Circulating Tumor Cells as a Tool of Minimal Residual Disease Can Predict Lung Cancer Recurrence: A longitudinal, Prospective Trial

Ching-Yang Wu, Chia-Lin Lee, Ching-Feng Wu, Jui-Ying Fu, Cheng-Ta Yang, Chi-Tsung Wen, Yun-Hen Liu, Hui-Ping Liu, Jason Chia-Hsun Hsieh, Ching-Yang Wu, Chia-Lin Lee, Ching-Feng Wu, Jui-Ying Fu, Cheng-Ta Yang, Chi-Tsung Wen, Yun-Hen Liu, Hui-Ping Liu, Jason Chia-Hsun Hsieh

Abstract

Background: The role of circulating tumor cells (CTCs) for predicting the recurrence of cancer in lung cancer patients after surgery remains unclear.

Methods: A negatively selected protocol of CTC identification was applied. For all the enrolled patients, CTC testing was performed before and after surgery on the operation day (day 0), postoperative day 1, and day 3. The daily decline and trend of CTCs were analyzed to correlate with cancer relapse. The mixed model repeated measures (MMRM) adjusted by cancer characteristics was applied for statistical significance.

Results: Fifty patients with lung mass undergoing surgery were enrolled. Among 41 primary lung cancers, 26 (63.4%) were pathological stage Tis and I. A total of 200 CTC tests were performed. MMRM analysis indicated that surgery could contribute to a CTC decline after surgery in all patients with statistical significance (p = 0.0005). The daily decrease of CTCs was statistically different between patients with and without recurrence (p = 0.0068). An early rebound of CTC counts on postoperative days 1 and 3 was associated with recurrence months later.

Conclusion: CTC testing can potentially serve as a tool for minimal residual disease detection in early-staged lung cancer after curative surgery.

Keywords: circulating tumor cells; early detection; early-stage; minimal residual disease’ mixed model repeated measures; non-small cell lung cancer.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
The study flow of the prospective design. (CTC, circulating tumor cell; POD1, postoperative day 1; POD3, postoperative day 3).
Figure 2
Figure 2
The positivity of circulating tumor cells (CTCs) in different clinical scenarios. The CTC positivity is not different among four subgroups before curative surgery and is statistically different immediately after the surgery (postoperative day 0, p = 0.04). A possible false positivity of 25.0% can be observed in the benign group (n = 4), but it goes down to zero after surgery. On the other hand, the CTC positivity of the oligometastasis group (n = 6) remains 40.0–80.0% though the operation has removed even the visible tumor(s).
Figure 3
Figure 3
The rebound of circulating tumor cell counts correlates to minimal residual disease. In this figure, a rebound of mean CTC number was observed on day three after surgery in both oligometastasis group (red) and primary lung cancer with recurrence group (green), indicating possible minimal residual tumor signals in blood after surgery.
Figure 4
Figure 4
Boxplots of circulating tumor cells within 3 days of surgery.

