Next Article in Journal
Beyond the Influence of IDH Mutations: Exploring Epigenetic Vulnerabilities in Chondrosarcoma
Previous Article in Journal
Angiopoietin-2 Combined with Radiochemotherapy Impedes Glioblastoma Recurrence by Acting in an Autocrine and Paracrine Manner: A Preclinical Study
Previous Article in Special Issue
A Multi-Analyte Approach for Improved Sensitivity of Liquid Biopsies in Prostate Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Characteristics and Predictive Biomarkers on Circulating Tumor Cells for Therapeutic Strategy

1
Leading Center for the Development and Research of Cancer Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
2
Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
3
Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
4
Departments of Pathology and Oncology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(12), 3588; https://doi.org/10.3390/cancers12123588
Submission received: 14 October 2020 / Revised: 27 November 2020 / Accepted: 28 November 2020 / Published: 30 November 2020
(This article belongs to the Special Issue Circulating Tumor Cells' Heterogeneity and Precision Oncology)

Abstract

:

Simple Summary

Circulating tumor cells (CTCs) are the seeds that spread through the circulatory system and generate the metastatic sites. Among the various phenotypes exhibited by CTCs, epithelial-to-mesenchymal transition (EMT) has extensively gained attention because of its contribution to the acquisition of invasiveness and motility of CTCs, which is critical for the successful metastasis process. So far, many clinical studies have demonstrated that expressions of mesenchymal features in CTCs are associated with poor clinical outcomes in many types of solid cancer. In this review, the clinical significance of CTCs as a liquid biopsy is described in terms of the mesenchymal characteristics, expression of predictive biomarkers, and their correlations. Detailed analysis of these multivariate targets in CTCs could improve therapeutic decision-making.

Abstract

Metastasis-related events are the primary cause of cancer-related deaths, and circulating tumor cells (CTCs) have a pivotal role in metastatic relapse. CTCs include a variety of subtypes with different functional characteristics. Interestingly, the epithelial–mesenchymal transition (EMT) markers expressed in CTCs are strongly associated with poor clinical outcome and related to the acquisition of circulating tumor stem cell (CTSC) features. Recent studies have revealed the existence of CTC clusters, also called circulating tumor microemboli (CTM), which have a high metastatic potential. In this review, we present current opinions regarding the clinical significance of CTCs and CTM with a mesenchymal phenotype as clinical surrogate markers, and we summarize the therapeutic strategy according to phenotype characterization of CTCs in various types of cancers for future precision medicine.

1. Introduction

The major cause of death in ~80% of cancer cases is a metastatic cancer lesion-related complication. Increasing numbers of circulating tumor cells (CTCs) are now utilized as surrogate endpoint markers [1,2,3,4,5], and many clinical studies have demonstrated that the presence of CTCs in the blood of patients associate with short recurrence-free survival and high risk of metastasis for several types of solid cancers [6,7,8,9,10,11,12,13]. Thus, CTCs are a glaringly obvious direct seed for forming future micro-metastatic colonies in distal organs. However, micro-metastatic colonization is a challenging process as the minorities of the surviving cells are exposed to significant shear force, innate immunity, chemotherapy, and oxidative stress under anoikis (detachment-induced apoptosis) in the bloodstream. In this respect, epithelial-to-mesenchymal transition (EMT) is related to survival by allowing cancer cells to avoid apoptosis and acquire the subsequent chemoresistance even under anoikis in the bloodstream [14]. The monitoring of CTCs could lead to the prediction of the future clinical time course of cancer development and/or early recurrence period at the onset of initial cancer development and to the detection of micro-metastatic lesions [15]. Therefore, the significance of CTCs as clinical surrogate markers is different from that of other diverse liquid biopsies (i.e., circulating tumor DNA, ctDNA, exosomes, or diverse serum liquid biomarkers) [16,17,18,19]. Among these, ctDNA measurement is the most popular diagnostic and prognostic tool that averages the status of the whole tumor burden and provides information on the response/resistance to chemotherapy, targeted therapy, and immunotherapy; therefore, it is useful for decision-making [17,18]. However, these biomarkers are considered as indirect markers released from tumor tissues into the bloodstream. In contrast, the detection of CTCs and circulating tumor microemboli (CTM) provide direct evidence for the existence of cancer lesions, and thus their measurement and phenotypic characterization could be a reliable diagnostic tool for cancer. The mesenchymal features of CTCs have been reported and, based on the expression level of epithelial and/or mesenchymal markers in CTCs, three different CTC subtypes—epithelial CTCs, hybrid CTCs that involve both epithelial and mesenchymal markers, and mesenchymal CTCs—were identified [20] (Figure 1). CellSearch® is the most commonly used detection method for CTCs worldwide, utilizing antibodies against the epithelial markers epithelial cell adhesion molecule (EpCAM) and cytokeratin. CTC detection by the CellSearch® system (generally two or more CTCs/7.5 mL blood as positive) does not have a high sensitivity in non-small cell lung cancer (NSCLC) and pancreatic ductal adenocarcinoma (PDAC), giving a result of only 20% even in the advanced metastatic stage [21]. This contributes to the underestimation of CTC number and could be, in part, due to the high number of CTCs with a mesenchymal phenotype undergoing EMT (i.e., from hybrid CTCs to EMT-CTCs) [22,23]. Similarly, a study conducted on malignant pleural mesothelioma using the CellSearch® system showed a low detection rate of 33%, suggesting a need for efficient EpCAM-independent detection systems of CTCs [24]. CTCs morphologically and biologically involve different phenotypes and consist of single cells and/or cell clusters with or without the EMT feature, which plays an essential role in the metastatic process. CTM is also seen in patients with advanced cancers [25] and is detected as multicellular cancer fragments, ranging from 2 to 100 cancer cells (with most clusters comprising between 20 and 40 cells), in the blood circulation of patients with diverse metastatic cancers [25,26,27,28,29,30]. Cluster CTCs also contain cancer-associated fibroblasts, platelets, and immune cells [31]. CTM appears to be more aggressive than single CTCs in developing cancer invasion [32,33,34]. However, it remains unclear which types of CTCs (single versus clusters) play a critical role in the metastasis process.
In the past decade, the detection of CTCs as clinical biomarkers has been reported using diverse, highly sensitive methods. A randomized trial including patients with metastatic breast cancer with a persistent increase in CTCs reported that an early switch to an alternative cytotoxic therapy after the failure of the first-line chemotherapy prolonged the overall survival (OS) (SWOG S0500) [5]. Recent CTC-related studies focused on further detailed approaches, including phenotypic characterization of predictable therapeutic response markers through genotypic and immunophenotypic analysis. These approaches can provide useful predictive information for the decision-making in precision medicine, but CTCs represent a heterogeneous population of tumor cells with the potential for metastases [35]. Thus, CTC measurements to predict treatment efficacy are still insufficient in providing useful clinical biomarkers with clear evidence.
Here, we summarize the clinical significance of CTCs, including the mesenchymal phenotype and the potential treatment response markers for future precision medicine.

2. The Significance of EMT in CTCs as Prognostic and Predictive Biomarkers

It is widely known that EMT contributes to the generation of CTCs by facilitating intravasation into the blood circulation [36]. EMT is induced by transcriptional factors, and the Twist–Snail axis plays a vital role in the transcriptional regulation of EMT and metastasis to maintain the stability and survival of CTCs in the bloodstream by avoiding anoikis suppression [37]. Zeb1, which acts as a repressor for E-cadherin transcription, protects anoikis-induced apoptosis and causes metastatic tumors in vivo [38]. Thus, these transcriptional regulators of EMT are also utilized as detection markers for EMT-CTCs. Vimentin, a cytoplasmic protein comprising intermediate filaments, is known to be upregulated in EMT-induced cancer cells and correlated with cancer progression [39,40,41]. In addition, some vimentin molecules expressed in cancer cells migrate to the cell surface and have been identified as cancer-specific EMT markers, called cell-surface vimentin (CSV), and were shown to correlate with highly invasive and metastatic properties of cancers [42,43,44,45,46,47]. Subsequent mesenchymal–epithelial transition (MET), the reverse process of EMT, in the metastatic cascade for extravasation into secondary metastatic sites [48] reportedly induces the re-expression of E-cadherin [49]. EMT leads not only to recurrent metastasis but also to chemoresistance. Yu et al. followed the epithelial/mesenchymal marker status of hybrid CTCs. When patients with breast cancer responded to chemotherapy, epithelial marker-dominant CTCs were dramatically reduced, and when patients developed recurrence, the amount of mesenchymal marker-dominant CTCs was significantly increased [26]. Furthermore, blocking EMT contributed to enhanced chemosensitivity and increased the OS of genetically engineered cancer mouse models [50]. Patients with advanced gastric cancer and breast cancer showed an increase in mesenchymal CTCs in the post-chemotherapy blood specimens, which was associated with the progressive disease, suggesting that monitoring the EMT status of CTCs can provide reliable information to predict the therapy response and cancer recurrence for future precision medicine [22,26,51,52]. In a study conducted for primary breast cancer, 77 (18.0%) of 427 primary breast cancer patients were found to have EMT-CTCs characterized by TWIST, SNAIL1, SLUG, and ZEB1 expression levels, and patients without EMT-CTCs had a longer disease-free survival than those with EMT-CTCs [53]. Positive EMT-CTCs at the baseline are associated with poor clinical outcome for treatment responsiveness in patients with advanced NSCLC [23]. The presence of mesenchymal markers, Twist1+, vimentin+, or CSV+, in CTCs predicts a worse prognosis than the expression of epithelial markers alone in metastatic cancers [46,47,54]. A study of metastatic breast cancer has demonstrated that overexpression of EMT-inducing transcription factors is associated with shorter progression-free survival (PFS), and EMT-CTCs are associated with recurrence [55]. Furthermore, patients with metastatic diseases have a high incidence of EMT-CTCs compared to those with early-stage diseases [56,57,58,59]. In contrast, EMT-CTCs have been reported as dispensable biomarkers to predict prognosis in patients with castrate-resistant prostate cancer (CRPC), PDAC, and breast cancer [60,61,62]. Furthermore, inhibiting EMT by overexpressing the microRNA miR-200 does not affect lung metastasis but abrogates EMT-mediated chemoresistance [63]. Patients with PDAC show KRAS mutations in CTCs [64,65], indicating a prognostic value, which does not depend on the expression of ZEB1, the EMT marker [66]. These negative results for the value of EMT-CTCs as prognostic markers are probably due to the diverse mechanisms that induce EMT and the lack of ability to detect EMT-CTC subtypes in clinical settings [67]. The clinical significances of CTCs with mesenchymal phenotype are summarized in Table 1.
Since mesenchymal markers such as N-cadherin and vimentin are frequently expressed in blood cells, it is necessary to remove these blood cells during the enrichment and detection processes of CTCs. One approach to enable blood cell removal is the negative selection with blood cell-specific markers, such as CD45, not expressed in cancer cells. Although there are no universal markers for specific enrichment and detection of mesenchymal CTCs, various detection systems have combined negative selection of blood cells with positive selection by mesenchymal markers to achieve highly sensitive EMT-CTC detection [85,86,87,88,89].

2.1. The Early Occurrence of EMT in CTCs

A large meta-analysis of 19 studies enrolling 2993 early-stage breast cancer patients demonstrated the detection of the CTCs as a stable prognosticator in early-stage breast cancer [90]. Several studies have identified CTCs expressing EMT markers in patients with early-stage breast cancer [57,91,92]. Mego et al. reported that CTCs exhibiting EMT phenotype are more frequent than those expressing only epithelial markers [91]. Furthermore, RT-PCR characterization of CTCs isolated from early-stage breast cancer patients, where Twist1 expression was used as an EMT marker, revealed the presence of EMT-CTCs in 31% of patients [93], whereas immunostaining experiments revealed that EMT-CTC expressing Twist1 and vimentin were identified in 73% and 77% of patients with early-stage breast cancer, respectively [57]. The occult micro-metastatic tumor cells establish cytokeratin-positive cells in the lymph nodes, and the overexpression of EMT markers, occurring at an early stage, is associated with a worse OS even in patients with curatively resected stage I NSCLC [94,95,96]. CTC-positive patients with NSCLC and breast cancer at the time of definitive surgery showed short recurrence-free periods [97,98,99]. Thus, CTCs isolated from early stages of lung cancer are predictors of poor prognosis and can be used to determine biomarkers predictive of early recurrence [35]. EMT-CTCs were identified in patients with early-stage NSCLC before receiving surgical operation, and the population of EMT-CTCs was recognized at an almost 50% lower rate than that in patients with some other solid tumors [23,68]. In a previous study, CTCs with EMT markers were detected in a high-risk lung cancer group without clinically detectable tumors before the occurrence of early-stage lung cancer [100]. Thus, the development of a system with EMT-CTC detection capability will enable high-sensitivity CTC detection from early stages of cancer to advanced metastatic stages compared to systems that use only epithelial markers to detect CTCs such as the CellSearch® system.

