Skip to main content
Erschienen in: BMC Cancer 1/2016

Open Access 01.12.2016 | Technical advance

A cell transportation solution that preserves live circulating tumor cells in patient blood samples

verfasst von: Steingrimur Stefansson, Daniel L. Adams, William B. Ershler, Huyen Le, David H. Ho

Erschienen in: BMC Cancer | Ausgabe 1/2016

Abstract

Background

Circulating tumor cells (CTCs) are typically collected into CellSave fixative tubes, which kills the cells, but preserves their morphology. Currently, the clinical utility of CTCs is mostly limited to their enumeration. More detailed investigation of CTC biology can be performed on live cells, but obtaining live CTCs is technically challenging, requiring blood collection into biocompatible solutions and rapid isolation which limits transportation options. To overcome the instability of CTCs, we formulated a sugar based cell transportation solution (SBTS) that stabilizes cell viability at ambient temperature. In this study we examined the long term viability of human cancer cell lines, primary cells and CTCs in human blood samples in the SBTS for transportation purposes.

Methods

Four cell lines, 5 primary human cells and purified human PBMCs were tested to determine the viability of cells stored in the transportation solution at ambient temperature for up to 7 days. We then demonstrated viability of MCF-7 cells spiked into normal blood with SBTS and stored for up to 7 days. A pilot study was then run on blood samples from 3 patients with metastatic malignancies stored with or without SBTS for 6 days. CTCs were then purified by Ficoll separation/microfilter isolation and identified using CTC markers. Cell viability was assessed using trypan blue or CellTracker™ live cell stain.

Results

Our results suggest that primary/immortalized cell lines stored in SBTS remain ~90 % viable for > 72 h. Further, MCF-7 cells spiked into whole blood remain viable when stored with SBTS for up to 7 days. Finally, live CTCs were isolated from cancer patient blood samples kept in SBTS at ambient temperature for 6 days. No CTCs were isolated from blood samples stored without SBTS.

Conclusions

In this proof of principle pilot study we show that viability of cell lines is preserved for days using SBTS. Further, this solution can be used to store patient derived blood samples for eventual isolation of viable CTCs after days of storage. Therefore, we suggest an effective and economical transportation of cancer patient blood samples containing live CTCs can be achieved.
Hinweise

Competing interests

SS and DHH are employees of HeMemics Biotechnologies. DLA is an employee of Creatv MicroTech, Inc. WBE and HL declare that they have no competing interests.

Authors’ contributions

SS performed experiments and wrote the paper. DLA performed experiments and conceptualized the data. WBE coordinated patient sample studies and the IRB at Inova Fairfax. HL and DHH oversaw the project. All authors have read the paper and approve its publication.
Abkürzungen
CK
Cytokeratin
CMAC
7-amino-4-chloromethylcoumarin
CTC
Circulating tumor cells
EpCAM
Epithelial cell adhesion molecule
FBS
Fetal bovine serum
SBTS
sugar based transportation solution

Background

The primary mechanism of metastatic spread begins with dissemination, or shedding, of cancerous epithelial cells from tumor sites into the circulation. These circulating tumor cells (CTCs) travel throughout the body, adhere to organ vascular beds, infiltrate the tissue, grow and impair organ function [13].
Animal studies have shown that organ colonization of injected tumor cells is very efficient, i.e. ~80 % of injected tumor cells extravasate into organs [47]. However, the majority of those extravasated cells do not form tumors, thus their metastatic potential through CTC dissemination is, in most cases, very inefficient. Despite ~106 tumor cells are being shed into the circulation per gram of tumor tissue every 24 h, less than 1 % of shed CTCs remain alive 24 h after dissemination [811]. This loss of CTC viability has been attributed to many factors including fragility, shear stresses in the circulation, anoikis and destruction by the immune system [1216]. Interestingly, studies imply that the injected cancer cells that form primary tumors have different biological properties than their progeny populating the metastatic tumors [17, 18].
Analyzing human CTCs is technically challenging because of their extreme rarity in cancer patient blood samples (commonly ≤ 10 CTCs among 109 total blood cells), their inherent heterogeneity and instability [16, 1921]. CellSearch® is the only FDA approved and clinically validated CTC assay that isolates CTCs, used as a prognostic indicator of survival for breast, prostate, and colorectal cancer patients. This system was developed based on affinity-based isolation procedures and biomarker presence [2225]. The CellSearch® CTC Test requires blood samples to be collected in CellSave tubes which contain a fixative solution that allows storage of blood samples for up to 3 days, but also kills the CTCs. The need for CTC fixation is necessitated because of their inherent fragility [1921, 26, 27], but besides killing the CTCs, fixation also crosslinks extra- and intracellular biomolecules that can compromise some molecular analysis [28, 29].
Examining live CTCs has the potential of advancing the knowledge of cancer metastasis by interrogating the underlying biological activity of cells which cannot be accomplished on dead cells, i.e. mRNA profiling, culturing, etc. [2834]. While many devices and techniques have been developed to capture live CTCs from patient blood samples, the blood sample is usually processed on-site and within hours of collection to retain viability [30, 3541]. Therefore, transportation of live CTCs in blood samples between research institutions is often not possible due to the high rate of CTC senescence in whole blood.
We first examined the ability of the SBTS to preserve viability in primary cells, cell lines and cancer cells spiked into blood cells at ambient temperature for storage/transportation purposes. We then extended our studies, examining the effect of the SBTS on patient blood samples with live CTCs. Our data suggest that cells can be stored for days without harming the viability of the cells and that CTCs in patient blood samples can also be stored for 6 days at room temperature and retain viability. While further studies on the potential use of the SBTS to preserve live CTCs in transporting blood samples is needed, we suggest that cells can be kept viable when stored long term in blood at ambient temperatures.

