Skip to main content
Cancer Biology & Medicine logoLink to Cancer Biology & Medicine
. 2016 Mar;13(1):19–40. doi: 10.28092/j.issn.2095-3941.2016.0007

The potential for liquid biopsies in the precision medical treatment of breast cancer

Victoria A Forte 1,2, Dany K Barrak 2,3, Mostafa Elhodaky 2,4, Lily Tung 2,3, Anson Snow 1,2, Julie E Lang 2,3
PMCID: PMC4850125  PMID: 27144060

Abstract

Currently the clinical management of breast cancer relies on relatively few prognostic/predictive clinical markers (estrogen receptor, progesterone receptor, HER2), based on primary tumor biology. Circulating biomarkers, such as circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs) may enhance our treatment options by focusing on the very cells that are the direct precursors of distant metastatic disease, and probably inherently different than the primary tumor's biology. To shift the current clinical paradigm, assessing tumor biology in real time by molecularly profiling CTCs or ctDNA may serve to discover therapeutic targets, detect minimal residual disease and predict response to treatment. This review serves to elucidate the detection, characterization, and clinical application of CTCs and ctDNA with the goal of precision treatment of breast cancer.

Keywords: Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), cell free DNA (cfDNA), biomarker, cancer

Introduction

The concept of personalized medicine in oncology, or customizing cancer treatment based on specific biomarkers predictive of drug response, is rapidly progressing based on innovative translational research strategies and the implementation of biomarkers in clinical trial design. The key to personalizing medicine is to identify biomarkers which are easily accessible and could be repeatedly accessed to inform physicians about an individual's tumor biology, predict which therapeutic interventions may be of greatest benefit, monitor treatment responses to therapy, identify mechanisms of therapeutic resistance to guide subsequent therapies, and assay for microscopic relapse1.

Primary tumor gene expression profiling is prognostic in terms of probability of recurrence; however, breast cancer patients would benefit greatly from a blood test that could provide a real time assessment of the cancer cells that are most likely to produce distant metastatic disease. Circulating biomarkers may provide insights into how to better treat breast cancer patients by focusing on the very cells that are the direct precursors of distant metastatic disease, and probably very different than the primary tumor's biology. Two circulating biomarkers through which personalized medicine can be achieved are the study of circulating tumor cells (CTCs) and that of circulating tumor DNA (ctDNA). These have been promoted as potential real time "liquid biopsies", minimally invasive biomarkers capable of providing a global picture of the tumor burden and vital genetic information about the targeted tumor from a blood draw2-4. However, challenges still remain in the identification,isolation, and ultimate harnessing of these methodologies in order to effectively guide treatment. This review serves to identify the detection, characterization, and application of ctDNA and CTCs with the goal of precision treatment of breast cancer.

CTCs

CTCs are recognized to be rare cells that are shed from the primary or metastatic tumor and can be isolated from the peripheral blood of patients with solid and liquid tumors5. CTCs were first reported in 1869 in the blood of a man with metastatic cancer by scientist Thomas Ashworth, who performed a thorough comparison of the morphology of the circulating cells to tumor cells leading him to conclude that "one thing is certain, that if they (CTCs) came from an existing cancer structure, they must have passed through the greater part of the circulatory system"6,7. CTCs are an independent prognostic factor in all stages of breast cancer8-12. CTC's rarity and short half-life, probably measured in hours, renders isolation a challenge; therefore a successful assay should ideally be able to process a large amount of cells in a relatively short amount of time while being able to specifically enumerate and capture malignant cells from a vast blood background - requiring the ability to detect one CTC per 105-107 mononuclear cells13. Such assays include a large panel of technologies based on their multiple physical and biological properties such as size, deformability, surface protein expression, viability, and invasion capacity properties distinguishing them from the surrounding hematopoietic cell lineage5,14. Our group has previously reviewed the state of this scientific field and speculated that CTCs would gain considerable interest as molecular techniques that allowed for their characterization developed and could permit translation to the clinic15.

CTCs have been molecularly profiled via several approaches, including using next-generation sequencing (NGS)16,17. These advances have revealed some mechanisms of drug resistance including epithelial to mesenchymal transition (EMT)18-20. In addition, these breakthroughs can help us characterize those cancer cells capable of metastasis and optimize targeted therapies based on each patient's unique tumor biology, not simply rely on the standard four therapeutic groups of breast cancer: (1) hormone receptor (HR) positive patients who have estrogen and/or progesterone-dependent tumors but are HER2 (human epidermal growth factor type 2) negative (ER/PR+HER2−); (2) HR positive and HER2 positive (ER/PR+HER2+) patients who have amplification or overexpression of HER2, in addition to estrogen and/or progesterone-dependent tumors; (3) HR negative group with HER2 overexpression/ amplification (ER/PR−HER2+), and (4) triple-negative breast cancers (TNBCs), which are defined by a lack of expression of ER, PR, and HER221,22. Optimization of detection techniques is required in order to molecularly characterize CTCs and incorporate this information into the current clinical landscape.

CTC detection techniques

Over the last two decades there have been several methods developed for isolating and analyzing CTCs. Overall they may be separated into affinity based, physical properties based, and functional assays. Many affinity-based techniques isolate cells based on expression of cell-surface markers, such as the epithelial cell adhesion molecule (EpCAM), and include the CellSearch assay (the only FDA approved method of detecting CTCs in breast cancer), the Herringbone-CTC chip, and flow cytometry-based approaches. Many of these selection technologies are criticized for their reliance on cell-surface expression of EpCAM to capture (and define) CTCs because some tumors down-regulate expression of this marker during EMT. In addition, preanalytic variables, such as time to assay are significant factors in CTC detection and may be more important than EpCAM high status, with the exception of claudin low tumors23. To address this limitation, physical property and functional based technologies have been developed that isolate CTCs on the basis of physico-chemical properties-such as size, density, surface charges, unique functional characteristics-that distinguish them from other blood cells24. Please see Table 1 for a summary of the various isolation techniques being utilized25-93.

Table1.

A summary of the various isolation techniques being utilized

Technique Description Reference
Physical properties based assays
Dean Flow Fractionation (DFF) The device maintains to process 3 mL of whole blood in an hour using centrifugal forces with >90% CTC recovery. The continuous collection of sorted CTCs and short residence time in the device significantly shortens the CTCs exposure time to constant shear in the channel, thus minimizing any undesirable shear induced changes to the CTCs' phenotype. 25
Cell density-based enrichment Density gradient separation of CTCs from other cells in the blood may be performed using commercially available density gradient liquid separation kits. This process generates a layered separation of cell types based on their density. Limitations include a possible loss of CTCs due to an unwanted migration into the plasma fraction, as well as the formation of nonspecific aggregates containing CTCs at the bottom of the gradient. 26-28
Size-based cell enrichment by filtration Direct enrichment of epithelial cells by filtration is based on the observation that the vast majority of peripheral blood cells are among the smallest cells in the human body. They can be eliminated by blood filtration using polycarbonate membrane calibrated pore filters. This method is quite simple, involving one single step. 29-34
Selective size amplification (SSA) It offers advantages not only in resolving the trade-off between recovery rate and purity—optimizing both—but also in reducing the mechanical stress exerted upon the CTCs during filter transit. The major reasons for this enhanced performance include distinctive size discrimination between WBCs and CTCs as well as the benefits of the solid microbeads mitigating cell deformation within the MOA filter gap. 35
3D microfiltration This device consists of two parylene membrane layers with pores and a gap precisely defined by photolithography. The positions of the pores are shifted between the top and bottom membranes. The bottom membrane supports captured cells and minimizes stress, which is concentrated on the cell membrane and sustains cell viability during filtration under very low pressure. 36
ISET (isolation by size of epithelial tumor cells) Size-based enrichments of CTCs have been described by membrane filter devices such as ISET. 29,30,37
NanoVelcro CTC Chip By switching device temperature in a physiologically endurable range (i.e. 4-37°C), thermoresponsive conformational changes of nanosubstrate-grafted polymer brushes alter the accessibility of capture agent to specifically capture (37°C) and release (4°C) CTCs to give viable CTCs in desired purity. 38
Telomescan A novel cancer detection platform that measures telomerase activity from viable CTCs captured on a parylene-C slot microfilter. Using a constant low pressure delivery system, the new microfilter platform is capable of cell capture from 1 mL of whole blood in less than 5 min, achieving 90% capture efficiency. Addition of an adenovirus-containing GFP to peripheral blood assay, incubation with cancer cells allows precise enumeration and visualization of CTCs. 39,40
Affinity based assays
CellSearch The only FDA-approved technology for CTC detection is based on immunomagnetic enrichment. It employs an immunomagnetic enrichment step to isolate cells that express the epithelial cells' adhesion molecule (EpCAM). Additionally, to be identified as a CTC, the cell must contain a nucleus, express cytoplasmic cytokeratin, and have a diameter larger than 5 μm. This technology has demonstrated the prognostic utility of enumerating and monitoring CTC counts in patients with metastatic breast, prostate, and colorectal cancers. Semiautomated analyzer enriches CTCs with ferrofluid nanoparticles coated with anti-EpCAM antibodies, then CD45-, CK8+, CK18+ and CK19+ cells are counted by a four-color semiautomated fluorescence microscope. 41-47
CTC-chip Capture of CTCs by EpCAM-coated microposts under strict manipulation of velocity and shear force. It enables a high yield of capture (median, 50 CTCs per milliliter) and purity (ranging from 10% to 50%), most likely caused by the gentle one-step microfluidic processing. Captured cells remain viable after capture, although the absence of cell fixation currently limits the time allowed between blood collection and microfluidic analysis to a few hours. Captured CTCs are visualized by staining with antibodies against cytokeratin or tissue-specific markers. For CTC enumeration, the entire device is imaged at multiple planes using a semiautomated imaging system while on-chip lysis allows for DNA and RNA extraction and molecular analyses. Nuclear fluorescence and CK stain for positive selection and CD45 stain for negative selection; CTCs captured are directly recognized by cameras, based on morphology, viability and expression of tumor markers. It has a total of 98% cell viability and high detection rate, making further analysis possible. 48-51
Herringbone-chip Its chambers were made of transparent materials, allowing imaging of the captured CTCs, using traditional histopathological stains, transmitted light microscopy and immunofluorescence-conjugated antibodies. The Herringbone-chip has been tested in metastatic prostate and lung cancer patients, verifying results with those obtained with the CTC-chip method of analysis: Herringbone-chip shows higher flow rates and higher CTC capture efficiency and purity. 52,53
AdnaTest Immunomagnetic separation with EpCAMs and MUC1 coupled antibodies; further analysis by isolation, direct lysis, mRNA extraction and application of multiplexed RT-PCR for HER2, EpCAM and MUC-1. Possibility to characterize CTCs for stem cells and epithelial mesenchymal transition. It lacks flexibility and automation. Cannot enumerate cells due to lysis. False-positive results due to the expression of the same antigens on nontumor cells; false-negative results due to loss of antigens on CTCs. 54-58
EPISPOT (Epithelial ImmunoSPOT) Detects only viable cells after the depletion of CD45- positive cells, and was introduced for CTC analyses. Avoiding direct contact with the target cells, this technique assesses the presence of CTCs based on secreted or released proteins during 48h of short-term culture. 59,60
Collagen Adhesion Matrix (CAM) assay It has been reported in breast, prostate and ovarian cancer: CAM ingestion and epithelial immunostaining identifies CTCs based on their invasive properties in vitro. 61
MAINTRAC A specialized laser scanning cytometer provides another EpCAM-based approach. 62
Biocept Utilizes proprietary antibody based enrichment technique to detect rare CTCs found in a patient’s blood sample (1 in 1 million). 63
Photoacoustic flowmetry Making use of the broadband absorption spectrum of melanin, it has been tested to detect melanoma cells and has been combined with nanoparticles targeting cell surface antigens to broaden its applicability in CTC detection. 64,65
MagSweeper A magnetic stir bar coated with an antibody to EpCAM. The device can process 9 mL of blood per hour and purified cells of interest can be individually selected for subsequent molecular analysis, since the MagSweeper technology preserves cell function and does not perturb gene expression. 66-70
DEPArray (Silicon Biosystems) An automated system with fluorescence imaging that captures cells in a chip based upon electric movement. DEPArray achieved 100% purity, eliminating all white blood cells (WBC), in the isolation of a mixed population of tumor cell lines downstream of CellSearch enrichment. This enabled molecular profiling of pure tumor cells from whole blood spiked tumor cell lines. 71
CTC-iChip Whole blood is now processed through a microscale system at speeds of 8 mL/hour while preserving the high sensitivity afforded by microfluidic isolation techniques. Furthermore, rapid and gentle isolation of CTCs, as well as their collection in suspension, increases the integrity of these cells and their RNA quality. Moreover, the system can be run in either a positive selection or a negative depletion mode. The robustness of this platform was demonstrated by staining CTCs per clinical pathology protocols, which yielded high-quality diagnostic images. The negCTC-iChip allowed for isolation of CTCs from a nonepithelial cancer (melanoma) and from cancer that has undergone EMT and lost virtually all detectable EpCAM expression (TNBC). Limitations: low CTC purity to facilitate routine molecular analyses of CTCs and total blood volume needed to enable early cancer detection. 72
Negative depletion CTC enrichment strategy Relies on the removal of normal cells using immunomagnetic separation in the blood of cancer patients. This method is based on the combination of magnetic and fluid forces in an axial, laminar flow in long cylinders placed in quadrapole magnets. 73
Millennium Sciences IsoFlux The blood is centrifuged. Immunomagnetic particles are added to the PMBC layer that target the cells of interest. It is then transferred into a microfluidic cartridge. A permanent magnet is placed on the roof of the channel to attract the labeled target cells. 74
Cynvenio Liquid Biopsy platform This platform uses high throughput sheath flow microfluidics for the positive selection of CTC populations. Furthermore the platform quantitatively isolates cells useful for molecular methods such as detection of mutations in 50 oncogenes. 75
Photoacoustic flowmetry Making use of the broadband absorption spectrum of melanin, it has been tested to detect melanoma cells and has been combined with nanoparticles targeting cell surface antigens to broaden its applicability in CTC detection. 64,65
Cytometric assays
FACS (Fluorescence-activated cell sorting) It enables simultaneous analysis of multiparameters, such as size, viability, DNA content and expression of different markers for CTCs detection. It has high specificity, but low sensitivity. 76-79
Slide-based automated scanning microscopes (Ikoniscope and Ariol) Maximizes scanner utilization with brightfield-multi-channel fluorescent and FISH capture capabilities. Introduced for detecting CTCs; still need to be validated. 80,81
Fiber-optic array-scanning technology (FAST) It involves deposition of nucleated cells on the surface of a large glass slide, with scanning of cells positive for epithelial or tumor-specific antigens. Ultra-high-speed automated digital microscopy using fiber-optic array scanning technology has been developed to detect CTCs mounted directly on a slide that are labeled by antibodies with fluorescent conjugates. 82,83
Multiphoton intravital flow cytometry It detects CTCs tagged in vivo using injected fluorescent ligands as they flow through the vasculature. 48
Functional based assays
Folate-conjugated nanotubes and magnetic uPA-conjugated nanoparticles + photoacoustic flow cytometry assay This assay has been validated in a mouse model. Most cancer cells express folate receptors and high levels of the urokinase plasminogen activator (uPA) receptors. Thus, CTCs can be dually targeted in vivo (in the bloodstream) with folate-conjugated nanotubes and magnetic uPA-conjugated nanoparticles and subsequently detected with two-color photoacoustic flow cytometry. Future studies on humans will inform whether this new platform can diagnose tumor cell dissemination. 84
Molecular detection
RT-PCR It allows the analysis of expression of candidate genes specific to epithelial tumor cells by mRNA evaluation, often combined with other enrichment techniques. It has high sensitivity. Disadvantages include RNA degradation, false-positive results due to nonspecific amplification, contaminations and pseudogenes; false negative results due to low expression levels. 85-91
Enzyme-linked immunosorbent spot technology Immunological assay based on the ELISA (identification and count of cells able to secrete proteins like MUC1 and CK19 in short-term culture), after immunomagnetic depletion of CD45+ cells. Disadvantages include: CTC isolation not possible, further analysis not available, need of active protein secretion and technically challenging. 55-57
QuantiGene ViewRNA CTC Platform CTC is isolated by size; sample is prepared (fixed, baked, permeabilized and protease digested) to enable RNA accessibility. Target RNA Probe Sets are hybridized followed by a sequential hybridization of signal amplification and detection components. Once processed, filters are transferred to a microscope slide for image processing and analysis. 92
CK19 mRNA Assay Assays targeting specific mRNAs are the most widely used alternative to immunological assays to identify CTCs. In breast cancer, the CK19 mRNA has been most frequently used in clinical studies. Many transcripts (e.g. encoding CK18, CK19, CK20, Mucin-1, prostate-specific antigen and carcinoembryonic antigen), however, are also expressed at low levels in normal blood and BM cells 93, so quantitative RT-PCR assays with validated cutoff values are required to overcome this problem. 93