2.2. The Metastatic Ability of CTCs and CTC Clusters with a Mesenchymal Phenotype

The increase in the percentage of CTCs positive for the mesenchymal marker is higher in metastatic breast cancer than in early-stage breast cancer [57], denoting a more aggressive and metastatic potential of mesenchymal CTCs than those with epithelial markers alone [56,73,76,78,79,80,101,102]. Hybrid CTCs, cytokeratins+/vimentin+/CD45, are also associated with disease progression and the presence of distant metastasis [103,104]. EMT-CTCs have specific markers that allow a more accurate prediction of worse prognosis and metastasis. BCAT1-induced EMT-CTCs may be an important biomarker of hepatocellular carcinoma metastasis [105]. Twist+ CTCs with a higher expression of glypican-3 are closely correlated with the rate of metastasis, recurrence, and mortality [105]. EMT-CTCs with high LGR5 expression significantly correlate with the occurrence of metastasis [105]. These findings suggest that EMT-CTCs play a crucial role in developing initial metastatic colonization at distal organs. A model for cancer metastasis has been proposed in which EMT promotes the release of both EMT and non-EMT cells from the primary tumor and vascular invasion, and non-EMT cancer cells in the bloodstream collaborate with EMT cells to establish colonies in the secondary sites [106]. CTC clusters correlate with higher metastatic potential and shorter survival times than single CTCs [107,108,109,110,111]. CTC clusters increase the metastatic potential by approximately 50-fold compared with single CTCs in experimental models [33]. CTM maintains the epithelial feature of preserving cell–cell junctions, and CTM displaying plakoglobin, a cell adhesion protein that contributes to the CTC cluster formation, is an independent prognostic factor, particularly for distant-metastasis-free survival in patients with breast and prostate cancer [33]. Platelets bound to CTC clusters in the blood circulation release TGF-β, which leads to EMT in the CTC cluster, and this partial EMT status in the hybrid CTC cluster may have a higher metastatic potential and a survival advantage during colonization at the distant site [26,112].

3. Cancer Stem Cells (CSCs) with a Mesenchymal Phenotype

CTCs expressing CSC markers indicate tumor recurrence and exhibit significant resistance to conventional chemotherapy [113]. EMT promotes the acquisition of CSC-like properties [114], and the CSC phenotype also expresses genes associated with the EMT features, resulting in enhanced metastatic ability [115]. Cases of gastric cancer with CD44+ in CK+ epithelial CTCs are significantly more common among patients with distant metastases and associated with shorter survival than that observed in the case of patients with CK+CD44 CTCs [71]. NSCLC patients with an increased ratio of CD133+ CTCs to pan-CK-positive cell type (stem cell-like to epithelial ratio) and those that have mesenchymal N-cadherin-positive CTCs are significantly associated with shortened PFS [116]. The co-expression of the stem cell marker CD133 in hybrid CTCs (CK+, vimentin+, and N-cadherin+) has also been detected at a high frequency in metastatic breast cancer [117,118]. Aldehyde dehydrogenase 1 (ALDH1), a CSC marker, has potential diagnostic and therapeutic implications [119], and ALDH1+ CTCs positivity in patients with NSCLC was found to be associated with the disease stage [120]. Additionally, ALDH1+ EMT-CTCs (vimentin+ and fibronectin+) are isolated from patients at a higher frequency with metastatic breast cancer and correlated with the disease stage [121]. The expression of both ALDH1 (ALDH 1high) and nuclear Twist (Twistnuc) CTCs in patients with metastatic breast cancer is higher than that in patients with early-stage breast cancer [92]. CTCs with positive EMT markers and/or ALDH1 expression indicate therapy resistance in patients with metastatic breast cancer [122]. The expression of the stemness markers CD44+/CD24 and ALDH1+ in CTCs is associated with an enhanced tumorigenic potential in breast cancer [123]. The glioblastoma-derived CTC expressing the CSC phenotype, which includes the stemness-associated genes SOX2, OCT4, and NANOG, contributes to local tumorigenesis and recurrence by the process of self-seeding [124]. These studies indicate that the CTCs that acquire CSC properties, namely circulating cancer stem cells (CTSCs), are more frequent in advanced-stage cancers and are associated with poor clinical outcomes. The clinical significances of CTSCs are summarized in Table 2.

4. The Concordance between the Characteristics of CTCs and Primary Tumors

Detection of CTC driver mutation as a predictable molecular targeted therapy response, which reflects the concordance with primary tissue status, is essential for non-invasive therapeutic decision-making. The concordance of KRAS status, a biomarker for predicting the therapeutic efficiency of anti-epidermal growth factor receptor (EGFR) therapy, between CTCs and the corresponding colorectal cancer tumor tissues was 77% [125]. Similarly, Buim et al. also showed that KRAS mutations (codon 12 and 13) in CTCs and matched primary tumor from patients with colorectal cancer showed a high grade of concordance of 71% (p = 0.017), which suggests that CTCs can be used as surrogates of primary tumors in clinical practice [126]. Several other studies have also shown similar results [126,127,128,129]. Kalikaki et al. characterized CTCs from 31 metastatic colorectal cancer patients for KRAS mutations on codons 12 and 13 and showed 45% and 16.7% of patients with mutant and wild-type primary tumors, respectively, have detectable mutations in their CTCs [130]. They further revealed that individual patients’ CTCs in serial blood samples exhibited different mutational statuses of KRAS during treatment [130]. Furthermore, it has been shown that the frequency of at least one matching KRAS mutation between CTCs and PDAC was 58% [64], and the discordant KRAS mutations in CTCs and corresponding tumors were not associated with OS [64,66]. These studies show the importance of utilizing CTCs as a dynamic source of liquid biopsy for real-time genotyping of tumor cells during treatment.
In breast cancer, the human epidermal growth factor receptor 2 (HER2) oncogene has been shown to play an important role in growth and progression [131,132]. A good correspondence of HER2 amplification was found between CTCs and the primary tumor as well as the immunohistochemistry results [133], suggesting that the status of HER2 on CTCs reflects the status of the primary tumor. However, several studies have demonstrated that HER2 status might change during disease progression [134,135,136,137]. Moreover, discordance between the expression of HER2 and the estrogen receptor (ER) in CTCs and that in breast cancer tumor tissues occurs frequently [138], and thus has no prognostic impact on patients with metastatic breast cancer with a HER2-negative primary tumor receiving endocrine therapy [134,138,139]. These discordances could be, in part, due to the inability of traditional biopsies that sample a small piece of tumor tissue to reflect the heterogeneity of cancer cells in tumor tissue leading to failure in cancer prognosis and prediction. In contrast, several studies have provided evidence that the heterogeneity of CTCs reflects the cellular heterogeneity of the primary and metastatic tumor [140,141,142]; moreover, CTCs often acquire different characteristics from their original tissue during the metastatic process. Therefore, a detailed characterization of the expression of CTC markers could provide useful information in predicting prognosis and treatment response as a real-time biopsy [139,143].

5. The Potential Markers on CTCs for Precision Medicine

Detection of predictive biomarkers for treatment response on CTCs can improve clinical decision-making, and many studies have been conducted to identify reliable biomarkers. These potential treatment response markers for future precision medicine are summarized in Table 3.

5.1. The Analysis of Molecular Target Expression in CTCs as a Predictive Biomarker for Treatment Response

HER2 gene amplification in CTCs was determined during cancer progression, even in patients with HER2-negative primary tumors [144]. Detected HER2-positive CTCs in patients with early-stage breast cancer [145,146] and patients that had HER2-positive CTCs before neoadjuvant therapy were associated with shortened disease-free survival and OS [35]. The predictive treatment response values of HER2-positive CTC status have been demonstrated in patients receiving anti-HER2 therapy [147]. However, trastuzumab, an anti-HER2 antibody therapy, did not decrease the detection rate of CTCs in patients with non-HER2-amplified early breast cancer following surgery after chemotherapy [148]. A digital RNA analysis of CTCs demonstrated that the failure to suppress ER signaling within CTCs after endocrine therapy predicts early progression, and therefore, scoring the CTC-RNA signatures could guide the therapeutic options [149]. CTCs with synaptophysin expression are also a useful biomarker of both the treatment efficacy of and resistance to androgen receptor-targeted therapies in patients with metastatic CRPC [150]. Other specific and predictable chemoresponse markers in CTCs have also been reported. In patients with metastatic breast cancer, a decrease in the topoisomerase 1 (Top1) level in CTCs after the initial cycle of administration of the Top1 inhibitor etirinotecan pegol was associated with an increased OS [151]. During treatment, Bcl-2+/CD45 CTCs seemed to be a dynamic biomarker for predicting the treatment efficacy and clinical outcome in patients with chemotherapy-naive small cell lung cancer (SCLC) [152].

5.2. The Analysis of Oncogenic Driver Mutation in CTCs as a Predictive Biomarker for Treatment Response

The efficacy and prognosis of EGFR-tyrosine kinase inhibitor (TKI) treatment for patients with EGFR driver mutations were reportedly better in the low CTC group than in the high CTC group in patients with NSCLC [153]. However, CTCs contain multiple EGFR mutations due to CTC heterogeneity, as evidenced by next-generation sequencing [154]. In patients with EGFR-mutant tumors progressing on EGFR-TKI therapy, 70% of CTCs contained the EGFR T790M mutation and acquired EGFR-TKI resistance [153]. Anaplastic lymphoma kinase (ALK) rearrangement with aberrant ALK-fluorescence in situ hybridization (FISH) patterns can be detected in CTCs in concordance with patients having ALK-positive NSCLC, enabling both diagnostic testing and decision-making for ALK inhibitors. These ALK-rearranged CTCs also express a mesenchymal phenotype and decrease the response to crizotinib [155]. Decreasing ALK copy number in CTCs is associated with treatment response and a longer PFS [156]. Single-cell sequencing of CTCs from ALK-rearranged patients may provide information to assess mutations that cause therapeutic resistance to ALK inhibitors [157]. Androgen receptor (AR) amplification is conserved between CTCs and tumor biopsies in patients with metastatic CRPC, and CTCs can serve as a non-invasive surrogate to document AR amplification [158]. It has been demonstrated that AR splice variant 7 (AR-V7) expression in CTCs and CTC clusters predicts poor treatment response in metastatic CRPC patients treated with novel hormonal therapies, such as abiraterone or enzalutamide [159,160]. CTCs expressing AR-V7 and ARv567es+, as well as the percentage of androgen receptor nuclear localization, are associated with sensitivity to taxane therapy [161,162]. Phenotypic profiling of CTCs isolated from patients with metastatic CRPC (mCRPC) has revealed elevated levels of AR-V7 and the mesenchymal marker, N-cadherin, in patients with progressive cancer [163].

5.3. The Immunophenotype Analysis of CTCs as Predictive Biomarkers for Immune Checkpoint Therapy Response

Programmed death-ligand 1 (PD-L1) is widely implicated in tumor immune evasion, and PD-L1 expression in tumor tissues has been reported to be associated with the responses to the current immune checkpoint therapy of melanoma and NSCLC [164,165,166,167,168]. Therefore, PD-L1 expression in CTCs is used as a predictive response value for PD-1/PD-L1 inhibitor immune checkpoint therapies. However, the concordance of PD-L1 expression between tumor tissues and CTCs is not high [169,170,171]. This might support the conclusion that the presence of PD-L1+CTC is not significantly correlated with the treatment outcomes of nivolumab, a PD-1 inhibitor, and a higher baseline PD-L1+CTC number does not lead to a response in patients with advanced NSCLC [172]. Interestingly, the levels of EMT-CTCs are negatively correlated with T-cell immunity, and the levels of CD3+ and CD8+ T cells in NSCLC suggest resistance to cancer immunity [173]. Patients who show an increase in PD-L1+CTCs are predicted to have resistance to PD-1/PD-L1 inhibitors, while no changes or a decrease in PD-L1+CTCs is observed in responding patients [170]. Patients with advanced melanoma and gastrointestinal tumors with detectable PD-L1+CTCs also responded to the PD-1 inhibitor and longer PFS [174,175]. A higher number of CSV+PD-L1+ CTCs are significantly associated with short survival and poor therapeutic responses in gastric cancer patients [46].

5.4. The Combination Analysis of EMT and Predictive Markers for Treatment Decision-Making

The existence of EMT-CTCs was found to correlate with drug resistance [54,184]. Therefore, the detection and monitoring of EMT as a marker of treatment resistance before and during treatment is important to improve the quality of treatment management. High EMT marker expression has also been associated with high expression of immune checkpoint markers, such as PD1, PD-L1, CTLA4, OX40L, and PD-L2 [185,186,187,188]. For example, several studies have shown that EMT in cancer cells increases the expression of PD-L1 and consequently confers immune tolerance in cancer cells [72,189,190,191,192]. Analyses of CTCs in NSCLC patients revealed that PD-L1 expression on EMT-CTCs associated with significantly poorer survival after curative surgery, and PD-L1 and EMT markers were expressed at significantly higher proportions in CTCs than patient-matched NSCLC tissues [72,190]. Furthermore, it was reported that PD-L1 expressed in the nucleus of vimentin-positive CTCs predicts poor prognosis in colorectal and prostate cancer patients [193]. Several studies suggest complex bidirectional regulation between EMT and PD-L1 signaling, leading to tumor immune escape [194,195,196]. The combined analysis of EMT and PD-L1 expression in CTCs could be an effective method for predicting and monitoring the outcome of immunotherapy. EMT-related genes in CTCs are also candidate markers for combination analysis. For example, in metastatic prostate cancer, IGF1, IGF2, EGFR, FOXP3, and TGFB3 were commonly observed in CTCs of castration-sensitive cases, whereas an additional subset of EMT-related genes (e.g., PTPRN2, ALDH1, ESR2, and WNT5A) were expressed in CTCs of castration-resistant cases [197]. Mesenchymal phenotype and increased expression level of AR-V7 in CTCs from patients with mCRPC have been observed for those with a limited response to abiraterone and enzalutamide (androgen-signaling targeted inhibitors) treatment [163]. Further detailed molecular analysis of the relationship between EMT and predictive biomarkers could lead to a better understanding of the mechanisms of treatment resistance and create a reliable basis for therapeutic decision-making.

