51
|
Wu M, Huang Y, Zhou Y, Zhao H, Lan Y, Yu Z, Jia C, Cong H, Zhao J. The Discovery of Novel Circulating Cancer-Related Cells in Circulation Poses New Challenges to Microfluidic Devices for Enrichment and Detection. SMALL METHODS 2022; 6:e2200226. [PMID: 35595707 DOI: 10.1002/smtd.202200226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/19/2022] [Indexed: 06/15/2023]
Abstract
Circulating tumor cells (CTCs) enumeration has been widely used as a surrogate predictive marker for early diagnoses, the evaluation of chemotherapy efficacy, and cancer prognosis. Microfluidic technologies for CTCs enrichment and detection have been developed and commercialized as automation platforms. Currently, in addition to CTCs, some new types of circulating cancer-related cells (e.g., CCSCs, CTECs, CAMLs, and heterotypic CTC clusters) in circulation are also reported to be correlated to cancer diagnosis, metastasis, or prognosis. And they widely differ from the conventional CTCs in positive markers, cellular morphology, or size, which presents a new technological challenge to microfluidic devices that use affinity-based capture methods or size-based filtration methods for CTCs detection. This review focuses on the biological and physical properties as well as clinical significance of the novel circulating cancer-related cells, and discusses the challenges of their discovery to microfluidic chip for enrichment. Finally, the current challenges of CTCs detection in clinical application and future opportunities are also discussed.
Collapse
Affiliation(s)
- Man Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuhang Huang
- Shanghai Normal University, Shanghai, 200030, China
| | - Yang Zhou
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuwei Lan
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhibin Yu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunping Jia
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
52
|
Han J, Lu C, Shen M, Sun X, Mo X, Yang G. Fast, Reusable, Cell Uniformly Distributed Membrane Filtration Device for Separation of Circulating Tumor Cells. ACS OMEGA 2022; 7:20761-20767. [PMID: 35755342 PMCID: PMC9219081 DOI: 10.1021/acsomega.2c01153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
Isolation of circulating tumor cells (CTCs) is of great significance for the diagnosis, prognosis, and treatment of metastatic cancer. Among CTC capture methods independent of antibodies, membrane filtration-based methods have the advantages of simplicity, rapidity, and high throughput but usually have problems such as clogging, high pressure drop, and impaired cell viability. In this study, we designed and tested a reusable device that used horizontal rotor and fluid-assisted separation to capture CTCs by centrifugal membrane filtration, achieving simple, fast, highly efficient, and viable cell capture on traditional centrifuge. The average capture efficiency was 95.8% for different types of cancer cells with >90% survival, and the removal of white blood cells can reach 99.72% under four times cleaning of the membrane after filtration. A further clinic demo was performed using the device to detect residual leukemic cells in patients; the results showed a 10-fold enrichment of the leukemic cells in peripheral blood samples. Taken together, the simple, robust, and efficient CTC capture device may have the potential for clinic routine detection and analysis of circulating tumor cells.
Collapse
Affiliation(s)
- Jintao Han
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Chunyang Lu
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Mengzhu Shen
- Beijing
Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking
University People’s Hospital, Beijing 100044, China
| | - Xiaoyi Sun
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Xiaodong Mo
- Beijing
Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Peking
University People’s Hospital, Beijing 100044, China
| | - Gen Yang
- State
Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
- Wenzhou
Institute, University of Chinese Academy
of Sciences, Wenzhou 352001, China
| |
Collapse
|
53
|
Hu Y, Chen D, Napoleon JV, Srinivasarao M, Singhal S, Savran CA, Low PS. Efficient capture of circulating tumor cells with low molecular weight folate receptor-specific ligands. Sci Rep 2022; 12:8555. [PMID: 35595733 PMCID: PMC9122947 DOI: 10.1038/s41598-022-12118-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/29/2022] [Indexed: 11/09/2022] Open
Abstract
Retrieval of circulating tumor cells (CTC) has proven valuable for assessing a patient's cancer burden, evaluating response to therapy, and analyzing which drug might treat a cancer best. Although most isolation methods retrieve CTCs based on size, shape, or capture by tumor-specific antibodies, we explore here the use of small molecule tumor-specific ligands linked to magnetic beads for CTC capture. We have designed folic acid-biotin conjugates with different linkers for the capture of folate receptor (FR) + tumor cells spiked into whole blood, and application of the same technology to isolate FR + CTCs from the peripheral blood of both tumor-bearing mice and non-small cell lung patients. We demonstrate that folic acid linked via a rigid linker to a flexible PEG spacer that is in turn tethered to a magnetic bead enables optimal CTC retrieval, reaching nearly 100% capture when 100 cancer cells are spiked into 1 mL of aqueous buffer and ~ 90% capture when the same quantity of cells is diluted into whole blood. In a live animal model, the same methodology is shown to efficiently retrieve CTCs from tumor-bearing mice, yielding cancer cell counts that are proportional to total tumor burden. More importantly, the same method is shown to collect ~ 29 CTCs/8 mL peripheral blood from patients with non-small cell lung cancer. Since the ligand-presentation strategy optimized here should also prove useful in targeting other nanoparticles to other cells, the methods described below should have general applicability in the design of nanoparticles for cell-specific targeting.
Collapse
Affiliation(s)
- Yingwen Hu
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Danyang Chen
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - John V Napoleon
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Madduri Srinivasarao
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cagri A Savran
- School of Mechanical Engineering, Birck Nanotechnology Center, Purdue Center for Cancer Research, Purdue University, 1205 W. State St., West Lafayette, IN, 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
54
|
Chen B, Zheng J, Gao K, Hu X, Guo SS, Zhao XZ, Liao F, Yang Y, Liu W. Noninvasive Optical Isolation and Identification of Circulating Tumor Cells Engineered by Fluorescent Microspheres. ACS APPLIED BIO MATERIALS 2022; 5:2768-2776. [PMID: 35537085 DOI: 10.1021/acsabm.2c00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Circulating tumor cells (CTCs) are rare, meaning that current isolation strategies can hardly satisfy efficiency and cell biocompatibility requirements, which hinders clinical applications. In addition, the selected cells require immunofluorescence identification, which is a time-consuming and expensive process. Here, we developed a method to simultaneously separate and identify CTCs by the integration of optical force and fluorescent microspheres. Our method achieved high-purity separation of CTCs without damage through light manipulation and avoided additional immunofluorescence staining procedures, thus achieving rapid identification of sorted cells. White blood cells (WBCs) and CTCs are similar in size and density, which creates difficulties in distinguishing them optically. Therefore, fluorescent PS microspheres with high refractive index (RI) are designed here to capture the CTCs (PS-CTCs) and increase the average index of refraction of PS-CTCs. In optofluidic chips, PS-CTCs were propelled to the collection channel from the sample mixture, under the radiation of light force. Cells from the collection outlet were easily identified under a fluorescence microscope due to the fluorescence signals of PS microspheres. This method provides an approach for the sorting and identification of CTCs, which holds great potential for clinical applications in early diagnosis of disease.
Collapse
Affiliation(s)
- Bei Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Jingjing Zheng
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Kefan Gao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xuejia Hu
- Department of Electronic Engineering School of Electronic Science and Engineering, Xiamen University, Xiamen, Fujian Province 361005, China
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Fei Liao
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yi Yang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.,Wuhan Institute of Quantum Technology, Wuhan 430206, China.,Hubei Luojia Laboratory, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
55
|
Gioe E, Uddin MR, Kim JH, Chen X. Deterministic Lateral Displacement (DLD) Analysis Tool Utilizing Machine Learning towards High-Throughput Separation. MICROMACHINES 2022; 13:661. [PMID: 35630129 PMCID: PMC9145823 DOI: 10.3390/mi13050661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/01/2023]
Abstract
Deterministic lateral displacement (DLD) is a microfluidic method for the continuous separation of particles based on their size. There is growing interest in using DLD for harvesting circulating tumor cells from blood for further assays due to its low cost and robustness. While DLD is a powerful tool and development of high-throughput DLD separation devices holds great promise in cancer diagnostics and therapeutics, much of the experimental data analysis in DLD research still relies on error-prone and time-consuming manual processes. There is a strong need to automate data analysis in microfluidic devices to reduce human errors and the manual processing time. In this work, a reliable particle detection method is developed as the basis for the DLD separation analysis. Python and its available packages are used for machine vision techniques, along with existing identification methods and machine learning models. Three machine learning techniques are implemented and compared in the determination of the DLD separation mode. The program provides a significant reduction in video analysis time in DLD separation, achieving an overall particle detection accuracy of 97.86% with an average computation time of 25.274 s.
