Article Search
검색
검색 팝업 닫기

Metrics

Help

  • 1. Aims and Scope

    Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE

  • 2. Editorial Board

    Editor-in-Chief + MORE

    Editor-in-Chief
    Yong Chan Lee Professor of Medicine
    Director, Gastrointestinal Research Laboratory
    Veterans Affairs Medical Center, Univ. California San Francisco
    San Francisco, USA

    Deputy Editor

    Deputy Editor
    Jong Pil Im Seoul National University College of Medicine, Seoul, Korea
    Robert S. Bresalier University of Texas M. D. Anderson Cancer Center, Houston, USA
    Steven H. Itzkowitz Mount Sinai Medical Center, NY, USA
  • 3. Editorial Office
  • 4. Articles
  • 5. Instructions for Authors
  • 6. File Download (PDF version)
  • 7. Ethical Standards
  • 8. Peer Review

    All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
    The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.

    The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.

Search

Search

Year

to

Article Type

Review

Split Viewer

Clinical Application of Circulating Tumor Cells in Gastric Cancer

Moon Won Lee , Gwang Ha Kim , Hye Kyung Jeon , Su Jin Park

Department of Internal Medicine, Pusan National University School of Medicine, and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea

Correspondence to: Gwang Ha Kim
Department of Internal Medicine, Pusan National University School of Medicine, and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan 49241, Korea
Tel: +82-51-240-7869, Fax: +82-51-244-8180, E-mail: doc0224@pusan.ac.kr

Received: October 30, 2018; Revised: November 19, 2018; Accepted: November 23, 2018

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Gut Liver 2019;13(4):394-401. https://doi.org/10.5009/gnl18484

Published online July 15, 2019, Published date February 27, 2019

Copyright © Gut and Liver.

Early detection and accurate monitoring of cancer is important for improving clinical outcomes. Endoscopic biopsy and/or surgical resection specimens are the gold standard for diagnosing gastric cancer and are also useful for selecting therapeutic strategies based on the analysis of genomic/immune parameters. However, these approaches cannot be easily performed because of their invasiveness and because these specimens do not always reflect tumor dynamics and drug sensitivities during therapeutic processes, especially chemotherapy. Accordingly, many researchers have tried to develop noninvasive novel biomarkers that can monitor real-time tumor dynamics for early diagnosis, prognostic evaluation, and prediction of recurrence and therapeutic efficacy. Circulating tumor cells (CTCs) are metastatic cells that are released from the primary tumors into the blood stream and comprise a crucial step in hematogenous metastasis. CTCs, as a liquid biopsy, have received a considerable amount of attention from researchers since they are easily accessible in peripheral blood, avoiding the invasiveness associated with traditional biopsy techniques; they can also be used to derive clinical information for monitoring disease status. In this review, with respect to CTCs, we summarize the metastatic cascade, detection methods, clinical applications, and prospects for patients with gastric cancer.

Keywords: Circulating tumor cells, Liquid biopsy, Neoplasms, Stomach neoplasms

Gastric cancer (GC) is the 6th most prevalent cancer (1,033,701 new cases per year) and the 2nd most common cause of cancer-related deaths worldwide in 2018.1 Despite the development of diagnostic and therapeutic modalities, more than 40% of patients with advanced GC have a poor prognosis and exhibit no response to chemotherapy or resistance to chemotherapy during treatment.2 Approximately 30% to 40% of GC patients experience tumor recurrence and metastasis even after curative resection.3,4 This suggests the presence of potential metastatic cells that cannot be detected based on conventional diagnostic methods.

Traditional tumor markers such as carcinoembryonic antigen (CEA) and cancer antigen 19-9 (CA19-9) are known biomarkers of GC. However, the positivity rate of these markers is less than 40% in GC patients, and the sensitivity and specificity of assays to detect these markers are insufficient for diagnostic and prognostic approaches.5 Generally, the diagnosis of GC is based on endoscopic biopsy and/or surgical resection specimens, but these approaches cannot be easily performed due to their invasiveness. Furthermore, these specimens do not represent the intratumoral heterogeneity and mechanisms of tumor progression and resistance to treatment. Therefore, the development of new modalities that can predict recurrence and metastasis, and which can be used for diagnosis and evaluation of therapeutic response, is important to improve clinical outcomes for GC patients.6

Circulating tumor cells (CTCs) are disseminated tumor cells in the peripheral blood of cancer patients. The first report of CTCs occurred in 1869 from Thomas Ashworth, who described the presence of tumor-derived epithelial cells in the blood compartment of a patient with end-stage metastatic cancer.7 However, because the number of cancer cells in circulation ranges from only approximately around 1–1,000 cells/10 mL,8 conventional methods are not adequate for their detection. Recently, numerous new approaches have been established to isolate, enumerate, and characterize CTCs in patients with various cancers.9 CTCs have strengths such as accessibility and superiority over conventional tumor markers; moreover, based on the analysis of CTCs, researchers can elucidate genetic and phenotypic differences between primary and metastatic tumors. Herein, we summarize the metastatic cascade associated with CTCs, provide an overview of the detection methods for CTCs, and suggest clinical applications for CTCs in GC patients; we then discuss their future use in the clinical practice.

Cancer metastasis is a multi-step process that includes the loss of intercellular connections, invasion into the basal membrane and surrounding tissues, intravasation into venous or lymphatic vessels, which generates CTCs, survival in the peripheral system, extravasation, and proliferation at secondary sites.1012 Epithelial tumor cells are immobile due to strong cell-to-cell and cell-to-extracellular adhesions, which consist of adherent junctions, tight junctions, and desmosomes. Moreover, conditions in the blood stream are too harsh for the survival of epithelial tumor cells. Therefore, these cells undergo epithelial-to-mesenchymal transition (EMT), which is associated with the loss of adhesion, increased plasticity, and the capacity for migration and invasion. EMT is a molecular and cellular change that includes the down-regulation of epithelial proteins such as E-cadherin, claudins and cytokeratins and the up-regulation of mesenchymal proteins such as N-cadherin, fibronectin and vimentin, which confers increased motility and invasiveness to the cells.1012 EMT promotes the generation of CTCs by increasing tumor cell invasiveness,1315 promoting tumor cell intravasation,16,17 and facilitating tumor cell survival in the peripheral system.18,19

The correlation between EMT and tumor cell invasiveness or CTCs has been confirmed, but the details of the metastatic cascade are still controversial. Two hypotheses have been proposed for the metastatic model associated with EMT: EMT/mesenchymal-epithelial migration (MET) model and collective migration model (Fig. 1).20 In the EMT/MET model, at first, epithelial cancer cells must undergo EMT to become invasive and motile and to generate CTCs; after that, CTCs circulate throughout the entire body and extravasate to distant secondary sites. After extravasation to secondary sites, cancer cells need to recover their epithelial properties via MET, the inverse process of EMT, and then finally colonize at the distant site to complete the metastasis.2124 This metastatic model is the first reported and most widely known model, and also explains why the histological characteristics of the primary tumor are consistent with those of the secondary tumor.22 Furthermore, recent studies have reported a switch between EMT and MET marker phenotypic plasticity in some cancer types.25,26 In the collective migration model, epithelial-like and mesenchymal-like cancer cells can cooperate during collective migration to achieve metastasis instead of undergoing the difficult process of EMT and subsequent MET.27,28 Instead of migrating as a single cell, cancer cells undergoing various degrees of EMT coexist as multi-cellular clusters that migrate collectively. More motile, invasive mesenchymal-like cells aggregate at the invasive front of multi-cellular clusters to facilitate this process, whereas epithelial-like cells follow behind and exploit the opportunity to proliferate and colonize at the distant sites after extravasation.28 Accordingly, multi-cellular cancer fragments, or so-called tumor micro-emboli or CTC clusters, have been observed and correlate with distant metastasis.29,30 Of note, the EMT/MET and collective migration models might not be independent or opposing. Notably, tumor cells might use both mechanisms alternately under certain circumstances, or the two mechanisms could synergistically contribute to metastases.