Methods

Antibodies and reagents

The sugar based cell transportation solution (SBTS) is a non-toxic proprietary mixture of high and low molecular weight carbohydrates (HemSol™). CellSieve™ CTC micro-filtration system from Creatv Microtech (Rockville, MD) using a low-pressure vacuum system which isolates CTCs based on size exclusion, > 7 micron, as previously described [4244]. Stains used on the CTCs were CellTracker™ Blue CMAC live cell stain (Thermo-Fisher). FITC labeled Pan-cytokeratin clone C11 (Sigma), which recognizes human cytokeratins 4, 5, 6, 8, 10, 13 and 18. Phycoerythrin (PE) labeled anti-human epithelial cell adhesion molecule (EpCAM), clone 1B7 (eBioscience). Alexa Fluor® 594 labeled anti-human CD45, clone 2D1 (Novus) and DRAQ5™ fluorescent DNA probe (Thermo-Fisher). Exposure times (and ex/em wavelengths of the Leica microscope filters), respectively, were: Blue CMAC: 35 msec, (350 nm/460 nm); FITC-CK, 500 msec (470 nm/525 nm); PE-EpCAM, 500 msec (546 nm/585 nm); Alexa Fluor594 500 msec (594 nm/645 nm); . DRAQ5 600 msec (640 nm/690 nm). Samples were analyzed using a Leica fluorescent microscope and imaged with a Leica camera and Leica Microsystems imaging software.

Cell lines, primary cells and blood samples

Chinese hamster ovary cells (CHO), human embryonic kidney cells (HEK 293), human umbilical vein endothelial cells (HUVEC) and human epithelial colorectal adenocarcinoma cells (CACO-2) human breast cancer epithelial cells (MCF-7), in addition to primary human cells were purchased from ATCC. Peripheral blood mononuclear cells (PBMCs) were isolated from whole blood samples obtained from healthy volunteers using Ficoll separation (GE Healthcare) as described by the manufacturer. Healthy volunteer blood samples were procured with signed informed consent and IRB approval by Western IRB. Patient blood samples were collected with signed informed consent and an IRB with Inova Fairfax Hospital.

Incubation of cells and cell lines with HemSol™ transportation solution

HemSol™ preservation experiments with primary human hepatocytes, B-Cells, kidney cells, mesenchymal stem cells and non-small cell (NSC) lung carcinoma were performed in collaborations with AscentGene Inc. (Gaithersburg, MD). Approximately 105–107 cells were kept in their respective growth media prior to treatment with HemSol™. Live cells were enumerated by trypan blue exclusion. Cells were then mixed with concentrated HemSol™ (2-6X) for a final concentration of 1X HemSol™. The cells were then stored in the solution for the indicated time at ambient temperature, after which the HemSol™ was washed away using the respective cell media and live cells determined using trypan blue exclusion and noted in Table 1.
Table 1
Viability of cells kept in SBTS for the indicated timea at ambient temperature. Cells (105-107), were mixed with SBTS and cell viability was assessed using trypan-blue exclusion after washing and resuspending cells in their respective serum containing mediab
Cell Type
Cell Condition
Time in SBTS (Days)a
% Live Cellsb
CHO
Suspension
4
95–98
CHO
Adherent
3
95–99
HEK 293
Adherent
3
92–98
HUVEC
Suspension
3
90–98
HUVEC
Adherent
3
93–96
CACO-2
Adherent
3
92–98
Primary hepatocytes
Adherent
7
85–92
PBMC
Suspension
3
> 95
B-Cells
Suspension
4
95–98
Primary kidney cells
Suspension
7
85–90
Mesenchymal stem cells
Suspension
9
92–98
NSC lung carcinoma
Suspension
3
80–92
Live cells were determined from at least 3 experiments

Analysis of MCF-7 cells spiked into normal whole blood with HemSol™ transportation solution

Human breast cancer cell line (MCF-7) was labeled with CellTracker™ Green (Invitrogen) according to manufacturer’s protocols and cells were then washed to remove remaining free dye. CellTracker™ Green is an intracellular stain that only labels live cells and is retained by cell progeny, allowing for multigenerational tracking. Approximately 10,000 of the labeled MCF-7 cells were added to 8 ml of normal whole blood. 4 ml of that blood sample was then mixed with HemSol™ and 4 ml was incubated without the HemSol™ at RT for 4 days. After the incubation, the blood was filtered and the filters put into tissue culture with DMEM, 10 % serum at 37 °C, 5 % CO2 and imaged after 3 days in culture. A second set of labeled MCF-7 cells were kept in whole blood for 7 days at RT followed by filtration using the CellSieve™ CTC micro-filtration system from Creatv Microtech, as previously described [4244] and imaged.

