Skip to main content
Erschienen in: BMC Cancer 1/2012

Open Access 01.12.2012 | Research article

Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition

verfasst von: Tobias M Gorges, Ingeborg Tinhofer, Michael Drosch, Lars Röse, Thomas M Zollner, Thomas Krahn, Oliver von Ahsen

Erschienen in: BMC Cancer | Ausgabe 1/2012

Abstract

Background

Circulating tumour cells (CTCs) have shown prognostic relevance in metastatic breast, prostate, colon and pancreatic cancer. For further development of CTCs as a biomarker, we compared the performance of different protocols for CTC detection in murine breast cancer xenograft models (MDA-MB-231, MDA-MB-468 and KPL-4). Blood samples were taken from tumour bearing animals (20 to 200 mm2) and analysed for CTCs using 1. an epithelial marker based enrichment method (AdnaTest), 2. an antibody independent technique, targeting human gene transcripts (qualitative PCR), and 3. an antibody-independent approach, targeting human DNA-sequences (quantitative PCR). Further, gene expression changes associated with epithelial-to-mesenchymal transition (EMT) were determined with an EMT-specific PCR assay.

Methods

We used the commercially available Adna Test, RT-PCR on human housekeeping genes and a PCR on AluJ sequences to detect CTCs in xenografts models. Phenotypic changes in CTCs were tested with the commercially available “Human Epithelial to Mesenchymal Transition RT-Profiler PCR Array”.

Results

Although the AdnaTest detects as few as 1 tumour cell in 1 ml of mouse blood spiking experiments, no CTCs were detectable with this approach in vivo despite visible metastasis formation. The presence of CTCs could, however, be demonstrated by PCR targeting human transcripts or DNA-sequences - without epithelial pre-enrichment. The failure of CTC detection by the AdnaTest resulted from downregulation of EpCAM, whereas mesenchymal markers like Twist and EGFR were upregulated on CTCs. Such a change in the expression profile during metastatic spread of tumour cells has already been reported and was linked to a biological program termed epithelial-mesenchymal transition (EMT).

Conclusions

The use of EpCAM-based enrichment techniques leads to the failure to detect CTC populations that have undergone EMT. Our findings may explain clinical results where low CTC numbers have been reported even in patients with late metastatic cancers. These results are a starting point for the identification of new markers for detection or capture of CTCs, including the mesenchymal-like subpopulations.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-12-178) contains supplementary material, which is available to authorized users.
Presented by Oliver von Ahsen

Competing interests

All authors, except Ingeborg Tinhofer are full time employees of Bayer Pharma AG and hold company shares.

Authors’ contributions

TG: Performed all experiments except FACS analysis. IT: Manuscript writing and scientific discussion. MD: FACS analysis and scientific discussion. LR: Manuscript writing and scientific discussion. TMZ: Administration, funding and scientific discussion. TK: Administration, funding and scientific discussion. OA: Supervision and manuscript writing. All authors read and approved the final manuscript.

Background

Metastasis is a multistep process in the course of which tumour cells migrate into the circulation, generating circulating tumour cells (CTCs) in the blood stream or disseminated tumour cells (DTCs) that home to the bone marrow [1]. It is thought that epithelial tumour cells have to undergo phenotypic changes to overcome the successive barriers to intravasation, survival in the blood, extravasation and secondary tumour formation [2]. These changes are accompanied by a reduction of cell-cell adhesion, loss of apical-basolateral polarity, and loss of epithelial marker expression whereas the expression of mesenchymal-associated genes is induced [3]. This developmental program, described as epithelial-to-mesenchymal transition (EMT), may be a prerequisite for invasive growth and metastatic spread [4]. Although the role of EMT for CTCs is still under debate, recent findings support the hypothesis that CTC profile changes occur during tumour cell dissemination [57]. As EMT processes in human samples are hard to follow and because of the difficulty of detecting CTCs in humans [8], animal models could help to evaluate and dissect the functional implications of EMT specific processes in vivo.
CTCs could already be detected in blood samples of an orthotopic breast cancer model (MDA-MB-231) using a modified version of the FDA approved CellSearch technique [9]. In xenograft models of prostate cancer, however, CTCs could only be detected after a second round of tumour cell inoculation after one in vivo passage [10]. CTCs isolated from MDA-MB-231 breast cancer xenografts display a more aggressive phenotype than the original cells, indicating the impact of CTCs for metastatic formation [11]. In addition, phenotypic changes on CTCs in animal models were also demonstrated [12], showing that CTCs are technically suitable for functional characterisation. We compared different breast cancer models (MDA-MB-231, MDA-MB-468 and KPL-4) in order to test their suitability to serve as model system for CTC formation. The fast emergence of mesenchymal-related CTC populations led to identification of new markers for CTC detection.

Methods

Cell culture

Human breast cancer cell lines MDA-MB-231 (ECACC, #92020424), MDA-MB-468 (ATCC, #HTB-132) and KPL-4 (Kawasaki Medical School) were cultured in DMEM/F12 + 200 mM glutamine (MDA-MB-231, MDA-MB-468) or RPMI + Glutamax (KPL-4) supplemented with 10% fetal calf serum (FCS) and 1% penicillin/streptomycin. Cells were maintained at 37°C in a humidified incubator with 5% CO2. Adherent cells were harvested at 80% confluency by washing with sterile Dulbecco’s phosphate buffered saline (PBS) followed by trypsinisation for 5 min at 37°C.

Western blot

For the detection of EpCAM (epithelial cell adhesion molecule) and MUC-1 (mucin-1) expression, Western Blot analysis was performed. 20 μg of total cell protein were separated using SDS polyacrylamide gel electrophoresis (SDS-PAGE), and proteins were transferred to a nitrocellulose membrane. The membrane was blocked with PBS containing 0.1% Tween-20 (PBS-T) and 5% skimmed milk powder for 1 h at room temperature (RT). Following the blocking step, the EpCAM (abcam; [E144] (ab32392), 1 : 100) and MUC-1 (abcam; [EP1024Y] (ab45167), 1 : 1,000) primary antibodies were added and incubated over night at 4°C. The blot was washed three times with PBS-T. Next, horseradish peroxidase-conjugated secondary antibody was added to the blocking solution (GE Healthcare: NIF824, 1 : 5,000) and incubated for 2 h at RT. After three washing steps with PBS-T, detection was carried out using Amersham ECL Western Blotting Systems (RPN2232) and Imagequant software. Tubulin (Sigma: T6074, 1:2000) detection was included as loading control. The secondary antibody was purchased from GE Healthcare (NA93IVS).

