Abstract
Circulating tumor cells (CTCs) are tumor cells detached from the original lesion and getting into the blood and lymphatic circulation systems. They potentially establish new tumors in remote areas, namely, metastasis. Isolation of CTCs and following biological molecular analysis facilitate investigating cancer and coming out treatment. Since CTCs carry important information on the primary tumor, they are vital in exploring the mechanism of cancer, metastasis, and diagnosis. However, CTCs are very difficult to separate due to their extreme heterogeneity and rarity in blood. Recently, advanced technologies, such as nanosurfaces, quantum dots, and Raman spectroscopy, have been integrated with microfluidic chips. These achievements enable the next generation isolation technologies and subsequent biological analysis of CTCs. In this review, we summarize CTCs’ separation with microfluidic chips based on the principle of immunomagnetic isolation of CTCs. Fundamental insights, clinical applications, and potential future directions are discussed.
I. INTRODUCTION
Circulating tumor cells (CTCs),1 discovered by an Austrian physicist, Thomas Ashworth, in 1869,2 are cancer cells shed from the primary or secondary lesion. They enter into the blood and lymphatic circulation systems, spreading and forming distant tissues.3–5 The behavior of those CTCs might finally lead to cancer metastasis, responsible for almost 90% death of cancer patients.6–8 Thus, the detection of CTCs in the blood sample indicates cancer and possible metastasis. Also, since CTCs carrying significant molecular information from the primary tumor,9 detecting and characterizing CTCs would contribute to the understanding of cancer and its metastasis mechanism. Further, the number of CTCs is related to the overall survival of patients and the severity of cancer,10–14 as less number indicates disease improvement or effective treatment. Thus, the precise enumeration of CTCs has endowed more significant values in prognosis and treatment procedures. It could be seen that CTCs have prognostic relevance and clinical meaning.15–18
As seen in Fig. 1, microfluidic chips are non-invasive or conservative “liquid biopsy” detectors.19–21 This technology would minimize the pain in cancer detection. However, CTCs are extremely rare and heterogeneous, with approximately 1–10 CTCs amid millions of white blood cells (WBCs) and billions of red blood cells (RBCs) in 1 ml whole blood.22,23
FIG. 1.
Schematic of all kinds of microfluidic chips for isolating circulating tumor cells.
Until now, Cellsearch™ (developed by Veridex Raritan, NJ, USA) is the only device approved by the US Food and Drug Administration (FDA) for successful clinical CTC enumeration system in breast, prostate, and colorectal cancers.24–27 As reported, the efficiency of this system was approximately 80%.28 In this device, immunomagnetic technology is through ferrofluidic nanoparticles labeled with antibodies targeting epithelial cell adhesion. CTCs are identified through fluorescent staining of cytokeratins proteins of keratin-containing intermediate filaments in the intracytoplasmic cytoskeleton of epithelial tissue. Leukocytes are excluded by CD45 (Leucocyte Common Antigen) antibody staining. To our knowledge, the main limitation of CellSearch™ lies in the ineffectiveness of cells with low expression of epithelial markers. This ineffectiveness affects cell viability and further downstream characterization. Also, some leukocytes of epithelial expression would cause false-positive results. Besides, it is still semi-automatic with low efficiency and high cost.
Besides the commercialized products, more advanced progress has been made toward capturing and characterizing CTCs with microfluidic chip-based technology. Microfluidic chips have unique advantages over conventional approaches. For example, in the separation of CTCs using density gradient centrifugation, the fluid media, Ficoll–Hypaque, is toxic to cells.29 Flow cytometry always requires pre-enrichment. But the cell loss may occasionally happen in the pre-separation stage. On the other hand, microfluidic chips could integrate different stages on a small chip, which exhibits high potential applications in relevant technologies such as DNA sequencing,30,31 protein detection,32,33 cell manipulation, and molecular analysis.34,35 Besides, materials for microfluidic chips, such as PDMS, are always biocompatible and low-cost. All these merits make microfluidic chips one of the best candidates for cell-scale CTCs sorting.
Until now, there have been few reviews about the immunomagnetic separation of CTCs with microfluidic chips and their clinical application. We analyze each typical microfluidic chips and its working principle as well as clinical assays. Some viewpoints described in the manuscript, such as reproducibility, clinical assay, advantages and disadvantages, and future prospective, are new. From those points of view, this review manuscript is unique. In this paper, we reviewed emerging immunomagnetic-based microfluidic chips designed for capturing and isolating CTCs. Prospective potential research interests of CTCs’ capture based on this principle are presented and discussed.
II. CURRENT MICROFLUIDIC CHIP-BASED METHODOLOGIES FOR ISOLATION OF CTCs
A. Current technologies
To date, the CTC microfluidic chips can be classified into the following three types according to different working principles:
-
(1)
Immunoaffinity-based, which depends on the specific binding of antigens of CTCs to antibodies modified on microchannels of the microfluidic chip.36–54
-
(2)
Physical-based, which depends on the physical property differences, such as size/deformability discrepancy between CTCs and hematological cells.55–73
-
(3)
Immunomagnetic-based, in which CTCs are bonded with immunomagnetic beads with size amplification and magnetization.74–82
Most technologies, such as dielectrophoresis-based,83–86 acoustic-based, electrophoresis-based,87,88 hydrodynamic and cross-flow filtration,89,90 deterministic lateral displacement (DLD),91,92 and inertial focusing systems,93–95 including the viscoelasticity aided method,96 fall into these three categories.
B. Separation evaluation
Parameters used to evaluate the efficiency of microfluidic chips are as follows:
| (1a) |
| (1b) |
| (2) |
| (3) |
| (4) |
The reason for formula (1b) seldom getting involved in capture efficiency (CE) calculation, but more precise is due to sedimentation. From rich experience, we could conclude that formula (1b) could better reflect the real value. As CTCs are usually heavier than blood constitutes with bigger size, CTCs tend to precipitate in the front part of the syringe.97 In particular, in the case of slow flow, it would cause the total amount of CTCs captured and escaped from the outlet is less than the number of CTCs spiked in. The sedimentation phenomenon could be observed if tumor cells coated with dark immunomagnetic beads.
Toward the clinical trial, it is significant to evaluate new technologies based on technical specifications. For example, capture efficiency is the most critical parameter in CTCs’ isolation. It describes how many CTCs are captured by the microfluidic chip with respect to spiked CTCs. Precisely enumerating almost every single CTC is significant due to the rarity of CTCs in a limited blood sample. Purity, another critical parameter, is how many CTCs captured among hematological cells not purposely captured. High purity would contribute to preventing leukocyte buildup on the surfaces of the microfluidic chip. Specificity is the ability to recognize CTCs even with some disturbance such as WBCs and RBCs. Specificity has to be high for CTCs to be identified among interference. Specificity has to be acceptable with minimum non-specific adhesion. Viability is whether CTCs are viable after captured. Viability would facilitate the following culture and further treatment. A low CTC damage rate is preferred, which avoids a sharp edge, and high shear rates are recommended. Thus, tumor cells would be kept viable for following biological molecular analysis and reculture. High throughput is to process more blood in unit time. Owing to the short half-life of CTCs, 2–4 h,23 high throughput is favorable to shorten processing time to keep the viability of CTCs captured. Usually, a high flow rate is required to isolate CTCs faster. Another significant metric in CTCs’ isolation, reproducibility, is key for clinical application and usually highly neglected. That is, whether the assay is a success for randomly chosen test statistically. High efficiently detecting CTCs should not be occasional, but it should be successful for each test in the sense of statistics based on responsibility on every patient. The metric of reproducibility should be significantly strengthened for future research.
III. IMMUNOMAGNETIC SEPARATION OF CTCs
Affinity ligand-functionalized micro-/nanoparticles, such as epithelial cell adhesion molecule (EpCAM) antibody-coated magnetic microbeads, are utilized for on-chip CTCs segregation.98–110 Target cells are coated with microbeads modified with an antibody. They are incubated through mixing and shaking. Through this procedure, CTCs are magnetizing and could be attracted by a magnetic field. During the release, removing the magnetic field, magnetic attraction disappears. CTCs captured could be enriched. Examples of immunomagnetic-based isolation of CTCs are listed as follows.
A. Non-hybrid microfluidic chips
1. An intravascular magnetic wire for retrieving CTCs with high throughput
In 2018, Ophir Vermesh and co-workers proposed an intravascular magnetic wire to retrieve rare biomarkers such as CTCs from the subject's blood in vivo at a rich yield.98 The MagWIRE could be inserted and removed through an intravenous catheter to capture biomarkers. Those biomarkers are prelabeled with injected magnetic particles. In an experiment to evaluate the MagWIRE in a live porcine model, in vivo labeling could be achieved, and single-pass capture of viable model CTCs could be performed in less than 10 s. The results are so satisfied with the amount of CTCs 10–80 times the amount in the 5-ml blood draw. Compared with the commercially available Gilupi CellCollector the enrichments are 500–5000 times. For conventional microfluidic chips isolating CTCs, the number of CTCs for 5 ml patient blood is few. Thus, the requirement for capture efficiency is high such as to 90%. However, more number of CTCs enriched brings benefit to re-culture for following biological molecular analysis. This MagWIRE could be performed with such a high throughput to yield more number of CTCs. This design would ease following molecular analysis and therapy. But the disadvantage of this approach is kind of invasive to input this MagWIRE to intravenous catheter to realize capture. Almost the majority of microfluidic chips in isolating CTCs are “non-invasive” and extracorporeal. Non-invasive methods are much safer and easy to operate. Only CTCs coated with magnetic particles could be captured. Therefore, it would harm the viability of CTCs for subsequent culture.
