DNA Nanospheres with Microfluidics: a Promising Platform for Cancer Diagnosis?
Z Hugh Fan,Weihong Tan
DOI: https://doi.org/10.2217/nnm.13.163
2013-01-01
Abstract:NanomedicineVol. 8, No. 11 EditorialFree AccessDNA nanospheres with microfluidics: a promising platform for cancer diagnosis?Z Hugh Fan & Weihong TanZ Hugh Fan* Author for correspondenceInterdisciplinary Microsystems Group, Department of Mechanical & Aerospace Engineering, J Crayton Pruitt Family Department of Biomedical Engineering, & Department of Chemistry, University of Florida, PO Box 116250, Gainesville, FL 32611, USA. Search for more papers by this authorEmail the corresponding author at hfan@ufl.edu & Weihong TanCenter for Research at the Bio/Nano Interface, Department of Chemistry, Department of Physiology & Functional Genomics & Shands Cancer Center, University of Florida, Gainesville, FL 32611-7200, USA.Search for more papers by this authorPublished Online:24 Oct 2013https://doi.org/10.2217/nnm.13.163AboutSectionsPDF/EPUB ToolsAdd to favoritesDownload CitationsTrack CitationsPermissionsReprints ShareShare onFacebookTwitterLinkedInRedditEmail Keywords: aptamercirculating tumor cellmicrofluidicsmultivalencynanoparticleThe mortality rate from cancer in the USA reached more than 570,000 in 2012, or equal to more than 1500 Americans each day [1]. More than 90% of cancer deaths result from metastasis [2], hence methods for early-stage cancer detection are vital for the reduction of mortality and they will have a significant medical and social impact. However, according to the Early Detection Research Network of the National Cancer Institute, ‘there are no validated molecular biomarker tests for the early detection of any cancer’ [3], even though a handful of biomarkers have been approved by the US FDA.It is known that, during metastasis, some cancer cells escape from the primary tumor through exfoliation, entering the bloodstream and becoming circulating tumor cells (CTCs) in the peripheral blood stream [2,4]. Some of these CTCs acquire the capability to colonize secondary sites, spreading tumors to distant organs [2,4]. Therefore, CTCs have the potential to serve as important biomarkers for early diagnosis of cancer metastasis [2,4]. An additional benefit is that CTC enumeration is less invasive than biopsy, while providing a quantifiable method for cancer diagnosis and prognosis.However, CTCs are extremely rare; the number of CTCs is in the order of one to 100 cells in 1 ml of blood [4]. For comparison, other cells in 1 ml of blood include approximately 5 × 109 erythrocytes, 7.8 × 106 leukocytes and approximately 3.5 × 108 platelets [5]. Therefore, detection of CTCs in peripheral blood is a needle-in-a-haystack challenge. Currently, CellSearch® (Veridex LLC, NJ, USA) is the only FDA-approved method, and it relies on antibody-coated magnetic particles for isolation of CTCs from blood [4]. One of its key drawbacks is low capture efficiency.Microfluidics could play an important role in enhancing the capture efficiency of CTCs. Microfluidics is a multidisciplinary field that involves microfabrication to make devices, microflows to study fluids in the micron scale and a variety of other applications. One aim of microfluidics is to shrink an assembly of laboratory instruments onto a device the size of a computer chip, known as a ‘lab-on-a-chip’. This is analogous to reducing a room-sized computer of the 1950s to a laptop today. Examples of commercially available microfluidics-enabled apparatuses include the Bioanalyzer 2100 from Agilent Technologies (CA, USA) and the BioMark™ HD System from Fluidigm Corp. (CA, USA).The application of microfluidics to cell capture affords a number of advantages. First, cells in a sample must diffuse from the bulk of the solution to the solid surface before interacting with the immobilized antibodies, as in CellSearch. Diffusion distance is in the millimeter scale; however, the diffusion distance in microchannels is in the micrometer scale, significantly increasing the interaction opportunities between CTCs and capture agents. As a result, the capture efficiency should increase, along with decreased incubation time. Second, microchannels offer a significantly higher surface-to-volume ratio than macroscale reaction vessels. Consequently, the amount of capture agents on the surfaces could be in excess, driving their reactions with CTCs to completion. In addition, micropillars and other geometries may be created to enhance the surface areas. Third, microfluidics enables integration with other components, such as mixers and detectors. Mixing can propel CTCs to the proximity of channel surfaces, thus, enhancing capture efficiency. Detection components may allow the sorted cells to be further analyzed using on-chip biological assays, speeding up the process compared with conventional methods in which sorting is performed first, followed by analysis in another apparatus.Applying microfluidics to CTC isolation has been recently demonstrated by a few research groups [6–12]. Using the same CTC definition as in CellSearch, Nagrath and coworkers isolated CTCs in blood samples from breast cancer patients; they showed that >50 CTCs/ml were enumerated in 80% of the cohort [6]. However, <7 CTCs/ml (or 50 CTCs/7.5 ml) were obtained in 88% of breast cancer patients when CellSearch was used [4]. These results suggest that the use of microfluidics is advantageous for CTC detection, with higher capture efficiency, as discussed above.The capture agents most commonly used in CTC research are antibodies. An alternative to antibodies is aptamers. Aptamers are oligonucleic acid sequences, such as short DNA or RNA, with a high binding affinity to a target of interest. Aptamers possess binding capability comparable with that of monoclonal antibodies, and they have been shown to bind to a variety of targets, including small molecules, proteins, nucleic acids and cells [13,14]. In contrast to antibodies, aptamers have the following advantages: complete engineering in test tubes rather than in animals; straightforward production by chemical synthesis through DNA synthesizers; and long-term stability during storage [13,14].Aptamers are selected through an approach called systematic evolution of ligands by exponential enrichment [15]. In their pioneering work, Tuerk and Gold found two RNA sequences with a dissociation constant of 5 × 10-9 M with T4 DNA polymerase [15]. Cell-based systematic evolution of ligands by exponential enrichment is a newer hybrid process developed to select aptamers for target cells [16]. A wide range of DNA aptamers have been selected and shown to have strong binding affinity with a variety of