1
|
Gupta S, Singh B, Abhishek R, Gupta S, Sachan M. The emerging role of liquid biopsy in oral squamous cell carcinoma detection: advantages and challenges. Expert Rev Mol Diagn 2024; 24:311-331. [PMID: 38607339 DOI: 10.1080/14737159.2024.2340997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Oral Squamous Cell Carcinoma (OSCC), the sixth most widespread malignancy in the world, accounts for 90% of all cases of oral cancer. The primary risk factors are tobacco chewing, alcohol consumption, viral infection, and genetic modifications. OSCC has a high morbidity rate due to the lack of early diagnostic methods. Nowadays, liquid biopsy plays a vital role in the initial diagnosis of oral cancer. ctNAs extracted from saliva and serum/plasma offer meaningful insights into tumor genetics and dynamics. The interplay of these elements in saliva and serum/plasma showcases their significance in advancing noninvasive, effective OSCC detection and monitoring. AREAS COVERED This review mainly focused on the role of liquid biopsy as an emerging point in the diagnosis and prognosis of OSCC and the current advancements and challenges associated with liquid biopsy. EXPERT OPINION Liquid biopsy is regarded as a new, minimally invasive, real-time monitoring tool for cancer diagnosis and prognosis. Many biomolecules found in bodily fluids, including ctDNA, ctRNA, CTCs, and EVs, are significant biomarkers to identify cancer in its early stages. Despite these groundbreaking strides, challenges persist. Standardization of sample collection, isolation, processing, and detection methods is imperative for ensuring result reproducibility across diverse studies.
Collapse
Affiliation(s)
- Sudha Gupta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Brijesh Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Rajul Abhishek
- Department of Surgical Oncology, Motilal Nehru Medical College, Prayagraj, India
| | - Sameer Gupta
- Department of Surgical Oncology, King George Medical University, Lucknow, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| |
Collapse
|
2
|
Sbrana A, Mazzini G, Comolli G, Antonuzzo A, Danova M. The contribution of automated cytometry in immuno-oncology. Methods Cell Biol 2023; 195:23-37. [PMID: 40180453 DOI: 10.1016/bs.mcb.2023.03.005] [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: 04/05/2025]
Abstract
Cancer immunotherapy has been a real revolution and has given many survival chances to several patients. However, the understanding of resistance to immunotherapy is still an unmet need in clinical practice. Monitoring of immune mechanisms could be a tool to better understand this phenomenon. FCM and CyTOF could be used in this field, since they allow the simultaneous analysis of several protein expressions pattern, thus possibly understanding the functions of several immune cell populations, such as T cells, and their interactions with tumor cells and tumor microenvironment. Furthermore, automated cytometry could be used to understand the interaction of drugs with their target through the analysis of receptor occupancy. Spectral overlap, however, could be a limit for multiple simultaneous analyses. Other possible limitations of these techniques are a low number of cells in samples and the need for viable cells (with the possible interference of cell debris). The lack of standardized protocols, and thus the difficult reproducibility, have been the major limit to their application in clinical practice, so international efforts have been made to get to shared guidelines. Ongoing trials are to answer to the possibility of clinical application of these techniques.
Collapse
Affiliation(s)
- Andrea Sbrana
- Department of Surgical, Medical and Molecular Pathology and Critical Care Area, University of Pisa, Pisa, Italy; Service of Pneumo-Oncology, Unit of Pneumology, Pisa, Italy
| | | | - Giuditta Comolli
- Department of Microbiology and Virology and Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy
| | | | - Marco Danova
- Unit of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, Pavia, Italy; LIUC University, Castellanza, Varese, Italy.
| |
Collapse
|
3
|
Ren J, Liu R. The Implication of Liquid Biopsy in the Non-small Cell Lung Cancer: Potential and Expectation. Methods Mol Biol 2023; 2695:145-163. [PMID: 37450117 DOI: 10.1007/978-1-0716-3346-5_10] [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: 07/18/2023]
Abstract
Nowadays, lung cancer has remained the most lethal cancer, despite great advances in diagnosis and treatment. However, a large proportion of patients were diagnosed with locally advanced or metastatic disease and have poor prognosis. Immunotherapy and targeted drugs have greatly improved the survival and prognosis of patients with advanced lung cancer. However, how to identify the optimal patients to accept those therapies and how to monitor therapeutic efficacy are still in dispute. In the past few decades, tissue biopsy, including percutaneous fine needle biopsy and surgical excision, has still been the gold standard for examining the gene mutation such as EGFR, ALK, ROS, and PD-1/PD/L1, which can indicate the follow-up treatment. Nevertheless, the biopsy techniques mentioned above were invasive and unrepeatable, which were not suitable for advanced patients. Liquid biopsy, accounting for heterogeneity compared with tissue biopsy, is an alternative technique for monitoring the mutation, and a large quantity of research has demonstrated its feasibility to detect the circulating tumor cell, cell-free DNA, circulating tumor DNA, and extracellular vesicles from peripheral venous blood. The proposal of the concept of precision medicine brings a novel medical model developed with the rapid progress of genome sequencing technology and the cross-application of bioinformation, which was based on personalized medicine. The emerging method of liquid biopsy might contribute to promoting the development of precision medicine. In this review, we intend to describe the liquid biopsy in non-small cell lung cancer in detail in the aspect of screening, diagnosis, monitoring, treatment, and drug resistance.
Collapse
Affiliation(s)
- Jianghao Ren
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ruijun Liu
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
4
|
Advances in microfluidics devices and its applications in personalized medicines. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 186:191-201. [PMID: 35033284 DOI: 10.1016/bs.pmbts.2021.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Microfluidics is an exponentially growing area and is being used for numerous applications from basic science to advanced biotechnology and medicines. Microfluidics provides a platform to the research community for studying and building new strategies for the diagnosis and therapeutics applications. In the last decade, microfluidic have enriched the field of diagnostics by providing new solutions which was not possible with conventional detection and treatment methods. Microfluidics has the ability to precisely control and perform high-throughput functions. It has been proven as an efficient and rapid method for biological sample preparation, analysis and controlled drug delivery system. Microfluidics plays significant role in personalized medicine. These personalized medicines are used for medical decisions, practices and other interventions as well as for individual patients based on their predicted response or risk of disease. This chapter highlights microfluidics in developing personalized medical applications for its applications in diseases such as cancer, cardiovascular disease, diabetes, pulmonary disease and several others.