Analysis of cancer patient blood samples incubated with HemSol™ transportation solution

To determine the preservation of live CTCs in HemSol™, duplicate whole blood samples (7.5 ml) from 1 breast cancer patient, 1 pancreatic cancer patient and 1 lung cancer patient were collected into EDTA vacutainers and the contents of one tube immediately transferred to a 15 ml conical tube containing 6X concentrated HemSol™, which was diluted to 1X HemSol™ with the blood and mixed for complete dispersal. The duplicate blood sample was transferred to a conical tube containing PBS at the same volume as the concentrated HemSol™, and mixed similarly. The blood samples were kept for 6 days at RT without agitation and then fractionated by mixing the samples 1:1 with PBS and overlaying the samples onto Ficoll. The samples were centrifuged at 400 × g for 30 min at 18 °C in 50-mL centrifuge tubes according to manufacturer’s instructions. HemSol™ did not interfere with the density separation of red blood cells from the PBMC buffy coat layer. The collected buffy coat layer was pelleted by centrifugation and resuspended in 1 ml PBS with CellTracker™ Blue CMAC Dye cell live stain (Invitrogen) for 45 min at RT according to manufacturer’s instructions. After incubation, unbound dye was removed by centrifugation and resuspension of the cell pellet in 2 ml PBS followed by re-centrifugation. The resulting cell pellet was then resuspended in PBS containing 1 % paraformaldehyde (PFA), 5 mM EDTA and incubated for 20 min at RT. After fixation, the cells were filtered using CellSieve™ micro-filtration system (Creatv Microtech) [4245] set at a flow rate of 5 ml/min. The captured cells were then permeabilized with PBS containing 0.4 % Triton X-100 in PBS, washed twice with 4 ml PBS and stained as previouly described [4245].

Results and discussion

Analyzing physical parameters of cancer patient blood samples are routine procedures performed in clinical labs around the world. A recent addition to the arsenal of clinical blood testing procedures in oncology practices is the enumeration of circulating tumor cells (CTCs). These are cancerous cells of epithelial origin that are shed from solid tumors and found in the circulation of many cancer patients which can be used as indicators of patient survival. However, CTCs are fragile cells that die and disintegrate rapidly in whole blood samples [16, 1921]. Therefore, enumeration of CTCs requires patient blood collection into specialized tubes containing fixatives that preserve CTC morphology, but sacrifice viability. The blood samples collected into fixatives keep cell integrity for at least 3 days, allowing them to be transported to other labs for enumeration and further analysis. However, many biological analyses are compromised as cells are dead and imbued with fixative [28, 29]. Keeping CTCs alive in whole blood samples long enough for timely and economical transportation is a challenge as whole blood is an exceptionally harsh media for non-hematopoietic cells.
The SBTS is a proprietary mixture of simple and complex carbohydrates that was initially formulated to stabilize human red blood cell (RBC) structure and antigen properties upon desiccation for use in reference labs [46]. Oxygen binding properties of desiccated RBCs and platelet structure and function stored at 4 °C for up to 2 weeks are also protected in the presence of these carbohydrates [47, 48]. Those results prompted us to examine whether the SBTS could also stabilize the structure of live nucleated cells. Table 1 shows stabilization of live cell kept in SBTS for 3–9 days at RT.
These data show that immortalized, and primary cells both adherent and in suspension are kept live in SBTS for at least 3 days at ambient temperature. Of special notice is that anchorage dependent cells, including human umbilical vein endothelial cells (HUVEC), human primary kidney cells, Chinese hamster ovary cells (CHO) and human non-small cell (NSC) lung carcinoma cells remain viable in suspension for at least 3 days in SBTS. The positive results of these studies prompted us to examine whether SBTS can preserve cells in other inhospitable media, such as whole blood. Many CTCs are of epithelial origin, which means that they are generally anchorage dependent and undergo apoptosis when in suspension.
Before analyzing CTCs from patient blood samples, we first performed experiments with live MCF-7 epithelial breast cancer cells, which are also anchorage dependent, spiked into whole blood as described [42]. CellTracker™ Green pre-labeled MCF-7 cells were spiked into 4 ml of normal whole blood with or without SBTS. This CellTracker™ dye in a non-fluorescent cell permeable esterase substrate that becomes fluorescent after cleavage by intracellular esterases. The cleavage traps the fluorescent dye inside the cells and it is retained even after cell divisions, however the dye is lost when the integrity of the cell is compromised.
After 4 days in whole blood at RT, the MCF-7 cells were isolated using CellSieve™ microfilters and the filters were put into cell culture media and allowed to grow for 3 days. Figure 1a shows that MCF-7 cells spiked into whole peripheral blood treated with SBTS were able to proliferate and formed colonies post-isolation. In contrast, MCF-7 cells isolated from whole blood in the absence of SBTS had far fewer viable cells and none of the cells proliferated, even after 7 days in culture (Fig. 1b). Interestingly, in the absence of SBTS, live MCF-7 cells isolated from blood did not proliferate.
Additionally, live MCF-7 cells were further kept in normal whole blood for 7 days at RT followed by filtration and imaging. Figure 2a shows live MCF-7 cells in the SBTS treated blood sample with very few live MCF-7 cells observed in whole blood in the absence of SBTS (Fig. 2b).
We then proceeded to analyze whether the SBTS was able to keep CTCs alive in cancer patient blood samples. Live CTCs are fragile cells that generally need to be isolated rapidly from blood samples [1921, 26, 27]. Therefore, extending the life of CTCs in patient blood samples for days will increase the availability of these rare cells, for use in real-time downstream analysis using next generation sequencing and/or proteomic analysis. Three cancer patient blood samples (breast, prostate and lung) were divided and incubated with or without SBTS for 6 days at RT. Blood samples were separated using ficoll, which has been previously described in isolating viable CTC from blood samples [41, 4952]. The buffy coat layer was removed and incubated with CellTracker™ Blue CMAC cell viability stain as recommended by the manufacturer, then cells were centrifuged to remove free dye. Blue CMAC is a membrane permeable stain that is well retained by live cells and is fixable by formaldehyde. The cells were then filtered CellSieve™ microfilters, fixed and stained with the standard CTC marker antibodies (see Methods).
The prostate and lung cancer samples had no live CTCs as detected by our isolation procedure, whereas the breast cancer sample had 4 detectable live CTCs (CMAC Blue+, CK+, EpCAM+, CD45-) after 6 days in SBTS. The lack of CTCs isolated from the other patient samples can be attributed to a number of factors, including the fact that some cancer patients do not have detectable CTCs in their blood samples and that the ficoll separation step can lead to losses of CTCs [5356].
Figure 3, panels a and b show 2 live CTCs isolated from a breast cancer patient sample treated with SBTS and stored for 6 days at room temperature. The cells are alive and intact since Blue CMAC stain is retained within the cells. Cells are also positive for cytokeratin (green) and EpCAM (yellow), negative for CD45 and have an intact nucleus (red).
Duplicate patient blood samples treated with buffer and stored for 6 days at room temperature were also filtered and stained. Neither live nor dead CK+, EpCAM+ cells were found on the filters of blood samples stored without SBTS. The other 2 CTCs isolated from the breast cancer patient sample retained the Blue CMAC viability stain, but showed cell damage (not shown).
The data presented demonstrates that the SBTS preserves cell viability, even in harsh medium such as whole blood, which is detrimental to CTCs survival. Although the CTCs in the breast cancer patient sample were viable, we did not attempt their propagation in cell culture because culture conditions that allows for reproducible CTC growth without altering their phenotype has yet to be described. However, these initial experiments which show viability of CTCs suggest that when culture techniques for CTCs are developed, the SBTS is well suited for long term storage and transport of patient derived blood samples.