Flow cytometry

5 x 105 cells were detached by trypsinisation, washed and stained for 30 min at RT in the dark. For the analysis of EpCAM expression tumour cells were stained with an APC-labeled (allophycocyanin) anti-EpCAM antibody (BD Bioscience, Cat. no. 347200; 1:200). FACS (fluorescence activated cell sorting) analysis was performed (FACScalibur) with addition of SYTOX® Red Dead Cell Stain (Invitrogen, Cat. No. S34859) to exclude dead cells. 10,000 cells were counted for the analysis.

AdnaTest system

The AdnaTest System is a method based on immunomagnetic enrichment of epithelial tumour cells using antibodies against EpCAM and MUC-1 followed by mRNA isolation via oligo(dT)-conjugated magnetic particles for the analysis of tumour associated gene expression by RT-PCR (EpCAM, MUC-1, Her2 (human epithelial growth factor receptor 2), and Actin). This assay was developed for the detection of CTCs in 5 ml of human blood. Since much smaller amounts of blood can be collected from murine models (800 to 1,000 μl, or ~1/5 of the recommended blood volume), we reduced the BreastSelect Beads (BreastCancerSelect, Article no. T-1-508) to 25 μl instead of 100 μl per sample. The rest of the protocol was carried out according to the manufacturer’s instructions. In the PCR reaction (run on a Veriti Thermal Cycler, Applied Biosystems), amplicons of the following sizes were generated: EpCAM: 395 base pairs (bp), MUC-1: 293 bp, Her2: 270 bp, and Actin: 114 bp. Visualisation of the PCR products was carried out with a 2100 Bioanalyser using DNA 1000 LabChips (Agilent Technologies), as recommended by AdnaGen.

Qualitative CTC detection - targeting human gene transcripts

100 μl blood samples were collected from mice in EDTA-tubes (S-Monovette, Sarstedt) and transferred to RNAprotect Animal Blood Tubes (Qiagen Cat. No. 76544) for immediate stabilisation of cellular RNA. Next, total RNA was purified using the RNeasy Protect Animal Blood Kit (Qiagen, Cat. No. 73224). cDNA synthesis was performed with 800 ng of RNA using the High Capacity RNA-to-cDNA Kit from Applied Biosystems (Cat. No. 4387406). For CTC detection, we designed human-specific primers targeting mRNA of the housekeeping genes glycerinaldehyd-3-phosphat-dehydrogenase (GAPDH) [Fwd 5`-ATCATCCCTGCCTCTACTGG-3`/Rev 5`-GTCAGGTCCACCACTGACAC-3`] and peptidylprolyl isomerase A (PPIA) [Fwd 5`-TTCATCTGCACTGCCAAGAC-3`/Rev 5`-TCGAGTTGTCCACAGTCAGC-3`] and the mesenchymal marker Vimentin [Fwd 5`-CGAAAACACCCTGCAATCTT-3`/Rev 5`-TCCAGCAGCTTCCTGTAGGT-3`]. PCR Master Mix was prepared by mixing 5 μl cDNA together with 12.5 μl HotStarTaq Master Mix Kit (Qiagen, Cat. No. 203443, 250 U). Primers were added to a final concentration of 0.5 μM and the samples were filled up with RNase-free water to a total volume of 25 μl. Additional amplifications with 2 μl of the BreastDetect primer mix from the AdnaTest (BreastDetect, Adna Article no. T-1-509) were performed to analyse EpCAM, MUC-1 and Her2 gene expression. PCR was performed under the following conditions: after 15 min denaturation at 95°C, 42 cycles were carried out by denaturation at 94°C for 40 s, annealing/extension at 60°C for 40 s and elongation for 30 to 60 s at 72°C. A final elongation step at 72°C for 10 min was followed by storage of the samples at 4°C. The PCR generates products of the following sizes: Vimentin: 618 bp, EpCAM, MUC-1 and Her2 (as mentioned before), GAPDH: 138 bp, and PPIA: 133 bp. Visualisation was done with a Agilent 2100 Bioanalyser and peaks with a concentration above 1 ng/μl were defined as positive (Agilent Assay properties: normal). The actin primers of the AdnaTest could not be evaluated in this setting since they are not human-specific.

Quantitative CTC detection - targeting human DNA (alu-sequences)

200 μl of murine blood were collected and red blood cells were lysed using RBC lysis solution from Qiagen for 10 min at 4°C (Cat. No. 158904). Next, samples were centrifuged for 10 min (450 g), followed by two washing steps using 1 ml of sterile PBS. DNA isolation was performed using DNeasy Mini Kits (Qiagen, Cat. No. 51306). 5 μl of the eluate were taken for qRT-PCR targeting human Alu-sequences [For 5`- CACCTGTAATCCCAGCACTTT-3`/Rev 5`-CCCAGGCTGGAGTGCAGT-3`] [13]. Primers were added to a final concentration of 0.5 μM and the samples were filled up with RNase free water to a total volume of 25 μl. PCR (Real-Time PCR System 7500, Applied Biosystems) using the QuantiTec SYBR Green PCR Kit (Qiagen, Cat. No. 204143) was done as follows: after 15 min denaturation at 95°C, 40 cycles were carried out by denaturation at 94°C for 60 s, annealing/extension at 60°C for 30 s and elongation for 30 at 72°C. Amplification curves were analysed and the number of CTCs were calculated by spiking experiments which were included in every run. DNA from murine blood and water were included as negative controls.

Spiking experiments

Spiking experiments were done to test the sensitivity of the different methods. Fresh EDTA-blood was collected from female athymic NCr nude mice (nu/nu; Taconic). Cultured cells were harvested by trypsinisation and spiked into 1 ml of murine blood for the AdnaTest or 100 to 200 μl for detection of CTCs without antibody pre-enrichment. 1 to 50 cells were collected by pipetting under microscopic control, whereas higher numbers (1,000 and 100,000 cells) were prepared by dilution series. As negative controls, 20 murine blood samples were tested for each approach.

Murine xenografts

5 to 6 week old female NCr nude mice were used throughout all xenograft studies. All experiments were approved by the local regulatory agency Landesamt für Gesundheit und Soziales Berlin (approval number: 1403). Mice were housed according to institutional guidelines with highest animal welfare standards [14]. For in vivo inoculation, harvested cells were counted with a CASY DT Cell Counter and Analyser (Innovatis AG). Cell number and viability were determined using the Analyser Software. Cells were kept at 4°C and resuspended in cold PBS or mixed with Matrigel (1 : 1) (BD Biosciences). 1 x 105 to 2 x 106 viable cells (MDA-MB-231, -468, and KPL-4) were injected orthotopically into mammary fat pads (m.f.p.) of the mice in a volume of 50 μl. Subsequently, mice were monitored for tumour growth.
Alternatively, for testing the stability of marker expression in CTCs, 1 x 105 to 5 x 105 cells of the KPL-4 and MDA-MB-231 line were injected into the tail vein or into the heart of female nude mice in a volume of 150 μl PBS.

