Cancer panel analysis of circulating tumor cells in patients with breast cancer

  • Authors:
    • Cham Han Lee
    • Soo Jeong Lee
    • Sung Ho Choi
    • Sei Hyun Ahn
    • Byung Ho Son
    • Jong Won Lee
    • Jong Han Yu
    • Nak‑Jung Kwon
    • Woo Chung Lee
    • Kap‑Seok Yang
    • Dong Hyoung Lee
    • Du Yeol Han
    • Mi So Choi
    • Pyeong‑Soo Park
    • Hyun Kyung Lee
    • Myoung Shin Kim
    • Jinseon Lee
    • Byung Hee Jeon
  • View Affiliations

  • Published online on: May 7, 2018     https://doi.org/10.3892/ol.2018.8646
  • Pages: 612-618
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liquid biopsy using circulating tumor cells (CTCs) is a noninvasive and repeatable procedure, and is therefore useful for molecular assays. However, the rarity of CTCs remains a challenge. To overcome this issue, our group developed a novel technology for the isolation of CTCs on the basis of cell size difference. The present study isolated CTCs from patients with breast cancer using this method, and then used these cells for cancer gene panel analysis. Blood samples from eight patients with breast cancer were collected, and CTCs were enriched using size‑based filtration. Enriched CTCs were counted using immunofluorescent staining with an epithelial cell adhesion molecule (EpCAM) and CD45 antibodies. CTC genomic DNA was extracted, amplified, and screened for mutations in 400 genes using the Ion AmpliSeq Comprehensive Cancer Panel. White blood cells (WBCs) from the same patient served as a negative control, and mutations in CTCs and WBCs were compared. EpCAM+ cells were detected in seven out of eight patients, and the average number of EpCAM+ cells was 8.6. The average amount of amplified DNA was 32.7 µg, and the percentage of reads mapped to any targeted region relative to all reads mapped to the reference was 98.6%. The detection rate of CTC‑specific mutations was 62.5%. The CTC‑specific mutations were enhancer of zeste polycomb repressive complex 2 subunit, notch 1, AT‑rich interaction domain 1A, serine/threonine kinase 11, fms related tyrosine kinase 3, MYCN proto‑oncogene, bHLH transcription factor, APC, WNT signaling pathway regulator, and phosphatase and tensin homolog. The technique used by the present study was demonstrated to be effective at isolating CTCs at a sufficiently high purity for genomic analysis, and supported the use of comprehensive cancer panel analysis as a potential application for precision medicine.

Introduction

Breast cancer is the most common cancer in women, with the fifth highest mortality rate among all types of cancer and the highest occurrence rate among female cancers globally (1). Furthermore, the mortality rate of breast cancer in patients aged 25–45 years in Korea is the highest worldwide (2). Despite the fact that it is possible to treat hormone receptor-positive breast cancer with a wide variety of effective regimens in the early stages of disease and obtain relatively improved survival rates, a significant number of patients may experience tumor recurrence and metastasis (3).

The genomic characteristics of a metastatic tumor are different from those of a primary tumor, owing to the time interval between recurrence and metastasis, and the occurrence of the primary tumor. Furthermore, this genomic difference is intensified after treatment, including chemotherapy (4).

Trastuzumab, a targeted therapeutic agent, markedly improves progression-free and overall survival rates in patients with metastatic breast cancer, who have a poor prognosis, in the short term. However, long-term observation over 30 months demonstrated similar recurrence and mortality rates in patients treated with general chemotherapy and targeted therapy (5). Previous reports provided several hypotheses to explain the resistance to trastuzumab caused by genomic changes in tumor cells during treatment (6), and other therapies for metastatic cancer that are resistant to trastuzumab have been reported (7,8).

There is an increasing necessity to monitor the genomic profiles of tumor cells during cancer onset, recurrence, and metastasis. However, repeated tumor tissue biopsy is not always practical. Circulating tumor cells (CTCs) that have shed from a primary tumor are present in the blood circulation and may cause tumor metastases (9,10). Liquid biopsy using CTCs is noninvasive and repeatable; therefore, it is useful for counting tumor cells, pathological characterization and molecular assays. Furthermore, it is possible to use a liquid biopsy with CTCs to replace metastatic tissue biopsy for the prediction of drug sensitivity and resistance, monitoring of drug responsiveness, and detection of metastasis (11,12).

