1
|
Nian M, Chen B, He M, Hu B. A Cascaded Phase-Transfer Microfluidic Chip with Magnetic Probe for High-Activity Sorting, Purification, Release, and Detection of Circulating Tumor Cells. Anal Chem 2024; 96:766-774. [PMID: 38158582 DOI: 10.1021/acs.analchem.3c03971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Microfluidic chips have emerged as a promising tool for sorting and enriching circulating tumor cells (CTCs) in blood, while the efficacy and purity of CTC sorting greatly depend on chip design. Herein, a novel cascaded phase-transfer microfluidic chip was developed for high-efficiency sorting, purification, release, and detection of MCF-7 cells (as a model CTC) in blood samples. MCF-7 cells were specifically captured by EpCAM aptamer-modified magnetic beads and then introduced into the designed cascaded phase-transfer microfluidic chip that consisted of three functional regions (sorting, purification, and release zone). In the sorting zone, the MCF-7 cells moved toward the inner wall of the channel and entered the purification zone for primary separation from white blood cells; in the purification zone, the MCF-7 cells were transferred to the phosphate-buffered saline flow under the interaction of Dean forces and central magnetic force, achieving high purification of MCF-7 cells from blood samples; in the release zone, MCF-7 cells were further transferred into the nuclease solution and fixed in groove by the strong magnetic force and hydrodynamic force, and the continuously flowing nuclease solution cleaved the aptamer on the trapped MCF-7 cells, causing gentle release of MCF-7 cells for subsequent inductively coupled plasma mass spectrometry (ICP-MS) detection or further cultivation. By measurement of the endogenous element Zn in the cells using ICP-MS for cell counting, an average cell recovery of 84% for MCF-7 cells was obtained in spiked blood samples. The developed method was applied in the analysis of real blood samples from healthy people and breast cancer patients, and CTCs were successfully detected in all tested patient samples (16/16). Additionally, the removal of the magnetic probes on the cell surface significantly improved cell viability up to 99.3%. Therefore, the developed cascaded phase-transfer microfluidic chip ICP-MS system possessed high integration for CTCs analysis with high cell viability, cell recovery, and purity, showing great advantages in early clinical cancer diagnosis.
Collapse
Affiliation(s)
- Miaoxiang Nian
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Beibei Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Man He
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
2
|
R G, Kar S, Nagai M, Mahapatra PS, Santra TS. Massively Parallel High-Throughput Single-Cell Patterning and Large Biomolecular Delivery in Mammalian Cells Using Light Pulses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303053. [PMID: 37548122 DOI: 10.1002/smll.202303053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/21/2023] [Indexed: 08/08/2023]
Abstract
The recent advancements of single-cell analysis have significantly enhanced the ability to understand cellular physiology when compared to bulk cellular analysis. Here a massively parallel single-cell patterning and very large biomolecular delivery is reported. Micro-pillar polydimethyl siloxane stamp with different diameters (40-100 µm with 1 cm × 1 cm patterning area) is fabricated and then imprint distinct proteins and finally pattern single-cell to small clusters of cells depending on the micro-pillar diameters. The maximum patterning efficiency is achieved 99.7% for SiHa, 96.75% for L929, and 98.6% for MG63 cells, for the 100 µm micro-pillar stamp. For intracellular delivery of biomolecules into the patterned cells, a titanium micro-dish device is aligned on top of the cells and exposed by infrared light pulses. The platform successfully delivers small to very large biomolecules such as PI dyes (668 Da), dextran 3000 Da, siRNA (20-24 bp), and large size enzymes (464 KDa) in SiHa, L929 and MG63 cells. The delivery efficiency for PI dye, Dextran 3000, siRNA, and enzyme for patterned cells are ≈95 ± 3%, 97 ± 1%, 96 ± 1% and 94 ± 3%, with cell viability of 98 ± 1%. Thus, the platform is compact, robust, easy for printing, and potentially applicable for single-cell therapy and diagnostics.
Collapse
Affiliation(s)
- Gayathri R
- Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai, 600036, India
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Srabani Kar
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, 600036, India
- Department of Physics, Indian Institute of Science Education and Research, Tirupati, 517507, India
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, 441-8580, Japan
| | - Pallab Sinha Mahapatra
- Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, 600036, India
| |
Collapse
|
3
|
Lenhart A, Kennedy RT. Evaluation of Surface Treatments of PDMS Microfluidic Devices for Improving Small-Molecule Recovery with Application to Monitoring Metabolites Secreted from Islets of Langerhans. ACS MEASUREMENT SCIENCE AU 2023; 3:380-389. [PMID: 37868359 PMCID: PMC10588933 DOI: 10.1021/acsmeasuresciau.3c00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 10/24/2023]
Abstract
Microfluidic devices are becoming an important tool for bioanalysis with applications including studying cell secretion, cell growth, and drug delivery. Small molecules such as drugs, cell products, or nutrients may partition into polydimethylsiloxane (PDMS), a commonly used material for microfluidic devices, potentially leading to poor recovery or inaccurate delivery of such chemicals. To decrease small-molecule partitioning, surface and bulk PDMS treatments have been developed; however, these have been tested on few analytes, or their biocompatibility are unknown. Studies often focus on one analyte, whereas a diversity of chemicals are of interest and possibly affected. In this study, 11 device treatments are tested and applied to 21 biologically relevant small molecules with a variety of chemical structures. Device treatments are characterized using water contact angle measurements and evaluated by measuring recovery of the 21 target analytes using liquid chromatography-mass spectrometry. 1,5-Dimethyl-1,5-diazaundecamethylene polymethobromide (polybrene), a positively charged polymer, produced the least hydrophilic surface and was found to provide the best recovery with most of the analytes having >50% recovery and up to 92% recovery; however, recovery varied by analyte highlighting the importance of analyte diversity rather than targeting a single analyte in evaluating treatments. A polybrene-treated device was applied to investigate secretion from pancreatic islets, which are micro-organs involved in glucose homeostasis and diabetes. Islets secrete small molecules that have been shown to modulate the secretion of islets' main functional products, glucose-regulating hormones. The polybrene treatment enabled the detection of 20 target analytes from islets-on-chip during isosmotic and hypo-osmotic glucose perfusions and resulted in detection of more significant secretion changes compared to untreated PDMS.
Collapse
Affiliation(s)
- Ashley
E. Lenhart
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
4
|
Surappa S, Multani P, Parlatan U, Sinawang PD, Kaifi J, Akin D, Demirci U. Integrated "lab-on-a-chip" microfluidic systems for isolation, enrichment, and analysis of cancer biomarkers. LAB ON A CHIP 2023; 23:2942-2958. [PMID: 37314731 PMCID: PMC10834032 DOI: 10.1039/d2lc01076c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liquid biopsy has garnered considerable attention as a complementary clinical tool for the early detection, molecular characterization and monitoring of cancer over the past decade. In contrast to traditional solid biopsy techniques, liquid biopsy offers a less invasive and safer alternative for routine cancer screening. Recent advances in microfluidic technologies have enabled handling of liquid biopsy-derived biomarkers with high sensitivity, throughput, and convenience. The integration of these multi-functional microfluidic technologies into a 'lab-on-a-chip' offers a powerful solution for processing and analyzing samples on a single platform, thereby reducing the complexity, bio-analyte loss and cross-contamination associated with multiple handling and transfer steps in more conventional benchtop workflows. This review critically addresses recent developments in integrated microfluidic technologies for cancer detection, highlighting isolation, enrichment, and analysis strategies for three important sub-types of cancer biomarkers: circulating tumor cells, circulating tumor DNA and exosomes. We first discuss the unique characteristics and advantages of the various lab-on-a-chip technologies developed to operate on each biomarker subtype. This is then followed by a discussion on the challenges and opportunities in the field of integrated systems for cancer detection. Ultimately, integrated microfluidic platforms form the core of a new class of point-of-care diagnostic tools by virtue of their ease-of-operation, portability and high sensitivity. Widespread availability of such tools could potentially result in more frequent and convenient screening for early signs of cancer at clinical labs or primary care offices.
Collapse
Affiliation(s)
- Sushruta Surappa
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Priyanka Multani
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Ugur Parlatan
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jussuf Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
5
|
Recent advances in integrated microfluidics for liquid biopsies and future directions. Biosens Bioelectron 2022; 217:114715. [PMID: 36174359 DOI: 10.1016/j.bios.2022.114715] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 07/20/2022] [Accepted: 09/09/2022] [Indexed: 12/12/2022]
Abstract
Liquid biopsies have piqued the interest of researchers as a new tumor diagnosis technique due to their unique benefits of non-invasiveness, sensitivity, and convenience. Recent advances in microfluidic technology have integrated separation, purification, and detection, allowing for high-throughput, high-sensitivity, and high-controllability detection of specific biomarkers in liquid biopsies. With the increasing demand for tumor detection and individualized treatment, new challenges are emerging for the ever-improving microfluidic technology. The state-of-the-art microfluidic design and fabrications have been reviewed in this manuscript, and how this technology can be applied to liquid biopsies from the point of view of the detection process. The primary discussion objectives are circulating tumor cells (CTCs), exosomes, and circulating nucleic acid (ctDNA). Furthermore, the challenges and future direction of microfluidic technology in detecting liquid biomarkers have been discussed.
