1
|
Wu P, He X, Fan J, Tai Y, Zheng D, Yao Y, Sun S, Luo Y, Chen J, Hu WW, Ying B, Luo F, Niu Q, Sun X, Li Y. Electrochemical cytosensors for non-invasive liquid biopsy: Detection procedures and technologies for circulating tumor cells. Biosens Bioelectron 2025; 267:116818. [PMID: 39353368 DOI: 10.1016/j.bios.2024.116818] [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: 06/04/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Each year, millions of new cancer cases and cancer-related deaths underscore the urgent need for effective, affordable screening methods. Circulating tumor cells (CTCs), which derived from tumors and shedding into bloodstream, are considered promising biomarkers for liquid biopsy due to their unique biological significance and the substantial volume of supporting research. Among many advanced CTCs detection methods, electrochemical sensing is rapidly developing due to their high selectivity, high sensitivity, low cost, and rapid detection capability, well meeting the growing demand for non-invasive liquid biopsy. This review focuses on the entire procedure of detecting CTCs using electrochemical cytosensors, starting from sample preparation, detailing bio-recognition elements for capturing CTCs, highlighting design strategies of cytosensor, and discussing the prospects and challenges of electrochemical cytosensor applications.
Collapse
Affiliation(s)
- Peilin Wu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Jiwen Fan
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Yunze Tai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Dongdong Zheng
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yongchao Yao
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Shengjun Sun
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yao Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Binwu Ying
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qian Niu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China; College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yi Li
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Yang X, Zhang Z, Bi X. A nomogram for predicting colorectal cancer liver metastasis using circulating tumor cells from the first drainage vein. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108579. [PMID: 39121633 DOI: 10.1016/j.ejso.2024.108579] [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: 10/02/2023] [Revised: 03/05/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE To use circulating tumor cells (CTC) from the first drainage vein (FDV) of the primary lesion and other clinically relevant parameters to construct a nomogram for predicting liver metastasis in colorectal cancer (CRC) patients, and to provide a theoretical basis for clinical diagnosis and treatment. METHODS Information from 343 CRC patients was collected and a database was established. Multivariate logistic analysis was used to identify independent factors for colorectal cancer liver metastasis(mCRC) and nomograms were constructed. Receiver operating characteristic curves(ROC), calibration plots, and decision curve analysis (DCA) were used to assess discrimination, agreement with actual risk, and the clinical utility of the prediction model, respectively. RESULT CTC levels in FDV were significantly higher in patients with liver metastasis than in those without liver metastasis. Logistic multivariate analysis showed that vascular invasion, T stage, carcinoembryonic antigen (CEA), CA19-9, and CTC could be used as predictors to construct nomograms. The nomograms showed good discriminatory ability in predicting mCRC, with area under the curve (AUC) values of 0.871 [95 % CI: 0.817-0.924) and 0.891 (95 % CI: 0.817-0.964) for the training and validation sets, respectively.] The calibration curves of both the training and validation sets showed that the model was effective in predicting the probability of mCRC. DCA was used to evaluate this predictive model and showed good net clinical benefit. CONCLUSION We developed and validated a nomogram model based on the combination of CTC in the FDV with other clinical parameters to better predict the occurrence of mCRC.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China
| | - Zhongguo Zhang
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China.
| | - Xue Bi
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, PR China.
| |
Collapse
|
3
|
Wang J, Jiang X, Wang Q, Zhao T, Shen H, Liu X, Feng D, Shen R, Wang Y, Yang W, Wei B. Detection and identification of circulating tumor cells in parathyroid tumors and correlation analysis with clinicopathological features. Endocrine 2024; 85:1357-1364. [PMID: 38730070 DOI: 10.1007/s12020-024-03831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024]
Abstract
INTRODUCTION The differential diagnosis of parathyroid carcinoma (PC)/parathyroid adenoma (PA) in parathyroid tumors is critical for their management and prognosis. Circulating tumor cells (CTCs) identification in the peripheral blood of parathyroid tumors remains unknown. In this study, we proposed to investigate the differences of CTCs in PC/PA and the relationship with clinicopathologic features to assess its relevance to PC and value in identifying PC/PA. METHODS AND MATERIALS Peripheral blood was collected from 27 patients with PC and 37 patients with PA treated in our hospital, and the number of chromosome 8 aberrant CTCs was detected by negative magnetic bead sorting fluorescence in situ hybridization (NE-FISH). The differences of CTCs in PC/PA peripheral blood were compared and their diagnostic efficacy was evaluated, and the correlation between CTCs and clinicopathological features of PC was further explored. RESULTS CTCs differed significantly in PC/PA (p = 0.0008) and were up-regulated in PC, with good diagnostic efficacy. CTCs combined with alkaline phosphatase (ALP) assay improved the diagnostic efficacy in identifying PC/PA (AUC = 0.7838, p = 0.0001). The number of CTCs was correlated with tumor dimensions, but not significantly correlated with clinical markers such as calcium and PTH and pathological features such as vascular invasion, lymph node metastasis and distant metastasis. CONCLUSION As a non-invasive liquid biopsy method, CTCs test combined with ALP test can be used as an important reference basis for timely and accurate identification and treatment of PC. It is of great significance to improve the current situation of PC diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Jiacheng Wang
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xingran Jiang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Qian Wang
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Teng Zhao
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hong Shen
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xing Liu
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dalin Feng
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Rongfang Shen
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuting Wang
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wenjing Yang
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Bojun Wei
- Department of Thyroid and Neck Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
4
|
Andresen NK, Røssevold AH, Borgen E, Schirmer CB, Gilje B, Garred Ø, Lømo J, Stensland M, Nordgård O, Falk RS, Mathiesen RR, Russnes HG, Kyte JA, Naume B. Circulating tumor cells in metastatic breast cancer patients treated with immune checkpoint inhibitors - a biomarker analysis of the ALICE and ICON trials. Mol Oncol 2024. [PMID: 38978352 DOI: 10.1002/1878-0261.13675] [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: 02/14/2024] [Revised: 04/16/2024] [Accepted: 05/27/2024] [Indexed: 07/10/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been introduced in breast cancer (BC) treatment and better biomarkers are needed to predict benefit. Circulating tumor cells (CTCs) are prognostic in BC, but knowledge is limited on CTCs in the context of ICI therapy. In this study, serial sampling of CTCs (CellSearch system) was evaluated in 82 patients with metastatic BC enrolled in two randomized trials investigating ICI plus chemotherapy. Programmed death-ligand 1 (PD-L1) expression on CTCs was also measured. Patients with ≥ 2 CTCs per 7.5 mL at baseline had gene expression profiles in tumor suggestive of increased T-cell activity, including increased tumor inflammation signature (TIS) in both triple-negative (P = 0.010) and hormone receptor-positive (P = 0.024) disease. Patients with luminal A BC had higher CTC levels. The association between CTC status and outcome was most apparent 4 weeks into therapy. PD-L1 expression in CTCs was observed in 6/17 CTC-positive patients and was associated with inferior survival. In conclusion, our study indicates that CTC numbers may inform on tumor immune composition, as well as prognosis. These findings suggest a potential of using CTCs as an accessible biomarker source in BC patients treated with immunotherapy.
