Abstract

BackgroundCirculating tumor cells (CTCs) have shown prognostic relevance in many cancer types. However, the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population.MethodsWe developed a microfluidic CTC capture chip that incorporated a nanoroughened glass substrate for capturing CTCs from blood samples. Our CTC capture chip utilized the differential adhesion preference of cancer cells to nanoroughened etched glass surfaces as compared to normal blood cells and thus did not depend on the physical size or surface protein expression of CTCs.ResultsThe microfluidic CTC capture chip was able to achieve a superior capture yield for both epithelial cell adhesion molecule positive (EpCAM+) and EpCAM- cancer cells in blood samples. Additionally, the microfluidic CTC chip captured CTCs undergoing transforming growth factor beta-induced epithelial-to-mesenchymal transition (TGF-β-induced EMT) with dynamically down-regulated EpCAM expression. In a mouse model of human breast cancer using EpCAM positive and negative cell lines, the number of CTCs captured correlated positively with the size of the primary tumor and was independent of their EpCAM expression. Furthermore, in a syngeneic mouse model of lung cancer using cell lines with differential metastasis capability, CTCs were captured from all mice with detectable primary tumors independent of the cell lines’ metastatic ability.ConclusionsThe microfluidic CTC capture chip using a novel nanoroughened glass substrate is broadly applicable to capturing heterogeneous CTC populations of clinical interest independent of their surface marker expression and metastatic propensity. We were able to capture CTCs from a non-metastatic lung cancer model, demonstrating the potential of the chip to collect the entirety of CTC populations including subgroups of distinct biological and phenotypical properties. Further exploration of the biological potential of metastatic and presumably non-metastatic CTCs captured using the microfluidic chip will yield insights into their relevant differences and their effects on tumor progression and cancer outcomes.Electronic supplementary materialThe online version of this article (doi:10.1186/s12885-016-2638-x) contains supplementary material, which is available to authorized users.

Highlights

  • Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types

  • In our prior work [31], we described that a nanorough glass substrate generated by reactiveion etching (RIE) without any positive-selection antibodies exhibits significantly improved cancer cell capture efficiency owing to enhanced adherent interactions between the nanoscale topological features on the glass substrate and the nanoscale cellular adhesion apparatus [21]

  • In this work we showed that the microfluidic CTC capture chip could capture > 80 % of breast and lung cancer cells spiked in whole blood samples independent of the cell lines’ epithelial cell adhesion molecule (EpCAM) expression

Read more

Summary

Introduction

Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types. the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population. The spread of cancer systemically relies upon the critical step of the hematogenous spread of cancer cells [2] These circulating tumor cells (CTCs) in the bloodstream are shed from primary and metastatic lesions and are believed to be key agents in the metastatic process [2,3,4]. The challenge being as a tumor progresses down the metastatic cascade, cancer cells are known to express diverse molecular phenotypes in a dynamic fashion, which complicates the isolation of CTCs for further study [6, 8,9,10,11,12,13] Other cells such as fibroblasts and non-cancerous epithelial cells are shed into the circulation further complicating the identification of the true potentially metastatic cells

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call