4,459
Views
47
CrossRef citations to date
0
Altmetric
Theme: Nanotechnology & Single-Cell Analysis - Review

Colorectal cancer diagnostics: biomarkers, cell-free DNA, circulating tumor cells and defining heterogeneous populations by single-cell analysis

, , , &
Pages 581-599 | Published online: 09 Jan 2014

Abstract

Reliable biomarkers are needed to guide treatment of colorectal cancer, as well as for surveillance to detect recurrence and monitor therapeutic response. In this review, the authors discuss the use of various biomarkers in addition to serum carcinoembryonic antigen, the current surveillance method for metastatic recurrence after resection. The clinical relevance of mutations including microsatellite instability, KRAS, BRAF and SMAD4 is addressed. The role of circulating tumor cells and cell-free DNA with regards to their implementation into clinical use is discussed, as well as how single-cell analysis may fit into a monitoring program. The detection and characterization of circulating tumor cells and cell-free DNA in colorectal cancer patients will not only improve the understanding of the development of metastasis, but may also supplant the use of other biomarkers.

Colorectal carcinoma is the third most common cancer diagnosed worldwide and the fourth most common cause of cancer death Citation[1,2]. Oncologic outcomes have improved with advances in surgical technique and adjuvant therapy. Overall 5-year survival for colorectal cancer is excellent for disease localized to the colon (95% for stage I and 82% for stage II) , but decreases to 61% for patients with regional spread to the lymph nodes (stage III) and only 8% for patients with distant metastases (stage IV) Citation[3,4]. Fewer than 40% of the patients in the USA are diagnosed with stage I or II disease, for which surgery alone can be curative. Patients with stage III disease, comprising 36–54%, are candidates for adjuvant chemotherapy after surgical resection. Patients with stage IV disease comprise 20–31% and usually undergo first-line chemotherapy, potentially followed by surgical resection Citation[5]. Metastasis occurs most commonly to the liver in a synchronous fashion in 14.5% and in a metachronous fashion in 60% Citation[6,7]. Synchronous metastasis to the peritoneal surface occurs in 9–18% of the patients, to the lung in 10–14% and to the bone in 0.7–1.6% Citation[8]. There are several regimens used for the treatment of stage III and IV colorectal cancer, including 5-fluorouracil, leucovorin and oxaliplatin (FOLFOX); 5-fluorouracil, leucovorin and irinotecan (FOLFIRI); and capecitabine (the oral form of 5-fluorouracil, otherwise known as Xeloda®) plus oxaliplatin (XELOX). Advances in molecular diagnostics have allowed the development of more targeted therapeutic agents to reduce the risk of metastases and to treat metastases if they do occur.

The most widely used tumor marker in colorectal cancer is serum carcinoembryonic antigen (CEA), which is helpful in the detection of liver metastases developing after tumor resection. In addition, tumor-specific markers such as KRAS and microsatellite instability (MSI) are useful for both prognosis and directing chemotherapy.

Circulating tumor cells (CTCs) have been identified as a necessary component of the metastatic cascade. Numerous methods of CTC detection have been developed. Studies demonstrating that CTCs have prognostic and predictive value have spawned interest in potential clinical applications. Additionally, single-cell profiling of CTCs has opened a new frontier in personalized oncologic therapy. Cell-free DNA has also been studied for its prognostic and predictive value.

Tumor markers in colorectal cancer

CEA is the current serum biomarker used in clinical practice, with its main utility for indicating disease recurrence in the liver. Tumor biomarkers identified in colorectal cancer tissue have been used to guide chemotherapy regimens and include KRAS, BRAF, MSI and SMAD4 Citation[9].

CEA

Since its identification in colon adenocarcinoma in 1965, CEA has become the standard tumor marker in the clinical care of patients with colorectal carcinoma Citation[10]. It is correlated to disease stage and preoperative CEA level is related to prognosis Citation[11–16]. Following resection of the primary tumor, serial CEA levels are routinely obtained every 2–3 months for at least 2 years to monitor for disease recurrence or metastasis Citation[17–19]. There is no consensus on what change in CEA level would trigger further evaluation for recurrence Citation[20]. Increases of CEA by at least 50% has a sensitivity of 76% and specificity of 90% for predicting progression of disease in patients with metastatic disease undergoing palliative chemotherapy, and a drop of at least 30% can exclude disease progression Citation[21].

MSI

MSI occurs due to defects in DNA mismatch repair genes, and while it is typically associated with hereditary nonpolyposis colorectal cancer, most MSI-high tumors occur sporadically Citation[22,23]. Despite their resistance to alkylating agents and cisplatin, MSI-high tumors have better recurrence-free and overall survival Citation[23]. In patients with stage II disease, MSI-high status was found to confer the same advantage in long-term outcomes as that conferred by stage T3 over T4 Citation[24].

KRAS

Mutant KRAS is associated with resistance to anti-EGFR monoclonal antibody immunotherapy with agents such as cetuximab or panitumumab. Randomized trials (CRYSTAL, OPUS and PRIME) demonstrated the efficacy of EGFR inhibitors when added to FOLFOX or 5-fluorouracil, leucovorin and irinotecan chemotherapy regimens for patients with KRAS wild-type tumors, especially with significantly improved progression-free survival, response rates and R0 resection rates compared with chemotherapy alone Citation[25–28].

However, the MRC COIN and NORDIC-VII trials subsequently failed to show the same benefit, demonstrating increased response rate without improvement in progression-free or overall survival when cetuximab was added to first-line chemotherapy for metastatic disease Citation[29,30]. Current clinical practice for patients with metastatic colorectal cancer screens tumor tissue for KRAS mutations so that only patients with wild-type KRAS tumors receive anti-EGFR therapy.

KRAS mutations occur more frequently in right-sided tumors, low-grade tumors, and MSI-low or microsatellite-stable (MSS) tumors. It does not appear to have prognostic value for recurrence-free and overall survival Citation[31].

BRAF

Efforts to find other tumor mutations that will allow more specific targeting of patients who will benefit from anti-EGFR immunotherapy have shown that the presence of an activating mutation in BRAF in the KRAS wild-type population is associated with poor response Citation[32,33]. However, the CRYSTAL and OPUS trials that supported the use of cetuximab in KRAS wild-type metastatic colorectal cancer also found that while BRAF mutation status could not be used in the same fashion as a predictive biomarker for chemotherapy, it did have an association with poor prognosis Citation[34]. Similarly, the NORDIC-VII study that did not support the use of cetuximab in this same patient population also found that BRAF mutation was a strong negative prognostic factor Citation[30].

Right-sided colon cancers with BRAF mutation and microsatellite stability have a particularly poor prognosis with regard to overall and disease-free survival. These tumors tend to have adverse histologic features such as lymphatic and perineural invasion, tumor budding and mucinous differentiation Citation[35]. BRAF mutation in stage II and III disease has been shown to be prognostic for overall survival in the MSI-low or MSS population Citation[24]. BRAF mutations occur more frequently in right-sided tumors, MSI-high tumors, high-grade tumors, patients over 60 years of age and female patients Citation[31]. In preclinical studies, vemurafenib (PLX4032), a B-Raf kinase inhibitor approved by the US FDA for use in late-stage melanoma patients with BRAF mutations, showed poor efficacy in colorectal cancer (CRC) patients with a BRAF mutation Citation[36]. This resistance to BRAF inhibitors can be overcome by using it in combination with phosphoinositide 3-kinase (PI3K) inhibitors, AKT inhibitors or standard regimens like capecitabine and bevacizumab or cetuximab and irinotecan; this has been demonstrated by in vitro as well as in vivo animal studies Citation[37,38].

Loss of heterozygosity & SMAD4

The SMAD gene products are transcriptional mediators in the TGF-β signaling pathway. The genes encoding SMADs are located at chromosome 18q21. Initial studies demonstrated that loss of heterozygosity at chromosome 18q was a strong prognostic factor for worse survival rates in patients with stage II and III colorectal cancer Citation[9,39,40]. Subsequent studies have demonstrated that loss of heterozygosity is less important in prognosis than previously thought, with no association with cancer-specific or overall survival, although it is independently associated with high tumor grade and KRAS mutation Citation[24,41,42].

However, loss of SMAD4 expression, one of several genes encoded at 18q21, has been shown to be a significant independent prognostic factor for worse recurrence-free and overall survival, particularly in patients with stage III disease Citation[24,43,44]. Mouse studies demonstrate that loss of SMAD4 expression changes the role of TGF-β from growth suppressor to growth promoter, thus increasing the tumorigenic and metastatic potential of colorectal cancer cells Citation[45]. Patients with stage III disease and intact SMAD4 expression with MSI were found to have similar outcomes compared with patients with stage II disease, whereas patients with stage II disease and loss of SMAD4 expression without MSI status had outcomes similar to patients with stage III disease Citation[24]. Retention of SMAD4 expression has also been found to be a predictive marker for a threefold increase in benefit from 5-fluorouracil-based chemotherapy Citation[46]. Loss of SMAD activity occurs in 10% of the colorectal cancers and is associated with advanced-stage disease, the presence of lymph node metastases and shorter overall survival Citation[47].

