51
|
Cell immunocapture microfluidic chip based on high-affinity recombinant protein binders. Biosens Bioelectron 2021; 172:112784. [PMID: 33161292 DOI: 10.1016/j.bios.2020.112784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/30/2020] [Accepted: 10/30/2020] [Indexed: 12/29/2022]
Abstract
Cell immunocapture microfluidic devices represent a rapidly developing field with many potential applications in medical diagnostics. The core of such approach lies in the cell binding to antibody coated surfaces through their surface receptors. Here we show, that the small recombinant protein binders (PBs) can be used for this purpose as well, with the advantage of their constructional flexibility, possibility of fusion with range of tags and cheap mass production. For this purpose, two different PBs derived from Albumin Binding Domain (ABDwt) of streptococcal protein G, so called REX and ARS ligands with proved high affinity and selectivity to the human interleukin-23 (IL-23R) and IL-17 receptor A were used. Four PBs variants recognizing two different epitopes on two different receptors and two PBs variants binding to the same epitope on one receptor but having different peptide spacer with Avitag sequence necessary for their immobilization on sensor surface were tested for cell-capture efficiency. The glass microfluidic Y-system with planar immunocapture channel working in so-called stop-flow dynamic regime was designed. Up to 60-74% immunocapture efficiency of model THP-1 cells on REX/ARS surfaces and practically no cell binding on control ABDwt surfaces was achieved. Moreover, the specific immunocapture of THP-1 cells from mixture with IL-17RA negative DU-145 cells was demonstrated. We discuss the role of the epitope, affinity and immobilization spacer of PBs as well as the influence of stop-flow dynamic regime on the effectivity of THP-1 cell immunocapture. Results can be further exploited in design of microfluidic devices for rare cells immunocapture.
Collapse
|
52
|
Pang TCY, Po JW, Becker TM, Goldstein D, Pirola RC, Wilson JS, Apte MV. Circulating tumour cells in pancreatic cancer: A systematic review and meta-analysis of clinicopathological implications. Pancreatology 2021; 21:103-114. [PMID: 33309014 DOI: 10.1016/j.pan.2020.11.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The detection and quantification of circulating tumour cells (CTCs) in pancreatic cancer (PC) has the potential to provide prognostic information. The aim of this review was to provide an overview of the literature surrounding CTCs in PC. METHODS A systematic literature review on CTCs in PC between 2005-2020 was performed. Data based on peripheral vein samples were used to determine the positivity rate of CTCs, their prognostic significance and their relative numbers compared to portal vein (PV) samples. RESULTS The overall CTC detection rate in forty-four articles was 65% (95%CI: 55-75%). Detection rate for CellSearch was 26% (95%CI: 14-38%), which was lower than for both filtration and microfluidic techniques. In nine studies with >50 patients, overall survival was worse with CTC positivity (HR 1.82; 95%CI: 1.61-2.05). Five of seven studies which described PV CTC collection provided patient-level data. PV CTC yield was 7.7-fold (95%CI 1.35-43.9) that of peripheral blood. CONCLUSIONS CTCs were detected in the peripheral circulation of most patients with PC and may be related to prognosis and disease stage. PV blood contains more CTCs than peripheral blood sampling. This review points to the maturation of techniques of CTC enrichment, and its evidence base for eventual clinical deployment.
Collapse
Affiliation(s)
- Tony C Y Pang
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Joseph W Po
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Therese M Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia
| | - David Goldstein
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Romano C Pirola
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia.
| |
Collapse
|
53
|
Dong J, Zhang RY, Sun N, Hu J, Smalley MD, Zhou A, Yue H, Rothermich W, Chen M, Chen J, Ye J, Teng PC, Qi D, Toretsky JA, Tomlinson JS, Li M, Weiss PS, Jonas SJ, Federman N, Wu L, Zhao M, Tseng HR, Zhu Y. Coupling Nanostructured Microchips with Covalent Chemistry Enables Purification of Sarcoma-Derived Extracellular Vesicles for Downstream Functional Studies. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2003237. [PMID: 34220409 PMCID: PMC8248519 DOI: 10.1002/adfm.202003237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Indexed: 05/18/2023]
Abstract
Tumor-derived extracellular vesicles (EVs) play essential roles in intercellular communication during tumor growth and metastatic evolution. Currently, little is known about the possible roles of tumor-derived EVs in sarcoma because the lack of specific surface markers makes it technically challenging to purify sarcoma-derived EVs. In this study, a specific purification system is developed for Ewing sarcoma (ES)-derived EVs by coupling covalent chemistry-mediated EV capture/ release within a nanostructure-embedded microchip. The purification platform-ES-EV Click Chip-takes advantage of specific anti-LINGO-1 recognition and sensitive click chemistry-mediated EV capture, followed by disulfide cleavage-driven EV release. Since the device is capable of specific and efficient purification of intact ES EVs with high purity, ES-EV Click Chip is ideal for conducting downstream functional studies of ES EVs. Absolute quantification of the molecular hallmark of ES (i.e., EWS rearrangements) using reverse transcription Droplet Digital PCR enables specific quantification of ES EVs. The purified ES EVs can be internalized by recipient cells and transfer their mRNA cargoes, exhibiting their biological intactness and potential role as biological shuttles in intercellular communication.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Ryan Y Zhang
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Na Sun
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Junhui Hu
- Department of Molecular and Medical Pharmacology David Geffen School of Medicine UCLA 650 Charles E Young Dr., Los Angeles, CA 90095, USA
| | - Matthew D Smalley
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Anqi Zhou
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Hua Yue
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Winston Rothermich
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Mengxiang Chen
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jiayuan Chen
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jinglei Ye
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Pai-Chi Teng
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Dongping Qi
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jeffrey A Toretsky
- Departments of Oncology and Pediatrics Georgetown University 3970 Reservoir Rd NW, Washington, DC 20057, USA
| | - James S Tomlinson
- Department of Surgery UCLA 200 Medical Plaza, Los Angeles, CA 90024, USA
| | - Mengyuan Li
- Beijing National Laboratory for Molecular Sciences MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering College of Chemistry and Molecular Engineering Peking University 202 Chengfu Road, Haidian District, Beijing 100871, China
| | - Paul S Weiss
- California NanoSystems Institute Departments of Chemistry and Biochemistry Bioengineering, and Materials Science and Engineering UCLA 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Steven J Jonas
- Department of Pediatrics Ronald Reagan UCLA Medical Center UCLA Mattel Children's Hospital 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Noah Federman
- Department of Pediatrics Ronald Reagan UCLA Medical Center UCLA Mattel Children's Hospital 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Lily Wu
- Department of Molecular and Medical Pharmacology David Geffen School of Medicine UCLA 650 Charles E Young Dr., Los Angeles, CA 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering College of Chemistry and Molecular Engineering Peking University 202 Chengfu Road, Haidian District, Beijing 100871, China
| | - Hsian-Rong Tseng
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute Crump Institute for Molecular Imaging Department of Molecular and Medical Pharmacology University of California Los Angeles (UCLA) 570 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
54
|
Ozensoy Guler O, Supuran CT, Capasso C. Carbonic anhydrase IX as a novel candidate in liquid biopsy. J Enzyme Inhib Med Chem 2020; 35:255-260. [PMID: 31790601 PMCID: PMC6896409 DOI: 10.1080/14756366.2019.1697251] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 12/15/2022] Open
Abstract
Among the diagnostic techniques for the identification of tumour biomarkers, the liquid biopsy is considered one that offers future research on precision diagnosis and treatment of tumours in a non-invasive manner. The approach consists of isolating tumor-derived components, such as circulating tumour cells (CTC), tumour cell-free DNA (ctDNA), and extracellular vesicles (EVs), from the patient peripheral blood fluids. These elements constitute a source of genomic and proteomic information for cancer treatment. Within the tumour-derived components of the body fluids, the enzyme indicated with the acronym CA IX and belonging to the superfamily of carbonic anhydrases (CA, EC 4.2.1.1) is a promising aspirant for checking tumours. CA IX is a transmembrane-CA isoform that is strongly overexpressed in many cancers being not much diffused in healthy tissues except the gastrointestinal tract. Here, it is summarised the role of CA IX as tumour-associated protein and its putative relationship in liquid biopsyfor diagnosing and monitoring cancer progression.
Collapse
Affiliation(s)
- Ozen Ozensoy Guler
- Department of Medical Biology, Faculty of Medicine, Yildirim Beyazit University, Ankara, Turkey
| | - Claudiu. T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Firenze, Italy
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, Institute of Biosciences and Bioresources, CNR, Napoli, Italy
| |
Collapse
|
55
|
Lee J, Kwak B. Simultaneous on-chip isolation and characterization of circulating tumor cell sub-populations. Biosens Bioelectron 2020; 168:112564. [PMID: 32892118 DOI: 10.1016/j.bios.2020.112564] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 12/24/2022]
Abstract
The diagnosis of tumor metastasis using circulating tumor cells (CTCs) has been considered an important developmental target for several decades but remains a formidable challenge because of the rarity and heterogeneity of CTCs. Additional downstream analysis is required after isolating CTCs on-chip for subtype verification. To solve those problems, we have developed microfluidic based integrated system which uses magnetic field gradient and immune-fluorescence differences to on-chip isolation and discrimination of CTCs simultaneously. The system presented in the present study can isolate CTCs with an efficiency of >99% by utilizing magnetic nanoparticles conjugated to CTC membranes. Furthermore, the statuses of three biomarkers can be determined on-chip simultaneously. The devised microfluidic system can differentiate eight different subtypes of heterogenic CTCs by on-chip isolation and based on the statuses of three biomarkers (HER2, ER, and PR) which are critical variables to five-year overall survivals for breast cancer patients.