Collapse
|
5
|
Cho HY, Choi JH, Lim J, Lee SN, Choi JW. Microfluidic Chip-Based Cancer Diagnosis and Prediction of Relapse by Detecting Circulating Tumor Cells and Circulating Cancer Stem Cells. Cancers (Basel) 2021; 13:1385. [PMID: 33803846 PMCID: PMC8003176 DOI: 10.3390/cancers13061385] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Detecting circulating tumor cells (CTCs) has been considered one of the best biomarkers in liquid biopsy for early diagnosis and prognosis monitoring in cancer. A major challenge of using CTCs is detecting extremely low-concentrated targets in the presence of high noise factors such as serum and hematopoietic cells. This review provides a selective overview of the recent progress in the design of microfluidic devices with optical sensing tools and their application in the detection and analysis of CTCs and their small malignant subset, circulating cancer stem cells (CCSCs). Moreover, discussion of novel strategies to analyze the differentiation of circulating cancer stem cells will contribute to an understanding of metastatic cancer, which can help clinicians to make a better assessment. We believe that the topic discussed in this review can provide brief guideline for the development of microfluidic-based optical biosensors in cancer prognosis monitoring and clinical applications.
Collapse
Affiliation(s)
- Hyeon-Yeol Cho
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea;
- Interdisciplinary Program for Bio-health Convergence, Kookmin University, Seoul 02707, Korea
| | - Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
- School of Chemical Engineering, Jeonbuk National University, Jeonju 54896, Korea
| | - Joungpyo Lim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| | - Sang-Nam Lee
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| |
Collapse
|
6
|
|
7
|
Zhang X, Lu X, Gao W, Wang Y, Jia C, Cong H. A label-free microfluidic chip for the highly selective isolation of single and cluster CTCs from breast cancer patients. Transl Oncol 2021; 14:100959. [PMID: 33248414 PMCID: PMC7704402 DOI: 10.1016/j.tranon.2020.100959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/31/2020] [Accepted: 11/13/2020] [Indexed: 11/21/2022] Open
Abstract
Our results showed that this new chip had an important clinical value of patients with metastatic breast cancer. CTCs count showed good prospects in monitoring cancer prognosis and guiding future individualized treatment. We have manufactured a new microfluidic chip to isolate and identify CTCs and CTCs clusters with high throughput.
Background Circulating tumor cells (CTCs) existing in peripheral blood can be used to predict the prognosis and survival of cancer patients. The study was designed to detect circulating tumor cells and circulating tumor single cell genes by applying microfluidic chip technology. It was used to explore the clinical application value in breast cancer. Methods We have developed a size-based CTCs sorting microfluidic chip, which contains a hexagonal array and a micro-pipe channel array to isolate and confirm both single CTCs and CTCs clusters. The sorting performance of the as-fabricated chip was tested by analyzing the clinical samples collected from 129 breast cancer patients and 50 healthy persons. Results In this study, the chip can detect different immunophenotypes of CTCs in breast cancer patients. It was found that the new microfluidic device had high sensitivity (73.6%) and specificity (82.0%) in detecting CTCs. By detecting the blood samples of 129 breast cancer patients and 50 healthy blood donors, it was found that the number of CTCs was not associated with clinical factors such as age, gender, pathological type, and tumor size of breast cancer patients (P > 0.05), but was associated with TNM staging of breast cancer, with or without metastasis (P < 0.005). There was a statistically significant difference in the number of CTCs between luminal A (ER+/PR+/HER2-) and HER-2+ (ER-/PR-/HER2+) (P < 0.05). The best cut-off level distinguished by CTC between the breast cancer patients and the healthy persons was 3.5 cells/mL, with 0.845 for AUC-ROC, 0.790–0.901 for 95% CI, 73.6% for sensitivity, and 82% for specificity (P = 0.000). The combination of CTC, CEA, CA125 and CA153 can provide more effective breast cancer screening. Conclusions The CTCs analysis method presented here doesn't rely on the specific antibody, such as anti-EpCAM, which would avoid the missed inspection caused by antibody-relied methods and offer more comprehensive biological information for clinical breast cancer diagnosis and treatment.
Collapse
|
8
|
Wilson RE, O’Connor R, Gallops CE, Kwizera EA, Noroozi B, Morshed BI, Wang Y, Huang X. Immunomagnetic Capture and Multiplexed Surface Marker Detection of Circulating Tumor Cells with Magnetic Multicolor Surface-Enhanced Raman Scattering Nanotags. ACS APPLIED MATERIALS & INTERFACES 2020; 12:47220-47232. [PMID: 32966038 PMCID: PMC11864881 DOI: 10.1021/acsami.0c12395] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circulating tumor cells (CTCs) have substantial clinical implications in cancer diagnosis and monitoring. Although significant progress has been made in developing technologies for CTC detection and counting, the ability to quantitatively detect multiple surface protein markers on individual tumor cells remains very limited. In this work, we report a multiplexed method that uses magnetic multicolor surface-enhanced Raman scattering (SERS) nanotags in conjunction with a chip-based immunomagnetic separation to quantitatively and simultaneously detect four surface protein markers on individual tumor cells in whole blood. Four-color SERS nanotags were prepared using magnetic-optical iron oxide-gold core-shell nanoparticles with different Raman reporters to recognize four different cancer markers with respective antibodies. A microfluidic device was fabricated to magnetically capture the nanoparticle-bound tumor cells and to perform online negative staining and single-cell optical detection. The level of each targeted protein was obtained by signal deconvolution of the mixed SERS signals from individual tumor cells using the classic least squares regression method. The method was tested with spiked tumor cells in human whole blood with three different breast cancer cell lines and compared with the results of purified cancer cells suspended in a phosphate buffer solution. The method, with either spiked cancer cells in blood or purified cancer cells, showed a strong correlation with purified cancer cells by enzyme-linked immunosorbent assay, suggesting the potential of our method for the reliable detection of multiple surface markers on CTCs. Combining immunomagnetic enrichment with high specificity, multiplexed targeting for the capture of CTC subpopulations, multicolor SERS detection with high sensitivity and specificity, microfluidics for handling rare cells and magnetic-plasmonic nanoparticles for dual enrichment and detection, our method provides an integrated, yet a simple and an efficient platform that has the potential to more sensitively detect and monitor cancer metastasis.
