Open access peer-reviewed chapter

The Selection Strategy for Circulating Tumor Cells (CTCs) Isolation and Enumeration: Technical Features, Methods, and Clinical Applications

Written By

Jason Chia‐Hsun Hsieh and Tyler Ming‐Hsien Wu

Submitted: 16 June 2016 Reviewed: 05 July 2016 Published: 14 September 2016

DOI: 10.5772/64812

From the Edited Volume

Tumor Metastasis

Edited by Ke Xu

Chapter metrics overview

2,054 Chapter Downloads

View Full Metrics

Abstract

The key aim of the proposed chapter is to provide readers a brief description for the most important parts of the field of circulating tumor cells (CTCs): the core techniques, including negative and positive selection‐based CTC isolation, and the differences between them. Most importantly, we will also review the clinical applications and important findings in clinical trials. The evidence‐based review will not only help clinicians use CTCs to predict recurrence and foresee the disease‐related outcomes but also to inspire the researchers in this field to conduct further investigations.

Keywords

  • circulating tumor cells
  • negative selection
  • cancer
  • stem cell
  • liquid biopsy

1. Introduction of circulating tumor cells (CTCs)

1.1. Brief history of CTC researches

Circulating tumor cells (CTCs) are cells shedding from primary tumor(s) into the adjacent vasculature and are floating around in the circulation throughout the human body. The cells, as seeds for the subsequent initiation of distant site metastases, are responsible for the cancer‐related deaths [1]. For the first time, CTCs were described via the observation in the blood of cancer patient by Dr. Thomas Ramsden Ashworth, who postulated that “cells identical with those of the cancer itself being seen in the blood may tend to throw some light upon the mode of origin of multiple tumors existing in the same person” in 1869 [2]. In 1906, Goldmann reported that visible venous invasion by cancer in approximately 20% of 500 necropsies and microscopic invasion of vasculature in nearly 10% [3]. In very early 1900s, several reports of observation of free cancer cells by morphology have been discussed in patients with melanoma, gastric cancer [46], and lung cancer [7]. However, long being in the technical limitation on isolation of these rare cells in circulation, the realization of CTCs isolated from living cancer patients and analysis of their clinical impacts began since 1930s [810]. One of the first systemic surveys in 40 living cancer patients was done by Pool and Dunlop in 1934 [11]. In this period, morphology and cytochemical characteristics remained the most important method to identify “abnormal” or “atypical” cells. Tumor cell embolization was observed [12] and widely accepted to be one of the major mechanisms for dissemination of cancer [13, 14]. Since 1986 with the development of polymerase chain reaction (PCR), investigators began to utilize these nucleic acid‐based detection methods to help identify CTCs, including circulating tumor mutated DNA and mRNAs [1523]. In early 2000s, semi‐automated devices appeared and facilitate the advances of CTC testing in clinical study enrolling healthy subjects and patients with various types of cancer [2426], given the fact that numerous previous methods were relatively operator‐dependent and often lacked of validated sensitivity, specificity, coefficient of variation and reproducibility [22, 2731]. In 2004, CellSearch™ (Veridex, Janssen Diagnostics, USA) got approval from United States (US) Food and Drug Administration (FDA) for testing in patients with breast, colorectal, and prostate cancer [32, 33]. Since 2012, a rapid exploration of number of CTC isolation devices having nearly fully automated design emerged [3440].

Recently, CTC studies are focusing on devices harboring high sensitivity, high specificity, reduced sample requirement, label‐free isolation, and the ability to catch living CTCs for in‐vitro culture. Owing to the less invasive nature than conventional cancer tissue biopsy, CTCs, as serum circulating tumor DNA (ctDNA) and microRNA (miRNA), are termed to be “liquid biopsies.”

1.2. Natures of CTCs

It has been a long time after CTCs were noticed and efficiently captured by many methods; however, little is known about the behavior of CTCs [41]. Investigators have observed some phenomenon about what CTCs look like, how CTCs shed, migrate, live, defense human immune system, and initiate distant metastases.

First, CTCs were believed to be larger than normal blood cells, which contributed the development size‐based isolation strategy. Marrinucci et al. [42] supported that the fact of CTCs being larger than white blood cells and having high nuclear to cytoplasmic ratios with voluminous cytoplasm. In addition, the morphology of CTCs is highly similar to that of cells from original biopsied cancer tissues. Numerous devices were developed on the basis of this characteristics and collect cells with larger size (often >15 μm), including dielectrophoresis (DEP) [43, 44], optically induced dielectrophorestic (ODEP) [45] force‐dependent devices, and filter‐based systems [4661]. However, other investigators found that the size of real CTCs could be greatly differed from cell lines [62], and might even vary interindividually and intraindividually [63, 64]. The size criteria of CTC definition remain in debate.

Second, CTCs exist in almost all staged cancer and could be detected in the course of the disease [27]. In 1995, Hansen et al. analyzed the blood samples drawn from surgical fields during 61 oncologic surgeries and 93.4% samples found tumor cells [65], which suggest one of the possible routes of early dissemination of cancer cells. In 2000, Yamashita et al. found that signals of CTCs (carcinoembryonic antigen messenger RNA, CEA mRNA) from preoperatively negative to postsurgically positive might suggest a specific type of surgery could contribute to the cancer cell dissemination [66]. Similar results were reported by other investigators to support the findings in various types of cancer (but breast cancer mainly) [6773]. Although CTCs were found in early‐stage cancer patients, the cells do not result in metastasis all the times. The clearance of human immune systems and inadequate “soil” of distant organs are one of the plausible explanations. To look on the bright side, CTCs in very early stage cancer could help early diagnosis of cancer and prevent overwhelming dissemination and cancer death [74].

Third, CTCs would form cell clusters, clumps, or circulating tumor microemboli (CTM) and were found to highly correlate to cancer progression [7579] and resistance to systemic anticancer therapies [78, 80], which was established on the basis of many animal model and preclinical reports [8185]. Recently, Sarioglu et al. [86] designed a Cluster‐Chip for efficient capture of CTC clusters and enable the detailed characterization of the biological properties and role of CTC clusters in metastasis [86]. Studies of cancer metastasis have emphasized the novel concept of “seed and soil” as a key determinant of metastatic propensity [87]. This model matches the importance of mutated genetic drivers within tumor cells conferring proliferative and invasive properties, with that of the microenvironment of the distant organ or “niche,” which may facilitate metastasis occurrence. However, the physical characteristics of single CTCs and CTC clusters may also contribute to metastatic propensity, especially as they impact the ability of epithelial tumor cells to survive the loss of cell adherence and shear forces in the blood stream, i.e., different survival signals among the cancer cell “seeds” may be important. For instance, in a mouse endogenous pancreatic cancer model, noncanonical Wnt signaling is elevated within CTCs, where it appears to suppress anoikis [88], while in a subcutaneous tumor xenograft model, the admixture of tumor and stromal cells within microemboli may contribute to stromal‐derived survival signals [77, 89, 90].

Fourth, CTCs could also be detected in cancer patients who underwent curative surgery, indicating minimal residual disease in the circulation [9199] and suggesting the correlation to disease recurrence in the following months [92, 95, 100, 101]. van Dalum et al. [99] found that the presence of CTC in blood drawn pre and one and two years after surgery, but not postsurgery is associated with shorter Relapse-free survival (RFS) and OS for stages I–III breast cancer, which could partially answer the question of how frequent and how long oncologists should follow patients’ CTCs up and the timing of CTC testing after curative surgery. This phenomenon indicates that postsurgical adjuvant therapy might be required in specific population on the basis of CTC testing which remains uncertain to date.

Fifth, the captured CTCs according to their method of isolation, sometimes are alive for in‐vitro culture [48, 61, 102, 103] and might play a very important role to continuously obtain primary cancer cell lines in the near future. Furthermore, CTC‐derived cell lines and xenografts might reveal new therapeutic targets and can be used for drug screening [104, 105]. The phenomenon is the main difference of CTCs from ctDNA possibly being released from dead cancer cells. In addition, living CTCs can also colonize their tumors of origin, in a process that is call “tumor self‐seeding.” Kim et al. [106] successfully revealed the self‐seeding phenomenon in breast cancer, colon cancer, and melanoma tumors in mice model, which was predominantly mediated by CTCs with aggressive features, including those with bone, lung, or brain metastatic tropism. The cancer‐derived cytokines IL‐6 and IL‐8 acted as CTC attractants and the markers MMP1/collagenase‐1 and the actin cytoskeleton component fascin‐1 as mediators of CTC infiltration into mammary tumors. The important findings of tumor self‐seeding phenomenon could explain the relationships between tumor size, anaplasia, vascularity and prognosis, and local recurrence seeded by disseminated cells following ostensibly complete tumor excision.

Sixth, CTCs could represent a merged status of a whole tumor mass, including static and active parts with expression of specific functional markers [107110] and could serve as a multifunctional biomarker [108, 111]. Functional analyses on CTCs might provide the possibility to identify the biological characteristics of metastatic cancer cells, including the identification of metastasis‐initiating cells [104].

1.3. CTCs in cancer progression

1.3.1. Cancer migration, invasion, epithelial‐mesenchymal transition (EMT), mesenchymal‐epithelial transition (MET) and cancer stem cells (CSCs)

As mentioned above, Hansen et al. [65] found that CTCs exist in the 93% blood samples drawn from surgical fields. That correlated to one of the two common routes of cancer migration: hematologic and lymphatic spreading. In clinical aspect, tumor migration and invasion means tumor growth or progression and can be analyzed via the time from disease‐free status to recurrence or time from baseline to enlargement of tumor size. Early in 1999, Palmieri et al. have found a significant correlation among clinical stages, tumor progression, and presence of circulating cancer‐associated antigens in stages I–III melanoma patients [112]. In other cancer types, investigators widely agreed with the observation that the higher CTCs signals indicate to higher cancer stage and recurrence rate, suggesting larger number of CTCs might promote cancer progression [113117]. However, not only CTC count but also the specific properties of cancer cells matter. Two of them have been widely reported are epithelial‐mesenchymal transitions (EMTs) or stem‐like properties of CTCs [118].

In many animal species, EMTs normally occur during critical phases of embryonic development. The formation of mesenchymal cells (nonepithelial) that are loosely embedded in an extracellular matrix from a primitive epithelium is an important feature of most metazoans [119]. During this transition, mesenchymal cells acquire a morphology that is appropriate for migration in an extracellular environment and settlement in areas that are involved in organ formation, which involves interactions between epithelial and mesenchymal cells. Mesenchymal cells can also participate in the formation of epithelial organs through mesenchymal‐epithelial transition (MET) [119]. CTCs may also undergo phenotypic EMT changes, which allow them to travel to the site of metastasis formation without getting affected by conventional treatment [118, 120, 121]. The acquired molecular changes by CTCs undergoing EMT that facilitates cancer progression and resistance to conventional therapies [122, 123]. EMT markers, including vimentin, twist, ZEB1, ZEB2, snail, slug, and N‐cadherin in CTCs, primary HCC tumors and adjacent nontumoral liver tissues were evaluated by Li et al. [123] and the twist and vimentin expression levels in CTCs could be promising biomarkers for evaluating metastasis and prognosis in liver cancer patients. Most importantly, CTCs would abandon their epithelial properties (EpCAM) [88, 124] and escape from CTC capture by positive selection strategy (will discuss below) and become one of the main downsides of the strategy of CTC isolation. Several investigators have noticed that phenomenon and suggested that there is an urgent need for optimizing CTCs detection methods through the inclusion of EMT markers [120, 125129]. Deeper understanding of those processes is of fundamental importance for the development of new strategies of early cancer detection and effective cancer treatment approaches that will be translated into clinical practice [122].

Stemness features of CTCs, sometimes termed as circulating cancer stem cells (CSCs), have also been getting noticed as EMT of CTCs in recent years. The CSC hypothesis claims that a small subset of cells within a tumor has the ability of both tumor initiation and sustaining tumor growth [130132]. These cells with expression of stemness markers are capable of forming floating spheres in serum‐free medium, a property associated with stem cells and are able to differentiate into an aberrant cell phenotype constituting tumor heterogeneity [133]. Among all the possible molecular markers of stemness feature, CD133, CD44, ICAM‐1, and CXCR4 are common used antibodies for labeling the subpopulation from other CTCs and actually technically available [134136]. These are not the only markers to identify CSCs and depend on cancer types. Sun et al. [137] found that stem cell‐like phenotypes (labeled with CD133 and ABCG2) in EpCAM‐positive CTCs, and a preoperative CTC of more than 2 cells/7.5 ml blood is a novel predictor for tumor recurrence in HCC patients after surgery, especially in patient subgroups with AFP levels of less than 400 ng/ml or low tumor recurrence risk. Many other studies in various types of cancer have come across with the similar conclusions, including breast, colorectal, gastric, liver, and NSCLC, etc. [120, 123, 138145].

Therefore, the subpopulation of CTCs, CSCs, and CTC with EMT features is probably the one of the key determinants of future CTC and cancer metastasis investigations.

1.4. The impact of CTCs on multidrug resistance

In 2011, Gradilone et al. reported an interesting study aiming to test the hypothesis that drug‐resistant CTCs might have predictive value in metastatic breast cancer (MBC) and possibly retain stem‐like properties [146]. As the study presented, the extraction of mRNA from CTCs for multiple drug resistance proteins (MRPs) analysis are most commonly used protocol. They also found the expression status of MRP1 and MRP2 in CTCs was found to correlate to response to anthracyclines (doxorubicin or epirubicin) [147]. In 2013, Nadal et al. found an interesting phenomenon that a relative enrichment of cytokeratin CK(+)/CD133(+) CTCs in triple negative and HER2‐amplified tumors was found. While CK(+)/CTCs decreases after chemotherapy when analyzing the whole population, CK(+)/CD133(+) CTCs were enriched in posttreatment samples in nonluminal BC subtypes. These findings suggest the potential role of CD133 as a promising marker of chemoresistance in nonluminal BC patients [148]. Similar results were also reported in recent years and the authors have come across with the same conclusion that multiple drug resistance profiling (MRPs mainly, sometimes with CD133 [148], ALDH1 [149], and ERCC1 [150]) of CTCs could predict the responses to given chemotherapies [146, 148, 150152].

One direct proof of CTC exhibiting drug resistance comes from a study in 2014. Pavese et al. observed that CTC and DTC cell lines, established from mice bearing human prostate cancer orthotopic implants, exhibit increased cellular invasion in vitro, increased metastasis in mice, and express increased EMT biomarkers. In addition, CTC cell lines are selectively resistant to growth inhibition by mitoxantrone‐like agents. The findings are important and suggested that CTC formation is accompanied by phenotypic progression without obligate reversion. Their increased metastatic potential, selective therapeutic resistance, and differential expression of potential therapeutic targets provide a rational basis to test further interventions [153].

Therefore, developing an in‐vitro chemosensitivity test on CTCs is not impossible though it required large‐scale clinical trials to test and validate. Yu et al. applied pharmacogenomic (PGx) modeling testing on CTCs, while PGx testing was used on cancer tissue to predict the efficacy of chemotherapeutic agents in preclinical cancer models, and reported the feasibility in 2014. In the report, clinical benefit was seen for study participants treated with chemotherapy regimens predicted to be effective versus chemotherapy regimens predicted to be ineffective with regard to progression‐free (10.4 months versus 3.6 months; P < 0.0001; HR, 0.14) and overall survival (17.2 months versus 8.3 months; P < 0.0249; HR, 0.29) [151]. In another study, thymidylate synthase expression in CTCs could possibly serve as a new tool to predict 5‐fluorouracil resistance in metastatic colorectal cancer patients [152]. Other than conventional imaging studies evaluating two‐dimensional tumor size every 8–12 weeks for routine tumor assessment during anticancer therapy, CTCs could possibly serve as a rapid responding biomarker to real‐time change of cancer cells, including the early response or resistance to given therapeutic drugs [154156].

Advertisement

2. The strategies for CTC isolation and enumeration

There are hundreds of methods/protocols reported to be able to efficiently detect or isolate CTCs. In a simple way to discuss here, we have several common strategies of CTC isolation could be worthy of development in the future. The first one is label‐free isolation strategy, including size‐based, physical properties‐based, morphology‐based isolation strategy; the second one is positive selection strategy, including positively identification of cancer‐specific markers on nucleus or cell surface, or specific DNA mutation(s), mRNA(s) overexpression; and the third one is negative selection strategy, consisting of depletion of red and white blood cells by any means. Finally, the fourth one is combination of two or more strategies mentioned above.

2.1. Label‐free isolation strategy

Several novel studies using size as a key criterion of CTC identification were reported [157160]. Early in 2004, ISET system was used for a well‐designed clinical trial evaluating 44 patients with primary liver cancer and without metastases, 30 patients with chronic active hepatitis, 39 with liver cirrhosis, and 38 healthy individuals, and all participants were followed up for a mean period of 1 year. Both the presence (P = 0.01) and number (P = 0.02) of CTCs and microemboli were significantly associated with a shorter overall survival. Beta‐catenin mutations could be found in 3 of 60 CTCs which might be suggesting their impact on the initiation of cancer cells invasion [161]. Similar positive findings by size‐based CTC isolation were reported in melanoma [162164], gastric cancer [76, 165], prostate cancer [166, 167], lung cancer [168170], pancreatic cancer [103], liver cancer [127], sarcoma [171], and breast cancer [172]. Separation by physical properties, i.e., gravity, density gradients, using microfluidic technology [45, 46, 56, 60, 173186], or microfiltration [53, 172, 187, 188] were also reported to be able to capture CTCs efficiently.

By means of label‐free isolation, combined molecular analysis could be easily performed after CTC isolation owing to no chemicals exposure and less procedures done during isolation. For instance, Zheng et al. [189] reported a novel device designed based on membrane microfilter device to isolate CTCs and then send them to PCR‐based genomic analysis by performing on‐membrane electrolysis with embedded electrodes reaching each of the individual 16,000 filtering pores. Immunocytochemistry and FISH assays following label‐free isolation were reported to be successfully performed directly on the filter system [157, 176, 190, 191]. Interestingly, some investigators compared the isolation efficiency of ISET and CellSearch™ systems [76, 93] and one team concluded that a combination of ISET plus CellSearch™ would have better performance in CTC detection in NCSCL patients than ISET or CellSearch™ alone [93].

There are several disadvantages of physical methods should be noticed. First, the isolation process based on physical properties can cause the deformation and damage of CTCs by filter pores [192]. Second, larger size cells could not always be cancer cells and the isolated population often mixed up with megakaryocytes, which are very common to see in the circulation of cancer patients just underwent chemotherapy. Third, small‐size CTCs would be inevitably missed by this isolation strategy.

2.2. Positive versus negative selection‐based CTC isolation

2.2.1. Positive selection methods

Positive selection strategy is the most commonly used method of CTC isolation in the literature. CellSearch™ is the most evidenced and the only one device having class III approval from US FDA since 2004; therefore, hundreds of clinical trials chose to apply the device for CTC testing for validation [114, 193204], mainly in patients with breast, colorectal, and prostate cancer. Other representative positive selection platforms are magnetic‐activated cell sorting system (MACS) and Isoflux. The main process of positive selection is to label targets cells by anti‐CK (AE1/AE3) antibody with ferric beads and immunofluorescence dye. The approximate sensitivity of detection is 10-7 (CTCs/hematologic cells). Another system, MACS used 50–100 nm‐sized ferric beads. However, lower sensitivity and lower recovery rate of CTCs were observed. The device was firstly introduced in 1998 [205] and then CTCs obtained by the system could correlated with breast cancer stages [206] and could correlate with progression‐free survival in colorectal cancer patients [207]. These systems are all based on immunomagnetic beads technology for CTC isolation and have long been limited by relatively low efficiency of antibody conjugation due to tumor heterogeneity [192]. This limitation further causes the difficulty of molecular analysis [208]. Fortunately, the technique of single CTC isolation and analysis has been much more mature in recent years [166, 209, 210]. However, fewer sampling (CTCs) could greatly contribute to the bias for prediction of target population behavior (the whole tumors in the body). Another downside of positive selection strategy was the limitation of EpCAM‐dependent nature. Hyun et al. [124] demonstrated that EMT‐induced breast cancer cells maintained in prolonged mammosphere culture conditions possess increased EMT markers and cancer stem cell markers, as well as reduced cell mass and size by quantitative phase microscopy. In addition, EpCAM expression is dramatically decreased in these cells. Moreover, CTCs isolated from breast cancer patients using a label‐free microfluidic flow fractionation device had differing expression patterns of EpCAM, indicating that affinity approaches reliant on EpCAM expression may underestimate CTC number and potentially miss critical subpopulations.