Collapse
Affiliation(s)
- Nikolai Kragøe Andresen
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Norway
| | | | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Norway
| | - Jon Lømo
- Department of Pathology, Oslo University Hospital, Norway
| | - Marius Stensland
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
- Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, Norway
| | - Ragnhild Sørum Falk
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Norway
| | | | - Hege G Russnes
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Pathology, Oslo University Hospital, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Jon Amund Kyte
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Oncology, Oslo University Hospital, Norway
| |
Collapse
|
5
|
Shrestha P, Kao S, Cheung VK, Cooper WA, van Zandwijk N, Rasko JEJ, Yeo D. Circulating tumor cells: advancing personalized therapy in small cell lung cancer patients. Mol Oncol 2024. [PMID: 38956984 DOI: 10.1002/1878-0261.13696] [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: 10/20/2023] [Revised: 03/27/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive cancer with a dismal 5-year survival of < 7%, despite the addition of immunotherapy to first-line chemotherapy. Specific tumor biomarkers, such as delta-like ligand 3 (DLL3) and schlafen11 (SLFN11), may enable the selection of more efficacious, novel immunomodulating targeted treatments like bispecific T-cell engaging monoclonal antibodies (tarlatamab) and chemotherapy with PARP inhibitors. However, obtaining a tissue biopsy sample can be challenging in SCLC. Circulating tumor cells (CTCs) have the potential to provide molecular insights into a patient's cancer through a "simple" blood test. CTCs have been studied for their prognostic ability in SCLC; however, their value in guiding treatment decisions is yet to be elucidated. This review explores novel and promising targeted therapies in SCLC, summarizes current knowledge of CTCs in SCLC, and discusses how CTCs can be utilized for precision medicine.
Collapse
Affiliation(s)
- Prajwol Shrestha
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Steven Kao
- Faculty of Medicine and Health, University of Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Veronica K Cheung
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Wendy A Cooper
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
- School of Medicine, University of Western Sydney, Australia
| | - Nico van Zandwijk
- Faculty of Medicine and Health, University of Sydney, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Concord Repatriation General Hospital, Sydney Local Health District, Concord, Australia
| | - John E J Rasko
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Dannel Yeo
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| |
Collapse
|
6
|
Gao S, Li X, Hu Z, Wang Z, Hao X. Dual targeting negative enrichment strategy for highly sensitive and purity detection of CTCs. Front Chem 2024; 12:1400988. [PMID: 38831912 PMCID: PMC11144890 DOI: 10.3389/fchem.2024.1400988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 06/05/2024] Open
Abstract
Circulating tumor cells (CTCs) have significant clinical value in early tumor detection, dynamic monitoring and immunotherapy. CTC detection stands out as a leading non-invasive approach for tumor diagnostics and therapeutics. However, the high heterogeneity of CTCs and the occurrence of epithelial-mesenchymal transition (EMT) during metastasis pose challenges to methods relying on EpCAM-positive enrichment. To address these limitations, a method based on negative enrichment of CTCs using specific leukocyte targets has been developed. In this study, aiming to overcome the low purity associated with immunomagnetic beads targeting solely the leukocyte common antigen CD45, we introduced CD66b-modified immunomagnetic beads. CD66b, a specific target for neutrophils with abundant residues, was chosen as a complementary approach. The process involved initial collection of nucleated cells from whole blood samples using density gradient centrifugation. Subsequently, magnetically labeled leukocytes were removed by magnetic field, enabling the capture of CTCs with higher sensitivity and purity while retaining their activity. Finally, we selected 20 clinical blood samples from patients with various cancers to validate the effectiveness of this strategy, providing a new generalized tool for the clinical detection of CTCs.
Collapse
Affiliation(s)
- Siying Gao
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Xuejie Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Zhiyuan Hu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- School of Nanoscience and Technology, SinoDanish College, University of Chinese Academy of Sciences, Beijing, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaopeng Hao
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Abusamra SM, Barber R, Sharafeldin M, Edwards CM, Davis JJ. The integrated on-chip isolation and detection of circulating tumour cells. SENSORS & DIAGNOSTICS 2024; 3:562-584. [PMID: 38646187 PMCID: PMC11025039 DOI: 10.1039/d3sd00302g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/12/2024] [Indexed: 04/23/2024]
Abstract
Circulating tumour cells (CTCs) are cancer cells shed from a primary tumour which intravasate into the blood stream and have the potential to extravasate into distant tissues, seeding metastatic lesions. As such, they can offer important insight into cancer progression with their presence generally associated with a poor prognosis. The detection and enumeration of CTCs is, therefore, critical to guiding clinical decisions during treatment and providing information on disease state. CTC isolation has been investigated using a plethora of methodologies, of which immunomagnetic capture and microfluidic size-based filtration are the most impactful to date. However, the isolation and detection of CTCs from whole blood comes with many technical barriers, such as those presented by the phenotypic heterogeneity of cell surface markers, with morphological similarity to healthy blood cells, and their low relative abundance (∼1 CTC/1 billion blood cells). At present, the majority of reported methods dissociate CTC isolation from detection, a workflow which undoubtedly contributes to loss from an already sparse population. This review focuses on developments wherein isolation and detection have been integrated into a single-step, microfluidic configuration, reducing CTC loss, increasing throughput, and enabling an on-chip CTC analysis with minimal operator intervention. Particular attention is given to immune-affinity, microfluidic CTC isolation, coupled to optical, physical, and electrochemical CTC detection (quantitative or otherwise).