Collapse
|
6
|
Addanki S, Meas S, Sarli VN, Singh B, Lucci A. Applications of Circulating Tumor Cells and Circulating Tumor DNA in Precision Oncology for Breast Cancers. Int J Mol Sci 2022; 23:ijms23147843. [PMID: 35887191 PMCID: PMC9315812 DOI: 10.3390/ijms23147843] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
Liquid biopsies allow for the detection of cancer biomarkers such as circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Elevated levels of these biomarkers during cancer treatment could potentially serve as indicators of cancer progression and shed light on the mechanisms of metastasis and therapy resistance. Thus, liquid biopsies serve as tools for cancer detection and monitoring through a simple, non-invasive blood draw, allowing multiple longitudinal sampling. These circulating markers have significant prospects for use in assessing patients’ prognosis, monitoring response to therapy, and developing precision medicine. In addition, single-cell omics of these liquid biopsy markers can be potential tools for identifying tumor heterogeneity and plasticity as well as novel therapeutic targets. In this review, we focus on our current understanding of circulating tumor biomarkers, especially in breast cancer, and the scope of novel sequencing technologies and diagnostic methods for better prognostication and patient stratification to improve patient outcomes.
Collapse
Affiliation(s)
- Sridevi Addanki
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
| | - Salyna Meas
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanessa Nicole Sarli
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Balraj Singh
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anthony Lucci
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
7
|
Li C, He W, Wang N, Xi Z, Deng R, Liu X, Kang R, Xie L, Liu X. Application of Microfluidics in Detection of Circulating Tumor Cells. Front Bioeng Biotechnol 2022; 10:907232. [PMID: 35646880 PMCID: PMC9133555 DOI: 10.3389/fbioe.2022.907232] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/11/2022] [Indexed: 12/22/2022] Open
Abstract
Tumor metastasis is one of the main causes of cancer incidence and death worldwide. In the process of tumor metastasis, the isolation and analysis of circulating tumor cells (CTCs) plays a crucial role in the early diagnosis and prognosis of cancer patients. Due to the rarity and inherent heterogeneity of CTCs, there is an urgent need for reliable CTCs separation and detection methods in order to obtain valuable information on tumor metastasis and progression from CTCs. Microfluidic technology is increasingly used in various studies of CTCs separation, identification and characterization because of its unique advantages, such as low cost, simple operation, less reagent consumption, miniaturization of the system, rapid detection and accurate control. This paper reviews the research progress of microfluidic technology in CTCs separation and detection in recent years, as well as the potential clinical application of CTCs, looks forward to the application prospect of microfluidic technology in the treatment of tumor metastasis, and briefly discusses the development prospect of microfluidic biosensor.
Collapse
Affiliation(s)
- Can Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei He
- Department of Clinical Medical Engineering, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nan Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhipeng Xi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rongrong Deng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiyu Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ran Kang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Orthopedics, Nanjing Lishui Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Lin Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
An Alternative Low-Cost Strategy for Simultaneous Sensitive Detection of Adjacent ESR1 Mutations in Single Circulating Tumor Cell. JOURNAL OF ANALYSIS AND TESTING 2022. [DOI: 10.1007/s41664-022-00216-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
9
|
Palacio-Castañeda V, Velthuijs N, Le Gac S, Verdurmen WPR. Oxygen control: the often overlooked but essential piece to create better in vitro systems. LAB ON A CHIP 2022; 22:1068-1092. [PMID: 35084420 DOI: 10.1039/d1lc00603g] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Variations in oxygen levels play key roles in numerous physiological and pathological processes, but are often not properly controlled in in vitro models, introducing a significant bias in experimental outcomes. Recent developments in microfluidic technology have introduced a paradigm shift by providing new opportunities to better mimic physiological and pathological conditions, which is achieved by both regulating and monitoring oxygen levels at the micrometre scale in miniaturized devices. In this review, we first introduce the nature and relevance of oxygen-dependent pathways in both physiological and pathological contexts. Subsequently, we discuss strategies to control oxygen in microfluidic devices, distinguishing between engineering approaches that operate at the device level during its fabrication and chemical approaches that involve the active perfusion of fluids oxygenated at a precise level or supplemented with oxygen-producing or oxygen-scavenging materials. In addition, we discuss readout approaches for monitoring oxygen levels at the cellular and tissue levels, focusing on electrochemical and optical detection schemes for high-resolution measurements directly on-chip. An overview of different applications in which microfluidic devices have been utilized to answer biological research questions is then provided. In the final section, we provide our vision for further technological refinements of oxygen-controlling devices and discuss how these devices can be employed to generate new fundamental insights regarding key scientific problems that call for emulating oxygen levels as encountered in vivo. We conclude by making the case that ultimately emulating physiological or pathological oxygen levels should become a standard feature in all in vitro cell, tissue, and organ models.
Collapse
Affiliation(s)
- Valentina Palacio-Castañeda
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| | - Niels Velthuijs
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, Organ-on-a-chip Centre, University of Twente, Postbus 217, 7500 AE Enschede, The Netherlands.
| | - Wouter P R Verdurmen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Kalita-de Croft P, Joshi V, Saunus JM, Lakhani SR. Emerging Biomarkers for Diagnosis, Prevention and Treatment of Brain Metastases-From Biology to Clinical Utility. Diseases 2022; 10:11. [PMID: 35225863 PMCID: PMC8884016 DOI: 10.3390/diseases10010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Primary malignancies of the lung, skin (melanoma), and breast have higher propensity for metastatic spread to the brain. Advances in molecular tumour profiling have aided the development of targeted therapies, stereotactic radiotherapy, and immunotherapy, which have led to some improvement in patient outcomes; however, the overall prognosis remains poor. Continued research to identify new prognostic and predictive biomarkers is necessary to further impact patient outcomes, as this will enable better risk stratification at the point of primary cancer diagnosis, earlier detection of metastatic deposits (for example, through surveillance), and more effective systemic treatments. Brain metastases exhibit considerable inter- and intratumoural heterogeneity-apart from distinct histology, treatment history and other clinical factors, the metastatic brain tumour microenvironment is incredibly variable both in terms of subclonal diversity and cellular composition. This review discusses emerging biomarkers; specifically, the biological context and potential clinical utility of tumour tissue biomarkers, circulating tumour cells, extracellular vesicles, and circulating tumour DNA.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Vaibhavi Joshi
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Jodi M. Saunus
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
- Pathology Queensland, The Royal Brisbane and Women’s Hospital Herston, Herston, QLD 4029, Australia
| |
Collapse
|
11
|
He W, Wang Q, Tian X, Pan G. Recapitulating dynamic ECM ligand presentation at biomaterial interfaces: Molecular strategies and biomedical prospects. EXPLORATION (BEIJING, CHINA) 2022; 2:20210093. [PMID: 37324582 PMCID: PMC10191035 DOI: 10.1002/exp.20210093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 06/14/2023]
Abstract
The extracellular matrix (ECM) provides not only physical support for the tissue structural integrity, but also dynamic biochemical cues capable of regulating diverse cell behaviors and functions. Biomaterial surfaces with dynamic ligand presentation are capable of mimicking the dynamic biochemical cues of ECM, showing ECM-like functions to modulate cell behaviors. This review paper described an overview of present dynamic biomaterial interfaces by focusing on currently developed molecular strategies for dynamic ligand presentation. The paradigmatic examples for each strategy were separately discussed. In addition, the regulation of some typical cell behaviors on these dynamic biointerfaces including cell adhesion, macrophage polarization, and stem cell differentiation, and their potential applications in pathogenic cell isolation, single cell analysis, and tissue engineering are highlighted. We hope it would not only clarify a clear background of this field, but also inspire to exploit novel molecular strategies and more applications to match the increasing demand of manipulating complex cellular processes in biomedicine.
