4.6 Article

Nanoroughened adhesion-based capture of circulating tumor cells with heterogeneous expression and metastatic characteristics

Journal

BMC CANCER
Volume 16, Issue -, Pages -

Publisher

BMC
DOI: 10.1186/s12885-016-2638-x

Keywords

Circulating tumor cells; Adhesion; Microfluidics; Metastasis; Breast cancer; Lung cancer

Categories

Funding

  1. National Science Foundation [CMMI 1536087]
  2. UM Comprehensive Cancer Center Prostate SPORE Pilot Project [NIH/NCI P50 CA069568]
  3. National Cancer Institute F30 fellowship program [NIH/NCI F30 CA173910]
  4. Breast Cancer Research Foundation
  5. Avon Foundation
  6. American Heart Association Predoctoral Fellowship
  7. New York University Research Challenge Fund
  8. NIH/NCI [CA132571-01]
  9. Elizabeth A. Crary Fund
  10. NSF
  11. Directorate For Engineering
  12. Div Of Civil, Mechanical, & Manufact Inn [1536087] Funding Source: National Science Foundation

Ask authors/readers for more resources

Background: Circulating 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. Methods: We 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. Results: The 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-beta-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. Conclusions: The 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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available