Collapse
Affiliation(s)
- Sophia M Abusamra
- Nuffield Department of Surgical Sciences, University of Oxford Oxford OX3 9DU UK
| | - Robert Barber
- Department of Chemistry, University of Oxford Oxford OX1 3QZ UK
| | | | - Claire M Edwards
- Nuffield Department of Surgical Sciences, University of Oxford Oxford OX3 9DU UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Systems, University of Oxford Oxford UK
| | - Jason J Davis
- Department of Chemistry, University of Oxford Oxford OX1 3QZ UK
| |
Collapse
|
8
|
Kim YJ, Min J. Hydrogel-based technologies in liquid biopsy for the detection of circulating clinical markers: challenges and prospects. Anal Bioanal Chem 2024; 416:2065-2078. [PMID: 37963993 DOI: 10.1007/s00216-023-05025-7] [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: 07/27/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023]
Abstract
Liquid biopsy, which promises noninvasive detection of tumor-derived material, has recently been highlighted because of its potential to lead us to an era of precision medicine. However, its development has encountered challenges owing to the extremely low frequency and low purity of circulating tumor markers, such as circulating tumor cells (CTCs), circulating exosomes, and circulating tumor nucleic acids (ctNAs). Much effort has been made to overcome this limitation over the last decade, and an increasing number of studies have shown interest in the special characteristics of hydrogels. This hydrophilic and biocompatible polymeric network, which absorbs a large amount of water, can aid in the isolation, protection, and analysis of these low-abundance and short-lived circulating biomarkers. The role of hydrogels in liquid biopsy is extensive and ranges from enrichment to encapsulation. This review provides an overview of hydrogel-based technologies to pave the way in liquid biopsy.
Collapse
Affiliation(s)
- Young Jun Kim
- School of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
9
|
Liu S, Chen S, Xiao L, Zhang K, Qi Y, Li H, Cheng Y, Hu Z, Lin C. Unraveling the motion and deformation characteristics of red blood cells in a deterministic lateral displacement device. Comput Biol Med 2024; 168:107712. [PMID: 38006825 DOI: 10.1016/j.compbiomed.2023.107712] [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: 07/13/2023] [Revised: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 11/27/2023]
Abstract
Deterministic Lateral Displacement (DLD) device has gained widespread recognition and trusted for filtering blood cells. However, there remains a crucial need to explore the complex interplay between deformable cells and flow within the DLD device to improve its design. This paper presents an approach utilizing a mesoscopic cell-level numerical model based on dissipative particle dynamics to effectively capture this complex phenomenon. To establish the model's credibility, a series of numerical simulations were conducted and the numerical results were validated with nominal experimental data from the literature. These include single cell stretching experiment, comparisons of the morphological characteristics of cells in DLD, and comparison the specific row-shift fraction of DLD required to initiate the zigzag mode. Additionally, we investigate the effect of cell rigidity, which serves as an indicator of cell health, on average flow velocity, trajectory, and asphericity. Moreover, we extend the existing theory of predicting zigzag mode for solid spherical particles to encompass the behavior of red blood cells. To achieve this, we introduce a new concept of effective diameter and demonstrate its applicability in providing highly accurate predictions across a wide range of conditions.
Collapse
Affiliation(s)
- Shuai Liu
- School of Aerospace Engineering and Applied Mechanics, Tongji University, Shanghai, 200092, China
| | - Shuo Chen
- School of Aerospace Engineering and Applied Mechanics, Tongji University, Shanghai, 200092, China.
| | - Lanlan Xiao
- School of Mechanical and Automotive Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Kaixuan Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuan Qi
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China
| | - Hao Li
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China
| | - Yuan Cheng
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China
| | - Zixin Hu
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China; Fudan Zhangjiang Institute, Shanghai, 201203, China; Shanghai Pudong Hospital, Shanghai, 201399, China
| | - Chensen Lin
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China; Fudan Zhangjiang Institute, Shanghai, 201203, China; Shanghai Pudong Hospital, Shanghai, 201399, China.
| |
Collapse
|
10
|
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: 6.0] [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
|
11
|
Einloth KR, Gayfield S, McMaster T, Didier A, Dworkin L, Creeden JF. The application, safety, and future of ex vivo immune cell therapies and prognosis in different malignancies. BIOIMPACTS : BI 2023; 13:439-455. [PMID: 38022382 PMCID: PMC10676524 DOI: 10.34172/bi.2023.27521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 12/01/2023]
Abstract
Introduction Immunotherapy has revolutionized how cancer is treated. Many of these immunotherapies rely on ex vivo expansion of immune cells, classically T cells. Still, several immunological obstacles remain, including tumor impermeability by immune cells and the immunosuppressive nature of the tumor microenvironment (TME). Logistically, high costs of treatment and variable clinical responses have also plagued traditional T cell-based immunotherapies. Methods To review the existing literature on cellular immunotherapy, the PubMed database was searched for publications using variations of the phrases "cancer immunotherapy", "ex vivo expansion", and "adoptive cell therapy". The Clinicaltrials.gov database was searched for clinical trials related to ex vivo cellular therapies using the same phrases. The National Comprehensive Cancer Network guidelines for cancer treatment were also referenced. Results To circumvent the challenges of traditional T cell-based immunotherapies, researchers have developed newer therapies including tumor infiltrating lymphocyte (TIL), chimeric antigen receptor (CAR), T cell receptor (TCR) modified T cell, and antibody-armed T cell therapies. Additionally, newer immunotherapeutic strategies have used other immune cells, including natural killer (NK) and dendritic cells (DC), to modulate the T cell immune response to cancers. From a prognostic perspective, circulating tumor cells (CTC) have been used to predict cancer morbidity and mortality. Conclusion This review highlights the mechanism and clinical utility of various types of ex vivo cellular therapies in the treatment of cancer. Comparing these therapies or using them in combination may lead to more individualized and less toxic chemotherapeutics.