Collapse
Affiliation(s)
- Wenbo He
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Qinghe Wang
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Xiaohua Tian
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
- School of Chemistry and Chemical EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Guoqing Pan
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| |
Collapse
|
12
|
Burr R, Edd JF, Chirn B, Mishra A, Haber DA, Toner M, Maheswaran S. Negative-Selection Enrichment of Circulating Tumor Cells from Peripheral Blood Using the Microfluidic CTC-iChip. Methods Mol Biol 2022; 2471:309-321. [PMID: 35175606 DOI: 10.1007/978-1-0716-2193-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The ability to isolate and analyze rare circulating tumor cells (CTCs) holds the potential to increase our understanding of cancer evolution and allows monitoring of disease and therapeutic responses through a relatively non-invasive blood-based biopsy. While many methods have been described to isolate CTCs from the blood, the vast majority rely on size-based sorting or positive selection of CTCs based on surface markers, which introduces bias into the downstream product by making assumptions about these heterogenous cells. Here we describe a negative-selection protocol for enrichment of CTCs through removal of blood components including red blood cells, platelets, and white blood cells. This procedure results in a product that is amenable to downstream single-cell analytics including RNA-Seq, ATAC-Seq and DNA methylation, droplet digital PCR (ddPCR) for tumor specific transcripts, staining and extensive image analysis, and ex vivo culture of patient-derived CTCs.
Collapse
Affiliation(s)
- Risa Burr
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
| | - Jon F Edd
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Chirn
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
| | - Avanish Mishra
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A Haber
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA
- Howard Hughes Medical Institute, Bethesda, MD, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, MA, USA.
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Volovetskiy AB, Malinina PA, Kapitannikova AY, Smetanina SV, Kruglova IA, Maslennikova AV. Isolation of Circulating Tumor Cells from Peripheral Blood Samples of Cancer Patients Using Microfluidic Technology. Sovrem Tekhnologii Med 2021; 12:62-68. [PMID: 34796020 PMCID: PMC8596232 DOI: 10.17691/stm2020.12.6.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Indexed: 11/23/2022] Open
Abstract
The aim of the investigation was to study the potential of an innovative microfluidic technology for the isolation of circulating tumor cells (CTCs) from the peripheral blood samples of cancer patients.
Collapse
Affiliation(s)
- A B Volovetskiy
- Researcher, Laboratory of Nanotheranostics, Institute for Molecular Medicine, Biomedical Science and Technology Park; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Malaya Trubetskaya St., Moscow, 119991, Russia
| | - P A Malinina
- Clinical Resident, Department of Oncology, Radiation Therapy and Radiation Diagnostics; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - A Y Kapitannikova
- Researcher, Laboratory of Nanoteranology, Institute for Molecular Medicine, Biomedical Science and Technology Park; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Malaya Trubetskaya St., Moscow, 119991, Russia
| | - S V Smetanina
- Head of the Сytological Laboratory; Nizhny Novgorod Regional Oncologic Dispensary, 11/1 Delovaya St., Nizhny Novgorod, 603163, Russia
| | - I A Kruglova
- Physician of Clinical Laboratory Diagnostics; City Hospital No.35, 47 Respublikanskaya St., Nizhny Novgorod, 603089, Russia
| | - A V Maslennikova
- Head of the Department of Oncology, Radiation Therapy and Radiation Diagnostics; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Professor, Department of Biophysics National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| |
Collapse
|
14
|
Li R, Gong Z, Liu Y, Zhao X, Guo S. Detection of circulating tumor cells and single cell extraction technology: principle, effect and application prospect. NANO FUTURES 2021; 5:032002. [DOI: 10.1088/2399-1984/ac1325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Bucheli OTM, Sigvaldadóttir I, Eyer K. Measuring single-cell protein secretion in immunology: Technologies, advances, and applications. Eur J Immunol 2021; 51:1334-1347. [PMID: 33734428 PMCID: PMC8252417 DOI: 10.1002/eji.202048976] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/12/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
The dynamics, nature, strength, and ultimately protective capabilities of an active immune response are determined by the extracellular constitution and concentration of various soluble factors. Generated effector cells secrete such mediators, including antibodies, chemo‐ and cytokines to achieve functionality. These secreted factors organize the individual immune cells into functional tissues, initiate, orchestrate, and regulate the immune response. Therefore, a single‐cell resolved analysis of protein secretion is a valuable tool for studying the heterogeneity and functionality of immune cells. This review aims to provide a comparative overview of various methods to characterize immune reactions by measuring single‐cell protein secretion. Spot‐based and cytometry‐based assays, such as ELISpot and flow cytometry, respectively, are well‐established methods applied in basic research and clinical settings. Emerging novel technologies, such as microfluidic platforms, offer new ways to measure and exploit protein secretion in immune reactions. Further technological advances will allow the deciphering of protein secretion in immunological responses with unprecedented detail, linking secretion to functionality. Here, we summarize the development and recent advances of tools that allow the analysis of protein secretion at the single‐cell level, and discuss and contrast their applications within immunology.
Collapse
Affiliation(s)
- Olivia T M Bucheli
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| | - Ingibjörg Sigvaldadóttir
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| | - Klaus Eyer
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
16
|
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
|
17
|
Kumar T, Soares RRG, Ali Dholey L, Ramachandraiah H, Aval NA, Aljadi Z, Pettersson T, Russom A. Multi-layer assembly of cellulose nanofibrils in a microfluidic device for the selective capture and release of viable tumor cells from whole blood. NANOSCALE 2020; 12:21788-21797. [PMID: 33103175 DOI: 10.1039/d0nr05375a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
According to reports by the World Health Organization (WHO), cancer-related deaths reached almost 10 million in 2018. Nearly 65% of these deaths occurred in low- to middle-income countries, a trend that is bound to increase since cancer diagnostics are not currently considered a priority in resource-limited settings (RLS). Thus, cost-effective and specific cancer screening and diagnostics tools are in high demand, particularly in RLS. The selective isolation and up-concentration of rare cells while maintaining cell viability and preventing phenotypic changes is a powerful tool to allow accurate and sensitive downstream analysis. Here, multi-layer cellulose nanofibril-based coatings functionalized with anti-EpCAM antibodies on the surface of disposable microfluidic devices were optimized for specific capture of target cells, followed by efficient release without significant adverse effects. HCT 116 colon cancer cells were captured in a single step with >97% efficiency at 41.25 μL min-1 and, when spiked in whole blood, an average enrichment factor of ∼200-fold relative to white blood cells was achieved. The release of cells was performed by enzymatic digestion of the cellulose nanofibrils which had a negligible impact on cell viability. In particular, >80% of the cells were recovered with at least 97% viability in less than 30 min. Such performance paves the way to expand and improve clinical diagnostic applications by simplifying the isolation of circulating tumor cells (CTCs) and other rare cells directly from whole blood.
Collapse
Affiliation(s)
- Tharagan Kumar
- KTH Royal Institute of Technology, Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, Solna, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Yang L, George J, Wang J. Deep Profiling of Cellular Heterogeneity by Emerging Single-Cell Proteomic Technologies. Proteomics 2020; 20:e1900226. [PMID: 31729152 PMCID: PMC7225074 DOI: 10.1002/pmic.201900226] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/14/2019] [Indexed: 12/20/2022]
Abstract
The ability to comprehensively profile cellular heterogeneity in functional proteome is crucial in advancing the understanding of cell behavior, organism development, and disease mechanisms. Conventional bulk measurement by averaging the biological responses across a population often loses the information of cellular variations. Single-cell proteomic technologies are becoming increasingly important to understand and discern cellular heterogeneity. The well-established methods for single-cell protein analysis based on flow cytometry and fluorescence microscopy are limited by the low multiplexing ability owing to the spectra overlap of fluorophores for labeling antibodies. Recent advances in mass spectrometry (MS), microchip, and reiterative staining-based techniques for single-cell proteomics have enabled the evaluation of cellular heterogeneity with high throughput, increased multiplexity, and improved sensitivity. In this review, the principles, developments, advantages, and limitations of these advanced technologies in analysis of single-cell proteins, along with their biological applications to study cellular heterogeneity, are described. At last, the remaining challenges, possible strategies, and future opportunities that will facilitate the improvement and broad applications of single-cell proteomic technologies in cell biology and medical research are discussed.
Collapse
Affiliation(s)
- Liwei Yang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Justin George
- Department of Chemistry, State University of New York, University at Albany, Albany, NY 12222
| | - Jun Wang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY 11794
| |
Collapse
|
19
|
Armbrecht L, Rutschmann O, Szczerba BM, Nikoloff J, Aceto N, Dittrich PS. Quantification of Protein Secretion from Circulating Tumor Cells in Microfluidic Chambers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903237. [PMID: 32537399 PMCID: PMC7284199 DOI: 10.1002/advs.201903237] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 05/05/2023]
Abstract
Cancer cells can be released from a cancerous lesion and migrate into the circulatory system, from whereon they may form metastases at distant sites. Today, it is possible to infer cancer progression and treatment efficacy by determining the number of circulating tumor cells (CTCs) in the patient's blood at multiple time points; further valuable information about CTC phenotypes remains inaccessible. In this article, a microfluidic method for integrated capture, isolation, and analysis of membrane markers as well as quantification of proteins secreted by single CTCs and CTC clusters is introduced. CTCs are isolated from whole blood with extraordinary efficiencies above 95% using dedicated trapping structures that allow co-capture of functionalized magnetic beads to assess protein secretion. The patform is tested with multiple breast cancer cell lines spiked into human blood and mouse-model-derived CTCs. In addition to immunostaining, the secretion level of granulocyte growth stimulating factor (G-CSF), which is shown to be involved in neutrophil recruitment, is quantified The bead-based assay provides a limit of detection of 1.5 ng mL-1 or less than 3700 molecules per cell. Employing barcoded magnetic beads, this platform can be adapted for multiplexed analysis and can enable comprehensive functional CTC profiling in the future.