Previously, our group developed a novel technology to enrich and isolate CTCs on the basis of differences in cell size (13). In the present study, CTCs were isolated from patients with breast cancer using this method and a cancer panel analysis of isolated CTCs was performed. Furthermore, the genetic mutations of CTCs were compared with those of white blood cells (WBCs) from the same patient in order to evaluate cancer-specific mutations.

Materials and methods

Cell culture

H358-GFP, MCF7, PC9 and KG-1 cell lines (American Type Culture Collection, Manassas, VA, USA), were maintained in RPMI (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% Antibiotic-Antimycotic (Gibco; Thermo Fisher Scientific, Inc.). Cells were cultured 37°C at 5% CO2 in an incubator.

Clinical background of patients with breast cancer

From February 2015 to March 2015, 8 female patients with breast cancer, with a median age of 45 years (range, 28–48 years), and 4 female healthy volunteers with a median age of 34 years (range, 25–45 years) from the Asan Medical Center (Seoul, Korea) were included in the present study. A total of 4 patients (CG 237–240) received neoadjuvant systemic therapy and 4 patients (CG 242–245) did not receive treatment. Cancer stage was evaluated on the basis of the seventh American Joint Committee on Cancer Tumor, Node, and Metastasis Classification (Table I) (14). All blood samples and medical data used in the present study were irreversibly anonymized. The present study was approved by the Institutional Review Board of Asan Medical Center (IRB no. 2013-1048).

Table I.

Clinical information of patients with breast cancer, and Whole Genome Amplification results.

Table I.

Clinical information of patients with breast cancer, and Whole Genome Amplification results.

Patient IDAge, yearsAJCC TNM stageEpCAM+ cells, naCell typeDNA amount, µgPurity of DNA (A260/A280)On target, %b
CG 23741IIA20CTC35.971.897.83
WBC33.221.8498.89
CG 23845IIIA  1CTC30.691.8898.93
WBC32.781.8998.91
CG 23948IIA  7CTC31.351.8999.17
WBC33.551.9198.54
CG 24047IIB23CTC30.581.8498.81
WBC34.11.8799.13
CG 24248IIIA  2CTC33.441.8898.67
WBC30.691.9298.74
CG 24330IIA  6CTC31.021.9198.21
WBC31.91.8698.64
CG 24445IIA  0CTC31.021.8898.87
WBC34.431.8598.42
CG 24528IIA  1CTC35.091.8698.19
WBC33.551.8898.19

a Per 5 ml of blood.

b Reads mapped to any targeted region relative to all reads mapped to the reference. AJCC, the seventh American Joint Committee on Cancer; TNM, Tumor Lymph Node Metastasis; EpCAM, epithelial cell adhesion molecule; CTC, circulating tumor cell; WBC, white blood cell.

Blood collection and CTC enrichment

Blood from each patient (10 ml) was collected in BD Vacutainer acid citrate dextrose-solution A tubes and processed within 4 h. The blood samples were divided into two groups as follows: One for immunofluorescent staining, and the other for the cancer panel analysis of CTCs. The samples were processed using the same procedure, with a CTC isolation kit (cat. no. CIKW10; Cytogen, Inc.) used according to the protocol. Briefly, blood samples were incubated for 20 min with an antibody cocktail against WBCs and red blood cells, and then mixed with preactivation buffer prior to density gradient centrifugation (400 × g for 30 min at 25°C). A cell suspension containing CTCs was collected and gradually diluted with dilution buffer. The diluted cell suspensions were then filtered through a high-density microporous (HDM) chip as previously described (13). Cells on the HDM chip were retrieved and transferred to a microtube. For immunofluorescent staining, isolated cells were fixed in 4% paraformaldehyde for 5 min at room temperature. For cancer panel analysis, isolated cells were pelleted and kept at −80°C until further processing. From the same patient, 500 µl blood was layered onto a density gradient medium (Ficoll-Paque PLUS; GE Healthcare Life Sciences, Little Chalfont, UK) and centrifuged (400 × g for 30 min at 25°C). From the peripheral blood mononuclear cell layer, 100 WBCs were isolated as a negative control for cancer panel analysis. In addition, different numbers (5, 10, 20 and 100) of MCF7 cells were spiked into 1 ml blood from healthy volunteers, isolated using the same CTC isolation procedure, and used as a positive control for evaluation of the Cytogen protocol.

Immunofluorescent staining

Cells on slides were permeabilized with 0.2% Triton-X 100 in PBS for 10 min, and quenched with 0.3% hydrogen peroxide for 1 h. Cells were then blocked with 1% bovine serum albumin (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) in PBS for 30 min, and incubated with primary antibodies followed by secondary antibodies for 1 h each at room temperature. The primary antibodies were as follows: Mouse anti-epithelial cell adhesion molecule (EpCAM; dilution 1:200; cat. no. #2929; Cell Signaling Technology, Inc., Danvers, MA, USA) and rabbit anti-CD45 (dilution 1:10; cat. no. SC-25590; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). EpCAM signals were amplified using the Tyramide Signal Amplification system (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The secondary antibody for CD45 was Alexa 594-conjugated goat anti-rabbit immunoglobulin G (H+L) (dilution 1:200; cat. no. A11012; Invitrogen; Thermo Fisher Scientific, Inc.). The slides were mounted with Fluoroshield with DAPI (Immunobioscience Corporation, Mukilteo, WA, USA). Stained cells were observed and photographed 3 fields using a fluorescence microscope (Eclipse Ti; Nikon Corporation, Tokyo, Japan) at a magnification of ×400.

Whole genome amplification

The cell pellets that were kept at −80°C were amplified using the REPLI-g Single Cell kit (Qiagen GmbH, Hilden, Germany) according to the manufacturer's protocol. Briefly, the cell pellets were mixed with a denaturation buffer (included in the kit) and incubated at 65°C for 10 min. Following the addition of a stop solution (included in the kit), the denatured DNA samples were mixed with REPLI-g sc DNA polymerase and a reaction buffer (included in the kit) and incubated at 30°C for 8 h and then at 65°C for 3 min.

Ion AmpliSeq comprehensive cancer panel (CCP) analysis

Genomic mutations were analyzed using the Ion AmpliSeq CCP (Thermo Fisher Scientific, Inc.), which is a next-generation sequencing assay that provides all-exon coverage of 409 oncogenes and tumor suppressor genes. The Ion AmpliSeq CCP was designed to target all exons of key tumor suppressor genes and oncogenes most frequently cited and most frequently mutated. Briefly, genomic DNA was amplified using the Ion AmpliSeq Cancer Panel and the amplicons were purified using Agencourt AM-Pure XP (Beckman Coulter, Inc., Brea, CA, USA). This was followed by end repairing and ligation with Ion Xpress barcode adapters (Thermo Fisher Scientific, Inc.). The median fragment size and concentration of the final library were detected using a BioAnalyzer instrument with a high sensitivity chip (Agilent Technologies, Inc., Santa Clara, CA, USA). The library was diluted to 10 pM by low TE buffer included in the kit; and the library (5 µl) was used for emulsion PCR reactions using the Ion PI™ Hi-Q™ OT2 200 kit (Invitrogen; Thermo Fisher Scientific, Inc.). The following thermocycling conditions were used: 80°C for 3 min, followed by 18 cycles of 99°C for 20 sec, 58°C for 30 sec, 72°C for 1 min, 99°C for 20 sec, 56°C for 30 sec and 70°C for 1 min, and 10 cycles of 99°C for 20 sec and 58°C for extended durations from 3–20 min. The emulsion PCR product was enriched using Dynabeads MyOne Streptavidin C1 beads (Invitrogen; Thermo Fisher Scientific, Inc.). The final enriched Ion spheres were mixed with a sequencing primer (included in the kit) and polymerase (included in the kit) and loaded onto a total of five chips of Ion 316™ Chip kit. Base calling was generated by Torrent Suite 3.0 software (Thermo Fisher Scientific, Inc.), using tmap-f3 on the Ion Torrent server for further analysis. Bam and FASTQ alignment files were generated on the basis of the base calling result and were used to report the variant calling, including single nucleotide polymorphisms and insertions/deletions.

Catalogue of somatic mutations in cancer (COSMIC) database

COSMIC is an online database of somatically acquired mutations in human cancer. It is the most comprehensive resource for exploring the impact of somatic mutations in human cancer (15).

Statistical analysis

Correlation analysis was performed by simple linear regression analysis. The equation used was ‘Y=0.6179× + 5.6398’, and was calculated using Microsoft Excel 2010 (Microsoft Corporation, Redmond, WA, USA).

Results

Mutations are detectable in purified MCF7 cells

Fig. 1 depicts the methodologies employed by the present study. Immunofluorescent cells were counted to determine the number of EpCAM-positive cells. The purity of MCF7 cells increased with the number of cells (Fig. 2A). The recovery rate of our method using H358-GFP cell lines was 84% (data not shown). Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) mutation, which is a known mutation in MCF7, was detected in MCF7 cells isolated through the Cytogen protocol. The frequencies of mutation were increased when purity was high; however even in samples with low purity, mutations were detected (Fig. 2B). Therefore, R2 (between the purity and frequency of PIK3CA mut. in cancer panel results) of Fig. 2B indicates that the panel analysis was reliable.

Successful isolation of CTCs

CTCs were defined as EpCAM+ and CD45 cells (Fig. 3). EpCAM+ cells were detected in 7/8 patients, and the average number of EpCAM+ cells was 8.6 (1–23; Table I). PC9 (EpCAM+) and KG-1 (CD45+) cell lines were used as positive controls during immunostaining. CTCs isolated for cancer panel analysis were amplified using whole genome amplification, and the average DNA amount was 32.7 µg with high purity (A260/A280 above 1.80). On target, the percentage of reads mapped to any targeted region relative to all reads mapped to the reference was 98.6% (range 97.8–99.2%; Table I).

Cancer gene panel analysis

COSMIC was used to confirm the CCP results. When mutations were in the WBCs analyzed as negative controls, they were considered germ line mutations. These mutations in CTCs were excluded from the analysis. CTC-specific mutations were validated by comparing mutations between CTCs and WBCs (Table II). CTC-specific mutations had a detection rate of 62.5%, and these were enhancer of zeste polycomb repressive complex 2 subunit (EZH2), notch 1 (NOTCH1), AT-rich interaction domain 1A, serine/threonine kinase 11, fms related tyrosine kinase 3, MYCN proto-oncogene, bHLH transcription factor, APC, WNT signaling pathway regulator, and phosphatase and tensin homolog (PTEN).

Table II.

Comprehensive cancer panel analysis results.

Table II.

Comprehensive cancer panel analysis results.

WBCCTCCTC specific



Patient IDGeneAA mutationGeneAA mutationGeneAA mutation
CG 237MSH2Unknown
ARID1Ap.D1850fs*4
HNF1Ap.G292fs*25
EZH2p.D730fs*1EZH2p.D730fs*1
NOTCH1p.D1698DNOTCH1p.D1698D
CG 238PDGFRAp.V824VPDGFRAp.V824V
NOTCH1p.D1698DNOTCH1p.D1698D
RETp.T278NRETp.T278N
NF2p.N371NNF2p.N371N
MSH2Unknown
PTCH1p.C727fs*11
CG 239NOTCH1p.D1698D
CG 240MSH2UnknownMSH2Unknown
PDGFRAp.V824VPDGFRAp.V824V
FLT3p.L561LFLT3p.L561L
STK11p.T32TSTK11p.T32T
SMARCB1p.P383fs*4
SMARCB1p.P383fs
NOTCH1p.A2463fs*14NOTCH1p.A2463fs*14
NOTCH1p.D1698DNOTCH1p.D1698D
CG 242PDGFRAp.V824VPDGFRAp.V824V
FLT3p.L561LFLT3p.L561L
MSH2Unknown
EZH2p.D730fs*1
NOTCH1p.D1698D
ARID1Ap.D1850fs*4ARID1Ap.D1850fs*4
CG 243ARID1Ap.D1850fs*4ARID1Ap.D1850fs*4
FLT3p.L561LFLT3p.L561L
SMARCB1p.T372TSMARCB1p.T372T
NOTCH1p.D1698DNOTCH1p.D1698D
STK11p.L282fs*3STK11p.L282fs*3
CG 244MSH2Unknown
PDGFRAp.V824V
STK11p.L282fs*3
ARID1Ap.K1072fs*21ARID1Ap.K1072fs*21
NOTCH1p.D1698DNOTCH1p.D1698D
FLT3p.L561LFLT3p.L561L
CG 245MSH2Unknown
STK11p.L282fs*3
MYCNp.P358L
APCp.R554*
NOTCH1p.D1698D
PTENp.L57fs*6