2. A two-dimensional immunomagnetic nano-net
In 2019, Chian-Hui Lai and co-workers proposed a two-dimensional immunomagnetic “nano-net,” which was designed and synthesized for efficient isolation of CTCs from whole blood (Fig. 2).111 The nano-net, namely, Ab@Lipo-MNP-GO, was composed of conjugated antibody molecules on a lipid coated magnetic nanoparticle-graphene oxide sheet complex. Superconducting quantum interference device (SQUID), Fourier-transform infrared spectrometer (FTIR), thermal gravimetric analysis (TGA), dynamic light scattering (DLS), and scanning electron microscope (SEM) were used to characterize the magnetism, chemical composition, and the morphology of the construct. Superior properties prove the high possibility of capture of synthetic “nano-net.” Clinical patient samples of oral, colon, and lung cancers were utilized to test the performance. The characteristic feature is a stable, patch-like multivalent matrix nano-net instead of point contact. From clinical evaluation, the magnetic capture with “nano-net” showed superiority to conventional magnetic capture. Contact areas are greatly enlarged compared to traditional point-to-point contact. Thus, the contact possibility is highly enhanced. However, from the following diagram, it could be seen that the synthetic procedure of this “nano-net” is very complicated. The antibody is very expensive. Comparing with usual “attraction” of magnetic nanoparticles, point-to-point contact has been transferred to area-to-area contact creating more strong chances. Conjugating becomes relatively easy. However, it is difficult for captured CTCs to get rid of this “nano-net.” The aim of CTCs capture is following biological molecular analysis. This strong attachment inhibits cell viability and sets up a barricade for subsequent analysis. Therefore, although capture efficiency is highly improved, the “nano-net” is not suitable for clinical application.
FIG. 2.
Schematic illustration of CTCs capture.111 Reproduced from Lai et al., Nanoscale 11(44), 21119 (2019). Copyright 2019 American Chemical Society. (a) Immunomagnetic beads, Ab@beads, with point contacts. (b) Patch-like immunomagnetic nano-nets, Ab@Lipo-MNP-GO. (c) Synthetic steps of the nano-net (GO:graphene oxide; MNP@GO: magnetic nanoparticle-GO; Lipo@MNP-GO: lipid bilayer coated MNP-GO; Ab@Lipo-MNP-GO: antibody conjugated lipid bilayer-MNP-GO complex).
3. Microfluidic immunomagnetic detecting chamber
In 2011, Hoshino et al. demonstrated the immunomagnetic detection of CTCs.112 A microchip-based immunomagnetic CTCs’ detection described cancer cells labeled with magnetic nanoparticles, entering the chamber, deposited at the bottom of a chamber with arrayed magnets located below (Fig. 3). Customized Fe3O4 magnetic nanoparticles conjugated to anti- EpCAM antibodies were added to the blood samples to label cancer cells. In artificial patient blood, capture rates are at 90% and 86% for COLO205 and SKBR3 cells, respectively, even at 10 ml/h. But this magnetic “attraction” would fail into a valid one since the key requirement of every CTC bonded with magnetic beads was not guaranteed. A large number of hematological cells disturb the conjugating of CTCs with magnetic beads. Those CTCs without coating with magnetic nanoparticles escape away, especially at a high flow rate.
FIG. 3.
Microchip design for immunomagnetic detection of the cancer cells.112 Reproduced with permission from Hoshino et al., Lab Chip 11, 3499 (2011). Copyright 2011 American Chemical Society. (a) The principle of immunomagnetic capture of CTCs. (b) The dimension of the microchannel. (c) Schematic of the pneumatic flow system. (d) and (e) TEM images of Fe3O4 magnetic nanoparticles.
4. A combined micromagnetic–microfluidic device
In 2012, Kang et al. reported a combined micromagnetic–microfluidic device for rapid capture and culture of rare CTCs (Fig. 4).113 The device contained a main microfluidic channel and redundant “double collection” channel lined by two rows of dead-end side chambers with a permanent magnet placed directly beneath the lower row of the side chamber to realize tumor cells captured. NdFeB N52 magnet with a magnetic flux density of 14.5 KGs was displaced below the lower row of the collection side chamber of 500 μm. Isolation efficiency was as high as 87% for M6C cells with better than 90% of the isolated CTCs keeping viable after directly tested in the side chambers of the device. Retrieving isolated CTCs from the channels was achieved by shifting the position of the magnet to the opposite side of the device and flushing with PBS. This high-efficient cell separation technique was validated for CTCs circulating in blood of transgenic FVB C3(1)-SV40T-antigen transgenic mice that experience progression from normal mammary glands to metastatic mammary tumors. As a result, the number of CTCs isolated from the blood of transgenic mice increases with cancer progression. Isolated CTCs from the blood of mice mixing with 4T1 breast tumors proliferated for seven days on a tissue culture plate. In this capture, the side chamber played a significant role in sorting and recovering CTCs, but slim chances still exist for CTCs missing in the main chamber. The reason is that if CTCs could not bond with magnetic nanoparticles, those CTCs would be lost.
FIG. 4.
Design and operation of the immunomagnetic microfluidic chip.113 Reproduced with permission from Kang et al., Lab Chip 12, 2175 (2012). Copyright 2012 American Chemical Society. (a) The principle of operation. The device contains an angled inlet channel that connects to the main channel followed by a dual collection channel before going out through the outlet. Rows of dead-end side chambers line the main channel and double collection channel. Permanent magnets are placed beneath the lower row of side chambers to collect magnetic bead-bound cells. (b) Magnified view of part (a). (c) A photograph of a PDMS device.
5. Self-assembled magnetic bead patterns
In 2010, Sivagnanam demonstrated self-assembled magnetic bead patterns in a microfluidic chip to isolate and on-chip culture of CTCs.114 Arrays of positively charged (3-aminopropyl)triethoxysilane (APTES) microdots were patterned on a glass substrate and then negatively charged streptavidin-coated magnetic beads were self-assembled on the APTES dots by the external magnetic field. The attached microbeads were modified with biotinylated 5D10 monoclonal antibodies (mAb) and biotinylated fibronectin (FN) in order to capture and culture MCF-7. A NdFeB was placed underneath the microfluidic chip producing a magnetic field of ∼100 mT on top of the glass substrate. Capture efficiency could reach 85 ± 10% for cultured MCF-7 cells to be selectively captured from Jurkat cells.
6. Self-assembled magnetic arrays, “Ephesia”
Saliba and co-workers demonstrated an “Ephesia,” the self-assembly of super-paramagnetic beads into an array of vertical bead columns (Fig. 5).115 “Ephesia,” formed by dipole–dipole interaction on the magnetic dots from microcontact printing of a ferrofluid onto the glass substrate, trap tumor cells among mixture at a capture yield better than 94%. Magnetic beads with a diameter of 4.5 μm coated anti-CD19 mAb could be self-assembled to form bead columns. Cultivating in situ the captured cells were also possible [Fig. 4(b)]. Clinically, this chip is validated with different types of lymphomas and leukemia: chronic lymphocytic leukemia (CLL) (n = number of patients = 4), mantle cell lymphoma (n = 1), and follicular lymphoma (n = 2), and two healthy volunteers. Magnetic bead patterns and “Ephesia” are formed in such a fantastic way, automatically producing microposts on the substrate to realize capturing, isolating, and even culture. However, those “artificial” microposts are not strong enough to sustain capture comparing with usual ones, especially when the flow rates have been increased.
FIG. 5.
Principle and practical implementation of Ephesia system.115 Reproduced with permission from Saliba et al., Proc. Natl. Acad. Sci. U.S.A. 107, 14524 (2010). Copyright 2010 American Chemical Society. (A) Principle of magnetic self-assembly. Without a magnetic field, beads coated with an antibody are randomly distributed. They would form a regular array of bead columns on top of the ink dots under an external vertical magnetic field. (B) Two levels PDMS integrated microchip. (C) A magnetically assembled array of columns of 4.5 μm beads coated with anti-CD19 mAb. (D) Optical micrograph of the columns after the passage of 1000 Jurkat cells. (E) Captured tumor cells around the columns after the passage of 400 Raji cells.
7. A surface-enhanced Raman scattering (SERS)-coding microsphere suspension chip
Except for those magnetic attractions, magnetic nanoparticles are gradually and extensively applied to combine with light characteristics. Li et al. also creatively figured out a surface-enhanced Raman scattering (SERS)-coding microsphere suspension chip.116 Folate modified the surfaces of magnetic composite microspheres to capture Hela cells achieving capturing efficiency of 95%. The magnetic supraparticles (MSPs) are composed of high magnetic-responsive supraparticles as a core and a hydrophilic polymeric gel containing targeting molecules as shell. The modified folate (∼200 nm in diameter) can be attached to the polymethacrylic acid (PMAA) shell to form MSPs@PMMA-SS-FA. 90% cells could be recovered within 20 min with glutathione solution.
