 102340. [Google Scholar] [CrossRef] [PubMed]
  33. Asleh, K.; Riaz, N.; Nielsen, T.O. Heterogeneity of triple negative breast cancer: Current advances in subtyping and treatment implications. J. Exp. Clin. Cancer Res. 2022, 41, 265. [Google Scholar] [CrossRef]
  34. Saoudi González, N.; Salvà, F.; Ros, J.; Baraibar, I.; Rodríguez-Castells, M.; García, A.; Alcaráz, A.; Vega, S.; Bueno, S.; Tabernero, J.; et al. Unravelling the Complexity of Colorectal Cancer: Heterogeneity, Clonal Evolution, and Clinical Implications. Cancers 2023, 15, 4020. [Google Scholar] [CrossRef] [PubMed]
  35. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef]
  36. Fearon, E.R.; Bert, V. A genetic model for colorectal tumorigenesis. Cell 1991, 61, 759–767. [Google Scholar] [CrossRef]
  37. Kitamura, T.; Kometani, K.; Hashida, H.; Matsunaga, A.; Miyoshi, H.; Hosogi, H.; Aoki, M.; Oshima, M.; Hattori, M.; Takabayashi, A.; et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat. Genet. 2007, 39, 467–475. [Google Scholar] [CrossRef]
  38. Alberto, B.; Siena, S. Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J. Clin. Oncol. 2010, 28, 1254–1261. [Google Scholar]
  39. Rajagopalan, H.; Bardelli, A.; Lengauer, C.; Kinzler, K.W.; Vogelstein, B.; Velculescu, V.E. RAF/RAS oncogenes and mismatch-repair status. Nature 2002, 418, 934. [Google Scholar] [CrossRef]
  40. 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 in ex vivo culture. Gut 2017, 66, 1802–1810. [Google Scholar] [CrossRef]
  41. Nomura, M.; Miyake, Y.; Inoue, A.; Yokoyama, Y.; Noda, N.; Kouda, S.; Hata, T.; Ogino, T.; Miyoshi, N.; Takahashi, H.; et al. Single-Cell Analysis of Circulating Tumor Cells from Patients with Colorectal Cancer Captured with a Dielectrophoresis-Based Micropore System. Biomedicines 2023, 11, 203. [Google Scholar] [CrossRef] [PubMed]
  42. Misale, S.; Yaeger, R.; Hobor, S.; Scala, E.; Janakiraman, M.; Liska, D.; Valtorta, E.; Schiavo, R.; Buscarino, M.; Siravegna, G.; et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012, 486, 532–536. [Google Scholar] [CrossRef]
  43. Diaz Jr, L.A.; Williams, R.T.; Wu, J.; Kinde, I.; Hecht, J.R.; Berlin, J.; Allen, B.; Bozic, I.; Reiter, J.G.; Nowak, M.A.; et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012, 486, 537–540. [Google Scholar] [CrossRef] [PubMed]
  44. Jeselsohn, R.; Buchwalter, G.; De Angelis, C.; Brown, M.; Schiff, R. ESR1 mutations--a mechanism for acquired endocrine resistance in breast cancer. Nat. Rev. Clin. Oncol. 2015, 12, 573–583. [Google Scholar] [CrossRef]
  45. De Mattos-Arruda, L.; Weigelt, B.; Cortes, J.; Won, H.H.; Ng, C.K.Y.; Nuciforo, P.; Bidard, F.-C.; Aura, C.; Saura, C.; Peg, V.; et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: A proof-of-principle. Ann. Oncol. 2014, 25, 1729–1735. [Google Scholar] [CrossRef]
  46. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor–Positive Advanced Breast Cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef]
  47. Robinson, D.R.; Wu, Y.-M.; Vats, P.; Su, F.; Lonigro, R.J.; Cao, X.; Kalyana-Sundaram, S.; Wang, R.; Ning, Y.; Hodges, L.; et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat. Genet. 2013, 45, 1446–1451. [Google Scholar] [CrossRef] [PubMed]
  48. Jones, S.; Chen, W.-D.; Parmigiani, G.; Diehl, F.; Beerenwinkel, N.; Antal, T.; Traulsen, A.; Nowak, M.A.; Siegel, C.; Velculescu, V.E.; et al. Comparative lesion sequencing provides insights into tumor evolution. Proc. Natl. Acad. Sci. USA 2008, 105, 4283–4288. [Google Scholar] [CrossRef]