Conclusions

A number of technologies have been developed to isolate live CTCs. However, since CTCs are rare and unstable in whole blood, isolation procedures must be performed rapidly after blood collection [1921, 26, 27]. A six day time point for blood storage SBTS was chosen based on our results with cell culture (Table 1) and live MCF-7 cells spiked into normal whole blood (Figs. 1 and 2). Additionally, the 6 day period was chosen to represent an extreme case for transcontinental transportation of blood samples. The benefits of having a simple, low cost and non-toxic treatment of patient blood samples that preserves live CTC in blood samples for up to 6 days at ambient temperature will greatly enhance the ability to analyze CTCs, because clinicians and researchers will have more time to process these samples or ship them between research facilities. M. Ignatiadis et al. [57] succinctly wrote in their paper about this problem: “However, an important limitation of our study is that all patients were recruited in one center and the CTC analysis was done in one laboratory. An international, prospective, multicenter trial with different participating laboratories in which issues of stability during shipment of the samples, interlaboratory reproducibility of the multimarker assay, and validation of our results in a diverse patient population is urgently needed.”

Acknowledgements

The authors would like to thank the patients at Inova Fairfax that donated blood samples for this study, Jenille Giaimo at Inova Fairfax for collecting patient samples and Dr. Cha-Mei Tang at Creatv Microtech Inc. (Rockville MD) for support with CellSieve™ CTC micro-filtration system. Non-support was provided by the NIH.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

SS and DHH are employees of HeMemics Biotechnologies. DLA is an employee of Creatv MicroTech, Inc. WBE and HL declare that they have no competing interests.