Collapse
Affiliation(s)
- Jaehun Lee
- Korea Institute of Machinery and Materials, Daegu Research Center for Medical Devices and Rehab. Engineering, Department of Medical Device, 330 Techno Sunhwan-ro, Yuga-eup, Dalsung-gun, Daegu, 42994, Republic of Korea; Kyungpook National University, College of IT Engineering, School of Electronics Engineering, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea
| | - Bongseop Kwak
- Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
56
|
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: 57] [Impact Index Per Article: 11.4] [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
|
57
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
58
|
Garcia-Cordero JL, Maerkl SJ. Microfluidic systems for cancer diagnostics. Curr Opin Biotechnol 2020; 65:37-44. [DOI: 10.1016/j.copbio.2019.11.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
|
59
|
Sun N, Lee YT, Zhang RY, Kao R, Teng PC, Yang Y, Yang P, Wang JJ, Smalley M, Chen PJ, Kim M, Chou SJ, Bao L, Wang J, Zhang X, Qi D, Palomique J, Nissen N, Han SHB, Sadeghi S, Finn RS, Saab S, Busuttil RW, Markovic D, Elashoff D, Yu HH, Li H, Heaney AP, Posadas E, You S, Yang JD, Pei R, Agopian VG, Tseng HR, Zhu Y. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat Commun 2020; 11:4489. [PMID: 32895384 PMCID: PMC7477161 DOI: 10.1038/s41467-020-18311-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
We report a covalent chemistry-based hepatocellular carcinoma (HCC)-specific extracellular vesicle (EV) purification system for early detection of HCC by performing digital scoring on the purified EVs. Earlier detection of HCC creates more opportunities for curative therapeutic interventions. EVs are present in circulation at relatively early stages of disease, providing potential opportunities for HCC early detection. We develop an HCC EV purification system (i.e., EV Click Chips) by synergistically integrating covalent chemistry-mediated EV capture/release, multimarker antibody cocktails, nanostructured substrates, and microfluidic chaotic mixers. We then explore the translational potential of EV Click Chips using 158 plasma samples of HCC patients and control cohorts. The purified HCC EVs are subjected to reverse-transcription droplet digital PCR for quantification of 10 HCC-specific mRNA markers and computation of digital scoring. The HCC EV-derived molecular signatures exhibit great potential for noninvasive early detection of HCC from at-risk cirrhotic patients with an area under receiver operator characteristic curve of 0.93 (95% CI, 0.86 to 1.00; sensitivity = 94.4%, specificity = 88.5%).
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 215123, Suzhou, P.R. China
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Ryan Y Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Rueihung Kao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Pai-Chi Teng
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Yingying Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Peng Yang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Jasmine J Wang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Pin-Jung Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Minhyung Kim
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Shih-Jie Chou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Lirong Bao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Jing Wang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Xinyue Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Juvelyn Palomique
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Nicolas Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steven-Huy B Han
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Saeed Sadeghi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Richard S Finn
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Sammy Saab
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Ronald W Busuttil
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Daniela Markovic
- Department of Medicine, Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - David Elashoff
- Department of Medicine, Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Hsiao-Hua Yu
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, 11529, Taipei, Taiwan
| | - Huiying Li
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Anthony P Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Edwin Posadas
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ju Dong Yang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Digestive and Liver Diseases, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 215123, Suzhou, P.R. China.
| | - Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
60
|
Chen K, Amontree J, Varillas J, Zhang J, George TJ, Fan ZH. Incorporation of lateral microfiltration with immunoaffinity for enhancing the capture efficiency of rare cells. Sci Rep 2020; 10:14210. [PMID: 32848184 PMCID: PMC7450051 DOI: 10.1038/s41598-020-71041-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/22/2020] [Indexed: 02/03/2023] Open
Abstract
The methods for isolating rare cells such as circulating tumor cells (CTCs) can be generally classified into two categories: those based on physical properties (e.g., size) and methods based on biological properties (e.g., immunoaffinity). CellSearch, the only FDA-approved method for the CTC-based cancer prognosis, relies on immunoaffinity interactions between CTCs and antibodies immobilized on magnetic particles. Immunoaffinity-based CTC isolation has also been employed in microfluidic devices, which show higher capture efficiency than CellSearch. We report here our investigation of combining size-based microfiltration into a microfluidic device with immunoaffinity for enhanced capture efficiency of CTCs. The device consists of four serpentine main channels, and each channel contains an array of lateral filters that create a two-dimensional flow. The main flow is through the serpentine channel, allowing the majority of the sample to pass by while the secondary flow goes through the lateral filters. The device design is optimized to make all fluid particles interact with filters. The filter sizes range from 24 to 12 µm, being slightly larger than or having similar dimension of CTCs. These filters are immobilized with antibodies specific to CTCs and thus they function as gates, allowing normal blood cells to pass by while forcing the interactions between CTCs and antibodies on the filter surfaces. The hydrodynamic force experienced by a CTC was also studied for optimal experimental conditions to ensure immunoaffinity-enabled cell capture. The device was evaluated by capturing two types of tumor cells spiked in healthy blood or a buffer, and we found that their capture efficiency was between 87.2 and 93.5%. The platform was further validated by isolating CTCs from blood samples of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Kangfu Chen
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jacob Amontree
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL, 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA.
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA.
| |
Collapse
|
61
|
Habli Z, AlChamaa W, Saab R, Kadara H, Khraiche ML. Circulating Tumor Cell Detection Technologies and Clinical Utility: Challenges and Opportunities. Cancers (Basel) 2020; 12:cancers12071930. [PMID: 32708837 PMCID: PMC7409125 DOI: 10.3390/cancers12071930] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/03/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
The potential clinical utility of circulating tumor cells (CTCs) in the diagnosis and management of cancer has drawn a lot of attention in the past 10 years. CTCs disseminate from tumors into the bloodstream and are believed to carry vital information about tumor onset, progression, and metastasis. In addition, CTCs reflect different biological aspects of the primary tumor they originate from, mainly in their genetic and protein expression. Moreover, emerging evidence indicates that CTC liquid biopsies can be extended beyond prognostication to pharmacodynamic and predictive biomarkers in cancer patient management. A key challenge in harnessing the clinical potential and utility of CTCs is enumerating and isolating these rare heterogeneous cells from a blood sample while allowing downstream CTC analysis. That being said, there have been serious doubts regarding the potential value of CTCs as clinical biomarkers for cancer due to the low number of promising outcomes in the published results. This review aims to present an overview of the current preclinical CTC detection technologies and the advantages and limitations of each sensing platform, while surveying and analyzing the published evidence of the clinical utility of CTCs.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Walid AlChamaa
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Raya Saab
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, 77030 TX, USA;
| | - Massoud L. Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
- Correspondence:
| |
Collapse
|
62
|
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: 3.4] [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
|
63
|
Genna A, Vanwynsberghe AM, Villard AV, Pottier C, Ancel J, Polette M, Gilles C. EMT-Associated Heterogeneity in Circulating Tumor Cells: Sticky Friends on the Road to Metastasis. Cancers (Basel) 2020; 12:E1632. [PMID: 32575608 PMCID: PMC7352430 DOI: 10.3390/cancers12061632] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transitions (EMTs) generate hybrid phenotypes with an enhanced ability to adapt to diverse microenvironments encountered during the metastatic spread. Accordingly, EMTs play a crucial role in the biology of circulating tumor cells (CTCs) and contribute to their heterogeneity. Here, we review major EMT-driven properties that may help hybrid Epithelial/Mesenchymal CTCs to survive in the bloodstream and accomplish early phases of metastatic colonization. We then discuss how interrogating EMT in CTCs as a companion biomarker could help refine cancer patient management, further supporting the relevance of CTCs in personalized medicine.
Collapse
Affiliation(s)
- Anthony Genna
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Aline M. Vanwynsberghe
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Amélie V. Villard
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Charles Pottier
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
- Department of Medical Oncology, University Hospital of Liège, 4000 Liège, Belgium
| | - Julien Ancel
- CHU (Centre Hopitalier Universitaire) de Reims, Hôpital Maison Blanche, Service de Pneumologie, 51092 Reims, France;
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
| | - Myriam Polette
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
- CHU de Reims, Hôpital Maison Blanche, Laboratoire de Pathologie, 51092 Reims, France
| | - Christine Gilles
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| |
Collapse
|
64
|
Ashtari B, Shams A, Esmaeilzadeh N, Tanbakooei S, Koruji M, Moghadam MJ, Ansari JM, Moghadam AJ, Shabani R. Separating mouse malignant cell line (EL4) from neonate spermatogonial stem cells utilizing microfluidic device in vitro. Stem Cell Res Ther 2020; 11:191. [PMID: 32448280 PMCID: PMC7245899 DOI: 10.1186/s13287-020-01671-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/25/2020] [Accepted: 04/07/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Some children who have survived cancer will be azoospermic in the future. Performing isolation and purification procedures for spermatogonial stem cells (SSC) is very critical. In this regard, performing the process of decontamination of cancerous cells is the initial step. The major objective of the present study is to separate the malignant EL4 cell line in mice and spermatogonial stem cells in vitro. METHODS The spermatogonial stem cells of sixty neonatal mice were isolated, and the procedure of co-culturing was carried out by EL4 which were classified into 2 major groups: (1) the control group (co-culture in a growth medium) and (2) the group of co-cultured cells which were separated using the microfluidic device. The percentage of cells was assessed using flow cytometry technique and common laboratory technique of immunocytochemistry and finally was confirmed through the laboratory technique of reverse transcription-polymerase chain reaction (RT-PCR). RESULTS The actual percentage of EL4 and SSC after isolation was collected at two outlets: the outputs for the smaller outlet were 0.12% for SSC and 42.14% for EL4, while in the larger outlet, the outputs were 80.38% for SSC and 0.32% for EL4; in the control group, the percentages of cells were 21.44% for SSC and 23.28% for EL4 (based on t test (p ≤ 0.05)). CONCLUSIONS The present study demonstrates that the use of the microfluidic device is effective in separating cancer cells from spermatogonial stem cells.