Collapse
Affiliation(s)
- Lucas Armbrecht
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Ophélie Rutschmann
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Barbara Maria Szczerba
- Department of BiomedicineCancer Metastasis LabUniversity of Basel and University Hospital BaselMattenstrasse 28BaselCH‐4058Switzerland
| | - Jonas Nikoloff
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Nicola Aceto
- Department of BiomedicineCancer Metastasis LabUniversity of Basel and University Hospital BaselMattenstrasse 28BaselCH‐4058Switzerland
| | - Petra S. Dittrich
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| |
Collapse
|
20
|
Wan L, Neumann CA, LeDuc PR. Tumor-on-a-chip for integrating a 3D tumor microenvironment: chemical and mechanical factors. LAB ON A CHIP 2020; 20:873-888. [PMID: 32025687 PMCID: PMC7067141 DOI: 10.1039/c9lc00550a] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Tumor progression, including metastasis, is significantly influenced by factors in the tumor microenvironment (TME) such as mechanical force, shear stress, chemotaxis, and hypoxia. At present, most cancer studies investigate tumor metastasis by conventional cell culture methods and animal models, which are limited in data interpretation. Although patient tissue analysis, such as human patient-derived xenografts (PDX), can provide important clinical relevant information, they may not be feasible for functional studies as they are costly and time-consuming. Thus, in vitro three-dimensional (3D) models are rapidly being developed that mimic TME and allow functional investigations of metastatic mechanisms and drug responses. One of those new 3D models is tumor-on-a-chip technology that provides a powerful in vitro platform for cancer research, with the ability to mimic the complex physiological architecture and precise spatiotemporal control. Tumor-on-a-chip technology can provide integrated features including 3D scaffolding, multicellular culture, and a vasculature system to simulate dynamic flow in vivo. Here, we review a select set of recent achievements in tumor-on-a-chip approaches and present potential directions for tumor-on-a-chip systems in the future for areas including mechanical and chemical mimetic systems. We also discuss challenges and perspectives in both biological factors and engineering methods for tumor-on-a-chip progress. These approaches will allow in the future for the tumor-on-a-chip systems to test therapeutic approaches for individuals through using their cancerous cells gathered through approaches like biopsies, which then will contribute toward personalized medicine treatments for improving their outcomes.
Collapse
Affiliation(s)
- L Wan
- Department of Mechanical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213 US.
| | - C A Neumann
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, Magee Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213 US.
| | - P R LeDuc
- Department of Mechanical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213 US.
| |
Collapse
|
21
|
Fung CW, Chan SN, Wu AR. Microfluidic single-cell analysis-Toward integration and total on-chip analysis. BIOMICROFLUIDICS 2020; 14:021502. [PMID: 32161631 PMCID: PMC7060088 DOI: 10.1063/1.5131795] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
Various types of single-cell analyses are now extensively used to answer many biological questions, and with this growth in popularity, potential drawbacks to these methods are also becoming apparent. Depending on the specific application, workflows can be laborious, low throughput, and run the risk of contamination. Microfluidic designs, with their advantages of being high throughput, low in reaction volume, and compatible with bio-inert materials, have been widely used to improve single-cell workflows in all major stages of single-cell applications, from cell sorting to lysis, to sample processing and readout. Yet, designing an integrated microfluidic chip that encompasses the entire single-cell workflow from start to finish remains challenging. In this article, we review the current microfluidic approaches that cover different stages of processing in single-cell analysis and discuss the prospects and challenges of achieving a full integrated workflow to achieve total single-cell analysis in one device.
Collapse
Affiliation(s)
- Cheuk Wang Fung
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shek Nga Chan
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Angela Ruohao Wu
- Author to whom correspondence should be addressed:. Tel.: +852 3469-2577
| |
Collapse
|
22
|
Sukumar P, Deliorman M, Brimmo AT, Alnemari R, Elsori D, Chen W, Qasaimeh MA. Airplug-mediated isolation and centralization of single T cells in rectangular microwells for biosensing. ADVANCED THERAPEUTICS 2020; 3:1900085. [PMID: 33117882 PMCID: PMC7591138 DOI: 10.1002/adtp.201900085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Indexed: 11/09/2022]
Abstract
Sorting cells in a single cell per microwell format is of great interest to basic biology studies, biotherapeutics, and biosensing including cell phenotyping. For instance, isolation of individual immune T cells in rectangular microwells has been shown to empower the multiplex cytokine profiling at the single cell level for therapeutics applications. The present study, however, shows that there is an existing bias in temporal cytokine sensing that originates from random "unpredicted" positions of loaded cells within the rectangular microwells. To eliminate this bias, the isolated cells need to be well-aligned with each other and relative to the sensing elements. Hence, an approach that utilizes the in situ formation and release of airplugs to localize cells towards the center of the rectangular microwells is reported. The chip includes 2250 microwells (each 500 × 50 × 20 μm3) arranged in 9 rows. Results showed 20% efficiency in trapping single T cells per microwells, where cells are localized within ±3% of the center of microwells. The developed platform could provide real-time dynamic and unbiased multiplex cytokine detection from single T cells for phenotyping and biotherapeutics studies.
Collapse
Affiliation(s)
- Pavithra Sukumar
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Muhammedin Deliorman
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Ayoola T Brimmo
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Roaa Alnemari
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Deena Elsori
- Department of Applied Sciences and Mathematics, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, UAE
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Mohammad A Qasaimeh
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| |
Collapse
|
23
|
Godino N, Pfisterer F, Gerling T, Guernth-Marschner C, Duschl C, Kirschbaum M. Combining dielectrophoresis and computer vision for precise and fully automated single-cell handling and analysis. LAB ON A CHIP 2019; 19:4016-4020. [PMID: 31746875 DOI: 10.1039/c9lc00800d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
With the advent of single-cell technologies comes the necessity for efficient protocols to process single cells. We combine dielectrophoresis with open source computer vision programming to automatically control the trajectories of single cells inside a microfluidic device. Using real-time image analysis, individual cells are automatically selected, isolated and spatially arranged.
Collapse
Affiliation(s)
- Neus Godino
- Fraunhofer IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | - Felix Pfisterer
- Fraunhofer IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | - Tobias Gerling
- Fraunhofer IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | | | - Claus Duschl
- Fraunhofer IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | | |
Collapse
|
24
|
Abstract
Abstract
Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. This can be achieved by leveraging omics information for accurate molecular characterization of tumors. Tumor tissue biopsies are currently the main source of information for molecular profiling. However, biopsies are invasive and limited in resolving spatiotemporal heterogeneity in tumor tissues. Alternative non-invasive liquid biopsies can exploit patient’s body fluids to access multiple layers of tumor-specific biological information (genomes, epigenomes, transcriptomes, proteomes, metabolomes, circulating tumor cells, and exosomes). Analysis and integration of these large and diverse datasets using statistical and machine learning approaches can yield important insights into tumor biology and lead to discovery of new diagnostic, predictive, and prognostic biomarkers. Translation of these new diagnostic tools into standard clinical practice could transform oncology, as demonstrated by a number of liquid biopsy assays already entering clinical use. In this review, we highlight successes and challenges facing the rapidly evolving field of cancer biomarker research.
Lay Summary
Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. The discovery of biomarkers for precision oncology has been accelerated by high-throughput experimental and computational methods, which can inform fine-grained characterization of tumors for clinical decision-making. Moreover, advances in the liquid biopsy field allow non-invasive sampling of patient’s body fluids with the aim of analyzing circulating biomarkers, obviating the need for invasive tumor tissue biopsies. In this review, we highlight successes and challenges facing the rapidly evolving field of liquid biopsy cancer biomarker research.