8. A multifunctional biocompatible graphene oxide quantum dots (GOQDs) coated, magnetic nanoplatform
In 2015, Shi and colleagues presented a multifunctional magnetic nanoplatform to selectively separate HepG2 live cancer tumor CTCs from Glypican-3(GPC3)-expression117 using graphene oxide quantum dots (GOQDs) coated technology. Captured HepG2 tumor cells could be identified on the anti-GPC3-antibody-attached GOQDs-coated magnetic nanoplatform through a two-photon luminescence platform with 960 nm light. The GPC3-specific monoclonal anti-GPC3 antibody, specific to HCC tumor cells, was utilized to attach to the amine-functionalized PEG-coated GOODs-attached magnetic nanoplatforms. Capture efficiency is as high as 97%, and 98% of cells were kept viable after 24 h.
9. Highly qualitatively and quantifiably capture CTCs combining quantum dots (QDs) and magnetic beads
Innovatively, Min et al. reported highly qualitatively and quantifiably captured CTCs combining Quantum Dots (QDs) and Magnetic Beads.118 Anti-EpCAM antibody-conjugated QDs (anti-EpCAM-QDs) target the EpCAM antigen on the surface of the cancer cell for seizing cells. Anti-mouse IgG-modified-magnetic beads (anti-IgG-MBs) function as isolating CTCs with the aid of a magnetic field. Specifically, anti-IgG-MBs could bind to anti-EpCAM antibodies on QDs (anti-EpCAM-QDs). Construction of the topographical objects of QDs could enhance the roughness of CTCs surfaces. This increase enables the multivalent binding of anti-IgG-MBs. As a result, the capture efficiency is 80% for the cells ranging from 100 to 20 cells ml−1, and the capture efficiency for SK-Br3 achieved 70% for 3000 cells mixed with HL-60 cells of 1.0 × 105 in 1 ml of serum-free medium, respectively, with a purity of 18% to 23%. It not only realizes magnetic-based capturing but also technically and creatively introduces QDs to change the morphology of CTCs to serve better sorting. Simple quantification of captured CTCs could be achieved based on the fluorescence intensity of the quantum dots. They use a model cell line, SK-Br3, with a number distributed from 4.0 × 102 to 5.0 × 104. Initially following this conjugation, the fluorescence intensity from the QD-attached cells was measured. A linear relationship was obtained for fluorescence intensity with the number of cells. With this line, the unknown number of cells could be estimated by measuring fluorescence intensity. The detection limit was 390 cells ml−1. This detection and characterization approach is new, and the capture efficiency is increased through enhancing binding with magnetic beads. However, for the clinical patient sample, there are only 1–100 CTCs in 7.5 ml patient blood. Those tumor cells used are beyond the actual number of CTCs, and the capture efficiency is not high enough to perform clinical tests.
10. CTCs’ isolation and detection based on ZnS:Mn2+ quantum dots and magnetic nanocomposites
Cui presented rapid and efficient isolation and detection of circulating tumor cells based on ZnS:Mn2+ quantum dots and magnetic nanocomposites (Fig. 6).119 Instead of conventional three-color immunofluorescence identification, simultaneously captured CTCs could be achievable due to immunonanocomposites [ZnS:Mn2+ quantum dots (QDs) and Fe3O4/SiO2] equipped with permanent fluorescent and magnetic properties. A multifunctional nanocomposite was synthesized by encapsulating ZnS: Mn2+ quantum dots (QDs) and Fe3O4 nanoparticles into SiO2 nanospheres and bio-conjugating tumor-specific anti-EpCAM antibodies onto the surface. The synthesized nanocomposite had a high tumor cell binding ability. Fe3O4 nanoparticles had a rapid magnetic response that enabled capture of CTCs from patients' blood within minutes. Moreover, the yellow-orange light emitted by the ZnS: Mn2+ quantum dots would recognize the cell-immunonanocomposites complexes. CTCs were labeled without utilizing the complicated and destructive procedure utilized in traditional CTCs identification. Up to 90.8% capture efficiency was successfully achieved. Clinically, the specific fluorescence labeling of CTCs was accomplished in nine clinical breast cancer patients' samples.
FIG. 6.
Scheme showing modifications of the composite material and the procedures for capturing circulating tumor cells.119 Reproduced with permission from Cui et al., Talanta 202, 230 (2019). Copyright 2019 American Chemical Society.
B. Hybrid microfluidic chips
1. Tumor antigen-independent microfluidic CTC-iChip
Karabacak et al. presented a CTC-iChip for marker-free isolation of CTCs from blood samples.120 CTC-iChip is composed of two separate microfluidic devices.120 CTC-iChip1 designed an array of posts utilizing continuous deterministic lateral displacement (DLD) to remove nucleated cells and size-based separation of WBCs and tumor cells from whole blood. CTC-iChip2 used inertial focusing to precisely position these cells in a micro-channel and then immunomagnetic isolated CTCs from “magnetic” WBCs coated with magnetic beads. This system could sort up to 107 cells/s. Through using two-stage magnetophoresis and depletion antibodies against leukocytes, performance could be achieved of 3.8-log depletion of white blood cells and a 97% yield of rare cells with a sample processing rate of 8 ml of whole blood/h. From the design, we could see that this CTC-iChip structure is a little complex. For DLD separation to isolate CTCs and WBCs for whole blood, CTCs could be lost and there is still RBCs contamination since the number of RBCs is enormous. For negative magnetic isolation, the conjugation of WBCs and magnetic beads is not complete although both CD45 and CD66b are used as leukocyte markers.
2. A hybrid magnetic/size-sorting (HMSS) chip for negative enrichment and size-sorting CTCs
This technology is a combination of negative magnetic depletion of leucocytes and the size-selective capture of CTCs (Fig. 7). Chung et al. proposed a hybrid magnetic/size-sorting chip. The device named HMSS chip is composed of two parts, a self-assembled magnet filter and a cell-size sorter.121 The magnetic filter is formed from a layer of magnetic grains (125 μm, NdFeB) self-assembling into an array of anti-aligned magnetic dipoles. A strong magnetic field is produced on the array surface. On top of the device, there is a herringbone structure. The size-based cell sorter is composed of weir-style physical barrier structure with an underpass-gap allowing the passing of small particles such as RBCs. The width of the capture sites is 5 μm. CTCs are trapped at the specific capture sites rendering in situ analysis of single cells. Magnetic nanoparticles were attached to leucocytes and mixed at 220 μl/ml cells for 10 min. The flow rate is 3 ml/h. The average recovery rate was 87% for 50 cells and increased to 96% for 250 cells. The technology is a negative depletion of WBCs using incubating WBCs with magnetic nanoparticles. Then, they utilized magnetic “attraction” to get rid of leucocytes and a following size-based sorter to capture tumor cells. Red blood cell lysis (RBL) was used to preprocess the blood. If clinical patient blood was used, the processing procedure is still complicated. CTCs have possibilities to be missed during this procedure, loss in red blood cell lysis (RBL), mixed with WBCs, and remained in the first part of the chip, and pass through the 5 μm gap.
FIG. 7.
Design of a hybrid magnetic/size-sorting chip.121 Reproduced with permission from Chung et al., Biomicrofluidics 7, 054107 (2013). Copyright 2013 AIP Publishing LLC. (a) A herringbone pattern was integrated on main parallel channels. (b) Self-assembled magnetic grains to generate a strong magnetic field. (c) HMSS sorter. (d) Size-based capture structure. (e) A micrograph of a HMSS sorter.
3. A two-stage microfluidic chip for negative enrichment
In 2014, Hyun et al. presented a two-stage microfluidic chip for negative enrichment, eluting non-target cells initially (Fig. 8).122 The first stage included a microfluidic magnetic-activated cell sorting (μ-MACS) chip to eliminate WBCs. Incubate WBCs with magnetic nanoparticles and let the cell suspension pass through a strong magnetic field to remove magnetic WBCs. Then, CTCs enter the second stage to be segregated. The second stage adopted a geometrically activated surface interaction (GASI) chip for selectively isolating CTCs. Modify the microfluidic chip with antibody, anti-EpCAM. CTCs with antigen, EpCAM positive CTCs would bind with antibody modified on the microchannel and be captured. EpCAM negative CTCs without bonding could be collected from the outlet. Deleting WBCs after RBL, then using sorting is a wise technique maximum reducing contamination. However, preprocessing seemed complex such as coating WBCs with magnetic nanoparticles. If not accomplished completely, it would still cause low isolating purity. Utilizing magnets to get rid of WBCs is challenging to perform thoroughly. For the second stage, relying on anti-EpCAM to isolate EpCAM positive CTCs, the capture efficiency is not high enough to distinguish two kinds of CTCs still with WBCs disturbance.
FIG. 8.