Collapse
Affiliation(s)
- Behnaz Ashtari
- Shahdad Ronak Commercialization Company, Pasdaran Street, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azar Shams
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Narges Esmaeilzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Tanbakooei
- School of Mechanical Engineering, Iran University of Science & Technology, Tehran, Iran
| | - Morteza Koruji
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- School of Mechanical Engineering, Iran University of Science & Technology, Tehran, Iran
| | | | - Javad Mohajer Ansari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Adel Johari Moghadam
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Ronak Shabani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- School of Mechanical Engineering, Iran University of Science & Technology, Tehran, Iran.
| |
Collapse
|
65
|
Pahattuge TN, Jackson JM, Digamber R, Wijerathne H, Brown V, Witek MA, Perera C, Givens RS, Peterson BR, Soper SA. Visible photorelease of liquid biopsy markers following microfluidic affinity-enrichment. Chem Commun (Camb) 2020; 56:4098-4101. [PMID: 32163053 PMCID: PMC7469076 DOI: 10.1039/c9cc09598e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We detail a heterobifunctional, 7-aminocoumarin photocleavable (PC) linker with unique properties to covalently attach Abs to surfaces and subsequently release them with visible light (400-450 nm). The PC linker allowed rapid (2 min) and efficient (>90%) release of CTCs and EVs without damaging their molecular cargo.
Collapse
Affiliation(s)
- Thilanga N Pahattuge
- Center of BioModular Multi-Scale Systems, Department of Chemistry, University of Kansas, 1567 Irving Hill Rd., Lawrence, KS 66045, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Wang Y, Liu Q, Men T, Liang Y, Niu H, Wang J. A microfluidic system based on the monoclonal antibody BCMab1 specifically captures circulating tumor cells from bladder cancer patients. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:1199-1210. [PMID: 32275489 DOI: 10.1080/09205063.2020.1748332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Circulating bladder tumor cells provide significant information for cancer diagnosis, tumor staging and personalized cancer therapy. Previous studies have reported various methods for capturing circulating tumor cells; however, capturing circulating tumor cells remain a challenge. Here, we present a microfluidic chip with high specificity and capture yields that we refer to as a bladder cancer diagnosis chip. We show that this chip can be used to effectively capture circulating bladder cancer cells based on antibody-BCMab1, a monoclonal antibody that binds to aberrantly glycosylated integrin a3b1. This capture platform is composed of a polydimethylsiloxane (PDMS) chip, whose microchannels are functionalized with biotinylated BCMab1. To change the direction of flow to increase cell-substrate contact, we also introduced a herringbone or chevron channel pattern into the chip. Using this system, we were able to capture bladder cancer cells with high specificity. The capture rates of the bladder cancer diagnosis chip were evaluated at different flow rates and cell concentrations. We found that 90% of the cancer cells were successfully captured at flow rates of 10 μL/min and at various cell concentrations. This highly specific microfluidic chip is a novel tool for bladder cancer diagnosis and offers an opportunity for personalized treatment.
Collapse
Affiliation(s)
- Yunchao Wang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qing Liu
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tongyi Men
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Ye Liang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haitao Niu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianning Wang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
67
|
Lee NJ, Maeng S, Kim HU, Roh YH, Hwang C, Kim J, Hwang KT, Bong KW. Affinity-Enhanced CTC-Capturing Hydrogel Microparticles Fabricated by Degassed Mold Lithography. J Clin Med 2020; 9:E301. [PMID: 31973077 PMCID: PMC7073783 DOI: 10.3390/jcm9020301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Technologies for the detection and isolation of circulating tumor cells (CTCs) are essential in liquid biopsy, a minimally invasive technique for early diagnosis and medical intervention in cancer patients. A promising method for CTC capture, using an affinity-based approach, is the use of functionalized hydrogel microparticles (MP), which have the advantages of water-like reactivity, biologically compatible materials, and synergy with various analysis platforms. In this paper, we demonstrate the feasibility of CTC capture by hydrogel particles synthesized using a novel method called degassed mold lithography (DML). This technique increases the porosity and functionality of the MPs for effective conjugation with antibodies. Qualitative fluorescence analysis demonstrates that DML produces superior uniformity, integrity, and functionality of the MPs, as compared to conventional stop flow lithography (SFL). Analysis of the fluorescence intensity from porosity-controlled MPs by each reaction step of antibody conjugation elucidates that more antibodies are loaded when the particles are more porous. The feasibility of selective cell capture is demonstrated using breast cancer cell lines. In conclusion, using DML for the synthesis of porous MPs offers a powerful method for improving the cell affinity of the antibody-conjugated MPs.
Collapse
Affiliation(s)
- Nak Jun Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea; (N.J.L.); (H.U.K.); (Y.H.R.); (C.H.)
| | - Sejung Maeng
- Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 07061, Korea; (S.M.); (J.K.)
| | - Hyeon Ung Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea; (N.J.L.); (H.U.K.); (Y.H.R.); (C.H.)
| | - Yoon Ho Roh
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea; (N.J.L.); (H.U.K.); (Y.H.R.); (C.H.)
| | - Changhyun Hwang
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea; (N.J.L.); (H.U.K.); (Y.H.R.); (C.H.)
| | - Jongjin Kim
- Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 07061, Korea; (S.M.); (J.K.)
| | - Ki-Tae Hwang
- Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 07061, Korea; (S.M.); (J.K.)
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea; (N.J.L.); (H.U.K.); (Y.H.R.); (C.H.)
| |
Collapse
|
68
|
Li N, Zuo H, Chen L, Liu H, Zhou J, Yao Y, Xu B, Gong H, Weng Y, Hu Q, Song Q, Peng M, Cheng Y. Circulating Tumor Cell Detection In Epithelial Ovarian Cancer Using Dual-Component Antibodies Targeting EpCAM And FRα. Cancer Manag Res 2019; 11:10939-10948. [PMID: 32021417 PMCID: PMC6978676 DOI: 10.2147/cmar.s211455] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/02/2019] [Indexed: 12/25/2022] Open
Abstract
Purpose Circulating tumor cell (CTC) detection methods based on epithelial cell adhesion molecule (EpCAM) have low detection rates in epithelial ovarian cancer (EOC). Meanwhile, folate receptor alpha (FRα) has high expression in EOC cells. We explored the feasibility of combining FRα and EpCAM as CTC capture targets in EOC. Patients and methods EpCAM and FRα antibodies were linked to magnetic nanospheres (MNs) using the principle of carbodiimide chemistry. Blood samples from healthy donor spiked with A2780 ovarian cancer cells were used for detecting the capture rate. Ninety-five blood samples from 30 patients with EOC were used for comparing the positive rate of detection when using anti-EpCAM-MNs alone with that when using combination of anti-EpCAM-MNs and anti-FRα-MNs. Samples from 28 patients initially diagnosed with EOC and 20 patients with ovarian benign disease were used for evaluating the sensitivity and specificity of combination of anti-EpCAM-MNs and anti-FRα-MNs. Results Regression analysis between the number of recovered and that of spiked A2780 cells revealed yEpCAM = 0.535x (R2 = 0.99), yFRα = 0.901x (R2 = 0.99), and yEpCAM+FRα = 0.928x (R2 = 0.99). In mixtures of A2780 and MCF7 cells, the capture rate was 92% using the combination of anti-EpCAM-MNs and anti-FRα-MNs, exceeding the rate when using anti-EpCAM-MNs or anti-FRα-MNs alone by approximately 20% (P < 0.01). The combination of anti-EpCAM-MNs and anti-FRα-MNs showed a significantly increased positive rate of CTC detection in EOC patients compared with anti-EpCAM-MNs alone (χ2 = 14.45, P < 0.001). Sensitivity values were 0.536 and 0.75 and specificity values were 0.9 and 0.85 when using anti-EpCAM-MNs alone and when using the combination of anti-EpCAM-MNs and anti-FRα-MNs, respectively. Conclusion The combination of FRα and EpCAM is feasible as a CTC capture target of CTC detection in patients with EOC.
Collapse
Affiliation(s)
- Na Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hao Zuo
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Luojun Chen
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Huali Liu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Jin Zhou
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yi Yao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Bin Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hongyun Gong
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yiming Weng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Qibin Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
69
|
Liu H, Koch C, Haller A, Joosse SA, Kumar R, Vellekoop MJ, Horst LJ, Keller L, Babayan A, Failla AV, Jensen J, Peine S, Keplinger F, Fuchs H, Pantel K, Hirtz M. Evaluation of Microfluidic Ceiling Designs for the Capture of Circulating Tumor Cells on a Microarray Platform. ACTA ACUST UNITED AC 2019; 4:e1900162. [DOI: 10.1002/adbi.201900162] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Hui‐Yu Liu
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT) 76344 Eggenstein‐Leopoldshafen Germany
| | - Claudia Koch
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Anna Haller
- Institute of Sensor and Actuator SystemsTU Wien 1040 Vienna Austria
| | - Simon A. Joosse
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Ravi Kumar
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT) 76344 Eggenstein‐Leopoldshafen Germany
| | - Michael J. Vellekoop
- Institute for MicrosensorsMicroactuators and Microsystems (IMSAS)Microsystems Center Bremen MCBUniversity of Bremen 28359 Bremen Germany
| | - Ludwig J. Horst
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Laura Keller
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Anna Babayan
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Antonio Virgilio Failla
- Microscopy Imaging Facility (UMIF)University Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Jana Jensen
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Sven Peine
- Department of Transfusion MedicineUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Franz Keplinger
- Institute of Sensor and Actuator SystemsTU Wien 1040 Vienna Austria
| | - Harald Fuchs
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT) 76344 Eggenstein‐Leopoldshafen Germany
- Physical Institute and Center for Nanotechnology (CeNTech)University of Münster 48149 Münster Germany
| | - Klaus Pantel
- Department of Tumor BiologyUniversity Medical Center Hamburg‐Eppendorf 20246 Hamburg Germany
| | - Michael Hirtz
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF)Karlsruhe Institute of Technology (KIT) 76344 Eggenstein‐Leopoldshafen Germany
| |
Collapse
|
70
|
Jiménez-Zenteno AK, Cerf A. Liquid Biopsy Based on Circulating Cancer-Associated Cells: Bridging the Gap from an Emerging Concept to a Mainstream Tool in Precision Medicine. ACTA ACUST UNITED AC 2019; 4:e1900164. [PMID: 32293131 DOI: 10.1002/adbi.201900164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/15/2019] [Indexed: 01/01/2023]
Abstract
The concept of liquid biopsy and the isolation and analysis of circulating biomarkers from blood samples is proposed as a surrogate to solid biopsies and can have the potential to revolutionize the management of patients with cancer. The relevance of circulating tumor cells (CTCs) and the importance of the information they carry is acknowledged by the medical community. But what are the barriers to clinical adoption? This review draws a panorama of the biological implications of CTCs, their physical and biochemical properties, and the current technological bottlenecks for their analysis in relation with the medical needs. Keys and considerations to bridge the technological and clinical gaps that still need to be overcome to be able to introduce CTCs in clinical routine are finally synthesized.
