1
|
Zhao Q, Li S, Krall L, Li Q, Sun R, Yin Y, Fu J, Zhang X, Wang Y, Yang M. Deciphering cellular complexity: advances and future directions in single-cell protein analysis. Front Bioeng Biotechnol 2025; 12:1507460. [PMID: 39877263 PMCID: PMC11772399 DOI: 10.3389/fbioe.2024.1507460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Single-cell protein analysis has emerged as a powerful tool for understanding cellular heterogeneity and deciphering the complex mechanisms governing cellular function and fate. This review provides a comprehensive examination of the latest methodologies, including sophisticated cell isolation techniques (Fluorescence-Activated Cell Sorting (FACS), Magnetic-Activated Cell Sorting (MACS), Laser Capture Microdissection (LCM), manual cell picking, and microfluidics) and advanced approaches for protein profiling and protein-protein interaction analysis. The unique strengths, limitations, and opportunities of each method are discussed, along with their contributions to unraveling gene regulatory networks, cellular states, and disease mechanisms. The importance of data analysis and computational methods in extracting meaningful biological insights from the complex data generated by these technologies is also highlighted. By discussing recent progress, technological innovations, and potential future directions, this review emphasizes the critical role of single-cell protein analysis in advancing life science research and its promising applications in precision medicine, biomarker discovery, and targeted therapeutics. Deciphering cellular complexity at the single-cell level holds immense potential for transforming our understanding of biological processes and ultimately improving human health.
Collapse
Affiliation(s)
- Qirui Zhao
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Shan Li
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Leonard Krall
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Qianyu Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Rongyuan Sun
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuqi Yin
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Jingyi Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xu Zhang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Yonghua Wang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Mei Yang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| |
Collapse
|
2
|
Lin P, Gan YB, He J, Lin SE, Xu JK, Chang L, Zhao LM, Zhu J, Zhang L, Huang S, Hu O, Wang YB, Jin HJ, Li YY, Yan PL, Chen L, Jiang JX, Liu P. Advancing skeletal health and disease research with single-cell RNA sequencing. Mil Med Res 2024; 11:33. [PMID: 38816888 PMCID: PMC11138034 DOI: 10.1186/s40779-024-00538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
Orthopedic conditions have emerged as global health concerns, impacting approximately 1.7 billion individuals worldwide. However, the limited understanding of the underlying pathological processes at the cellular and molecular level has hindered the development of comprehensive treatment options for these disorders. The advent of single-cell RNA sequencing (scRNA-seq) technology has revolutionized biomedical research by enabling detailed examination of cellular and molecular diversity. Nevertheless, investigating mechanisms at the single-cell level in highly mineralized skeletal tissue poses technical challenges. In this comprehensive review, we present a streamlined approach to obtaining high-quality single cells from skeletal tissue and provide an overview of existing scRNA-seq technologies employed in skeletal studies along with practical bioinformatic analysis pipelines. By utilizing these methodologies, crucial insights into the developmental dynamics, maintenance of homeostasis, and pathological processes involved in spine, joint, bone, muscle, and tendon disorders have been uncovered. Specifically focusing on the joint diseases of degenerative disc disease, osteoarthritis, and rheumatoid arthritis using scRNA-seq has provided novel insights and a more nuanced comprehension. These findings have paved the way for discovering novel therapeutic targets that offer potential benefits to patients suffering from diverse skeletal disorders.
Collapse
Grants
- 2022YFA1103202 National Key Research and Development Program of China
- 82272507 National Natural Science Foundation of China
- 32270887 National Natural Science Foundation of China
- 32200654 National Natural Science Foundation of China
- CSTB2023NSCQ-ZDJO008 Natural Science Foundation of Chongqing
- BX20220397 Postdoctoral Innovative Talent Support Program
- SFLKF202201 Independent Research Project of State Key Laboratory of Trauma and Chemical Poisoning
- 2021-XZYG-B10 General Hospital of Western Theater Command Research Project
- 14113723 University Grants Committee, Research Grants Council of Hong Kong, China
- N_CUHK472/22 University Grants Committee, Research Grants Council of Hong Kong, China
- C7030-18G University Grants Committee, Research Grants Council of Hong Kong, China
- T13-402/17-N University Grants Committee, Research Grants Council of Hong Kong, China
- AoE/M-402/20 University Grants Committee, Research Grants Council of Hong Kong, China
Collapse
Affiliation(s)
- Peng Lin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi-Bo Gan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jian He
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, the General Hospital of Western Theater Command, Chengdu, 610031, China
| | - Si-En Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Jian-Kun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Li-Ming Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Sacramento, CA, 94305, USA
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Zhang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Sha Huang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ying-Bo Wang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huai-Jian Jin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yang-Yang Li
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Pu-Lin Yan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jian-Xin Jiang
- Wound Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
3
|
Qiao Z, Teng X, Liu A, Yang W. Novel Isolating Approaches to Circulating Tumor Cell Enrichment Based on Microfluidics: A Review. MICROMACHINES 2024; 15:706. [PMID: 38930676 PMCID: PMC11206030 DOI: 10.3390/mi15060706] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Circulating tumor cells (CTCs), derived from the primary tumor and carrying genetic information, contribute significantly to the process of tumor metastasis. The analysis and detection of CTCs can be used to assess the prognosis and treatment response in patients with tumors, as well as to help study the metastatic mechanisms of tumors and the development of new drugs. Since CTCs are very rare in the blood, it is a challenging problem to enrich CTCs efficiently. In this paper, we provide a comprehensive overview of microfluidics-based enrichment devices for CTCs in recent years. We explore in detail the methods of enrichment based on the physical or biological properties of CTCs; among them, physical properties cover factors such as size, density, and dielectric properties, while biological properties are mainly related to tumor-specific markers on the surface of CTCs. In addition, we provide an in-depth description of the methods for enrichment of single CTCs and illustrate the importance of single CTCs for performing tumor analyses. Future research will focus on aspects such as improving the separation efficiency, reducing costs, and increasing the detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Zezheng Qiao
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Xiangyu Teng
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| | - Anqin Liu
- School of Mechanical and Electrical Engineering, Yantai Institute of Technology, Yantai 264005, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China; (Z.Q.); (X.T.)
| |
Collapse
|
4
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
5
|
Oladokun R, Adekanmbi EO, An V, Gangavaram I, Srivastava SK. Dielectrophoretic profiling of erythrocytes to study the impacts of metabolic stress, temperature, and storage duration utilizing a point-and-planar microdevice. Sci Rep 2023; 13:17281. [PMID: 37828082 PMCID: PMC10570315 DOI: 10.1038/s41598-023-44022-9] [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] [Received: 08/08/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Dielectrophoresis (DEP) is widely utilized for trapping and sorting various types of cells, including live and dead cells and healthy and infected cells. This article focuses on the dielectric characterization of erythrocytes (red blood cells or RBCs) by quantifying DEP crossover frequency using a novel point-and-planar microwell device platform. Numerical simulations using COMSOL Multiphysics software demonstrate that the distribution of the DEP force is influenced by factors such as the shape of the point electrode, spacing between the point and planar electrodes, and the type of bioparticle being investigated. The dependency on electrode spacing is experimentally evaluated by analyzing the DEP crossover response of erythrocytes. Furthermore, the results are validated against the traditional electrical characterization technique called electrorotation, which typically requires laborious fabrication and operation using quadrupole electrodes. Other significant factors, including erythrocyte storage age and the changes in cell properties over time since collection, osmolarity, and temperature, are also assessed to determine the optimal conditions for erythrocyte characterization. The findings indicate a significant difference between fresh and stored erythrocyte samples (up to 4 days), highlighting the importance of maintaining an isotonic medium for cell storage.
Collapse
Affiliation(s)
- Raphael Oladokun
- Department of Chemical and Biomedical Engineering, West Virginia University, 1306 Evansdale Dr., PO Box 6102, Morgantown, WV, 26506-6102, USA
| | | | - Vanessa An
- Summer 2022 High School Intern, Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV, USA
| | - Isha Gangavaram
- Summer 2022 High School Intern, Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV, USA
| | - Soumya K Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, 1306 Evansdale Dr., PO Box 6102, Morgantown, WV, 26506-6102, USA.
| |
Collapse
|
6
|
Xu Y, Chen B, He M, Cui Z, Hu B. All-in-One Microfluidic Chip for Online Labeling, Separating, and Focusing Rare Circulating Tumor Cells from Blood Samples Followed by Inductively Coupled Plasma Mass Spectrometry Detection. Anal Chem 2023; 95:14061-14067. [PMID: 37677145 DOI: 10.1021/acs.analchem.3c02680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Circulating tumor cell (CTC) detection is essential for early cancer diagnosis and evaluating treatment efficacy. Despite the growing interest in isolating CTCs and further quantifying surface biomarkers at the single-cell level, highly efficient separation of rare CTCs from massive blood cells is still a big challenge. Here, we developed an all-in-one microfluidic chip system for the immunolabeling, magnetic separation, and focusing of HepG2 cells (as a CTC model) and online combined it with single cell-inductively coupled plasma mass spectrometry (SC-ICP-MS) for quantitative analysis of the asialoglycoprotein receptor (ASGPR) on single HepG2 cells. Lanthanide-labeled anti-ASGPR monoclonal antibody and antiepithelial cell adhesion molecule-modified magnetic beads were prepared as signal and magnetic probes, respectively. Target cells were highly efficiently labeled with signal and magnetic probes in the mixing zone of the microfluidic chip and then focused and sorted in the separation zone by specific magnetic separation techniques to avoid matrix contamination. The average cell recovery of HepG2 cells was derived to be 94.1 ± 5.7% with high separation efficiency and purity. The sorted cells with signal probes were detected for enumeration and quantification of ASGPR on their surface by SC-ICP-MS. The developed method showed good specificity and high sensitivity, detecting an average of (1.0 ± 0.2) × 105 ASGPR molecules per cell surface. This method can be used for absolute quantitative analysis of ASGPR on the surface of single hepatocellular carcinoma cells in real-world samples, providing a highly efficient analytical platform for studying targeted drug delivery in cancer therapy.