  49. Kozuka, M.; Battaglin, F.; Jayachandran, P.; Wang, J.; Arai, H.; Soni, S.; Zhang, W.; Hirai, M.; Matsusaka, S.; Lenz, H.-J. Clinical Significance of Circulating Tumor Cell Induced Epithelial-Mesenchymal Transition in Patients with Metastatic Colorectal Cancer by Single-Cell RNA-Sequencing. Cancers 2021, 13, 4862. [Google Scholar] [CrossRef]
  50. Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112.e114. [Google Scholar] [CrossRef]
  51. 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] [PubMed]
  52. Sarioglu, A.F.; Aceto, N.; Kojic, N.; Donaldson, M.C.; Zeinali, M.; Hamza, B.; Engstrom, A.; Zhu, H.; Sundaresan, T.K.; Miyamoto, D.T.; et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat. Methods 2015, 12, 685–691. [Google Scholar] [CrossRef]
  53. Jordan, N.V.; Bardia, A.; Wittner, B.S.; Benes, C.; Ligorio, M.; Zheng, Y.; Yu, M.; Sundaresan, T.K.; Licausi, J.A.; Desai, R.; et al. HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature 2016, 537, 102–106. [Google Scholar] [CrossRef]
  54. Aceto, N.; Bardia, A.; Wittner, B.S.; Donaldson, M.C.; O’Keefe, R.; Engstrom, A.; Bersani, F.; Zheng, Y.; Comaills, V.; Niederhoffer, K.; et al. AR Expression in Breast Cancer CTCs Associates with Bone Metastases. Mol. Cancer Res. 2018, 16, 720–727. [Google Scholar] [CrossRef]
  55. Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [PubMed]
  56. Satelli, A.; Mitra, A.; Cutrera, J.J.; Devarie, M.; Xia, X.; Ingram, D.R.; Dibra, D.; Somaiah, N.; Torres, K.E.; Ravi, V.; et al. Universal Marker and Detection Tool for Human Sarcoma Circulating Tumor Cells. Cancer Res. 2014, 74, 1645–1650. [Google Scholar] [CrossRef]
  57. Satelli, A.; Mitra, A.; Brownlee, Z.; Xia, X.; Bellister, S.; Overman, M.J.; Kopetz, S.; Ellis, L.M.; Meng, Q.H.; Li, S. Epithelial–Mesenchymal Transitioned Circulating Tumor Cells Capture for Detecting Tumor Progression. Clin. Cancer Res. 2015, 21, 899–906. [Google Scholar] [CrossRef]
  58. Satelli, A.; Batth, I.S.; Brownlee, Z.; Rojas, C.; Meng, Q.H.; Kopetz, S.; Li, S. Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci. Rep. 2016, 6, 28910. [Google Scholar] [CrossRef] [PubMed]
  59. Yu, J.; Yang, M.; Peng, T.; Liu, Y.; Cao, Y. Evaluation of cell surface vimentin positive circulating tumor cells as a prognostic biomarker for stage III/IV colorectal cancer. Sci. Rep. 2023, 13, 18791. [Google Scholar] [CrossRef]
  60. Muller, V.; Stahmann, N.; Riethdorf, S.; Rau, T.; Zabel, T.; Goetz, A.; Jänicke, F.; Pantel, K. Circulating tumor cells in breast cancer: Correlation to bone marrow micrometastases, heterogeneous response to systemic therapy and low proliferative activity. Clin. Cancer Res. 2005, 11, 3678–3685. [Google Scholar] [CrossRef]
  61. Spiliotaki, M.; Mavroudis, D.; Kapranou, K.; Markomanolaki, H.; Kallergi, G.; Koinis, F.; Kalbakis, K.; Georgoulias, V.; Agelaki, S. Evaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancy. Breast Cancer Res. 2014, 16, 485. [Google Scholar] [CrossRef] [PubMed]
  62. Powell, A.A.; Talasaz, A.H.; Zhang, H.; Coram, M.A.; Reddy, A.; Deng, G.; Telli, M.L.; Advani, R.H.; Carlson, R.W.; Mollick, J.A.; et al. Single Cell Profiling of Circulating Tumor Cells: Transcriptional Heterogeneity and Diversity from Breast Cancer Cell Lines. PLoS ONE 2012, 7, e33788. [Google Scholar] [CrossRef]