In general, CTCs are rare in the peripheral blood of cancer patients. The enumeration of these CTCs is a great technological challenge. Thus, the enrichment methods require sufficient sensitivity and reproducibility. Current methods for separating CTCs include those based on immunoaffinity differences, using an antibody that binds to a specific protein marker on the surface of cancer cells,3133 and differences in physical properties such as the sizes, densities, and electrical characteristics of cancer cells and blood cells (Fig. 2).3439

Immunoaffinity-based separation is the most widely-used strategy. This method uses specific biomarkers such as epithelial cell adhesion molecule (EpCAM) and CD45 which are expressed on the cell surface, to capture cells. The antibodies used for selection are typically linked to the surface of a device or to a magnetic substance allowing them to be captured by a magnetic field (i.e., immunomagnetic capture). During positive selection, tumor-associated cell surface antigens such as EpCAM are targeted, whereas negative selection removes background cells by antigens not expressed on CTCs (e.g., CD45). Specifically, the CellSearchTM platform (Veridex LLC, Huntingdon Valley, PA, USA) was approved by the U.S. Food and Drug Administration (FDA) for the diagnosis and prognostic evaluation of patients with metastatic breast cancer, colorectal cancer, and prostate cancer between 2004 and 2008.40 However, the expression level of CTCs is extremely variable, and CTCs with low target antibody expression are more difficult to capture.41 Moreover, separation processes for CTCs include complex steps and are time consuming with respect to achieving sufficient reactions between antigens and antibodies. In addition, the heterogeneous array of surface markers expressed by CTCs has made it impossible to identify a universal CTC-specific antigen.

In the density gradient method, blood samples are diluted and laid over the media, and then separated by centrifugation.42 With this, it is easy to remove red blood cells with a high density, but it is difficult to separate CTCs and white blood cells due to their similar densities. Size-based filtration has been used to enrich CTCs based on the fact that they are larger than other blood cells. However, CTCs are generally 8 to 20 μm in size, whereas leukocytes are 6 to 10 μm; thus, leukocytes will be present to some extent in samples treated by the size-based separation methods, which makes it difficult to capture high-purity CTCs.43 In the inertia sorting method, the CTCs are isolated using a spiral channel microfluidic chip, which separates different-sized cells with different Dean forces in a curved channel. With this, it is possible to capture CTCs exhibiting different expression levels of specific surface proteins, and this technique has several advantages such as fast separation time, ease of use, and low cost. However, size-based filtration has the disadvantage of potentially capturing contaminated cells.44

Dielectrophoresis (DEP) is an electrokinetic method that exploits the dielectric properties of floating cells for identification and separation. This method isolates CTCs based on different responses to DEP due to differences in size and membrane properties. Further, it is advantageous for the separation of CTCs because of its simple experimental set-up, much higher operational flow rate compared to other separator methods, and lack of dependence on cell surface markers.45 However, for efficient isolation, whole blood cannot be used directly; there is also limitations associated with the use of a special buffer optimized for electrical conductivity to increase the efficiency of cancer cell isolation.

With advances in technology, next generation microfluidic platforms that enrich CTCs have been developed for cost-effectiveness and high levels of purity. Therefore, if these platforms with high sensitivity and specificity can be further developed, they could be used extensively in clinical practice as well as in the studies on CTCs.

1. Diagnostic and prognostic value of circulating tumor cells

Based on a meta-analysis of the diagnostic accuracy of various CTC detection methods, the pooled sensitivity and specificity of CTCs for GC were 42% (95% confidence interval [CI], 21% to 67%) and 99% (95% CI, 96% to 100%), respectively;46 the authors of this study concluded that CTC detection has a limited role in screening test for GC, but might be used as a non-invasive method for the confirmation of GC diagnosis. Seventeen of 20 studies included in this meta-analysis used reverse transcriptase-polymerase chain reaction (RT-PCR) for cytokeratin or CEA to detect CTCs and the results varied among different detection methods. RT-PCR is one molecular approach for the detection of CTCs; however, an ideal method should focus on the tumor cells directly rather than indirectly-linked surrogate markers via a molecular approach.47 To date, the CellSearchTM system (Veridex LLC) is the only CTC detection technique approved by the U.S. FDA. Using this platform, it is possible to obtain highly reproducible quantitative results in different laboratories. Therefore, this review will focus further on studies reporting significant data regarding the role of CTCs in GC that were obtained by the cytometric methods including the CellSearchTM system.4860 The results of these studies are summarized in Table 1.61

In our previous study,56 CTCs were measured in 116 GC patients and 31 healthy individuals using a centrifugal microfluidic system with a new fluid-assisted separation technique, specifically based on size-selective CTC isolation.56 When using a CTC level of ≥2 per 7.5 mL of blood, the sensitivity and specificity for differentiating GC patients from healthy controls was 85.3% (95% CI, 81.4% to 87.2%) and 90.3% (95% CI, 75.6% to 97.4%), respectively. In addition, CTCs were detected in more than 80% of early stage (T1 or N0) GC patients. Although CTCs were not associated with any clinicopathologic features such as staging, histologic type, or mucin phenotype, our results suggest that CTCs could be an early diagnostic biomarker for GC. In a recent study, using a novel wedge-shaped microfluidic chip, CTCs were detected in 75% (30/40) of GC patients, whereas CTCs were not detected in 25 healthy donors.60 In this study, CTCs were associated with tumor differentiation, lymphovascular invasion, and staging. These differences in the association between CTCs and clinicopathologic characteristics might be attributed to heterogeneity in the baseline clinicopathologic characteristics and the use of different CTC detection methods.

The measurement of CTCs in GC patients can be useful for predicting patient survival and prognosis. In a prospective study including 251 patients with advanced GC using the CellSearchTM system, CTCs were detected in 16 patients (16/148, 10.8%) in the resection group with a higher relapse rate and in 62 patients (62/103, 60.2%) in the non-resectable group, but were not detected in healthy volunteer. Thus, the evaluation of CTCs might be a useful strategy to predict tumor progression and prognosis in GC patients. Based on a meta-analysis (including 26 studies comprising 2,566 GC patients) evaluating the prognostic significance of CTCs in GC patients, the frequency of CTC detection was higher in advanced GC than early GC, in poorly differentiated GC than well/moderately differentiated GC, and in GC with lymphatic metastasis than that without lymphatic metastasis; it was also significantly related to the disease-free and overall survival of patients.62

In addition to being diagnostic and prognostic indicators, CTCs can be used to monitor chemotherapy response. The measurement of CTCs using peripheral blood is non-invasive and can be performed repeatedly. Thus, periodically monitoring CTCs could be helpful to predict the efficacy of chemotherapeutics. Although CEA and CA19-9 are frequently used as markers in GC, they are not appropriate for this application because temporary increases can be detected after chemotherapy and many patients with advanced GC have normal levels of CEA and CA19-9. Matsusaka et al.50 measured CTC levels at baseline, 2 weeks, and 4 weeks in 52 patients with advanced GC undergoing S-1-based chemotherapy. Patients with ≥4 CTCs at 2- and 4-weeks post-chemotherapy had a shorter median progression-free survival (PFS) and overall survival (OS).50 In another study measuring CTC levels at baseline and after 6 weeks of chemotherapy in 136 patients with advanced GC, patients with ≥3 CTCs after chemotherapy had a shorter PFS and OS; moreover, an elevated number of CTCs after chemotherapy corresponded to ineffective therapeutic responses.55 Therefore, monitoring dynamic changes in CTCs in response to therapy might be a useful alternative for assessing treatment resistance in GC patients.

2. Value of circulating tumor cells for selecting therapeutic agents

Overcoming tumor heterogeneity is a major challenge for the personalized treatment of GC. Therefore, the further characterization of CTCs might help to identify specific morphological, phenotypic, and molecular features of different cancer types based on time, disease stage, and therapeutic definitions.63 As stated earlier, CTCs that have undergone the EMT exhibit enhanced cell motility rather than growth, resulting in the ability of cancer cells to exit the vasculature. In this situation, most CTCs are not responsive to cytotoxic chemotherapy because they do not undergo cell division.64,65 Therefore, the addition of CTC-targeted treatment to conventional chemotherapy might be helpful to improve prognosis for cancer patients. In non-small-cell lung cancers, a therapeutic agent targeting an epidermal growth factor receptor (EGFR) mutation has been developed, therefore, it is important to also detect this EGFR mutation in CTCs. Currently, there are several clinical reports suggesting that detection rate of the EGFR mutation in the CTCs of patients with non-small-cell lung cancer is comparable to that in tissues, confirming a high rate of concordance.6668

For GC, trastuzumab, which bind the human epidermal growth factor receptor 2 (HER2) gene, and ramucirumab, which bind vascular endothelial growth factor receptor-2, have been developed. Especially, the overexpression of HER2 is a selective biomarker for treatment with the monoclonal antibody bevacizumab in metastatic GC. Recently, Iwatsuki et al.69 evaluated CTCs and their HER2 status in patients with gastrointestinal cancer. Among the 62 CTC-positive cases, 22 (35.5%) showed discordance in terms of HER2 status between primary tumors and CTCs. Among the HER2-negative primary tumors, 17 of 54 developed HER2-positive CTCs, and among the HER2-positive primary tumors, five of eight were associated with HER2-negative CTCs. These results suggest that for primary HER2-negative tumors, CTCs could acquire HER2 gene amplification during cancer progression, and therefore, it might be necessary to determine the HER2 status of CTCs, as a liquid biopsy, to suggest personalized treatment strategies for GC. Thus, it is expected that CTCs derived from GC patients could be used to predict responses to targeted therapy. These CTCs require precise medicine, which will not only help to select the appropriate chemotherapy but will also allow for the periodic monitoring of mutations associated with resistance.