Authors’ contributions

SS performed experiments and wrote the paper. DLA performed experiments and conceptualized the data. WBE coordinated patient sample studies and the IRB at Inova Fairfax. HL and DHH oversaw the project. All authors have read the paper and approve its publication.
Literatur
1.
Zurück zum Zitat Paterlini-Brechot P, Naoual LB. Circulating tumor cells (CTC) detection: clinical impact and future directions. Cancer Lett. 2007;253(2):180–204.CrossRefPubMed Paterlini-Brechot P, Naoual LB. Circulating tumor cells (CTC) detection: clinical impact and future directions. Cancer Lett. 2007;253(2):180–204.CrossRefPubMed
2.
Zurück zum Zitat Hou JM, Krebs MG, Lancashire L, Sloane R, Backen A, Swain RK, et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. 2012;30(5):525–32.CrossRefPubMed Hou JM, Krebs MG, Lancashire L, Sloane R, Backen A, Swain RK, et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. 2012;30(5):525–32.CrossRefPubMed
3.
Zurück zum Zitat Ring A, Smith IE, Dowsett M. Circulating tumor cells in breast cancer. Lancet. 2004;5:79–88.CrossRefPubMed Ring A, Smith IE, Dowsett M. Circulating tumor cells in breast cancer. Lancet. 2004;5:79–88.CrossRefPubMed
4.
Zurück zum Zitat Koop S, MacDonald IC, Luzzi K, Schmidt EE, Morris VL, Grattan M, et al. Fate of melanoma cells entering the microcirculation: over 80 % survive and extravasate. Cancer Res. 1995;55:2520–3.PubMed Koop S, MacDonald IC, Luzzi K, Schmidt EE, Morris VL, Grattan M, et al. Fate of melanoma cells entering the microcirculation: over 80 % survive and extravasate. Cancer Res. 1995;55:2520–3.PubMed
5.
Zurück zum Zitat Cameron MD, Schmidt EE, Kerkvliet N, Nadkarni KV, Morris VL, Groom AC, et al. Temporal progression of metastasis in lung: cell survival, dormancy, and location dependence of metastatic inefficiency. Cancer Res. 2000;60(9):2541–6.PubMed Cameron MD, Schmidt EE, Kerkvliet N, Nadkarni KV, Morris VL, Groom AC, et al. Temporal progression of metastasis in lung: cell survival, dormancy, and location dependence of metastatic inefficiency. Cancer Res. 2000;60(9):2541–6.PubMed
6.
Zurück zum Zitat Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Chambers AF, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153(3):865–73.CrossRefPubMedPubMedCentral Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Chambers AF, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153(3):865–73.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Naumov GN, MacDonald IC, Weinmeister PM, Kerkvliet N, Nadkarni KV, Wilson SM, Chambers AF. Persistence of solitary mammary carcinoma cells in a secondary site a possible contributor to dormancy. Cancer Res. 2002;62(7):2162–8.PubMed Naumov GN, MacDonald IC, Weinmeister PM, Kerkvliet N, Nadkarni KV, Wilson SM, Chambers AF. Persistence of solitary mammary carcinoma cells in a secondary site a possible contributor to dormancy. Cancer Res. 2002;62(7):2162–8.PubMed
8.
Zurück zum Zitat Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2-deoxyuridine. J Natl Cancer Inst. 1970;45:773–82.PubMed Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2-deoxyuridine. J Natl Cancer Inst. 1970;45:773–82.PubMed
9.
Zurück zum Zitat Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res. 1975;35(3):512–6.PubMed Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res. 1975;35(3):512–6.PubMed
10.
Zurück zum Zitat 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. 2000;97(26):14608–13.CrossRefPubMedPubMedCentral 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. 2000;97(26):14608–13.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Juacaba SF, Horak E, Price JE, Tarin D. Tumor cell dissemination patterns and metastasis of murine mammary carcinoma. Cancer Res. 1989;49(3):570–5.PubMed Juacaba SF, Horak E, Price JE, Tarin D. Tumor cell dissemination patterns and metastasis of murine mammary carcinoma. Cancer Res. 1989;49(3):570–5.PubMed
12.
Zurück zum Zitat Berezovskaya O, Schimmer AD, Glinskii AB, Pinilla C, Hoffman RM, Reed JC, et al. Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res. 2005;65(6):2378–86.CrossRefPubMed Berezovskaya O, Schimmer AD, Glinskii AB, Pinilla C, Hoffman RM, Reed JC, et al. Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res. 2005;65(6):2378–86.CrossRefPubMed
13.
Zurück zum Zitat Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235–9.CrossRefPubMedPubMedCentral Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235–9.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Hanna NA, Burton RC. Definitive evidence that natural killer (NK) cells inhibit experimental tumor metastases in vivo. J Immunol. 1981;127(5):1754–8.PubMed Hanna NA, Burton RC. Definitive evidence that natural killer (NK) cells inhibit experimental tumor metastases in vivo. J Immunol. 1981;127(5):1754–8.PubMed
15.
Zurück zum Zitat Rossi E, Basso U, Celadin R, Zilio F, Pucciarelli S, Aieta, et al. M30 neoepitope expression in epithelial cancer: quantification of apoptosis in circulating tumor cells by Cell Search analysis. Clin Cancer Res. 2010;16(21):5233–43.CrossRefPubMed Rossi E, Basso U, Celadin R, Zilio F, Pucciarelli S, Aieta, et al. M30 neoepitope expression in epithelial cancer: quantification of apoptosis in circulating tumor cells by Cell Search analysis. Clin Cancer Res. 2010;16(21):5233–43.CrossRefPubMed
16.
Zurück zum Zitat Weiss L, Harlos JP, Elkin G. Mechanism of mechanical trauma to Ehrlich ascites tumor cells in vitro and its relationship to rapid intravascular death during metastasis. Int J Cancer. 1989;44:143–8.CrossRefPubMed Weiss L, Harlos JP, Elkin G. Mechanism of mechanical trauma to Ehrlich ascites tumor cells in vitro and its relationship to rapid intravascular death during metastasis. Int J Cancer. 1989;44:143–8.CrossRefPubMed
17.