Collapse
|
56
|
Li M, Li D, Song Y, Li D. Tunable particle/cell separation across aqueous two-phase system interface by electric pulse in microfluidics. J Colloid Interface Sci 2022; 612:23-34. [PMID: 34974255 DOI: 10.1016/j.jcis.2021.12.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 01/16/2023]
Abstract
HYPOTHESIS Separations of particles and cells are indispensable in many microfluidic systems and have numerous applications in chemistry and biomedicine. The interface of aqueous two-phase system (ATPS) can act as a liquid filter. Under electric field stimuli, the selective transfer of targets across the liquid-liquid interface are expected for particles and cells separation. EXPERIMENTS The separations of particles and cells based on ATPS electrophoresis in a microfluidic chip were investigated. A systematical study of the mechanism of ATPS electrophoresis was performed first by employing polystyrene (PS) particles. Subsequently, the separations of particles and microalgae cells were demonstrated. FINDINGS The electrophoretic transfer of particles across the interface of ATPS is determined by multi-parameters, including the strength of electric pulse, particle size, zeta potential, and hydrophobicity of the particle. The continuous separations of particles/cells can be achieved through the controllable transfer of target particles/cells across the interface under electric pulses in a microfluidic chip. By simply turning the magnitude of the applied electric pulse, the technique is suitable for different purposes, for example, the separations of particles and cells, purification of cells, and viability identification of cells. This tunable separation approach opens opportunities in multidimensional particle and cell sorting for the fields of seed selection of microorganisms, environmental assessment, and biomedical research.
Collapse
Affiliation(s)
- Mengqi Li
- Department of Marine Engineering, Dalian Maritime University, Dalian 116026, China
| | - Deyu Li
- Department of Marine Engineering, Dalian Maritime University, Dalian 116026, China
| | - Yongxin Song
- Department of Marine Engineering, Dalian Maritime University, Dalian 116026, China
| | - Dongqing Li
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1.
| |
Collapse
|
57
|
Xu X, Lin J, Guo Y, Wu X, Xu Y, Zhang D, Zhang X, Yujiao X, Wang J, Yao C, Yao J, Xing J, Cao Y, Li Y, Ren W, Chen T, Ren Y, Wu A. TiO2-based Surface-Enhanced Raman Scattering bio-probe for efficient circulating tumor cell detection on microfilter. Biosens Bioelectron 2022; 210:114305. [DOI: 10.1016/j.bios.2022.114305] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/05/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022]
|
58
|
Chen B, Wang G, Huang C, Sun Y, Zhang J, Chai Z, Guo SS, Zhao XZ, Yuan Y, Liu W. A light-induced hydrogel responsive platform to capture and selectively isolate single circulating tumor cells. NANOSCALE 2022; 14:3504-3512. [PMID: 35171188 DOI: 10.1039/d1nr06876h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Isolation of circulating tumor cells (CTCs) from patients is a challenge due to the rarity of CTCs. Recently, various platforms to capture and release CTCs for downstream analysis have been developed. However, most of the reported release methods provide external stimuli to release all captured cells, which lead to lack of specificity in the pool of collected cells, and the external stimuli may affect the activity of the released cells. Here, we presented a simple method for single-cell recovery to overcome the shortcomings, which combined the nanostructures with a photocurable hydrogel, chondroitin sulfate methacryloyl (CSMA). In brief, we synthesized gelatin nanoparticles (Gnps) and modified them on flat glass (Gnp substrate) for the specific capture of CTCs. A 405 nm laser was projected onto the selected cells, and then CSMA was cured to encapsulate the selected CTCs. Unselected cells were removed with MMP-9 enzyme solution, and selected CTCs were recovered using a microcapillary. Finally, the photocurable hydrogel-encapsulated cells were analyzed by nucleic acid detection. In addition, the results suggested that the isolation platform showed good biocompatibility and successfully achieved the isolation of selected cells. In summary, our light-induced hydrogel responsive platform holds certain potential for clinical applications.
Collapse
Affiliation(s)
- Bei Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Ganggang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Chunyu Huang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Yue Sun
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Jing Zhang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Zhuomin Chai
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, China.
| |
Collapse
|
59
|
Feng T, Karges J, Liao X, Ji L, Chao H. Engineered exosomes as a natural nanoplatform for cancer targeted delivery of metal-based drugs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
60
|
Dotse E, Lim KH, Wang M, Wijanarko KJ, Chow KT. An Immunological Perspective of Circulating Tumor Cells as Diagnostic Biomarkers and Therapeutic Targets. Life (Basel) 2022; 12:323. [PMID: 35207611 PMCID: PMC8878951 DOI: 10.3390/life12020323] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 11/19/2022] Open
Abstract
Immune modulation is a hallmark of cancer. Cancer-immune interaction shapes the course of disease progression at every step of tumorigenesis, including metastasis, of which circulating tumor cells (CTCs) are regarded as an indicator. These CTCs are a heterogeneous population of tumor cells that have disseminated from the tumor into circulation. They have been increasingly studied in recent years due to their importance in diagnosis, prognosis, and monitoring of treatment response. Ample evidence demonstrates that CTCs interact with immune cells in circulation, where they must evade immune surveillance or modulate immune response. The interaction between CTCs and the immune system is emerging as a critical point by which CTCs facilitate metastatic progression. Understanding the complex crosstalk between the two may provide a basis for devising new diagnostic and treatment strategies. In this review, we will discuss the current understanding of CTCs and the complex immune-CTC interactions. We also present novel options in clinical interventions, targeting the immune-CTC interfaces, and provide some suggestions on future research directions.
Collapse
Affiliation(s)
- Eunice Dotse
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China; (E.D.); (K.H.L.); (M.W.)
| | - King H. Lim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China; (E.D.); (K.H.L.); (M.W.)
| | - Meijun Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China; (E.D.); (K.H.L.); (M.W.)
| | - Kevin Julio Wijanarko
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3010, Australia;
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Kwan T. Chow
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 999077, China; (E.D.); (K.H.L.); (M.W.)
| |
Collapse
|
61
|
Sabirova A, Florica CF, Pisig F, Syed A, Buttner U, Li X, Nunes SP. Nanoporous membrane fabrication by nanoimprint lithography for nanoparticle sieving. NANOSCALE ADVANCES 2022; 4:1119-1124. [PMID: 36131770 PMCID: PMC9417922 DOI: 10.1039/d1na00812a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/22/2021] [Indexed: 06/15/2023]
Abstract
An isoporous membrane with strictly controlled pore size, shape and distribution could provide an efficient, precise and mild sieving of particles in nanotechnology and biomedical applications. However there is a lack of highly porous polymeric membranes combining isoporosity and high permeance in the range below 500 nm. Track-etched membranes are practically the only commercial option. Membranes prepared by phase inversion typically have a broad pore size distribution. Most nanofabrication methods have limited the preparation of membranes with pores in the micrometer range. In this work, we present a nanotechnology-based fabrication methodology to manufacture a stable and flexible nanoporous polymeric membrane with 300 nm isopores using UV nanoimprint lithography. The highly porous membrane has a pore density of 4 × 109 pores per cm2 and stable permeance of 108 000 L m-2 h-1 bar-1. Uniform ZIF-8 nanoparticles were synthesized and the isoporous membrane successfully demonstrated as high as 100% rejection and size-based sieving performance of nanoparticles.
Collapse
Affiliation(s)
- Ainur Sabirova
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Advanced Membranes and Porous Materials Center 23955-6900 Thuwal Saudi Arabia
| | - Camelia F Florica
- King Abdullah University of Science and Technology (KAUST), Nanofabrication Core Laboratory 23955-6900 Thuwal Saudi Arabia
| | - Florencio Pisig
- King Abdullah University of Science and Technology (KAUST), Nanofabrication Core Laboratory 23955-6900 Thuwal Saudi Arabia
| | - Ahad Syed
- King Abdullah University of Science and Technology (KAUST), Nanofabrication Core Laboratory 23955-6900 Thuwal Saudi Arabia
| | - Ulrich Buttner
- King Abdullah University of Science and Technology (KAUST), Nanofabrication Core Laboratory 23955-6900 Thuwal Saudi Arabia
| | - Xiang Li
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Advanced Membranes and Porous Materials Center 23955-6900 Thuwal Saudi Arabia
| | - Suzana P Nunes
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE) Division, Advanced Membranes and Porous Materials Center 23955-6900 Thuwal Saudi Arabia
| |
Collapse
|
62
|
Zhao K, Liu Y, Wang H, Song Y, Chen X, Huang C, Niu Q, Cao J, Chen X, Wang W, Wu L, Yang C. Selective, user-friendly, highly porous, efficient, and rapid (SUPER) filter for isolation and analysis of rare tumor cells. LAB ON A CHIP 2022; 22:367-376. [PMID: 34918732 DOI: 10.1039/d1lc00886b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Rapid, efficient, and selective separation of tumor cells from complex body fluids is urgently needed for clinical application of tumor-cell-based liquid biopsy. Herein, a size-selective affinity filtration system, named selective, user-friendly, highly porous, efficient, and rapid filter (SUPER Filter), was developed for high-performance tumor cell isolation and analysis. SUPER Filter enabled selective interaction of tumor cells with size-optimized and antibody-coated micropore walls during filtration, achieving a high efficiency of 91.0 ± 6.1% in buffer and 83.7 ± 6.4% in whole blood. Meanwhile, its larger micropore size than those of traditional filtration devices greatly reduced the nonspecific capture of background cells (55-126 cells per mL blood) with enrichment factors of 1.1 × 104-1.0 × 105 and a purity of 52.7 ± 4.2%. Moreover, its high porosity enabled ultra-fast (<5 s for 1 mL of blood or 10 mL of buffer samples) and user-friendly gravity-driven filtration. Finally, SUPER Filter demonstrated rapid, efficient, and selective separation of tumor cells from blood and large-volume pleural and ascetic fluid samples from cancer patients for morphological and molecular analysis. We expect that this size-selective affinity filtration strategy facilitates the clinical application of tumor-cell-based liquid biopsy.