References

    1. Torre L.A., Siegel R.L., Ward E.M., Jemal A. Global cancer incidence and mortality rates and trends—an update. Cancer Epidemiol. Prev. Biomark. 2016;25:16–27. doi: 10.1158/1055-9965.EPI-15-0578.
    1. National Comprehensive Cancer Network Non-Small Cell Lung Cancer (Version 2. 2020) [(accessed on 6 March 2020)];2019 Available online: .
    1. Wu C.F., Wu C.Y., Fu J.Y., Wang C.W., Liu Y.H., Hsieh M.J., Wu Y.C. Prognostic value of metastatic N1 lymph node ratio and angiolymphatic invasion in patients with pathologic stage IIA non-small cell lung cancer. Medicine. 2014;93:e102. doi: 10.1097/MD.0000000000000102.
    1. Wu C.F., Fu J.Y., Yeh C.J., Liu Y.H., Hsieh M.J., Wu Y.C., Wu C.Y., Tsai Y.H., Chou W.C. Recurrence Risk Factors Analysis for Stage I Non-small Cell Lung Cancer. Medicine. 2015;94:e1337. doi: 10.1097/MD.0000000000001337.
    1. Wu C.-Y., Fu J.-Y., Wu C.-F., Hsieh M.-J., Liu Y.-H., Wu Y.-C., Yang C.-T., Tsai Y.-H. Survival Prediction Model Using Clinico-Pathologic Characteristics for Nonsmall Cell Lung Cancer Patients After Curative Resection. Medicine. 2015;94:45. doi: 10.1097/MD.0000000000002013.
    1. Hsieh C.P., Hsieh M.J., Wu C.F., Fu J.Y., Liu Y.H., Wu Y.C., Yang C.T., Wu C.Y. Prognostic factors in non-small cell lung cancer patients who received neoadjuvant therapy and curative resection. J. Thorac. Dis. 2016;8:1477–1486. doi: 10.21037/jtd.2016.05.57.
    1. Dahlbom M., Hoffman E.J., Hoh C.K., Schiepers C., Rosenqvist G., Hawkins R.A., Phelps M.E. Whole-body positron emission tomography: Part I. Methods and performance characteristics. J. Nucl. Med. 1992;33:1191–1199.
    1. Nolop K.B., Rhodes C.G., Brudin L.H., Beaney R.P., Krausz T., Jones T., Hughes J.M. Glucose utilization in vivo by human pulmonary neoplasms. Cancer. 1987;60:2682–2689. doi: 10.1002/1097-0142(19871201)60:11<2682::AID-CNCR2820601118>;2-H.
    1. Brown R.S., Leung J.Y., Kison P.V., Zasadny K.R., Flint A., Wahl R.L. Glucose transporters and FDG uptake in untreated primary human non-small cell lung cancer. J. Nucl. Med. 1999;40:556–565.
    1. Pieterman R.M., van Putten J.W., Meuzelaar J.J., Mooyaart E.L., Vaalburg W., Koëter G.H., Fidler V., Pruim J., Groen H.J. Preoperative staging of non–small-cell lung cancer with positron-emission tomography. N. Engl. J. Med. 2000;343:254–261. doi: 10.1056/NEJM200007273430404.
    1. Funama Y., Awai K., Liu D., Oda S., Yanaga Y., Nakaura T., Kawanaka K., Shimamura M., Yamashita Y. Detection of nodules showing ground-glass opacity in the lungs at low-dose multidetector computed tomography: Phantom and clinical study. J. Comput. Assist. Tomogr. 2009;33:49–53. doi: 10.1097/RCT.0b013e31815e6291.
    1. Boedeker K.L., Cooper V.N., McNitt-Gray M.F. Application of the noise power spectrum in modern diagnostic MDCT: Part I. Measurement of noise power spectra and noise equivalent quanta. Phys. Med. Biol. 2007;52:4027–4046. doi: 10.1088/0031-9155/52/14/002.
    1. Schwartz L.H., Litière S., de Vries E., Ford R., Gwyther S., Mandrekar S., Shankar L., Bogaerts J., Chen A., Dancey J. RECIST 1.1—Update and clarification: From the RECIST committee. Eur. J. Cancer. 2016;62:132–137. doi: 10.1016/j.ejca.2016.03.081.
    1. Bueno J., Landeras L., Chung J.H. Updated Fleischner Society Guidelines for Managing Incidental Pulmonary Nodules: Common Questions and Challenging Scenarios. Radiographics. 2018;38:1337–1350. doi: 10.1148/rg.2018180017.
    1. Gupta G.P., Massague J. Cancer metastasis: Building a framework. Cell. 2006;127:679–695. doi: 10.1016/j.cell.2006.11.001.
    1. Fidler I.J. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer. 2003;3:453. doi: 10.1038/nrc1098.
    1. Masuda T., Hayashi N., Iguchi T., Ito S., Eguchi H., Mimori K. Clinical and biological significance of circulating tumor cells in cancer. Mol. Oncol. 2016;10:408–417. doi: 10.1016/j.molonc.2016.01.010.
