DOI QR코드

DOI QR Code

Meta-analysis of Circulating Tumor Cells as a Prognostic Marker in Lung Cancer

  • Ma, Xue-Lei (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Xiao, Zhi-Lan (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Liu, Lei (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Liu, Xiao-Xiao (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Nie, Wen (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Li, Ping (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Chen, Nian-Yong (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University) ;
  • Wei, Yu-Quan (State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University)
  • Published : 2012.04.30

Abstract

Introduction: Recent studies have shown that circulating tumor cells (CTCs) play potential roles as diagnostic and prognostic biomarkers with various cancer types. The aim of this study was to comprehensively and quantitatively summarize the evidence for the use of CTCs to predict the survival outcome of lung cancer patients. Materials and Methods: Relevant literature was identified using Medline and EMBASE. Patients' clinical characteristics, overall survival (OS) and progression-free survival (PFS) together with CTC positive rates at different time points (before, during and after treatment) were extracted. A meta-analysis was performed to clarify the prognostic role of CTCs and the correlation between the CTC appearance and clinical characteristics. Results: A total of 12 articles containing survival outcomes and clinical characteristics and 15 articles containing only clinical characteristics were included for the global meta-analysis. The hazard ratio (HR) for OS predicted by pro-treatment CTCs was 2.61 [1.82, 3.74], while the HR for PFS was 2.37 [1.41, 3.99]. The HR for OS predicted by post-treatment CTCs was 4.19 [2.92, 6.00], while the HR for PFS was 4.97 [3.05, 8.11]. Subgroup analyses were conducted according to histological classification and detection method. Odds ratio (OR) showed the appearance of pro-treatment CTCs correlated with the lymph node status, distant metastasis, and TNM staging, while post-treatment CTCs correlated with TNM staging only. Conclusion: Detection of CTCs in the peripheral blood indicates a poor prognosis in patients with lung cancer.