Collapse
Affiliation(s)
| | - Aline Cerf
- Université de Toulouse, CNRS, 7 Avenue du Colonel Roche, 31400, Toulouse, France
| |
Collapse
|
71
|
Kamal M, Saremi S, Klotz R, Iriondo O, Amzaleg Y, Chairez Y, Tulpule V, Lang JE, Kang I, Yu M. PIC&RUN: An integrated assay for the detection and retrieval of single viable circulating tumor cells. Sci Rep 2019; 9:17470. [PMID: 31767951 PMCID: PMC6877641 DOI: 10.1038/s41598-019-53899-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Circulating tumor cells (CTCs) shed from solid tumors can serve as a minimally invasive liquid biopsy for monitoring disease progression. Because CTCs are rare and heterogeneous, their biological properties need to be investigated at the single cell level, which requires efficient ways to isolate and analyze live single CTCs. Current methods for CTC isolation and identification are either performed on fixed and stained cells or need multiple procedures to isolate pure live CTCs. Here, we used the AccuCyte-RareCyte system to develop a Protocol for Integrated Capture and Retrieval of Ultra-pure single live CTCs using Negative and positive selection (PIC&RUN). The positive selection module of PIC&RUN identifies CTCs based on detection of cancer surface markers and exclusion of immune markers. Combined with a two-step cell picking protocol to retrieve ultrapure single CTCs, the positive selection module is compatible for downstream single cell transcriptomic analysis. The negative selection module of PIC&RUN identifies CTCs based on a live cell dye and the absence of immune markers, allowing retrieval of viable CTCs that are suitable for ex vivo culture. This new assay combines the CTC capture and retrieval in one integrated platform, providing a valuable tool for downstream live CTC analyses.
Collapse
Affiliation(s)
- Mohamed Kamal
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
- Department of Zoology, Faculty of Science, University of Benha, Benha, Egypt
| | - Shahin Saremi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
- MS Biotechnology program, California State University Channel Islands, Camarillo, CA, 93012, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Oihana Iriondo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Yonatan Amzaleg
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Yvonne Chairez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Varsha Tulpule
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Julie E Lang
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
- Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Irene Kang
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA.
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, 90033, USA.
| |
Collapse
|
72
|
Lee SJ, Sim TS, Shin HY, Lee J, Kim MY, Sunoo J, Lee JG, Yea K, Kim YZ, van Noort D, Park SK, Kim WH, Park KW, Kim MS. Microslit on a chip: A simplified filter to capture circulating tumor cells enlarged with microbeads. PLoS One 2019; 14:e0223193. [PMID: 31647823 PMCID: PMC6812780 DOI: 10.1371/journal.pone.0223193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/15/2019] [Indexed: 12/11/2022] Open
Abstract
Microchips are widely used to separate circulating tumor cells (CTCs) from whole blood by virtues of sophisticated manipulation for microparticles. Here, we present a chip with an 8 μm high and 27.9 mm wide slit to capture cancer cells bound to 3 μm beads. Apart from a higher purity and recovery rate, the slit design allows for simplified fabrication, easy cell imaging, less clogging, lower chamber pressure and, therefore, higher throughput. The beads were conjugated with anti-epithelial cell adhesion molecules (anti-EpCAM) to selectively bind to breast cancer cells (MCF-7) used to spike the whole blood. The diameter of the cell-bead construct was in average 23.1 μm, making them separable from other cells in the blood. As a result, the cancer cells were separated from 5 mL of whole blood with a purity of 52.0% and a recovery rate of 91.1%, and also we confirmed that the device can be applicable to clinical samples of human breast cancer patients. The simple design with microslit, by eliminating any high-aspect ratio features, is expected to reduce possible defects on the chip and, therefore, more suitable for mass production without false separation outputs.
Collapse
Affiliation(s)
- Seung Joon Lee
- Department of New Biology, DGIST, Daegu, Republic of Korea
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Tae Seok Sim
- Samsung Electronics, Ltd., Maetan3-dong, Youngtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | | | - Jungmin Lee
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Min Young Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Joseph Sunoo
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jeong-Gun Lee
- Samsung Electronics, Ltd., Maetan3-dong, Youngtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Danny van Noort
- Department of New Biology, DGIST, Daegu, Republic of Korea
- Division of Biotechnology, IFM, Linköping University, Linköping, Sweden
| | - Soo Kyung Park
- CytoDx, Pangyo-ro, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Woon-Hae Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Kyun Woo Park
- Daejeon Wellness Hospital, Beon-gil, Dongseo-daero, Daedeok-gu, Daejeon, Republic of Korea
| | - Minseok S. Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
- Translational Responsive Medicine Center, DGIST, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
73
|
Integration of Hierarchical Micro-/Nanostructures in a Microfluidic Chip for Efficient and Selective Isolation of Rare Tumor Cells. MICROMACHINES 2019; 10:mi10100698. [PMID: 31615080 PMCID: PMC6843196 DOI: 10.3390/mi10100698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022]
Abstract
Circulating tumor cells (CTCs) are important clinical markers for both cancer early diagnosis and prognosis. Various techniques have been developed in the past decade to isolate and quantify these cells from the blood while microfluidic technology attracts significant attention due to better controlled microenvironment. When combined with advanced nanotechnologies, CTC isolation performance in microfluidic devices can be further improved. In this article, by extending the wavy-herringbone concept developed earlier in our team, we prepared a hierarchical microfluidic chip by introducing a uniform coating of nanoparticles with anti-epithelial cell adhesion molecule (EpCAM) on wavy microgrooves. This hierarchical structured platform not only maintains the capture purity of the wavy-herringbone structure but improves the capture efficiency thanks to the larger surface area to volume ratio brought by nanoparticles. Our results demonstrated a capture efficiency of almost 100% at a low shear rate of 60/s. Even at a higher shear rate of 400/s, the hierarchical micro/nanostructures demonstrated an enhancement of up to ~3-fold for capture efficiency (i.e., 70%) and ~1.5-fold for capture purity (i.e., 68%), compared to wavy-herringbone structures without nanoparticle coating. With these promising results, this hierarchical structured platform represents a technological advancement for CTC isolation and cancer care.
Collapse
|
74
|
Cheng SB, Chen MM, Wang YK, Sun ZH, Xie M, Huang WH. Current techniques and future advance of microfluidic devices for circulating tumor cells. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
75
|
Lee J, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol 2019; 13:1623-1650. [PMID: 31243883 PMCID: PMC6670020 DOI: 10.1002/1878-0261.12537] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/07/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Reliable biomarkers are required to evaluate and manage pancreatic ductal adenocarcinoma. Circulating tumor cells and circulating tumor DNA are shed into blood and can be relatively easily obtained from minimally invasive liquid biopsies for serial assays and characterization, thereby providing a unique potential for early diagnosis, forecasting disease prognosis, and monitoring of therapeutic response. In this review, we provide an overview of current technologies used to detect circulating tumor cells and circulating tumor DNA and describe recent advances regarding the multiple clinical applications of liquid biopsy in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jee‐Soo Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Sung Sup Park
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Young Kyung Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineHallym University College of MedicineAnyangKorea
| | - Jeffrey A. Norton
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | | |
Collapse
|
76
|
Dong J, Jan YJ, Cheng J, Zhang RY, Meng M, Smalley M, Chen PJ, Tang X, Tseng P, Bao L, Huang TY, Zhou D, Liu Y, Chai X, Zhang H, Zhou A, Agopian VG, Posadas EM, Shyue JJ, Jonas SJ, Weiss PS, Li M, Zheng G, Yu HH, Zhao M, Tseng HR, Zhu Y. Covalent chemistry on nanostructured substrates enables noninvasive quantification of gene rearrangements in circulating tumor cells. SCIENCE ADVANCES 2019; 5:eaav9186. [PMID: 31392269 PMCID: PMC6669017 DOI: 10.1126/sciadv.aav9186] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/26/2019] [Indexed: 05/07/2023]
Abstract
Well-preserved mRNA in circulating tumor cells (CTCs) offers an ideal material for conducting molecular profiling of tumors, thereby providing a noninvasive diagnostic solution for guiding treatment intervention and monitoring disease progression. However, it is technically challenging to purify CTCs while retaining high-quality mRNA.Here, we demonstrate a covalent chemistry-based nanostructured silicon substrate ("Click Chip") for CTC purification that leverages bioorthogonal ligation-mediated CTC capture and disulfide cleavage-driven CTC release. This platform is ideal for CTC mRNA assays because of its efficient, specific, and rapid purification of pooled CTCs, enabling downstream molecular quantification using reverse transcription Droplet Digital polymerase chain reaction. Rearrangements of ALK/ROS1 were quantified using CTC mRNA and matched with those identified in biopsy specimens from 12 patients with late-stage non-small cell lung cancer. Moreover, CTC counts and copy numbers of ALK/ROS1 rearrangements could be used together for evaluating treatment responses and disease progression.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu Jen Jan
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ju Cheng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Meng Meng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin-Jung Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xinghong Tang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lirong Bao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tzu-Yang Huang