In addition to conventional immunomagnetic bead separation methods, density separation and flow cytometry or cell sorting systems have been postulated to be potential tools of CTC isolation and identification considering their high sensitivity and purity since 1998 [27]. This method could be seen as a combination of negative selection strategy and a positive confirmation with surface markers, such as EpCAM or cytokeratins. Later in 2011, leukapheresis and fluorescence‐activated cell sorting (FACS) elutriation were also reported to be effective for large volume blood process for CTC isolation with molecular analysis [211]. Recently, many microscale on‐chip sorting systems were developed considering the high purity of isolation for CTC culture or tumor related genetic analysis. In 2014, Kim et al. have postulated an on‐chip multi‐imaging flow cytometry system to obtain morphometric parameters of cell clusters such as cell number, perimeter, total cross‐sectional area, number of nuclei, and size of clusters as “imaging biomarkers,” with simultaneous acquisition and analysis of both bright‐field and fluorescent images at 200 frames per second [212]. Moreover, laser scanning cytometry is also a novel innovation developed to help identify CTCs [213, 214]. These methods, are mainly based on flow cytometry and sorting techniques, which could possibly yield an extremely high purity of CTCs (more than 80%). However, one of the drawbacks of the cytometric systems is operator‐dependent and multiple quality and internal controls are often required when setting a criterion of CTC identification. Another downside of conventional sorting systems is cell damage and decreased viability after sorting process.

Recently, by the advances of nanotechnology, nanoplates [215], nanowires [216], for positively trapping of CTCs are becoming hot devices with theoretically higher sensitive capturing efficiency than conventional ones. These techniques are often developed by biomedical engineers who are good at medicine, biology and engineering; however, the devices seem to be still in proof‐on‐concept phase. Hopefully, these new devices would facilitate the development of easy hands‐on CTC testing and validation in clinical trials in the near future. In brief, positive selection methods hold the greatest clinical application to date.

2.2.2. Negative selection methods

Negative selection methods are developed on the basis of the disadvantages of positive selection methods‐losing non‐EpCAM or CK‐expressing CTCs and relatively poor recovery rate. The principle of negative selection strategy is to remove all the cells other than CTCs as its first step. Owing to the sequence of isolation has changed, in the negative depletion processing, the cancer information was preserved as possible, which makes the phenomenon that the number of CTCs isolated by a negative method would generally (but not always) larger than those by a positive one. In addition, by the CD45 depletion procedures, CTCs without expression of epithelial markers could be isolated though further clarification of the clinical significance of these cell populations is required [217]. The nature of the isolation strategy increased the sensitivity, recovery rate of CTCs but decrease the specificity with inevitable “background noises.”

In the developing history of the negative selection methods, Naume et al. [29, 218] have proposed to use CD45 (a common antigen of leukocytes) coated beads to remove white blood cells from tumor cells and red blood cells depleted by lysis buffer or density separation processing for CTC isolation. Based on the concepts, Balasubramanian et al. [219] also successfully demonstrated positive staining images for cytokeratin‐positive CTC identification after negative selection processes in 32 cancer blood samples. Among all the negative selection systems, one of the representative systems is epithelial immunospot (EPISPOT) [220224]. By the procedures, CTCs in the blood sample are enriched by anti‐CD45 immunomagnetic beads. The isolated CTCs are then cultured in tissue culture plates precoated with antibodies which capture cathepsin D, MUC1, or CK19 protein [225]. After the incubation period, cells are washed out and the released protein spots are detected by the incubation with a fluorochrome‐conjugated antibody and counted. Each spot corresponds to one viable CTC. The device focused on the expression of CK19‐expressing cells, which were found to be detectable in up to 65 and 70% of colorectal cancer and breast cancer patients, respectively, and correlated with status of metastasis and poor survival in breast cancer [226, 227]. However, a single sample processing in EPISPOT system requires three days for analysis, which prohibits of its clinical use considering the time‐consuming problem.

Nevertheless, the background cells in a negatively isolated sample are often mixed with numerous white blood cells and red blood cells if the process is not well‐performed, thus prohibit the following molecular analyses. For this disadvantages of conventional negative selection method, a better depletion process for red and white blood cells depletion are warranted. Another device developed by Wu et al. [228], CanPatrol™ CTC enrichment, they reported a recovery rate more than 80%. Interestingly, FISH assay could be successfully performed for ALK gene rearrangement from CTC samples; however, further validation in prospective clinical trials was still required.

Similarly, Lin et al. [229231] postulated a protocol and a device (PowerMag) to perform red blood cell lysis and immunomagnetic beads conjugation for CD45‐positve cells and identify EpCAM‐positive cells (defined as CTCs) from the blood samples. The protocol was proven to effectively isolate CTCs from patients with colorectal, head and neck cancer and thyroid cancer. Furthermore, the CTCs isolated by this negative selection method are further proven to be alive and are capable of being cultivated for at least several weeks [229].

2.3. Other methodologies

In addition to pure positive and negative or label‐free methods, some investigators proposed their prototypes for CTC isolation. For example, Qin et al. [50] performed CTC isolation by the size and deformability based separation from castrate resistant prostate cancer patients using resettable cell traps. Compared with CellSearch™, the method could capture more than 10 times of CTCs for subsequent analyses. Basically, it is a label‐free method and could be rapidly processed.

Synchrotron X‐ray microimaging techniques, high‐resolution images of individual flowing tumor cells, and nanotechnology were also proposed to help identification of CTCs. Positively charged gold nanoparticles (AuNPs) which were inappropriate for incorporation into human red blood cells were selectively incorporated into tumor cells to enhance the image contrast, which was reported by Jung et al. [232]. This new technology for in vivo imaging of CTCs would contribute to improve cancer diagnosis and cancer therapy prognosis. Moreover, new chemical materials using a refined carbon‐coated pure iron‐based immunomagnetic nanoparticle‐enriched assay, and nested‐RT‐PCR was also reported to successfully isolate CTCs efficiently.

Furthermore, not only for general population of CTCs, Hosseini et al. [233] postulated an integrated nano‐electromechanical chip (NELMEC) to isolate CTCs and CTCs with EMT features from white blood cells. These new technologies hold great promising on automation, which might greatly ameliorate current problems in CTC field.

2.3.1. Comparison of different strategies

In comparison between positive selection strategy and negative selection methods, the former is most commonly used CTC isolation platform and widely validated by prospective clinical trials. Several articles of meta‐analysis confirmed the clinical impacts of CTCs obtained by CellSearch™ [107, 234237]. However, it is relatively costly and device‐dependent. Interestingly, some investigators compared the ISET and CellSearch™ systems for their performance on CTC isolation [76, 93] and one team concluded that a combination of ISET plus CellSearch™ would have better performance in CTC detection in NCSCL patients than ISET or CellSearch™ alone [93]. That intriguing conclusion supports the combination method in the following eras; however, a long processing time of combined platforms also causes cell damage or loss which is a problem the combined systems should be noticed.

In comparison between positive selection method and label‐free strategy, Konigsberg et al. [238] compared the efficiency of CTC isolation of MACS (positive selection system) with OncoQuick (label‐free system) and found EpCAM‐negative CTCs cannot be detected by EpCAM‐dependent enrichment methods. EpCAM‐independent enrichment technologies seem to be superior to detect the entire CTC population.

Enrichment strategy System Detection markers Pros Cons
Positive selection CellSearch EpCAM, CKs, CD45,
DAPI
FDA cleared; reliable; reproducible; visual identification; clinical relevance in metastatic
breast, colorectal and
prostate cancer; semi‐automated processing; capable of detecting smaller CTCs; standardized kits
EpCAM‐positivity dependent, expensive; cells losing EpCAM could not be detected; limited number of markers
CTC chip EpCAM, CKs, CD45,
DAPI
High detection rate; visual identification EpCAM‐positivity dependent, cells losing EpCAM could not be detected; require clinical trial validation
Ariol system EpCAM, CKs, CD45,
DAPI
High detection rate (versus CellSearch) EpCAM‐positivity dependent
Laser‐scanning cytometer Automated microscopic procedure; high detection rate EpCAM‐positivity dependent
Adna test EpCAM, MUC1,
mucin‐1, HER2
High sensitivity; rapid processing No morphology confirmation; EpCAM and MUC1‐positivity dependent
Negative selection EPISPOT
assay
CD45, CK19, mucin‐1, cathepsin‐D Can detect viable CTCs Lack of enough clinical trials for validation
PowerMag CD45 depletion for 4 repeated times, EpCAM, Hoechst Clinically validated in
several cancer types; viable CTCs
Background noise, subjective judgment of CTCs, labor‐intensive; limited markers can be used for a sample
Negative +
flow
cytometry, FACS
CD45, EpCAM, CKs, CD133, CD44, Syto62 High sensitivity for multiple markers, high purity of isolation Controls, cell aggregations, laser compensation, operator‐dependent
Label‐free (size) CTC‐filtering devices; Size, CKs, Her2/neu, ALDH1, CD44, CD24 Rapid processing; multiplexed imaging and genetic analysis Limited by size of CTCs variation
ISET Size, CKs, EGFR, VE‐cadherin, ki67 Rapid processing; non‐antigen dependent; able to isolate CTM; cell illustrated by IHC staining, able to perform FISH, DNA/RNA analysis Size‐dependent (may miss cells less than 8 μm); require more clinical validation trials; manual processing
DEP force Size, surface electricity, viability Rapid processing, can
isolate single cell very precisely
Low throughput; time‐consuming
ODEP force Size, surface electricity, viability Rapid processing, can
isolate single cell very precisely; can differ viable from dead cells
Relatively low throughput
Tracheal
carina‐
inspired bifurcated (TRAB) microfilter system
Size High recovery rate, acceptable purity; viable isolation Require clinical trial validation
Label‐free (gradient) Ficoll + RT‐PCR CK‐19, HER2, h‐MAM, CEA, maspin, GABA A, B726P High sensitivity No morphology confirmation; not really capture CTCs
Ficoll + RT
‐qPCR
CK‐19, BST1, PTPRC High sensitivity; quantification No morphology confirmation; not really capture CTCs
OncoQuick CCNE2, DKFZp762E1312, EMP2 High sensitivity; quantification No morphology confirmation; not really capture CTCs
MAL2, PPIC and
SLC6A8, hMAM, and
EpCAM
High sensitivity; quantification No morphology confirmation; not really capture CTCs

Table 1.

Overview of analytical methodologies for the detection and molecular characterization of CTCs.

Another report addressed the differences between positive and label‐free method was reported by Qin et al. [50]. They designed a micropore filtration platform (using resettable cell traps) to perform CTC isolation by the characteristic of CTCs (size and deformability) from patients with castrate resistant prostate cancer. Compared with CellSearch™, the method could capture CTCs 10 times more than CellSearch™ can achieve. The method was also proven to be able to perform subsequent molecular analyses.

Interestingly, some investigators compared the isolation efficiency of ISET and CellSearch™ systems [76, 93] and one team concluded that a combination of ISET plus CellSearch™ would have better performance in CTC detection in NCSCL patients than ISET or CellSearch™ alone [93].

One question which is often and needed to be asked is that how to choose a best platform for upcoming studies and trials. Before answering the question, the readers/investigators should fully understand the differences, pros and cons among these methods. Then you should choose a platform wisely according to future directions of investigations (i.e., clinical trial, CTC culture, patient‐derived xerograft model from CTCs) (i.e., genetic analysis, single cell, physical properties) and the requirements of the study materials (i.e., cells with high purity, with high cell numbers, viability, expression with specific marker, etc.). Table 1 demonstrated the brief comparison among novel platforms. In our opinion, for genetic analysis and future personalized medicine, we need a large number of CTCs captured for cultivate, whole genome or transcriptome sequencing and avoid sampling errors by hyper‐selection of few cells to represent the whole populations of cancer. Therefore, negative selection as first step is currently most suitable strategy among all the methods.

Advertisement

3. The main elements in negative selection plus microfluidic CTCs isolation

3.1. Immunomagnetic beads‐based methods

The method is in fact derived from conventional cytological diagnostics for bone marrow and hematologic malignancies. However, when investigators attempted to apply this method to CTC filed, they faced a big problem—the CTCs were so rare to identify in thousands of blood smear slides. Therefore, an alternative method was to exam samples after series of centrifugation, density separation (i.e., in buffy coat or peripheral blood mononuclear cells, PBMC layer), and red blood cells removal. The vast majority of the following detection techniques of CTCs in these prepared samples has long been based on sensitive immunocytochemical (ICC) analysis using antibodies against different epithelial antigens [29, 31, 239242]. Whether positive selection or negative selection procedures using immunomagnetic beads before ICC analysis are both helpful and critical for efficient CTC identification [29, 31]. Zigeuner et al. [31] found that immunomagnetic cell enrichment significantly improves the sensitivity of detection of CTCs cells added to mononuclear cells compared to immunocytochemistry method.

Although the exact procedures of immunomagnetic beads separation protocol was variable with the beads and antibiotics but they have general principles and we would take the procedures of Dynabeads as an example (modified from Naume et al.’s work in 1997 [29]). The main procedures are preparation of beads, incubation with samples and beads, using a magnetic field or column (depends on chosen systems) for target cells isolation by washing out other cells which did not conjugated with beads. If the target cells are those we do not want to analyze, the procedure is defined to be a negative selection. Conversely, if the cells are the targets in the study, it is a positive selection.

3.1.1. Preparation of the magnetic beads

Rat antimouse (RAM) IgGl‐coated M280 Dynabeads coupled to BerEP4 mAb (Product No. 112.07), M450 Dynabeads coated with an anti‐CD45 mAb that recognizes all isoforms of CD45 (Product No. 111.19), and Neodynium Magnetic Particle Concentrators were sup plied by Dynal (Oslo, Norway). Coating of the M280 Dynabeads with antiepithelial mAb was performed according to the manufacturer’s instructions. Briefly, the RAM M280 Dynabeads were incubated with either BerEP4, 9189, or MOC31 mAbs at a concentration of 1/u.g/107 beads for 30 min at 4°C under gentle rotation, followed by three magnet washes in PBS/0.1% HSA and then stored at 4°C. Before use, the Dynabeads were washed once with separation medium.

3.1.2. Positive immunomagnetic separation technique

A total of 1 × 107 peripheral blood mononuclear cells (PBMNC) were resuspended in cold separation medium to a concentration of 2 ×107 MNC/ml (0.5 ml volume) and incubated with RAM IgGl M280 Dynabeads coated with either BerEP4 317G5, or MOC31 mAb. The bead concentration varied from 2.5 to 40 × 106 b/ml, as described in individual experiments. The bead/cell suspension was incubated under gentle rotation for 30 min at 4°C. The sample was then diluted to 3 ml and placed against a magnet for 7 min to recover the rosetted cells, followed by two additional washes as follows. The supernatant was removed, and the rosetted cells were resuspended in 3 ml separation medium, followed by treatment with the magnet (7 min). To facilitate ICC TC detection, the positive LMS product was finally resuspended to contain 5–7 × 106 beads/ml, and 0.5 ml aliquots were centrifuged onto each cytospin slide for further immunocytochemical analysis.

3.1.3. Negative immunomagnetic separation technique

A total of 1 × 107 peripheral blood mononuclear cells (PBMNC) were resuspended in cold separation medium to a concentration of 2 × 107 MNC/ml (0.5 ml volume) and incubated with anti‐CD45‐conjugated M450 Dynabeads at a bead/cell ratio of 2.5:1, 5:1, or 10:1. The bead/cell suspension was incubated under gentle rotation for 45 min. The solution was then diluted to about 30 ml, and the magnet was applied for 5 min, with initial rotation of the tubes onto the magnet to reduce trapping of tumor cells. The supernatant was collected and centrifuged at 450g for 10 min, counted, and resuspended in 10% FBS in PBS to 1 × 106 cells/ml. Then, cytospins containing 5 × 105 cells were prepared. All the slides were air‐dried overnight and stained by immunocytochemistry.

3.2. Microfluidic‐based methods for the high purity CTC isolation

In novel era of huge advances of microfluidic devices as mentioned in the section of “Label‐free isolation strategy” [45, 46, 56, 60, 173186]. In fact, the vast majority of microfluidic devices were designed based on EpCAM‐ or CK‐identifying mechanism, which is positive selection method. The CTC‐Chip [25, 243], and the herringbone chip [244, 245] have been proven effective to isolate CTCs with both high CTC purity (50–62%) [25, 245] and high recovery rate (90–95%) [244, 245]. There are several microfluidic devices designed to use positive selection strategy for proof‐of‐concept purpose [34, 57, 59, 124, 184, 210, 246269] and for specific cancer in clinical trials, (e.g., breast [270, 271], pancreas [272, 273], ovarian [274], prostate [275], esophageal cancer [270], gastric [271], colorectal cancer [276], cancer of unknown primary [277]) and for mutational analysis [278]. Moreover, combined preparation using positive, negative, or label‐free selection methods with microfluidic devices for better performance is also feasible and have been reported [93].

However, there are several drawbacks or limitations of microfluidic devices reported [267]. First of all, reports in literature, however, have revealed that EpCAM or CKs are not expressed in all cancer cells (e.g., sarcoma, melanoma, or CTCs bearing EMT), and therefore some kinds of CTCs cannot be targeted via the positive selection‐based microfluidic device [279]. Secondly, several microfluidic chips could identify with microtubes or micropoles with or without EpCAM conjugation. It seems to be difficult to release captured CTCs from the chips. The efficiency of identification will not be equal to recovered cells for further molecular or genetic analysis. Thirdly, almost more than 80% of microfluidic devices are still in proof‐of‐concepts phase and comes from a single team or laboratory. It might be because that the advances of new innovation always come up faster than validation reports. However, we do need well‐designed and well‐conducted prospective clinical studies to critically elucidate the clinical impacts of the microfluidic devices. The investigators could consider to learn from the developing history of CellSearch™ system.

3.3. Perspective for future of CTC technology: combinations of several methods

Our perspectives for future CTC isolation is mainly combined methodologies instead of conventional ones based on a single isolation strategy.

Yamamoto et al. [49] displayed a combination of size‐based filtration plus a magnetic column method for CTC isolation. The combined use of the column and filter decreased the required time for the spiked cancer cell capture, and the recovery rate of the spiked cancer cells from blood was significantly higher using the combination process (80.7%) than that using the filter alone (64.7%). Moreover, the recovered CTCs are more abundant by the combination process. Another combination was ISET and CellSearch™ systems [76, 93] and the combination had better performance in CTC detection in non‐small cell lung cancer (NCSCL) patients than ISET or CellSearch™ alone [93]. Furthermore, density separation plus flow cytometry or cell sorting systems have been postulated to be potential tools of CTC isolation and identification considering their high sensitivity and purity since 1998 [27]. This method could be seen as a combination of negative selection strategy and a positive confirmation with surface markers, such as EpCAM or cytokeratins. Later in 2011, leukapheresis plus fluorescence‐activated cell sorting (FACS) elutriation were also reported to be effective for large volume blood process for CTC isolation with molecular analysis [211]. These studies illustrated the possibility and better efficacy the combination can achieve, therefore, in our opinion, to find a suitable combination of CTC isolation protocols considering the balance of efficiency, time, sample and costs is very important in the future CTC field.

Intriguingly, several liquid biopsies, as aforementioned, could be combined to be tested in a single sample and at the same time. To realize the goals and minimize the blood sample required, Chudziak et al. [248] reported a novel device, Parsortix system, could negatively select CTCs and perform cfDNA analysis simultaneously. The system recovered more CTCs than CellSearch™ system in the comparison.

