Skip to main content

Advertisement

Log in

CTC clusters in cancer progression and metastasis

  • Review Article
  • Published:
Medical Oncology Aims and scope Submit manuscript

Abstract

Circulating tumor cells (CTC) in the blood of cancer patients are regarded as potential metastatic seeds, and their detailed characterization holds great promises for more exact prognosis, better diagnosis and therapy of the metastatic cancer. Circulating tumor cell clusters represent different class of CTCs, with specific properties, including high metastatic potential. In this review, we present current opinions on differences between single CTCs and CTC clusters, their mode of dissemination, methods of detection and clinical importance in various types of cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

CTC:

Circulating tumor cells

EMT:

Epithelial–mesenchymal transition

MET:

Mesenchymal–epithelial transition

ISET:

Isolation by size of epithelial tumor cells

FMSA:

Flexible micro spring array

FDA:

Food and Drug Administration

MBC:

Metastatic breast cancer

MCA:

Microcavity array

CEA:

Carcinoembryonic antigen

CK:

Cytokeratins

CTM:

Circulating tumor microemboli

TGF-β:

Transforming growth factor β

EpCAM:

Epithelial adhesion molecule

DDF:

Dean flow fractionation

HER2:

Human epidermal growth factor

mCRPC:

Metastatic castration-resistant prostate cancer

DAPI:

4′,6-diamidino-2-phenylindole

IF:

Immunofluorescence

PFS:

Progression-free survival

OS:

Overall survival

NSCLC:

Non-small cell lung cancer

FU:

Follow-up

ccRCC:

Clear cell renal cell carcinoma

PDAC:

Pancreatic ductal adenocarcinoma

References

  1. Massague J, Obenauf AC. Metastatic colonization by circulating tumour cells. Nature. 2016;529(7586):298–306. doi:10.1038/nature17038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ashworth T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J. 1869;14(3):146–9.

    Google Scholar 

  3. Watanabe S. The metastasizability of tumor cells. Cancer. 1954;7(2):215–23.

    Article  CAS  PubMed  Google Scholar 

  4. Paget G. Remarks on a case of alternate partial anaesthesia. Br Med J. 1889;1(1462):1–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–9. doi:10.1056/NEJM198612253152606.

    Article  CAS  PubMed  Google Scholar 

  6. Duda DG, Duyverman AM, Kohno M, Snuderl M, Steller EJ, Fukumura D, et al. Malignant cells facilitate lung metastasis by bringing their own soil. Proc Natl Acad Sci USA. 2010;107(50):21677–82. doi:10.1073/pnas.1016234107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158(5):1110–22. doi:10.1016/j.cell.2014.07.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Aceto N, Toner M, Maheswaran S, Haber DA. En route to metastasis: circulating tumor cell clusters and epithelial-to-mesenchymal transition. Trends Cancer. 2015;1(1):44–52. doi:10.1016/j.trecan.2015.07.006.

    Article  Google Scholar 

  9. Hong Y, Fang F, Zhang Q. Circulating tumor cell clusters: what we know and what we expect (review). Int J Oncol. 2016;. doi:10.3892/ijo.2016.3747.

    Google Scholar 

  10. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339(6119):580–4. doi:10.1126/science.1228522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cheung KJ, Padmanaban V, Silvestri V, Schipper K, Cohen JD, Fairchild AN, et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc Natl Acad Sci USA. 2016;113(7):E854–63. doi:10.1073/pnas.1508541113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Au SH, Storey BD, Moore JC, Tang Q, Chen YL, Javaid S, et al. Clusters of circulating tumor cells traverse capillary-sized vessels. Proc Natl Acad Sci USA. 2016;113(18):4947–52. doi:10.1073/pnas.1524448113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Koren S, Bentires-Alj M. Breast tumor heterogeneity: source of Fitness. Hurdle for Therapy. Mol Cell. 2015;60(4):537–46. doi:10.1016/j.molcel.2015.10.031.