Collapse
Affiliation(s)
- Kaifeng Zhao
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yaoping Liu
- Institute of Microelectronics, Peking University, Beijing, 100871, China.
| | - Hua Wang
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaofeng Chen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chen Huang
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Qi Niu
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jiao Cao
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Xin Chen
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Wei Wang
- Institute of Microelectronics, Peking University, Beijing, 100871, China.
| | - Lingling Wu
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Chaoyong Yang
- Institute of Molecular Medicine, Clinical Laboratory, Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
63
|
Cheng J, Zhang L, Zhang Y, Ye Y, Zhao W, Zhang L, Li Y, Liu Y, Zhang W, Guo H, Li M, Zhao Y, Huang C. 3D spiral channels combined with flexible micro-sieve for high-throughput rare tumor cell enrichment and assay from clinical pleural effusion samples. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
64
|
Design and Numerical Simulation of Biomimetic Structures to Capture Particles in a Microchannel. FLUIDS 2022. [DOI: 10.3390/fluids7010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The study of separating different sizes of particles through a microchannel has been an interest in recent years and the primary attention of this study is to isolate the particles to the specific outlets. The present work highly focuses on the design and numerical analysis of a microchip and the microparticles capture using special structures like corrugated dragonfly wing structure and cilia walls. The special biomimetic structured corrugated wing is taken from the cross-sectional area of the dragonfly wing and cilia structure is obtained from the epithelium terminal bronchioles to the larynx from the human body. Parametric studies were conducted on different sizes of microchip scaled and tested up in the range between 2–6 mm and the thickness was assigned as 80 µm in both dragonfly wing structure and cilia walls. The microflow channel is a low Reynolds number regime and with the help of the special structures, the flow inside the microchannel is pinched and a sinusoidal waveform pattern is observed. The pinched flow with sinusoidal waveform carries the particles downstream and induces the particles trapped in desired outlets. Fluid particle interaction (FPI) with a time-dependent solver in COMSOL Multiphysics was used to carry out the numerical study. Two particle sizes of 5 µm and 20 µm were applied, the inlet velocity of 0.52 m/s with an inflow angle of 50° was used throughout the study and it suggested that: the microchannel length of 3 mm with corrugated dragonfly wing structure had the maximum particle capture rate of 20 µm at the mainstream outlet. 80% capture rate for the microchannel length of 3 mm with corrugated dragonfly wing structure and 98% capture rate for the microchannel length of 2 mm with cilia wall structure were observed. Numerical simulation results showed that the cilia walled microchip is superior to the corrugated wing structure as the mainstream outlet can conduct most of the 20 µm particles. At the same time, the secondary outlet can laterally capture most of the 5 µm particles. This biomimetic microchip design is expected to be implemented using the PDMS MEMS process in the future.
Collapse
|
65
|
Methods for the Detection of Circulating Biomarkers in Cancer Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:525-552. [DOI: 10.1007/978-3-031-04039-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
66
|
Xu Y, Zhang D, Lin J, Wu X, Xu X, Akakuru OU, Zhang H, Zhang Z, Xie Y, Wu A, Shao G. Ultrahigh SERS Activity of TiO2@Ag Nanostructure leveraged for Accurately Detecting CTCs in peripheral blood. Biomater Sci 2022; 10:1812-1820. [PMID: 35234756 DOI: 10.1039/d1bm01821c] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Circulating tumor cells (CTCs) usually shed from primary and metastatic tumors serve as an important tumor marker, and easily cause fatal distant metastasis in cancer patients. Accurately and effectively detecting...
Collapse
Affiliation(s)
- Yanping Xu
- Second clinical college, Zhejiang Chinese Medical University, Hang Zhou 310053, China
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Dinghu Zhang
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Jie Lin
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Xiaoxia Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Xiawei Xu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Ozioma Udochukwu Akakuru
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Hao Zhang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Zhewei Zhang
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Yujiao Xie
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P.R. China
| | - Guoliang Shao
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
67
|
Antibody-engineered red blood cell interface for high-performance capture and release of circulating tumor cells. Bioact Mater 2021; 11:32-40. [PMID: 34938910 DOI: 10.1016/j.bioactmat.2021.09.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs), as important liquid biopsy target, can provide valuable information for cancer progress monitoring and individualized treatment. However, current isolation platforms incapable of balancing capture efficiency, specificity, cell viability, and gentle release have restricted the clinical applications of CTCs. Herein, inspired by the structure and functional merits of natural membrane interfaces, we established an antibody-engineered red blood cell (RBC-Ab) affinity interface on microfluidic chip for high-performance isolation and release of CTCs. The lateral fluidity, pliability, and anti-adhesion property of the RBC microfluidic interface enabled efficient CTCs capture (96.5%), high CTCs viability (96.1%), and high CTCs purity (average 4.2-log depletion of leukocytes). More importantly, selective lysis of RBCs by simply changing the salt concentration was utilized to destroy the affinity interface for efficient and gentle release of CTCs without nucleic acid contamination. Using this chip, CTCs were successfully detected in colon cancer samples with 90% sensitivity and 100% specificity (20 patients and 10 healthy individuals). After the release process, KRAS gene mutations of CTCs were identified from all the 5 cancer samples, which was consistent with the results of tissue biopsy. We expect this RBC interface strategy will inspire further biomimetic interface construction for rare cell analysis.
Collapse
|
68
|
Li F, Xu H, Zhao Y. Magnetic particles as promising circulating tumor cell catchers assisting liquid biopsy in cancer diagnosis: A review. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
69
|
He S, Wei J, Ding L, Yang X, Wu Y. State-of-the-arts techniques and current evolving approaches in the separation and detection of circulating tumor cell. Talanta 2021; 239:123024. [PMID: 34952370 DOI: 10.1016/j.talanta.2021.123024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells that shed from the primary tumor and then enter the circulatory system, a small part of which may evolve into metastatic cancer under appropriate microenvironment conditions. The detection of CTCs is a truly noninvasive, dynamic monitor for disease changes, which has considerable clinical implications in the selection of targeted drugs. However, their inherent rarity and heterogeneity pose significant challenges to their isolation and detection. Even the "gold standard", CellSearch™, suffers from high expenses, low capture efficiency, and the consumption of time. With the advancement of CTCs analysis technologies in recent years, the yield and efficiency of CTCs enrichment have gradually been improved, as well as detection sensitivity. In this review, the isolation and detection strategies of CTCs have been completely described and the potential directions for future research and development have also been highlighted through analyzing the challenges faced by current strategies.
Collapse
Affiliation(s)
- Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Jinlan Wei
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
70
|
Smith KJ, Jana JA, Kaehr A, Purcell E, Opdycke T, Paoletti C, Cooling L, Thamm DH, Hayes DF, Nagrath S. Inertial focusing of circulating tumor cells in whole blood at high flow rates using the microfluidic CTCKey™ device for CTC enrichment. LAB ON A CHIP 2021; 21:3559-3572. [PMID: 34320046 DOI: 10.1039/d1lc00546d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Circulating tumor cells (CTCs) are extremely rare cells shed from tumors into the blood stream. These cells can provide valuable information about their tumor of origin and direct treatment decisions to improve patient outcomes. Current technologies isolate CTCs from a limited blood volume and often require pre-processing that leads to CTC loss, making it difficult to isolate enough CTCs to perform in-depth tumor analysis. Many inertial microfluidic devices have been developed to isolate CTCs at high flow rates, but they typically require either blood dilution, pre-processing to remove red blood cells, or a sheath buffer rather than being able to isolate cells directly from whole blood. To decrease the need for pre-processing while increasing CTC yield, we developed an inertial device, the CTCKey™, to focus CTCs in whole blood at high throughput yielding a concentrated product stream enriched for CTCs. The CTCKey™ consists of two sections to create CTC enriched blood that can be further processed using any CTC isolation device to selectively isolate the CTCs. A thorough analysis was performed using the MCF7 breast cancer cell line spiked into bovine serum albumin (BSA) solutions of varying concentrations, as well as whole blood to characterize the focusing patterns of the CTCKey™. At the optimal flow rate of 2.4 mL min-1, the CTCKey™ reduces the CTC containing blood volume by 78%; the CTCs from 1 mL of blood are now in 0.22 mL of blood. The CTCKey's™ ability to concentrate CTCs from a large original blood volume to a smaller, highly concentrated volume enables a much greater blood volume to be interrogated by downstream isolation and characterization methods despite their low volume input limitations.
Collapse
Affiliation(s)
- Kaylee Judith Smith
- Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, USA.
| | | | - Anna Kaehr
- Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, USA.
| | - Emma Purcell
- Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, USA.
| | - Tyler Opdycke
- Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, USA.
| | - Costanza Paoletti
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Laura Cooling
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas H Thamm
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel F Hayes
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Sunitha Nagrath
- Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, USA.