Advertisement

4. The applications of CTC testing in clinical cancer researches

As aforementioned, CTC testing are designed to help the diagnosis, early detection and monitoring for response and disease status of cancer patients. Clinical trials to evaluate and validate are inevitable during the developing of any CTC testing. Here, we introduce several important clinical validated studies for the clinical impacts of CTC testing in different cancer types.

4.1. Breast cancer

One meta‐analysis reported by Liao et al. [107], 14 studies with 2336 patients were enrolled and found that presence of CTCs in peripheral blood was significantly associated with the size of tumor [OR 0.68, 95% confidence interval (CI) (0.54, 0.87), P = 0.002], tumor grade [OR 0.71, 95% CI (0.55, 0.91), P = 0.006], estrogen receptor (ER) status [OR 0.72, 95% CI (0.57, 0.91), P = 0.007], and progesterone receptor (PR) of tumor status [OR 0.78, 95% CI (0.61, 0.98), P = 0.04]. In addition, the presence of CTCs is highly correlated with tumor size, tumor grade, ER, and PR status in patients with breast cancer. Although the analysis did not consider the method of isolation which might be one of the downsides and biases of the analysis, the results suggested a trend of physical (tumor size), functional (tumor grade) and status of drugable targets (ER, PR status), which are very useful clinically.

In Zhao et al. [234] performed a meta‐analysis collecting 24 trials with 4013 breast cancer patients and 1333 controls. Poor overall survival was found to be associated with the positive CTC detection (HR = 3.00 [95% CI 2.29–3.94], P < 0.0001) and recurrence‐free survival as well (HR = 2.67 [95% CI 2.09–3.42], P < 0.0001). CTC‐positive breast cancers were significantly associated with high histological grade (HR = 1.21 [95% CI 1.09–1.35], P < 0.0001), tumor size (>2 cm) (HR = 1.12 [95% CI 1.02–1.22], P = 0.01), and nodal status (≥1) (HR = 1.10 [95% CI 1.00–1.21], P = 0.037). The studies, different to that of Liao et al. [107], mentioned about prognostic values of CTC testing. However, the two reports did not mention about the isolation methods and might neglect the biases from CTC number is highly correlated to the method of isolation.

For the purpose of technical standardization, Janni et al. [235] conducted a pooled analysis of individual data from 3173 patients with nonmetastatic (stages I–III) breast cancer from five breast cancer institutions. The prevalence and numbers of CTCs were assessed at the time of primary diagnosis with the FDA‐cleared CellSearch System. Results confirmed that ≥1 CTC(s) were detected in 20.2% of the patients and CTC‐positive patients had larger tumors, increased lymph node involvement, and a higher histologic tumor grade than did CTC‐negative patients (all P < 0.002). Multivariate Cox regressions confirmed that the presence of CTCs was an independent prognostic factor for disease‐free survival [HR, 1.82; 95% confidence interval (CI), 1.47–2.26], distant disease‐free survival (HR, 1.89; 95% CI, 1.49–2.40), breast cancer‐specific survival (HR, 2.04; 95% CI, 1.52–2.75), and overall survival (HR, 1.97; 95% CI, 1.51–2.59). The study addressed the clinical impacts of CellSearch™ system in breast cancer patients and it has confirmed the positive results from a large pooled database.

For a subset in breast cancers, Rack et al. [91] addressed the role of CTCs isolated by CellSearch™ in a prospective trial enrolling 2026 early average‐to‐high risk breast cancer patients and found an independent prognostic relevance of CTCs both before and after adjuvant chemotherapy. The study successfully proved the prognostic role of CTCs in adjuvant settings. The next direction of future studies should be designed to answer the question that whether if extended adjuvant therapy is needed and whether if the extended therapy did reduce the risk of recurrence or not.

For the role of CTCs in a novel and specific therapy in breast cancer, Paoletti et al. [280] reported that heterogeneous mechanisms of resistance to fulvestrant, including estrogen receptor alpha gene (ESR1) mutation. CTC enumeration, phenotyping, and genotyping might identify patients who would benefit from fulvestrant dose escalation versus switching to alternative therapies. The CTCs could possibly help find the resistance genes during the therapy and warn the clinicians to change therapy in time before the tumor already gets progression.

In triple negative breast cancer (TNBC) who lacks of drugable targets (hormone therapy) in breast cancer, Hall et al. (2015) enrolled 44 TNBC patients using CellSearch™ for CTC testing and found that ≥1 CTC in each sample was identified in 30% of patients completing neoadjuvant chemotherapy (NACT). Multivariate analysis demonstrated that detection of ≥1 CTC predicted decreased RFS (log‐rank P = 0.03, HR 5.25, 95% CI 1.34–20.56) and OS (log‐rank P = 0.03, HR 7.04, 95% CI 1.26–39.35). The results suggested a modification of clinical management for TNBC patients with positive CTC detection after NACT, including extension of NACT or adding another anti‐cancer therapy before tumor recurs.

4.2. Lung cancer

In a meta‐analysis reported in 2013, pooled results from a total of 20 studies, comprising 1576 nonsmall cell lung cancer (NSCLC) patients showed that CTCs were associated with lymph node metastasis (OR = 2.06; 95% CI: 1.18–3.62; Z = 2.20; P = 0.027) and tumor stage (OR = 1.95; 95% CI: 1.08–3.54; Z = 2.53; P = 0.011). CTCs were significantly associated with shorter overall survival (relative risk [RR] = 2.19; 95% CI: 1.53–3.12; Z = 4.32; P < 0.0001) and progression‐free/disease‐free survival (RR = 2.14; 95% CI: 1.36–3.38; Z = 3.28; P < 0.0001) [281]. Another study reported the ability to recurrence prediction after curative surgery is positive [282].

For small cell lung cancer, a relatively aggressive subtype with poor prognosis population, a total of seven papers covering 440 SCLC patients were combined in the final analysis. The meta‐analysis revealed that CTCs were significantly associated with shorter overall survival (HR = 1.9; 95% CI: 1.19–3.04; Z = 2.67; P < 0.0001) and progression‐free survival (HR = 2.6; 95% CI: 1.9–3.54; Z = 6.04; P < 0.0001) [283].

Interestingly, in a molecular era nowadays, cancer therapy often relies on genetic or molecular information from cancer tissues, CTCs as well. Das et al. [105] checked the status of ERCC1 expression on captured and found that low expression of ERCC1 on CTCs correlates with progression‐free survival (PFS) in patients with metastatic NSCLC receiving platinum‐based therapy. ERCC1 expression was conventionally checked on NSCLC cancer tissue to predict the response to platinum therapy, which has been the first line standard chemotherapy in patients without active EGFR mutation responding to tyrosine kinase inhibitors (TKIs). The impacts of the study suggested that analysis of ERCC1 expression on CTCs in lung cancer patients could predict the chemotherapy responses. It is a predictive role could possible direct therapy in the future if the findings were confirmed in another large‐scale phase III clinical trials. In addition. Yanagita et al. [284] evaluated CTCs and cfDNA in EGFR‐mutant NSCLC patients treated with erlotinib until progression. Among the enrolled 60 patients, rebiopsy was performed in 35/44 patients (80%), with paired CTC/cfDNA analysis in 41/44 samples at baseline and 36/44 samples at progression. T790M was identified in 23/35 (66%) of tissue biopsies and 9/39 (23%) of cfDNA samples. At diagnosis, high levels of cfDNA but not high levels of CTCs correlated with progression‐free survival. Therefore, cfDNA and CTCs are complementary, noninvasive assays for evaluation of acquired resistance to first‐line EGFR TKIs. Recently, ALK rearrangement on CTCs are successfully performed and compared with cancer tissues [51, 285]. Chromosome instability and ROS‐1 rearrangement on CTCs were also proved to be successful [51]. Immune cells analysis, tumor‐associated macrophages (TAMs) accompanied with CTCs analysis were also proven to be possible and CTCs are competent to specifically manipulate TAMs to increase cancer invasiveness, angiogenesis, immunosuppression and possibly lipid catabolism in lung cancer patients [286]. These studies pointed to the driven mutation detection and would directly benefit to NSCLC patients under targeted therapies.

4.3. Gastrointestinal tract cancer

In 2014, a meta‐analysis comprised 26 studies with peripheral blood samples of 1950 cases for final analysis. The pooled results showed that gastric cancer (GC) patients with detectable CTCs (including circulating miRNAs) had a tendency to experience shortened RFS (HR = 2.91, 95% CI [1.84–4.61], I2 = 52.18%). As for patient deaths, we found a similar association of CTC (including circulating miRNAs) presence with worse OS (HR = 1.78, 95% CI [1.49–2.12], I2 = 30.71%, n = 30). Additionally, subgroup analyses indicated strong prognostic powers of CTCs, irrespective of geographical, methodological, detection time and sample size differences of the studies [287]. In addition, the role of EMT status on CTCs correlates with poor treatment outcomes was also revealed. CTCs expressing CD44 were also found to be prognostic and indicated to malignant behaviors of gastric cancer [288].

For pancreatic cancer, a prospective study addressing the role of CTCs, CTMs in 63 pancreatic ductal adenocarcinoma (PDAC) patients before treatment using anti‐EpCAM (epithelial cell adhesion molecule)‐conjugated supported lipid bilayer‐coated microfluidic chips. CTM was an independent prognostic factor of overall survival (OS) and progression free survival (PFS). Patients were stratified into unfavorable and favorable CTM groups on the basis of CTM more or less than 30 per 2 ml blood, respectively. Patients with baseline unfavorable CTM, compared with patients with favorable CTM, had shorter PFS (2.7 versus 12.1 months; P < 0.0001) and OS (6.4 versus 19.8 months; P < 0.0001). Differences persisted if we stratified patients into early and advanced diseases. The number of CTM before treatment was an independent predictor of PFS and OS after adjustment for clinically significant factors. Therefore, in conclusion, the number of CTM, instead of CTCs, before treatment is an independent predictor of PFS and OS in patients with PDAC [272].

In molecular analysis to predict treatment response, Abdallah et al. [152] found that thymidylate synthase expression in circulating tumor cells can be useful tool as a 5‐FU resistance predictor biomarker in patients with colorectal cancer while other studies elucidate the prognostic and predictive roles of CTCs [198, 289291]. Recently, KRAS and BRAF were successfully detected on CTCs by high‐resolution melt (HRM) and allele‐specific PCR (ASPCR) and KRAS‐codon 12/13‐ and BRAF‐codon 600‐specific assays. Comparing tumor tissues and CTCs mutation status using HRM, Mohamed Suhaimi et al. [292] reported that a 84.1% concordance in KRAS genotype (P = 0.000129) and a 90.9% (P = 0.174) concordance in BRAF genotype. Another report utilized ISET system plus PCR for KRAS codons 12 and 13 mutation with a 71% concordance between cancer tissue and CTCs from colorectal cancer patients [293]. In gastrointestinal stroma tumor, Li et al. [294] conducted a trial to elucidate the role of CTCs expressing ANO1(DOG1) in GIST. ANO1s were more frequently detected in unresectable patients. Tumor size, mitotic count, and risk level were associated with ANO1 detection in resectable GIST patients. The presence of ANO1 significantly correlated with poor disease‐free survival (15.3 versus 19.6 months, P = 0.038). Most patients turned ANO1‐negative after surgery and inversely, all 21 patients with recurrence turned ANO1‐positive with high ANO1 expression levels. Moreover, in the neoadjuvant setting, decline of ANO1 expression level correlated with the response of imatinib. In the near future, these results would possibly promote the genetic analysis on mutation‐driven cancer therapies although they have not become routine screen tools in CRC patients to date.

4.4. Head and neck cancer

Grobe et al. [92] used CellSearch™ for CTC isolation in 80 oral cavity cancer patients and found that 12.5% patients harbored CTCs in peripheral blood, whereas in 20.0% patients DTCs in bone marrow could be detected. Significant correlations could be found for CTCs and tumor size (P = 0.04), nodal status and DTCs (P = 0.02), and distant metastasis with CTCs (P = 0.004) and DTCs (P = 0.005). Univariate and multivariate analyses revealed that CTCs and DTCs were significant and independent predictors of recurrence‐free survival (P < 0.001) as well as in other findings in HNSCC, including the ability of prediction 6‐month death [231]. In 2015, Oliveira‐Costa et al. reported that immunohistochemistry was performed in cancer tissues and in CTCs by immunofluorescence and Nanostring. Correlation was shown between PD‐L1 and tumor size and lymph node metastasis, HOXB9 and tumor size, BLNK and perineural invasion, and between ZNF813 and perineural invasion. PD‐L1 positivity was an independent prognostic factor in this cohort (P = 0.044, HH = 0.426) in OSCC patients [295]. The results could possibly apply to current immune‐oncology studies.

Wu et al. [296] reported a meta‐analysis conducted a computerized retrieval of literatures. Twenty‐two retrieved studies were eligible for systematic review, of which nine conformed for the diagnostic test meta‐analysis and five for the prognostic analysis. Subgroup analysis showed 24.6% pooled sensitivity and 100% pooled specificity of detections by using positive selection strategy, which moreover presented low heterogeneity. The presence of CTC was significantly associated with shorter disease free survival (DFS, HR 4.62, 95% CI 2.51–8.52). The presence of CTC indicates a worse DFS.

4.5. Liver cancer

Early in 2004, Vona et al. have reported that the presence (P = 0.01) and number (P = 0.02) of CTCs and microemboli (CTMs) were significantly associated with a shorter survival [161]. Fan et al. [297] reported a meta‐analysis consisting of 23 trials and found that CTC positivity was significantly associated with RFS (HR 3.03, 95% CI: [1.89–4.86]; P < 0.00001) and overall survival (OS) (HR 2.45, 95% CI: [1.73–3.48]; P < 0.00001). CTC positivity were also significantly associated with TNM Stage (RR 1.30, 95% CI: [1.02–1.65]; P = 0.03), Tumor size (RR 1.36, 95% CI: [1.09–1.69]; P = 0.006), Vascular invasion (RR 1.99, 95% CI: [1.43–2.77]; P < 0.0001), Portal vein tumor thrombus (RR 1.73, 95% CI: [1.42–2.11]; P = 0.0001), Serum alpha‐fetoprotein (AFP) level (RR 2.05; P = 0.01) [297]. Sun et al. found that Stem cell‐like phenotypes are observed in EpCAM+ CTCs, and a preoperative CTCs of ≥2 is a novel predictor for tumor recurrence in hepatocellular carcinoma (HCC) patients after surgery, especially in patient subgroups with AFP levels of ≤400 ng/ml or low tumor recurrence risk. EpCAM+ CTCs could serve as a real‐time parameter for monitoring treatment response and a therapeutic target in HCC recurrence [137]. The prognostic value of overall survival of CTCs in HCC patients has been also revealed [298].

4.6. Genitourinary tract cancer

Rink et al. (2012) found that using CellSearch™, CTC were detected in 23 of 100 patients (23%) with nonmetastatic urothelial carcinoma of urinary bladder. CTC‐positive patients had significantly higher risks of disease recurrence and cancer‐specific and overall mortality (P values ≤ 0.001). After adjusting for effects of standard clinicopathologic features, CTC positivity remained an independent predictor for all end points (hazard ratios: 4.6, 5.2, and 3.5, respectively; P values ≤ 0.003). HER2 positivity was found in 3 of 22 patients (14%). There was concordance between CTC, primary tumors, and lymph node metastases in all CTC‐positive cases (100%).

4.7. Skin cancer and melanoma

Conventionally, melanoma cells lack of cytokeratin or EpCAM expression and CTCs by definition are very difficult to identify. However, investigators broke through the strait by combination with CTCs plus cfDNA. Salvianti et al. [299] enrolled 84 melanoma patients and 68 healthy controls for CTC and cell‐free DNA (cfDNA) testing to assess the diagnostic performance of a tumor‐related methylated cfDNA marker in melanoma patients and to compare this parameter with the presence of CTCs. The percentage of cases with methylated RASSF1A promoter in cfDNA was significantly higher in each class of melanoma patients (in situ, invasive and metastatic) than in healthy subjects (P < 0.001). The concentration of RASSF1A methylated cfDNA in the subjects with a detectable quantity of methylated alleles was significantly higher in melanoma patients than in controls. When the CTCs plus RASSF1A cfDNA are jointly considered, a higher sensitivity of the detection of positive cases in invasive and metastatic melanomas could be obtained. A similar finding was obtained to suggest combine cfDNA (GNAQ/GNA11 mutations) and CTCs to identify uveal melanoma patients with poor prognosis [300]. In another reports, a phase III trial of adjuvant immunotherapy after complete resection of stage IV melanoma, quantitative real‐time reverse‐transcriptase polymerase chain reaction (qPCR) for expression of CTC‐specific MART‐1, MAGE‐A3, and PAX3 mRNA biomarkers were found to be not associated with known prognostic factors or treatment arm. In multivariate analysis, pretreatment CTC (>0 versus 0 biomarker) status was significantly associated with disease‐free survival (DFS; HR 1.64, P = 0.002) and overall survival (OS; HR 1.53, P = 0.028). Serial CTC (>0 versus 0 biomarker) status was also significantly associated with DFS (HR 1.91, P = 0.02) and OS (HR 2.57, P = 0.012) [301]. The report suggested CTCT could be a new risk factor other than any conventional known factors, which might change the staging systems if the evidence gets solid and validated.

4.8. Other cancers

For ovarian cancer, Romero‐Laorden et al. [302] performed a meta‐analysis enrolling 14 studies. Results showed the presence of CTCs and DTCs is associated with adverse clinicopathological characteristics and poor clinical outcomes in ovarian cancer patients. They noticed that different CTC number obtained by different devices could not be compared. Using size‐based isolation strategy (MetaCell®) in 118 ovarian cancer patients, CTCs might have add‐on values on current staging system and the cells could be cultivated after isolation [303, 304]. Furthermore, in another meta‐analysis, eight studies of 1184 ovarian cancer patients were included in the final analysis. In the PB group, it showed that patients with positive CTCs had significantly shorter overall survival (OS) and disease‐free survival (DFS) than patients with negative CTCs (HR, 2.09; CI, 1.13–3.88 and HR, 1.72; CI, 1.32–2.25, respectively). The same result was shown with DTCs in the BM group (HR, 1.61; CI, 1.27–2.04 and HR, 1.44; CI, 1.15–1.80, respectively) [305].

For carcinoma of unknown primary (CUP), Matthew et al. [306] used a real‐time, single‐cell multiplex immunophenotyping of CTCs to inform diagnosis of tissue of origin in CUP patients. CellSearch™ plus multiplexed Q‐dot or DyLight conjugated antibodies were used for cytokeratin 7 (CK7), cytokeratin 20 (CK20), thyroid transcription factor 1 (TTF‐1), estrogen receptor (ER), or prostate‐specific antigen (PSA) expression. The feasibility of staining multiple markers in CTCs presented in this work suggested CTCs could possibly have a non‐inferior role as that of cancer tissues in diagnostics.

Advertisement

5. Unanswered questions in the field of CTCs, technically and clinically

The unanswered question is that the optimal protocol or device has not been found or validated. Many investigators have clearly realized that the number of CTCs cannot easily compare with that counted by another system, but some are not. Recently, Fina et al. [307] compared two CTC isolation methods in a clinical trial. AdnaTest EMT‐1/ and EMT‐2/Stem CellSelect/Detect kits, and ScreenCell Cyto devices were both performed for all samples. Higher CTC detection rates were obtained with the AdnaTest approach when using for CTC‐enrichment antibodies against ERBB2 and EGFR in addition to MUC1 and the classical epithelial surface marker EPCAM (13% versus 48%). When the physical properties of tumor cells were exploited, CTCs were detected at higher percentages than with positive‐selection‐based methods. The results supported that different approaches for CTC detection probably identify distinct tumor cell subpopulations. Technical standardization before clinical validity would be the most urgent issue we have to solve.