[i] WBC, white blood cell; CTC, circulating tumor cell; AA, amino acid; CG, patient ID; MSH2, mutS homolog 2; PDGFRA, platelet derived growth factor receptor α; FLT3, fms related tyrosine kinase 3; STK11, serine/threonine kinase 11; SMARCB1, SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1; NOTCH1, notch 1; EZH2, enhancer of zeste polycomb repressive complex 2 subunit; ARID1A, AT-rich interaction domain 1A; MYCN, MYCN proto-oncogene, bHLH transcription factor; APC, APC, WNT signaling pathway regulator; PTEN, phosphatase and tensin homolog.

Discussion

Patients with early-stage hormone receptor-positive breast cancer may have several effective treatment options; however, multiple patients also develop recurrence and metastasis. Therefore, the early diagnosis of cancer, prognostication and monitoring of the genomic characteristics of tumor cells are essential (3). However, biopsies of tumor tissues are not always easy to repeat. CTCs may be able to overcome this limitation of tumor tissue biopsies as CTCs have similar characteristics to those of primary tumors, and may cause metastasis (16). Due to the presence of CTCs at low concentrations (1 in 1×109) (17), it is important to enrich or isolate them from the blood effectively. CellSearch (Menarini Silicon Biosystems, Bologna, Italy), a well-known commercial device, isolates CTCs through EpCAM+ selection (18). It is not possible to achieve this EpCAM+ selection technique when tumor cells downregulate EpCAM expression (19). In addition, a previous study reported that a significant portion of CTCs are EpCAM (20).

Our group has developed a novel CTC enrichment technique based on cell size difference and double negative selection, which removes nontargeted cells with an antibody complex against WBCs (13). Using this technique, CTCs were effectively isolated at a purity that was sufficient for genomic analysis (Fig. 2). In addition, COSMIC mutations were detected even in patients lacking EpCAM+ cells (Table I), demonstrating that this technology was able to effectively isolate EpCAM CTCs. Among the CTC-specific COSMIC gene mutations that were identified, EZH2, NOTCH1 and PTEN have been reported to affect breast cancer status (2124). EZH2 mutations cause abnormal DNA methylation and promote mammary stem cell expansion and metastasis (21). NOTCH1 has been reported to regulate the epithelial-mesenchymal transition and to promote the migration and invasion of breast cancer cells (22). Furthermore, NOTCH1 expression in breast tumor tissues is higher than in normal tissues (23). Mutated PTEN is not able to inhibit the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin pathway, thereby losing its tumor suppressor activity (24).

There was a previous report on single-gene mutation analysis of CTCs and WBCs (25), and another study performed cancer panel analysis of CTCs without WBCs as controls (26). However, to the best of our knowledge, the present study is the first attempt at a CCP using CTCs in conjunction with WBCs. CTC-specific COSMIC mutations were identified, and genomic information that may be useful for precision medicine was provided.

In conclusion, the CTC isolation technique used by the present study was effective, providing sufficient purity for genomic analysis, and demonstrated that CCP analysis is a potential application for precision medicine.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the National R&D Program, Ministry of Trade, Industry and Energy, Republic of Korea (grant no., 10045947).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

SHC, MSK JL and BHJ directed and designed the study with contributions from CHL and SJL. DHL, DYH and PSP analyzed the circulating tumor cells from the patients. MSC and HKL maintained the cell lines and performed the spike tests. SHA, BHS, JWL and JHY provided patients' blood samples and clinical information. NJK, WCL and KSY performed the CCP (comprehensive cancer panel) analyses. CHL and MSK wrote the manuscript with contributions from JL and BHJ. All authors have read and approved the manuscript.