Blood collection and autopsy

We obtained EDTA-blood from either the jugular vein, the inferior vena cava or by cardiac puncture as tumours reached sizes between 20 to 200 mm2. Tumours, lymph nodes, lungs and livers were taken out and immediately frozen in liquid nitrogen for the detection of metastases by PCR of human transcripts.

RNA isolation and mRNA amplification from tumours and tissues

Tissue was homogenised using Lysing Matrix D (MP Biomedicals, Cat. No. 116913500) and Fast Prep-24 System (MP Biomedicals, Cat. No. 116003500). RNA was purified using the RNeasy Mini Kit from Qiagen (Cat. no. 74104) and cDNA synthesis was performed with 400 ng of RNA input using the High Capacity RNA-to-cDNA Kit. PCR Master Mix was prepared as followed: 4 μl cDNA, 12.5 μl HotStarTaq Master Mix Kit and 2 μl of the primers from the AdnaTest kit or 0.5 μM in case of GAPDH, PPIA and vimentin. Each sample was filled up to 25 μl with RNase free water. Again, actin was excluded from analysis as signals were obtained in lysed tissues of tumour-free mice (data not shown). The Thermocycler was used for the PCR. Conditions were chosen as follows: 15 min denaturation at 95°C, 40 cycles of PCR were carried out by denaturation at 94°C for 40 s, annealing at 60°C for 40 s and elongation for 60 s at 72°C. Subsequently, termination of the reaction was carried out at 72°C for 10 min followed by storage of the samples at 4°C. 20 tumour-free (naive) mice were analysed as negative controls. Visualisation was done with an Agilent 2100 Bioanalyser and peaks with a concentration above 1 ng/μl were defined as positive (Assay properties set to “normal”).

Gene expression analysis of CTCs

EMT-like changes on CTCs during tumour growth were analysed in orthotopic models using the “Human Epithelial to Mesenchymal Transition RT-Profiler PCR Array” System (SABiosciences, Cat. No: PAHS-090 C). This array profiles the expression of 84 genes that have been associated with EMT. 500 μl of blood were collected and transferred to RNAprotect Animal Blood Tubes. Total RNA was purified using the RNeasy Protect Animal Blood Kit and cDNA synthesis was performed using the RT2 First Strand Kit (SABiosciences, Cat. No. C-03/330401). A pre-amplification step was included using the PreAMP cDNA Synthesis Primer Mix (SABiosciences Cat. No. PBH-9090). Next, sample cocktails were loaded onto the array and amplification was performed as recommended by the vendor. Two murine blood samples were included as negative controls to test the species specificity of the assay. Although the array is intended to be human specific, only 14 genes could be included for data analysis in our study since mRNA signals for the remaining 70 genes were detected in blood samples from control mice. Genes with a cycle threshold (Ct) value below 36 in the control setting were excluded from expression analysis. As positive control, 10 cells of the KPL-4 cell line with epithelial phenotype were spiked into 500 μl of native blood. ΔCt values were calculated by subtracting the Ct value of the target gene from the Ct value of the house-keeping gene (GAPDH). ΔCt values of blood samples taken from tumour-bearing animals (KPL-4, n = 5) were subtracted from ΔCt values of the epithelial control (ΔΔCt). Negative ΔΔCt (relative quantification) values represent upregulated genes whereas positive values indicate downregulation of the markers.

Results

Detection of CTCs using the AdnaTest

To establish sensitivity and specificity of the AdnaTest System, 1 to 50 cells of human breast cancer cell lines MDA-MB-231, MDA-MB-468, and KPL-4 were spiked into 1 ml of blood collected from tumour-free mice. We could detect as few as 1 human cancer cell/1 ml blood when spiking cells of the MDA-MB-468 and KPL-4 line, which express high levels of EpCAM. In contrast, the detection limit for EpCAMlow cells (MDA-MB-231) was reached at 10 cells/ml (Figure 1a - c). We conclude that the detection limit of this assay is dependent on the EpCAM expression, since comparable MUC-1 RNA levels were detected in all three cell lines by a faint band in the samples containing 10 spiked cells (Figure 1a) and more relevant, similar protein expression levels were observed in all three cell lines (Figure 1c). Analysis of non-spiked blood samples revealed no positive signal, confirming the specificity of the assay.
Having validated the threshold, we next tried to detect CTCs in orthotopic breast cancer models (n = 96) bearing tumours of 20 to 200 mm2 in size (MDA-MB-231, MDA-MB-468 and KPL-4). To analyse possible metastatic spread, draining lymph nodes (axillary: LN1), remote lymph nodes (inguinal: LN2), lungs and livers were analysed for the presence of human mRNA. Such evidence for metastases was found in nearly all xenografted animals. The lymph nodes located next to the primary tumour or the lungs were infiltrated first during tumour growth (Figure 2a) and with increasing tumour size, metastases in livers and distant lymph nodes became visible as well. Most of the primary tumours and metastases were positive for EpCAM, MUC-1 and Her2 but in some cases, EpCAM and especially MUC-1 expression disappeared (Figure 2a - d). Despite extensive tumour vascularisation (Figure 2e) and metastatic spread, the AdnaTest system revealed no positive signal for CTCs in blood of any sample collected from jugular vein, inferior vena cava or by cardiac puncture (Figure 2a - d).

