Circulating tumor cells detected by lab-on-a-disc: Role in early diagnosis of gastric cancer

Hwa Mi Kang, Gwang Ha Kim, Hye Kyung Jeon, Dae Hwan Kim, Tae Yong Jeon, Do Youn Park, Hyunjin Jeong, Won Joo Chun, Mi-Hyun Kim, Juhee Park, Minji Lim, Tae-Hyeong Kim, Yoon-Kyung Cho, Hwa Mi Kang, Gwang Ha Kim, Hye Kyung Jeon, Dae Hwan Kim, Tae Yong Jeon, Do Youn Park, Hyunjin Jeong, Won Joo Chun, Mi-Hyun Kim, Juhee Park, Minji Lim, Tae-Hyeong Kim, Yoon-Kyung Cho

Abstract

Background: The use of circulating tumor cells (CTCs) as an early diagnostic biomarker and prognostic indicator after surgery or chemotherapy has been suggested for various cancers. This study aimed to evaluate CTCs in patients who underwent gastrectomy for gastric cancer and to explore their clinical usefulness in the early diagnosis of gastric cancer.

Methods: A total of 116 patients with gastric cancer who underwent gastrectomy and 31 healthy volunteers were prospectively included between 2014 and 2015. Peripheral blood samples were collected before gastrectomy, and CTCs were examined using a centrifugal microfluidic system with a new fluid-assisted separation technique.

Results: After creating a receiver operating characteristic curve to identify the discriminative CTC value needed differentiate patients with gastric cancer from healthy volunteers, sensitivity and specificity were nearly optimized at a CTC threshold of 2 per 7.5 mL of blood. Of the 102 persons with a CTC level ≥2 per 7.5 mL of blood, 99 (97.1%) had gastric cancer, and of the 45 persons with a CTC level <2 per 7.5 mL of blood, 28 (62.2%) were healthy controls. Accordingly, the sensitivity and specificity for the differentiation of patients with gastric cancer from healthy controls were 85.3% and 90.3%, respectively. However, the presence of CTCs was not associated with any clinicopathologic features such as staging, histologic type, or mucin phenotype.

Conclusion: Although we could not prove the clinical feasibility of CTCs for gastric cancer staging, our results suggest a potential role of CTCs as an early diagnostic biomarker of gastric cancer.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Circulating tumor cells (CTCs).
Fig 1. Circulating tumor cells (CTCs).
CTCs were defined as captured cells that were CK+ or EpCAM+, CD45-, and DAPI+ and had a diameter >8 μm.
Fig 2. Circulating tumor cells (CTCs) in…
Fig 2. Circulating tumor cells (CTCs) in healthy controls and patients with gastric cancer.
CTCs were identified in 3 of 31 healthy controls and 105 of 116 patients with gastric cancer.
Fig 3. Receiver operating characteristic (ROC) curve.
Fig 3. Receiver operating characteristic (ROC) curve.
The value of circulating tumor cells (CTCs) showed an area under the ROC curve of 0.928 (95% confidence interval: 0.885–0.971, P < 0.001). To identify the optimal CTC threshold value for differentiating patients with gastric cancer from healthy controls, the sensitivity and specificity were optimized using a threshold count of 2 CTCs per 7.5 mL of blood.

