Elsevier

Biomaterials

Volume 35, Issue 8, March 2014, Pages 2454-2461
Biomaterials

A microfluidic 3D in vitro model for specificity of breast cancer metastasis to bone

https://doi.org/10.1016/j.biomaterials.2013.11.050Get rights and content

Abstract

Cancer metastases arise following extravasation of circulating tumor cells with certain tumors exhibiting high organ specificity. Here, we developed a 3D microfluidic model to analyze the specificity of human breast cancer metastases to bone, recreating a vascularized osteo-cell conditioned microenvironment with human osteo-differentiated bone marrow-derived mesenchymal stem cells and endothelial cells. The tri-culture system allowed us to study the transendothelial migration of highly metastatic breast cancer cells and to monitor their behavior within the bone-like matrix. Extravasation, quantified 24 h after cancer cell injection, was significantly higher in the osteo-cell conditioned microenvironment compared to collagen gel-only matrices (77.5 ± 3.7% vs. 37.6 ± 7.3%), and the migration distance was also significantly greater (50.8 ± 6.2 μm vs. 31.8 ± 5.0 μm). Extravasated cells proliferated to form micrometastases of various sizes containing 4 to more than 60 cells by day 5. We demonstrated that the breast cancer cell receptor CXCR2 and the bone-secreted chemokine CXCL5 play a major role in the extravasation process, influencing extravasation rate and traveled distance. Our study provides novel 3D in vitro quantitative data on extravasation and micrometastasis generation of breast cancer cells within a bone-like microenvironment and demonstrates the potential value of microfluidic systems to better understand cancer biology and screen for new therapeutics.

Introduction

The systemic nature of cancer metastases coupled with the resistance to most current therapeutic agents explains why metastases are responsible for as much as 90% of cancer-related mortality [1], [2]. The dissemination of circulating tumor cells (CTCs) represents a “hidden” process leading to micrometastases where quiescent cells can survive for prolonged periods before their activation [3], [4].

In order to generate secondary tumors, CTCs must survive in the circulation and undergo a process known as extravasation [5], [6], [7]. Extravasation into the parenchyma of distant tissues represents a multistep sequence within the metastatic cascade, in which cancer cells establish transient, metastable contacts with the endothelium [8], [9], [10], firmly adhere to the vascular walls [11] and finally transmigrate across the endothelial and pericyte layers [12] as microcolonies or isolated cells [13], [14].

Although it is well known that circulatory patterns play a pivotal role in the spread of metastatic cells to secondary sites, the cross-talk between specific cancer cell types and receptive environments also preferentially guides the dissemination process [15]. In this context, it has been shown that breast cancer metastasizes to bone, liver, lung and brain while prostate cancer frequently disseminates to bone [3]. Particularly, autoptic studies have demonstrated that 70% of advanced breast cancer patients have skeletal metastases, leading to pain, due to spinal cord compression and fractures, and often mortality [16], [17].

Despite the clinical importance of metastases, research has largely focused on the oncogenic transformations leading to the development of primary tumors and much remains to be learned about the metastatic process [5]. Moreover, a deeper understanding of the metastatic cascade and particularly of extravasation to a specific organ could promote the development of new therapeutic strategies, thus improving cancer survival rates [12].

In vivo and ex vivo models have been developed to study the extravasation process in mice and zebrafish embryos through intravital microscopy [13], [18], [19] and advanced models of bone metastasis employ intravenous, intracardiac or direct skeletal injection of breast cancer cells [20], [21]. Although these experiments replicate physiological conditions, they cannot model all aspects of the interaction and cross-talk between human cancer cells, human endothelial cells and human tissue parenchyma. Moreover, strictly regulated, reproducible parametric studies are difficult to perform.

In vitro models, although unable to fully replicate the in vivo situation, can overcome some of these limitations by using human cells throughout and providing highly controllable environments where single culture parameters can be modified [22], [23]. Traditional assays (e.g. Boyden chamber, wound assay, and others) have been widely used to study cell migration in response to chemotactic gradients, particularly cancer cell invasion and migration. However, they do not provide tight control over the local environment, complex interactions cannot be accurately analyzed, and imaging is limited [24], [25], [26].

Microfluidics can provide useful model systems to investigate complex phenomena under combination of multiple controllable biochemical and biophysical microenvironments, coupled with high resolution real time imaging [27], [28], [29], [30]. The synthesis of these features is technically impossible with traditional assays as the Boyden chamber [31], [32]. Toward this goal, several microfluidic devices have been developed to investigate cancer cell transition to invasion and migration from a primary site [33], [34], [35], cell transition effects across mechanical barriers [36], intravasation [37], adhesion [38] and extravasation [39], [40], [41], [42], [43], [44] processes.

However, despite supporting experimental evidence, none of the previously reported in vitro systems has reproduced the specific cross-talk among several cell types in a complex cancer microenvironment during extravasation and none has gone beyond the study of transendothelial migration towards a non-organ-specific extracellular matrix (ECM). Indeed, the importance of organ-specific cancer models lies in the possibility to better clarify the mutual interactions between different cell populations in a well-defined microenvironment, in order to develop highly focused and more effective therapies.