6. Conclusions

The detailed molecular characteristics, including mesenchymal and cancer stem cell features, in single CTC and CTM are required to develop reliable and predictable biomarkers as prognostic, diagnostic, and therapeutic strategies. However, CTCs have a diverse and heterogeneous phenotype, and thus, the clinical value of CTCs and CTM with mesenchymal phenotype still does not translate into clinical applications. Furthermore, the expression of some pharmaco-molecular targets in CTCs is different from that in primary tumors, making it difficult to standardize the role of CTCs as a companion diagnostic tool. On the contrary, the CTCs and CTM with a mesenchymal phenotype are associated with poor clinical outcomes and provide information on recurrence periods and therapeutic resistance, rendering them as a potential source of predictable biomarkers. Therefore, detailed analyses of multivariate targets in CTCs could improve clinical decision-making as a real-time liquid biopsy. Furthermore, molecular studies of the relationship between EMT and immune checkpoint will lead to new cancer immunotherapies based on EMT markers and immune checkpoint status. This study suggests that, for the development of specific and reliable therapeutic markers based on CTCs and CTM, further clinical prospective studies should be undertaken to increase the clinical value of CTC and CTM measurements for future personalized treatments.

Author Contributions

Conceptualization, A.O., S.T., and K.T.; writing—original draft preparation, T.O. and S.T.; data curation and table preparation, Y.F., J.W., and I.S.; review and editing, A.O. and K.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI Grant Numbers JP18K07303 and JP19K17684.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bidard, F.C.; Huguet, F.; Louvet, C.; Mineur, L.; Bouche, O.; Chibaudel, B.; Artru, P.; Desseigne, F.; Bachet, J.B.; Mathiot, C.; et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: The ancillary CirCe 07 study to the LAP 07 trial. Ann. Oncol. 2013, 24, 2057–2061. [Google Scholar] [CrossRef] [PubMed]
  2. Nakamura, S.; Yagata, H.; Ohno, S.; Yamaguchi, H.; Iwata, H.; Tsunoda, N.; Ito, Y.; Tokudome, N.; Toi, M.; Kuroi, K.; et al. Multi-center study evaluating circulating tumor cells as a surrogate for response to treatment and overall survival in metastatic breast cancer. Breast Cancer 2010, 17, 199–204. [Google Scholar] [CrossRef] [PubMed]
  3. Muinelo-Romay, L.; Vieito, M.; Abalo, A.; Nocelo, M.A.; Barón, F.; Anido, U.; Brozos, E.; Vázquez, F.; Aguín, S.; Abal, M.; et al. Evaluation of Circulating Tumor Cells and Related Events as Prognostic Factors and Surrogate Biomarkers in Advanced NSCLC Patients Receiving First-Line Systemic Treatment. Cancers 2014, 6, 153–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Scher, H.I.; Heller, G.; Molina, A.; Attard, G.; Danila, D.C.; Jia, X.; Peng, W.; Sandhu, S.K.; Olmos, D.; Riisnaes, R.; et al. Circulating tumor cell biomarker panel as an individual-level surrogate for survival in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2015, 33, 1348–1355. [Google Scholar] [CrossRef]
  5. 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]
  6. Chen, J.F.; Ho, H.; Lichterman, J.; Lu, Y.T.; Zhang, Y.; Garcia, M.A.; Chen, S.F.; Liang, A.J.; Hodara, E.; Zhau, H.E.; et al. Subclassification of prostate cancer circulating tumor cells by nuclear size reveals very small nuclear circulating tumor cells in patients with visceral metastases. Cancer 2015, 121, 3240–3251. [Google Scholar] [CrossRef] [Green Version]
  7. Danila, D.C.; Heller, G.; Gignac, G.A.; Gonzalez-Espinoza, R.; Anand, A.; Tanaka, E.; Lilja, H.; Schwartz, L.; Larson, S.; Fleisher, M.; et al. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin. Cancer Res. 2007, 13, 7053–7058. [Google Scholar] [CrossRef] [Green Version]
  8. Dorff, T.B.; Groshen, S.; Tsao-Wei, D.D.; Xiong, S.; Gross, M.E.; Vogelzang, N.; Quinn, D.I.; Pinski, J.K. A Phase II trial of a combination herbal supplement for men with biochemically recurrent prostate cancer. Prostate Cancer Prostatic Dis. 2014, 17, 359–365. [Google Scholar] [CrossRef] [Green Version]
  9. Iinuma, H.; Watanabe, T.; Mimori, K.; Adachi, M.; Hayashi, N.; Tamura, J.; Matsuda, K.; Fukushima, R.; Okinaga, K.; Sasako, M.; et al. Clinical significance of circulating tumor cells, including cancer stem-like cells, in peripheral blood for recurrence and prognosis in patients with Dukes’ stage B and C colorectal cancer. J. Clin. Oncol. 2011, 29, 1547–1555. [Google Scholar] [CrossRef]
  10. 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] [Green Version]
  11. Franken, B.; de Groot, M.R.; Mastboom, W.J.; Vermes, I.; van der Palen, J.; Tibbe, A.G.; Terstappen, L.W. Circulating tumor cells, disease recurrence and survival in newly diagnosed breast cancer. Breast Cancer Res. 2012, 14, R133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Allen-Mersh, T.G.; McCullough, T.K.; Patel, H.; Wharton, R.Q.; Glover, C.; Jonas, S.K. Role of circulating tumour cells in predicting recurrence after excision of primary colorectal carcinoma. Br. J. Surg. 2007, 94, 96–105. [Google Scholar] [CrossRef] [PubMed]
  13. Hoshimoto, S.; Shingai, T.; Morton, D.L.; Kuo, C.; Faries, M.B.; Chong, K.; Elashoff, D.; Wang, H.J.; Elashoff, R.M.; Hoon, D.S. Association between circulating tumor cells and prognosis in patients with stage III melanoma with sentinel lymph node metastasis in a phase III international multicenter trial. J. Clin. Oncol. 2012, 30, 3819–3826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tiwari, N.; Gheldof, A.; Tatari, M.; Christofori, G. EMT as the ultimate survival mechanism of cancer cells. Semin. Cancer Biol. 2012, 22, 194–207. [Google Scholar] [CrossRef] [PubMed]
  15. Castro-Giner, F.; Aceto, N. Tracking cancer progression: From circulating tumor cells to metastasis. Genome Med. 2020, 12, 31. [Google Scholar] [CrossRef] [Green Version]
  16. Helman, E.; Nguyen, M.; Karlovich, C.A.; Despain, D.; Choquette, A.K.; Spira, A.I.; Yu, H.A.; Camidge, D.R.; Harding, T.C.; Lanman, R.B.; et al. Cell-Free DNA Next-Generation Sequencing Prediction of Response and Resistance to Third-Generation EGFR Inhibitor. Clin. Lung Cancer 2018, 19, 518–530. [Google Scholar] [CrossRef] [Green Version]
  17. Chaudhuri, A.A.; Chabon, J.J.; Lovejoy, A.F.; Newman, A.M.; Stehr, H.; Azad, T.D.; Khodadoust, M.S.; Esfahani, M.S.; Liu, C.L.; Zhou, L.; et al. Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. Cancer Discov. 2017, 7, 1394–1403. [Google Scholar] [CrossRef] [Green Version]
  18. Kruger, S.; Heinemann, V.; Ross, C.; Diehl, F.; Nagel, D.; Ormanns, S.; Liebmann, S.; Prinz-Bravin, I.; Westphalen, C.B.; Haas, M.; et al. Repeated mutKRAS ctDNA measurements represent a novel and promising tool for early response prediction and therapy monitoring in advanced pancreatic cancer. Ann. Oncol. 2018, 29, 2348–2355. [Google Scholar] [CrossRef]
  19. Bronkhorst, A.J.; Ungerer, V.; Holdenrieder, S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol. Detect. Quantif. 2019, 17, 100087. [Google Scholar] [CrossRef]
  20. Lecharpentier, A.; Vielh, P.; Perez-Moreno, P.; Planchard, D.; Soria, J.C.; Farace, F. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br. J. Cancer 2011, 105, 1338–1341. [Google Scholar] [CrossRef] [Green Version]
  21. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.; Uhr, J.W.; Terstappen, L.W. 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] [Green Version]
  22. Papadaki, M.A.; Stoupis, G.; Theodoropoulos, P.A.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Circulating tumor cells with stemness and epithelial-to-mesenchymal transition features are chemoresistant and predictive of poor outcome in metastatic breast cancer. Mol. Cancer Ther. 2018. [Google Scholar] [CrossRef] [Green Version]
  23. Togo, S.; Katagiri, N.; Namba, Y.; Tulafu, M.; Nagahama, K.; Kadoya, K.; Takamochi, K.; Oh, S.; Suzuki, K.; Sakurai, F.; et al. Sensitive detection of viable circulating tumor cells using a novel conditionally telomerase-selective replicating adenovirus in non-small cell lung cancer patients. Oncotarget 2017, 8, 34884–34895. [Google Scholar] [CrossRef] [Green Version]
  24. Yoneda, K.; Tanaka, F.; Kondo, N.; Hashimoto, M.; Takuwa, T.; Matsumoto, S.; Okumura, Y.; Tsubota, N.; Sato, A.; Tsujimura, T.; et al. Circulating tumor cells (CTCs) in malignant pleural mesothelioma (MPM). Ann. Surg. Oncol. 2014, 21, S472–S480. [Google Scholar] [CrossRef] [PubMed]
  25. Stott, S.L.; Hsu, C.H.; Tsukrov, D.I.; Yu, M.; Miyamoto, D.T.; Waltman, B.A.; Rothenberg, S.M.; Shah, A.M.; Smas, M.E.; Korir, G.K.; et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl. Acad. Sci. USA 2010, 107, 18392–18397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Molnar, B.; Ladanyi, A.; Tanko, L.; Sreter, L.; Tulassay, Z. Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients. Clin. Cancer Res. 2001, 7, 4080–4085. [Google Scholar]
  28. Fidler, I.J. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur. J. Cancer 1973, 9, 223–227. [Google Scholar] [CrossRef]
  29. Cho, E.H.; Wendel, M.; Luttgen, M.; Yoshioka, C.; Marrinucci, D.; Lazar, D.; Schram, E.; Nieva, J.; Bazhenova, L.; Morgan, A.; et al. Characterization of circulating tumor cell aggregates identified in patients with epithelial tumors. Phys. Biol. 2012, 9, 016001. [Google Scholar] [CrossRef] [Green Version]
  30. Mizukoshi, K.; Okazawa, Y.; Haeno, H.; Koyama, Y.; Sulidan, K.; Komiyama, H.; Saeki, H.; Ohtsuji, N.; Ito, Y.; Kojima, Y.; et al. Metastatic seeding of human colon cancer cell clusters expressing the hybrid epithelial/mesenchymal state. Int. J. Cancer 2020, 146, 2547–2562. [Google Scholar] [CrossRef]
  31. Aceto, N.; Toner, M.; Maheswaran, S.; Haber, D.A. En Route to Metastasis: Circulating Tumor Cell Clusters and Epithelial-to-Mesenchymal Transition. Trends Cancer 2015, 1, 44–52. [Google Scholar] [CrossRef] [PubMed]
  32. Cheung, K.J.; Padmanaban, V.; Silvestri, V.; Schipper, K.; Cohen, J.D.; Fairchild, A.N.; Gorin, M.A.; Verdone, J.E.; Pienta, K.J.; Bader, J.S.; et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc. Natl. Acad. Sci. USA 2016, 113, E854–E863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; 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] [Green Version]
  34. Aceto, N. Bring along your friends: Homotypic and heterotypic circulating tumor cell clustering to accelerate metastasis. Biomed. J. 2020, 43, 18–23. [Google Scholar] [CrossRef]
  35. Riethdorf, S.; Muller, V.; Loibl, S.; Nekljudova, V.; Weber, K.; Huober, J.; Fehm, T.; Schrader, I.; Hilfrich, J.; Holms, F.; et al. Prognostic Impact of Circulating Tumor Cells for Breast Cancer Patients Treated in the Neoadjuvant "Geparquattro" Trial. Clin. Cancer Res. 2017, 23, 5384–5393. [Google Scholar] [CrossRef] [Green Version]