Additionally, there are various pre-analytical conditions to take into consideration such as the time interval between blood draw and assay, type of tube utilized, use of fixatives or preservatives, and temperature. Our group has focused on pre-analytic variables pertaining to the amplification of picogram quantities of RNA as well as time to CTC assay influencing the number of cells recovered23,94.

EMT

According to recent findings, more invasive CTCs may lose their epithelial antigens by the EMT process, rendering detection via EpCAM based technologies challenging. Through the EMT process, epithelial cells lose cell-cell contacts and cell polarity, downregulate epithelial-associated genes, acquire mesenchymal gene expression, and undergo major changes in their cytoskeleton. This cellular process culminates in a mesenchymal appearance and increased motility and invasiveness95,96.

In the actuation of the EMT program, epithelial markers such as E-cadherin and cytokeratins are downregulated, whereas mesenchymal markers, such as vimentin and fibronectin are frequently overexpressed. Furthermore, intermediate phenotypes between epithelial and mesenchymal differentiation are described to co-exist in human cancer97. Cancer cells can be induced to undergo EMT by several signaling pathways, most notably those involving the cooperation between TGF-β 1 signaling and oncogenic RAS or other receptor tyrosine kinases, as well as Wnt, Notch, and the signaling activated by Hedgehog98, which may be potential drug targets.

In addition, certain transcription factors (TF), including TWIST1, SNAIL1, SLUG, ZEB1, and FOXC2 can induce EMT in mammary epithelial cells and/or breast cancer cells99. Moreover, blocking the expression of TWIST1 in the highly metastatic 4T1 murine mammary cell line reduced both metastatic burden and the number of CTCs in mice bearing xenograft mammary tumors, thus linking EMT, metastasis, and the presence of CTCs99. These findings suggest that the expression of epithelial-cell surface markers, such as EpCAM, may not be optimal for detecting a heterogeneous population of CTCs including those with a mesenchymal phenotype. Evidence exists that EpCAM-negative CTCs might have undergone EMT54.

Raimondi et al.95 investigated the expression of EMT and stem cell markers in CTCs from 92 metastatic breast cancer patients. CTCs were isolated by CELLection Dynabeads coated with the monoclonal antibody toward EpCAM. Samples positive for CTCs presence (CD45-/CK+) were evaluated for the expression of ER alpha, HER2, ALDH1, vimentin, and fibronectin. Samples negative for CTCs presence (CD45-/CK-) were also evaluated for the expression of vimentin and fibronectin, used as markers of EMT. In 34% of patients, they detected cells with negative CK/CD45 expression but positive expression of vimentin and fibronectin95. This mesenchymal phenotype is more common for the basal-like molecular subtype of breast cancer100. However, further analysis is needed to classify CTC expression with known subtypes of breast cancer for both validation and clinical relevance.

One study on mRNA analysis shows that the low abundance of EpCAM expression is associated with increased vimentin in basal-like breast cancer101. Another study further demonstrated dynamic changes in epithelial and mesenchymal composition in circulating breast cancer cells with mixed probes using RNA in situ hybridization, particularly in CTC clusters. Although rare in the circulation compared with single CTCs, CTC-clusters have 23 to 50-fold increased metastatic potential. CTC clusters are oligoclonal in origin, derived from groupings of primary tumor cells that together enter the circulation102. They can be captured via several techiniques such as microfluidic devices, including a specifically designed cluster chip which operates at sub-physiological flow rates, preventing these highly deformable cell groupings from squeezing through small pores under higher flow pressures103, direct precipitation of all blood cells onto specially prepared slides, followed by high-speed microscopic scanning104, as well as enrichment based techniques105,106.

Claudin-low tumor cells

The most commonly used markers to identify carcinoma cells are cytokeratins, which have become a standard detection marker for CTCs56,57,93,107. Since hematopoietic cells rarely express cytokeratin proteins detectable by immunostaining108,109, specific detection can be achieved for cells from epithelial tumors, such as breast carcinomas from blood cells. However, if tumor cells undergo EMT to migrate and invade the body, the cytoskeleton is rearranged and epithelial markers such as E-cadherin, claudins, and cytokeratins are downregulation110. Many detection methods using cytokeratin antibodies are not able to detect all CTCs secondary to this downregulation111. If indeed not all CTCs are detected by the common detection methods, false-negative results would hamper the clinical implementation of CTCs as a diagnostic marker. To improve CTC detection of claudin low tumors, the use of CD146 and CD49f as selection markers has been shown to improve detection of those cell lines showing EMT-features112. Our group has recently demonstrated that EpCAM based capture of CTC mimics using a panel of ten cell lines recovered all intrinsic subtypes of breast cancer except the claudin-low group23. Furthermore, detection techniques not dependent on epithelial markers may be used for detection of these cells, see Table 1.

Apoptotic CTCs

As tumors increases in volume, so too does the cellular turnover and hence the number of apoptotic and necrotic cells. Under normal physiologic circumstances, apoptotic and necrotic cellular remains are cleared by infiltrating phagocytes. In theory this does not happen efficiently within the tumoral mass, leading to CTC escape as well as the accumulation of cellular debris and its inevitable release into the circulation(ctDNA)113,114.

Although many CTCs migrate early from the primary tumor into the circulation, many may be cleared within a few days115. According to the seed and soil hypothesis, the survival of these cells depends on their distinctive biologic characteristics as well as on the microenvironment at the secondary site116. Only rare subsets of cells finally succeed in establishing a cross-talk with stromal cells in secondary organs that promotes tumor cell survival, angiogenesis, and metastatic outgrowth. This could be related to the induction of senescence in CTCs, to a low proliferative potential, or to the presence of apoptotic cells. Mehes et al.117 first noted apoptotic cells significantly contribute to the circulating tumor cell fraction in breast cancer patients after he performed a detailed microscopic analysis. An "inclusion type" cytokeratin staining pattern and nuclear condensation indicated apoptosis in the CTCs isolated. Furthermore, apoptosis-related DNA strand breaks could be demonstrated by applying the TdT-uridine nick end labeling assay in these cells117. The monoclonal antibody targeting the neoepitope M30, revealed by caspase cleavage of CK18 in early apoptosis, has also been shown to be a marker for apoptotic CTCs118. Kallergi et al.119 noted the presence of exclusively apoptotic CTCs in a patient may represent a favorable prognostic factor, whereas the preponderance of proliferating cells (determined by Ki-67) could be related to poor patient outcome in breast cancer. The median percentage of apoptotic CTCs per patient in his study was lower in patients with advanced disease compared with those with early disease. Adjuvant chemotherapy reduced both the number of CTCs per patient and the number of proliferating CTCs. Kallergi et al.119 concluded that the detection of CTCs that survive despite adjuvant therapy implies that CTC elimination should be attempted using agents targeting their distinctive molecular characteristics.

CTC enumeration clinical trials

Numerous studies including Bidard et al.12 pooled analysis reveal that CTC count is independently prognostic of progression-free survival and overall survival in metastatic breast cancer (MBC) patients120. Other trials by Lucci et al.9 and a meta analysis by Zhang et al.10 have further demonstrated CTC detection is prognostic in non-metastatic breast cancer as well11.

Additionally, The Southwest Oncology group (SWOG) completed a prospective clinical trial (SO500) to evaluate the utility in changing therapy versus maintaining therapy in MBC patients who have elevated CTCs after one cycle of a new first-line chemotherapy121. Although CTC status was prognostic, simple enumeration did not predict for a benefit in switching to an alternate cytotoxic therapy, likely because simple enumeration has a low predictive value and cannot predict a specific course of treatment. Other limitations to this trial include chemo/cross resistance and statistical limitations.

The early DETECT trials revealed that serial CTC measurements before and after chemotherapy are prognostic and can be used to monitor treatment benefit122,123. Subsequent trials being conducted by the DETECT study group are evaluating targeted agents based on phenotypes of CTC in patients with MBC124. To fully realize the potential of CTCs as a useful biomarker, simple enumeration is not sufficient. Detailed molecular profiling of CTCs must be performed to inform the discovery of therapeutic predictors and actionable targets, important steps forward in personalizing medicine.

CTC molecular profiling

Several mechanisms exist to profile CTCs including protein expression, genomic, gene expression and functional characterization125,126. They have been cultured in vitro, used in xenotransplantation in vivo16,127, and assessed using next-generation sequencing (NGS)16,17. Several studies have shown that recurrent/metastatic breast tumors and CTCs show discordance in hormone receptors (ER and PR) and HER2 expression from the primary tumor112,128-130. Furthermore, patients with TNBC do not have identified targeted therapies and are currently treated by chemotherapy alone and have very limited treatment options upon distant metastasis. Therefore, comparing genomic alterations in cancer-related genes between primary, metastatic breast tumors and CTCs and may provide insights into mechanisms of tumor metastases and drug resistance that may help guide targeted therapeutics131. Recent technical advancements in massively parallel or high-throughput NGS offer multigene mutational profiling that provides comprehensive genetic information on breast cancer molecular pathology, paving the road for newer and more effective therapeutic targets131. NGS has changed the perspective on genome profiling, since DNA sequencing has the potential to identify structural changes, including gene fusions and even point mutations, in addition to copy number. Furthermore, NGS using RNA Seq would permit full genomic and transcriptomic profiling, maximizing the ability to study individual genes, pathway perturbations, and chromosomal losses/gains. Additionally, NGS panels for inherited cancer has increased the potential to identify pathogenic variants in genes that would not typically have been tested (in breast cancer, genes other than BRCA1/2) and to offer more tailored preventative management to patients and family members132. Each sequencing technique has specific advantages and disadvantages (Table 2)133-144.

Table2.

Advantages and disadvantages of DNA sequencing

Method Basic technique Advantages Disadvantages Reference
Sanger-chain termination method (first generation sequencing) Fluorescent dye-labeled bases; DNA fragments separated by capillary electrophoresis High sensitivity, gold standard complete sequence Very time consuming; cannot detect deletions, translocations or copy number changes 133-136
Pyrosequencing-sequencing by synthesis method Chemiluminescent detection; DNA polymerase synthesizes cDNA to a target template; pyrophosphate release is detected at each base addition More sensitive than Sanger; provides % of mutated vs. wild-type DNA; works well with fragmented DNA Short read length limits technique to hot spots. Limited accuracy at detecting changes in homopolymer runs. Scalability is limited compared with other NGS methods 136-140
Allele-specific RT-PCR Primers span DNA sites of interest and probes detect specific mutations Very high sensitivity widely used for clinical testing for oncogene mutations in CRC and NSCLC Scalability constraints limit application to hot spots 136,141-143
RT-PCR melting curve analysis Heterogeneous DNA PCR products melt at different temperatures than homogenous DNA/PCR products High sensitivity provides percentage of mutated versus wild-type DNA Often difficult to resolve differences in melt curves. Difficult to standardize. Multiplex capability is limited 136,144

In addition there are a variety of NGS platforms available including second generation, sequencing of an ensemble of DNA molecules with wash-and-scan techniques and third generation, sequencing single DNA molecules without the need to halt between read steps, whether enzymatic or otherwise145 (Table 3)136,146-160.