Collapse
|
25
|
Lim SB, Lim CT, Lim WT. Single-Cell Analysis of Circulating Tumor Cells: Why Heterogeneity Matters. Cancers (Basel) 2019; 11:cancers11101595. [PMID: 31635038 PMCID: PMC6826423 DOI: 10.3390/cancers11101595] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/31/2022] Open
Abstract
Unlike bulk-cell analysis, single-cell approaches have the advantage of assessing cellular heterogeneity that governs key aspects of tumor biology. Yet, their applications to circulating tumor cells (CTCs) are relatively limited, due mainly to the technical challenges resulting from extreme rarity of CTCs. Nevertheless, recent advances in microfluidics and immunoaffinity enrichment technologies along with sequencing platforms have fueled studies aiming to enrich, isolate, and sequence whole genomes of CTCs with high fidelity across various malignancies. Here, we review recent single-cell CTC (scCTC) sequencing efforts, and the integrated workflows, that have successfully characterized patient-derived CTCs. We examine how these studies uncover DNA alterations occurring at multiple molecular levels ranging from point mutations to chromosomal rearrangements from a single CTC, and discuss their cellular heterogeneity and clinical consequences. Finally, we highlight emerging strategies to address key challenges currently limiting the translation of these findings to clinical practice.
Collapse
Affiliation(s)
- Su Bin Lim
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore 117456, Singapore.
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore.
| | - Chwee Teck Lim
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore 117456, Singapore.
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore.
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore.
| | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore.
- Office of Academic and Clinical Development, Duke-NUS Medical School, Singapore 169857, Singapore.
- IMCB NCC MPI Singapore Oncogenome Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore.
| |
Collapse
|
26
|
Abstract
Microfluidics is an emerging field in diagnostics that allows for extremely precise fluid control and manipulation, enabling rapid and high-throughput sample processing in integrated micro-scale medical systems. These platforms are well-suited for both standard clinical settings and point-of-care applications. The unique features of microfluidics-based platforms make them attractive for early disease diagnosis and real-time monitoring of the disease and therapeutic efficacy. In this chapter, we will first provide a background on microfluidic fundamentals, microfluidic fabrication technologies, microfluidic reactors, and microfluidic total-analysis-systems. Next, we will move into a discussion on the clinical applications of existing and emerging microfluidic platforms for blood analysis, and for diagnosis and monitoring of cancer and infectious disease. Together, this chapter should elucidate the potential that microfluidic systems have in the development of effective diagnostic technologies through a review of existing technologies and promising directions.
Collapse
Affiliation(s)
- Alison Burklund
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Amogha Tadimety
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Yuan Nie
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Nanjing Hao
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States; Norris Cotton Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States.
| |
Collapse
|
27
|
Abstract
As an alternative target to surgically resected tissue specimens, liquid biopsy has gained much attention over the past decade. Of the various circulating biomarkers, circulating tumor cells (CTCs) have particularly opened new windows into the metastatic cascade, with their functional, biochemical, and biophysical properties. Given the extreme rarity of intact CTCs and the associated technical challenges, however, analyses have been limited to bulk-cell strategies, missing out on clinically significant sources of information from cellular heterogeneity. With recent technological developments, it is now possible to probe genetic material of CTCs at the single-cell resolution to study spatial and temporal dynamics in circulation. Here, we discuss recent transcriptomic profiling efforts that enabled single-cell characterization of patient-derived CTCs spanning diverse cancer types. We further highlight how expression data of these putative biomarkers have advanced our understanding of metastatic spectrum and provided a basis for the development of CTC-based liquid biopsies to track, monitor, and predict the efficacy of therapy and any emergent resistance.
Collapse
|
28
|
Liu T, Ma Q, Zhang Y, Wang X, Xu K, Yan K, Dong W, Fan Q, Zhang Y, Qiu X. Self-seeding circulating tumor cells promote the proliferation and metastasis of human osteosarcoma by upregulating interleukin-8. Cell Death Dis 2019; 10:575. [PMID: 31366916 PMCID: PMC6668432 DOI: 10.1038/s41419-019-1795-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 01/21/2023]
Abstract
Most circulating tumor cells (CTCs) die during the process of metastasis, but self-seeding CTCs can invade the primary tumor or form clinically meaningful metastases. This study aimed to evaluate the capacity of self-seeding CTCs to promote osteosarcoma growth and lung metastasis and to clarify the specific role of interleukin (IL)-8 in CTC self-seeding. We successfully isolated and cultured self-seeding CTCs through a self-seeding nude mouse model established using green fluorescent protein (GFP)-labeled F5M2 cells and found that self-seeding CTCs exhibit increased cellular proliferation, migration, and invasion in vitro, increased tumor growth and lung metastasis in mice, and increased IL-8 expression. Furthermore, suppressing IL-8 inhibited tumor growth and metastasis and reduced CTC seeding in primary tumors in vitro and in vivo. In osteosarcoma patients, IL-8 levels significantly correlated with the Enneking stage and metastasis. These findings demonstrate that self-seeding osteosarcoma CTCs can promote tumor growth and lung metastasis through IL-8. Their increased metastatic potential and elevated IL-8 expression suggest a novel strategy for future therapeutic interventions to prevent osteosarcoma progression and metastasis.
Collapse
Affiliation(s)
- Tao Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, Shanxi, China.,Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Qiong Ma
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Yinglong Zhang
- Department of Orthopaedics, First Affiliated Hospital of PLA General Hospital, 100048, Beijing, China
| | - Xin Wang
- Rehabilitation Center of Lintong Sanatorium of PLA, No. 32 Huaqing Road, Lintong District, Xi'an, 710600, Shanxi, China
| | - Kui Xu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Kang Yan
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Wengang Dong
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Qingyu Fan
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, Shanxi, China.
| | - Xiuchun Qiu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710032, Shanxi, China.
| |
Collapse
|
29
|
Chen D, Wen J, Zeng S, Ma H. DNA fragment‐assisted microfluidic chip for capture and release of circulating tumor cells. Electrophoresis 2019; 40:2845-2852. [DOI: 10.1002/elps.201900165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Dengyi Chen
- College of Laboratory MedicineDalian Medical University Dalian Liaoning Province P. R. China
| | - Jing Wen
- College of Laboratory MedicineDalian Medical University Dalian Liaoning Province P. R. China
| | - Shaojiang Zeng
- College of Laboratory MedicineDalian Medical University Dalian Liaoning Province P. R. China
| | - Huipeng Ma
- College of Laboratory MedicineDalian Medical University Dalian Liaoning Province P. R. China
| |
Collapse
|
30
|
Kshitiz, Ellison DD, Suhail Y, Afzal J, Woo L, Kilic O, Spees J, Levchenko A. Dynamic secretome of bone marrow-derived stromal cells reveals a cardioprotective biochemical cocktail. Proc Natl Acad Sci U S A 2019; 116:14374-14383. [PMID: 31239339 PMCID: PMC6628676 DOI: 10.1073/pnas.1902598116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transplanted stromal cells have demonstrated considerable promise as therapeutic agents in diverse disease settings. Paracrine signaling can be an important mediator of these therapeutic effects at the sites of acute or persistent injury and inflammation. As many stromal cell types, including bone marrow-derived stromal cells (BMSCs), display tissue-specific responses, there is a need to explore their secretory dynamics in the context of tissue and injury type. Paracrine signals are not static, and could encode contextual dynamics in the kinetic changes of the concentrations of the secreted ligands. However, precise measurement of dynamic and context-specific cellular secretory signatures, particularly in adherent cells, remains challenging. Here, by creating an experimental and computational analysis platform, we reconstructed dynamic secretory signatures of cells based on a very limited number of time points. By using this approach, we demonstrate that the secretory signatures of CD133-positive BMSCs are uniquely defined by distinct biological contexts, including signals from injured cardiac cells undergoing oxidative stress, characteristic of cardiac infarction. Furthermore, we show that the mixture of recombinant factors reproducing the dynamics of BMSC-generated secretion can mediate a highly effective rescue of cells injured by oxidative stress and an improved cardiac output. These results support the importance of the dynamic multifactorial paracrine signals in mediating remedial effects of stromal stem cells, and pave the way for stem cell-inspired cell-free treatments of cardiac and other injuries.
