Skip to content
BY 4.0 license Open Access Published by De Gruyter September 27, 2021

Circulating tumor cells in bladder cancer: a new horizon of liquid biopsy for precision medicine

  • Felice Crocetto , Amelia Cimmino EMAIL logo , Matteo Ferro and Daniela Terracciano ORCID logo EMAIL logo

Clinical management of bladder cancer (BC) patients offers several challenges such as poor outcome because of elevated recurrence rates and lack of response to chemotherapy [1]. So, there is a need of noninvasive prognostic and predictive tools able to allow risk category assessment and real-time supervision of drug response [2]. Recently, circulating tumor cells (CTCs) have been proposed as prognostic tool able to improve cancer patients’ clinical management [3], [4], [5], [6]. CTCs detached from the primary tumor, enter the bloodstream and colonize distant organ, promoting cancer dissemination [7]. Emerging technologies are available to isolate CTC from patient’s blood to provide a “liquid biopsy”. Such a tool provides a molecular picture of the metastatic disease, useful to assess the cause of drug resistance onset [3, 6, 8], [9], [10], [11], [12], [13], [14]. CTC are very scarce in the blood, so robust methods are still needed for their routine use in laboratory practice [3, 11]. Several technologies have been developed in the last few years [11, 12] and several studies have been performed on the potential use of CTCs in bladder cancer patient clinical management.

CTCs, derived from both primary or metastatic tumors by passive shedding or dynamic stromal invasion, are considered responsible of disease dissemination. Once in the circulatory system, CTC survive to the sheer stress and escape to the immune system to reach distant organs. The microenvironment of the reached site will influence their state of quiescent or proliferation developing metastatic foci. Isolation from blood is challenging because CTC account for less than 0.004% of all mononucleated blood cells. For this reason, various methods of isolation and count assessment including immunoaffinity, biophysical and direct molecular approaches, have been developed to enrich CTCs from many millions of normal blood cells. CellSearch (Janssen Diagnostics, USA) has been the most used platform for CTC-based studies in BC. This is the only FDA-approved method for monitoring CTC in patients with metastatic breast, colorectal, and prostate cancer, but not yet for BC. This assay is based on the characteristic of CTC that are nucleated EpCAM+/CK+/CD45 – cells, different from healthy EpCAM-/CK-/CD45+ blood cells. This test is based on the positive selection of CTCs using antibodies against the epithelial cell adhesion molecule (EpCAM) antigen. Thus, blood samples are first incubated with ferroparticles coated in antiEpCAM antibodies. Then ferroparticle bound cells are then captured in a magnetic field, following the flow cytometry analysis of cells fluorescently labelled with antibodies against CK8, CK18, CK19, and CD45 and cytokeratins 8, 18, and 19. By using CellSearch assay, a CTC is defined as any nucleated cell that is positive for cytokeratin expression, but negative for CD45 typically expressed on leukocytes.

Currently, several studies have evaluated the CTCs by using CellSearch system, in nonmetastatic BC patients. Regarding the studies enrolling nonmetastatic patient’s cohorts, eight were able to detect CTC in 17–30% of patients [15], [16], [17], [18], [19], [20], whereas the remaining two were unable to detect CTC in the blood [21, 22]. Such low yields could be associated with the lack of sensitivity and specificity of the assay for diagnostic use. Several studies showed that CTCs were associated with staging, grading, and metastasis and CTC-positive patients have poorer progression-free, cancer-specific, and overall-survival compared to CTC-negative subjects [18, 23], [24], [25], [26]. Worth of attention is that the detection of a single CTC was significantly associated with a decreased survival in patients with BC [27]. Moreover, some authors demonstrated the relevance of the molecular characterization of CTCs. Osman et al. reported that patients presenting uroplakin/EGFR-positive CTCs after radical cystectomy had a higher risk of recurrence [28]. Gudemann et al. found that the expression of CK20 on CTCs was significantly associated with staging and tumor burden [29]. Anantharaman et al. [30] showed that programed death-ligand 1 (PDL-1) expression was significantly associated with reduced overall survival. Predicting which individuals will respond to a specific treatment is still challenging for BC clinical management. Alvea et al. [31] collected blood samples from 20 patients with BC who were eligible for cisplatin based neoadjuvant chemotherapy. CTCs were detected by using IsoFlux method and compared to CellSearch, showing an improvement in the CTCs identification capacity. It was demonstrated that high (>30 CTCs) counts prior to neoadjuvant chemotherapy are associated with unfavorable tumor features at radical cystectomy. In the adjuvant chemotherapy (AC) setting a meta-analysis including 945 patients found an overall survival and disease-free survival benefit in patients with muscle-invasive BC (MIBC) receiving adjuvant cisplatin-based chemotherapy after radical cystectomy [32]. In a prospective study including 185 patients, Soave et al. investigated the potential role of the CTC status regarding decision making on AC administration in BC patients, following RC. In this study, the presence of CTC was associated with inferior outcomes in patients without administration of AC, but not in patients with administration of AC [19]. The authors concluded that CTC may be useful for decision-making pro or contra AC. The in SWOG S1314, a randomized phase II trial testing a gene panel derived by coexpression extrapolation (COXEN) [33], was initiated to evaluate prospectively the potential benefit of AC for localized MIBC. If successful, COXEN score derived from CTC RNA can predict treatment response to AC as RNA from bladder tumor biopsy.