cells, including acute lymphoblastic leukemia, lymphoma and hepatocellular carcinoma cells, as well as bacterial pathogens [14,16].Aptamers have now been incorporated with microfluidic devices for tumor cell isolation from unprocessed whole blood [12]. It was found that a compromise must be made between capture efficiency and cell purity by optimizing key parameters such as device geometry and flow rate. Capture efficiency is defined as the ratio of the number of target cells captured to the number of target cells introduced. Cell purity is defined as the ratio of the number of target cells captured to the number of total cells captured. For instance, capture efficiency decreases with increasing flow rate as a result of larger shear force at a higher flow rate, and reduced interaction time between cells and binding agents on channel surfaces. However, cell purity increases with increasing flow rate owing to the fact that cells bound nonspecifically are washed away with a stronger shear force at a higher flow rate. Therefore, an optimal flow rate exists that results in satisfactory capture efficiency with decent cell purity.To further enhance capture efficiency, we recently reported the development of a platform combining DNA nanospheres with microfluidics for CTC isolation [17]. Each nanosphere consisted of a gold nanoparticle (AuNP) that was conjugated with a number of DNA aptamers. Conjugation between gold and thio-ended DNA has been well established, and attachment of multiple DNA oligonucleotides to a AuNP has been demonstrated for various applications [18,19]. Up to 95 aptamers were attached to each AuNP, forming a DNA nanosphere (AuNP-aptamers). Multiple aptamers on each AuNP were able to interact simultaneously with several receptors on a tumor cell, resulting in multivalent binding between AuNP-aptamers and the tumor cell. It was found that the binding affinity of AuNP-aptamers was 39-times higher than aptamers themselves alone. The capture efficiency of human acute leukemia cells increased from 49% using aptamers alone to 92% using AuNP-aptamers. Results show that combining DNA nanospheres with microfluidics are a promising platform for CTC isolation and cancer diagnosis and prognosis.Future studies will include applying this platform to the isolation of CTCs from peripheral blood samples of patients with metastatic cancer. For example, CTCs have been enumerated for a study investigating the efficacy of a chemotherapy drug combination for pancreatic cancer. Preliminary results of this double-blind study indicate that our CTC counts correlate very well with the clinical data. We currently use the same CTC definition as in the FDA-approved CellSearch: nucleated cells lacking CD45 and expressing EpCAM and cytokeratin (CK) [4]. However, it is very likely that this definition of CTCs, that is EpCAM-positive/CK-positive/DAPI-positive/CD45-negative cells, is too narrow because those tumor cells with little or no EpCAM expression are overlooked. During metastasis, tumor cells undergo epithelial-to-mesenchymal transition, in which tumor cells change from epithelial to mesenchymal morphology to acquire increased migratory and invasive capabilities [2,20]. Importantly, during epithelial-to-mesenchymal transition, some cells lose biomarkers associated with epithelial origin, such as EpCAM [2,20]. As a result, affinity ligands are needed to isolate both EpCAM-positive and EpCAM-negative cells. From this perspective, aptamers are advantageous since a panel of aptamers can be selected for both types of CTCs.AcknowledgementsThe authors wish to thank W Sheng and T Chen for their work related to this editorial.Financial & competing interests disclosureThe authors are grateful for the financial support from the NIH (K25CA149080, R21GM103535, GM079359 and CA133086). A provisional patent application has been filed by the authors’ employer, the University of Florida. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.No writing assistance was utilized in the production of this manuscript.References1 American Cancer Society. Cancer Facts and Figures 2012. American Cancer Society, GA, USA (2012).Google Scholar2 Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science331(6024),1559–1564 (2011).Crossref, Medline, CAS, Google Scholar3 National Cancer Institute. The Early Detection Research Network Fourth Report. NIH, MD, USA (2008).Google Scholar4 Cristofanilli M, Budd GT, Ellis MJ et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med.351(8),781–791 (2004).Crossref, Medline, CAS, Google Scholar5 Henry JB. Clinical Diagnosis and Management by Laboratory Methods (19th Edition). Saunders, PA, USA (1996).Google Scholar6 Nagrath S, Sequist LV, Maheswaran S et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature450(7173),1235–1239 (2007).Crossref, Medline, CAS, Google Scholar7 Adams AA, Okagbare PI, Feng J et al. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. J. Am. Chem. Soc.130(27),8633–8641 (2008).Crossref, Medline, CAS, Google Scholar8 Stott SL, Hsu CH, Tsukrov DI et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl Acad. Sci. USA107(43),18392–18397 (2010).Crossref, Medline, CAS, Google Scholar9 Gleghorn JP, Pratt ED, Denning D et al. Capture of circulating tumor cells from whole blood of prostate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip10(1),27–29 (2010).Crossref, Medline, CAS, Google Scholar10 Wang S, Liu K, Liu J et al. Highly efficient capture of circulating tumor cells by using nanostructured silicon substrates with integrated chaotic micromixers. Angew. Chem. Int. Ed. Engl.50(13),3084–3088 (2011).Crossref, Medline, CAS, Google Scholar11 Schiro PG, Zhao M, Kuo JS, Koehler KM, Sabath DE, Chiu DT. Sensitive and high-throughput isolation of rare cells from peripheral blood with ensemble-decision aliquot ranking. Angew. Chem. Int. Ed. Engl.51(19),4618–4622 (2012).Crossref, Medline, CAS, Google Scholar12 Sheng W, Chen T, Kamath R, Xiong X, Tan W, Fan ZH. Aptamer-enabled efficient isolation of cancer cells from whole blood using a microfluidic device. . Anal. Chem. 84(9),4199–4206 (2012).Crossref, Medline, CAS, Google Scholar13 Famulok M, Hartig JS, Mayer G. Functional aptamers and aptazymes in biotechnology, diagnostics, and therapy. Chem. Rev.107(9),3715–3743 (2007).Crossref, Medline, CAS, Google Scholar14 Fang X, Tan W. Aptamers generated from cell-SELEX for molecular medicine: a chemical biology approach. Acc. Chem. Res.43(1),48–57 (2010).Crossref, Medline, CAS, Google Scholar15 Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science249(4968),505–510 (1990).Crossref, Medline, Google Scholar16 Shangguan D, Li Y, Tang Z et al. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc. Natl Acad. Sci. USA103(32),11838–11843 (2006).Crossref, Medline, CAS, Google Scholar17 Sheng W, Chen T, Tan W, Fan ZH. Multivalent DNA nanospheres for enhanced capture of cancer cells in microfluidic devices. ACS Nano7(8),7067–7076 (2013).Crossref, Medline, CAS, Google Scholar18 Mirkin CA, Letsinger RL, Mucic RC, Storhoff JJ. A DNA-based method for rationally assembling nanoparticles into macroscopic materials. Nature382(6592),607–609 (1996).Crossref, Medline, CAS, Google Scholar19 Cutler JI, Auyeung E, Mirkin CA. Spherical nucleic acids. J. Am. Chem. Soc.134(3),1376–1391 (2012).Crossref, Medline, CAS, Google Scholar20 Lianidou ES, Markou A. Circulating tumor cells in breast cancer: detection systems, molecular characterization, and future challenges. Clin. Chem.57(9),1242–1255 (2011).Crossref, Medline, CAS, Google ScholarFiguresReferencesRelatedDetailsCited ByMagnetic nanoparticles in microfluidics-based diagnostics: an appraisalSmriti Sharma & Vinayak Bhatia24 May 2021 | Nanomedicine, Vol. 16, No. 15Microfluidic devices and drug delivery systems3D models in the new era of immune oncology: focus on T cells, CAF and ECM22 March 2019 | Journal of Experimental & Clinical Cancer Research, Vol. 38, No. 1Programmable and Multifunctional DNA-Based Materials for Biomedical Applications1 February 2018 | Advanced Materials, Vol. 30, No. 24Reduction in Migratory Phenotype in a Metastasized Breast Cancer Cell Line via Downregulation of S100A4 and GRM314 June 2017 | Scientific Reports, Vol. 7, No. 1Competitive Volumetric Bar-Chart Chip with Real-Time Internal Control for Point-of-Care Diagnostics18 March 2015 | Analytical Chemistry, Vol. 87, No. 7Lab on a chip and circulating tumor cells1 January 2014 | Lab Chip, Vol. 14, No. 1 Vol. 8, No. 11 Follow us on social media for the latest updates Metrics History Published online 24 October 2013 Published in print November 2013 Information© Future Medicine LtdKeywordsaptamercirculating tumor cellmicrofluidicsmultivalencynanoparticleAcknowledgementsThe authors wish to thank W Sheng and T Chen for their work related to this editorial.Financial & competing interests disclosureThe authors are grateful for the financial support from the NIH (K25CA149080, R21GM103535, GM079359 and CA133086). A provisional patent application has been filed by the authors’ employer, the University of Florida. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.No writing assistance was utilized in the production of this manuscript.PDF download
What problem does this paper attempt to address?
-
Multivalent Dna Nanospheres For Enhanced Capture Of Cancer Cells In Microfluidic Devices
Weian Sheng,Tao Chen,Weihong Tan,Z. Hugh Fan
DOI: https://doi.org/10.1021/nn4023747
IF: 17.1
2013-01-01
ACS Nano
Abstract:Isolation of circulating tumor cells (CTCs), from from peripheral blood or cancer cells from bone marrow has significant applications in cancer diagnosis, therapy monitoring, and drug development. CTCs are cancer cells shed from primary tumors; they circulate in the bloodstream, leading to metastasis. The extraordinary rarity of CTCs in the bloodstream makes their isolation a significant technological challenge. Herein, we report the development of a platform combining multivalent DNA aptamer nanospheres with microfluidic devices for efficient isolation of cancer cells from blood. Gold nanoparticles (AuNPs) were used as an efficient platform for assembling a number of aptamers for high-efficiency cell capture. Up to 95 aptamers were attached onto each AuNP, resulting in enhanced molecular recognition capability. An increase of 39-fold in binding affinity was confirmed by flow cytometry for AuNP-aptamer conjugates (AuNP-aptamer) when compared with aptamer alone. With a laminar flow flat channel microfluidic device, the capture efficiency of human acute leukemia cells from a cell mixture in buffer increased from 49% using aptamer alone to 92% using AuNP-aptamer. We also employed AuNP-aptamer in a microfluidic device with herringbone mixing microstructures for isolation of leukemia cells in whole blood. The cell capture efficiency was also significantly increased with the AuNP-aptamer over aptamer alone, especially at high flow rates. Our results show that the platform combining DNA nanostructures with microfluidics has a great potential for sensitive isolation of CTCs and is promising for cancer diagnosis and prognosis.
-
Accurate Isolation of Circulating Tumor Cells via a Heterovalent DNA Framework Recognition Element-Functionalized Microfluidic Chip
Yan Liu,Zhun Lin,Ziwei Zheng,Yuanqing Zhang,Lingling Shui
DOI: https://doi.org/10.1021/acssensors.1c02692
IF: 8.9
2022-02-03
ACS Sensors
Abstract:Detection of circulating tumor cells (CTCs) has provided a noninvasive and efficient approach for early diagnosis, treatment, and prognosis of cancer. However, efficient capture of CTCs in the clinical environment is very challenging because of the extremely rare and heterogeneous expression of CTCs. Herein, we fabricated a multimarker microfluidic chip for the enrichment of heterogeneous CTCs from peripheral blood samples of breast cancer patients. The multimarker aptamer cocktail DNA nanostructures (TP-multimarker) were modified on a deterministic lateral displacement (DLD)-patterned microfluidic chip to enhance the capture efficiency through the size selection effect of DLD arrays and the synergistic effect of multivalent aptamers. As compared to a monovalent aptamer-modified chip, the multimarker chip exhibits enhanced capture efficiency toward both high and low epithelial cell adhesion molecule expression cell lines, and the DNA nanostructure-functionalized chip enables the accurate capture of different phenotypes of CTCs. In addition, the DNA nanoscaffold makes nucleases more accessible to the aptamers to release cells with molecular integrity and outstanding cell viability.The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acssensors.1c02692.Details on cell lines and reagents, assembly and characterization of DNA nanostructures, agarose gel electrophoresis analysis, AFM images of the TP multimarker, fluorescence microscopy images of the TP nanostructured chip and DiI-labeled target cells, optimization of the flow rate, cell capture yield toward WBCs, and fluorescence images of cell viability, DNA sequences for the assembly of DNA nanostructures, and data on the number of CTCs captured from breast cancer patients' blood samples (PDF)This article has not yet been cited by other publications.
chemistry, multidisciplinary, analytical,nanoscience & nanotechnology
-
Aptamer-enabled efficient isolation of cancer cells from whole blood using a microfluidic device.