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Dongjing Zhou
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Yupin Liu
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Xiaoshu Chai
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Haibo Zhang
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Anqi Zhou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vatche G. Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Edwin M. Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jing-Jong Shyue
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Steven J. Jonas
- Department of Pediatrics, David Geffen School of Medicine, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, and Children’s Discovery and Innovation Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul S. Weiss
- California NanoSystems Institute, Departments of Chemistry and Biochemistry and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mengyuan Li
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Guangjuan Zheng
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China
| | - Hsiao-hua Yu
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
77
|
Kamyabi N, Huang J, Lee JJ, Bernard V, Semaan A, Stephens B, Hurd MW, Vanapalli SA, Maitra A, Guerrero PA. A microfluidic device for label-free isolation of tumor cell clusters from unprocessed blood samples. BIOMICROFLUIDICS 2019; 13:044111. [PMID: 31462955 PMCID: PMC6701978 DOI: 10.1063/1.5111888] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/01/2019] [Indexed: 05/05/2023]
Abstract
Primary cancers disseminate both single circulating tumor cells (CTCs) and CTC "clusters," the latter of which have been shown to demonstrate greater metastatic propensity and adverse impact on prognosis. Many devices developed to isolate single CTCs also capture CTC clusters, but there is translational potential for a platform specifically designed to isolate CTC clusters. Herein, we introduce our microfluidic device for isolating CTC clusters ("Microfluidic Isolation of CTC Clusters" or MICC), which is equipped with ∼10 000 trap chambers that isolate tumor cell clusters based on their large sizes and dynamic force balance against a pillar obstacle in the trap chamber. Whole blood is injected, followed by a wash step to remove blood cells and a final backflush to release intact clusters for downstream analysis. Using clusters from tumor cell-line and confocal microscopy, we verified the ability of the MICC platform to specifically capture tumor cell clusters in the trap chambers. Our flow rate optimization experiments identified 25 μl/min for blood injection, 100 μl/min as wash flow rate, and 300 μl/min as the release flow rate - indicating that 1 ml of whole blood can be processed in less than an hour. Under these optimal flow conditions, we assessed the MICC platform's capture and release performance using blood samples spiked with different concentrations of clusters, revealing a capture efficiency of 66%-87% and release efficiency of 76%-90%. The results from our study suggest that the MICC platform has the potential to isolate CTC clusters from cancer patient blood, enabling it for clinical applications in cancer management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Siva A. Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| | | | | |
Collapse
|
78
|
Insights on CTC Biology and Clinical Impact Emerging from Advances in Capture Technology. Cells 2019; 8:cells8060553. [PMID: 31174404 PMCID: PMC6627072 DOI: 10.3390/cells8060553] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/01/2023] Open
Abstract
Circulating tumor cells (CTCs) and circulating tumor microemboli (CTM) have been shown to correlate negatively with patient survival. Actual CTC counts before and after treatment can be used to aid in the prognosis of patient outcomes. The presence of circulating tumor materials (CTMat) can advertise the presence of metastasis before clinical presentation, enabling the early detection of relapse. Importantly, emerging evidence is indicating that cancer treatments can actually increase the incidence of CTCs and metastasis in pre-clinical models. Subsequently, the study of CTCs, their biology and function are of vital importance. Emerging technologies for the capture of CTC/CTMs and CTMat are elucidating vitally important biological and functional information that can lead to important alterations in how therapies are administered. This paves the way for the development of a "liquid biopsy" where treatment decisions can be informed by information gleaned from tumor cells and tumor cell debris in the blood.
Collapse
|
79
|
Chen K, Dopico P, Varillas J, Zhang J, George TJ, Fan ZH. Integration of Lateral Filter Arrays with Immunoaffinity for Circulating-Tumor-Cell Isolation. Angew Chem Int Ed Engl 2019; 58:7606-7610. [PMID: 30958635 PMCID: PMC6534423 DOI: 10.1002/anie.201901412] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Indexed: 01/06/2023]
Abstract
Circulating tumor cells (CTCs) are an important biomarker for cancer prognosis and treatment monitoring. However, the heterogeneity of the physical and biological properties of CTCs limits the efficiency of various approaches used to isolate small numbers of CTCs from billions of normal blood cells. To address this challenge, we developed a lateral filter array microfluidic (LFAM) device to integrate size-based separation with immunoaffinity-based CTC isolation. The LFAM device consists of a serpentine main channel, through which most of a sample passes, and an array of lateral filters for CTC isolation. The unique device design produces a two-dimensional flow, which reduces nonspecific, geometric capture of normal cells as typically observed in vertical filters. The LFAM device was further functionalized by immobilizing antibodies that are specific to the target cells. The resulting devices captured pancreatic cancer cells spiked in blood samples with (98.7±1.2) % efficiency and were used to isolate CTCs from patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Kangfu Chen
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Pablo Dopico
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
80
|
Kim TJ, Moon HW, Kang S, Yang J, Hong SH, Lee JY, Ha US. Urovysion FISH Could Be Effective and Useful Method to Confirm the Identity of Cultured Circulating Tumor Cells from Bladder Cancer Patients. J Cancer 2019; 10:3259-3266. [PMID: 31289598 PMCID: PMC6603370 DOI: 10.7150/jca.30079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/28/2019] [Indexed: 12/17/2022] Open
Abstract
Objective: To explore whether cultured CTC from bladder-cancer patients originate from bladder cancer and share chromosomal abnormalities, by means of a fluorescence in situ hybridization (FISH) test. Methods: A total of 15 ml of blood was collected from the patients with bladder cancer before treatment began. Isolated CTCs were divided into 5 ml for CTC enumeration and 10 ml for CTC culture. CTCs were counted by immunofluorescent staining with vimentin, cytokeratin, CD45, and DAPI antibody. CTCs were cultured using isolated CTCs in 96-well plates of Mesenchymal Stem Cell Growth Medium for 16~18 days. The resulting cultured CTCs from 20 men with bladder cancer were analyzed by Urovysion FISH. Results: Common gains were on chromosome 3, 7, and 17 in 20 (74.1%), 14 (51.9%), and 20 (74.1%) of 27 patients, respectively. Polysomy was detected on chromosomes 3 and 7 in 9 patients (33.3%). Polysomy involving two chromosomes was observed in 16 (59.3%, chromosome 3 and 17) and 9 patients (33.3%, chromosome 7 and 17) in the same cell. Among the patients with isolated gain, 17 (63.0%) met the positive criteria for Urovysion FISH. Homozygous deletion of 9p21, 5 (18.5%) involved more than 12 cells. Among the different patient cohorts, positive results based on the Urovysion criteria were obtained in cultured CTCs derived from 19 (70.4%) patients. Conclusion: Application of FISH Urovysion to cultured CTCs from bladder cancer could be an effective first step to confirm their origin and sharing of chromosomal abnormalities.
Collapse
Affiliation(s)
- Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyong Woo Moon
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungmin Kang
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jonghyup Yang
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - U-Syn Ha
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
81
|
Bankó P, Lee SY, Nagygyörgy V, Zrínyi M, Chae CH, Cho DH, Telekes A. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol 2019; 12:48. [PMID: 31088479 PMCID: PMC6518774 DOI: 10.1186/s13045-019-0735-4] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
The importance of early cancer diagnosis and improved cancer therapy has been clear for years and has initiated worldwide research towards new possibilities in the care strategy of patients with cancer using technological innovations. One of the key research fields involves the separation and detection of circulating tumor cells (CTC) because of their suggested important role in early cancer diagnosis and prognosis, namely, providing easy access by a liquid biopsy from blood to identify metastatic cells before clinically detectable metastasis occurs and to study the molecular and genetic profile of these metastatic cells. Provided the opportunity to further progress the development of technology for treating cancer, several CTC technologies have been proposed in recent years by various research groups and companies. Despite their potential role in cancer healthcare, CTC methods are currently mainly used for research purposes, and only a few methods have been accepted for clinical application because of the difficulties caused by CTC heterogeneity, CTC separation from the blood, and a lack of thorough clinical validation. Therefore, the standardization and clinical application of various developed CTC technologies remain important subsequent necessary steps. Because of their suggested future clinical benefits, we focus on describing technologies using whole blood samples without any pretreatment and discuss their advantages, use, and significance. Technologies using whole blood samples utilize size-based, immunoaffinity-based, and density-based methods or combinations of these methods as well as positive and negative enrichment during separation. Although current CTC technologies have not been truly implemented yet, they possess high potential as future clinical diagnostic techniques for the individualized therapy of patients with cancer. Thus, a detailed discussion of the clinical suitability of these new advanced technologies could help prepare clinicians for the future and can be a foundation for technologies that would be used to eliminate CTCs in vivo.