The role of CTCs in the metastatic cascade

CTCs are defined as tumor cells that have separated from either the primary tumor or metastases, and are circulating in the peripheral blood. CTCs have been detected in almost all cancers, including colon, breast, prostate, lung, ovary, pancreas, liver, gastric, esophageal, renal, bladder, thyroid, nasopharyngeal and melanoma; they are extremely rare in patients without malignancy, and can be detected before the development of metastases Citation[48,49]. An early step in the metastatic process is the sporadic shedding of malignant cells from the primary tumor, after which these cells must access the circulation Citation[50]. It has been observed that up to 1 million cells enter the circulation daily per gram of tumor tissue Citation[51]. However, tumor cell embolism does not necessarily lead to metastasis, due to a process of elimination of most of these cells, called ‘metastatic inefficiency’ Citation[52,53]. For example, trauma from mechanical shear forces causing lethal deformation can cause cancer cells in the peripheral blood to undergo apoptosis Citation[54–56]. The majority of CTCs undergo anoikis, which is apoptosis induced by cell detachment; cells with anoikis resistance are more likely to survive in the circulation Citation[57]. CTCs have been shown to possess physical properties that potentially aid their ability to intravasate into the peripheral blood, such as increased cell elasticity Citation[58]. Even after hematogenous dissemination, tumor cells are highly inefficient in the processes of extravasation and subsequent growth. A small proportion of extravasated cells might form micrometastases, most of which disappear while only a few progress to form macroscopic tumors Citation[53,56]. This process of extravasation and growth involves the attachment of tumor cells to the vascular endothelium followed by cancer cell aggregation at these sites of attachment Citation[59,60].

Another contribution CTCs may make to the progression of malignancy is the development of highly metastatic phenotypes due to genetic exchange between tumor cells Citation[61,62]. The epithelial–mesenchymal transition (EMT) may also be a key step for metastatic spread to occur by way of CTCs Citation[63–69]. Single-cell analysis has been used to identify and characterize CTCs, and in breast and prostate cancer, CTCs have been found to express EMT-related genes, with these cells more commonly associated with metastatic disease Citation[58]. Interactions with platelets also play a role in allowing tumor cells to make the transition to an invasive mesenchymal phenotype leading to metastasis Citation[70].

For formation of metastasis, cells undergo a reversed process referred to as mesenchymal–epithelial transition (MET) Citation[71]. However, it is not yet clearly established whether EMT is a necessary step leading to the development of metastases as there is evidence that cell invasion can occur in the absence of EMT, and whether MET plays an important role Citation[72,73]. Nevertheless, recent work demonstrates that a small subset of CTCs is indeed capable of inducing metastasis. These cells express epithelial cell adhesion molecule (EpCAM), CD44, CD47 and MET in patients with luminal breast cancers Citation[74]. Further investigation is needed to fully understand this process.

As demonstrated by genomic analysis of single cells from primary and metastatic tumors, there is genetic diversity within primary tumors, and mutated tumor cells in the primary tumor can then undergo clonal expansion in the metastatic site Citation[75,76]. There is substantial lack of concordance between the mutation phenotypes of primary and metastatic tumors in colorectal cancer, seen in up to 23% for KRAS mutations. KRAS and BRAF mutations can be identified in CTCs as well, although the mutation phenotype of CTCs has not been shown to necessarily correlate with that of metastatic tumors Citation[77,78]. Mutational analysis of KRAS, BRAF and PIK3CA in single CTCs demonstrate heterogeneity among CTCs from the same patient, which may explain the inconsistent response to EGFR inhibitors and emphasizing the importance of single-cell analysis for identifying different subpopulations of CTCs that may respond to different therapies Citation[79,80]. Not all CTCs are necessarily reflective of the population of cells that has successfully caused metastasis. Thus, the characteristics of CTCs may differ from those of the cells of metastatic tumors Citation[81].

CTC enrichment & capture technologies

The laboratory definition of a CTC is an epithelial cell found in a cancer patient’s blood which has an intact nucleus and shows expression of cytokeratin, but not CD45 (a white blood cell marker) Citation[82]. A wide variety of technologies have been developed to accomplish the feat of isolating a single CTC from approximately 5 × 106 nucleated leukocytes and 5 × 109 red blood cells per ml Citation[48].

The potential sources of blood for CTC analysis in patients with colorectal cancer are mesenteric, portal or peripheral. In a large prospective study, the detection efficiency of CTCs in the central venous blood compartment and mesenteric venous blood compartment was compared, demonstrating that CTCs were found at a markedly higher rate in the mesenteric venous blood compartment than in the central venous blood compartment Citation[83]. However, since a peripheral blood draw is much more convenient and can be obtained at different time points of treatment and disease progression, most studies analyze CTCs using this approach.

The majority of CTC detection methods require an enrichment step, which may be in the form of red blood cell lysis and/or density-gradient centrifugation for elimination of red blood cells, followed by a separation from contaminating leukocytes prior to isolation and characterization Citation[84,85]. CTC enrichment methods for colorectal cancer fall into two categories: detection according to expression of surface proteins which can be EpCAM-based or non-EpCAM-based, and detection according to other cell characteristics such as size and morphology, physical characteristics or functional properties .

Enrichment according to expression of surface proteins

EpCAM is a glycosylated membrane protein initially discovered as the dominant antigen on colon cancer cells and mediates epithelial-specific intercellular cell adhesion. High expression of EpCAM has been found in most adenocarcinomas, metastases and cancer stem cells and thus is considered an optimal tumor surface antigen for the detection of CTCs Citation[86,87]. The CellSearch System (Veridex, NJ, USA) is the most widely used EpCAM-based system for CTC detection, and the only one that is FDA-approved for use in patients with metastatic colorectal, breast and prostate cancer Citation[88–94]. For the purposes of prognostic and predictive value, the cut-off value separating favorable and unfavorable CTC in metastatic colorectal cancer is three CTCs per 7.5 ml of blood Citation[94].

An EpCAM-based system developed by our group is the MagSweeper, which is an immunomagnetic cell separation device that isolates live cells with extremely high purity, allowing not only enumeration, but also downstream single-cell molecular characterization and growth of CTCs Citation[95]. Multiple other systems based on EpCAM have been developed, including the Adna Test Citation[96], microfluidic chips such as the Herringbone Chip or the newly developed iChip, which also can be used in a negative selection mode (non-EpCAM based) Citation[97–101], the Isoflux system (Fluxion) Citation[102], the GILUPI Nanodetector (insertable wire) Citation[103], as well as magnetic-activated cell separation Citation[104]. Furthermore, there are several non-EpCAM-based methods that allow enrichment of CTCs through negative selection by exclusion of CD45-positive leukocytes Citation[105–107].

Enrichment according to other cell characteristics

The following methods allow the detection of CTCs by using their different size, deformability, density and electric charges compared with normal blood cells. For isolation of CTCs by size, several different filtration methods have been developed (ISET Citation[108,109], ScreenCell® Citation[110], the CREATV MicroTech filter [CellSieve] Citation[111] and a Parylene filter Citation[112]) that enable rapid isolation of CTCs on removable filters, which can immediately be used for downstream analysis. Other technologies isolate CTCs according to their cellular functionality (Vitatex) Citation[113], by microfluidics by the use of label-free biochips Citation[114] or Dean Flow Fractionation Citation[115], as well as by the use of physical cell properties using dielectrophoretic field flow fractionation Citation[116,117]. The fiber-optic array scanning technology allows detailed cytomorphologic analysis of cells Citation[118].

CTC detection methods

Even after enrichment of CTCs, a considerable number of contaminating leukocytes remain in the CTC specimen, so a second step is needed for identification of CTCs. Detection and characterization of single CTCs can be carried out by performing immunocytochemistry (ICC), RT-PCR or the epithelial immunospot (EPISPOT) assay.

ICC

ICC analysis is used to identify CTCs, since there often is remaining leukocyte contamination after enrichment. Most methods use antibodies against cytokeratin, CD45 (leukocyte antigen used as a negative marker), and 4′,6-diamidino-2-phenylindole (DAPI nuclear stain) for fluorescent staining of CTCs.

RT-PCR

Using RT-PCR, the gene expression analysis of at least one of the epithelial markers KRT-7, -8, -18 and/or -19 as well as reference genes (GAPDH, ACTB) confirms the existence of a CTC. Cells expressing CD45 have to be excluded Citation[119,120]. RT-PCR has also been used to detect CEA and CK20 from peripheral blood before and after curative resection, and from mesenteric tumor drainage blood. Positive rates of marker genes in tumor drainage blood are 3.3-times higher than that of peripheral blood Citation[121,122].

EPISPOT assay

The EPISPOT assay is derived from the ELISA and can be used after any kind of enrichment method to detect only live CTCs by detecting proteins released from epithelial cancer cells. Enriched and captured cells are cultured for 48 h on an antibody-coated membrane. Those antibodies capture released proteins, which in a second step can be detected by adding secondary antibodies labeled with fluorochromes. Apoptotic tumor cells do not secrete measurable amounts of proteins and therefore are not detected Citation[123].

