Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics

Abstract

Circulating tumor cells (CTCs) are rare cancer cells that are shed from primary or metastatic tumors into the peripheral blood circulation. Phenotypic and genetic characterization of these rare cells can provide important information to guide cancer staging and treatment, and thus further research into their characteristics and properties is an area of considerable interest. In this protocol, we describe detailed procedures for the production and use of a label-free spiral microfluidic device to allow size-based isolation of viable CTCs using hydrodynamic forces that are present in curvilinear microchannels. This spiral system enables us to achieve ≥85% recovery of spiked cells across multiple cancer cell lines and 99.99% depletion of white blood cells in whole blood. The described spiral microfluidic devices can be produced at an extremely low cost using standard microfabrication and soft lithography techniques (2–3 d), and they can be operated using two syringe pumps for lysed blood samples (7.5 ml in 12.5 min for a three-layered multiplexed chip). The fast processing time and the ability to collect CTCs from a large patient blood volume allows this technique to be used experimentally in a broad range of potential genomic and transcriptomic applications.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Overview of CTC isolation using spiral microfluidics.
Figure 2: Schematic illustration and working principle of particle or cell focusing in straight and curvilinear microchannels.
Figure 3: Illustration of different steps for fabrication of spiral microfluidic chips.
Figure 4: Characterization of the spiral biochips for CTC isolation.
Figure 5: Immunostaining of enriched CTCs from clinical patient blood samples.
Figure 6: Validation of the spiral inertial biochip for clinical analysis.

Similar content being viewed by others

References

  1. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    Google Scholar 

  2. Esmaeilsabzali, H., Beischlag, T.V., Cox, M.E., Parameswaran, A.M. & Park, E.J. Detection and isolation of circulating tumor cells: principles and methods. Biotechnol. Adv. 31, 1063–1084 (2013).

    Article  CAS  Google Scholar 

  3. Gupta, G.P. & Massagué, J. Cancer metastasis: building a framework. Cell 127, 679–695 (2006).

    Article  CAS  Google Scholar 

  4. van de Stolpe, A., Pantel, K., Sleijfer, S., Terstappen, L.W. & den Toonder, J.M. Circulating tumor cell isolation and diagnostics: toward routine clinical use. Cancer Res. 71, 5955–5960 (2011).

    Article  CAS  Google Scholar 

  5. Ashworth, T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Australian Med. J. 14, 146–149 (1869).

    Google Scholar 

  6. Wittekind, C. & Neid, M. Cancer invasion and metastasis. Oncology 69, 14–16 (2005).

    Article  Google Scholar 

  7. Racila, E. et al. Detection and characterization of carcinoma cells in the blood. Proc. Natl. Acad. Sci. 95, 4589–4594 (1998).

    Article  CAS  Google Scholar 

  8. Adalsteinsson, V.A. & Love, J.C. Toward engineered processes for sequencing-based analysis of single circulating tumor cells. Curr. Opin. Chem. Eng. 4, 97–104 (2014).

    Article  Google Scholar 

  9. Majid, E.W. & Lim, C.T. Microfluidic Platforms for Human Disease Cell Mechanics Studies (Springer, 2013).

  10. Lara, O., Tong, X., Zborowski, M. & Chalmers, J.J. Enrichment of rare cancer cells through depletion of normal cells using density and flow-through, immunomagnetic cell separation. Exp. Hematol. 32, 891–904 (2004).

    Article  Google Scholar 

  11. De Giorgi, V. et al. Application of a filtration-and isolation-by-size technique for the detection of circulating tumor cells in cutaneous melanoma. J. Invest. Dermatol. 130, 2440–2447 (2010).

    Article  CAS  Google Scholar 

  12. Warkiani, M.E. et al. Isoporous micro/nanoengineered membranes. ACS Nano 7, 1882–1904 (2013).

    Article  CAS  Google Scholar 

  13. Matas, J.-P., Morris, J.F. & Guazzelli, E. Inertial migration of rigid spherical particles in Poiseuille flow. J. Fluid Mech. 515, 171–195 (2004).