Weian Sheng,Tao Chen,Rahul Kamath,Xiangling Xiong,Weihong Tan,Z Hugh Fan
DOI: https://doi.org/10.1021/ac3005633
IF: 7.4
2012-01-01
Analytical Chemistry
Abstract:Circulating tumor cells (CTC) in the peripheral blood could provide important information for diagnosis of cancer metastasis and monitoring treatment progress. However, CTC are extremely rare in the bloodstream, making their detection and characterization technically challenging. We report here the development of an aptamer-mediated, micropillar-based microfluidic device that is able to efficiently isolate tumor cells from unprocessed whole blood. High affinity aptamers were used as an alternative to antibodies for cancer cell isolation. The microscope-slide-sized device consists of >59 000 micropillars, which enhanced the probability of the interactions between aptamers and target cancer cells. The device geometry and the flow rate were investigated and optimized by studying their effects on the isolation of target leukemia cells from a cell mixture. The device yielded a capture efficiency of similar to 95% with purity of similar to 81% at the optimum flow rate of 600 nL/s. Further, we exploited the device for isolating colorectal tumor cells from unprocessed whole blood; as few as 10 tumor cells were captured from 1 mL of whole blood. We also addressed the question of low throughput of a typical microfluidic device by processing 1 mL of blood within 28 mm. In addition, we found that, similar to 93% of the captured cells were viable, making them suitable for subsequent molecular and cellular studies.
-
Antibody-Functional Microspheres Integrated Filter Chip with Inertial Microflow for Size-Immune-Capturing and Digital Detection of Circulating Tumor Cells.
Yi Su,Qingchang Tian,Dingyi Pan,Lanlan Hui,Yanni Chen,Qi Zhang,Wei Tian,Jie Yu,Shen Hu,Yang Gao,Dahong Qian,Tian Xie,Ben Wang
DOI: https://doi.org/10.1021/acsami.9b09655
IF: 9.5
2019-01-01
ACS Applied Materials & Interfaces
Abstract:Circulating tumor cells (CTCs) in blood is the direct cause of tumor metastasis. The isolation and detection of CTCs in whole blood is very important and of clinical value in early diagnosis, postoperative review and personalized treatment. It is difficult to separate all types of CTCs that efficiently rely on a single path due to cancer cell heterogenicity. Here, we designed a new kind of "filter chip" for the retention of CTCs with very high efficiency by integrating the effects of cell size and specific antigens on the surface of tumor cells. The "filter chip" consists of a semicircle arc and arrays and can separate large-scale CTC-microspheres, which combined with CTCs automatically. We synthesized interfacial zinc oxide coating with nanostructure on the surface of the microsphere to increase specific surface area to enhance capturing efficiency of CTCs. Microspheres, trapped in the arrays, would entrap CTCs, too. The combination of the three kinds of strategies resulted in above 90% of capture efficiency for different tumor cell lines. Furthermore, it is easy to find and isolate the circulating tumor cells from the chip as tumor cells would be fixed inside the structure of "filter chip". To avoid the high background contamination when a few CTCs are surrounded by millions of nontarget cells, a digital detection method was applied to improve detection sensitivity. CTCs in whole blood were specifically labeled by the antibody-DNA conjugates and were detected via the DNA of the conjugates with a signal amplification. The strategy of antibody-functional microspheres integrated microchip for cell sorting and the detection of CTCs may find broad implications that favor fundamental cancer biology research, the precise diagnosis and monitoring of cancer in the clinics.
-
Gold Nanoparticle-Based Microfluidic Chips for Capture and Detection of Circulating Tumor Cells
Valber A Pedrosa,Kangfu Chen,Thomas J George,Z Hugh Fan
DOI: https://doi.org/10.3390/bios13070706
2023-07-04
Abstract:Liquid biopsy has progressed to its current use to diagnose and monitor cancer. Despite the recent advances in investigating cancer detection and diagnosis strategies, there is still a room for improvements in capturing CTCs. We developed an efficient CTC detection system by integrating gold nanoparticles with a microfluidic platform, which can achieve CTC capture within 120 min. Here, we report our development of a simple and effective way to isolate CTCs using antibodies attached on gold nanoparticles to the surface of a lateral filter array (LFA) microdevice. Our method was optimized using three pancreatic tumor cell lines, enabling the capture with high efficiency (90% ± 3.2%). The platform was further demonstrated for isolating CTCs from patients with metastatic pancreatic cancer. Our method and platform enables the production of functionalized, patterned surfaces that interact with tumor cells, enhancing the selective capture of CTCs for biological assays.
-
Nano/Micro Fluidic Systems for Circulating Tumor Cells (ctcs) Rapid Detection and Diagnosis
Fan‐Gang Tseng
2013-01-01
Journal of Aeronautics & Aerospace Engineering
Abstract:A B S T R A C T Despite the recent advancement of biotechnology and pharmaceutical research, cancers remain the leading cause of human mortality. It is vital to diagnose cancers at an early stage when treatment can dramatically improve prognosis. So far, low-cost and easy to operate devices, which allow efficient isolation and sensitive detection of circulating tumor cells (CTCs) for routine blood screening, remain lacking. This talk will introduce a novel micro fluidic platform which can isolate CTCs from the real blood sample in 30 minutes: this system includes a high throughput blood cell separation chip which can separate white blood cells with CTCs from red blood cells and platelets by inertial and suction actions; a nano structured surface which can allow higher retention rate of CTCs on the surface for sample enrichment by 100 folds from 1/107 upto 1/105 CTCs/WBCs.and the enriched sample will go through a final cells self-assembly process into a densed monolayer on a cell assembly chip for in parallel inspection at high speed. As a result, the CTCs can be identified in 30 minutes by the integration of these three chips altogether. Isolated CTCs will still be in vital and can be further characterized and cultivated for the identification of cancer stem cells for prognosis
-
Nanomaterial-Based Immunocapture Platforms for the Recognition, Isolation, and Detection of Circulating Tumor Cells
Yichao Liu,Rui Li,Lingling Zhang,Shishang Guo
DOI: https://doi.org/10.3389/fbioe.2022.850241
IF: 5.7
2022-03-14
Frontiers in Bioengineering and Biotechnology
Abstract:Circulating tumor cells (CTCs) are a type of cancer cells that circulate in the peripheral blood after breaking away from solid tumors and are essential for the establishment of distant metastasis. Up to 90% of cancer-related deaths are caused by metastatic cancer. As a new type of liquid biopsy, detecting and analyzing CTCs will provide insightful information for cancer diagnosis, especially the in-time disease status, which would avoid some flaws and limitations of invasive tissue biopsy. However, due to the extremely low levels of CTCs among a large number of hematologic cells, choosing immunocapture platforms for CTC detection and isolation will achieve good performance with high purity, selectivity, and viability. These properties are directly associated with precise downstream analysis of CTC profiling. Recently, inspired by the nanoscale interactions of cells in the tissue microenvironment, platforms based on nanomaterials have been widely explored to efficiently enrich and sensitively detect CTCs. In this review, various immunocapture platforms based on different nanomaterials for efficient isolation and sensitive detection of CTCs are outlined and discussed. First, the design principles of immunoaffinity nanomaterials are introduced in detail. Second, the immunocapture and release of platforms based on nanomaterials ranging from nanoparticles, nanostructured substrates, and immunoaffinity microfluidic chips are summarized. Third, recent advances in single-cell release and analysis of CTCs are introduced. Finally, some perspectives and challenges are provided in future trends of CTC studies.