CTCs are highly heterogeneous in their surface antigens and genetic characteristics. The only device that has received FDA approval is the CellSearch System. However, because this method is EpCAM-based, it has a large drawback in its inability to capture tumor cells undergoing EMT, as the expression of EpCAM and cytokeratins, used to identify CTCs, is downregulated, while markers of EMT (e.g., vimentin and N-cadherin) are expressed. Therefore, negative selection methods by eliminating leukocytes by CD45-depletion or label-free devices might seem more favorable for maximizing CTC enrichment. In addition, the development of novel markers of CTCs such as Plastin3 that is not repressed during EMT has successfully been shown to increase the yield of CTCs. CTCs identified by this method were also shown to be prognostically significant Citation[124].

The large number of methods for enrichment and detection of CTCs and their use in various studies with different tumor entities renders fair comparison of results, specificity, sensitivity and reproducibility impossible. Assays successfully tested on cell-line tumor cells cannot be expected to show equal results to patient blood samples due to the heterogeneity of tumor cells even within a single patient.

The use of anti-EpCAM antibodies for the isolation of CTCs in colorectal cancer is applicable as 98% of the colorectal cancers are adenocarcinomas Citation[125]. Furthermore, in patients suffering from hepatocellular carcinoma, EpCAM-positive CTCs have a high tumorigenic potential compared with EpCAM-negative CTCs Citation[126]. Nevertheless, a label-free device may be more effective in the isolation of CTCs undergoing phenotypic changes, and the analysis of these cells is likely to lead to a better understanding of tumor biology and the metastatic process.

Another limitation during CTC detection is the small volume of blood screened for rare CTCs in most assays. To overcome this problem, the newly developed GILUPI Nanodetector can be functionalized with antibodies, such as EpCAM, for a highly selective enrichment of CTCs in a blood volume as high as 1.5 l. Finally, CTC detection may also be affected by trauma of the tumor during resection that induces cell shedding; thus, surgical manipulation may be a confounding factor affecting the number of CTCs detected perioperatively Citation[127–129]. This is further illustrated by the differences in CTC detection among different blood compartments within the same patient Citation[83]. Further studies on larger patient cohorts are necessary to be able to determine which device can most effectively and reliably detect CTCs.

Defining heterogeneous populations by single-cell analysis

Single-cell profiling techniques have been developed to analyze the genome of CTCs, not only at the DNA level, but also at the RNA level, corresponding to gene expression. Comparative genome hybridization, for example, analyzes DNA copy number variation and can be performed in single cells using PCR-based whole genome amplification Citation[130–132]. Next-generation sequencing, fluorescence-activating cell sorting and single-nucleus sequencing are other techniques that have been developed to not only enable the identification of tumor cells, but also the characterization of the genome of those individual cells Citation[133,134].

The minute quantities of RNA within a single cell poses a great technical challenge for the sensitivity of detection. For transcriptional analysis, after converting RNA to cDNA, amplification is necessary, after which gene expression analysis using a microarray or next-generation sequencing can be performed Citation[82,135].

In a pioneering study, Dalerba et al. demonstrated the importance of single-cell expression analysis for improving the understanding of tumor heterogeneity and clonality Citation[136]. Cells from normal and neoplastic human colon epithelia were isolated and fluorescence-activating cell sorting applied using established markers of differentiation to distinguish mature cells on the top of colon epithelial crypts from immature stem cells at the bottom of the crypt. In a second step, single-cell analysis by gene-expression profiling was performed. By principal component analysis, the gene expression data were then divided into different groups, defining the gene expression profiles of the differentiated enterocytes at the top of the crypts and of the immature, stem and goblet cells at the bottom of the crypts. Furthermore, a colon cancer mouse xenograft model was developed by injection of a single colorectal cancer cell into the flanks of the mice. Only cells highly expressing EpCAM and CD44, which are known to be cancer stem cell markers, resulted in tumors. By performing gene-expression profiling of these tumors and comparison with the primary tumor from which the implanted cells originated, a very similar heterogeneous cell population was found. In addition, the investigation and comparison of colorectal adenoma gene expression with healthy colon tissue demonstrated that the tumor was comprised of the exact cell populations, which shows that tumor heterogeneity is due to multilineage differentiation.

In another recent study, CTCs from 15 patients with advanced-stage colorectal cancer were isolated using the CellSearch device. The genomic profiles of the primary tumor were compared with the profiles of liver metastasis and CTCs in each patient by performing array comparative genome hybridization. This revealed that CTCs had some similarities in copy number changes to the primary tumor, as well as other similarities to the copy number changes in the liver metastasis, especially in known colorectal driver genes (e.g., KRAS, APC, PIK3CA), but that they also had additional different copy number changes. By deep sequencing of the primary and corresponding liver metastasis, it was discovered that the mutations that were found uniquely in CTCs were detectable in the primary and metastatic tissue at subclonal levels Citation[137]. Analysis of the marker profiles of CTCs and primary tumors in nonmetastatic breast cancer patients using immunohistochemistry and FISH demonstrated no correlation between the two profiles Citation[80]. These studies emphasize the importance of isolation and characterization of CTCs, as these cells can carry mutations that have developed in the clinical course of cancer patients and can serve as liquid biopsies to monitor therapeutic response and also help make decisions concerning therapeutic regimens for an individual patient. CTCs, however, do not necessarily carry the same mutations as the primary tumor, and this discordance may explain the current variability of treatment response to chemotherapy. The differences in profile between CTCs and primary tumors in breast cancer has been demonstrated and a similar difference in colorectal cancer is likely Citation[80].

In the near future, it will be possible to use droplet-based microfluidics for isolation and high-throughput analysis of approximately 1 million single cells in parallel, as well as enable cell growth, which could significantly facilitate single-cell profiling. The droplets are only picoliters or nanoliters in size and are compartmentalized in the microfluidic system, leading to a faster accumulation and thus quicker detection of secreted molecules Citation[138].

Clinical significance of CTCs

CTCs have been shown in multiple studies to correlate not only with worse clinicopathologic features of disease, but also to have prognostic and predictive value. Prognosis refers to the likely course and outcome of the disease regardless of therapy, and is measured by cancer-specific survival, disease-free survival and overall survival. The predictive value of a biomarker refers to the effect of therapy on a patient’s outcome Citation[139].

As a marker of advanced disease

The correlation of CTC levels with clinicopathologic features that indicate more advanced disease has been observed in both metastatic and nonmetastatic disease, at various sampling points before and after surgical or systemic treatment, and in both the peripheral and mesenteric compartment. CTC detection as determined by CEA/CK20 expression in both preoperative peripheral blood and preresection tumor drainage blood is significantly associated with depth of tumor invasion, venous invasion, lymph node metastasis, liver metastasis and stage Citation[121]. CTC levels obtained from the peripheral blood after resection of the primary tumor have been shown to have a significant correlation with regional lymph node involvement and stage of disease Citation[140]. Similarly, CTCs obtained from the venous drainage blood after curative CRC resection also correlate with lymph node positivity Citation[141]. For patients with metastatic disease not undergoing resection, unfavorable baseline CTC levels (>3 CTCs per 7.5 ml blood using the CellSearch System) correlate with more advanced disease. Among patients with metastatic colorectal carcinoma, those with liver metastases and poorer performance status had higher baseline CTC levels Citation[90,94]. Another marker of CTCs, Plastin3-, has recently been shown to have significant clinical relevance. Plastin3-positivity in the peripheral blood was found to be associated with clinicopathologic risk factors of greater depth of invasion (>=T3), lymph node metastasis, liver metastasis, peritoneal dissemination, recurrence rate and Dukes stage. Plastin3 expression was also detected in all patients with recurrent disease and at a higher level compared with prerecurrence levels and to patients without recurrence Citation[124].

As a prognostic factor

Detection of CTCs portends poor prognosis in patients with metastatic colorectal cancer. A meta-analysis of 16 studies to include 1491 patients with metastatic colorectal cancer demonstrated that patients in whom circulating tumor cells are detected have a 2.5-fold increased mortality risk and a twofold increased risk of disease progression or recurrence Citation[142,143]. A higher percentage of patients with metastatic colorectal cancer who had disease progression or death had unfavorable CTCs at 3–5 weeks after treatment than patients whose disease did not progress. Baseline CTC is an independent prognostic factor in metastatic colorectal cancer. Patients with unfavorable levels of CTCs at baseline had significantly shorter median disease-free and overall survival than patients with fewer CTCs Citation[90,144,145]. The effect of favorable baseline CTCs was seen in significantly longer progression-free survival time for patients receiving irinotecan, as well as in a near doubling of overall survival time for all patients regardless of whether they received oxaliplatin, irinotecan or bevacizumab. This overall effect was more pronounced in patients over 65 years of age with poor ECOG performance status Citation[144].

For patients with nonmetastatic disease (Dukes’ stage B or C), overall and disease-free survival of patients in whom CTCs were detected were significantly worse than those of patients in whom CTCs were not detected. This difference was not seen in patients with Dukes’ stage A disease Citation[146]. In combination with lymph node staging, CTC detection 24 h after curative resection has even greater sensitivity for predicting recurrence Citation[122].