  36. Banyard, J.; Bielenberg, D.R. The role of EMT and MET in cancer dissemination. Connect. Tissue Res. 2015, 56, 403–413. [Google Scholar] [CrossRef] [Green Version]
  37. Smit, M.A.; Geiger, T.R.; Song, J.Y.; Gitelman, I.; Peeper, D.S. A Twist-Snail axis critical for TrkB-induced epithelial-mesenchymal transition-like transformation, anoikis resistance, and metastasis. Mol. Cell Biol. 2009, 29, 3722–3737. [Google Scholar] [CrossRef] [Green Version]
  38. Smit, M.A.; Peeper, D.S. Zeb1 is required for TrkB-induced epithelial-mesenchymal transition, anoikis resistance and metastasis. Oncogene 2011, 30, 3735–3744. [Google Scholar] [CrossRef]
  39. Nakamura, M.; Tokura, Y. Epithelial-mesenchymal transition in the skin. J. Dermatol. Sci. 2011, 61, 7–13. [Google Scholar] [CrossRef]
  40. Lin, K.; Baritaki, S.; Militello, L.; Malaponte, G.; Bevelacqua, Y.; Bonavida, B. The Role of B-RAF Mutations in Melanoma and the Induction of EMT via Dysregulation of the NF-kappaB/Snail/RKIP/PTEN Circuit. Genes Cancer 2010, 1, 409–420. [Google Scholar] [CrossRef]
  41. Kaufhold, S.; Bonavida, B. Central role of Snail1 in the regulation of EMT and resistance in cancer: A target for therapeutic intervention. J. Exp. Clin. Cancer Res. 2014, 33, 62. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed] [Green Version]
  43. Satelli, A.; Li, S. Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol. Life Sci. 2011, 68, 3033–3046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mitra, A.; Satelli, A.; Xia, X.; Cutrera, J.; Mishra, L.; Li, S. Cell-surface Vimentin: A mislocalized protein for isolating csVimentin(+) CD133(-) novel stem-like hepatocellular carcinoma cells expressing EMT markers. Int. J. Cancer 2015, 137, 491–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Batth, I.S.; Dao, L.; Satelli, A.; Mitra, A.; Yi, S.; Noh, H.; Li, H.; Brownlee, Z.; Zhou, S.; Bond, J.; et al. Cell surface vimentin-positive circulating tumor cell-based relapse prediction in a long-term longitudinal study of postremission neuroblastoma patients. Int. J. Cancer 2020. [Google Scholar] [CrossRef]
  46. Liu, M.; Wang, R.; Sun, X.; Liu, Y.; Wang, Z.; Yan, J.; Kong, X.; Liang, S.; Liu, Q.; Zhao, T.; et al. Prognostic significance of PD-L1 expression on cell-surface vimentin-positive circulating tumor cells in gastric cancer patients. Mol. Oncol. 2020, 14, 865–881. [Google Scholar] [CrossRef]
  47. Satelli, A.; Batth, I.; Brownlee, Z.; Mitra, A.; Zhou, S.; Noh, H.; Rojas, C.R.; Li, H.; Meng, Q.H.; Li, S. EMT circulating tumor cells detected by cell-surface vimentin are associated with prostate cancer progression. Oncotarget 2017, 8, 49329–49337. [Google Scholar] [CrossRef] [Green Version]
  48. Chaffer, C.L.; Brennan, J.P.; Slavin, J.L.; Blick, T.; Thompson, E.W.; Williams, E.D. Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: Role of fibroblast growth factor receptor-2. Cancer Res. 2006, 66, 11271–11278. [Google Scholar] [CrossRef] [Green Version]
  49. Chao, Y.L.; Shepard, C.R.; Wells, A. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition. Mol. Cancer 2010, 9, 179. [Google Scholar] [CrossRef] [Green Version]
  50. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
  51. Horimoto, Y.; Tokuda, E.; Murakami, F.; Uomori, T.; Himuro, T.; Nakai, K.; Orihata, G.; Iijima, K.; Togo, S.; Shimizu, H.; et al. Analysis of circulating tumour cell and the epithelial mesenchymal transition (EMT) status during eribulin-based treatment in 22 patients with metastatic breast cancer: A pilot study. J. Transl. Med. 2018, 16, 287. [Google Scholar] [CrossRef] [PubMed]
  52. Li, T.T.; Liu, H.; Li, F.P.; Hu, Y.F.; Mou, T.Y.; Lin, T.; Yu, J.; Zheng, L.; Li, G.X. Evaluation of epithelial-mesenchymal transitioned circulating tumor cells in patients with resectable gastric cancer: Relevance to therapy response. World J. Gastroenterol. 2015, 21, 13259–13267. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. Agnoletto, C.; Corra, F.; Minotti, L.; Baldassari, F.; Crudele, F.; Cook, W.J.J.; Di Leva, G.; d’Adamo, A.P.; Gasparini, P.; Volinia, S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers 2019, 11, 483. [Google Scholar] [CrossRef] [Green Version]
  55. Mego, M.; Gao, H.; Lee, B.N.; Cohen, E.N.; Tin, S.; Giordano, A.; Wu, Q.; Liu, P.; Nieto, Y.; Champlin, R.E.; et al. Prognostic Value of EMT-Circulating Tumor Cells in Metastatic Breast Cancer Patients Undergoing High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation. J. Cancer 2012, 3, 369–380. [Google Scholar] [CrossRef]
  56. Gradilone, A.; Raimondi, C.; Nicolazzo, C.; Petracca, A.; Gandini, O.; Vincenzi, B.; Naso, G.; Agliano, A.M.; Cortesi, E.; Gazzaniga, P. Circulating tumour cells lacking cytokeratin in breast cancer: The importance of being mesenchymal. J. Cell Mol. Med. 2011, 15, 1066–1070. [Google Scholar] [CrossRef] [Green Version]
  57. Kallergi, G.; Papadaki, M.A.; Politaki, E.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 2011, 13, R59. [Google Scholar] [CrossRef] [Green Version]
  58. Alonso-Alconada, L.; Muinelo-Romay, L.; Madissoo, K.; Diaz-Lopez, A.; Krakstad, C.; Trovik, J.; Wik, E.; Hapangama, D.; Coenegrachts, L.; Cano, A.; et al. Molecular profiling of circulating tumor cells links plasticity to the metastatic process in endometrial cancer. Mol. Cancer 2014, 13, 223. [Google Scholar] [CrossRef] [Green Version]
  59. Xu, L.; Mao, X.; Guo, T.; Chan, P.Y.; Shaw, G.; Hines, J.; Stankiewicz, E.; Wang, Y.; Oliver, R.T.D.; Ahmad, A.S.; et al. The Novel Association of Circulating Tumor Cells and Circulating Megakaryocytes with Prostate Cancer Prognosis. Clin. Cancer Res. 2017, 23, 5112–5122. [Google Scholar] [CrossRef] [Green Version]
  60. Kallergi, G.; Agelaki, S.; Kalykaki, A.; Stournaras, C.; Mavroudis, D.; Georgoulias, V. Phosphorylated EGFR and PI3K/Akt signaling kinases are expressed in circulating tumor cells of breast cancer patients. Breast Cancer Res. 2008, 10, R80. [Google Scholar] [CrossRef] [Green Version]
  61. Pal, S.K.; He, M.; Wilson, T.; Liu, X.; Zhang, K.; Carmichael, C.; Torres, A.; Hernandez, S.; Lau, C.; Agarwal, N.; et al. Detection and phenotyping of circulating tumor cells in high-risk localized prostate cancer. Clin. Genitourin Cancer 2015, 13, 130–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Polioudaki, H.; Agelaki, S.; Chiotaki, R.; Politaki, E.; Mavroudis, D.; Matikas, A.; Georgoulias, V.; Theodoropoulos, P.A. Variable expression levels of keratin and vimentin reveal differential EMT status of circulating tumor cells and correlation with clinical characteristics and outcome of patients with metastatic breast cancer. BMC Cancer 2015, 15, 399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef] [PubMed]
  64. Kulemann, B.; Rosch, S.; Seifert, S.; Timme, S.; Bronsert, P.; Seifert, G.; Martini, V.; Kuvendjiska, J.; Glatz, T.; Hussung, S.; et al. Pancreatic cancer: Circulating Tumor Cells and Primary Tumors show Heterogeneous KRAS Mutations. Sci. Rep. 2017, 7, 4510. [Google Scholar] [CrossRef] [Green Version]
  65. Kulemann, B.; Pitman, M.B.; Liss, A.S.; Valsangkar, N.; Fernández-Del Castillo, C.; Lillemoe, K.D.; Hoeppner, J.; Mino-Kenudson, M.; Warshaw, A.L.; Thayer, S.P. Circulating tumor cells found in patients with localized and advanced pancreatic cancer. Pancreas 2015, 44, 547–550. [Google Scholar] [CrossRef] [Green Version]
  66. Kulemann, B.; Liss, A.S.; Warshaw, A.L.; Seifert, S.; Bronsert, P.; Glatz, T.; Pitman, M.B.; Hoeppner, J. KRAS mutations in pancreatic circulating tumor cells: A pilot study. Tumour Biol. 2016, 37, 7547–7554. [Google Scholar] [CrossRef]
  67. Li, W.; Kang, Y. Probing the Fifty Shades of EMT in Metastasis. Trends Cancer 2016, 2, 65–67. [Google Scholar] [CrossRef] [Green Version]
  68. Lindsay, C.R.; Faugeroux, V.; Michiels, S.; Pailler, E.; Facchinetti, F.; Ou, D.; Bluthgen, M.V.; Pannet, C.; Ngo-Camus, M.; Bescher, G.; et al. A prospective examination of circulating tumor cell profiles in non-small-cell lung cancer molecular subgroups. Ann. Oncol. 2017, 28, 1523–1531. [Google Scholar] [CrossRef]
  69. Hu, B.; Tian, X.; Li, Y.; Liu, Y.; Yang, T.; Han, Z.; An, J.; Kong, L.; Li, Y. Epithelial-mesenchymal transition may be involved in the immune evasion of circulating gastric tumor cells via downregulation of ULBP1. Cancer Med. 2020, 9, 2686–2697. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, Y.; Liu, Y.; Zhang, L.; Tong, L.; Gao, Y.; Hu, F.; Lin, P.P.; Li, B.; Zhang, T. Vimentin expression in circulating tumor cells (CTCs) associated with liver metastases predicts poor progression-free survival in patients with advanced lung cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 2911–2920. [Google Scholar] [CrossRef] [Green Version]
  71. Szczepanik, A.; Sierzega, M.; Drabik, G.; Pituch-Noworolska, A.; Kolodziejczyk, P.; Zembala, M. CD44(+) cytokeratin-positive tumor cells in blood and bone marrow are associated with poor prognosis of patients with gastric cancer. Gastric Cancer 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Manjunath, Y.; Upparahalli, S.V.; Avella, D.M.; Deroche, C.B.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Smith, C.J.; Li, G.; Kaifi, J.T. PD-L1 Expression with Epithelial Mesenchymal Transition of Circulating Tumor Cells Is Associated with Poor Survival in Curatively Resected Non-Small Cell Lung Cancer. Cancers 2019, 11, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhang, X.; Wei, L.; Li, J.; Zheng, J.; Zhang, S.; Zhou, J. Epithelialmesenchymal transition phenotype of circulating tumor cells is associated with distant metastasis in patients with NSCLC. Mol. Med. Rep. 2019, 19, 601–608. [Google Scholar] [CrossRef] [Green Version]
  74. de Wit, S.; Manicone, M.; Rossi, E.; Lampignano, R.; Yang, L.; Zill, B.; Rengel-Puertas, A.; Ouhlen, M.; Crespo, M.; Berghuis, A.M.S.; et al. EpCAM(high) and EpCAM(low) circulating tumor cells in metastatic prostate and breast cancer patients. Oncotarget 2018, 9, 35705–35716. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, W.; Wan, L.; Wu, S.; Yang, J.; Zhou, Y.; Liu, F.; Wu, Z.; Cheng, Y. Mesenchymal marker and LGR5 expression levels in circulating tumor cells correlate with colorectal cancer prognosis. Cell Oncol. 2018, 41, 495–504. [Google Scholar] [CrossRef]
  76. Han, D.; Chen, K.; Che, J.; Hang, J.; Li, H. Detection of Epithelial-Mesenchymal Transition Status of Circulating Tumor Cells in Patients with Esophageal Squamous Carcinoma. Biomed. Res. Int. 2018, 2018, 7610154. [Google Scholar] [CrossRef] [Green Version]
  77. Kallergi, G.; Aggouraki, D.; Zacharopoulou, N.; Stournaras, C.; Georgoulias, V.; Martin, S.S. Evaluation of alpha-tubulin, detyrosinated alpha-tubulin, and vimentin in CTCs: Identification of the interaction between CTCs and blood cells through cytoskeletal elements. Breast Cancer Res. 2018, 20, 67. [Google Scholar] [CrossRef]