We develop here a new tri-culture microfluidic 3D in vitro model demonstrating the key role played by an osteo-cell conditioned microenvironment, a collagen gel with embedded osteo-differentiated human bone marrow-derived mesenchymal stem cells (hBM-MSCs) [45] and lined with endothelium, in the extravasation process of highly-metastatic MDA-MB-231 human breast cancer cells [16], [46].

Section snippets

Microfluidic system

A previously developed microfluidic device consisting of 3 media channels and 4 independent gel channels was adopted in the present study. Specifications and microfabrication details of the system were previously described [47], [48]. Inlet and outlet ports of the PDMS (poly-dimethyl-siloxane; Silgard 184, Dow Chemical) devices were bored using disposable biopsy punches and the PDMS layer was bonded to a cover glass to create microfluidic channels 150 μm deep with oxygen plasma treatment. Eight

Generation of the osteo-cell conditioned microenvironment

The originality and relevance of the present work lie in the ability to specifically recreate an organ-specific tumor microenvironment by combining a well known biocompatible material, collagen gel type I, conditioned by bone-specific proteins secreted directly from primary human cells, thus modeling the extravasation process of breast cancer cells into an osteo-cell conditioned microenvironment. A key point of our system is that the extracellular matrix with secreted proteins from

Conclusions

We have presented an advanced human organ-specific microfluidic 3D in vitro model to analyze the extravasation of highly metastatic breast cancer cells into an osteo-cell conditioned microenvironment. We have provided unique quantitative results concerning the interplay between a specific “seed and soil couple” by computing extravasation rate and extravasated distance of breast cancer cells in the presence of an attractive matrix. Moreover, we have shown how the molecular pathway involving

Acknowledgments

Support from the National Cancer Institute (R33 CA174550-01 and R21 CA140096) and the Italian Ministry of Health, fellowship support to S. Bersini provided by the Fondazione Fratelli Agostino and Enrico Rocca through the Progetto Rocca Doctoral Fellowship and support to J.S. Jeon provided by Repligen Fellowship in Cancer Research and Draper Fellowship are gratefully acknowledged. S. Chung was supported by NRF (2012-022481) and KETEP (20124010203250) of Korea.

References (63)

  • F.Q. Nie et al.

    On-chip cell migration assay using microfluidic channels

    Biomaterials

    (2007)
  • K. Yang et al.

    A microfluidic array for quantitative analysis of human neural stem cell self-renewal and differentiation in three-dimensional hypoxic microenvironment

    Biomaterials

    (2013)
  • E. Leclerc et al.

    Study of osteoblastic cells in a microfluidic environment

    Biomaterials

    (2006)
  • J.L. Halpern et al.

    Mesenchymal stem cells promote mammary cancer cell migration in vitro via the CXCR2 receptor

    Cancer Lett

    (2011)
  • G. Cserni et al.

    Variations in sentinel node isolated tumour cells/micrometastasis and non-sentinel node involvement rates according to different interpretations of the TNM definitions

    Eur J Cancer

    (2008)
  • C.L. Chaffer et al.

    A perspective on cancer cell metastasis

    Science

    (2011)
  • A.F. Chambers et al.

    Dissemination and growth of cancer cells in metastatic sites

    Nat Rev Cancer

    (2002)
  • I.J. Fidler

    The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited

    Nat Rev Cancer

    (2003)
  • J.A. Joyce et al.

    Microenvironmental regulation of metastasis

    Nat Rev Cancer

    (2009)
  • M. Fukuda et al.

    C-type lectins and sialyl Lewis X oligosaccharides. Versatile roles in cell-cell interaction

    J Cell Biol

    (1999)
  • F.L. Miles et al.

    Stepping out of the flow: capillary extravasation in cancer metastasis

    Clin Exp Metastasis

    (2008)
  • A. Zipin et al.

    Tumor-microenvironment interactions: the fucose-generating FX enzyme controls adhesive properties of colorectal cancer cells

    Cancer Res

    (2004)
  • E.T. Roussos et al.

    Chemotaxis in cancer

    Nat Rev Cancer

    (2011)
  • A.B. Al-Mehdi et al.

    Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis

    Nat Med

    (2000)
  • S. Paget

    The distribution of secondary growths in cancer of the breast. 1889

    Cancer Metastasis Rev

    (1989)
  • K.M. Bussard et al.

    The bone microenvironment in metastasis; what is special about bone?

    Cancer Metastasis Rev

    (2008)
  • R.E. Coleman

    Skeletal complications of malignancy

    Cancer

    (1997)
  • G.N. Naumov et al.

    Cellular expression of green fluorescent protein, coupled with high-resolution in vivo videomicroscopy, to monitor steps in tumor metastasis

    J Cell Sci.

    (1999)
  • K. Stoletov et al.

    Visualizing extravasation dynamics of metastatic tumor cells

    J Cell Sci

    (2010)
  • R.H. Goldstein et al.

    Of mice and (wo)men: mouse models of breast cancer metastasis to bone

    J Bone Miner Res

    (2010)
  • C. Kuperwasser et al.

    A mouse model of human breast cancer metastasis to human bone

    Cancer Res

    (2005)
  • Cited by (0)

    1

    S.B. and J.S.J. contributed equally to the present work.

    2

    M. Moretti and R.D. Kamm equally contributed.

    View full text