  63. Van Der Toom, E.E.; Verdone, J.E.; Gorin, M.A.; Pienta, K.J. Technical challenges in the isolation and analysis of circulating tumor cells. Oncotarget 2016, 7, 62754–62766. [Google Scholar] [CrossRef] [PubMed]
  64. Zhao, L.; Wu, X.; Li, T.; Luo, J.; Dong, D. ctcRbase: The gene expression database of circulating tumor cells and microemboli. Database 2020, 2020, baaa020. [Google Scholar] [CrossRef]
  65. Wang, L.; Zhou, S.; Zhang, W.; Wang, J.; Wang, M.; Hu, X.; Liu, F.; Zhang, Y.; Jiang, B.; Yuan, H. Circulating tumor cells as an independent prognostic factor in advanced colorectal cancer: A retrospective study in 121 patients. Int. J. Color. Dis. 2019, 34, 589–597. [Google Scholar] [CrossRef]
  66. Bidard, F.-C.; Kiavue, N.; Ychou, M.; Cabel, L.; Stern, M.-H.; Madic, J.; Saliou, A.; Rampanou, A.; Decraene, C.; Bouché, O.; et al. Circulating Tumor Cells and Circulating Tumor DNA Detection in Potentially Resectable Metastatic Colorectal Cancer: A Prospective Ancillary Study to the Unicancer Prodige-14 Trial. Cells 2019, 8, 516. [Google Scholar] [CrossRef]
  67. Wang, D.; Yang, Y.; Jin, L.; Wang, J.; Zhao, X.; Wu, G.; Zhang, J.; Kou, T.; Yao, H.; Zhang, Z. Prognostic models based on postoperative circulating tumor cells can predict poor tumor recurrence-free survival in patients with stage II-III colorectal cancer. J. Cancer 2019, 10, 4552–4563. [Google Scholar] [CrossRef]
  68. Su, P.; Lai, W.; Liu, L.; Zeng, Y.; Xu, H.; Lan, Q.; Chu, Z.; Chu, Z. Mesenchymal and Phosphatase of Regenerating Liver-3 Status in Circulating Tumor Cells May Serve as a Crucial Prognostic Marker for Assessing Relapse or Metastasis in Postoperative Patients With Colorectal Cancer. Clin. Transl. Gastroenterol. 2020, 11, e00265. [Google Scholar] [CrossRef] [PubMed]
  69. Matsusaka, S.; Suenaga, M.; Mishima, Y.; Kuniyoshi, R.; Takagi, K.; Terui, Y.; Mizunuma, N.; Hatake, K. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in Japanese patients with metastatic colorectal cancer. Cancer Sci. 2011, 102, 1188–1192. [Google Scholar] [CrossRef]
  70. Aggarwal, C.; Meropol, N.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.A.; Mitchell, E.; et al. Relationship among circulating tumor cells, CEA and overall survival in patients with metastatic colorectal cancer. Ann. Oncol. 2013, 24, 420–428. [Google Scholar] [CrossRef]
  71. Sastre, J.; Vidaurreta, M.; Gómez, A.; Rivera, F.; Massutí, B.; López, M.R.; Abad, A.; Gallen, M.; Benavides, M.; Aranda, E.; et al. Prognostic Value of the Combination of Circulating Tumor Cells Plus KRAS in Patients With Metastatic Colorectal Cancer Treated With Chemotherapy Plus Bevacizumab. Clin. Color. Cancer 2013, 12, 280–286. [Google Scholar] [CrossRef]
  72. Aranda, E.; Viéitez, J.M.; Gómez-España, A.; Gil Calle, S.; Salud-Salvia, A.; Graña, B.; Garcia-Alfonso, P.; Rivera, F.; Quintero-Aldana, G.A.; Reina-Zoilo, J.J.; et al. FOLFOXIRI plus bevacizumab versus FOLFOX plus bevacizumab for patients with metastatic colorectal cancer and ≥3 circulating tumour cells: The randomised phase III VISNÚ-1 trial. ESMO Open 2020, 5, e000944. [Google Scholar] [CrossRef]
  73. Krebs, M.G.; Renehan, A.G.; Backen, A.; Gollins, S.; Chau, I.; Hasan, J.; Valle, J.W.; Morris, K.; Beech, J.; Ashcroft, L.; et al. Circulating Tumor Cell Enumeration in a Phase II Trial of a Four-Drug Regimen in Advanced Colorectal Cancer. Clin. Color. Cancer 2015, 14, 115–122.e2. [Google Scholar] [CrossRef] [PubMed]