multidisciplinary sciences
-
Designer tetrahedral DNA framework-based microfluidic technology for multivalent capture and release of circulating tumor cells
Chenguang Wang,Yi Xu,Shuainan Li,Yi Zhou,Qiuling Qian,Yifan Liu,Xianqiang Mi
DOI: https://doi.org/10.1016/j.mtbio.2022.100346
IF: 8.2
2022-07-20
Materials Today Bio
Abstract:Circulating tumor cells (CTCs) have been recognized as a general biomarker for the early detection, diagnosis and therapy monitoring of cancer. Due to their extreme rarity in peripheral blood, the isolation and analysis of CTCs with high efficiency, high purity and high viability remains a tremendous technological challenge. Herein, we combined tetrahedral DNA framework (TDFs), herringbone channel (HB) chip, together with aptamer-triggered hybridization chain reaction (apt-HCR) to develop an efficient microfluidic system (T-μFS) for capture and release of simulated CTCs. The capture efficiency of MCF-7 cells was from 83.3% to 94.2% when the cell numbers ranged from 10 to 10 3 using our T-μFS in the whole blood. The release efficiency of the MCF-7 cells was 96.2% and the MCF-7 cell viability after release was 94.6% using our T-μFS in PBS buffer. Reculture and RT-qPCR studies showed that there was almost no damage by the capture and release treatment for the MCF-7 cells viability. These results revealed that our T-μFS could be developed as an integrated and automatic technical platform with great performance for multivalent capture and release of CTCs and have a wide application prospect for tumor liquid biopsy.
engineering, biomedical,materials science, biomaterials
-
Microchip-based immunomagnetic detection of circulating tumor cells
Kazunori Hoshino,Yu-Yen Huang,Nancy Lane,Michael Huebschman,Jonathan W Uhr,Eugene P Frenkel,Xiaojing Zhang
DOI: https://doi.org/10.1039/c1lc20270g
2011-10-21
Lab Chip
Abstract:Screening for circulating tumor cells (CTCs) in blood has been an object of interest for evidence of progressive disease, status of disease activity, recognition of clonal evolution of molecular changes and for possible early diagnosis of cancer. We describe a new method of microchip-based immunomagnetic CTC detection, in which the benefits of both immunomagnetic assay and the microfluidic device are combined. As the blood sample flows through the microchannel closely above arrayed magnets, cancer cells labeled with magnetic nanoparticles are separated from blood flow and deposited at the bottom wall of the glass coverslip, which allows direct observation of captured cells with a fluorescence microscope. A polydimethylsiloxane (PDMS)-based microchannel fixed on a glass coverslip was used to screen blood samples. The thin, flat dimensions of the microchannel, combined with the sharp magnetic field gradient in the vicinity of arrayed magnets with alternate polarities, lead to an effective capture of labeled cells. Compared to the commercially available CellSearch™ system, fewer (25%) magnetic particles are required to achieve a comparable capture rate, while the screening speed (at an optimal blood flow rate of 10 mL h(-1)) is more than five times faster than those reported previously with a microchannel-based assay. For the screening experiment, blood drawn from healthy subjects into CellSave™ tubes was spiked with cultured cancer cell lines of COLO205 and SKBR3. The blood was then kept at room temperature for 48 hours before the screening, emulating the actual clinical cases of blood screening. Customized Fe(3)O(4) magnetic nanoparticles (Veridex Ferrofluid™) conjugated to anti-epithelial cell adhesion molecule (EpCAM) antibodies were introduced into the blood samples to label cancer cells, and the blood was then run through the microchip device to capture the labelled cells. After capture, the cells were stained with fluorescent labelled anti-cytokeratin, DAPI and anti-CD45. Subsequent immunofluorescence images were taken for the captured cells, followed by comprehensive computer aided analysis based on fluorescence intensities and cell morphology. Rare cancer cells (from ∼1000 cells down to ∼5 cells per mL) with very low tumor cell to blood cell ratios (about 1 : 10(7) to 10(9), including red blood cells) were successfully detected. Cancer cell capture rates of 90% and 86% were demonstrated for COLO205 and SKBR3 cells, respectively.
-
Real-Time Detection of Tumor Cells during Capture on a Filter Element Significantly Enhancing Detection Rate
Astrid Lux,Hannah Bott,Nisar Peter Malek,Roland Zengerle,Tanja Maucher,Jochen Hoffmann
DOI: https://doi.org/10.3390/bios11090312
2021-09-03
Abstract:Circulating tumor cells (CTCs) that enter the bloodstream play an important role in the formation of metastases. The prognostic significance of CTCs as biomarkers obtained from liquid biopsies is intensively investigated and requires accurate methods for quantification. The purpose of this study was the capture of CTCs on an optically accessible surface for real-time quantification. A filtration device was fabricated from a transparent material so that capturing of cells could be observed microscopically. Blood samples were spiked with stained tumor cells and the sample was filtrated using a porous structure with pore sizes of 7.4 µm. The possible removal of lysed erythrocytes and the retention of CTCs were assessed. The filtration process was observed in real-time using fluorescence microscopy, whereby arriving cells were counted in order to determine the number of CTCs present in the blood. Through optimization of the microfluidic channel design, the cell retention rate could be increased by 13% (from 76% ± 7% to 89% ± 5%). Providing the possibility for real-time detection significantly improved quantification efficiency even for the smallest cells evaluated. While end-point evaluation resulted in a detection rate of 63% ± 3% of the spiked cells, real-time evaluation led to an increase of 21% to 84% ± 4%. The established protocol provides an advantageous and efficient method for integration of fully automated sample preparation and CTC quantification into a lab-on-a-chip system.