Advertisement

6. Concluding remarks

We suggest the investigators to combine different isolation methods to achieve the most optimal performance of CTC isolation and clinical trials for solid validation. The cooperation among medical oncologists and biomedical engineers are critically important for the future advances in CTC field. Genetic or molecular analysis, such as PCR for epigenetics or mutation of specific gene(s) or next‐generation sequencing for whole genome, whole exon, or chosen targeted genes will be the major directions for personalized cancer therapies. The advances of microfluidic devices will quickly solve the conventional problems of time‐consuming, sample‐consuming, operator‐dependent, and marker‐dependent limitations.

References

  1. 1. G.P. Gupta, J. Massague, Cancer metastasis: building a framework, Cell, 127 (2006) 679–695.
  2. 2. T.R. Ashworth, A case of cancer in which cells similar to those in the tumours were seen in the blood after death, Aust Med J, 14 (1989) 146–147
  3. 3. E. Goldmann, Relation of cancer cells to blood vessels andducts, Lancet, 1 (1906) 23.
  4. 4. R.A. Sellwood, S.W. Kuper, J.I. Burn, E.N. Wallace, Circulating cancer cells, Br Med J, 1 (1964) 1683–1686.
  5. 5. W.R. McCune, E.P. Galleher, C. Wood, Circulating “cancer cells”, JAMA, 189 (1964) 852.
  6. 6. S. Roberts, A. Watne, G.R. Mc, G.E. Mc, W.H. Cole, Technique and results of isolation of cancer cells from the circulating blood, AMA Arch Surg, 76 (1958) 334–346.
  7. 7. H. Marcus, Krebszellen im strömenden Blut (Cancerous cells in the circulating blood.), Z Krebsforsch, 16 (1919) 217.
  8. 8. H.C. Engell, Cancer cells in the circulating blood; a clinical study on the occurrence of cancer cells in the peripheral blood and in venous blood draining the tumour area at operation, Acta Chir Scand Suppl, 201 (1955) 1–70.
  9. 9. T.M. Scheinin, A.P. Koivuniemi, Large benign cells in circulating blood and their significance in the identification of cancer cells, Cancer, 15 (1962) 972–977.
  10. 10. S. Roberts, O.L.L. Jonasson, G.R. Mc, G.E. Mc, W.H. Cole, Clinical significance of cancer cells in the circulating blood: two‐ to five‐year survival, Ann Surg, 154 (1961) 362–371.
  11. 11. E.H. Pool, G.R. Dunlop, Cancer cells in the blood stream, Am J Cancer, 21 (1934) 99–102.
  12. 12. S. Warren, O. Gates, The fate of intravenously injected tumor cells, Am J Cancer, 27 (1936) 485–492.
  13. 13. J. Ewing, Neoplastic diseases, 4th ed. Philadelphia: W.B. Saunders, 1940.
  14. 14. O. Saphir, The fate of carcinoma emboli in the lung, Am J Pathol 23 (1947) 245–253.
  15. 15. D. Billadeau, L. Quam, W. Thomas, N. Kay, P. Greipp, R. Kyle, M.M. Oken, B. Van Ness, Detection and quantitation of malignant cells in the peripheral blood of multiple myeloma patients, Blood, 80 (1992) 1818–1824.
  16. 16. K. Pittman, S. Burchill, B. Smith, J. Southgate, J. Joffe, M. Gore, P. Selby, Reverse transcriptase‐polymerase chain reaction for expression of tyrosinase to identify malignant melanoma cells in peripheral blood, Ann Oncol, 7 (1996) 297–301.
  17. 17. F.W. Cremer, K. Kiel, C. Sucker, J. Wacker, A. Atzberger, R. Haas, H. Goldschmidt, M. Moos, A rationale for positive selection of peripheral blood stem cells in multiple myeloma: highly purified CD34+ cell fractions of leukapheresis products do not contain malignant cells, Leukemia, 11 (Suppl 5) (1997) S41–S46.
  18. 18. M. Kawakami, T. Okaneya, K. Furihata, O. Nishizawa, T. Katsuyama, Detection of prostate cancer cells circulating in peripheral blood by reverse transcription‐PCR for hKLK2, Cancer Res, 57 (1997) 4167–4170.
  19. 19. M. Probst‐Kepper, A. Schrader, J. Buer, J. Grosse, M. Volkenandt, H.J. Illiger, B. Metzner, J. Kadar, S. Duensing, B. Hertenstein, A. Ganser, J. Atzpodien, Detection of melanoma cells in peripheral blood stem cell harvests of patients with progressive metastatic malignant melanoma, Br J Haematol, 98 (1997) 488–490.
  20. 20. K. Peck, Y.P. Sher, J.Y. Shih, S.R. Roffler, C.W. Wu, P.C. Yang, Detection and quantitation of circulating cancer cells in the peripheral blood of lung cancer patients, Cancer Res, 58 (1998) 2761–2765.
  21. 21. J.E. Hardingham, D. Kotasek, R.E. Sage, M.C. Eaton, V.H. Pascoe, A. Dobrovic, Detection of circulating tumor cells in colorectal cancer by immunobead‐PCR is a sensitive prognostic marker for relapse of disease, Mol Med, 1 (1995) 789–794.
  22. 22. J.E. Hardingham, D. Kotasek, B. Farmer, R.N. Butler, J.X. Mi, R.E. Sage, A. Dobrovic, Immunobead‐PCR: a technique for the detection of circulating tumor cells using immunomagnetic beads and the polymerase chain reaction, Cancer Res, 53 (1993) 3455–3458.
  23. 23. I. Leotsakos, P. Dimopoulos, E. Gkioka, P. Msaouel, A. Nezos, K.G. Stravodimos, M. Koutsilieris, C.A. Constantinides, Detection of circulating tumor cells in bladder cancer using multiplex PCR assays, Anticancer Res, 34 (2014) 7415–7424.
  24. 24. W.J. Allard, J. Matera, M.C. Miller, M. Repollet, M.C. Connelly, C. Rao, A.G. Tibbe, J.W. Uhr, L.W. Terstappen, Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases, Clin Cancer Res, 10 (2004) 6897–6904.
  25. 25. S. Nagrath, L.V. Sequist, S. Maheswaran, D.W. Bell, D. Irimia, L. Ulkus, M.R. Smith, E.L. Kwak, S. Digumarthy, A. Muzikansky, P. Ryan, U.J. Balis, R.G. Tompkins, D.A. Haber, M. Toner, Isolation of rare circulating tumour cells in cancer patients by microchip technology, Nature, 450 (2007) 1235–1239.
  26. 26. I. Van der Auwera, D. Peeters, I.H. Benoy, H.J. Elst, S.J. Van Laere, A. Prove, H. Maes, P. Huget, P. van Dam, P.B. Vermeulen, L.Y. Dirix, Circulating tumour cell detection: a direct comparison between the CellSearch System, the AdnaTest and CK‐19/mammaglobin RT‐PCR in patients with metastatic breast cancer, Br J Cancer, 102 (2010) 276–284.
  27. 27. E. Racila, D. Euhus, A.J. Weiss, C. Rao, J. McConnell, L.W. Terstappen, J.W. Uhr, Detection and characterization of carcinoma cells in the blood, Proc Natl Acad Sci USA, 95 (1998) 4589–4594.
  28. 28. A.A. Ross, B.W. Cooper, H.M. Lazarus, W. Mackay, T.J. Moss, N. Ciobanu, M.S. Tallman, M.J. Kennedy, N.E. Davidson, D. Sweet, et al., Detection and viability of tumor cells in peripheral blood stem cell collections from breast cancer patients using immunocytochemical and clonogenic assay techniques, Blood, 82 (1993) 2605–2610.
  29. 29. B. Naume, E. Borgen, K. Beiske, T.K. Herstad, G. Ravnas, A. Renolen, S. Trachsel, K. Thrane‐Steen, S. Funderud, G. Kvalheim, Immunomagnetic techniques for the enrichment and detection of isolated breast carcinoma cells in bone marrow and peripheral blood, J Hematother, 6 (1997) 103–114.
  30. 30. U. Bilkenroth, H. Taubert, D. Riemann, U. Rebmann, H. Heynemann, A. Meye, Detection and enrichment of disseminated renal carcinoma cells from peripheral blood by immunomagnetic cell separation, Int J Cancer, 92 (2001) 577–582.
  31. 31. R.E. Zigeuner, R. Riesenberg, H. Pohla, A. Hofstetter, R. Oberneder, Immunomagnetic cell enrichment detects more disseminated cancer cells than immunocytochemistry in vitro, J Urol, 164 (2000) 1834–1837.
  32. 32. Available at: http://www.fda.gov/MedicalDevices/ProductsandMedicalProcedures/DeviceApprovalsandClearances/Recently‐ApprovedDevices/ucm081239.htm. Clearance Date: January 21, 2004 (Approved Evaluation of Automatic Class III Designation).
  33. 33. M. Cristofanilli, Circulating tumor cells, disease progression, and survival in metastatic breast cancer, Semin Oncol, 33 (2006) S9–S14.
  34. 34. J.M. Park, M.S. Kim, H.S. Moon, C.E. Yoo, D. Park, Y.J. Kim, K.Y. Han, J.Y. Lee, J.H. Oh, S.S. Kim, W.Y. Park, W.Y. Lee, N. Huh, Fully automated circulating tumor cell isolation platform with large‐volume capacity based on lab‐on‐a‐disc, Anal Chem, 86 (2014) 3735–3742.
  35. 35. P. Gogoi, S. Sepehri, Y. Zhou, M.A. Gorin, C. Paolillo, E. Capoluongo, K. Gleason, A. Payne, B. Boniface, M. Cristofanilli, T.M. Morgan, P. Fortina, K.J. Pienta, K. Handique, Y. Wang, Development of an automated and sensitive microfluidic device for capturing and characterizing circulating tumor cells (CTCs) from clinical blood samples, PLoS One, 11 (2016) e0147400.
  36. 36. C.M. Svensson, R. Hubler, M.T. Figge, Automated classification of circulating tumor cells and the impact of interobsever variability on classifier training and performance, J Immunol Res, 2015 (2015) 573165.
  37. 37. D.E. Campton, A.B. Ramirez, J.J. Nordberg, N. Drovetto, A.C. Clein, P. Varshavskaya, B.H. Friemel, S. Quarre, A. Breman, M. Dorschner, S. Blau, C.A. Blau, D.E. Sabath, J.L. Stilwell, E.P. Kaldjian, High‐recovery visual identification and single‐cell retrieval of circulating tumor cells for genomic analysis using a dual‐technology platform integrated with automated immunofluorescence staining, BMC Cancer, 15 (2015) 360.
  38. 38. C.M. Svensson, S. Krusekopf, J. Lucke, M. Thilo Figge, Automated detection of circulating tumor cells with naive Bayesian classifiers, Cytometry A, 85 (2014) 501–511.
  39. 39. M. Zhao, P.G. Schiro, J.S. Kuo, K.M. Koehler, D.E. Sabath, V. Popov, Q. Feng, D.T. Chiu, An automated high‐throughput counting method for screening circulating tumor cells in peripheral blood, Anal Chem, 85 (2013) 2465–2471.
  40. 40. T.M. Scholtens, F. Schreuder, S.T. Ligthart, J.F. Swennenhuis, J. Greve, L.W. Terstappen, Automated identification of circulating tumor cells by image cytometry, Cytometry A, 81 (2012) 138–148.
  41. 41. S.M. Leong, K.M. Tan, H.W. Chua, D. Tan, D. Fareda, S. Osmany, M.H. Li, S. Tucker, E.S. Koay, Sampling circulating tumor cells for clinical benefits: how frequent?, J Hematol Oncol, 8 (2015) 75.
  42. 42. D. Marrinucci, K. Bethel, M. Luttgen, R.H. Bruce, J. Nieva, P. Kuhn, Circulating tumor cells from well‐differentiated lung adenocarcinoma retain cytomorphologic features of primary tumor type, Arch Pathol Lab Med, 133 (2009) 1468–1471.
  43. 43. P.R. Gascoyne, S. Shim, Isolation of circulating tumor cells by dielectrophoresis, Cancers (Basel), 6 (2014) 545–579.
  44. 44. S. Shim, K. Stemke‐Hale, A.M. Tsimberidou, J. Noshari, T.E. Anderson, P.R. Gascoyne, Antibody‐independent isolation of circulating tumor cells by continuous‐flow dielectrophoresis, Biomicrofluidics, 7 (2013) 11807.
  45. 45. S.B. Huang, M.H. Wu, Y.H. Lin, C.H. Hsieh, C.L. Yang, H.C. Lin, C.P. Tseng, G.B. Lee, High‐purity and label‐free isolation of circulating tumor cells (CTCs) in a microfluidic platform by using optically‐induced‐dielectrophoretic (ODEP) force, Lab Chip, 13 (2013) 1371–1383.
  46. 46. M.E. Warkiani, B.L. Khoo, L. Wu, A.K. Tay, A.A. Bhagat, J. Han, C.T. Lim, Ultra‐fast, label‐free isolation of circulating tumor cells from blood using spiral microfluidics, Nat Protoc, 11 (2016) 134–148.
  47. 47. H. Li, P. Song, B. Zou, M. Liu, K. Cui, P. Zhou, S. Li, B. Zhang, Circulating tumor cell analyses in patients with esophageal squamous cell carcinoma using epithelial marker‐dependent and ‐independent approaches, Medicine (Baltimore), 94 (2015) e1565.
  48. 48. L. Xu, X. Mao, A. Imrali, F. Syed, K. Mutsvangwa, D. Berney, P. Cathcart, J. Hines, J. Shamash, Y.J. Lu, Optimization and evaluation of a novel size based circulating tumor cell isolation system, PLoS One, 10 (2015) e0138032.
  49. 49. S. Yamamoto, J. Fei, M. Okochi, K. Shimizu, A. Yusa, N. Kondo, H. Iwata, H. Nakanishi, H. Honda, Efficient capturing of circulating tumor cells using a magnetic capture column and a size‐selective filter, Bioprocess Biosyst Eng, 38 (2015) 1693–1704.
  50. 50. X. Qin, S. Park, S.P. Duffy, K. Matthews, R.R. Ang, T. Todenhofer, H. Abdi, A. Azad, J. Bazov, K.N. Chi, P.C. Black, H. Ma, Size and deformability based separation of circulating tumor cells from castrate resistant prostate cancer patients using resettable cell traps, Lab Chip, 15 (2015) 2278–2286.
  51. 51. E. Pailler, N. Auger, C.R. Lindsay, P. Vielh, A. Islas‐Morris‐Hernandez, I. Borget, M. Ngo‐Camus, D. Planchard, J.C. Soria, B. Besse, F. Farace, High level of chromosomal instability in circulating tumor cells of ROS1‐rearranged non‐small‐cell lung cancer, Ann Oncol, 26 (2015) 1408–1415.
  52. 52. J.T. Kaifi, M. Kunkel, A. Das, R.A. Harouaka, D.T. Dicker, G. Li, J. Zhu, G.A. Clawson, Z. Yang, M.F. Reed, N.J. Gusani, E.T. Kimchi, K.F. Staveley‐O’Carroll, S.Y. Zheng, W.S. El‐Deiry, Circulating tumor cell isolation during resection of colorectal cancer lung and liver metastases: a prospective trial with different detection techniques, Cancer Biol Ther, 16 (2015) 699–708.
  53. 53. X. Fan, C. Jia, J. Yang, G. Li, H. Mao, Q. Jin, J. Zhao, A microfluidic chip integrated with a high‐density PDMS‐based microfiltration membrane for rapid isolation and detection of circulating tumor cells, Biosens Bioelectron, 71 (2015) 380–386.
  54. 54. J.F. Chen, H. Ho, J. Lichterman, Y.T. Lu, Y. Zhang, M.A. Garcia, S.F. Chen, A.J. Liang, E. Hodara, H.E. Zhau, S. Hou, R.S. Ahmed, D.J. Luthringer, J. Huang, K.C. Li, L.W. Chung, Z. Ke, H.R. Tseng, E.M. Posadas, Subclassification of prostate cancer circulating tumor cells by nuclear size reveals very small nuclear circulating tumor cells in patients with visceral metastases, Cancer, 121 (2015) 3240–3251.
  55. 55. C.L. Chang, W. Huang, S.I. Jalal, B.D. Chan, A. Mahmood, S. Shahda, B.H. O’Neil, D.E. Matei, C.A. Savran, Circulating tumor cell detection using a parallel flow micro‐aperture chip system, Lab Chip, 15 (2015) 1677–1688.
  56. 56. Y. Tang, J. Shi, S. Li, L. Wang, Y.E. Cayre, Y. Chen, Microfluidic device with integrated microfilter of conical‐shaped holes for high efficiency and high purity capture of circulating tumor cells, Sci Rep, 4 (2014) 6052.
  57. 57. E. Sollier, D.E. Go, J. Che, D.R. Gossett, S. O’Byrne, W.M. Weaver, N. Kummer, M. Rettig, J. Goldman, N. Nickols, S. McCloskey, R.P. Kulkarni, D. Di Carlo, Size‐selective collection of circulating tumor cells using Vortex technology, Lab Chip, 14 (2014) 63–77.
  58. 58. V.K. Liadov, M.A. Skrypnikova, O.P. Popova, Isolation of circulating tumor cells in blood by means of “Isolation by SizE of Tumor cells (ISET)”, Vopr Onkol, 60 (2014) 548–552.
  59. 59. A. Lee, J. Park, M. Lim, V. Sunkara, S.Y. Kim, G.H. Kim, M.H. Kim, Y.K. Cho, All‐in‐one centrifugal microfluidic device for size‐selective circulating tumor cell isolation with high purity, Anal Chem, 86 (2014) 11349–11356.
  60. 60. T. Huang, C.P. Jia, Y. Jun, W.J. Sun, W.T. Wang, H.L. Zhang, H. Cong, F.X. Jing, H.J. Mao, Q.H. Jin, Z. Zhang, Y.J. Chen, G. Li, G.X. Mao, J.L. Zhao, Highly sensitive enumeration of circulating tumor cells in lung cancer patients using a size‐based filtration microfluidic chip, Biosens Bioelectron, 51 (2014) 213–218.
  61. 61. V. Bobek, R. Matkowski, R. Gurlich, K. Grabowski, J. Szelachowska, R. Lischke, J. Schutzner, T. Harustiak, A. Pazdro, A. Rzechonek, K. Kolostova, Cultivation of circulating tumor cells in esophageal cancer, Folia Histochem Cytobiol, 52 (2014) 171–177.
  62. 62. S. Park, R.R. Ang, S.P. Duffy, J. Bazov, K.N. Chi, P.C. Black, H. Ma, Morphological differences between circulating tumor cells from prostate cancer patients and cultured prostate cancer cells, PLoS One, 9 (2014) e85264.
  63. 63. I. Nel, H.A. Baba, J. Ertle, F. Weber, B. Sitek, M. Eisenacher, H.E. Meyer, J.F. Schlaak, A.C. Hoffmann, Individual profiling of circulating tumor cell composition and therapeutic outcome in patients with hepatocellular carcinoma, Transl Oncol, 6 (2013) 420–428.
  64. 64. I. Nel, U. Jehn, T. Gauler, A.C. Hoffmann, Individual profiling of circulating tumor cell composition in patients with non‐small cell lung cancer receiving platinum based treatment, Transl Lung Cancer Res, 3 (2014) 100–106.
  65. 65. E. Hansen, N. Wolff, R. Knuechel, J. Ruschoff, F. Hofstaedter, K. Taeger, Tumor cells in blood shed from the surgical field, Arch Surg, 130 (1995) 387–393.