  74. Delgado-Ureña, M.; Ortega, F.G.; De Miguel-Pérez, D.; Rodriguez-Martínez, A.; García-Puche, J.L.; Ilyine, H.; Lorente, J.A.; Exposito-Hernandez, J.; Garrido-Navas, M.C.; Delgado-Ramirez, M.; et al. Circulating tumor cells criteria (CyCAR) versus standard RECIST criteria for treatment response assessment in metastatic colorectal cancer patients. J. Transl. Med. 2018, 16, 251. [Google Scholar] [CrossRef]
  75. Troncarelli Flores, B.C.; Souza E Silva, V.; Ali Abdallah, E.; Mello, C.A.L.; Gobo Silva, M.L.; Gomes Mendes, G.; Camila Braun, A.; Aguiar Junior, S.; Thomé Domingos Chinen, L. Molecular and Kinetic Analyses of Circulating Tumor Cells as Predictive Markers of Treatment Response in Locally Advanced Rectal Cancer Patients. Cells 2019, 8, 641. [Google Scholar] [CrossRef] [PubMed]
  76. Mego, M.; Karaba, M.; Minarik, G.; Benca, J.; Silvia, J.; Sedlackova, T.; Manasova, D.; Kalavska, K.; Pindak, D.; Cristofanilli, M.; et al. Circulating Tumor Cells With Epithelial–to–mesenchymal Transition Phenotypes Associated with Inferior Outcomes in Primary Breast Cancer. Anticancer Res. 2019, 39, 1829–1837. [Google Scholar] [CrossRef] [PubMed]
  77. Magbanua, M.J.M.; Yau, C.; Wolf, D.M.; Lee, J.S.; Chattopadhyay, A.; Scott, J.H.; Bowlby-Yoder, E.; Hwang, E.S.; Alvarado, M.; Ewing, C.A.; et al. Synchronous Detection of Circulating Tumor Cells in Blood and Disseminated Tumor Cells in Bone Marrow Predicts Adverse Outcome in Early Breast Cancer. Clin. Cancer Res. 2019, 25, 5388–5397. [Google Scholar] [CrossRef]
  78. Bortolini Silveira, A.; Bidard, F.-C.; Tanguy, M.-L.; Girard, E.; Trédan, O.; Dubot, C.; Jacot, W.; Goncalves, A.; Debled, M.; Levy, C.; et al. Multimodal liquid biopsy for early monitoring and outcome prediction of chemotherapy in metastatic breast cancer. NPJ Breast Cancer 2021, 7, 115. [Google Scholar] [CrossRef]
  79. Magbanua, M.J.M.; Hendrix, L.H.; Hyslop, T.; Barry, W.T.; Winer, E.P.; Hudis, C.; Toppmeyer, D.; Carey, L.A.; Partridge, A.H.; Pierga, J.-Y.; et al. Serial Analysis of Circulating Tumor Cells in Metastatic Breast Cancer Receiving First-Line Chemotherapy. JNCI J. Natl. Cancer Inst. 2021, 113, 443–452. [Google Scholar] [CrossRef]
  80. Bidard, F.-C.; Michiels, S.; Riethdorf, S.; Mueller, V.; Esserman, L.J.; Lucci, A.; Naume, B.; Horiguchi, J.; Gisbert-Criado, R.; Sleijfer, S.; et al. Circulating Tumor Cells in Breast Cancer Patients Treated by Neoadjuvant Chemotherapy: A Meta-analysis. J. Natl. Cancer Inst. 2018, 110, 560–567. [Google Scholar] [CrossRef]
  81. Paoletti, C.; Miao, J.; Dolce, E.M.; Darga, E.P.; Repollet, M.I.; Doyle, G.V.; Gralow, J.R.; Hortobagyi, G.N.; Smerage, J.B.; Barlow, W.E.; et al. Circulating Tumor Cell Clusters in Patients with Metastatic Breast Cancer: A SWOG S0500 Translational Medicine Study. Clin. Cancer Res. 2019, 25, 6089–6097. [Google Scholar] [CrossRef] [PubMed]
  82. Magbanua, M.J.M.; Savenkov, O.; Asmus, E.J.; Ballman, K.V.; Scott, J.H.; Park, J.W.; Dickler, M.; Partridge, A.; Carey, L.A.; Winer, E.P.; et al. Clinical Significance of Circulating Tumor Cells in Hormone Receptor-positive Metastatic Breast Cancer Patients who Received Letrozole with or Without Bevacizumab. Clin. Cancer Res. 2020, 26, 4911–4920. [Google Scholar] [CrossRef] [PubMed]