For the use of CTCs to become a clinical analytical method that complements tissue biopsies, there are many issues that need to be solved. Regarding the CellSearchTM system, cells that are positive for EpCAM are isolated, which can be considered a false negative for CTCs undergoing EMT. Therefore, it is necessary to find markers to accurately detect CTCs with an EMT phenotype. In addition, it is difficult to directly compare the sensitivity, specificity, and reproducibility of CTC separation methods because the types and preparations of various CTC isolation methods currently under development are different and have not been standardized. Therefore, it is important to develop a standardized isolation method to establish appropriate criteria for the use of CTCs as diagnostic and prognostic indicators of GC, and to confirm their clinical significance through large scale prospective clinical studies.

Technology associated with liquid biopsy was globally recognized as one of the world top 10 technologies in 2017 and comprises a highly competitive field for the development of methods that enable the most simple and accurate diagnosis of cancer. Recently, several studies have shown that peripheral blood CTCs are useful to predict prognosis and monitor therapy in GC patients. Future CTC studies are expected to provide additional information to identify mechanisms that are important for the proliferation and metastasis of malignant tumors. Furthermore, these studies are expected to play a major role in precision medicine by uncovering molecular biologic targets related to drug sensitivity and resistance in GC patients.

Fig. 1.Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) in the emergence and progression of carcinoma. Adapted from Jie XX, et al. Oncotarget 2017;8:81558–81571.
Fig. 2.Enrichment technologies for circulating tumor cells (CTCs). A: Immunoaffinity method using immunomagnetic particles that bind to the CTC surface antigen. B: Density gradient method separating other blood cells from CTCs based on their density. C: Dielectrophoresis (DEP) utilizing the dielectric properties of CTCs under an electric field. D: Size-based filtration resulting in enrichment based on the larger size of CTCs compared with the sizes of other cells. E: Inertia sorting method using Dean force at microfluidic channels. Adapted from Alix-Panabières C and Pantel K. Nat Rev Cancer 2014;14:623–631.

Summary of Studies on Circulating Tumor Cells Using Cytometric Methods in Patients with Gastric Cancer

YearAuthorNo. of patientsDetection methodsMarkersPositive definitionCTC detection, No. (%)OutcomesClinical significance
2007Pituch-Noworolska et al.4857FACSCK8,18,19≥3 CTCs31 (54)OSNo
2008Hiraiwa et al.49Non-metastatic, 14CellSearchEpCAM≥2 CTCs2 (14)OSYes
Metastatic, 27CK8,18,1915 (56)Monitoring
2010Matsusaka et al.50Baseline, 52CellSearchEpCAM≥4 CTCs17 (33)OS, PFSYes
2-wk, 51CK8,18,197 (14)Monitoring
4-wk, 489 (19)
2013Uenosono et al.51Resection, 148CellSearchEpCAM≥1 CTC16 (11)OS, RFSYes
Non-resectable, 103CK8,18,1962 (60)
2015Okabe et al.52136CellSearchEpCAM≥1 CTC25 (18)PFSYes
CK8,18,19
2015Yuan et al.5331FACSCD44+ CD45−-14 (45)--
2016Kolostova et al.5422MetaCellCK7,18,19, 20-13 (59)--
EpCAM, MUC1, HER2, EGFR
2016Li et al.55Baseline, 136CellSearchEpCAM≥3 CTCs57 (42)OS, PFSYes
6-wk, 106CK8,18,1926 (25)Monitoring
2017Kang et al.56116FASTEpCAM/CK≥2 CTCs99 (85)--
2017Liu et al.5759CELLectionEpCAM>2 CTCs36 (61)OS, PFSYes
CK7,8,18,19Monitoring
2017Pernot et al.58Baseline, 106CellSearchEpCAM≥2 CTCs49 (46)OS, PFSYes
4-wk, 65CK8,18,1912 (18)Monitoring
2018Brungs et al.5943IsoFluxEpCAM≥17 CTCs20 (47)OSYes
platformMonitoring
2018Yang et al.6040Microfluidic chipCK≥1 CTC20 (75)-Yes