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
71
|
Pan Y, Lu B, Peng Y, Wang M, Deng Y, Yin Y, Yang J, Li G. A simple method to assay tumor cells based on target-initiated steric hindrance. Chem Commun (Camb) 2021; 57:6522-6525. [PMID: 34105555 DOI: 10.1039/d1cc02532e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have proposed a simple electrochemical method in this work for the assay of tumor cells through their own steric hindrance effect. Specifically, tumor cells can block the catalysis of terminal deoxynucleotidyl transferase to the aptamer previously immobilized on the electrode surface. By making use of the hindrance effect, cancer cells can be quantitatively analyzed in the range from 1.6 × 102 to 1.6 × 106 cells per mL without complicated design or cumbersome operation, while the detection limit can be about 53 cells per mL. This method can also show satisfactory performance in complex environments, indicating its potential in clinical application.
Collapse
Affiliation(s)
- Yanhong Pan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Bing Lu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Peng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Minghui Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Deng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yongmei Yin
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China and Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
72
|
Recent Development of Nanomaterials-Based Cytosensors for the Detection of Circulating Tumor Cells. BIOSENSORS-BASEL 2021; 11:bios11080281. [PMID: 34436082 PMCID: PMC8391755 DOI: 10.3390/bios11080281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
The accurate analysis of circulating tumor cells (CTCs) holds great promise in early diagnosis and prognosis of cancers. However, the extremely low abundance of CTCs in peripheral blood samples limits the practical utility of the traditional methods for CTCs detection. Thus, novel and powerful strategies have been proposed for sensitive detection of CTCs. In particular, nanomaterials with exceptional physical and chemical properties have been used to fabricate cytosensors for amplifying the signal and enhancing the sensitivity. In this review, we summarize the recent development of nanomaterials-based optical and electrochemical analytical techniques for CTCs detection, including fluorescence, colorimetry, surface-enhanced Raman scattering, chemiluminescence, electrochemistry, electrochemiluminescence, photoelectrochemistry and so on.
Collapse
|
73
|
Dunphy K, O’Mahoney K, Dowling P, O’Gorman P, Bazou D. Clinical Proteomics of Biofluids in Haematological Malignancies. Int J Mol Sci 2021; 22:ijms22158021. [PMID: 34360786 PMCID: PMC8348619 DOI: 10.3390/ijms22158021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Since the emergence of high-throughput proteomic techniques and advances in clinical technologies, there has been a steady rise in the number of cancer-associated diagnostic, prognostic, and predictive biomarkers being identified and translated into clinical use. The characterisation of biofluids has become a core objective for many proteomic researchers in order to detect disease-associated protein biomarkers in a minimally invasive manner. The proteomes of biofluids, including serum, saliva, cerebrospinal fluid, and urine, are highly dynamic with protein abundance fluctuating depending on the physiological and/or pathophysiological context. Improvements in mass-spectrometric technologies have facilitated the in-depth characterisation of biofluid proteomes which are now considered hosts of a wide array of clinically relevant biomarkers. Promising efforts are being made in the field of biomarker diagnostics for haematologic malignancies. Several serum and urine-based biomarkers such as free light chains, β-microglobulin, and lactate dehydrogenase are quantified as part of the clinical assessment of haematological malignancies. However, novel, minimally invasive proteomic markers are required to aid diagnosis and prognosis and to monitor therapeutic response and minimal residual disease. This review focuses on biofluids as a promising source of proteomic biomarkers in haematologic malignancies and a key component of future diagnostic, prognostic, and disease-monitoring applications.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Kelly O’Mahoney
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
- Correspondence:
| |
Collapse
|
74
|
Delikoyun K, Yaman S, Yilmaz E, Sarigil O, Anil-Inevi M, Telli K, Yalcin-Ozuysal O, Ozcivici E, Tekin HC. HologLev: A Hybrid Magnetic Levitation Platform Integrated with Lensless Holographic Microscopy for Density-Based Cell Analysis. ACS Sens 2021; 6:2191-2201. [PMID: 34124887 DOI: 10.1021/acssensors.0c02587] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In clinical practice, a variety of diagnostic applications require the identification of target cells. Density has been used as a physical marker to distinguish cell populations since metabolic activities could alter the cell densities. Magnetic levitation offers great promise for separating cells at the single cell level within heterogeneous populations with respect to cell densities. Traditional magnetic levitation platforms need bulky and precise optical microscopes to visualize levitated cells. Moreover, the evaluation process of cell densities is cumbersome, which also requires trained personnel for operation. In this work, we introduce a device (HologLev) as a fusion of the magnetic levitation principle and lensless digital inline holographic microscopy (LDIHM). LDIHM provides ease of use by getting rid of bulky and expensive optics. By placing an imaging sensor just beneath the microcapillary channel without any lenses, recorded holograms are processed for determining cell densities through a fully automated digital image processing scheme. The device costs less than $100 and has a compact design that can fit into a pocket. We perform viability tests on the device by levitating three different cell lines (MDA-MB-231, U937, D1 ORL UVA) and comparing them against their dead correspondents. We also tested the differentiation of mouse osteoblastic (7F2) cells by monitoring characteristic variations in their density. Last, the response of MDA-MB-231 cancer cells to a chemotherapy drug was demonstrated in our platform. HologLev provides cost-effective, label-free, fully automated cell analysis in a compact design that could be highly desirable for laboratory and point-of-care testing applications.
Collapse
Affiliation(s)
- Kerem Delikoyun
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Sena Yaman
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Esra Yilmaz
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Oyku Sarigil
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Muge Anil-Inevi
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Kubra Telli
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Ozden Yalcin-Ozuysal
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Engin Ozcivici
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - H. Cumhur Tekin
- Faculty of Engineering, Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
- METU MEMS Center, Ankara 06520, Turkey
| |
Collapse
|
75
|
Advances in aptamer-based nanomaterials for separation and analysis of non-genetic biomarkers in biofluids. Sci China Chem 2021. [DOI: 10.1007/s11426-020-9955-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
76
|
Hou J, Liu X, Zhou S. Programmable materials for efficient CTCs isolation: From micro/nanotechnology to biomimicry. VIEW 2021. [DOI: 10.1002/viw.20200023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Jianwen Hou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xia Liu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
| |
Collapse
|
77
|
Biagioni V, Sow AL, Adrover A, Cerbelli S. Brownian Sieving Effect for Boosting the Performance of Microcapillary Hydrodynamic Chromatography. Proof of Concept. Anal Chem 2021; 93:6808-6816. [PMID: 33890769 PMCID: PMC8253478 DOI: 10.1021/acs.analchem.1c00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microcapillary hydrodynamic chromatography (MHDC) is a well-established technique for the size-based separation of suspensions and colloids, where the characteristic size of the dispersed phase ranges from tens of nanometers to micrometers. It is based on hindrance effects which prevent relatively large particles from experiencing the low velocity region near the walls of a pressure-driven laminar flow through an empty microchannel. An improved device design is here proposed, where the relative extent of the low velocity region is made tunable by exploiting a two-channel annular geometry. The geometry is designed so that the core and the annular channel are characterized by different average flow velocities when subject to one and the same pressure drop. The channels communicate through openings of assigned cut-off length, say A. As they move downstream the channel, particles of size bigger than A are confined to the core region, whereas smaller particles can diffuse through the openings and spread throughout the entire cross section, therein attaining a spatially uniform distribution. By using a classical excluded-volume approach for modeling particle transport, we perform Lagrangian-stochastic simulations of particle dynamics and compare the separation performance of the two-channel and the standard (single-channel) MHDC. Results suggest that a quantitative (up to thirtyfold) performance enhancement can be obtained at operating conditions and values of the transport parameters commonly encountered in practical implementations of MHDC. The separation principle can readily be extended to a multistage geometry when the efficient fractionation of an arbitrary size distribution of the suspension is sought.
Collapse
Affiliation(s)
- Valentina Biagioni
- Dipartimento di Ingegneria Chimica Materiali Ambiente, Sapienza Università di Roma, Via Eudossiana 18, Roma 00184, Italy
| | - Alpha L Sow
- Dipartimento di Ingegneria Chimica Materiali Ambiente, Sapienza Università di Roma, Via Eudossiana 18, Roma 00184, Italy
| | - Alessandra Adrover
- Dipartimento di Ingegneria Chimica Materiali Ambiente, Sapienza Università di Roma, Via Eudossiana 18, Roma 00184, Italy
| | - Stefano Cerbelli
- Dipartimento di Ingegneria Chimica Materiali Ambiente, Sapienza Università di Roma, Via Eudossiana 18, Roma 00184, Italy
| |
Collapse
|
78
|
Hu X, Zang X, Lv Y. Detection of circulating tumor cells: Advances and critical concerns. Oncol Lett 2021; 21:422. [PMID: 33850563 PMCID: PMC8025150 DOI: 10.3892/ol.2021.12683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the main cause of cancer-related death and the major challenge in cancer treatment. Cancer cells in circulation are termed circulating tumor cells (CTCs). Primary tumor metastasis is likely due to CTCs released into the bloodstream. These CTCs extravasate and form fatal metastases in different organs. Analyses of CTCs are clarifying the biological understanding of metastatic cancers. These data are also helpful to monitor disease progression and to inform the development of personalized cancer treatment-based liquid biopsy. However, CTCs are a rare cell population with 1-10 CTCs per ml and are difficult to isolate from blood. Numerous approaches to detect CTCs have been developed based on the physical and biological properties of the cells. The present review summarizes the progress made in detecting CTCs.