Collapse
Affiliation(s)
- Katelyn R. Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Scott Gayfield
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Thomas McMaster
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Alexander Didier
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
12
|
Martel A, Mograbi B, Romeo B, Gastaud L, Lalvee S, Zahaf K, Fayada J, Nahon-Esteve S, Bonnetaud C, Salah M, Tanga V, Baillif S, Bertolotto C, Lassalle S, Hofman P. Assessment of Different Circulating Tumor Cell Platforms for Uveal Melanoma: Potential Impact for Future Routine Clinical Practice. Int J Mol Sci 2023; 24:11075. [PMID: 37446253 DOI: 10.3390/ijms241311075] [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: 06/02/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Liquid biopsy and circulating tumor cell (CTC) screening has gained interest over the last two decades for detecting almost all solid malignancies. To date, the major limitation in terms of the applicability of CTC screening in daily clinical practice is the lack of reproducibility due to the high number of platforms available that use various technologies (e.g., label-dependent versus label-free detection). Only a few studies have compared different CTC platforms. The aim of this study was to compare the efficiency of four commercially available CTC platforms (Vortex (VTX-1), ClearCell FX, ISET, and Cellsearch) for the detection and identification of uveal melanoma cells (OMM 2.3 cell line). Tumor cells were seeded in RPMI medium and venous blood from healthy donors, and then processed similarly using these four platforms. Melan-A immunochemistry was performed to identify tumor cells, except when the Cellsearch device was used (automated identification). The mean overall recovery rates (with mean recovered cells) were 39.2% (19.92), 22.2% (11.31), 8.9% (4.85), and 1.1% (0.20) for the ISET, Vortex (VTX-1), ClearCell FX, and CellSearch platforms, respectively. Although paramount, the recovery rate is not sufficient to assess a CTC platform. Other parameters, such as the purpose for using a platform (diagnosis, genetics, drug sensitivity, or patient-derived xenograft models), reproducibility, purity, user-friendliness, cost-effectiveness, and ergonomics, should also be considered before they can be used in daily clinical practice and are discussed in this article.
Collapse
Affiliation(s)
- Arnaud Martel
- Ophthalmology Department, University Hospital of Nice, Cote d'Azur University, 06 000 Nice, France
- Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, Cote d'Azur University, 06 000 Nice, France
| | - Baharia Mograbi
- Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, Cote d'Azur University, 06 000 Nice, France
| | - Barnabe Romeo
- Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, Cote d'Azur University, 06 000 Nice, France
| | - Lauris Gastaud
- Oncology Department, Antoine Lacassagne Cancer Center, 06 000 Nice, France
| | - Salome Lalvee
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Julien Fayada
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Sacha Nahon-Esteve
- Ophthalmology Department, University Hospital of Nice, Cote d'Azur University, 06 000 Nice, France
- Inserm, Biology and Pathologies of Melanocytes, Team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, 06 100 Nice, France
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Myriam Salah
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Virginie Tanga
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Stéphanie Baillif
- Ophthalmology Department, University Hospital of Nice, Cote d'Azur University, 06 000 Nice, France
| | - Corine Bertolotto
- Inserm, Biology and Pathologies of Melanocytes, Team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, 06 100 Nice, France
| | - Sandra Lassalle
- Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, Cote d'Azur University, 06 000 Nice, France
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| | - Paul Hofman
- Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, Cote d'Azur University, 06 000 Nice, France
- Laboratory of Clinical and Experimental Pathology, University Hospital of Nice, FHU OncoAge, Cote d'Azur University, Biobank BB-0033-00025, 06 000 Nice, France
| |
Collapse
|
13
|
Massimino M, Martorana F, Stella S, Vitale SR, Tomarchio C, Manzella L, Vigneri P. Single-Cell Analysis in the Omics Era: Technologies and Applications in Cancer. Genes (Basel) 2023; 14:1330. [PMID: 37510235 PMCID: PMC10380065 DOI: 10.3390/genes14071330] [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: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer molecular profiling obtained with conventional bulk sequencing describes average alterations obtained from the entire cellular population analyzed. In the era of precision medicine, this approach is unable to track tumor heterogeneity and cannot be exploited to unravel the biological processes behind clonal evolution. In the last few years, functional single-cell omics has improved our understanding of cancer heterogeneity. This approach requires isolation and identification of single cells starting from an entire population. A cell suspension obtained by tumor tissue dissociation or hematological material can be manipulated using different techniques to separate individual cells, employed for single-cell downstream analysis. Single-cell data can then be used to analyze cell-cell diversity, thus mapping evolving cancer biological processes. Despite its unquestionable advantages, single-cell analysis produces massive amounts of data with several potential biases, stemming from cell manipulation and pre-amplification steps. To overcome these limitations, several bioinformatic approaches have been developed and explored. In this work, we provide an overview of this entire process while discussing the most recent advances in the field of functional omics at single-cell resolution.
Collapse
Affiliation(s)
- Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Cristina Tomarchio
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-S. Marco", 95123 Catania, Italy
- Humanitas Istituto Clinico Catanese, University Oncology Department, 95045 Catania, Italy
| |
Collapse
|
14
|
Strati A, Economopoulou P, Lianidou E, Psyrri A. Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy. Biomedicines 2023; 11:1768. [PMID: 37371863 DOI: 10.3390/biomedicines11061768] [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: 05/31/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The approval of monoclonal antibodies against programmed death-ligand 1 (PD-L1) and programmed cell death protein (PD1) has changed the landscape of cancer treatment. To date, many immune checkpoint inhibitors (ICIs) have been approved by the FDA for the treatment of metastatic cancer as well as locally recurrent advanced cancer. However, immune-related adverse events (irAEs) of ICIs highlight the need for biomarker analysis with strong predictive value. Liquid biopsy is an important tool for clinical oncologists to monitor cancer patients and administer or change appropriate therapy. CTCs frequently express PD-L1, and this constitutes a clinically useful and non-invasive method to assess PD-L1 status in real-time. This review summarizes all the latest findings about the clinical significance of CTC for the management of cancer patients during the administration of immunotherapy and mainly focuses on the assessment of PD-L1 expression in CTCs.