    Article  CAS  PubMed  Google Scholar 

  14. Maddipati R, Stanger BZ. Pancreatic cancer metastases harbor evidence of polyclonality. Cancer Discov. 2015;5(10):1086–97. doi:10.1158/2159-8290.CD-15-0120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–54. doi:10.1038/nrc822.

    Article  CAS  PubMed  Google Scholar 

  16. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472–6. doi:10.1038/nature15748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial–mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20(5):576–90. doi:10.1016/j.ccr.2011.09.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cima I, Kong SL, Sengupta D, Tan IB, Phyo WM, Lee D, et al. Tumor-derived circulating endothelial cell clusters in colorectal cancer. Sci Transl Med. 2016;8(345):345ra89. doi:10.1126/scitranslmed.aad7369.

    Article  PubMed  Google Scholar 

  19. Cheung KJ, Ewald AJ. A collective route to metastasis: seeding by tumor cell clusters. Science. 2016;352(6282):167–9. doi:10.1126/science.aaf6546.

    Article  CAS  PubMed  Google Scholar 

  20. Alunni-Fabbroni M, Sandri MT. Circulating tumour cells in clinical practice: methods of detection and possible characterization. Methods. 2010;50(4):289–97. doi:10.1016/j.ymeth.2010.01.027.

    Article  CAS  PubMed  Google Scholar 

  21. Harouaka R, Kang Z, Zheng SY, Cao L. Circulating tumor cells: advances in isolation and analysis, and challenges for clinical applications. Pharmacol Ther. 2014;141(2):209–21. doi:10.1016/j.pharmthera.2013.10.004.

    Article  CAS  PubMed  Google Scholar 

  22. Mohamed H, Murray M, Turner JN, Caggana M. Isolation of tumor cells using size and deformation. J Chromatogr A. 2009;1216(47):8289–95. doi:10.1016/j.chroma.2009.05.036.

    Article  CAS  PubMed  Google Scholar 

  23. Tan SJ, Yobas L, Lee GY, Ong CN, Lim CT. Microdevice for the isolation and enumeration of cancer cells from blood. Biomed Microdevices. 2009;11(4):883–92. doi:10.1007/s10544-009-9305-9.

    Article  PubMed  Google Scholar 

  24. Zheng S, Lin H, Liu JQ, Balic M, Datar R, Cote RJ, et al. Membrane microfilter device for selective capture, electrolysis and genomic analysis of human circulating tumor cells. J Chromatogr A. 2007;1162(2):154–61. doi:10.1016/j.chroma.2007.05.064.

    Article  CAS  PubMed  Google Scholar 

  25. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo RA, Tan DS, et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci Rep. 2013;3:1259. doi:10.1038/srep01259.

    PubMed  PubMed Central  Google Scholar 

  26. Harouaka RA, Zhou MD, Yeh YT, Khan WJ, Das A, Liu X, et al. Flexible micro spring array device for high-throughput enrichment of viable circulating tumor cells. Clin Chem. 2014;60(2):323–33. doi:10.1373/clinchem.2013.206805.

    Article  CAS  PubMed  Google Scholar 

  27. Rosenberg R, Gertler R, Friederichs J, Fuehrer K, Dahm M, Phelps R, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry. 2002;49(4):150–8. doi:10.1002/cyto.10161.

    Article  CAS  PubMed  Google Scholar 

  28. Gertler R, Rosenberg R, Fuehrer K, Dahm M, Nekarda H, Siewert JR. Detection of circulating tumor cells in blood using an optimized density gradient centrifugation. Recent Results Cancer Res. 2003;162:149–55.

    Article  PubMed  Google Scholar 

  29. Naume B, Borgen E, Tossvik S, Pavlak N, Oates D, Nesland JM. Detection of isolated tumor cells in peripheral blood and in BM: evaluation of a new enrichment method. Cytotherapy. 2004;6(3):244–52. doi:10.1080/14653240410006086.