CTC detection without pre-enrichment

We hypothesised that phenotypic changes associated with the epithelial-to-mesenchymal transition (EMT) and downregulation of the epithelial surface marker EpCAM could be responsible for our failure to detect CTCs using the AdnaTest. Therefore, we established two EpCAM-independent methods for CTC detection. The methods were based either on mRNA amplification of human gene transcripts (GAPDH, PPIA, EpCAM, MUC-1, Her2 and Vimentin) or amplification of human DNA (Alu-sequences).
One to 10,000 human breast cancer cells (MDA-MB-231, MDA-MB-468 and KPL-4) were spiked into the blood of naive mice for assay validation. No false positive result was seen in blood from tumour-free control mice (n = 20), proving that the used PCR primers were specific to human sequences and therefore did not give any background signals, for example for Vimentin that would be expected in mesenchymal blood cells. All spiked samples showed positive signals for GAPDH and PPIA. As few as 2 tumour cells in 100 μl mouse blood could be reproducibly detected by expression of human housekeeping genes. EpCAM signals were detectable from 2 tumour cells or more for EpCAMhigh cells (MDA-MB-468, KPL-4) but the detection limit was 10 tumour cells in case of EpCAMlow cells (MDA-MB-231) (Figure 3). Vimentin expression was detectable in cells of the basal like MDA-MB-231 line (2 cells/100 μl) and weakly in the MDA-MB-468 line (from 1,000 cells on) whereas no signals for Vimentin were seen in blood samples spiked with cells of the KPL-4 line (Figure 3). The cells also show the described Her2 expression pattern, very strong signals in KPL4 cells and very weak signals in MDA-MB-231 and MDA-MB-468.
For comparison, we also tested the detection of human cells by amplification of repetitive DNA sequences of the AluJ type. This approach could be more robust than testing RNA and should enable the quantification of cells due to the fixed copy number of these sequences. Using this approach, however, signals from 1 tumour cell in 200 μl blood could be detected in only 5 out of 8 (60%) spiking experiments (Figure 4). Signals from 10 cells or more were traceable in 100% of the analysed samples. Thus, the higher abundance of house-keeping gene mRNA leads to higher sensitivity of the RNA based CTC detection.
Having validated the methods, we next tested the performance of these CTC detection protocols in vivo. Using detection of GAPDH and PPIA transcripts, CTCs could be detected in nearly all blood samples collected by cardiac puncture (Figure 5). Positive signals were found in mice bearing tumours of 50 mm2 size, consistent with the appearance of lymph node metastases at this stage (Figure 2). In contrast, no signals for EpCAM, MUC-1 or Her2 transcripts could be detected in these samples, with the exception of one sample (Figure 5). Vimentin expression was, however, found in blood samples of mice bearing MDA-MB-231 tumours and surprisingly, in mice bearing xenografts from the KPL-4 cell line indicating EMT-like changes in this model, since no vimentin-specific transcripts could be detected in spiking experiments using cells taken directly from cell cultures (Figures 3 and 5).
Quantitative analysis revealed that blood samples contain CTCs at numbers which should be sufficient for detection with the AdnaTest, given that this assay detected as few as 1 cell/ml in spiking experiments (Table 1). Tumour cell numbers between 5 and 20 cells/ml (MDA-MB-231 and KPL-4) could be detected by targeting human Alu-sequences. However, no CTCs could be found when MDA-MB-231 cells were transplanted subcutaneously (Table 1).
Table 1
Quantitative detection of CTCs in tumour bearing animals
 Model
Tumour size
CTC
(Application)
(Animals)
(cells/ml)
MDA-MB-231
20mm2
0/5
(orthotopic)
(5)
 
MDA-MB-231
50 mm2
0/5
(orthotopic)
(5)
 
MDA-MB-231
80 mm2
3/5
(orthotopic)
(5)
(5,10,and 15)
MDA-MB-231
120 mm2
1/5
(orthotopic)
(5)
(7)
MDA-MB-231
20-120 mm2
0/20
(subcutaneous)
(20)
 
 KPL-4
50 mm2
0/3
(orthotopic)
(3)
 
 KPL-4
80 mm2
1/2
(orthotopic)
(2)
(20)
 KPL-4
120 mm2
1/4
(orthotopic)
(4)
(15)
CTCs could be quantified by qRT-PCR targeting human Alu-sequences in 6 out of 29 blood samples gained from xenografted animals. CTCs were detectable at tumour sizes between 80 to 140 mm2 (MDA-MB-231 and KPL-4). No CTCs could be detected in a subcutaneous MDA-MB-231 model. Number of CTCs were calculated by spiking rows which were included in every run.

Phenotypic changes of CTCs after intravenous and intracardiac injection

As we found a number of changes in gene expression comparing cell lines grown in vitro and in xenografts, we investigated the time course for changes in EpCAM and MUC-1 gene expression. First, we spiked 10 cells of the KPL-4 line into 100 μl or 1 ml of mouse blood. 30 min to 4 h later gene expression was assessed by qualitative PCR or the AdnaTest. Spiked cells were detected with both methods at all time points indicating stable mRNA and protein levels of EpCAM (ex vivo) (Figure 6a). Next, 100,000 cells of the KPL-4 line were injected into the tail vein of female mice and blood was collected by cardiac puncture for cell detection after 30 min to 4 h. No cells could be detected by the AdnaTest although 5 to 25 cells/ml were detected by amplification of human Alu-sequences (data not shown).
Tumour cells showed dynamic changes and down-regulated EpCAM and Her2 while circulating through the bloodstream (Figure 6a). Interestingly, cells adhering in the lungs kept EpCAM and Her2 mRNA expression (Figure 6b). To confirm these findings, we injected 500,000 cells of the MDA-MB-231 line into the heart of female mice. 45 min - 5 h after injection, blood was taken by cardiac puncture. Again, down-regulation of epithelial EpCAM and MUC-1 was observed, whereas transcripts of the mesenchymal marker vimentin and the house-keeping genes were detected in 100% of the blood samples (Figure 6c).
A more detailed comparative analysis of KPL-4 cells taken from cell culture and circulating KPL-4 cells in blood from tumour-bearing mice revealed changes in expression of EMT-associated genes with upregulation of Twist, Collagen-alpha-2(V)-chain (COL5A2), epidermal growth factor receptor (EGFR), moesin (MSN) and beta-type platelet-derived growth factor receptor (PDGFRB) in the latter. In contrast, expression of epithelial markers like cytokeratin-19 (CK-19) and osteopontin (SPP1) was slightly reduced in CTCs (Figure 7).