  78. Qi, L.N.; Xiang, B.D.; Wu, F.X.; Ye, J.Z.; Zhong, J.H.; Wang, Y.Y.; Chen, Y.Y.; Chen, Z.S.; Ma, L.; Chen, J.; et al. Circulating Tumor Cells Undergoing EMT Provide a Metric for Diagnosis and Prognosis of Patients with Hepatocellular Carcinoma. Cancer Res. 2018, 78, 4731–4744. [Google Scholar] [CrossRef] [Green Version]
  79. Zhao, R.; Cai, Z.; Li, S.; Cheng, Y.; Gao, H.; Liu, F.; Wu, S.; Liu, S.; Dong, Y.; Zheng, L.; et al. Expression and clinical relevance of epithelial and mesenchymal markers in circulating tumor cells from colorectal cancer. Oncotarget 2017, 8, 9293–9302. [Google Scholar] [CrossRef] [Green Version]
  80. Wu, F.; Zhu, J.; Mao, Y.; Li, X.; Hu, B.; Zhang, D. Associations between the Epithelial-Mesenchymal Transition Phenotypes of Circulating Tumor Cells and the Clinicopathological Features of Patients with Colorectal Cancer. Dis. Markers 2017, 2017, 9474532. [Google Scholar] [CrossRef] [Green Version]
  81. Wang, Z.; Luo, L.; Cheng, Y.; He, G.; Peng, B.; Gao, Y.; Jiang, Z.S.; Pan, M. Correlation Between Postoperative Early Recurrence of Hepatocellular Carcinoma and Mesenchymal Circulating Tumor Cells in Peripheral Blood. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2018, 22, 633–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Chebouti, I.; Kasimir-Bauer, S.; Buderath, P.; Wimberger, P.; Hauch, S.; Kimmig, R.; Kuhlmann, J.D. EMT-like circulating tumor cells in ovarian cancer patients are enriched by platinum-based chemotherapy. Oncotarget 2017, 8, 48820–48831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Guan, X.; Ma, F.; Liu, S.; Wu, S.; Xiao, R.; Yuan, L.; Sun, X.; Yi, Z.; Yang, H.; Xu, B. Analysis of the hormone receptor status of circulating tumor cell subpopulations based on epithelial-mesenchymal transition: A proof-of-principle study on the heterogeneity of circulating tumor cells. Oncotarget 2016, 7, 65993–66002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lindsay, C.R.; Le Moulec, S.; Billiot, F.; Loriot, Y.; Ngo-Camus, M.; Vielh, P.; Fizazi, K.; Massard, C.; Farace, F. Vimentin and Ki67 expression in circulating tumour cells derived from castrate-resistant prostate cancer. BMC Cancer 2016, 16, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Giordano, A.; Gao, H.; Anfossi, S.; Cohen, E.; Mego, M.; Lee, B.N.; Tin, S.; De Laurentiis, M.; Parker, C.A.; Alvarez, R.H.; et al. Epithelial-mesenchymal transition and stem cell markers in patients with HER2-positive metastatic breast cancer. Mol. Cancer Ther. 2012, 11, 2526–2534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lustberg, M.; Jatana, K.R.; Zborowski, M.; Chalmers, J.J. Emerging technologies for CTC detection based on depletion of normal cells. Recent Results Cancer Res. 2012, 195, 97–110. [Google Scholar] [CrossRef] [Green Version]
  87. Watanabe, M.; Kenmotsu, H.; Ko, R.; Wakuda, K.; Ono, A.; Imai, H.; Taira, T.; Naito, T.; Murakami, H.; Abe, M.; et al. Isolation and molecular analysis of circulating tumor cells from lung cancer patients using a microfluidic chip type cell sorter. Cancer Sci. 2018, 109, 2539–2548. [Google Scholar] [CrossRef] [Green Version]
  88. Morimoto, A.; Mogami, T.; Watanabe, M.; Iijima, K.; Akiyama, Y.; Katayama, K.; Futami, T.; Yamamoto, N.; Sawada, T.; Koizumi, F.; et al. High-Density Dielectrophoretic Microwell Array for Detection, Capture, and Single-Cell Analysis of Rare Tumor Cells in Peripheral Blood. PLoS ONE 2015, 10, e0130418. [Google Scholar] [CrossRef]
  89. Li, X.; Li, Y.; Shao, W.; Li, Z.; Zhao, R.; Ye, Z. Strategies for enrichment of circulating tumor cells. Transl. Cancer Res. 2020, 9, 2012–2025. [Google Scholar] [CrossRef]
  90. Zhang, L.; Riethdorf, S.; Wu, G.; Wang, T.; Yang, K.; Peng, G.; Liu, J.; Pantel, K. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin. Cancer Res. 2012, 18, 5701–5710. [Google Scholar] [CrossRef] [Green Version]
  91. Mego, M.; Cierna, Z.; Janega, P.; Karaba, M.; Minarik, G.; Benca, J.; Sedlácková, T.; Sieberova, G.; Gronesova, P.; Manasova, D.; et al. Relationship between circulating tumor cells and epithelial to mesenchymal transition in early breast cancer. BMC Cancer 2015, 15, 533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Papadaki, M.A.; Kallergi, G.; Zafeiriou, Z.; Manouras, L.; Theodoropoulos, P.A.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Co-expression of putative stemness and epithelial-to-mesenchymal transition markers on single circulating tumour cells from patients with early and metastatic breast cancer. BMC Cancer 2014, 14, 651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Barriere, G.; Riouallon, A.; Renaudie, J.; Tartary, M.; Rigaud, M. Mesenchymal and stemness circulating tumor cells in early breast cancer diagnosis. BMC Cancer 2012, 12, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Jiang, W.; Pang, X.G.; Wang, Q.; Shen, Y.X.; Chen, X.K.; Xi, J.J. Prognostic role of Twist, Slug, and Foxc2 expression in stage I non-small-cell lung cancer after curative resection. Clin. Lung Cancer 2012, 13, 280–287. [Google Scholar] [CrossRef]
  95. Hung, J.J.; Yang, M.H.; Hsu, H.S.; Hsu, W.H.; Liu, J.S.; Wu, K.J. Prognostic significance of hypoxia-inducible factor-1alpha, TWIST1 and Snail expression in resectable non-small cell lung cancer. Thorax 2009, 64, 1082–1089. [Google Scholar] [CrossRef] [Green Version]
  96. Osaki, T.; Oyama, T.; Gu, C.D.; Yamashita, T.; So, T.; Takenoyama, M.; Sugio, K.; Yasumoto, K. Prognostic impact of micrometastatic tumor cells in the lymph nodes and bone marrow of patients with completely resected stage I non-small-cell lung cancer. J. Clin. Oncol. 2002, 20, 2930–2936. [Google Scholar] [CrossRef]
  97. Lucci, A.; Hall, C.S.; Lodhi, A.K.; Bhattacharyya, A.; Anderson, A.E.; Xiao, L.; Bedrosian, I.; Kuerer, H.M.; Krishnamurthy, S. Circulating tumour cells in non-metastatic breast cancer: A prospective study. Lancet Oncol. 2012, 13, 688–695. [Google Scholar] [CrossRef]
  98. Rack, B.; Schindlbeck, C.; Juckstock, J.; Andergassen, U.; Hepp, P.; Zwingers, T.; Friedl, T.W.; Lorenz, R.; Tesch, H.; Fasching, P.A.; et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef]
  99. Sawabata, N.; Okumura, M.; Utsumi, T.; Inoue, M.; Shiono, H.; Minami, M.; Nishida, T.; Sawa, Y. Circulating tumor cells in peripheral blood caused by surgical manipulation of non-small-cell lung cancer: Pilot study using an immunocytology method. Gen. Thorac. Cardiovasc. Surg. 2007, 55, 189–192. [Google Scholar] [CrossRef]
  100. Ilie, M.; Hofman, V.; Long-Mira, E.; Selva, E.; Vignaud, J.M.; Padovani, B.; Mouroux, J.; Marquette, C.H.; Hofman, P. "Sentinel" circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS ONE 2014, 9, e111597. [Google Scholar] [CrossRef] [Green Version]
  101. Sun, Y.; Wu, G.; Cheng, K.S.; Chen, A.; Neoh, K.H.; Chen, S.; Tang, Z.; Lee, P.F.; Dai, M.; Han, R.P.S. CTC phenotyping for a preoperative assessment of tumor metastasis and overall survival of pancreatic ductal adenocarcinoma patients. EBioMedicine 2019, 46, 133–149. [Google Scholar] [CrossRef] [PubMed]
  102. Markiewicz, A.; Ksiazkiewicz, M.; Welnicka-Jaskiewicz, M.; Seroczynska, B.; Skokowski, J.; Szade, J.; Zaczek, A.J. Mesenchymal phenotype of CTC-enriched blood fraction and lymph node metastasis formation potential. PLoS ONE 2014, 9, e93901. [Google Scholar] [CrossRef]
  103. Nel, I.; Baba, H.A.; Ertle, J.; Weber, F.; Sitek, B.; Eisenacher, M.; Meyer, H.E.; Schlaak, J.F.; Hoffmann, A.C. Individual profiling of circulating tumor cell composition and therapeutic outcome in patients with hepatocellular carcinoma. Transl. Oncol. 2013, 6, 420–428. [Google Scholar] [CrossRef] [PubMed]
  104. 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] [PubMed] [Green Version]
  105. Yin, L.C.; Luo, Z.C.; Gao, Y.X.; Li, Y.; Peng, Q.; Gao, Y. Twist Expression in Circulating Hepatocellular Carcinoma Cells Predicts Metastasis and Prognoses. Biomed. Res. Int. 2018, 2018, 3789613. [Google Scholar] [CrossRef]
  106. Tsuji, T.; Ibaragi, S.; Hu, G.F. Epithelial-mesenchymal transition and cell cooperativity in metastasis. Cancer Res. 2009, 69, 7135–7139. [Google Scholar] [CrossRef] [Green Version]
  107. Chui, M.H. Insights into cancer metastasis from a clinicopathologic perspective: Epithelial-Mesenchymal Transition is not a necessary step. Int. J. Cancer 2013, 132, 1487–1495. [Google Scholar] [CrossRef]
  108. Wu, G.; Zhu, R.; Li, Y.; Zhao, Y.; Dai, M. Prognostic significance of circulating tumor microemboli in patients with pancreatic ductal adenocarcinoma. Oncol. Lett. 2018, 15, 7376–7382. [Google Scholar] [CrossRef] [Green Version]
  109. Larsson, A.M.; Jansson, S.; Bendahl, P.O.; Levin Tykjaer Jorgensen, C.; Loman, N.; Graffman, C.; Lundgren, L.; Aaltonen, K.; Ryden, 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]
  110. Wang, C.; Mu, Z.; Chervoneva, I.; Austin, L.; Ye, Z.; Rossi, G.; Palazzo, J.P.; Sun, C.; Abu-Khalaf, M.; Myers, R.E.; et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res. Treat. 2017, 161, 83–94. [Google Scholar] [CrossRef]
  111. Costa, C.; Muinelo-Romay, L.; Cebey-López, V.; Pereira-Veiga, T.; Martínez-Pena, I.; Abreu, M.; Abalo, A.; Lago-Lestón, R.M.; Abuín, C.; Palacios, P.; et al. Analysis of a Real-World Cohort of Metastatic Breast Cancer Patients Shows Circulating Tumor Cell Clusters (CTC-clusters) as Predictors of Patient Outcomes. Cancers 2020, 12, 1111. [Google Scholar] [CrossRef] [PubMed]
  112. Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef] [PubMed]
  114. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
  115. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
  116. Nel, I.; Jehn, U.; Gauler, T.; Hoffmann, A.C. Individual profiling of circulating tumor cell composition in patients with non-small cell lung cancer receiving platinum based treatment. Transl. Lung Cancer Res. 2014, 3, 100–106. [Google Scholar] [CrossRef]
  117. Armstrong, A.J.; Marengo, M.S.; Oltean, S.; Kemeny, G.; Bitting, R.L.; Turnbull, J.D.; Herold, C.I.; Marcom, P.K.; George, D.J.; Garcia-Blanco, M.A. Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol. Cancer Res. 2011, 9, 997–1007. [Google Scholar] [CrossRef] [Green Version]
  118. Kasimir-Bauer, S.; Hoffmann, O.; Wallwiener, D.; Kimmig, R.; Fehm, T. Expression of stem cell and epithelial-mesenchymal transition markers in primary breast cancer patients with circulating tumor cells. Breast Cancer Res. 2012, 14, R15. [Google Scholar] [CrossRef] [Green Version]
  119. Douville, J.; Beaulieu, R.; Balicki, D. ALDH1 as a functional marker of cancer stem and progenitor cells. Stem Cells Dev. 2009, 18, 17–25. [Google Scholar] [CrossRef]
  120. Hanssen, A.; Wagner, J.; Gorges, T.M.; Taenzer, A.; Uzunoglu, F.G.; Driemel, C.; Stoecklein, N.H.; Knoefel, W.T.; Angenendt, S.; Hauch, S.; et al. Characterization of different CTC subpopulations in non-small cell lung cancer. Sci. Rep. 2016, 6, 28010. [Google Scholar] [CrossRef]