  83. Shliakhtunou, Y.A. CTCs-oriented adjuvant personalized cytostatic therapy non-metastatic breast cancer patients: Continuous non-randomized prospective study and prospective randomized controlled study. Breast Cancer Res. Treat. 2021, 186, 439–451. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, C.; Mu, Z.; Ye, Z.; Zhang, Z.; Abu-Khalaf, M.M.; Silver, D.P.; Palazzo, J.P.; Jagannathan, G.; Fellin, F.M.; Bhattacharya, S.; et al. Prognostic value of HER2 status on circulating tumor cells in advanced-stage breast cancer patients with HER2-negative tumors. Breast Cancer Res. Treat. 2020, 181, 679–689. [Google Scholar] [CrossRef]
  85. Bidard, F.-C.; Peeters, D.J.; Fehm, T.; Nolé, F.; Gisbert-Criado, R.; Mavroudis, D.; Grisanti, S.; Generali, D.; Garcia-Saenz, J.A.; Stebbing, J.; et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: A pooled analysis of individual patient data. Lancet Oncol. 2014, 15, 406–414. [Google Scholar] [CrossRef]
  86. Smerage, J.B.; Barlow, W.E.; Hortobagyi, G.N.; Winer, E.P.; Leyland-Jones, B.; Srkalovic, G.; Tejwani, S.; Schott, A.F.; O’Rourke, M.A.; Lew, D.L.; et al. Circulating Tumor Cells and Response to Chemotherapy in Metastatic Breast Cancer: SWOG S0500. J. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef]
  87. Stergiopoulou, D.; Markou, A.; Strati, A.; Zavridou, M.; Tzanikou, E.; Mastoraki, S.; Kallergi, G.; Georgoulias, V.; Lianidou, E. Comprehensive liquid biopsy analysis as a tool for the early detection of minimal residual disease in breast cancer. Sci. Rep. 2023, 13, 1258. [Google Scholar] [CrossRef]
  88. Radovich, M.; Jiang, G.; Hancock, B.A.; Chitambar, C.; Nanda, R.; Falkson, C.; Lynce, F.C.; Gallagher, C.; Isaacs, C.; Blaya, M.; et al. Association of Circulating Tumor DNA and Circulating Tumor Cells After Neoadjuvant Chemotherapy With Disease Recurrence in Patients With Triple-Negative Breast Cancer. JAMA Oncol. 2020, 6, 1410. [Google Scholar] [CrossRef] [PubMed]
  89. Gerratana, L.; Davis, A.A.; Polano, M.; Zhang, Q.; Shah, A.N.; Lin, C.; Basile, D.; Toffoli, G.; Wehbe, F.; Puglisi, F.; et al. Understanding the organ tropism of metastatic breast cancer through the combination of liquid biopsy tools. Eur. J. Cancer 2021, 143, 147–157. [Google Scholar] [CrossRef]
  90. Patelli, G.; Vaghi, C.; Tosi, F.; Mauri, G.; Amatu, A.; Massihnia, D.; Ghezzi, S.; Bonazzina, E.; Bencardino, K.; Cerea, G.; et al. Liquid Biopsy for Prognosis and Treatment in Metastatic Colorectal Cancer: Circulating Tumor Cells vs Circulating Tumor DNA. Target. Oncol. 2021, 16, 309–324. [Google Scholar] [CrossRef]
  91. Tzanikou, E.; Markou, A.; Politaki, E.; Koutsopoulos, A.; Psyrri, A.; Mavroudis, D.; Georgoulias, V.; Lianidou, E. PIK3CA hotspot mutations in circulating tumor cells and paired circulating tumor DNA in breast cancer: A direct comparison study. Mol. Oncol. 2019, 13, 2515–2530. [Google Scholar] [CrossRef] [PubMed]
  92. Hashimoto, T.; Nakamura, Y.; Oki, E.; Kobayashi, S.; Yuda, J.; Shibuki, T.; Bando, H.; Yoshino, T. Bridging horizons beyond CIRCULATE-Japan: A new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int. J. Clin. Oncol. 2024, 29, 495–511. [Google Scholar] [CrossRef] [PubMed]
  93. Black, J.R.M.; Bartha, G.; Abbott, C.W.; Boyle, S.M.; Karasaki, T.; Li, B.; Chen, R.; Harris, J.; Veeriah, S.; Colopi, M.; et al. Ultrasensitive ctDNA detection for preoperative disease stratification in early-stage lung adenocarcinoma. Nat. Med. 2025, 31, 70–76. [Google Scholar] [CrossRef]
  94. Japanese Promotion Council for Laboratory Testing. Perspectives on Quality Assurance of ctDNA Testing. 2022. Available online: https://www.jpclt.org/message/#ctDNA (accessed on 14 April 2025).