Collapse
Affiliation(s)
| | - Ryan O’Connor
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | | | | | - Babak Noroozi
- Department of Computer Science, Texas Tech University, Lubbock, TX 79409
| | - Bashir I. Morshed
- Department of Computer Science, Texas Tech University, Lubbock, TX 79409
| | - Yongmei Wang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| |
Collapse
|
9
|
Chelakkot C, Ryu J, Kim MY, Kim JS, Kim D, Hwang J, Park SH, Ko SB, Park JW, Jung MY, Kim RN, Song K, Kim YJ, Choi YL, Lee HS, Shin YK. An Immune-Magnetophoretic Device for the Selective and Precise Enrichment of Circulating Tumor Cells from Whole Blood. MICROMACHINES 2020; 11:mi11060560. [PMID: 32486306 PMCID: PMC7345362 DOI: 10.3390/mi11060560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
Here, we validated the clinical utility of our previously developed microfluidic device, GenoCTC, which is based on bottom magnetophoresis, for the isolation of circulating tumor cells (CTCs) from patient whole blood. GenoCTC allowed 90% purity, 77% separation rate, and 80% recovery of circulating tumor cells at a 90 μL/min flow rate when tested on blood spiked with epithelial cell adhesion molecule (EpCAM)-positive Michigan Cancer Foundation-7 (MCF7) cells. Clinical studies were performed using blood samples from non-small cell lung cancer (NSCLC) patients. Varying numbers (2 to 114) of CTCs were found in each NSCLC patient, and serial assessment of CTCs showed that the CTC count correlated with the clinical progression of the disease. The applicability of GenoCTC to different cell surface biomarkers was also validated in a cholangiocarcinoma patient using anti-EPCAM, anti-vimentin, or anti-tyrosine protein kinase MET (c-MET) antibodies. After EPCAM-, vimentin-, or c-MET-positive cells were isolated, CTCs were identified and enumerated by immunocytochemistry using anti-cytokeratin 18 (CK18) and anti-CD45 antibodies. Furthermore, we checked the protein expression of PDL1 and c-MET in CTCs. A study in a cholangiocarcinoma patient showed that the number of CTCs varied depending on the biomarker used, indicating the importance of using multiple biomarkers for CTC isolation and enumeration.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jiyeon Ryu
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Mi Young Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea; (M.Y.K.); (J.-S.K.)
| | - Dohyeong Kim
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Juhyun Hwang
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Sung Hoon Park
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Seok Bum Ko
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
| | - Jeong Won Park
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Moon Youn Jung
- IT Convergence Technology Research Laboratory, Electronic and Telecommunications Research Institute, Daejon 34129, Korea; (J.W.P.); (M.Y.J.)
| | - Ryong Nam Kim
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea;
| | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yu Jin Kim
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul 08394, Korea; (K.S.); (Y.J.K.)
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 08394, Korea;
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hun Seok Lee
- Technical Research Center, Genobio Corp., Seoul 08394, Korea; (C.C.); (J.R.); (D.K.); (J.H.); (S.H.P.); (S.B.K.)
- Correspondence: (H.S.L.); (Y.K.S.)
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul 08826, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
- The Center for Anti-Cancer Companion Diagnostics, Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Korea
- Correspondence: (H.S.L.); (Y.K.S.)
| |
Collapse
|
10
|
Zhou J, Kulasinghe A, Bogseth A, O’Byrne K, Punyadeera C, Papautsky I. Isolation of circulating tumor cells in non-small-cell-lung-cancer patients using a multi-flow microfluidic channel. MICROSYSTEMS & NANOENGINEERING 2019; 5:8. [PMID: 31057935 PMCID: PMC6387977 DOI: 10.1038/s41378-019-0045-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/04/2018] [Accepted: 12/22/2018] [Indexed: 05/03/2023]
Abstract
Circulating tumor cells (CTCs) carry a wealth of information on primary and metastatic tumors critical for precise cancer detection, monitoring, and treatment. Numerous microfluidic platforms have been developed in the past few years to capture these rare cells in patient bloodstream for deciphering the critical information needed. However, the practical need for a high-quality method of CTC isolation remains to be met. Herein, we demonstrate a novel multi-flow microfluidic device that is able to sensitively provide high purity (>87%) of separation outcome without labeling. Our device is constructed and configured based on the phenomenal effect of size-dependent inertial migration. The recovery rate of >93% has been achieved using spiked cancer cells at clinically relevant concentrations (10 cells per 5 mL and above). We have also successfully detected CTCs from 6 out of 8 non-small-cell-lung-cancer (NSCLC) patients, while none for 5 healthy control subjects. With these results, we envision our approach is a promising alternative for reliable CTC capture, and thus for facilitating the progress of extracting information from CTCs to personalize treatment strategies for solid tumor patients.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607 USA
- University of Illinois Cancer Center, Chicago, IL 60612 USA
| | - Arutha Kulasinghe
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD Australia
- Translational Research Institute, Brisbane, QLD Australia
| | - Amanda Bogseth
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607 USA
| | - Ken O’Byrne
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD Australia
- Princess Alexandra Hospital, Woolloongabba, QLD Australia
| | - Chamindie Punyadeera
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD Australia
- Translational Research Institute, Brisbane, QLD Australia
| | - Ian Papautsky
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607 USA
- University of Illinois Cancer Center, Chicago, IL 60612 USA
| |
Collapse
|
11
|
Danova M, Torchio M, Comolli G, Sbrana A, Antonuzzo A, Mazzini G. The role of automated cytometry in the new era of cancer immunotherapy. Mol Clin Oncol 2018; 9:355-361. [PMID: 30233791 DOI: 10.3892/mco.2018.1701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
The introduction in the clinical practice of several new approaches to cancer immunotherapy has greatly increased the interest in analytical methodologies that can define the immunological profile of patients in the clinical setting. This requires huge effort to obtain reliable monitoring tools that could be used to improve the patient's clinical outcome. The clinical applications of flow cytometry (FCM) in oncology started with the measurement of DNA content for the evaluation of both ploidy and cell cycle profile as potential prognostic parameters in the majority of human solid cancer types. The availability of monoclonal antibodies widely broadened the spectrum of clinical applications of this technique, which rapidly became a fundamental tool for the diagnosis and prognosis of malignant hematological diseases. Among the emerging clinical applications of FCM, the study of minimal residual disease in hematological malignancies, the quantification of blood dendritic cells in various types of tumors, the study of metastatic spread in solid tumors throughout both the analysis of circulating endothelial progenitor cells and the identification and characterization of circulating tumor cells, all appear very promising. More recently, an advanced single cell analysis technique has been developed that combines the precision of mass spectrometry with the unique advantages of FCM. This approach, termed mass cytometry, utilizes antibodies conjugated to heavy metal ions for the analysis of cellular proteins by a mass spectrometer. It provides measurement of over 100 simultaneous cellular parameters in a single sample and has evolved from a promising technology to a high recognized platform for multi-dimensional single-cell analysis. Should a careful standardization of the analytical procedures be reached, both FCM and mass cytometry could effectively become ideal tools for the optimization of new immunotherapeutic approaches in cancer patients.