Discussion

We confirm here previous reports that the AdnaTest is a very sensitive assay for detection of epithelial cells. We also demonstrate that the sensitivity of the assay was significantly dependent on the expression levels of EpCAM, which was causally linked to its failure to detect any positive CTC sample in murine breast cancer xenograft models. We were able to show that EMT occurs in vivo during formation of metastases in tumour-bearing mice. These changes include the down-regulation of EpCAM. We showed for the first time that gene expression changes in CTCs are very fast and seem to depend on contact with endothelial surfaces or organ systems in vivo, since even i.v. injected tumour cells quickly down-regulated EpCAM. The surprisingly fast loss of EpCAM could be explained by cleavage and shedding of the extracellular domain as described [15, 16]. Besides the down-regulation of EpCAM, we identified up-regulation of mesenchymal markers like COL5A2, EGFR, MSN, PDGFRB and Twist in CTCs, supporting the epithelial-to-mesenchymal transition as the cause for down-regulation of EpCAM. Confirming our findings, COL52A has been identified as marker for EMT, since TGFβ/TNFα-treatment induced a mesenchymal cell phenotype accompanied with increased expression of this gene [17]. EGF signaling has also been previously linked to tumour progression and metastasis [1821]. We found dramatically increased EGFR expression in CTCs, confirming the role of this pathway in the process of metastatic spread. It has been shown that EGF can induce EMT-like effects including upregulation of Twist through the EGFR pathway [22]. Twist expression has been described as a crucial regulator of EMT and metastasis formation [23]. CTCs also showed elevated Twist expression levels in our model, confirming these findings [23]. The modified gene expression profile that we report here could provide the basis for alternative methods for isolation and detection of CTCs from clinical samples. The surface marker EGFR may be useful for capturing mesenchymal-like CTC subpopulations, whereas the intracellular proteins like vimentin, collagen-alpha-2 and Twist could be used for improved detection of CTCs by immunostaining. Such approaches will be especially useful to confirm the identity of CTC populations isolated by size exclusion as recently described [24]. The isolation of CTCs based on their size is independent of epithelial marker expression and therefore leads to higher CTC yields. A larger set of tumor-specific markers will be useful in confirming the identity of CTC populations enriched by filtration methods.
In contrast to this study, CTC detection has previously been described using an EpCAM-based method in a murine xenograft model [9]. In addition, CTC-positive samples were found in murine prostate cancer xenografts using a CTC-chip [10], which has been previously described and evaluated [25]. Either EpCAM expression was not downregulated in the PC3 model or the CTC chip captures even cells with very little residual EpCAM expression. This hypothesis is supported by the fact that low EpCAM cells are captured late on the CTC-chip whereas cells with high EpCAM expression are bound already in the beginning of the flowpath (pers. communication S. Nagrath). Down-regulation of MUC-1 and Her2 on CTCs has also been noted by this group, in keeping with our findings. More recently, increased vimentin expression, indicative of EMT processes in CTCs in another orthotopic breast cancer model, has been published [26]. Taken together, these results demonstrate that phenotypic changes occur in CTCs. For this reason, we believe that new methods are needed to ensure that the complete phenotypic variability of CTC populations can be described.
Although the prognostic value of CTC counts have been demonstrated using epithelial marker based CTC detection methods [2730], our findings indicate that numerous cells may be lost in many blood samples of cancer patients due to EMT processes. This may explain why even in late stage metastatic patients, only around 50% of the patients with breast or prostate cancer have more than 5 CTCs and in case of colorectal cancer only 26% more than 3 CTCs in 7.5 ml blood. Other authors failed to detect subtypes of CTCs resembling normal type breast cancer using the CellSearch technique [31]. Further, significantly higher numbers of CTCs could be detected using an EpCAM independent detection method compared to EpCAM based enrichment technology (69.2% vs. 42.3%) in breast cancer patients [32], suggesting that a mixture of EpCAM positive and EpCAM negative tumour cells circulate in the blood. In a study of neuroendocrine cancer patients using CellSearch, CTCs were found in only 21-43% of the patients, although 100% of the tumours showed strong membrane EpCAM expression by IHC [33]. Down-regulation of EpCAM has been reported independently for DTCs in bone marrow and CTCs in peripheral blood of patients with breast cancer and for other solid tumors [34, 35]. These publications demonstrate that the use of anti-EpCAM antibody-based enrichment strategies limits the clinical utility the CellSearch System, the AdnaTest or other EpCAM based procedures. Our own data and several recent studies support the hypothesis that EMT-like shifts on CTCs occur during metastatic spread [57, 36]. These results indicate that CTCs will be lost using immuno-affinity enrichment that relies only on the level of EpCAM expression on the cell surface [37].
Taken together, our findings explain low CTC numbers in clinical studies even in metastatic cancer. The use of EpCAM-based enrichment techniques leads to the selective loss of CTC populations undergoing epithelial-to-mesenchymal transition. We could show that new technologies have to be developed which securely isolate all CTCs. Methods for the analysis of CTCs in clinical samples should be designed to detect both epithelial and mesenchymal-like subpopulations. Our findings could be used as a starting point for the qualification of new markers for CTC detection or capture including the mesenchymal-like subpopulations. Future studies should consider the heterogeneity of CTC populations and find out which one is prognostic for the course of the disease and which population may give best hints towards therapeutic options.

Conclusion

As we have shown in this work, the use of single surface markers for isolation of CTCs can lead to the loss of CTC populations having undergone phenotypic changes like the epithelial to mesenchymal transition. This finding may explain the clinical results with low CTC counts or even absence of detectable CTCs in many even late metastatic cancer patients. Future research has to identify new selection markers that are robustly expressed and not subject to phenotypic changes. An alternative is the enrichment of CTCs based on size and a detailed molecular analysis of the enriched cell population especially in respect to predictive markers like target expression and/or specific mutations.

Acknowledgements

We thank Tanja Rose, Daniela Foerster and Elke Schmid for expert technical assistance, Dr. David Henderson, Dr. Christoph Schatz and Dr. Marcus Conrad for many stimulating discussions and the Kawasaki Medical School for providing the KPL-4 breast cancer cell line.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

All authors, except Ingeborg Tinhofer are full time employees of Bayer Pharma AG and hold company shares.

Authors’ contributions

TG: Performed all experiments except FACS analysis. IT: Manuscript writing and scientific discussion. MD: FACS analysis and scientific discussion. LR: Manuscript writing and scientific discussion. TMZ: Administration, funding and scientific discussion. TK: Administration, funding and scientific discussion. OA: Supervision and manuscript writing. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Fidler IJ: The pathogenesis of cancer metastasis: the seed and soil hypothesis revisited. Nat Rev Cancer. 2003, 3: 453-458. 10.1038/nrc1098.CrossRefPubMed Fidler IJ: The pathogenesis of cancer metastasis: the seed and soil hypothesis revisited. Nat Rev Cancer. 2003, 3: 453-458. 10.1038/nrc1098.CrossRefPubMed
2.
Zurück zum Zitat Bonnomet A, Brysse A, Tachsidis A, Waltham M, Thompson EW, Polette M, Gilles C: Epithelial-to-mesenchymal transitions and circulating tumour cells. J Mammary Gland Biol Neoplasia. 2010, 15: 261-273. 10.1007/s10911-010-9174-0.CrossRefPubMed Bonnomet A, Brysse A, Tachsidis A, Waltham M, Thompson EW, Polette M, Gilles C: Epithelial-to-mesenchymal transitions and circulating tumour cells. J Mammary Gland Biol Neoplasia. 2010, 15: 261-273. 10.1007/s10911-010-9174-0.CrossRefPubMed
3.
Zurück zum Zitat May CD, Sphyris N, Evans KW, Werden SJ, Guo W, Mani SA: Epithelial-mesenchymal transition and cancer stem cells: a dangerously dynamic duo in breast cancer progression. Breast Cancer Res. 2011, 13: 202-CrossRefPubMedPubMedCentral May CD, Sphyris N, Evans KW, Werden SJ, Guo W, Mani SA: Epithelial-mesenchymal transition and cancer stem cells: a dangerously dynamic duo in breast cancer progression. Breast Cancer Res. 2011, 13: 202-CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Yang J, Weinberg RA: Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008, 14: 818-829. 10.1016/j.devcel.2008.05.009. ReviewCrossRefPubMed Yang J, Weinberg RA: Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008, 14: 818-829. 10.1016/j.devcel.2008.05.009. ReviewCrossRefPubMed
5.
Zurück zum Zitat Hou J-M, Krebs M, Ward T, Sloane R, Priest L, Hughes H, Clack G, Ranson M, Blackhall F, Dive C: Circulating Tumour Cells as a Window on Metastasis Biology in Lung Cancer. Am J Pathol. 2011, 178: 989-996. 10.1016/j.ajpath.2010.12.003.CrossRefPubMedPubMedCentral Hou J-M, Krebs M, Ward T, Sloane R, Priest L, Hughes H, Clack G, Ranson M, Blackhall F, Dive C: Circulating Tumour Cells as a Window on Metastasis Biology in Lung Cancer. Am J Pathol. 2011, 178: 989-996. 10.1016/j.ajpath.2010.12.003.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Mego M, Mani SA, Lee BN, Li1 C, Evans Kw, Cohen EN, Gaol H, Jackson SA, Giordano A, Hortobagyi GN, Cristofanilli M, Lucci A, Reuben JM: Expression of epithelial-mesenchymal transition-inducing transcription factors in primary breast cancer: the effect of neoadjuvant therapy. Int J Cancer. 2012, 130: 808-816. 10.1002/ijc.26037.CrossRefPubMed Mego M, Mani SA, Lee BN, Li1 C, Evans Kw, Cohen EN, Gaol H, Jackson SA, Giordano A, Hortobagyi GN, Cristofanilli M, Lucci A, Reuben JM: Expression of epithelial-mesenchymal transition-inducing transcription factors in primary breast cancer: the effect of neoadjuvant therapy. Int J Cancer. 2012, 130: 808-816. 10.1002/ijc.26037.CrossRefPubMed
7.
Zurück zum Zitat 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. 2011, 13: R59-10.1186/bcr2896.CrossRefPubMedPubMedCentral 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. 2011, 13: R59-10.1186/bcr2896.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Pantel K, Alix-Panabieres C: Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010, 16: 398-406. 10.1016/j.molmed.2010.07.001.CrossRefPubMed Pantel K, Alix-Panabieres C: Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010, 16: 398-406. 10.1016/j.molmed.2010.07.001.CrossRefPubMed
9.
Zurück zum Zitat Eliane JP, Repollet M, Luker KE, Brown M, Rae JM, Dontu G, Schott AF, Wicha M, Doyle GV, Hayes DF, Luker GD: Monitoring serial changes in circulating human breast cancer cells in murine xenograft models. Cancer Res. 2008, 68: 5529-5532. 10.1158/0008-5472.CAN-08-0630.CrossRefPubMedPubMedCentral Eliane JP, Repollet M, Luker KE, Brown M, Rae JM, Dontu G, Schott AF, Wicha M, Doyle GV, Hayes DF, Luker GD: Monitoring serial changes in circulating human breast cancer cells in murine xenograft models. Cancer Res. 2008, 68: 5529-5532. 10.1158/0008-5472.CAN-08-0630.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat He H, Yang X, Davidson AJ, Wu D, Marshall FF, Chung LW, Zhau HE, Wang R: Progressive epithelial to mesenchymal transitions in ARCaPE prostate cancer cells during xenograft tumour formation and metastasis. Prostate. 2010, 70: 518-520.CrossRefPubMedPubMedCentral He H, Yang X, Davidson AJ, Wu D, Marshall FF, Chung LW, Zhau HE, Wang R: Progressive epithelial to mesenchymal transitions in ARCaPE prostate cancer cells during xenograft tumour formation and metastasis. Prostate. 2010, 70: 518-520.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ameri K, Luong R, Zhang H, Powell AA, Montgomery KD, Espinosa I, Bouley DM, Harris AL, Jeffrey SS: Circulating tumour cells demonstrate an altered response to hypoxia and an aggressive phenotype. Brit J Cancer. 2010, 102: 561-569. 10.1038/sj.bjc.6605491.CrossRefPubMedPubMedCentral Ameri K, Luong R, Zhang H, Powell AA, Montgomery KD, Espinosa I, Bouley DM, Harris AL, Jeffrey SS: Circulating tumour cells demonstrate an altered response to hypoxia and an aggressive phenotype. Brit J Cancer. 2010, 102: 561-569. 10.1038/sj.bjc.6605491.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Helzer KT, Barness HE, Day L, Harvey J, Billings PR, Forsyth A: Circulating tumour cells are transcriptionally similar to the primary tumour in a murine prostate model. Cancer Res. 2009, 69: 7860-7866. 10.1158/0008-5472.CAN-09-0801.CrossRefPubMed Helzer KT, Barness HE, Day L, Harvey J, Billings PR, Forsyth A: Circulating tumour cells are transcriptionally similar to the primary tumour in a murine prostate model. Cancer Res. 2009, 69: 7860-7866. 10.1158/0008-5472.CAN-09-0801.CrossRefPubMed
13.
Zurück zum Zitat Schneider T, Osl F, Friess T, Stockinger H, Scheuer WV: Quantification of human Alu sequences by real-time PCR - an improved method to measure therapeutic efficacy of anti-metastatic drugs in human xenotransplants. Clin Exp Metastasis. 2002, 19: 571-582. 10.1023/A:1020992411420.CrossRefPubMed Schneider T, Osl F, Friess T, Stockinger H, Scheuer WV: Quantification of human Alu sequences by real-time PCR - an improved method to measure therapeutic efficacy of anti-metastatic drugs in human xenotransplants. Clin Exp Metastasis. 2002, 19: 571-582. 10.1023/A:1020992411420.CrossRefPubMed