Collapse
Affiliation(s)
- Kshitiz
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516;
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030
| | - David D Ellison
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030
| | - Junaid Afzal
- Department of Cardiology, University of California, San Francisco, CA 94115
| | - Laura Woo
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Onur Kilic
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516
| | - Jeffrey Spees
- Department of Cellular Molecular and Biomedical Sciences, University of Vermont, Burlington, VT 05405
| | - Andre Levchenko
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516;
| |
Collapse
|
31
|
Chen Z, Chen JJ, Fan R. Single-Cell Protein Secretion Detection and Profiling. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:431-449. [PMID: 30978293 DOI: 10.1146/annurev-anchem-061318-115055] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Secreted proteins play important roles in mediating various biological processes such as cell-cell communication, differentiation, migration, and homeostasis at the population or tissue level. Here, we review bioanalytical technologies and devices for detecting protein secretions from single cells. We begin by discussing conventional approaches followed by detailing the latest advances in microengineered systems for detecting single-cell protein secretions with an emphasis on multiplex measurement. These platforms include droplet microfluidics, micro-/nanowell-based assays, and microchamber-based assays, among which the advantages and limitations are compared. Microscale systems also enable the tracking of protein secretion dynamics in single cells, further empowering the study of the cell-cell communication network. Looking forward, we discuss the remaining challenges and future opportunities that will transform basic research of cellular secretion functions at the systems level and the clinical applications for immune monitoring and cancer treatment.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520, USA;
| | - Jonathan J Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520, USA;
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520, USA;
- Yale Cancer Center, Yale Stem Cell Center, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
32
|
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
|
33
|
Liu Y, Chen X, Zhang Y, Liu J. Advancing single-cell proteomics and metabolomics with microfluidic technologies. Analyst 2019; 144:846-858. [PMID: 30351310 DOI: 10.1039/c8an01503a] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent advances in single-cell analysis have unraveled substantial heterogeneity among seemingly identical cells at genomic and transcriptomic levels. These discoveries have urged scientists to develop new tools that are capable of investigating single cells from a broader set of "omics". Proteomics and metabolomics, for instance, are of particular interest as they are closely correlated with a dynamic picture of cellular behaviors and phenotypic identities. The development of such tools requires highly efficient isolation and processing of a large number of individual cells, where techniques such as microfluidics are extremely useful. Here, we review the recent advances in single-cell proteomics and metabolomics, with a focus on microfluidics-based platforms. We highlight a vast array of emerging microfluidic formats for single-cell isolation and manipulation, and how the state-of-the-art analytical tools are coupled with such platforms for proteomic and metabolomic profiling.
Collapse
Affiliation(s)
- Yifan Liu
- Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu Province 215123, China.
| | | | | | | |
Collapse
|
34
|
Andree KC, Mentink A, Nguyen AT, Goldsteen P, van Dalum G, Broekmaat JJ, van Rijn CJM, Terstappen LWMM. Tumor cell capture from blood by flowing across antibody-coated surfaces. LAB ON A CHIP 2019; 19:1006-1012. [PMID: 30762848 DOI: 10.1039/c8lc01158c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The load of circulating tumor cells (CTC) is related to poor outcomes in cancer patients. A sufficient number of these cells would enable a full characterization of the cancer. An approach to probe larger blood volumes, allowing for the detection of more of these very rare CTC, is the use of leukapheresis. Currently available techniques allow only the analysis of a small portion of leukapheresis products. Here, we present a method that uses flow rather than static conditions which allows processing of larger volumes. We evaluated the conditions needed to isolate tumor cells from blood while passing antibody coated surfaces. Results show that our set-up efficiently captures cancer cells from whole blood. Results show that the optimal velocity at which cells are captured from blood is 0.6 mm s-1. Also, it can be concluded that the VU1D9 antibody targeting the EpCAM antigen has very high capture efficiency. When using an antibody that does not capture 100% of all cells, combining multiple antibodies on the capture surface is very beneficial leading to an increase in cell capture and is therefore worthwhile considering in any cancer cell capture methodology.
Collapse
Affiliation(s)
- K C Andree
- Medical Cell Biophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Wang C, Yang L, Wang Z, He J, Shi Q. Highly multiplexed profiling of cell surface proteins on single circulating tumor cells based on antibody and cellular barcoding. Anal Bioanal Chem 2019; 411:5373-5382. [PMID: 30820628 DOI: 10.1007/s00216-019-01666-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 01/26/2019] [Accepted: 02/04/2019] [Indexed: 11/27/2022]
Abstract
Circulating tumor cells (CTCs) are extraordinarily rare in blood samples and represent a real-time "liquid biopsy" of tumors. Although genetic and transcriptional sequencing of single CTCs has been reported, these methods fail to provide phenotypic and functional information of CTCs such as protein levels of surface proteins. Studies of single-cell proteomic assays of CTCs have been rare because of a lack of single-cell proteomic methods to handle and analyze rare cells in a high background of non-target cells with high sensitivity, throughput, and multiplexing capacity. Here, we develop a microchip-assisted single-cell proteomic method for profiling surface proteins of CTCs based on antibody and cellular DNA barcoding strategy. We combine DNA-encoded antibody tags and cell indexes to profile 15 proteins in ~ 100 single rare cells simultaneously, and use high-throughput sequencing as the readout to generate surface protein profiles of CTCs according to their cell indexes and antibody-derived protein barcodes. A 6400-well microchip and the automated puncher are used to rapidly retrieve single CTCs from enriched CTC population with minimal cell loss (~ 10%). This technological platform integrates reliable isolation and proteomic analysis of single CTCs and can be extendable to ~ 100 proteins in hundreds of rare cells with single-cell precision.
Collapse
Affiliation(s)
- Chunying Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Minhang District, Shanghai, 200240, China
| | - Liu Yang
- Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Zhuo Wang
- Minhang Branch, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 201100, China
| | - Jianjun He
- Abmart (Shanghai) Inc., Shanghai, 200233, China
| | - Qihui Shi
- Minhang Branch, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 201100, China.
| |
Collapse
|
36
|
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
|
37
|
He E, Cai L, Zheng F, Zhou Q, Guo D, Zhou Y, Zhang X, Li Z. Rapid Quantitative Fluorescence Detection of Copper Ions with Disposable Microcapsule Arrays Utilizing Functional Nucleic Acid Strategy. Sci Rep 2019; 9:36. [PMID: 30631123 PMCID: PMC6328549 DOI: 10.1038/s41598-018-36842-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/14/2018] [Indexed: 12/21/2022] Open
Abstract
In this work, an economical and easy-to-use microcapsule array fabricated by ice printing technique has been realized for ultrasensitive fluorescence quantification of copper ions employing functional nucleic acid strategy. With ice printing, the detection reagents are sealed by polystyrene (PS) film isolation and photopolymer, which guarantees a stable and contamination-free environment for functional nucleic acid reaction. Our microcapsule arrays have shown long-term stability (20 days) under -20 °C storage in frozen form before use. During the Cu2+ on-site detection, 1 μL sample is simply injected into the thawy microcapsule by a microliter syringe under room temperature, and after 20 minutes the fluorescence result can be obtained by an LED transilluminator. This method can realize the detection limit to 100 nM (100 fmol/μL) with high specificity.
Collapse
Affiliation(s)
- Enqi He
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.,Center for Nano and Micro Mechanics, Tsinghua University, Beijing, 100084, China
| | - Liangyuan Cai
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry, Peking University, Beijing, 100871, China
| | - Fengyi Zheng
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Institute of Microelectronics, Peking University, Beijing, 100871, China
| | - Qianyu Zhou
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry, Peking University, Beijing, 100871, China
| | - Dan Guo
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yinglin Zhou
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry, Peking University, Beijing, 100871, China.
| | - Xinxiang Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry, Peking University, Beijing, 100871, China.
| | - Zhihong Li
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Institute of Microelectronics, Peking University, Beijing, 100871, China. .,Center for Nano and Micro Mechanics, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
38
|
Markiewicz A, Topa J, Nagel A, Skokowski J, Seroczynska B, Stokowy T, Welnicka-Jaskiewicz M, Zaczek AJ. Spectrum of Epithelial-Mesenchymal Transition Phenotypes in Circulating Tumour Cells from Early Breast Cancer Patients. Cancers (Basel) 2019; 11:cancers11010059. [PMID: 30634453 PMCID: PMC6356662 DOI: 10.3390/cancers11010059] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 01/05/2023] Open
Abstract
Circulating tumour cells (CTCs) can provide valuable prognostic information in a number of epithelial cancers. However, their detection is hampered due to their molecular heterogeneity, which can be induced by the epithelial-mesenchymal transition (EMT) process. Therefore, current knowledge about CTCs from clinical samples is often limited due to an inability to isolate wide spectrum of CTCs phenotypes. In the current work, we aimed at isolation and molecular characterization of CTCs with different EMT status in order to establish their clinical significance in early breast cancer patients. We have obtained CTCs-enriched blood fraction from 83 breast cancer patients in which we have tested the expression of epithelial, mesenchymal and general breast cancer CTCs markers (MGB1/HER2/CK19/CDH1/CDH2/VIM/PLS3), cancer stem cell markers (CD44, NANOG, ALDH1, OCT-4, CD133) and cluster formation gene (plakoglobin). We have shown that in the CTCs-positive patients, epithelial, epithelial-mesenchymal and mesenchymal CTCs markers were detected at a similar rate (in 28%, 24% and 24%, respectively). Mesenchymal CTCs were characterized by the most aggressive phenotype (significantly higher expression of CXCR4, uPAR, CD44, NANOG, p < 0.05 for all), presence of lymph node metastases (p = 0.043), larger tumour size (p = 0.023) and 7.33 higher risk of death in the multivariate analysis (95% CI 1.06–50.41, p = 0.04). Epithelial-mesenchymal subtype, believed to correspond to highly plastic and aggressive state, did not show significant impact on survival. Gene expression profile of samples with epithelial-mesenchymal CTCs group resembled pure epithelial or pure mesenchymal phenotypes, possibly underlining degree of EMT activation in particular patient’s sample. Molecular profiling of CTCs EMT phenotype provides more detailed and clinically informative results, proving the role of EMT in malignant cancer progression in early breast cancer.