Table3.

Next-generation sequencing platforms

Device Method Generation Major uses Run time Length of reads Reference
CCD: charge-coupled device; NA: not applicable; NGS: next-generation sequencing; WES: whole exome sequencing; WGS: whole genome sequencing; SNP: single nucleotide polymorphism; WTA: whole transcriptome analysis.
Illumina Hi Seq 4000 Flow cell-based, reversible dye termination and four-color optical imaging 2nd WES WGS SNP WTA <1-3.5 days 2×150 bp 136,146-151
Roche 454 pyro-sequencing GS FLX+ Emulsion PCR with bead-based pyro-sequencing and CCD light imaging 2nd WGS WTA Targeted Seq SNP 10 h Up to 1000 bp 136,146,151-153
Life Technologies SOLiD 5500xl Sequential dinucleotide ligation; flow cell-based four-color optical imaging 2nd WES WGS SNP 6 days 2×60 bp 136,146,150,151,154
Life Tech-nologies Ion Torrent PGM Semi-conductor based nonoptical detection; standard dNTP sequencing chemistry 2nd Targeted panel Demand Sequenc-ing <1 day 400 bp 136,151,155
Complete Genomics CGA platform DNA nanoball arrays coupled with combinatori-al probe anchor ligation 2nd WGS Slow: weeks to months ~70 bases 136,151,156
Pacific Biosciences PacBioRSII Zero-mode waveguide, individual polymerase; single molecule sequencing using fluorescent dNTPs 3rd Long read, full microbial genome <1-6 h 10-15 kb 136,151,157,158
Seqll Heliscope True Single Molecule Sequencing, massive parallel sequencing without amplification 3rd Targeted WGS WES WTA ~8 h 25-60 bp 136,151,156,159
Nanopore Minion Nanopore exonuclease sequencing, no need for amplification 3rd Long read, full microbial 1 min-48 h 230-300kb 160
Illumina Hi Seq 4000 Flow cell-based, reversible dye termination and four-color optical imaging 2nd WES WGS SNP WTA <1-3.5 days 2×150 bp 136,146-151
Roche 454 pyro-sequencing GS FLX+ Emulsion PCR with bead-based pyro-sequencing and CCD light imaging 2nd WGS WTA Targeted Seq SNP 10 h Up to 1000 bp 136,146,151-153
Life Technologies SOLiD 5500xl Sequential dinucleotide ligation; flow cell-based four-color optical imaging 2nd WES WGS SNP 6 days 2×60 bp 136,146,150,151,154
Life Tech-nologies Ion Torrent PGM Semi-conductor based nonoptical detection; standard dNTP sequencing chemistry 2nd Targeted panel Demand Sequenc-ing <1 day 400 bp 136,151,155
Complete Genomics CGA platform DNA nanoball arrays coupled with combinatorial probe anchor ligation 2nd WGS Slow: weeks to months ~70 bases 136,151,156
Pacific Biosciences PacBioRSII Zero-mode waveguide, individual polymerase; single molecule sequencing using fluorescent dNTPs 3rd Long read, full microbial genome <1-6 h 10-15 kb 136,151,157,158
Seqll Heliscope True Single Molecule Sequencing, massive parallel sequencing without amplification 3rd Targeted WGS WES WTA ~8 h 25-60 bp 136,151,156,159
Nanopore Minion Nanopore exonuclease sequencing, no need for amplification 3rd Long read, full microbial 1 min-48 h 230-300kb 160

The advantages of sequencing CTCs include: (1) the ability to fully sequence large numbers of genes (hundreds to thousands) in a single blood test and simultaneously detect deletions, insertions, copy number variations, translocations, fusion events and exome-wide base substitutions (including known "hot-spot mutations") in all known cancer-related genes in a cell(s) most likely to cause metastases, which may be important for predicting response to select therapies; (2) results from sequencing are presented as either positive or negative and do not appear as gradations as with immunofluorescence; (3) analysis can be automated to reduce interpreter bias; and (4) allows us to evaluate tumor heterogeneity161.

Molecular profiling of CTCs has been shown to predict response and or resistance to therapy. RNA sequencing via CTC-iChip revealed activating mutations in PIK3CA, FGFR2, and ESR-1 in breast cancer patients. Drug sensitivity testing revealed that the selective estrogen receptor modulators (SERMs) tamoxifen and raloxifene, and the selective ER degrader (SERD) fulvestrant, were ineffective in ESR-1 mutant cells, either alone or in the clinically approved combination with inhibitors of the phosphatidylinositol 3-kinase-mammalian target of rapamycin (PI3K-mTOR) pathway (everolimus). Cultured CTCs were highly sensitive to the PIK3CA inhibitor BYL719 and the FGFR2 inhibitor AZD4547127.

A trial conducted by Gradilone et al.162 revealed that in MBC patients the presence of CTCs expressing multidrug-resistance-related proteins, and ALDH1, is predictive of response to chemotherapy. Liu et al.163 reported on a patient affected by chemo-refractory metastatic HER2-positive breast cancer receiving lapatinib. The detection and characterization of CTCs was evaluated and depletion of the EGFR-positive CTC pool in the blood was associated with tumor response, whereas disease progression was related to a recurrence in CTCs, indicating expression of EGFR may predict response to lapatinib-based treatments.

Sequencing techniques also have several disadvantages including: (1) limitations with regards to sensitivity that make single cell analysis difficult164,165, including amplification bias166; and (2) leukocyte contamination which may cause false positive/negative results due to impaired visualization of amplified transcripts. However, several groups have been able to overcome these challenges via various techniques, such as micromanipulation, laser-capture microdissection, and flow cytometry with promising results167,168. For instance Magbanua et al.169 demonstrated that it is feasible to isolate CTCs away from hematopoietic cells with high purity through immunomagnetic enrichment followed by fluorescence activated cell sorting (IE-FACS) and profile them via array comparative genomic hybridization analysis. With newer amplification techniques such as whole genome amplification, in combination with NGS and FACS, some researchers have shown it is possible to profile copy number in a single cell in various cancer types, including in breast cancer170-173. Single cell analysis has identified clinically significant genomic disparity between primary tumors and CTCs3,174-176 therefore analysis of CTCs in this fashion may surmount the plaguing concern of tumor heterogeneity.

Tumor heterogeneity

Tumor heterogeneity can exist within the same site of disease or between sites of disease177,178 and contamination with adjacent normal tissue can lead to false negative results or confound successful targeted therap179. As Navin and Hicks168 pointed out, tumor heterogeneity exists in breast cancer because the malignant cells often arise from ductal tissue and are constrained by the duct structure until they begin to invade surrounding stromal tissue. They exhibit regions of growth, regions of hypoxia and necrosis, and regions of interaction with blood vessels and lymph ducts. Given these variable selection pressures, tumor cells within a single mass are not identical180. This heterogeneity in the molecular characterization of single cells, including CTCs could potentially identify rare driver mutations that are diluted out when profiling bulk tumors, including their stroma168. Navin et al.172 revealed that by comparing multiple single-cell copy number profiles, they could provide highly accurate measures of genomic heterogeneity within solid tumors. Furthermore, by comparing multiple single-cell profiles, they showed that it was possible to reconstruct the evolutionary lineage of a tumor and understand its pattern of progression.

In addition, Heitzer et al.181 has described a CTC sequencing approach for single-cell analysis, suggesting that contamination with normal cells may be reduced. Secondly, although the sequencing of CTCs does not change the fact that the readout is still at the level of the genome, we anticipate that, especially in cases in which metastatic lesions are inaccessible, CTC sequencing will strengthen conclusions regarding mutations that are drivers versus those that are passengers as they may be present not only in the primary/metastatic lesion but also in the cells that were able to escape into the circulation. The conserved nature of these mutations may suggest an important functional contribution to disease progression. Heitzer et al.181 demonstrated the sequencing of CTCs may identify relevant private mutations, present but not detected in tumor tissue. In contrast to CTCs, single cell analysis is not possible when evaluating ctDNA as a biomarker.

CtDNA

Circulating DNA was first identified by Mandel and Metais182 in 1948 but no association with disease was hypothesized. Only 30 years later, in 1977, Leon et al.183 found circulating tumor DNA (ctDNA) in plasma of patients affected by lung cancer. Cell free DNA (cfDNA) was identified in the peripheral blood of healthy individuals however patients with cancerous tumors have higher quantities and detection is associated with poorer prognosis4,184. Due to reduced cell turnover and more efficient removal of defective cells from the circulation by phagocytes, the concentration of cfDNA is lower in healthy individuals. CtDNA, or soluble nucleic acids shed into the bloodstream in patients with cancer, are linked to apoptosis and necrosis of cancer cells in the tumor microenvironment. Secretion within exosomes has also been suggested as a potential source of cfDNA. The released tumor cells are usually phagocytosed by macrophages which engulf necrotic cells and release digested DNA into the tissue environment185. Additionally, tumors usually harbor a mixture of different cancer cell clones that justify their genomic and epigenomic heterogeneity, together with other normal cell types, such as hematopoietic and stromal cells. Thus, during tumor development and turnover both tumor-derived and wild-type cfDNA can be released into the blood. Consequently, the proportion of cfDNA that originates from tumor cells varies according to the state and size of the tumor. The amount of cfDNA is also affected by physiological factors such as clearance, degradation and filtering events of the blood and lymphatic system. Nucleic acids have a half-life in the circulation ranging from 15 minutes to several hours depending on the rate of clearance from the blood by the spleen, liver and kidney186,187.

The characteristics of ctDNA suggest they are in part derived from apoptotic cells. For instance, when the lengths of ctDNA strands are measured, they often assume the classic ladder pattern in integer multiples of 180 base pairs, characteristic of the apoptotic process. In fact, most ctDNA fragments measure between 180 and 200 base pairs, suggesting that apoptosis likely produces the majority of ctDNA in circulation113,114.

Pre-analytic considerations

Cell lysis can occur hours after venipuncture when using standard tubes for blood collection, leading to an increase in contaminating cellular DNA that may hinder analysis of ctDNA. This normal DNA from dying blood cells will contaminate the specimens and dilute ctDNA. This is the reason why plasma is recommended, since its analysis avoids the simultaneous testing of material originally associated with hematopoietic cells. However, centrifugation speed, amount of blood collected, a delay in blood processing and storage temperature (contaminating cells can be removed after storage at -20°C)can influence the amount of cfDNA extracted from plasma187-189. Toro et al.190 conducted a study comparing the properties of two cell stabilizing reagents in blood samples from MBC patients and measured genome equivalents of plasma DNA by droplet digital PCR. They compared wild type PIK3CA genome equivalents and also assayed for two PIK3CA hotspot mutations, E545K and H1047R. Their results demonstrated that blood stored for 7 days in BCT tubes did not show evidence of cell lysis, whereas PAX gene tubes showed an order of magnitude increase in genome equivalents, indicative of considerable cellular lysis190. Several kits exist for DNA extraction with some recent studies comparing the kits. The QIAamp DNA Blood Mini and Circulating Nucleic Acid kits from Qiagen (Hilden, Germany) gave the highest recovery of cfDNA191,192. Currently there is no uniform standardization and each study has its own protocol regarding pre-analytical conditions for processing specimens.

Furthermore, levels of cfDNA might reflect physiological and pathological processes that are not tumor-specific. CfDNA yields are higher in patients with malignant lesions than in patients with resected tumors, but increased levels have also been quantified in patients with benign lesions, inflammatory diseases and tissue trauma193. Please see Table 4 for the advantages and limitations of CTCs vs. cfDNA5-20,113,114,183-187.

Table4.

Advantages and limitations of CTCs vs. cfDNA

Advantages Limitations
CTC Allows in depth assessment of viable metastatic tumor cells, DNA, RNA, and protein; may reflect mechanisms of resistance; may reflect treatment efficacy; allows functionally analysis (in vitro/in vivo testing) Few and fragile; requires sophisticated isolation techniques; EMT may cause false negative results; tumor heterogeneity within CTCs; unclear which CTCs cause metastases
Cell free cfDNA May reflect mechanisms of resistance; may reflect treatment efficacy; sensitive biomarker; available from other sources: urine, plasma, ascitic fluid Present in inflammatory states and aging (not cancer specific); contamination with DNA from lysed cells; unclear if clinically relevant given being released from dying tumor cells; predetermined somatic alterations need to be identified; does not permit single cell analysis

CfDNA detection and molecular profiling

Several sensitive techniques exist to detect such mutations in cfDNA including droplet digital PCR (ddPCR)194, CAPP-Seq195 and Guardant360196, Sanger sequencing197, amplification refractory mutation system (ARMS)198, pyrosequencing199, pyrophophorolysis-activated polymerization (PAP)200, tagged-amplicon deep sequencing (TAM-Seq)201, beads, emulsion, amplification and magnetics (BEAMing)202, modified semi-nested or nested methylation-specific PCR203, utilization of microsatellite biomarkers followed by post-PCR product analysis using capillary array electrophoresis204 among others. It is possible to evaluate nucleic acids for fusion events, copy number variants, indels, and single nucleotide polymorphisms.