Collapse
Affiliation(s)
- Xiuxiu Hu
- School of Medical Technology, Jiangsu College of Nursing, Huai'an, Jiangsu 22300, P.R. China
| | - Xiaojuan Zang
- Department of Ultrasonography, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| | - Yanguan Lv
- Clinical Medical Laboratory, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
79
|
Research Progress for the Clinical Application of Circulating Tumor Cells in Prostate Cancer Diagnosis and Treatment. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6230826. [PMID: 33506020 PMCID: PMC7814947 DOI: 10.1155/2021/6230826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Prostate cancer is a life-threatening and highly heterogeneous malignancy. In the past decade, circulating tumor cells (CTCs) have been suggested to play a critical role in the occurrence and progression of prostate cancer. In particular, as the “seed” of the cancer metastasis cascade, CTCs determine numerous biological behaviors, such as tumor invasion into adjacent tissues and migration to distant organs. Many studies have shown that CTCs are necessary in the processes of tumor progression, including tumorigenesis, invasion, metastasis, and colonization. Furthermore, CTCs express various biomarkers relevant to prostate cancer and thus can be applied clinically in noninvasive tests. Moreover, CTCs can serve as potential prognostic targets in prostate cancer due to their roles in regulating many processes associated with cancer metastasis. In this review, we discuss the isolation and detection of CTCs as predictive markers of prostate cancer, and we discuss their clinical application in the diagnosis and prognosis of prostate cancer and in monitoring the response to treatment and the prediction of metastasis.
Collapse
|
80
|
Mondelo-Macía P, García-González J, León-Mateos L, Castillo-García A, López-López R, Muinelo-Romay L, Díaz-Peña R. Current Status and Future Perspectives of Liquid Biopsy in Small Cell Lung Cancer. Biomedicines 2021; 9:48. [PMID: 33430290 PMCID: PMC7825645 DOI: 10.3390/biomedicines9010048] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 01/08/2023] Open
Abstract
Approximately 19% of all cancer-related deaths are due to lung cancer, which is the leading cause of mortality worldwide. Small cell lung cancer (SCLC) affects approximately 15% of patients diagnosed with lung cancer. SCLC is characterized by aggressiveness; the majority of SCLC patients present with metastatic disease, and less than 5% of patients are alive at 5 years. The gold standard of SCLC treatment is platinum and etoposide-based chemotherapy; however, its effects are short. In recent years, treatment for SCLC has changed; new drugs have been approved, and new biomarkers are needed for treatment selection. Liquid biopsy is a non-invasive, rapid, repeated and alternative tool to the traditional tumor biopsy that could allow the most personalized medicine into the management of SCLC patients. Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) are the most commonly used liquid biopsy biomarkers. Some studies have reported the prognostic factors of CTCs and cfDNA in SCLC patients, independent of the stage. In this review, we summarize the recent SCLC studies of CTCs, cfDNA and other liquid biopsy biomarkers, and we discuss the future utility of liquid biopsy in the clinical management of SCLC.
Collapse
Affiliation(s)
- Patricia Mondelo-Macía
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
| | - Jorge García-González
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Luis León-Mateos
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | | | - Rafael López-López
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Roberto Díaz-Peña
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
81
|
Design and Clinical Application of an Integrated Microfluidic Device for Circulating Tumor Cells Isolation and Single-Cell Analysis. MICROMACHINES 2021; 12:mi12010049. [PMID: 33401770 PMCID: PMC7824094 DOI: 10.3390/mi12010049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022]
Abstract
Circulating tumor cells (CTCs) have been considered as an alternative to tissue biopsy for providing both germline-specific and tumor-derived genetic variations. Single-cell analysis of CTCs enables in-depth investigation of tumor heterogeneity and individualized clinical assessment. However, common CTC enrichment techniques generally have limitations of low throughput and cell damage. Herein, based on micropore-arrayed filtration membrane and microfluidic chip, we established an integrated CTC isolation platform with high-throughput, high-efficiency, and less cell damage. We observed a capture rate of around 85% and a purity of 60.4% by spiking tumor cells (PC-9) into healthy blood samples. Detection of CTCs from lung cancer patients demonstrated a positive detectable rate of 87.5%. Additionally, single CTCs, ctDNA and liver biopsy tissue of a representative advanced lung cancer patient were collected and sequenced, which revealed comprehensive genetic information of CTCs while reflected the differences in genetic profiles between different biological samples. This work provides a promising tool for CTCs isolation and further analysis at single-cell resolution with potential clinical value.
Collapse
|
82
|
Chow T, Wutami I, Lucarelli E, Choong PF, Duchi S, Di Bella C. Creating In Vitro Three-Dimensional Tumor Models: A Guide for the Biofabrication of a Primary Osteosarcoma Model. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:514-529. [PMID: 33138724 DOI: 10.1089/ten.teb.2020.0254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is a highly aggressive primary bone tumor. The mainstay for its treatment is multiagent chemotherapy and surgical resection, with a 50-70% 5-year survival rate. Despite the huge effort made by clinicians and researchers in the past 30 years, limited progress has been made to improve patient outcomes. As novel therapeutic approaches for OS become available, such as monoclonal antibodies, small molecules, and immunotherapies, the need for OS preclinical model development becomes equally pressing. Three-dimensional (3D) OS models represent an alternative system to study this tumor: In contrast to two-dimensional monolayers, 3D matrices can recapitulate key elements of the tumor microenvironment (TME), such as the cellular interaction with the bone mineralized matrix. The advancement of tissue engineering and biofabrication techniques enables the incorporation of specific TME aspects into 3D models, to investigate the contribution of individual components to tumor progression and enhance understanding of basic OS biology. The use of biomaterials that mimic the extracellular matrix could also facilitate the testing of drugs targeting the TME itself, allowing a larger range of therapeutics to be tested, while averting the ethical implications and high cost associated with in vivo preclinical models. This review aims at serving as a practical guide by delineating the OS TME ("what it is like") and, in turn, propose various biofabrication strategies to create a 3D model ("how to recreate it"), to improve the in vitro representation of the OS tumor and ultimately generate more accurate drug response profiles.
Collapse
Affiliation(s)
- Thomas Chow
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia.,BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Ilycia Wutami
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia.,BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Enrico Lucarelli
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Peter F Choong
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Serena Duchi
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Claudia Di Bella
- BioFab3D-ACMD, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| |
Collapse
|
83
|
Cess CG, Finley SD. Multi-scale modeling of macrophage-T cell interactions within the tumor microenvironment. PLoS Comput Biol 2020; 16:e1008519. [PMID: 33362239 PMCID: PMC7790427 DOI: 10.1371/journal.pcbi.1008519] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/07/2021] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Within the tumor microenvironment, macrophages exist in an immunosuppressive state, preventing T cells from eliminating the tumor. Due to this, research is focusing on immunotherapies that specifically target macrophages in order to reduce their immunosuppressive capabilities and promote T cell function. In this study, we develop an agent-based model consisting of the interactions between macrophages, T cells, and tumor cells to determine how the immune response changes due to three macrophage-based immunotherapeutic strategies: macrophage depletion, recruitment inhibition, and macrophage reeducation. We find that reeducation, which converts the macrophages into an immune-promoting phenotype, is the most effective strategy and that the macrophage recruitment rate and tumor proliferation rate (tumor-specific properties) have large impacts on therapy efficacy. We also employ a novel method of using a neural network to reduce the computational complexity of an intracellular signaling mechanistic model.
Collapse
Affiliation(s)
- Colin G. Cess
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, United States of America
| | - Stacey D. Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, United States of America
- Department of Quantitative and Biological Sciences, University of Southern California, Los Angeles, California, United States of America
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
84
|
Gao T, Ding P, Li W, Wang Z, Lin Q, Pei R. Isolation of DNA aptamers targeting N-cadherin and high-efficiency capture of circulating tumor cells by using dual aptamers. NANOSCALE 2020; 12:22574-22585. [PMID: 33174555 DOI: 10.1039/d0nr06180h] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
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 Kd 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.
Collapse
Affiliation(s)
- Tian Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | | | | | | | | | | |
Collapse
|
85
|
On-Chip Multiple Particle Velocity and Size Measurement Using Single-Shot Two-Wavelength Differential Image Analysis. MICROMACHINES 2020; 11:mi11111011. [PMID: 33212970 PMCID: PMC7698501 DOI: 10.3390/mi11111011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 12/11/2022]
Abstract
Precise and quick measurement of samples' flow velocities is essential for cell sorting timing control and reconstruction of acquired image-analyzed data. We developed a simple technique for the single-shot measurement of flow velocities of particles simultaneously in a microfluidic pathway. The speed was calculated from the difference in the particles' elongation in an acquired image that appeared when two wavelengths of light with different irradiation times were applied. We ran microparticles through an imaging flow cytometer and irradiated two wavelengths of light with different irradiation times simultaneously to those particles. The mixture of the two wavelength transmitted lights was divided into two wavelengths, and the images of the same microparticles for each wavelength were acquired in a single shot. We estimated the velocity from the difference of its elongation divided by the difference of irradiation time by comparing these two images. The distribution of polystyrene beads' velocity was parabolic and highest at the center of the flow channel, consistent with the expected velocity distribution of the laminar flow. Applying the calculated velocity, we also restored the accurate shapes and cross-sectional areas of particles in the images, indicating this simple method for improving of imaging flow cytometry and cell sorter for diagnostic screening of circulating tumor cells.