Based on currently available data, the detection of circulating tumor cells in BC patients represents a promising noninvasive tool for prognostic assessment and to guide personalized treatment (Figure 1). Further studies are encouraged to better clarify the clinical impact of CTCs enumeration and molecular characterization in clinical practice.

Figure 1: 
Potential of circulating tumor cells in bladder cancer patient clinical management.
Figure 1:

Potential of circulating tumor cells in bladder cancer patient clinical management.


Corresponding authors: Daniela Terracciano, Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini, 5, 80131, Naples, Italy, Phone: +39817463617, E-mail: ; and Amelia Cimmino, Institute of Genetics and Biophysics, National Research Council, Via Pietro Castellino, 111, 80131, Napoli, NA, Italy, E-mail:

Acknowledgments

Not applicable.

  1. Research funding: None to declare.

  2. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: Authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: Not applicable.

References

1. Azevedo, R, Peixoto, A, Gaiteiro, C, Fernandes, E, Neves, M, Lima, L, et al.. Over forty years of bladder cancer glycobiology: where do glycans stand facing precision oncology? Oncotarget 2017;8:91734–64. https://doi.org/10.18632/oncotarget.19433.Search in Google Scholar PubMed PubMed Central

2. Ferro, M, Baba, DF, de Cobelli, O, Musi, G, Lucarelli, G, Terracciano, D, et al.. Neutrophil percentage-to-albumin ratio predicts mortality in bladder cancer patients treated with neoadjuvant chemotherapy followed by radical cystectomy. Future Sci OA 2021;7:FSO709. https://doi.org/10.2144/fsoa-2021-0008.Search in Google Scholar PubMed PubMed Central

3. Hu, F, Mao, X, Zhang, Y, Zheng, X, Gu, P, Wang, H, et al.. Reliability of using circulating tumor cells for detecting epidermal growth factor receptor mutation status in advanced non-small-cell lung cancer patients: a meta-analysis and systematic review. OncoTargets Ther 2018;11:1373–84. https://doi.org/10.2147/ott.s158479.Search in Google Scholar PubMed PubMed Central

4. Huang, H, Shi, Y, Huang, J, Wang, X, Zhang, R, Chen, H. Circulating tumor cells as a potential biomarker in diagnosis of lung cancer: a systematic review and meta-analysis. Clin Respir J 2018;12:639–45. https://doi.org/10.1111/crj.12573.Search in Google Scholar PubMed

5. Tan, Y, Wu, H. The significant prognostic value of circulating tumor cells in colorectal cancer: a systematic review and meta-analysis. Curr Probl Canc 2018;42:95–106. https://doi.org/10.1016/j.currproblcancer.2017.11.002.Search in Google Scholar PubMed

6. Chalopin, A, Tellez-Gabriel, M, Brown, HK, Vallette, F, Heymann, MF, Gouin, F, et al.. Isolation of circulating tumor cells in a preclinical model of osteosarcoma: effect of chemotherapy. J Bone Oncol 2018;12:83–90. https://doi.org/10.1016/j.jbo.2018.07.002.Search in Google Scholar PubMed PubMed Central

7. Maheswaran, S, Haber, DA. Circulating tumor cells: a window into cancer biology and metastasis. Curr Opin Genet Dev 2010;20:96–9. https://doi.org/10.1016/j.gde.2009.12.002.Search in Google Scholar PubMed PubMed Central

8. Bhagwat, N, Dulmage, K, Pletcher, CHJr., Wang, L, DeMuth, W, Sen, M, et al.. An integrated flow cytometry-based platform for isolation and molecular characterization of circulating tumor single cells and clusters. Sci Rep 2018;8:5035. https://doi.org/10.1038/s41598-018-23217-5.Search in Google Scholar PubMed PubMed Central