    1. Young R., Pailler E., Billiot F., Drusch F., Barthelemy A., Oulhen M., Besse B., Soria J.C., Farace F., Vielh P. Circulating tumor cells in lung cancer. Acta Cytol. 2012;56:655–660. doi: 10.1159/000345182.
    1. Tognela A., Spring K.J., Becker T., Caixeiro N.J., Bray V.J., Yip P.Y., Chua W., Lim S.H., de Souza P. Predictive and prognostic value of circulating tumor cell detection in lung cancer: A clinician’s perspective. Crit. Rev. Oncol. Hematol. 2015;93:90–102. doi: 10.1016/j.critrevonc.2014.10.001.
    1. Tanaka F., Yoneda K., Kondo N., Hashimoto M., Takuwa T., Matsumoto S., Okumura Y., Rahman S., Tsubota N., Tsujimura T. Circulating tumor cell as a diagnostic marker in primary lung cancer. Clin. Cancer Res. 2009;15:6980–6986. doi: 10.1158/1078-0432.CCR-09-1095.
    1. O’Flaherty J.D., Gray S., Richard D., Fennell D., O’Leary J.J., Blackhall F.H., O’Byrne K.J. Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer. 2012;76:19–25. doi: 10.1016/j.lungcan.2011.10.018.
    1. Yoon S.O., Kim Y.T., Jung K.C., Jeon Y.K., Kim B.H., Kim C.W. 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. 2011;71:209–216. doi: 10.1016/j.lungcan.2010.04.017.
    1. Nieva J., Wendel M., Luttgen M.S., Marrinucci D., Bazhenova L., Kolatkar A., Santala R., Whittenberger B., Burke J., Torrey M., et al. High-definition imaging of circulating tumor cells and associated cellular events in non-small cell lung cancer patients: A longitudinal analysis. Phys. Biol. 2012;9:016004. doi: 10.1088/1478-3975/9/1/016004.
    1. Maheswaran S., Sequist L.V., Nagrath S., Ulkus L., Brannigan B., Collura C.V., Inserra E., Diederichs S., Iafrate A.J., Bell D.W., et al. Detection of mutations in EGFR in circulating lung-cancer cells. N. Engl. J. Med. 2008;359:366–377. doi: 10.1056/NEJMoa0800668.
    1. Carbone D.P., Salmon J.S., Billheimer D., Chen H., Sandler A., Roder H., Roder J., Tsypin M., Herbst R.S., Tsao A.S., et al. VeriStrat classifier for survival and time to progression in non-small cell lung cancer (NSCLC) patients treated with erlotinib and bevacizumab. Lung Cancer. 2010;69:337–340. doi: 10.1016/j.lungcan.2009.11.019.
    1. Chen Y.Y., Xu G.B. Effect of circulating tumor cells combined with negative enrichment and CD45-FISH identification in diagnosis, therapy monitoring and prognosis of primary lung cancer. Med. Oncol. 2014;31:240. doi: 10.1007/s12032-014-0240-0.
    1. Mascalchi M., Falchini M., Maddau C., Salvianti F., Nistri M., Bertelli E., Sali L., Zuccherelli S., Vella A., Matucci M. Prevalence and number of circulating tumour cells and microemboli at diagnosis of advanced NSCLC. J. Cancer Res. Clin. Oncol. 2016;142:195–200. doi: 10.1007/s00432-015-2021-3.
    1. Su P.-J., Wu M.-H., Wang H.-M., Lee C.-L., Huang W.-K., Wu C.-E., Chang H.-K., Chao Y.-K., Tseng C.-K., Chiu T.-K. Circulating tumour cells as an independent prognostic factor in patients with advanced oesophageal squamous cell carcinoma undergoing chemoradiotherapy. Sci. Rep. 2016;6:31423. doi: 10.1038/srep31423.
    1. Thornblade L.W., Mulligan M.S., Odem-Davis K., Hwang B., Waworuntu R.L., Wolff E.M., Kessler L., Wood D.E., Farjah F. Challenges in Predicting Recurrence After Resection of Node-Negative Non-Small Cell Lung Cancer. Ann. Thorac. Surg. 2018;106:1460–1467. doi: 10.1016/j.athoracsur.2018.06.022.
    1. Okumura Y., Tanaka F., Yoneda K., Hashimoto M., Takuwa T., Kondo N., Hasegawa S. Circulating tumor cells in pulmonary venous blood of primary lung cancer patients. Ann. Thorac. Surg. 2009;87:1669–1675. doi: 10.1016/j.athoracsur.2009.03.073.
    1. Dziedzic D.A., Rudzinski P., Langfort R., Orlowski T., Group P.L.C.S. Risk Factors for Local and Distant Recurrence After Surgical Treatment in Patients with Non–Small-Cell Lung Cancer. Clin. Lung Cancer. 2016;17:e157–e167. doi: 10.1016/j.cllc.2015.12.013.