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424.
    Pubmed CrossRef
  2. Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric cancer. Lancet 2009;374:477-490.
    Pubmed KoreaMed CrossRef
  3. Lim DH, Kim DY, Kang MK, et al. Patterns of failure in gastric carcinoma after D2 gastrectomy and chemoradiotherapy: a radiation oncologist’s view. Br J Cancer 2004;91:11-17.
    Pubmed KoreaMed CrossRef
  4. Marrelli D, De Stefano A, de Manzoni G, Morgagni P, Di Leo A, Roviello F. Prediction of recurrence after radical surgery for gastric cancer: a scoring system obtained from a prospective multicenter study. Ann Surg 2005;241:247-255.
    Pubmed KoreaMed CrossRef
  5. Li Y, Yang Y, Lu M, Shen L. Predictive value of serum CEA, CA19-9 and CA72.4 in early diagnosis of recurrence after radical resection of gastric cancer. Hepatogastroenterology 2011;58:2166-2170.
    Pubmed CrossRef
  6. Kang EA, Han YM, Park JM, Yoo IK, Hong SP, Hahm KB. Liquid biopsy: current status and future perspective in gastric cancer and helicobacter infection. Korean J Helicobacter Up Gastrointest Res 2018;18:150-156.
    CrossRef
  7. Ashworth T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J 1869;14:146.
  8. Nieva JJ, Kuhn P. Fluid biopsy for solid tumors: a patient’s companion for lifelong characterization of their disease. Future Oncol 2012;8:989-998.
    Pubmed KoreaMed CrossRef
  9. Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer 2004;4:448-456.
    Pubmed CrossRef
  10. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-1428.
    Pubmed KoreaMed CrossRef
  11. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2:442-454.
    Pubmed CrossRef
  12. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009;139:871-890.
    Pubmed CrossRef
  13. Liberko M, Kolostova K, Bobek V. Essentials of circulating tumor cells for clinical research and practice. Crit Rev Oncol Hematol 2013;88:338-356.
    Pubmed CrossRef
  14. Derksen PW, Liu X, Saridin F, et al. Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 2006;10:437-449.
    Pubmed CrossRef
  15. Hulit J, Suyama K, Chung S, et al. N-cadherin signaling potentiates mammary tumor metastasis via enhanced extracellular signal-regulated kinase activation. Cancer Res 2007;67:3106-3116.
    Pubmed CrossRef
  16. Qi J, Wang J, Romanyuk O, Siu CH. Involvement of Src family kinases in N-cadherin phosphorylation and beta-catenin dissociation during transendothelial migration of melanoma cells. Mol Biol Cell 2006;17:1261-1272.
    Pubmed KoreaMed CrossRef
  17. Drake JM, Strohbehn G, Bair TB, Moreland JG, Henry MD. ZEB1 enhances transendothelial migration and represses the epithelial phenotype of prostate cancer cells. Mol Biol Cell 2009;20:2207-2217.
    Pubmed KoreaMed CrossRef
  18. Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 2010;29:4741-4751.
    Pubmed KoreaMed CrossRef
  19. Tiwari N, Gheldof A, Tatari M, Christofori G. EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012;22:194-207.
    Pubmed CrossRef
  20. Jie XX, Zhang XY, Xu CJ. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: mechanisms and clinical applications. Oncotarget 2017;8:81558-81571.
    Pubmed KoreaMed CrossRef
  21. Hugo H, Ackland ML, Blick T, et al. Epithelial: mesenchymal and mesenchymal. Epithelial transitions in carcinoma progression. J Cell Physiol 2007;213:374-383.
    Pubmed CrossRef
  22. Chaffer CL, Thompson EW, Williams ED. Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs 2007;185:7-19.
    Pubmed CrossRef
  23. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science 2011;331:1559-1564.
    Pubmed CrossRef
  24. Banyard J, Bielenberg DR. The role of EMT and MET in cancer dissemination. Connect Tissue Res 2015;56:403-413.
    Pubmed KoreaMed CrossRef
  25. Brabletz T, Jung A, Reu S, et al. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci U S A 2001;98:10356-10361.
    Pubmed KoreaMed CrossRef
  26. Hudson LG, Zeineldin R, Stack MS. Phenotypic plasticity of neoplastic ovarian epithelium: unique cadherin profiles in tumor progression. Clin Exp Metastasis 2008;25:643-655.
    Pubmed KoreaMed CrossRef
  27. Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer 2003;3:362-374.
    Pubmed CrossRef
  28. Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol 2009;10:445-457.
    Pubmed CrossRef
  29. Chui MH. Insights into cancer metastasis from a clinicopathologic perspective: epithelial-mesenchymal transition is not a necessary step. Int J Cancer 2013;132:1487-1495.
    Pubmed CrossRef
  30. Wang C, Mu Z, Chervoneva I, et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res Treat 2017;161:83-94.
    Pubmed CrossRef
  31. Riethdorf S, Fritsche H, Müller V, 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 2007;13:920-928.
    Pubmed CrossRef
  32. Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol 2010;2010 617421.
    Pubmed KoreaMed CrossRef
  33. Harb W, Fan A, Tran T, et al. Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay. Transl Oncol 2013;6:528-538.
    Pubmed KoreaMed CrossRef
  34. Desitter I, Guerrouahen BS, Benali-Furet N, et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res 2011;31:427-441.
    Pubmed
  35. Kim TH, Lim M, Park J, et al. FAST: size-selective, clog-free isolation of rare cancer cells from whole blood at a liquid-liquid interface. Anal Chem 2017;89:1155-1162.
    Pubmed CrossRef
  36. Gogoi P, Sepehri S, Zhou Y, et al. Development of an automated and sensitive microfluidic device for capturing and characterizing circulating tumor cells (CTCs) from clinical blood samples. PLoS One 2016;11:e0147400.
    Pubmed KoreaMed CrossRef
  37. Hodgkinson CL, Morrow CJ, Li Y, et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat Med 2014;20:897-903.
    Pubmed CrossRef
  38. Lim M, Cho YK. Current methods of circulating tumor cell detection. Korean J Helicobacter Up Gastrointest Res 2018;18:157-161.
    CrossRef
  39. Alix-Panabières C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer 2014;14:623-631.
    Pubmed CrossRef
  40. Adams DL, Stefansson S, Haudenschild C, et al. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the CellSearch(®) CTC test. Cytometry A 2015;87:137-144.
    Pubmed CrossRef
  41. Gradilone A, Iacovelli R, Cortesi E, et al. Circulating tumor cells and “suspicious objects” evaluated through CellSearch® in meta-static renal cell carcinoma. Anticancer Res 2011;31:4219-4221.
    Pubmed
  42. Rosenberg R, Gertler R, Friederichs J, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry 2002;49:150-158.
    Pubmed CrossRef
  43. Coumans FA, van Dalum G, Beck M, Terstappen LW. Filter characteristics influencing circulating tumor cell enrichment from whole blood. PLoS One 2013;8:e61770.
    Pubmed KoreaMed CrossRef
  44. Teo J, Mirenska A, Tan M, et al. A preliminary study for the assessment of PD-L1 and PD-L2 on circulating tumor cells by microfluidic-based chipcytometry. Future Sci OA 2017;3:FSO244.
    Pubmed KoreaMed CrossRef
  45. Moon HS, Kwon K, Kim SI, et al. Continuous separation of breast cancer cells from blood samples using multi-orifice flow fractionation (MOFF) and dielectrophoresis (DEP). Lab Chip 2011;11:1118-1125.
    Pubmed CrossRef
  46. Tang L, Zhao S, Liu W, et al. Diagnostic accuracy of circulating tumor cells detection in gastric cancer: systematic review and meta-analysis. BMC Cancer 2013;13:314.
    Pubmed KoreaMed CrossRef
  47. Ghossein RA, Bhattacharya S, Rosai J. Molecular detection of micrometastases and circulating tumor cells in solid tumors. Clin Cancer Res 1999;5:1950-1960.
    Pubmed
  48. Pituch-Noworolska A, Kolodziejczyk P, Kulig J, et al. Circulating tumour cells and survival of patients with gastric cancer. Anticancer Res 2007;27:635-640.
    Pubmed
  49. Hiraiwa K, Takeuchi H, Hasegawa H, et al. Clinical significance of circulating tumor cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol 2008;15:3092-3100.
    Pubmed CrossRef
  50. Matsusaka S, Chìn K, Ogura M, et al. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci 2010;101:1067-1071.
    Pubmed CrossRef
  51. Uenosono Y, Arigami T, Kozono T, et al. Clinical significance of circulating tumor cells in peripheral blood from patients with gastric cancer. Cancer 2013;119:3984-3991.
    Pubmed CrossRef
  52. Okabe H, Tsunoda S, Hosogi H, et al. Circulating tumor cells as an independent predictor of survival in advanced gastric cancer. Ann Surg Oncol 2015;22:3954-3961.
    Pubmed CrossRef
  53. Yuan D, Chen L, Li M, et al. Isolation and characterization of circulating tumor cells from human gastric cancer patients. J Cancer Res Clin Oncol 2015;141:647-660.
    Pubmed CrossRef
  54. Kolostova K, Matkowski R, Gürlich R, et al. Detection and cultivation of circulating tumor cells in gastric cancer. Cytotechnology 2016;68:1095-1102.
    Pubmed KoreaMed CrossRef
  55. Li Y, Gong J, Zhang Q, et al. Dynamic monitoring of circulating tumour cells to evaluate therapeutic efficacy in advanced gastric cancer. Br J Cancer 2016;114:138-145.
    Pubmed KoreaMed CrossRef
  56. Kang HM, Kim GH, Jeon HK, et al. Circulating tumor cells detected by lab-on-a-disc: role in early diagnosis of gastric cancer. PLoS One 2017;12:e0180251.
    Pubmed KoreaMed CrossRef
  57. Liu Y, Ling Y, Qi Q, et al. Prognostic value of circulating tumor cells in advanced gastric cancer patients receiving chemotherapy. Mol Clin Oncol 2017;6:235-242.
    Pubmed KoreaMed CrossRef
  58. Pernot S, Badoual C, Terme M, et al. Dynamic evaluation of circulating tumour cells in patients with advanced gastric and oesogastric junction adenocarcinoma: prognostic value and early assessment of therapeutic effects. Eur J Cancer 2017;79:15-22.
    Pubmed CrossRef
  59. Brungs D, Lynch D, Luk AW, et al. Cryopreservation for delayed circulating tumor cell isolation is a valid strategy for prognostic association of circulating tumor cells in gastroesophageal cancer. World J Gastroenterol 2018;24:810-818.
    Pubmed KoreaMed CrossRef
  60. Yang C, Zhang N, Wang S, et al. Wedge-shaped microfluidic chip for circulating tumor cells isolation and its clinical significance in gastric cancer. J Transl Med 2018;16:139.
    Pubmed KoreaMed CrossRef
  61. Jeon HK, Kim GH. Clinical significance of circulating tumor cells in gastric cancer. Korean J Helicobacter Up Gastrointest Res 2018;18:162-167.
    CrossRef
  62. Huang X, Gao P, Sun J, et al. Clinicopathological and prognostic significance of circulating tumor cells in patients with gastric cancer: a meta-analysis. Int J Cancer 2015;136:21-33.
    Pubmed CrossRef
  63. Parkinson DR, Dracopoli N, Petty BG, et al. Considerations in the development of circulating tumor cell technology for clinical use. J Transl Med 2012;10:138.
    Pubmed KoreaMed CrossRef
  64. Müller V, Stahmann N, Riethdorf S, et al. Circulating tumor cells in breast cancer: correlation to bone marrow micrometastases, heterogeneous response to systemic therapy and low proliferative activity. Clin Cancer Res 2005;11:3678-3685.
    Pubmed CrossRef
  65. Pantel K, Schlimok G, Braun S, et al. Differential expression of proliferation-associated molecules in individual micrometastatic carcinoma cells. J Natl Cancer Inst 1993;85:1419-1424.
    Pubmed CrossRef
  66. Ke Z, Lin M, Chen JF, et al. Programming thermoresponsiveness of NanoVelcro substrates enables effective purification of circulating tumor cells in lung cancer patients. ACS Nano 2015;9:62-70.
    Pubmed KoreaMed CrossRef
  67. Sundaresan TK, Sequist LV, Heymach JV, et al. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res 2016;22:1103-1110.
    Pubmed KoreaMed CrossRef
  68. Yeo T, Tan SJ, Lim CL, et al. Microfluidic enrichment for the single cell analysis of circulating tumor cells. Sci Rep 2016;6:22076.
    Pubmed KoreaMed CrossRef
  69. Iwatsuki M, Toyoshima K, Watanabe M, et al. Frequency of HER2 expression of circulating tumour cells in patients with metastatic or recurrent gastrointestinal cancer. Br J Cancer 2013;109:2829-2832.
    Pubmed KoreaMed CrossRef

Article

Review

Gut and Liver 2019; 13(4): 394-401

Published online February 27, 2019 https://doi.org/10.5009/gnl18484

Copyright © Gut and Liver.