    Article  Google Scholar 

  14. Whitesides, G.M. The origins and the future of microfluidics. Nature 442, 368–373 (2006).

    Article  CAS  Google Scholar 

  15. Schwartz, M. Molecular characterization of CTCs. Genet. Eng. Biotechnol. News 33, 36–37 (2013).

    Article  Google Scholar 

  16. Nagrath, S. et al. Isolation of rare circulating tumor cells in cancer patients by microchip technology. Nature 450, 1235–1239 (2007).

    Article  CAS  Google Scholar 

  17. Gleghorn, J.P. et al. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip 10, 27–29 (2010).

    Article  CAS  Google Scholar 

  18. Chen, W. et al. Nanoroughened surfaces for efficient capture of circulating tumor cells without using capture antibodies. ACS Nano 7, 566–575 (2012).

    Article  Google Scholar 

  19. Wang, S. et al. Highly efficient capture of circulating tumor cells by using nanostructured silicon substrates with integrated chaotic micromixers. Angew. Chem. Int. Ed. Engl. 50, 3084–3088 (2011).

    Article  CAS  Google Scholar 

  20. Allan, A.L. & Keeney, M. Circulating tumor cell analysis: technical and statistical considerations for application to the clinic. J. Oncol. 2010, 426218 (2010).

    Article  Google Scholar 

  21. Thiery, J.P. & Lim, C.T. Tumor dissemination: an EMT affair. Cancer Cell 23, 272–273 (2013).

    Article  CAS  Google Scholar 

  22. Prang, N. et al. Cellular and complement-dependent cytotoxicity of EpCAM-specific monoclonal antibody MT201 against breast cancer cell lines. Br. J. cancer 92, 342–349 (2005).

    Article  CAS  Google Scholar 

  23. Maheswaran, S. et al. Detection of mutations in EGFR in circulating lung-cancer cells. N. Engl. J. Med. 359, 366–377 (2008).

    Article  CAS  Google Scholar 

  24. Riethdorf, S. et al. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin. Cancer Res. 16, 2634–2645 (2010).

    Article  CAS  Google Scholar 

  25. Lacroix, M. Significance, detection and markers of disseminated breast cancer cells. Endocr. Relat. Cancer 13, 1033–1067 (2006).

    Article  CAS  Google Scholar 

  26. Kemmner, W. Currently used markers for CTC isolation-advantages, limitations and impact on cancer prognosis. J. Clin. Exp. Pathol. 1, 102 (2011).

    Google Scholar 

  27. Pecot, C.V. et al. A novel platform for detection of CK+ and CK CTCs. Cancer Discov. 1, 580–586 (2011).

    Article  CAS  Google Scholar 

  28. Sieuwerts, A.M. et al. Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumor cells. J. Natl. Cancer Inst. 101, 61–66 (2009).

    Article  CAS  Google Scholar 

  29. Fehm, T. et al. Methods for isolating circulating epithelial cells and criteria for their classification as carcinoma cells. Cytotherapy 7, 171–185 (2005).

    Article  CAS  Google Scholar 

  30. Pachmann, K. et al. Standardized quantification of circulating peripheral tumor cells from lung and breast cancer. Clin. Chem. Lab. Med. 43, 617–627 (2005).

    Article  CAS  Google Scholar 

  31. Tkaczuk, K.H.R. et al. The significance of circulating epithelial cells in breast cancer patients by a novel negative selection method. Breast Cancer Res. Treat. 111, 355–364 (2008).

    Article  Google Scholar 

  32. Alix-Panabières, C. et al. Detection of circulating prostate-specific antigen–secreting cells in prostate cancer patients. Clin. Chem. 51, 1538–1541 (2005).

    Article  Google Scholar 

  33. Ozkumur, E. et al. Inertial focusing for tumor antigen–dependent and–independent sorting of rare circulating tumor cells. Sci. Transl. Med. 5, 179ra47 (2013).