Collapse
Affiliation(s)
- Yan Xu
- 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
| | - Zewei Cui
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
7
|
Orrapin S, Thongkumkoon P, Udomruk S, Moonmuang S, Sutthitthasakul S, Yongpitakwattana P, Pruksakorn D, Chaiyawat P. Deciphering the Biology of Circulating Tumor Cells through Single-Cell RNA Sequencing: Implications for Precision Medicine in Cancer. Int J Mol Sci 2023; 24:12337. [PMID: 37569711 PMCID: PMC10418766 DOI: 10.3390/ijms241512337] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Circulating tumor cells (CTCs) hold unique biological characteristics that directly involve them in hematogenous dissemination. Studying CTCs systematically is technically challenging due to their extreme rarity and heterogeneity and the lack of specific markers to specify metastasis-initiating CTCs. With cutting-edge technology, single-cell RNA sequencing (scRNA-seq) provides insights into the biology of metastatic processes driven by CTCs. Transcriptomics analysis of single CTCs can decipher tumor heterogeneity and phenotypic plasticity for exploring promising novel therapeutic targets. The integrated approach provides a perspective on the mechanisms underlying tumor development and interrogates CTCs interactions with other blood cell types, particularly those of the immune system. This review aims to comprehensively describe the current study on CTC transcriptomic analysis through scRNA-seq technology. We emphasize the workflow for scRNA-seq analysis of CTCs, including enrichment, single cell isolation, and bioinformatic tools applied for this purpose. Furthermore, we elucidated the translational knowledge from the transcriptomic profile of individual CTCs and the biology of cancer metastasis for developing effective therapeutics through targeting key pathways in CTCs.
Collapse
Affiliation(s)
- Santhasiri Orrapin
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Patcharawadee Thongkumkoon
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Sasimol Udomruk
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Sutpirat Moonmuang
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Songphon Sutthitthasakul
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Petlada Yongpitakwattana
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Dumnoensun Pruksakorn
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Parunya Chaiyawat
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| |
Collapse
|
8
|
Wang Z. Assessing Tumorigenicity in Stem Cell-Derived Therapeutic Products: A Critical Step in Safeguarding Regenerative Medicine. Bioengineering (Basel) 2023; 10:857. [PMID: 37508884 PMCID: PMC10376867 DOI: 10.3390/bioengineering10070857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells hold promise in regenerative medicine due to their ability to proliferate and differentiate into various cell types. However, their self-renewal and multipotency also raise concerns about their tumorigenicity during and post-therapy. Indeed, multiple studies have reported the presence of stem cell-derived tumors in animal models and clinical administrations. Therefore, the assessment of tumorigenicity is crucial in evaluating the safety of stem cell-derived therapeutic products. Ideally, the assessment needs to be performed rapidly, sensitively, cost-effectively, and scalable. This article reviews various approaches for assessing tumorigenicity, including animal models, soft agar culture, PCR, flow cytometry, and microfluidics. Each method has its advantages and limitations. The selection of the assay depends on the specific needs of the study and the stage of development of the stem cell-derived therapeutic product. Combining multiple assays may provide a more comprehensive evaluation of tumorigenicity. Future developments should focus on the optimization and standardization of microfluidics-based methods, as well as the integration of multiple assays into a single platform for efficient and comprehensive evaluation of tumorigenicity.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
9
|
Wang Q, Šabanović B, Awada A, Reina C, Aicher A, Tang J, Heeschen C. Single-cell omics: a new perspective for early detection of pancreatic cancer? Eur J Cancer 2023; 190:112940. [PMID: 37413845 DOI: 10.1016/j.ejca.2023.112940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 07/08/2023]
Abstract
Pancreatic cancer is one of the most lethal cancers, mostly due to late diagnosis and limited treatment options. Early detection of pancreatic cancer in high-risk populations bears the potential to greatly improve outcomes, but current screening approaches remain of limited value despite recent technological advances. This review explores the possible advantages of liquid biopsies for this application, particularly focusing on circulating tumour cells (CTCs) and their subsequent single-cell omics analysis. Originating from both primary and metastatic tumour sites, CTCs provide important information for diagnosis, prognosis and tailoring of treatment strategies. Notably, CTCs have even been detected in the blood of subjects with pancreatic precursor lesions, suggesting their suitability as a non-invasive tool for the early detection of malignant transformation in the pancreas. As intact cells, CTCs offer comprehensive genomic, transcriptomic, epigenetic and proteomic information that can be explored using rapidly developing techniques for analysing individual cells at the molecular level. Studying CTCs during serial sampling and at single-cell resolution will help to dissect tumour heterogeneity for individual patients and among different patients, providing new insights into cancer evolution during disease progression and in response to treatment. Using CTCs for non-invasive tracking of cancer features, including stemness, metastatic potential and expression of immune targets, provides important and readily accessible molecular insights. Finally, the emerging technology of ex vivo culturing of CTCs could create new opportunities to study the functionality of individual cancers at any stage and develop personalised and more effective treatment approaches for this lethal disease.
Collapse
Affiliation(s)
- Qi Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Azhar Awada
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy; Molecular Biotechnology Center, University of Turin (UniTO), Turin, Italy
| | - Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Jiajia Tang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China; South Chongqing Road 227, Shanghai, China.
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy; South Chongqing Road 227, Shanghai, China.
| |
Collapse
|
10
|
Brockley LJ, Souza VGP, Forder A, Pewarchuk ME, Erkan M, Telkar N, Benard K, Trejo J, Stewart MD, Stewart GL, Reis PP, Lam WL, Martinez VD. Sequence-Based Platforms for Discovering Biomarkers in Liquid Biopsy of Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:2275. [PMID: 37190212 PMCID: PMC10136462 DOI: 10.3390/cancers15082275] [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: 03/07/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Lung cancer detection and monitoring are hampered by a lack of sensitive biomarkers, which results in diagnosis at late stages and difficulty in tracking response to treatment. Recent developments have established liquid biopsies as promising non-invasive methods for detecting biomarkers in lung cancer patients. With concurrent advances in high-throughput sequencing technologies and bioinformatics tools, new approaches for biomarker discovery have emerged. In this article, we survey established and emerging biomarker discovery methods using nucleic acid materials derived from bodily fluids in the context of lung cancer. We introduce nucleic acid biomarkers extracted from liquid biopsies and outline biological sources and methods of isolation. We discuss next-generation sequencing (NGS) platforms commonly used to identify novel biomarkers and describe how these have been applied to liquid biopsy. We highlight emerging biomarker discovery methods, including applications of long-read sequencing, fragmentomics, whole-genome amplification methods for single-cell analysis, and whole-genome methylation assays. Finally, we discuss advanced bioinformatics tools, describing methods for processing NGS data, as well as recently developed software tailored for liquid biopsy biomarker detection, which holds promise for early diagnosis of lung cancer.
Collapse
Affiliation(s)
- Liam J. Brockley
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Vanessa G. P. Souza
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
| | - Aisling Forder
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Michelle E. Pewarchuk
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Melis Erkan
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| | - Nikita Telkar
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Katya Benard
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Jessica Trejo
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Matt D. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Greg L. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Patricia P. Reis
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Victor D. Martinez
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
11
|
Mentink A, Isebia KT, Kraan J, Terstappen LWMM, Stevens M. Measuring antigen expression of cancer cell lines and circulating tumour cells. Sci Rep 2023; 13:6051. [PMID: 37055551 PMCID: PMC10101999 DOI: 10.1038/s41598-023-33179-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/08/2023] [Indexed: 04/15/2023] Open
Abstract
When evaluating EpCAM-based enrichment technologies for circulating tumour cells (CTCs), the cell lines used should closely resemble real CTCs, meaning the EpCAM expression of CTCs needs to be known, but also the EpCAM expression of cell lines at different institutions and times is important. As the number of CTCs in the blood is low, we enriched CTCs through the depletion of leukocytes from diagnostic leukapheresis products of 13 prostate cancer patients and measured EpCAM expression using quantitative flow cytometry. Antigen expression was compared between multiple institutions by measuring cultures from each institution. Capture efficiency was also measured for one of the used cell lines. Results show CTCs derived from castration-sensitive prostate cancer patients have varying but relatively low EpCAM expression, with median expression per patient ranging from 35 to 89,534 (mean 24,993) molecules per cell. A large variation in the antigen expression of identical cell lines cultured at different institutions was found, resulting in recoveries when using the CellSearch system ranging from 12 up to 83% for the same cell line. We conclude that large differences in capture efficiency can occur while using the same cell line. To closely resemble real CTCs from castration-sensitive prostate cancer patients, a cell line with a relatively low EpCAM expression should be used, and its expression should be monitored frequently.