Patients with metastatic disease and high baseline CTC levels who demonstrate a decrease to favorable CTC levels after 3–5 weeks of chemotherapy experience the same disease-free survival as those who started with favorable baseline CTC levels, although this effect was not seen in overall survival. Patients with persistently unfavorable CTC levels prior to and after treatment have significantly worse disease-free survival and overall survival than those in whom the CTCs converted to favorable levels after treatment Citation[90,147].

Plastin3-positivity was also associated with worse prognosis. Stage for stage, patients in whom Plastin3-positive CTCs were detected had significantly shorter overall survival than Plastin3-negative patients; patients without synchronous distant metastasis had significant shorter disease-free survival. For Dukes’ stage B and C disease, Plastin3-expression was independently associated with decreased overall survival and disease-free survival, whereas clinicopathological factors such as tumor size, lymphatic invasion, venous invasion, histologic type and depth of invasion were not Citation[124].

In addition to CTC detection in the peripheral blood, CTC detection in tumor drainage venous blood before and after resection is also associated with prognosis. CTC detection in tumor drainage blood obtained just prior to resection has been demonstrated to correlate with overall and disease-free survival Citation[121]. CTC levels obtained from tumor drainage venous blood after resection in Dukes’ stage B and C colorectal cancer patients was also shown to be an independent prognostic factor in both disease-free and overall survival Citation[119]. Intraoperative CTC levels appear to be affected by surgical manipulation of tumors, whereas postoperative CTC levels are more correlated to disease-free survival Citation[143]. Another meta-analysis examining the presence of CTCs in the venous drainage of curative colorectal cancer resections demonstrated an increased hepatic metastases rate in patients with CTCs compared with those without. Similarly, disease-free survival was significantly worse in patients with CTCs in the venous drainage blood of the tumor than those without Citation[141].

As a predictive factor (predicting response to specific therapies)

In a trial comparing maintenance therapy for metastatic disease with either single-agent bevacizumab or XELOX plus bevacizumab after induction chemotherapy with XELOX plus bevacizumab, baseline CTC counts did not predict the response to chemotherapy. However, patients with a low CTC count after 3 months of treatment had significantly higher response rates to chemotherapy than those with a high post-therapy CTC count, resulting in a longer progression-free survival and overall survival in patients receiving chemotherapy plus bevacizumab. Patients with low baseline and post-treatment CTC counts had longer progression-free and overall survival than patients with an initially high baseline CTC count which decreased after chemotherapy. As expected, patients with persistently high baseline and post-therapy CTC counts fared the worst Citation[145].

CTC assessments have also been shown to correlate with tumor responsiveness as determined by radiographic imaging in patients undergoing chemotherapy for metastatic colorectal cancer Citation[120]. Combining CTC counts with tumor response as determined by CT imaging may enhance the potential of CTC as a marker of response to treatment Citation[147].

Cell-free DNA

Cell-free DNA (cfDNA) – also known as circulating tumor DNA (ctDNA), or plasma DNA – can be detected in the blood of colorectal cancer patients. There are different hypothesis concerning the origin of cfDNA. It may emerge from tumor necrosis, apoptosis or even active secretion, but the exact mechanism is unknown Citation[148]. It has been shown that colorectal cancer patients carrying a KRAS mutation are resistant to EGFR inhibitors, for example, cetuximab and panitumumab. By analyzing the plasma DNA concentration of 28 CRC patients receiving monotherapy with panitumumab because of an initially detectable wild-type KRAS tumor, a subset of patients showed detectable KRAS mutations, which can explain the resistance acquired after a certain period of time, in most cases after 5–6 months of therapy Citation[149]. These mutations are detectable months before imaging reveals disease progression, implying that the analysis of KRAS mutation status in plasma can be used to evaluate therapeutic response with a simple blood draw and modulate therapy accordingly Citation[150].

The prognostic value of cfDNA detection in plasma of CRC patients has also been demonstrated, as the 2-year survival rate was 100% in patients without evidence of cfDNA with KRAS2 mutations or p16 gene promoter hypermethylation, which can be found in 40% or 20–50% of CRC patients, respectively. The patients with detectable cfDNA had an overall 2-year survival rate of only 48% and disease recurrence Citation[151].

In a study analyzing the plasma DNA concentrations of 32 patients with advanced-stage CRC, some showed biphasic size distributions of plasma DNA fragments, which was linked to higher numbers of CTCs and higher levels of plasma DNA mutations. Interestingly, not all patients had detectable levels of cfDNA in their circulation despite their advanced tumor stage, which could be due to the short mean half-life of cfDNA of 16 min; thus, patients might have had higher levels of cfDNA prior to the blood draw Citation[152].

To elucidate the kinetics of cfDNA release into the circulation, another group implanted colorectal cancer cells subcutaneously into rats and performed measurements of cfDNA concentrations at different time points. They found that cfDNA was detectable at an early stage, 1 week after tumor inoculation, but that the size of the resulting tumors did not significantly correlate with the concentration of detectable cfDNA. Furthermore, cfDNA could only be detected in 50% of animals that developed metastases and the concentrations did not show significant differences when compared with the ones of animals without metastases Citation[153].

In several studies, the evaluation of methylated SEPT9 in plasma has been shown to be useful for the detection of asymptomatic patients with average risk of CRC as well as patients of all stages of CRC. This would be an ideal method of cancer detection particularly for patients who are resistant or unable to undergo colonoscopy for screening Citation[154–156].

Despite all of these findings, the utilization of cfDNA for therapeutic monitoring in the colorectal cancer field is not yet ready for widespread clinical use, as there are large numbers of genes that can be mutated and different studies have used different laboratory techniques for their detection. There is also no correlation among the presence and concentration of cfDNA and tumor stage, and further studies are needed to elucidate the role of cfDNA and to define specific cfDNA markers that can be detected in the majority of CRC patients. Nevertheless, cfDNA has been shown to be a powerful tool for monitoring patients who undergo therapy with EGFR inhibitors and early identification of developed drug resistance in these patients. In particular, because of the multiplicity of CTC technologies that have yet to be clinically validated in prospective large-scale trials, it may be more straightforward to monitor cfDNA for a panel of mutations that may indicate therapeutic failure. Once the presence of mutations are identified that suggest it is time to switch therapies, single-cell CTC analysis may be performed to identify whether both wild-type and mutant subpopulations of cancer cells coexist that may require specific drugs targeted to the different subpopulations.

Expert commentary

Inclusion of molecular marker assessment such as MSI status, KRAS, SMAD4, as well as CTC and cfDNA detection, may refine our ability to offer prognostic information, which is currently based mainly on TNM staging. The decision to use EGFR inhibitors such as cetuximab should be informed by KRAS mutation status. Testing for MSI is currently performed on tumors based on the revised Bethesda guidelines which screen for hereditary nonpolyposis colorectal cancer Citation[157]. However, given that MSI can occur in the setting of sporadic colorectal cancer and has prognostic and predictive potential, routine MSI testing on all colorectal cancers does have clinical use. In particular, patients with T3N1 (stage III) disease with MSI-high tumors without loss of SMAD4 expression have similar survival to those with T3N0 (stage IIA) disease. If this survival benefit is seen in patients who do not undergo chemotherapy, then it is possible that 11% of the T3N1 population may not need to undergo adjuvant chemotherapy Citation[24]. However, genetic heterogeneity of the cells within a single tumor may explain a suboptimal response to targeted chemotherapy Citation[76].

CEA, despite being the current biomarker used for surveillance in colorectal cancer, has no correlation with CTCs Citation[92]. The prognostic information that CTCs offer can be added to CEA at baseline and during treatment at 2–3-month intervals, similar to CEA testing. However, we anticipate that they may eventually be used to guide selection of chemotherapeutic agents, either by characterizing CTC populations that contain different markers, such as specific mutations that predict sensitivity or insensitivity to specific drug regimens, or by performing drug testing on CTCs grown in culture or captured fresh Citation[158,159].

For predictive purposes, CTC levels obtained at baseline and at various time points during chemotherapy treatment can correlate with disease progression or response to chemotherapy. Conversion to favorable levels of CTCs during treatment results in significantly longer progression-free and overall survival compared with persistent unfavorable levels of CTCs. Peripheral blood seems to be the best source for CTCs in colorectal cancer, contrary to what we have seen in primary breast cancer where bone marrow seems to be a better source of disseminating tumor cells.

The existence of CTCs in the peripheral blood reveals the existence of a vascularized primary tumor and perhaps metastases, but there are still many aspects about CTCs that need to be investigated to elucidate the metastatic process, and to identify the small population of cells that are highly aggressive and capable of forming metastases or cancer recurrence Citation[160]. Only by administration of therapies that eliminate this subpopulation of cells can a cancer can be cured Citation[161,162]. As a biopsy or resection or administration of therapy may influence CTCs levels, multiple measurements of CTCs are necessary in order to adjust the therapeutic regimen accordingly. Thus, it is still unclear whether a single CTC within a CTC microembolus is responsible for forming metastasis, or whether a CTC invades into the target organ and then releases substances that induce mutations in surrounding cells Citation[160].