    Article  Google Scholar 

  34. Yang, L. et al. Optimization of an enrichment process for circulating tumor cells from the blood of head and neck cancer patients through depletion of normal cells. Biotechnol. Bioeng. 102, 521–534 (2009).

    Article  CAS  Google Scholar 

  35. Munz, M. et al. Side-by-side analysis of five clinically tested anti-EpCAM monoclonal antibodies. Cancer Cell Int. 10, 44 (2010).

    Article  Google Scholar 

  36. Chaudhuri, P.K., Ebrahimi Warkiani, E., Jing, T., Kenry & Lim, C.T. Microfluidics for research and applications in oncology. Analyst http://dx.doi.org/10.1039/C5AN00382B (2015).

  37. Hyun, K.A., Kwon, K., Han, H., Kim, S.I. & Jung, H.I. Microfluidic flow fractionation device for label-free isolation of circulating tumor cells (CTCs) from breast cancer patients. Biosens. Bioelectron. 40, 206–212 (2013).

    Article  CAS  Google Scholar 

  38. Tan, S.J. et al. Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients. Biosens. Bioelectron. 26, 1701–1705 (2010).

    Article  CAS  Google Scholar 

  39. Warkiani, M.E. et al. Slanted spiral microfluidics for the ultra-fast, label-free isolation of circulating tumor cells. Lab Chip 14, 128–137 (2014).

    Article  CAS  Google Scholar 

  40. Gertler, R. et al. Detection of circulating tumor cells in blood using an optimized density gradient centrifugation. Recent Results Cancer Res. 162, 149–155 (2003).

    Article  Google Scholar 

  41. Hur, S.C., Henderson-MacLennan, N.K., McCabe, E.R. & Di Carlo, D. Deformability-based cell classification and enrichment using inertial microfluidics. Lab Chip 11, 912–920 (2011).

    Article  CAS  Google Scholar 

  42. Moon, H.S. et al. Continuous separation of breast cancer cells from blood samples using multi-orifice flow fractionation (MOFF) and dielectrophoresis (DEP). Lab Chip 11, 1118–1125 (2011).

    Article  CAS  Google Scholar 

  43. Huang, S.B. et al. High-purity and label-free isolation of circulating tumor cells (CTCs) in a microfluidic platform by using optically-induced-dielectrophoretic (ODEP) force. Lab Chip 13, 1371–1383 (2013).

    Article  CAS  Google Scholar 

  44. Cima, I. et al. Label-free isolation of circulating tumor cells in microfluidic devices: current research and perspectives. Biomicrofluidics 7, 011810 (2013).

    Article  Google Scholar 

  45. Gao, D. et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014).

    Article  CAS  Google Scholar 

  46. Sollier, E. et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip 14, 63–77 (2014).

    Article  CAS  Google Scholar 

  47. Karabacak, N.M. et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat. Protoc. 9, 694–710 (2014).

    Article  CAS  Google Scholar 

  48. Hou, H. et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 3, 1259 (2013).

    Article  Google Scholar 

  49. Warkiani, M.E. et al. An ultra-high-throughput spiral microfluidic biochip for the enrichment of circulating tumor cells. Analyst 139, 3245–3255 (2014).

    Article  CAS  Google Scholar 

  50. Khoo, B.L. et al. Clinical validation of an ultra-high-throughput spiral microfluidics for the detection and enrichment of viable circulating tumor cells. PLoS ONE 9, e99409 (2014).

    Article  Google Scholar 

  51. Segre, G. Radial particle displacements in Poiseuille flow of suspensions. Nature 189, 209–210 (1961).

    Article  Google Scholar 

  52. Segre, G. & Silberberg, A. Behaviour of macroscopic rigid spheres in Poiseuille flow Part 2. Experimental results and interpretation. J. Fluid Mech. 14, 136–157 (1962).