  66. 66. J.I. Yamashita, Y. Kurusu, N. Fujino, T. Saisyoji, M. Ogawa, Detection of circulating tumor cells in patients with non‐small cell lung cancer undergoing lobectomy by video‐assisted thoracic surgery: a potential hazard for intraoperative hematogenous tumor cell dissemination, J Thorac Cardiovasc Surg, 119 (2000) 899–905.
  67. 67. T. Nakagawa, S.R. Martinez, Y. Goto, K. Koyanagi, M. Kitago, T. Shingai, D.A. Elashoff, X. Ye, F.R. Singer, A.E. Giuliano, D.S. Hoon, Detection of circulating tumor cells in early‐stage breast cancer metastasis to axillary lymph nodes, Clin Cancer Res, 13 (2007) 4105–4110.
  68. 68. M.P. Raynor, S.A. Stephenson, K.B. Pittman, D.C. Walsh, M.A. Henderson, A. Dobrovic, Identification of circulating tumour cells in early stage breast cancer patients using multi marker immunobead RT‐PCR, J Hematol Oncol, 2 (2009) 24.
  69. 69. R.M. Reddy, V. Murlidhar, L. Zhao, S. Grabauskiene, Z. Zhang, N. Ramnath, J. Lin, A.C. Chang, P. Carrott, W. Lynch, M.B. Orringer, D.G. Beer, S. Nagrath, Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early‐stage lung cancer, J Thorac Cardiovasc Surg, 151 (2016) 852–858.
  70. 70. G. Sinha, Circulating tumor cells in early‐stage breast cancer, J Natl Cancer Inst, 104 (2012) 1693–1694.
  71. 71. M. Tewes, S. Kasimir‐Bauer, A. Welt, M. Schuler, R. Kimmig, B. Aktas, Detection of disseminated tumor cells in bone marrow and circulating tumor cells in blood of patients with early‐stage male breast cancer, J Cancer Res Clin Oncol, 141 (2015) 87–92.
  72. 72. F. Miyazono, S. Natsugoe, S. Takao, K. Tokuda, F. Kijima, K. Aridome, S. Hokita, M. Baba, Y. Eizuru, T. Aikou, Surgical maneuvers enhance molecular detection of circulating tumor cells during gastric cancer surgery, Ann Surg, 233 (2001) 189–194.
  73. 73. W.S. Chen, M.Y. Chung, J.H. Liu, J.M. Liu, J.K. Lin, Impact of circulating free tumor cells in the peripheral blood of colorectal cancer patients during laparoscopic surgery, World J Surg, 28 (2004) 552–557.
  74. 74. M. Ilie, V. Hofman, E. Long‐Mira, E. Selva, J.M. Vignaud, B. Padovani, J. Mouroux, C.H. Marquette, P. Hofman, “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease, PLoS One, 9 (2014).
  75. 75. J.M. Hou, M.G. Krebs, L. Lancashire, R. Sloane, A. Backen, R.K. Swain, L.J. Priest, A. Greystoke, C. Zhou, K. Morris, T. Ward, F.H. Blackhall, C. Dive, Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small‐cell lung cancer, J Clin Oncol, 30 (2012) 525–532.
  76. 76. M.G. Krebs, J.M. Hou, R. Sloane, L. Lancashire, L. Priest, D. Nonaka, T.H. Ward, A. Backen, G. Clack, A. Hughes, M. Ranson, F.H. Blackhall, C. Dive, Analysis of circulating tumor cells in patients with non‐small cell lung cancer using epithelial marker‐dependent and ‐independent approaches, J Thorac Oncol, 7 (2012) 306–315.
  77. 77. N. Aceto, A. Bardia, D.T. Miyamoto, M.C. Donaldson, B.S. Wittner, J.A. Spencer, M. Yu, A. Pely, A. Engstrom, H. Zhu, B.W. Brannigan, R. Kapur, S.L. Stott, T. Shioda, S. Ramaswamy, D.T. Ting, C.P. Lin, M. Toner, D.A. Haber, S. Maheswaran, Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis, Cell, 158 (2014) 1110–1122.
  78. 78. G. Kats‐Ugurlu, E. Oosterwijk, S. Muselaers, J. Oosterwijk‐Wakka, C. Hulsbergen‐van de Kaa, M. de Weijert, H. van Krieken, I. Desar, C. van Herpen, C. Maass, R. de Waal, P. Mulders, W. Leenders, Neoadjuvant sorafenib treatment of clear cell renal cell carcinoma and release of circulating tumor fragments, Neoplasia, 16 (2014) 221–228.
  79. 79. L. Lu, H. Zeng, X. Gu, W. Ma, Circulating tumor cell clusters‐associated gene plakoglobin and breast cancer survival, Breast Cancer Res Treat, 151 (2015) 491–500.
  80. 80. B. Molnar, A. Ladanyi, L. Tanko, L. Sreter, Z. Tulassay, Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients, Clin Cancer Res, 7 (2001) 4080–4085.
  81. 81. I.J. Fidler, The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis, Eur J Cancer, 9 (1973) 223–227.
  82. 82. L.A. Liotta, M.G. Saidel, J. Kleinerman, The significance of hematogenous tumor cell clumps in the metastatic process, Cancer Res, 36 (1976) 889–894.
  83. 83. P. Friedl, D. Gilmour, Collective cell migration in morphogenesis, regeneration and cancer, Nat Rev Mol Cell Biol, 10 (2009) 445–457.
  84. 84. O. Ilina, P. Friedl, Mechanisms of collective cell migration at a glance, J Cell Sci, 122 (2009) 3203–3208.
  85. 85. K.J. Cheung, V. Padmanaban, V. Silvestri, K. Schipper, J.D. Cohen, A.N. Fairchild, M.A. Gorin, J.E. Verdone, K.J. Pienta, J.S. Bader, A.J. Ewald, Polyclonal breast cancer metastases arise from collective dissemination of keratin 14‐expressing tumor cell clusters, Proc Natl Acad Sci USA, 113 (2016) E854–E863.
  86. 86. A.F. Sarioglu, N. Aceto, N. Kojic, M.C. Donaldson, M. Zeinali, B. Hamza, A. Engstrom, H. Zhu, T.K. Sundaresan, D.T. Miyamoto, X. Luo, A. Bardia, B.S. Wittner, S. Ramaswamy, T. Shioda, D.T. Ting, S.L. Stott, R. Kapur, S. Maheswaran, D.A. Haber, M. Toner, A microfluidic device for label‐free, physical capture of circulating tumor cell clusters, Nat Methods, 12 (2015) 685–691.
  87. 87. I.J. Fidler, The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited, Nat Rev Cancer, 3 (2003) 453–458.
  88. 88. M. Yu, A. Bardia, B.S. Wittner, S.L. Stott, M.E. Smas, D.T. Ting, S.J. Isakoff, J.C. Ciciliano, M.N. Wells, A.M. Shah, K.F. Concannon, M.C. Donaldson, L.V. Sequist, E. Brachtel, D. Sgroi, J. Baselga, S. Ramaswamy, M. Toner, D.A. Haber, S. Maheswaran, Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition, Science, 339 (2013) 580–584.
  89. 89. D.G. Duda, A.M. Duyverman, M. Kohno, M. Snuderl, E.J. Steller, D. Fukumura, R.K. Jain, Malignant cells facilitate lung metastasis by bringing their own soil, Proc Natl Acad Sci USA, 107 (2010) 21677–21682.
  90. 90. C. Alix‐Panabieres, K. Pantel, Circulating tumor cells: liquid biopsy of cancer, Clin Chem, 59 (2013) 110–118.
  91. 91. B. Rack, C. Schindlbeck, J. Juckstock, U. Andergassen, P. Hepp, T. Zwingers, T.W. Friedl, R. Lorenz, H. Tesch, P.A. Fasching, T. Fehm, A. Schneeweiss, W. Lichtenegger, M.W. Beckmann, K. Friese, K. Pantel, W. Janni, S.S. Group, Circulating tumor cells predict survival in early average‐to‐high risk breast cancer patients, J Natl Cancer Inst, 106 (2014).
  92. 92. A. Grobe, M. Blessmann, H. Hanken, R.E. Friedrich, G. Schon, J. Wikner, K.E. Effenberger, L. Kluwe, M. Heiland, K. Pantel, S. Riethdorf, Prognostic relevance of circulating tumor cells in blood and disseminated tumor cells in bone marrow of patients with squamous cell carcinoma of the oral cavity, Clin Cancer Res, 20 (2014) 425–433.
  93. 93. V. Hofman, M.I. Ilie, E. Long, E. Selva, C. Bonnetaud, T. Molina, N. Venissac, J. Mouroux, P. Vielh, P. Hofman, Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non‐small‐cell lung carcinoma: comparison of the efficacy of the CellSearch Assay and the isolation by size of epithelial tumor cell method, Int J Cancer, 129 (2011) 1651–1660.
  94. 94. C. Ren, P. He, J. Zhang, Z. Zheng, Y. Qiao, X. Zhao, Malignant characteristics of circulating tumor cells and corresponding primary tumor in a patient with esophageal squamous cell carcinoma before and after surgery, Cancer Biol Ther, 11 (2011) 633–638.
  95. 95. M. Pesta, J. Fichtl, V. Kulda, O. Topolcan, V. Treska, Monitoring of circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer, Anticancer Res, 33 (2013) 2239–2243.
  96. 96. E. Magni, E. Botteri, P.S. Ravenda, M.C. Cassatella, E. Bertani, A. Chiappa, F. Luca, L. Zorzino, P.P. Bianchi, L. Adamoli, M.T. Sandri, M.G. Zampino, Detection of circulating tumor cells in patients with locally advanced rectal cancer undergoing neoadjuvant therapy followed by curative surgery, Int J Colorectal Dis, 29 (2014) 1053–1059.
  97. 97. J. Inhestern, K. Oertel, V. Stemmann, H. Schmalenberg, A. Dietz, N. Rotter, J. Veit, M. Gorner, H. Sudhoff, C. Junghanss, C. Wittekindt, K. Pachmann, O. Guntinas‐Lichius, Prognostic role of circulating tumor cells during Induction chemotherapy followed by curative surgery combined with postoperative radiotherapy in patients with locally advanced oral and oropharyngeal squamous cell cancer, PLoS One, 10 (2015) e0132901.
  98. 98. W. Li, X. Zhou, Z. Huang, H. Zhang, L. Zhang, C. Shang, Y. Chen, Laparoscopic surgery minimizes the release of circulating tumor cells compared to open surgery for hepatocellular carcinoma, Surg Endosc, 29 (2015) 3146–3153.
  99. 99. G. van Dalum, G.J. van der Stam, A.G. Tibbe, B. Franken, W.J. Mastboom, I. Vermes, M.R. de Groot, L.W. Terstappen, Circulating tumor cells before and during follow‐up after breast cancer surgery, Int J Oncol, 46 (2015) 407–413.
  100. 100. B. Biggers, S. Knox, M. Grant, J. Kuhn, J. Nemunatitis, T. Fisher, J. Lamont, Circulating tumor cells in patients undergoing surgery for primary breast cancer: preliminary results of a pilot study, Ann Surg Oncol, 16 (2009) 969–971.
  101. 101. L.E. Lowes, M. Lock, G. Rodrigues, D. D’Souza, G. Bauman, B. Ahmad, V. Venkatesan, A.L. Allan, T. Sexton, The significance of circulating tumor cells in prostate cancer patients undergoing adjuvant or salvage radiation therapy, Prostate Cancer Prostatic Dis, 18 (2015) 358–364.
  102. 102. V. Bobek, G. Kacprzak, A. Rzechonek, K. Kolostova, Detection and cultivation of circulating tumor cells in malignant pleural mesothelioma, Anticancer Res, 34 (2014) 2565–2569.
  103. 103. V. Bobek, R. Gurlich, P. Eliasova, K. Kolostova, Circulating tumor cells in pancreatic cancer patients: enrichment and cultivation, World J Gastroenterol, 20 (2014) 17163–17170.
  104. 104. K. Pantel, C. Alix‐Panabieres, Functional studies on viable circulating tumor cells, Clin Chem, 62 (2016) 328–334.
  105. 105. M. Das, J.W. Riess, P. Frankel, E. Schwartz, R. Bennis, H.B. Hsieh, X. Liu, J.C. Ly, L. Zhou, J.J. Nieva, H.A. Wakelee, R.H. Bruce, ERCC1 expression in circulating tumor cells (CTCs) using a novel detection platform correlates with progression‐free survival (PFS) in patients with metastatic non‐small‐cell lung cancer (NSCLC) receiving platinum chemotherapy, Lung Cancer, 77 (2012) 421–426.
  106. 106. M.Y. Kim, T. Oskarsson, S. Acharyya, D.X. Nguyen, X.H. Zhang, L. Norton, J. Massague, Tumor self‐seeding by circulating cancer cells, Cell, 139 (2009) 1315–1326.
  107. 107. Y. Liao, S.Y. Wang, X.Y. Meng, J. Yang, M.J. Shi, H.L. Liu, F.F. Chen, B. Xiong, Circulating tumor cells in breast cancer and its association with tumor clinicopathological characteristics: a meta‐analysis, Med Oncol, 31 (2014) 343.
  108. 108. T.A. Yap, D. Lorente, A. Omlin, D. Olmos, J.S. de Bono, Circulating tumor cells: a multifunctional biomarker, Clin Cancer Res, 20 (2014) 2553–2568.
  109. 109. C. Alix‐Panabieres, K. Bartkowiak, K. Pantel, Functional studies on circulating and disseminated tumor cells in carcinoma patients, Mol Oncol, 10 (2016) 443–449.
  110. 110. B.J. Kirby, M. Jodari, M.S. Loftus, G. Gakhar, E.D. Pratt, C. Chanel‐Vos, J.P. Gleghorn, S.M. Santana, H. Liu, J.P. Smith, V.N. Navarro, S.T. Tagawa, N.H. Bander, D.M. Nanus, P. Giannakakou, Functional characterization of circulating tumor cells with a prostate‐cancer‐specific microfluidic device, PLoS One, 7 (2012) e35976.
  111. 111. B. Paiva, T. Paino, J.M. Sayagues, M. Garayoa, L. San‐Segundo, M. Martin, I. Mota, M.L. Sanchez, P. Barcena, I. Aires‐Mejia, L. Corchete, C. Jimenez, R. Garcia‐Sanz, N.C. Gutierrez, E.M. Ocio, M.V. Mateos, M.B. Vidriales, A. Orfao, J.F. San Miguel, Detailed characterization of multiple myeloma circulating tumor cells shows unique phenotypic, cytogenetic, functional, and circadian distribution profile, Blood, 122 (2013) 3591–3598.
  112. 112. G. Palmieri, M. Strazzullo, P.A. Ascierto, S.M. Satriano, A. Daponte, G. Castello, Polymerase chain reaction‐based detection of circulating melanoma cells as an effective marker of tumor progression. Melanoma Cooperative Group, J Clin Oncol, 17 (1999) 304–311.
  113. 113. I.H. Wong, Transcriptional profiling of circulating tumor cells: quantification and cancer progression (review), Oncol Rep, 10 (2003) 229–235.
  114. 114. M. Cristofanilli, G.T. Budd, M.J. Ellis, A. Stopeck, J. Matera, M.C. Miller, J.M. Reuben, G.V. Doyle, W.J. Allard, L.W. Terstappen, D.F. Hayes, Circulating tumor cells, disease progression, and survival in metastatic breast cancer, N Engl J Med, 351 (2004) 781–791.
  115. 115. T. Bauernhofer, S. Zenahlik, G. Hofmann, M. Balic, M. Resel, R. Pirchmoser, P. Regitnig, P. Ambros, N. Dandachi, H. Samonigg, Association of disease progression and poor overall survival with detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer, Oncol Rep, 13 (2005) 179–184.
  116. 116. D.F. Hayes, M. Cristofanilli, G.T. Budd, M.J. Ellis, A. Stopeck, M.C. Miller, J. Matera, W.J. Allard, G.V. Doyle, L.W. Terstappen, Circulating tumor cells at each follow‐up time point during therapy of metastatic breast cancer patients predict progression‐free and overall survival, Clin Cancer Res, 12 (2006) 4218–4224.
  117. 117. A. Poveda, S.B. Kaye, R. McCormack, S. Wang, T. Parekh, D. Ricci, C.A. Lebedinsky, J.C. Tercero, P. Zintl, B.J. Monk, Circulating tumor cells predict progression free survival and overall survival in patients with relapsed/recurrent advanced ovarian cancer, Gynecol Oncol, 122 (2011) 567–572.
  118. 118. B. Aktas, M. Tewes, T. Fehm, S. Hauch, R. Kimmig, S. Kasimir‐Bauer, Stem cell and epithelial‐mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients, Breast Cancer Res, 11 (2009) R46.
  119. 119. J.P. Thiery, Epithelial‐mesenchymal transitions in tumour progression, Nat Rev Cancer, 2 (2002) 442–454.
  120. 120. C. Raimondi, A. Gradilone, G. Naso, B. Vincenzi, A. Petracca, C. Nicolazzo, A. Palazzo, R. Saltarelli, F. Spremberg, E. Cortesi, P. Gazzaniga, Epithelial‐mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients, Breast Cancer Res Treat, 130 (2011) 449–455.
  121. 121. M. Bourcy, M. Suarez‐Carmona, J. Lambert, M.E. Francart, H. Schroeder, C. Delierneux, N. Skrypek, E.W. Thompson, G. Jerusalem, G. Berx, M. Thiry, S. Blacher, B.G. Hollier, A. Noel, C. Oury, M. Polette, C. Gilles, Tissue factor induced by epithelial‐mesenchymal transition triggers a pro‐coagulant state that drives metastasis of circulating tumor cells, Cancer Res, (2016). DOI: 10.1158/0008-5472.CAN-15-2263.
  122. 122. M. Ksiazkiewicz, A. Markiewicz, A.J. Zaczek, Epithelial‐mesenchymal transition: a hallmark in metastasis formation linking circulating tumor cells and cancer stem cells, Pathobiology, 79 (2012) 195–208.
  123. 123. Y.M. Li, S.C. Xu, J. Li, K.Q. Han, H.F. Pi, L. Zheng, G.H. Zuo, X.B. Huang, H.Y. Li, H.Z. Zhao, Z.P. Yu, Z. Zhou, P. Liang, Epithelial‐mesenchymal transition markers expressed in circulating tumor cells in hepatocellular carcinoma patients with different stages of disease, Cell Death Dis, 4 (2013) e831.
  124. 124. K.A. Hyun, K.B. Goo, H. Han, J. Sohn, W. Choi, S.I. Kim, H.I. Jung, Y.S. Kim, Epithelial‐to‐mesenchymal transition leads to loss of EpCAM and different physical properties in circulating tumor cells from metastatic breast cancer, Oncotarget, 7 (2016) 24677‐24687
  125. 125. G. Barriere, M. Tartary, M. Rigaud, Epithelial mesenchymal transition: a new insight into the detection of circulating tumor cells, ISRN Oncol, 2012 (2012) 382010.
  126. 126. S. Kasimir‐Bauer, O. Hoffmann, D. Wallwiener, R. Kimmig, T. Fehm, Expression of stem cell and epithelial‐mesenchymal transition markers in primary breast cancer patients with circulating tumor cells, Breast Cancer Res, 14 (2012) R15.
  127. 127. Y.K. Liu, B.S. Hu, Z.L. Li, X. He, Y. Li, L.G. Lu, An improved strategy to detect the epithelial‐mesenchymal transition process in circulating tumor cells in hepatocellular carcinoma patients, Hepatol Int, 10 (2016) 640‐646.