Zurück zum Zitat Swartz MA, Kristensen CA, Melder RJ, Roberge S, Calautti E, Fukumura D, et al. Cells shed from tumours show reduced clonogenicity, resistance to apoptosis, and in vivo tumorigenicity. Br J Cancer. 1999;81(5):756.CrossRefPubMedPubMedCentral Swartz MA, Kristensen CA, Melder RJ, Roberge S, Calautti E, Fukumura D, et al. Cells shed from tumours show reduced clonogenicity, resistance to apoptosis, and in vivo tumorigenicity. Br J Cancer. 1999;81(5):756.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Goodison S, Kawai K, Hihara J, Jiang P, Yang M, Urquidi V, et al. Prolonged dormancy and site-specific growth potential of cancer cells spontaneously disseminated from nonmetastatic breast tumors as revealed by labeling with green fluorescent protein. Clin Cancer Res. 2003;9(10):3808–14.PubMed Goodison S, Kawai K, Hihara J, Jiang P, Yang M, Urquidi V, et al. Prolonged dormancy and site-specific growth potential of cancer cells spontaneously disseminated from nonmetastatic breast tumors as revealed by labeling with green fluorescent protein. Clin Cancer Res. 2003;9(10):3808–14.PubMed
19.
Zurück zum Zitat Hofman V, Ilie M, Long E, Guibert N, Selva E, Washetine K, et al. Detection of circulating tumor cells from lung cancer patients in the era of targeted therapy: promises, drawbacks and pitfalls. Curr Mol Med. 2014;14(4):440–56.CrossRefPubMed Hofman V, Ilie M, Long E, Guibert N, Selva E, Washetine K, et al. Detection of circulating tumor cells from lung cancer patients in the era of targeted therapy: promises, drawbacks and pitfalls. Curr Mol Med. 2014;14(4):440–56.CrossRefPubMed
20.
Zurück zum Zitat Larson CJ, Moreno JG, Pienta KJ, Gross S, Repollet M, O’Hara SM, et al. Apoptosis of circulating tumor cells in prostate cancer patients. Cytometry A. 2004;62(1):46–53.CrossRefPubMed Larson CJ, Moreno JG, Pienta KJ, Gross S, Repollet M, O’Hara SM, et al. Apoptosis of circulating tumor cells in prostate cancer patients. Cytometry A. 2004;62(1):46–53.CrossRefPubMed
21.
Zurück zum Zitat Qin J, Alt JR, Hunsley BA, Williams TL, Fernando MR. Stabilization of circulating tumor cells in blood using a collection device with a preservative reagent. Cancer Cell Int. 2014;14(1):1–6.CrossRef Qin J, Alt JR, Hunsley BA, Williams TL, Fernando MR. Stabilization of circulating tumor cells in blood using a collection device with a preservative reagent. Cancer Cell Int. 2014;14(1):1–6.CrossRef
22.
Zurück zum Zitat Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897–904.CrossRefPubMed Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897–904.CrossRefPubMed
23.
Zurück zum Zitat Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–91.CrossRefPubMed Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–91.CrossRefPubMed
24.
Zurück zum Zitat de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302–9.CrossRefPubMed de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302–9.CrossRefPubMed
25.
Zurück zum Zitat Cohen SJ, Alpaugh RK, Gross S, O’Hara SM, Smirnov DA, Terstappen LW, et al. Isolation and characterization of circulating tumor cells in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2006;6:125–32.CrossRefPubMed Cohen SJ, Alpaugh RK, Gross S, O’Hara SM, Smirnov DA, Terstappen LW, et al. Isolation and characterization of circulating tumor cells in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2006;6:125–32.CrossRefPubMed
26.
Zurück zum Zitat Sheth RA, Hesketh R, Deipolyi AR, Oklu R. Circulating tumor cells: personalized medicine in interventional oncology? J Vasc Interv Radiol. 2013;24(2):221–8.CrossRefPubMed Sheth RA, Hesketh R, Deipolyi AR, Oklu R. Circulating tumor cells: personalized medicine in interventional oncology? J Vasc Interv Radiol. 2013;24(2):221–8.CrossRefPubMed
27.
Zurück zum Zitat Flores LM, Kindelberger DW, Ligon AH, Capelletti M, Fiorentino M, Loda M, et al. Improving the yield of circulating tumour cells facilitates molecular characterisation and recognition of discordant HER2 amplification in breast cancer. Br J Cancer. 2010;102(10):1495–502.CrossRefPubMedPubMedCentral Flores LM, Kindelberger DW, Ligon AH, Capelletti M, Fiorentino M, Loda M, et al. Improving the yield of circulating tumour cells facilitates molecular characterisation and recognition of discordant HER2 amplification in breast cancer. Br J Cancer. 2010;102(10):1495–502.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Swennenhuis JF, Reumers J, Thys K, Aerssens J, Terstappen LW. Efficiency of whole genome amplification of Single Circulating Tumor Cells enriched by Cell Search and sorted by FACS. Genome Med. 2013;5:106.CrossRefPubMedPubMedCentral Swennenhuis JF, Reumers J, Thys K, Aerssens J, Terstappen LW. Efficiency of whole genome amplification of Single Circulating Tumor Cells enriched by Cell Search and sorted by FACS. Genome Med. 2013;5:106.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Adalsteinsson VA, Love JC. Toward engineered processes for sequencing-based analysis of single circulating tumor cells. Curr Opin Chem Eng. 2014;4:97–104.CrossRefPubMedPubMedCentral Adalsteinsson VA, Love JC. Toward engineered processes for sequencing-based analysis of single circulating tumor cells. Curr Opin Chem Eng. 2014;4:97–104.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Powell AA, Talasaz AH, Zhang H, Coram MA, Reddy A, Deng G, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One. 2012;7(5):e33788.CrossRefPubMedPubMedCentral Powell AA, Talasaz AH, Zhang H, Coram MA, Reddy A, Deng G, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One. 2012;7(5):e33788.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Chen CL, Mahalingam D, Osmulski P, Jadhav RR, Wang CM, Leach RJ, et al. Single‐cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT‐related genes in metastatic prostate cancer. Prostate. 2013;73(8):813–26.CrossRefPubMed Chen CL, Mahalingam D, Osmulski P, Jadhav RR, Wang CM, Leach RJ, et al. Single‐cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT‐related genes in metastatic prostate cancer. Prostate. 2013;73(8):813–26.CrossRefPubMed