    Article  Google Scholar 

  53. Karnis, A., Goldsmith, H. & Mason, S. The flow of suspensions through tubes: V. Inertial effects. Can. J. Chem. Eng. 44, 181–193 (1966).

    Article  CAS  Google Scholar 

  54. Tachibana, M. On the behaviour of a sphere in the laminar tube flows. Rheol. Acta 12, 58–69 (1973).

    Article  Google Scholar 

  55. Choi, Y.-S., Seo, K.-W. & Lee, S.-J. Lateral and cross-lateral focusing of spherical particles in a square microchannel. Lab Chip 11, 460–465 (2011).

    Article  CAS  Google Scholar 

  56. Di Carlo, D., Irimia, D., Tompkins, R.G. & Toner, M. Continuous inertial focusing, ordering, and separation of particles in microchannels. Proc. Natl. Acad. Sci. 104, 18892–18897 (2007).

    Article  CAS  Google Scholar 

  57. Bhagat, A.A.S., Kuntaegowdanahalli, S.S. & Papautsky, I. Continuous particle separation in spiral microchannels using dean flows and differential migration. Lab Chip 8, 1906–1914 (2008).

    Article  CAS  Google Scholar 

  58. Dean, W. Fluid motion in a curved channel. Proc. R. Soci. Lond. 121, 402–420 (1928).

    Article  Google Scholar 

  59. Di Carlo, D. Inertial microfluidics. Lab Chip 9, 3038–3046 (2009).

    Article  CAS  Google Scholar 

  60. Hayes, D.F. & Smerage, J.B. Circulating tumor cells. Prog. Mol. Biol. Transl. Sci. 95, 95–112 (2009).

    Article  Google Scholar 

  61. Kuntaegowdanahalli, S.S., Bhagat, A.A.S., Kumar, G. & Papautsky, I. Inertial microfluidics for continuous particle separation in spiral microchannels. Lab Chip 9, 2973–2980 (2009).

    Article  CAS  Google Scholar 

  62. Khoo, B. et al. Ultra-high throughput enrichment of viable circulating tumor cells. in The 15th International Conference on Biomedical Engineering (ICBME 2013, 4th 43, 1–4 (Springer).

  63. Cristofanilli, M. et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 23, 1420–1430 (2005).

    Article  Google Scholar 

  64. Smerage, J. & Hayes, D. The measurement and therapeutic implications of circulating tumour cells in breast cancer. Br. J. Cancer 94, 8–12 (2006).

    Article  CAS  Google Scholar 

  65. Yu, M. et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339, 580–584 (2013).

    Article  CAS  Google Scholar 

  66. Payne, R. et al. Viable circulating tumour cell detection using multiplex RNA in situ hybridisation predicts progression-free survival in metastatic breast cancer patients. Br. J. Cancer 106, 1790–1797 (2012).

    Article  CAS  Google Scholar 

  67. Yu, M. et al. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 345, 216–220 (2014).

    Article  CAS  Google Scholar 

  68. Zhang, L. et al. The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci. Transl. Med. 5, 180ra148 (2013).

    Article  Google Scholar 

  69. Khoo, B.L. et al. Short-term expansion of breast circulating cancer cells predicts response to anti-cancer therapy. Oncotarget 6, 15578–15593 (2015).

    Article  Google Scholar 

  70. Dittrich, P.S. & Manz, A. Lab-on-a-chip: microfluidics in drug discovery. Nat. Rev. Drug Discov. 5, 210–218 (2006).

    Article  CAS  Google Scholar 

  71. Wu, T.-L. et al. Cell-free DNA: measurement in various carcinomas and establishment of normal reference range. Clin. Chim. Acta 321, 77–87 (2002).

    Article  CAS  Google Scholar 

  72. Aceto, N. et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 158, 1110–1122 (2014).

    Article  CAS  Google Scholar 

  73. Borgen, E. et al. Standardization of the immunocytochemical detection of cancer cells in BM and blood: I. establishment of objective criteria for the evaluation of immunostained cells. Cytotherapy 1, 377–388 (1999).