μ-MACS + GASI.122 Reproduced with permission from Hyun et al., Biosens. Bioelectron. 67, 86 (2015). Copyright 2015 American Chemical Society. (a) A two-stage enrichment chip for selective isolation of CTCs. μ-MACS + GASI. (b) μ-MACS was used for negative enrichment to get rid of WBCs. (c) GASI was used for positive enrichment to selectively separate cells based on their surface protein expression. (d) In GASI, anti-EpCAM was coated to microchannel to capture EpCAM positive tumor cells and EpCAM negative tumor cells were flowed away and collected.
4. An integrated ferrohydrodynamic cell separation (iFCS) system
Zhao et al. proposed an iFCS system (Fig. 9).123 The enrichment process is composed of two stages integrated on a single iFCS device. The procedure is cell-size variation inclusive and tumor antigen-independent enrichment of viable CTCs. At the same time, WBCs could be depleted. WBCs are bonded with magnetic microbeads through leukocyte surface biomarkers. Therefore, the overall magnetization of the WBC-bead conjugates was more significant than surrounding ferrofluids. The magnetization of the unlabeled CTCs was less than ferrofluids. In the first stage, a magnetic field gradient was generated. Unlabeled and sheath-focused CTCs were pushed to maintain at the upper boundary of a microchannel. However, unbound magnetic beads and WBCs conjugated with ≥3 microbeads were attracted to be depleted toward a waste outlet. In this stage, a high percentage of WBCs and beads are removed to mitigate bead aggregation. In the second stage, a symmetric magnetic field was produced with the highest value in the middle. The “magnetic” WBCs were still attracted at the center of the channel. Unlabeled CTCs were moving along upper and lower boundaries to be collected. Each magnet had a remnant magnetization of 1.48 T. The average recovery rate across eight cancer cell lines was 99.08%. Cell viability of HCC 1806 BrC cells before and after enrichment was determined to be 98.30 ± 0.56% and 97.69 ± .56% for low CTC occurrence rate (1–10 cells ml−1). A blood processing throughput is 12 ml/h. For clinical tests, sizes of CTCs and WBCs were measured for the effective diameter (maximum ferret diameter of cells from their bright-field images) for three breast cancer patients' samples. Analysis of surface antigens expression of individual CTCs was carried out to reveal a high heterogeneity of epithelial and mesenchymal characteristics in these cells. All the design, basic ideas, and experimental results are excellent. However, the whole procedure is relatively complex. RBL is the first one. Magnetic depletion for two stages depends on whether WBCs could conjugate with magnetic beads. Since the amount of WBCs is massive all WBCs would not definitely bond with magnetic nanoparticles.
FIG. 9.
Overview of an integrated ferrohydrodynamic cell separation (iFCS) system and its working principle.123 Reproduced with permission from Zhao et al., Lab Chip 19, 1860 (2019). Copyright 2019 American Chemical Society. (a) Top: schematic of an unlabeled CTC and its moving. Bottom: schematic of a magnetic bead labeled white blood cell (WBC) and its movement. (b) Two enrichment stages for CTCs and magnetic depletion of WBCs. (c) Top-view of the iFCS microchannel. (d) A photo of prototype microchannel (left) and assembled iFCS device with four permanent magnets in quadrupole configuration inside a holder (right).
The immunomagnetic separation of CTCs of all kinds of microfluidic chips has been summarized in Table I.
TABLE I.
Summary of magnetic-based method for CTCs isolation (MNP, magnetic nanoparticles, CE, capture efficiency).
| Structure | Antibody/Aptamer | Cell line (sample) | CE | Viability | Throughput | Reference |
|---|---|---|---|---|---|---|
| An intravascular magnetic wire97 | Biotinylated anti-human CD326 (anti-EpCAM) antibody Clone 9C4 | H1650 NSCLC cells, Human PCS-201 fibroblasts cells | 49% | … | 5 cm−1 | 97 |
| DNA mimic of double-sided tape (DST)102 | A long single-stranded DNA containing multiple copies of aptamers | T47D, MDA-MB-231 | 85% | … | … | 102 |
| Microfluidic chip (PDMS), NPs112 | Anti-EpCAM | COLO-205 in blood | 90% | 10 ml/h | 112 | |
| Self-assembled magnetic bead patterns in a microfluidic chip114 | 5D10 mAb or biotinylated FN | MCF-7, human leukemia cell line Jurkat | … | 0.18–0.36 ml/h | 114 | |
| Ephesia115 | Anti-CD19 mAb | Jurkat cell line and Raji cell line | … | 115 | ||
| ZnS:Mn2+ quantum dots and magnetic nanocomposites119 | Anti-EpCAM | SW480, MCF-7 | 90.8 | … | … | 119 |
| μ-MACS + GASI, CD45 antibody-conjugated MNP122 | CD45 depletion cocktail, Anti-EpCAM | MCF-7,MDA-MB-231,SK-BR-3 | 98.81% | … | 2.4, 6 ml/h | 122 |
| A semi-integrated electrical biosensor123 | Anti-EpCAM | MCF-7 in blood | >70% | … | 1.2 ml/h | 123 |
| A combined microfluidic micromagnetic device124 | Anti-EpCAM | M6C, 4T1 in blood | 90% | >90% | 1.2 ml/h | 113 |
| Immunomagnetic gold hybrid nanoparticles124 | Anti-EpCAM | MCF-7, leukocytes, Jurkat T cells | 79% | … | … | 124 |
| A chip-based DMR system, Mn-MNP125 | Anti-HER2/neu, anti-EGFR, anti-EpCAM | HCT116, SkBr3, MDA-MB-231 in leukocytes | … | … | … | 125 |
| Miniaturized diagnostic magnetic resonance (DMR), MNP126 | Her2/neu, EGFR | 3T3, SK-BR-3, MDA-MB-231 | … | … | … | 126 |
| A micro-nuclear magnetic resonance system127 | EpCAM,MUC-1, Her2, EGFR, B7-H3, CK18, Ki-67, p53 | 50 patients | … | … | … | 127 |
| An electrical biosensor128 | Anti-EpCAM | MCF-7 | >88% | … | 9 ml/h | 128 |
| Magnetic sifters, MNP129 | Anti-EpCAM | H-1650 (NSCLC) | 95.7% | … | 10 ml/h | 129 |
| A magnetic capture column and a size-selective filter130 | GCIY-EGFP, RPMI1788 | 97.7%, 97.5% | 97.6% | 30–1200 ml/h | 130 | |
| Magnetic capture (Apt-MBs)131 | Aptamer conjugated magnetic beads | DLD-1, CCRF-CEM | 73%, 55% | … | … | 131 |
The summary of commercially available systems is listed in Table II.
TABLE II.
Summary of commercially available systems.
| CTC commercial system | Principle | Reference |
|---|---|---|
| CellSearch | The CellSearch system (Veridex, Raritan, NJ, USA) is the only US FDA-approved CTC enumeration system.132 Utilizing ferrofluid nanoparticles with antibodies that target epithelial cell adhesion, CTCs are magnetically separated from the bulk of other cells in the blood. This system could successfully enumerate breast, prostate, and colorectal cancers through immunomagnetic technology. Disadvantages are high cost and low efficiency. | 132 |
| IsoFlux | The IsoFlux System (Fluxion Biosciences Inc., South San Francisco, CA) could provide high-sensitivity rare cell isolation coupled with a novel cell retrieval mechanism.133 The system employs immunomagnetic beads that facilitate use of single or multiple capture antibodies to target cells of a specific pathology. With an externally applied magnetic field the sample flow over a microfluidic device that contains an isolation zone to capture CTCs on the upper surface of the cartridge. The roof of the microfluidic channel could detach from the rest of the cartridge and transfers off-chip with the CTCs remained on its surface, providing near perfect transfer efficiency. It is possible for the IsoFlex System to molecular characterize intact viable CTCs or isolated DNA, RNA, or protein. | 133 |
| ImageStream | The ImageStream (Amnis, Seattle, WA, USA) is an imaging cytometry device that combines the strengths of flow cytometry and fluorescent microscopy in a single platform and has potential in applications to CTC counting.134 ImageStream platform for CTC enumeration potentially has a value for the early diagnosis of disseminated disease. Disadvantage is precision needed for the enumeration of low number of CTC. | 134 |
| Clearbridge BioMedics' ClearCell FX System | The ClearCell FX1 System works as a leading, automated, label-free liquid biopsy device for the enrichment of target cells from blood.79 The system employs the innovative microfluidic CTChip to isolate and retrieve intact, viable target cells from a patient's blood sample. This is a spiral microfluidic chip that could size-based separate and retrieve CTCs from blood with high throughput using the Dean drag force coupled with inertial microfluidics phenomenon. | 79 |
| Celsee Diagnostics CTC system | Key component of this system is a microfluidic chip with individual compartment in the shape of a bowl as the cross section and with a 7.5 μm width tunnel located below as filter.135 Blood sample was injected from top and enriched in the bowl and hematological cells and waste flow away from the bottom. It is based on size and deformability. The Celsee PREP 400™ system (Celsee Diagnostics, Plymouth, MI) is an automated system of this microfluidic device and contains a parallel network of four microfluidic chips each of which has approximately 56 320 capture chambers. The downstream of identification of individual CTCs can use a wide array of antibodies and DNA/RNA-based probes. | 135 |
| Parsortix cell separation system from ANGLE plc Company | Parsortix™ Cell Separation System (ANGLE North America, Inc., King of Prussia, PA) is a microfluidic based technology capturing CTCs based on size and deformability.136 It has reproducibly high capture efficiency and obtains highly enriched, viable (viability dependent on preservative used) CTCs that are amenable to a multitude of downstream analysis even for single cells. There exists a Parsortix GEN3 Cell Separation Cassette showing how the blood flows into the cassette, over the step structures and through the critical gap. The separation cassette is with a 6.5 μm critical gap size. | 136 |
IV. DISCUSSION
The immunomagnetic separation described above adopted a complicated approach of conjugating magnetic nanoparticles. Some of them are through bonding WBCs with magnetic beads and then utilizing magnetic “attraction” to get rid of WBCs. Modifying the microchannel with antibodies such as anti-EpCAM to capture EpCAM expressed CTCs and separately collected mesenchymal-expressed CTCs. Then only mesenchymal-expressed CTCs are left. Therefore, we collect them to achieve separation of two kinds of CTCs. Positively, magnetic beads connected with CTCs. In order to increase contact chances, CTCs conjugate with QD first to increase roughness and then connect with magnetic beads. In order to increase the contact area, traditional point-to-point contact has been transferred to “nanonet.” From the description above, it could be seen complex processing procedure has been taken. QD, GOQD, ZnS:Mn2+ quantum dots, SERS mark, and magnetic beads modifying with kinds of antibodies are to increase affinity contact. Our aim to perform rich CTCs experiments is to carry out clinical assays and come out following treatment. High-cost antibody and complex processing processes hinder clinical tests, especially without reproducibility. Due to the lack of clinical trials to validate the microfluidic chips, the significance of the design and experiments is questionable. Clinical validation requires the success of each patient blood test, which puts a high requirement of the reproducibility of microfluidic chips. However, reproducibility is almost neglected in those assays and most CTC chips. With advanced progress of CTC chips, other easy-operate and fast approach has been introduced into this area such as laser detection. Utilizing laser irradiating on the blood samples or processed ones, the characteristic spectrum would tell everything. The location of characteristic peaks can be used to identify cancer type, and the intensity can be determined by the number of CTCs. Magnetic separation combining with light technology would bring “revolutionary” CTCs’ isolation. Utilizing “magnetic beads,” we would further purify the blood sample to go a step further to serve light identification better. Laser identifying needs more number of CTCs with satisfactory purity. With magnetic attraction and other technology to get rid of most hematological cells such as RBCs and WBCs, light identifying that even could be accomplished within several seconds could be realized and come earlier.