-
Nanotechnology-Assisted Isolation and Analysis of Circulating Tumor Cells on Microfluidic Devices
Jie Cheng,Yang Liu,Yang Zhao,Lina Zhang,Lingqian Zhang,Haiyang Mao,Chengjun Huang
DOI: https://doi.org/10.3390/mi11080774
IF: 3.4
2020-08-14
Micromachines
Abstract:Circulating tumor cells (CTCs), a type of cancer cell that spreads from primary tumors into human peripheral blood and are considered as a new biomarker of cancer liquid biopsy. It provides the direction for understanding the biology of cancer metastasis and progression. Isolation and analysis of CTCs offer the possibility for early cancer detection and dynamic prognosis monitoring. The extremely low quantity and high heterogeneity of CTCs are the major challenges for the application of CTCs in liquid biopsy. There have been significant research endeavors to develop efficient and reliable approaches to CTC isolation and analysis in the past few decades. With the advancement of microfabrication and nanomaterials, a variety of approaches have now emerged for CTC isolation and analysis on microfluidic platforms combined with nanotechnology. These new approaches show advantages in terms of cell capture efficiency, purity, detection sensitivity and specificity. This review focuses on recent progress in the field of nanotechnology-assisted microfluidics for CTC isolation and detection. Firstly, CTC isolation approaches using nanomaterial-based microfluidic devices are summarized and discussed. The different strategies for CTC release from the devices are specifically outlined. In addition, existing nanotechnology-assisted methods for CTC downstream analysis are summarized. Some perspectives are discussed on the challenges of current methods for CTC studies and promising research directions.
chemistry, analytical,nanoscience & nanotechnology,instruments & instrumentation,physics, applied
-
Rapid Capture of Rare Cancer Cells Using a High-Performance Microfluidic Chip
Weian Sheng,Tao Chen,Weihong Tan,Hugh Fan
DOI: https://doi.org/10.1115/imece2013-62952
2014-01-01
Abstract:Rare circulating tumor cells (CTCs) are cancer cells that detached from either a primary tumor or metastatic sites. CTCs circulate into bloodstream and become the origin of metastasis, the spread of cancer to distant organs, which is the primary cause of cancer-induced death. This paper describes our development of a microfluidic chip with unique micropillar geometry for rapid capture of CTCs from whole blood. The microscope-slide-sized microchip contains tens of thousands of isotropically-etched elliptical micropillars, which enhanced the interactions between cells and chip surfaces. The microchip was coated with DNA aptamer, an antibody-like molecule which can specifically bind with their target cells. With optimized channel geometry and flow rate, the microchip yielded a capture efficiency of >95% and a purity of >81% when capturing leukemia cells from a cell mixture. Then, the device was applied to capture colorectal tumor cells from whole blood; as few as 10 tumor cells can be efficiently isolated from 1 mL blood in 28 min. We envision that this high-performance microchip is promising for the detection, enrichment and isolation of rare CTCs, and will open up new opportunities for cancer diagnosis and monitoring cancer treatment.
-
Abstract 3307: Label-free high throughput microfluidic device for the isolation and single cell multiplex gene expression analysis of circulating tumor cells from breast cancer patients
Eric Lin,Lianette Rivera,Shamileh Fouladdel,Hyeun Joong Yoon,Stephanie Guthrie,Jacob Weiner,Yadwinder S. Deol,Evan Keller,Vaibhav Sahai,Diane M. Simeone,Monika L. Burness,Ebrahim Azizi,Max S. Wicha,Sunitha Nagrath
DOI: https://doi.org/10.1158/1538-7445.am2016-3307
2016-07-15
Tumor Biology
Abstract:Abstract Introduction and Objective: The metastasis of cancer is preceded by the dissemination of cancer cells from the primary tumor site to remote sites via the blood circulation. The presence of circulating tumor cells (CTCs) in the peripheral blood represents a strong and independent prognostic factor for decreased disease-free and overall survival. Immune-affinity based capture, although being the most commonly used method for the isolation of CTCs, offers low throughput (∼1mL/hr) and have considerably cell loss caused by the heterogeneous expression of biomarkers on CTCs. Various label-free approaches utilizing the physical properties of CTCs have been developed to overcome the limitations, such as micro-filters, microscale laminar vortices, inertial migration of particles, and integrated systems. Here we present an inertial microfluidic-based separation technique for high throughput and label-free isolation of CTCs yielding the highest throughput with high CTC recovery and high blood cell removal among all the label-free technologies. The isolated CTC populations were further analyzed for single cell multiplex gene expression analysis. Methods: The PDMS-made inertial microfluidic device has 637 mm in length with 56 corners, 500 μm in width, and 100 μm in height. The separation of CTCs is driven by two main forces: (i) inertial force that focuses the cells into streamlines, and (ii) drag force from Dean flow that migrates the focused cells to various positions based on size. Device is optimized with MCF-7 and Panc-1 cell line within PBS buffer solution and diluted blood, and is tested in patients with breast cancer on an average of 5 mL of whole blood processed through double devices in series. CTCs isolated were analyzed for tumor specific protein markers and genomic characterization is done using singe cell analysis techniques. Results: Samples are processed through the inertial microfluidic device and CTCs are enriched in second outlet based on size difference between CTCs and blood cells. Device is optimized to operate at an extremely high throughput of 2500 μL/min with high recovery (greater than 90%) and high white blood cells (WBCs) removal (5 log orders). In patient samples, we identified CTCs in 38 of 40 (95%) breast patients with metastatic disease (5.4±4.6 CTC/mL) with low WBC contamination (663±647 WBC/mL). Based on the gene expression, both inter and intra patient heterogeneity of CTCs at the single cell level were discovered among the tested patient samples. Conclusion: The study of CTCs could have a direct impact upon society by presenting novel ways to address one of the major hurdles in cancer research - early detection - and will foster the advancement of science and engineering via the exploration of new druggable targets approaches and the further understanding of the pharmacodynamics. Citation Format: Eric Lin, Lianette Rivera, Shamileh Fouladdel, Hyeun Joong Yoon, Stephanie Guthrie, Jacob Weiner, Yadwinder S. Deol, Evan Keller, Vaibhav Sahai, Diane M. Simeone, Monika L. Burness, Ebrahim Azizi, Max S. Wicha, Sunitha Nagrath. Label-free high throughput microfluidic device for the isolation and single cell multiplex gene expression analysis of circulating tumor cells from breast cancer patients. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3307.