Ethics approval and consent to participate

The present study was approved by the Institutional Review Board of Asan Medical Center (Institutional Review Board no. 2013-1048). All blood samples and medical data used in the present study were irreversibly anonymized.

Consent for publication

Not applicable.

Competing interests

CHL, SJL, SHC, DHL, DYH, MSC, PSP, HKL, MSK, JL and BHJ are employees of Cytogen, Inc. (Seoul, Korea), and the CTC isolation kit was supplied courtesy of Cytogen, Inc. The authors report no other competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Jung KW, Won YJ, Kong HJ, Oh CM, Cho H, Lee DH and Lee KH: Cancer statistics in Korea: Incidence, mortality, survival, and prevalence in 2012. Cancer Res Treat. 47:127–141. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Lorusso G and Rüegg C: New insights into the mechanisms of organ-specific breast cancer metastasis. Semin Cancer Boil. 22:226–233. 2012. View Article : Google Scholar

4 

Suzuki M and Tarin D: Gene expression profiling of human lymph node metastases and matched primary breast carcinomas: Clinical implications. Mol Oncol. 1:172–180. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Von Minckwitz G, du Bois A, Schmidt M, Maass N, Cufer T, de Jongh FE, Maartense E, Zielinski C, Kaufmann M, Bauer W, et al: Trastuzumab beyond progression in human epidermal growth factor receptor 2-positive advanced breast cancer: A german breast group 26/breast international group 03–05 study. J Clin Oncol. 27:1999–2006. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Peake BF and Nahta R: Resistance to HER2-targeted therapies: A potential role for FOXM1. Breast Cancer Manag. 3:423–431. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Blackwell KL, Burstein HJ, Storniolo AM, Rugo H, Sledge G, Koehler M, Ellis C, Casey M, Vukelja S, Bischoff J, et al: Randomized study of Lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol. 28:1124–1130. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Modi S, Stopeck A, Linden H, Solit D, Chandarlapaty S, Rosen N, D'Andrea G, Dickler M, Moynahan ME, Sugarman S, et al: HSP90 inhibition is effective in breast cancer: A phase II trial of tanespimycin (17-AAG) plus trastuzumab in patients with HER2-positive metastatic breast cancer progressing on trastuzumab. Clin Cancer Res. 17:5132–5139. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Franken B, de Groot MR, Mastboom WJ, Vermes I, van der Palen J, Tibbe AG and Terstappen LW: Circulating tumor cells, disease recurrence and survival in newly diagnosed breast cancer. Breast Cancer Res. 14:R1332012. View Article : Google Scholar : PubMed/NCBI

10 

Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse M, Mitchell E, Miller MC, et al: Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 26:3213–3221. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Van de Stolpe A, Pantel K, Sleijfer S, Terstappen LW and den Toonder JM: Circulating tumor cell isolation and diagnostics: Toward routine clinical use. Cancer Res. 71:5955–5960. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Giuliano M, Giordano A, Jackson S, De Giorgi U, Mego M, Cohen EN, Gao H, Anfossi S, Handy BC, Ueno NT, et al: Circulating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic dissemination. Breast Cancer Res. 16:4402014. View Article : Google Scholar : PubMed/NCBI

13 

Kim EH, Lee JK, Kim BC, Rhim SH, Kim JW, Kim KH, Jung SM, Park PS, Park HC, Lee J, et al: Enrichment of cancer cells from whole blood using a microfabricated porous filter. Anal Biochem. 440:114–116. 2013. View Article : Google Scholar : PubMed/NCBI

14 

American Joint Committee on Cancer: AJCC Cancer Staging Manual. 7th edition. Springer; 2010

15 

Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al: COSMIC: Mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res. 39:D945–D950. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Lang JM, Casavant BP and Beebe DJ: Circulating tumor cells: Getting more from less. Sci Transl Med. 4:141ps132012. View Article : Google Scholar : PubMed/NCBI