Collapse
|
86
|
Lu C, Xu J, Han J, Li X, Xue N, Li J, Wu W, Sun X, Wang Y, Ouyang Q, Yang G, Luo C. A novel microfluidic device integrating focus-separation speed reduction design and trap arrays for high-throughput capture of circulating tumor cells. LAB ON A CHIP 2020; 20:4094-4105. [PMID: 33089845 DOI: 10.1039/d0lc00631a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Isolation and analysis of circulating tumor cells (CTCs) from peripheral blood provides a potential way to detect and characterize cancer. Existing technologies to separate or capture CTCs from whole blood still have issues with sample throughput, separation efficiency or stable efficiency at different flow rates. Here, we proposed a new concept to capture rare CTCs from blood by integrating a triangular prism array-based capture apparatus with streamline-based focus-separation speed reduction design. The focus-separation design could focus and maintain CTCs, while removing a considerable proportion of liquid (98.9%) containing other blood cells to the outlet, therefore, a high CTC capture efficiency could be achieved in the trap arrays with a high initial flow rate. It is worth mentioning that the new design works well over a wide range of flow rates, so it does not require the stability of the flow rate. The results showed that this novel integrated chip can achieve a sample throughput from 5 to 40 mL h-1 with a stable and high CTC capture efficiency (up to 94.8%) and high purity (up to 4 log white blood cells/WBC depletion). The clinical experiment showed that CTCs including CTC clusters were detected in 11/11 (100.0%) patients (mean = 31 CTCs mL-1, median = 25 CTCs mL-1). In summary, our chip enriches and captures CTCs based on physical properties, and it is simple, cheap, fast, and efficient and has low requirements on flow rate, which is very suitable for large-scale application of CTC testing in clinics.
Collapse
Affiliation(s)
- Chunyang Lu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Jian Xu
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China.
| | - Jintao Han
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Xiao Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ningtao Xue
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Jinsong Li
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Wenhua Wu
- Jining No. 2 People's Hospital, Jining 272049, China
| | - Xinlei Sun
- Jining Tumor Hospital, Jining 272007, China
| | - Yugang Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Qi Ouyang
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China. and Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China.
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China. and Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
87
|
Pei H, Li L, Han Z, Wang Y, Tang B. Recent advances in microfluidic technologies for circulating tumor cells: enrichment, single-cell analysis, and liquid biopsy for clinical applications. LAB ON A CHIP 2020; 20:3854-3875. [PMID: 33107879 DOI: 10.1039/d0lc00577k] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) detach from primary or metastatic lesions and circulate in the peripheral blood, which is considered to be the cause of distant metastases. CTC analysis in the form of liquid biopsy, enumeration and molecular analysis provide significant clinical information for cancer diagnosis, prognosis and therapeutic strategies. Despite the great clinical value, CTC analysis has not yet entered routine clinical practice due to lack of efficient technologies to perform CTC isolation and single-cell analysis. Taking the rarity and inherent heterogeneity of CTCs into account, reliable methods for CTC isolation and detection are in urgent demand for obtaining valuable information on cancer metastasis and progression from CTCs. Microfluidic technology, featuring microfabricated structures, can precisely control fluids and cells at the micrometer scale, thus making itself a particularly suitable method for rare CTC manipulation. Besides the enrichment function, microfluidic chips can also realize the analysis function by integrating multiple detection technologies. In this review, we have summarized the recent progress in CTC isolation and detection using microfluidic technologies, with special attention to emerging direct enrichment and enumeration in vivo. Further, few insights into single CTC molecular analysis are also demonstrated. We have provided a review of potential clinical applications of CTCs, ranging from early screening and diagnosis, tumor progression and prognosis, treatment and resistance monitoring, to therapeutic evaluation. Through this review, we conclude that the clinical utility of CTCs will be expanded as the isolation and analysis techniques are constantly improving.
Collapse
Affiliation(s)
- Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | | | | | | | | |
Collapse
|
88
|
Park K, Lew D, Chapman C, Wachsman A, Bloom M, Bancila L, Perry R, Wang Q, Jamil L, Pandol S, Lo S. Feasibility and safety study of 22-gauge endoscopic ultrasound (EUS) needles for portal vein sampling in a swine model. Endosc Int Open 2020; 8:E1717-E1724. [PMID: 33140030 PMCID: PMC7581479 DOI: 10.1055/a-1264-7206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/20/2020] [Indexed: 11/09/2022] Open
Abstract
Background and study aims Endoscopic ultrasound (EUS) has been used for portal vein sampling in patients with pancreaticobiliary cancers for enumerating circulating tumor cells but is not yet a standard procedure. Further evaluation is needed to refine the methodology. Therefore, we evaluated the feasibility and safety of 19-gauge (19G) versus a 22-gauge (22 G) EUS fine-needle aspiration needles for portal vein sampling in a swine model. Methods Celiotomy was performed on two farm pigs. Portal vein sampling occurred transhepatically. We compared 19 G and 22 G needles coated interiorly with saline, heparin or ethylenediaminetetraacetic acid (EDTA). Small- (10 mL) and large- (25 mL) volume blood collections were evaluated. Two different collection methods were tested: direct-to-vial and suction syringe. A bleeding risk trial for saline-coated 19 G and 22 G needles was performed by puncturing the portal vein 20 times. Persistent bleeding after 3 minutes was considered significant. Results All small-volume collection trials were successful except for 22 G saline-coated needles with direct-to-vial method. All large-volume collection trials were successful when using suction syringe; direct-to-vial method for both 19 G and 22 G needles were unsuccessful. Collection times were shorter for 19 G vs. 22 G needles for both small and large-volume collections ( P < 0.05). Collection times for saline-coated 22 G needles were longer compared to heparin/EDTA-coated ( P < 0.05). Bleeding occurred in 10 % punctures with 19 G needles compared to 0 % with 22 G needles. Conclusion The results of this animal study demonstrate the feasibility and the safety of using 22 G needles for portal vein sampling and can form the basis for a pilot study in patients.
Collapse
Affiliation(s)
- Kenneth Park
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Daniel Lew
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Christopher Chapman
- University of Chicago Medical Center, Center for Endoscopic Research and Therapeutics, Chicago, Illinois, United States
| | - Ashley Wachsman
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Matthew Bloom
- Cedars-Sinai Medical Center – Surgery, Los Angeles, California, United States
| | - Liiana Bancila
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Rachel Perry
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Qiang Wang
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Laith Jamil
- William Beaumont Hospital – Royal Oak, Gastroenterology and Hepatology, Royal Oak, Michigan, United States
| | - Stephen Pandol
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| | - Simon Lo
- Cedars-Sinai Medical Center, Division of Digestive Diseases, Los Angeles, California
| |
Collapse
|
89
|
Özkayar G, Mutlu E, Şahin Ş, Demircan Yalçın Y, Töral T, Külah H, Yildirim E, Zorlu Ö, Özgür E. A Novel Microfluidic Method Utilizing a Hydrofoil Structure to Improve Circulating Tumor Cell Enrichment: Design and Analytical Validation. MICROMACHINES 2020; 11:mi11110981. [PMID: 33143378 PMCID: PMC7693848 DOI: 10.3390/mi11110981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/14/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
Being one of the major pillars of liquid biopsy, isolation and characterization of circulating tumor cells (CTCs) during cancer management provides critical information on the evolution of cancer and has great potential to increase the success of therapies. In this article, we define a novel strategy to effectively enrich CTCs from whole blood based on size, utilizing a spiral microfluidic channel embedded with a hydrofoil structure at the downstream of the spiral channel. The hydrofoil increases the distance between the streams of CTCs and peripheral blood cells, which are already distributed about two focal axes by the spiral channel, thereby improving the resolution of the separation. Analytical validation of the system has been carried out using Michigan Cancer Foundation-7 (MCF7) breast cancer cell lines spiked into blood samples from healthy donors, and the performance of the system in terms of white blood cell (WBC) depletion, CTC recovery rate and cell viability has been shown in single or two-step process: by passing the sample once or twice through the microfluidic chip. Single step process yielded high recovery (77.1%), viable (84.7%) CTCs. When the collected cell suspension is re-processed by the same chip, recovery decreases to 65.5%, while the WBC depletion increases to 88.3%, improving the purity. Cell viability of >80% was preserved after two-step process. The novel microfluidic chip is a good candidate for CTC isolation applications requiring high recovery rate and viability, including functional downstream analyses for variety of cancer types.
Collapse
Affiliation(s)
- Gürhan Özkayar
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Ege Mutlu
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Şebnem Şahin
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Yağmur Demircan Yalçın
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Taylan Töral
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Haluk Külah
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
- Department of Electrical and Electronics Engineering, Middle East Technical University (METU), Ankara 06530, Turkey
| | - Ender Yildirim
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
- Department of Mechanical Engineering, Middle East Technical University (METU), Ankara 06530, Turkey
| | - Özge Zorlu
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
| | - Ebru Özgür
- Mikro Biyosistemler A.Ş., ODTÜ Teknokent MET Yerleskesi, No:280/B/10, Ankara 06530, Turkey; (G.Ö.); (E.M.); (Ş.Ş.); (Y.D.Y.); (T.T.); (H.K.); (E.Y.); (Ö.Z.)