9. Khetani, S, Mohammadi, M, Nezhad, AS. Filter-based isolation, enrichment, and characterization of circulating tumor cells. Biotechnol Bioeng 2018;115:2504–29. https://doi.org/10.1002/bit.26787.Search in Google Scholar PubMed

10. Park, ES, Yan, JP, Ang, RA, Lee, JH, Deng, X, Duffy, SP, et al.. Isolation and genome sequencing of individual circulating tumor cells using hydrogel encapsulation and laser capture microdissection. Lab Chip 2018;18:1736–49. https://doi.org/10.1039/c8lc00184g.Search in Google Scholar PubMed

11. Waheed, W, Alazzam, A, Mathew, B, Christoforou, N, Abu-Nada, E. Lateral fluid flow fractionation using dielectrophoresis (LFFF-DEP) for size-independent, label-free isolation of circulating tumor cells. J Chromatogr B Analyt Technol Biomed Life Sci 2018;1087–1088:133–7. https://doi.org/10.1016/j.jchromb.2018.04.046.Search in Google Scholar PubMed

12. Watanabe, M, Kenmotsu, H, Ko, R, Wakuda, K, Ono, A, Imai, H, et al.. Isolation and molecular analysis of circulating tumor cells from lung cancer patients using a microfluidic chip type cell sorter. Canc Sci 2018;109:2539–48.10.1111/cas.13692Search in Google Scholar PubMed PubMed Central

13. Yang, C, Zhang, N, Wang, S, Shi, D, Zhang, C, Liu, K, et al.. Wedge-shaped microfluidic chip for circulating tumor cells isolation and its clinical significance in gastric cancer. J Transl Med 2018;16:139. https://doi.org/10.1186/s12967-018-1521-8.Search in Google Scholar PubMed PubMed Central

14. Ferro, M, La Civita, E, Liotti, A, Cennamo, M, Tortora, F, Buonerba, C, et al.. Liquid biopsy biomarkers in urine: a route towards molecular diagnosis and personalized medicine of bladder cancer. J Personalized Med 2021;11:237.10.3390/jpm11030237Search in Google Scholar PubMed PubMed Central

15. Flaig, TW, Wilson, S, van Bokhoven, A, Varella-Garcia, M, Wolfe, P, Maroni, P, et al.. Detection of circulating tumor cells in metastatic and clinically localized urothelial carcinoma. Urology 2011;78:863–7. https://doi.org/10.1016/j.urology.2011.05.045.Search in Google Scholar PubMed PubMed Central

16. Gazzaniga, P, Gradilone, A, de Berardinis, E, Busetto, GM, Raimondi, C, Gandini, O, et al.. Prognostic value of circulating tumor cells in nonmuscle invasive bladder cancer: a CellSearch analysis. Ann Oncol 2012;23:2352–6. https://doi.org/10.1093/annonc/mdr619.Search in Google Scholar PubMed

17. Guzzo, TJ, McNeil, BK, Bivalacqua, TJ, Elliott, DJ, Sokoll, LJ, Schoenberg, MP. The presence of circulating tumor cells does not predict extravesical disease in bladder cancer patients prior to radical cystectomy. Urol Oncol 2012;30:44–8. https://doi.org/10.1016/j.urolonc.2009.10.008.Search in Google Scholar PubMed

18. Rink, M, Chun, FK, Minner, S, Friedrich, M, Mauermann, O, Heinzer, H, et al.. Detection of circulating tumour cells in peripheral blood of patients with advanced non-metastatic bladder cancer. BJU Int 2011;107:1668–75. https://doi.org/10.1111/j.1464-410x.2010.09562.x.Search in Google Scholar PubMed

19. Soave, A, Riethdorf, S, Dahlem, R, von Amsberg, G, Minner, S, Weisbach, L, et al.. A nonrandomized, prospective, clinical study on the impact of circulating tumor cells on outcomes of urothelial carcinoma of the bladder patients treated with radical cystectomy with or without adjuvant chemotherapy. Int J Canc 2017;140:381–9. https://doi.org/10.1002/ijc.30445.Search in Google Scholar PubMed

20. Abrahamsson, J, Aaltonen, K, Engilbertsson, H, Liedberg, F, Patschan, O, Rydén, L, et al.. Circulating tumor cells in patients with advanced urothelial carcinoma of the bladder: association with tumor stage, lymph node metastases, FDG-PET findings, and survival. Urol Oncol 2017;35:606 e9-16. https://doi.org/10.1016/j.urolonc.2017.05.021.Search in Google Scholar PubMed