-
Combining hybrid cell membrane modified magnetic nanoparticles and inverted microfluidic chip for in situ CTCs capture and inactivation
Jiao Sun,Songrui Han,Rui Yang,Lihua Guo,Jiawei Li,Chunxia Li,Lin Xu,Haipeng Liu,Biao Dong
DOI: https://doi.org/10.1016/j.bios.2024.116575
2024-11-01
Abstract:Circulating tumor cells (CTCs) serve as crucial indicators for tumor occurrence, progression, and prognosis monitoring. However, achieving high sensitivity and high purity capture of CTCs remains challenging. Additionally, in situ capture and synchronous clearance hold promise as methods to impede tumor metastasis, but further exploration is needed. In this study, biomimetic cell membrane-coated magnetic nanoparticles (NPs) were designed to address the issue of nonspecific adsorption of capture probes by the immune system during blood circulation. Membranes from human breast cancer cells (tumor cell membranes, TMs) and leukocytes (white blood cell membranes, WMs) were extracted and fused to form a hybrid membrane (HM), which was further modified onto the surface of porous magnetic NPs loaded with indocyanine green (ICG). The incorporation of TM enhanced the material's target specificity, thus increasing capture efficiency, while WM coating reduced interference from homologous white blood cells (WBCs), further enhancing capture purity. Additionally, in conjunction with our novel inverted microfluidic chip, this work introduces the first use of polymer photonic crystals as the capture interface for CTCs. Besides providing an advantageous surface structure for CTC attachment, the 808 nm photonic bandgap effectively amplifies the 808 nm excitation light at the capture surface position. Therefore, upon capturing CTCs, the ICG molecules in the probes facilitate enhanced photothermal (PTT) and photodynamic (PDT) synergistic effects, directly inactivating the captured CTCs. This method achieves capture efficiency and purity exceeding 95% and permits in situ inactivation post-capture, providing an important approach for future research on impeding tumor metastasis in vivo.
-
Highly Efficient Circulating Tumor Cell Isolation from Whole Blood and Label-Free Enumeration Using Polymer-Based Microfluidics with an Integrated Conductivity Sensor
André A. Adams,Paul I. Okagbare,Juan Feng,Matuesz L. Hupert,Don Patterson,Jost Göttert,Robin L. McCarley,Dimitris Nikitopoulos,Michael C. Murphy,Steven A. Soper,André A. Adams,Jost Göttert
DOI: https://doi.org/10.1021/ja8015022
IF: 15
2008-06-17
Journal of the American Chemical Society
Abstract:A novel microfluidic device that can selectively and specifically isolate exceedingly small numbers of circulating tumor cells (CTCs) through a monoclonal antibody (mAB) mediated process by sampling large input volumes (>/=1 mL) of whole blood directly in short time periods (<37 min) was demonstrated. The CTCs were concentrated into small volumes (190 nL), and the number of cells captured was read without labeling using an integrated conductivity sensor following release from the capture surface. The microfluidic device contained a series (51) of high-aspect ratio microchannels (35 mum width x 150 mum depth) that were replicated in poly(methyl methacrylate), PMMA, from a metal mold master. The microchannel walls were covalently decorated with mABs directed against breast cancer cells overexpressing the epithelial cell adhesion molecule (EpCAM). This microfluidic device could accept inputs of whole blood, and its CTC capture efficiency was made highly quantitative (>97%) by designing capture channels with the appropriate widths and heights. The isolated CTCs were readily released from the mAB capturing surface using trypsin. The released CTCs were then enumerated on-device using a novel, label-free solution conductivity route capable of detecting single tumor cells traveling through the detection electrodes. The conductivity readout provided near 100% detection efficiency and exquisite specificity for CTCs due to scaling factors and the nonoptimal electrical properties of potential interferences (erythrocytes or leukocytes). The simplicity in manufacturing the device and its ease of operation make it attractive for clinical applications requiring one-time use operation.
chemistry, multidisciplinary
-
Enhancing the efficiency of lung cancer cell capture using microfluidic dielectrophoresis and aptamer‐based surface modification
Shu‐Hui Lin,Tzu‐Cheng Su,Shuo Jie Huang,Chun‐Ping Jen
DOI: https://doi.org/10.1002/elps.202300206
2024-01-06
Electrophoresis
Abstract:Metastasis remains a significant cause to cancer‐related mortality, underscoring the critical need for early detection and analysis of circulating tumor cells (CTCs). This study presents a novel microfluidic chip designed to efficiently capture A549 lung cancer cells by combining dielectrophoresis (DEP) and aptamer‐based binding, thereby enhancing capture efficiency and specificity. The microchip features interdigitated electrodes made of indium‐tin‐oxide that generate a nonuniform electric field to manipulate CTCs. Following three chip design, scenarios were investigated: (A) bare glass surface, (B) glass modified with gold nanoparticles (AuNPs) only, and (C) glass modified with both AuNPs and aptamers. Experimental results demonstrate that AuNPs significantly enhance capture efficiency under DEP, with scenarios (B) and (C) exhibiting similar performance. Notably, scenario (C) stands out as aptamer‐functionalized surfaces resisting fluid shear forces, achieving CTCs retention even after electric field deactivation. Additionally, an innovative reverse pumping method mitigates inlet clogging, enhancing experimental efficiency. This research offers valuable insights into optimizing surface modifications and understanding key factors influencing cell capture, contributing to the development of efficient cell manipulation techniques with potential applications in cancer research and personalized treatment options.