  128. 128. S. Wu, S. Liu, Z. Liu, J. Huang, X. Pu, J. Li, D. Yang, H. Deng, N. Yang, J. Xu, Classification of circulating tumor cells by epithelial‐mesenchymal transition markers, PLoS One, 10 (2015) e0123976.
  129. 129. G. Barriere, P. Fici, G. Gallerani, F. Fabbri, W. Zoli, M. Rigaud, Circulating tumor cells and epithelial, mesenchymal and stemness markers: characterization of cell subpopulations, Ann Transl Med, 2 (2014) 109.
  130. 130. C.T. Jordan, M.L. Guzman, M. Noble, Cancer stem cells, N Engl J Med, 355 (2006) 1253–1261.
  131. 131. J.E. Visvader, G.J. Lindeman, Cancer stem cells in solid tumours: accumulating evidence and unresolved questions, Nat Rev Cancer, 8 (2008) 755–768.
  132. 132. T. Dittmar, C. Heyder, E. Gloria‐Maercker, W. Hatzmann, K.S. Zanker, Adhesion molecules and chemokines: the navigation system for circulating tumor (stem) cells to metastasize in an organ‐specific manner, Clin Exp Metastasis, 25 (2008) 11–32.
  133. 133. M. Locke, M. Heywood, S. Fawell, I.C. Mackenzie, Retention of intrinsic stem cell hierarchies in carcinoma‐derived cell lines, Cancer Res, 65 (2005) 8944–8950.
  134. 134. L. Zhu, W. Zhang, J. Wang, R. Liu, Evidence of CD90+CXCR4+ cells as circulating tumor stem cells in hepatocellular carcinoma, Tumour Biol, 36 (2015) 5353–5360.
  135. 135. S. Katoh, T. Goi, T. Naruse, Y. Ueda, H. Kurebayashi, T. Nakazawa, Y. Kimura, Y. Hirono, A. Yamaguchi, Cancer stem cell marker in circulating tumor cells: expression of CD44 variant exon 9 is strongly correlated to treatment refractoriness, recurrence and prognosis of human colorectal cancer, Anticancer Res, 35 (2015) 239–244.
  136. 136. S. Liu, N. Li, X. Yu, X. Xiao, K. Cheng, J. Hu, J. Wang, D. Zhang, S. Cheng, S. Liu, Expression of intercellular adhesion molecule 1 by hepatocellular carcinoma stem cells and circulating tumor cells, Gastroenterology, 144 (2013) 1031–1041 e1010.
  137. 137. Y.F. Sun, Y. Xu, X.R. Yang, W. Guo, X. Zhang, S.J. Qiu, R.Y. Shi, B. Hu, J. Zhou, J. Fan, Circulating stem cell‐like epithelial cell adhesion molecule‐positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection, Hepatology, 57 (2013) 1458–1468.
  138. 138. M.A. Papadaki, G. Kallergi, Z. Zafeiriou, L. Manouras, P.A. Theodoropoulos, D. Mavroudis, V. Georgoulias, S. Agelaki, 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, 14 (2014) 651.
  139. 139. F. Wang, Y.C. Li, L.P. Liu, H.M. Zhang, S. Tong, Circulating Tumor Cells and Tumor Stem Cells Detection in the Peripheral Blood Mononuclear Cells of Breast Cancer, J Clin Lab Anal, (2016). DOI: 10.1002/jcla.21911
  140. 140. I. Tinhofer, M. Saki, F. Niehr, U. Keilholz, V. Budach, Cancer stem cell characteristics of circulating tumor cells, Int J Radiat Biol, 90 (2014) 622–627.
  141. 141. I. Nel, P. David, G.G. Gerken, J.F. Schlaak, A.C. Hoffmann, Role of circulating tumor cells and cancer stem cells in hepatocellular carcinoma, Hepatol Int, 8 (2014) 321–329.
  142. 142. M. Li, B. Zhang, Z. Zhang, X. Liu, X. Qi, J. Zhao, Y. Jiang, H. Zhai, Y. Ji, D. Luo, Stem cell‐like circulating tumor cells indicate poor prognosis in gastric cancer, Biomed Res Int, 2014 (2014) 981261.
  143. 143. N. Krawczyk, F. Meier‐Stiegen, M. Banys, H. Neubauer, E. Ruckhaeberle, T. Fehm, Expression of stem cell and epithelial‐mesenchymal transition markers in circulating tumor cells of breast cancer patients, Biomed Res Int, 2014 (2014) 415721.
  144. 144. G. Pirozzi, V. Tirino, R. Camerlingo, A. La Rocca, N. Martucci, G. Scognamiglio, R. Franco, M. Cantile, N. Normanno, G. Rocco, Prognostic value of cancer stem cells, epithelial‐mesenchymal transition and circulating tumor cells in lung cancer, Oncol Rep, 29 (2013) 1763–1768.
  145. 145. D. Yuan, H. Xia, Y. Zhang, L. Chen, W. Leng, T. Chen, Q. Chen, Q. Tang, X. Mo, M. Liu, F. Bi, P‐Akt/miR200 signaling regulates epithelial‐mesenchymal transition, migration and invasion in circulating gastric tumor cells, Int J Oncol, 45 (2014) 2430–2438.
  146. 146. A. Gradilone, G. Naso, C. Raimondi, E. Cortesi, O. Gandini, B. Vincenzi, R. Saltarelli, E. Chiapparino, F. Spremberg, M. Cristofanilli, L. Frati, A.M. Agliano, P. Gazzaniga, Circulating tumor cells (CTCs) in metastatic breast cancer (MBC): prognosis, drug resistance and phenotypic characterization, Ann Oncol, 22 (2011) 86–92.
  147. 147. P. Gazzaniga, G. Naso, A. Gradilone, E. Cortesi, O. Gandini, W. Gianni, M.A. Fabbri, B. Vincenzi, F. di Silverio, L. Frati, A.M. Agliano, M. Cristofanilli, Chemosensitivity profile assay of circulating cancer cells: prognostic and predictive value in epithelial tumors, Int J Cancer, 126 (2010) 2437–2447.
  148. 148. R. Nadal, F.G. Ortega, M. Salido, J.A. Lorente, M. Rodriguez‐Rivera, M. Delgado‐Rodriguez, M. Macia, A. Fernandez, J.M. Corominas, J.L. Garcia‐Puche, P. Sanchez‐Rovira, F. Sole, M.J. Serrano, CD133 expression in circulating tumor cells from breast cancer patients: potential role in resistance to chemotherapy, Int J Cancer, 133 (2013) 2398–2407.
  149. 149. A. Gradilone, C. Raimondi, G. Naso, I. Silvestri, L. Repetto, A. Palazzo, W. Gianni, L. Frati, E. Cortesi, P. Gazzaniga, How circulating tumor cells escape from multidrug resistance: translating molecular mechanisms in metastatic breast cancer treatment, Am J Clin Oncol, 34 (2011) 625–627.
  150. 150. J.D. Kuhlmann, P. Wimberger, A. Bankfalvi, T. Keller, S. Scholer, B. Aktas, P. Buderath, S. Hauch, F. Otterbach, R. Kimmig, S. Kasimir‐Bauer, ERCC1‐positive circulating tumor cells in the blood of ovarian cancer patients as a predictive biomarker for platinum resistance, Clin Chem, 60 (2014) 1282–1289.
  151. 151. K.H. Yu, M. Ricigliano, M. Hidalgo, G.K. Abou‐Alfa, M.A. Lowery, L.B. Saltz, J.F. Crotty, K. Gary, B. Cooper, R. Lapidus, M. Sadowska, E.M. O’Reilly, Pharmacogenomic modeling of circulating tumor and invasive cells for prediction of chemotherapy response and resistance in pancreatic cancer, Clin Cancer Res, 20 (2014) 5281–5289.
  152. 152. E.A. Abdallah, M.F. Fanelli, M.E. Buim, M.C. Machado Netto, J.L. Gasparini Junior, E.S.V. Souza, A.L. Dettino, N.B. Mingues, J.V. Romero, L.M. Ocea, B.M. Rocha, V.S. Alves, D.V. Araujo, L.T. Chinen, Thymidylate synthase expression in circulating tumor cells: a new tool to predict 5‐fluorouracil resistance in metastatic colorectal cancer patients, Int J Cancer, 137 (2015) 1397–1405.
  153. 153. J.M. Pavese, R.C. Bergan, Circulating tumor cells exhibit a biologically aggressive cancer phenotype accompanied by selective resistance to chemotherapy, Cancer Lett, 352 (2014) 179–186.
  154. 154. S.J. Cohen, C.J. Punt, N. Iannotti, B.H. Saidman, K.D. Sabbath, N.Y. Gabrail, J. Picus, M. Morse, E. Mitchell, M.C. Miller, G.V. Doyle, H. Tissing, L.W. Terstappen, N.J. Meropol, Relationship of circulating tumor cells to tumor response, progression‐free survival, and overall survival in patients with metastatic colorectal cancer, J Clin Oncol, 26 (2008) 3213–3221.
  155. 155. S. Yalcin, S. Kilickap, O. Portakal, C. Arslan, G. Hascelik, T. Kutluk, Determination of circulating tumor cells for detection of colorectal cancer progression or recurrence, Hepatogastroenterology, 57 (2010) 1395–1398.
  156. 156. K. Pachmann, O. Camara, A. Kohlhase, C. Rabenstein, T. Kroll, I.B. Runnebaum, K. Hoeffken, Assessing the efficacy of targeted therapy using circulating epithelial tumor cells (CETC): the example of SERM therapy monitoring as a unique tool to individualize therapy, J Cancer Res Clin Oncol, 137 (2011) 821–828.
  157. 157. I. Desitter, B.S. Guerrouahen, N. Benali‐Furet, J. Wechsler, P.A. Janne, Y. Kuang, M. Yanagita, L. Wang, J.A. Berkowitz, R.J. Distel, Y.E. Cayre, A new device for rapid isolation by size and characterization of rare circulating tumor cells, Anticancer Res, 31 (2011) 427–441.
  158. 158. E.S. Lianidou, A. Markou, Circulating tumor cells in breast cancer: detection systems, molecular characterization, and future challenges, Clin Chem, 57 (2011) 1242–1255.
  159. 159. J.M. Park, J.Y. Lee, J.G. Lee, H. Jeong, J.M. Oh, Y.J. Kim, D. Park, M.S. Kim, H.J. Lee, J.H. Oh, S.S. Lee, W.Y. Lee, N. Huh, Highly efficient assay of circulating tumor cells by selective sedimentation with a density gradient medium and microfiltration from whole blood, Anal Chem, 84 (2012) 7400–7407.
  160. 160. R. Harouaka, Z. Kang, S.Y. Zheng, L. Cao, Circulating tumor cells: advances in isolation and analysis, and challenges for clinical applications, Pharmacol Ther, 141 (2014) 209–221.
  161. 161. G. Vona, L. Estepa, C. Beroud, D. Damotte, F. Capron, B. Nalpas, A. Mineur, D. Franco, B. Lacour, S. Pol, C. Brechot, P. Paterlini‐Brechot, Impact of cytomorphological detection of circulating tumor cells in patients with liver cancer, Hepatology, 39 (2004) 792–797.
  162. 162. V. De Giorgi, P. Pinzani, F. Salvianti, J. Panelos, M. Paglierani, A. Janowska, M. Grazzini, J. Wechsler, C. Orlando, M. Santucci, T. Lotti, M. Pazzagli, D. Massi, Application of a filtration‐ and isolation‐by‐size technique for the detection of circulating tumor cells in cutaneous melanoma, J Invest Dermatol, 130 (2010) 2440–2447.
  163. 163. P. Pinzani, C. Mazzini, F. Salvianti, D. Massi, R. Grifoni, C. Paoletti, F. Ucci, E. Molinara, C. Orlando, M. Pazzagli, B. Neri, Tyrosinase mRNA levels in the blood of uveal melanoma patients: correlation with the number of circulating tumor cells and tumor progression, Melanoma Res, 20 (2010) 303–310.
  164. 164. C. Mazzini, P. Pinzani, F. Salvianti, C. Scatena, M. Paglierani, F. Ucci, M. Pazzagli, D. Massi, Circulating tumor cells detection and counting in uveal melanomas by a filtration‐based method, Cancers (Basel), 6 (2014) 323–332.
  165. 165. K. Kolostova, R. Matkowski, R. Gurlich, K. Grabowski, K. Soter, R. Lischke, J. Schutzner, V. Bobek, Detection and cultivation of circulating tumor cells in gastric cancer, Cytotechnology, 68 (2015) 1095‐1102.
  166. 166. C.L. Chen, D. Mahalingam, P. Osmulski, R.R. Jadhav, C.M. Wang, R.J. Leach, T.C. Chang, S.D. Weitman, A.P. Kumar, L. Sun, M.E. Gaczynska, I.M. Thompson, T.H. Huang, Single‐cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT‐related genes in metastatic prostate cancer, Prostate, 73 (2013) 813–826.
  167. 167. K. Kolostova, M. Broul, J. Schraml, M. Cegan, R. Matkowski, M. Fiutowski, V. Bobek, Circulating tumor cells in localized prostate cancer: isolation, cultivation in vitro and relationship to T‐stage and Gleason score, Anticancer Res, 34 (2014) 3641–3646.
  168. 168. M. Hosokawa, H. Kenmotsu, Y. Koh, T. Yoshino, T. Yoshikawa, T. Naito, T. Takahashi, H. Murakami, Y. Nakamura, A. Tsuya, T. Shukuya, A. Ono, H. Akamatsu, R. Watanabe, S. Ono, K. Mori, H. Kanbara, K. Yamaguchi, T. Tanaka, T. Matsunaga, N. Yamamoto, Size‐based isolation of circulating tumor cells in lung cancer patients using a microcavity array system, PLoS One, 8 (2013) e67466.
  169. 169. M. Hosokawa, T. Yoshikawa, R. Negishi, T. Yoshino, Y. Koh, H. Kenmotsu, T. Naito, T. Takahashi, N. Yamamoto, Y. Kikuhara, H. Kanbara, T. Tanaka, K. Yamaguchi, T. Matsunaga, Microcavity array system for size‐based enrichment of circulating tumor cells from the blood of patients with small‐cell lung cancer, Anal Chem, 85 (2013) 5692–5698.
  170. 170. A. Fiorelli, M. Accardo, E. Carelli, D. Angioletti, M. Santini, M. Di Domenico, Circulating tumor cells in diagnosing lung cancer: clinical and morphologic analysis, Ann Thorac Surg, 99 (2015) 1899–1905.
  171. 171. L.T. Chinen, C.A. Mello, E.A. Abdallah, L.M. Ocea, M.E. Buim, N.M. Breve, J.L.J. Gasparini, M.F. Fanelli, P. Paterlini‐Brechot, Isolation, detection, and immunomorphological characterization of circulating tumor cells (CTCs) from patients with different types of sarcoma using isolation by size of tumor cells: a window on sarcoma‐cell invasion, Onco Targets Ther, 7 (2014) 1609–1617.
  172. 172. R. Riahi, P. Gogoi, S. Sepehri, Y. Zhou, K. Handique, J. Godsey, Y. Wang, A novel microchannel‐based device to capture and analyze circulating tumor cells (CTCs) of breast cancer, Int J Oncol, 44 (2014) 1870–1878.
  173. 173. M. Hosokawa, T. Hayata, Y. Fukuda, A. Arakaki, T. Yoshino, T. Tanaka, T. Matsunaga, Size‐selective microcavity array for rapid and efficient detection of circulating tumor cells, Anal Chem, 82 (2010) 6629–6635.
  174. 174. H.K. Lin, S. Zheng, A.J. Williams, M. Balic, S. Groshen, H.I. Scher, M. Fleisher, W. Stadler, R.H. Datar, Y.C. Tai, R.J. Cote, Portable filter‐based microdevice for detection and characterization of circulating tumor cells, Clin Cancer Res, 16 (2010) 5011–5018.
  175. 175. S.J. Tan, R.L. Lakshmi, P. Chen, W.T. Lim, L. Yobas, C.T. Lim, Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients, Biosens Bioelectron, 26 (2010) 1701–1705.
  176. 176. L.S. Lim, M. Hu, M.C. Huang, W.C. Cheong, A.T. Gan, X.L. Looi, S.M. Leong, E.S. Koay, M.H. Li, Microsieve lab‐chip device for rapid enumeration and fluorescence in situ hybridization of circulating tumor cells, Lab Chip, 12 (2012) 4388–4396.
  177. 177. H.W. Hou, M.E. Warkiani, B.L. Khoo, Z.R. Li, R.A. Soo, D.S. Tan, W.T. Lim, J. Han, A.A. Bhagat, C.T. Lim, Isolation and retrieval of circulating tumor cells using centrifugal forces, Sci Rep, 3 (2013) 1259.
  178. 178. K.A. Hyun, K. Kwon, H. Han, S.I. Kim, H.I. Jung, Microfluidic flow fractionation device for label‐free isolation of circulating tumor cells (CTCs) from breast cancer patients, Biosens Bioelectron, 40 (2013) 206–212.
  179. 179. M.X. Lin, K.A. Hyun, H.S. Moon, T.S. Sim, J.G. Lee, J.C. Park, S.S. Lee, H.I. Jung, Continuous labeling of circulating tumor cells with microbeads using a vortex micromixer for highly selective isolation, Biosens Bioelectron, 40 (2013) 63–67.
  180. 180. E. Ozkumur, A.M. Shah, J.C. Ciciliano, B.L. Emmink, D.T. Miyamoto, E. Brachtel, M. Yu, P.I. Chen, B. Morgan, J. Trautwein, A. Kimura, S. Sengupta, S.L. Stott, N.M. Karabacak, T.A. Barber, J.R. Walsh, K. Smith, P.S. Spuhler, J.P. Sullivan, R.J. Lee, D.T. Ting, X. Luo, A.T. Shaw, A. Bardia, L.V. Sequist, D.N. Louis, S. Maheswaran, R. Kapur, D.A. Haber, M. Toner, Inertial focusing for tumor antigen‐dependent and ‐independent sorting of rare circulating tumor cells, Sci Transl Med, 5 (2013) 179ra147.
  181. 181. W. Sun, C. Jia, T. Huang, W. Sheng, G. Li, H. Zhang, F. Jing, Q. Jin, J. Zhao, G. Li, Z. Zhang, High‐performance size‐based microdevice for the detection of circulating tumor cells from peripheral blood in rectal cancer patients, PLoS One, 8 (2013) e75865.
  182. 182. T.H. Kim, H.J. Yoon, P. Stella, S. Nagrath, Cascaded spiral microfluidic device for deterministic and high purity continuous separation of circulating tumor cells, Biomicrofluidics, 8 (2014) 064117.
  183. 183. M.E. Warkiani, B.L. Khoo, D.S. Tan, A.A. Bhagat, W.T. Lim, Y.S. Yap, S.C. Lee, R.A. Soo, J. Han, C.T. Lim, An ultra‐high‐throughput spiral microfluidic biochip for the enrichment of circulating tumor cells, Analyst, 139 (2014) 3245–3255.
  184. 184. K.A. Hyun, T.Y. Lee, S.H. Lee, H.I. Jung, Two‐stage microfluidic chip for selective isolation of circulating tumor cells (CTCs), Biosens Bioelectron, 67 (2015) 86–92.
  185. 185. J. Che, V. Yu, M. Dhar, C. Renier, M. Matsumoto, K. Heirich, E.B. Garon, J. Goldman, J. Rao, G.W. Sledge, M.D. Pegram, S. Sheth, S.S. Jeffrey, R.P. Kulkarni, E. Sollier, D. Di Carlo, Classification of large circulating tumor cells isolated with ultra‐high throughput microfluidic vortex technology, Oncotarget, 7 (2016) 12748–12760.
  186. 186. G.E. Hvichia, Z. Parveen, C. Wagner, M. Janning, J. Quidde, A. Stein, V. Muller, S. Loges, R.P. Neves, N.H. Stoecklein, H. Wikman, S. Riethdorf, K. Pantel, T.M. Gorges, A novel microfluidic platform for size and deformability based separation and the subsequent molecular characterization of viable circulating tumor cells, Int J Cancer, 138 (2016) 2894–2904.