  95. Riethdorf, S. Detection of Circulating Tumor Cells in Peripheral Blood of Patients with Metastatic Breast Cancer: A Validation Study of the CellSearch System. Clin. Cancer Res. 2007, 13, 920–928. [Google Scholar] [CrossRef]
  96. Wang, P.X.; Sun, Y.F.; Jin, W.X.; Cheng, J.W.; Peng, H.X.; Xu, Y.; Zhou, K.Q.; Chen, L.M.; Huang, K.; Wu, S.Y.; et al. Circulating tumor cell detection and single-cell analysis using an integrated workflow based on ChimeraX®-i120 Platform: A prospective study. Mol. Oncol. 2021, 15, 2345–2362. [Google Scholar] [CrossRef]
  97. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.J.; Uhr, J.W.; Terstappen, L.W.M.M. Tumor Cells Circulate in the Peripheral Blood of All Major Carcinomas but not in Healthy Subjects or Patients With Nonmalignant Diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef] [PubMed]
  98. Ciccioli, M.; Kim, K.; Khazan, N.; Khoury, J.D.; Cooke, M.J.; Miller, M.C.; O’Shannessy, D.J.; Pailhes-Jimenez, A.-S.; Moore, R.G. Identification of circulating tumor cells captured by the FDA-cleared Parsortix® PC1 system from the peripheral blood of metastatic breast cancer patients using immunofluorescence and cytopathological evaluations. J. Exp. Clin. Cancer Res. 2024, 43, 240. [Google Scholar] [CrossRef]
  99. Oshiro, K.; Wakizaka, Y.; Takano, M.; Itoi, T.; Ohge, H.; Koba, K.; Yarimizu, K.; Fujiyoshi, S.; Maruyama, F. Fabrication of a new all-in-one microfluidic dielectrophoresis integrated chip and living cell separation. iScience 2022, 25, 103776. [Google Scholar] [CrossRef]
  100. Warkiani, M.E.; Khoo, B.L.; Wu, L.; Tay, A.K.P.; Bhagat, A.A.S.; Han, J.; Lim, C.T. Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics. Nat. Protoc. 2016, 11, 134–148. [Google Scholar] [CrossRef]
  101. Yang, C.; Zou, K.; Zheng, L.; Xiong, B. Prognostic and clinicopathological significance of circulating tumor cells detected by RT-PCR in non-metastatic colorectal cancer: A meta-analysis and systematic review. BMC Cancer 2017, 17, 725. [Google Scholar] [CrossRef]
  102. 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]
  103. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977, 37, 646–650. [Google Scholar]
  104. Descamps, L.; Le Roy, D.; Deman, A.-L. Microfluidic-Based Technologies for CTC Isolation: A Review of 10 Years of Intense Efforts towards Liquid Biopsy. Int. J. Mol. Sci. 2022, 23, 1981. [Google Scholar] [CrossRef]
  105. Paterlini-Brechot, P.; Linda Benali, N. Circulating tumor cells (CTC) detection: Clinical impact and future directions. Cancer Lett. 2007, 253, 180–204. [Google Scholar] [CrossRef]
  106. Lelievre, L.; Paterlini-Brechot, P.; Camatte, S.; Tartour, E.; Aggerbeck, M.; Vilde, F.; Lecuru, F. Effect of laparoscopy versus laparotomy on circulating tumor cells using isolation by size of epithelial tumor cells. Int. J. Gynecol. Cancer 2004, 14, 229–233. [Google Scholar] [CrossRef] [PubMed]
  107. Ozkumur, E.; Shah, A.M.; Ciciliano, J.C.; Emmink, B.L.; Miyamoto, D.T.; Brachtel, E.; Yu, M.; Chen, P.-I.; Morgan, B.; Trautwein, J.; et al. Inertial Focusing for Tumor Antigen–Dependent and –Independent Sorting of Rare Circulating Tumor Cells. Sci. Transl. Med. 2013, 5, 179ra47. [Google Scholar] [CrossRef]
  108. Warkiani, M.E.; Guan, G.; Luan, K.B.; Lee, W.C.; Bhagat, A.A.S.; Kant Chaudhuri, P.; Tan, D.S.-W.; Lim, W.T.; Lee, S.C.; Chen, P.C.Y.; et al. Slanted spiral microfluidics for the ultra-fast, label-free isolation of circulating tumor cells. Lab Chip 2014, 14, 128–137. [Google Scholar] [CrossRef] [PubMed]
  109. Huang, L.R.; Cox, E.C.; Austin, R.H.; Sturm, J.C. Continuous Particle Separation Through Deterministic Lateral Displacement. Science 2004, 304, 987–990. [Google Scholar] [CrossRef]
  110. Antolovic, D.; Galindo, L.; Carstens, A.; Rahbari, N.; Büchler, M.W.; Weitz, J.; Koch, M. Heterogeneous detection of circulating tumor cells in patients with colorectal cancer by immunomagnetic enrichment using different EpCAM-specific antibodies. BMC Biotechnol. 2010, 10, 35. [Google Scholar] [CrossRef]
  111. Balic, M.; Dandachi, N.; Hofmann, G.; Samonigg, H.; Loibner, H.; Obwaller, A.; Van Der Kooi, A.; Tibbe, A.G.J.; Doyle, G.V.; Terstappen, L.W.M.M.; et al. Comparison of two methods for enumerating circulating tumor cells in carcinoma patients. Cytom. Part B Clin. Cytom. 2005, 68B, 25–30. [Google Scholar] [CrossRef]
  112. Kong, L.X.; Perebikovsky, A.; Moebius, J.; Kulinsky, L.; Madou, M. Lab-on-a-CD: A Fully Integrated Molecular Diagnostic System. SLAS Technol. 2016, 21, 323–355. [Google Scholar] [CrossRef]
  113. Nagrath, S.; Sequist, L.V.; Maheswaran, S.; Bell, D.W.; Irimia, D.; Ulkus, L.; Smith, M.R.; Kwak, E.L.; Digumarthy, S.; Muzikansky, A.; et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007, 450, 1235–1239. [Google Scholar] [CrossRef]
