Open access peer-reviewed chapter

Profiling Circulating Tumour Cells for Clinical Applications

Written By

Kah Yee Goh and Wan-Teck Lim

Submitted: 25 January 2018 Reviewed: 31 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.79228

From the Edited Volume

Liquid Biopsy

Edited by Ilze Strumfa and Janis Gardovskis

Chapter metrics overview

1,215 Chapter Downloads

View Full Metrics

Abstract

Circulating tumour cells (CTCs) refer to cells found in the peripheral blood, which are derived from the primary or secondary tumour. They serve as an alternative to study the biology of the primary tumour especially when tissue biopsy is not available. However, major challenges in CTC analysis are the rarity of these cells and the purity of the isolated population. The advancement in technologies allows detection and enrichment of sufficiently pure CTCs at the single-cell level, facilitating downstream molecular characterisation. Single CTC analysis allows detection of key mutations that may be critical to disease management and helps to address the intercellular differences among tumour cells. In this chapter, we discuss the technologies for CTC isolation and the use of CTCs in achieving early detection and prognosis of cancer, real-time monitoring of cancer therapy and tailoring of personalised treatments.

Keywords

  • cancer
  • CTC
  • single-cell analysis
  • liquid biopsy
  • personalised treatment

1. Introduction

Cancer is a leading cause of death in many countries [1]. According to World Health Organisation, approximately one in six deaths is attributable to cancer. The development of cancer is a multistage process. Briefly, normal cells undergo transformation into tumour cells, which are defined by various hallmarks including the ability to sustain proliferative signals, evade growth suppressors, promote replicative immortality, avoid cell death and immune destruction, induce angiogenesis and activate invasion and metastasis [2]. This cellular transformation results in uncontrolled proliferation and enables tumour cells to migrate from their primary organ to a distant organ, a process known as metastasis. In 90% of cancer patients, death occurs because of metastasis [3, 4].

According to Cancer Research UK, 46% of patients in England are diagnosed in the advance stages of disease, leading to less effective treatment outcomes. To reduce the number of deaths caused by cancer, detection at an early stage of disease development is critical so that clinical intervention can come in place to improve the chances of survival for cancer patients. The diagnosis of cancer involves multiple tests including tissue biopsy, liquid biopsy, imaging scans, genetic tests and an examination of medical history. Tissue biopsy is regarded as the gold standard for the clinical diagnosis of cancer [5, 6], despite the invasiveness and inconvenience of collecting the biopsy sample.

In recent years, liquid biopsy is increasing being used for the detection of cancer because it only requires a routine draw of blood and is less invasive compared to tissue biopsy, which may also not be repeatedly done safely or feasibly [7]. Liquid biopsy includes the analysis of circulating tumour cells (CTCs), circulating cell-free DNA (cfDNA) or exosomes present in the patient’s blood [8]. CTCs, in particular, have garnered much attention for its potential clinical utility. CTCs are cells disseminated from the primary or secondary tumour into the peripheral blood and are associated with the development of metastasis. They are precursors of secondary tumour formation and may carry key information relating to the mechanism of metastasis. They are approximately 12–25 μm [9] and present in extremely low numbers (typically 1–10 CTCs per 10 ml of blood or ~1–100 CTCs per 109 blood cells) [10, 11, 12]. The number of CTCs found in the blood varies with the type and stage of cancer and the treatment provided [13]. Typically, patients in the advanced stages of cancers have higher number of CTCs [14]. CTCs of different cellular morphology may exist in the blood and these include the epithelial CTCs, epithelial-to-mesenchymal (EMT) CTCs and mesenchymal CTCs. Moreover, the mutations found in CTCs are often concordant with the primary tumour [15, 16, 17], suggesting that the genetic composition of CTCs is similar to the primary tumour. Thus, CTCs has the potential to be used as a ‘surrogate’ to study the biology of cancer cells.

In cancer treatment, drug resistance is a major concern. The failure of chemotherapeutic drugs to work in patients lies in the heterogeneity and complexity of cancer cells [18, 19]. Cancer stem cells are resistant to chemotherapy and contribute to the intra-tumoural heterogeneity [20]. Therefore, there is a need for molecular profiling of tumour cells at the single-cell level to better address the intra- and inter-cellular differences in cancer cells and enable clinicians to have a better picture of the disease complexity. While paired tumour/normal tissues is the gold standard for molecular analyses of tumour [21], CTCs may provide information on the dynamic changes in tumour cells when blood is extracted at different times, which cannot be achieved in tissue biopsy. In addition, where tissue is not easily accessible, CTCs may provide a diagnostic window.

However, the main challenges of CTC research are that these cells are extremely rare and the population of isolated CTCs may not be pure due to contamination with white blood cells (WBCs). Therefore, highly sensitive and specific technologies are required to isolate CTCs efficiently. Over the past decade, microfluidics technology has greatly advanced the enrichment and isolation of CTCs from whole blood containing red blood cells (RBCs) and WBCs [22]. Microfluidics deals with the behaviour of fluid passing through the microchannels [23, 24]. It makes use of the laminar flow of fluid in the microchannels to manipulate the fluid to achieve cell separation. Furthermore, because of the small space and short flow distance in the microdevice, microfluidics-based technologies consume small amounts of reagents and greatly increase the speed and throughput of blood sample processing, allowing clinical adoption. The use of microfluidics facilitates the integration of downstream molecular characterisation of CTCs, which will enhance our understanding on the complexity of cancer development and enable clinicians to develop better therapeutic strategies to eradicate cancer cells and improve the overall survival of patients. In this chapter, we discuss the technologies for CTC isolation and the use of single-cell analysis in achieving early detection and prognosis of cancer, real-time monitoring of cancer therapy and tailoring of personalised treatments.

Advertisement

2. Technologies for enrichment and isolation of CTCs

Many technologies have been developed to enrich and isolate CTCs from the peripheral blood. In most methods, CTCs are separated from the blood cells based on their biological properties and/or physical properties such as size, deformability, density and electric charge. Conventional CTC enrichment systems such as fluorescence activated cell sorters (FACs) have been used to separate CTCs from whole blood based on the expression of cell surface protein markers [25]. Technologies that isolate CTCs based on physical properties also exist. For instance, ISET and ScreenCell use a filtration system to separate the slightly larger CTCs (12–25 μm) from the smaller WBCs (7–15 μm) and RBCs (8 μm) [9]. CTCs and mononuclear cells have a density (<1.077 g/ml) lower than other blood cells (>1.077 g/ml), allowing layered separation of CTCs [6]. CTCs and blood cells exhibit differences in deformability, allowing them to be separated [26]. The dielectric properties of CTCs are different from normal blood cells, allowing separation of CTCs when the cells are subjected to a non-uniform electric field [6, 27]. However, these conventional CTC enrichment methods suffer from limited ability to process large volumes of blood, limited detection sensitivity, inherent losses and poor recovery of viable CTCs, low throughput and insufficient purity due to contamination with WBCs [6].

2.1. Use of microfluidics in CTC enrichment

To overcome these limitations, microfluidics technology offers an alternative platform for isolating CTCs with improved detection sensitivity, high recovery rate, high efficiency and throughput. Each microfluidics platform has its advantages and limitations (Table 1). In 2004, US Food and Drug Administration approved the clinical use of CellSearch system (Veridex) for CTC detection in epithelial cancer types such as breast [28], colorectal [29] and prostate cancer [30] for purposes of prognostication. This system uses immunomagnetic and fluorescence imaging technology to enrich and enumerate CTCs from 7.5 ml of whole blood based on the expression of specific proteins in CTCs [31, 32]. CTCs are first separated from other blood cells using magnetic iron nanoparticles coated with antibodies targeted against EpCAM (an epithelial cell adhesion molecule present on the cell surface of CTCs). Subsequently, cells are stained with antibodies targeted against cytokeratin (CK; a protein found in the cytoplasm of CTCs) and CD45 (a cell surface protein found exclusively on WBCs) to differentiate CTCs from contaminating WBCs. DAPI (4′,6-diamidino-2-phenylindole) is also used to stain the nuclei of CTCs and WBCs. Finally, a magnetic field is applied to collect the CTCs, which are identified by positive expression of EpCAM and CK and negative expression of CD45.