- Correspondence:
| |
Collapse
|
90
|
Electrochemical Detection and Point-of-Care Testing for Circulating Tumor Cells: Current Techniques and Future Potentials. SENSORS 2020; 20:s20216073. [PMID: 33114569 PMCID: PMC7663783 DOI: 10.3390/s20216073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) are tumor cells that escaped from the primary tumor or the metastasis into the blood and they play a major role in the initiation of metastasis and tumor recurrence. Thus, it is widely accepted that CTC is the main target of liquid biopsy. In the past few decades, the separation of CTC based on the electrochemical method has attracted widespread attention due to its convenience, rapidness, low cost, high sensitivity, and no need for complex instruments and equipment. At present, CTC detection is not widely used in the clinic due to various reasons. Point-of-care CTC detection provides us with a possibility, which is sensitive, fast, cheap, and easy to operate. More importantly, the testing instrument is small and portable, and the testing does not require specialized laboratories and specialized clinical examiners. In this review, we summarized the latest developments in the electrochemical-based CTC detection and point-of-care CTC detection, and discussed the challenges and possible trends.
Collapse
|
91
|
Mendelaar PAJ, Kraan J, Van M, Zeune LL, Terstappen LWMM, Oomen-de Hoop E, Martens JWM, Sleijfer S. Defining the dimensions of circulating tumor cells in a large series of breast, prostate, colon, and bladder cancer patients. Mol Oncol 2020; 15:116-125. [PMID: 32949099 PMCID: PMC7782084 DOI: 10.1002/1878-0261.12802] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/24/2020] [Accepted: 09/10/2020] [Indexed: 01/06/2023] Open
Abstract
Circulating tumor cells (CTCs) in the blood of cancer patients are of high clinical relevance. Since detection and isolation of CTCs often rely on cell dimensions, knowledge of their size is key. We analyzed the median CTC size in a large cohort of breast (BC), prostate (PC), colorectal (CRC), and bladder (BLC) cancer patients. Images of patient‐derived CTCs acquired on cartridges of the FDA‐cleared CellSearch® method were retrospectively collected and automatically re‐analyzed using the accept software package. The median CTC diameter (μm) was computed per tumor type. The size differences between the different tumor types and references (tumor cell lines and leukocytes) were nonparametrically tested. A total of 1962 CellSearch® cartridges containing 71 612 CTCs were included. In BC, the median computed diameter (CD) of patient‐derived CTCs was 12.4 μm vs 18.4 μm for cultured cell line cells. For PC, CDs were 10.3 μm for CTCs vs 20.7 μm for cultured cell line cells. CDs for CTCs of CRC and BLC were 7.5 μm and 8.6 μm, respectively. Finally, leukocytes were 9.4 μm. CTC size differed statistically significantly between the four tumor types and between CTCs and the reference data. CTC size differences between tumor types are striking and CTCs are smaller than cell line tumor cells, whose size is often used as reference when developing CTC analysis methods. Based on our data, we suggest that the size of CTCs matters and should be kept in mind when designing and optimizing size‐based isolation methods.
Collapse
Affiliation(s)
- Pauline A J Mendelaar
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| | - Mai Van
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| | - Leonie L Zeune
- Department of Medical Cell BioPhysics, University of Twente, Enschede, The Netherlands
| | - Leon W M M Terstappen
- Department of Medical Cell BioPhysics, University of Twente, Enschede, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC University Medical Center, Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
92
|
Litti L, Colusso A, Pinto M, Ruli E, Scarsi A, Ventura L, Toffoli G, Colombatti M, Fracasso G, Meneghetti M. SERRS multiplexing with multivalent nanostructures for the identification and enumeration of epithelial and mesenchymal cells. Sci Rep 2020; 10:15805. [PMID: 32978492 PMCID: PMC7519640 DOI: 10.1038/s41598-020-72911-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
Liquid biopsy represents a new frontier of cancer diagnosis and prognosis, which allows the isolation of tumor cells released in the blood stream. The extremely low abundance of these cells needs appropriate methodologies for their identification and enumeration. Herein we present a new protocol based on surface enhanced resonance Raman scattering (SERRS) gold multivalent nanostructures to identify and enumerate tumor cells with epithelial and mesenchimal markers. The validation of the protocol is obtained with spiked samples of peripheral blood mononuclear cells (PBMC). Gold nanostructures are functionalized with SERRS labels and with antibodies to link the tumor cells. Three types of such nanosystems were simultaneously used and the protocol allows obtaining the identification of all individual tumor cells with the help of a Random Forest ensemble learning method.
Collapse
Affiliation(s)
- Lucio Litti
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Andrea Colusso
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Marcella Pinto
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Erlis Ruli
- Department of Statistical Sciences, University of Padova, via Battisti 241, 35121, Padua, Italy
| | - Alessia Scarsi
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy
| | - Laura Ventura
- Department of Statistical Sciences, University of Padova, via Battisti 241, 35121, Padua, Italy
| | - Giuseppe Toffoli
- SOC Farmacologia Sperimentale e Clinica, Centro di Riferimento Oncologico, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Marco Colombatti
- Department of Medicine, University of Verona, P.le L.A. Scuro, 37134, Verona, Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona, P.le L.A. Scuro, 37134, Verona, Italy.
| | - Moreno Meneghetti
- Department of Chemical Science, University of Padova, via Marzolo 1, 35131, Padua, Italy.
| |
Collapse
|
93
|
Wang X, Deng L, Gjertsen BT. A microfluidic device for differential capture of heterogeneous rare tumor cells with epithelial and mesenchymal phenotypes. Anal Chim Acta 2020; 1129:1-11. [PMID: 32891378 DOI: 10.1016/j.aca.2020.06.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Accumulating evidence suggests that the features associated with epithelial to mesenchymal transition (EMT) in circulating tumor cells (CTCs) reflect intrinsic metastatic potential and associate with therapy resistance. Thus, profiling the EMT phenotypes of CTCs is increasingly important for cancer diagnosis and prediction of therapeutic responses. However, rapid assessment of the EMT status of a global CTC population is still a challenge due to the difficulty in enriching and phenotyping CTCs simultaneously. Here, we report a microfluidic device consisting of an enrichment section and a capture section for differential capture of rare tumor cells from blood according to their EMT phenotypes. A row of micropillars was constructed in the enrichment section to provide cross-flows for the size-dependent filtration of cells. Thus, MCF-7 and MDA-MB-231 tumor cells mimicking CTCs were first separated from white blood cells through the micropillars and continually flowed into the capture section at a reduced velocity under a differential hydrodynamic pressure. In the capture section, the heterogeneous tumor cells were then phenotypically sorted and captured in two cascaded compartments functionalized with either an anti-EpCAM antibody or a cocktail of antibodies against mesenchymal markers including Axl, PD-L1, and EGFR. Direct counting of the captured cells in each compartment resulted in the enumeration of epithelial and mesenchymal subpopulations of the tumor cells without additional labeling. Furthermore, the captured tumor cells were successfully maintained for up to six days in the device with high viability and marked proliferation for downstream analysis. Thus, this integrated microfluidic device may have great potential in phenotyping EMT status of CTCs for precision cancer therapy.
Collapse
Affiliation(s)
- Xiang Wang
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China; Centre of Cancer Biomarkers CCBIO, Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China
| | - Bjørn Tore Gjertsen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
94
|
Han L, Peng R, Jiang W, Xu T, Zhang C, Chen K, Zhang Y, Song H, Jia L. Coordination-driven reversible surfaces with site-specifically immobilized nanobody for dynamic cancer cell capture and release. J Mater Chem B 2020; 8:7511-7520. [PMID: 32677632 DOI: 10.1039/d0tb00574f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Selective isolation of circulating tumor cells (CTCs) from blood provides a non-invasive avenue for the diagnosis, prognosis and personalized treatment for patients with cancer. The specific capture of CTCs is conventionally based on the immunoaffinity recognition between antibody and receptor on cell membranes. However, using a traditional antibody for high-efficiency isolation of CTCs remains a challenge due to the limited loading capacity of the large antibodies on material surfaces. Herein, using a small-sized nanobody (Nb), we developed a widely applicable strategy to construct reversible site-specifically immobilized Nb surfaces for the capture and release of epidermoid cancer cell line A431 cells. Coordination interaction between the histidine tag (His-tag) of the nanobody (Nb) and Ni2+ ions that chelated to the NTA-modified poly(2-hydroxyethyl methacrylate) (PHEMA) brushes was used to achieve site-specific immobilization of EGFR Nb (PHEMA-aEGFR surfaces). The high-density immobilized nanobody possessing maximized activity resulted in the high-efficiency capture of 81% rare A431 cells within just 30 min, showing a higher capture yield and shorter capture time compared with that achieved by the conventional antibody immobilized on the flat surface. Additionally, the PHEMA-aEGFR surfaces exhibited low capture limit (1 cell mL-1), cytocompatibility for captured cells, as well as negligible non-specific adhesion of PBMCs. With a one-step treatment using imidazole for competitive coordination, 86% of the captured cells were effectively released. This multifunctional and dynamic site-specifically immobilized nanobody strategy paves a new path in the development of materials and instruments for the high-efficiency capture and release of rare cells at a low cost.