  114. Augustsson, P.; Magnusson, C.; Nordin, M.; Lilja, H.; Laurell, T. Microfluidic, Label-Free Enrichment of Prostate Cancer Cells in Blood Based on Acoustophoresis. Anal. Chem. 2012, 84, 7954–7962. [Google Scholar] [CrossRef] [PubMed]
  115. Fabbri, F.; Carloni, S.; Zoli, W.; Ulivi, P.; Gallerani, G.; Fici, P.; Chiadini, E.; Passardi, A.; Frassineti, G.L.; Ragazzini, A.; et al. Detection and recovery of circulating colon cancer cells using a dielectrophoresis-based device: KRAS mutation status in pure CTCs. Cancer Lett. 2013, 335, 225–231. [Google Scholar] [CrossRef]
  116. Mahabadi, S.; Labeed, F.H.; Hughes, M.P. Effects of cell detachment methods on the dielectric properties of adherent and suspension cells. Electrophoresis 2015, 36, 1493–1498. [Google Scholar] [CrossRef]
  117. Mohammadali, R.; Bayareh, M.; Nadooshan, A.A. Performance optimization of a DLD microfluidic device for separating deformable CTCs. Electrophoresis 2024, 45, 1775–1784. [Google Scholar] [CrossRef] [PubMed]
  118. Lemaire, C.A.; Liu, S.Z.; Wilkerson, C.L.; Ramani, V.C.; Barzanian, N.A.; Huang, K.-W.; Che, J.; Chiu, M.W.; Vuppalapaty, M.; Dimmick, A.M.; et al. Fast and Label-Free Isolation of Circulating Tumor Cells from Blood: From a Research Microfluidic Platform to an Automated Fluidic Instrument, VTX-1 Liquid Biopsy System. SLAS Technol. 2018, 23, 16–29. [Google Scholar] [CrossRef] [PubMed]
  119. He, W.; Kularatne, S.A.; Kalli, K.R.; Prendergast, F.G.; Amato, R.J.; Klee, G.G.; Hartmann, L.C.; Low, P.S. Quantitation of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands. Int. J. Cancer 2008, 123, 1968–1973. [Google Scholar] [CrossRef]
  120. Schmitt, M.; Foekens, J.A. Circulating tumor cells in blood of primary breast cancer patients assessed by a novel RT-PCR test kit and comparison with status of bone marrow-disseminated tumor cells. Breast Cancer Res. 2009, 11, 109. [Google Scholar] [CrossRef]
  121. Yang, J.; Antin, P.; Berx, G.; Blanpain, C.; Brabletz, T.; Bronner, M.; Campbell, K.; Cano, A.; Casanova, J.; Christofori, G.; et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2020, 21, 341–352. [Google Scholar] [CrossRef]
  122. Hsu, C.-L.; Tsai, T.-H.; Huang, C.-K.; Yang, C.-Y.; Liao, W.-Y.; Ho, C.-C.; Ruan, S.-Y.; Chen, K.-Y.; Shih, J.-Y.; Yang, P.-C. Monitoring levels of vimentin-positive circulating cancer stem cells and tumor cells in patients with advanced EGFR-mutated non-small cell lung cancer. Lung Cancer 2021, 156, 50–58. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, Y.-P.; Giret, T.M.; Cote, R.J. Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers 2021, 13, 2723. [Google Scholar] [CrossRef] [PubMed]
  124. National Center for Biotechnology Information, National Library of Medicine. ClinicalTrial.gov. Available online: https://clinicaltrials.gov/ (accessed on 14 May 2025).
  125. Jiang, Z.F.; Cristofanilli, M.; Shao, Z.M.; Tong, Z.S.; Song, E.W.; Wang, X.J.; Liao, N.; Hu, X.C.; Liu, Y.; Wang, Y.; et al. Circulating tumor cells predict progression-free and overall survival in Chinese patients with metastatic breast cancer, HER2-positive or triple-negative (CBCSG004): A multicenter, double-blind, prospective trial. Ann. Oncol. 2013, 24, 2766–2772. [Google Scholar] [CrossRef] [PubMed]
  126. Wallwiener, M.; Riethdorf, S.; Hartkopf, A.D.; Modugno, C.; Nees, J.; Madhavan, D.; Sprick, M.R.; Schott, S.; Domschke, C.; Baccelli, I.; et al. Serial enumeration of circulating tumor cells predicts treatment response and prognosis in metastatic breast cancer: A prospective study in 393 patients. BMC Cancer 2014, 14, 512. [Google Scholar] [CrossRef]
  127. Souza E Silva, V.; Ali Abdallah, E.; Lopes de Mello, C.A.; Tariki, M.S.; Calsavara, V.F.; Chinen, L.T.D. Prospective study with circulating tumor cells as potential prognosis biomarker in metastatic colorectal cancer. J. Clin. Oncol. 2020, 38, 203. [Google Scholar] [CrossRef]
  128. Bidard, F.-C.; Bidard, F.-C.; Kiavue, N.; Jacot, W.; Bachelot, T.; Dureau, S.; Bourgeois, H.; Goncalves, A.; Brain, E.; Ladoire, S.; et al. Overall Survival With Circulating Tumor Cell Count–Driven Choice of Therapy in Advanced Breast Cancer: A Randomized Trial. J. Clin. Oncol. 2024, 42, 383–389. [Google Scholar] [CrossRef]
Figure 1. Overview of tumor dissemination and blood collection for CTC analysis. Colorectal (yellow, panel (a)) and breast (pink, panel (b)) carcinomas invade surrounding tissues, reach nearby veins (red purple, or blue), and enter the bloodstream as CTCs (yellow or pink circles). In panel (a), CTCs of colorectal cancer are frequently transported to the liver (brown) via the mesenteric and portal veins (red-purple) and subsequently reach the lungs (dark purple). In contrast, lower rectal cancer cells primarily metastasize to the lungs (dark purple) via the inferior rectal vein (blue), inferior vena cava, and heart. In panel (b), breast cancer cells are transported to the bones (not shown), lungs (dark purple), and liver (brown) through the venous circulation and heart.