  187. 187. M.S. Kim, J. Kim, W. Lee, S.J. Cho, J.M. Oh, J.Y. Lee, S. Baek, Y.J. Kim, T.S. Sim, H.J. Lee, G.E. Jung, S.I. Kim, J.M. Park, J.H. Oh, O. Gurel, S.S. Lee, J.G. Lee, A trachea‐inspired bifurcated microfilter capturing viable circulating tumor cells via altered biophysical properties as measured by atomic force microscopy, Small, 9 (2013) 3103–3110.
  188. 188. J. Wang, W. Lu, C. Tang, Y. Liu, J. Sun, X. Mu, L. Zhang, B. Dai, X. Li, H. Zhuo, X. Jiang, Label‐free isolation and mRNA detection of circulating tumor cells from patients with metastatic lung cancer for disease diagnosis and monitoring therapeutic efficacy, Anal Chem, 87 (2015) 11893–11900.
  189. 189. S. Zheng, H. Lin, J.Q. Liu, M. Balic, R. Datar, R.J. Cote, Y.C. Tai, Membrane microfilter device for selective capture, electrolysis and genomic analysis of human circulating tumor cells, J Chromatogr A, 1162 (2007) 154–161.
  190. 190. W. Xu, L. Cao, L. Chen, J. Li, X.F. Zhang, H.H. Qian, X.Y. Kang, Y. Zhang, J. Liao, L.H. Shi, Y.F. Yang, M.C. Wu, Z.F. Yin, Isolation of circulating tumor cells in patients with hepatocellular carcinoma using a novel cell separation strategy, Clin Cancer Res, 17 (2011) 3783–3793.
  191. 191. F. Ge, H. Zhang, D.D. Wang, L. Li, P.P. Lin, Enhanced detection and comprehensive in situ phenotypic characterization of circulating and disseminated heteroploid epithelial and glioma tumor cells, Oncotarget, 6 (2015) 27049–27064.
  192. 192. S.W. Lee, K.A. Hyun, S.I. Kim, J.Y. Kang, H.I. Jung, Continuous enrichment of circulating tumor cells using a microfluidic lateral flow filtration chip, J Chromatogr A, 1377 (2015) 100–105.
  193. 193. D.J. Gallagher, M.I. Milowsky, N. Ishill, A. Trout, M.G. Boyle, J. Riches, M. Fleisher, D.F. Bajorin, Detection of circulating tumor cells in patients with urothelial cancer, Ann Oncol, 20 (2009) 305–308.
  194. 194. P. Gazzaniga, A. Gradilone, E. de Berardinis, G.M. Busetto, C. Raimondi, O. Gandini, C. Nicolazzo, A. Petracca, B. Vincenzi, A. Farcomeni, V. Gentile, E. Cortesi, L. Frati, Prognostic value of circulating tumor cells in nonmuscle invasive bladder cancer: a CellSearch analysis, Ann Oncol, 23 (2012) 2352–2356.
  195. 195. A. Giordano, M. Giuliano, M. De Laurentiis, G. Arpino, S. Jackson, B.C. Handy, N.T. Ueno, E. Andreopoulou, R.H. Alvarez, V. Valero, S. De Placido, G.N. Hortobagyi, J.M. Reuben, M. Cristofanilli, Circulating tumor cells in immunohistochemical subtypes of metastatic breast cancer: lack of prediction in HER2‐positive disease treated with targeted therapy, Ann Oncol, 23 (2012) 1144–1150.
  196. 196. T.J. Hiltermann, M.M. Pore, A. van den Berg, W. Timens, H.M. Boezen, J.J. Liesker, J.H. Schouwink, W.J. Wijnands, G.S. Kerner, F.A. Kruyt, H. Tissing, A.G. Tibbe, L.W. Terstappen, H.J. Groen, Circulating tumor cells in small‐cell lung cancer: a predictive and prognostic factor, Ann Oncol, 23 (2012) 2937–2942.
  197. 197. M. Mego, U. De Giorgi, L. Hsu, N.T. Ueno, V. Valero, S. Jackson, E. Andreopoulou, S.W. Kau, J.M. Reuben, M. Cristofanilli, Circulating tumor cells in metastatic inflammatory breast cancer, Ann Oncol, 20 (2009) 1824–1828.
  198. 198. M.J. Sotelo, J. Sastre, M.L. Maestro, S. Veganzones, J.M. Vieitez, V. Alonso, C. Gravalos, P. Escudero, R. Vera, E. Aranda, P. Garcia‐Alfonso, J. Gallego‐Plazas, C. Lopez, C. Pericay, A. Arrivi, P. Vicente, P. Ballesteros, E. Elez, A. Lopez‐Ladron, E. Diaz‐Rubio, Role of circulating tumor cells as prognostic marker in resected stage III colorectal cancer, Ann Oncol, 26 (2015) 535–541.
  199. 199. S. Dawood, K. Broglio, V. Valero, J. Reuben, B. Handy, R. Islam, S. Jackson, G.N. Hortobagyi, H. Fritsche, M. Cristofanilli, Circulating tumor cells in metastatic breast cancer: from prognostic stratification to modification of the staging system?, Cancer, 113 (2008) 2422–2430.
  200. 200. W.J. Allard, L.W. Terstappen, CCR 20th anniversary commentary: paving the way for circulating tumor cells, Clin Cancer Res, 21 (2015) 2883–2885.
  201. 201. A. Goldkorn, B. Ely, D.I. Quinn, C.M. Tangen, L.M. Fink, T. Xu, P. Twardowski, P.J. Van Veldhuizen, N. Agarwal, M.A. Carducci, J.P. Monk, 3rd, R.H. Datar, M. Garzotto, P.C. Mack, P. Lara, Jr., C.S. Higano, M. Hussain, I.M. Thompson, Jr., R.J. Cote, N.J. Vogelzang, Circulating tumor cell counts are prognostic of overall survival in SWOG S0421: a phase III trial of docetaxel with or without atrasentan for metastatic castration‐resistant prostate cancer, J Clin Oncol, 32 (2014) 1136–1142.
  202. 202. H.I. Scher, M.J. Morris, E. Basch, G. Heller, End points and outcomes in castration‐resistant prostate cancer: from clinical trials to clinical practice, J Clin Oncol, 29 (2011) 3695–3704.
  203. 203. C.R. Boland, A. Goel, Prognostic subgroups among patients with stage II colon cancer, N Engl J Med, 374 (2016) 277–278.
  204. 204. A.C. Voogd, K. van Gestel, M.F. Ernst, Circulating epithelial cells in breast cancer, N Engl J Med, 351 (2004) 2452–2454; author reply 2452–2454.
  205. 205. Z. Dombovari, B. Molnar, J. Bocsi, I. Lang, K. Papik, J. Feher, Z. Tulassay, Biologic detection methods in the comparison of circulating tumor cells and micrometastases, Orv Hetil, 139 (1998) 1793–1797.
  206. 206. X.C. Hu, Y. Wang, D.R. Shi, T.Y. Loo, L.W. Chow, Immunomagnetic tumor cell enrichment is promising in detecting circulating breast cancer cells, Oncology, 64 (2003) 160–165.
  207. 207. R. Konigsberg, M. Gneist, D. Jahn‐Kuch, G. Pfeiler, G. Hager, M. Hudec, C. Dittrich, R. Zeillinger, Circulating tumor cells in metastatic colorectal cancer: efficacy and feasibility of different enrichment methods, Cancer Lett, 293 (2010) 117–123.
  208. 208. Z.S. Lalmahomed, J. Kraan, J.W. Gratama, B. Mostert, S. Sleijfer, C. Verhoef, Circulating tumor cells and sample size: the more, the better, J Clin Oncol, 28 (2010) e288–289; author reply e290.
  209. 209. T. Yeo, S.J. Tan, C.L. Lim, D.P. Lau, Y.W. Chua, S.S. Krisna, G. Iyer, G.S. Tan, T.K. Lim, D.S. Tan, W.T. Lim, C.T. Lim, Microfluidic enrichment for the single cell analysis of circulating tumor cells, Sci Rep, 6 (2016) 22076.
  210. 210. Y. Deng, Y. Zhang, S. Sun, Z. Wang, M. Wang, B. Yu, D.M. Czajkowsky, B. Liu, Y. Li, W. Wei, Q. Shi, An integrated microfluidic chip system for single‐cell secretion profiling of rare circulating tumor cells, Sci Rep, 4 (2014) 7499.
  211. 211. R.L. Eifler, J. Lind, D. Falkenhagen, V. Weber, M.B. Fischer, R. Zeillinger, Enrichment of circulating tumor cells from a large blood volume using leukapheresis and elutriation: proof of concept, Cytometry B Clin Cytom, 80 (2011) 100–111.
  212. 212. H. Kim, H. Terazono, Y. Nakamura, K. Sakai, A. Hattori, M. Odaka, M. Girault, T. Arao, K. Nishio, Y. Miyagi, K. Yasuda, Development of on‐chip multi‐imaging flow cytometry for identification of imaging biomarkers of clustered circulating tumor cells, PLoS One, 9 (2014) e104372.
  213. 213. L.E. Lowes, D. Goodale, M. Keeney, A.L. Allan, Image cytometry analysis of circulating tumor cells, Methods Cell Biol, 102 (2011) 261–290.
  214. 214. S.K. Lee, G.S. Kim, Y. Wu, D.J. Kim, Y. Lu, M. Kwak, L. Han, J.H. Hyung, J.K. Seol, C. Sander, A. Gonzalez, J. Li, R. Fan, Nanowire substrate‐based laser scanning cytometry for quantitation of circulating tumor cells, Nano Lett, 12 (2012) 2697–2704.
  215. 215. S.I. Han, K.H. Han, Electrical detection method for circulating tumor cells using graphene nanoplates, Anal Chem, 87 (2015) 10585–10592.
  216. 216. D.J. Kim, W.Y. Lee, N.W. Park, G.S. Kim, K.M. Lee, J. Kim, M.K. Choi, G.H. Lee, W. Han, S.K. Lee, Drug response of captured BT20 cells and evaluation of circulating tumor cells on a silicon nanowire platform, Biosens Bioelectron, 67 (2015) 370–378.
  217. 217. Z. Liu, A. Fusi, E. Klopocki, A. Schmittel, I. Tinhofer, A. Nonnenmacher, U. Keilholz, Negative enrichment by immunomagnetic nanobeads for unbiased characterization of circulating tumor cells from peripheral blood of cancer patients, J Transl Med, 9 (2011) 70.
  218. 218. B. Naume, New methods for early detection of breast cancer metastasis, Tidsskr Nor Laegeforen, 118 (1998) 354.
  219. 219. P. Balasubramanian, L. Yang, J.C. Lang, K.R. Jatana, D. Schuller, A. Agrawal, M. Zborowski, J.J. Chalmers, Confocal images of circulating tumor cells obtained using a methodology and technology that removes normal cells, Mol Pharm, 6 (2009) 1402–1408.
  220. 220. C. Alix‐Panabieres, J.P. Vendrell, M. Slijper, O. Pelle, E. Barbotte, G. Mercier, W. Jacot, M. Fabbro, K. Pantel, Full‐length cytokeratin‐19 is released by human tumor cells: a potential role in metastatic progression of breast cancer, Breast Cancer Res, 11 (2009) R39.
  221. 221. E. Deneve, S. Riethdorf, J. Ramos, D. Nocca, A. Coffy, J.P. Daures, T. Maudelonde, J.M. Fabre, K. Pantel, C. Alix‐Panabieres, Capture of viable circulating tumor cells in the liver of colorectal cancer patients, Clin Chem, 59 (2013) 1384–1392.
  222. 222. S. Kruck, G. Gakis, A. Stenzl, Disseminated and circulating tumor cells for monitoring chemotherapy in urological tumors, Anticancer Res, 31 (2011) 2053–2057.
  223. 223. K. Pantel, E. Deneve, D. Nocca, A. Coffy, J.P. Vendrell, T. Maudelonde, S. Riethdorf, C. Alix‐Panabieres, Circulating epithelial cells in patients with benign colon diseases, Clin Chem, 58 (2012) 936–940.
  224. 224. J.M. Ramirez, T. Fehm, M. Orsini, L. Cayrefourcq, T. Maudelonde, K. Pantel, C. Alix‐Panabieres, Prognostic relevance of viable circulating tumor cells detected by EPISPOT in metastatic breast cancer patients, Clin Chem, 60 (2014) 214–221.
  225. 225. M.J. Magbanua, E.V. Sosa, R. Roy, L.E. Eisenbud, J.H. Scott, A. Olshen, D. Pinkel, H.S. Rugo, J.W. Park, Genomic profiling of isolated circulating tumor cells from metastatic breast cancer patients, Cancer Res, 73 (2013) 30–40.
  226. 226. C. Alix‐Panabieres, J.P. Brouillet, M. Fabbro, H. Yssel, T. Rousset, T. Maudelonde, G. Choquet‐Kastylevsky, J.P. Vendrell, Characterization and enumeration of cells secreting tumor markers in the peripheral blood of breast cancer patients, J Immunol Methods, 299 (2005) 177–188.
  227. 227. C. Alix‐Panabieres, S. Riethdorf, K. Pantel, Circulating tumor cells and bone marrow micrometastasis, Clin Cancer Res, 14 (2008) 5013–5021.
  228. 228. S. Wu, Z. Liu, S. Liu, L. Lin, W. Yang, J. Xu, Enrichment and enumeration of circulating tumor cells by efficient depletion of leukocyte fractions, Clin Chem Lab Med, 52 (2014) 243–251.
  229. 229. H.C. Lin, H.C. Hsu, C.H. Hsieh, H.M. Wang, C.Y. Huang, M.H. Wu, C.P. Tseng, A negative selection system PowerMag for effective leukocyte depletion and enhanced detection of EpCAM positive and negative circulating tumor cells, Clin Chim Acta, 419 (2013) 77–84.
  230. 230. H.C. Lin, M.J. Liou, H.L. Hsu, J.C. Hsieh, Y.A. Chen, C.P. Tseng, J.D. Lin, Combined analysis of circulating epithelial cells and serum thyroglobulin for distinguishing disease status of the patients with papillary thyroid carcinoma, Oncotarget, 7 (2016) 17242–17253.
  231. 231. J.C. Hsieh, H.C. Lin, C.Y. Huang, H.L. Hsu, T.M. Wu, C.L. Lee, M.C. Chen, H.M. Wang, C.P. Tseng, Prognostic value of circulating tumor cells with podoplanin expression in patients with locally advanced or metastatic head and neck squamous cell carcinoma, Head Neck, 37 (2015) 1448–1455.
  232. 232. F.R. Li, Q. Li, H.X. Zhou, H. Qi, C.Y. Deng, Detection of circulating tumor cells in breast cancer with a refined immunomagnetic nanoparticle enriched assay and nested‐RT‐PCR, Nanomedicine, 9 (2013) 1106–1113.
  233. 233. S.A. Hosseini, M. Abdolahad, S. Zanganeh, M. Dahmardeh, M. Gharooni, H. Abiri, A. Alikhani, S. Mohajerzadeh, O. Mashinchian, Nanoelectromechanical chip (NELMEC) combination of nanoelectronics and microfluidics to diagnose epithelial and mesenchymal circulating tumor cells from leukocytes, Small, 12 (2016) 883–891.
  234. 234. S. Zhao, Y. Liu, Q. Zhang, H. Li, M. Zhang, W. Ma, W. Zhao, J. Wang, M. Yang, The prognostic role of circulating tumor cells (CTCs) detected by RT‐PCR in breast cancer: a meta‐analysis of published literature, Breast Cancer Res Treat, 130 (2011) 809–816.
  235. 235. W.J. Janni, B. Rack, L.W. Terstappen, J.Y. Pierga, F.A. Taran, T. Fehm, C. Hall, M.R. de Groot, F.C. Bidard, T.W. Friedl, P.A. Fasching, S.Y. Brucker, K. Pantel, A. Lucci, Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer, Clin Cancer Res, 22 (2016) 2583–2593.
  236. 236. Y. Zhou, B. Bian, X. Yuan, G. Xie, Y. Ma, L. Shen, Prognostic value of circulating tumor cells in ovarian cancer: a meta‐analysis, PLoS One, 10 (2015) e0130873.
  237. 237. Q. Lv, L. Gong, T. Zhang, J. Ye, L. Chai, C. Ni, Y. Mao, Prognostic value of circulating tumor cells in metastatic breast cancer: a systemic review and meta‐analysis, Clin Transl Oncol, 18 (2016) 322–330.
  238. 238. R. Konigsberg, E. Obermayr, G. Bises, G. Pfeiler, M. Gneist, F. Wrba, M. de Santis, R. Zeillinger, M. Hudec, C. Dittrich, Detection of EpCAM positive and negative circulating tumor cells in metastatic breast cancer patients, Acta Oncol, 50 (2011) 700–710.
  239. 239. I.J. Diel, M. Kaufmann, R. Goerner, S.D. Costa, S. Kaul, G. Bastert, Detection of tumor cells in bone marrow of patients with primary breast cancer: a prognostic factor for distant metastasis, J Clin Oncol, 10 (1992) 1534–1539.
  240. 240. B. Naume, E. Borgen, G. Kvalheim, R. Karesen, H. Qvist, T. Sauer, T. Kumar, J.M. Nesland, Detection of isolated tumor cells in bone marrow in early‐stage breast carcinoma patients: comparison with preoperative clinical parameters and primary tumor characteristics, Clin Cancer Res, 7 (2001) 4122–4129.
  241. 241. R.J. Cote, P.P. Rosen, M.L. Lesser, L.J. Old, M.P. Osborne, Prediction of early relapse in patients with operable breast cancer by detection of occult bone marrow micrometastases, J Clin Oncol, 9 (1991) 1749–1756.
  242. 242. K. Pantel, G. Schlimok, M. Angstwurm, D. Weckermann, W. Schmaus, H. Gath, B. Passlick, J.R. Izbicki, G. Riethmuller, Methodological analysis of immunocytochemical screening for disseminated epithelial tumor cells in bone marrow, J Hematother, 3 (1994) 165–173.
  243. 243. S. Maheswaran, L.V. Sequist, S. Nagrath, L. Ulkus, B. Brannigan, C.V. Collura, E. Inserra, S. Diederichs, A.J. Iafrate, D.W. Bell, S. Digumarthy, A. Muzikansky, D. Irimia, J. Settleman, R.G. Tompkins, T.J. Lynch, M. Toner, D.A. Haber, Detection of mutations in EGFR in circulating lung‐cancer cells, N Engl J Med, 359 (2008) 366–377.
  244. 244. J.P. Gleghorn, E.D. Pratt, D. Denning, H. Liu, N.H. Bander, S.T. Tagawa, D.M. Nanus, P.A. Giannakakou, B.J. Kirby, Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate‐specific antibody, Lab Chip, 10 (2010) 27–29.
  245. 245. S.L. Stott, C.H. Hsu, D.I. Tsukrov, M. Yu, D.T. Miyamoto, B.A. Waltman, S.M. Rothenberg, A.M. Shah, M.E. Smas, G.K. Korir, F.P. Floyd, Jr., A.J. Gilman, J.B. Lord, D. Winokur, S. Springer, D. Irimia, S. Nagrath, L.V. Sequist, R.J. Lee, K.J. Isselbacher, S. Maheswaran, D.A. Haber, M. Toner, Isolation of circulating tumor cells using a microvortex‐generating herringbone‐chip, Proc Natl Acad Sci USA, 107 (2010) 18392–18397.
  246. 246. S. Wang, A. Thomas, E. Lee, S. Yang, X. Cheng, Y. Liu, Highly efficient and selective isolation of rare tumor cells using a microfluidic chip with wavy‐herringbone micro‐patterned surfaces, Analyst, 141 (2016) 2228–2237.
  247. 247. M. Tang, C.Y. Wen, L.L. Wu, S.L. Hong, J. Hu, C.M. Xu, D.W. Pang, Z.L. Zhang, A chip assisted immunomagnetic separation system for the efficient capture and in situ identification of circulating tumor cells, Lab Chip, 16 (2016) 1214–1223.
  248. 248. J. Chudziak, D.J. Burt, S. Mohan, D.G. Rothwell, B. Mesquita, J. Antonello, S. Dalby, M. Ayub, L. Priest, L. Carter, M.G. Krebs, F. Blackhall, C. Dive, G. Brady, Clinical evaluation of a novel microfluidic device for epitope‐independent enrichment of circulating tumour cells in patients with small cell lung cancer, Analyst, 141 (2016) 669–678.