Collapse
Affiliation(s)
- Anouk Mentink
- Medical Cell Biophysics Group, Techmed Center, Faculty of Science and Technology, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands
| | - Khrystany T Isebia
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Leon W M M Terstappen
- Medical Cell Biophysics Group, Techmed Center, Faculty of Science and Technology, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands
| | - Michiel Stevens
- Medical Cell Biophysics Group, Techmed Center, Faculty of Science and Technology, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands.
| |
Collapse
|
12
|
Armakolas A, Kotsari M, Koskinas J. Liquid Biopsies, Novel Approaches and Future Directions. Cancers (Basel) 2023; 15:1579. [PMID: 36900369 PMCID: PMC10000663 DOI: 10.3390/cancers15051579] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Cancer is among the leading causes of death worldwide. Early diagnosis and prognosis are vital to improve patients' outcomes. The gold standard of tumor characterization leading to tumor diagnosis and prognosis is tissue biopsy. Amongst the constraints of tissue biopsy collection is the sampling frequency and the incomplete representation of the entire tumor bulk. Liquid biopsy approaches, including the analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating miRNAs, and tumor-derived extracellular vesicles (EVs), as well as certain protein signatures that are released in the circulation from primary tumors and their metastatic sites, present a promising and more potent candidate for patient diagnosis and follow up monitoring. The minimally invasive nature of liquid biopsies, allowing frequent collection, can be used in the monitoring of therapy response in real time, allowing the development of novel approaches in the therapeutic management of cancer patients. In this review we will describe recent advances in the field of liquid biopsy markers focusing on their advantages and disadvantages.
Collapse
Affiliation(s)
- Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- B' Department of Medicine, Hippokration Hospital, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Maria Kotsari
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - John Koskinas
- B' Department of Medicine, Hippokration Hospital, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
13
|
CTC-5: A novel digital pathology approach to characterise circulating tumour cell biodiversity. Heliyon 2023; 9:e13044. [PMID: 36747925 PMCID: PMC9898658 DOI: 10.1016/j.heliyon.2023.e13044] [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: 06/26/2022] [Revised: 12/17/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Metastatic progression and tumor evolution complicates the clinical management of cancer patients. Circulating tumor cell (CTC) characterization is a growing discipline that aims to elucidate tumor metastasis and evolution processes. CTCs offer the clinical potential to monitor cancer patients for therapy response, disease relapse, and screen 'at risk' groups for the onset of malignancy. However, such clinical utility is currently limited to breast, prostate, and colorectal cancer patients. Further understanding of the basic CTC biology of other malignancies is required to progress them towards clinical utility. Unfortunately, such basic clinical research is often limited by restrictive characterization methods and high-cost barrier to entry for CTC isolation and imaging infrastructure. As experimental clinical results on applications of CTC are accumulating, it is becoming clear that a two-tier system of CTC isolation and characterization is required. The first tier is to facilitate basic research into CTC characterization. This basic research then informs a second tier specialised in clinical prognostic and diagnostic testing. This study presented in this manuscript describes the development and application of a low-cost, CTC isolation and characterization pipeline; CTC-5. This approach uses an established 'isolation by size' approach (ScreenCell Cyto) and combines histochemical morphology stains and multiparametric immunofluorescence on the same isolated CTCs. This enables capture and characterization of CTCs independent of biomarker-based pre-selection and accommodates both single CTCs and clusters of CTCs. Additionally, the developed open-source software is provided to facilitate the synchronization of microscopy data from multiple sources (https://github.com/CTC5/). This enables high parameter histochemical and immunofluorescent analysis of CTCs with existing microscopy infrastructure without investment in CTC specific imaging hardware. Our approach confirmed by the number of successful tests represents a potential major advance towards highly accessible low-cost technology aiming at the basic research tier of CTC isolation and characterization. The biomarker independent approach facilitates closing the gap between malignancies with poorly, and well-defined CTC phenotypes. As is currently the case for some of the most commonly occurring breast, prostate and colorectal cancers, such advances will ultimately benefit the patient, as early detection of relapse or onset of malignancy strongly correlates with their prognosis.
Collapse
|
14
|
Saez J, Garcia-Hernando M, Savva A, Owens RM, Benito-Lopez F, Basabe-Desmonts L. Capture and Release of Cancer Cells Through Smart Bioelectronics. Methods Mol Biol 2023; 2679:305-314. [PMID: 37300625 DOI: 10.1007/978-1-0716-3271-0_21] [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: 06/12/2023]
Abstract
Noninvasive collection of target cells such as circulating tumor cells (CTCs) is crucial for biology and medicine research. Conventional methods of cell collection are often complex, requiring either size-dependent sorting or invasive enzymatic reactions. Here, we show the development of a functional polymer film, which combines the thermoresponsive poly(N-isopropylacrylamide) and the conducting poly(3,4-ethylenedioxythiopene)/poly(styrene sulfonate), and its use for the capture and release of CTCs. When coated onto microfabricated gold electrodes, the proposed polymer films are capable of noninvasively capturing and controllably releasing cells while, at the same time, monitoring these processes with conventional electrical measurements.
Collapse
Affiliation(s)
- Janire Saez
- Microfluidics Cluster UPV/EHU, BIOMICs microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.
- Basque Foundation of Science, IKERBASQUE, Bilbao, Spain.
- Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain.
| | - Maite Garcia-Hernando
- Microfluidics Cluster UPV/EHU, BIOMICs microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, UK
| | - Roisin M Owens
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, UK
| | - Fernando Benito-Lopez
- Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Lourdes Basabe-Desmonts
- Microfluidics Cluster UPV/EHU, BIOMICs microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Basque Foundation of Science, IKERBASQUE, Bilbao, Spain
- Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain
| |
Collapse
|
15
|
Chavez‐Pineda OG, Rodriguez‐Moncayo R, Cedillo‐Alcantar DF, Guevara‐Pantoja PE, Amador‐Hernandez JU, Garcia‐Cordero JL. Microfluidic systems for the analysis of blood‐derived molecular biomarkers. Electrophoresis 2022; 43:1667-1700. [DOI: 10.1002/elps.202200067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 12/19/2022]
Affiliation(s)
- Oriana G. Chavez‐Pineda
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Roberto Rodriguez‐Moncayo
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Diana F. Cedillo‐Alcantar
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Pablo E. Guevara‐Pantoja
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Josue U. Amador‐Hernandez
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Jose L. Garcia‐Cordero
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
- Roche Institute for Translational Bioengineering (ITB) Roche Pharma Research and Early Development, Roche Innovation Center Basel Basel Switzerland
| |
Collapse
|
16
|
Szymborski TR, Czaplicka M, Nowicka AB, Trzcińska-Danielewicz J, Girstun A, Kamińska A. Dielectrophoresis-Based SERS Sensors for the Detection of Cancer Cells in Microfluidic Chips. BIOSENSORS 2022; 12:681. [PMID: 36140065 PMCID: PMC9496591 DOI: 10.3390/bios12090681] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 06/16/2023]
Abstract
The detection of freely circulating cancer cells (CTCs) is one of the greatest challenges of modern medical diagnostics. For several years, there has been increased attention on the use of surface-enhanced Raman spectroscopy (SERS) for the detection of CTCs. SERS is a non-destructive, accurate and precise technique, and the use of special SERS platforms even enables the amplification of weak signals from biological objects. In the current study, we demonstrate the unique arrangement of the SERS technique combined with the deposition of CTCs cells on the surface of the SERS platform via a dielectrophoretic effect. The appropriate frequencies of an alternating electric field and a selected shape of the electric field can result in the efficient deposition of CTCs on the SERS platform. The geometry of the microfluidic chip, the type of the cancer cells and the positive dielectrophoretic phenomenon resulted in the trapping of CTCs on the surface of the SERS platform. We presented results for two type of breast cancer cells, MCF-7 and MDA-MB-231, deposited from the 0.1 PBS solution. The limit of detection (LOD) is 20 cells/mL, which reflects the clinical potential and usefulness of the developed approach. We also provide a proof-of-concept for these CTCs deposited on the SERS platform from blood plasma.