Collapse
Affiliation(s)
- Aleksandra Markiewicz
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology of the University of Gdansk and Medical University of Gdansk, 80-211 Gdańsk, Poland.
| | - Justyna Topa
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology of the University of Gdansk and Medical University of Gdansk, 80-211 Gdańsk, Poland.
| | - Anna Nagel
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology of the University of Gdansk and Medical University of Gdansk, 80-211 Gdańsk, Poland.
| | - Jaroslaw Skokowski
- Department of Surgical Oncology, Medical University of Gdansk, 80-210 Gdańsk, Poland.
- Department of Medical Laboratory Diagnostics-Biobank, Medical University of Gdansk, 80-210 Gdańsk, Poland.
- Biobanking and Biomolecular Resources Research Infrastructure (BBMRI.PL), 80-210 Gdansk, Poland.
| | - Barbara Seroczynska
- Department of Medical Laboratory Diagnostics-Biobank, Medical University of Gdansk, 80-210 Gdańsk, Poland.
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | | | - Anna J Zaczek
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology of the University of Gdansk and Medical University of Gdansk, 80-211 Gdańsk, Poland.
| |
Collapse
|
39
|
Wu J, Lin JM. Microfluidic Technology for Single-Cell Capture and Isolation. MICROFLUIDICS FOR SINGLE-CELL ANALYSIS 2019. [DOI: 10.1007/978-981-32-9729-6_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Tellez-Gabriel M, Cochonneau D, Cadé M, Jubellin C, Heymann MF, Heymann D. Circulating Tumor Cell-Derived Pre-Clinical Models for Personalized Medicine. Cancers (Basel) 2018; 11:cancers11010019. [PMID: 30586936 PMCID: PMC6356998 DOI: 10.3390/cancers11010019] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022] Open
Abstract
The main cause of death from cancer is associated with the development of metastases, resulting from the inability of current therapies to cure patients at metastatic stages. Generating preclinical models to better characterize the evolution of the disease is thus of utmost importance, in order to implement effective new cancer biomarkers and therapies. Circulating Tumor Cells (CTCs) are good candidates for generating preclinical models, making it possible to follow up the spatial and temporal heterogeneity of tumor tissues. This method is a non-invasive liquid biopsy that can be obtained at any stage of the disease. It partially summarizes the molecular heterogeneity of the corresponding tumors at a given time. Here, we discuss the CTC-derived models that have been generated so far, from simplified 2D cultures to the most complex CTC-derived explants (CDX models). We highlight the challenges and strengths of these preclinical tools, as well as some of the recent studies published using these models.
Collapse
Affiliation(s)
- Marta Tellez-Gabriel
- RNA and Molecular Pathology Research Group, Department of Medical Biology, The Artic University of Norway, N-9037 Tromsø, Norway.
| | - Denis Cochonneau
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
| | - Marie Cadé
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
| | - Camille Jubellin
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
| | - Marie-Françoise Heymann
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
| | - Dominique Heymann
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
- Department of Oncology & Metabolism, The Medical School, Beech Hill Road, Sheffield S10 2RX, UK.
| |
Collapse
|
41
|
Shashni B, Matsuura H, Saito R, Hirata T, Ariyasu S, Nomura K, Takemura H, Akimoto K, Aikawa N, Yasumori A, Aoki S. Simple and Convenient Method for the Isolation, Culture, and Re-collection of Cancer Cells from Blood by Using Glass-Bead Filters. ACS Biomater Sci Eng 2018; 5:438-452. [DOI: 10.1021/acsbiomaterials.8b01335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Babita Shashni
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hidehiko Matsuura
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Nijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Riku Saito
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Nijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Takuma Hirata
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Nijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Shinya Ariyasu
- Center for Technologies against Cancer, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kenta Nomura
- Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hiroshi Takemura
- Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kazunori Akimoto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Naoyuki Aikawa
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Nijuku, Katsushika-ku, Tokyo 125-8585, Japan
- Center for Technologies against Cancer, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Atsuo Yasumori
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Nijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Center for Technologies against Cancer, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Imaging Frontier Center, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
42
|
Tang W, Jiang D, Li Z, Zhu L, Shi J, Yang J, Xiang N. Recent advances in microfluidic cell sorting techniques based on both physical and biochemical principles. Electrophoresis 2018; 40:930-954. [DOI: 10.1002/elps.201800361] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/28/2018] [Accepted: 09/30/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Wenlai Tang
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
- Nanjing Institute of Intelligent High-end Equipment Industry Co., Ltd.; P. R. China
| | - Di Jiang
- School of Mechanical and Electronic Engineering; Nanjing Forestry University; P. R. China
| | - Zongan Li
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Liya Zhu
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Jianping Shi
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Jiquan Yang
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
- Nanjing Institute of Intelligent High-end Equipment Industry Co., Ltd.; P. R. China
| | - Nan Xiang
- School of Mechanical Engineering; Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments; Southeast University; P. R. China
| |
Collapse
|
43
|
Sinha N, Subedi N, Tel J. Integrating Immunology and Microfluidics for Single Immune Cell Analysis. Front Immunol 2018; 9:2373. [PMID: 30459757 PMCID: PMC6232771 DOI: 10.3389/fimmu.2018.02373] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/24/2018] [Indexed: 12/16/2022] Open
Abstract
The field of immunoengineering aims to develop novel therapies and modern vaccines to manipulate and modulate the immune system and applies innovative technologies toward improved understanding of the immune system in health and disease. Microfluidics has proven to be an excellent technology for analytics in biology and chemistry. From simple microsystem chips to complex microfluidic designs, these platforms have witnessed an immense growth over the last decades with frequent emergence of new designs. Microfluidics provides a highly robust and precise tool which led to its widespread application in single-cell analysis of immune cells. Single-cell analysis allows scientists to account for the heterogeneous behavior of immune cells which often gets overshadowed when conventional bulk study methods are used. Application of single-cell analysis using microfluidics has facilitated the identification of several novel functional immune cell subsets, quantification of signaling molecules, and understanding of cellular communication and signaling pathways. Single-cell analysis research in combination with microfluidics has paved the way for the development of novel therapies, point-of-care diagnostics, and even more complex microfluidic platforms that aid in creating in vitro cellular microenvironments for applications in drug and toxicity screening. In this review, we provide a comprehensive overview on the integration of microsystems and microfluidics with immunology and focus on different designs developed to decode single immune cell behavior and cellular communication. We have categorized the microfluidic designs in three specific categories: microfluidic chips with cell traps, valve-based microfluidics, and droplet microfluidics that have facilitated the ongoing research in the field of immunology at single-cell level.
Collapse
Affiliation(s)
- Nidhi Sinha
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nikita Subedi
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
44
|
Wang Z, Wu W, Wang Z, Tang Y, Deng Y, Xu L, Tian J, Shi Q. Ex vivo expansion of circulating lung tumor cells based on one-step microfluidics-based immunomagnetic isolation. Analyst 2018; 141:3621-5. [PMID: 26887792 DOI: 10.1039/c5an02554k] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We describe a one-step microfludics-based immunomagnetic isolation method to isolate CTCs directly from the whole blood of lung adenocarcinoma patients. This method avoids harsh sample preparation and enrichment steps, and therefore preserves the viability of CTCs during the in vitro isolation. Importantly, isolated, magnetic bead-bearing CTCs are concentrated in a small volume of culture medium with a high CTC density. High cell viability and culturing density promote the ex vivo expansion of limited numbers of CTCs. Expanded CTCs are characterized at the genetic, protein and metabolic levels.
