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:

On-chip human microvasculature assay for visualization and quantification of tumor cell extravasation dynamics

Abstract

Distant metastasis, which results in >90% of cancer-related deaths, is enabled by hematogenous dissemination of tumor cells via the circulation. This requires the completion of a sequence of complex steps including transit, initial arrest, extravasation, survival and proliferation. Increased understanding of the cellular and molecular players enabling each of these steps is key to uncovering new opportunities for therapeutic intervention during early metastatic dissemination. As a protocol extension, this article describes an adaptation to our existing protocol describing a microfluidic platform that offers additional applications. This protocol describes an in vitro model of the human microcirculation with the potential to recapitulate discrete steps of early metastatic seeding, including arrest, transendothelial migration and early micrometastases formation. The microdevice features self-organized human microvascular networks formed over 4–5 d, after which the tumor can be perfused and extravasation events are easily tracked over 72 h via standard confocal microscopy. Contrary to most in vivo and in vitro extravasation assays, robust and rapid scoring of extravascular cells, combined with high-resolution imaging, can be easily achieved because of the confinement of the vascular network to one plane close to the surface of the device. This renders extravascular cells clearly distinct and allows tumor cells of interest to be identified quickly as compared with those in thick tissues. The ability to generate large numbers of devices (36) per experiment further allows for highly parametric studies, which are required when testing multiple genetic or pharmacological perturbations. This is coupled with the capability for live tracking of single-cell extravasation events, allowing both tumor and endothelial morphological dynamics to be observed in high detail with a moderate number of data points.

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

Access options

Buy this article

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

Figure 1: Schematics of device fabrication and cell–fibrin gel seeding protocol.
Figure 2: Microvessel bed formation and tumor cell perfusion procedure.
Figure 3: Scoring and quantification of extravasation efficiency.
Figure 4: Tracking and quantification of extravasation kinetics.
Figure 5: Resolving temporal–spatial organization of endothelial and tumor cell proteins at a single-cell level.
Figure 6: Beyond the endothelium: the basement membrane.
Figure 7: Addition of noncancer host cells.
Figure 8: Tracking tumor cell proliferation post extravasation.

Similar content being viewed by others

References

  1. Nguyen, D.X., Bos, P.D. & Massagué, J. Metastasis: from dissemination to organ-specific colonization. Nat. Rev. Cancer 9, 274–284 (2009).

    Article  CAS  Google Scholar 

  2. Shin, Y. et al. Microfluidic assay for simultaneous culture of multiple cell types on surfaces or within hydrogels. Nat. Protoc. 7, 1247–1259 (2012).

    Article  CAS  Google Scholar 

  3. Chen, M.B., Whisler, J.A., Jeon, J.S. & Kamm, R.D. Mechanisms of tumor cell extravasation in an in vitro microvascular network platform. Integr. Biol. 5, 1262–1271 (2013).

    Article  CAS  Google Scholar 

  4. Whisler, J.A., Chen, M.B. & Kamm, R.D. Control of perfusable microvascular network morphology using a multiculture microfluidic system. Tissue Eng. Part C. Methods 20, 543–552 (2014).

    Article  CAS  Google Scholar 

  5. Ehsan, S.M. et al. A three-dimensional in vitro model of tumor cell intravasation. Integr. Biol. 6, 603–610 (2015).

    Article  Google Scholar 

  6. Ghajar, C.M. et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013).

    Article  CAS  Google Scholar 

  7. Hsu, Y.-H. et al. Full range physiological mass transport control in 3D tissue cultures. Lab Chip 13, 81–89 (2012).

    Article  Google Scholar 

  8. Kim, S., Lee, H., Chung, M. & Jeon, N.L. Engineering of functional, perfusable 3D microvascular networks on a chip. Lab Chip 13, 1489–1500 (2013).

    Article  CAS  Google Scholar 

  9. Kim, J. et al. Implantable microfluidic device for the formation of three-dimensional vasculature by human endothelial progenitor cells. Biotechnol. Bioprocess Eng. 19, 379–385 (2014).