32.
Zurück zum Zitat Court CM, Ankeny JS, Hou S, Tseng HR, Tomlinson JS. Improving pancreatic cancer diagnosis using circulating tumor cells: prospects for staging and single-cell analysis. Expert review of molecular diagnostics. 2015;15(11):1491–1504. Court CM, Ankeny JS, Hou S, Tseng HR, Tomlinson JS. Improving pancreatic cancer diagnosis using circulating tumor cells: prospects for staging and single-cell analysis. Expert review of molecular diagnostics. 2015;15(11):1491–1504.
33.
Zurück zum Zitat Fabbri F, Carloni S, Zoli W, Ulivi P, Gallerani G, Fici P, et al. Detection and recovery of circulating colon cancer cells using a dielectrophoresis-based device: KRAS mutation status in pure CTCs. Cancer Lett. 2013;335(1):225–31.CrossRefPubMed Fabbri F, Carloni S, Zoli W, Ulivi P, Gallerani G, Fici P, et al. Detection and recovery of circulating colon cancer cells using a dielectrophoresis-based device: KRAS mutation status in pure CTCs. Cancer Lett. 2013;335(1):225–31.CrossRefPubMed
34.
Zurück zum Zitat Zhang C, Zhang C, Chen S, Yin X, Pan X, Lin G, et al. A single cell level based method for copy number variation analysis by low coverage massively parallel sequencing. PLoS One. 2013;8(1):e54236.CrossRefPubMedPubMedCentral Zhang C, Zhang C, Chen S, Yin X, Pan X, Lin G, et al. A single cell level based method for copy number variation analysis by low coverage massively parallel sequencing. PLoS One. 2013;8(1):e54236.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Sheng W, Ogunwobi OO, Chen T, Zhang J, George TJ, Liu C, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip. 2014;14(1):89–98.CrossRefPubMed Sheng W, Ogunwobi OO, Chen T, Zhang J, George TJ, Liu C, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip. 2014;14(1):89–98.CrossRefPubMed
36.
Zurück zum Zitat 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. 2012;12(12):2175–81.CrossRefPubMed 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. 2012;12(12):2175–81.CrossRefPubMed
37.
Zurück zum Zitat Coumans FA, van Dalum G, Beck M, Terstappen LW. Filter characteristics influencing circulating tumor cell enrichment from whole blood. PLoS One. 2013;8(4):e61770.CrossRefPubMedPubMedCentral Coumans FA, van Dalum G, Beck M, Terstappen LW. Filter characteristics influencing circulating tumor cell enrichment from whole blood. PLoS One. 2013;8(4):e61770.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Payne RE, Wang F, Su N, Krell J, Zebrowski A, Yagüe E, et al. Viable circulating tumour cell detection using multiplex RNA in situ hybridisation predicts progression-free survival in metastatic breast cancer patients. Br J Cancer. 2012;106(11):1790–7.CrossRefPubMedPubMedCentral Payne RE, Wang F, Su N, Krell J, Zebrowski A, Yagüe E, et al. Viable circulating tumour cell detection using multiplex RNA in situ hybridisation predicts progression-free survival in metastatic breast cancer patients. Br J Cancer. 2012;106(11):1790–7.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Reyes EE, VanderWeele DJ, Isikbay M, Duggan R, Campanile A, Stadler WM, et al. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J Transl Med. 2014;12(1):1–5.CrossRef Reyes EE, VanderWeele DJ, Isikbay M, Duggan R, Campanile A, Stadler WM, et al. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J Transl Med. 2014;12(1):1–5.CrossRef
40.
Zurück zum Zitat Xu MJ, Cooke M, Steinmetz D, Karakousis G, Saxena D, Bartlett E, et al. A novel approach for the detection and genetic analysis of live melanoma circulating tumor cells. PLoS One. 2015;10(3):e0123376.CrossRefPubMedPubMedCentral Xu MJ, Cooke M, Steinmetz D, Karakousis G, Saxena D, Bartlett E, et al. A novel approach for the detection and genetic analysis of live melanoma circulating tumor cells. PLoS One. 2015;10(3):e0123376.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Shao C, Liao CP, Hu P, Chu CY, Zhang L, Bui MH, Ng CS. Detection of live circulating tumor cells by a class of near-infrared heptamethine carbocyanine dyes in patients with localized and metastatic prostate cancer. PLoS One. 2014;9:e88967.CrossRefPubMedPubMedCentral Shao C, Liao CP, Hu P, Chu CY, Zhang L, Bui MH, Ng CS. Detection of live circulating tumor cells by a class of near-infrared heptamethine carbocyanine dyes in patients with localized and metastatic prostate cancer. PLoS One. 2014;9:e88967.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Adams DL, Zhu P, Makarova OV, Martin SS, Charpentier M, Chumsri S, et al. The systematic study of circulating tumor cell isolation using lithographic microfilters. RSC Adv. 2014;4(9):4334–42.CrossRef Adams DL, Zhu P, Makarova OV, Martin SS, Charpentier M, Chumsri S, et al. The systematic study of circulating tumor cell isolation using lithographic microfilters. RSC Adv. 2014;4(9):4334–42.CrossRef
43.
Zurück zum Zitat Adams DL, Stefansson S, Haudenschild C, Martin SS, Charpentier M, Chumsri S, et al. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the cellsearch® CTC test. Cytometry A. 2015;87(2):137–44.CrossRefPubMed Adams DL, Stefansson S, Haudenschild C, Martin SS, Charpentier M, Chumsri S, et al. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the cellsearch® CTC test. Cytometry A. 2015;87(2):137–44.CrossRefPubMed
44.
Zurück zum Zitat Adams DL, Martin SS, Alpaugh RK, Charpentier M, Tsai S, Bergan RC, et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc Natl Acad Sci. 2014;111(9):3514–9.CrossRefPubMedPubMedCentral Adams DL, Martin SS, Alpaugh RK, Charpentier M, Tsai S, Bergan RC, et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc Natl Acad Sci. 2014;111(9):3514–9.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Adams DL, Alpaugh RK, Martin SS, Charpentier M, Chumsri S, Cristofanilli M, et al. Precision Microfilters as an all in one System for Multiplex Analysis of Circulating Tumor Cells. RSC Adv., 2016, Accepted Manuscript DOI: 10.1039/C5RA21524B. Adams DL, Alpaugh RK, Martin SS, Charpentier M, Chumsri S, Cristofanilli M, et al. Precision Microfilters as an all in one System for Multiplex Analysis of Circulating Tumor Cells. RSC Adv., 2016, Accepted Manuscript DOI: 10.​1039/​C5RA21524B.