Finally, cfDNA seems to be an ideal tool for screening and monitoring therapeutic response, especially in colorectal cancer patients receiving anti-EGFR therapy in form of cetuximab or panitumumab, because KRAS mutation, as an evidence of developed resistance, can be detected in the patient’s serum. Once response failure is identified, then single-cell analysis of CTC populations may take precedence.

Five-year view

CTCs clearly have clinical relevance and prognostic significance, as demonstrated by multiple studies. An important next step is standardization of the various approaches of CTC detection, with regards not only to the assays and markers used to detect them but also to the optimal time point in relation to surgical resection. Prospective multi-institutional trials are needed to validate the use of CTCs as prognostic biomarkers and biomarkers of recurrence, much the way that CEA is currently used.

Further exploration of the predictive potential of CTCs is warranted. If high CTC levels in patients with stage I or II disease is a poor prognostic factor for disease recurrence and adjuvant chemotherapy is shown to prolong disease-free survival in these patients, then CTCs can be used as a predictive biomarker to guide therapy. Conversely, selected patients with stage III disease with very low CTC levels and thus a low risk of disease recurrence may not warrant adjuvant chemotherapy.

Obtaining CTC levels at various time points during chemotherapy treatment is a way of tracking response to the treatment before one would typically perform follow-up imaging. Thus, patients who fail to demonstrate treatment response may be spared unnecessary toxicity to ineffective treatment and greater success by changing to a different regimen. CTC levels may also identify patients that are appropriate for a treatment break Citation[90].

CTC detection as well as genomic analysis of the individual cells can inform adjuvant therapy decisions. Single-cell analysis of CTCs has been performed in other cancers and if applied to colorectal cancer, may be rather useful not only for predicting response to chemotherapy, as well as identifying new genes against which therapy can be developed Citation[131,132,163]. Analysis of mutations in CTCs, which may differ from mutations in the primary tumor, has potential to direct the chemotherapeutic agents used for adjuvant therapy Citation[77]. Whereas our current practice is to use the resected primary tumor’s mutation profile to direct adjuvant therapy, the mutation profile of the CTCs may be more appropriate for directing therapy. In addition, single-cell profiling of CTCs has the potential to play a significant role in the development of novel targeted therapies for colorectal and other cancers Citation[82].

As detection of CTCs in peripheral blood has been shown to correlate with disease recurrence, prospective studies to measure CTC levels along with CEA levels are required to determine if CTCs can be used clinically to screen for metastases.

Circulating angiogenic factors as a potential prognostic factor is another important area of study. Low levels of PIGF, which belongs to the VEGF family of growth factors and may have a role in pathologic neovascularization, are associated with poor recurrence-free survival Citation[164].

Since CTCs are present in patients with localized cancers, CTC detection in conjunction with single-cell sequencing also has the potential to be a diagnostic or screening test for cancer. Thus, for colorectal cancer, a blood draw could even replace the invasive colonoscopy as the preferred mode for screening and surveillance Citation[133].

Finally, cfDNA may eventually supplant CTCs as a monitoring tool Citation[165]. For example, it has been shown that KRAS analysis can be performed in plasma and is a feasible alternative to tissue analysis, thus offering another way to predict response to chemotherapy agents Citation[166]. Another application of cfDNA is monitoring for colorectal cancer recurrence after curative resection Citation[167]. However, unlike CTCs, cfDNA cannot be analyzed for cancer tumor cell heterogeneity nor used for drug testing; therefore, it is likely that both tools will be used to advance personalized therapy of CRC.

Table 1. TNM staging of colon and rectal cancer, American Joint Committee on Cancer Manual, 7th edition.

Table 2. Enrichment of circulating tumor cells by positive (using anti-EpCAM antibodies) and negative (non-EpCAM-based) selection.

Table 3. Enrichment of circulating tumor cells by utilization of other cell characteristics.

Key issues

  • • Serum measurement of carcinoembryonic antigen is a proven method of detecting liver metastases from colorectal cancer. It has limited prognostic and predictive value.

  • • Tumor-specific markers have demonstrated predictive value that can direct adjuvant chemotherapy. KRAS mutation status predicts poor response to EGFR inhibitors such as cetuximab. Intact SMAD4 expression predicts improved response to 5-fluorouracil-based chemotherapy.

  • • Tumor-specific markers also have prognostic value. Colorectal cancers carrying the BRAF mutation have worse prognosis. While loss of heterozygosity at chromosome 18q does not have prognostic significance, loss of SMAD4, which is encoded by 18q, does confer worse prognosis.

  • • Circulating tumor cells (CTCs) play a critical role in the process of metastasis. Genetic exchange among tumor cells can lead to the development of highly metastatic phenotypes within the same patient.

  • • The heterogeneity of the genetic profiles of cells from the primary tumor, metastatic tumors and CTCs may be an explanation for variable response to EGFR-inhibitor chemotherapy.

  • • CTCs are not only a marker for advanced disease, but also have prognostic potential. In addition, CTCs have predictive power, as a decrease in CTC level during chemotherapy is correlated with improved response to chemotherapy.

  • • There are numerous methods for the enrichment and detection of CTCs. Further studies are required for harmonization of these methods before CTCs can be adapted for widespread clinical use.

  • • Single-cell analysis of CTCs, as well as cells from primary and metastatic tumors, has the potential to enable more targeted therapy for colorectal cancer by identifying different subpopulations of cancer cells responsible for metastatic growth and spread that may require different treatments.

  • • Cell-free DNA has the potential to be an easily obtainable, powerful alternative for monitoring therapeutic response in colorectal cancer patients.

  • • As for CTCs, there is also a large number of different detection methods for cell-free DNA, which requires standardization and clinical trial validation.

Financial & competing interests disclosure

This article was supported in part by the John and Marva Warnock Cancer Research Fund (SS Jeffrey) and fellowship funds from Natalie and Vladimir Ermakoff (E Kidess). SS Jeffrey is an inventor of the MagSweeper technology discussed in this manuscript. Stanford University has licensed this technology to Illumina, Inc., and receives licensing royalties; SS Jeffrey has donated her royalties to support student programs at The Jackson Laboratory, a nonprofit biomedical research institution. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

No writing assistance was utilized in the production of this manuscript.