    Article  CAS  Google Scholar 

  10. Labelle, M. & Hynes, R.O. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2, 1091–1099 (2012).

    Article  CAS  Google Scholar 

  11. Labelle, M., Begum, S. & Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 20, 576–590 (2011).

    Article  CAS  Google Scholar 

  12. Kienast, Y. et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 16, 116–122 (2010).

    Article  CAS  Google Scholar 

  13. Francia, G., Cruz-munoz, W., Man, S., Xu, P. & Kerbel, R.S. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat. Rev. Cancer 11, 135–141 (2011).

    Article  CAS  Google Scholar 

  14. Kitamura, T. et al. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J. Exp. Med. 212, 1043–1059 (2015).

    Article  CAS  Google Scholar 

  15. Qian, B. et al. A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS One 4, e6562 (2009).

    Article  Google Scholar 

  16. Stoletov, K., Montel, V., Lester, R.D., Gonias, S.L. & Klemke, R. High-resolution imaging of the dynamic tumor cell vascular interface in transparent zebrafish. Proc. Natl. Acad. Sci. USA 104, 17406–17411 (2007).

    Article  CAS  Google Scholar 

  17. Leong, H.S. et al. Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis. Cell Rep. 8, 1558–1570 (2014).

    Article  CAS  Google Scholar 

  18. Koop, S. et al. Fate of melanoma cells entering the microcirculation: over 80% survive and extravasate. Cancer Res. 55, 2520–2523 (1995).

    CAS  PubMed  Google Scholar 

  19. Koop, S. et al. Independence of metastatic ability and extravasation: metastatic ras-transformed and control fibroblasts extravasate equally well. Proc. Natl. Acad. Sci. USA 93, 11080–11084 (1996).

    Article  CAS  Google Scholar 

  20. Labelle, M., Begum, S. & Hynes, R.O. Platelets guide the formation of early metastatic niches. Proc. Natl. Acad. Sci. USA 111, E3053–E3061 (2014).

    Article  CAS  Google Scholar 

  21. Roussos, E.T., Condeelis, J.S. & Patsialou, A. Chemotaxis in cancer. Nat. Rev. Cancer 11, 573–587 (2011).

    Article  CAS  Google Scholar 

  22. Albini, A. & Benelli, R. The chemoinvasion assay: a method to assess tumor and endothelial cell invasion and its modulation. Nat. Protoc. 2, 504–511 (2007).

    Article  CAS  Google Scholar 

  23. Mierke, C.T. Cancer cells regulate biomechanical properties of human microvascular endothelial cells. J. Biol. Chem. 286, 40025–40037 (2011).

    Article  CAS  Google Scholar 

  24. Chrobak, K.M., Potter, D.R. & Tien, J. Formation of perfused, functional microvascular tubes in vitro. Microvasc. Res. 71, 185–196 (2006).

    Article  CAS  Google Scholar 

  25. Zheng, Y. et al. In vitro microvessels for the study of angiogenesis and thrombosis. Proc. Natl. Acad. Sci. USA 109, 9342–9347 (2012).

    Article  CAS  Google Scholar 

  26. Kolesky, D.B., Homan, K.A., Skylar-Scott, M.A & Lewis, J.A. Three-dimensional bioprinting of thick vascularized tissues. Proc. Natl. Acad. Sci. USA 113, 3179–3184 (2016).

    Article  CAS  Google Scholar 

  27. Shin, M.K., Kim, S.K. & Jung, H. Integration of intra- and extravasation in one cell-based microfluidic chip for the study of cancer metastasis. Lab Chip 11, 3880–3887 (2011).

    Article  CAS  Google Scholar 

  28. Song, J.W. et al. Microfluidic endothelium for studying the intravascular adhesion of metastatic breast cancer cells. PLoS One 4, e5756 (2009).

