Circulating Tumor Cells as a Screening and Diagnostic Marker for Early-Stage Non-Small Cell Lung Cancer

Guo-Chen Duan, Xiao-Peng Zhang, Hui-En Wang, Zhi-Kang Wang, Hua Zhang, Lei Yu, Wen-Fei Xue, Zhi-Fei Xin, Zhong-Hui Hu, Qing-Tao Zhao, Guo-Chen Duan, Xiao-Peng Zhang, Hui-En Wang, Zhi-Kang Wang, Hua Zhang, Lei Yu, Wen-Fei Xue, Zhi-Fei Xin, Zhong-Hui Hu, Qing-Tao Zhao

Abstract

Background: Circulating tumor cells (CTCs) have become potential diagnostic biomarker for several types of cancer, including lung cancer. In this study, we aim to determine whether CTCs detected by CellCollector can be used for early-stage diagnosis of lung cancer.

Methods: In this study, we recruited 64 volunteers, among whom 44 were suspected lung cancer patients requiring surgical treatment and 20 were healthy volunteers. We simultaneously analyzed PD-L1 expression in CTCs isolated using the GILUPI CellCollector and copy number variation by next-generation sequencing (NGS).

Results: We enrolled a total of 44 patients with suspected lung cancer who required surgery and 20 healthy volunteers. The patients were classified into 4 groups based on their pathological results: benign disease, in situ cancer, microinvasive, and invasive. The CTCs detection rate for each group was 10.00% (1/10), 45% (5/11), 50% (7/14), and 67% (6/9), respectively. Among the patients with lung cancer, the CTCs detection rate increased with disease progression. The rate of CTCs positivity was 52.94% (18/34) in patients who were diagnosed with lung cancer by pathology and 10% (1/10) in patients with benign disease. CTCs were not detected in the control group. The area under the receiver operating characteristic (ROC) curve, a measure for distinguishing patients with primary lung cancer, was 0.715 (95% CI 0.549-0.880, P=0.041). The sensitivity and specificity of the in vivo CTCs detection strategy for the diagnosis of early-stage lung cancer were 52.94% and 90%, respectively. CTCs were associated with clinical pathology but not with the size and location of the nodules.

Conclusion: CTCs isolation using the CellCollector in vivo detection method might be effective for distinguishing between benign and malignant nodules and may be used for early-stage diagnosis of lung cancer.

Keywords: CellCollector; circulating tumor cells; early-stage diagnosis; in vivo; lung cancer.

Conflict of interest statement

The authors have no conflicts of interest to declare.

© 2020 Duan et al.

Figures

Figure 1
Figure 1
Experimental flow chart.
Figure 2
Figure 2
Collection and determination of CTCs. (A) CellCollector in vivo CTCs detection system. A medical stainless steel wire with a 2-cm-long functional domain coated with EpCAM antibody and a hydrogel stratum. The sampling probe was inserted into cubital vein peripheral blood through a 20G catheter with the functional domain exposed to the peripheral blood and placed in vivo for 30 min to capture tumor cells. (B) Determination of tumor cells by CK7/19/panCK (green channel). Hoechst was used for nuclear counterstaining (blue channel), and white cells were determined by CD45 staining (red channel). PD-L1 expression in CTCs was determined by PD-L1 staining (orange channel). Scale bar: 10 μm. WBC indicates white blood cell. (C) Collection and determination of CTCs in the recruited patients by CellCollector. Two CTCs were collected in patient number 36, 3 CTCs in patient number 39, and 1 CTC in patient numbers 42 and 44. P indicates patient. (D) Imaging and pathological results for patient number 39. A pulmonary nodule was found in the right lung. The patient was diagnosed with invasive adenocarcinoma by pathology.
Figure 3
Figure 3
CTCs detection in suspected lung cancer patients and healthy controls. (A) CTCs counts and detection rate in suspected lung cancer patients and healthy controls. (B) CTCs counts and detection rate in patients diagnosed with lung cancer and benign disease by pathology. (C) CTCs counts and detection rate in different groups of patients (benign disease group, in situ cancer group, microinvasive group, and invasive group). Bar values represent the median and interquartile range. (D) ROC curve using CTC counts to distinguish benign and malignant nodules.

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(6):394–424. doi:10.3322/caac.v68.6
    1. Goldstraw P, Chansky K, Crowley J, et al. The IASLC lung cancer staging project: proposals for revision of the TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11(1):39–51. doi:10.1016/j.jtho.2015.09.009
    1. National Lung Screening Trial Research T, Aberle DR, Adams AM, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):395–409.
    1. Patz EF Jr, Pinsky P, Gatsonis C, et al. Overdiagnosis in low-dose computed tomography screening for lung cancer. JAMA Intern Med. 2014;174(2):269–274. doi:10.1001/jamainternmed.2013.12738
    1. Wicha MS, Hayes DF. Circulating tumor cells: not all detected cells are bad and not all bad cells are detected. J Clin Oncol. 2011;29(12):1508–1511. doi:10.1200/JCO.2010.34.0026
    1. Alix-Panabieres C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem. 2013;59(1):110–118. doi:10.1373/clinchem.2012.194258
    1. Patz EF Jr., Campa MJ, Gottlin EB, Kusmartseva I, Guan XR, Herndon JE 2nd. Panel of serum biomarkers for the diagnosis of lung cancer. J Clin Oncol. 2007;25(35):5578–5583. doi:10.1200/JCO.2007.13.5392
    1. Cho IH, JY. Lung cancer biomarkers. Adv Clin Chem. 2015;72:107.
    1. Fujiwara K, Fujimoto N, Tabata M, et al. Identification of epigenetic aberrant promoter methylation in serum DNA is useful for early detection of lung cancer. Clin Cancer Res. 2005;11(3):1219–1225.
    1. Weiss G, Schlegel A, Kottwitz D, Konig T, Tetzner R. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J Thorac Oncol. 2017;12(1):77–84. doi:10.1016/j.jtho.2016.08.123
    1. Santarpia M, Liguori A, D’aveni A, et al. Liquid biopsy for lung cancer early detection. J Thorac Dis. 2018;10(Suppl 7):S882–S897.
    1. Diamandis EP, Fiala C. Can circulating tumor DNA be used for direct and early stage cancer detection? F1000Res. 2017;6:2129.
    1. Zhang H, Gong S, Liu Y, et al. Enumeration and molecular characterization of circulating tumor cell using an in vivo capture system in squamous cell carcinoma of head and neck. Chin J Cancer Res. 2017;29(3):196–203. doi:10.21147/j.issn.1000-9604.2017.03.05
    1. Markou A, Lazaridou M, Paraskevopoulos P, et al. Multiplex gene expression profiling of in vivo isolated circulating tumor cells in high-risk prostate cancer patients. Clin Chem. 2017;64(2):297–306. doi:10.1373/clinchem.2017.275503
    1. Mandair D, Vesely C, Ensell L, et al. A comparison of CellCollector with CellSearch in patients with neuroendocrine tumours. Endocr Relat Cancer. 2016;23(10):L29–32. doi:10.1530/ERC-16-0201
    1. Gorges TM, Penkalla N, Schalk T, et al. Enumeration and molecular characterization of tumor cells in lung cancer patients using a novel in vivo device for capturing circulating tumor cells. Clin Cancer Res. 2016;22(9):2197–2206. doi:10.1158/1078-0432.CCR-15-1416
    1. Li JB, Geng CZ, Yan M, et al. Circulating tumor cells in patients with breast tumors were detected by a novel device: a multicenter, clinical trial in China (in Chinese). Zhonghua Yi Xue Za Zhi. 2017;97(24):1857–1861. doi:10.3760/cma.j.issn.0376-2491.2017.24.003
    1. Chen S, Tauber G, Langsenlehner T, et al. In vivo detection of circulating tumor cells in high-risk non-metastatic prostate cancer patients undergoing radiotherapy. Cancers (Basel). 2019;11(7). doi:10.3390/cancers11070933.
    1. Cieslikowski WA, Budna-tukan J, Swierczewska M, et al. Circulating tumor cells as a marker of disseminated disease in patients with newly diagnosed high-risk prostate cancer. Cancers (Basel). 2020;12(1):160. doi:10.3390/cancers12010160
    1. He Y, Shi J, Shi G, et al. Using the new cellcollector to capture circulating tumor cells from Blood in Different Groups of pulmonary disease: a cohort study. Sci Rep. 2017;7(1):9542. doi:10.1038/s41598-017-09284-0
    1. Tanaka F, Yoneda K, Kondo N, et al. Circulating tumor cell as a diagnostic marker in primary lung cancer. Clin Cancer Res. 2009;15(22):6980–6986. doi:10.1158/1078-0432.CCR-09-1095
    1. Wang X, Sun Q, Liu Q, Wang C, Yao R, Wang Y. CTC immune escape mediated by PD-L1. Med Hypotheses. 2016;93:138–139. doi:10.1016/j.mehy.2016.05.022
    1. Nicolazzo C, Raimondi C, Mancini M, et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci Rep. 2016;6:31726. doi:10.1038/srep31726
    1. Satelli A, Batth IS, Brownlee Z, et al. Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci Rep. 2016;6:28910. doi:10.1038/srep28910
    1. Alix-panabieres C, Riethdorf S, Pantel K. Circulating tumor cells and bone marrow micrometastasis. Clin Cancer Res. 2008;14(16):5013–5021. doi:10.1158/1078-0432.CCR-07-5125
    1. Hosseini H, Obradovic MM, Hoffmann M, et al. Early dissemination seeds metastasis in breast cancer. Nature. 2016;540:552–558. doi:10.1038/nature20785
    1. Chemi F, Rothwell DG, McGranahan N, et al. Pulmonary venous circulating tumor cell dissemination before tumor resection and disease relapse. Nat Med. 2019;25(10):1534–1539. doi:10.1038/s41591-019-0593-1
    1. Zeng Y, Zhu J, Dan S, et al. MicroRNA-205 targets SMAD4 in non-small cell lung cancer and promotes lung cancer cell growthin vitroandin vivo. Oncotarget. 2017;8(19):30817–30829. doi:10.18632/oncotarget.10339
    1. Singh A, Singh N, Behera D, Sharma S. Genetic investigation of polymorphic OGG1 and MUTYH genes towards increased susceptibility in lung adenocarcinoma and its impact on overall survival of lung cancer patients treated with platinum based chemotherapy. Pathol Oncol Res. 2017;1–14.
    1. Chuanliang P, Yunpeng Z, Yingtao H, Qifeng S, Xiaogang Z, Bo C. Syk expression in non-small-cell lung cancer and its relation with angiogenesis. J Can Res Ther. 2016;12(2):663–666. doi:10.4103/0973-1482.154082
    1. Udyavar AR, Hoeksema MD, Clark JE, et al. Co-expression network analysis identifies Spleen Tyrosine Kinase (SYK) as a candidate oncogenic driver in a subset of small-cell lung cancer. BMC Syst Biol. 2013;7(S5):S1. doi:10.1186/1752-0509-7-S5-S1
    1. Gu J, Chen J, Feng J, et al. Overexpression of ADAMTS5 can regulate the migration and invasion of non-small cell lung cancer. Tumor Biol. 2016;37(7):1–9. doi:10.1007/s13277-015-4573-x

Source: PubMed

3
구독하다