Collapse
Affiliation(s)
- Petra Bankó
- Department of Biochemical Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Sun Young Lee
- Department of Radiation Oncology, Chonbuk National University Hospital, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | | | - Miklós Zrínyi
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Chang Hoon Chae
- Laboratory of Nanochemistry, Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Dong Hyu Cho
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical, Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
- Department of Obstetrics and Gynecology, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - András Telekes
- Department of Oncology, St. Lazarus Hospital, Salgótarján, Hungary
| |
Collapse
|
82
|
Chen K, Dopico P, Varillas J, Zhang J, George TJ, Fan ZH. Integration of Lateral Filter Arrays with Immunoaffinity for Circulating‐Tumor‐Cell Isolation. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201901412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Kangfu Chen
- Interdisciplinary Microsystems Group (IMG)Department of Mechanical and Aerospace EngineeringUniversity of Florida P.O. BOX 116250 Gainesville FL 32611 USA
| | - Pablo Dopico
- Interdisciplinary Microsystems Group (IMG)Department of Mechanical and Aerospace EngineeringUniversity of Florida P.O. BOX 116250 Gainesville FL 32611 USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of Florida Gainesville FL 32611 USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group (IMG)Department of Mechanical and Aerospace EngineeringUniversity of Florida P.O. BOX 116250 Gainesville FL 32611 USA
| | - Thomas J. George
- Department of MedicineUniversity of Florida Gainesville FL 32610 USA
| | - Z. Hugh Fan
- Interdisciplinary Microsystems Group (IMG)Department of Mechanical and Aerospace EngineeringUniversity of Florida P.O. BOX 116250 Gainesville FL 32611 USA
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of Florida Gainesville FL 32611 USA
| |
Collapse
|
83
|
Dong J, Zhang RY, Sun N, Smalley M, Wu Z, Zhou A, Chou SJ, Jan YJ, Yang P, Bao L, Qi D, Tang X, Tseng P, Hua Y, Xu D, Kao R, Meng M, Zheng X, Liu Y, Vagner T, Chai X, Zhou D, Li M, Chiou SH, Zheng G, Di Vizio D, Agopian VG, Posadas E, Jonas SJ, Ju SP, Weiss PS, Zhao M, Tseng HR, Zhu Y. Bio-Inspired NanoVilli Chips for Enhanced Capture of Tumor-Derived Extracellular Vesicles: Toward Non-Invasive Detection of Gene Alterations in Non-Small Cell Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2019; 11:13973-13983. [PMID: 30892008 PMCID: PMC6545291 DOI: 10.1021/acsami.9b01406] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Tumor-derived extracellular vesicles (EVs) present in bodily fluids are emerging liquid biopsy markers for non-invasive cancer diagnosis and treatment monitoring. Because the majority of EVs in circulation are not of tumor origin, it is critical to develop new platforms capable of enriching tumor-derived EVs from the blood. Herein, we introduce a biostructure-inspired NanoVilli Chip, capable of highly efficient and reproducible immunoaffinity capture of tumor-derived EVs from blood plasma samples. Anti-EpCAM-grafted silicon nanowire arrays were engineered to mimic the distinctive structures of intestinal microvilli, dramatically increasing surface area and enhancing tumor-derived EV capture. RNA in the captured EVs can be recovered for downstream molecular analyses by reverse transcription Droplet Digital PCR. We demonstrate that this assay can be applied to monitor the dynamic changes of ROS1 rearrangements and epidermal growth factor receptor T790M mutations that predict treatment responses and disease progression in non-small cell lung cancer patients.
Collapse
Affiliation(s)
- Jiantong Dong
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, P. R. China
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Zipeng Wu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Anqi Zhou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Shih-Jie Chou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- Institute of Pharmacology, National Yang-Ming University, Taipei 112, Taiwan
| | - Yu Jen Jan
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Peng Yang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Lirong Bao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Xinghong Tang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Patrick Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Yue Hua
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Dianwen Xu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Rueihung Kao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Meng Meng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, P. R. China
| | - Xirun Zheng
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| | - Ying Liu
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| | - Tatyana Vagner
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California 90048, United States
| | - Xiaoshu Chai
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| | - Dongjing Zhou
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| | - Mengyuan Li
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang-Ming University, Taipei 112, Taiwan
| | - Guangjuan Zheng
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| | - Dolores Di Vizio
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California 90048, United States
| | - Vatche G. Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Edwin Posadas
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Steven J. Jonas
- Department of Pediatrics, David Geffen School of Medicine, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, and Children’s Discovery and Innovation Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute and Departments of Chemistry and Biochemistry, and of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Shin-Pon Ju
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Paul S. Weiss
- California NanoSystems Institute and Departments of Chemistry and Biochemistry, and of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, P. R. China
| |
Collapse
|
84
|
|
85
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
86
|
Wang X, Sun L, Zhang H, Wei L, Qu W, Zeng Z, Liu Y, Zhu Z. Microfluidic chip combined with magnetic-activated cell sorting technology for tumor antigen-independent sorting of circulating hepatocellular carcinoma cells. PeerJ 2019; 7:e6681. [PMID: 30972256 PMCID: PMC6448555 DOI: 10.7717/peerj.6681] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/25/2019] [Indexed: 12/29/2022] Open
Abstract
Purpose We aimed to generate a capture platform that integrates a deterministic lateral displacement (DLD) microfluidic structure with magnetic-activated cell sorting (MACS) technology for miniaturized, efficient, tumor antigen-independent circulating tumor cell (CTC) separation. Methods The microfluidic structure was based on the theory of DLD and was designed to remove most red blood cells and platelets. Whole Blood CD45 MicroBeads and a MACS separator were then used to remove bead-labeled white blood cells. We established HepG2 human liver cancer cells overexpressing green fluorescent protein by lentiviral transfection to simulate CTCs in blood, and these cells were then used to determine the CTC isolation efficiency of the device. The performance and clinical value of our platform were evaluated by comparison with the Abnova CytoQuest™ CR system in the separating of blood samples from 12 hepatocellular carcinoma patients undergoing liver transplantation in a clinical follow-up experiment. The isolated cells were stained and analyzed by confocal laser scanning microscopy. Results Using our integrated platform at the optimal flow rates for the specimen (60 µl/min) and buffer (100 µl/min per chip), we achieved an CTC yield of 85.1% ± 3.2%. In our follow-up of metastatic patients, CTCs that underwent epithelial–mesenchymal transition were found. These CTCs were missed by the CytoQuest™ CR bulk sorting approach, whereas our platform displayed increased sensitivity to EpCAMlow CTCs. Conclusions Our platform, which integrates microfluidic and MACS technology, is an attractive method for high-efficiency CTC isolation regardless of surface epitopes.
Collapse
Affiliation(s)
- Xuebin Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Liying Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Haiming Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Lin Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Wei Qu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhigui Zeng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Ying Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Zhijun Zhu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Liver Transplantation Center, National Clinical Research Center for Digestive Diseases (NCRC-DD), Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
87
|
Cho H, Kim J, Song H, Sohn KY, Jeon M, Han KH. Microfluidic technologies for circulating tumor cell isolation. Analyst 2019; 143:2936-2970. [PMID: 29796523 DOI: 10.1039/c7an01979c] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metastasis is the main cause of tumor-related death, and the dispersal of tumor cells through the circulatory system is a critical step in the metastatic process. Early detection and analysis of circulating tumor cells (CTCs) is therefore important for early diagnosis, prognosis, and effective treatment of cancer, enabling favorable clinical outcomes in cancer patients. Accurate and reliable methods for isolating and detecting CTCs are necessary to obtain this clinical information. Over the past two decades, microfluidic technologies have demonstrated great potential for isolating and detecting CTCs from blood. The present paper reviews current advanced microfluidic technologies for isolating CTCs based on various biological and physical principles, and discusses their fundamental advantages and drawbacks for subsequent cellular and molecular assays. Owing to significant genetic heterogeneity among CTCs, microfluidic technologies for isolating individual CTCs have recently been developed. We discuss these single-cell isolation methods, as well as approaches to overcoming the limitations of current microfluidic CTC isolation technologies. Finally, we provide an overview of future innovative microfluidic platforms.
Collapse
Affiliation(s)
- Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae 621-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
88
|
Rostami P, Kashaninejad N, Moshksayan K, Saidi MS, Firoozabadi B, Nguyen NT. Novel approaches in cancer management with circulating tumor cell clusters. JOURNAL OF SCIENCE: ADVANCED MATERIALS AND DEVICES 2019; 4:1-18. [DOI: 10.1016/j.jsamd.2019.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
89
|
Abalde-Cela S, Piairo P, Diéguez L. The Significance of Circulating Tumour Cells in the Clinic. Acta Cytol 2019; 63:466-478. [PMID: 30820013 DOI: 10.1159/000495417] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/08/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Despite the hype about circulating tumour cells (CTCs) in the early 2000s and their potential in the diagnosis of metastasis, in recent years, the hope for personalised cancer management relies more on circulating tumour (ct)DNA that has entered the clinic in a much more efficient way. So far, approved methods for CTCs in the clinic only provide the counting of CTCs, which enables monitoring of the progression of metastatic breast, prostate, and colorectal cancer patients with therapy. Approved methods for ctDNA facilitate the analysis of specific mutations in lung cancer, thereby providing indications for potentially successful treatments. This situation inclined the balance towards molecular analysis in liquid biopsy, leveraged by new technologies and companies providing broader mutation and gene expression analysis towards the early diagnosis of cancer. STUDY DESIGN We conducted a search for the studies published to date that provide details about the significance of CTCs in the clinic. RESULTS Many studies and clinical trials have demonstrated the potential of CTCs in patient screening, early diagnosis, therapy resistance, and patient prognosis. CONCLUSIONS Large multi-centre studies are still needed to formally validate the clinical relevance of CTCs. Meticulous design of the clinical trials is a crucial point to achieve this long-sought objective.
Collapse
Affiliation(s)
- Sara Abalde-Cela
- Medical Devices Research Group, Department of Life Sciences, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Paulina Piairo
- Medical Devices Research Group, Department of Life Sciences, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal
- iMM- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | - Lorena Diéguez
- Medical Devices Research Group, Department of Life Sciences, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal,
| |
Collapse
|
90
|
Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat Biomed Eng 2019; 3:438-451. [PMID: 31123323 PMCID: PMC6556143 DOI: 10.1038/s41551-019-0356-9] [Citation(s) in RCA: 381] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 01/16/2019] [Indexed: 12/21/2022]
Abstract
The performance of current microfluidic methods for exosome detection is constrained by boundary conditions, as well as fundamental limits to microscale mass transfer and interfacial exosome binding. Here, we show that a microfluidic chip designed with self-assembled three-dimensional herringbone nanopatterns can detect low levels of tumour-associated exosomes in plasma (10 exosomes μl-1, or approximately 200 vesicles per 20 μl of spiked sample) that would otherwise be undetectable by standard microfluidic systems for biosensing. The nanopatterns promote microscale mass transfer, increase surface area and probe density to enhance the efficiency and speed of exosome binding, and permit drainage of the boundary fluid to reduce near-surface hydrodynamic resistance, thus promoting particle-surface interactions for exosome binding. We used the device for the detection-in 2 μl plasma samples from 20 ovarian cancer patients and 10 age-matched controls-of exosome subpopulations expressing CD24, epithelial cell adhesion molecule and folate receptor alpha proteins, and suggest exosomal folate receptor alpha as a potential biomarker for early detection and progression monitoring of ovarian cancer. The nanolithography-free nanopatterned device should facilitate the use of liquid biopsies for cancer diagnosis.