Collapse
Affiliation(s)
- Areti Strati
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Panagiota Economopoulou
- Department of Internal Medicine, Section of Medical Oncology, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Amanda Psyrri
- Department of Internal Medicine, Section of Medical Oncology, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| |
Collapse
|
15
|
Kurniali PC, Storandt MH, Jin Z. Utilization of Circulating Tumor Cells in the Management of Solid Tumors. J Pers Med 2023; 13:jpm13040694. [PMID: 37109080 PMCID: PMC10145886 DOI: 10.3390/jpm13040694] [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: 03/16/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells shed from the primary tumor into circulation, with clusters of CTCs responsible for cancer metastases. CTC detection and isolation from the bloodstream are based on properties distinguishing CTCs from normal blood cells. Current CTC detection techniques can be divided into two main categories: label dependent, which depends upon antibodies that selectively bind cell surface antigens present on CTCs, or label-independent detection, which is detection based on the size, deformability, and biophysical properties of CTCs. CTCs may play significant roles in cancer screening, diagnosis, treatment navigation, including prognostication and precision medicine, and surveillance. In cancer screening, capturing and evaluating CTCs from peripheral blood could be a strategy to detect cancer at its earliest stage. Cancer diagnosis using liquid biopsy could also have tremendous benefits. Full utilization of CTCs in the clinical management of malignancies may be feasible in the near future; however, several challenges still exist. CTC assays currently lack adequate sensitivity, especially in early-stage solid malignancies, due to low numbers of detectable CTCs. As assays improve and more trials evaluate the clinical utility of CTC detection in guiding therapies, we anticipate increased use in cancer management.
Collapse
Affiliation(s)
- Peter C Kurniali
- Sanford Cancer Center, 701 E Rosser Ave, Bismarck, ND 58501, USA
- Department of Internal Medicine, Division of Hematology/Oncology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | | | | |
Collapse
|
16
|
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: 18] [Impact Index Per Article: 18.0] [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
|
17
|
Zhou Q, Liu X, Li J, Tong B, Xu Y, Chen M, Liu X, Gao X, Shi Y, Zhao J, Zhong W, Wang M. Circulating tumor cells PD-L1 expression detection and correlation of therapeutic efficacy of immune checkpoint inhibition in advanced non-small-cell lung cancer. Thorac Cancer 2023; 14:470-478. [PMID: 36630992 PMCID: PMC9925337 DOI: 10.1111/1759-7714.14767] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION This study investigated whether programmed death-ligand 1 (PD-L1) expression of circulating tumor cells (CTCs) in peripheral blood can serve as a predictive biomarker for immunotherapy efficacy in patients with advanced non-small-cell lung cancer (NSCLC). METHODS We employed a negative enrichment method to isolate CTCs. We identified PD-L1 + CTCs as PD-L1+/4',6-diamidino-2-phenylindole (DAPI)+/CD45-circulating tumor cells through an immunofluorescence method. Tumor tissue PD-L1 expression was determined by immunohistochemical staining. The correlation between CTC PD-L1 expression and patients' prognostic features was estimated through the Kaplan-Meier method. RESULTS CTCs released a higher detection rate of PD-L1 expression than tumor tissues (53.0% vs. 42.1%). No correlation was observed between them. Forty-nine NSCLC patients received anti-PD-1/PD-L1 immunotherapy (three with combined anti-PD-1/PD-L1 and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), two with four cycles of combined immune checkpoint inhibitors [ICIs] plus chemotherapy and ICI monotherapy for maintenance). Patients with PD-L1 expression on tissue or CTCs had a median progression-free survival (mPFS) of 5.6 months (n = 36, 95% confidence interval [CI] 3.6-7.5 months), significantly longer than those without PD-L1 detection (n = 9, mPFS of 1.4 months, 95% CI 1.3-1.5 months, log-rank p = 0.032). The multivariable Cox proportional-hazard model suggested that the tissue or CTC PD-L1 expression was associated with a lower risk of progression (hazard ratio 0.45, 95% CI 0.21-0.98, p = 0.043). CONCLUSIONS CTCs and tumor tissues reveal heterogeneous expression of PD-L1 in NSCLC patients. Patients with baseline PD-L1 expression on CTCs or tissue showed prolonged mPFS and may help to identify the subsets of patients who potentially benefit from immunotherapy.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Xiangning Liu
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Ji Li
- Department of PathologyPeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Bing Tong
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Yan Xu
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Minjiang Chen
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Xiaoyan Liu
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Xiaoxing Gao
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Yuequan Shi
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Jing Zhao
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Wei Zhong
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Mengzhao Wang
- Department of Respiratory MedicinePeking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
18
|
Hasanzadeh Kafshgari M, Hayden O. Advances in analytical microfluidic workflows for differential cancer diagnosis. NANO SELECT 2023. [DOI: 10.1002/nano.202200158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Morteza Hasanzadeh Kafshgari
- Heinz‐Nixdorf‐Chair of Biomedical Electronics Campus Klinikum München rechts der Isar TranslaTUM Technical University of Munich Munich Germany
| | - Oliver Hayden
- Heinz‐Nixdorf‐Chair of Biomedical Electronics Campus Klinikum München rechts der Isar TranslaTUM Technical University of Munich Munich Germany
| |
Collapse
|
19
|
Noubissi Nzeteu GA, Geismann C, Arlt A, Hoogwater FJH, Nijkamp MW, Meyer NH, Bockhorn M. Role of Epithelial-to-Mesenchymal Transition for the Generation of Circulating Tumors Cells and Cancer Cell Dissemination. Cancers (Basel) 2022; 14:5483. [PMID: 36428576 PMCID: PMC9688619 DOI: 10.3390/cancers14225483] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Tumor-related death is primarily caused by metastasis; consequently, understanding, preventing, and treating metastasis is essential to improving clinical outcomes. Metastasis is mainly governed by the dissemination of tumor cells in the systemic circulation: so-called circulating tumor cells (CTCs). CTCs typically arise from epithelial tumor cells that undergo epithelial-to-mesenchymal transition (EMT), resulting in the loss of cell-cell adhesions and polarity, and the reorganization of the cytoskeleton. Various oncogenic factors can induce EMT, among them the transforming growth factor (TGF)-β, as well as Wnt and Notch signaling pathways. This entails the activation of numerous transcription factors, including ZEB, TWIST, and Snail proteins, acting as transcriptional repressors of epithelial markers, such as E-cadherin and inducers of mesenchymal markers such as vimentin. These genetic and phenotypic changes ultimately facilitate cancer cell migration. However, to successfully form distant metastases, CTCs must primarily withstand the hostile environment of circulation. This includes adaption to shear stress, avoiding being trapped by coagulation and surviving attacks of the immune system. Several applications of CTCs, from cancer diagnosis and screening to monitoring and even guided therapy, seek their way into clinical practice. This review describes the process leading to tumor metastasis, from the generation of CTCs in primary tumors to their dissemination into distant organs, as well as the importance of subtyping CTCs to improve personalized and targeted cancer therapy.
Collapse
Affiliation(s)
- Gaetan Aime Noubissi Nzeteu
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, 26129 Oldenburg, Germany
| | - Claudia Geismann
- Laboratory of Molecular Gastroenterology & Hepatology, Department of Internal Medicine I, UKSH-Campus Kiel, 24118 Kiel, Germany
| | - Alexander Arlt
- Department for Gastroenterology and Hepatology, University Hospital Oldenburg, Klinikum Oldenburg AöR, European Medical School (EMS), 26133 Oldenburg, Germany
| | - Frederik J. H. Hoogwater
- Section of HPB Surgery & Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Maarten W. Nijkamp
- Section of HPB Surgery & Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - N. Helge Meyer
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, 26129 Oldenburg, Germany
| | - Maximilian Bockhorn
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, 26129 Oldenburg, Germany
| |
Collapse
|
20
|
Petrik J, Verbanac D, Fabijanec M, Hulina-Tomašković A, Čeri A, Somborac-Bačura A, Petlevski R, Grdić Rajković M, Rumora L, Krušlin B, Štefanović M, Ljubičić N, Baršić N, Hanžek A, Bočkor L, Ćelap I, Demirović A, Barišić K. Circulating Tumor Cells in Colorectal Cancer: Detection Systems and Clinical Utility. Int J Mol Sci 2022; 23:13582. [PMID: 36362369 PMCID: PMC9654465 DOI: 10.3390/ijms232113582] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The high mortality from CRC is mainly related to metastasis affecting distant organs and their function. Dissemination of tumor cells from the primary tumor and hematogeneous spread are considered crucial in the formation of tumor metastases. The analysis of circulating tumor cells (CTCs) and CTC clusters in the blood can be used for the early detection of invasive cancer. Moreover, CTCs have a prognostic significance in the monitoring of a malignant disease or the response to chemotherapy. This work presents an overview of the research conducted on CTCs with the aim of finding suitable detection systems and assessing the possibility of clinical applications in patients with CRC.
Collapse
Affiliation(s)
- József Petrik
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Donatella Verbanac
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Marija Fabijanec
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Andrea Hulina-Tomašković
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Andrea Čeri
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Anita Somborac-Bačura
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Roberta Petlevski
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Marija Grdić Rajković
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Lada Rumora
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Božo Krušlin
- School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
- Department of Pathology and Cytology “Ljudevit Jurak”, University Hospital Centre “Sestre milosrdnice”, University of Zagreb, Vinogradska 29, 10000 Zagreb, Croatia
| | - Mario Štefanović
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
- Department of Clinical Chemistry, University Hospital Centre “Sestre milosrdnice”, University of Zagreb, Vinogradska 29, 10000 Zagreb, Croatia
| | - Neven Ljubičić
- School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
- Department of Internal Medicine, University Hospital Centre “Sestre milosrdnice”, Division of Gastroenterology and Hepatology, University of Zagreb, Vinogradska 29, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, Gundulićeva 5, 10000 Zagreb, Croatia
| | - Neven Baršić
- School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
- Department of Internal Medicine, University Hospital Centre “Sestre milosrdnice”, Division of Gastroenterology and Hepatology, University of Zagreb, Vinogradska 29, 10000 Zagreb, Croatia
| | - Antonija Hanžek
- UPR CHROME, University of Nimes, 7 Place Gabriel Peri, 30000 Nîmes, France
| | - Luka Bočkor
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Ljudevita Gaja 32, 10000 Zagreb, Croatia
| | - Ivana Ćelap
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Alma Demirović
- Department of Pathology and Cytology “Ljudevit Jurak”, University Hospital Centre “Sestre milosrdnice”, University of Zagreb, Vinogradska 29, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, Gundulićeva 5, 10000 Zagreb, Croatia
| | - Karmela Barišić
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| |
Collapse
|
21
|
Yao Y, Zhu X, Liu W, Jiang J, Jiang H. Meta-analysis of the prognostic value of circulating tumor cells in gastrointestinal cancer. Medicine (Baltimore) 2022; 101:e31099. [PMID: 36281182 PMCID: PMC9592416 DOI: 10.1097/md.0000000000031099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Detecting circulating tumor cells (CTCs) has become a new strategy for predicting the prognosis of cancer patients. However, limited systematic research evidence is available for the detection of CTCs in various gastrointestinal tumors such as esophageal cancer (EC), colorectal cancer (CRC) and gastric cancer (GC). This topic was addressed to assess the prognostic significance of CTCs in gastrointestinal tumors. METHODS We conducted a literature search according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist (from November 20, 2021). We performed a meta-analysis using the random effects model and Review Manager 5.3 software (The Cochrane Collaboration, Copenhagen, Denmark) according to the inclusion and exclusion criteria, data extraction and evaluation methods. RESULTS Twenty-four articles met the inclusion criteria for this study, and they included 3803 EC, CRC and GC patients, including 1189 CTC-positive and 2462 CTC-negative cases. The meta-analysis showed that the presence of CTCs was associated with worse OS (HR = 2.05, 95% CI = 1.75-2.40, P = .060) and PFS (HR = 2.27, 95% CI = 1.79-2.89, P < .001). Further meta-regression and subgroup analyses showed that CTC-positive patients also showed worse OS and PFS in different subgroups. CONCLUSION Our meta-analysis suggests that detecting CTCs in peripheral blood may be an important tool for improving the prognosis of patients with gastrointestinal tumors. Moreover, CTCs detection results could be used to develop personalized treatment plans in the future.
Collapse
Affiliation(s)
- Yuming Yao
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Xiang Zhu
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Weixin Liu
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Jiayi Jiang
- Mathematics Major, New York University, New York, NY, USA
| | - Han Jiang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- * Correspondence: Han Jiang, Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu, Nanchang 330006, Jiangxi, China (e-mail: )
| |
Collapse
|
22
|
Rodríguez‐Pena A, Armendariz E, Oyarbide A, Morales X, Ortiz‐Espinosa S, Ruiz‐Fernández de Córdoba B, Cochonneau D, Cornago I, Heymann D, Argemi J, D'Avola D, Sangro B, Lecanda F, Pio R, Cortés‐Domínguez I, Ortiz‐de‐Solórzano C. Design and validation of a tunable inertial microfluidic system for the efficient enrichment of circulating tumor cells in blood. Bioeng Transl Med 2022; 7:e10331. [PMID: 36176621 PMCID: PMC9472016 DOI: 10.1002/btm2.10331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 11/08/2022] Open
Abstract
The analysis of circulating tumor cells (CTCs) in blood is a powerful noninvasive alternative to conventional tumor biopsy. Inertial-based separation is a promising high-throughput, marker-free sorting strategy for the enrichment and isolation of CTCs. Here, we present and validate a double spiral microfluidic device that efficiently isolates CTCs with a fine-tunable cut-off value of 9 μm and a separation range of 2 μm. We designed the device based on computer simulations that introduce a novel, customized inertial force term, and provide practical fabrication guidelines. We validated the device using calibration beads, which allowed us to refine the simulations and redesign the device. Then we validated the redesigned device using blood samples and a murine model of metastatic breast cancer. Finally, as a proof of principle, we tested the device using peripheral blood from a patient with hepatocellular carcinoma, isolating more than 17 CTCs/ml, with purity/removal values of 96.03% and 99.99% of white blood cell and red blood cells, respectively. These results confirm highly efficient CTC isolation with a stringent cut-off value and better separation results than the state of the art.
Collapse
Affiliation(s)
- Alejandro Rodríguez‐Pena
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | | | - Alvaro Oyarbide
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Xabier Morales
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Sergio Ortiz‐Espinosa
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Department of Biochemistry and Genetics, School of SciencesUniversity of NavarraPamplonaSpain
| | - Borja Ruiz‐Fernández de Córdoba
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Denis Cochonneau
- Institut de Cancérologie de l'Ouest, “Tumor Heterogeneity and Precision Medicine” Lab., Blvd Jacques MonodSaint‐HerblainFrance
| | - Iñaki Cornago
- Automotive and Mechatronics R&D Foundation (Naitec)PamplonaSpain
| | - Dominique Heymann
- Institut de Cancérologie de l'Ouest, “Tumor Heterogeneity and Precision Medicine” Lab., Blvd Jacques MonodSaint‐HerblainFrance
- Nantes Université, CNRS, US2B, UMR 6286NantesFrance
| | - Josepmaría Argemi
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Delia D'Avola
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Bruno Sangro
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Fernando Lecanda
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Department of Pathology, Anatomy and PhysiologyUniversity of NavarraPamplonaSpain
| | - Ruben Pio
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Department of Biochemistry and Genetics, School of SciencesUniversity of NavarraPamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
| | - Iván Cortés‐Domínguez
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Carlos Ortiz‐de‐Solórzano
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
23
|
Chang TK, Tung PC, Lee MJ, Lee W. A liquid-crystal aptasensing platform for label-free detection of a single circulating tumor cell. Biosens Bioelectron 2022; 216:114607. [PMID: 35969962 DOI: 10.1016/j.bios.2022.114607] [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: 05/26/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022]
Abstract
Circulating tumor cells (CTCs), which are shed from a primary site into the bloodstream and lead to distal metastases, are pivotal as a prognostic marker for evaluating the treatment response of cancer patients. One of the major challenges of detecting CTCs is their scarcity in blood. We report herein a label-free liquid crystal (LC) cytosensor by adopting an aptamer against epithelial cell adhesion molecule (EpCAM) to capture EpCAM-positive cancer cells. The optical and dielectric signals transduced from the interaction between LC and different numbers of captured breast cancer cells were investigated. A limit of detection (LOD) of 5 CTCs was resulted from the optical biosensing approach relying on texture observation and image analysis of the optical signal in polarizing micrographs. The LOD was further lowered to a single CTC in the dielectric approach by studying the real- and imaginary-part dielectric constants of LC at 1 kHz and 30 Hz as well as the relaxation frequency. The LC-based EpCAM-specific dielectric cytosensor was successfully applied to single-cell CTC detection in cancer cell-spiked human serum and whole blood. This platform demonstrates the potential of LC-based biosensing technologies in cellular-level detection and quantitation, which is crucial to the early diagnosis of cancer metastasis and progression.
Collapse
Affiliation(s)
- Tsung-Keng Chang
- College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan; National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Pei-Chi Tung
- Department of Bioscience Technology, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan
| | - Mon-Juan Lee
- Department of Bioscience Technology, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan; Department of Medical Science Industries, Chang Jung Christian University, Guiren Dist, Tainan, 711301, Taiwan.
| | - Wei Lee
- College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan; Institute of Imaging and Biomedical Photonics, College of Photonics, National Yang Ming Chiao Tung University, Guiren Dist, Tainan, 711010, Taiwan.
| |
Collapse
|
24
|
Khan T, Becker TM, Po JW, Chua W, Ma Y. Single-Circulating Tumor Cell Whole Genome Amplification to Unravel Cancer Heterogeneity and Actionable Biomarkers. Int J Mol Sci 2022; 23:ijms23158386. [PMID: 35955517 PMCID: PMC9369222 DOI: 10.3390/ijms23158386] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
The field of single-cell analysis has advanced rapidly in the last decade and is providing new insights into the characterization of intercellular genetic heterogeneity and complexity, especially in human cancer. In this regard, analyzing single circulating tumor cells (CTCs) is becoming particularly attractive due to the easy access to CTCs from simple blood samples called “liquid biopsies”. Analysis of multiple single CTCs has the potential to allow the identification and characterization of cancer heterogeneity to guide best therapy and predict therapeutic response. However, single-CTC analysis is restricted by the low amounts of DNA in a single cell genome. Whole genome amplification (WGA) techniques have emerged as a key step, enabling single-cell downstream molecular analysis. Here, we provide an overview of recent advances in WGA and their applications in the genetic analysis of single CTCs, along with prospective views towards clinical applications. First, we focus on the technical challenges of isolating and recovering single CTCs and then explore different WGA methodologies and recent developments which have been utilized to amplify single cell genomes for further downstream analysis. Lastly, we list a portfolio of CTC studies which employ WGA and single-cell analysis for genetic heterogeneity and biomarker detection.
Collapse
Affiliation(s)
- Tanzila Khan
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.K.); (T.M.B.); (W.C.)
- Medical Oncology, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia
- Centre of Circulating Tumor Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Therese M. Becker
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.K.); (T.M.B.); (W.C.)
- Medical Oncology, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia
- Centre of Circulating Tumor Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia;
- South West Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Joseph W. Po
- Centre of Circulating Tumor Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia;
- Surgical Innovations Unit, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.K.); (T.M.B.); (W.C.)
- Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Yafeng Ma
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.K.); (T.M.B.); (W.C.)
- Medical Oncology, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia
- Centre of Circulating Tumor Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia;
- South West Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- Correspondence:
| |
Collapse
|
25
|
Powering single-cell genomics to unravel circulating tumour cell subpopulations in non-small cell lung cancer patients. J Cancer Res Clin Oncol 2022; 149:1941-1950. [PMID: 35896898 PMCID: PMC10097753 DOI: 10.1007/s00432-022-04202-y] [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: 04/05/2022] [Accepted: 07/09/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Circulating tumour cells (CTCs) are attractive "liquid biopsy" candidates that could provide insights into the different phenotypes of tumours present within a patient. The epithelial-to-mesenchymal transition (EMT) of CTCs is considered a critical step in tumour metastasis; however, it may confound traditional epithelial feature-based CTC isolation and detection. We applied single-cell copy number alteration (CNA) analysis for the identification of genomic alterations to confirm the neoplastic nature of circulating cells with only mesenchymal phenotypes. METHODS We isolated CTCs from blood samples collected from 46 NSCLC patients using the Parsortix system. Enriched cells were subjected to immunofluorescent staining for CTC identification using a multi-marker panel comprising both epithelial and mesenchymal markers. A subset of isolated CTCs was subjected to whole genome amplification (WGA) and low-pass whole-genome sequencing (LP-WGS) for the analysis of copy number alterations (CNAs). RESULTS CTCs were detected in 16/46 (34.8%) patients, inclusive of CK+/EpCAM+ CTCs (3/46, 6.5%) and Vim+ CTCs (13/46, 28.3%). Clusters of Vim+ cells were detected in 8 samples, which constitutes 50% of the total number of NSCLC patients with CTCs. No patients had detectable hybrid CK+/EpCAM+/Vim+ cells. All of the tested CK+/EpCAM+ CTCs and 7/8 Vim+ CTCs or CTC clusters carried CNAs confirming their neoplastic nature. Notably, the Vim+ cluster with no CNAs was characterised by spindle morphology and, therefore, defined as normal mesenchymal circulating cells. CONCLUSION Our results revealed that CK-negative, vimentin-expressing cells represent a large proportion of CTCs detected in NSCLC patients, which are likely missed by standard epithelial-marker-dependent CTC categorisation.
Collapse
|
26
|
Chaperonin containing TCP1 as a marker for identification of circulating tumor cells in blood. PLoS One 2022; 17:e0264651. [PMID: 35749519 PMCID: PMC9232171 DOI: 10.1371/journal.pone.0264651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
Herein we report the use of Chaperonin-Containing TCP-1 (CCT or TRiC) as a marker to detect circulating tumor cells (CTCs) that are shed from tumors during oncogenesis. Most detection methods used in liquid biopsy approaches for enumeration of CTCs from blood, employ epithelial markers like cytokeratin (CK). However, such markers provide little information on the potential of these shed tumor cells, which are normally short-lived, to seed metastatic sites. To identify a marker that could go beyond enumeration and provide actionable data on CTCs, we evaluated CCT. CCT is a protein-folding complex composed of eight subunits. Previously, we found that expression of the second subunit (CCT2 or CCTβ) inversely correlated with cancer patient survival and was essential for tumorigenesis in mice, driving tumor-promoting processes like proliferation and anchorage-independent growth. In this study, we examined CCT2 expression in cancer compared to normal tissues and found statistically significant increases in tumors. Because not all blood samples from cancer patients contain detectable CTCs, we used the approach of spiking a known number of cancer cells into blood from healthy donors to test a liquid biopsy approach using CCT2 to distinguish rare cancer cells from the large number of non-cancer cells in blood. Using a clinically validated method for capturing CTCs, we evaluated detection of intracellular CCT2 staining for visualization of breast cancer and small cell lung (SCLC) cancer cells. We demonstrated that CCT2 staining could be incorporated into a CTC capture and staining protocol, providing biologically relevant information to improve detection of cancer cells shed in blood. These results were confirmed with a pilot study of blood from SCLC patients. Our studies demonstrate that detection of CCT2 could identify rare cancer cells in blood and has application in liquid biopsy approaches to enhance the use of minimally invasive methods for cancer diagnosis.
Collapse
|
27
|
Yekula A, Tracz J, Rincon-Torroella J, Azad T, Bettegowda C. Single-Cell RNA Sequencing of Cerebrospinal Fluid as an Advanced Form of Liquid Biopsy for Neurological Disorders. Brain Sci 2022; 12:brainsci12070812. [PMID: 35884620 PMCID: PMC9313114 DOI: 10.3390/brainsci12070812] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/16/2022] Open
Abstract
Diagnosis and longitudinal monitoring of neurological diseases are limited by the poor specificity and limited resolution of currently available techniques. Analysis of circulating cells in cerebrospinal fluid (CSF) has emerged as a promising strategy for the diagnosis, molecular characterization, and monitoring of neurological disease. In comparison to bulk sequencing analysis, single-cell sequencing studies can provide novel insights into rare cell populations and uncover heterogeneity in gene expression at a single-cell resolution, which has several implications for understanding disease pathology and treatment. Parallel development of standardized biofluid collection protocols, pre-processing strategies, reliable single-cell isolation strategies, downstream genomic analysis, and robust computational analysis is paramount for comprehensive single-cell sequencing analysis. Here we perform a comprehensive review of studies focusing on single-cell sequencing of cells in the CSF of patients with oncological or non-oncological diseases of the central nervous system.
Collapse
Affiliation(s)
- Anudeep Yekula
- Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Jovanna Tracz
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (J.T.); (J.R.-T.); (T.A.)
| | - Jordina Rincon-Torroella
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (J.T.); (J.R.-T.); (T.A.)
| | - Tej Azad
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (J.T.); (J.R.-T.); (T.A.)
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (J.T.); (J.R.-T.); (T.A.)
- Correspondence:
| |
Collapse
|
28
|
Hu Y, Chen D, Napoleon JV, Srinivasarao M, Singhal S, Savran CA, Low PS. Efficient capture of circulating tumor cells with low molecular weight folate receptor-specific ligands. