609
Views
14
CrossRef citations to date
0
Altmetric
Review

Circulating tumor cell clusters: Insights into tumour dissemination and metastasis

, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1139-1147 | Received 12 Sep 2020, Accepted 02 Nov 2020, Published online: 26 Nov 2020

References

  • Paget S. The distribution of secondary growths in cancer of the breast. Lancet. 1889;133:571–573.
  • Massagué J, Obenauf AC. Metastatic colonization by circulating tumor cells. Nature. 2016;529(7586):298–306.
  • Fidler IJ. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur J Cancer (1965).. 1973;9(3):223–227.
  • Liotta LA, Kleinerman J, Saldel GM. The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res. 1976;36(3):889–894.
  • Cho EH, Wendel M, Luttgen M, et al. Characterization of circulating tumor cell aggregates identified in patients with epithelial tumors. Phys Biol. 2012;9(1):016001.
  • Suo Y, Xie C, Zhu X, et al. Proportion of circulating tumor cell clusters increases during cancer metastasis. Cytometry Part A. 2017;91(3):250–253.
  • Aceto N, Bardia A, Miyamoto DT, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158(5):1110–1122.•• Key paper in the field highlighting the higher metastatic potential of CTC clusters compared to single CTCs
  • Harris TJ, Tepass U. Adherens junctions: from molecules to morphogenesis. Nat Rev Mol Cell Biol. 2010;11(7):502–514.
  • Gkountela S, Castro-Giner F, Szczerba BM, et al. Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell. 2019;176(1–2):98–112. e14.
  • Yu M, Bardia A, Wittner BS, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. science. 2013;339(6119):580–584.
  • Aceto N. Bring along your friends: homotypic and heterotypic circulating tumor cell clustering to accelerate metastasis. Biomed J. 2020;43(1):18–23.
  • Rianna C, Radmacher M, Kumar S. Direct evidence that tumor cells soften when navigating confined spaces. Mol Biol Cell. 2020;31(16):1726–1734
  • Moro A, Foresta E, Gasparini G, et al. Ameloblastic carcinoma of the maxilla: A case report and an updated review of the literature. Oncol Lett. 2016;12(6):4339–4350.
  • Au SH, Storey BD, Moore JC, et al. Clusters of circulating tumor cells traverse capillary-sized vessels. Proc Nat Acad Sci. 2016;113(18):4947–4952.•• An important paper demonstrating that CTC clusters can travel through narrow capillaries in a single-file and reform the cluster
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. cell. 2011;144(5):646–674.
  • Sawabata N. Circulating tumor cells in lung cancer: cluster circulating tumor cells as hybrid epithelial-mesenchymal transition/mesenchymal-epithelial transition (E/M). J Thorac Dis. 2017;9(10):3547–3550.
  • Lee JM, Dedhar S, Kalluri R, et al. The epithelial–mesenchymal transition: new insights in signaling, development, and disease. J cell Biol. 2006;172(7):973–981.
  • Zheng Q-M, Chen X-Y, Bao Q-F, et al. ILK enhances migration and invasion abilities of human endometrial stromal cells by facilitating the epithelial–mesenchymal transition. Gynecological Endocrinol. 2018;34(12):1091–1096.
  • Van Es JH, Barker N, Clevers H. You Wnt some, you lose some: oncogenes in the Wnt signaling pathway. Curr Opin Genet Dev. 2003;13(1):28–33.
  • Thompson EW, Newgreen DF. Carcinoma invasion and metastasis: a role for epithelial-mesenchymal transition? Cancer Res. 2005;65(14):5991–5995.
  • Zavadil J, Böttinger EP. TGF-β and epithelial-to-mesenchymal transitions. Oncogene. 2005;24(37):5764–5774.
  • Jin H, Varner J. Integrins: roles in cancer development and as treatment targets. Br J Cancer. 2004;90(3):561–565.
  • Thomas JR, Moore KM, Sproat C, et al. Integrin αVβ6-EGFR crosstalk regulates bidirectional force transmission and controls breast cancer invasion. bioRxiv. 2018;407908.
  • Mani SA, Guo W, Liao M-J, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715.
  • Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9(4):265–273.
  • Yochum ZA, Cades J, Wang H, et al. Targeting the EMT transcription factor TWIST1 overcomes resistance to EGFR inhibitors in EGFR-mutant non-small-cell lung cancer. Oncogene. 2019;38(5):656–670.
  • Khoo BL, Warkiani ME, Tan DS-W, et al. Clinical validation of an ultra high-throughput spiral microfluidics for the detection and enrichment of viable circulating tumor cells. PloS One. 2014;9(7):e99409.
  • De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013;13(2):97–110.
  • Vetter M, Landin J, Szczerba BM, et al. Denosumab treatment is associated with the absence of circulating tumor cells in patients with breast cancer. Breast Cancer Res. 2018;20(1):141.
  • Friedlander TW, Premasekharan G, Paris PL. Looking back, to the future of circulating tumor cells. Pharmacol Ther. 2014;142(3):271–280.
  • Szczerba BM, Castro-Giner F, Vetter M, et al. Neutrophils escort circulating tumor cells to enable cell cycle progression. Nature. 2019;566(7745):553–557.•• An important paper describing the composition of heterotypic CTC clusters and the presence of neutrophils within CTC clusters
  • Rostami P, Kashaninejad N, Moshksayan K, et al. Novel approaches in cancer management with circulating tumor cell clusters. J Sci. 2019;4(1):1–18.
  • Sprouse ML, Welte T, Boral D, et al. PMN-MDSCs enhance CTC metastatic properties through reciprocal interactions via ROS/Notch/Nodal signaling. Int J Mol Sci. 2019;20(8):1916.
  • Cheung KJ, Padmanaban V, Silvestri V, et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc Nat Acad Sci. 2016;113(7):E854–E863.
  • Brandt B, Junker R, Griwatz C, et al. Isolation of prostate-derived single cells and cell clusters from human peripheral blood. Cancer Res. 1996;56(20):4556–4561.
  • Jansson S, Bendahl P-O, Larsson A-M, et al. 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(1):433.
  • Murlidhar V, Reddy RM, Fouladdel S, et al. Poor prognosis indicated by venous circulating tumor cell clusters in early-stage lung cancers. Cancer Res. 2017;77(18):5194–5206.
  • Chistiakov DA, Chekhonin VP. Circulating tumor cells and their advances to promote cancer metastasis and relapse, with focus on glioblastoma multiforme. Exp Mol Pathol. 2018;105(2):166–174.
  • Mego M, Gao H, Cohen EN, et al. Circulating tumor cells (CTCs) are associated with abnormalities in peripheral blood dendritic cells in patients with inflammatory breast cancer. Oncotarget. 2017 May 30;8(22):35656–35668. PubMed PMID: 27374101; PubMed Central PMCID: PMCPMC5482606. eng.
  • Scheel C, Weinberg RA, Scheel C, et al., editors. Cancer stem cells and epithelial–mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012;22:396–403. Elsevier.
  • Kim MS, Sim TS, Kim YJ, et al. SSA-MOA: a novel CTC isolation platform using selective size amplification (SSA) and a multi-obstacle architecture (MOA) filter. Lab Chip. 2012;12(16):2874–2880.
  • Luzzi KJ, MacDonald IC, Schmidt EE, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153(3):865–873.
  • Wang C, Mu Z, Chervoneva I, et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res Treat. 2017;161(1):83–94.
  • Jacot W, Mazel M, Mollevi C, et al. Expression of PD-L1 on circulating breast cancer cells: correlation with clinicopathologic data and impact on prognosis. AACR. 2018;78(13 Suppl):Abstract nr 4589.
  • Kulasinghe A, Perry C, Kenny L, et al. PD-L1 expressing circulating tumor cells in head and neck cancers. BMC Cancer. 2017;17(1):333.
  • Brahmer J. PD-1-targeted immunotherapy: recent clinical findings. Clin Adv Hematol Oncol. 2012;10(10):674.
  • Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454.
  • Mazel M, Jacot W, Pantel K, et al. Frequent expression of PD-L1 on circulating breast cancer cells. Mol Oncol. 2015;9(9):1773–1782.
  • Strati A, Koutsodontis G, Papaxoinis G, et al. Prognostic significance of PD-L1 expression on circulating tumor cells in patients with head and neck squamous cell carcinoma. Ann Oncol. 2017;28(8):1923–1933.