Clinical Application of Circulating Tumor Cells in Gastric Cancer

Moon Won Lee , Gwang Ha Kim , Hye Kyung Jeon , Su Jin Park

Department of Internal Medicine, Pusan National University School of Medicine, and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea

Correspondence to:Gwang Ha Kim
Department of Internal Medicine, Pusan National University School of Medicine, and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan 49241, Korea
Tel: +82-51-240-7869, Fax: +82-51-244-8180, E-mail: doc0224@pusan.ac.kr

Received: October 30, 2018; Revised: November 19, 2018; Accepted: November 23, 2018

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Early detection and accurate monitoring of cancer is important for improving clinical outcomes. Endoscopic biopsy and/or surgical resection specimens are the gold standard for diagnosing gastric cancer and are also useful for selecting therapeutic strategies based on the analysis of genomic/immune parameters. However, these approaches cannot be easily performed because of their invasiveness and because these specimens do not always reflect tumor dynamics and drug sensitivities during therapeutic processes, especially chemotherapy. Accordingly, many researchers have tried to develop noninvasive novel biomarkers that can monitor real-time tumor dynamics for early diagnosis, prognostic evaluation, and prediction of recurrence and therapeutic efficacy. Circulating tumor cells (CTCs) are metastatic cells that are released from the primary tumors into the blood stream and comprise a crucial step in hematogenous metastasis. CTCs, as a liquid biopsy, have received a considerable amount of attention from researchers since they are easily accessible in peripheral blood, avoiding the invasiveness associated with traditional biopsy techniques; they can also be used to derive clinical information for monitoring disease status. In this review, with respect to CTCs, we summarize the metastatic cascade, detection methods, clinical applications, and prospects for patients with gastric cancer.

Keywords: Circulating tumor cells, Liquid biopsy, Neoplasms, Stomach neoplasms

INTRODUCTION

Gastric cancer (GC) is the 6th most prevalent cancer (1,033,701 new cases per year) and the 2nd most common cause of cancer-related deaths worldwide in 2018.1 Despite the development of diagnostic and therapeutic modalities, more than 40% of patients with advanced GC have a poor prognosis and exhibit no response to chemotherapy or resistance to chemotherapy during treatment.2 Approximately 30% to 40% of GC patients experience tumor recurrence and metastasis even after curative resection.3,4 This suggests the presence of potential metastatic cells that cannot be detected based on conventional diagnostic methods.

Traditional tumor markers such as carcinoembryonic antigen (CEA) and cancer antigen 19-9 (CA19-9) are known biomarkers of GC. However, the positivity rate of these markers is less than 40% in GC patients, and the sensitivity and specificity of assays to detect these markers are insufficient for diagnostic and prognostic approaches.5 Generally, the diagnosis of GC is based on endoscopic biopsy and/or surgical resection specimens, but these approaches cannot be easily performed due to their invasiveness. Furthermore, these specimens do not represent the intratumoral heterogeneity and mechanisms of tumor progression and resistance to treatment. Therefore, the development of new modalities that can predict recurrence and metastasis, and which can be used for diagnosis and evaluation of therapeutic response, is important to improve clinical outcomes for GC patients.6

Circulating tumor cells (CTCs) are disseminated tumor cells in the peripheral blood of cancer patients. The first report of CTCs occurred in 1869 from Thomas Ashworth, who described the presence of tumor-derived epithelial cells in the blood compartment of a patient with end-stage metastatic cancer.7 However, because the number of cancer cells in circulation ranges from only approximately around 1–1,000 cells/10 mL,8 conventional methods are not adequate for their detection. Recently, numerous new approaches have been established to isolate, enumerate, and characterize CTCs in patients with various cancers.9 CTCs have strengths such as accessibility and superiority over conventional tumor markers; moreover, based on the analysis of CTCs, researchers can elucidate genetic and phenotypic differences between primary and metastatic tumors. Herein, we summarize the metastatic cascade associated with CTCs, provide an overview of the detection methods for CTCs, and suggest clinical applications for CTCs in GC patients; we then discuss their future use in the clinical practice.

CIRCULATING TUMOR CELLS AND METASTASIS

Cancer metastasis is a multi-step process that includes the loss of intercellular connections, invasion into the basal membrane and surrounding tissues, intravasation into venous or lymphatic vessels, which generates CTCs, survival in the peripheral system, extravasation, and proliferation at secondary sites.1012 Epithelial tumor cells are immobile due to strong cell-to-cell and cell-to-extracellular adhesions, which consist of adherent junctions, tight junctions, and desmosomes. Moreover, conditions in the blood stream are too harsh for the survival of epithelial tumor cells. Therefore, these cells undergo epithelial-to-mesenchymal transition (EMT), which is associated with the loss of adhesion, increased plasticity, and the capacity for migration and invasion. EMT is a molecular and cellular change that includes the down-regulation of epithelial proteins such as E-cadherin, claudins and cytokeratins and the up-regulation of mesenchymal proteins such as N-cadherin, fibronectin and vimentin, which confers increased motility and invasiveness to the cells.1012 EMT promotes the generation of CTCs by increasing tumor cell invasiveness,1315 promoting tumor cell intravasation,16,17 and facilitating tumor cell survival in the peripheral system.18,19

The correlation between EMT and tumor cell invasiveness or CTCs has been confirmed, but the details of the metastatic cascade are still controversial. Two hypotheses have been proposed for the metastatic model associated with EMT: EMT/mesenchymal-epithelial migration (MET) model and collective migration model (Fig. 1).20 In the EMT/MET model, at first, epithelial cancer cells must undergo EMT to become invasive and motile and to generate CTCs; after that, CTCs circulate throughout the entire body and extravasate to distant secondary sites. After extravasation to secondary sites, cancer cells need to recover their epithelial properties via MET, the inverse process of EMT, and then finally colonize at the distant site to complete the metastasis.2124 This metastatic model is the first reported and most widely known model, and also explains why the histological characteristics of the primary tumor are consistent with those of the secondary tumor.22 Furthermore, recent studies have reported a switch between EMT and MET marker phenotypic plasticity in some cancer types.25,26 In the collective migration model, epithelial-like and mesenchymal-like cancer cells can cooperate during collective migration to achieve metastasis instead of undergoing the difficult process of EMT and subsequent MET.27,28 Instead of migrating as a single cell, cancer cells undergoing various degrees of EMT coexist as multi-cellular clusters that migrate collectively. More motile, invasive mesenchymal-like cells aggregate at the invasive front of multi-cellular clusters to facilitate this process, whereas epithelial-like cells follow behind and exploit the opportunity to proliferate and colonize at the distant sites after extravasation.28 Accordingly, multi-cellular cancer fragments, or so-called tumor micro-emboli or CTC clusters, have been observed and correlate with distant metastasis.29,30 Of note, the EMT/MET and collective migration models might not be independent or opposing. Notably, tumor cells might use both mechanisms alternately under certain circumstances, or the two mechanisms could synergistically contribute to metastases.

METHODS TO DETECT CICRULATING TUMOR CELLS

In general, CTCs are rare in the peripheral blood of cancer patients. The enumeration of these CTCs is a great technological challenge. Thus, the enrichment methods require sufficient sensitivity and reproducibility. Current methods for separating CTCs include those based on immunoaffinity differences, using an antibody that binds to a specific protein marker on the surface of cancer cells,3133 and differences in physical properties such as the sizes, densities, and electrical characteristics of cancer cells and blood cells (Fig. 2).3439

Immunoaffinity-based separation is the most widely-used strategy. This method uses specific biomarkers such as epithelial cell adhesion molecule (EpCAM) and CD45 which are expressed on the cell surface, to capture cells. The antibodies used for selection are typically linked to the surface of a device or to a magnetic substance allowing them to be captured by a magnetic field (i.e., immunomagnetic capture). During positive selection, tumor-associated cell surface antigens such as EpCAM are targeted, whereas negative selection removes background cells by antigens not expressed on CTCs (e.g., CD45). Specifically, the CellSearchTM platform (Veridex LLC, Huntingdon Valley, PA, USA) was approved by the U.S. Food and Drug Administration (FDA) for the diagnosis and prognostic evaluation of patients with metastatic breast cancer, colorectal cancer, and prostate cancer between 2004 and 2008.40 However, the expression level of CTCs is extremely variable, and CTCs with low target antibody expression are more difficult to capture.41 Moreover, separation processes for CTCs include complex steps and are time consuming with respect to achieving sufficient reactions between antigens and antibodies. In addition, the heterogeneous array of surface markers expressed by CTCs has made it impossible to identify a universal CTC-specific antigen.