biochemical research methods,chemistry, analytical
-
Isolation of DNA aptamers targeting N-cadherin and high-efficiency capture of circulating tumor cells by using dual aptamers
Tian Gao,Pi Ding,Wenjing Li,Zhili Wang,Qiao Lin,Renjun Pei
DOI: https://doi.org/10.1039/d0nr06180h
IF: 6.7
2020-01-01
Nanoscale
Abstract:Circulating tumor cells (CTCs) acquire mesenchymal markers (e.g., N-cadherin) and lose epithelial markers (e.g., epithelial cell adhesion molecule, EpCAM) during the epithelial-mesenchymal transition (EMT) and are therefore ideal biomarkers of tumor metastasis. However, it is still a challenge to efficiently capture and detect circulating tumor cells with different phenotypes simultaneously. In this work, to obtain aptamers targeting N-cadherin in the native conformation on live cells, we established stable N-cadherin overexpressing cells (N-cadherin cells) and used these cells to identify a panel of N-cadherin-specific aptamers through the cell-SELEX approach. Two aptamer candidates obtained after 12 rounds of selection showed a low equilibrium dissociation constant in the nanomolar range, indicating high binding affinity. The truncated aptamer candidate NC3S showed the highest binding affinity to N-cadherin cells with a low K-d value of 20.08 nM. The SYL3C aptamer was reported to target cancer cell surface biomarker EpCAM. Then, we synthesized two kinds of aptamer-modified magnetic nanoparticles (SYL3C-MNPs and NC3S-MNPs). Both SYL3C and NC3S aptamers possess excellent capture specificity and efficiency for the target cells. The aptamer-MNP cocktail exhibits a considerable capture efficiency and sensitivity for rare cancer cells of epithelial and mesenchymal phenotypes. Furthermore, no CTCs were found in blood samples from healthy donors, while CTCs were successfully isolated by using the aptamer-MNP cocktail for 15 out of 16 samples collected from patients. In summary, the two kinds of aptamer-modified MNPs could be utilized as a promising tool for capturing CTCs from clinical samples.
-
On-chip Circulating Tumor Cells Isolation Based on Membrane Filtration and Immuno-Magnetic Bead Clump Capture
Shuai Zhang,Yue Wang,Chaoqiang Yang,Junwen Zhu,Xiongying Ye,Wenhui Wang
DOI: https://doi.org/10.1063/10.0009560
2022-01-01
Nanotechnology and Precision Engineering
Abstract:Isolating rare circulating tumor cells (CTCs) from blood is critical for the downstream analysis that is important in cancer-related research, diagnosis, and medicine, and efforts are ongoing to increase the efficiency and purity of CTC isolation in microfluidics. Reported in this paper is a two-stage integrated microfluidic chip for coarse-to-fine CTC isolation from whole blood. First, blood cells are removed by filtration using a micropore-array membrane, then CTCs and other cells that are trapped in the micropores are peeled off the membrane by a novel release method based on air–liquid interfacial tension, which significantly increases the recovery rate of CTCs. The second stage is CTC capture based on an on-chip dense immuno-magnetic-bead clump, which offers high capture efficiency and purity. Both the micropore filtration and immuno-magnetic-bead capture are validated and optimized experimentally. Overall, the integrated microfluidic chip can realize a recovery rate of 85.5% and a purity of 37.8% for rare cancer cells spiked in whole blood.
-
Affinity-Enhanced CTC-Capturing Hydrogel Microparticles Fabricated by Degassed Mold Lithography
Nak Jun Lee,Sejung Maeng,Hyeon Ung Kim,Yoon Ho Roh,Changhyun Hwang,Jongjin Kim,Ki-Tae Hwang,Ki Wan Bong
DOI: https://doi.org/10.3390/jcm9020301
IF: 3.9
2020-01-21
Journal of Clinical Medicine
Abstract:Technologies for the detection and isolation of circulating tumor cells (CTCs) are essential in liquid biopsy, a minimally invasive technique for early diagnosis and medical intervention in cancer patients. A promising method for CTC capture, using an affinity-based approach, is the use of functionalized hydrogel microparticles (MP), which have the advantages of water-like reactivity, biologically compatible materials, and synergy with various analysis platforms. In this paper, we demonstrate the feasibility of CTC capture by hydrogel particles synthesized using a novel method called degassed mold lithography (DML). This technique increases the porosity and functionality of the MPs for effective conjugation with antibodies. Qualitative fluorescence analysis demonstrates that DML produces superior uniformity, integrity, and functionality of the MPs, as compared to conventional stop flow lithography (SFL). Analysis of the fluorescence intensity from porosity-controlled MPs by each reaction step of antibody conjugation elucidates that more antibodies are loaded when the particles are more porous. The feasibility of selective cell capture is demonstrated using breast cancer cell lines. In conclusion, using DML for the synthesis of porous MPs offers a powerful method for improving the cell affinity of the antibody-conjugated MPs.
medicine, general & internal
-
Highly Efficient Capture of Circulating Tumor Cells with Low Background Signals by Using Pyramidal Microcavity Array
Jiaxiang Yin,Lei Mou,Mingzhu Yang,Wenwu Zou,Chang Du,Wei Zhang,Xingyu Jiang
DOI: https://doi.org/10.1016/j.aca.2019.01.054
IF: 6.911
2019-01-01
Analytica Chimica Acta
Abstract:This report demonstrates that a microfluidic device with integrated silicon filter exhibits outstanding capture efficiency and superior enrichment purity when employed to separate tumor cells from whole blood samples. We fabricate the silicon filter with pyramidal microcavity array (MCA) by micro-fabrication. We design the structure of the cavity to efficiently enrich tumor cells, while allowing hematologic cells to deform and pass through. The capture efficiency of MCF-7, SW620 and Hela cells spiked in 1 mL of whole blood are approximately 80%. Unwanted white blood cells (WBCs) trapped on the MCA are below 0.003%. In addition, this microfluidic device successfully identifies circulating tumor cells (CTCs) in 5 of 6 patients' blood samples, with a range of 5-86 CTCs per mL. These results reveal that the disposable microfluidic device can effectively enrich tumor cells with different sizes and various morphologies, while maintaining high capture efficiency and purity. Therefore, this label-free technique can serve as a versatile platform to facilitate CTCs analysis in diverse biochemical applications. (C) 2019 Elsevier B.V. All rights reserved.