References

  • Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127(12), 2893–2917 (2010).
  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA. Cancer J. Clin. 63(1), 11–30 (2013).
  • Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA. Cancer J. Clin. 55(2), 74–108 (2005).
  • Hayat MJ, Howlader N, Reichman ME, Edwards BK. Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology, and End Results (SEER) Program. Oncologist 12(1), 20–37 (2007).
  • Maringe C, Walters S, Rachet B et al.; ICBP Module 1 Working Group. Stage at diagnosis and colorectal cancer survival in six high-income countries: A population-based study of patients diagnosed during 2000-2007. Acta Oncol. 52(5), 919–932 (2013).
  • Mantke R, Schmidt U, Wolff S, Kube R, Lippert H. Incidence of synchronous liver metastases in patients with colorectal cancer in relationship to clinico-pathologic characteristics. Results of a German prospective multicentre observational study. Eur. J. Surg. Oncol. 38(3), 259–265 (2012).
  • Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J, Bouvier AM. Epidemiology and management of liver metastases from colorectal cancer. Ann. Surg. 244(2), 254–259 (2006).
  • Mitry E, Guiu B, Cosconea S, Jooste V, Faivre J, Bouvier AM. Epidemiology, management and prognosis of colorectal cancer with lung metastases: a 30-year population-based study. Gut 59(10), 1383–1388 (2010).
  • Watanabe T, Wu TT, Catalano PJ et al. Molecular predictors of survival after adjuvant chemotherapy for colon cancer. N. Engl. J. Med. 344(16), 1196–1206 (2001).
  • Locker GY, Hamilton S, Harris J et al.; ASCO. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J. Clin. Oncol. 24(33), 5313–5327 (2006).
  • Wanebo HJ, Rao B, Pinsky CM et al. Preoperative carcinoembryonic antigen level as a prognostic indicator in colorectal cancer. N. Engl. J. Med. 299(9), 448–451 (1978).
  • Goslin R, Steele G Jr, Macintyre J et al. The use of preoperative plasma CEA levels for the stratification of patients after curative resection of colorectal cancers. Ann. Surg. 192(6), 747–751 (1980).
  • Wolmark N, Fisher B, Wieand HS et al. The prognostic significance of preoperative carcinoembryonic antigen levels in colorectal cancer. Results from NSABP (National Surgical Adjuvant Breast and Bowel Project) clinical trials. Ann. Surg. 199(4), 375–382 (1984).
  • Laubert T, Bente V, Freitag-Wolf S et al. Aneuploidy and elevated CEA indicate an increased risk for metachronous metastasis in colorectal cancer. Int. J. Colorectal Dis. 28(6), 767–775 (2013).
  • Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinical score for predicting recurrence after hepatic resection for metastatic colorectal cancer: analysis of 1001 consecutive cases. Ann. Surg. 230(3), 309–18; discussion 318 (1999).
  • Nordlinger B, Guiguet M, Vaillant JC et al. Surgical resection of colorectal carcinoma metastases to the liver. A prognostic scoring system to improve case selection, based on 1568 patients. Association Française de Chirurgie. Cancer 77(7), 1254–1262 (1996).
  • Wichmann MW, Müller C, Lau-Werner U et al. The role of carcinoembryonic antigen for the detection of recurrent disease following curative resection of large-bowel cancer. Langenbecks. Arch. Surg. 385(4), 271–275 (2000).
  • Desch CE, Benson AB 3rd, Somerfield MR et al.; American Society of Clinical Oncology. Colorectal cancer surveillance: 2005 update of an American Society of Clinical Oncology practice guideline. J. Clin. Oncol. 23(33), 8512–8519 (2005).
  • Goldstein MJ, Mitchell EP. Carcinoembryonic antigen in the staging and follow-up of patients with colorectal cancer. Cancer Invest. 23(4), 338–351 (2005).
  • Körner H, Söreide K, Stokkeland PJ, Söreide JA. Diagnostic accuracy of serum-carcinoembryonic antigen in recurrent colorectal cancer: a receiver operating characteristic curve analysis. Ann. Surg. Oncol. 14(2), 417–423 (2007).
  • Hanke B, Riedel C, Lampert S et al. CEA and CA 19-9 measurement as a monitoring parameter in metastatic colorectal cancer (CRC) under palliative first-line chemotherapy with weekly 24-hour infusion of high-dose 5-fluorouracil (5-FU) and folinic acid (FA). Ann. Oncol. 12(2), 221–226 (2001).
  • Prolla TA. DNA mismatch repair and cancer. Curr. Opin. Cell Biol. 10(3), 311–316 (1998).
  • Marra G, Jiricny J. DNA mismatch repair and colon cancer. Adv. Exp. Med. Biol. 570, 85–123 (2005).
  • Roth AD, Delorenzi M, Tejpar S et al. Integrated analysis of molecular and clinical prognostic factors in stage II/III colon cancer. J. Natl Cancer Inst. 104(21), 1635–1646 (2012).
  • Van Cutsem E, Köhne CH, Hitre E et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N. Engl. J. Med. 360(14), 1408–1417 (2009).
  • Van Cutsem E, Köhne CH, Láng I et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J. Clin. Oncol. 29(15), 2011–2019 (2011).
  • Bokemeyer C, Bondarenko I, Hartmann JT et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann. Oncol. 22(7), 1535–1546 (2011).
  • Douillard JY, Siena S, Cassidy J et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J. Clin. Oncol. 28(31), 4697–4705 (2010).
  • Maughan TS, Adams RA, Smith CG et al.; MRC COIN Trial Investigators. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised Phase 3 MRC COIN trial. Lancet 377(9783), 2103–2114 (2011).
  • Tveit KM, Guren T, Glimelius B et al. Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (Nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: the NORDIC-VII study. J. Clin. Oncol. 30(15), 1755–1762 (2012).
  • Roth AD, Tejpar S, Delorenzi M et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J. Clin. Oncol. 28(3), 466–474 (2010).
  • De Roock W, Claes B, Bernasconi D et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 11(8), 753–762 (2010).
  • Tol J, Dijkstra JR, Klomp M et al. Markers for EGFR pathway activation as predictor of outcome in metastatic colorectal cancer patients treated with or without cetuximab. Eur. J. Cancer 46(11), 1997–2009 (2010).
  • Bokemeyer C, Van Cutsem E, Rougier P et al. Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur. J. Cancer 48(10), 1466–1475 (2012).
  • Pai RK, Jayachandran P, Koong AC et al. BRAF-mutated, microsatellite-stable adenocarcinoma of the proximal colon: an aggressive adenocarcinoma with poor survival, mucinous differentiation, and adverse morphologic features. Am. J. Surg. Pathol. 36(5), 744–752 (2012).
  • Corcoran RB, Ebi H, Turke AB et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2(3), 227–235 (2012).
  • Mao M, Tian F, Mariadason JM et al. Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clin. Cancer Res. 19(3), 657–667 (2013).
  • Yang H, Higgins B, Kolinsky K et al. Antitumor activity of BRAF inhibitor vemurafenib in preclinical models of BRAF-mutant colorectal cancer. Cancer Res. 72(3), 779–789 (2012).
  • Jen J, Kim H, Piantadosi S et al. Allelic loss of chromosome 18q and prognosis in colorectal cancer. N. Engl. J. Med. 331(4), 213–221 (1994).
  • Popat S, Houlston RS. A systematic review and meta-analysis of the relationship between chromosome 18q genotype, DCC status and colorectal cancer prognosis. Eur. J. Cancer 41(14), 2060–2070 (2005).
  • Barratt PL, Seymour MT, Stenning SP et al.; UKCCCR AXIS trial collaborators. Adjuvant X-ray and Fluorouracil Infusion Study. DNA markers predicting benefit from adjuvant fluorouracil in patients with colon cancer: a molecular study. Lancet 360(9343), 1381–1391 (2002).
  • Ogino S, Nosho K, Irahara N et al. Prognostic significance and molecular associations of 18q loss of heterozygosity: a cohort study of microsatellite stable colorectal cancers. J. Clin. Oncol. 27(27), 4591–4598 (2009).
  • Alazzouzi H, Alhopuro P, Salovaara R et al. SMAD4 as a prognostic marker in colorectal cancer. Clin. Cancer Res. 11(7), 2606–2611 (2005).
  • Alhopuro P, Alazzouzi H, Sammalkorpi H et al. SMAD4 levels and response to 5-fluorouracil in colorectal cancer. Clin. Cancer Res. 11(17), 6311–6316 (2005).
  • Zhang B, Halder SK, Kashikar ND et al. Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology 138(3), 969–80.e1 (2010).
  • Boulay JL, Mild G, Lowy A et al. SMAD4 is a predictive marker for 5-fluorouracil-based chemotherapy in patients with colorectal cancer. Br. J. Cancer 87(6), 630–634 (2002).
  • Xie W, Rimm DL, Lin Y, Shih WJ, Reiss M. Loss of Smad signaling in human colorectal cancer is associated with advanced disease and poor prognosis. Cancer J. 9(4), 302–312 (2003).
  • Allard WJ, Matera J, Miller MC 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. 10(20), 6897–6904 (2004).
  • Glaves D. Correlation between circulating cancer cells and incidence of metastases. Br. J. Cancer 48(5), 665–673 (1983).
  • Mayhew E, Glaves D. Quantitation of tumorigenic disseminating and arrested cancer cells. Br. J. Cancer 50(2), 159–166 (1984).
  • Chang YS, di Tomaso E, McDonald DM, Jones R, Jain RK, Munn LL. Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc. Natl Acad. Sci. USA 97(26), 14608–14613 (2000).
  • Weiss L, Mayhew E, Rapp DG, Holmes JC. Metastatic inefficiency in mice bearing B16 melanomas. Br. J. Cancer 45(1), 44–53 (1982).
  • Luzzi KJ, MacDonald IC, Schmidt EE et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 153(3), 865–873 (1998).
  • Wong CW, Lee A, Shientag L et al. Apoptosis: an early event in metastatic inefficiency. Cancer Res. 61(1), 333–338 (2001).
