DOI QR코드

DOI QR Code

Clinical Significance of Circulating Tumor Cells in Gastric Cancer

위암에서 순환종양세포의 임상적 의의

  • Jeon, Hye Kyung (Department of Internal Medicine, Pusan National University School of Medicine) ;
  • Kim, Gwang Ha (Department of Internal Medicine, Pusan National University School of Medicine)
  • 전혜경 (부산대학교 의과대학 내과학교실) ;
  • 김광하 (부산대학교 의과대학 내과학교실)
  • Received : 2018.02.05
  • Accepted : 2018.03.22
  • Published : 2018.09.30

Abstract

Cancer specimens obtained via surgical resection or biopsy are generally used to understand tumor-associated alterations; however, those approaches cannot always be performed because of their invasive nature, and they may fail to reflect current tumor dynamics and drug sensitivity, which may change during the therapeutic process. Therefore, many research groups have focused on developing a non-invasive biomarker with the ability to monitor tumor dynamics. Circulating tumor cells (CTCs) are metastatic cells released from the primary tumor into the bloodstream. Hematogenous spreading of CTCs is a crucial step in the metastatic cascade, which leads to the formation of overt metastases. CTCs have attracted considerable attention because of their easy accessibility and their superiority over conventional tumor markers. Detecting CTCs is considered a valuable modality to determine prognosis and monitor response to systemic therapies in patients with gastric cancer. Moreover, molecular analyses of CTCs may provide important biological information for individual patients with cancer, which may lead to the development of personalized cancer treatment. In this article, we review potential roles and clinical applications of CTCs in patients with gastric cancer.

Keywords

Acknowledgement

Supported by : Pusan National University Hospital

References

  1. Jung KW, Won YJ, Oh CM, Kong HJ, Lee DH, Lee KH. Cancer Statistics in Korea: incidence, mortality, survival, and prevalence in 2014. Cancer Res Treat 2017;49:292-305. https://doi.org/10.4143/crt.2017.118
  2. Nam SY, Choi IJ, Park KW, et al. Effect of repeated endoscopic screening on the incidence and treatment of gastric cancer in health screenees. Eur J Gastroenterol Hepatol 2009;21:855-860. https://doi.org/10.1097/MEG.0b013e328318ed42
  3. National Cancer Information Center. Common cancer sites in Korea [Internet]. Goyang: National Cancer Information Center, [cited 2018 May 18]. Available from: https://www.cancer.go.kr/lay1/S1T648C652/contents.do.
  4. Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric cancer. Lancet 2009;374:477-490. https://doi.org/10.1016/S0140-6736(09)60617-6
  5. 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. https://doi.org/10.1038/sj.bjc.6601896
  6. 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. https://doi.org/10.1097/01.sla.0000152019.14741.97
  7. Onuigbo WI. An index of the fate of circulating cancer cells. Lancet 1963;2:828-831.
  8. Robinson KP, McGrath R, McGrew E. Circulating cancer cells in patients with lung tumors. Surgery 1963;53:630-636.
  9. Ashworth T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aus Med J 1869;14:146-149.
  10. Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev 1989;8:98-101.
  11. Fidler IJ. Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Res 1978;38:2651-2660.
  12. Riethdorf S, Wikman H, Pantel K. Review: biological relevance of disseminated tumor cells in cancer patients. Int J Cancer 2008;123:1991-2006. https://doi.org/10.1002/ijc.23825
  13. Pantel K, Alix-Panabieres C. Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med 2010;16:398-406. https://doi.org/10.1016/j.molmed.2010.07.001
  14. Bernards R, Weinberg RA. A progression puzzle. Nature 2002;418:823. https://doi.org/10.1038/418823a
  15. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2:442-454. https://doi.org/10.1038/nrc822
  16. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-1428. https://doi.org/10.1172/JCI39104
  17. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science 2011;331:1559-1564. https://doi.org/10.1126/science.1203543
  18. Allard WJ, Matera J, Miller MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res 2004;10:6897-6904. https://doi.org/10.1158/1078-0432.CCR-04-0378
  19. Bertazza L, Mocellin S, Marchet A, et al. Survivin gene levels in the peripheral blood of patients with gastric cancer independently predict survival. J Transl Med 2009;7:111. https://doi.org/10.1186/1479-5876-7-111
  20. Yie SM, Lou B, Ye SR, et al. Detection of survivin-expressing circulating cancer cells (CCCs) in peripheral blood of patients with gastric and colorectal cancer reveals high risks of relapse. Ann Surg Oncol 2008;15:3073-3082. https://doi.org/10.1245/s10434-008-0069-x
  21. Andree KC, van Dalum G, Terstappen LW. Challenges in circulating tumor cell detection by the CellSearch system. Mol Oncol 2016;10:395-407. https://doi.org/10.1016/j.molonc.2015.12.002
  22. Nagrath S, Sequist LV, Maheswaran S, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007;450:1235-1239. https://doi.org/10.1038/nature06385
  23. Kolostova K, Matkowski R, Gurlich R, et al. Detection and cultivation of circulating tumor cells in gastric cancer. Cytotechnology 2016;68:1095-1102. https://doi.org/10.1007/s10616-015-9866-9
  24. Lee A, Park J, Lim M, et al. All-in-one centrifugal microfluidic device for size-selective circulating tumor cell isolation with high purity. Anal Chem 2014;86:11349-11356. https://doi.org/10.1021/ac5035049
  25. 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.
  26. 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. https://doi.org/10.1245/s10434-008-0122-9
  27. Matsusaka S, Chin 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. https://doi.org/10.1111/j.1349-7006.2010.01492.x
  28. 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. https://doi.org/10.1002/cncr.28309
  29. 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. https://doi.org/10.1245/s10434-015-4483-6
  30. 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. https://doi.org/10.1007/s00432-014-1814-0
  31. 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. https://doi.org/10.1038/bjc.2015.417
  32. 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. https://doi.org/10.1371/journal.pone.0180251
  33. 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. https://doi.org/10.3892/mco.2017.1125
  34. 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. https://doi.org/10.1016/j.ejca.2017.03.036
  35. Kim HJ, Lee KW, Kim YJ, et al. Chemotherapy-induced transient CEA and CA19-9 surges in patients with metastatic or recurrent gastric cancer. Acta Oncol 2009;48:385-390. https://doi.org/10.1080/02841860802446761
  36. Muller 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. https://doi.org/10.1158/1078-0432.CCR-04-2469
  37. 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. https://doi.org/10.1093/jnci/85.17.1419
  38. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783-792. https://doi.org/10.1056/NEJM200103153441101
  39. Wulfing P, Borchard J, Buerger H, et al. HER2-positive circulating tumor cells indicate poor clinical outcome in stage I to III breast cancer patients. Clin Cancer Res 2006;12:1715-1720. https://doi.org/10.1158/1078-0432.CCR-05-2087
  40. Riethdorf S, Muller V, Zhang L, et al. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res 2010;16:2634-2645. https://doi.org/10.1158/1078-0432.CCR-09-2042

Cited by

  1. Clinical Application of Circulating Tumor Cells in Gastric Cancer vol.13, pp.4, 2019, https://doi.org/10.5009/gnl18484
  2. Clinical Significance of TWIST-Positive Circulating Tumor Cells in Patients with Esophageal Squamous Cell Carcinoma vol.15, pp.4, 2018, https://doi.org/10.5009/gnl20194
  3. Circulating Tumor Cells and TWIST Expression in Patients with Metastatic Gastric Cancer: A Preliminary Study vol.10, pp.19, 2018, https://doi.org/10.3390/jcm10194481