17 

Mostert B, Sleijfer S, Foekens JA and Gratama JW: Circulating tumor cells (CTCs): Detection methods and their clinical relevance in breast cancer. Cancer Treat Rev. 35:463–474. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, Janni W, Coith C, Beck K, Jänicke F, et al: Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: A validation study of the cellsearch system. Clin Cancer Res. 13:920–928. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Casavant BP, Mosher R, Warrick JW, Maccoux LJ, Berry SM, Becker JT, Chen V, Lang JM, McNeel DG and Beebe DJ: A negative selection methodology using a microfluidic platform for the isolation and enumeration of circulating tumor cells. Methods. 64:137–143. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Giordano A, Gao H, Anfossi S, Cohen E, Mego M, Lee BN, Tin S, De Laurentiis M, Parker CA, Alvarez RH, et al: Epithelial-mesenchymal transition and stem cell markers in patients with HER2-positive metastatic breast cancer. Mol Cancer Ther. 11:2526–2534. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Wu J and Crowe DL: The histone methyltransferase EZH2 promotes mammary stem and luminal progenitor cell expansion, metastasis and inhibits estrogen receptor-positive cellular differentiation in a model of basal breast cancer. Oncol Rep. 34:455–460. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Shao S and Zhao X, Zhang X, Luo M, Zuo X, Huang S, Wang Y, Gu S and Zhao X: Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer. 14:282015. View Article : Google Scholar : PubMed/NCBI

23 

Yuan X, Zhang M, Wu H, Xu H, Han N, Chu Q, Yu S, Chen Y and Wu K: Expression of Notch1 correlates with breast cancer progression and prognosis. PLoS One. 10:e01316892015. View Article : Google Scholar : PubMed/NCBI

24 

Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, et al: Crucial role of p53-dependent cellular senescence in suppression of PTEN-deficient tumorigenesis. Nature. 436:725–730. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Fernandez SV, Bingham C, Fittipaldi P, Austin L, Palazzo J, Palmer G, Alpaugh K and Cristofanilli M: TP53 mutations detected in circulating tumor cells present in the blood of metastatic triple negative breast cancer patients. Breast Cancer Res. 16:4452014. View Article : Google Scholar : PubMed/NCBI

26 

Liu S, Wang H, Zhang L, Tang C, Jones L, Ye H, Ban L, Wang A, Liu Z, Lou F, et al: Rapid detection of genetic mutations in individual breast cancer patients by next-generation DNA sequencing. Hum Genomics. 9:22015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 16 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee CH, Lee SJ, Choi SH, Ahn SH, Son BH, Lee JW, Yu JH, Kwon NJ, Lee WC, Yang KS, Yang KS, et al: Cancer panel analysis of circulating tumor cells in patients with breast cancer. Oncol Lett 16: 612-618, 2018
APA
Lee, C.H., Lee, S.J., Choi, S.H., Ahn, S.H., Son, B.H., Lee, J.W. ... Jeon, B.H. (2018). Cancer panel analysis of circulating tumor cells in patients with breast cancer. Oncology Letters, 16, 612-618. https://doi.org/10.3892/ol.2018.8646
MLA
Lee, C. H., Lee, S. J., Choi, S. H., Ahn, S. H., Son, B. H., Lee, J. W., Yu, J. H., Kwon, N., Lee, W. C., Yang, K., Lee, D. H., Han, D. Y., Choi, M. S., Park, P., Lee, H. K., Kim, M. S., Lee, J., Jeon, B. H."Cancer panel analysis of circulating tumor cells in patients with breast cancer". Oncology Letters 16.1 (2018): 612-618.
Chicago
Lee, C. H., Lee, S. J., Choi, S. H., Ahn, S. H., Son, B. H., Lee, J. W., Yu, J. H., Kwon, N., Lee, W. C., Yang, K., Lee, D. H., Han, D. Y., Choi, M. S., Park, P., Lee, H. K., Kim, M. S., Lee, J., Jeon, B. H."Cancer panel analysis of circulating tumor cells in patients with breast cancer". Oncology Letters 16, no. 1 (2018): 612-618. https://doi.org/10.3892/ol.2018.8646