NGS has been developed to target the genome at various scales (whole genome205, whole exome206, and targeted panels207-209) and are a key component toward realizing personalized care in oncology. A genome-wide screen for copy number changes has the advantage that it is an untargeted approach, which does not require any prior knowledge about characteristics of the primary tumor genome or its metastatic deposits. Panel-based targeted sequencing of selected cancer genes or mutational hotspots is a popular approach, and can generally be classified on the basis of the target enrichment method used (e.g. amplicon PCR versus hybridization capture), sequencing chemistry, and scale of the sequencing platform. There is considerable variability across targeted NGS panels implemented in different clinical laboratories in terms of the number and identities of genes tested, and sample throughput. The number of genes targeted varies depending on the technology. Most vendor solutions are generally amplicon PCR-based and target selected mutation hotspots in 1 to 50 cancer genes, and these include the Ion- Torrent AmpliSeq (Life Technologies, Carlsbad, CA)210, and the Illumina TruSeq Amplicon Cancer Panel (Illumina, San Diego, CA)207 among others. Targeted enrichment using hybridization capture can incorportate more genes without risk a of problematic primer. Via a customized hybrization capture technique the MSK-IMPACT panel targets 341 genes208,209. Foundation Medicine targets 287 genes (however on, formalin-fixed, paraffin-embedded (FFPE)tissue, use of these genomic targets as a ctDNA assay is currently being validated on clinicaltrials.gov protocol NCT02620527)211,212.

CtDNA clinical trials

For instance, Schiavon et al.215 conducted a study on 171 breast cancer patients with advanced breast disease evaluating ESR1 mutations by ultra high-sensitivity multiplex digital polymerase chain reaction assaysin ctDNA. ESR1 mutations were found to be a major mechanism of resistance to aromatase inhibitors, as evidenced by shorter progression-free survival (PFS) in those patients with the mutation, and more common in those with metastatic disease.

Additionally in the BOLERO-2 trial, Chandarlapaty et al.216 demonstrated that patients with the ESR1 mutation D538G in ctDNA derived a 3.1 month PFS benefit from the addition of everolimus (an mTOR inhibitor), whereas those with the Y537S mutation showed stable PFS.

Additionally, it was demonstrated that persistent declines in the levels of the AKT1 E17K mutation in ctDNA is associated with durable tumor regression to the AKT inhibitor AZD5363217.

Another trial assessing mutations in ctDNA as an indicator of therapeutic resistance is the phase 3 BELLE 2 trial. PIK3CA status in cfDNA predicted efficacy of the inhibitor buparlisib in combination with fulvestrant in postmenopausal women with endocrine resistant advanced breast cancer. In patients with PIK3CA mutations identified by ctDNA, median PFS was 7.0 months in patients treated with buparlisib vs. 3.2 months in those treated with placebo. In those with non-mutant PIK3CA by liquid biopsy, there was no difference in PFS218.

Murtaza et al.206 presented an extensive comparison of biopsy and plasma samples collected from a MBC patient over a 3-year clinical course, in which whole exome sequencing (WES) and deep sequencing of plasma DNA and tumor biopsies revealed resistance to targeted agents such as lapatinib, a tyrosine kinase inhibitor of EGFR (epidermal growth factor receptor) and HER2. Thus deep sequencing of plasma DNA, applied to selected samples with high tumor burden in blood, may help identify the mutations associated with drug resistance193,219.

The role of ctDNA in evaluating minimal residual disease has also been evaluated by Schiavon et al.215 in a prospective cohort of 55 early breast cancer patients in which detection of ctDNA in plasma after completion of apparently curative treatment-either at a single postsurgical time point or with serial follow-up plasma samples-predicted metastatic relapse with high accuracy. The concept of tumor dormancy may in part account for minimal residual disease in patients.

Tumor dormancy

Experimental biology studies have documented two types of tumor dormancy: cellular dormancy with solitary cancer cells held in cell-cycle arrest and micrometastatic dormancy with a balanced state of apoptosis and proliferation220. Dormant CTCs appear to account for late disease recurrence even after prolonged disease free survival in breast cancer221. CTCs are detectable in as many as one-third of patients who are prolonged survivors222. Payne et al.223 confirmed the ability to detect minimal residual disease in breast cancer patients by assessing CTCs, DTCs and ctDNA. Shaw et al.224 revealed that copy number variations (CNV) detected in ctDNA, mirror the primary tumor up to 12 years after diagnosis in one-fifth of breast cancer patients. SNP/CNV analysis of ctDNA was able to distinguish between patients with breast cancer and healthy controls during routine follow-up. Given these findings, the detection of minimal residual disease in breast cancer via CTCs or ctDNA is possible, and molecular profiling may be able to identify therapeutic targets in the future.

Circulating exosomes

Exosomes are small membrane vesicles (30-100 nm) containing functional biomolecules (proteins, lipids, RNA and DNA) that are released by most cell types upon fusion of multivesicular bodies with the plasma membrane, presumably as a vehicle for cell-free intercellular communication225. Circulating tumor derived exosomes were demonstrated to prepare a favorable microenvironment at future metastatic sites and mediate non-random patterns of metastasis via vascular leakiness, stromal cell education at organotropic sites, bone-marrow-derived cell education, and recruitment necessary to complete pre-metastatic niche evolution226-228. Furthermore, exosomal DNA was found to reflect the mutational status of parental tumor cells. Therefore future studies on exosomes as a biomarkers may be useful not only to predict metastatic propensity in breast cancer229, but also to determine organ sites of future metastasis227 and to monitor treatment response230.

Circulating microRNAs

MicroRNAs (miRs) are evolutionary conserved, small non-coding RNA molecules consisting of approximately 22 nucleotides. One miR has binding affinity to hundreds of different mRNAs and hence, miRs are involved in the regulation of various cellular processes, such as development, differentiation, and proliferation231. MiRs are released into the blood circulation by apoptotic and necrotic cells or active secretion232. They can exist either extracellularly, in association with Argonaut proteins, or in exosomes in the blood circulation225. To date, numerous miRs have been identified, and varied circulating miRs concentrations are found in different breast cancer subtypes and some are associated with prognosis and treatment response233-235. Given there is already quite a substantial literature in this emerging field, miRs are beyond the scope of this review.

Circulating biomarker characterization and clinical implications

Detailed molecular profiling of tumors and circulating tumor biomarkers such as CTCs and ctDNA has already been changing the therapeutic landscape of cancer across several cancer types. Currently it is standard clinical practice to evaluate genetic alterations in many cancers. For instance, in non-small cell lung cancer (NSCLC), the detection of the somatic activating mutations in EGFR predicts for sensitivity to EGFR inhibitors such as Tarceva49,236,237. In melanoma, the presence of BRAF V600E mutation predicts sensitivity to BRAF inhibitors such as vemurafenib238. In colon cancer the presence of KRAS mutations has been established as a negative predictive marker for treatment with EGF receptor (EGFR) inhibitors239,240. However, unlike drug therapy matched somatic mutations in NSCLC, melanoma, and colon cancer, drug therapy matched to the presence of a somatic mutation has yet to be robustly established as a standard approach in breast cancer.

In breast cancer, mutations in the estrogen receptor ESR1 are a potential driver of endocrine resistance215,216,241. Furthermore multiple alterations in genes related to the PI3K-mTOR pathway may confer resistance to trastuzumab218,242. A recent trial in breast cancer established a CTC PD-L1 assay that can be used for liquid biopsy in future clinical trials for stratification and monitoring of cancer patients undergoing immune checkpoint blockade243.

As demonstrated in other tumor types and in breast cancer, a better understanding of mechanisms of treatment resistance and biomarkers to predict for response are required to reduce cancer related mortality. The key to standardizing the assessment of somatic mutations in breast cancer would be to integrate tumor, CTCs and ctDNA sequencing data into a systematic and broad-based plan with clinical endpoints. Currently there are several trials investigating anti-HER2 therapies in patients with HER2 negative primary tumors, yet HER2 positive CTCs such as the DETECT 3 trial244, evaluating lapatinib, CareMore-Trastuzumab244 evaluating trastuzumab, a monoclonal antibody directed against HER2 in breast cancer and CirCe TDM-1 evaluating TDM-1, trastuzumab linked to the cytotoxic agent DM-1245. Incorporating molecular profiling of CTCs/ctDNA in trials such as Molecular Analysis for Therapy Choice (MATCH), a genomic pre-screening study, the National Cancer Institute (NCI) aims to explore the efficacy of existing targeted agents against specific molecular aberrations, and to evaluate if these same therapies have comparable activities across different tumor subtypes246. One challenge to this approach is that there are relatively few targeted therapies anticipated to be effective in some solid tumors, such as breast cancer. NGS of circulating biomarkers may help discover new potential rational targeted therapy options.

Conclusions

Profiling primary tumor biology has led to many important insights about the biology of breast cancer, yet relatively few signatures predict treatment sensitivity. A critical barrier to progress may be that MBC and the circulating cells leading to macrometastasis are inherently different than primary breast cancer. In contrast, primary tumors shed CTCs and cfDNA into the systemic circulation where they are subject to metastatic inefficiency and potentially after a period of tumor dormancy may lead to macrometastases247,248. The ultimate goal of detecting and molecularly characterizing CTCs and ctDNA would be to shift the current clinical paradigm that incorporates relatively few prognostic/predictive clinical markers (estrogen receptor, progesterone receptor, HER2) to a landscape in which clinicians could measure tumor biology in real time by profiling CTCs or ctDNA in order to discover therapeutic targets, detect minimal residual disease, and predict response to treatment. In order to attain such a goal, CTCs and ctDNA sequencing data must be integrated into systematic and broad-based trials with clinical endpoints. Several trials discussed in this paper strive toward this goal, with the DETECT 3, BOLERO-2, BELLE 2 trials and others focusing on this very concept by evaluating therapeutic response based on phenotypes of CTCs or ctDNA in patients with breast cancer124,216,218. In summary, once further established, the molecular characterization of circulating biomarkers has the potential to provide the ideal mechanism of personalizing treatment in breast cancer for the best clinical results by predicting treatment response, evaluating minimal residual disease and allowing for the rational selection of targeted therapies.

Conflict of interest statement

No potential conflicts of interest are disclosed.