    1. Godoy M.C., Naidich D.P. Subsolid pulmonary nodules and the spectrum of peripheral adenocarcinomas of the lung: Recommended interim guidelines for assessment and management. Radiology. 2009;253:606–622. doi: 10.1148/radiol.2533090179.
    1. Yu Y.-H., Liao C.-C., Hsu W.-H., Chen H.-J., Liao W.-C., Muo C.-H., Sung F.-C., Chen C.-Y. Increased lung cancer risk among patients with pulmonary tuberculosis: A population cohort study. J. Thorac. Oncol. 2011;6:32–37. doi: 10.1097/JTO.0b013e3181fb4fcc.
    1. Luo Y.-H., Wu C.-H., Wu W.-S., Huang C.-Y., Su W.-J., Tsai C.-M., Lee Y.-C., Perng R.-P., Chen Y.-M. Association between tumor epidermal growth factor receptor mutation and pulmonary tuberculosis in patients with adenocarcinoma of the lungs. J. Thorac. Oncol. 2012;7:299–305. doi: 10.1097/JTO.0b013e31823c588d.
    1. Park C.M., Goo J.M., Kim T.J., Lee H.J., Lee K.W., Lee C.H., Kim Y.T., Kim K.G., Lee H.Y., Park E.-A. Pulmonary nodular ground-glass opacities in patients with extrapulmonary cancers: What is their clinical significance and how can we determine whether they are malignant or benign lesions? Chest. 2008;133:1402–1409. doi: 10.1378/chest.07-2568.
    1. Hashimoto M., Tanaka F., Yoneda K., Takuwa T., Matsumoto S., Okumura Y., Kondo N., Tsubota N., Tsujimura T., Tabata C., et al. Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact. Cardiovasc. Thorac. Surg. 2014;18:775–783. doi: 10.1093/icvts/ivu048.
    1. Crosbie P.A., Shah R., Krysiak P., Zhou C., Morris K., Tugwood J., Booton R., Blackhall F., Dive C. Circulating Tumor Cells Detected in the Tumor-Draining Pulmonary Vein Are Associated with Disease Recurrence after Surgical Resection of NSCLC. J. Thorac. Oncol. 2016;11:1793–1797. doi: 10.1016/j.jtho.2016.06.017.
    1. Reddy R.M., Murlidhar V., Zhao L., Grabauskiene S., Zhang Z., Ramnath N., Lin J., Chang A.C., Carrott P., Lynch W. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J. Thorac. Cardiovasc. Surg. 2016;151:852–858. doi: 10.1016/j.jtcvs.2015.09.126.
    1. Sakurai F., Narii N., Tomita K., Togo S., Takahashi K., Machitani M., Tachibana M., Ouchi M., Katagiri N., Urata Y., et al. Efficient detection of human circulating tumor cells without significant production of false-positive cells by a novel conditionally replicating adenovirus. Mol. Ther. Methods Clin. Dev. 2016;3:16001. doi: 10.1038/mtm.2016.1.
    1. Kolbl A.C., Jeschke U., Andergassen U. The Significance of Epithelial-to-Mesenchymal Transition for Circulating Tumor Cells. Int. J. Mol. Sci. 2016;17:1308. doi: 10.3390/ijms17081308.
    1. Koebel C.M., Vermi W., Swann J.B., Zerafa N., Rodig S.J., Old L.J., Smyth M.J., Schreiber R.D. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450:903–907. doi: 10.1038/nature06309.
    1. De Rubis G., Rajeev Krishnan S., Bebawy M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends Pharmacol. Sci. 2019;40:172–186. doi: 10.1016/j.tips.2019.01.006.
    1. Jahr S., Hentze H., Englisch S., Hardt D., Fackelmayer F.O., Hesch R.D., Knippers R. DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–1665.
    1. Lo Y.M., Zhang J., Leung T.N., Lau T.K., Chang A.M., Hjelm N.M. Rapid clearance of fetal DNA from maternal plasma. Am. J. Hum. Genet. 1999;64:18–24. doi: 10.1086/302205.
    1. Lathia J.D., Liu H. Overview of Cancer Stem Cells and Stemness for Community Oncologists. Target. Oncol. 2017;12:387–399. doi: 10.1007/s11523-017-0508-3.
    1. Chaudhuri A.A., Chabon J.J., Lovejoy A.F., Newman A.M., Stehr H., Azad T.D., Khodadoust M.S., Esfahani M.S., Liu C.L., Zhou L., et al. Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. Cancer Discov. 2017;7:1394–1403. doi: 10.1158/-17-0716.
    1. Comino-Mendez I., Turner N. Predicting Relapse with Circulating Tumor DNA Analysis in Lung Cancer. Cancer Discov. 2017;7:1368–1370. doi: 10.1158/-17-1086.

Source: PubMed

3
구독하다