Keywords

References

  1. Ashworth TR (1869). A case of cancer in which cells similar to those in the tumours were seen in the blood after death. 14,146-149.
  2. Begg CB (1994). Publication bias. 25, 299-409.
  3. Castaldo G, Tomaiuolo R, Sanduzzi A, et al (1997). Lung cancer metastatic cells detected in blood by reverse transcriptasepolymerase chain reaction and dot-blot analysis. J Clin Oncol, 15, 3388-93.
  4. Chen TF, Jiang GL, Fu XL, et al (2007). CK19 mRNA expression measured by reverse-transcription polymerase chain reaction (RT-PCR) in the peripheral blood of patients with non-small cell lung cancertreated by chemo-radiation: an independent prognostic factor. Lung Cancer, 56, 105-14. https://doi.org/10.1016/j.lungcan.2006.11.006
  5. Cohen SJ, Punt CJ, Iannotti N, et al (2008). Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol, 26, 3213-21. https://doi.org/10.1200/JCO.2007.15.8923
  6. Cristofanilli M (2006). Circulating tumor cells, disease progression, and survival in metastatic breast cancer. Semin Oncol, 33, S9-14.
  7. Devriese LA, Bosma AJ, de Heuvel MM, v Heemsbergen W, Voest EE, Schellens JH (2012). Circulating tumor cell detection in advanced non-small cell lung cancer patients by multi-marker QPCR analysis. Lung Cancer, 75, 242-7. https://doi.org/10.1016/j.lungcan.2011.07.003
  8. Farace F, Massard C, Vimond N, et al (2011). A direct comparison of CellSearch and ISET for circulating tumourcell detection in patients with metastatic carcinomas. Br J Cancer, 105, 847-53. https://doi.org/10.1038/bjc.2011.294
  9. Guo Y, Wang J, Huang P (2009). [Clinical Significance of CK20, CK19, CEA mRNAs in Peripheral Blood from Lung Cancer Patients.]. Zhongguo Fei Ai Za Zhi, 12, 1013-7.
  10. Hayes DC, Secrist H, Bangur CS, et al (2006). Multigene realtime PCR detection of circulating tumor cells in peripheral blood of lung cancer patients. Anticancer Res, 26, 1567-75.
  11. Hayes DF, Isaacs C, Stearns V (2001). Prognostic factors in breast cancer: current and new predictors of metastasis. J Mammary Gland Biol Neoplasia, 6, 375-92. https://doi.org/10.1023/A:1014778713034
  12. Hedges LV, Vevea JL (1998). Fixed-and random-effects models in meta-analysis. Psychol Methods, 3, 486. https://doi.org/10.1037/1082-989X.3.4.486
  13. Helo P, Cronin AM, Danila DC, et al (2009). Circulating prostate tumor cells detected by reverse transcription-PCR in men with localized or castration-refractory prostate cancer: concordance with CellSearch assay and association with bone metastases and with survival. Clin Chem, 55, 765-73. https://doi.org/10.1373/clinchem.2008.117952
  14. Higgins JP, Thompson SG, Deeks JJ, Altman DG (2003). Measuring inconsistency in meta-analyses. BMJ, 327, 557-60. https://doi.org/10.1136/bmj.327.7414.557
  15. Hofman V, Bonnetaud C, Ilie MI, et al (2011). Preoperative circulating tumor cell detection using the isolation by size of epithelial tumor cell method for patients with lung cancer is a new prognostic biomarker. Clin Cancer Res, 17, 827-35. https://doi.org/10.1158/1078-0432.CCR-10-0445
  16. Hofman V, Ilie MI, Long E, et al (2011). Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch Assay and the isolation by size of epithelial tumor cell method. Int J Cancer, 129, 1651-60. https://doi.org/10.1002/ijc.25819
  17. Hofman V, Long E, Ilie M, et al (2012). Morphological analysis of circulating tumour cells in patients undergoing surgeryfor non-small cell lung carcinoma using the isolation by size of epithelialtumour cell (ISET) method. Cytopathology, 23, 30-8. https://doi.org/10.1111/j.1365-2303.2010.00835.x
  18. Hofman V, Long E, Ilie M, et al (2012). Morphological analysis of circulating tumour cells in patients undergoing surgeryfor non-small cell lung carcinoma using the isolation by size of epithelialtumour cell (ISET) method. Cytopathology, 23, 30-8. https://doi.org/10.1111/j.1365-2303.2010.00835.x
  19. Hou JM, Greystoke A, Lancashire L, et al (2009). Evaluation of circulating tumor cells and serological cell death biomarkers in small cell lung cancer patients undergoing chemotherapy. Am J Pathol, 175, 808-16. https://doi.org/10.2353/ajpath.2009.090078
  20. Hou JM, Krebs M, Ward T, et al (2011). Circulating tumor cells as a window on metastasis biology in lung cancer. Am J Pathol, 178, 989-96. https://doi.org/10.1016/j.ajpath.2010.12.003
  21. Hou JM, Krebs MG, Lancashire L, et al (2012). Clinical significance and molecular characteristics of circulating tumor cellsand circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol, 30, 525-32. https://doi.org/10.1200/JCO.2010.33.3716
  22. Huang TH, Wang Z, Li Q, Li FR, Qi H, Zhou HX (2007). [Clinical significance of enrichment and detection of circulating tumor cells in NSCLC patients with immunomagnetic beads]. Zhonghua Zhong Liu Za Zhi, 29, 676-80.
  23. Jemal A, Bray F, Center MM, et al (2011). Global cancer statistics. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
  24. Kim MY, Oskarsson T, Acharyya S, et al (2009). Tumor selfseeding by circulating cancer cells. Cell, 139, 1315-26. https://doi.org/10.1016/j.cell.2009.11.025
  25. Krebs MG, Sloane R, Priest L, et al (2011). Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol, 29, 1556-63. https://doi.org/10.1200/JCO.2010.28.7045
  26. Krebs MG, Sloane R, Priest L, et al (2011). Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol, 29, 1556-63. https://doi.org/10.1200/JCO.2010.28.7045
  27. Kubuschok B, Passlick B, Izbicki JR, Thetter O, Pantel K (1999). Disseminated tumor cells in lymph nodes as a determinant for survival in surgically resected non-small-cell lung cancer. J Clin Oncol, 17,19-24.
  28. Kurusu Y, Yamashita J, Ogawa M (1999). Detection of circulating tumor cells by reverse transcriptase-polymerase chain reaction in patients with resectable non-small-cell lung cancer. Surgery, 126, 820-6. https://doi.org/10.1016/S0039-6060(99)70020-6
  29. Kurusu Y, Yamashita J, Ogawa M (1999). Detection of circulating tumor cells by reverse transcriptase-polymerase chainreaction in patients with resectable non-small-cell lung cancer. Surgery, 126, 820-6. https://doi.org/10.1016/S0039-6060(99)70020-6
  30. Labelle M, Begum S, Hynes RO (2011). Direct signaling between platelets and cancer cells induces an epithelial mesenchymal-like transition and promotes metastasis. Cancer Cell, 20, 576-90. https://doi.org/10.1016/j.ccr.2011.09.009
  31. Li J, Sun YE, Sheng QM, Yan LD, Lu XC (2005). [Detection of blood dissemination during the operation of lung cancer and its significance]. Zhonghua Wai Ke Za Zhi, 43, 76-9.
  32. Liu L, Liao GQ, He P, et al (2008). Detection of circulating cancer cells in lung cancer patients with a panel of marker genes. Biochem Biophys Res Commun, 372, 756-60. https://doi.org/10.1016/j.bbrc.2008.05.101
  33. Mocellin S, Hoon D, Ambrosi A, Nitti D, Rossi CR (2006). The prognostic value of circulating tumor cells in patients with melanoma: asystematic review and meta-analysis. Clin Cancer Res, 12,4605-13. https://doi.org/10.1158/1078-0432.CCR-06-0823
  34. Nieva J, Wendel M, Luttgen MS, et al (2012). High-definition imaging of circulating tumor cells and associated cellular eventsin non-small cell lung cancer patients: a longitudinal analysis. Phys Biol, 9, 016004. https://doi.org/10.1088/1478-3975/9/1/016004
  35. Okumura Y, Tanaka F, Yoneda K, et al (2009). Circulating tumor cells in pulmonary venous blood of primary lung cancer patients. Ann Thorac Surg, 87, 1669-75. https://doi.org/10.1016/j.athoracsur.2009.03.073
  36. Pantel K, Alix-Panabieres C (2010). Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med, 16, 398-406. https://doi.org/10.1016/j.molmed.2010.07.001
  37. Parmar MK, Torri V, Stewart L (1998). Extracting summary statistics to perform meta-analyses of the publishedliterature for survival endpoints. Stat Med, 17, 2815-34. https://doi.org/10.1002/(SICI)1097-0258(19981230)17:24<2815::AID-SIM110>3.0.CO;2-8
  38. Peck K, Sher YP, Shih JY, et al (1998). Detection and quantitation of circulating cancer cells in the peripheral blood oflung cancer patients. Cancer Res, 58, 2761-5.
  39. Rahbari NN, Aigner M, Thorlund K, et al (2010). Metaanalysis shows that detection of circulating tumor cells indicates poorprognosis in patients with colorectal cancer. Gastroenterology, 138, 1714-26. https://doi.org/10.1053/j.gastro.2010.01.008
  40. Sawabata N, Okumura M, Utsumi T, et al (2007). Circulating tumor cells in peripheral blood caused by surgical manipulation of non-small-cell lung cancer: pilot study using an immunocytology method. Gen Thorac Cardiovasc Surg, 55, 189-92. https://doi.org/10.1007/s11748-007-0101-2
  41. Sher YP, Shih JY, Yang PC, et al (2005). Prognosis of non-small cell lung cancer patients by detecting circulating cancer cells in the peripheral blood with multiple marker genes. Clin Cancer Res, 11, 173-9.
  42. Sheu CC, Yu YP, Tsai JR, et al (2006). Development of a membrane array-based multimarker assay for detection ofcirculating cancer cells in patients with non-small cell lung cancer. Int J Cancer, 119, 1419-26. https://doi.org/10.1002/ijc.21999
  43. Tanaka F, Yoneda K, Kondo N, et al (2009). Circulating tumor cell as a diagnostic marker in primary lung cancer. Clin Cancer Res, 15, 6980-6. https://doi.org/10.1158/1078-0432.CCR-09-1095
  44. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR (2007). Practical methods for incorporating summary time-to-event data intometa-analysis. Trials, 8, 16. https://doi.org/10.1186/1745-6215-8-16
  45. Wang FB, Yang XQ, Yang S, et al (2011). A Higher Number of Circulating Tumor Cells (CTC) in Peripheral Blood Indicates Poor Prognosis in Prostate Cancer Patients - A Meta-analysis. Asian Pac J Cancer Prev, 12, 2629-35.
  46. Wendel M, Bazhenova L, Boshuizen R, et al (2012). Fluid biopsy for circulating tumor cell identification in patients with earlyandlate- stage non-small cell lung cancer: a glimpse into lung cancer biology. Phys Biol, 9, 016005. https://doi.org/10.1088/1478-3967/9/1/016005
  47. Williamson PR, Smith CT, Hutton JL, Marson AG (2002). Aggregate data meta-analysis with time-to-event outcomes. Stat Med, 21, 3337-51. https://doi.org/10.1002/sim.1303
  48. Wu C, Hao H, Li L, et al (2009). Preliminary investigation of the clinical significance of detecting circulatingtumor cells enriched from lung cancer patients. J Thorac Oncol, 4, 30-6. https://doi.org/10.1097/JTO.0b013e3181914125
  49. Yamashita J, Matsuo A, Kurusu Y, et al (2002). Preoperative evidence of circulating tumor cells by means of reverse transcriptase-polymerase chain reaction for carcinoembryonic antigen messenger RNA is an independent predictor of survival in non-small cell lung cancer: a prospective study. J Thorac Cardiovasc Surg, 124, 299-305. https://doi.org/10.1067/mtc.2002.124370
  50. Yamashita JI, Kurusu Y, Fujino N, Saisyoji T, Ogawa M (2000). Detection of circulating tumor cells in patients with non-small cell lung cancer undergoing lobectomy by video-assisted thoracic surgery: a potential hazard for intraoperative hematogenous tumor cell dissemination. J Thorac Cardiovasc Surg, 119, 899-905. https://doi.org/10.1016/S0022-5223(00)70084-5
  51. Yoon SO, Kim YT, Jung KC, et al (2011). TTF-1 mRNA-positive circulating tumor cells in the peripheral blood predict poor prognosis in surgically resected non-small cell lung cancer patients. Lung Cancer, 71, 209-16. https://doi.org/10.1016/j.lungcan.2010.04.017
  52. Zhao S, Liu Y, Zhang Q, et al (2011). The prognostic role of circulating tumor cells (CTCs) detected by RT-PCR inbreast cancer: a meta-analysis of published literature. Breast Cancer Res Treat, 130, 809-16. https://doi.org/10.1007/s10549-011-1379-4

Cited by

  1. Meta-analysis shows that circulating tumor cells including circulating microRNAs are useful to predict the survival of patients with gastric cancer vol.14, pp.1, 2014, https://doi.org/10.1186/1471-2407-14-773
  2. Prognostic Role of Circulating Tumor Cells in Patients with Pancreatic Cancer: a Meta-analysis vol.15, pp.15, 2014, https://doi.org/10.7314/APJCP.2014.15.15.6015
  3. Circulating Tumor Cells are Associated with Bone Metastasis of Lung Cancer vol.15, pp.15, 2014, https://doi.org/10.7314/APJCP.2014.15.15.6369
  4. How to Explain the Contradiction of microRNA 200c Expression and Survival in Solid Tumors?: a Meta-analysis vol.15, pp.8, 2014, https://doi.org/10.7314/APJCP.2014.15.8.3687
  5. Patients with CD133-Negative Colorectal Liver Metastasis Have a Poor Prognosis After Hepatectomy vol.21, pp.6, 2014, https://doi.org/10.1245/s10434-014-3549-1
  6. Prognostic role of circulating tumor cells and disseminated tumor cells in patients with prostate cancer: a systematic review and meta-analysis vol.35, pp.6, 2014, https://doi.org/10.1007/s13277-014-1731-5
  7. Carboxybetaine Methacrylate-Modified Nylon Surface for Circulating Tumor Cell Capture vol.6, pp.6, 2014, https://doi.org/10.1021/am500394j
  8. Cancer stem cell characteristics of circulating tumor cells vol.90, pp.8, 2014, https://doi.org/10.3109/09553002.2014.886798
  9. Prognostic value of circulating tumor cells in patients with squamous cell carcinoma of the head and neck: a systematic review and meta-analysis vol.32, pp.5, 2015, https://doi.org/10.1007/s12032-015-0579-x
  10. Phenotype of circulating tumor cell: face-off between epithelial and mesenchymal masks vol.37, pp.5, 2016, https://doi.org/10.1007/s13277-016-4796-5
  11. Differential impact of circulating tumor cells on disease recurrence and survivals in patients with head and neck squamous cell carcinomas: An updated meta-analysis vol.13, pp.9, 2018, https://doi.org/10.1371/journal.pone.0203758