Collapse
|
91
|
Varillas JI, Zhang J, Chen K, Barnes II, Liu C, George TJ, Fan ZH. Microfluidic Isolation of Circulating Tumor Cells and Cancer Stem-Like Cells from Patients with Pancreatic Ductal Adenocarcinoma. Theranostics 2019; 9:1417-1425. [PMID: 30867841 PMCID: PMC6401494 DOI: 10.7150/thno.28745] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) requires multimodal therapeutic approaches and disease monitoring for effective treatment. Liquid biopsy biomarkers, including circulating tumor cells (CTCs) and cancer stem-like cells (CSCs), hold promise for evaluating treatment response promptly and guiding therapeutic modifications. Methods: From 24 patients with metastatic PDAC (stage IV, M1) undergoing active systemic treatment, we collected 78 blood samples at different time points for CTC and CSC isolation using a microfluidic platform functionalized with antibodies against a CTC biomarker, epithelial cell adhesion molecule (EpCAM), or a CSC biomarker, CD133. These isolated cells were further verified, via fluorescent staining and imaging, using cytokeratin (CK), CD45, and nucleic acid stain 4',6-diamidino-2-phenylindole (DAPI). Results: The majority (84.4%) of patient blood samples were positive for CTCs (EpCAM+CK+CD45-DAPI+) and 70.8% of patient blood samples were positive for CSCs (CD133+CK+CD45-DAPI+), using the highest baseline value of healthy samples as threshold. The CTC subtypes (EpCAM+CK+CD45-DAPI+CD133+ and EpCAM+CK+CD45-DAPI+CD133-) and CSC subtypes (CD133+CK+CD45-DAPI+EpCAM+ and CD133+CK+CD45-DAPI+EpCAM-) were also analyzed using immunochemical methods. In several cases, CSCs exhibited cytokeratin expression that did not express EpCAM, indicating that they will not be detected using EpCAM-based isolation. Conclusion: The microfluidic platform enabled the reliable isolation of CTCs and CSCs from PDAC patient samples, as well as their subtypes. Complementary assessment of both CTCs and CSCs appears advantageous to assess the profile of tumor progressing in some cases. This research has important implications for the application and interpretation of approved methods to detect CTCs.
Collapse
|
92
|
LeValley PJ, Tibbitt MW, Noren B, Kharkar P, Kloxin AM, Anseth KS, Toner M, Oakey J. Immunofunctional photodegradable poly(ethylene glycol) hydrogel surfaces for the capture and release of rare cells. Colloids Surf B Biointerfaces 2019; 174:483-492. [PMID: 30497010 PMCID: PMC6545105 DOI: 10.1016/j.colsurfb.2018.11.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) play a central role in cancer metastasis and represent a rich source of data for cancer prognostics and therapeutic guidance. Reliable CTC recovery from whole blood therefore promises a less invasive and more sensitive approach to cancer diagnosis and progression tracking. CTCs, however, are exceedingly rare in whole blood, making their quantitative recovery challenging. Several techniques capable of isolating these rare cells have been introduced and validated, yet most suffer from low CTC purity or viability, both of which are essential to develop a clinically viable CTC isolation platform. To address these limitations, we introduce a patterned, immunofunctional, photodegradable poly(ethylene glycol) (PEG) hydrogel capture surface for the isolation and selective release of rare cell populations. Flat and herringbone capture surfaces were successfully patterned via PDMS micromolding and photopolymerization of photolabile PEG hydrogels. Patterned herringbone surfaces, designed to convectively transport cells to the capture surface, exhibited improved capture density relative to flat surfaces for target cell capture from buffer, buffy coat, and whole blood. Uniquely, captured cells were released for collection by degrading the hydrogel capture surface with either bulk or targeted irradiation with cytocompatible doses of long wavelength UV light. Recovered cells remained viable following capture and release and exhibited similar growth rates as untreated control cells. The implementation of molded photodegradable PEG hydrogels as a CTC capture surface provides a micropatternable, cytocompatible platform that imparts the unique ability to recover pure, viable CTC samples by selectively releasing target cells.
Collapse
Affiliation(s)
- Paige J LeValley
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States; Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mark W Tibbitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder,Boulder, CO 80309, United States; Department of Mechanical and Process Engineering, ETH Zürich, Zürich, 8092, Switzerland
| | - Ben Noren
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States
| | - Prathamesh Kharkar
- Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - April M Kloxin
- Department of Biomolecular and Chemical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder,Boulder, CO 80309, United States; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, United States; Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Mehmet Toner
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children and Harvard Medical School, Boston, MA 02114, United States
| | - John Oakey
- Department of Chemical Engineering, University of Wyoming, Laramie, WY 82071, United States.
| |
Collapse
|
93
|
Que Z, Luo B, Zhou Z, Dong C, Jiang Y, Wang L, Shi Q, Tian J. Establishment and characterization of a patient-derived circulating lung tumor cell line in vitro and in vivo. Cancer Cell Int 2019; 19:21. [PMID: 30718976 PMCID: PMC6352330 DOI: 10.1186/s12935-019-0735-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/08/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) have been described as a population of cells that may seed metastasis, which is a reliable target for the prevention of metastases in lung cancer patients at the early stage. The culturing of CTCs in vitro can be used to study the mechanism of lung cancer metastasis and to screen antimetastasis drugs. This study aims to establish CTC cell line in vitro and explore the potential mechanism of its metastasis. METHODS A mixture of EpCAM- and EGFR-coated immunomagnetic microbeads in microfluidic Herringbone-Chip was used to capture CTCs. The CTCs, 95-D and A549 cells was evaluated by cell proliferation assays, clonal formation assays, migration assays and drug resistance. Flow cytometry and cytokine protein chip were used to detect the difference in phenotype and cytokine secretion between CTCs, 95-D and A549 cells. The NOD/SCID mice were used to study tumorigenicity, lung organ colonization and metastasis of CTCs. The H&E staining, immunohistochemistry and immunofluorescence assay were used to detect the pathological status of CTCs. RESULTS The number of EpCAM(+)/EGFR(+)/CK(+)/CD45(-) lung CTCs showed a weak negative correlation with clinical stages in patients with non-small cell lung cancer (NSCLC). In a phase IIa lung cancer patient, we successfully establish a permanent CTC cell line, named CTC-TJH-01. In vitro studies showed the CTC-TJH-01 cells were in the intermediate stage of epithelial to mesenchymal transition (EMT), had stem cell characteristics and were drug resistant. In vivo studies showed that CTC-TJH-01 cells can induce tumorigenesis, lung organ colonization and metastasis after xenografting in immunodeficient mice. In addition, the low expression level of CX3CL1 and high expression level of CXCL5 in the CTC-TJH-01 cells may be an important mechanism for their metastasis. CONCLUSIONS We successfully established a permanent CTC cell line with metastatic ability, which can be used to screen antimetastatic drugs and study the mechanism of lung cancer metastasis.
Collapse
Affiliation(s)
- Zujun Que
- Institute of Traditional Chinese Medicine Oncology, Shanghai Institute of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Bin Luo
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Zhiyi Zhou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Changsheng Dong
- Institute of Traditional Chinese Medicine Oncology, Shanghai Institute of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Yi Jiang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Lin Wang
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| | - Qihui Shi
- Key Laboratory of Medical Epigenetics and Metabolism, Minhang Branch, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433 People’s Republic of China
| | - Jianhui Tian
- Institute of Traditional Chinese Medicine Oncology, Shanghai Institute of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032 People’s Republic of China
| |
Collapse
|
94
|
Kumar SA, Delgado M, Mendez VE, Joddar B. Applications of stem cells and bioprinting for potential treatment of diabetes. World J Stem Cells 2019; 11:13-32. [PMID: 30705712 PMCID: PMC6354103 DOI: 10.4252/wjsc.v11.i1.13] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/26/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023] Open
Abstract
Currently, there does not exist a strategy that can reduce diabetes and scientists are working towards a cure and innovative approaches by employing stem cell-based therapies. On the other hand, bioprinting technology is a novel therapeutic approach that aims to replace the diseased or lost β-cells, insulin-secreting cells in the pancreas, which can potentially regenerate damaged organs such as the pancreas. Stem cells have the ability to differentiate into various cell lines including insulin-producing cells. However, there are still barriers that hamper the successful differentiation of stem cells into β-cells. In this review, we focus on the potential applications of stem cell research and bioprinting that may be targeted towards replacing the β-cells in the pancreas and may offer approaches towards treatment of diabetes. This review emphasizes on the applicability of employing both stem cells and other cells in 3D bioprinting to generate substitutes for diseased β-cells and recover lost pancreatic functions. The article then proceeds to discuss the overall research done in the field of stem cell-based bioprinting and provides future directions for improving the same for potential applications in diabetic research.
Collapse
Affiliation(s)
- Shweta Anil Kumar
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory, Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968, United States
| | - Monica Delgado
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory, Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968, United States
| | - Victor E Mendez
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory, Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968, United States
| | - Binata Joddar
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory, Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968, United States
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968, United States.
| |
Collapse
|
95
|
Kulasinghe A, Zhou J, Kenny L, Papautsky I, Punyadeera C. Capture of Circulating Tumour Cell Clusters Using Straight Microfluidic Chips. Cancers (Basel) 2019; 11:E89. [PMID: 30646614 PMCID: PMC6356955 DOI: 10.3390/cancers11010089] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/29/2022] Open
Abstract
Circulating tumour cells (CTCs) are the metastatic precursors to distant disease in head and neck cancers (HNCs). Whilst the prognostic and predictive value of single CTCs have been well documented, the role of CTC clusters, which potentially have a higher metastatic capacity are limited. In this study, the authors used a novel straight microfluidic chip to focus and capture CTCs. The chip offers high cell recoveries with clinically relevant numbers (10⁻500 cells/mL) without the need for further purification. Single CTCs were identified in 10/21 patient samples (range 2⁻24 CTCs/mL), CTC clusters in 9/21 patient samples (range 1⁻6 CTC clusters/mL) and circulating tumour microemboli (CTM) in 2/21 samples. This study demonstrated that CTC clusters contain EGFR amplified single CTCs within the cluster volume. This novel microfluidic chip demonstrates the efficient sorting and preservation of single CTCs, CTC clusters and CTMs. The authors intend to expand this study to a larger cohort to determine the clinical implication of the CTC subsets in HNC.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| | - Jian Zhou
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Liz Kenny
- School of Medicine, University of Queensland, QLD 4029, Australia.