In the density gradient method, blood samples are diluted and laid over the media, and then separated by centrifugation.42 With this, it is easy to remove red blood cells with a high density, but it is difficult to separate CTCs and white blood cells due to their similar densities. Size-based filtration has been used to enrich CTCs based on the fact that they are larger than other blood cells. However, CTCs are generally 8 to 20 μm in size, whereas leukocytes are 6 to 10 μm; thus, leukocytes will be present to some extent in samples treated by the size-based separation methods, which makes it difficult to capture high-purity CTCs.43 In the inertia sorting method, the CTCs are isolated using a spiral channel microfluidic chip, which separates different-sized cells with different Dean forces in a curved channel. With this, it is possible to capture CTCs exhibiting different expression levels of specific surface proteins, and this technique has several advantages such as fast separation time, ease of use, and low cost. However, size-based filtration has the disadvantage of potentially capturing contaminated cells.44

Dielectrophoresis (DEP) is an electrokinetic method that exploits the dielectric properties of floating cells for identification and separation. This method isolates CTCs based on different responses to DEP due to differences in size and membrane properties. Further, it is advantageous for the separation of CTCs because of its simple experimental set-up, much higher operational flow rate compared to other separator methods, and lack of dependence on cell surface markers.45 However, for efficient isolation, whole blood cannot be used directly; there is also limitations associated with the use of a special buffer optimized for electrical conductivity to increase the efficiency of cancer cell isolation.

With advances in technology, next generation microfluidic platforms that enrich CTCs have been developed for cost-effectiveness and high levels of purity. Therefore, if these platforms with high sensitivity and specificity can be further developed, they could be used extensively in clinical practice as well as in the studies on CTCs.

CLINICAL DATA REGARDING CIRCULATING TUMOR CELLS IN PATIENTS WITH GASTRIC CANCER

1. Diagnostic and prognostic value of circulating tumor cells

Based on a meta-analysis of the diagnostic accuracy of various CTC detection methods, the pooled sensitivity and specificity of CTCs for GC were 42% (95% confidence interval [CI], 21% to 67%) and 99% (95% CI, 96% to 100%), respectively;46 the authors of this study concluded that CTC detection has a limited role in screening test for GC, but might be used as a non-invasive method for the confirmation of GC diagnosis. Seventeen of 20 studies included in this meta-analysis used reverse transcriptase-polymerase chain reaction (RT-PCR) for cytokeratin or CEA to detect CTCs and the results varied among different detection methods. RT-PCR is one molecular approach for the detection of CTCs; however, an ideal method should focus on the tumor cells directly rather than indirectly-linked surrogate markers via a molecular approach.47 To date, the CellSearchTM system (Veridex LLC) is the only CTC detection technique approved by the U.S. FDA. Using this platform, it is possible to obtain highly reproducible quantitative results in different laboratories. Therefore, this review will focus further on studies reporting significant data regarding the role of CTCs in GC that were obtained by the cytometric methods including the CellSearchTM system.4860 The results of these studies are summarized in Table 1.61

In our previous study,56 CTCs were measured in 116 GC patients and 31 healthy individuals using a centrifugal microfluidic system with a new fluid-assisted separation technique, specifically based on size-selective CTC isolation.56 When using a CTC level of ≥2 per 7.5 mL of blood, the sensitivity and specificity for differentiating GC patients from healthy controls was 85.3% (95% CI, 81.4% to 87.2%) and 90.3% (95% CI, 75.6% to 97.4%), respectively. In addition, CTCs were detected in more than 80% of early stage (T1 or N0) GC patients. Although CTCs were not associated with any clinicopathologic features such as staging, histologic type, or mucin phenotype, our results suggest that CTCs could be an early diagnostic biomarker for GC. In a recent study, using a novel wedge-shaped microfluidic chip, CTCs were detected in 75% (30/40) of GC patients, whereas CTCs were not detected in 25 healthy donors.60 In this study, CTCs were associated with tumor differentiation, lymphovascular invasion, and staging. These differences in the association between CTCs and clinicopathologic characteristics might be attributed to heterogeneity in the baseline clinicopathologic characteristics and the use of different CTC detection methods.

The measurement of CTCs in GC patients can be useful for predicting patient survival and prognosis. In a prospective study including 251 patients with advanced GC using the CellSearchTM system, CTCs were detected in 16 patients (16/148, 10.8%) in the resection group with a higher relapse rate and in 62 patients (62/103, 60.2%) in the non-resectable group, but were not detected in healthy volunteer. Thus, the evaluation of CTCs might be a useful strategy to predict tumor progression and prognosis in GC patients. Based on a meta-analysis (including 26 studies comprising 2,566 GC patients) evaluating the prognostic significance of CTCs in GC patients, the frequency of CTC detection was higher in advanced GC than early GC, in poorly differentiated GC than well/moderately differentiated GC, and in GC with lymphatic metastasis than that without lymphatic metastasis; it was also significantly related to the disease-free and overall survival of patients.62

In addition to being diagnostic and prognostic indicators, CTCs can be used to monitor chemotherapy response. The measurement of CTCs using peripheral blood is non-invasive and can be performed repeatedly. Thus, periodically monitoring CTCs could be helpful to predict the efficacy of chemotherapeutics. Although CEA and CA19-9 are frequently used as markers in GC, they are not appropriate for this application because temporary increases can be detected after chemotherapy and many patients with advanced GC have normal levels of CEA and CA19-9. Matsusaka et al.50 measured CTC levels at baseline, 2 weeks, and 4 weeks in 52 patients with advanced GC undergoing S-1-based chemotherapy. Patients with ≥4 CTCs at 2- and 4-weeks post-chemotherapy had a shorter median progression-free survival (PFS) and overall survival (OS).50 In another study measuring CTC levels at baseline and after 6 weeks of chemotherapy in 136 patients with advanced GC, patients with ≥3 CTCs after chemotherapy had a shorter PFS and OS; moreover, an elevated number of CTCs after chemotherapy corresponded to ineffective therapeutic responses.55 Therefore, monitoring dynamic changes in CTCs in response to therapy might be a useful alternative for assessing treatment resistance in GC patients.

2. Value of circulating tumor cells for selecting therapeutic agents

Overcoming tumor heterogeneity is a major challenge for the personalized treatment of GC. Therefore, the further characterization of CTCs might help to identify specific morphological, phenotypic, and molecular features of different cancer types based on time, disease stage, and therapeutic definitions.63 As stated earlier, CTCs that have undergone the EMT exhibit enhanced cell motility rather than growth, resulting in the ability of cancer cells to exit the vasculature. In this situation, most CTCs are not responsive to cytotoxic chemotherapy because they do not undergo cell division.64,65 Therefore, the addition of CTC-targeted treatment to conventional chemotherapy might be helpful to improve prognosis for cancer patients. In non-small-cell lung cancers, a therapeutic agent targeting an epidermal growth factor receptor (EGFR) mutation has been developed, therefore, it is important to also detect this EGFR mutation in CTCs. Currently, there are several clinical reports suggesting that detection rate of the EGFR mutation in the CTCs of patients with non-small-cell lung cancer is comparable to that in tissues, confirming a high rate of concordance.6668

For GC, trastuzumab, which bind the human epidermal growth factor receptor 2 (HER2) gene, and ramucirumab, which bind vascular endothelial growth factor receptor-2, have been developed. Especially, the overexpression of HER2 is a selective biomarker for treatment with the monoclonal antibody bevacizumab in metastatic GC. Recently, Iwatsuki et al.69 evaluated CTCs and their HER2 status in patients with gastrointestinal cancer. Among the 62 CTC-positive cases, 22 (35.5%) showed discordance in terms of HER2 status between primary tumors and CTCs. Among the HER2-negative primary tumors, 17 of 54 developed HER2-positive CTCs, and among the HER2-positive primary tumors, five of eight were associated with HER2-negative CTCs. These results suggest that for primary HER2-negative tumors, CTCs could acquire HER2 gene amplification during cancer progression, and therefore, it might be necessary to determine the HER2 status of CTCs, as a liquid biopsy, to suggest personalized treatment strategies for GC. Thus, it is expected that CTCs derived from GC patients could be used to predict responses to targeted therapy. These CTCs require precise medicine, which will not only help to select the appropriate chemotherapy but will also allow for the periodic monitoring of mutations associated with resistance.

FUTURE DIRECTIONS

For the use of CTCs to become a clinical analytical method that complements tissue biopsies, there are many issues that need to be solved. Regarding the CellSearchTM system, cells that are positive for EpCAM are isolated, which can be considered a false negative for CTCs undergoing EMT. Therefore, it is necessary to find markers to accurately detect CTCs with an EMT phenotype. In addition, it is difficult to directly compare the sensitivity, specificity, and reproducibility of CTC separation methods because the types and preparations of various CTC isolation methods currently under development are different and have not been standardized. Therefore, it is important to develop a standardized isolation method to establish appropriate criteria for the use of CTCs as diagnostic and prognostic indicators of GC, and to confirm their clinical significance through large scale prospective clinical studies.