Collapse
Affiliation(s)
- Zhihua Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Wenjun Wu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Zhuo Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Ying Tang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Ling Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jianhui Tian
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qihui Shi
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China. and State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Khan M, Mao S, Li W, Lin J. Microfluidic Devices in the Fast‐Growing Domain of Single‐Cell Analysis. Chemistry 2018; 24:15398-15420. [DOI: 10.1002/chem.201800305] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Mashooq Khan
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry, & Chemical Biology Tsinghua University Beijing 100084 China
| | - Sifeng Mao
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry, & Chemical Biology Tsinghua University Beijing 100084 China
| | - Weiwei Li
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry, & Chemical Biology Tsinghua University Beijing 100084 China
| | - Jin‐Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry, & Chemical Biology Tsinghua University Beijing 100084 China
| |
Collapse
|
46
|
A photoelectrochemical platform for the capture and release of rare single cells. Nat Commun 2018; 9:2288. [PMID: 29895867 PMCID: PMC5997639 DOI: 10.1038/s41467-018-04701-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 05/18/2018] [Indexed: 01/05/2023] Open
Abstract
For many normal and aberrant cell behaviours, it is important to understand the origin of cellular heterogeneity. Although powerful methods for studying cell heterogeneity have emerged, they are more suitable for common rather than rare cells. Exploring the heterogeneity of rare single cells is challenging because these rare cells must be first pre-concentrated and undergo analysis prior to classification and expansion. Here, a versatile capture & release platform consisting of an antibody-modified and electrochemically cleavable semiconducting silicon surface for release of individual cells of interest is presented. The captured cells can be interrogated microscopically and tested for drug responsiveness prior to release and recovery. The capture & release strategy was applied to identify rare tumour cells from whole blood, monitor the uptake of, and response to, doxorubicin and subsequently select cells for single-cell gene expression based on their response to the doxorubicin. Many cell capture systems exist but the characterisation and controlled release of single cells is a challenge. Here, the authors report on the development of a duel trigger release system using a combination of photo and electro triggers to allow for light based analysis without unwanted release.
Collapse
|
47
|
Tadimety A, Closson A, Li C, Yi S, Shen T, Zhang JXJ. Advances in liquid biopsy on-chip for cancer management: Technologies, biomarkers, and clinical analysis. Crit Rev Clin Lab Sci 2018; 55:140-162. [PMID: 29388456 PMCID: PMC6101655 DOI: 10.1080/10408363.2018.1425976] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Liquid biopsy, as a minimally invasive method of gleaning insight into the dynamics of diseases through a patient fluid sample, has been growing in popularity for cancer diagnosis, prognosis, and monitoring. While many technologies have been developed and validated in research laboratories, there has also been a push to expand these technologies into other clinical settings and as point of care devices. In this article, we discuss and evaluate microchip-based technologies for circulating tumor cell (CTC), exosome, and circulating tumor nucleic acid (ctNA) capture, detection, and analysis. Such integrated systems streamline otherwise multiple-step, manual operations to get a sample-to-answer quantitation. In addition, analysis of disease biomarkers is suited to point of care settings because of ease of use, low consumption of sample and reagents, and high throughput. We also cover the basics of biomarkers and their detection in biological fluid samples suitable for liquid biopsy on-chip. We focus on emerging technologies that process a small patient sample with high spatial-temporal resolution and derive clinically meaningful results through on-chip biomarker sensing and downstream molecular analysis in a simple workflow. This critical review is meant as a resource for those interested in developing technologies for capture, detection, and analysis platforms for liquid biopsy in a variety of settings.
Collapse
Affiliation(s)
- Amogha Tadimety
- a Thayer School of Engineering , Dartmouth College , Hanover , NH , USA
| | - Andrew Closson
- a Thayer School of Engineering , Dartmouth College , Hanover , NH , USA
| | - Cathy Li
- a Thayer School of Engineering , Dartmouth College , Hanover , NH , USA
| | - Song Yi
- b Nanolite Systems , Austin , TX , USA
| | - Ting Shen
- b Nanolite Systems , Austin , TX , USA
| | - John X J Zhang
- a Thayer School of Engineering , Dartmouth College , Hanover , NH , USA
- c Dartmouth-Hitchcock Medical Center , Lebanon , NH , USA
| |
Collapse
|
48
|
Ji Q, Zhang JM, Liu Y, Li X, Lv P, Jin D, Duan H. A Modular Microfluidic Device via Multimaterial 3D Printing for Emulsion Generation. Sci Rep 2018; 8:4791. [PMID: 29556013 PMCID: PMC5859176 DOI: 10.1038/s41598-018-22756-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022] Open
Abstract
3D-printing (3DP) technology has been developing rapidly. However, limited studies on the contribution of 3DP technology, especially multimaterial 3DP technology, to droplet-microfluidics have been reported. In this paper, multimaterial 3D-printed devices for the pneumatic control of emulsion generation have been reported. A 3D coaxial flexible channel with other rigid structures has been designed and printed monolithically. Numerical and experimental studies have demonstrated that this flexible channel can be excited by the air pressure and then deform in a controllable way, which can provide the active control of droplet generation. Furthermore, a novel modular microfluidic device for double emulsion generation has been designed and fabricated, which consists of three modules: function module, T-junction module, and co-flow module. The function module can be replaced by (1) Single-inlet module, (2) Pneumatic Control Unit (PCU) module and (3) Dual-inlet module. Different modules can be easily assembled for different double emulsion production. By using the PCU module, double emulsions with different number of inner droplets have been successfully produced without complicated operation of flow rates of different phases. By using single and dual inlet module, various double emulsions with different number of encapsulated droplets or encapsulated droplets with different compositions have been successfully produced, respectively.
Collapse
Affiliation(s)
- Qinglei Ji
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, 29 Yudao Street, Nanjing, 210016, Jiangsu, People's Republic of China.,State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Jia Ming Zhang
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Ying Liu
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Xiying Li
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Pengyu Lv
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Dongping Jin
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, 29 Yudao Street, Nanjing, 210016, Jiangsu, People's Republic of China
| | - Huiling Duan
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing, 100871, People's Republic of China. .,CAPT, HEDPS and IFSA Collaborative Innovation Center of MoE, Peking University, Beijing, 100871, People's Republic of China.
| |
Collapse
|
49
|
McDonald MP, Gemeinhardt A, König K, Piliarik M, Schaffer S, Völkl S, Aigner M, Mackensen A, Sandoghdar V. Visualizing Single-Cell Secretion Dynamics with Single-Protein Sensitivity. NANO LETTERS 2018; 18:513-519. [PMID: 29227108 DOI: 10.1021/acs.nanolett.7b04494] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cellular secretion of proteins into the extracellular environment is an essential mediator of critical biological mechanisms, including cell-to-cell communication, immunological response, targeted delivery, and differentiation. Here, we report a novel methodology that allows for the real-time detection and imaging of single unlabeled proteins that are secreted from individual living cells. This is accomplished via interferometric detection of scattered light (iSCAT) and is demonstrated with Laz388 cells, an Epstein-Barr virus (EBV)-transformed B cell line. We find that single Laz388 cells actively secrete IgG antibodies at a rate of the order of 100 molecules per second. Intriguingly, we also find that other proteins and particles spanning ca. 100 kDa-1 MDa are secreted from the Laz388 cells in tandem with IgG antibody release, likely arising from EBV-related viral proteins. The technique is general and, as we show, can also be applied to studying the lysate of a single cell. Our results establish label-free iSCAT imaging as a powerful tool for studying the real-time exchange between cells and their immediate environment with single-protein sensitivity.
Collapse
Affiliation(s)
- Matthew P McDonald
- Nano-Optics Division, Max Planck Institute for the Science of Light , Staudtstraße 2, 91058 Erlangen, Germany
| | - André Gemeinhardt
- Nano-Optics Division, Max Planck Institute for the Science of Light , Staudtstraße 2, 91058 Erlangen, Germany
| | - Katharina König
- Nano-Optics Division, Max Planck Institute for the Science of Light , Staudtstraße 2, 91058 Erlangen, Germany
- Department of Physics , Friedrich Alexander University Erlangen-Nuremberg , Schloßplatz 4, 91054 Erlangen, Germany
| | - Marek Piliarik
- Nano-Optics Division, Max Planck Institute for the Science of Light , Staudtstraße 2, 91058 Erlangen, Germany
| | - Stefanie Schaffer
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg , Ulmenweg 18, 91054 Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg , Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg , Ulmenweg 18, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg , Ulmenweg 18, 91054 Erlangen, Germany
| | - Vahid Sandoghdar
- Nano-Optics Division, Max Planck Institute for the Science of Light , Staudtstraße 2, 91058 Erlangen, Germany
- Department of Physics , Friedrich Alexander University Erlangen-Nuremberg , Schloßplatz 4, 91054 Erlangen, Germany
| |
Collapse
|
50
|
Abstract
Quantification of single-cell proteomics provides key insights in the field of cellular heterogeneity. This chapter discusses the emerging techniques that are being used to measure the protein copy numbers at the single-cell level, which includes flow cytometry, mass cytometry, droplet cytometry, microengraving, and single-cell barcoding microchip. The advantages and limitations of each technique are compared, and future research opportunities are highlighted.
Collapse
|