  • Tian M, Zhang Y, Liu Z, et al. The PD-1/PD-L1 inhibitory pathway is altered in pre-eclampsia and regulates T cell responses in pre-eclamptic rats. Sci Rep. 2016;6(1):1–14.
  • DiDomenico J, Lamano JB, Oyon D, et al. The immune checkpoint protein PD-L1 induces and maintains regulatory T cells in glioblastoma. Oncoimmunology. 2018;7(7):e1448329.
  • Lin A, Yan W-H. Human leukocyte antigen-G (HLA-G) expression in cancers: roles in immune evasion, metastasis and target for therapy. Mol Med. 2015;21(1):782–791.
  • Leblanc R, Peyruchaud O. Metastasis: new functional implications of platelets and megakaryocytes. Blood J Am Soc Hematol. 2016;128(1):24–31.
  • Placke T, Örgel M, Schaller M, et al. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012;72(2):440–448.
  • Palumbo JS, Talmage KE, Massari JV, et al. Platelets and fibrin (ogen) increase metastatic potential by impeding natural killer cell–mediated elimination of tumor cells. Blood. 2005;105(1):178–185.
  • Gruber I, Landenberger N, Staebler A, et al. Relationship between circulating tumor cells and peripheral T-cells in patients with primary breast cancer. Anticancer Res. 2013;33(5):2233–2238.
  • Lou X-L, Sun J, Gong S-Q, et al. Interaction between circulating cancer cells and platelets: clinical implication. Chin J Cancer Res. 2015;27(5):450.
  • Piskounova E, Agathocleous M, Murphy MM, et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature. 2015;527(7577):186–191.
  • Alix-Panabières C. EPISPOT assay: detection of viable DTCs/CTCs in solid tumor patients. In: Minimal residual disease and circulating tumor cells in breast cancer. Vol. 195, Recent Results in Cancer Research. Berlin, Heidelberg: Springer; 2012. p. 69–76.
  • Yu M, Stott S, Toner M, et al. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192(3):373–382.
  • Königsberg R, Obermayr E, Bises G, et al. Detection of EpCAM positive and negative circulating tumor cells in metastatic breast cancer patients. Acta Oncologica. 2011;50(5):700–710.
  • Kulasinghe A, Zhou J, Kenny L, et al. Capture of circulating tumor cell clusters using straight microfluidic chips. Cancers (Basel). 2019;11(1):89.
  • Pantel K, Alix-Panabières C. Circulating tumor cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010;16(9):398–406.
  • Zeinali M, Lee M, Nadhan A, et al. High-throughput label-free isolation of heterogeneous circulating tumor cells and CTC clusters from non-small-cell lung cancer patients. Cancers (Basel). 2020;12(1):127.
  • Alix-Panabières C, Pantel K. Challenges in circulating tumor cell research. Nat Rev Cancer. 2014;14(9):623–631.
  • Poudineh M, Sargent EH, Pantel K, et al. Profiling circulating tumor cells and other biomarkers of invasive cancers. Nat Biomed Eng. 2018;2(2):72–84.
  • Alshareef M, Metrakos N, Juarez Perez E, et al. Separation of tumor cells with dielectrophoresis-based microfluidic chip. Biomicrofluidics. 2013;7(1):011803.
  • Tang Y, Shi J, Li S, et al. Microfluidic device with integrated microfilter of conical-shaped holes for high efficiency and high purity capture of circulating tumor cells. Sci Rep. 2014;4:6052.
  • Wan S, Kim TH, Smith KJ, et al. New labyrinth microfluidic device detects circulating tumor cells expressing cancer stem cell marker and circulating tumor microemboli in hepatocellular carcinoma. Sci Rep. 2019 Dec 09;9(1):18575.
  • Fachin F, Spuhler P, Martel-Foley JM, et al. Monolithic chip for high-throughput blood cell depletion to sort rare circulating tumor cells. Sci Rep. 2017;7(1):1–11.
  • Zhou J, Kulasinghe A, Bogseth A, et al. Isolation of circulating tumor cells in non-small-cell-lung-cancer patients using a multi-flow microfluidic channel. Microsys Nanoeng. 2019;5(1):1–12.
  • Qian W, Zhang Y, Chen W. Capturing cancer: emerging microfluidic technologies for the capture and characterization of circulating tumor cells. Small. 2015;11(32):3850–3872.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.