Collapse
Affiliation(s)
- Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Agashe R, Kurzrock R. Circulating Tumor Cells: From the Laboratory to the Cancer Clinic. Cancers (Basel) 2020; 12:cancers12092361. [PMID: 32825548 PMCID: PMC7564158 DOI: 10.3390/cancers12092361] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells that are shed from tumors into the bloodstream. Cell enrichment and isolation technology as well as molecular profiling via next-generation sequencing have allowed for a greater understanding of tumor cancer biology via the interrogation of CTCs. CTC detection can be used to predict cancer relapse, progression, and survival; evaluate treatment effectiveness; and explore the ex vivo functional impact of agents. Detection methods can be by either immunoaffinity (positive or negative enrichment strategies) or biophysical strategies. CTC characterization, which is performed by DNA, RNA, and/or protein techniques, can predict metastatic potential. Currently, CTC-derived explant models may mimic patient response to chemotherapy and help with studying druggable targets and testing treatments. The Food and Drug Administration has cleared a CTC blood test to enumerate CTCs derived from breast, prostate, and colorectal cancers. In conclusion, liquid biopsies via CTCs provide a non-invasive way to obtain important diagnostic, prognostic, and predictive information in patients with cancer.
Collapse
|
96
|
Cheng J, Liu Y, Zhao Y, Zhang L, Zhang L, Mao H, Huang C. Nanotechnology-Assisted Isolation and Analysis of Circulating Tumor Cells on Microfluidic Devices. MICROMACHINES 2020; 11:E774. [PMID: 32823926 PMCID: PMC7465711 DOI: 10.3390/mi11080774] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Jie Cheng
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Liu
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Lina Zhang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China;
| | - Lingqian Zhang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
| | - Chengjun Huang
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China; (J.C.); (Y.L.); (Y.Z.); (L.Z.); (H.M.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
97
|
Clark C, Turiello R, Cotton R, Landers JP. Analytical approaches to differential extraction for sexual assault evidence. Anal Chim Acta 2020; 1141:230-245. [PMID: 33248657 DOI: 10.1016/j.aca.2020.07.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022]
Abstract
Many forensic laboratories face growing demands for the processing of DNA evidence from sexual assault investigations. In these cases, evidence collected from the crime scene or from the victim in the form of a Sexual Assault and Evidence Collection Kit (SAECK) typically contains a mixture of cells from at least two donors. Isolation of DNA contributions to link a sample to an alleged offender requires precise chemical treatment of each sample with the goal of separating epithelial cells from non-sperm cells. Currently, the vast majority of laboratories employ differential chemical lysis methods that require lengthy incubations and several manual steps, preventing complete automation. Numerous alternative methods for the differential extraction (DE) of sexual assault evidence have been developed to provide a solution to the growing backlog of samples observed in the US and other countries. Here, we will discuss the predominant methodology for the DE of DNA from sexual assault samples and review alternative approaches from literature. We illustrate three criteria that provide a measure of success in performing these types of chemical separations and examine all methods based upon these expectations. We conclude by providing some general insight into the application of DE techniques in forensic laboratories and discuss the potential future directions of alternative technologies.
Collapse
Affiliation(s)
- Charles Clark
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Rachelle Turiello
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States.
| | - Robin Cotton
- Department of Pathology, University of Virginia Health Science Center, Charlottesville, VA, United States
| | - James P Landers
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States; Department of Forensic Science, Boston University, Boston, MA, United States; Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
98
|
Chu PY, Hsieh CH, Wu MH. The Combination of Immunomagnetic Bead-Based Cell Isolation and Optically Induced Dielectrophoresis (ODEP)-Based Microfluidic Device for the Negative Selection-Based Isolation of Circulating Tumor Cells (CTCs). Front Bioeng Biotechnol 2020; 8:921. [PMID: 32903713 PMCID: PMC7438881 DOI: 10.3389/fbioe.2020.00921] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/17/2020] [Indexed: 01/19/2023] Open
Abstract
Negative selection-based circulating tumor cell (CTC) isolation is able to harvest viable, label-free, and clinically meaningful CTCs from the cancer patients' blood. Nevertheless, its main shortcoming is its inability to isolate high-purity CTCs, restricting subsequent CTC-related analysis. To address this issue, this study proposed a two-step optically-induced dielectrophoresis (ODEP) cell manipulation to process the cell sample harvested by negative selection-/immunomagnetic microbeads-based CTC isolation. The working mechanism is that the ODEP force acting on the cells with and without magnetic microbeads binding is different. Accordingly, the use of ODEP cell manipulation in a microfluidic system was designed to first separate and then isolate the cancer cells from other magnetic microbead-bound cells. Immunofluorescent microscopic observation and ODEP cell manipulation were then performed to refine the purity of the cancer cells. In this study, the optimum operating conditions for effective cell isolation were determined experimentally. The results revealed that the presented method was able to further refine the purity of cancer cell in the sample obtained after negative selection-based CTC isolation with high cell purity (81.6~86.1%). Overall, this study proposed the combination of immunomagnetic bead-based cell isolation and ODEP cell manipulation for the negative selection-based isolation of CTCs.
Collapse
Affiliation(s)
- Po-Yu Chu
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Hsun Hsieh
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital (Linkou), Taoyuan City, Taiwan
- Division of Haematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Min-Hsien Wu
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital (Linkou), Taoyuan City, Taiwan
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
99
|
Nasiri R, Shamloo A, Akbari J, Tebon P, R. Dokmeci M, Ahadian S. Design and Simulation of an Integrated Centrifugal Microfluidic Device for CTCs Separation and Cell Lysis. MICROMACHINES 2020; 11:E699. [PMID: 32698447 PMCID: PMC7407509 DOI: 10.3390/mi11070699] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/02/2023]
Abstract
Separation of circulating tumor cells (CTCs) from blood samples and subsequent DNA extraction from these cells play a crucial role in cancer research and drug discovery. Microfluidics is a versatile technology that has been applied to create niche solutions to biomedical applications, such as cell separation and mixing, droplet generation, bioprinting, and organs on a chip. Centrifugal microfluidic biochips created on compact disks show great potential in processing biological samples for point of care diagnostics. This study investigates the design and numerical simulation of an integrated microfluidic device, including a cell separation unit for isolating CTCs from a blood sample and a micromixer unit for cell lysis on a rotating disk platform. For this purpose, an inertial microfluidic device was designed for the separation of target cells by using contraction-expansion microchannel arrays. Additionally, a micromixer was incorporated to mix separated target cells with the cell lysis chemical reagent to dissolve their membranes to facilitate further assays. Our numerical simulation approach was validated for both cell separation and micromixer units and corroborates existing experimental results. In the first compartment of the proposed device (cell separation unit), several simulations were performed at different angular velocities from 500 rpm to 3000 rpm to find the optimum angular velocity for maximum separation efficiency. By using the proposed inertial separation approach, CTCs, were successfully separated from white blood cells (WBCs) with high efficiency (~90%) at an angular velocity of 2000 rpm. Furthermore, a serpentine channel with rectangular obstacles was designed to achieve a highly efficient micromixer unit with high mixing quality (~98%) for isolated CTCs lysis at 2000 rpm.
Collapse
Affiliation(s)
- Rohollah Nasiri
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
| | - Javad Akbari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
100
|
Vafaei S, Roudi R, Madjd Z, Aref AR, Ebrahimi M. Potential theranostics of circulating tumor cells and tumor-derived exosomes application in colorectal cancer. Cancer Cell Int 2020; 20:288. [PMID: 32655320 PMCID: PMC7339440 DOI: 10.1186/s12935-020-01389-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background At the present time, colorectal cancer (CRC) is still known as a disease with a high mortality rate. Theranostics are flawless scenarios that link diagnosis with therapy, including precision medicine as a critical platform that relies on the development of biomarkers particularly “liquid biopsy”. Circulating tumor cells (CTCs) and tumor-derived exosomes (TDEs) in a liquid biopsy approach are of substantial importance in comparison with traditional ones, which cannot generally be performed to determine the dynamics of the tumor due to its wide restriction of range. Thus, recent attempts has shifted towards minimally noninvasive methods. Main text CTCs and TDEs, as significant signals emitted from the tumor microenvironment, which are also detectable in the blood, prove themselves to be promising novel biomarkers for cancer diagnosis, prognosis, and treatment response prediction. The therapeutic potential of them is still limited, and studies are at its infancy. One of the major challenges for the implementation of CTCs and TDEs which are new trends in translational medicine is the development of isolation and characterization; a standardizable approach. This review highlights and discusses the current challenges to find the bio fluids application in CRC early detection and clinical management. Conclusion Taken together, CTCs and TDEs as silent drivers of metastasis can serve in the management of cancer patient treatment and it is of the upmost importance to expand our insight into this subject. However, due to the limited data available from clinical trials, further validations are required before addressing their putative application in oncology.![]()
Collapse
Affiliation(s)
- Somayeh Vafaei
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|