  121. Raimondi, C.; Gradilone, A.; Naso, G.; Vincenzi, B.; Petracca, A.; Nicolazzo, C.; Palazzo, A.; Saltarelli, R.; Spremberg, F.; Cortesi, E.; et al. Epithelial-mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients. Breast Cancer Res. Treat. 2011, 130, 449–455. [Google Scholar] [CrossRef] [PubMed]
  122. Aktas, B.; Tewes, M.; Fehm, T.; Hauch, S.; Kimmig, R.; Kasimir-Bauer, S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009, 11, R46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Theodoropoulos, P.A.; Polioudaki, H.; Agelaki, S.; Kallergi, G.; Saridaki, Z.; Mavroudis, D.; Georgoulias, V. Circulating tumor cells with a putative stem cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett. 2010, 288, 99–106. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, T.; Xu, H.; Huang, M.; Ma, W.; Saxena, D.; Lustig, R.A.; Alonso-Basanta, M.; Zhang, Z.; DM, O.R.; Zhang, L.; et al. Circulating glioma cells exhibit stem cell-like properties. Cancer Res. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Denis, J.A.; Patroni, A.; Guillerm, E.; Pepin, D.; Benali-Furet, N.; Wechsler, J.; Manceau, G.; Bernard, M.; Coulet, F.; Larsen, A.K.; et al. Droplet digital PCR of circulating tumor cells from colorectal cancer patients can predict KRAS mutations before surgery. Mol. Oncol. 2016, 10, 1221–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Buim, M.E.; Fanelli, M.F.; Souza, V.S.; Romero, J.; Abdallah, E.A.; Mello, C.A.; Alves, V.; Ocea, L.M.; Mingues, N.B.; Barbosa, P.N.; et al. Detection of KRAS mutations in circulating tumor cells from patients with metastatic colorectal cancer. Cancer Biol. Ther. 2015, 16, 1289–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Kondo, Y.; Hayashi, K.; Kawakami, K.; Miwa, Y.; Hayashi, H.; Yamamoto, M. KRAS mutation analysis of single circulating tumor cells from patients with metastatic colorectal cancer. BMC Cancer 2017, 17, 311. [Google Scholar] [CrossRef] [Green Version]
  128. 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]
  129. Mostert, B.; Jiang, Y.; Sieuwerts, A.M.; Wang, H.; Bolt-de Vries, J.; Biermann, K.; Kraan, J.; Lalmahomed, Z.; van Galen, A.; de Weerd, V.; et al. KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. Int. J. Cancer 2013, 133, 130–141. [Google Scholar] [CrossRef]
  130. Kalikaki, A.; Politaki, H.; Souglakos, J.; Apostolaki, S.; Papadimitraki, E.; Georgoulia, N.; Tzardi, M.; Mavroudis, D.; Georgoulias, V.; Voutsina, A. KRAS genotypic changes of circulating tumor cells during treatment of patients with metastatic colorectal cancer. PLoS ONE 2014, 9, e104902. [Google Scholar] [CrossRef]
  131. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef] [PubMed]
  132. Yu, D. Mechanisms of ErbB2-mediated paclitaxel resistance and trastuzumab-mediated paclitaxel sensitization in ErbB2-overexpressing breast cancers. Semin. Oncol. 2001, 28, 12–17. [Google Scholar] [CrossRef]
  133. Pinzani, P.; Salvadori, B.; Simi, L.; Bianchi, S.; Distante, V.; Cataliotti, L.; Pazzagli, M.; Orlando, C. Isolation by size of epithelial tumor cells in peripheral blood of patients with breast cancer: Correlation with real-time reverse transcriptase-polymerase chain reaction results and feasibility of molecular analysis by laser microdissection. Hum. Pathol. 2006, 37, 711–718. [Google Scholar] [CrossRef] [PubMed]
  134. Fehm, T.; Müller, V.; Aktas, B.; Janni, W.; Schneeweiss, A.; Stickeler, E.; Lattrich, C.; Löhberg, C.R.; Solomayer, E.; Rack, B.; et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: A prospective, multicenter trial. Breast Cancer Res. Treat. 2010, 124, 403–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zidan, J.; Dashkovsky, I.; Stayerman, C.; Basher, W.; Cozacov, C.; Hadary, A. Comparison of HER-2 overexpression in primary breast cancer and metastatic sites and its effect on biological targeting therapy of metastatic disease. Br. J. Cancer 2005, 93, 552–556. [Google Scholar] [CrossRef] [Green Version]
  136. Aitken, S.J.; Thomas, J.S.; Langdon, S.P.; Harrison, D.J.; Faratian, D. Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases. Ann. Oncol. 2010, 21, 1254–1261. [Google Scholar] [CrossRef]
  137. Flores, L.M.; Kindelberger, D.W.; Ligon, A.H.; Capelletti, M.; Fiorentino, M.; Loda, M.; Cibas, E.S.; Jänne, P.A.; Krop, I.E. Improving the yield of circulating tumour cells facilitates molecular characterisation and recognition of discordant HER2 amplification in breast cancer. Br. J. Cancer 2010, 102, 1495–1502. [Google Scholar] [CrossRef] [Green Version]
  138. Beije, N.; Onstenk, W.; Kraan, J.; Sieuwerts, A.M.; Hamberg, P.; Dirix, L.Y.; Brouwer, A.; de Jongh, F.E.; Jager, A.; Seynaeve, C.M.; et al. Prognostic Impact of HER2 and ER Status of Circulating Tumor Cells in Metastatic Breast Cancer Patients with a HER2-Negative Primary Tumor. Neoplasia 2016, 18, 647–653. [Google Scholar] [CrossRef] [Green Version]
  139. Jaeger, B.A.S.; Neugebauer, J.; Andergassen, U.; Melcher, C.; Schochter, F.; Mouarrawy, D.; Ziemendorff, G.; Clemens, M.; Abel, E.V.; Heinrich, G.; et al. The HER2 phenotype of circulating tumor cells in HER2-positive early breast cancer: A translational research project of a prospective randomized phase III trial. PLoS ONE 2017, 12, e0173593. [Google Scholar] [CrossRef] [Green Version]
  140. Tellez-Gabriel, M.; Heymann, M.F.; Heymann, D. Circulating Tumor Cells as a Tool for Assessing Tumor Heterogeneity. Theranostics 2019, 9, 4580–4594. [Google Scholar] [CrossRef]
  141. Mohamed Suhaimi, N.A.; Foong, Y.M.; Lee, D.Y.; Phyo, W.M.; Cima, I.; Lee, E.X.; Goh, W.L.; Lim, W.Y.; Chia, K.S.; Kong, S.L.; et al. Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients. Mol. Oncol. 2015, 9, 850–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Bingham, C.; Fernandez, S.V.; Fittipaldi, P.; Dempsey, P.W.; Ruth, K.J.; Cristofanilli, M.; Katherine Alpaugh, R. Mutational studies on single circulating tumor cells isolated from the blood of inflammatory breast cancer patients. Breast Cancer Res. Treat. 2017, 163, 219–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wang, Q.; Zhao, L.; Han, L.; Tuo, X.; Ma, S.; Wang, Y.; Feng, X.; Liang, D.; Sun, C.; Wang, Q.; et al. The Discordance of Gene Mutations between Circulating Tumor Cells and Primary/Metastatic Tumor. Mol. Ther. Oncolytics 2019, 15, 21–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Meng, S.; Tripathy, D.; Shete, S.; Ashfaq, R.; Haley, B.; Perkins, S.; Beitsch, P.; Khan, A.; Euhus, D.; Osborne, C.; et al. HER-2 gene amplification can be acquired as breast cancer progresses. Proc. Natl. Acad. Sci. USA 2004, 101, 9393–9398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wang, C.H.; Chang, C.J.; Yeh, K.Y.; Chang, P.H.; Huang, J.S. The Prognostic Value of HER2-Positive Circulating Tumor Cells in Breast Cancer Patients: A Systematic Review and Meta-Analysis. Clin. Breast Cancer 2017, 17, 341–349. [Google Scholar] [CrossRef] [PubMed]
  146. Apostolaki, S.; Perraki, M.; Kallergi, G.; Kafousi, M.; Papadopoulos, S.; Kotsakis, A.; Pallis, A.; Xenidis, N.; Kalmanti, L.; Kalbakis, K.; et al. Detection of occult HER2 mRNA-positive tumor cells in the peripheral blood of patients with operable breast cancer: Evaluation of their prognostic relevance. Breast Cancer Res. Treat. 2009, 117, 525–534. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, S.; Li, L.; Wang, T.; Bian, L.; Hu, H.; Xu, C.; Liu, B.; Liu, Y.; Cristofanilli, M.; Jiang, Z. Real-time HER2 status detected on circulating tumor cells predicts different outcomes of anti-HER2 therapy in histologically HER2-positive metastatic breast cancer patients. BMC Cancer 2016, 16, 526. [Google Scholar] [CrossRef] [Green Version]
  148. Ignatiadis, M.; Litiere, S.; Rothe, F.; Riethdorf, S.; Proudhon, C.; Fehm, T.; Aalders, K.; Forstbauer, H.; Fasching, P.A.; Brain, E.; et al. Trastuzumab versus observation for HER2 nonamplified early breast cancer with circulating tumor cells (EORTC 90091-10093, BIG 1-12, Treat CTC): A randomized phase II trial. Ann. Oncol. 2018, 29, 1777–1783. [Google Scholar] [CrossRef]
  149. Kwan, T.T.; Bardia, A.; Spring, L.M.; Giobbie-Hurder, A.; Kalinich, M.; Dubash, T.; Sundaresan, T.; Hong, X.; LiCausi, J.A.; Ho, U.; et al. A Digital RNA Signature of Circulating Tumor Cells Predicting Early Therapeutic Response in Localized and Metastatic Breast Cancer. Cancer Discov. 2018, 8, 1286–1299. [Google Scholar] [CrossRef] [Green Version]
  150. Pal, S.K.; He, M.; Chen, L.; Yang, L.; Pillai, R.; Twardowski, P.; Hsu, J.; Kortylewski, M.; Jones, J.O. Synaptophysin expression on circulating tumor cells in patients with castration resistant prostate cancer undergoing treatment with abiraterone acetate or enzalutamide. Urol. Oncol. 2018, 36, 162.e1–162.e6. [Google Scholar] [CrossRef]
  151. Rugo, H.S.; Cortes, J.; Awada, A.; O’Shaughnessy, J.; Twelves, C.; Im, S.A.; Hannah, A.; Lu, L.; Sy, S.; Caygill, K.; et al. Change in Topoisomerase 1-Positive Circulating Tumor Cells Affects Overall Survival in Patients with Advanced Breast Cancer after Treatment with Etirinotecan Pegol. Clin. Cancer Res. 2018, 24, 3348–3357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Messaritakis, I.; Nikolaou, M.; Politaki, E.; Koinis, F.; Lagoudaki, E.; Koutsopoulos, A.; Georgoulia, N.; Georgoulias, V.; Kotsakis, A. Bcl-2 expression in circulating tumor cells (CTCs) of patients with small cell lung cancer (SCLC) receiving front-line treatment. Lung Cancer 2018, 124, 270–278. [Google Scholar] [CrossRef] [PubMed]
  153. Sundaresan, T.K.; Sequist, L.V.; Heymach, J.V.; Riely, G.J.; Janne, P.A.; Koch, W.H.; Sullivan, J.P.; Fox, D.B.; Maher, R.; Muzikansky, A.; et al. Detection of T790M, the Acquired Resistance EGFR Mutation, by Tumor Biopsy versus Noninvasive Blood-Based Analyses. Clin. Cancer Res. 2016, 22, 1103–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Marchetti, A.; Del Grammastro, M.; Felicioni, L.; Malatesta, S.; Filice, G.; Centi, I.; De Pas, T.; Santoro, A.; Chella, A.; Brandes, A.A.; et al. Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next generation sequencing: Toward a real-time liquid biopsy for treatment. PLoS ONE 2014, 9, e103883. [Google Scholar] [CrossRef]
  155. Pailler, E.; Adam, J.; Barthelemy, A.; Oulhen, M.; Auger, N.; Valent, A.; Borget, I.; Planchard, D.; Taylor, M.; Andre, F.; et al. Detection of circulating tumor cells harboring a unique ALK rearrangement in ALK-positive non-small-cell lung cancer. J. Clin. Oncol. 2013, 31, 2273–2281. [Google Scholar] [CrossRef] [Green Version]
  156. Pailler, E.; Oulhen, M.; Borget, I.; Remon, J.; Ross, K.; Auger, N.; Billiot, F.; Ngo Camus, M.; Commo, F.; Lindsay, C.R.; et al. Circulating Tumor Cells with Aberrant ALK Copy Number Predict Progression-Free Survival during Crizotinib Treatment in ALK-Rearranged Non-Small Cell Lung Cancer Patients. Cancer Res. 2017, 77, 2222–2230. [Google Scholar] [CrossRef] [Green Version]
  157. Pailler, E.; Faugeroux, V.; Oulhen, M.; Mezquita, L.; Laporte, M.; Honore, A.; Lecluse, Y.; Queffelec, P.; NgoCamus, M.; Nicotra, C.; et al. Acquired Resistance Mutations to ALK Inhibitors Identified by Single Circulating Tumor Cell Sequencing in ALK-Rearranged Non-Small-Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 6671–6682. [Google Scholar] [CrossRef] [Green Version]
  158. Podolak, J.; Eilers, K.; Newby, T.; Slottke, R.; Tucker, E.; Olson, S.B.; Lue, H.W.; Youngren, J.; Aggarwal, R.; Small, E.J.; et al. Androgen receptor amplification is concordant between circulating tumor cells and biopsies from men undergoing treatment for metastatic castration resistant prostate cancer. Oncotarget 2017, 8, 71447–71455. [Google Scholar] [CrossRef] [Green Version]
  159. Okegawa, T.; Ninomiya, N.; Masuda, K.; Nakamura, Y.; Tambo, M.; Nutahara, K. AR-V7 in circulating tumor cells cluster as a predictive biomarker of abiraterone acetate and enzalutamide treatment in castration-resistant prostate cancer patients. Prostate 2018, 78, 576–582. [Google Scholar] [CrossRef]
  160. Antonarakis, E.S.; Lu, C.; Luber, B.; Wang, H.; Chen, Y.; Zhu, Y.; Silberstein, J.L.; Taylor, M.N.; Maughan, B.L.; Denmeade, S.R.; et al. Clinical Significance of Androgen Receptor Splice Variant-7 mRNA Detection in Circulating Tumor Cells of Men With Metastatic Castration-Resistant Prostate Cancer Treated With First- and Second-Line Abiraterone and Enzalutamide. J. Clin. Oncol. 2017, 35, 2149–2156. [Google Scholar] [CrossRef]
  161. Tagawa, S.T.; Antonarakis, E.S.; Gjyrezi, A.; Galletti, G.; Kim, S.; Worroll, D.; Stewart, J.; Zaher, A.; Szatrowski, T.P.; Ballman, K.V.; et al. Expression of AR-V7 and ARv567es in circulating tumor cells correlates with outcomes to taxane therapy in men with metastatic prostate cancer treated in TAXYNERGY. Clin. Cancer Res. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Antonarakis, E.S.; Tagawa, S.T.; Galletti, G.; Worroll, D.; Ballman, K.; Vanhuyse, M.; Sonpavde, G.; North, S.; Albany, C.; Tsao, C.K.; et al. Randomized, Noncomparative, Phase II Trial of Early Switch From Docetaxel to Cabazitaxel or Vice Versa, With Integrated Biomarker Analysis, in Men With Chemotherapy-Naive, Metastatic, Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2017, 35, 3181–3188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Green, B.J.; Nguyen, V.; Atenafu, E.; Weeber, P.; Duong, B.T.V.; Thiagalingam, P.; Labib, M.; Mohamadi, R.M.; Hansen, A.R.; Joshua, A.M.; et al. Phenotypic Profiling of Circulating Tumor Cells in Metastatic Prostate Cancer Patients Using Nanoparticle-Mediated Ranking. Anal. Chem. 2019, 91, 9348–9355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Incorvaia, L.; Fanale, D.; Badalamenti, G.; Barraco, N.; Bono, M.; Corsini, L.R.; Galvano, A.; Gristina, V.; Listì, A.; Vieni, S.; et al. Programmed Death Ligand 1 (PD-L1) as a Predictive Biomarker for Pembrolizumab Therapy in Patients with Advanced Non-Small-Cell Lung Cancer (NSCLC). Adv. Ther. 2019, 36, 2600–2617. [Google Scholar] [CrossRef] [Green Version]
  165. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
  166. Grosso, J.; Horak, C.E.; Inzunza, D.; Cardona, D.M.; Simon, J.S.; Gupta, A.K.; Sankar, V.; Park, J.-S.; Kollia, G.; Taube, J.M.; et al. Association of tumor PD-L1 expression and immune biomarkers with clinical activity in patients (pts) with advanced solid tumors treated with nivolumab (anti-PD-1; BMS-936558; ONO-4538). J. Clin. Oncol. 2013, 31, 3016. [Google Scholar] [CrossRef]
  167. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [Green Version]
  168. McDermott, D.F.; Kluger, H.; Sznol, M.; Carvajal, R.; Lawrence, D.; Topalian, S.L.; Atkins, M.B.; Powderly, J.D.; Sharfman, W.H.; Puzanov, I.; et al. 1088PD—Long-Term Survival of Ipilimumab-Naïve Patients (Pts) with Advanced Melanoma (Mel) Treated with Nivolumab (Anti-Pd-1; Bms-936558, Ono-4538) in a Phase 1 Trial. Ann. Oncol. 2014, 25, 375. [Google Scholar] [CrossRef] [Green Version]
  169. Chikamatsu, K.; Tada, H.; Takahashi, H.; Kuwabara-Yokobori, Y.; Ishii, H.; Ida, S.; Shino, M. Expression of immune-regulatory molecules in circulating tumor cells derived from patients with head and neck squamous cell carcinoma. Oral. Oncol. 2019, 89, 34–39. [Google Scholar] [CrossRef]
  170. Janning, M.; Kobus, F.; Babayan, A.; Wikman, H.; Velthaus, J.L.; Bergmann, S.; Schatz, S.; Falk, M.; Berger, L.A.; Bottcher, L.M.; et al. Determination of PD-L1 Expression in Circulating Tumor Cells of NSCLC Patients and Correlation with Response to PD-1/PD-L1 Inhibitors. Cancers 2019, 11, 835. [Google Scholar] [CrossRef] [Green Version]
  171. Chen, Y.L.; Huang, W.C.; Lin, F.M.; Hsieh, H.B.; Hsieh, C.H.; Hsieh, R.K.; Chen, K.W.; Yen, M.H.; Lee, J.; Su, S.; et al. Novel circulating tumor cell-based blood test for the assessment of PD-L1 protein expression in treatment-naive, newly diagnosed patients with non-small cell lung cancer. Cancer Immunol. Immunother. 2019, 68, 1087–1094. [Google Scholar] [CrossRef] [Green Version]
  172. Guibert, N.; Delaunay, M.; Lusque, A.; Boubekeur, N.; Rouquette, I.; Clermont, E.; Mourlanette, J.; Gouin, S.; Dormoy, I.; Favre, G.; et al. PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab. Lung Cancer 2018, 120, 108–112. [Google Scholar] [CrossRef] [PubMed]
  173. Sun, W.W.; Xu, Z.H.; Lian, P.; Gao, B.L.; Hu, J.A. Characteristics of circulating tumor cells in organ metastases, prognosis, and T lymphocyte mediated immune response. Onco Targets Ther. 2017, 10, 2413–2424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Khattak, M.A.; Reid, A.; Freeman, J.; Pereira, M.; McEvoy, A.; Lo, J.; Frank, M.H.; Meniawy, T.; Didan, A.; Spencer, I.; et al. PD-L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study. Oncologist 2020, 25, e520–e527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Yue, C.; Jiang, Y.; Li, P.; Wang, Y.; Xue, J.; Li, N.; Li, D.; Wang, R.; Dang, Y.; Hu, Z.; et al. Dynamic change of PD-L1 expression on circulating tumor cells in advanced solid tumor patients undergoing PD-1 blockade therapy. Oncoimmunology 2018, 7, e1438111. [Google Scholar] [CrossRef]
  176. Graf, R.P.; Hullings, M.; Barnett, E.S.; Carbone, E.; Dittamore, R.; Scher, H.I. Clinical Utility of the Nuclear-localized AR-V7 Biomarker in Circulating Tumor Cells in Improving Physician Treatment Choice in Castration-resistant Prostate Cancer. Eur. Urol. 2020, 77, 170–177. [Google Scholar] [CrossRef]
  177. Messaritakis, I.; Nikolaou, M.; Koinis, F.; Politaki, E.; Koutsopoulos, A.; Lagoudaki, E.; Vetsika, E.K.; Georgoulias, V.; Kotsakis, A. Characterization of DLL3-positive circulating tumor cells (CTCs) in patients with small cell lung cancer (SCLC) and evaluation of their clinical relevance during front-line treatment. Lung Cancer 2019, 135, 33–39. [Google Scholar] [CrossRef]
  178. Troncarelli Flores, B.C.; Souza, E.S.V.; Ali Abdallah, E.; Mello, C.A.L.; Gobo Silva, M.L.; Gomes Mendes, G.; Camila Braun, A.; Aguiar Junior, S.; Thome 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] [Green Version]
  179. Qiu, Z.L.; Wei, W.J.; Sun, Z.K.; Shen, C.T.; Song, H.J.; Zhang, X.Y.; Zhang, G.Q.; Chen, X.Y.; Luo, Q.Y. Circulating Tumor Cells Correlate with Clinicopathological Features and Outcomes in Differentiated Thyroid Cancer. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 48, 718–730. [Google Scholar] [CrossRef]
  180. Bao, H.; Bai, T.; Takata, K.; Yokobori, T.; Ohnaga, T.; Hisada, T.; Maeno, T.; Bao, P.; Yoshida, T.; Kumakura, Y.; et al. High expression of carcinoembryonic antigen and telomerase reverse transcriptase in circulating tumor cells is associated with poor clinical response to the immune checkpoint inhibitor nivolumab. Oncol. Lett. 2018, 15, 3061–3067. [Google Scholar] [CrossRef] [Green Version]
  181. Gasch, C.; Oldopp, T.; Mauermann, O.; Gorges, T.M.; Andreas, A.; Coith, C.; Muller, V.; Fehm, T.; Janni, W.; Pantel, K.; et al. Frequent detection of PIK3CA mutations in single circulating tumor cells of patients suffering from HER2-negative metastatic breast cancer. Mol. Oncol. 2016, 10, 1330–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Scher, H.I.; Lu, D.; Schreiber, N.A.; Louw, J.; Graf, R.P.; Vargas, H.A.; Johnson, A.; Jendrisak, A.; Bambury, R.; Danila, D.; et al. Association of AR-V7 on Circulating Tumor Cells as a Treatment-Specific Biomarker With Outcomes and Survival in Castration-Resistant Prostate Cancer. JAMA Oncol. 2016, 2, 1441–1449. [Google Scholar] [CrossRef] [PubMed]
  183. Nicolazzo, C.; Raimondi, C.; Mancini, M.; Caponnetto, S.; Gradilone, A.; Gandini, O.; Mastromartino, M.; Del Bene, G.; Prete, A.; Longo, F.; et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci. Rep. 2016, 6, 31726. [Google Scholar] [CrossRef] [PubMed]
  184. Genna, A.; Vanwynsberghe, A.M.; Villard, A.V.; Pottier, C.; Ancel, J.; Polette, M.; Gilles, C. EMT-Associated Heterogeneity in Circulating Tumor Cells: Sticky Friends on the Road to Metastasis. Cancers 2020, 12, 1632. [Google Scholar] [CrossRef]
  185. Mak, M.P.; Tong, P.; Diao, L.; Cardnell, R.J.; Gibbons, D.L.; William, W.N.; Skoulidis, F.; Parra, E.R.; Rodriguez-Canales, J.; Wistuba, I.I.; et al. A Patient-Derived, Pan-Cancer EMT Signature Identifies Global Molecular Alterations and Immune Target Enrichment Following Epithelial-to-Mesenchymal Transition. Clin. Cancer Res. 2016, 22, 609–620. [Google Scholar] [CrossRef] [Green Version]
  186. Bergmann, S.; Coym, A.; Ott, L.; Soave, A.; Rink, M.; Janning, M.; Stoupiec, M.; Coith, C.; Peine, S.; von Amsberg, G.; et al. Evaluation of PD-L1 expression on circulating tumor cells (CTCs) in patients with advanced urothelial carcinoma (UC). Oncoimmunology 2020, 9, 1738798. [Google Scholar] [CrossRef] [Green Version]
  187. Chae, Y.K.; Chang, S.; Ko, T.; Anker, J.; Agte, S.; Iams, W.; Choi, W.M.; Lee, K.; Cruz, M. Epithelial-mesenchymal transition (EMT) signature is inversely associated with T-cell infiltration in non-small cell lung cancer (NSCLC). Sci. Rep. 2018, 8, 2918. [Google Scholar] [CrossRef] [Green Version]
  188. Lou, Y.; Diao, L.; Cuentas, E.R.; Denning, W.L.; Chen, L.; Fan, Y.H.; Byers, L.A.; Wang, J.; Papadimitrakopoulou, V.A.; Behrens, C.; et al. Epithelial-Mesenchymal Transition Is Associated with a Distinct Tumor Microenvironment Including Elevation of Inflammatory Signals and Multiple Immune Checkpoints in Lung Adenocarcinoma. Clin. Cancer Res. 2016, 22, 3630–3642. [Google Scholar] [CrossRef] [Green Version]
  189. Song, N.; Bai, M.; Che, X.; Li, Z.; Jing, W.; Li, C.; Teng, Z.; Qu, X.; Liu, Y. PD-L1 upregulation accompanied with epithelial-mesenchymal transition attenuates sensitivity to ATR inhibition in p53 mutant pancreatic cancer cells. Med. Oncol. 2020, 37, 47. [Google Scholar] [CrossRef]
  190. Raimondi, C.; Carpino, G.; Nicolazzo, C.; Gradilone, A.; Gianni, W.; Gelibter, A.; Gaudio, E.; Cortesi, E.; Gazzaniga, P. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoimmunology 2017, 6, e1315488. [Google Scholar] [CrossRef] [Green Version]
  191. Ock, C.Y.; Kim, S.; Keam, B.; Kim, M.; Kim, T.M.; Kim, J.H.; Jeon, Y.K.; Lee, J.S.; Kwon, S.K.; Hah, J.H.; et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget 2016, 7, 15901–15914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Koh, Y.W.; Han, J.H.; Haam, S. Expression of PD-L1, cancer stem cell and epithelial-mesenchymal transition phenotype in non-small cell lung cancer. Pathology 2020. [Google Scholar] [CrossRef] [PubMed]
  193. 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]
  194. Qiu, X.Y.; Hu, D.X.; Chen, W.-Q.; Chen, R.Q.; Qian, S.R.; Li, C.Y.; Li, Y.J.; Xiong, X.X.; Liu, D.; Pan, F.; et al. PD-L1 confers glioblastoma multiforme malignancy via Ras binding and Ras/Erk/EMT activation. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1754–1769. [Google Scholar] [CrossRef] [PubMed]
  195. Jiang, Y.; Zhan, H. Communication between EMT and PD-L1 signaling: New insights into tumor immune evasion. Cancer Lett. 2020, 468, 72–81. [Google Scholar] [CrossRef]
  196. Terry, S.; Savagner, P.; Ortiz-Cuaran, S.; Mahjoubi, L.; Saintigny, P.; Thiery, J.P.; Chouaib, S. New insights into the role of EMT in tumor immune escape. Mol. Oncol. 2017, 11, 824–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Chen, C.L.; Mahalingam, D.; Osmulski, P.; Jadhav, R.R.; Wang, C.M.; Leach, R.J.; Chang, T.C.; Weitman, S.D.; Kumar, A.P.; Sun, L.; et al. Single-cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT-related genes in metastatic prostate cancer. Prostate 2013, 73, 813–826. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of circulating tumor cells (CTCs) during the metastatic process and characterization as a clinical biomarker after isolation. During the metastatic process, CTCs undergo gradual or full epithelial-to-mesenchymal transition (EMT) by which the primary epithelial tumor cells acquire mesenchymal characteristics and enhance metastatic and invasive abilities. Circulating tumor stem cells (CTSCs) exhibit the stem cell properties and the mesenchymal phenotype. Circulating tumor microemboli (CTM) are the CTC clusters, which often appear to be a heterogeneous cell population. The presence of CTCs and CTM with mesenchymal markers associate with chemoresistance and poor clinical outcomes, and the detection of these cells has significance as surrogate biomarkers for decision-making in therapeutic strategy.
Figure 1. Schematic representation of circulating tumor cells (CTCs) during the metastatic process and characterization as a clinical biomarker after isolation. During the metastatic process, CTCs undergo gradual or full epithelial-to-mesenchymal transition (EMT) by which the primary epithelial tumor cells acquire mesenchymal characteristics and enhance metastatic and invasive abilities. Circulating tumor stem cells (CTSCs) exhibit the stem cell properties and the mesenchymal phenotype. Circulating tumor microemboli (CTM) are the CTC clusters, which often appear to be a heterogeneous cell population. The presence of CTCs and CTM with mesenchymal markers associate with chemoresistance and poor clinical outcomes, and the detection of these cells has significance as surrogate biomarkers for decision-making in therapeutic strategy.
Cancers 12 03588 g001
Table 1. Mesenchymal markers expressed on circulating tumor cells (CTCs) from patients with different tumor types and clinical significance as surrogate biomarkers.
Table 1. Mesenchymal markers expressed on circulating tumor cells (CTCs) from patients with different tumor types and clinical significance as surrogate biomarkers.
Cancer TypeMolecular Marker on CTCDetection MethodSignificance as BiomarkerPositivity of Total PtsRef.
Advanced NSCLCVIMCellSearch®Increasing in the EGFR-mutated subgroup20.80%[68]
Gastric cancerVIMCyttel methodImmune evasion capacity of CTCs75.86%[69]
Gastric cancerCSV/PD-L1CSV magnetic positive selectionShort survival duration and poor therapeutic response71%[46]
Lung cancerVIMVimentin-iFISHShorter PFS19.70%[70]
Gastric cancerCK+/CD44+Flow cytometry sortingShortened survival10%[71]
NSCLCVIM/N-Cadherin/PD-L1Immunofluorescent stainingPoorer survival after curative surgery86.70%[72]
NSCLCVIM/TwistCanPatrol™Predictor of metastasis80.00%[73]
mBCALDH1/TWIST1Immunofluorescent stainingChemoresistance, lung metastases, and decreased PFS27.70%[22]
CRPCEpCAMlow-expressionCellSearch®Shorter OS in ≥5 EpCAMhigh-expression CTC28.00%[74]
mBCEpCAMlow-expressionCellSearch® Shorter OS in ≥5 EpCAMhigh-expression CTC36%[74]
Colorectal cancerLGR5+CanPatrol™Metastasis86.40%[75]
mBC (triple negative subtype)VIMMicrofluidic Chip deviceShorter PFS100%[51]
Esophageal squamous cell carcinomaVIM/TwistCanPatrol™Staging32.60%[76]
Hepatocellular carcinoma VIM/TwistCanPatrol™Shortened postoperative disease-free survival38.79%[75]
Breast cancerTUB/VIM/GLUISETMetastases8.24% (VIM)[77]
Hepatocellular carcinomaVIM/TwistCanPatrol™Metastasis90.18%[78]
Colorectal cancerAKT2/SNAIL1CanPatrol™Staging and metastasis56.90%[79]
Colorectal cancerVIMCanPatrol™Metastasis42.06%[80]
Hepatocellular carcinoma VIM/TwistCanPatrol™Shortened postoperative disease-free survival69.20%[81]
Prostate cancer CK/VIMParsortix systemTumor aggressiveness and poorer survival73%[59]
NSCLCVIMTelomeScan®Shorter PFS46%[23]
Ovarian cancerPI3Kα/Akt-2/TwistAdnaTest OvarianCancer and EMT-1Suggested therapeutic resistance30%[82]
mBCVIM/TwistCanPatrol™Increasing in the negative hormone receptor subgroup93%[83]
mCRPCVIMCellSearch®Shorter OS32.30%[84]
VIM: vimentin; CSV: cell-surface vimentin; CK: cytokeratin; mCRPC: metastatic castration-resistant prostate cancer; NSCLC: non-small cell lung cancer; SCLC: small cell lung cancer; mBC: metastatic breast cancer; Pts: patients.
Table 2. Circulating cancer stem cell (CTSC) markers from different tumor types and clinical significance as surrogate biomarkers.
Table 2. Circulating cancer stem cell (CTSC) markers from different tumor types and clinical significance as surrogate biomarkers.
Cancer TypeMolecular Marker on CTCDetection MethodClinical Significance as BiomarkerPositivity of Total PtsRef.
Gastric cancerCK+/CD44+Flow cytometry sortingShortened survival10%[71]
NSCLCCD133Immunofluorescent stainingShortened survival84%[116]
CRPCCD133CellSearch®Metastases82%[117]
Breast cancerALDH1Immunomagnetic cell selectionRecurrence and survival14%[118]
NSCLCALDH1RT-PCR following immunomagnetic cell selectionProgressive metastases25%[120]
mBCALDH1+/VIM+/fibronectin+Immunomagnetic cell selectionMetastases34%[121]
mBCALDH1/nuclear TwistImmunofluorescent stainingDisease progression76%[92]
mBCEMT markers (TWIST, Akt2, PI3K) and/or ALDH1Immunomagnetic cell selectionTherapy resistant38%[122]
Breast cancerCD44+/CD24+ or low-expressionImmunofluorescent stainingMetastases23%[123]
CK: cytokeratin; ALDH1: aldehyde dehydrogenase-1; VIM: vimentin; NSCLC: non-small cell lung cancer; CRPC: castration-resistant prostate cancer; PFS: progression-free survival; EMT: epithelial-to-mesenchymal transition; mBC: metastatic breast cancer; Pts: patients.
Table 3. The potential treatment response markers expressed on CTCs for future precision medicine.
Table 3. The potential treatment response markers expressed on CTCs for future precision medicine.
Cancer TypeMolecular Marker on CTCDetection MethodClinical Significance as BiomarkerPositivity of Total PtsRef.
mCRPCNuclear-localized AR-V7Immunofluorescent stainingTherapeutic response of taxane22%[176]
Metastatic melanomaPD-L1Multiparametric flow cytometryTherapeutic response of pembrolizumab and longer PFS64%[174]
SCLCDLL3+/CD45CellSearch®Therapeutic response of etoposide/platinum and decreased PFS74.10%[177]
Rectal cancerTYMS/RAD23BISET®Predict resistance of neoadjuvant chemoradiotherapy100% (TYMS mRNA)/75% (RAD protein)[178]
NSCLCPD-L1CellSearch®Resistance to PD-1/PD-L1 blockade therapies47%[170]
Metastatic thyroid cancerEpCAM-/CD45-/DAPI+/CEP8AneuploidyPoor response to 131I treatment and worse prognosis86.11%[179]
Advanced gastrointestinal tumorPD-L1Immunofluorescent stainingTherapeutic response of PD-1/PD-L1 blockade therapies74%[175]
mBCTopoisomerase 1ApoStream (enrich for CTCs)Therapeutic response of topoisomerase 1 inhibitor etirinotecan pegol52%[151]
Advanced NSCLCCEA/hTERTCTC chipTherapeutic response of nivolumab, PD-1 inhibitorOnly selected patients[180]
Advanced NSCLCPD-L1ISETPoor response to nivolumab, PD-1 inhibitor and shorter PFS83%[172]
mCRPCSynaptophysinCellSearch®Resistance to AR-targeted therapies, abiraterone, and enzalutamideOnly selected patients[150]
SCLCBcl-2+Immunofluorescent stainingPrognostic and treatment efficacy72.70%[152]
mCRPCAR splice variantsCellSearch®Therapeutic response of taxane67%: AR-V7+, 78%: ARv567es+[161]
mCRPCAR splice variantsFlow cytometryTherapeutic response of AR-targeted therapies, abiraterone, and enzalutamide26.5%: CTC cluster(+)/AR-V7(+)[159]
mCRPCAR nuclear localization (%ARNL)CellSearch®Suggested resistance to taxane with increasing PSAOnly selected patients[162]
Advanced NSCLC with ALK rearrangementALK-CNGISETTherapeutic response of ALK inhibitor, crizotinib, by decreasing CTC of ALK-CNG100%[156]
mCRPCAR splice variantsEpCAM-based Prostate Cancer Select kitTherapeutic response of AR-targeted therapies, abiraterone. and enzalutamide17.8%: AR-V7+[160]
mBCPIK3CA mutationsCellSearch®Resistance to anti-HER2 therapy36.40%[181]
mBCHER2CellSearch®Therapeutic response of anti-HER2 therapy37.9[147]
mCRPCNuclear-localized AR-V7Immunofluorescent stainingResistance to AR inhibitors18%[182]
Advanced NSCLCPD-L1CellSearch®Resistance to nivolumab, PD-1 inhibitor93%[183]
mCRPC: metastatic castration-resistant prostate cancer; NSCLC: non-small cell lung cancer; SCLC: small cell lung cancer; mBC: metastatic breast cancer; AR: androgen receptor; ALK-CNG: ALK-copy number gain; PFS: progression-free survival; TYMS: thymidylate synthase; RAD23B: UV excision repair protein RAD23 homolog B; Pts: patients.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Okabe, T.; Togo, S.; Fujimoto, Y.; Watanabe, J.; Sumiyoshi, I.; Orimo, A.; Takahashi, K. Mesenchymal Characteristics and Predictive Biomarkers on Circulating Tumor Cells for Therapeutic Strategy. Cancers 2020, 12, 3588. https://doi.org/10.3390/cancers12123588

AMA Style

Okabe T, Togo S, Fujimoto Y, Watanabe J, Sumiyoshi I, Orimo A, Takahashi K. Mesenchymal Characteristics and Predictive Biomarkers on Circulating Tumor Cells for Therapeutic Strategy. Cancers. 2020; 12(12):3588. https://doi.org/10.3390/cancers12123588

Chicago/Turabian Style

Okabe, Takahiro, Shinsaku Togo, Yuichi Fujimoto, Junko Watanabe, Issei Sumiyoshi, Akira Orimo, and Kazuhisa Takahashi. 2020. "Mesenchymal Characteristics and Predictive Biomarkers on Circulating Tumor Cells for Therapeutic Strategy" Cancers 12, no. 12: 3588. https://doi.org/10.3390/cancers12123588

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