Figure 1. Overview of tumor dissemination and blood collection for CTC analysis. Colorectal (yellow, panel (a)) and breast (pink, panel (b)) carcinomas invade surrounding tissues, reach nearby veins (red purple, or blue), and enter the bloodstream as CTCs (yellow or pink circles). In panel (a), CTCs of colorectal cancer are frequently transported to the liver (brown) via the mesenteric and portal veins (red-purple) and subsequently reach the lungs (dark purple). In contrast, lower rectal cancer cells primarily metastasize to the lungs (dark purple) via the inferior rectal vein (blue), inferior vena cava, and heart. In panel (b), breast cancer cells are transported to the bones (not shown), lungs (dark purple), and liver (brown) through the venous circulation and heart.
Ijms 26 05539 g001
Table 1. Comparison between CTCs and ctDNA.
Table 1. Comparison between CTCs and ctDNA.
FactorCTCsctDNA
Discovery year1869 11977 2
Detection targetViable tumor cellsCell-free DNA fragments
Release mechanismIntravasation and circulationApoptosis or necrosis
SensitivityVaries by separation technique 3Relatively high
Amplification in detectionNot required (but available) 4Required
Monitoring MRDYesYes
HeterogeneityIntra- and intertumor cellsIntra-/intertumor + mutated benign cells
Pathology/cytologyYesNot applicable
Functional assayYesNot applicable
Gene expressionYesNot applicable
Gene alterationYesYes
Epigenetic changesYesYes (DNA methylation)
CharacteristicsPreserved cell morphology and phenotypeDNA sequence and methylation
Chemosensitivity testYesNo
Feasibility in hospital settingHigh (routine practice) 5Limited (specialized setting) 6
Outsourced serviceAvailable from commercial servicesAvailable from commercial services
1, [102]; 2, [103]; 3, [104]; 4, [105]; 5, [95,96,97]; 6, [94].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kakizaki, F.; Oshiro, K.; Enoki, Y.; Kawanishi, K.; Masuda, N.; Maekawa, H.; Matsubayashi, J.; Kawashima, M.; Miyoshi, H.; Takemura, Y.; et al. Precision Oncology Framework Using Circulating Tumor Cells. Int. J. Mol. Sci. 2025, 26, 5539. https://doi.org/10.3390/ijms26125539

AMA Style

Kakizaki F, Oshiro K, Enoki Y, Kawanishi K, Masuda N, Maekawa H, Matsubayashi J, Kawashima M, Miyoshi H, Takemura Y, et al. Precision Oncology Framework Using Circulating Tumor Cells. International Journal of Molecular Sciences. 2025; 26(12):5539. https://doi.org/10.3390/ijms26125539

Chicago/Turabian Style

Kakizaki, Fumihiko, Kyoichi Oshiro, Yuya Enoki, Kana Kawanishi, Norikazu Masuda, Hisatsugu Maekawa, Jun Matsubayashi, Masahiro Kawashima, Hiroyuki Miyoshi, Yukitoshi Takemura, and et al. 2025. "Precision Oncology Framework Using Circulating Tumor Cells" International Journal of Molecular Sciences 26, no. 12: 5539. https://doi.org/10.3390/ijms26125539

APA Style

Kakizaki, F., Oshiro, K., Enoki, Y., Kawanishi, K., Masuda, N., Maekawa, H., Matsubayashi, J., Kawashima, M., Miyoshi, H., Takemura, Y., & Obama, K. (2025). Precision Oncology Framework Using Circulating Tumor Cells. International Journal of Molecular Sciences, 26(12), 5539. https://doi.org/10.3390/ijms26125539

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