14.
Zurück zum Zitat Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ, Double JA, Everitt J, Farningham DA, Glennie MJ, Kelland LR, Robinson V, Stratford IJ, Tozer GM, Watson S, Wedge SR, Eccles SA, Committee of the National Cancer Research Institute: . Guidelines for the welfare and use of animals in cancer research. Br J Cancer. 2010, 102: 1555-1577. 10.1038/sj.bjc.6605642.CrossRefPubMedPubMedCentral Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ, Double JA, Everitt J, Farningham DA, Glennie MJ, Kelland LR, Robinson V, Stratford IJ, Tozer GM, Watson S, Wedge SR, Eccles SA, Committee of the National Cancer Research Institute: . Guidelines for the welfare and use of animals in cancer research. Br J Cancer. 2010, 102: 1555-1577. 10.1038/sj.bjc.6605642.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Denzel S, Maetzel D, Mack B, Eggert C, Bärr G, Gires O: Initial activation of EpCAM cleavage via cell-to-cell contact. BMC Cancer. 2009, 9: 402-10.1186/1471-2407-9-402.CrossRefPubMedPubMedCentral Denzel S, Maetzel D, Mack B, Eggert C, Bärr G, Gires O: Initial activation of EpCAM cleavage via cell-to-cell contact. BMC Cancer. 2009, 9: 402-10.1186/1471-2407-9-402.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, Kieu C, Papior P, Baeuerle PA, Munz M, Gires O: Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol. 2009, 11: 162-171. 10.1038/ncb1824.CrossRefPubMed Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, Kieu C, Papior P, Baeuerle PA, Munz M, Gires O: Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol. 2009, 11: 162-171. 10.1038/ncb1824.CrossRefPubMed
17.
Zurück zum Zitat Asiedu MK, James Ingle N, Marshall Behrens D, Derek Radisky C, Keith Knutson L: TGFβ/TNFα-Mediated Epithelial-Mesenchymal Transition Generates Breast Cancer Stem Cells with a Claudin-Low Phenotype. Cancer Res. 2011, 71: 4707-4719. 10.1158/0008-5472.CAN-10-4554.CrossRefPubMedPubMedCentral Asiedu MK, James Ingle N, Marshall Behrens D, Derek Radisky C, Keith Knutson L: TGFβ/TNFα-Mediated Epithelial-Mesenchymal Transition Generates Breast Cancer Stem Cells with a Claudin-Low Phenotype. Cancer Res. 2011, 71: 4707-4719. 10.1158/0008-5472.CAN-10-4554.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat VerbeeK BS, Adriaansen-Slot SS, Vroom TM, Beckers T, Rijksen G: Overexpression of EGFR and c-erb2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett. 1998, 425: 145-150. 10.1016/S0014-5793(98)00224-5.CrossRefPubMed VerbeeK BS, Adriaansen-Slot SS, Vroom TM, Beckers T, Rijksen G: Overexpression of EGFR and c-erb2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett. 1998, 425: 145-150. 10.1016/S0014-5793(98)00224-5.CrossRefPubMed
19.
Zurück zum Zitat Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ: Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res. 2000, 60: 2926-2935.PubMed Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ: Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res. 2000, 60: 2926-2935.PubMed
20.
Zurück zum Zitat Matsuo M, Sakurai H, Saiki I: ZD1839, a selective epidermal growth factor receptor tyrosine kinase inhibitor, shows antimetastatic activity using a hepatocellular carcinoma model. Mol Cancer Ther. 2003, 2: 557-561.CrossRefPubMed Matsuo M, Sakurai H, Saiki I: ZD1839, a selective epidermal growth factor receptor tyrosine kinase inhibitor, shows antimetastatic activity using a hepatocellular carcinoma model. Mol Cancer Ther. 2003, 2: 557-561.CrossRefPubMed
21.
Zurück zum Zitat Tsuji T, Ibaragi S, Shima K, Hu MG, Katsurano M, Sasaki A, Hu GF: Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumour cell local invasion but suppresses distant colony growth. Cancer Res. 2008, 68: 10377-10386. 10.1158/0008-5472.CAN-08-1444.CrossRefPubMedPubMedCentral Tsuji T, Ibaragi S, Shima K, Hu MG, Katsurano M, Sasaki A, Hu GF: Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumour cell local invasion but suppresses distant colony growth. Cancer Res. 2008, 68: 10377-10386. 10.1158/0008-5472.CAN-08-1444.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbruzzese JL, Hortobagyi GN, Hung MC: Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via upregulation of TWIST gene expression. Cancer Res. 2007, 67: 9066-9076. 10.1158/0008-5472.CAN-07-0575.CrossRefPubMedPubMedCentral Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbruzzese JL, Hortobagyi GN, Hung MC: Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via upregulation of TWIST gene expression. Cancer Res. 2007, 67: 9066-9076. 10.1158/0008-5472.CAN-07-0575.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA: Twist, a master regulator of morphogenesis, plays an essential role in tumour metastasis. Cell. 2004, 117: 927-939. 10.1016/j.cell.2004.06.006.CrossRefPubMed Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA: Twist, a master regulator of morphogenesis, plays an essential role in tumour metastasis. Cell. 2004, 117: 927-939. 10.1016/j.cell.2004.06.006.CrossRefPubMed
24.
Zurück zum Zitat Krebs MG, Hou JM, Sloane R, Lancashire L, Priest L, Nonaka D, Ward TH, Backen A, Clack G, Hughes A, Ranson M, Blackhall FH, Dive C: Analysis of Circulating Tumor Cells in Patients with Non-small Cell Lung Cancer Using Epithelial Marker-Dependent and -Independent Approaches. J Thorac Oncol. 2012, 7: 306-315. 10.1097/JTO.0b013e31823c5c16.CrossRefPubMed Krebs MG, Hou JM, Sloane R, Lancashire L, Priest L, Nonaka D, Ward TH, Backen A, Clack G, Hughes A, Ranson M, Blackhall FH, Dive C: Analysis of Circulating Tumor Cells in Patients with Non-small Cell Lung Cancer Using Epithelial Marker-Dependent and -Independent Approaches. J Thorac Oncol. 2012, 7: 306-315. 10.1097/JTO.0b013e31823c5c16.CrossRefPubMed
25.
Zurück zum Zitat Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A, Ryan P, Balis UJ, Tompkins RG, Haber DA, Toner M: Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007, 450: 1235-1239. 10.1038/nature06385.CrossRefPubMedPubMedCentral Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A, Ryan P, Balis UJ, Tompkins RG, Haber DA, Toner M: Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007, 450: 1235-1239. 10.1038/nature06385.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Bonnomet A, Syne L, Brysse A, Feyereisen E, Thompson EW, Noël A, Foidart JM, Birembaut P, Polette M, Gilles C: A dynamic in vivo model of epithelial-to-mesenchymal transitions in circulating tumor cells and metastases of breast cancer. Oncogene. 2011, 10.1038/onc.2011.540. [Epub ahead of print] Bonnomet A, Syne L, Brysse A, Feyereisen E, Thompson EW, Noël A, Foidart JM, Birembaut P, Polette M, Gilles C: A dynamic in vivo model of epithelial-to-mesenchymal transitions in circulating tumor cells and metastases of breast cancer. Oncogene. 2011, 10.1038/onc.2011.540. [Epub ahead of print]