References

    1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86. doi: .
    1. Digklia A, Wagner AD. Advanced gastric cancer: Current treatment landscape and future perspectives. World J Gastroenterol. 2016;22(8):2403–14. doi: ;.
    1. Correia M, Machado JC, Ristimaki A. Basic aspects of gastric cancer. Helicobacter. 2009;14 Suppl 1:36–40. doi: .
    1. Yeh KH, Chen YC, Yeh SH, Chen CP, Lin JT, Cheng AL. Detection of circulating cancer cells by nested reverse transcription-polymerase chain reaction of cytokeratin-19 (K19)—possible clinical significance in advanced gastric cancer. Anticancer Res. 1998;18(2B):1283–6. .
    1. Uenosono Y, Arigami T, Kozono T, Yanagita S, Hagihara T, Haraguchi N, et al. Clinical significance of circulating tumor cells in peripheral blood from patients with gastric cancer. Cancer. 2013;119(22):3984–91. doi: .
    1. Kubisch I, de Albuquerque A, Schuppan D, Kaul S, Schaich M, Stolzel U. Prognostic Role of a Multimarker Analysis of Circulating Tumor Cells in Advanced Gastric and Gastroesophageal Adenocarcinomas. Oncology. 2015;89(5):294–303. doi: .
    1. Bertazza L, Mocellin S, Marchet A, Pilati P, Gabrieli J, Scalerta R, et al. Survivin gene levels in the peripheral blood of patients with gastric cancer independently predict survival. J Transl Med. 2009;7:111 doi: ;.
    1. Ghossein RA, Bhattacharya S, Rosai J. Molecular detection of micrometastases and circulating tumor cells in solid tumors. Clin Cancer Res. 1999;5(8):1950–60. .
    1. Mimori K, Fukagawa T, Kosaka Y, Ishikawa K, Iwatsuki M, Yokobori T, et al. A large-scale study of MT1-MMP as a marker for isolated tumor cells in peripheral blood and bone marrow in gastric cancer cases. Ann Surg Oncol. 2008;15(10):2934–42. doi: .
    1. Kim TH, Lim M, Park J, Oh JM, Kim H, Jeong H, et al. FAST: Size-Selective, Clog-Free Isolation of Rare Cancer Cells from Whole Blood at a Liquid-Liquid Interface. Anal Chem. 2017; 89(2):1155–62. doi: .
    1. Lee A, Park J, Lim M, Sunkara V, Kim SY, Kim GH, et al. All-in-one centrifugal microfluidic device for size-selective circulating tumor cell isolation with high purity. Anal Chem. 2014;86(22):11349–56. doi: .
    1. Mimori K, Fukagawa T, Kosaka Y, Kita Y, Ishikawa K, Etoh T, et al. Hematogenous metastasis in gastric cancer requires isolated tumor cells and expression of vascular endothelial growth factor receptor-1. Clin Cancer Res. 2008;14(9):2609–16. doi: .
    1. Wu CH, Lin SR, Hsieh JS, Chen FM, Lu CY, Yu FJ, et al. Molecular detection of disseminated tumor cells in the peripheral blood of patients with gastric cancer: evaluation of their prognostic significance. Dis Markers. 2006;22(3):103–9. ;. doi:
    1. Lee HJ, Kim GH, Park do Y, Lee BE, Jeon HK, Jhi JH, et al. Is endoscopic submucosal dissection safe for papillary adenocarcinoma of the stomach? World J Gastroenterol. 2015;21(13):3944–52. doi: ;.
    1. Japanese Gastric Cancer A. Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer. 2011;14(2):101–12. doi: .
    1. Edge SB, Byrd DR, Compton CC. AJCC cancer staging handbook, 7th ed New York: Springer-Verlag; 2010.
    1. Park DY, Srivastava A, Kim GH, Mino-Kenudson M, Deshpande V, Zukerberg LR, et al. Adenomatous and foveolar gastric dysplasia: distinct patterns of mucin expression and background intestinal metaplasia. Am J Surg Pathol. 2008;32(4):524–33. doi: .
    1. Ha Kim G, Am Song G, Youn Park D, Han Lee S, Hyun Lee D, Oh Kim T, et al. CDX2 expression is increased in gastric cancers with less invasiveness and intestinal mucin phenotype. Scand J Gastroenterol. 2006;41(8):880–6. doi: .
    1. Kim DH, Shin N, Kim GH, Song GA, Jeon TY, Kim DH, et al. Mucin expression in gastric cancer: reappraisal of its clinicopathologic and prognostic significance. Arch Pathol Lab Med. 2013;137(8):1047–53. doi: .
    1. Huang X, Gao P, Sun J, Chen X, Song Y, Zhao J, et al. Clinicopathological and prognostic significance of circulating tumor cells in patients with gastric cancer: a meta-analysis. Int J Cancer. 2015;136(1):21–33. doi: .
    1. Witzig TE, Bossy B, Kimlinger T, Roche PC, Ingle JN, Grant C, et al. Detection of circulating cytokeratin-positive cells in the blood of breast cancer patients using immunomagnetic enrichment and digital microscopy. Clin Cancer Res. 2002;8(5):1085–91. .
    1. Beeharry MK, Liu WT, Yan M, Zhu ZG. New blood markers detection technology: A leap in the diagnosis of gastric cancer. World J Gastroenterol. 2016;22(3):1202–12. doi: ;.
    1. Farace F, Massard C, Vimond N, Drusch F, Jacques N, Billiot F, et al. A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas. Br J Cancer. 2011;105(6):847–53. doi: ;.
    1. Hosokawa M, Yoshikawa T, Negishi R, Yoshino T, Koh Y, Kenmotsu H, et al. Microcavity array system for size-based enrichment of circulating tumor cells from the blood of patients with small-cell lung cancer. Anal Chem. 2013;85(12):5692–8. doi: .
    1. Tang Y, Shi J, Li S, Wang L, Cayre YE, Chen Y. Microfluidic device with integrated microfilter of conical-shaped holes for high efficiency and high purity capture of circulating tumor cells. Sci Rep. 2014;4:6052 doi: .
    1. Coumans FA, van Dalum G, Beck M, Terstappen LW. Filtration parameters influencing circulating tumor cell enrichment from whole blood. PLoS One. 2013;8(4):e61774 doi: ;.
    1. Zhou MD, Hao S, Williams AJ, Harouaka RA, Schrand B, Rawal S, et al. Separable bilayer microfiltration device for viable label-free enrichment of circulating tumour cells. Sci Rep. 2014;4:7392 doi: ;.
    1. Adams DL, Zhu P, Makarova OV, Martin SS, Charpentier M, Chumsri S, et al. The systematic study of circulating tumor cell isolation using lithographic microfilters. RSC Adv. 2014;9:4334–42. doi: ;.
    1. Sarioglu AF, Aceto N, Kojic N, Donaldson MC, Zeinali M, Hamza B, et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat Methods. 2015;12(7):685–91. doi: ;.
    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10(20):6897–904. doi: .
    1. Hiraiwa K, Takeuchi H, Hasegawa H, Saikawa Y, Suda K, Ando T, et al. Clinical significance of circulating tumor cells in blood from patients with gastrointestinal cancers. Ann Surg Oncol. 2008;15(11):3092–100. doi: .
    1. Baccelli I, Schneeweiss A, Riethdorf S, Stenzinger A, Schillert A, Vogel V, et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat Biotechnol. 2013;31(6):539–44. doi: .
    1. Li M, Zhang B, Zhang Z, Liu X, Qi X, Zhao J, et al. Stem cell-like circulating tumor cells indicate poor prognosis in gastric cancer. Biomed Res Int. 2014;2014:981261 doi: ;.
    1. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15. doi: ;.
    1. Joosse SA, Pantel K. Biologic challenges in the detection of circulating tumor cells. Cancer Res. 2013;73(1):8–11. doi: .

Source: PubMed

3
Abonner