- Central Integrated Regional Cancer Services, Royal Brisbane and Women's Hospital, Queensland Health, QLD 4029, Australia.
| | - Ian Papautsky
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Chamindie Punyadeera
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| |
Collapse
|
96
|
Sun M, Xu J, Shamul JG, Lu X, Husain S, He X. Creating a capture zone in microfluidic flow greatly enhances the throughput and efficiency of cancer detection. Biomaterials 2019; 197:161-170. [PMID: 30660052 DOI: 10.1016/j.biomaterials.2019.01.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/09/2018] [Accepted: 01/05/2019] [Indexed: 12/11/2022]
Abstract
Efficient capture of rare circulating tumor cells (CTCs) from blood samples is valuable for early cancer detection to improve the management of cancer. In this work, we developed a highly efficient microfluidics-based method for detecting CTCs in human blood. This is achieved by creating separate capture and flow zones in the microfluidic device (ZonesChip) and using patterned dielectrophoretic force to direct cells from the flow zone into the capture zone. This separation of the capture and flow zones minimizes the negative impact of high flow speed (and thus high throughput) and force in the flow zone on the capture efficiency, overcoming a major bottleneck of contemporary microfluidic approaches using overlapping flow and capture zones for CTC detection. When the flow speed is high (≥0.58 mm/s) in the flow zone, the separation of capture and flow zones in our ZonesChip could improve the capture efficiency from ∼0% (for conventional device without separating the two zones) to ∼100%. Our ZonesChip shows great promise as an effective platform for the detection of CTCs in blood from patients with early/localized-stage colorectal tumors.
Collapse
Affiliation(s)
- Mingrui Sun
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jiangsheng Xu
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Syed Husain
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
97
|
Hwang JY, Kim ST, Kwon J, Lee J, Chun YO, Han JS, Han HS. Ultrasensitive Fluorescence Monitoring and in Vivo Live Imaging of Circulating Tumor Cell-Derived miRNAs Using Molecular Beacon System. ACS Sens 2018; 3:2651-2659. [PMID: 30426747 DOI: 10.1021/acssensors.8b01095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circulating tumor cells (CTCs) have considerable clinical significance in cancer progression and prognosis. In this context, CTC-derived microRNAs (miRs) in blood and tissues have been proposed as the novel noninvasive biomarkers for monitoring tumor progression, especially at the early stages. To monitor circulating miRs, a tool should have high sensitivity, be a simple procedure, and allow detection in very small volumes. Thus, we designed a sensing tool for sensitive monitoring of blood or tissue miRs using a fluorophore-quencher probe-based molecular beacon (MB). This MB-based tool displayed an ultrasensitive limit of detection (LOD) level of 6.7 × 10-17 M and 8.7 × 10-17 for metastasis-derived miR-21a and miR-221, respectively. It also can discriminate miR-21a/221 from both guide strand miRs and its precursor forms (pre-miR). Furthermore, the tool discriminated between blood and tissue-related miR-21a/221-expression and detected metastasis and epithelial-mesenchymal transition and also describe a noninvasive miR fluorescence imaging of CTCs in a mouse model, showing the potential for clinical diagnosis and prognosis.
Collapse
Affiliation(s)
- Ji Yeon Hwang
- Preclinical Research Center, Biomedical Research Institute, Seoul National University Bundang Hospital, Gyeonggi-do 13620, Republic of Korea
- The Institute for the 3Rs, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Sang Tae Kim
- Department of Surgery, Seoul National University Bundang Hospital, Gyeonggi-do 13620, Republic of Korea
- Biomedical Research Institute, Seoul National University Bundang Hospital, Gyeonggi-do 13605, Republic of Korea
| | - Junyoung Kwon
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46240, Republic of Korea
| | - Jaebeom Lee
- Department of Chemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Ok Chun
- Biomedical Research Institute, Seoul National University Bundang Hospital, Gyeonggi-do 13605, Republic of Korea
- Department of Molecular Biology, DanKook University, Gyeonggi-do 16890, Republic of Korea
| | - Jin Soo Han
- The Institute for the 3Rs, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Ho-Seong Han
- Department of Surgery, Seoul National University Bundang Hospital, Gyeonggi-do 13620, Republic of Korea
| |
Collapse
|
98
|
Shen Q, Yang H, Peng C, Zhu H, Mei J, Huang S, Chen B, Liu J, Wu W, Cao S. Capture and biological release of circulating tumor cells in pancreatic cancer based on peptide-functionalized silicon nanowire substrate. Int J Nanomedicine 2018; 14:205-214. [PMID: 30636873 PMCID: PMC6307685 DOI: 10.2147/ijn.s187892] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Efficient and precise circulating tumor cells' (CTCs) capture and release with minimal effect on cell viability for CTCs' analysis are general requirements of CTCs' detection device in clinical application. However, these two essential factors are difficult to be achieved simultaneously. Methods In order to reach the aforementioned goal, we integrated multiple strategies and technologies of staggered herringbone structure, nanowires' substrate, peptides, enzymatic release, specific cell staining, and gene sequencing into microfluidic device and the sandwich structure peptide-silicon nanowires' substrate was termed as Pe-SiNWS. Results The Pe-SiNWS demonstrated excellent capture efficiency (95.6%) and high release efficiency (92.6%). The good purity (28.5%) and cell viability (93.5%) of CTCs could be obtained through specific capture and biological release by using Pe-SiNWS. The good purity of CTCs facilitated precise and quick biological analysis, and five types of KRAS mutation were detected in 16 pancreatic cancer patients but not in healthy donors. Conclusion The results proved that the effective capture, minor damage release, and precise analysis of CTCs could be realized simultaneously by our novel strategy. The successful clinical application indicated that our work was anticipated to open up new opportunities for the design of CTC microfluidic device.
Collapse
Affiliation(s)
- Qinglin Shen
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, .,Cancer Center, Renmin Hospital, Wuhan University, Wuhan, China,
| | - Haitao Yang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China,
| | - Caixia Peng
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Central Laboratory, The Central Hospital of Wuhanper, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhu
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jia Mei
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Shan Huang
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Bin Chen
- Central Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jue Liu
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbo Wu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China,
| | - Shaokui Cao
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China,
| |
Collapse
|
99
|
Rofi E, Vivaldi C, Del Re M, Arrigoni E, Crucitta S, Funel N, Fogli S, Vasile E, Musettini G, Fornaro L, Falcone A, Danesi R. The emerging role of liquid biopsy in diagnosis, prognosis and treatment monitoring of pancreatic cancer. Pharmacogenomics 2018; 20:49-68. [PMID: 30520336 DOI: 10.2217/pgs-2018-0149] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor DNA, circulating tumor cells and tumor-related exosomes may offer new opportunities to provide insights into the biological and clinical characteristics of a neoplastic disease. They represent alternative routes for diagnostic and prognostic purposes, and for predicting and longitudinally monitoring response to treatment and disease progression. Hence, circulating biomarkers represent promising noninvasive tools in the scenario of pancreatic cancer, where neither molecular nor clinical predictors of treatment benefit have been identified yet. This review aims to provide an overview of the current status of circulating biomarker research in pancreatic cancer, and discusses their potential clinical utility to facilitate clinical decision-making.
Collapse
Affiliation(s)
- Eleonora Rofi
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Caterina Vivaldi
- Unit of Medical Oncology, Department of Translational Research & New Technologies in Medicine, University of Pisa, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Elena Arrigoni
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Niccola Funel
- Department of Translational Research & The New Technologies in Medicine & Surgery, University of Pisa, Pisa, Italy
| | - Stefano Fogli
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Enrico Vasile
- Unit of Medical Oncology, Department of Translational Research & New Technologies in Medicine, University of Pisa, Italy
| | - Gianna Musettini
- Unit of Medical Oncology, Department of Translational Research & New Technologies in Medicine, University of Pisa, Italy
| | - Lorenzo Fornaro
- Unit of Medical Oncology, Department of Translational Research & New Technologies in Medicine, University of Pisa, Italy
| | - Alfredo Falcone
- Unit of Medical Oncology, Department of Translational Research & New Technologies in Medicine, University of Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology & Pharmacogenetics, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
100
|
Samandari M, Julia MG, Rice A, Chronopoulos A, Del Rio Hernandez AE. Liquid biopsies for management of pancreatic cancer. Transl Res 2018; 201:98-127. [PMID: 30118658 DOI: 10.1016/j.trsl.2018.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/17/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the main causes of cancer-related deaths worldwide. It is asymptomatic at an early stage, and most diagnosis occurs when the disease is already at a late stage, by which time the tumor is nonresectable. In order to increase the overall survival of patients with pancreatic cancer, as well as to decrease the cancer burden, it is necessary to perform early diagnosis, prognosis stratifications and cancer monitoring using accurate, minimally invasive, and cost-effective methods. Liquid biopsies seek to detect tumor-associated biomarkers in a variety of extractable body fluids and can help to monitor treatment response and disease progression, and even predict patient outcome. In patients with pancreatic cancer, tumor-derived materials, primarily circulating tumor DNA, circulating tumor cells and exosomes, are being studied for inclusion in the management of the disease. This review focuses on describing the biology of these biomarkers, methods for their enrichment and detection, as well as their potential for clinical application. Moreover, we discuss the future direction of liquid biopsies and introduce how they can be exploited toward point of care personalized medicine for the management of pancreatic cancer.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - María Gil Julia
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|