CONCLUSIONS

Technology associated with liquid biopsy was globally recognized as one of the world top 10 technologies in 2017 and comprises a highly competitive field for the development of methods that enable the most simple and accurate diagnosis of cancer. Recently, several studies have shown that peripheral blood CTCs are useful to predict prognosis and monitor therapy in GC patients. Future CTC studies are expected to provide additional information to identify mechanisms that are important for the proliferation and metastasis of malignant tumors. Furthermore, these studies are expected to play a major role in precision medicine by uncovering molecular biologic targets related to drug sensitivity and resistance in GC patients.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

ACKNOWLEDGEMENTS

This work was supported by a Busan Cancer Center Research Grant (2018), Pusan National University Hospital.

Fig 1.

Figure 1.Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) in the emergence and progression of carcinoma. Adapted from Jie XX, et al. Oncotarget 2017;8:81558–81571.
Gut and Liver 2019; 13: 394-401https://doi.org/10.5009/gnl18484

Fig 2.

Figure 2.Enrichment technologies for circulating tumor cells (CTCs). A: Immunoaffinity method using immunomagnetic particles that bind to the CTC surface antigen. B: Density gradient method separating other blood cells from CTCs based on their density. C: Dielectrophoresis (DEP) utilizing the dielectric properties of CTCs under an electric field. D: Size-based filtration resulting in enrichment based on the larger size of CTCs compared with the sizes of other cells. E: Inertia sorting method using Dean force at microfluidic channels. Adapted from Alix-Panabières C and Pantel K. Nat Rev Cancer 2014;14:623–631.
Gut and Liver 2019; 13: 394-401https://doi.org/10.5009/gnl18484

Table 1 Summary of Studies on Circulating Tumor Cells Using Cytometric Methods in Patients with Gastric Cancer

YearAuthorNo. of patientsDetection methodsMarkersPositive definitionCTC detection, No. (%)OutcomesClinical significance
2007Pituch-Noworolska et al.4857FACSCK8,18,19≥3 CTCs31 (54)OSNo
2008Hiraiwa et al.49Non-metastatic, 14CellSearchEpCAM≥2 CTCs2 (14)OSYes
Metastatic, 27CK8,18,1915 (56)Monitoring
2010Matsusaka et al.50Baseline, 52CellSearchEpCAM≥4 CTCs17 (33)OS, PFSYes
2-wk, 51CK8,18,197 (14)Monitoring
4-wk, 489 (19)
2013Uenosono et al.51Resection, 148CellSearchEpCAM≥1 CTC16 (11)OS, RFSYes
Non-resectable, 103CK8,18,1962 (60)
2015Okabe et al.52136CellSearchEpCAM≥1 CTC25 (18)PFSYes
CK8,18,19
2015Yuan et al.5331FACSCD44+ CD45−-14 (45)--
2016Kolostova et al.5422MetaCellCK7,18,19, 20-13 (59)--
EpCAM, MUC1, HER2, EGFR
2016Li et al.55Baseline, 136CellSearchEpCAM≥3 CTCs57 (42)OS, PFSYes
6-wk, 106CK8,18,1926 (25)Monitoring
2017Kang et al.56116FASTEpCAM/CK≥2 CTCs99 (85)--
2017Liu et al.5759CELLectionEpCAM>2 CTCs36 (61)OS, PFSYes
CK7,8,18,19Monitoring
2017Pernot et al.58Baseline, 106CellSearchEpCAM≥2 CTCs49 (46)OS, PFSYes
4-wk, 65CK8,18,1912 (18)Monitoring
2018Brungs et al.5943IsoFluxEpCAM≥17 CTCs20 (47)OSYes
platformMonitoring
2018Yang et al.6040Microfluidic chipCK≥1 CTC20 (75)-Yes

CTC, circulating tumor cell; FACS, fluorescence-activated cell sorting; CK, cytokeratin; OS, overall survival; EpCAM, epithelial cell adhesion molecule; PFS, progression-free survival; RFS, recurrence-free survival; HER2, human epidermal growth factor receptor 2; EGFR, epidermal growth factor receptor; FAST, fluid-assisted separation technique.

Adapted from Jeon HK and Kim GH. Korean J Helicobacter Up Gastrointest Res 2018;18:162–167.61