  • Weiss L, Nannmark U, Johansson BR, Bagge U. Lethal deformation of cancer cells in the microcirculation: a potential rate regulator of hematogenous metastasis. Int. J. Cancer 50(1), 103–107 (1992).
  • Barbera-Guillem E, Weiss L. Cancer-cell traffic in the liver. III Lethal deformation of B16 melanoma cells in liver sinusoids. Int. J. Cancer 54(5), 880–884 (1993).
  • Berezovskaya O, Schimmer AD, Glinskii AB et al. Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res. 65(6), 2378–2386 (2005).
  • Chen CL, Mahalingam D, Osmulski P et al. Single-cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT-related genes in metastatic prostate cancer. Prostate 73(8), 813–826 (2013).
  • Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L, Lee A, Muschel RJ. Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis. Nat. Med. 6(1), 100–102 (2000).
  • Glinsky VV, Glinsky GV, Glinskii OV et al. Intravascular metastatic cancer cell homotypic aggregation at the sites of primary attachment to the endothelium. Cancer Res. 63(13), 3805–3811 (2003).
  • Glinsky GV, Glinskii AB, Berezovskaya O et al. Dual-color-coded imaging of viable circulating prostate carcinoma cells reveals genetic exchange between tumor cells in vivo, contributing to highly metastatic phenotypes. Cell Cycle 5(2), 191–197 (2006).
  • Tome Y, Tsuchiya H, Hayashi K et al. In vivo gene transfer between interacting human osteosarcoma cell lines is associated with acquisition of enhanced metastatic potential. J. Cell. Biochem. 108(2), 362–367 (2009).
  • Kallergi G, Papadaki MA, Politaki E, Mavroudis D, Georgoulias V, Agelaki S. Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 13(3), R59 (2011).
  • Kasimir-Bauer S, Hoffmann O, Wallwiener D, Kimmig R, Fehm T. Expression of stem cell and epithelial-mesenchymal transition markers in primary breast cancer patients with circulating tumor cells. Breast Cancer Res. 14(1), R15 (2012).
  • Aktas B, Tewes M, Fehm T, Hauch S, Kimmig R, Kasimir-Bauer S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 11(4), R46 (2009).
  • Lecharpentier A, Vielh P, Perez-Moreno P, Planchard D, Soria JC, Farace F. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br. J. Cancer 105(9), 1338–1341 (2011).
  • Raimondi C, Gradilone A, Naso G et al. Epithelial-mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients. Breast Cancer Res. Treat. 130(2), 449–455 (2011).
  • Armstrong AJ, Marengo MS, Oltean S et al. Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol. Cancer Res. 9(8), 997–1007 (2011).
  • Yu M, Bardia A, Wittner BS et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339(6119), 580–584 (2013).
  • Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 20(5), 576–590 (2011).
  • Khoja L, Backen A, Sloane R et al. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br. J. Cancer 106(3), 508–516 (2012).
  • Tsuji T, Ibaragi S, Hu GF. Epithelial-mesenchymal transition and cell cooperativity in metastasis. Cancer Res. 69(18), 7135–7139 (2009).
  • Wicki A, Lehembre F, Wick N, Hantusch B, Kerjaschki D, Christofori G. Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell 9(4), 261–272 (2006).
  • Baccelli I, Schneeweiss A, Riethdorf S et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat. Biotechnol. (2013).
  • Navin N, Kendall J, Troge J et al. Tumour evolution inferred by single-cell sequencing. Nature 472(7341), 90–94 (2011).
  • Gerlinger M, Rowan AJ, Horswell S et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366(10), 883–892 (2012).
  • Mostert B, Jiang Y, Sieuwerts AM et al. KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. Int. J. Cancer 133(1), 130–141 (2013).
  • Thebo JS, Senagore AJ, Krygier S, Luchtefeld MA. K-ras mutational analysis of polyclonal colorectal cancers identifies uniclonal circulating tumor cells. Am. Surg. 67(8), 802–805 (2001).
  • Gasch C, Bauernhofer T, Pichler M et al. Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin. Chem. 59(1), 252–260 (2013).
  • Nadal R, Fernandez A, Sanchez-Rovira P et al. Biomarkers characterization of circulating tumour cells in breast cancer patients. Breast Cancer Res. 14(3), R71 (2012).
  • Carvalho FL, Simons BW, Antonarakis ES et al. Tumorigenic potential of circulating prostate tumor cells. Oncotarget 4(3), 413–421 (2013).
  • Powell AA, Talasaz AH, Zhang H et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS ONE 7(5), e33788 (2012).
  • Rahbari NN, Bork U, Kircher A et al. Compartmental differences of circulating tumor cells in colorectal cancer. Ann. Surg. Oncol. 19(7), 2195–2202 (2012).
  • Joosse SA, Pantel K. Biologic challenges in the detection of circulating tumor cells. Cancer Res. 73(1), 8–11 (2013).
  • Mostert B, Sleijfer S, Foekens JA, Gratama JW. Circulating tumor cells (CTCs): detection methods and their clinical relevance in breast cancer. Cancer Treat. Rev. 35(5), 463–474 (2009).
  • Patriarca C, Macchi RM, Marschner AK, Mellstedt H. Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat. Rev. 38(1), 68–75 (2012).
  • Munz M, Baeuerle PA, Gires O. The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res. 69(14), 5627–5629 (2009).
  • Neki K, Kawahara H, Watanabe K, Toyama Y, Akiba T, Yanaga K. Usefulness of circulating tumor cells after preliminary chemotherapy for prediction of response to further anticancer therapy in patients with initially unresectable metastatic colorectal cancer. Anticancer Res. 33(4), 1769–1772 (2013).
  • Cristofanilli M, Hayes DF, Budd GT et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 23(7), 1420–1430 (2005).
  • Cohen SJ, Punt CJ, Iannotti N et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J. Clin. Oncol. 26(19), 3213–3221 (2008).
  • de Bono JS, Scher HI, Montgomery RB et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 14(19), 6302–6309 (2008).
  • Aggarwal C, Meropol NJ, Punt CJ et al. Relationship among circulating tumor cells, CEA and overall survival in patients with metastatic colorectal cancer. Ann. Oncol. 24(2), 420–428 (2013).
  • Mego M, Gao H, Lee BN et al. Prognostic Value of EMT-Circulating Tumor Cells in Metastatic Breast Cancer Patients Undergoing High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation. J. Cancer 3, 369–380 (2012).
  • 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 (2010).
  • Talasaz AH, Powell AA, Huber DE 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. USA 106(10), 3970–3975 (2009).
  • Zieglschmid V, Hollmann C, Gutierrez B et al. Combination of immunomagnetic enrichment with multiplex RT-PCR analysis for the detection of disseminated tumor cells. Anticancer Res. 25(3A), 1803–1810 (2005).
  • Nagrath S, Sequist LV, Maheswaran S et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 450(7173), 1235–1239 (2007).
  • Stott SL, Hsu CH, Tsukrov DI et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl Acad. Sci. USA 107(43), 18392–18397 (2010).
  • Saliba AE, Saias L, Psychari E et al. Microfluidic sorting and multimodal typing of cancer cells in self-assembled magnetic arrays. Proc. Natl Acad. Sci. USA 107(33), 14524–14529 (2010).
  • Dharmasiri U, Njoroge SK, Witek MA et al. High-throughput selection, enumeration, electrokinetic manipulation, and molecular profiling of low-abundance circulating tumor cells using a microfluidic system. Anal. Chem. 83(6), 2301–2309 (2011).
  • Ozkumur E, Shah AM, Ciciliano JC et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci. Transl. Med. 5(179), 179ra47 (2013).
  • Conant CG, Schwartz MA, Ionescu-Zanetti C. Well plate-coupled microfluidic devices designed for facile image-based cell adhesion and transmigration assays. J. Biomol. Screen. 15(1), 102–106 (2010).
  • Saucedo-Zeni N, Mewes S, Niestroj R et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int. J. Oncol. (2012).
  • Königsberg R, Gneist M, Jahn-Kuch D et al. Circulating tumor cells in metastatic colorectal cancer: efficacy and feasibility of different enrichment methods. Cancer Lett. 293(1), 117–123 (2010).
  • Naume B, Borgen E, Tøssvik S, Pavlak N, Oates D, Nesland JM. Detection of isolated tumor cells in peripheral blood and in BM: evaluation of a new enrichment method. Cytotherapy 6(3), 244–252 (2004).
  • 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. 72(5), 310–323 (2007).
  • Gleghorn JP, Pratt ED, Denning D et al. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip 10(1), 27–29 (2010).
  • Vona G, Sabile A, Louha M et al. Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulatingtumor cells. Am. J. Pathol. 156(1), 57–63 (2000).
  • Farace F, Massard C, Vimond N et al. A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas. Br. J. Cancer 105(6), 847–853 (2011).
  • Desitter I, Guerrouahen BS, Benali-Furet N et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res. 31(2), 427–441 (2011).
  • Zhu P, Shelton DR, Li S et al. Detection of E. coli O157:H7 by immunomagnetic separation coupled with fluorescence immunoassay. Biosens. Bioelectron. 30(1), 337–341 (2011).
  • Lin HK, Zheng S, Williams AJ et al. Portable filter-based microdevice for detection and characterization of circulating tumor cells. Clin. Cancer Res. 16(20), 5011–5018 (2010).
  • Lu J, Fan T, Zhao Q et al. Isolation of circulating epithelial and tumor progenitor cells with an invasive phenotype from breast cancer patients. Int. J. Cancer 126(3), 669–683 (2010).
  • Tan SJ, Lakshmi RL, Chen P, Lim WT, Yobas L, Lim CT. Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients. Biosens. Bioelectron. 26(4), 1701–1705 (2010).