References

  1. Lang JM, Casavant BP, Beebe DJ. Circulating tumor cells: getting more from less. Sci Transl Med. 2012;4: 141ps13. doi: 10.1126/scitranslmed.3004261. [DOI] [PubMed] [Google Scholar]
  2. Pantel K, Alix-Panabières C. Real-time liquid biopsy in cancer patients: fact or fiction. Cancer Res. 2013;73: 6384–8. doi: 10.1158/0008-5472.CAN-13-2030. [DOI] [PubMed] [Google Scholar]
  3. Heitzer E, Auer M, Gasch C, Pichler M, Ulz P, Hoffmann EM, et al. Complex tumor genomes inferred from single circulating tumor cells by Array-CGH and next-generation sequencing. Cancer Res. 2013;73: 2965–75. doi: 10.1158/0008-5472.CAN-12-4140. [DOI] [PubMed] [Google Scholar]
  4. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368: 1199–209. doi: 10.1056/NEJMoa1213261. [DOI] [PubMed] [Google Scholar]
  5. Alix-Panabieres C, Schwarzenbach H, Pantel K. Circulating tumor cells and circulating tumor DNA. Annu Rev Med. 2012;63: 199–215. doi: 10.1146/annurev-med-062310-094219. [DOI] [PubMed] [Google Scholar]
  6. Ashworth TR. "A case of cancer in which cells similar to those in the tumours were seen in the blood after death". Aust Med J. 1869;14: 146–7. [Google Scholar]
  7. Fisher ER, Turnbull RB. The cytologic demonstration and significance of tumor cells in the mesenteric venous blood in patients with colorectal carcinoma. Surg Gynecol Obstet. 1955;100: 102–8. [PubMed] [Google Scholar]
  8. Cristofanilli M. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. Semin Oncol. 2006; 33, Supplement 9: 9-14.
  9. Lucci A, Hall CS, Lodhi AK, Bhattacharyya A, Anderson AE, Xiao L, et al. Circulating tumour cells in non-metastatic breast cancer: a prospective study. Lancet Oncol. 2012;13: 688–95. doi: 10.1016/S1470-2045(12)70209-7. [DOI] [PubMed] [Google Scholar]
  10. Zhang L, Riethdorf S, Wu G, Wang T, Yang K, Peng G, et al. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. 2012;18: 5701–10. doi: 10.1158/1078-0432.CCR-12-1587. [DOI] [PubMed] [Google Scholar]
  11. Rack B, Schindlbeck C, Jückstock J, Andergassen U, Hepp P, Zwingers T, et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J Natl Cancer Inst. 2014;106: 106. doi: 10.1093/jnci/dju106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bidard FC, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol. 2014;15: 406–14. doi: 10.1016/S1470-2045(14)70069-5. [DOI] [PubMed] [Google Scholar]
  13. Ross AA, Cooper BW, Lazarus HM, Mackay W, Moss TJ, Ciobanu N, 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. 1993;82: 2605–10. [PubMed] [Google Scholar]
  14. Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192: 373–82. doi: 10.1083/jcb.201010021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Lang JE, Hall CS, Singh B, Lucci A. Significance of micrometastasis in bone marrow and blood of operable breast cancer patients: research tool or clinical application. Expert Rev Anticancer Ther. 2007;7: 1463–72. doi: 10.1586/14737140.7.10.1463. [DOI] [PubMed] [Google Scholar]
  16. Helzer KT, Barnes HE, Day L, Harvey J, Billings PR, Forsyth A. Circulating tumor cells are transcriptionally similar to the primary tumor in a murine prostate model. Cancer Res. 2009;69: 7860–6. doi: 10.1158/0008-5472.CAN-09-0801. [DOI] [PubMed] [Google Scholar]
  17. Marchetti A, Del Grammastro M, Felicioni L, Malatesta S, Filice G, Centi I, et al. Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next-generation sequencing: toward a real-time liquid biopsy for treatment. PLoS One. 2014;9: e103883. doi: 10.1371/journal.pone.0103883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339: 580–4. doi: 10.1126/science.1228522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bednarz-Knoll N, Alix-Panabières C, Pantel K. Plasticity of disseminating cancer cells in patients with epithelial malignancies. Cancer Metastasis Rev. 2012;31: 673–87. doi: 10.1007/s10555-012-9370-z. [DOI] [PubMed] [Google Scholar]
  20. Kang Y, Pantel K. Tumor cell dissemination: emerging biological insights from animal models and cancer patients. Cancer Cell. 2013;23: 573–81. doi: 10.1016/j.ccr.2013.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Perou CM, Sorlie T, Eisen MB, Van De Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406: 747–52. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  22. Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98: 10869–74. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Ring A, Mineyev N, Zhu W, Park E, Lomas C, Punj V, et al. EpCAM based capture detects and recovers circulating tumor cells from all subtypes of breast cancer except claudin-low. Oncotarget. 2015;6: 44623–34. doi: 10.18632/oncotarget.5977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Moore LR, Williams PS, Nehl F, Abe K, Chalmers JJ, Zborowski M. Feasibility study of red blood cell debulking by magnetic field-flow fractionation with step-programmed flow. Anal Bioanal Chem. 2014;406: 1661–70. doi: 10.1007/s00216-013-7394-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo RA, Tan DS-W, et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci Rep. 2013;3: 1259. doi: 10.1038/srep01259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Rosenberg R, Gertler R, Friederichs J, Fuehrer K, Dahm M, Phelps R, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry. 2002;49: 150–8. doi: 10.1002/cyto.10161. [DOI] [PubMed] [Google Scholar]
  27. 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 Cancer Res. 2003;162: 149–55. doi: 10.1007/978-3-642-59349-9_13. [DOI] [PubMed] [Google Scholar]
  28. Danova M, Torchio M, Mazzini G. Isolation of rare circulating tumor cells in cancer patients: technical aspects and clinical implications. Expert Rev Mol Diagn. 2011;11: 473–85. doi: 10.1586/erm.11.33. [DOI] [PubMed] [Google Scholar]
  29. 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. J Chromatogr A. 2007;1162: 154–61. doi: 10.1016/j.chroma.2007.05.064. [DOI] [PubMed] [Google Scholar]
  30. Wong NS, Kahn HJ, Zhang L, Oldfield S, Yang LY, Marks A, et al. Prognostic significance of circulating tumour cells enumerated after filtration enrichment in early and metastatic breast cancer patients. Breast Cancer Res Treat. 2006;99: 63–9. doi: 10.1007/s10549-006-9181-4. [DOI] [PubMed] [Google Scholar]
  31. De Giorgi V, Pinzani P, Salvianti F, Panelos J, Paglierani M, Janowska A, et al. Application of a filtration- and isolation-by-size technique for the detection of circulating tumor cells in cutaneous melanoma. J Invest Dermatol. 2010;130: 2440–7. doi: 10.1038/jid.2010.141. [DOI] [PubMed] [Google Scholar]
  32. Tan SJ, Yobas L, Lee GY, Ong CN, Lim CT. Microdevice for the isolation and enumeration of cancer cells from blood. Biomed Microdevices. 2009;11: 883–92. doi: 10.1007/s10544-009-9305-9. [DOI] [PubMed] [Google Scholar]
  33. Chudziak J, Burt DJ, Mohan S, Rothwell DG, Mesquita B, Antonello J, et al. Clinical evaluation of a novel microfluidic device for epitope-independent enrichment of circulating tumour cells in patients with small cell lung cancer. Analyst. 2016;141: 669–78. doi: 10.1039/C5AN02156A. [DOI] [PubMed] [Google Scholar]
  34. Hvichia G, Parveen Z, Wagner C, Janning M, Quidde J, Stein A, et al. A novel microfluidic platform for size and deformability based separation and the subsequent molecular characterization of viable circulating tumor cells. Int J Cancer. 2016 Jan 20. doi: 10.1002/ijc.30007. [Epub ahead of print]
  35. Kim MS, Sim TS, Kim YJ, Kim SS, Jeong H, Park JM, et al. SSA-MOA: a novel CTC isolation platform using selective size amplification (SSA) and a multi-obstacle architecture (MOA) filter. Lab Chip. 2012;12: 2874–80. doi: 10.1039/c2lc40065k. [DOI] [PubMed] [Google Scholar]
  36. Zheng S, Lin HK, Lu B, Williams A, Datar R, Cote RJ, et al. 3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood. Biomed Microdevices. 2011;13: 203–13. doi: 10.1007/s10544-010-9485-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Pinzani P, Salvadori B, Simi L, Bianchi S, Distante V, Cataliotti L, et al. Isolation by size of epithelial tumor cells in peripheral blood of patients with breast cancer: correlation with real-time reverse transcriptase-polymerase chain reaction results and feasibility of molecular analysis by laser microdissection. Hum Pathol. 2006;37: 711–8. doi: 10.1016/j.humpath.2006.01.026. [DOI] [PubMed] [Google Scholar]
  38. Lu YT, Zhao LB, Shen QL, Garcia MA, Hou S, Song M, et al. NanoVelcro chip for CTC enumeration in prostate cancer patients. Methods. 2013;64: 144–52. doi: 10.1016/j.ymeth.2013.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Xu T, Lu B, Tai YC, Goldkorn A. A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter. Cancer Res. 2010;70: 6420–6. doi: 10.1158/0008-5472.CAN-10-0686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kojima T, Hashimoto Y, Watanabe Y, Kagawa S, Uno F, Kuroda S, et al. A simple biological imaging system for detecting viable human circulating tumor cells. J Clin Invest. 2009;119: 3172–81. doi: 10.1172/JCI38609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. 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. Clin Cancer Res. 2004;10: 6897–904. doi: 10.1158/1078-0432.CCR-04-0378. [DOI] [PubMed] [Google Scholar]
  42. Cristofanilli M. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. Semin Oncol. 2006; 33, Supplement 9: 9-14.
  43. Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol. 2010: 617421.
  44. Kraan J, Sleijfer S, Strijbos MH, Ignatiadis M, Peeters D, Pierga JY, et al. External quality assurance of circulating tumor cell enumeration using the CellSearch(®) system: a feasibility study. Cytometry B Clin Cytom. 2011;80: 112–8. doi: 10.1002/cyto.b.20573. [DOI] [PubMed] [Google Scholar]
  45. Mostert B, Kraan J, Bolt-De Vries J, Van Der Spoel P, Sieuwerts AM, Schutte M, et al. Detection of circulating tumor cells in breast cancer May improve through enrichment with anti-CD146. Breast Cancer Res Treat. 2011;127: 33–41. doi: 10.1007/s10549-010-0879-y. [DOI] [PubMed] [Google Scholar]
  46. Obermayr E, Sanchez-Cabo F, Tea M, Singer CF, Krainer M, Fischer MB, et al. Assessment of a six gene panel for the molecular detection of circulating tumor cells in the blood of female cancer patients. BMC Cancer. 2010;10: 666. doi: 10.1186/1471-2407-10-666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Pantel K, Alix-Panabières C, Riethdorf S. Cancer micrometastases. Nat Rev Clin Oncol. 2009;6: 339–51. doi: 10.1038/nrclinonc.2009.44. [DOI] [PubMed] [Google Scholar]
  48. 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: 1235–9. doi: 10.1038/nature06385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura CV, et al. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med. 2008;359: 366–77. doi: 10.1056/NEJMoa0800668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Sequist LV, Nagrath S, Toner M, Haber DA, Lynch TJ. The CTC-chip: an exciting new tool to detect circulating tumor cells in lung cancer patients. J Thorac Oncol. 2009;4: 281–3. doi: 10.1097/JTO.0b013e3181989565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. 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. Proc Natl Acad Sci U S A. 2010;107: 18392–7. doi: 10.1073/pnas.1012539107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Sheng W, Ogunwobi OO, Chen T, Zhang J, George TJ, Lui C, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Labchip. 2014;14: 89–98. doi: 10.1039/c3lc51017d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192: 373–82. doi: 10.1083/jcb.201010021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Fehm T, Hoffmann O, Aktas B, Becker S, Solomayer EF, Wallwiener D, et al. Detection and characterization of circulating tumor cells in blood of primary breast cancer patients by RT-PCR and comparison to status of bone marrow disseminated cells. Breast Cancer Res. 2009;11: R59. doi: 10.1186/bcr2349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Ross JS, Slodkowska EA. Circulating and disseminated tumor cells in the management of breast cancer. Am J Clin Pathol. 2009;132: 237–45. doi: 10.1309/AJCPJI7DEOLKCS6F. [DOI] [PubMed] [Google Scholar]
  56. Alix-Panabieres C, Riethdorf S, Pantel K. Circulating tumor cells and bone marrow micrometastasis. Clinical Cancer Research. 2008;14: 5013–21. doi: 10.1158/1078-0432.CCR-07-5125. [DOI] [PubMed] [Google Scholar]
  57. Pantel K, Alix-Panabieres C. The clinical significance of circulating tumor cells. Nat Clin Pract Oncol. 2007;4: 62–3. doi: 10.1038/ncponc0737. [DOI] [PubMed] [Google Scholar]
  58. Hauch S, Zimmermann S, Lankiewicz S, Zieglschmid V, Böcher O, Albert WH. The clinical significance of circulating tumour cells in breast cancer and colorectal cancer patients. Anticancer Res. 2007;27: 1337–41. [PubMed] [Google Scholar]
  59. Schwarzenbach H, Alix-Panabières C, Müller I, Letang N, Vendrell JP, Rebillard X, et al. Cell-free tumor DNA in blood plasma as a marker for circulating tumor cells in prostate cancer. Clin Cancer Res. 2009;15: 1032–8. doi: 10.1158/1078-0432.CCR-08-1910. [DOI] [PubMed] [Google Scholar]
  60. Alix-Panabières C, Vendrell JP, Slijper M, Pellé O, Barbotte E, Mercier G, et al. Full-length cytokeratin-19 is released by human tumor cells: a potential role in metastatic progression of breast cancer. Breast Cancer Res. 2009;11: R39. doi: 10.1186/bcr2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Lu J, Fan T, Zhao Q, Zeng W, Zaslavsky E, Chen JJ, et al. Isolation of circulating epithelial and tumor progenitor cells with an invasive phenotype from breast cancer patients. International Journal of Cancer. 2010;126: 669–83. doi: 10.1002/ijc.24814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Pachmann K, Camara O, Kavallaris A, Krauspe S, Malarski N, Gajda M, et al. Monitoring the response of circulating epithelial tumor cells to adjuvant chemotherapy in breast cancer allows detection of patients at risk of early relapse. J Clin Oncol. 2008;26: 1208–15. doi: 10.1200/JCO.2007.13.6523. [DOI] [PubMed] [Google Scholar]
  63. Mayer JA, Pham T, Wong KL, Scoggin J, Sales EV, Clarin T, et al. FISH-based determination of HER2 status in circulating tumor cells isolated with the microfluidic CEE™ platform. Cancer Genet. 2011;204: 589–95. doi: 10.1016/j.cancergen.2011.10.011. [DOI] [PubMed] [Google Scholar]