V. CONCLUSIONS
Immunomagnetic separation of CTCs is very sensitive, specific, and not limited to the flow rate with no need for modification of surfaces of the microchannel. The release is simple to be realized by removing the magnet. The only FDA-approved product of CellSearch is based on this approach. Nevertheless, target cells have to bind with affinity ligand-functionalized micro-nanoparticles. The bonding efficiency determines the capture effectiveness. A large number of hematological cells would interfere with CTCs bonded with immunomagnetic particles, causing ineffectiveness. To promote CTC-beads’ binding, red blood cell lysis is usually taken. Cell viability is also affected to some extent. Same as the affinity-based method, it would depend on the expression of certain surface markers. Therefore, it would be better to adopt a size and magnetic combination.
ACKNOWLEDGMENTS
This research work was supported by the Anhui Natural Science Foundation of China (No. 1908085MF197), the National Natural Science Foundation of China (NNSFC) (No. 21904003), the Natural Science Foundation of Anhui Province (No. 1908085QB66), and the Postdoctoral Research Funding (No. 2014M550794).
Contributor Information
Hongmei Chen, Email: .
Shuangshou Wang, Email: .
DATA AVAILABILITY
The data that support the findings of this study are available from the corresponding author upon reasonable request.
REFERENCES
- 1.Williams S. C., “Circulating tumor cells,” Proc. Natl. Acad. Sci. U.S.A. 110, 4861 (2013). 10.1073/pnas.1304186110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ashworth T. R., “A case of cancer in which cells similar to those in the tumours were seen in the blood after death,” Aust. Med. J. 14, 146–147 (1969). [Google Scholar]
- 3.Plaks V. et al. , “Circulating tumor cells,” Science 341, 1186–1188 (2013). 10.1126/science.1235226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Conteduca V. et al. , “Circulating tumor cells: Utopia or reality?,” Future Oncol. 9, 1337 (2013). 10.2217/fon.13.101 [DOI] [PubMed] [Google Scholar]
- 5.Pantel K. et al. , “Clinical relevance and specific biological properties of disseminating tumour cells,” Nat. Rev. Cancer 8, 329 (2008). 10.1038/nrc2375 [DOI] [PubMed] [Google Scholar]
- 6.Plaks V. et al. , “Towards the biological understanding of CTC: Capture technologies, definitions and potential to create metastasis,” Cancers 5, 1619 (2013). 10.3390/cancers5041619 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Joyce J. A. et al. , “Microenvironmental regulation of metastasis,” Nat. Rev. Cancer 9(4), 239–252 (2009). 10.1038/nrc2618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mehlen P. and Puisieux A., “Metastasis: A question of life or death,” Nat. Rev. Cancer 6, 449–458 (2006). 10.1038/nrc1886 [DOI] [PubMed] [Google Scholar]
- 9.O’Flaherty J. D. et al. , “Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer,” Lung Cancer 76, 19 (2012). 10.1016/j.lungcan.2011.10.018 [DOI] [PubMed] [Google Scholar]
- 10.Qu L. et al. , “Dual-aptamer modification generates a unique interface for highly sensitive and specific electrochemical detection of tumor cells,” ACS Appl. Mater. Interfaces 6, 7309–7315 (2014). 10.1021/am5006783 [DOI] [PubMed] [Google Scholar]
- 11.Zhang Y. et al. , “Circulating tumor cells in hepatocellular carcinoma: Detection techniques, clinical implications, and future perspectives,” Semin. Oncol. 39, 449–460 (2012). 10.1053/j.seminoncol.2012.05.012 [DOI] [PubMed] [Google Scholar]
- 12.Wu L. J. et al. , “Capturing circulating tumor cells of hepatocellular carcinoma,” Cancer Lett. 326, 17–22 (2012). 10.1016/j.canlet.2012.07.024 [DOI] [PubMed] [Google Scholar]
- 13.Xu W. et al. , “Isolation of circulating tumor cells in patients with hepatocellular carcinoma using a novel cell separation strategy,” Clin. Cancer Res. 17, 3783–3793 (2011). 10.1158/1078-0432.CCR-10-0498 [DOI] [PubMed] [Google Scholar]
- 14.Nellore B. P. V. et al. , “Aptamer-conjugated graphene oxide membranes for highly efficient capture and accurate identification of multiple types of circulating tumor cells,” Bioconjug. Chem. 26, 235–242 (2015). 10.1021/bc500503e [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pantel K. and Alix-Panabieres C., “Circulating tumour cells in cancer patient: Challenges and perspectives,” Trends Mol. Med. 16, 398 (2010). 10.1016/j.molmed.2010.07.001 [DOI] [PubMed] [Google Scholar]
- 16.Fehm T. et al. , “Patterns of aneusomy for three chromosomes in individual cells from breast cancer tumors,” Breast Cancer Res. Treat 75(3), 227 (2002). 10.1023/A:1019901010758 [DOI] [PubMed] [Google Scholar]
- 17.Cristofanilli M. et al. , “Circulating tumor cells, disease progression, and survival in metastatic breast cancer,” N. Engl. J. Med. 351(8), 781–791 (2004). 10.1056/NEJMoa040766 [DOI] [PubMed] [Google Scholar]
- 18.Fan T. et al. , “Clinical significance of circulating tumor cells detected by an invasion assay in peripheral blood of patients with ovarian cancer,” Gynecol. Oncol. 112(1), 185–191 (2009). 10.1016/j.ygyno.2008.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.De Mattos-Arruda L. et al. , “Circulating tumour cells and cell-free DNA as tools for managing breast cancer,” Nat. Rev. Clin. Oncol. 10, 377–389 (2013). 10.1038/nrclinonc.2013.80 [DOI] [PubMed] [Google Scholar]
- 20.Alix-Panabières C., Schwarzenbach H., and Pantel K., “Circulating tumor cells and circulating tumor DNA,” Ann. Rev. Med. 63, 199–215 (2012). 10.1146/annurev-med-062310-094219 [DOI] [PubMed] [Google Scholar]
- 21.Pierga J.-Y. et al. , “Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial,” Clin. Cancer Res. 14, 7004 (2008). 10.1158/1078-0432.CCR-08-0030 [DOI] [PubMed] [Google Scholar]
- 22.Stott S. L. et al. , “Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer,” Sci. Transl. Med. 2, 25ra23 (2010). 10.1126/scitranslmed.3000403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Chen H. et al. , “Highly-sensitive capture of circulating tumor cells using micro-ellipse filters,” Sci. Rep. 7, 610 (2017). 10.1038/s41598-017-18475-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Krebs M. G. et al. , “Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer,” J. Clin. Oncol. 29(12), 1556–1563 (2011). 10.1200/JCO.2010.28.7045 [DOI] [PubMed] [Google Scholar]
- 25.Khan M. S. et al. , “Circulating tumor cells as prognostic markers in neuroendocrine tumors,” J. Clin. Oncol. 31(3), 365–372 (2013). 10.1200/JCO.2012.44.2905 [DOI] [PubMed] [Google Scholar]
- 26.Khoja L. et al. , “Biomarker utility of circulating tumor cells in metastatic cutaneous melanoma,” J. Invest Dermatol. 133, 1582–1590 (2013). 10.1038/jid.2012.468 [DOI] [PubMed] [Google Scholar]
- 27.de Wit S. et al. , “The detection of EpCAM(+) and EpCAM(−) circulating tumor cells,” Sci. Rep. 5, 12270 (2015). 10.1038/srep12270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cohen S. J. et al. , “Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer,” Ann. Oncol. 20(7), 1223–1229 (2009). 10.1093/annonc/mdn786 [DOI] [PubMed] [Google Scholar]
- 29.de Bono J. S. et al. , “Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer,” Clin. Cancer Res. 14(19), 6302–6309 (2008). 10.1158/1078-0432.CCR-08-0872 [DOI] [PubMed] [Google Scholar]
- 30.Budd G. T. et al. , “Circulating tumor cells versus imaging—Predicting overall survival in metastatic breast cancer,” Clin. Cancer Res. 12(21), 6403–6409 (2006). 10.1158/1078-0432.CCR-05-1769 [DOI] [PubMed] [Google Scholar]
- 31.Wulfing P. et al. , “HER2-positive circulating tumor cells indicate poor clinical outcome in stage I to III breast cancer patients,” Clin. Cancer Res. 12(6), 1715–1720 (2006). 10.1158/1078-0432.CCR-05-2087 [DOI] [PubMed] [Google Scholar]
- 32.Danila D. C. et al. , “Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer,” Clin. Cancer Res. 13(23), 7053–7058 (2007). 10.1158/1078-0432.CCR-07-1506 [DOI] [PubMed] [Google Scholar]
- 33.Hyun K. A. and Jung H. I., “Advances and critical concerns with the microfluidic enrichments of circulating tumor cells,” Lab Chip 14(1), 45–56 (2014). 10.1039/C3LC50582K [DOI] [PubMed] [Google Scholar]
- 34.Sollier E. et al. , “Size-selective collection of circulating tumor cells using vortex technology,” Lab Chip 14, 63 (2014). 10.1039/C3LC50689D [DOI] [PubMed] [Google Scholar]
- 35.Pantel K., Alix-Panabieres C., and Riethdorf S., “Cancer micrometastases,” Nat. Rev. Clin. Oncol. 6(6), 339–351 (2009). 10.1038/nrclinonc.2009.44 [DOI] [PubMed] [Google Scholar]
- 36.Weissenstein U. et al. , “Detection of circulating tumor cells in blood of metastatic breast cancer patients using a combination of cytokeratin and EpCAM antibodies,” BMC Cancer 12, 206 (2012). 10.1186/1471-2407-12-206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yu M. et al. , “Circulating tumor cells: Approaches to isolation and characterization,” J. Cell Biol. 192(3), 373–382 (2011). 10.1083/jcb.201010021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Murlidhar V. et al. , “Radial flow microfluidic device for high-throughput affinity-based isolation of circulating tumor cells,” Small 10, 4895 (2014). 10.1002/smll.201400719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tan K. et al. , “Longitudinal monitoring reveals dynamic changes in circulating tumor cells and CTC-associated miRNAs in response to chemotherapy in metastatic colorectal cancer patients,” Cancer Lett. 423, 1 (2018). 10.1016/j.canlet.2018.02.039 [DOI] [PubMed] [Google Scholar]
- 40.Nagrath S. et al. , “Isolation of rare circulating tumor cells in cancer patients by microchip,” Nature 450, 1235 (2007). 10.1038/nature06385 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stott S. L. et al. , “Isolation of circulating tumor cells using a microvortex-generating herringbone-chip,” Proc. Natl. Acad. Sci. U.S.A. 107, 18392 (2010). 10.1073/pnas.1012539107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yoo J. et al. , “Sensitive capture of circulating tumor cells by functionalized grapheme oxide nanosheets,” Nat. Nanotechnol. 8, 735 (2013). 10.1038/nnano.2013.194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zheng F. et al. , “Aptamer-functionalized barcode particles for the capture and detection of multiple types of circulating tumor cells,” Adv. Mater. 26(43), 7333–7338 (2014). 10.1002/adma.201403530 [DOI] [PubMed] [Google Scholar]
- 44.Yoon H. J. et al. , “Tunable thermal-sensitive polymer-graphene oxide composite for efficient capture and release of viable circulating tumor cells,” Adv. Mater. 8(24), 4891–4897 (2016). 10.1002/adma.201600658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhao L. et al. , “Enhanced and differential capture of circulating tumor cells from lung cancer patients by microfluidic assays using aptamer cocktail,” Small 12(8), 1072–1081 (2016). 10.1002/smll.201503188 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Song Y. et al. , “Selection of DNA aptamers against epithelial cell adhesion molecule for cancer cell imaging and circulating tumor cell capture,” Anal. Chem. 85(8), 4141–4149 (2013). 10.1021/ac400366b [DOI] [PubMed] [Google Scholar]
- 47.Wan Y. et al. , “Nanotextured substrates with immobilized aptamers for cancer cell isolation and cytology,” Cancer 118, 1145 (2012). 10.1002/cncr.26349 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wan Y. et al. , “Capture, isolation and release of cancer cells with aptamer-functionalized glass bead array,” Lab Chip 12(22), 4693 (2012). 10.1039/c2lc21251j [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wan Y. et al. , “Surface-immobilized aptamers for cancer cell isolation and microscopic cytology,” Cancer Res. 70(22), 9371–9380 (2010). 10.1158/0008-5472.CAN-10-0568 [DOI] [PubMed] [Google Scholar]
- 50.Chen H., “Capturing and clinical applications of circulating tumor cells with wave microfluidic chip,” Appl. Biochem. Biotechnol. 190(4), 1470–1483 (2020). 10.1007/s12010-019-03199-4 [DOI] [PubMed] [Google Scholar]
- 51.Liu L. et al. , “Artificial antibody with site-enhanced multivalent aptamers for specific capture of circulating tumor cells,” Anal. Chem. 91(4), 2591–2594 (2019). 10.1021/acs.analchem.8b05259 [DOI] [PubMed] [Google Scholar]
- 52.Li W. M. et al. , “Selection of metastatic breast cancer cell-specific aptamers for the capture of CTCs with a metastatic phenotype by cell-SELEX,” Mol. Ther. Nucleic Acids 12, 707–717 (2018). 10.1016/j.omtn.2018.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chen K. et al. , “Tumor cell capture patterns around aptamer-immobilized microposts in microfluidic devices,” Biomicrofluidics 11(5), 054110 (2017). 10.1063/1.5000707 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chen H. et al. , “Combination of antibody-coated, physical-based microfluidic chip with wave-shaped arrays for isolating circulating tumor cells,” Biomed. Microdevices 19(3), 66 (2017). 10.1007/s10544-017-0202-3 [DOI] [PubMed] [Google Scholar]
- 55.Ishibashi R. et al. , “Detection of circulating colorectal cancer cells by a custom microfluid system before and after endoscopic metallic stent placement,” Oncol. Lett. 18(6), 6397–6404 (2019). 10.3892/ol.2019.11047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chen K. et al. , “Integration of lateral filter arrays with immunoaffinity for circulating-tumor-cell isolation,” Angew. Chem. Int. Ed. Engl. 58(23), 7606–7610 (2019). 10.1002/anie.201901412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zheng S. Y. et al. , “3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood,” Biomed. Microdevices 13, 203–213 (2011). 10.1007/s10544-010-9485-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Tan S. J. et al. , “Microdevice for the isolation and enumeration of cancer cells from blood,” Biomed. Microdevices 11, 883 (2009). 10.1007/s10544-009-9305-9 [DOI] [PubMed] [Google Scholar]
- 59.Lim L. S. et al. , “Microsieve lab-chip device for rapid enumeration and fluorescence in situ hybridization of circulating tumor cells,” Lab Chip 12(21), 4388–4396 (2012). 10.1039/c2lc20750h [DOI] [PubMed] [Google Scholar]
- 60.McFaul S. F., Lin B. K., and Ma H. S., “Cell separation based on size and deformability using microfluidic funnel ratchets,” Lab Chip 12, 2369 (2012). 10.1039/c2lc21045b [DOI] [PubMed] [Google Scholar]
- 61.Sarioglu A. F. et al. , “A microfluidic device for label-free, physical capture of circulating tumor cell clusters,” Nat. Methods 12, 685 (2015). 10.1038/nmeth.3404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xu T., Lu B., Tai Y. C., and Goldkorn A., “A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter,” Cancer Res. 70(16), 6420–6426 (2010). 10.1158/0008-5472.CAN-10-0686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lin H. K. et al. , “Portable filter-based microdevice for detection and characterization of circulating tumor cells,” Clin. Cancer Res. 16, 5011 (2010). 10.1158/1078-0432.CCR-10-1105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Preira P. et al. , “Passive circulating cell sorting by deformability using a microfluidic gradual filter,” Lab Chip 13(1), 161–170 (2013). 10.1039/C2LC40847C [DOI] [PubMed] [Google Scholar]
- 65.Lee S. W. et al. , “Continuous enrichment of circulating tumor cells using a microfluidic lateral flow filtration chip,” J. Chromatogr. A 1377, 100–105 (2015). 10.1016/j.chroma.2014.12.037 [DOI] [PubMed] [Google Scholar]
- 66.Magbanua J. et al. , “A novel strategy for detection and enumeration of circulating rare cell populations in metastatic cancer patients using automated microfluidic filtration and multiplex immunoassay,” Plos One 10, e0141166 (2015). 10.1371/journal.pone.0141166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Aya-Bonilla C. A. et al. , “Isolation and detection of circulating tumour cells from metastatic melanoma patients using a slanted spiral microfluidic device,” Oncotarget 8(40), 67355 (2017). 10.18632/oncotarget.18641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhou J. et al. , “Label-free separation and culture of tumor cells in a microfluidic biochip,” Analyst 145, 1706–1715 (2020). 10.1039/C9AN02092F [DOI] [PubMed] [Google Scholar]
- 69.Zhou J. et al. , “Isolation of circulating tumor cells in non-small-cell-lung-cancer patients using a multi-flow microfluidic channel,” Microsyst. Nanoeng. 5(1), 8 (2019). 10.1038/s41378-019-0045-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kulasinghe A. et al. , “Capture of circulating tumour cell clusters using straight microfluidic chips,” Cancers 11(1), 89 (2019). 10.3390/cancers11010089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhou J. et al. , “Isolation of cells from whole blood using shear-induced diffusion,” Sci. Rep. 8(1), 9411 (2018). 10.1038/s41598-018-27779-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Warkiani M. E. et al. , “Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics,” Nat. Protoc. 11(1), 134–148 (2016). 10.1038/nprot.2016.003 [DOI] [PubMed] [Google Scholar]
- 73.Zhang Z. et al. , “The effects of 3D channel geometry on CTC passing pressure-towards deformability-based cancer cell separation,” Lab on a Chip 14(14), 2576–2584 (2014). 10.1039/C4LC00301B [DOI] [PubMed] [Google Scholar]
- 74.Sun N. et al. , “High-purity capture of CTCs based on micro-beads enhanced isolation by size of epithelial tumor cells (ISET) method,” Biosens. Bioelectron. 102, 157–163 (2018). 10.1016/j.bios.2017.11.026 [DOI] [PubMed] [Google Scholar]
- 75.Kim S. et al. , “Circulating tumor cell microseparator based on lateral magnetophoresis and immunomagnetic nanobeads,” Anal. Chem. 85(5), 2779–2786 (2013). 10.1021/ac303284u [DOI] [PubMed] [Google Scholar]
- 76.Miltenyi S., Müller W., Weichel W., and Radbruch A., “High gradient magnetic cell separation with MACS,” Cytometry 11, 231–238 (1990). 10.1002/cyto.990110203 [DOI] [PubMed] [Google Scholar]
- 77.Wu X. et al. , “Magnetic cell centrifuge platform performance study with different microsieve pore geometries,” Sensors 20(1), E48 (2019). 10.3390/s20010048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Xiao Y. et al. , “Design of DNA aptamer-functionalized magnetic short nanofibers for efficient capture and release of circulating tumor cells,” Bioconjug. Chem. 31(1), 130–138 (2020). 10.1021/acs.bioconjchem.9b00816 [DOI] [PubMed] [Google Scholar]
- 79.Tang M. et al. , “Magnetic chip based extracorporeal circulation: A new tool for circulating tumor cell in vivo detection,” Anal. Chem. 91(23), 15260–15266 (2019). 10.1021/acs.analchem.9b04286 [DOI] [PubMed] [Google Scholar]
- 80.Wang Z. et al. , “High-efficiency isolation and rapid identification of heterogeneous circulating tumor cells (CTCs) using dual-antibody-modified fluorescent-magnetic nanoparticles,” ACS Appl. Mater. Interfaces 11(43), 39586–39593 (2019). 10.1021/acsami.9b14051 [DOI] [PubMed] [Google Scholar]
- 81.Labib M. et al. , “Tracking the expression of therapeutic protein targets in rare cells by antibody-mediated nanoparticle labelling and magnetic sorting,” Nat. Biomd. Eng. (2020). 10.1038/s41551-020-0590-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dou B. et al. , “Aptamer-functionalized and gold nanoparticle array-decorated magnetic grapheme nanosheets enable multiplexed and sensitive electrochemical detection of rare circulating tumor cells in whole blood,” Anal. Chem. 91(16), 10792–10799 (2019). 10.1021/acs.analchem.9b02403 [DOI] [PubMed] [Google Scholar]
- 83.Emrich C. A. et al. , “Microfabricated two-dimensional electrophoresis device for differential protein expression profiling,” Anal. Chem. 79, 7360–7360 (2007). 10.1021/ac0711485 [DOI] [PubMed] [Google Scholar]
- 84.Aghaamoo M. et al. , “On the design of deterministic dielectrophoresis for continuous separation of circulating tumor cells from peripheral blood cells,” Electrophoresis 40(10), 1486–1493 (2019). 10.1002/elps.201800459 [DOI] [PubMed] [Google Scholar]
- 85.lliescu F. S. et al. , “Highlighting the uniqueness in dielectrophoretic enrichment of circulating tumor cells,” Electrophoresis 40(10), 1457–1477 (2019). 10.1002/elps.201800446 [DOI] [PubMed] [Google Scholar]
- 86.Nguyen N. V. and Jen C. P., “Impedance detection integrated with dielectrophoresis enrichment platform for lung circulating tumor cells in a microfluidic channel,” Biosens. Bioelectron. 121, 10–18 (2018). 10.1016/j.bios.2018.08.059 [DOI] [PubMed] [Google Scholar]
- 87.Li P. et al. , “Acoustic separation of circulating tumor cells,” Proc. Natl. Acad. Sci. U.S.A. 112, 4970–4975 (2015). 10.1073/pnas.1504484112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Alshareef M. et al. , “Separation of tumor cells with dielectrophoresis-based microfluidic chip,” Biomicrofluidics 7, 011803 (2013). 10.1063/1.4774312 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Geislinger T. M. et al. , “Hydrodynamic and label-free sorting of circulating tumor cells from whole blood,” Appl. Phys. Lett. 107, 203702 (2015). 10.1063/1.4935563 [DOI] [Google Scholar]
- 90.Li X. et al. , “Continuous-flow microfluidic blood cell sorting for unprocessed whole blood using surface-micromachined microfiltration membranes,” Lab Chip 14, 2565–2575 (2014). 10.1039/C4LC00350K [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Liu Z. et al. , “Rapid isolation of cancer cells using microfluidic deterministic lateral displacement structure,” Biomicrofluidic 7, 011801 (2013). 10.1063/1.4774308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Okano H. et al. , “Enrichment of circulating tumor cells in tumour-bearing mouse blood by a deterministic lateral displacement microfluidic device,” Biomed. Microdevices 17, 59 (2015). 10.1007/s10544-015-9964-7 [DOI] [PubMed] [Google Scholar]
- 93.Haddadi H. et al. , “Separation of cancer cells using vertical microfluidic flows,” Biomicrofluidics 12, 014112 (2018). 10.1063/1.5009037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Zhang J. et al. , “Fundamentals and applications of inertial microfluidics: A review,” Lab Chip 16, 10–34 (2016). 10.1039/C5LC01159K [DOI] [PubMed] [Google Scholar]
- 95.Wang J. et al. , “Label-free isolation and mRNA detection of circulating tumor cells from patients with metastatic lung cancer for disease diagnosis and monitoring therapeutic efficacy,” Anal. Chem. 87, 11893–11900 (2015). 10.1021/acs.analchem.5b03484 [DOI] [PubMed] [Google Scholar]
- 96.Tian F. et al. , “Label-free isolation of rare tumor cells from untreated whole blood by interfacial viscoelastic microfluidics,” Lab Chip 18, 3436–3445 (2018). 10.1039/C8LC00700D [DOI] [PubMed] [Google Scholar]
- 97.Dong Y. et al. , “Microfluidics and circulating tumor cells,” J. Mol. Diagn. 15, 149 (2013). 10.1016/j.jmoldx.2012.09.004 [DOI] [PubMed] [Google Scholar]
- 98.Vermesh O. et al. , “An intravascular magnetic wire for the high-throughput retrieval of circulating tumour cells in vivo,” Nat. Biomed. Eng. 2(9), 696–705 (2018). 10.1038/s41551-018-0257-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Cui F. et al. , “A novel magnetic fluorescent biosensor based on graphene quantum dots for rapid, efficient, and sensitive separation and detection of circulating tumor cells,” Anal. Bioanal. Chem. 411(5), 985–995 (2019). 10.1007/s00216-018-1501-0 [DOI] [PubMed] [Google Scholar]
- 100.Liu C. et al. , “Capture and separation of circulating tumor cells using functionalized magnetic nanocomposites with simultaneous in situ chemotherapy,” Nanotechnology 30(28), 285706 (2019). 10.1088/1361-6528/ab0e25 [DOI] [PubMed] [Google Scholar]
- 101.Esmaeilsabzali H. et al. , “Development of a microfluidic platform for size-based hydrodynamic enrichment and PSMA-targeted immunomagnetic isolation of circulating tumour cells in prostate cancer,” Biomicrofluidics 13(1), 014110 (2019). 10.1063/1.5064473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chen Y. et al. , “Non-invasive isolation of rare circulating tumor cells with a DNA mimic of double-sided tape using multimeric aptamers,” Nanoscale 11(13), 5879–5883 (2019). 10.1039/C9NR00364A [DOI] [PubMed] [Google Scholar]