    Article  Google Scholar 

  29. Jeon, J.S., Zervantonakis, I.K., Chung, S., Kamm, R.D. & Charest, J.L. In vitro model of tumor cell extravasation. PLoS One 8, e56910 (2013).

    Article  CAS  Google Scholar 

  30. Zervantonakis, I.K. et al. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc. Natl. Acad. Sci. USA 109, 13515–13520 (2012).

    Article  CAS  Google Scholar 

  31. Chaw, K.C., Manimaran, M., Tay, E.H. & Swaminathan, S. Multi-step microfluidic device for studying cancer metastasis. Lab Chip 7, 1041–1047 (2007).

    Article  CAS  Google Scholar 

  32. Zhang, Q., Liu, T. & Qin, J. A microfluidic-based device for study of transendothelial invasion of tumor aggregates in realtime. Lab Chip 12, 2837–2842 (2012).

    Article  CAS  Google Scholar 

  33. Roberts, S.A., Waziri, A.E. & Agrawal, N. Development of a single-cell migration and extravasation platform through selective surface modification. Anal. Chem. 88, 2770–2776 (2016).

    Article  CAS  Google Scholar 

  34. Riahi, R. et al. A microfluidic model for organ-specific extravasation of circulating tumor cells. Biomicrofluidics 8, 024103 (2014).

    Article  CAS  Google Scholar 

  35. Kim, Y. et al. Quantification of cancer cell extravasation in vivo. Nat. Protoc. 11, 937–948 (2016).

    Article  CAS  Google Scholar 

  36. Chen, M.B., Lamar, J.M., Li, R., Hynes, R.O. & Kamm, R.D. Elucidation of the roles of tumor integrin β1 in the extravasation stage of the metastasis cascade. Cancer Res. 76, 2513–2524 (2016).

    Article  CAS  Google Scholar 

  37. Stoletov, K. et al. Visualizing extravasation dynamics of metastatic tumor cells. J. Cell Sci. 123, 2332–2341 (2010).

    Article  CAS  Google Scholar 

  38. Jeon, J.S. et al. Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation. Proc. Natl. Acad. Sci. USA 112, 214–219 (2015).

    Article  CAS  Google Scholar 

  39. Albelda, S.M. et al. Permeability characteristics of cultured endothelial cell monolayers. J. Appl. Physiol. 64, 308–322 (1988).

    Article  CAS  Google Scholar 

  40. Quail, D.F. & Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    Article  CAS  Google Scholar 

  41. Kitamura, T., Qian, B.-Z. & Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 15, 73–86 (2015).

    Article  CAS  Google Scholar 

  42. Levario, T.J., Zhan, M., Lim, B., Shvartsman, S.Y. & Lu, H. Microfluidic trap array for massively parallel imaging of Drosophila embryos. Nat. Protoc. 8, 721–736 (2013).

    Article  Google Scholar 

  43. Spiegel, A. et al. Neutrophils suppress intraluminal NK-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 6, 630–649 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Chung for scientific discussions and A. Boussommier-Calleja for critical reading of the manuscript. We thank B. Bista and R. Hynes of the Department of Biology, Massachusetts Institute of Technology and J. Massague of the Sloan-Kettering Institute for sharing cell lines. M.B.C. and R.D.K. acknowledge support from the National Cancer Institute (CA202177). R.D.K. and J.A.W. thank the National Science Foundation for support (CBET-0939511).

Author information

Authors and Affiliations

Authors

Contributions

M.B.C., J.A.W. and R.D.K. conceived the project and designed the experiments; M.B.C. and C.Y. performed the experiments; J.F. designed fluorescent cell lines; M.B.C. analyzed the data; R.D.K. supervised the project; Y.S. designed the figure schematics, M.B.C. and R.D.K. wrote the paper.

Corresponding author

Correspondence to Roger D Kamm.

Ethics declarations

Competing interests

R.K. is a cofounder of and has a substantial financial interest in AIM Biotech, a company that has commercialized microfluid assays of design similar to the one described in the present protocol. All the reported studies, however, were performed with devices designed and fabricated at the Kamm laboratory at MIT.