  64. Weight RM, Dale PS, Viator JA. Detection of circulating melanoma cells in human blood using photoacoustic flowmetry. Conf Proc IEEE Eng Med Biol Soc. 2009;2009: 106–9. doi: 10.1109/IEMBS.2009.5335145. [DOI] [PubMed] [Google Scholar]
  65. Galanzha EI, Shashkov EV, Spring PM, Suen JY, Zharov VP. In vivo, noninvasive, label-free detection and eradication of circulating metastatic melanoma cells using two-color photoacoustic flow cytometry with a diode laser. Cancer Res. 2009;69: 7926–34. doi: 10.1158/0008-5472.CAN-08-4900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Talasaz AH, Powell AA, Huber DE, Berbee JG, Roh KH, Yu W, et al. Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc Natl Acad Sci U S A. 2009;106: 3970–5. doi: 10.1073/pnas.0813188106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Tong X, Yang L, Lang JC, Zborowski M, Chalmers JJ. Application of immunomagnetic cell enrichment in combination with RT-PCR for the detection of rare circulating head and neck tumor cells in human peripheral blood. Cytometry B Clin Cytom. 2007;72: 310–23. doi: 10.1002/cyto.b.20177. [DOI] [PubMed] [Google Scholar]
  68. Tkaczuk KH, Goloubeva O, Tait NS, Feldman F, Tan M, Lum ZP, et al. The significance of circulating epithelial cells in Breast Cancer patients by a novel negative selection method. Breast Cancer Res Treat. 2008;111: 355–64. doi: 10.1007/s10549-007-9771-9. [DOI] [PubMed] [Google Scholar]
  69. Balasubramanian P, Yang L, Lang JC, Jatana KR, Schuller D, Agrawal A, et al. Confocal images of circulating tumor cells obtained using a methodology and technology that removes normal cells. Mol Pharm. 2009;6: 1402–8. doi: 10.1021/mp9000519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Yang L, Lang JC, Balasubramanian P, Jatana KR, Schuller D, Agrawal A, et al. Optimization of an enrichment process for circulating tumor cells from the blood of head and neck cancer patients through depletion of normal cells. Biotechnol Bioeng. 2009;102: 521–34. doi: 10.1002/bit.22066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Medoro G, Gross S, Manaresi N, Sergio M, Fontana F, Gianni S, et al. Use of the DEPArray platform to detect, isolate, and molecularly characterize pure tumor cells from peripheral blood samples enriched using the CellSearch system, 2011: 29.
  72. 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. Sci Transl Med. 2013;5: 179ra47. doi: 10.1126/scitranslmed.3005616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lustberg M, Jatana KR, Zborowski M, Chalmers JJ. Emerging technologies for CTC detection based on depletion of normal cells. Recent Results Cancer Res. 2012;195: 97–110. doi: 10.1007/978-3-642-28160-0_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. 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. Transl Oncol. 2013;6: 528–38. doi: 10.1593/tlo.13367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Winer-Jones JP, Vahidi B, Arquilevich N, Fang C, Ferguson S, Harkins D, et al. Circulating tumor cells: clinically relevant molecular access based on a novel CTC flow cell. PLoS One. 2014;9: e86717. doi: 10.1371/journal.pone.0086717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Simpson SJ, Vachula M, Kennedy MJ, Kaizer H, Coon JS, Ghalie R, et al. Detection of tumor cells in the bone marrow, peripheral blood, and apheresis products of breast cancer patients using flow cytometry. Exp Hematol. 1995;23: 1062–8. [PubMed] [Google Scholar]
  77. Cruz I, Ciudad J, Cruz JJ, Ramos M, Gómez-Alonso A, Adansa JC, et al. Evaluation of multiparameter flow cytometry for the detection of breast cancer tumor cells in blood samples. Am J Clin Pathol. 2005;123: 66–74. doi: 10.1309/WP3QWKVJFYDHHXQD. [DOI] [PubMed] [Google Scholar]
  78. Allan AL, Vantyghem SA, Tuck AB, Chambers AF, Chin-Yee IH, Keeney M. Detection and quantification of circulating tumor cells in mouse models of human breast cancer using immunomagnetic enrichment and multiparameter flow cytometry. Cytometry A. 2005;65: 4–14. doi: 10.1002/cyto.a.20132. [DOI] [PubMed] [Google Scholar]
  79. Da BJ, Ortyn WE, Liang L, Venkatachalam V, Morrissey P. Cellular image analysis and imaging by flow cytometry. Clin Lab Med. 2007;27: 653–70. doi: 10.1016/j.cll.2007.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Ntouroupi TG, Ashraf SQ, Mcgregor SB, Turney BW, Seppo A, Kim Y, et al. Detection of circulating tumour cells in peripheral blood with an automated scanning fluorescence microscope. Br J Cancer. 2008;99: 789–95. doi: 10.1038/sj.bjc.6604545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Deng G, Herrler M, Burgess D, Manna E, Krag D, Burke JF. Enrichment with anti-cytokeratin alone or combined with anti-EpCAM antibodies significantly increases the sensitivity for circulating tumor cell detection in metastatic breast cancer patients. Breast Cancer Res. 2008;10: R69. doi: 10.1186/bcr2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Krivacic RT, Ladanyi A, Curry DN, Hsieh HB, Kuhn P, Bergsrud DE, et al. A rare-cell detector for cancer. Proc Natl Acad Sci U S A. 2004;101: 10501–4. doi: 10.1073/pnas.0404036101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Marrinucci D, Bethel K, Bruce RH, Curry DN, Hsieh B, Humphrey M, et al. Case study of the morphologic variation of circulating tumor cells. Hum Pathol. 2007;38: 514–9. doi: 10.1016/j.humpath.2006.08.027. [DOI] [PubMed] [Google Scholar]
  84. Galanzha EI, Shashkov EV, Kelly T, Kim JW, Yang L, Zharov VP. In vivo magnetic enrichment and multiplex photoacoustic detection of circulating tumour cells. Nat Nanotechnol. 2009;4: 855–60. doi: 10.1038/nnano.2009.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Smith B, Selby P, Southgate J, Pittman K, Bradley C, Blair GE. Detection of melanoma cells in peripheral blood by means of reverse transcriptase and polymerase chain reaction. Lancet. 1991;338: 1227–9. doi: 10.1016/0140-6736(91)92100-G. [DOI] [PubMed] [Google Scholar]
  86. Sergeant G, Penninckx F, Topal B. Quantitative RT-PCR detection of colorectal tumor cells in peripheral blood--a systematic review. J Surg Res. 2008;150: 144–52. doi: 10.1016/j.jss.2008.02.012. [DOI] [PubMed] [Google Scholar]
  87. Ignatiadis M, Xenidis N, Perraki M, Apostolaki S, Politaki E, Kafousi M, et al. Different prognostic value of cytokeratin-19 mRNA positive circulating tumor cells according to estrogen receptor and HER2 status in early-stage breast cancer. J Clin Oncol. 2007;25: 5194–202. doi: 10.1200/JCO.2007.11.7762. [DOI] [PubMed] [Google Scholar]
  88. Xenidis N, Ignatiadis M, Apostolaki S, Perraki M, Kalbakis K, Agelaki S, et al. Cytokeratin-19 mRNA-positive circulating tumor cells after adjuvant chemotherapy in patients with early breast cancer. J Clin Oncol. 2009;27: 2177–84. doi: 10.1200/JCO.2008.18.0497. [DOI] [PubMed] [Google Scholar]
  89. Ignatiadis M, Kallergi G, Ntoulia M, Perraki M, Apostolaki S, Kafousi M, et al. Prognostic value of the molecular detection of circulating tumor cells using a multimarker reverse transcription-PCR assay for cytokeratin 19, mammaglobin A, and HER2 in early breast cancer. Clin Cancer Res. 2008;14: 2593–600. doi: 10.1158/1078-0432.CCR-07-4758. [DOI] [PubMed] [Google Scholar]
  90. Lambrechts AC, Bosma AJ, Klaver SG, Top B, Perebolte L, Van' T Veer LJ, et al. Comparison of immunocytochemistry, reverse transcriptase polymerase chain reaction, and nucleic acid sequence-based amplification for the detection of circulating breast cancer cells. Breast Cancer Res Treat. 1999;56: 219–31. doi: 10.1023/a:1006261731125. [DOI] [PubMed] [Google Scholar]
  91. Panteleakou Z, Lembessis P, Sourla A, Pissimissis N, Polyzos A, Deliveliotis C, et al. Detection of circulating tumor cells in prostate cancer patients: methodological pitfalls and clinical relevance. Mol Med. 2009;15: 101–14. doi: 10.2119/molmed.2008.00116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Yu M, Ting DT, Stott SL, Wittner BS, Ozsolak F, Paul S, et al. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature. 2012;487: 510–3. doi: 10.1038/nature11217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Pantel K, Brakenhoff RH, Brandt B. Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer. 2008;8: 329–40. doi: 10.1038/nrc2375. [DOI] [PubMed] [Google Scholar]
  94. Lang JE, Magbanua MJ, Scott JH, Makrigiorgos GM, Wang G, Federman S, et al. A comparison of RNA amplification techniques at sub-nanogram input concentration. BMC genomics. 2009;10: 326. doi: 10.1186/1471-2164-10-326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Raimondi C, Gradilone A, Naso G, Vincenzi B, Petracca A, Nicolazzo C, et al. Epithelial-mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients. Breast Cancer Res Treat. 2011;130: 449–55. doi: 10.1007/s10549-011-1373-x. [DOI] [PubMed] [Google Scholar]
  96. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14: 818–29. doi: 10.1016/j.devcel.2008.05.009. [DOI] [PubMed] [Google Scholar]
  97. Bonnomet A, Brysse A, Tachsidis A, Waltham M, Thompson EW, Polette M, et al. Epithelial-to-mesenchymal transitions and circulating tumor cells. J Mammary Gland Biol Neoplasia. 2010;15: 261–73. doi: 10.1007/s10911-010-9174-0. [DOI] [PubMed] [Google Scholar]
  98. Moustakas A, Heldin CH. Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98: 1512–20. doi: 10.1111/j.1349-7006.2007.00550.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117: 927–39. doi: 10.1016/j.cell.2004.06.006. [DOI] [PubMed] [Google Scholar]
  100. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339: 580–4. doi: 10.1126/science.1228522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Punnoose EA, Atwal SK, Spoerke JM, Savage H, Pandita A, Yeh RF, et al. Molecular biomarker analyses using circulating tumor cells. PLoS One. 2010;5: e12517. doi: 10.1371/journal.pone.0012517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158: 1110–22. doi: 10.1016/j.cell.2014.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Sarioglu AF, Aceto N, Kojic N, Donaldson MC, Zeinali M, Hamza B, et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat Methods. 2015;12: 685–91. doi: 10.1038/nmeth.3404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Nieva J, Wendel M, Luttgen MS, Marrinucci D, Bazhenova L, Kolatkar A, et al. High-definition imaging of circulating tumor cells and associated cellular events in non-small cell lung cancer patients: a longitudinal analysis. Phys Biol. 2012;9: 016004. doi: 10.1088/1478-3975/9/1/016004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Molnar B, Ladanyi A, Tanko L, Sréter L, Tulassay Z. Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients. Clin Cancer Res. 2001;7: 4080–5. [PubMed] [Google Scholar]
  106. Cho EH, Wendel M, Luttgen M, Yoshioka C, Marrinucci D, Lazar D, et al. Characterization of circulating tumor cell aggregates identified in patients with epithelial tumors. Phys Biol. 2012;9: 016001. doi: 10.1088/1478-3975/9/1/016001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Braun S, Pantel K, Muller P, Janni W, Hepp FA, Gastroph S, et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med. 2000;342: 525–33. doi: 10.1056/NEJM200002243420801. [DOI] [PubMed] [Google Scholar]
  108. Borgen E, Naume B, Nesland JM, Kvalheim G, Beiske K, Fodstad O, et al. Standardization of the immunocytochemical detection of cancer cells in BM and blood: I. Standardization of the immunocytochemical detection of cancer cells in BM and blood: I. establishment of objective criteria for the evaluation of immunostained cells. Cytotherapy. 1999;1: 377–88. doi: 10.1080/0032472031000141283. [DOI] [PubMed] [Google Scholar]
  109. Jung R, Petersen K, Krüger W, Wolf M, Wagener C, Zander A, et al. Detection of micrometastasis by cytokeratin 20 RT-PCR is limited due to stable background transcription in granulocytes. Br J Cancer. 1999;81: 870–3. doi: 10.1038/sj.bjc.6690778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Baum B, Settleman J, Quinlan MP. Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol. 2008;19: 294–308. doi: 10.1016/j.semcdb.2008.02.001. [DOI] [PubMed] [Google Scholar]
  111. Sieuwerts AM, Kraan J, Bolt J, Van Der Spoel P, Elstrodt F, Schutte M, et al. Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumor cells. J Natl Cancer Inst. 2009;101: 61–6. doi: 10.1093/jnci/djn419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Mostert B, Kraan J, Sieuwerts AM, Van Der Spoel P, Bolt-De Vries J, Prager-Van Der Smissen WJ, et al. CD49f-based selection of circulating tumor cells (CTCs) improves detection across breast cancer subtypes. Cancer Lett. 2012;319: 49–55. doi: 10.1016/j.canlet.2011.12.031. [DOI] [PubMed] [Google Scholar]
  113. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61: 1659–65. [PubMed] [Google Scholar]
  114. Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P. About the possible origin and mechanism of circulating DNA - Apoptosis and active DNA release. Clinica Chimica Acta. 2001;313: 139–42. doi: 10.1016/S0009-8981(01)00665-9. [DOI] [PubMed] [Google Scholar]
  115. Weinberg RA. Leaving home early: reexamination of the canonical models of tumor progression. Cancer Cell. 2008;14: 283–4. doi: 10.1016/j.ccr.2008.09.009. [DOI] [PubMed] [Google Scholar]
  116. Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3: 453–8. doi: 10.1038/nrc1098. [DOI] [PubMed] [Google Scholar]
  117. Méhes G, Witt A, Kubista E, Ambros PF. Circulating breast cancer cells are frequently apoptotic. Am J Pathol. 2001;159: 17–20. doi: 10.1016/S0002-9440(10)61667-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Rossi E, Basso U, Celadin R, Zilio F, Pucciarelli S, Aieta M, et al. M30 neoepitope expression in epithelial cancer: quantification of apoptosis in circulating tumor cells by CellSearch analysis. Clin Cancer Res. 2010;16: 5233–43. doi: 10.1158/1078-0432.CCR-10-1449. [DOI] [PubMed] [Google Scholar]
  119. Kallergi G, Konstantinidis G, Markomanolaki H, Papadaki MA, Mavroudis D, Stournaras C, et al. Apoptotic circulating tumor cells in early and metastatic breast cancer patients. Mol Cancer Ther. 2013;12: 1886–95. doi: 10.1158/1535-7163.MCT-12-1167. [DOI] [PubMed] [Google Scholar]
  120. Giordano A, Giuliano M, De Laurentiis M, Eleuteri A, Iorio F, Tagliaferri R, et al. Artificial neural network analysis of circulating tumor cells in metastatic breast cancer patients. Breast Cancer Res Treat. 2011;129: 451–8. doi: 10.1007/s10549-011-1645-5. [DOI] [PubMed] [Google Scholar]
  121. Smerage JB, Barlow WE, Hortobagyi GN, Winer EP, Leyland-Jones B, Srkalovic G, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol. 2014;32: 3483–9. doi: 10.1200/JCO.2014.56.2561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Müller V, Riethdorf S, Rack B, Janni W, Fasching PA, Solomayer E, et al. Prognostic impact of circulating tumor cells assessed with the CellSearch System™ and AdnaTest Breast™ in metastatic breast cancer patients: the DETECT study. Breast Cancer Res. 2012;14: R118. doi: 10.1186/bcr3243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Pierga JY, Hajage D, Bachelot T, Delaloge S, Brain E, Campone M, et al. High Independent prognostic and predictive value of circulating tumor cells compared with serum tumor markers in a large prospective trial in first-line chemotherapy for metastatic breast cancer patients. Ann Oncol. 2012;23: 618–24. doi: 10.1093/annonc/mdr263. [DOI] [PubMed] [Google Scholar]
  124. Schramm A, Friedl TW, Schochter F, Scholz C, De Gregorio N, Huober J, et al. Therapeutic intervention based on circulating tumor cell phenotype in metastatic breast cancer: concept of the DETECT study program. Arch Gynecol Obstet. 2016;293: 271–81. doi: 10.1007/s00404-015-3879-7. [DOI] [PubMed] [Google Scholar]
  125. Ignatiadis M, Rothé F, Chaboteaux C, Durbecq V, Rouas G, Criscitiello C, et al. HER2-positive circulating tumor cells in breast cancer. PLoS One 2011; 6: e15624.
  126. Tan SC, Yiap BC. DNA, RNA, and protein extraction: the past and the present. J Biomed Biotechnol 2009: 574398.
  127. Yu M, Bardia A, Aceto N, Bersani FA, Donaldson MC, Desai R, et al. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 2014;345: 216–20. doi: 10.1126/science.1253533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Park YH, Shin HT, Jung HH, Choi YL, Ahn T, Park K, et al. Role of HER2 mutations in refractory metastatic breast cancers: targeted sequencing results in patients with refractory breast cancer. Oncotarget. 2015;6: 32027–38. doi: 10.18632/oncotarget.5184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Liedtke C, Broglio K, Moulder S, Hsu L, Kau SW, Symmans WF, et al. Prognostic impact of discordance between triple-receptor measurements in primary and recurrent breast cancer. Ann Oncol. 2009;20: 1953–8. doi: 10.1093/annonc/mdp263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Dupont Jensen J, Laenkholm AV, Knoop A, Ewertz M, Bandaru R, Liu W, et al. PIK3CA mutations May be discordant between primary and corresponding metastatic disease in breast cancer. Clin Cancer Res. 2011;17: 667–77. doi: 10.1158/1078-0432.CCR-10-1133. [DOI] [PubMed] [Google Scholar]
  131. Roy-Chowdhuri S, De Melo Gagliato D, Routbort MJ, Patel KP, Singh RR, Broaddus R, et al. Multigene clinical mutational profiling of breast carcinoma using next-generation sequencing. Am J Clin Pathol. 2015;144: 713–21. doi: 10.1309/AJCPWDEQYCYC92JQ. [DOI] [PubMed] [Google Scholar]
  132. Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, et al. Pathogenic and likely pathogenic variant prevalence among the first 10, 000 patients referred for next-generation cancer panel testing. Genet Med.2015 Dec 17. doi: 10.1038/gim.2015.166.[Epub ahead of print]
  133. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A. 1977;74: 5463–7. doi: 10.1073/pnas.74.12.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Lander ES, Int Human Genome Sequencing Consortium, Linton LA, Nusbaum C, Zody MC, Baldwin J, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409: 860–921. doi: 10.1038/35057062. [DOI] [PubMed] [Google Scholar]
  135. Human GC. Finishing the euchromatic sequence of the human genome. Nature. 2004;431: 931–45. doi: 10.1038/nature03001. [DOI] [PubMed] [Google Scholar]
  136. Cronin M, Ross JS. Comprehensive next-generation cancer genome sequencing in the era of targeted therapy and personalized oncology. Biomark Med. 2011;5: 293–305. doi: 10.2217/bmm.11.37. [DOI] [PubMed] [Google Scholar]
  137. Ronaghi M, Uhlén M, Nyrén P. A sequencing method based on real-time pyrophosphate. Science. 1998;281: 363, 365. doi: 10.1126/science.281.5375.363. [DOI] [PubMed] [Google Scholar]
  138. Ahmadian A, Ehn M, Hober S. Pyrosequencing: history, biochemistry and future. Clin Chim Acta. 2006;363: 83–94. doi: 10.1016/j.cccn.2005.04.038. [DOI] [PubMed] [Google Scholar]
  139. Marsh S. Pyrosequencing applications. Methods Mol Biol. 2007;373: 15–24. doi: 10.1385/1-59745-377-3:15. [DOI] [PubMed] [Google Scholar]
  140. Ronaghi M, Shokralla S, Gharizadeh B. Pyrosequencing for discovery and analysis of DNA sequence variations. Pharmacogenomics. 2007;8: 1437–41. doi: 10.2217/14622416.8.10.1437. [DOI] [PubMed] [Google Scholar]
  141. Higuchi R, Fockler C, Dollinger G, Watson R. Kinetic PCR analysis: real-time monitoring of DNA amplification reactions. Biotechnology (N Y) 1993;11: 1026–30. doi: 10.1038/nbt0993-1026. [DOI] [PubMed] [Google Scholar]
  142. Heid CA, Stevens J, Livak KJ, Williams PM. Real time quantitative PCR. Genome Res. 1996;6: 986–994. doi: 10.1101/gr.6.10.986. [DOI] [PubMed] [Google Scholar]
  143. Clayton SJ, Scott FM, Walker J, Callaghan K, Haque K, Liloglou T, et al. K-ras point mutation detection in lung cancer: comparison of two approaches to somatic mutation detection using ARMS allele-specific amplification. Clin Chem. 2000;46: 1929–38. [PubMed] [Google Scholar]
  144. Ririe KM, Rasmussen RP, Wittwer CT. Product differentiation by analysis of DNA melting curves during the polymerase chain reaction. Anal Biochem. 1997;245: 154–60. doi: 10.1006/abio.1996.9916. [DOI] [PubMed] [Google Scholar]
  145. Niedringhaus TP, Milanova D, Kerby MB, Snyder MP, Barron AE. Landscape of next-generation sequencing technologies. Anal Chem. 2011;83: 4327–41. doi: 10.1021/ac2010857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Mardis ER. Next-generation DNA sequencing methods. Annu Rev Genomics Hum Genet. 2008;9: 387–402. doi: 10.1146/annurev.genom.9.081307.164359. [DOI] [PubMed] [Google Scholar]
  147. Erlich Y, Mitra PP, Delabastide M, Mccombie WR, Hannon GJ. Alta-Cyclic: a self-optimizing base caller for next-generation sequencing. Nat Methods. 2008;5: 679–82. doi: 10.1038/nmeth.1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Quail MA, Kozarewa I, Smith F, Scally A, Stephens PJ, Durbin R, et al. A large genome center's improvements to the Illumina sequencing system. Nat Methods. 2008;5: 1005–10. doi: 10.1038/nmeth.1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Ding L, Wendl MC, Koboldt DC, Mardis ER. Analysis of next-generation genomic data in cancer: accomplishments and challenges. Hum Mol Genet. 2010;19: R188–96. doi: 10.1093/hmg/ddq391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Metzker ML. Sequencing technologies-the next generation. Nat Rev Genet. 2010;11: 31–46. doi: 10.1038/nrg2626. [DOI] [PubMed] [Google Scholar]
  151. Church GM. Genomes for all. Sci Am. 2006;294: 46–54. doi: 10.1038/scientificamerican0106-46. [DOI] [PubMed] [Google Scholar]
  152. Tawfik DS, Griffiths AD. Man-made cell-like compartments for molecular evolution. Nat Biotechnol. 1998;16: 652–6. doi: 10.1038/nbt0798-652. [DOI] [PubMed] [Google Scholar]
  153. Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature. 2005;437: 376–80. doi: 10.1038/nature03959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Shendure J, Porreca GJ, Reppas NB, Lin X, Mccutcheon JP, Rosenbaum AM, et al. Accurate multiplex polony sequencing of an evolved bacterial genome. Science. 2005;309: 1728–32. doi: 10.1126/science.1117389. [DOI] [PubMed] [Google Scholar]
  155. Rusk N. Torrents of sequence. Nat Meth 2011; 8: 44.
  156. Drmanac R, Sparks AB, Callow MJ, Halpern AL, Burns NL, Kermani BG, et al. Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays. Science. 2010;327: 78–81. doi: 10.1126/science.1181498. [DOI] [PubMed] [Google Scholar]
  157. Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequencing from single polymerase molecules. Science. 2009;323: 133–8. doi: 10.1126/science.1162986. [DOI] [PubMed] [Google Scholar]
  158. Chin CS, Sorenson J, Harris JB, Robins WP, Charles RC, Jean-Charles RR, et al. The origin of the Haitian cholera outbreak strain. N Engl J Med. 2011;364: 33–42. doi: 10.1056/NEJMoa1012928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Jonsson P, Coarfa C, Mesmar F, Raz T, Rajapakshe K, Thompson JF, et al. Single-Molecule sequencing reveals Estrogen-Regulated clinically relevant lncRNAs in breast cancer. Mol Endocrinol. 2015;29: 1634–45. doi: 10.1210/me.2015-1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Ip CL, Loose M, Tyson JR, De Cesare M, Brown BL, Jain M, et al. MinION analysis and reference consortium: phase 1 data release and analysis, 2015: 1075.
  161. Brouwer A, De Laere B, Peeters D, Peeters M, Salgado R, Dirix L, et al. Evaluation and consequences of heterogeneity in the circulating tumor cell compartment. Oncotarget.2016 Mar 9. doi: 10.18632/oncotarget.8015.[Epub ahead of print]
  162. Gradilone A, Naso G, Raimondi C, Cortesi E, Gandini O, Vincenzi B, et al. Circulating tumor cells (CTCs) in metastatic breast cancer (MBC): prognosis, drug resistance and phenotypic characterization. Ann Oncol. 2011;22: 86–92. doi: 10.1093/annonc/mdq323. [DOI] [PubMed] [Google Scholar]
  163. Liu Z, Fusi A, Schmittel A, Tinhofer I, Schneider A, Keilholz U. Eradication of EGFR-positive circulating tumor cells and objective tumor response with lapatinib and capecitabine. Cancer Biol Ther. 2010;10: 860–4. doi: 10.4161/cbt.10.9.13323. [DOI] [PubMed] [Google Scholar]
  164. Mostert B, Jiang Y, Sieuwerts AM, Wang H, Bolt-De Vries J, Biermann K, et al. KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. International Journal of Cancer. 2013;133: 130–41. doi: 10.1002/ijc.27987. [DOI] [PubMed] [Google Scholar]
  165. Khan ZA, Jonas SK, Le-Marer N, Patel H, Wharton RQ, Tarragona A, et al. P53 mutations in primary and metastatic tumors and circulating tumor cells from colorectal carcinoma patients. Clin Cancer Res. 2000;6: 3499–504. [PubMed] [Google Scholar]
  166. Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J, et al. Interactive analysis and assessment of single-cell copy-number variations. Nat Methods. 2015;12: 1058–60. doi: 10.1038/nmeth.3578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Lowes LE, Allan AL. Recent advances in the molecular characterization of circulating tumor cells. Cancers (Basel) 2014;6: 595–624. doi: 10.3390/cancers6010595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Navin N, Hicks J. Future medical applications of single-cell sequencing in cancer. Genome Med. 2011;3: 31. doi: 10.1186/gm247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Magbanua MJ, Sosa EV, Roy R, Eisenbud LE, Scott JH, Olshen A, et al. Genomic profiling of isolated circulating tumor cells from metastatic breast cancer patients. Cancer Res. 2013;73: 30–40. doi: 10.1158/0008-5472.CAN-11-3017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Fuhrmann C, Schmidt-Kittler O, Stoecklein NH, Petat-Dutter K, Vay C, Bockler K, et al. High-resolution array comparative genomic hybridization of single micrometastatic tumor cells. Nucleic Acids Res. 2008;36: e39. doi: 10.1093/nar/gkn101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. 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: 441–53. doi: 10.1016/j.ccr.2008.04.005. [DOI] [PubMed] [Google Scholar]
  172. Navin N, Kendall J, Troge J, Andrews P, Rodgers L, Mcindoo J, et al. Tumour evolution inferred by single-cell sequencing. Nature. 2011;472: 90–4. doi: 10.1038/nature09807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Peeters DJ, Van Dam PJ, Van Den Eynden GG, Rutten A, Wuyts H, Pouillon L, et al. Detection and prognostic significance of circulating tumour cells in patients with metastatic breast cancer according to immunohistochemical subtypes. Br J Cancer. 2014;110: 375–83. doi: 10.1038/bjc.2013.743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Pestrin M, Salvianti F, Galardi F, De Luca F, Turner N, Malorni L, et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol. 2015;9: 749–57. doi: 10.1016/j.molonc.2014.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Lohr JG, Adalsteinsson VA, Cibulskis KA, Rosenberg M, Cruz-Gordillo PA, Zhang CZ, et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat Biotechnol. 2014;32: 479–84. doi: 10.1038/nbt.2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. 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 Mol Med. 2014;6: 1371–86. doi: 10.15252/emmm.201404033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Roukos DH. Next-generation, genome sequencing-based biomarkers: concerns and challenges for medical practice. Biomark Med. 2010;4: 583–6. doi: 10.2217/bmm.10.70. [DOI] [PubMed] [Google Scholar]
  178. Schrijver I, Aziz N, Farkas DH, Furtado M, Gonzalez AF, Greiner TC, et al. Opportunities and challenges associated with clinical diagnostic genome sequencing: a report of the Association for Molecular Pathology. J Mol Diagn. 2012;14: 525–40. doi: 10.1016/j.jmoldx.2012.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Russnes HG, Navin N, Hicks J, Borresen-Dale AL. Insight into the heterogeneity of breast cancer through next-generation sequencing. J Clin Invest. 2011;121: 3810–8. doi: 10.1172/JCI57088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Navin NE, Hicks J. Tracing the tumor lineage. Mol Oncol. 2010;4: 267–83. doi: 10.1016/j.molonc.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Heitzer E, Auer M, Gasch C, Pichler M, Ulz P, Hoffmann EM, et al. Complex tumor genomes inferred from single circulating tumor cells by Array-CGH and next-generation sequencing. Cancer Res. 2013;73: 2965–75. doi: 10.1158/0008-5472.CAN-12-4140. [DOI] [PubMed] [Google Scholar]
  182. Mandel P, Metais P. Comptes rendus des seances de la Societe de biologie et de ses filiales. 1948; 142: 241-3.
  183. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37: 646–50. [PubMed] [Google Scholar]
  184. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11: 426–37. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