- 103.Xie W. et al. , “Capture and “self-release” of circulating tumor cells using metal-organic framework materials,” Nanoscale 11(17), 8293–8303 (2019). 10.1039/C8NR09071H [DOI] [PubMed] [Google Scholar]
- 104.Wang K. et al. , “A microfluidic platform for high-purity separating circulating tumor cells at the single-cell level,” Talanta 200, 169–176 (2019). 10.1016/j.talanta.2019.03.035 [DOI] [PubMed] [Google Scholar]
- 105.Mishra A. et al. , “Ultrahigh-throughput magnetic sorting of large blood volumes for epitope-agnostic isolation of circulating tumor cells,” Pro. Natl. Acad. Sci. USA 117(29), 16839–16847 (2020). 10.1039/C9LC00210C [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chen H. et al. , “Hybrid magnetic and deformability based isolation of circulating tumor cells using microfluidics,” AIP Adv. 9, 025023 (2019). 10.1063/1.5081849 [DOI] [Google Scholar]
- 107.Zhang L. et al. , “Rapid prototyping of nanoroughened polydimethylsiloxane surfaces for the enhancement of immunomagnetic isolation and recovery of rare tumor cells,” Biomed. Microdevices 21(3), 58 (2019). 10.1007/s10544-019-0418-5 [DOI] [PubMed] [Google Scholar]
- 108.Green B. J. et al. , “Phenotypic profilin ranking,” Anal. Chem. 91(15), 9348–9355 (2019). 10.1021/acs.analchem.9b01697 [DOI] [PubMed] [Google Scholar]
- 109.Yang J. et al. , “Highly specific and sensitive point-of-care detection of rare circulating tumor cells in whole blood via a dual recognition strategy,” Biosens. Bioelectron. 143, 111604 (2019). 10.1016/j.bios.2019.111604 [DOI] [PubMed] [Google Scholar]
- 110.Shen C. et al. , “Electrochemical detection of circulating tumor cells based on DNA generated electrochemical current and rolling circle amplification,” Anal. Chem. 91(18), 11614–11619 (2019). 10.1021/acs.analchem.9b01897 [DOI] [PubMed] [Google Scholar]
- 111.Lai C. H. et al. , “A two-dimensional immunomagnetic nano-net for the efficient isolation of circulating tumor cells in whole blood,” Nanoscale 11(44), 21119–21127 (2019). 10.1039/C9NR06256D [DOI] [PubMed] [Google Scholar]
- 112.Hoshino K. et al. , “Microchip-based immunomagnetic detection of circulating tumor cell,” Lab Chip 11, 3449 (2011). 10.1039/c1lc20270g [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kang J. H. et al. , “A combined micromagnetic-microfluidic device for rapid capture and culture of rare circulating tumor cells,” Lab chip 12, 2175 (2012). 10.1039/c2lc40072c [DOI] [PubMed] [Google Scholar]
- 114.Sivagnanam V. et al. , “Selective breast cancer cell capture, culture, and immunocytochemical analysis using self-assembled magnetic bead patterns in a microfluidic chip,” Langmuir 26(9), 6091–6096 (2010). 10.1021/la9045572 [DOI] [PubMed] [Google Scholar]
- 115.Saliba A. et al. , “Microfluidic sorting and multimodal typing of cancer cells in self-assembled magnetic arrays,” Proc. Natl. Acad. Sci. U.S.A. 107, 14524 (2010). 10.1073/pnas.1001515107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Li D. et al. , “Selective capture and quick detection of targeting cells with SERS-coding microsphere suspension chip,” Small 11, 2200 (2015). 10.1002/smll.201402531 [DOI] [PubMed] [Google Scholar]
- 117.Shi Y. et al. , “Multifunctional biocompatible graphene oxide quantum dots decorated magnetic nanoplatform for efficient capture and Two-photon imaging of rare tumor cells,” ACS Appl. Mater Interfaces 7, 10935 (2015). 10.1021/acsami.5b02199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Min H. et al. , “Efficient capture and simple quantification of circulating tumor cells using quantum dots and magnetic beads,” Small 11(21), 2536–2542 (2015). 10.1002/smll.201403126 [DOI] [PubMed] [Google Scholar]
- 119.Cui H. et al. , “Rapid and efficient isolation and detection of circulating tumor cells based on ZnS:Mn2+ quantum dots and magnetic nanocomposite,” Talanta 202, 230–236 (2019). 10.1016/j.talanta.2019.05.001 [DOI] [PubMed] [Google Scholar]
- 120.Karabacak N. M. et al. , “Microfluidic, marker-free isolation of circulating tumor cells from blood samples,” Nat. Protocols 9(3), 694 (2014). 10.1038/nprot.2014.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Chung J. et al. , “Rare cell isolation and profiling on a hybrid magnetic/size-sorting chip,” Biomicrofluidics 7, 054107 (2013). 10.1063/1.4821923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hyun K. et al. , “Two-stage microfluidic chip for selective isolation of circulating tumor cells (CTCs),” Biosens. Bioelectron. 67, 86–92 (2015). 10.1016/j.bios.2014.07.019 [DOI] [PubMed] [Google Scholar]
- 123.Zhao W. et al. , “Tumor antigen-independent and cell size variation-inclusive enrichment of viable circulating tumor cells,” Lab Chip 19, 1860 (2019). 10.1039/C9LC00210C [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chang Z. et al. , “Biomimetric immunomagnetic gold hybrid nanoparticles coupled with inductively coupled plasma mass spectrometry for the detection of circulating tumor cells,” J. Mater. Chem. B 8(23), 5019–5025 (2020). 10.1039/D0TB00403K [DOI] [PubMed] [Google Scholar]
- 125.Lee H. et al. , “Rapid detection and profiling of cancer cells in fine-needle aspirates,” Proc. Natl. Acad. Sci. U.S.A. 106(30), 12459–12464 (2009). 10.1073/pnas.0902365106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lee H. et al. , “Chip-NMR biosensor for detection and molecular analysis of cells,” Nat. Med. 14(8), 869–874 (2008). 10.1038/nm.1711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Haun J. B. et al. , “Micro-NMR for rapid molecular analysis of human tumor samples,” Sci. Transl. Med. 3, 71ra16 (2011). 10.1126/scitranslmed.3002048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chung Y. K. et al. , “An electrical biosensor for the detection of circulating tumor cells,” Biosens. Bioelectron. 26(5), 2520–2252 (2011). 10.1016/j.bios.2010.10.048 [DOI] [PubMed] [Google Scholar]
- 129.Earhart C. M. et al. , “Isolation and mutational analysis of circulating tumor cells from lung cancer patients with magnetic sifters and biochips,” Lab Chip 14, 78 (2014). 10.1039/C3LC50580D [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yamamoto S. et al. , “Efficient capturing of circulating tumor cells using a magnetic capture column and a size-selective filter,” Bioprocess. Biosyst. Eng. 38, 1693 (2015). 10.1007/s00449-015-1412-9 [DOI] [PubMed] [Google Scholar]
- 131.Sun C. et al. , “A rapid and simple method for efficient capture and accurate discrimination of circulating tumor cells using aptamer conjugated magnetic beads and surface-enhanced Raman scattering imaging,” Anal. Bioanal. Chem. 407, 8883 (2015). 10.1007/s00216-015-9049-8 [DOI] [PubMed] [Google Scholar]
- 132.Wang L. et al. , “Nanostructured substrates for isolation of circulating tumor cells,” Nano Today 8(4), 374–387 (2013). 10.1016/j.nantod.2013.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Harb W. et al. , “Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay,” Transl. Oncol. 6(5), 528–538 (2013). 10.1593/tlo.13367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.López-Riquelme N. et al. , “Imaging cytometry for counting circulating tumor cells: Comparative analysis of the CellSearch vs ImageStream system,” Apmis 121(12), 1139–1143 (2013). 10.1111/apm.12061 [DOI] [PubMed] [Google Scholar]
- 135.Gogoi P. et al. , “Development of an automated and sensitive microfluidic device for capturing and characterizing circulating tumor cells (CTCs) from clinical blood samples,” Plos One 11(1), e0147400 (2016). 10.1371/journal.pone.0147400 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Miller M. C. et al. , “The parsortix™ cell separation system—A versatile liquid biopsy platform,” Cytom. Part A 93, 1234–1239 (2018). 10.1002/cyto.a.23571 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.