References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424.
    Pubmed CrossRef
  2. Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric cancer. Lancet 2009;374:477-490.
    Pubmed KoreaMed CrossRef
  3. Lim DH, Kim DY, Kang MK, et al. Patterns of failure in gastric carcinoma after D2 gastrectomy and chemoradiotherapy: a radiation oncologist’s view. Br J Cancer 2004;91:11-17.
    Pubmed KoreaMed CrossRef
  4. Marrelli D, De Stefano A, de Manzoni G, Morgagni P, Di Leo A, Roviello F. Prediction of recurrence after radical surgery for gastric cancer: a scoring system obtained from a prospective multicenter study. Ann Surg 2005;241:247-255.
    Pubmed KoreaMed CrossRef
  5. Li Y, Yang Y, Lu M, Shen L. Predictive value of serum CEA, CA19-9 and CA72.4 in early diagnosis of recurrence after radical resection of gastric cancer. Hepatogastroenterology 2011;58:2166-2170.
    Pubmed CrossRef
  6. Kang EA, Han YM, Park JM, Yoo IK, Hong SP, Hahm KB. Liquid biopsy: current status and future perspective in gastric cancer and helicobacter infection. Korean J Helicobacter Up Gastrointest Res 2018;18:150-156.
    CrossRef
  7. Ashworth T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J 1869;14:146.
  8. Nieva JJ, Kuhn P. Fluid biopsy for solid tumors: a patient’s companion for lifelong characterization of their disease. Future Oncol 2012;8:989-998.
    Pubmed KoreaMed CrossRef
  9. Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer 2004;4:448-456.
    Pubmed CrossRef
  10. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-1428.
    Pubmed KoreaMed CrossRef
  11. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2:442-454.
    Pubmed CrossRef
  12. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009;139:871-890.
    Pubmed CrossRef
  13. Liberko M, Kolostova K, Bobek V. Essentials of circulating tumor cells for clinical research and practice. Crit Rev Oncol Hematol 2013;88:338-356.
    Pubmed CrossRef
  14. Derksen PW, Liu X, Saridin F, et al. Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 2006;10:437-449.
    Pubmed CrossRef
  15. Hulit J, Suyama K, Chung S, et al. N-cadherin signaling potentiates mammary tumor metastasis via enhanced extracellular signal-regulated kinase activation. Cancer Res 2007;67:3106-3116.
    Pubmed CrossRef
  16. Qi J, Wang J, Romanyuk O, Siu CH. Involvement of Src family kinases in N-cadherin phosphorylation and beta-catenin dissociation during transendothelial migration of melanoma cells. Mol Biol Cell 2006;17:1261-1272.
    Pubmed KoreaMed CrossRef
  17. Drake JM, Strohbehn G, Bair TB, Moreland JG, Henry MD. ZEB1 enhances transendothelial migration and represses the epithelial phenotype of prostate cancer cells. Mol Biol Cell 2009;20:2207-2217.
    Pubmed KoreaMed CrossRef
  18. Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 2010;29:4741-4751.
    Pubmed KoreaMed CrossRef
  19. Tiwari N, Gheldof A, Tatari M, Christofori G. EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012;22:194-207.
    Pubmed CrossRef
  20. Jie XX, Zhang XY, Xu CJ. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: mechanisms and clinical applications. Oncotarget 2017;8:81558-81571.
    Pubmed KoreaMed CrossRef
  21. Hugo H, Ackland ML, Blick T, et al. Epithelial: mesenchymal and mesenchymal. Epithelial transitions in carcinoma progression. J Cell Physiol 2007;213:374-383.
    Pubmed CrossRef
  22. Chaffer CL, Thompson EW, Williams ED. Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs 2007;185:7-19.
    Pubmed CrossRef
  23. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science 2011;331:1559-1564.
    Pubmed CrossRef
  24. Banyard J, Bielenberg DR. The role of EMT and MET in cancer dissemination. Connect Tissue Res 2015;56:403-413.
    Pubmed KoreaMed CrossRef
  25. Brabletz T, Jung A, Reu S, et al. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci U S A 2001;98:10356-10361.
    Pubmed KoreaMed CrossRef
  26. Hudson LG, Zeineldin R, Stack MS. Phenotypic plasticity of neoplastic ovarian epithelium: unique cadherin profiles in tumor progression. Clin Exp Metastasis 2008;25:643-655.
    Pubmed KoreaMed CrossRef
  27. Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer 2003;3:362-374.
    Pubmed CrossRef
  28. Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol 2009;10:445-457.
    Pubmed CrossRef
  29. Chui MH. Insights into cancer metastasis from a clinicopathologic perspective: epithelial-mesenchymal transition is not a necessary step. Int J Cancer 2013;132:1487-1495.
    Pubmed CrossRef
  30. Wang C, Mu Z, Chervoneva I, et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res Treat 2017;161:83-94.
    Pubmed CrossRef
  31. Riethdorf S, Fritsche H, Müller V, 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 2007;13:920-928.
    Pubmed CrossRef
  32. Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol 2010;2010 617421.
    Pubmed KoreaMed CrossRef
  33. Harb W, Fan A, Tran T, et al. Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay. Transl Oncol 2013;6:528-538.
    Pubmed KoreaMed CrossRef
  34. Desitter I, Guerrouahen BS, Benali-Furet N, et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res 2011;31:427-441.
    Pubmed
  35. Kim TH, Lim M, Park J, et al. FAST: size-selective, clog-free isolation of rare cancer cells from whole blood at a liquid-liquid interface. Anal Chem 2017;89:1155-1162.
    Pubmed CrossRef
  36. Gogoi P, Sepehri S, Zhou Y, et al. Development of an automated and sensitive microfluidic device for capturing and characterizing circulating tumor cells (CTCs) from clinical blood samples. PLoS One 2016;11:e0147400.
    Pubmed KoreaMed CrossRef
  37. Hodgkinson CL, Morrow CJ, Li Y, et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat Med 2014;20:897-903.
    Pubmed CrossRef
  38. Lim M, Cho YK. Current methods of circulating tumor cell detection. Korean J Helicobacter Up Gastrointest Res 2018;18:157-161.
    CrossRef
  39. Alix-Panabières C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer 2014;14:623-631.
    Pubmed CrossRef
  40. Adams DL, Stefansson S, Haudenschild C, et al. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the CellSearch(®) CTC test. Cytometry A 2015;87:137-144.
    Pubmed CrossRef
  41. Gradilone A, Iacovelli R, Cortesi E, et al. Circulating tumor cells and “suspicious objects” evaluated through CellSearch® in meta-static renal cell carcinoma. Anticancer Res 2011;31:4219-4221.
    Pubmed
  42. Rosenberg R, Gertler R, Friederichs J, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry 2002;49:150-158.
    Pubmed CrossRef
  43. Coumans FA, van Dalum G, Beck M, Terstappen LW. Filter characteristics influencing circulating tumor cell enrichment from whole blood. PLoS One 2013;8:e61770.
    Pubmed KoreaMed CrossRef
  44. Teo J, Mirenska A, Tan M, et al. A preliminary study for the assessment of PD-L1 and PD-L2 on circulating tumor cells by microfluidic-based chipcytometry. Future Sci OA 2017;3:FSO244.
    Pubmed KoreaMed CrossRef
  45. Moon HS, Kwon K, Kim SI, et al. Continuous separation of breast cancer cells from blood samples using multi-orifice flow fractionation (MOFF) and dielectrophoresis (DEP). Lab Chip 2011;11:1118-1125.
    Pubmed CrossRef
  46. Tang L, Zhao S, Liu W, et al. Diagnostic accuracy of circulating tumor cells detection in gastric cancer: systematic review and meta-analysis. BMC Cancer 2013;13:314.
    Pubmed KoreaMed CrossRef
  47. Ghossein RA, Bhattacharya S, Rosai J. Molecular detection of micrometastases and circulating tumor cells in solid tumors. Clin Cancer Res 1999;5:1950-1960.
    Pubmed
  48. Pituch-Noworolska A, Kolodziejczyk P, Kulig J, et al. Circulating tumour cells and survival of patients with gastric cancer. Anticancer Res 2007;27:635-640.
    Pubmed
  49. Hiraiwa K, Takeuchi H, Hasegawa H, et al. Clinical significance of circulating tumor cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol 2008;15:3092-3100.
    Pubmed CrossRef
  50. Matsusaka S, Chìn K, Ogura M, et al. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci 2010;101:1067-1071.
    Pubmed CrossRef
  51. Uenosono Y, Arigami T, Kozono T, et al. Clinical significance of circulating tumor cells in peripheral blood from patients with gastric cancer. Cancer 2013;119:3984-3991.
    Pubmed CrossRef
  52. Okabe H, Tsunoda S, Hosogi H, et al. Circulating tumor cells as an independent predictor of survival in advanced gastric cancer. Ann Surg Oncol 2015;22:3954-3961.
    Pubmed CrossRef
  53. Yuan D, Chen L, Li M, et al. Isolation and characterization of circulating tumor cells from human gastric cancer patients. J Cancer Res Clin Oncol 2015;141:647-660.
    Pubmed CrossRef
  54. Kolostova K, Matkowski R, Gürlich R, et al. Detection and cultivation of circulating tumor cells in gastric cancer. Cytotechnology 2016;68:1095-1102.
    Pubmed KoreaMed CrossRef
  55. Li Y, Gong J, Zhang Q, et al. Dynamic monitoring of circulating tumour cells to evaluate therapeutic efficacy in advanced gastric cancer. Br J Cancer 2016;114:138-145.
    Pubmed KoreaMed CrossRef
  56. Kang HM, Kim GH, Jeon HK, et al. Circulating tumor cells detected by lab-on-a-disc: role in early diagnosis of gastric cancer. PLoS One 2017;12:e0180251.
    Pubmed KoreaMed CrossRef
  57. Liu Y, Ling Y, Qi Q, et al. Prognostic value of circulating tumor cells in advanced gastric cancer patients receiving chemotherapy. Mol Clin Oncol 2017;6:235-242.
    Pubmed KoreaMed CrossRef
  58. Pernot S, Badoual C, Terme M, et al. Dynamic evaluation of circulating tumour cells in patients with advanced gastric and oesogastric junction adenocarcinoma: prognostic value and early assessment of therapeutic effects. Eur J Cancer 2017;79:15-22.
    Pubmed CrossRef
  59. Brungs D, Lynch D, Luk AW, et al. Cryopreservation for delayed circulating tumor cell isolation is a valid strategy for prognostic association of circulating tumor cells in gastroesophageal cancer. World J Gastroenterol 2018;24:810-818.
    Pubmed KoreaMed CrossRef
  60. Yang C, Zhang N, Wang S, et al. Wedge-shaped microfluidic chip for circulating tumor cells isolation and its clinical significance in gastric cancer. J Transl Med 2018;16:139.
    Pubmed KoreaMed CrossRef
  61. Jeon HK, Kim GH. Clinical significance of circulating tumor cells in gastric cancer. Korean J Helicobacter Up Gastrointest Res 2018;18:162-167.
    CrossRef
  62. Huang X, Gao P, Sun J, et al. Clinicopathological and prognostic significance of circulating tumor cells in patients with gastric cancer: a meta-analysis. Int J Cancer 2015;136:21-33.
    Pubmed CrossRef
  63. Parkinson DR, Dracopoli N, Petty BG, et al. Considerations in the development of circulating tumor cell technology for clinical use. J Transl Med 2012;10:138.
    Pubmed KoreaMed CrossRef
  64. Müller V, Stahmann N, Riethdorf S, et al. Circulating tumor cells in breast cancer: correlation to bone marrow micrometastases, heterogeneous response to systemic therapy and low proliferative activity. Clin Cancer Res 2005;11:3678-3685.
    Pubmed CrossRef
  65. Pantel K, Schlimok G, Braun S, et al. Differential expression of proliferation-associated molecules in individual micrometastatic carcinoma cells. J Natl Cancer Inst 1993;85:1419-1424.
    Pubmed CrossRef
  66. Ke Z, Lin M, Chen JF, et al. Programming thermoresponsiveness of NanoVelcro substrates enables effective purification of circulating tumor cells in lung cancer patients. ACS Nano 2015;9:62-70.
    Pubmed KoreaMed CrossRef
  67. Sundaresan TK, Sequist LV, Heymach JV, et al. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res 2016;22:1103-1110.
    Pubmed KoreaMed CrossRef
  68. Yeo T, Tan SJ, Lim CL, et al. Microfluidic enrichment for the single cell analysis of circulating tumor cells. Sci Rep 2016;6:22076.
    Pubmed KoreaMed CrossRef
  69. Iwatsuki M, Toyoshima K, Watanabe M, et al. Frequency of HER2 expression of circulating tumour cells in patients with metastatic or recurrent gastrointestinal cancer. Br J Cancer 2013;109:2829-2832.
    Pubmed KoreaMed CrossRef
Gut and Liver

Vol.18 No.5
September, 2024

pISSN 1976-2283
eISSN 2005-1212

qrcode
qrcode

Share this article on :

  • line

Popular Keywords

Gut and LiverQR code Download
qr-code

Editorial Office