    Article  CAS  PubMed  Google Scholar 

  30. McNiece I, Briddell R, Stoney G, Kern B, Zilm K, Recktenwald D, et al. Large-scale isolation of CD34+ cells using the Amgen cell selection device results in high levels of purity and recovery. J Hematother. 1997;6(1):5–11. doi:10.1089/scd.1.1997.6.5.

    Article  CAS  PubMed  Google Scholar 

  31. Neurauter AA, Bonyhadi M, Lien E, Nokleby L, Ruud E, Camacho S, et al. Cell isolation and expansion using Dynabeads. Adv Biochem Eng Biotechnol. 2007;106:41–73. doi:10.1007/10_2007_072.

    CAS  PubMed  Google Scholar 

  32. Peters CE, Woodside SM, Eaves AC. Isolation of subsets of immune cells. Methods Mol Biol. 2005;302:95–116. doi:10.1385/1-59259-903-6:095.

    CAS  PubMed  Google Scholar 

  33. Lankiewicz S, Rivero BG, Bocher O. Quantitative real-time RT-PCR of disseminated tumor cells in combination with immunomagnetic cell enrichment. Mol Biotechnol. 2006;34(1):15–27. doi:10.1385/MB:34:1:15.

    Article  CAS  PubMed  Google Scholar 

  34. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351(8):781–91. doi:10.1056/NEJMoa040766.

    Article  CAS  PubMed  Google Scholar 

  35. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(19):3213–21. doi:10.1200/JCO.2007.15.8923.

    Article  PubMed  Google Scholar 

  36. Danila DC, Heller G, Gignac GA, Gonzalez-Espinoza R, Anand A, Tanaka E, et al. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13(23):7053–8. doi:10.1158/1078-0432.CCR-07-1506.

    Article  CAS  PubMed  Google Scholar 

  37. Cristofanilli M, Hayes DF, Budd GT, Ellis MJ, Stopeck A, Reuben JM, et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J Clin Oncol. 2005;23(7):1420–30. doi:10.1200/JCO.2005.08.140.

    Article  PubMed  Google Scholar 

  38. Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9(4):265–73. doi:10.1038/nrc2620.

    Article  CAS  PubMed  Google Scholar 

  39. Jansson S, Bendahl PO, Larsson AM, Aaltonen KE, Ryden L. Prognostic impact of circulating tumor cell apoptosis and clusters in serial blood samples from patients with metastatic breast cancer in a prospective observational cohort. BMC Cancer. 2016;16:433. doi:10.1186/s12885-016-2406-y.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Mu Z, Wang C, Ye Z, Austin L, Civan J, Hyslop T, et al. Prospective assessment of the prognostic value of circulating tumor cells and their clusters in patients with advanced-stage breast cancer. Breast Cancer Res Treat. 2015;154(3):563–71. doi:10.1007/s10549-015-3636-4.

    Article  CAS  PubMed  Google Scholar 

  41. Paoletti C, Li Y, Muniz MC, Kidwell KM, Aung K, Thomas DG, et al. Significance of circulating tumor cells in metastatic triple-negative breast cancer patients within a randomized, phase II trial: TBCRC 019. Clin Cancer Res. 2015;21(12):2771–9. doi:10.1158/1078-0432.CCR-14-2781.

    Article  CAS  PubMed  Google Scholar 

  42. Hou JM, Krebs MG, Lancashire L, Sloane R, Backen A, Swain RK, et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. 2012;30(5):525–32. doi:10.1200/JCO.2010.33.3716.

    Article  PubMed  Google Scholar 

  43. Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci USA. 2010;107(43):18392–7. doi:10.1073/pnas.1012539107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hosokawa M, Yoshikawa T, Negishi R, Yoshino T, Koh Y, Kenmotsu H, et al. Microcavity array system for size-based enrichment of circulating tumor cells from the blood of patients with small-cell lung cancer. Anal Chem. 2013;85(12):5692–8. doi:10.1021/ac400167x.