System Separation principle Strengths Weaknesses References
Antibody-based capture
CellSearch Positive selection for EpCAM, CK8/CK18/CK19 and negative selection for CD45
  • Food and Drug administration (FDA)-approved for clinical use

  • Unable to capture tumour cells that lack EpCAM expression

  • Only applicable to cancers with epithelial origin

  • Cells are not viable after isolation

[28, 29, 30]
CTC-chip EpCAM-based
  • Sample does not require pre-processing

  • Cells remain intact and viable after isolation

  • Unable to capture tumour cells that lack EpCAM expression

  • Only applicable to cancers with epithelial origin

  • Unable to recover tumour cells with 100% purity

[33]
CTC-ichip Size-based separation followed by negative depletion of white blood cells with CD45 and CD66b magnetic beads
  • Fast processing time (8 ml/h)

  • Has the potential to capture CTCs from any cancer type.

  • Cells remain intact and viable after isolation

  • Unable to capture tumour cells that are smaller or similar in size to blood cells.

  • Unable to recover tumour cells with 100% purity

[51, 52]
IsoFlux EpCAM-based
  • Higher sensitivity of detecting CTCs than CellSearch system

  • Unable to capture tumour cells that lack EpCAM expression

  • Only applicable to cancers with epithelial origin

[53]
Magnetic Sifter EpCAM-based
  • Allows rapid imaging of captured cells on a small area

  • Cells remain intact and viable after isolation

  • Reduces sample losses with minimal pre-processing

  • Unable to capture tumour cells that lack EpCAM expression

  • Only applicable to cancers with epithelial origin

[54]
GEDI microdevice Prostate-specific membrane antigen (PSMA)/HER2-based
  • Higher sensitivity of detecting CTCs than CellSearch system

  • Device geometry reduces capture of WBCs

  • Only applicable to prostate cancer, breast cancer, gastric cancer

  • Unable to recover tumour cells with 100% purity

[43, 55, 56]
Label-free capture
ClearCell FX
(spiral chip)
Size-based
  • Fast processing time (3 ml/h)

  • Cells remain intact and viable after isolation

  • Cost-effective

  • Has the potential to capture CTCs from various cancer types.

  • Unable to capture tumour cells that are smaller or similar in size to blood cells.

  • Unable to recover tumour cells with 100% purity

[11, 40]
Microfluidic biochip Size-based
  • Fast processing time (7.5 ml of blood in 3 h)

  • Allows single-cell isolation

  • Able to isolate viable CTCs with 100% purity

  • Has the potential to capture CTCs from various cancer types.

  • Limited number of cell chambers for imaging

  • Unable to capture tumour cells that are smaller or similar in size to blood cells.

[16, 17]
Vortex Size-based
  • Fast processing time (7.5 ml of blood in 20 min)

  • Cells remain intact and viable after isolation

  • Has the potential to capture CTCs from various cancer types.

  • Unable to recover tumour cells with 100% purity

  • Low CTC capture efficiency

  • Unable to capture tumour cells that are smaller or similar in size to blood cells.

[57]
Microfluidic device for deformability-based cell classification Size and deformability-based
  • Cells remain intact and viable after isolation

  • Cost-effective

  • Has the potential to capture CTCs from various cancer types.

  • Unable to recover tumour cells with 100% purity

[35]
DEPArray Electric charge-based
  • Allows single-cell isolation

  • Able to isolate viable CTCs with 100% purity

  • Has the potential to capture CTCs from various cancer types.

  • Limited throughput

  • Large amount of sample loses

[58, 59]

Table 1.

Comparison of selected microfluidics systems used for enriching CTCs.

Abbreviations used are: CTC, circulating tumour cell; EpCAM, epithelial cell adhesion molecule; CK, cytokeratin proteins.

In 2007, a microfluidics chip developed for CTC enrichment and isolation, known as CTC-chip, was introduced [33]. In this system, CTCs are captured as blood flows through the microchannel containing EpCAM antibody-coated microposts [33]. CTCs are captured with ~50% purity and sample processing takes 1–2 ml/h. Following CTC-chip, a broad range of microfluidic devices were generated to isolate CTCs based on physical size, density, deformability [34, 35, 36, 37, 38, 39, 40, 41] or antibody-mediated CTC capture in surface functionalised microchannels [33, 42, 43, 44, 45, 46, 47].

Although the affinity binding methods (e.g. CellSearch, CTC-chip) may isolate CTCs of better purity than the physical methods, the strong antibody-antigen interaction in the microdevice may affect the recovery of viable CTCs [11, 48] and hinder subsequent downstream analysis. Additionally, affinity binding methods will lose out on the subpopulation of CTCs that have down-regulated expression of epithelial markers (e.g. EpCAM) such as the mesenchymal CTCs, resulting in an underrepresentation of the actual CTC count in the blood [11, 49]. Therefore, the limitations imposed by affinity binding methods prompt the development of label-free microfluidic devices that can isolate CTCs with increased purity and viability.

To efficiently separate CTCs from the large pool of RBCs and WBCs, a spiral microfluidics chip was introduced in 2013 [11, 50]. The spiral microchannel (500 μm width × 160 μm height) consists of two inlets and two outlets over a length of ~10 cm. Blood (diluted 2–2.5×) is pumped into the outer inlet while sheath fluid is pumped through the inner inlet. The additional sheath fluid in the spiral chip facilitates the Dean migration of large volume of RBCs in a well-controlled manner, thus allowing high haematocrit samples (20–25%) to be processed. In spiral microchannel, CTCs and other blood cells experience Dean drag forces in addition to inertial lift forces and the combined effects cause hydrodynamic focusing of the cells to specific region of the microchannel. The larger CTCs are focused near the inner wall of the channel while smaller WBCs and RBCs are focused along the outer wall, leading to efficient size-based isolation of CTCs [50]. Furthermore, the spiral chip is able to recover >85% spiked cancer cells and deplete almost 100% of WBCs from the blood sample [11, 50]. Trypan blue staining showed that most of the recovered cells (>98%) are viable [11]. Therefore, the spiral chip allows continuous isolation of CTCs in a single step with high sensitivity, recovery and throughput (3 ml of blood can be processed in an hour) [11]. The spiral chip is also designed to have large microchannel dimensions and high flow rate to prevent non-specific binding of CTCs to the walls and eliminate any potential clogging issues [11].

2.2. Microfluidics and single CTC isolation

Most microfluidics systems isolate CTCs in bulk rather than individually. The bulk analysis of CTCs may mask the presence of key mutations that are critical to disease progression, indicating the need for single-cell analysis. To isolate and study CTCs at the single-cell level, approaches such as micropipette aspiration [60, 61] and laser microdissection [62] have been used to manually select the individual CTCs. However, these methods are laborious and suffer from low throughput, making them less suitable for clinical use. Commercial platforms such as DEPArray (Silicon Biosystems), which rely on the dielectric properties of CTC for single-cell isolation, suffer from large amount of sample losses [58].

To address these limitations, Yeo et al. developed a microfluidics device capable of performing high throughput, selective isolation of individual viable CTCs with 100% purity amidst a large population of WBCs [17]. The separation efficiency is high because as few as 1 CTC in 20,000 WBCs can be recovered. Prior to starting the run, the cell suspension is stained with specific fluorophore-conjugated antibodies such as CK or CD45 to facilitate the differentiation of CTCs from WBCs. The device works on the principle of hydrodynamic focusing to restrict cells to flow in a single stream and hold them passively in active control cell chambers that are positioned along the outer curvature of the channel. CTCs and other blood cells flowing through the channel will experience a slight centrifugal force that facilitates their entry into the cell chambers. Because the microfluidics biochip is integrated into a microscope, the cell sitting in the chambers can be observed under the microscope to determine whether it is a CTC or WBC based on size or staining outcome. Each chamber holds 1 cell at a time and each is connected to a control line that can be activated to eject the selected cell back into the main channel and into the collection well. In this manner, the cells isolated in the chambers can be ejected sequentially so that each cell can be recovered individually. To maximise the recovery of rare CTCs, the effluent of each run is recycled back into the device for three times. The device is able to process 7.5–8 ml of blood in 3 h [16], facilitating the enumeration of CTCs in clinical blood samples in a short period of time. This method also allows the collection of single CTCs for downstream molecular profiling.

Advertisement

3. Clinical applications of CTCs

Given the convenience and ease of obtaining blood samples from cancer patients, CTCs are currently being adopted for clinical practice. This is especially the case when tissue biopsy samples are not readily available. With current technologies, CTCs can be isolated in bulk or individually for studies on disease progression and therapeutic treatment. Single-cell analysis offers several advantages over pooled cell analysis. First, single-cell analysis is able to detect critical driver mutations for drug response that are present in low frequency, which may be masked in pooled cell analysis since the mutations may only be present in a subset of clones. For instance, in stage IV non-small cell lung cancer (NSCLC) patients who developed resistance after tyrosine kinase inhibitor (TKI) treatment, sequencing of single CTC revealed the presence of epidermal growth factor receptor (EGFR) mutation T790 M that confers resistance in a subset of CTCs isolated from each patient (0–3 CTCs carry the mutation among <10 CTCs isolated per patient) [17]. Determining the mutation status of EGFR in NSCLC patients is important for patient stratification and treatment (see Section 3.4). Second, single-cell analysis reveals the heterogeneity in gene mutations and chromosomal copy number aberrations (CNA) among tumour cells while bulk cell analysis would have average out the signals. In metastatic breast cancer, a sequencing analysis of 40 single CTCs from five patients demonstrated heterogeneous mutations in four genes, phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA), tumour protein P53 (TP53), oestrogen receptor 1 (ESR1) and KRAS [63]. Within the same metastatic breast cancer patient, not all CTCs harbour the particular mutation and different patients harbour different gene mutations [63]. In small cell lung cancer (SCLC) patients, chromosomal CNA profiling of individual CTCs obtained at pre-treatment can predict whether the patient is sensitive or refractory to subsequent chemotherapy at an accuracy of 83.3% [64], facilitating clinical decision-making. Third, single-cell analysis facilitates the study of clonal diversity and mutation evolution over the course of chemotherapy, which is difficult to achieve in bulk cell analysis [65, 66]. In triple negative breast cancer (TNBC) patients, single-cell DNA sequencing revealed that chemoresistant patients carry pre-existing mutations and CNA that were adaptively selected in response to chemotherapy [65]. Single-cell RNA sequencing also found that chemotherapy induces transcriptional reprogramming to favour the resistant phenotype in TNBC patients who develop chemoresistance [65]. Thus, single-cell analysis provides a better resolution of the tumour profile than pooled sample analysis. More importantly, single-cell analysis of CTC can address the heterogeneous profile of cancer cells at the DNA, RNA and protein level and provide insights in the mechanism of metastasis and drug resistance. Here, we discuss the applications of CTCs that aid in the management of cancer and how single CTC analysis can contribute to achieving personalised medicine in the future.

3.1. Biomarker for early detection of cancer

The presence of CTCs in the peripheral blood acts as a biomarker for the early detection of cancer. Thus, the enumeration of CTC is important for cancer screening, especially in patients who at a higher risk of developing cancer due to genetic predisposition or disease state. The effective use of CTC enumeration in the detection of early-stage cancer has been demonstrated in patients with chronic obstructive pulmonary disease (COPD), who are at a high risk of developing lung cancer [5, 67]. Because of the risk, COPD patients are monitored annually for the development of lung cancer using computed tomography (CT) scan. In a subset of COPD patients without cancer diagnosis, CTCs were found in the peripheral blood. After 1–4 years of CTC detection, lung nodules were indeed detected by CT scan, leading to the diagnosis of early-stage lung cancer. Large-scale clinical studies are being done to further validate the use of CTCs as a diagnostic tool for the early detection of cancer [5]. Therefore, CTC can serve as a potential biomarker for the early detection of cancer so that prompt treatment intervention can come in place to improve the overall health of patients.

3.2. Prognostic marker for overall survival and metastasis

The number of CTCs found in the blood can indicate the state of the disease. Patients in later stages of cancer (e.g. metastatic cancer patients) have higher number of CTCs as compared to patients in the early stages of cancer [14, 68, 69]. Furthermore, the number of CTCs can vary with chemotherapy treatment, allowing clinicians to use CTC counts to determine the treatment efficacy and estimate the overall survival and risk of metastatic relapse. A study on early breast cancer patients [70] measured the number of CTCs before and after adjuvant chemotherapy demonstrated that the persistent presence of CTCs after chemotherapy is associated with poor disease-free survival and overall survival. Moreover, prognosis was worst in patients with >5 CTCs per 30 ml of blood [70]. Similarly, studies in lung cancer patients also showed that higher number of CTCs was significantly correlated to shorter survival [32, 71, 72, 73]. Specifically, the presence of >8 CTCs per 7.5 ml of blood after treatment strongly correlated with worse survival in small cell lung cancer patients [74]. In prostate cancer, therapeutic treatment resulting in CTC level dropping from >5 to <5 in 7.5 ml of blood is indicative of better overall survival [55]. Thus, the prognostic cut-off value for CTC is dependent on the type of cancer.

Additionally, CTC enumeration can be used for predicting the risk of metastasis. In non-metastatic colorectal cancer patients, those with >5 CTCs per 2 ml of blood were more likely to develop distant metastasis than those with <5 CTCs [69]. The strong correlation between CTC count and metastasis relapse have been shown in other types of cancers as well including bladder cancer [75, 76], liver cancer [77] and oesophageal cancer [78]. Thus, these patients may benefit more from early treatment.

3.3. Monitoring treatment response and disease progression

Sequential tracking of CTC number during treatment may inform on treatment response and disease progression, providing important information to clinicians on whether the treatment is suitable for the cancer patient. In advanced NSCLC patients, a significant decrease in CTC count after the second cycle of chemotherapy strongly correlated with better overall survival and progression-free survival [32, 79, 80]. However, a lack of decline in the number of CTCs after chemotherapy may suggest that the patient has developed resistance against the specific drug. Therefore, alternative treatment strategies have to be adopted to curb disease progression and improve patient survival. Another potential application may be the detection of early relapse with regular monitoring of the CTC count in post-surgical cancer patients. This may allow early detection and timely clinical intervention to treat the disease when the disease burden is less.

3.4. Identification of therapeutic targets and drug resistance

Given the continuous improvement in microfluidics technology, Khoo et al. demonstrated that patient-derived CTCs could be cultured into CTC clusters in vitro using a microfluidic culture device [81]. Prior enrichment of CTCs and supplements of growth factors are not required, thereby shortening the processing time. Using blood samples from the same patient, CTCs are co-cultured with immune cells in specially formulated microwells to promote the formation of CTC cluster within 2 weeks. The success rate of CTC cluster formation is approximately 50%. With the development of this platform, drug screening can be readily conducted and this can facilitate the discovery and testing of novel drugs that are more efficacious in the treatment of cancer. Furthermore, drug responses can be monitored with varying doses of drug to determine the optimal dose for individual patient.

With the advent of next-generation sequencing, the molecular profile of single CTCs can be obtained at the DNA, RNA and protein level. The genomic profile of single CTCs can be compared to normal or non-malignant cells to identify genes that are differentially expressed, which may mediate the process of metastasis and become potential therapeutic targets. The whole genome sequencing of individual CTCs can reveal genetic alterations such as mutations, copy number variations and single nucleotide polymorphisms that may confer selective advantage to tumour cells [82]. The presence of specific gene mutations in CTCs confers drug resistance and determines the type of treatment to be given to patients. This information is particularly useful when the mutation profile of CTC is concordant with the primary tumour. For example, the CTCs of NSCLC patients harbouring the EGFR T790 M mutation confer resistance to TKI treatment (e.g. gefitinib) [83]. Tracking changes in the CTC count and mutation frequency over the course of treatment allows real-time monitoring of treatment sensitivity and resistance. The early detection of these mutations may provide alternative treatment strategies for NSCLC patients and optimise disease management, leading to improved clinical outcomes [17]. Thus, determining the mutation status of EGFR is crucial since it allows clinicians to select patients who will benefit from TKI treatment. Furthermore, understanding the key mutations behind drug resistance may help to decipher the mechanism and signalling pathways involved in resistance.

The transcriptome of single CTCs also provides information on the identification of therapeutic targets and drug resistance. In prostate cancer, analysis of the mRNA profile in CTCs is required to determine drug sensitivity or resistance. Specifically, the expression of Arv7 mRNA, a truncated form of androgen receptor that remains constitutively active, in CTCs is predictive of anti-androgen therapy failure with enzalutamide and abiraterone [84, 85]. In prostate cancer patients with Arv7 expression, alternative drugs such as taxanes are used for treatment [86, 87, 88, 89]. Additionally, RNA-seq of CTCs can reveal miRNAs that are dysregulated in cancers, making these miRNAs potential targets for cancer therapy.

The expression profile of proteins in single CTCs also plays a role in determining the anti-cancer treatment. For example, oestrogen receptor (ER) is a primary target in the treatment of breast cancer patients. Thus, primary tumours of breast cancer patients are stratified as ER+ or ER- and hormonal therapy is given based on the status of ER expression in the primary tumour. However, breast cancer patients with ER+ primary tumours can harbour ER- CTCs in the blood, which may escape the hormonal therapy [90]. Similarly, metastatic breast cancer patients with HER2+ primary tumours can carry HER2- CTCs in the blood [91, 92, 93]. Because of this discordance, HER2-targeted therapies may only be effective against the primary tumour but not the CTCs. Thus, cancer cells will not be fully eradicated and this may lead to metastatic cancer relapse. In this situation, additional treatment strategies have to be adopted to target the CTCs, on top of the primary tumour. Therefore, the monitoring of genetic aberrations is important in identifying acquired mutations that confer resistance to drug therapy.

The expression status of programmed death ligand 1 (PD-L1) in CTCs aids in identifying the groups of patients who are likely to benefit from the immunotherapy as well as predicting the response to the immunotherapy. Tumour cells express PD-L1 that binds to PD-1 receptor found on the surface of activated T cells and B cells to induce an immunosuppressive effect by reducing cytokine production and immune cells proliferation [94, 95]. It was previously shown that metastatic tumour cells have higher PD-L1 expression than primary tumour cells [96]. In breast cancer, the detection of CTCs expressing PD-L1 indicates that patients carry metastatic cells that have the potential to evade immune destruction [94]. Breast cancer patients with a high frequency of PD-L1(+) CTCs are more likely to benefit from anti-PD-L1 immunotherapy than patients with PD-L1(−) CTCs [94]. In a study on NSCLC patients, PD-L1 expression on CTCs was monitored throughout the course of immunotherapy [97]. After 6 months of therapy, patients with PD-L1 expression in CTCs had poor prognosis while patients without PD-L1 expression in CTCs benefitted from the therapy [97].

Given the intra-tumoural and inter-tumoural heterogeneity and dynamic nature of cancer, single-cell analysis of CTC in circulation may provide information on the evolution of tumour and how they evade drug therapy and immune response (Figure 1). This enables clinicians to have a more holistic view of the disease complexity and more efficient targeting of cancer cells, moving towards the development of personalised therapy for individual patients.

Figure 1.

A proposed scheme of how single-cell analysis of CTCs may address tumour heterogeneity. Heterogeneous tumour cells are colour-coded. Heterogeneity stems from genetic or epigenetic changes that confer selective advantage to the tumour cells. Several strategies of single-cell analysis can be adopted to dissect the heterogeneity of tumour cells. Sequential analyses of individual CTCs aids in the monitoring of therapeutic response, tumour evolution and detection of treatment-resistant cells.

3.5. Comparing CTC with cell-free DNA and exosome

Apart from CTCs, other liquid biopsy markers that confer diagnostic and prognostic relevance include the cell-free DNA (cfDNA) and exosomes. Each marker has its own strengths and weaknesses (Table 2). cfDNA refers to DNA released from necrotic and apoptotic cells and they can be found in blood plasma. Cancer patients usually have higher concentration of cfDNA compared to healthy individuals [98, 99]. Most cfDNAs in the blood plasma are around 70–200 bp long [100, 101]. cfDNA includes the circulating tumour cell DNA (ctDNA), which are released from CTCs. The fraction of ctDNA contributing to cfDNA is small although usually higher in late stage cancer patients (>5–10%) than patients in the early stages of cancer (<1%) [98, 102, 103]. A large fraction of cfDNA comes from non-malignant cells that contain wild type DNA. Because of this issue, highly specific and sensitive technologies are required to isolate cfDNA that originates from tumour cells.

Liquid biopsy Strengths Weaknesses
Circulating tumour cell (CTC)
  • Allows downstream molecular analysis and in vivo or in vitro functional studies

  • Able to evaluate the DNA, RNA and protein profile of tumour cells

  • Able to study the cellular phenotype, morphology and protein localisation

  • Extremely rare and challenging to isolate

  • Heterogeneous population of CTCs may lead to false positive and false negative results

Cell-free DNA (cfDNA)
  • High sensitivity in detecting genetic aberrations

  • Challenging to isolate pure population of cfDNAs

  • Molecular characterisation is limited to genomic DNA and unable to evaluate the RNA and protein profile

  • Unable to perform phenotypic and functional studies

Exosome
  • Abundant in the plasma

  • Allows downstream molecular analysis and functional studies

  • Able to evaluate the DNA, RNA and protein profile of tumour cells

  • Allows analysis of inflammatory, stromal and other systemic changes

  • Challenging to isolate pure populations of tumour-derived exosomes

  • Unable to perform phenotypic studies

Table 2.

Comparison of the liquid biopsy markers.

The isolated cfDNAs are subjected to sequencing analysis using next-generation sequencing platforms to identify tumour-associated genetic mutations or epigenetic changes. For example, in a large-scale study of NSCLC patients on gefitinib treatment, cfDNA was used as a surrogate of tissue biopsy to identify the EGFR mutations, demonstrating its clinical utility [100, 104]. A comparison on the frequency of mutation detection between cfDNA and CTC revealed that cfDNA showed a higher frequency of the mutation from the same patient [100, 102], suggesting that cfDNA is more effective in detecting these genetic changes. Another advantage that cfDNAs have over CTCs is that cfDNAs can be obtained from bio-banked fluids such as frozen plasma whereas CTCs can only be obtained from peripheral blood [100]. However, the use of cfDNAs as a liquid biopsy marker also poses several limitations. First, because cfDNA can originate from any cell type including normal cells and tumour cells, there will be a high background of wild type DNA and thus isolating a pure population of the rare tumour-derived cfDNA is technically challenging. The abundance of wild type DNA may mask the detection of low copy genetic mutations that could be important for early detection of cancer or drug resistance. Second, molecular profiling of cfDNAs is restricted to the DNA level as the characterisation of the transcriptome and proteome is not possible [100].

Exosomes are membrane-bound microvesicles derived from multivesicular bodies (MVBs) and secreted into the extracellular environment through fusion of MVB to the plasma membrane [8, 100]. They mediate cell-cell communication by transferring biomolecules such as DNA, RNA, proteins and lipids from the donor cell to the recipient cell [105]. Exosomes can originate from many cell types including tumour cells, epithelial cells, fibroblasts, neuronal cells, haematopoietic cells and adipocytes [8, 106]. Most exosomes are around 30–200 nm and are present in large quantities in biological fluids including serum, plasma, urine and saliva [8, 100]. Tumour cells release tens of thousands of exosomes in a day, resulting in hundreds of billions of exosomes per ml of plasma [100, 107]. Since exosomes are abundant, they are easier to isolate compared to CTCs and cfDNAs. However, there is a lack of efficient tumour exosome enrichment method [108] because the isolation of exosomes is largely based on exosome-specific surface markers [100, 109], which do not distinguish between exosomes derived from tumour cells and normal cells.

Because exosomes carry the DNA, RNA and protein content from tumour cells (i.e. cell of origin), they are useful diagnostic and prognostic tools of cancer. Similar to CTCs and cfDNAs, exosomes can be subjected to DNA analysis to determine the genetic aberrations and mutational landscape of tumour cells. Additionally, the RNA and protein profiles of tumour exosomes can be characterised to provide insights into the biology of tumour cells. For example, analysis of exosomal mRNA and proteins allows real-time monitoring of therapeutic response and drug resistance in glioblastoma patients [110, 111]. The up-regulated expression of a panel of serum-derived exosomal miRNAs (miR-1246, miR-4644, miR-3976, miR-4306) serves as a biomarker for the diagnosis of pancreatic cancer [112]. The down-regulated expression of miR-92a is associated with high risk of cancer relapse in hepatocellular carcinoma patients [113]. Furthermore, previous reports showed that tumour exosomes play a role in suppressing immune response, promoting tumour cell growth, angiogenesis and metastasis [100, 114, 115, 116]. Similar to CTCs, molecular profiling of exosomes provides insights into the mechanism of metastasis and drug resistance.

3.6. Barriers to adoption of CTC as a clinical test

Although CTCs have numerous potential clinical applications, the incorporation of CTCs into routine clinical practice still faces several challenges. First, there is a lack of reproducibility in CTC enumeration when different measurements were taken from the same patient [117]. This variation is likely caused by the extremely low frequency of CTCs in peripheral blood, where >90% of patients with localised diseases and up to 30–40% of patients with metastatic disease do not have >5 CTCs per 7.5 ml of blood [117, 118]. A difference of 1 CTC may lead to different stratification and prognostic outcome [117]. A possible solution to improve the CTC yield is to process larger volume of blood sample from cancer patients [117, 119]. Second, there is a lack of standardisation across the myriad of CTC detection platforms in defining and isolating CTCs [120, 121], resulting in variability of CTC count. Studies comparing EpCAM-dependent and EpCAM-independent CTC enrichment methods using blood samples from the same patient showed that EpCAM-independent methods generate a higher CTC count compared to the CellSearch system [117]. Thus, a universal quality control system is required for detecting and isolating CTCs across the various platforms so as to benchmark the reliability of these methods [117, 120]. Cross-validation studies on CTC enumeration from different laboratories will also minimise the inter-observer variation [117]. Lastly, the current CTC isolation technologies have limited sensitivity and are not applicable to all types of cancer [117]. Therefore, before CTCs can be fully adopted for routine clinical use, well-designed appropriately powered validation studies are required.

Advertisement

4. Conclusion

Late clinical diagnosis and chemotherapy resistance are the main factors leading to reduced chances of survival for cancer patients. To combat cancer, CTCs provide invaluable information on the status of the disease and the likely outcome of chemotherapeutic treatment. The enumeration of CTCs allows early detection of cancer, prognosis and real-time monitoring of chemotherapy treatment. The single-cell analysis of CTCs provides a wealth of genetic information that enables better understanding of the disease complexity for individual patients and provides the opportunity for the development of personalised treatment. To aid in delivering better therapeutic medicine, current technologies allow CTCs to be cultured in vitro for the identification of novel therapeutic targets and optimal drug dosage for individual patients. Therefore, the molecular characterisation of CTCs is important for improving the clinical outcomes in cancer patients.

References

  1. 1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: A Cancer Journal for Clinicians. 2011;61(2):69-90. DOI: 10.3322/caac.20107
  2. 2. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144(5):646-674. DOI: 10.1016/j.cell.2011.02.013
  3. 3. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science (New York, N.Y.). 2011;331(6024):1559-1564. DOI: 10.1126/science.1203543
  4. 4. Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. Critical Reviews in Oncogenesis. 2013;18(1-2):43-73
  5. 5. Ilie M, Hofman V, Long-Mira E, Selva E, Vignaud JM, Padovani B, et al. “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS One. 2014;9(10):e111597. DOI: 10.1371/journal.pone.0111597
  6. 6. Zhang J, Chen K, Fan ZH. Circulating tumor cell isolation and analysis. Advances in Clinical Chemistry. 2016;75:1-31. DOI: 10.1016/bs.acc.2016.03.003
  7. 7. Ilie M, Hofman P. Pros: Can tissue biopsy be replaced by liquid biopsy? Translational Lung Cancer Research. 2016;5(4):420-423. DOI: 10.21037/tlcr.2016.08.06
  8. 8. Zhang W, Xia W, Lv Z, Ni C, Xin Y, Yang L. Liquid biopsy for cancer: Circulating tumor cells, circulating free DNA or exosomes? Cellular Physiology and Biochemistry: International Journal of Experimental Cellular Physiology, Biochemistry, and Pharmacology. 2017;41(2):755-768. DOI: 10.1159/000458736
  9. 9. Gkountela S, Szczerba B, Donato C, Aceto N. Recent advances in the biology of human circulating tumour cells and metastasis. ESMO Open. 2016;1(4):e000078. DOI: 10.1136/esmoopen-2016-000078
  10. 10. Alix-Panabieres C, Pantel K. Challenges in circulating tumour cell research. Nature Reviews Cancer. 2014;14(9):623-631. DOI: 10.1038/nrc3820
  11. 11. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo RA, Tan DS, et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Scientific Reports. 2013;3:1259. DOI: 10.1038/srep01259
  12. 12. Williams SC. Circulating tumor cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(13):4861. DOI: 10.1073/pnas.1304186110
  13. 13. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clinical cancer research: An official journal of the American Association for Cancer Research. 2004;10(20):6897-6904. DOI: 10.1158/1078-0432.ccr-04-0378
  14. 14. Fan T, Zhao Q, Chen JJ, Chen WT, Pearl ML. Clinical significance of circulating tumor cells detected by an invasion assay in peripheral blood of patients with ovarian cancer. Gynecologic Oncology. 2009;112(1):185-191. DOI: 10.1016/j.ygyno.2008.09.021
  15. 15. Kalinsky K, Mayer JA, Xu X, Pham T, Wong KL, Villarin E, et al. Correlation of hormone receptor status between circulating tumor cells, primary tumor, and metastasis in breast cancer patients. Clinical & Translational Oncology: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico. 2015;17(7):539-546. DOI: 10.1007/s12094-015-1275-1
  16. 16. Tan SJ, Yeo T, Sukhatme SA, Kong SL, Lim WT, Lim CT. Personalized treatment through detection and monitoring of genetic aberrations in single circulating tumor cells. Advances in Experimental Medicine and Biology. 2017;994:255-273. DOI: 10.1007/978-3-319-55947-6_14
  17. 17. Yeo T, Tan SJ, Lim CL, Lau DP, Chua YW, Krisna SS, et al. Microfluidic enrichment for the single cell analysis of circulating tumor cells. Scientific Reports. 2016;6:22076. DOI: 10.1038/srep22076
  18. 18. Janku F. Tumor heterogeneity in the clinic: Is it a real problem? Therapeutic Advances in Medical Oncology. 2014;6(2):43-51. DOI: 10.1177/1758834013517414
  19. 19. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: Past, present, and the future. Cell. 2017;168(4):613-628. DOI: 10.1016/j.cell.2017.01.018
  20. 20. Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Molecular Cancer. 2017;16(1):41. DOI: 10.1186/s12943-017-0600-4
  21. 21. Krishnamurthy N, Spencer E, Torkamani A, Nicholson L. Liquid biopsies for cancer: Coming to a patient near you. Journal of Clinical Medicine. 2017;6(1):3. DOI: 10.3390/jcm6010003
  22. 22. Dong Y, Skelley AM, Merdek KD, Sprott KM, Jiang C, Pierceall WE, et al. Microfluidics and circulating tumor cells. The Journal of Molecular Diagnostics. 2013;15(2):149-157. DOI: 10.1016/j.jmoldx.2012.09.004
  23. 23. Autebert J, Coudert B, Bidard FC, Pierga JY, Descroix S, Malaquin L, et al. Microfluidic: An innovative tool for efficient cell sorting. Methods (San Diego, Calif). 2012;57(3):297-307. DOI: 10.1016/j.ymeth.2012.07.002
  24. 24. Whitesides GM. The origins and the future of microfluidics. Nature. 2006;442(7101):368-373. DOI: 10.1038/nature05058
  25. 25. Moreno JG, O’Hara SM, Gross S, Doyle G, Fritsche H, Gomella LG, et al. Changes in circulating carcinoma cells in patients with metastatic prostate cancer correlate with disease status. Urology. 2001;58(3):386-392
  26. 26. Shaw Bagnall J, Byun S, Begum S, Miyamoto DT, Hecht VC, Maheswaran S, et al. Deformability of tumor cells versus. Blood Cells. Scientific Reports. 2015;5:18542. DOI: 10.1038/srep18542
  27. 27. Gascoyne PR, Shim S. Isolation of circulating tumor cells by dielectrophoresis. Cancer. 2014;6(1):545-579. DOI: 10.3390/cancers6010545
  28. 28. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. The New England Journal of Medicine. 2004;351(8):781-791. DOI: 10.1056/NEJMoa040766
  29. 29. Negin BP, Cohen SJ. Circulating tumor cells in colorectal cancer: Past, present, and future challenges. Current Treatment Options in Oncology. 2010;11(1-2):1-13. DOI: 10.1007/s11864-010-0115-3
  30. 30. Resel Folkersma L, Olivier Gomez C, San Jose Manso L, Veganzones de Castro S, Galante Romo I, Vidaurreta Lazaro M, et al. Immunomagnetic quantification of circulating tumoral cells in patients with prostate cancer: Clinical and pathological correlation. Archivos Espanoles De Urologia. 2010;63(1):23-31
  31. 31. Millner LM, Strotman LN. The future of precision medicine in oncology. Clinics in Laboratory Medicine. 2016;36(3):557-573. DOI: 10.1016/j.cll.2016.05.003
  32. 32. Truini A, Alama A, Dal Bello MG, Coco S, Vanni I, Rijavec E, et al. Clinical applications of circulating tumor cells in lung cancer patients by CellSearch system. Frontiers in Oncology. 2014;4:242. DOI: 10.3389/fonc.2014.00242
  33. 33. Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235-1239. DOI: 10.1038/nature06385
  34. 34. Gertler R, Rosenberg R, Fuehrer K, Dahm M, Nekarda H, Siewert JR. Detection of circulating tumor cells in blood using an optimized density gradient centrifugation. Recent Results in Cancer Research Fortschritte der Krebsforschung Progres dans les recherches sur le cancer. 2003;162:149-155
  35. 35. Hur SC, Henderson-MacLennan NK, McCabe ER, Di Carlo D. Deformability-based cell classification and enrichment using inertial microfluidics. Lab on a Chip. 2011;11(5):912-920. DOI: 10.1039/c0lc00595a
  36. 36. Hyun KA, Kwon K, Han H, Kim SI, Jung HI. Microfluidic flow fractionation device for label-free isolation of circulating tumor cells (CTCs) from breast cancer patients. Biosensors & Bioelectronics. 2013;40(1):206-212. DOI: 10.1016/j.bios.2012.07.021
  37. 37. Mohamed H, Murray M, Turner JN, Caggana M. Isolation of tumor cells using size and deformation. Journal of Chromatography A. 2009;1216(47):8289-8295. DOI: 10.1016/j.chroma.2009.05.036
  38. 38. Tan SJ, Lakshmi RL, Chen P, Lim WT, Yobas L, Lim CT. Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients. Biosensors & Bioelectronics. 2010;26(4):1701-1705. DOI: 10.1016/j.bios.2010.07.054
  39. 39. Tan SJ, Yobas L, Lee GY, Ong CN, Lim CT. Microdevice for the isolation and enumeration of cancer cells from blood. Biomedical Microdevices. 2009;11(4):883-892. DOI: 10.1007/s10544-009-9305-9
  40. 40. Warkiani ME, Guan G, Luan KB, Lee WC, Bhagat AA, Chaudhuri PK, et al. Slanted spiral microfluidics for the ultra-fast, label-free isolation of circulating tumor cells. Lab on a Chip. 2014;14(1):128-137. DOI: 10.1039/c3lc50617g
  41. 41. Zheng S, Lin H, Liu JQ, Balic M, Datar R, Cote RJ, et al. Membrane microfilter device for selective capture, electrolysis and genomic analysis of human circulating tumor cells. Journal of Chromatography A. 2007;1162(2):154-161. DOI: 10.1016/j.chroma.2007.05.064
  42. 42. Adams AA, Okagbare PI, Feng J, Hupert ML, Patterson D, Gottert J, et al. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. Journal of the American Chemical Society. 2008;130(27):8633-8641. DOI: 10.1021/ja8015022
  43. 43. Gleghorn JP, Pratt ED, Denning D, Liu H, Bander NH, Tagawa ST, et al. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab on a Chip. 2010;10(1):27-29. DOI: 10.1039/b917959c
  44. 44. Saliba AE, Saias L, Psychari E, Minc N, Simon D, Bidard FC, et al. Microfluidic sorting and multimodal typing of cancer cells in self-assembled magnetic arrays. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(33):14524-14529. DOI: 10.1073/pnas.1001515107
  45. 45. Sheng W, Ogunwobi OO, Chen T, Zhang J, George TJ, Liu C, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab on a Chip. 2014;14(1):89-98. DOI: 10.1039/c3lc51017d
  46. 46. Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(43):18392-18397. DOI: 10.1073/pnas.1012539107
  47. 47. Xu Y, Phillips JA, Yan J, Li Q, Fan ZH, Tan W. Aptamer-based microfluidic device for enrichment, sorting, and detection of multiple cancer cells. Analytical Chemistry. 2009;81(17):7436-7442. DOI: 10.1021/ac9012072
  48. 48. Spizzo G, Went P, Dirnhofer S, Obrist P, Simon R, Spichtin H, et al. High Ep-CAM expression is associated with poor prognosis in node-positive breast cancer. Breast Cancer Research and Treatment. 2004;86(3):207-213. DOI: 10.1023/b:brea.0000036787.59816.01
  49. 49. Pecot CV, Bischoff FZ, Mayer JA, Wong KL, Pham T, Bottsford-Miller J, et al. A novel platform for detection of CK+ and CK− CTCs. Cancer Discovery. 2011;1(7):580-586. DOI: 10.1158/2159-8290.cd-11-0215
  50. 50. Warkiani ME, Khoo BL, Wu L, Tay AK, Bhagat AA, Han J, et al. Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics. Nature Protocols. 2016;11(1):134-148. DOI: 10.1038/nprot.2016.003
  51. 51. Karabacak NM, Spuhler PS, Fachin F, Lim EJ, Pai V, Ozkumur E, et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nature Protocols. 2014;9(3):694-710. DOI: 10.1038/nprot.2014.044
  52. 52. Ozkumur E, Shah AM, Ciciliano JC, Emmink BL, Miyamoto DT, Brachtel E, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Science Translational Medicine. 2013;5(179):179ra47. DOI: 10.1126/scitranslmed.3005616
  53. 53. Harb W, Fan A, Tran T, Danila DC, Keys D, Schwartz M, et al. Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay. Translational Oncology. 2013;6(5):528-538
  54. 54. Earhart CM, Hughes CE, Gaster RS, Ooi CC, Wilson RJ, Zhou LY, et al. Isolation and mutational analysis of circulating tumor cells from lung cancer patients with magnetic sifters and biochips. Lab on a Chip. 2014;14(1):78-88. DOI: 10.1039/c3lc50580d
  55. 55. Galletti G, Portella L, Tagawa ST, Kirby BJ, Giannakakou P, Nanus DM. Circulating tumor cells in prostate cancer diagnosis and monitoring: An appraisal of clinical potential. Molecular Diagnosis & Therapy. 2014;18(4):389-402. DOI: 10.1007/s40291-014-0101-8
  56. 56. Kirby BJ, Jodari M, Loftus MS, Gakhar G, Pratt ED, Chanel-Vos C, et al. Functional characterization of circulating tumor cells with a prostate-cancer-specific microfluidic device. PLoS One. 2012;7(4):e35976. DOI: 10.1371/journal.pone.0035976
  57. 57. Sollier E, Go DE, Che J, Gossett DR, O’Byrne S, Weaver WM, et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab on a Chip. 2014;14(1):63-77. DOI: 10.1039/c3lc50689d
  58. 58. Peeters DJ, De Laere B, Van den Eynden GG, Van Laere SJ, Rothe F, Ignatiadis M, et al. Semiautomated isolation and molecular characterisation of single or highly purified tumour cells from CellSearch enriched blood samples using dielectrophoretic cell sorting. British Journal of Cancer. 2013;108(6):1358-1367. DOI: 10.1038/bjc.2013.92
  59. 59. Polzer B, Medoro G, Pasch S, Fontana F, Zorzino L, Pestka A, et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Molecular Medicine. 2014;6(11):1371-1386. DOI: 10.15252/emmm.201404033
  60. 60. Dey SS, Kester L, Spanjaard B, Bienko M, van Oudenaarden A. Integrated genome and transcriptome sequencing of the same cell. Nature Biotechnology. 2015;33(3):285-289. DOI: 10.1038/nbt.3129
  61. 61. Stoecklein NH, Hosch SB, Bezler M, Stern F, Hartmann CH, Vay C, et al. Direct genetic analysis of single disseminated cancer cells for prediction of outcome and therapy selection in esophageal cancer. Cancer Cell. 2008;13(5):441-453. DOI: 10.1016/j.ccr.2008.04.005
  62. 62. Kehr J. Single cell technology. Current Opinion in Plant Biology. 2003;6(6):617-621
  63. 63. Shaw JA, Guttery DS, Hills A, Fernandez-Garcia D, Page K, Rosales BM, et al. Mutation analysis of cell-free DNA and single circulating tumor cells in metastatic breast cancer patients with high circulating tumor cell counts. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2017;23(1):88-96. DOI: 10.1158/1078-0432.ccr-16-0825
  64. 64. Carter L, Rothwell DG, Mesquita B, Smowton C, Leong HS, Fernandez-Gutierrez F, et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nature Medicine. 2017;23(1):114-119. DOI: 10.1038/nm.4239
  65. 65. Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell. 2018;173(4):879-93.e13. DOI: 10.1016/j.cell.2018.03.041
  66. 66. Wang Y, Waters J, Leung ML, Unruh A, Roh W, Shi X, et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature. 2014;512(7513):155-160. DOI: 10.1038/nature13600
  67. 67. Alix-Panabieres C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discovery. 2016;6(5):479-491. DOI: 10.1158/2159-8290.cd-15-1483
  68. 68. Tanaka F, Yoneda K, Kondo N, Hashimoto M, Takuwa T, Matsumoto S, et al. Circulating tumor cell as a diagnostic marker in primary lung cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2009;15(22):6980-6986. DOI: 10.1158/1078-0432.ccr-09-1095
  69. 69. Tsai WS, Chen JS, Shao HJ, Wu JC, Lai JM, Lu SH, et al. Circulating tumor cell count correlates with colorectal neoplasm progression and is a prognostic marker for distant metastasis in non-metastatic patients. Scientific Reports. 2016;6:24517. DOI: 10.1038/srep24517
  70. 70. Rack B, Schindlbeck C, Juckstock J, Andergassen U, Hepp P, Zwingers T, et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. Journal of the National Cancer Institute. 2014;106(5):dju066. DOI: 10.1093/jnci/dju066
  71. 71. Hiltermann TJ, Pore MM, van den Berg A, Timens W, Boezen HM, Liesker JJ, et al. Circulating tumor cells in small-cell lung cancer: A predictive and prognostic factor. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2012;23(11):2937-2942. DOI: 10.1093/annonc/mds138
  72. 72. Hou JM, Greystoke A, Lancashire L, Cummings J, Ward T, Board R, et al. Evaluation of circulating tumor cells and serological cell death biomarkers in small cell lung cancer patients undergoing chemotherapy. The American Journal of Pathology. 2009;175(2):808-816. DOI: 10.2353/ajpath.2009.090078
  73. 73. Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2011;29(12):1556-1563. DOI: 10.1200/jco.2010.28.7045
  74. 74. Naito T, Tanaka F, Ono A, Yoneda K, Takahashi T, Murakami H, et al. Prognostic impact of circulating tumor cells in patients with small cell lung cancer. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2012;7(3):512-519. DOI: 10.1097/JTO.0b013e31823f125d
  75. 75. Gazzaniga P, De Berardinis E, Raimondi C, Gradilone A, Busetto GM, De Falco E, et al. Circulating tumor cells detection has independent prognostic impact in high-risk non-muscle invasive bladder cancer. International Journal of Cancer. 2014;135(8):1978-1982. DOI: 10.1002/ijc.28830
  76. 76. Rink M, Chun FK, Dahlem R, Soave A, Minner S, Hansen J, et al. Prognostic role and HER2 expression of circulating tumor cells in peripheral blood of patients prior to radical cystectomy: A prospective study. European Urology. 2012;61(4):810-817. DOI: 10.1016/j.eururo.2012.01.017
  77. 77. Schulze K, Gasch C, Staufer K, Nashan B, Lohse AW, Pantel K, et al. Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma. International Journal of Cancer. 2013;133(9):2165-2171. DOI: 10.1002/ijc.28230
  78. 78. Vashist YK, Effenberger KE, Vettorazzi E, Riethdorf S, Yekebas EF, Izbicki JR, et al. Disseminated tumor cells in bone marrow and the natural course of resected esophageal cancer. Annals of Surgery. 2012;255(6):1105-1112. DOI: 10.1097/SLA.0b013e3182565b0b
  79. 79. Muinelo-Romay L, Vieito M, Abalo A, Nocelo MA, Baron F, Anido U, et al. Evaluation of circulating tumor cells and related events as prognostic factors and surrogate biomarkers in advanced NSCLC patients receiving first-line systemic treatment. Cancer. 2014;6(1):153-165. DOI: 10.3390/cancers6010153
  80. 80. Punnoose EA, Atwal S, Liu W, Raja R, Fine BM, Hughes BG, et al. Evaluation of circulating tumor cells and circulating tumor DNA in non-small cell lung cancer: Association with clinical endpoints in a phase II clinical trial of pertuzumab and erlotinib. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2012;18(8):2391-2401. DOI: 10.1158/1078-0432.ccr-11-3148
  81. 81. Khoo BL, Grenci G, Lim YB, Lee SC, Han J, Lim CT. Expansion of patient-derived circulating tumor cells from liquid biopsies using a CTC microfluidic culture device. Nature Protocols. 2018;13(1):34-58. DOI: 10.1038/nprot.2017.125
  82. 82. Ni X, Zhuo M, Su Z, Duan J, Gao Y, Wang Z, et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(52):21083-21088. DOI: 10.1073/pnas.1320659110
  83. 83. Sequist LV, Joshi VA, Janne PA, Muzikansky A, Fidias P, Meyerson M, et al. Response to treatment and survival of patients with non-small cell lung cancer undergoing somatic EGFR mutation testing. The Oncologist. 2007;12(1):90-98
  84. 84. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. The New England Journal of Medicine. 2014;371(11):1028-1038. DOI: 10.1056/NEJMoa1315815
  85. 85. Steinestel J, Luedeke M, Arndt A, Schnoeller T, Lennerz JK, Maier C, et al. Detecting predictive androgen receptor modifications in circulating prostate cancer cells. Journal of Clinical Oncology. 2015;33(15_suppl):5067. DOI: 10.1200/jco.2015.33.15_suppl.5067
  86. 86. Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Nakazawa M, et al. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncology. 2015;1(5):582-591. DOI: 10.1001/jamaoncol.2015.1341
  87. 87. Nakazawa M, Lu C, Chen Y, Paller CJ, Carducci MA, Eisenberger MA, et al. Serial blood-based analysis of AR-V7 in men with advanced prostate cancer. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2015;26(9):1859-1865. DOI: 10.1093/annonc/mdv282
  88. 88. Onstenk W, Sieuwerts AM, Kraan J, Van M, Nieuweboer AJ, Mathijssen RH, et al. Efficacy of cabazitaxel in castration-resistant prostate cancer is independent of the presence of AR-V7 in circulating tumor cells. European Urology. 2015;68(6):939-945. DOI: 10.1016/j.eururo.2015.07.007
  89. 89. Thadani-Mulero M, Portella L, Sun S, Sung M, Matov A, Vessella RL, et al. Androgen receptor splice variants determine taxane sensitivity in prostate cancer. Cancer Research. 2014;74(8):2270-2282. DOI: 10.1158/0008-5472.can-13-2876
  90. 90. Babayan A, Hannemann J, Spotter J, Muller V, Pantel K, Joosse SA. Heterogeneity of estrogen receptor expression in circulating tumor cells from metastatic breast cancer patients. PLoS One. 2013;8(9):e75038. DOI: 10.1371/journal.pone.0075038
  91. 91. Fehm T, Muller V, Aktas B, Janni W, Schneeweiss A, Stickeler E, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: A prospective, multicenter trial. Breast Cancer Research and Treatment. 2010;124(2):403-412. DOI: 10.1007/s10549-010-1163-x
  92. 92. Ignatiadis M, Rothe F, Chaboteaux C, Durbecq V, Rouas G, Criscitiello C, et al. HER2-positive circulating tumor cells in breast cancer. PLoS One. 2011;6(1):e15624. DOI: 10.1371/journal.pone.0015624
  93. 93. Riethdorf S, Muller V, Zhang L, Rau T, Loibl S, Komor M, et al. Detection and HER2 expression of circulating tumor cells: Prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2010;16(9):2634-2645. DOI: 10.1158/1078-0432.ccr-09-2042
  94. 94. Mazel M, Jacot W, Pantel K, Bartkowiak K, Topart D, Cayrefourcq L, et al. Frequent expression of PD-L1 on circulating breast cancer cells. Molecular Oncology. 2015;9(9):1773-1782. DOI: 10.1016/j.molonc.2015.05.009
  95. 95. Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Molecular Immunology. 2015;67(2 Pt A):4-17. DOI: 10.1016/j.molimm.2015.02.009
  96. 96. Jilaveanu LB, Shuch B, Zito CR, Parisi F, Barr M, Kluger Y, et al. PD-L1 expression in clear cell renal cell carcinoma: An analysis of nephrectomy and sites of metastases. Journal of Cancer. 2014;5(3):166-172. DOI: 10.7150/jca.8167
  97. 97. Nicolazzo C, Raimondi C, Mancini M, Caponnetto S, Gradilone A, Gandini O, et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Scientific Reports. 2016;6:31726. DOI: 10.1038/srep31726
  98. 98. Pantel K, Alix-Panabieres C. Liquid biopsy in 2016: Circulating tumour cells and cell-free DNA in gastrointestinal cancer. Nature Reviews Gastroenterology & Hepatology. 2017;14(2):73-74. DOI: 10.1038/nrgastro.2016.198
  99. 99. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nature Reviews Cancer. 2011;11(6):426-437. DOI: 10.1038/nrc3066
  100. 100. Brock G, Castellanos-Rizaldos E, Hu L, Coticchia C, Skog J. Liquid biopsy for cancer screening, patient stratification and monitoring. Translational Cancer Research. 2015;4(3):280-290
  101. 101. Jiang P, Chan CW, Chan KC, Cheng SH, Wong J, Wong VW, et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(11):E1317-E1325. DOI: 10.1073/pnas.1500076112
  102. 102. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Science Translational Medicine. 2014;6(224):224ra24. DOI: 10.1126/scitranslmed.3007094
  103. 103. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nature Medicine. 2014;20(5):548-554. DOI: 10.1038/nm.3519
  104. 104. Douillard JY, Ostoros G, Cobo M, Ciuleanu T, Cole R, McWalter G, et al. Gefitinib treatment in EGFR mutated caucasian NSCLC: Circulating-free tumor DNA as a surrogate for determination of EGFR status. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2014;9(9):1345-1353. DOI: 10.1097/jto.0000000000000263
  105. 105. Soung YH, Ford S, Zhang V, Chung J. Exosomes in cancer diagnostics. Cancer. 2017;9(1):8. DOI: 10.3390/cancers9010008
  106. 106. van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacological Reviews. 2012;64(3):676-705. DOI: 10.1124/pr.112.005983
  107. 107. Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, Breakefield XO, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nature Communications. 2011;2:180. DOI: 10.1038/ncomms1180
  108. 108. Wang J, Chang S, Li G, Sun Y. Application of liquid biopsy in precision medicine: Opportunities and challenges. Frontiers of Medicine. 2017;11(4):522-527. DOI: 10.1007/s11684-017-0526-7
  109. 109. He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab on a Chip. 2014;14(19):3773-3780. DOI: 10.1039/c4lc00662c
  110. 110. Shao H, Chung J, Balaj L, Charest A, Bigner DD, Carter BS, et al. Protein typing of circulating microvesicles allows real-time monitoring of glioblastoma therapy. Nature Medicine. 2012;18(12):1835-1840. DOI: 10.1038/nm.2994
  111. 111. Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nature Communications. 2015;6:6999. DOI: 10.1038/ncomms7999
  112. 112. Madhavan B, Yue S, Galli U, Rana S, Gross W, Muller M, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. International Journal of Cancer. 2015;136(11):2616-2627. DOI: 10.1002/ijc.29324
  113. 113. Masyuk AI, Masyuk TV, Larusso NF. Exosomes in the pathogenesis, diagnostics and therapeutics of liver diseases. Journal of Hepatology. 2013;59(3):621-625. DOI: 10.1016/j.jhep.2013.03.028
  114. 114. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology. 2015;17(6):816-826. DOI: 10.1038/ncb3169
  115. 115. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nature Medicine. 2012;18(6):883-891. DOI: 10.1038/nm.2753
  116. 116. Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biology. 2008;10(12):1470-1476. DOI: 10.1038/ncb1800
  117. 117. Leon-Mateos L, Vieito M, Anido U, Lopez Lopez R, Muinelo Romay L. Clinical application of circulating tumour cells in prostate cancer: From bench to bedside and back. International Journal of Molecular Sciences. 2016;17(9):1580. DOI: 10.3390/ijms17091580
  118. 118. Maestro LM, Sastre J, Rafael SB, Veganzones SB, Vidaurreta M, Martin M, et al. Circulating tumor cells in solid tumor in metastatic and localized stages. Anticancer Research. 2009;29(11):4839-4843
  119. 119. Lalmahomed ZS, Kraan J, Gratama JW, Mostert B, Sleijfer S, Verhoef C. Circulating tumor cells and sample size: The more, the better. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2010;28(17):e288-e289 (author reply e90). DOI: 10.1200/jco.2010.28.2764
  120. 120. Lianidou ES. Circulating tumor cells--new challenges ahead. Clinical Chemistry. 2012;58(5):805-807. DOI: 10.1373/clinchem.2011.180646
  121. 121. Lianidou ES, Mavroudis D, Georgoulias V. Clinical challenges in the molecular characterization of circulating tumour cells in breast cancer. British Journal of Cancer. 2013;108(12):2426-2432. DOI: 10.1038/bjc.2013.265

Written By

Kah Yee Goh and Wan-Teck Lim

Submitted: 25 January 2018 Reviewed: 31 May 2018 Published: 05 November 2018