Integrated supplementary information

Supplementary Figure 1 Shear stress determination in microvascular networks.

A pressure drop of ~4 mmH2O is applied across the vascular network using a suspension of 2 micron red polystyrene spheres in EGM. Velocity of beads (those near the centerline only) are calculated using the “streak-length method”1, and diameters of vessels are estimated via corresponding phase contrast images using Image J. Viscosity of media is assumed to be ~0.0008 Pa s. (A) Example fluorescent images of flowing beads taken at 50 fps with 20 ms exposure (note images shown are post-processed to be oversaturated to clearly show streaks. Actual quantification should be done with raw (properly exposed) images to ensure streak length calculations are correct). (B) Distribution of shear stresses found in 1 device over 30 vessel segments. Mean velocity is taken as half of the centerline velocity. Pipe flow is assumed as an approximation. (C) Table of individual values of diameter, centerline bead velocity and corresponding shear stress estimated for individual vessel segments in a single device. (D) Table of average shear values of 20 vessels per device, for a total of 10 separate devices. (E) Table of the average shear over 10 devices per experiment (20 vessels per device), for a total of 5 experiments.

1. Al-Khazraji, B. K., Novielli, N. M., Goldman, D., Medeiros, P. J. & Jackson, D. N. A Simple 'Streak Length Method' for Quantifying and Characterizing Red Blood Cell Velocity Profiles and Blood Flow in Rat Skeletal Muscle Arterioles. Microcirculation 19, 327–335 (2012).

Supplementary Figure 2 Lumens are surrounded on all sides by hydrogel.

Confocal reconstruction of various lumens formed in micro devices (white=reflectance; red=HUVEC; green=MDA-MB-213 LifeAct GFP). While most lumens lie in roughly in one plane, the surface of lumens are at least >30 microns away from the bottom glass and top PDMS layers.

Supplementary Figure 3 Determining perfusability of microvascular networks.

A perfusable device satisfies 2 criteria: (1) 50% of interpost regions on one side allow for tumor cell entry and (2) more than 25% of tumor cells in the network are distributed beyond the centerline of the gel region. (A) Histogram of the number of devices (49 devices over 3 experiments) with different numbers of perfusable interpost regions. Perfusable interpost regions are counted for each device via bright field microscopy during tumor cell perfusion. Out of the 49 devices, 43 showed more than 10 (50%) perfusable interpost regions. (B) 40 out of 43 of these devices showed a distribution of tumor cells across the vascular network of more than 25% past the centerline of the gel. In these set of experiments, the perfusability is thus ~82% of total devices. (C) Phase contrast images (20X) of typical perfusable openings. (D) 10X phase contrast images of a good device with many openings (device 1) and a poor device with few openings (device 2).

Supplementary information

Supplementary Figures and Table

Supplementary Figures 1–3, Supplementary Methods and Supplementary Table 1. (PDF 727 kb)

Supplementary Data

Photo-mask with microdevice design. (ZIP 106 kb)

Time-lapse video of extravasating tumor cell.

Time-lapse video of a transmigrating MDA-MB-231 cell (LifeAct GFP, green) from a microvessel (HUVEC, red). Frames are 40 min apart. (MOV 237 kb)

Flow of tumor cells within microvascular networks.

Real-time video via phase-contrast at 10× magnification, depicting the flow of MDA-MB-231 cells and human platelets within microvascular networks upon introduction of a 4-mm H2O hydrostatic pressure drop. Tumor cells are seen to decelerate, arrest and at times dislodge from the capillaries under flow conditions. (MOV 3909 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, M., Whisler, J., Fröse, J. et al. On-chip human microvasculature assay for visualization and quantification of tumor cell extravasation dynamics. Nat Protoc 12, 865–880 (2017). https://doi.org/10.1038/nprot.2017.018

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.018

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