Collapse
Affiliation(s)
- Tomasz R. Szymborski
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Marta Czaplicka
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Ariadna B. Nowicka
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
- Faculty of Materials Engineering and Technical Physics, Poznan University of Technology, Piotrowo 3, 60-965 Poznan, Poland
| | - Joanna Trzcińska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Agnieszka Kamińska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
17
|
Słomka A, Wang B, Mocan T, Horhat A, Willms AG, Schmidt-Wolf IGH, Strassburg CP, Gonzalez-Carmona MA, Lukacs-Kornek V, Kornek MT. Extracellular Vesicles and Circulating Tumour Cells - complementary liquid biopsies or standalone concepts? Theranostics 2022; 12:5836-5855. [PMID: 35966579 PMCID: PMC9373826 DOI: 10.7150/thno.73400] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Liquid biopsies do promise a lot, but are they keeping it? In the past decade, additional novel biomarkers qualified to be called like that, of which, some took necessary hurdles resulting in FDA approval and clinical use. Some others are since a while around, well known and were once regarded to be a game changer in cancer diagnosis or cancer screening. But, during their clinical use limitations were observed from statistical significance and questions raised regarding their robustness, that eventually led to be dropped from associated clinical guidelines for certain applications including cancer diagnosis. The purpose of this review isn't to give a broad overview of all current liquid biopsy as biomarkers, weight them and promise a brighter future in cancer prevention, but rather to take a deeper look on two of those who do qualify to be called liquid biopsies now or then. These two are probably of greatest interest conceptually and methodically, and likely have the highest chances to be in clinical use soon, with a portfolio extension over their original conceptual usage. We aim to dig deeper beyond cancer diagnosis or cancer screening. Actually, we aim to review in depth extracellular vesicles (EVs) and compare with circulating tumour cells (CTCs). The latter methodology is partially FDA approved and in clinical use. We will lay out similarities as taking advantage of surface antigens on EVs and CTCs in case of characterization and quantification. But drawing readers' attention to downstream application based on capture/isolation methodology and simply on their overall nature, here apparently being living material eventually recoverable as CTCs are vs. dead material with transient effects on recipient cell as in case of EVs. All this we try to bring in perspective, compare and conclude towards which future direction we are aiming for, or should aim for. Do we announce a winner between CTCs vs EVs? No, but we provide good reasons to intensify research on them.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-067 Bydgoszcz, Poland
| | - Bingduo Wang
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Tudor Mocan
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Adelina Horhat
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Arnulf G Willms
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Department of General, Visceral and Vascular Surgery, German Armed Forces Hospital Hamburg, 22049 Hamburg, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Maria A Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Miroslaw T Kornek
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| |
Collapse
|
18
|
Sen M, Hausler RM, Dulmage K, Black TA, Murphy W, Pletcher Jr CH, Wang L, Chen C, Yee SS, Bornheimer SJ, Maxwell KN, Stanger BZ, Moore JS, Thompson JC, Carpenter EL. Transcriptional profiling of single tumour cells from pleural effusions reveals heterogeneity of epithelial to mesenchymal transition and extra-cellular matrix marker expression. Clin Transl Med 2022; 12:e888. [PMID: 35811459 PMCID: PMC9271990 DOI: 10.1002/ctm2.888] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Moen Sen
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ryan M. Hausler
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Keely Dulmage
- Life Science InnovationBD Technologies and InnovationsDurhamNorth CarolinaUSA
| | - Taylor A. Black
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - William Murphy
- Department of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Charles H. Pletcher Jr
- Department of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ling Wang
- Life Science InnovationBD Technologies and InnovationsDurhamNorth CarolinaUSA
| | - Chang Chen
- Life Science InnovationBD Technologies and InnovationsDurhamNorth CarolinaUSA
| | - Stephanie S. Yee
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Kara N. Maxwell
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ben Z. Stanger
- Department of Medicine, Division of Gastroenterology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jonni S. Moore
- Department of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jeffrey C. Thompson
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology GroupUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Erica L. Carpenter
- Department of Medicine, Division of Hematology and Oncology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Heins A, Hoang MD, Weuster‐Botz D. Advances in automated real-time flow cytometry for monitoring of bioreactor processes. Eng Life Sci 2022; 22:260-278. [PMID: 35382548 PMCID: PMC8961054 DOI: 10.1002/elsc.202100082] [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: 07/09/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
Flow cytometry and its technological possibilities have greatly advanced in the past decade as analysis tool for single cell properties and population distributions of different cell types in bioreactors. Along the way, some solutions for automated real-time flow cytometry (ART-FCM) were developed for monitoring of bioreactor processes without operator interference over extended periods with variable sampling frequency. However, there is still great potential for ART-FCM to evolve and possibly become a standard application in bioprocess monitoring and process control. This review first addresses different components of an ART-FCM, including the sampling device, the sample-processing unit, the unit for sample delivery to the flow cytometer and the settings for measurement of pre-processed samples. Also, available algorithms are presented for automated data analysis of multi-parameter fluorescence datasets derived from ART-FCM experiments. Furthermore, challenges are discussed for integration of fluorescence-activated cell sorting into an ART-FCM setup for isolation and separation of interesting subpopulations that can be further characterized by for instance omics-methods. As the application of ART-FCM is especially of interest for bioreactor process monitoring, including investigation of population heterogeneity and automated process control, a summary of already existing setups for these purposes is given. Additionally, the general future potential of ART-FCM is addressed.
Collapse
Affiliation(s)
- Anna‐Lena Heins
- Institute of Biochemical EngineeringTechnical University of MunichGarchingGermany
| | - Manh Dat Hoang
- Institute of Biochemical EngineeringTechnical University of MunichGarchingGermany
| | - Dirk Weuster‐Botz
- Institute of Biochemical EngineeringTechnical University of MunichGarchingGermany
| |
Collapse
|
20
|
Cha H, Fallahi H, Dai Y, Yuan D, An H, Nguyen NT, Zhang J. Multiphysics microfluidics for cell manipulation and separation: a review. LAB ON A CHIP 2022; 22:423-444. [PMID: 35048916 DOI: 10.1039/d1lc00869b] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multiphysics microfluidics, which combines multiple functional physical processes in a microfluidics platform, is an emerging research area that has attracted increasing interest for diverse biomedical applications. Multiphysics microfluidics is expected to overcome the limitations of individual physical phenomena through combining their advantages. Furthermore, multiphysics microfluidics is superior for cell manipulation due to its high precision, better sensitivity, real-time tunability, and multi-target sorting capabilities. These exciting features motivate us to review this state-of-the-art field and reassess the feasibility of coupling multiple physical processes. To confine the scope of this paper, we mainly focus on five common forces in microfluidics: inertial lift, elastic, dielectrophoresis (DEP), magnetophoresis (MP), and acoustic forces. This review first explains the working mechanisms of single physical phenomena. Next, we classify multiphysics techniques in terms of cascaded connections and physical coupling, and we elaborate on combinations of designs and working mechanisms in systems reported in the literature to date. Finally, we discuss the possibility of combining multiple physical processes and associated design schemes and propose several promising future directions.
Collapse
Affiliation(s)
- Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Hedieh Fallahi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Yuchen Dai
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Dan Yuan
- Centre for Regional and Rural Futures, Deakin University, Geelong, Victoria 3216, Australia
| | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
21
|
Abstract
Magnetic cell separation has become a key methodology for the isolation of target cell populations from biological suspensions, covering a wide spectrum of applications from diagnosis and therapy in biomedicine to environmental applications or fundamental research in biology. There now exists a great variety of commercially available separation instruments and reagents, which has permitted rapid dissemination of the technology. However, there is still an increasing demand for new tools and protocols which provide improved selectivity, yield and sensitivity of the separation process while reducing cost and providing a faster response. This review aims to introduce basic principles of magnetic cell separation for the neophyte, while giving an overview of recent research in the field, from the development of new cell labeling strategies to the design of integrated microfluidic cell sorters and of point-of-care platforms combining cell selection, capture, and downstream detection. Finally, we focus on clinical, industrial and environmental applications where magnetic cell separation strategies are amongst the most promising techniques to address the challenges of isolating rare cells.
Collapse
|
22
|
Ruan Q, Yang J, Zou F, Chen X, Zhang Q, Zhao K, Lin X, Zeng X, Yu X, Wu L, Lin S, Zhu Z, Yang C. Single-Cell Digital Microfluidic Mass Spectrometry Platform for Efficient and Multiplex Genotyping of Circulating Tumor Cells. Anal Chem 2021; 94:1108-1117. [PMID: 34964350 DOI: 10.1021/acs.analchem.1c04194] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Gene mutation profiling of heterogeneous circulating tumor cells (CTCs) offers comprehensive and real-time molecular information of tumors for targeted therapy guidance, but the lack of efficient and multiplex genotyping techniques for single-CTC analysis greatly hinders its development and clinical application. This paper reports a single-CTC mass spectrometry analysis method for efficient and multiplex mutation profiling based on digital microfluidics. Digital microfluidics affords integrated single-CTC manipulation, from single-CTC isolation to high-performance whole genome amplification, via nanoliter droplet-based wettability trapping and hydrodynamic adjustment of cell distribution. Coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, multiplex mutation information of individual CTCs can be efficiently and accurately identified by the inherent mass differences of different DNA sequences. This platform achieves Kirsten rat sarcoma viral oncogene mutation profiling of heterogeneous CTCs at the single-cell level from cancer patient samples, offering new avenues for genotype profiling of single CTCs and cancer therapy guidance.