27.
Zurück zum Zitat Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW, Hayes DF: Circulating tumour cells, disease progression and survival in metastatic breast cancer. New Engl J Med. 2004, 351: 781-791. 10.1056/NEJMoa040766.CrossRefPubMed Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW, Hayes DF: Circulating tumour cells, disease progression and survival in metastatic breast cancer. New Engl J Med. 2004, 351: 781-791. 10.1056/NEJMoa040766.CrossRefPubMed
28.
Zurück zum Zitat De Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, Doyle GV, Terstappen LW, Pienta KJ, Raghavan D: Circulating tumour cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Canc Res. 2008, 14: 6302-6309. 10.1158/1078-0432.CCR-08-0872.CrossRef De Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, Doyle GV, Terstappen LW, Pienta KJ, Raghavan D: Circulating tumour cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Canc Res. 2008, 14: 6302-6309. 10.1158/1078-0432.CCR-08-0872.CrossRef
29.
Zurück zum Zitat Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse MA, Mitchell E, Miller MC, Doyle GV, Tissing H, Terstappen LW, Meropol NJ: Prognostic significance of circulating tumour cells in patients with metastatic colorectal cancer. Ann Onc. 2009, 20: 1223-1229. 10.1093/annonc/mdn786.CrossRef Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse MA, Mitchell E, Miller MC, Doyle GV, Tissing H, Terstappen LW, Meropol NJ: Prognostic significance of circulating tumour cells in patients with metastatic colorectal cancer. Ann Onc. 2009, 20: 1223-1229. 10.1093/annonc/mdn786.CrossRef
30.
Zurück zum Zitat Tewes M, Aktas B, Welt A, Mueller S, Hauch S, Kimmig R, Kasimir-Bauer S: Molecular profiling and predictive value of circulating tumour cells in patients with metastatic breast cancer: an option for monitoring response to breast cancer related therapies. Breast Cancer Res Treat. 2009, 115: 581-590. 10.1007/s10549-008-0143-x.CrossRefPubMed Tewes M, Aktas B, Welt A, Mueller S, Hauch S, Kimmig R, Kasimir-Bauer S: Molecular profiling and predictive value of circulating tumour cells in patients with metastatic breast cancer: an option for monitoring response to breast cancer related therapies. Breast Cancer Res Treat. 2009, 115: 581-590. 10.1007/s10549-008-0143-x.CrossRefPubMed
31.
Zurück zum Zitat Sieuwerts AM, Kraan J, Bolt J, van der Spoel P, Elstrodt F, Schutte M, Martens JW, Gratama JW, Sleijfer S, Foekens JA: Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumour cells. J Natl Cancer Inst. 2009, 101: 61-66.CrossRefPubMedPubMedCentral Sieuwerts AM, Kraan J, Bolt J, van der Spoel P, Elstrodt F, Schutte M, Martens JW, Gratama JW, Sleijfer S, Foekens JA: Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumour cells. J Natl Cancer Inst. 2009, 101: 61-66.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Königsberg R, Obermayr E, Bises G, Pfeiler G, Gneist M, Wrba F, De Santis M, Zeillinger R, Hudec M, Dittrich C: Detection of EpCAM positive and negative circulating tumour cells in metastatic breast cancer patients. Acta Oncol. 2011, 50: 700-710. 10.3109/0284186X.2010.549151.CrossRefPubMed Königsberg R, Obermayr E, Bises G, Pfeiler G, Gneist M, Wrba F, De Santis M, Zeillinger R, Hudec M, Dittrich C: Detection of EpCAM positive and negative circulating tumour cells in metastatic breast cancer patients. Acta Oncol. 2011, 50: 700-710. 10.3109/0284186X.2010.549151.CrossRefPubMed
33.
Zurück zum Zitat Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV, Caplin M, Meyer T: Circulating Tumour Cells and EpCAM Expression in Neuroendocrine Tumours. Clin Cancer Res. 2011, 17: 337-345. 10.1158/1078-0432.CCR-10-1776.CrossRefPubMed Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV, Caplin M, Meyer T: Circulating Tumour Cells and EpCAM Expression in Neuroendocrine Tumours. Clin Cancer Res. 2011, 17: 337-345. 10.1158/1078-0432.CCR-10-1776.CrossRefPubMed
34.
Zurück zum Zitat Thurm H, Ebel S, Kentenich C, Hemsen A, Riethdorf S, Coith C, Wallwiener D, Braun S, Oberhoff C, Jänicke F, Pantel K: Rare expression of epithelial cell adhesion molecule on residual micrometastatic breast cancer cells after adjuvant chemotherapy. Clin Cancer Res. 2003, 9: 2598-2604.PubMed Thurm H, Ebel S, Kentenich C, Hemsen A, Riethdorf S, Coith C, Wallwiener D, Braun S, Oberhoff C, Jänicke F, Pantel K: Rare expression of epithelial cell adhesion molecule on residual micrometastatic breast cancer cells after adjuvant chemotherapy. Clin Cancer Res. 2003, 9: 2598-2604.PubMed
35.
Zurück zum Zitat Rao CG, Chianese D, Doyle GV, Miller MC, Russell T, Sanders RA, Terstappen LW: Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumours. Int J Oncol. 2005, 27: 49-57.PubMed Rao CG, Chianese D, Doyle GV, Miller MC, Russell T, Sanders RA, Terstappen LW: Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumours. Int J Oncol. 2005, 27: 49-57.PubMed
36.
Zurück zum Zitat Armstrong AJ, Marengo MS, Oltean S, Kemeny G, Bitting RL, Turnbull JD, Herold CI, Marcom PK, George DJ, Garcia-Blanco MA: Circulating tumour cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol Cancer Res. 2011, 9: 997-1007. 10.1158/1541-7786.MCR-10-0490.CrossRefPubMedPubMedCentral Armstrong AJ, Marengo MS, Oltean S, Kemeny G, Bitting RL, Turnbull JD, Herold CI, Marcom PK, George DJ, Garcia-Blanco MA: Circulating tumour cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol Cancer Res. 2011, 9: 997-1007. 10.1158/1541-7786.MCR-10-0490.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Punnoose EA, Atwal SK, Spoerke JM, Savage H, Pandita A, Yeh RF, Pirzkall A, Fine BM, Amler LC, Chen DS, Lackner MR: Molecular biomarker analyses using circulating tumour cells. PLoS One. 2010, 5: 1-11.CrossRef Punnoose EA, Atwal SK, Spoerke JM, Savage H, Pandita A, Yeh RF, Pirzkall A, Fine BM, Amler LC, Chen DS, Lackner MR: Molecular biomarker analyses using circulating tumour cells. PLoS One. 2010, 5: 1-11.CrossRef
Metadaten
Titel
Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition
verfasst von
Tobias M Gorges
Ingeborg Tinhofer
Michael Drosch
Lars Röse
Thomas M Zollner
Thomas Krahn
Oliver von Ahsen
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2012
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-12-178

Weitere Artikel der Ausgabe 1/2012

BMC Cancer 1/2012 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.