    Article  CAS  Google Scholar 

  74. Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. & Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 100, 3983–3988 (2003).

    Article  CAS  Google Scholar 

  75. Li, X. et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl. Cancer Inst. 100, 672–679 (2008).

    Article  CAS  Google Scholar 

  76. Friedl, P. & Gilmour, D. Collective cell migration in morphogenesis, regeneration and cancer. Nat. Rev. Mol. Cell Biol. 10, 445–457 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We express our sincere gratitude to all patients and healthy volunteers who participated in this trial and donated blood samples for characterization of our device. Financial support by the Singapore-MIT Alliance for Research and Technology (SMART) Center (BioSyM IRG) is gratefully acknowledged. This work is also supported by the use of NTU's Micro-Machine Center (MMC) facilities for wafer fabrication and the lab facilities at the Mechanobiology Institute (MBI) and the Nano Biomechanics Laboratory at the National University of Singapore. The clinical sample and data collection was supported by the Singapore National Medical Research Council grant no. NMRC 1225/2009.

Author information

Authors and Affiliations

Authors

Contributions

M.E.W., B.L.K., L.W., A.K.P.T., A.A.S.B. and C.T.L. contributed to the design of the spiral biochips. M.E.W., B.L.K., L.W., A.K.P.T. and A.A.S.B. prepared the manuscript and J.H. and C.T.L. commented on the manuscript.

Corresponding authors

Correspondence to Majid Ebrahimi Warkiani, Jongyoon Han or Chwee Teck Lim.

Ethics declarations

Competing interests

A.A.S.B. works for Clearbridge Biomedics Pte Ltd, which is commercializing the spiral biochips for cancer cell enrichment.

Integrated supplementary information

Supplementary Figure 1 Recovery efficiency of spiral biochip device.

(a) Histogram plot indicating a high separation efficiency of ~ 90% for different cancer cell lines tested. All error bars represent standard deviation (SD) of triplicates. (b) Diagram showing the recovery of MCF-7 cells spiked into lysed blood at clinically relevant concentrations.

Supplementary Figure 2 Optical image of a metallic mold made by conventional micromilling in stainless steel.

Supplementary Figure 3 The workstation setup for CTC separation from lysed blood.

The lysed blood and one sheath flow are pumped through the single (or multiplexed) spiral biochip using 2 different syringe pumps where CTCs are separated from other blood components rapidly and efficiently.

Supplementary Figure 4 Schematic diagram illustrating the correct assembly of single (or multiplexed) spiral biochip for CTC isolation from lysed blood.

For the multiplexed biochip (i.e., 3 spirals), the flow rate of sample and sheath flow must be multiplied by three.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4, Supplementary Tables 1–3 (PDF 808 kb)

Supplementary Data 1

Spiral CAD design for manufacture of a biochip. (ZIP 946 kb)

High-speed video illustrating the complete focusing of WBCs, platelets and RBC residuals after lysis to the outer wall of the channel from a healthy blood sample.

WBCs are clearly distinguished from platelets and RBC residuals based on size/morphology and phase contrast. (AVI 588 kb)

41596_2016_BFnprot2016003_MOESM283_ESM.avi

High-speed video illustrating the complete isolation of MDA-MB-231 cells from WBCs (lysed blood) at the device outlet using a single spiral biochip. (AVI 706 kb)

High-speed video captured at the outlet of a spiral biochip showing isolation of a CTC cluster from the peripheral blood of a patient with advanced metastatic lung cancer.

This movie clearly demonstrates the performance of our device for efficient enrichment of CTCs and micro-clusters from blood samples. (AVI 375 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Warkiani, M., Khoo, B., Wu, L. et al. Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics. Nat Protoc 11, 134–148 (2016). https://doi.org/10.1038/nprot.2016.003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2016.003

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer