skip to main content


Title: Microfluidic techniques for isolation, formation, and characterization of circulating tumor cells and clusters
Circulating tumor cell (CTC) clusters that are shed from the primary tumor into the bloodstream are associated with a poor prognosis, elevated metastatic potential, higher proliferation rate, and distinct molecular features compared to single CTCs. Studying CTC clusters may give us information on the differences in the genetic profiles, somatic mutations, and epigenetic changes in circulating cells compared to the primary tumor and metastatic sites. Microfluidic systems offer the means of studying CTC clusters through the ability to efficiently isolate these rare cells from the whole blood of patients in a liquid biopsy. Microfluidics can also be used to develop in vitro models of CTC clusters and make possible their characterization and analysis. Ultimately, microfluidic systems can offer the means to gather insight on the complexities of the metastatic process, the biology of cancer, and the potential for developing novel or personalized therapies. In this review, we aim to discuss the advantages and challenges of the existing microfluidic systems for working with CTC clusters. We hope that an improved understanding of the role microfluidics can play in isolation, formation, and characterization of CTC clusters, which can lead to increased sophistication of microfluidic platforms in cancer research.  more » « less
Award ID(s):
1841509 1841473
NSF-PAR ID:
10390289
Author(s) / Creator(s):
; ; ;
Date Published:
Journal Name:
APL Bioengineering
Volume:
6
Issue:
3
ISSN:
2473-2877
Page Range / eLocation ID:
031501
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Abstract

    Most cancer patients die from metastatic disease as a result of a circulating tumor cell (CTC) spreading from a primary tumor through the blood circulation to distant organs. Many studies have demonstrated the tremendous potential of using CTC counts as prognostic markers of metastatic development and therapeutic efficacy. However, it is only the viable CTCs capable of surviving in the blood circulation that can create distant metastasis. To date, little progress has been made in understanding what proportion of CTCs is viable and what proportion is in an apoptotic state. Here, we introduce a novel approach toward in situ characterization of CTC apoptosis status using a multicolor in vivo flow cytometry platform with fluorescent detection for the real‐time identification and enumeration of such cells directly in blood flow. The proof of concept was demonstrated with two‐color fluorescence flow cytometry (FFC) using breast cancer cells MDA‐MB‐231 expressing green fluorescein protein (GFP), staurosporine as an activator of apoptosis, Annexin‐V apoptotic kit with orange dye color, and a mouse model. The future application of this new platform for real‐time monitoring of antitumor drug efficiency is discussed. © 2019 International Society for Advancement of Cytometry

     
    more » « less
  2. Nguyen, N. T. (Ed.)

    Single-cell analysis provides a wealth of information regarding the molecular landscape of the tumor cells responding to extracellular stimulations, which has greatly advanced the research in cancer biology. In this work, we adapt such a concept for the analysis of inertial migration of cells and clusters, which is promising for cancer liquid biopsy, by isolation and detection of circulating tumor cells (CTCs) and CTC clusters. Using high-speed camera tracking live individual tumor cells and cell clusters, the behavior of inertial migration was profiled in unprecedented detail. We found that inertial migration is heterogeneous spatially, depending on the initial cross-sectional location. The lateral migration velocity peaks at about 25% of the channel width away from the sidewalls for both single cells and clusters. More importantly, while the doublets of the cell clusters migrate significantly faster than single cells (~two times faster), cell triplets unexpectedly have similar migration velocities to doublets, which seemingly disagrees with the size-dependent nature of inertial migration. Further analysis indicates that the cluster shape or format (for example, triplets can be in string format or triangle format) plays a significant role in the migration of more complex cell clusters. We found that the migration velocity of a string triplet is statistically comparable to that of a single cell while the triangle triplets can migrate slightly faster than doublets, suggesting that size-based sorting of cells and clusters can be challenging depending on the cluster format. Undoubtedly, these new findings need to be considered in the translation of inertial microfluidic technology for CTC cluster detection.

     
    more » « less
  3. null (Ed.)
    We demonstrate a label free and high-throughput microbubble-based acoustic microstreaming technique to isolate rare circulating cells such as circulating cancer associated fibroblasts (cCAFs) in addition to circulating tumor cells (CTCs) and immune cells ( i.e. leukocytes) from clinically diagnosed patients with a capture efficiency of 94% while preserving cell functional integrity within 8 minutes. The microfluidic device is self-pumping and was optimized to increase flow rate and achieve near perfect capturing of rare cells enabled by having a trapping capacity above the acoustic vortex saturation concentration threshold. Our approach enables rapid isolation of CTCs, cCAFs and their associated clusters from blood samples of cancer patients at different stages. By examining the combined role of cCAFs and CTCs in early cancer onset and metastasis progression, the device accurately diagnoses both cancer and the metastatic propensity of breast cancer patients. This was confirmed by flow cytometry where we observed that metastatic breast cancer blood samples had significantly higher percentage of exhausted CD8 + T cells expressing programmed cell death protein 1 (PD1), higher number of CD4 + T regulatory cells and T helper cells. We show for the first time that our lateral cavity acoustic transducers (LCATs)-based approach can thus be developed into a metastatic propensity assay for clinical usage by elucidating cancer immunological responses and the complex relationships between CTCs and its companion tumor microenvironment. 
    more » « less
  4. Abstract

    Circulating tumor cells (CTCs) are shed from primary tumors, circulate in the bloodstream and are capable of initiating metastasis at distant anatomical sites. The detection and molecular characterization of CTCs are pivotal for early-stage cancer diagnosis and prognosis. Recently, microfluidic technology has achieved significant progress in the separation of cells from complex and heterogeneous mixtures for many biomedical applications. Conventional microfluidic platforms exploit the difference in size between the particles to achieve separation, which makes them ineffective for sorting overlapping-sized CTCs. To address this issue, we propose a method using a spiral channel for label-free, and high throughput separation of CTCs coupling Dielectrophoresis (DEP) with inertial microfluidics. A numerical model has been developed to investigate the separation effectiveness of the device over a range of electrical voltage and flow rates. The presented channel is shown to effectively isolate similar-sized CTCs from the white blood cells (WBCs) in a single-stage separation process. Subsequently, optimum working parameters to enhance separation efficiency have been proposed. The hybrid microfluidic device can provide valuable insight into the development of a robust, inexpensive, and efficient platform for cell separation with reduced analysis time for future cancer research and treatment.

     
    more » « less
  5. Profiling circulating tumour cells (CTCs) in cancer patients' blood samples is critical to understand the complex and dynamic nature of metastasis. This task is challenged by the fact that CTCs are not only extremely rare in circulation but also highly heterogeneous in their molecular programs and cellular functions. Here we report a combinational approach for the simultaneous biochemical and functional phenotyping of patient-derived CTCs, using an integrated inertial ferrohydrodynamic cell separation (i 2 FCS) method and a single-cell microfluidic migration assay. This combinatorial approach offers unique capability to profile CTCs on the basis of their surface expression and migratory characteristics. We achieve this using the i 2 FCS method that successfully processes whole blood samples in a tumor cell marker and size agnostic manner. The i 2 FCS method enables an ultrahigh blood sample processing throughput of up to 2 × 10 5 cells s −1 with a blood sample flow rate of 60 mL h −1 . Its short processing time (10 minutes for a 10 mL sample), together with a close-to-complete CTC recovery (99.70% recovery rate) and a low WBC contamination (4.07-log depletion rate by removing 99.992% of leukocytes), results in adequate and functional CTCs for subsequent studies in the single-cell migration device. For the first time, we employ this new approach to query CTCs with single-cell resolution in accordance with their expression of phenotypic surface markers and migration properties, revealing the dynamic phenotypes and the existence of a high-motility subpopulation of CTCs in blood samples from metastatic lung cancer patients. This method could be adopted to study the biological and clinical value of invasive CTC phenotypes. 
    more » « less