Collapse
Affiliation(s)
- Marco Danova
- Department of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, ASST of Pavia, I-27029 Vigevano, Italy
| | - Martina Torchio
- Department of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, ASST of Pavia, I-27029 Vigevano, Italy
| | - Giuditta Comolli
- Department of Microbiology and Virology and Biotechnology Laboratories, IRCCS San Matteo Foundation, I-27100 Pavia, Italy
| | - Andrea Sbrana
- Department of Medical Oncology 2, University Hospital of Pisa, I-56126 Pisa, Italy
| | - Andrea Antonuzzo
- Department of Medical Oncology 2, University Hospital of Pisa, I-56126 Pisa, Italy
| | | |
Collapse
|
12
|
Yadav DK, Bai X, Yadav RK, Singh A, Li G, Ma T, Chen W, Liang T. Liquid biopsy in pancreatic cancer: the beginning of a new era. Oncotarget 2018; 9:26900-26933. [PMID: 29928492 PMCID: PMC6003564 DOI: 10.18632/oncotarget.24809] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/25/2018] [Indexed: 12/21/2022] Open
Abstract
With dismal survival rate pancreatic cancer remains one of the most aggressive and devastating malignancy. Predominantly, due to the absence of a dependable methodology for early identification and limited therapeutic options for advanced disease. However, it takes over 17 years to develop pancreatic cancer from initiation of mutation to metastatic cancer; therefore, if diagnosed early; it may increase overall survival dramatically, thus, providing a window of opportunity for early detection. Recently, genomic expression analysis defined 4 subtypes of pancreatic cancer based on mutated genes. Hence, we need simple and standard, minimally invasive test that can monitor those altered genes or their associated pathways in time for the success of precision medicine, and liquid biopsy seems to be one answer to all these questions. Again, liquid biopsy has an ability to pair with genomic tests. Additionally, liquid biopsy based development of circulating tumor cells derived xenografts, 3D organoids system, real-time monitoring of genetic mutations by circulating tumor DNA and exosome as the targeted drug delivery vehicle holds lots of potential for the treatment and cure of pancreatic cancer. At present, diagnosis of pancreatic cancer is frantically done on the premise of CA19-9 and radiological features only, which doesn't give a picture of genetic mutations and epigenetic alteration involved. In this manner, the current diagnostic paradigm for pancreatic cancer diagnosis experiences low diagnostic accuracy. This review article discusses the current state of liquid biopsy in pancreatic cancer as diagnostic and therapeutic tools and future perspectives of research in the light of circulating tumor cells, circulating tumor DNA and exosomes.
Collapse
Affiliation(s)
- Dipesh Kumar Yadav
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Rajesh Kumar Yadav
- Department of Pharmacology, Gandaki Medical College, Tribhuwan University, Institute of Medicine, Pokhara 33700, Nepal
| | - Alina Singh
- Department of Surgery, Bir Hospital, National Academy of Medical Science, Kanti Path, Kathmandu 44600, Nepal
| | - Guogang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
13
|
Abstract
Circulating tumor cells are a hallmark of cancer metastasis which accounts for approximately 90% of all cancer-related deaths. Their detection and characterization have significant implications in cancer biology and clinical practice. However, CTCs are rare cells and consist of heterogeneous subpopulations, requiring highly sensitive and specific techniques to identify and isolate them with high efficiency. Nanomaterials, with unique structural and functional properties, have shown strong promise to meet the challenging demands. In this review, we discuss CTC capture and therapeutic targeting, emphasizing the significance of the nanomaterials being used for this purpose. The next generation of therapy for metastatic cancer may well involve capturing and even directly neutralizing CTCs using nanomaterials.
Collapse
Affiliation(s)
- Zhenjiang Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| |
Collapse
|
14
|
Abstract
In these last few decades the great explosion of the molecular approaches has casted a little shadow on the DNA quantitative analysis. Nevertheless DNA cytochemistry represented a long piece of history in cell biology since the advent of the Feulgen reaction. This discovery was really the milestone of the emerging quantitative cytochemistry, and scientists from all over the world produced a very large literature on this subject. This first era of quantitation (histochemistry followed by cytochemistry) started by means of absorption measurements (histophotometry and cytophotometry). The successive introduction of fluorescence microscopy gave a great boost to quantitation, making easier and faster the determination of cell components by means of cytofluorometry. The development of flow cytometry further contributed to the importance of quantitative cytochemistry. At its beginning, the mission of flow cytometry was still DNA quantitation. For a decade the Feulgen reaction had been the reference methodology for both conventional and flow cytofluorometry; the advent of Shiff-type reagents contributed to expand the variety of possible fluorochromes excitable in the entire visible spectrum as well as in the ultraviolet region. The fluorescence scenario was progressively enriched by new probes among which are the intercalating dyes which made DNA quantitation simple and fast, thus spreading it worldwide. The final explosion of cytofluorometry was made possible by the availability of a large variety of probes directly binding DNA structure. In addition, immunofluorescence allowed to correlate the cell cycle-related DNA content to other cell markers. In the clinical application of flow cytometry, this promoted the introduction of multiparametric analyses aimed at describing the cytokinetic characteristics of a given cell subpopulation defined by a specific immunophenotype setting.
Collapse
Affiliation(s)
- Giuliano Mazzini
- Institute of Molecular Genetics, CNR, Via Abbiategrasso 207, 27100, Pavia, Italy. .,Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.
| | - Marco Danova
- Department of Medicine, Azienda Socio-Sanitaria Territoriale of Pavia, Pavia, Italy
| |
Collapse
|
15
|
Huang X, O'Connor R, Kwizera EA. Gold Nanoparticle Based Platforms for Circulating Cancer Marker Detection. Nanotheranostics 2017; 1:80-102. [PMID: 28217434 PMCID: PMC5313055 DOI: 10.7150/ntno.18216] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Detection of cancer-related circulating biomarkers in body fluids has become a cutting-edge technology that has the potential to noninvasively screen cancer, diagnose cancer at early stage, monitor tumor progression, and evaluate therapy responses. Traditional molecular and cellular detection methods are either insensitive for early cancer intervention or technically costly and complicated making them impractical for typical clinical settings. Due to their exceptional structural and functional properties that are not available from bulk materials or discrete molecules, nanotechnology is opening new horizons for low cost, rapid, highly sensitive, and highly specific detection of circulating cancer markers. Gold nanoparticles have emerged as a unique nanoplatform for circulating biomarker detection owning to their advantages of easy synthesis, facile surface chemistry, excellent biocompatibility, and remarkable structure and environment sensitive optical properties. In this review, we introduce current gold nanoparticle-based technology platforms for the detection of four major classes of circulating cancer markers - circulating tumor cells, vesicles, nucleic acids, and proteins. The techniques will be summarized in terms of signal detection strategies. Distinctive examples are provided to highlight the state-of-the-art technologies that significantly advance basic and clinical cancer research.
Collapse
Affiliation(s)
- Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | - Ryan O'Connor
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | | |
Collapse
|
16
|
Abdouh M, Hamam D, Arena V, Arena M, Alamri H, Arena GO. Novel blood test to predict neoplastic activity in healthy patients and metastatic recurrence after primary tumor resection. J Circ Biomark 2016; 5:1849454416663661. [PMID: 28936263 PMCID: PMC5548309 DOI: 10.1177/1849454416663661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
We reported that single oncosuppressor-mutated (SOM) cells turn malignant when exposed to cancer patients’ sera. We tested the possibility to incorporate this discovery into a biological platform able to detect cancer in healthy individuals and to predict metastases after tumor resection. Blood was drawn prior to tumor resection and within a year after surgery. Blood samples from healthy individuals or metastatic patients were used as negative and positive controls, respectively. Patients at risk for cancer were included in the screening cohort. Once treated, cells were injected into nonobese diabetic/severe combined immunodeficiency mice to monitor tumor growth. All samples of sera coming from metastatic patients transformed SOM cells into malignant cells. Four samples from screened patients transformed SOM cells. Further clinical tests done on these patients showed the presence of early cancerous lesions despite normal tumor markers. Based on the xenotransplants size, we were able to predict metastasis in three patients before diagnostic tests confirmed the presence of the metastatic lesions. These data show that this serum-based platform has potentials to be used for cancer screening and for identification of patients at risks to develop metastases regardless of the Tumor Node Metastasis (TNM) stage or tumor markers level.
Collapse
Affiliation(s)
- Mohamed Abdouh
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Quebec, Canada
| | - Dana Hamam
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Quebec, Canada.,Department of Experimental Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Vincenzo Arena
- Department of Obstetrics and Gynecology, Santo Bambino Hospital, Catania, Italy
| | - Manuel Arena
- Department of Surgical Sciences, Organ Transplantation and Advances Technologies, University of Catania, Catania, Italy
| | - Hussam Alamri
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Quebec, Canada.,Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - Goffredo Orazio Arena
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Quebec, Canada.,Department of Surgery, McGill University, St Mary Hospital, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Forte VA, Barrak DK, Elhodaky M, Tung L, Snow A, Lang JE. The potential for liquid biopsies in the precision medical treatment of breast cancer. Cancer Biol Med 2016; 13:19-40. [PMID: 27144060 PMCID: PMC4850125 DOI: 10.28092/j.issn.2095-3941.2016.0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Currently the clinical management of breast cancer relies on relatively few prognostic/predictive clinical markers (estrogen receptor, progesterone receptor, HER2), based on primary tumor biology. Circulating biomarkers, such as circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs) may enhance our treatment options by focusing on the very cells that are the direct precursors of distant metastatic disease, and probably inherently different than the primary tumor's biology. To shift the current clinical paradigm, assessing tumor biology in real time by molecularly profiling CTCs or ctDNA may serve to discover therapeutic targets, detect minimal residual disease and predict response to treatment. This review serves to elucidate the detection, characterization, and clinical application of CTCs and ctDNA with the goal of precision treatment of breast cancer.
Collapse
Affiliation(s)
- Victoria A Forte
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Dany K Barrak
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Mostafa Elhodaky
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Stem Cell and Regenerative Medicine, USC, Los Angeles, CA 90033, USA
| | - Lily Tung
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| | - Anson Snow
- Department of Medicine, Division of Medical Oncology, University of Southern California (USC), Los Angeles, CA 90033, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Julie E Lang
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA; Department of Surgery, Division of Breast, Endocrine and Soft Tissue Surgery, USC, Los Angeles, CA 90033, USA
| |
Collapse
|
18
|
Gabriel MT, Calleja LR, Chalopin A, Ory B, Heymann D. Circulating Tumor Cells: A Review of Non–EpCAM-Based Approaches for Cell Enrichment and Isolation. Clin Chem 2016; 62:571-81. [DOI: 10.1373/clinchem.2015.249706] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/04/2016] [Indexed: 12/14/2022]
Abstract
Abstract
BACKGROUND
Circulating tumor cells (CTCs) are biomarkers for noninvasively measuring the evolution of tumor genotypes during treatment and disease progression. Recent technical progress has made it possible to detect and characterize CTCs at the single-cell level in blood.
CONTENT
Most current methods are based on epithelial cell adhesion molecule (EpCAM) detection, but numerous studies have demonstrated that EpCAM is not a universal marker for CTC detection because it fails to detect both carcinoma cells that undergo epithelial-mesenchymal transition (EMT) and CTCs of mesenchymal origin. Moreover, EpCAM expression has been found in patients with benign diseases. A large proportion of the current studies and reviews about CTCs describe EpCAM-based methods, but there is evidence that not all tumor cells can be detected using this marker. Here we describe the most recent EpCAM-independent methods for enriching, isolating, and characterizing CTCs on the basis of physical and biological characteristics and point out the main advantages and disadvantages of these methods.
SUMMARY
CTCs offer an opportunity to obtain key biological information required for the development of personalized medicine. However, there is no universal marker of these cells. To strengthen the clinical utility of CTCs, it is important to improve existing technologies and develop new, non–EpCAM-based systems to enrich and isolate CTCs.
Collapse
Affiliation(s)
- Marta Tellez Gabriel
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes, France
- Université de Nantes, Nantes Atlantique Universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Lidia Rodriguez Calleja
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes, France
- Université de Nantes, Nantes Atlantique Universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Antoine Chalopin
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes, France
- Université de Nantes, Nantes Atlantique Universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
- CHU de Nantes, Nantes, France
| | - Benjamin Ory
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes, France
- Université de Nantes, Nantes Atlantique Universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Dominique Heymann
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes, France
- Université de Nantes, Nantes Atlantique Universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
- CHU de Nantes, Nantes, France
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| |
Collapse
|
19
|
Caceres G, Puskas JA, Magliocco AM. Circulating Tumor Cells: A Window Into Tumor Development and Therapeutic Effectiveness. Cancer Control 2016; 22:167-76. [PMID: 26068761 DOI: 10.1177/107327481502200207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are an important diagnostic tool for understanding the metastatic process and the development of cancer. METHODS This review covers the background, relevance, and potential limitations of CTCs as a measurement of cancer progression and how information derived from CTCs may affect treatment efficacy. It also highlights the difficulties of characterizing these rare cells due to the limited cell surface molecules unique to CTCs and each particular type of cancer. RESULTS The analysis of cancer in real time, through the measure of the number of CTCs in a " liquid" biopsy specimen, gives us the ability to monitor the therapeutic efficacy of treatments and possibly the metastatic potential of a tumor. CONCLUSIONS Through novel and innovative techniques yielding encouraging results, including microfluidic techniques, isolating and molecularly analyzing CTCs are becoming a reality. CTCs hold promise for understanding how tumors work and potentially aiding in their demise.
Collapse
Affiliation(s)
- Gisela Caceres
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
20
|
Zmijan R, Jonnalagadda US, Carugo D, Kochi Y, Lemm E, Packham G, Hill M, Glynne-Jones P. High throughput imaging cytometer with acoustic focussing. RSC Adv 2015; 5:83206-83216. [PMID: 29456838 PMCID: PMC5782801 DOI: 10.1039/c5ra19497k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/23/2015] [Indexed: 11/25/2022] Open
Abstract
We demonstrate an imaging flow cytometer that uses acoustic levitation to assemble cells and other particles into a sheet structure. This technique enables a high resolution, low noise CMOS camera to capture images of thousands of cells with each frame. While ultrasonic focussing has previously been demonstrated for 1D cytometry systems, extending the technology to a planar, much higher throughput format and integrating imaging is non-trivial, and represents a significant jump forward in capability, leading to diagnostic possibilities not achievable with current systems. A galvo mirror is used to track the images of the moving cells permitting exposure times of 10 ms at frame rates of 50 fps with motion blur of only a few pixels. At 80 fps, we demonstrate a throughput of 208 000 beads per second. We investigate the factors affecting motion blur and throughput, and demonstrate the system with fluorescent beads, leukaemia cells and a chondrocyte cell line. Cells require more time to reach the acoustic focus than beads, resulting in lower throughputs; however a longer device would remove this constraint.
Collapse
Affiliation(s)
- Robert Zmijan
- Engineering Sciences, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Umesh S Jonnalagadda
- Engineering Sciences, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Dario Carugo
- Engineering Sciences, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Yu Kochi
- Japan Patent Office, 3-chome-4-3 Kasumigaseki, Chiyoda-ku Tokyo 100-8915, Japan
| | - Elizabeth Lemm
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton General Hospital, UK
- Experimental Cancer Medicine Centre, Southampton General Hospital, UK
| | - Graham Packham
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton General Hospital, UK
- Experimental Cancer Medicine Centre, Southampton General Hospital, UK
| | - Martyn Hill
- Engineering Sciences, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Peter Glynne-Jones
- Engineering Sciences, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
21
|
Mazzini G, Carpignano F, Surdo S, Aredia F, Panini N, Torchio M, Erba E, Danova M, Scovassi AI, Barillaro G, Merlo S. 3D Silicon Microstructures: A New Tool for Evaluating Biological Aggressiveness of Tumor Cells. IEEE Trans Nanobioscience 2015; 14:797-805. [PMID: 26353377 DOI: 10.1109/tnb.2015.2476351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this work, silicon micromachined structures (SMS), consisting of arrays of 3- μ m-thick silicon walls separated by 50- μm-deep, 5- μ m-wide gaps, were applied to investigate the behavior of eight tumor cell lines, with different origins and biological aggressiveness, in a three-dimensional (3D) microenvironment. Several cell culture experiments were performed on 3D-SMS and cells grown on silicon were stained for fluorescence microscopy analyses. Most of the tumor cell lines recognized in the literature as highly aggressive (OVCAR-5, A375, MDA-MB-231, and RPMI-7951) exhibited a great ability to enter and colonize the narrow deep gaps of the SMS, whereas less aggressive cell lines (OVCAR-3, Capan-1, MCF7, and NCI-H2126) demonstrated less penetration capability and tended to remain on top of the SMS. Quantitative image analyses of several fluorescence microscopy fields of silicon samples were performed for automatic cell recognition and count, in order to quantify the fraction of cells inside the gaps, with respect to the total number of cells in the examined field. Our results show that higher fractions of cells in the gaps are obtained with more aggressive cell lines, thus supporting in a quantitative way the observation that the behavior of tumor cells on the 3D-SMS depends on their aggressiveness level.
Collapse
|
22
|
Satelli A, Mitra A, Brownlee Z, Xia X, Bellister S, Overman MJ, Kopetz S, Ellis LM, Meng QH, Li S. Epithelial-mesenchymal transitioned circulating tumor cells capture for detecting tumor progression. Clin Cancer Res 2014; 21:899-906. [PMID: 25516888 DOI: 10.1158/1078-0432.ccr-14-0894] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE This study aimed to detect cell-surface vimentin (CSV) on the surface of epithelial-mesenchymal transitioned (EMT) circulating tumor cells (CTC) from blood of patients with epithelial cancers. EXPERIMENTAL DESIGN In this study, 101 patients undergoing postsurgery adjuvant chemotherapy for metastatic colon cancer were recruited. EMT CTCs were detected from blood of patients using the 84-1 monoclonal antibody against CSV as a marker. EMT CTCs isolated were characterized further using EMT-specific markers, fluorescent in situ hybridization, and single-cell mutation analysis. RESULTS Using the 84-1 antibody, we detected CSV exclusively on EMT CTCs from a variety of tumor types but not in the surrounding normal cells in the blood. The antibody exhibited very high specificity and sensitivity toward different epithelial cancer cells. With this antibody, we detected and enumerated EMT CTCs from patients. From our observations, we defined a cutoff of <5 or ≥5 EMT CTCs as the optimal threshold with respect to therapeutic response using ROC curves. Using this defined threshold, the presence of ≥5 EMT CTCs was associated with progressive disease, whereas patients with <5 EMT CTCs showed therapeutic response. CONCLUSION Taken together, the number of EMT CTCs detected correlated with the therapeutic outcome of the disease. These results establish CSV as a universal marker for EMT CTCs from a wide variety of tumor types and thus provide the foundation for emerging CTC detection technologies and for studying the molecular regulation of these EMT CTCs.
Collapse
Affiliation(s)
- Arun Satelli
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abhisek Mitra
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zachary Brownlee
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xueqing Xia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seth Bellister
- Department of Surgical Oncology, Division of Surgery and Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Surgical Oncology, Division of Surgery and Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lee M Ellis
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qing H Meng
- Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
23
|
In-vitro culture system for mesenchymal progenitor cells derived from waste human ovarian follicular fluid. Reprod Biomed Online 2014; 29:457-69. [PMID: 25131558 DOI: 10.1016/j.rbmo.2014.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 11/21/2022]
Abstract
To characterize different cell populations in the human ovary, morphological and functional characteristics of cell populations collected during routine IVF procedures were studied. Cells obtained from follicular fluid grew in vitro under minimal medium conditions, without growth factor, including leukaemia-inhibiting factor. Morphological analysis revealed a heterogeneous cell population, with cells displaying a fibroblast-like, epithelial-like and also neuron-like features. Morpho-functional characteristics of fibroblast-like cells were similar to mesenchymal stem cells, and, in particular, were positive for mesenchymal stemness markers, including CD90, CD44, CD105, CD73, but negative for epithelial proteins, such as cytokeratins, CD34 and CD45 antigens. Cell proliferation activity at different times and colony-forming unit capability were evaluated, and multipotency of a subset of granulosa cells was established by in-vitro differentiation studies (e.g. osteogenic, chondrogenic and adipogenic differentiation). This study suggests that cells provided by mesenchymal plasticity can be easily isolated by waste follicular fluid, avoiding scraping of human ovaries, and cultivated in minimal conditions. Successful growth of such progenitor cells on three-dimensional cryogel scaffold provides the basis for future developments in tissue engineering. This culture system may be regarded as an experimental model in which biological behaviour is not influenced by specific growth factors.
Collapse
|
24
|
Bhana S, Chaffin E, Wang Y, Mishra SR, Huang X. Capture and detection of cancer cells in whole blood with magnetic–optical nanoovals. Nanomedicine (Lond) 2014; 9:593-606. [DOI: 10.2217/nnm.13.77] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop a simple assay for the capture and detection of rare cancer cells in whole blood using iron oxide–gold (IO–Au) nanoparticles. Materials & methods: IO–Au nanoovals (NOVs) were synthesized, coated with Raman tags and linked with antibodies targeting breast cancer. An integrated system was constructed for on-line magnetic cell capture and surface-enhanced Raman scattering (SERS) detection. The capabilities of IO–Au SERS NOVs to capture and detect rare cancer cells in blood were investigated in the integrated system using circulating tumor cell-mimic SK-BR-3 cells. Results: SK-BR-3 cells in whole blood were magnetically captured under a flow condition using IO–Au SERS NOVs, followed by on-line SERS detection with a limit of detection of 1–2 cells/ml blood. Conclusion: We developed a sensitive method that can capture and detect cancer cells in whole blood with a single nanoconstruct, which is highly promising for the detection of circulating tumor cells in the clinic. Original submitted 7 December 2012; Revised submitted 25 March 2013
Collapse
Affiliation(s)
- Saheel Bhana
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA
| | - Elise Chaffin
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA
| | - Yongmei Wang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA
| | - Sanjay R Mishra
- Department of Physics, The University of Memphis, Memphis, TN 38152, USA
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
25
|
Chen Y, Li P, Huang PH, Xie Y, Mai JD, Wang L, Nguyen NT, Huang TJ. Rare cell isolation and analysis in microfluidics. LAB ON A CHIP 2014; 14:626-45. [PMID: 24406985 PMCID: PMC3991782 DOI: 10.1039/c3lc90136j] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Rare cells are low-abundance cells in a much larger population of background cells. Conventional benchtop techniques have limited capabilities to isolate and analyze rare cells because of their generally low selectivity and significant sample loss. Recent rapid advances in microfluidics have been providing robust solutions to the challenges in the isolation and analysis of rare cells. In addition to the apparent performance enhancements resulting in higher efficiencies and sensitivity levels, microfluidics provides other advanced features such as simpler handling of small sample volumes and multiplexing capabilities for high-throughput processing. All of these advantages make microfluidics an excellent platform to deal with the transport, isolation, and analysis of rare cells. Various cellular biomarkers, including physical properties, dielectric properties, as well as immunoaffinities, have been explored for isolating rare cells. In this Focus article, we discuss the design considerations of representative microfluidic devices for rare cell isolation and analysis. Examples from recently published works are discussed to highlight the advantages and limitations of the different techniques. Various applications of these techniques are then introduced. Finally, a perspective on the development trends and promising research directions in this field are proposed.