    Article  CAS  PubMed  Google Scholar 

  45. Hosokawa M, Kenmotsu H, Koh Y, Yoshino T, Yoshikawa T, Naito T, et al. Size-based isolation of circulating tumor cells in lung cancer patients using a microcavity array system. PLoS One. 2013;8(6):e67466. doi:10.1371/journal.pone.0067466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Molnar B, Ladanyi A, Tanko L, Sreter L, Tulassay Z. Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients. Clin Cancer Res. 2001;7(12):4080–5.

    CAS  PubMed  Google Scholar 

  47. McDaniel AS, Ferraldeschi R, Krupa R, Landers M, Graf R, Louw J, et al. Phenotypic diversity of circulating tumour cells in patients with metastatic castration-resistant prostate cancer. BJU Int. 2016;. doi:10.1111/bju.13631.

    PubMed  Google Scholar 

  48. Luo X, Mitra D, Sullivan RJ, Wittner BS, Kimura AM, Pan S, et al. Isolation and molecular characterization of circulating melanoma cells. Cell Rep. 2014;7(3):645–53. doi:10.1016/j.celrep.2014.03.039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sarioglu AF, Aceto N, Kojic N, Donaldson MC, Zeinali M, Hamza B, et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat Methods. 2015;12(7):685–91. doi:10.1038/nmeth.3404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cho EH, Wendel M, Luttgen M, Yoshioka C, Marrinucci D, Lazar D, et al. Characterization of circulating tumor cell aggregates identified in patients with epithelial tumors. Phys Biol. 2012;9(1):016001. doi:10.1088/1478-3975/9/1/016001.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kats-Ugurlu G, Roodink I, de Weijert M, Tiemessen D, Maass C, Verrijp K, et al. Circulating tumour tissue fragments in patients with pulmonary metastasis of clear cell renal cell carcinoma. J Pathol. 2009;219(3):287–93. doi:10.1002/path.2613.

    Article  CAS  PubMed  Google Scholar 

  52. Krebs MG, Hou JM, Sloane R, Lancashire L, Priest L, Nonaka D, et al. Analysis of circulating tumor cells in patients with non-small cell lung cancer using epithelial marker-dependent and -independent approaches. J Thorac Oncol. 2012;7(2):306–15. doi:10.1097/JTO.0b013e31823c5c16.

    Article  PubMed  Google Scholar 

  53. Liu MC, Shields PG, Warren RD, Cohen P, Wilkinson M, Ottaviano YL, et al. Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol. 2009;27(31):5153–9. doi:10.1200/JCO.2008.20.6664.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Molnar B, Floro L, Sipos F, Toth B, Sreter L, Tulassay Z. Elevation in peripheral blood circulating tumor cell number correlates with macroscopic progression in UICC stage IV colorectal cancer patients. Dis Markers. 2008;24(3):141–50.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Chang MC, Chang YT, Chen JY, Jeng YM, Yang CY, Tien YW, et al. Clinical significance of circulating tumor microemboli as a prognostic marker in patients with pancreatic ductal adenocarcinoma. Clin Chem. 2016;62(3):505–13. doi:10.1373/clinchem.2015.248260.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was funded by the National Science Centre (Grant 2014/14/M/NZ1/00437).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anna Fabisiewicz.

Ethics declarations

Conflict of interest

Anna Fabisiewicz declares that she has no conflict of interest. Ewa Grzybowska declares that she has no conflict of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Additional information

Anna Fabisiewicz and Ewa Grzybowska contributed equally to this paper.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fabisiewicz, A., Grzybowska, E. CTC clusters in cancer progression and metastasis. Med Oncol 34, 12 (2017). https://doi.org/10.1007/s12032-016-0875-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12032-016-0875-0

Keywords

Navigation