  249. 249. M. Zhao, B. Wei, W.C. Nelson, P.G. Schiro, D.T. Chiu, Simultaneous and selective isolation of multiple subpopulations of rare cells from peripheral blood using ensemble‐decision aliquot ranking (eDAR), Lab Chip, 15 (2015) 3391–3396.
  250. 250. Y. Yang, H.S. Rho, M. Stevens, A.G. Tibbe, H. Gardeniers, L.W. Terstappen, Microfluidic device for DNA amplification of single cancer cells isolated from whole blood by self‐seeding microwells, Lab Chip, 15 (2015) 4331–4337.
  251. 251. P. Xue, Y. Wu, J. Guo, Y. Kang, Highly efficient capture and harvest of circulating tumor cells on a microfluidic chip integrated with herringbone and micropost arrays, Biomed Microdevices, 17 (2015) 39.
  252. 252. C. Wang, M. Ye, L. Cheng, R. Li, W. Zhu, Z. Shi, C. Fan, J. He, J. Liu, Z. Liu, Simultaneous isolation and detection of circulating tumor cells with a microfluidic silicon‐nanowire‐array integrated with magnetic upconversion nanoprobes, Biomaterials, 54 (2015) 55–62.
  253. 253. C.E. Nwankire, A. Venkatanarayanan, T. Glennon, T.E. Keyes, R.J. Forster, J. Ducree, Label‐free impedance detection of cancer cells from whole blood on an integrated centrifugal microfluidic platform, Biosens Bioelectron, 68 (2015) 382–389.
  254. 254. D. Nieto, R. Couceiro, M. Aymerich, R. Lopez‐Lopez, M. Abal, M.T. Flores‐Arias, A laser‐based technology for fabricating a soda‐lime glass based microfluidic device for circulating tumour cell capture, Colloids Surf B Biointerfaces, 134 (2015) 363–369.
  255. 255. N.G. Maremanda, K. Roy, R.K. Kanwar, V. Shyamsundar, V. Ramshankar, A. Krishnamurthy, S. Krishnakumar, J.R. Kanwar, Quick chip assay using locked nucleic acid modified epithelial cell adhesion molecule and nucleolin aptamers for the capture of circulating tumor cells, Biomicrofluidics, 9 (2015) 054110.
  256. 256. D. Issadore, Point‐of‐care rare cell cancer diagnostics, Methods Mol Biol, 1256 (2015) 123–137.
  257. 257. I. Freitag, C. Matthaus, A. Csaki, J.H. Clement, D. Cialla‐May, K. Weber, C. Krafft, J. Popp, Differentiation of MCF‐7 tumor cells from leukocytes and fibroblast cells using epithelial cell adhesion molecule targeted multicore surface‐enhanced Raman spectroscopy labels, J Biomed Opt, 20 (2015) 55002.
  258. 258. R. Burger, D. Kurzbuch, R. Gorkin, G. Kijanka, M. Glynn, C. McDonagh, J. Ducree, An integrated centrifugo‐opto‐microfluidic platform for arraying, analysis, identification and manipulation of individual cells, Lab Chip, 15 (2015) 378–381.
  259. 259. X. Zheng, L. Jiang, J. Schroeder, A. Stopeck, Y. Zohar, Isolation of viable cancer cells in antibody‐functionalized microfluidic devices, Biomicrofluidics, 8 (2014) 024119.
  260. 260. J.P. Winer‐Jones, B. Vahidi, N. Arquilevich, C. Fang, S. Ferguson, D. Harkins, C. Hill, E. Klem, P.C. Pagano, C. Peasley, J. Romero, R. Shartle, R.C. Vasko, W.M. Strauss, P.W. Dempsey, Circulating tumor cells: clinically relevant molecular access based on a novel CTC flow cell, PLoS One, 9 (2014) e86717.
  261. 261. M. Watanabe, M. Serizawa, T. Sawada, K. Takeda, T. Takahashi, N. Yamamoto, F. Koizumi, Y. Koh, A novel flow cytometry‐based cell capture platform for the detection, capture and molecular characterization of rare tumor cells in blood, J Transl Med, 12 (2014) 143.
  262. 262. F.I. Thege, T.B. Lannin, T.N. Saha, S. Tsai, M.L. Kochman, M.A. Hollingsworth, A.D. Rhim, B.J. Kirby, Microfluidic immunocapture of circulating pancreatic cells using parallel EpCAM and MUC1 capture: characterization, optimization and downstream analysis, Lab Chip, 14 (2014) 1775–1784.
  263. 263. Z. Svobodova, J. Kucerova, J. Autebert, D. Horak, L. Bruckova, J.L. Viovy, Z. Bilkova, Application of an improved magnetic immunosorbent in an Ephesia chip designed for circulating tumor cell capture, Electrophoresis, 35 (2014) 323–329.
  264. 264. Y.J. Kim, G.B. Koo, J.Y. Lee, H.S. Moon, D.G. Kim, D.G. Lee, J.Y. Lee, J.H. Oh, J.M. Park, M.S. Kim, H.G. Woo, S.I. Kim, P. Kang, W. Choi, T.S. Sim, W.Y. Park, J.G. Lee, Y.S. Kim, A microchip filter device incorporating slit arrays and 3‐D flow for detection of circulating tumor cells using CAV1‐EpCAM conjugated microbeads, Biomaterials, 35 (2014) 7501–7510.
  265. 265. B.L. Khoo, M.E. Warkiani, D.S. Tan, A.A. Bhagat, D. Irwin, D.P. Lau, A.S. Lim, K.H. Lim, S.S. Krisna, W.T. Lim, Y.S. Yap, S.C. Lee, R.A. Soo, J. Han, C.T. Lim, Clinical validation of an ultra high‐throughput spiral microfluidics for the detection and enrichment of viable circulating tumor cells, PLoS One, 9 (2014) e99409.
  266. 266. S. Jeon, W. Hong, E.S. Lee, Y. Cho, High‐purity isolation and recovery of circulating tumor cells using conducting polymer‐deposited microfluidic device, Theranostics, 4 (2014) 1123–1132.
  267. 267. K.A. Hyun, H.I. Jung, Advances and critical concerns with the microfluidic enrichments of circulating tumor cells, Lab Chip, 14 (2014) 45–56.
  268. 268. M.L. Hupert, J.M. Jackson, H. Wang, M.A. Witek, J. Kamande, M.I. Milowsky, Y.E. Whang, S.A. Soper, Arrays of high‐aspect ratio microchannels for high‐throughput isolation of circulating tumor cells (CTCs), Microsyst Technol, 20 (2014) 1815–1825.
  269. 269. C. Huang, J.P. Smith, T.N. Saha, A.D. Rhim, B.J. Kirby, Characterization of microfluidic shear‐dependent epithelial cell adhesion molecule immunocapture and enrichment of pancreatic cancer cells from blood cells with dielectrophoresis, Biomicrofluidics, 8 (2014) 044107.
  270. 270. T. Ohnaga, Y. Shimada, K. Takata, T. Obata, T. Okumura, T. Nagata, H. Kishi, A. Muraguchi, K. Tsukada, Capture of esophageal and breast cancer cells with polymeric microfluidic devices for CTC isolation, Mol Clin Oncol, 4 (2016) 599–602.
  271. 271. G. Galletti, M.S. Sung, L.T. Vahdat, M.A. Shah, S.M. Santana, G. Altavilla, B.J. Kirby, P. Giannakakou, Isolation of breast cancer and gastric cancer circulating tumor cells by use of an anti HER2‐based microfluidic device, Lab Chip, 14 (2014) 147–156.
  272. 272. M.C. Chang, Y.T. Chang, J.Y. Chen, Y.M. Jeng, C.Y. Yang, Y.W. Tien, S.H. Yang, H.L. Chen, T.Y. Liang, C.F. Wang, E.Y. Lee, Y.C. Chang, W.H. Lee, Clinical significance of circulating tumor microemboli as a prognostic marker in patients with pancreatic ductal adenocarcinoma, Clin Chem, 62 (2016) 505–513.
  273. 273. R.J. Torphy, C.J. Tignanelli, J.W. Kamande, R.A. Moffitt, S.G. Herrera Loeza, S.A. Soper, J.J. Yeh, Circulating tumor cells as a biomarker of response to treatment in patient‐derived xenograft mouse models of pancreatic adenocarcinoma, PLoS One, 9 (2014) e89474.
  274. 274. Z. Zhao, Y. Yang, Y. Zeng, M. He, A microfluidic ExoSearch chip for multiplexed exosome detection towards blood‐based ovarian cancer diagnosis, Lab Chip, 16 (2016) 489–496.
  275. 275. J. Li, S.G. Gregory, M.A. Garcia‐Blanco, A.J. Armstrong, Using circulating tumor cells to inform on prostate cancer biology and clinical utility, Crit Rev Clin Lab Sci, 52 (2015) 191–210.
  276. 276. J. Coget, F. Borrini, S. Susman, J.C. Sabourin, Colorectal carcinomas in 2013: the search for powerful prognostic markers is still on the go!, Cancer Biomark, 14 (2014) 145–150.
  277. 277. S.H. Lu, W.S. Tsai, Y.H. Chang, T.Y. Chou, S.T. Pang, P.H. Lin, C.M. Tsai, Y.C. Chang, Identifying cancer origin using circulating tumor cells, Cancer Biol Ther, 17 (2016) 430–438.
  278. 278. J. Autebert, B. Coudert, J. Champ, L. Saias, E.T. Guneri, R. Lebofsky, F.C. Bidard, J.Y. Pierga, F. Farace, S. Descroix, L. Malaquin, J.L. Viovy, High purity microfluidic sorting and analysis of circulating tumor cells: towards routine mutation detection, Lab Chip, 15 (2015) 2090–2101.
  279. 279. S.D. Mikolajczyk, L.S. Millar, P. Tsinberg, S.M. Coutts, M. Zomorrodi, T. Pham, F.Z. Bischoff, T.J. Pircher, Detection of EpCAM‐negative and cytokeratin‐negative circulating tumor cells in peripheral blood, J Oncol, 2011 (2011) 252361.
  280. 280. C. Paoletti, J.M. Larios, M.C. Muniz, K. Aung, E.M. Cannell, E.P. Darga, K.M. Kidwell, D.G. Thomas, N. Tokudome, M.E. Brown, M.C. Connelly, D.A. Chianese, A.F. Schott, N.L. Henry, J.M. Rae, D.F. Hayes, Heterogeneous estrogen receptor expression in circulating tumor cells suggests diverse mechanisms of fulvestrant resistance, Mol Oncol, (2016). DOI: 10.1016/j.molonc.2016.04.006
  281. 281. J. Wang, K. Wang, J. Xu, J. Huang, T. Zhang, Prognostic significance of circulating tumor cells in non‐small‐cell lung cancer patients: a meta‐analysis, PLoS One, 8 (2013) e78070.
  282. 282. C. Bayarri‐Lara, F.G. Ortega, A. Cueto Ladron de Guevara, J.L. Puche, J. Ruiz Zafra, D. de Miguel‐Perez, A.S. Ramos, C.F. Giraldo‐Ospina, J.A. Navajas Gomez, M. Delgado‐Rodriguez, J.A. Lorente, M.J. Serrano, Circulating tumor cells identify early recurrence in patients with non‐small cell lung cancer undergoing radical resection, PLoS One, 11 (2016) e0148659.
  283. 283. J. Zhang, H.T. Wang, B.G. Li, Prognostic significance of circulating tumor cells in small‐‐cell lung cancer patients: a meta‐analysis, Asian Pac J Cancer Prev, 15 (2014) 8429–8433.
  284. 284. M. Yanagita, A.J. Redig, C.P. Paweletz, S.E. Dahlberg, A. O’Connell, N. Feeney, M. Taibi, D. Boucher, G.R. Oxnard, B.E. Johnson, D.B. Costa, D.M. Jackman, P.A. Janne, A prospective evaluation of circulating tumor cells and cell‐free DNA in EGFR mutant non‐small cell lung cancer patients treated with erlotinib on a phase II trial, Clin Cancer Res, (2016). DOI: 10.1158/1078-0432.CCR‐16‐0909
  285. 285. E. Pailler, J. Adam, A. Barthelemy, M. Oulhen, N. Auger, A. Valent, I. Borget, D. Planchard, M. Taylor, F. Andre, J.C. Soria, P. Vielh, B. Besse, F. Farace, Detection of circulating tumor cells harboring a unique ALK rearrangement in ALK‐positive non‐small‐cell lung cancer, J Clin Oncol, 31 (2013) 2273–2281.
  286. 286. G. Hamilton, B. Rath, L. Klameth, M.J. Hochmair, Small cell lung cancer: Recruitment of macrophages by circulating tumor cells, Oncoimmunology, 5 (2016) e1093277.
  287. 287. Z.Y. Zhang, Z.L. Dai, X.W. Yin, S.H. Li, S.P. Li, H.Y. Ge, Meta‐analysis shows that circulating tumor cells including circulating microRNAs are useful to predict the survival of patients with gastric cancer, BMC Cancer, 14 (2014) 773.
  288. 288. D. Yuan, L. Chen, M. Li, H. Xia, Y. Zhang, T. Chen, R. Xia, Q. Tang, F. Gao, X. Mo, M. Liu, F. Bi, Isolation and characterization of circulating tumor cells from human gastric cancer patients, J Cancer Res Clin Oncol, 141 (2015) 647–660.
  289. 289. S.J. Cohen, C.J. Punt, N. Iannotti, B.H. Saidman, K.D. Sabbath, N.Y. Gabrail, J. Picus, M.A. Morse, E. Mitchell, M.C. Miller, G.V. Doyle, H. Tissing, L.W. Terstappen, N.J. Meropol, Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer, Ann Oncol, 20 (2009) 1223–1229.
  290. 290. H. Iinuma, T. Watanabe, K. Mimori, M. Adachi, N. Hayashi, J. Tamura, K. Matsuda, R. Fukushima, K. Okinaga, M. Sasako, M. Mori, 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, 29 (2011) 1547–1555.
  291. 291. C. Aggarwal, N.J. Meropol, C.J. Punt, N. Iannotti, B.H. Saidman, K.D. Sabbath, N.Y. Gabrail, J. Picus, M.A. Morse, E. Mitchell, M.C. Miller, S.J. Cohen, Relationship among circulating tumor cells, CEA and overall survival in patients with metastatic colorectal cancer, Ann Oncol, 24 (2013) 420–428.
  292. 292. N.A. Mohamed Suhaimi, Y.M. Foong, D.Y. Lee, W.M. Phyo, I. Cima, E.X. Lee, W.L. Goh, W.Y. Lim, K.S. Chia, S.L. Kong, M. Gong, B. Lim, A.M. Hillmer, P.K. Koh, J.Y. Ying, M.H. Tan, Non‐invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients, Mol Oncol, 9 (2015) 850–860.
  293. 293. M.E. Buim, M.F. Fanelli, V.S. Souza, J. Romero, E.A. Abdallah, C.A. Mello, V. Alves, L.M. Ocea, N.B. Mingues, P.N. Barbosa, C.J. Tyng, R. Chojniak, L.T. Chinen, Detection of KRAS mutations in circulating tumor cells from patients with metastatic colorectal cancer, Cancer Biol Ther, 16 (2015) 1289–1295.
  294. 294. Q. Li, X. Zhi, J. Zhou, R. Tao, J. Zhang, P. Chen, O.D. Roe, L. Sun, L. Ma, Circulating tumor cells as a prognostic and predictive marker in gastrointestinal stromal tumors: a prospective study, Oncotarget, 7 (2016) 36645‐26654.
  295. 295. J.P. Oliveira‐Costa, A.F. de Carvalho, G.G. da Silveira da, P. Amaya, Y. Wu, K.J. Park, M.P. Gigliola, M. Lustberg, M.E. Buim, E.N. Ferreira, L.P. Kowalski, J.J. Chalmers, F.A. Soares, D.M. Carraro, A. Ribeiro‐Silva, Gene expression patterns through oral squamous cell carcinoma development: PD‐L1 expression in primary tumor and circulating tumor cells, Oncotarget, 6 (2015) 20902–20920.
  296. 296. X.L. Wu, Q. Tu, G. Faure, P. Gallet, C. Kohler, C. Bittencourt Mde, Diagnostic and Prognostic Value of Circulating Tumor Cells in Head and Neck Squamous Cell Carcinoma: a systematic review and meta‐analysis, Sci Rep, 6 (2016) 20210.
  297. 297. J.L. Fan, Y.F. Yang, C.H. Yuan, H. Chen, F.B. Wang, Circulating tumor cells for predicting the prognostic of patients with hepatocellular carcinoma: a meta analysis, Cell Physiol Biochem, 37 (2015) 629–640.
  298. 298. K. Schulze, C. Gasch, K. Staufer, B. Nashan, A.W. Lohse, K. Pantel, S. Riethdorf, H. Wege, Presence of EpCAM‐positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma, Int J Cancer, 133 (2013) 2165–2171.
  299. 299. F. Salvianti, C. Orlando, D. Massi, V. De Giorgi, M. Grazzini, M. Pazzagli, P. Pinzani, Tumor‐related methylated cell‐free DNA and circulating tumor cells in melanoma, Front Mol Biosci, 2 (2015) 76.
  300. 300. F.C. Bidard, J. Madic, P. Mariani, S. Piperno‐Neumann, A. Rampanou, V. Servois, N. Cassoux, L. Desjardins, M. Milder, I. Vaucher, J.Y. Pierga, R. Lebofsky, M.H. Stern, O. Lantz, Detection rate and prognostic value of circulating tumor cells and circulating tumor DNA in metastatic uveal melanoma, Int J Cancer, 134 (2014) 1207–1213.
  301. 301. S. Hoshimoto, M.B. Faries, D.L. Morton, T. Shingai, C. Kuo, H.J. Wang, R. Elashoff, N. Mozzillo, M.C. Kelley, J.F. Thompson, J.E. Lee, D.S. Hoon, Assessment of prognostic circulating tumor cells in a phase III trial of adjuvant immunotherapy after complete resection of stage IV melanoma, Ann Surg, 255 (2012) 357–362.
  302. 302. N. Romero‐Laorden, D. Olmos, T. Fehm, J. Garcia‐Donas, I. Diaz‐Padilla, Circulating and disseminated tumor cells in ovarian cancer: a systematic review, Gynecol Oncol, 133 (2014) 632–639.
  303. 303. K. Kolostova, R. Matkowski, M. Jedryka, K. Soter, M. Cegan, M. Pinkas, A. Jakabova, J. Pavlasek, J. Spicka, V. Bobek, The added value of circulating tumor cells examination in ovarian cancer staging, Am J Cancer Res, 5 (2015) 3363–3375.
  304. 304. K. Kolostova, J. Spicka, R. Matkowski, V. Bobek, Isolation, primary culture, morphological and molecular characterization of circulating tumor cells in gynecological cancers, Am J Transl Res, 7 (2015) 1203–1213.
  305. 305. L. Zeng, X. Liang, Q. Liu, Z. Yang, The predictive value of circulating tumor cells in ovarian cancer: a meta analysis, Int J Gynecol Cancer, (2015). DOI: 10.1097/IGC.0000000000000459
  306. 306. E.M. Matthew, L. Zhou, Z. Yang, D.T. Dicker, S.L. Holder, B. Lim, R. Harouaka, S.Y. Zheng, J.J. Drabick, N.E. Lamparella, C.I. Truica, W.S. El‐Deiry, A multiplexed marker‐based algorithm for diagnosis of carcinoma of unknown primary using circulating tumor cells, Oncotarget, 7 (2016) 3662–3676.
  307. 307. E. Fina, C. Reduzzi, R. Motta, S. Di Cosimo, G. Bianchi, A. Martinetti, J. Wechsler, V. Cappelletti, M.G. Daidone, Did circulating tumor cells tell us all they could? The missed circulating tumor cell message in breast cancer, Int J Biol Markers, 30 (2015) e429–433.

Written By

Jason Chia‐Hsun Hsieh and Tyler Ming‐Hsien Wu

Submitted: 16 June 2016 Reviewed: 05 July 2016 Published: 14 September 2016