46.
Zurück zum Zitat Ho D, Schierts J, Zimmerman Z, Gadsden I, Bruttig S. Comparison of frozen versus desiccated reference human red blood cells for hemagglutination assays. Transfusion. 2009;49(10):2173–80.CrossRefPubMed Ho D, Schierts J, Zimmerman Z, Gadsden I, Bruttig S. Comparison of frozen versus desiccated reference human red blood cells for hemagglutination assays. Transfusion. 2009;49(10):2173–80.CrossRefPubMed
47.
Zurück zum Zitat Stefansson S, Chung DS, Yoo WS, Park YW, Kim G, et al. Improving oxygen binding of desiccated human red blood cells. Adv Biosci Biotechnol. 2016;7(02):47.CrossRef Stefansson S, Chung DS, Yoo WS, Park YW, Kim G, et al. Improving oxygen binding of desiccated human red blood cells. Adv Biosci Biotechnol. 2016;7(02):47.CrossRef
48.
Zurück zum Zitat Ho D, Stefanson S, Ershler WD. Development of HemSol-P ™, a novel platelet storage solution permitting storage at 4 °C. Blood. 2013;122(21):3661. Ho D, Stefanson S, Ershler WD. Development of HemSol-P ™, a novel platelet storage solution permitting storage at 4 °C. Blood. 2013;122(21):3661.
49.
Zurück zum Zitat Xu T, Lu B, Tai YC, Goldkorn A. A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter. Cancer Res. 2010;70(16):6420–6.CrossRefPubMedPubMedCentral Xu T, Lu B, Tai YC, Goldkorn A. A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter. Cancer Res. 2010;70(16):6420–6.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Hughes AD, Mattison J, Western LT, Powderly JD, Greene BT, King MR. Microtube device for selectin-mediated capture of viable circulating tumor cells from blood. Clin Chem. 2012;58(5):846–53.CrossRefPubMed Hughes AD, Mattison J, Western LT, Powderly JD, Greene BT, King MR. Microtube device for selectin-mediated capture of viable circulating tumor cells from blood. Clin Chem. 2012;58(5):846–53.CrossRefPubMed
52.
Zurück zum Zitat Nastały P, Ruf C, Becker P, Bednarz-Knoll N, Stoupiec M, Kavsur R, Isbarn H, Matthies C, Wagner W, Höppner D, Fisch M. Circulating tumor cells in patients with testicular germ cell tumors. Clin Cancer Res. 2014;20(14):3830–41.CrossRefPubMed Nastały P, Ruf C, Becker P, Bednarz-Knoll N, Stoupiec M, Kavsur R, Isbarn H, Matthies C, Wagner W, Höppner D, Fisch M. Circulating tumor cells in patients with testicular germ cell tumors. Clin Cancer Res. 2014;20(14):3830–41.CrossRefPubMed
53.
Zurück zum Zitat Rosenberg R, Gertler R, Friederichs J, Fuehrer K, Dahm M, Phelps R, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry. 2002;49(4):150–8.CrossRefPubMed Rosenberg R, Gertler R, Friederichs J, Fuehrer K, Dahm M, Phelps R, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry. 2002;49(4):150–8.CrossRefPubMed
54.
Zurück zum Zitat Toss A, Mu Z, Fernandez S, Cristofanilli M. CTC enumeration and characterization: moving toward personalized medicine. Annals of translational medicine. 2014;2(11). Toss A, Mu Z, Fernandez S, Cristofanilli M. CTC enumeration and characterization: moving toward personalized medicine. Annals of translational medicine. 2014;2(11).
55.
Zurück zum Zitat Woelfle U, Breit E, Zafrakas K, Otte M, Schubert F, Müller V, et al. Bi-specific immunomagnetic enrichment of micrometastatic tumour cell clusters from bone marrow of cancer patients. J Immunol Methods. 2005;300(1):136–45.CrossRefPubMed Woelfle U, Breit E, Zafrakas K, Otte M, Schubert F, Müller V, et al. Bi-specific immunomagnetic enrichment of micrometastatic tumour cell clusters from bone marrow of cancer patients. J Immunol Methods. 2005;300(1):136–45.CrossRefPubMed
56.
Zurück zum Zitat Baker MK, Mikhitarian K, Osta W, Callahan K, Hoda R, Brescia F, et al. Molecular detection of breast cancer cells in the peripheral blood of advanced-stage breast cancer patients using multimarker real-time reverse transcription-polymerase chain reaction and a novel porous barrier density gradient centrifugation technology. Clin Cancer Res. 2003;9(13):4865–71.PubMed Baker MK, Mikhitarian K, Osta W, Callahan K, Hoda R, Brescia F, et al. Molecular detection of breast cancer cells in the peripheral blood of advanced-stage breast cancer patients using multimarker real-time reverse transcription-polymerase chain reaction and a novel porous barrier density gradient centrifugation technology. Clin Cancer Res. 2003;9(13):4865–71.PubMed
57.
Zurück zum Zitat Ignatiadis M, Kallergi G, Ntoulia M, Perraki M, Apostolaki S, Kafousi M, et al. Prognostic value of the molecular detection of circulating tumor cells using a multimarker reverse transcription-PCR assay for cytokeratin 19, mammaglobin A, and HER2 in early breast cancer. Clin Cancer Res. 2008;14(9):2593–600.CrossRefPubMed Ignatiadis M, Kallergi G, Ntoulia M, Perraki M, Apostolaki S, Kafousi M, et al. Prognostic value of the molecular detection of circulating tumor cells using a multimarker reverse transcription-PCR assay for cytokeratin 19, mammaglobin A, and HER2 in early breast cancer. Clin Cancer Res. 2008;14(9):2593–600.CrossRefPubMed
Metadaten
Titel
A cell transportation solution that preserves live circulating tumor cells in patient blood samples
verfasst von
Steingrimur Stefansson
Daniel L. Adams
William B. Ershler
Huyen Le
David H. Ho
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2016
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2330-1

Weitere Artikel der Ausgabe 1/2016

BMC Cancer 1/2016 Zur Ausgabe

Update Onkologie

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