Collapse
Affiliation(s)
- Yuchao Chen
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Peng Li
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Po-Hsun Huang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuliang Xie
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - John D. Mai
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, PR China
| | - Lin Wang
- Ascent Bio-Nano Technologies Inc., State College, PA 16801, USA
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Brisbane 4111, Australia
| | - Tony Jun Huang
- Fax: 814-865-9974; Tel: 814-863-4209; Fax: 61-(0)7-3735-8021; Tel: 61-(0)7-3735-3921;
| |
Collapse
|
26
|
Wang L, Asghar W, Demirci U, Wan Y. Nanostructured substrates for isolation of circulating tumor cells. NANO TODAY 2013; 8:347-387. [PMID: 24944563 PMCID: PMC4059613 DOI: 10.1016/j.nantod.2013.07.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating tumor cells (CTCs) originate from the primary tumor mass and enter into the peripheral bloodstream. CTCs hold the key to understanding the biology of metastasis and also play a vital role in cancer diagnosis, prognosis, disease monitoring, and personalized therapy. However, CTCs are rare in blood and hard to isolate. Additionally, the viability of CTCs can easily be compromised under high shear stress while releasing them from a surface. The heterogeneity of CTCs in biomarker expression makes their isolation quite challenging; the isolation efficiency and specificity of current approaches need to be improved. Nanostructured substrates have emerged as a promising biosensing platform since they provide better isolation sensitivity at the cost of specificity for CTC isolation. This review discusses major challenges faced by CTC isolation techniques and focuses on nanostructured substrates as a platform for CTC isolation.
Collapse
Affiliation(s)
- Lixue Wang
- Department of Oncology, The Second Affiliated Hospital of Southeast University, Southeast University, Nanjing, Jiangsu 210003, PR China
| | - Waseem Asghar
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Center for Biomedical Engineering, Renal Division and Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Center for Biomedical Engineering, Renal Division and Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-Massachusetts Institute of Technology (MIT), Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Yuan Wan
- Department of Oncology, The Second Affiliated Hospital of Southeast University, Southeast University, Nanjing, Jiangsu 210003, PR China
- Ian Wark Research Institute, University of South Australia, Mawson Lakes, Adelaide, SA 5095, Australia
| |
Collapse
|
27
|
Karimi A, Yazdi S, Ardekani AM. Hydrodynamic mechanisms of cell and particle trapping in microfluidics. BIOMICROFLUIDICS 2013; 7:21501. [PMID: 24404005 PMCID: PMC3631262 DOI: 10.1063/1.4799787] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/21/2013] [Indexed: 05/03/2023]
Abstract
Focusing and sorting cells and particles utilizing microfluidic phenomena have been flourishing areas of development in recent years. These processes are largely beneficial in biomedical applications and fundamental studies of cell biology as they provide cost-effective and point-of-care miniaturized diagnostic devices and rare cell enrichment techniques. Due to inherent problems of isolation methods based on the biomarkers and antigens, separation approaches exploiting physical characteristics of cells of interest, such as size, deformability, and electric and magnetic properties, have gained currency in many medical assays. Here, we present an overview of the cell/particle sorting techniques by harnessing intrinsic hydrodynamic effects in microchannels. Our emphasis is on the underlying fluid dynamical mechanisms causing cross stream migration of objects in shear and vortical flows. We also highlight the advantages and drawbacks of each method in terms of throughput, separation efficiency, and cell viability. Finally, we discuss the future research areas for extending the scope of hydrodynamic mechanisms and exploring new physical directions for microfluidic applications.
Collapse
Affiliation(s)
- A Karimi
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S Yazdi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - A M Ardekani
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
28
|
Zheng W, Zhang W, Jiang X. Precise control of cell adhesion by combination of surface chemistry and soft lithography. Adv Healthc Mater 2013. [PMID: 23184447 DOI: 10.1002/adhm.201200104] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The adhesion of cells on an extracellular matrix (ECM) (in vivo) or the surfaces of materials (in vitro) is a prerequisite for most cells to survive. The rapid growth of nano/microfabrication and biomaterial technologies has provided new materials with excellent surfaces with specific, desirable biological interactions with their surroundings. On one hand, the chemical and physical properties of material surfaces exert an extensive influence on cell adhesion, proliferation, migration, and differentiation. On the other hand, material surfaces are useful for fundamental cell biology research and tissue engineering. In this Review, an overview will be given of the chemical and physical properties of newly developed material surfaces and their biological effects, as well as soft lithographic techniques and their applications in cell biology research. Recent advances in the manipulation of cell adhesion by the combination of surface chemistry and soft lithography will also be highlighted.
Collapse
Affiliation(s)
- Wenfu Zheng
- National Center for NanoScience and Technology, Beijing, China
| | | | | |
Collapse
|
29
|
Chen J, Li J, Sun Y. Microfluidic approaches for cancer cell detection, characterization, and separation. LAB ON A CHIP 2012; 12:1753-67. [PMID: 22437479 DOI: 10.1039/c2lc21273k] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
This article reviews the recent developments in microfluidic technologies for in vitro cancer diagnosis. We summarize the working principles and experimental results of key microfluidic platforms for cancer cell detection, characterization, and separation based on cell-affinity micro-chromatography, magnetic activated micro-sorting, and cellular biophysics (e.g., cell size and mechanical and electrical properties). We examine the advantages and limitations of each technique and discuss future research opportunities for improving device throughput and purity, and for enabling on-chip analysis of captured cancer cells.
Collapse
Affiliation(s)
- Jian Chen
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | | | | |
Collapse
|
30
|
Jacob J, Krell J, Castellano L, Jiao LR, Stebbing J, Frampton AE. Determination of cut-offs for circulating tumor cell measurement in metastatic cancer. Expert Rev Anticancer Ther 2011; 11:1345-50. [PMID: 21929309 DOI: 10.1586/era.11.133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The metastatic transformation of epithelial tumors progresses through various steps leading to the generation of circulating tumor cells (CTCs). Measurement of CTCs in the peripheral blood is being increasingly recognized as a promising tool in breast oncology. Several studies have evaluated the prognostic significance of CTCs in newly diagnosed metastatic breast cancer (MBC) patients. The IC 2006-04 was a high-powered, prospective, multicenter, observational study conceived to assess CTC changes in women with MBC treated with first-line chemotherapy. Levels ≥ 5 CTCs/7.5 ml blood at baseline and before the second cycle of treatment were independent prognostic factors associated with shorter progression-free and overall survival. This study provides further level II evidence for the clinical and prognostic value of CTCs in MBC, confirming data from earlier small studies. It also provides proof that CTCs should be investigated in ongoing interventional trials to see if better patient outcomes can be attained by altering treatment based on CTC levels.
Collapse
Affiliation(s)
- Jimmy Jacob
- Dept. of Oncology, Dept. of Surgery and Cancer, 5th Floor, MRC Cyclotron Building, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | | | | | | | | | | |
Collapse
|