Collapse
Affiliation(s)
- Qingyu Ruan
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jian Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Fenxiang Zou
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaofeng Chen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qianqian Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Kaifeng Zhao
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaoye Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xi Zeng
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiyuan Yu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuichao Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.,Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
23
|
Crocetto F, Cimmino A, Ferro M, Terracciano D. Circulating tumor cells in bladder cancer: a new horizon of liquid biopsy for precision medicine. J Basic Clin Physiol Pharmacol 2021; 33:525-527. [PMID: 34563104 DOI: 10.1515/jbcpp-2021-0233] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Felice Crocetto
- Department of Neurosciences, Sciences of Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Amelia Cimmino
- Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| | - Matteo Ferro
- Department of Urology of European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
24
|
Bode D, Cull AH, Rubio-Lara JA, Kent DG. Exploiting Single-Cell Tools in Gene and Cell Therapy. Front Immunol 2021; 12:702636. [PMID: 34322133 PMCID: PMC8312222 DOI: 10.3389/fimmu.2021.702636] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Single-cell molecular tools have been developed at an incredible pace over the last five years as sequencing costs continue to drop and numerous molecular assays have been coupled to sequencing readouts. This rapid period of technological development has facilitated the delineation of individual molecular characteristics including the genome, transcriptome, epigenome, and proteome of individual cells, leading to an unprecedented resolution of the molecular networks governing complex biological systems. The immense power of single-cell molecular screens has been particularly highlighted through work in systems where cellular heterogeneity is a key feature, such as stem cell biology, immunology, and tumor cell biology. Single-cell-omics technologies have already contributed to the identification of novel disease biomarkers, cellular subsets, therapeutic targets and diagnostics, many of which would have been undetectable by bulk sequencing approaches. More recently, efforts to integrate single-cell multi-omics with single cell functional output and/or physical location have been challenging but have led to substantial advances. Perhaps most excitingly, there are emerging opportunities to reach beyond the description of static cellular states with recent advances in modulation of cells through CRISPR technology, in particular with the development of base editors which greatly raises the prospect of cell and gene therapies. In this review, we provide a brief overview of emerging single-cell technologies and discuss current developments in integrating single-cell molecular screens and performing single-cell multi-omics for clinical applications. We also discuss how single-cell molecular assays can be usefully combined with functional data to unpick the mechanism of cellular decision-making. Finally, we reflect upon the introduction of spatial transcriptomics and proteomics, its complementary role with single-cell RNA sequencing (scRNA-seq) and potential application in cellular and gene therapy.
Collapse
Affiliation(s)
- Daniel Bode
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Alyssa H. Cull
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Juan A. Rubio-Lara
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - David G. Kent
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
25
|
Garcia-Hernando M, Saez J, Savva A, Basabe-Desmonts L, Owens RM, Benito-Lopez F. An electroactive and thermo-responsive material for the capture and release of cells. Biosens Bioelectron 2021; 191:113405. [PMID: 34144472 DOI: 10.1016/j.bios.2021.113405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
Non-invasive collection of target cells is crucial for research in biology and medicine. In this work, we combine a thermo-responsive material, poly(N-isopropylacrylamide), with an electroactive material, poly(3,4-ethylene-dioxythiopene):poly(styrene sulfonate), to generate a smart and conductive copolymer for the label-free and non-invasive detection of the capture and release of cells on gold electrodes by electrochemical impedance spectroscopy. The copolymer is functionalized with fibronectin to capture tumor cells, and undergoes a conformational change in response to temperature, causing the release of cells. Simultaneously, the copolymer acts as a sensor, monitoring the capture and release of cancer cells by electrochemical impedance spectroscopy. This platform has the potential to play a role in top-notch label-free electrical monitoring of human cells in clinical settings.
Collapse
Affiliation(s)
- Maite Garcia-Hernando
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Barrio Sarriena S/n, 48940, Leioa, Spain; Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006, Vitoria-Gasteiz, Spain.
| | - Janire Saez
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK.
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK.
| | - Lourdes Basabe-Desmonts
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006, Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain; BCMaterials, Basque Centre for Materials, Micro and Nanodevices, UPV/EHU Science Park, 48940, Leioa, Spain; Basque Foundation of Science, IKERBASQUE, María Díaz Haroko Kalea, 3, 48013, Bilbao, Spain.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK.
| | - Fernando Benito-Lopez
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Barrio Sarriena S/n, 48940, Leioa, Spain; Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain; BCMaterials, Basque Centre for Materials, Micro and Nanodevices, UPV/EHU Science Park, 48940, Leioa, Spain.
| |
Collapse
|
26
|
Labib M, Kelley SO. Circulating tumor cell profiling for precision oncology. Mol Oncol 2021; 15:1622-1646. [PMID: 33448107 PMCID: PMC8169448 DOI: 10.1002/1878-0261.12901] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/19/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Analysis of circulating tumor cells (CTCs) collected from patient's blood offers a broad range of opportunities in the field of precision oncology. With new advances in profiling technology, it is now possible to demonstrate an association between the molecular profiles of CTCs and tumor response to therapy. In this Review, we discuss mechanisms of tumor resistance to therapy and their link to phenotypic and genotypic properties of CTCs. We summarize key technologies used to isolate and analyze CTCs and discuss recent clinical studies that examined CTCs for genomic and proteomic predictors of responsiveness to therapy. We also point out current limitations that still hamper the implementation of CTCs into clinical practice. We finally reflect on how these shortcomings can be addressed with the likely contribution of multiparametric approaches and advanced data analytics.
Collapse
Affiliation(s)
- Mahmoud Labib
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
| | - Shana O. Kelley
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
- Institute for Biomaterials and Biomedical EngineeringUniversity of TorontoCanada
- Department of BiochemistryUniversity of TorontoCanada
- Department of ChemistryUniversity of TorontoCanada
| |
Collapse
|
27
|
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
|
28
|
Lambert C, Yanikkaya Demirel G, Keller T, Preijers F, Psarra K, Schiemann M, Özçürümez M, Sack U. Flow Cytometric Analyses of Lymphocyte Markers in Immune Oncology: A Comprehensive Guidance for Validation Practice According to Laws and Standards. Front Immunol 2020; 11:2169. [PMID: 33042129 PMCID: PMC7528430 DOI: 10.3389/fimmu.2020.02169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Many anticancer therapies such as antibody-based therapies, cellular therapeutics (e.g., genetically modified cells, regulators of cytokine signaling, and signal transduction), and other biologically tailored interventions strongly influence the immune system and require tools for research, diagnosis, and monitoring. In flow cytometry, in vitro diagnostic (IVD) test kits that have been compiled and validated by the manufacturer are not available for all requirements. Laboratories are therefore usually dependent on modifying commercially available assays or, most often, developing them to meet clinical needs. However, both variants must then undergo full validation to fulfill the IVD regulatory requirements. Flow cytometric immunophenotyping is a multiparametric analysis of parameters, some of which have to be repeatedly adjusted; that must be considered when developing specific antibody panels. Careful adjustments of general rules are required to meet legal and regulatory requirements in the analysis of these assays. Here, we describe the relevant regulatory framework for flow cytometry-based assays and describe methods for the introduction of new antibody combinations into routine work including development of performance specifications, validation, and statistical methodology for design and analysis of the experiments. The aim is to increase reliability, efficiency, and auditability after the introduction of in-house-developed flow cytometry assays.
Collapse
Affiliation(s)
- Claude Lambert
- University Hospital, Immunology Laboratory, FRE-CNRS 3312, Saint-Etienne, France
| | | | | | - Frank Preijers
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Katherina Psarra
- Department of Immunology-Histocompatibility, Evangelismos Hospital, Athens, Greece
| | - Matthias Schiemann
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Mustafa Özçürümez
- Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Ulrich Sack
- Medizinische Fakultät, Institut für Klinische Immunologie, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
29
|
Exploration of Circulating Tumour Cell (CTC) Biology: A Paradigm Shift in Liquid Biopsy. Indian J Clin Biochem 2020; 36:131-142. [PMID: 33867703 PMCID: PMC7994460 DOI: 10.1007/s12291-020-00923-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/01/2020] [Indexed: 01/07/2023]
Abstract
Circulating tumour cells (CTCs), are disseminated tumour cells found in the blood in solid tumour malignancies. Identification of CTCs act as emerging tools in the field of the Liquid Biopsy. Majority of the studies focused on detection and enumeration of CTCs due to technological challenges those results from the rarity of CTCs in the blood. Enumeration of CTCs has already proven their value as prognostic as well as predictive biomarkers for disease prognosis. However, recent advances in technology permitted to study the molecular and functional features of CTCs and these features have the potential to change the diagnostic, prognostic and predictive landscape in oncology. In this review, we summarize the paradigm shift in the field of liquid biopsy-based cancer diagnostics using CTC isolation and detection. We have discussed recent advances in the technologies for molecular characterization of CTCs which have aided a shift from CTC enumeration to an in-depth analysis of the CTC genome, transcriptomes, proteins, epigenomes along with various functional features. Finally, as a prognosticating strategy, the potentials of CTCs as a tool of liquid biopsy to predict micrometastasis, monitor prognosis and how to use them as an additional tool for cancer staging has been discussed.
Collapse
|
30
|
Jariyal H, Gupta C, Bhat VS, Wagh JR, Srivastava A. Advancements in Cancer Stem Cell Isolation and Characterization. Stem Cell Rev Rep 2020; 15:755-773. [PMID: 31863337 DOI: 10.1007/s12015-019-09912-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Occurrence of stem cells (CSCs) in cancer is well established in last two decades. These rare cells share several properties including presence of common surface markers, stem cell markers, chemo- and radio- resistance and are highly metastatic in nature; thus, considered as valuable prognostic and therapeutic targets in cancer. However, the studies related to CSCs pave number of issues due to rare cell population and difficulties in their isolation ascribed to common stem cell marker. Various techniques including flow cytometry, laser micro-dissection, fluorescent nanodiamonds and microfluidics are used for the isolation of these rare cells. In this review, we have included the advance strategies adopted for the isolation of CSCs using above mentioned techniques. Furthermore, CSCs are primarily found in the core of the solid tumors and their microenvironment plays an important role in maintenance, self-renewal, division and tumor development. Therefore, in vivo tracking and model development become obligatory for functional studies of CSCs. Fluorescence and bioluminescence tagging has been widely used for transplantation assay and lineage tracking experiments to improve our understanding towards CSCs behaviour in their niche. Techniques such as Magnetic resonance imaging (MRI) and Positron emission tomography (PET) have proved useful for tracking of endogenous CSCs which could be helpful in their identification in clinical settings.
Collapse
Affiliation(s)
- Heena Jariyal
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Chanchal Gupta
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Vedika Sandeep Bhat
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Jayant Ramakant Wagh
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Device, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
31
|
Luo L, He Y. Magnetically driven microfluidics for isolation of circulating tumor cells. Cancer Med 2020; 9:4207-4231. [PMID: 32325536 PMCID: PMC7300401 DOI: 10.1002/cam4.3077] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) largely contribute to cancer metastasis and show potential prognostic significance in cancer isolation and detection. Miniaturization has progressed significantly in the last decade which in turn enabled the development of several microfluidic systems. The microfluidic systems offer a controlled microenvironment for studies of fundamental cell biology, resulting in the rapid development of microfluidic isolation of CTCs. Due to the inherent ability of magnets to provide forces at a distance, the technology of CTCs isolation based on the magnetophoresis mechanism has become a routine methodology. This historical review aims to introduce two principles of magnetic isolation and recent techniques, facilitating research in this field and providing alternatives for researchers in their study of magnetic isolation. Researchers intend to promote effective CTC isolation and analysis as well as active development of next-generation cancer treatment. The first part of this review summarizes the primary principles based on positive and negative magnetophoretic isolation and describes the metrics for isolation performance. The second part presents a detailed overview of the factors that affect the performance of CTC magnetic isolation, including the magnetic field sources, functionalized magnetic nanoparticles, magnetic fluids, and magnetically driven microfluidic systems.
Collapse
Affiliation(s)
- Laan Luo
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
| | - Yongqing He
- School of Chemical EngineeringKunming University of Science and TechnologyKunmingChina
- Chongqing Key Laboratory of Micro‐Nano System and Intelligent SensingChongqing Technology and Business UniversityChongqingChina
| |
Collapse
|
32
|
Zhang X, Wei X, Wei Y, Chen M, Wang J. The up-to-date strategies for the isolation and manipulation of single cells. Talanta 2020; 218:121147. [PMID: 32797903 DOI: 10.1016/j.talanta.2020.121147] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023]
Abstract
Due to the large cellular heterogeneity, the strategies for the isolation and manipulation of single cells have been pronounced indispensable in the fields of disease diagnostics, drug delivery, and cancer biology at the single-cell resolution. Herein, an overview of the up-to-date techniques for precise manipulation/separation and analysis of single-cell is accomplished, these include the various approaches for the isolation and detection of individual cells in flow cytometry, microfluidic systems, micromodule systems, and others. In addition, the advanced application of these protocols is discussed. In particular, a few designs are highlighted for visualization, non-invasion, and intelligentization in single cell analysis, i.e., imaging flow cytometry, label-free microfluidic platform, single-cell capillary probe, and other related techniques. At the present, the main barriers in the various schemes for single cell manipulation which limited their practical applications are their cumbersome construction and single-functionality. The future opportunities and outstanding challenges in the isolation/manipulation of single cells are depicted.
Collapse
Affiliation(s)
- Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Yujia Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Mingli Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning, 110819, China; Analytical and Testing Center, Northeastern University, Shenyang, Liaoning, 110819, China.
| | - Jianhua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning, 110819, China.
| |
Collapse
|
33
|
Relevance of CTC Clusters in Breast Cancer Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:93-115. [PMID: 32304082 DOI: 10.1007/978-3-030-35805-1_7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metastasis is the major cause of mortality in patients with breast cancer; however, the mechanisms of tumor cell dissemination and metastasis formation are not well established yet. The study of circulating tumour cells (CTCs), the metastatic precursors of distant disease, may help in this search. CTCs can be found in the blood of cancer patients as single cells or as tumor cell aggregates, known as CTC clusters. CTC clusters have differential biological features such as an enhanced survival and metastatic potential, and they hold great promises for the evaluation of prognosis, diagnosis and therapy of the metastatic cancer. The analysis of CTC clusters offers new insights into the mechanism of metastasis and can guide towards the development of new diagnostic and therapeutic strategies to suppress cancer metastasis. This has become possible thanks to the development of improved technologies for detection of CTCs and CTC clusters. However, more efficient methods are needed in order to address important questions regarding the metastatic potential of CTC and future clinical applications. In this chapter, we explore the current knowledge on the role of CTC clusters in breast cancer metastasis, their origin, metastatic advantages and clinical importance.
Collapse
|
34
|
Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, Kolahian S, Javaheri T, Zare P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal 2020; 18:59. [PMID: 32264958 PMCID: PMC7140346 DOI: 10.1186/s12964-020-0530-4] [Citation(s) in RCA: 1052] [Impact Index Per Article: 210.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
The dynamic interactions of cancer cells with their microenvironment consisting of stromal cells (cellular part) and extracellular matrix (ECM) components (non-cellular) is essential to stimulate the heterogeneity of cancer cell, clonal evolution and to increase the multidrug resistance ending in cancer cell progression and metastasis. The reciprocal cell-cell/ECM interaction and tumor cell hijacking of non-malignant cells force stromal cells to lose their function and acquire new phenotypes that promote development and invasion of tumor cells. Understanding the underlying cellular and molecular mechanisms governing these interactions can be used as a novel strategy to indirectly disrupt cancer cell interplay and contribute to the development of efficient and safe therapeutic strategies to fight cancer. Furthermore, the tumor-derived circulating materials can also be used as cancer diagnostic tools to precisely predict and monitor the outcome of therapy. This review evaluates such potentials in various advanced cancer models, with a focus on 3D systems as well as lab-on-chip devices. Video abstract.
Collapse
Affiliation(s)
- Roghayyeh Baghban
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khaled Seidi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committees, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognitive, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Kolahian
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tuebingen, Tuebingen, Germany
| | - Tahereh Javaheri
- Health Informatics Lab, Metropolitan College, Boston University, Boston, USA
| | - Peyman Zare
- Dioscuri Center of Chromatin Biology and Epigenomics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Faculty of Medicine, Cardinal Stefan Wyszyński University in Warsaw, 01-938 Warsaw, Poland
| |
Collapse
|
35
|
Dianat-Moghadam H, Azizi M, Eslami-S Z, Cortés-Hernández LE, Heidarifard M, Nouri M, Alix-Panabières C. The Role of Circulating Tumor Cells in the Metastatic Cascade: Biology, Technical Challenges, and Clinical Relevance. Cancers (Basel) 2020; 12:E867. [PMID: 32260071 PMCID: PMC7225923 DOI: 10.3390/cancers12040867] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Metastases and cancer recurrence are the main causes of cancer death. Circulating Tumor Cells (CTCs) and disseminated tumor cells are the drivers of cancer cell dissemination. The assessment of CTCs' clinical role in early metastasis prediction, diagnosis, and treatment requires more information about their biology, their roles in cancer dormancy, and immune evasion as well as in therapy resistance. Indeed, CTC functional and biochemical phenotypes have been only partially characterized using murine metastasis models and liquid biopsy in human patients. CTC detection, characterization, and enumeration represent a promising tool for tailoring the management of each patient with cancer. The comprehensive understanding of CTCs will provide more opportunities to determine their clinical utility. This review provides much-needed insights into this dynamic field of translational cancer research.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran; (H.D.-M.); (M.N.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Mehdi Azizi
- Proteomics Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| | - Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| | - Maryam Heidarifard
- Drug Applied Research Center, Tabriz University of Medical Sciences, 51368 Tabriz, Iran;
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran; (H.D.-M.); (M.N.)
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| |
Collapse
|
36
|
Circulating tumor cell as the functional aspect of liquid biopsy to understand the metastatic cascade in solid cancer. Mol Aspects Med 2020; 72:100816. [PMID: 31377345 DOI: 10.1016/j.mam.2019.07.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022]
|
37
|
Konczalla L, Wöstemeier A, Kemper M, Karstens KF, Izbicki J, Reeh M. Clinical Significance of Circulating Tumor Cells in Gastrointestinal Carcinomas. Diagnostics (Basel) 2020; 10:diagnostics10040192. [PMID: 32235479 PMCID: PMC7235836 DOI: 10.3390/diagnostics10040192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
The idea of a liquid biopsy to screen, surveil and treat cancer patients is an intensively discussed and highly awaited tool in the field of oncology. Despite intensive research in this field, the clinical application has not been implemented yet and further research has to be conducted. However, one component of the liquid biopsy is circulating tumor cells (CTCs) whose potential for clinical application is evaluated in the following. CTCs can shed from primary tumors to the peripheral blood at any time point during the progress of a malignant disease. Following, one single CTC can be the origin for distant metastasis at later cancer stage. Thus, CTCs have great potential to either be used in cancer diagnostics and patient stratification or to function as a target for new therapeutic approaches to stop tumor dissemination and metastasis at the very early beginning. Due to the biological fundamental role of CTCs in tumor progression, here, we provide an overview of CTCs in gastrointestinal cancers and their potential use in the clinical setting. In particular, we discuss the usage of CTC for screening and stratifying patients’ risk. Moreover, we will discuss the potential role of CTCs for treatment specification and treatment monitoring.