21. Naoe, M, Ogawa, Y, Morita, J, Omori, K, Takeshita, K, Shichijyo, T, et al.. Detection of circulating urothelial cancer cells in the blood using the CellSearch System. Cancer 2007;109:1439–45. https://doi.org/10.1002/cncr.22543.Search in Google Scholar

22. Okegawa, T, Hayashi, K, Hara, H, Nutahara, K, Higashihara, E. Immunomagnetic quantification of circulating tumor cells in patients with urothelial cancer. Int J Urol 2010;17:254–8. https://doi.org/10.1111/j.1442-2042.2010.02454.x.Search in Google Scholar

23. Zhang, Z, Fan, W, Deng, Q, Tang, S, Wang, P, Xu, P, et al.. The prognostic and diagnostic value of circulating tumor cells in bladder cancer and upper tract urothelial carcinoma: a meta-analysis of 30 published studies. Oncotarget 2017;8:59527–38. https://doi.org/10.18632/oncotarget.18521.Search in Google Scholar

24. Abbosh, PH, Rosenberg, JE, Plimack, ER. Circulating biomarkers to guide systemic therapy for urothelial carcinoma. Urol Oncol 2016;34:502–9. https://doi.org/10.1016/j.urolonc.2016.08.019.Search in Google Scholar

25. Cegan, M, Kolostova, K, Matkowski, R, Broul, M, Schraml, J, Fiutowski, M, et al.. In vitro culturing of viable circulating tumor cells of urinary bladder cancer. Int J Clin Exp Pathol 2014;7:7164–71.Search in Google Scholar

26. Gradilone, A, Petracca, A, Nicolazzo, C, Gianni, W, Cortesi, E, Naso, G, et al.. Prognostic significance of survivin-expressing circulating tumour cells in T1G3 bladder cancer. BJU Int 2010;106:710–5. https://doi.org/10.1111/j.1464-410x.2009.09130.x.Search in Google Scholar

27. Cowan, NC, Crew, JP. Imaging bladder cancer. Curr Opin Urol 2010;20:409–13. https://doi.org/10.1097/mou.0b013e32833cbcb9.Search in Google Scholar

28. Osman, I, Kang, M, Lee, A, Deng, FM, Polsky, D, Mikhail, M, et al.. Detection of circulating cancer cells expressing uroplakins and epidermal growth factor receptor in bladder cancer patients. Int J Canc 2004;111:934–9. https://doi.org/10.1002/ijc.20366.Search in Google Scholar

29. Gudemann, CJ, Weitz, J, Kienle, P, Lacroix, J, Wiesel, MJ, Soder, M, et al.. Detection of hematogenous micrometastasis in patients with transitional cell carcinoma. J Urol 2000;164:532–6. https://doi.org/10.1016/s0022-5347(05)67418-6.Search in Google Scholar

30. Anantharaman, A, Friedlander, T, Lu, D, Krupa, R, Premasekharan, G, Hough, J, et al.. Programmed death-ligand 1 (PD-L1) characterization of circulating tumor cells (CTCs) in muscle invasive and metastatic bladder cancer patients. BMC Canc 2016;16:744. https://doi.org/10.1186/s12885-016-2758-3.Search in Google Scholar PubMed PubMed Central

31. Alva, A, Friedlander, T, Clark, M, Huebner, T, Daignault, S, Hussain, M, et al.. Circulating tumor cells as potential biomarkers in bladder cancer. J Urol 2015;194:790–8. https://doi.org/10.1016/j.juro.2015.02.2951.Search in Google Scholar PubMed PubMed Central

32. Leow, JJ, Martin-Doyle, W, Rajagopal, PS, Patel, CG, Anderson, EM, Rothman, AT, et al.. Adjuvant chemotherapy for invasive bladder cancer: a 2013 updated systematic review and meta-analysis of randomized trials. Eur Urol 2014;66:42–54. https://doi.org/10.1016/j.eururo.2013.08.033.Search in Google Scholar PubMed

33. Flaig, TW, Tangen, CM, Daneshmand, S, Alva, A, Lerner, SP, Lucia, MS, et al.. A randomized phase II study of coexpression extrapolation (COXEN) with neoadjuvant chemotherapy for bladder cancer (SWOG S1314; NCT02177695). Clin Canc Res 2021;27:2435–41. https://doi.org/10.1158/1078-0432.ccr-20-2409.Search in Google Scholar

Published Online: 2021-09-27

© 2021 Felice Crocetto et al., published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 6.6.2024 from https://www.degruyter.com/document/doi/10.1515/jbcpp-2021-0233/html
Scroll to top button