  185. Sidransky D. Nucleic acid-based methods for the detection of cancer. Science. 1997;278: 1054–9. doi: 10.1126/science.278.5340.1054. [DOI] [PubMed] [Google Scholar]
  186. Heitzer E, Auer M, Ulz P, Geigl JB, Speicher MR. Circulating tumor cells and DNA as liquid biopsies. Genome Med. 2013;5: 73. doi: 10.1186/gm477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61: 112–23. doi: 10.1373/clinchem.2014.222679. [DOI] [PubMed] [Google Scholar]
  188. Swinkels DW, Wiegerinck E, Steegers EA, De Kok JB. Effects of blood-processing protocols on cell-free DNA quantification in plasma. Clin Chem. 2003;49: 525–6. doi: 10.1373/49.3.525. [DOI] [PubMed] [Google Scholar]
  189. Chan KC, Yeung SW, Lui WB, Rainer TH, Lo YM. Effects of preanalytical factors on the molecular size of cell-free DNA in blood. Clin Chem. 2005;51: 781–4. doi: 10.1373/clinchem.2004.046219. [DOI] [PubMed] [Google Scholar]
  190. Toro PV, Erlanger B, Beaver JA, Cochran RL, Vandenberg DA, Yakim E, et al. Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA. Clin Biochem. 2015;48: 993–8. doi: 10.1016/j.clinbiochem.2015.07.097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Devonshire AS, Whale AS, Gutteridge A, Jones G, Cowen S, Foy CA, et al. Towards standardisation of cell-free DNA measurement in plasma: controls for extraction efficiency, fragment size bias and quantification. Anal Bioanal Chem. 2014;406: 6499–512. doi: 10.1007/s00216-014-7835-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Page K, Guttery DS, Zahra N, Primrose L, Elshaw SR, Pringle JH, et al. Influence of plasma processing on recovery and analysis of circulating nucleic acids. PLoS One. 2013;8: e77963. doi: 10.1371/journal.pone.0077963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775: 181–232. doi: 10.1016/j.bbcan.2006.10.001. [DOI] [PubMed] [Google Scholar]
  194. Castellanos-Rizaldos E, Paweletz C, Song C, Oxnard GR, Mamon H, Jänne PA, et al. Enhanced ratio of signals enables digital mutation scanning for rare allele detection. J Mol Diagn. 2015;17: 284–92. doi: 10.1016/j.jmoldx.2014.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. 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. Nat Med. 2014;20: 548–54. doi: 10.1038/nm.3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Ko AH, Bekaii-Saab T, Van Ziffle J, Mirzoeva OM, Joseph NM, Talasaz A, et al. A multicenter, Open-Label phase II clinical trial of combined MEK plus EGFR inhibition for Chemotherapy-Refractory advanced pancreatic adenocarcinoma. Clin Cancer Res. 2016;22: 61–8. doi: 10.1158/1078-0432.CCR-15-0979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Jänne PA, Borras AM, Kuang Y, Rogers AM, Joshi VA, Liyanage H, et al. A rapid and sensitive enzymatic method for epidermal growth factor receptor mutation screening. Clin Cancer Res. 2006;12: 751–8. doi: 10.1158/1078-0432.CCR-05-2047. [DOI] [PubMed] [Google Scholar]
  198. Newton CR, Graham A, Heptinstall LE, Powell SJ, Summers C, Kalsheker N, et al. Analysis of any point mutation in DNA. Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS). Nucleic Acids Res. 1989;17: 2503–16. doi: 10.1093/nar/17.7.2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, Namgyal C, et al. Sensitive sequencing method for KRAS mutation detection by Pyrosequencing. J Mol Diagn. 2005;7: 413–21. doi: 10.1016/S1525-1578(10)60571-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Liu Q, Sommer SS. Pyrophosphorolysis-activated polymerization (PAP): application to allele-specific amplification. Biotechniques. 2000;29: 1072–6, 1078, 1080 passim. doi: 10.2144/00295rr03. [DOI] [PubMed] [Google Scholar]
  201. Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4: 136ra68. doi: 10.1126/scitranslmed.3003726. [DOI] [PubMed] [Google Scholar]
  202. Li M, Diehl F, Dressman D, Vogelstein B, Kinzler KW. BEAMing up for detection and quantification of rare sequence variants. Nat Methods. 2006;3: 95–7. doi: 10.1038/nmeth850. [DOI] [PubMed] [Google Scholar]
  203. Korabecna M, Opatrna S, Wirth J, Rulcova K, Eiselt J, Sefrna F, et al. Cell-free plasma DNA during peritoneal dialysis and hemodialysis and in patients with chronic kidney disease. Ann N Y Acad Sci. 2008;1137: 296–301. doi: 10.1196/annals.1448.014. [DOI] [PubMed] [Google Scholar]
  204. Nakamura T, Sunami E, Nguyen T, Hoon DS. Analysis of loss of heterozygosity in circulating DNA. Methods Mol Biol. 2009;520: 221–9. doi: 10.1007/978-1-60327-811-9_16. [DOI] [PubMed] [Google Scholar]
  205. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368: 1199–209. doi: 10.1056/NEJMoa1213261. [DOI] [PubMed] [Google Scholar]
  206. Murtaza M, Dawson SJ, Pogrebniak K, Rueda OM, Provenzano E, Grant J, et al. Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer. Nat Commun. 2015;6: 8760. doi: 10.1038/ncomms9760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Luthra R, Patel KP, Reddy NG, Haghshenas V, Routbort MJ, Harmon MA, et al. Next-generation sequencing-based multigene mutational screening for acute myeloid leukemia using MiSeq: applicability for diagnostics and disease monitoring. Haematologica. 2014;99: 465–73. doi: 10.3324/haematol.2013.093765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. De Mattos-Arruda L, Mayor R, Ng CK, Weigelt B, Martínez-Ricarte F, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6: 8839. doi: 10.1038/ncomms9839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, et al. Memorial Sloan Kettering-Integrated mutation profiling of actionable cancer targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015;17: 251–64. doi: 10.1016/j.jmoldx.2014.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Guttery DS, Page K, Hills A, Woodley L, Marchese SD, Rghebi B, et al. Noninvasive detection of activating estrogen receptor 1(ESR1) mutations in estrogen receptor-positive metastatic breast cancer. Clin Chem. 2015;61: 974–82. doi: 10.1373/clinchem.2015.238717. [DOI] [PubMed] [Google Scholar]
  211. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31: 1023–31. doi: 10.1038/nbt.2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co- occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles and therapeutic vulnerabilities. Cancer Discovery. 2015;5: 860–77. doi: 10.1158/2159-8290.CD-14-1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. 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. Sci Transl Med 2014; 6: 224ra24.
  214. Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497: 108–12. doi: 10.1038/nature12065. [DOI] [PubMed] [Google Scholar]
  215. Schiavon G, Hrebien S, Garcia-Murillas I, Cutts RJ, Pearson A, Tarazona N, et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2015;7: 313ra182. doi: 10.1126/scitranslmed.aac7551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Chandarlapaty SS, Chen D. FDNA analysis from BOLERO-2 plasma samples identifies a high rate of ESR1 mutations: exploratory analysis for prognostic and predictive correlation of mutations reveals different efficacy outcomes of endocrine-therapy-based regimens//San Antonio Breast Cancer Symposium 2015, 2015:
  217. [No authors listed] Targeting mutant AKT in cancer. Cancer Discov. 2016;6: OF7. doi: 10.1158/2159-8290.CD-NB2015-160. [DOI] [PubMed] [Google Scholar]
  218. PI3K inhibitor improves PFS in BELLE-2 trial. Cancer Discov. 2016;6: 115-6.
  219. Legendre C, Gooden GC, Johnson K, Martinez RA, Liang WS, Salhia B. Whole-genome bisulfite sequencing of cell-free DNA identifies signature associated with metastatic breast cancer. Clin Epigenetics. 2015;7: 100. doi: 10.1186/s13148-015-0135-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Mcgowan PM, Kirstein JM, Chambers AF. Micrometastatic disease and metastatic outgrowth: clinical issues and experimental approaches. Future Oncol. 2009;5: 1083–98. doi: 10.2217/fon.09.73. [DOI] [PubMed] [Google Scholar]
  221. Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, et al. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10: 8152–62. doi: 10.1158/1078-0432.CCR-04-1110. [DOI] [PubMed] [Google Scholar]
  222. Spiliotaki M, Mavroudis D, Kapranou K, Markomanolaki H, Kallergi G, Koinis F, et al. Evaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancy. Breast Cancer Res. 2014;16: 485. doi: 10.1186/s13058-014-0485-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Payne RE, Hava NL, Page K, Blighe K, Ward B, Slade M, et al. The presence of disseminated tumour cells in the bone marrow is inversely related to circulating free DNA in plasma in breast cancer dormancy. Br J Cancer. 2012;106: 375–82. doi: 10.1038/bjc.2011.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Shaw JA, Page K, Blighe K, Hava N, Guttery D, Ward B, et al. Genomic analysis of circulating cell-free DNA infers breast cancer dormancy. Genome Res. 2012;22: 220–31. doi: 10.1101/gr.123497.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6: 267–83. doi: 10.1586/epr.09.17. [DOI] [PubMed] [Google Scholar]
  226. Peinado H, Alečković 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. Nat Med. 2012;18: 883–91. doi: 10.1038/nm.2753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527: 329–35. doi: 10.1038/nature15756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Skog J, Würdinger 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. Nat Cell Biol. 2008;10: 1470–6. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Lu X, Kang Y. Organotropism of breast cancer metastasis. J Mammary Gland Biol Neoplasia. 2007;12: 153–62. doi: 10.1007/s10911-007-9047-3. [DOI] [PubMed] [Google Scholar]
  230. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24: 766–9. doi: 10.1038/cr.2014.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Cheng AM, Byrom MW, Shelton J, Ford LP. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res. 2005;33: 1290–7. doi: 10.1093/nar/gki200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Su Z, Yang Z, Xu Y, Chen Y, Yu Q. MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget. 2015;6: 8474–90. doi: 10.18632/oncotarget.3523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Stückrath I, Rack B, Janni W, Jäger B, Pantel K, Schwarzenbach H. Aberrant plasma levels of circulating miR-16, miR-107, miR-130a and miR-146a are associated with lymph node metastasis and receptor status of breast cancer patients. Oncotarget. 2015;6: 13387–401. doi: 10.18632/oncotarget.3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Blenkiron C, Goldstein LD, Thorne NP, Spiteri I, Chin SF, Dunning MJ, et al. MicroRNA expression profiling of human breast cancer identifies new markers of tumor subtype. Genome Biol. 2007;8: R214. doi: 10.1186/gb-2007-8-10-r214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Tang W, Zhu J, Su S, Wu W, Liu Q, Su F, et al. MiR-27 as a prognostic marker for breast cancer progression and patient survival. PLoS One. 2012;7: e51702. doi: 10.1371/journal.pone.0051702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Cataldo VD, Gibbons DL, Perez-Soler R, Quintas-Cardama A. Treatment of non-small-cell lung cancer with erlotinib or gefitinib. N Engl J Med. 2011;364: 947–55. doi: 10.1056/NEJMct0807960. [DOI] [PubMed] [Google Scholar]
  237. Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, et al. Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res. 2011;17: 7808–15. doi: 10.1158/1078-0432.CCR-11-1712. [DOI] [PubMed] [Google Scholar]
  238. Flaherty KT, Puzanov I, Kim KB, Ribas A, Mcarthur GA, Sosman JA, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363: 809–19. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Walther A, Johnstone E, Swanton C, Midgley R, Tomlinson I, Kerr D. Genetic prognostic and predictive markers in colorectal cancer. Nat Rev Cancer. 2009;9: 489–99. doi: 10.1038/nrc2645. [DOI] [PubMed] [Google Scholar]
  240. Mouliere F, Robert B, Arnau Peyrotte E, Del Rio M, Ychou M, Molina F, et al. High fragmentation characterizes tumour-derived circulating DNA. PLoS One. 2011;6: e23418. doi: 10.1371/journal.pone.0023418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Jeselsohn R, Yelensky R, Buchwalter G, Frampton G, Meric-Bernstam F, Gonzalez-Angulo AM, et al. Emergence of constitutively active estrogen receptor-α mutations in pretreated advanced estrogen receptor-positive breast cancer. Clin Cancer Res. 2014;20: 1757–67. doi: 10.1158/1078-0432.CCR-13-2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Gámez-Pozo A, Pérez Carrión RM, Manso L, Crespo C, Mendiola C, López-Vacas R, et al. The Long-HER study: clinical and molecular analysis of patients with HER2+ advanced breast cancer who become long-term survivors with trastuzumab-based therapy. PLoS One. 2014;9: e109611. doi: 10.1371/journal.pone.0109611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Mazel M, Jacot W, Pantel K, Bartkowiak K, Topart D, Cayrefourcq L, et al. Frequent expression of PD-L1 on circulating breast cancer cells. Mol Oncol. 2015;9: 1773–82. doi: 10.1016/j.molonc.2015.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Schramm A, Friedl TW, Schochter F, Scholz C, De Gregorio N, Huober J, et al. Therapeutic intervention based on circulating tumor cell phenotype in metastatic breast cancer: concept of the DETECT study program. Arch Gynecol Obstet. 2016;293: 271–81. doi: 10.1007/s00404-015-3879-7. [DOI] [PubMed] [Google Scholar]
  245. Senkus E, Kyriakides S, Penault-Llorca F, Poortmans P, Thompson A, Zackrisson S, et al. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol.2013;24 Suppl 6: vi7-23.
  246. Moore KN, Mannel RS. Is the NCI MATCH trial a match for gynecologic oncology. Gynecol Oncol. 2016;140: 161–6. doi: 10.1016/j.ygyno.2015.11.003. [DOI] [PubMed] [Google Scholar]
  247. Hatzis C, Pusztai L, Valero V, Booser DJ, Esserman L, Lluch A, et al. A genomic predictor of response and survival following taxane-anthracycline chemotherapy for invasive breast cancer. JAMA. 2011;305: 1873–81. doi: 10.1001/jama.2011.593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Hayes DF, Smerage J. Is there a role for circulating tumor cells in the management of breast cancer. Clin Cancer Res. 2008;14: 3646–50. doi: 10.1158/1078-0432.CCR-07-4481. [DOI] [PubMed] [Google Scholar]

Articles from Cancer Biology & Medicine are provided here courtesy of Chinese Anti-Cancer Association

RESOURCES