Collapse
|
38
|
Voronin DV, Kozlova AA, Verkhovskii RA, Ermakov AV, Makarkin MA, Inozemtseva OA, Bratashov DN. Detection of Rare Objects by Flow Cytometry: Imaging, Cell Sorting, and Deep Learning Approaches. Int J Mol Sci 2020; 21:E2323. [PMID: 32230871 PMCID: PMC7177904 DOI: 10.3390/ijms21072323] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/25/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022] Open
Abstract
Flow cytometry nowadays is among the main working instruments in modern biology paving the way for clinics to provide early, quick, and reliable diagnostics of many blood-related diseases. The major problem for clinical applications is the detection of rare pathogenic objects in patient blood. These objects can be circulating tumor cells, very rare during the early stages of cancer development, various microorganisms and parasites in the blood during acute blood infections. All of these rare diagnostic objects can be detected and identified very rapidly to save a patient's life. This review outlines the main techniques of visualization of rare objects in the blood flow, methods for extraction of such objects from the blood flow for further investigations and new approaches to identify the objects automatically with the modern deep learning methods.
Collapse
Affiliation(s)
- Denis V. Voronin
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- Department of Physical and Colloid Chemistry, National University of Oil and Gas (Gubkin University), 119991 Moscow, Russia
| | - Anastasiia A. Kozlova
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Roman A. Verkhovskii
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- School of Urbanistics, Civil Engineering and Architecture, Yuri Gagarin State Technical University of Saratov, 410054 Saratov, Russia
| | - Alexey V. Ermakov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- Department of Biomedical Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Mikhail A. Makarkin
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Olga A. Inozemtseva
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Daniil N. Bratashov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| |
Collapse
|
39
|
Klotz R, Yu M. Insights into brain metastasis: Recent advances in circulating tumor cell research. Cancer Rep (Hoboken) 2020; 5:e1239. [PMID: 33372393 PMCID: PMC9124503 DOI: 10.1002/cnr2.1239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/24/2022] Open
Abstract
Background How tumor cells disseminate to brain and establish brain metastasis remains partly an unsolved problem. This devastating complication of many cancers is initiated by a rare subset of the circulating tumor cells (CTCs) shed into the blood stream. Thus, the profiling of the molecular properties in these brain metastasis‐initiating CTCs is essential to uncover the mechanisms underlying brain metastasis. Recent Findings Important efforts to improve the enrichment and detection of CTCs enabled the detailed molecular and functional analysis of CTCs that drive brain metastasis. In this review, we highlight key findings on existing preclinical studies that provide insights toward a comprehensive picture of brain metastasis‐precursors in CTCs and the potential clinical implications. Conclusion A deeper understanding of the brain metastasis precursors should help to stratify high‐risk patients and improve preventive therapeutic strategies. Although all these preclinical evidences have yet to be translated into patients, they provide considerable hope to benefit patients with brain metastases in the future.
Collapse
Affiliation(s)
- Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California.,USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California.,USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
40
|
Amodio D, Santilli V, Zangari P, Cotugno N, Manno EC, Rocca S, Rossi P, Cancrini C, Finocchi A, Chassiakos A, Petrovas C, Palma P. How to dissect the plasticity of antigen-specific immune response: a tissue perspective. Clin Exp Immunol 2020; 199:119-130. [PMID: 31626717 PMCID: PMC6954674 DOI: 10.1111/cei.13386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2019] [Indexed: 12/01/2022] Open
Abstract
Generation of antigen-specific humoral responses following vaccination or infection requires the maturation and function of highly specialized immune cells in secondary lymphoid organs (SLO), such as lymph nodes or tonsils. Factors that orchestrate the dynamics of these cells are still poorly understood. Currently, experimental approaches that enable a detailed description of the function of the immune system in SLO have been mainly developed and optimized in animal models. Conversely, methodological approaches in humans are mainly based on the use of blood-associated material because of the challenging access to tissues. Indeed, only few studies in humans were able to provide a discrete description of the complex network of cytokines, chemokines and lymphocytes acting in tissues after antigenic challenge. Furthermore, even fewer data are currently available on the interaction occurring within the complex micro-architecture of the SLO. This information is crucial in order to design particular vaccination strategies, especially for patients affected by chronic and immune compromising medical conditions who are under-vaccinated or who respond poorly to immunizations. Analysis of immune cells in different human tissues by high-throughput technologies, able to obtain data ranging from gene signature to protein expression and cell phenotypes, is needed to dissect the peculiarity of each immune cell in a definite human tissue. The main aim of this review is to provide an in-depth description of the current available methodologies, proven evidence and future perspectives in the analysis of immune mechanisms following immunization or infections in SLO.
Collapse
Affiliation(s)
- D. Amodio
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - V. Santilli
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - P. Zangari
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - N. Cotugno
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - E. C. Manno
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - S. Rocca
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - P. Rossi
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - C. Cancrini
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - A. Finocchi
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - A. Chassiakos
- Vaccine Research CenterNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - C. Petrovas
- Vaccine Research CenterNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - P. Palma
- Research Unit in Congenital and Perinatal InfectionsImmune and Infectious Diseases DivisionAcademic Department of PediatricsOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| |
Collapse
|
41
|
Zhu S, Jiang F, Han Y, Xiang N, Ni Z. Microfluidics for label-free sorting of rare circulating tumor cells. Analyst 2020; 145:7103-7124. [DOI: 10.1039/d0an01148g] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A review discussing the working principles and performances of label-free CTC sorting methods.
Collapse
Affiliation(s)
- Shu Zhu
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Fengtao Jiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Yu Han
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Nan Xiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Zhonghua Ni
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| |
Collapse
|
42
|
Zheng Y, Li Q, Hu W, Liao J, Zheng G, Su M. Whole slide imaging of circulating tumor cells captured on a capillary microchannel device. LAB ON A CHIP 2019; 19:3796-3803. [PMID: 31621763 DOI: 10.1039/c9lc00412b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liquid biopsy with circulating tumor cells (CTCs) can aid in cancer detection at early stages and determine whether a certain treatment is effective or not. However, existing CTC techniques focus on one or two aspects of CTC management including sampling, enrichment, enumeration, and treatment selection. This paper reports an integrated capillary microchannel device that allows efficient capturing of CTCs with a wide microchannel, rapid enumeration of captured CTCs with whole slide cell imaging, and in situ drug testing with captured CTCs. Blood is drawn into the microchannel whose height is appropriate to the diameter of cancer cells, while its width is a thousand times larger than the diameter of cancer cells. The inner bottom surface of the microchannel is modified with long chain polymers that have cell adhesive ends to efficiently capture CTCs from blood. With this design, cells including CTCs are forced to move through the polymer coated microchannel, and the chance of cell adhesive ends interacting with specific antigens overexpressed on surfaces of cancer cells is significantly enhanced without a channel blockage issue. Captured CTCs are enumerated with a whole slide imaging platform via dual LED autofocusing technology then exposed to anti-cancer drugs, followed by live/dead assay and fluorescence imaging. Given its straightforward, easy and powerless operation, this device with whole slide imaging will be useful for cancer diagnosis, prognosis and point-of-care treatments.
Collapse
Affiliation(s)
- Yiting Zheng
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA.
| | - Qingxuan Li
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA.
| | - Weike Hu
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA.
| | - Jun Liao
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ming Su
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA.
| |
Collapse
|
43
|
Yu L, Sa S, Wang L, Dulmage K, Bhagwat N, Yee SS, Sen M, Pletcher CH, Moore JS, Saksena S, Dixon EP, Carpenter EL. An integrated enrichment system to facilitate isolation and molecular characterization of single cancer cells from whole blood. Cytometry A 2019; 93:1226-1233. [PMID: 30549400 DOI: 10.1002/cyto.a.23599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 01/11/2023]
Abstract
Circulating tumor cells (CTCs) carry valuable biological information. While enumeration of CTCs in peripheral blood is an FDA-approved prognostic indicator of survival in metastatic prostate and other cancers, analysis of CTC phenotypic and genomic markers is needed to identify cancer origin and elucidate pathways that can guide therapeutic selection for personalized medicine. Given the emergence of single-cell mRNA sequencing technologies, a method is needed to isolate CTCs with high sensitivity and specificity as well as compatibility with downstream genomic analysis. Flow cytometry is a powerful tool to analyze and sort single cells, but pre-enrichment is required prior to flow sorting for efficient isolation of CTCs due to the extreme low frequency of CTCs in blood (one in billions of blood cells). While current enrichment technologies often require many steps and result in poor recovery, we demonstrate a magnetic separator and acoustic microfluidic focusing chip integrated system that enriches rare cells in-line with FACS™ (fluorescent activated cell sorting) and single-cell sequencing. This system analyzes, isolates, and index sorts single cells directly into 96-well plates containing reagents for Molecular Indexing (MI) and transcriptional profiling of single cells. With an optimized workflow using the integrated enrichment-FACS system, we performed a proof-of-concept experiment with spiked prostate cancer cells in peripheral blood and achieved: (i) a rapid one-step process to isolate rare cancer cells from lysed whole blood; (ii) an average of 92% post-enrichment cancer cell recovery (R2 = 0.9998) as compared with 55% recovery for a traditional benchtop workflow; and (iii) detection of differentially expressed genes at a single cell level that are consistent with reported cell-type dependent expression signatures for prostate cancer cells. These model system results lay the groundwork for applying our approach to human blood samples from prostate and other cancer patients, and support the enrichment-FACS system as a flexible solution for isolation and characterization of CTCs for cancer diagnosis. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Liping Yu
- BD Biosciences, San Jose, California
| | - Silin Sa
- BD Biosciences, San Jose, California
| | - Ling Wang
- BD Technologies and Innovation, Durham, North Carolina
| | - Keely Dulmage
- BD Technologies and Innovation, Durham, North Carolina
| | - Neha Bhagwat
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephanie S Yee
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Moen Sen
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charles H Pletcher
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jonni S Moore
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Eric P Dixon
- BD Technologies and Innovation, Durham, North Carolina
| | - Erica L Carpenter
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Rare Event Phenotyping and Molecular Characterization: Circulating Tumor Cells. Methods Mol Biol 2019. [PMID: 31522422 DOI: 10.1007/978-1-4939-9650-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Noninvasive isolation of circulating tumor cells (CTCs) from patient blood samples allows for interrogation of valuable molecular and phenotypic information useful for disease diagnosis and monitoring response to therapy. However, CTCs are extremely rare relative to red and white blood cells (R/WBC), thus making CTC isolation from unmanipulated whole blood very time-consuming. Moreover, single CTC analysis often requires hand-picking, a step that can result in more CTC loss and compromised cell integrity. Here we describe an automated flow cytometry-based approach for isolation and analysis of single, viable CTCs that combines gentle RBC lysis and magnetic, no-wash negative-depletion of WBCs, followed by a highly adaptable sorting protocol for rare cells of interest. Multiparametric flow-cytometric panels allow probing of numerous extracellular markers for immunophenotyping, while whole transcriptome analysis contributes to molecular characterization of individual CTCs. Index sorting links single CTC proteogenomics information.
Collapse
|
45
|
Tavakoli H, Zhou W, Ma L, Perez S, Ibarra A, Xu F, Zhan S, Li X. Recent advances in microfluidic platforms for single-cell analysis in cancer biology, diagnosis and therapy. Trends Analyt Chem 2019; 117:13-26. [PMID: 32831435 PMCID: PMC7434086 DOI: 10.1016/j.trac.2019.05.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Understanding molecular, cellular, genetic and functional heterogeneity of tumors at the single-cell level has become a major challenge for cancer research. The microfluidic technique has emerged as an important tool that offers advantages in analyzing single-cells with the capability to integrate time-consuming and labour-intensive experimental procedures such as single-cell capture into a single microdevice at ease and in a high-throughput fashion. Single-cell manipulation and analysis can be implemented within a multi-functional microfluidic device for various applications in cancer research. Here, we present recent advances of microfluidic devices for single-cell analysis pertaining to cancer biology, diagnostics, and therapeutics. We first concisely introduce various microfluidic platforms used for single-cell analysis, followed with different microfluidic techniques for single-cell manipulation. Then, we highlight their various applications in cancer research, with an emphasis on cancer biology, diagnosis, and therapy. Current limitations and prospective trends of microfluidic single-cell analysis are discussed at the end.
Collapse
Affiliation(s)
- Hamed Tavakoli
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Wan Zhou
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Stefani Perez
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Andrea Ibarra
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center,
Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of
China
| | - Sihui Zhan
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
| | - XiuJun Li
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| |
Collapse
|
46
|
Greenblatt MB, Ono N, Ayturk UM, Debnath S, Lalani S. The Unmixing Problem: A Guide to Applying Single-Cell RNA Sequencing to Bone. J Bone Miner Res 2019; 34:1207-1219. [PMID: 31336008 PMCID: PMC6658136 DOI: 10.1002/jbmr.3802] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 12/24/2022]
Abstract
Bone is composed of a complex mixture of many dynamic cell types. Flow cytometry and in vivo lineage tracing have offered early progress toward deconvoluting this heterogeneous mixture of cells into functionally well-defined populations suitable for further studies. Single-cell sequencing is poised as a key complementary technique to better understand the cellular basis of bone metabolism and development. However, single-cell sequencing approaches still have important limitations, including transcriptional effects of cell isolation and sparse sampling of the transcriptome, that must be considered during experimental design and analysis to harness the power of this approach. Accounting for these limitations requires a deep knowledge of the tissue under study. Therefore, with the emergence of accessible tools for conducting and analyzing single-cell RNA sequencing (scRNA-seq) experiments, bone biologists will be ideal leaders in the application of scRNA-seq to the skeleton. Here we provide an overview of the steps involved with a single-cell sequencing analysis of bone, focusing on practical considerations needed for a successful study. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
- Research Division, Hospital for Special Surgery, New York,
NY, USA
| | - Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, MI,
USA
| | - Ugur M Ayturk
- Musculoskeletal Integrity Program, Hospital for Special
Surgery, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
| |
Collapse
|
47
|
Lusk JF, Miranda C, Howell M, Chrest M, Eshima J, Smith BS. Photoacoustic Flow System for the Detection of Ovarian Circulating Tumor Cells Utilizing Copper Sulfide Nanoparticles. ACS Biomater Sci Eng 2019; 5:1553-1560. [PMID: 33405628 DOI: 10.1021/acsbiomaterials.8b01217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The development of cell-specific photoacoustic (PA) contrast agents within systems of fluidic flow provides opportunities for the accurate detection of early stage cancer metastasis. Despite the promise of exogenous contrast agents for use in clinical settings, applications are currently limited by both material biocompatibility and target specificity. In this study, folic acid functionalized copper sulfide nanoparticles (FA-CuS NPs) are synthesized to enable ovarian-cancer-specific binding and PA detection in a custom flow system. Folate receptors, known to be overexpressed on the surface of ovarian cancer cells, have remained an ideal candidate for specific targeting through functionalization on nanoparticles and other contrast agents. In combination with copper sulfide nanoparticles' strong absorbance in the near-infrared (NIR), these FA-CuS NPs are an ideal contrast agent capable of being detected by photoacoustic flow cytometry. For the first time, this study shows a potential PA contrast agent to accurately identify ovarian circulating tumor cells in flow.
Collapse
Affiliation(s)
- Joel F Lusk
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| | - Christopher Miranda
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| | - Madeleine Howell
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| | - Matthew Chrest
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| | - Jarrett Eshima
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| | - Barbara S Smith
- School of Biological and Health Systems Engineering, Arizona State University, 550 E. Orange Street, Tempe, Arizona 85281, United States
| |
Collapse
|
48
|
Pantel K, Alix-Panabières C. Liquid biopsy and minimal residual disease — latest advances and implications for cure. Nat Rev Clin Oncol 2019; 16:409-424. [DOI: 10.1038/s41571-019-0187-3] [Citation(s) in RCA: 704] [Impact Index Per Article: 117.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
49
|
Hazra S, Jayaprakash KS, Pandian K, Raj A, Mitra SK, Sen AK. Non-inertial lift induced migration for label-free sorting of cells in a co-flowing aqueous two-phase system. Analyst 2019; 144:2574-2583. [DOI: 10.1039/c8an02267d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We present a novel label-free passive microfluidic technique for isolation of cancer cells (EpCAM+ and CD45−) from peripheral blood mononuclear cells (PBMCs) (CD45+ and EpCAM−) in aqueous two-phase system (ATPS).
Collapse
Affiliation(s)
- S. Hazra
- Department of Mechanical Engineering
- Indian Institute of Technology Madras
- Chennai-600036
- India
| | - K. S. Jayaprakash
- Department of Mechanical Engineering
- Indian Institute of Technology Madras
- Chennai-600036
- India
| | - K. Pandian
- Department of Mechanical Engineering
- Indian Institute of Technology Madras
- Chennai-600036
- India
| | - A. Raj
- Department of Mechanical Engineering
- Indian Institute of Technology Madras
- Chennai-600036
- India
| | - S. K. Mitra
- Waterloo Institute for Nanotechnology
- University of Waterloo
- Canada
| | - A. K. Sen
- Department of Mechanical Engineering
- Indian Institute of Technology Madras
- Chennai-600036
- India
| |
Collapse
|
50
|
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory reaction that includes immune cells, fibroblasts, extracellular matrix, vascular and lymphatic vessels, and nerves. Overwhelming evidence indicates that the pancreatic cancer microenvironment regulates cancer initiation, progression, and maintenance. Pancreatic cancer treatment has progressed little over the past several decades, and the prognosis remains one of the worst for any cancer. The contribution of the microenvironment to carcinogenesis is a key area of research, offering new potential targets for treating the disease. Here, we explore the composition of the pancreatic cancer stroma, discuss the network of interactions between different components, and describe recent attempts to target the stroma therapeutically. We also discuss current areas of active research related to the tumor microenvironment.
Collapse
Affiliation(s)
- Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|