  • Hou HW, Warkiani ME, Khoo BL et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 3, 1259 (2013).
  • Shim S, Gascoyne P, Noshari J, Hale KS. Dynamic physical properties of dissociated tumor cells revealed by dielectrophoretic field-flow fractionation. Integr. Biol. (Camb). 3(8), 850–862 (2011).
  • Gascoyne PR, Shim S, Noshari J, Becker FF, Stemke-Hale K. Correlations between the dielectric properties and exterior morphology of cells revealed by dielectrophoretic field-flow fractionation. Electrophoresis 34(7), 1042–1050 (2013).
  • Marrinucci D, Bethel K, Lazar D et al. Cytomorphology of circulating colorectal tumor cells:a small case series. J. Oncol. 2010, 861341 (2010).
  • Shimada R, Iinuma H, Akahane T, Horiuchi A, Watanabe T. Prognostic significance of CTCs and CSCs of tumor drainage vein blood in Dukes’ stage B and C colorectal cancer patients. Oncol. Rep. 27(4), 947–953 (2012).
  • de Albuquerque A, Kubisch I, Stölzel U et al. Prognostic and predictive value of circulating tumor cell analysis in colorectal cancer patients. J. Transl. Med. 10, 222 (2012).
  • Iinuma H, Okinaga K, Egami H et al. Usefulness and clinical significance of quantitative real-time RT-PCR to detect isolated tumor cells in the peripheral blood and tumor drainage blood of patients with colorectal cancer. Int. J. Oncol. 28(2), 297–306 (2006).
  • Allen-Mersh TG, McCullough TK, Patel H, Wharton RQ, Glover C, Jonas SK. Role of circulating tumour cells in predicting recurrence after excision of primary colorectal carcinoma. Br. J. Surg. 94(1), 96–105 (2007).
  • Alix-Panabières C. EPISPOT assay: detection of viable DTCs/CTCs in solid tumor patients. Recent Results Cancer Res. 195, 69–76 (2012).
  • Yokobori T, Iinuma H, Shimamura T et al. Plastin3 is a novel marker for circulating tumor cells undergoing the epithelial-mesenchymal transition and is associated with colorectal cancer prognosis. Cancer Res. 73(7), 2059–2069 (2013).
  • Went P, Vasei M, Bubendorf L et al. Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers. Br. J. Cancer 94(1), 128–135 (2006).
  • Sun YF, Xu Y, Yang XR et al. Circulating stem cell-like epithelial cell adhesion molecule-positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection. Hepatology 57(4), 1458–1468 (2013).
  • Pesta M, Fichtl J, Kulda V, Topolcan O, Treska V. Monitoring of circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer. Anticancer Res. 33(5), 2239–2243 (2013).
  • Papavasiliou P, Fisher T, Kuhn J, Nemunaitis J, Lamont J. Circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer. Proc. (Bayl. Univ. Med. Cent). 23(1), 11–14 (2010).
  • Jiao LR, Apostolopoulos C, Jacob J et al. Unique localization of circulating tumor cells in patients with hepatic metastases. J. Clin. Oncol. 27(36), 6160–6165 (2009).
  • Klein CA, Schmidt-Kittler O, Schardt JA, Pantel K, Speicher MR, Riethmüller G. Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc. Natl Acad. Sci. USA 96(8), 4494–4499 (1999).
  • Klein CA, Seidl S, Petat-Dutter K et al. Combined transcriptome and genome analysis of single micrometastatic cells. Nat. Biotechnol. 20(4), 387–392 (2002).
  • Fuhrmann C, Schmidt-Kittler O, Stoecklein NH et al. High-resolution array comparative genomic hybridization of single micrometastatic tumor cells. Nucleic Acids Res. 36(7), e39 (2008).
  • Navin N, Hicks J. Future medical applications of single-cell sequencing in cancer. Genome Med. 3(5), 31 (2011).
  • Cann GM, Gulzar ZG, Cooper S et al. mRNA-Seq of single prostate cancer circulating tumor cells reveals recapitulation of gene expression and pathways found in prostate cancer. PLoS ONE 7(11), e49144 (2012).
  • Tang F, Lao K, Surani MA. Development and applications of single-cell transcriptome analysis. Nat. Methods 8(Suppl. 4), S6–11 (2011).
  • Dalerba P, Kalisky T, Sahoo D et al. Single-cell dissection of transcriptional heterogeneity in human colon tumors. Nat. Biotechnol. 29(12), 1120–1127 (2011).
  • Heitzer E, Auer M, Gasch C et al. Complex tumor genomes inferred from single Circulating tumor Cells by array-CGH and next-generation sequencing. Cancer Res. 73(10), 2965–2975 (2013).
  • Mazutis L, Gilbert J, Ung WL, Weitz DA, Griffiths AD, Heyman JA. Single-cell analysis and sorting using droplet-based microfluidics. Nat. Protoc. 8(5), 870–891 (2013).
  • Oldenhuis CN, Oosting SF, Gietema JA, de Vries EG. Prognostic versus predictive value of biomarkers in oncology. Eur. J. Cancer 44(7), 946–953 (2008).
  • Gazzaniga P, Gianni W, Raimondi C et al. Circulating tumor cells in high-risk nonmetastatic colorectal cancer. Tumour Biol. (2013).
  • Katsuno H, Zacharakis E, Aziz O et al. Does the presence of circulating tumor cells in the venous drainage of curative colorectal cancer resections determine prognosis? A meta-analysis. Ann. Surg. Oncol. 15(11), 3083–3091 (2008).
  • Groot Koerkamp B, Rahbari NN, Büchler MW, Koch M, Weitz J. Circulating Tumor Cells and Prognosis of Patients with Resectable Colorectal Liver Metastases or Widespread Metastatic Colorectal Cancer: A Meta-Analysis. Ann. Surg. Oncol. (2013).
  • Rahbari NN, Aigner M, Thorlund K et al. Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterology 138(5), 1714–1726 (2010).
  • Cohen SJ, Punt CJ, Iannotti N et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann. Oncol. 20(7), 1223–1229 (2009).
  • Sastre J, Maestro ML, Gómez-España A et al. Circulating tumor cell count is a prognostic factor in metastatic colorectal cancer patients receiving first-line chemotherapy plus bevacizumab: a Spanish Cooperative Group for the Treatment of Digestive Tumors study. Oncologist 17(7), 947–955 (2012).
  • Iinuma H, Watanabe T, Mimori K et al. Clinical significance of circulating tumor cells, including cancer stem-like cells, in peripheral blood for recurrence and prognosis in patients with Dukes’ stage B and C colorectal cancer. J. Clin. Oncol. 29(12), 1547–1555 (2011).
  • Tol J, Koopman M, Miller MC et al. Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents. Ann. Oncol. 21(5), 1006–1012 (2010).
  • Jahr S, Hentze H, Englisch S et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 61(4), 1659–1665 (2001).
  • Diaz LA Jr, Williams RT, Wu J et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486(7404), 537–540 (2012).
  • Misale S, Yaeger R, Hobor S et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486(7404), 532–536 (2012).
  • Lecomte T, Berger A, Zinzindohoué F et al. Detection of free-circulating tumor-associated DNA in plasma of colorectal cancer patients and its association with prognosis. Int. J. Cancer 100(5), 542–548 (2002).
  • Heitzer E, Auer M, Hoffmann EM et al. Establishment of tumor-specific copy number alterations from plasma DNA of patients with cancer. Int. J. Cancer 133(2), 346–356 (2013).
  • García-Olmo DC, Picazo MG, Toboso I, Asensio AI, García-Olmo D. Quantitation of cell-free DNA and RNA in plasma during tumor progression in rats. Mol. Cancer 12, 8 (2013).
  • Church TR, Wandell M, Lofton-Day C et al.; for the PRESEPT Clinical Study Steering Committee, Investigators and Study Team. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut (2013).
  • Warren JD, Xiong W, Bunker AM et al. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 9, 133 (2011).
  • Grützmann R, Molnar B, Pilarsky C et al. Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS ONE 3(11), e3759 (2008).
  • Umar A, Boland CR, Terdiman JP et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J. Natl. Cancer Inst. 96(4), 261–268 (2004).
  • Ameri K, Luong R, Zhang H et al. Circulating tumour cells demonstrate an altered response to hypoxia and an aggressive phenotype. Br. J. Cancer 102(3), 561–569 (2010).
  • Kang JH, Krause S, Tobin H, Mammoto A, Kanapathipillai M, Ingber DE. A combined micromagnetic-microfluidic device for rapid capture and culture of rare circulating tumor cells. Lab Chip 12(12), 2175–2181 (2012).
  • Krebs MG, Hou JM, Ward TH, Blackhall FH, Dive C. Circulating tumour cells: their utility in cancer management and predicting outcomes. Ther. Adv. Med. Oncol. 2(6), 351–365 (2010).
  • Baumann M, Krause M, Hill R. Exploring the role of cancer stem cells in radioresistance. Nat. Rev. Cancer 8(7), 545–554 (2008).
  • Clarke MF, Dick JE, Dirks PB et al. Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66(19), 9339–9344 (2006).
  • Stoecklein NH, Hosch SB, Bezler M et al. Direct genetic analysis of single disseminated cancer cells for prediction of outcome and therapy selection in esophageal cancer. Cancer Cell 13(5), 441–453 (2008).
  • Rahbari NN, Reissfelder C, Mühlbayer M et al. Correlation of circulating angiogenic factors with circulating tumor cells and disease recurrence in patients undergoing curative resection for colorectal liver metastases. Ann. Surg. Oncol. 18(8), 2182–2191 (2011).
  • Gorges TM, Schiller J, Schmitz A et al. Cancer therapy monitoring in xenografts by quantitative analysis of circulating tumor DNA. Biomarkers 17(6), 498–506 (2012).
  • Spindler KL, Pallisgaard N, Vogelius I, Jakobsen A. Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clin. Cancer Res. 18(4), 1177–1185 (2012).
  • Levy M, Benesova L, Lipska L et al. Utility of cell-free tumour DNA for post-surgical follow-up of colorectal cancer patients. Anticancer Res. 32(5), 1621–1626 (2012).
  • AJCC Cancer Staging Manual (7th Edition). Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A (Eds). Springer, NY, USA (2010).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.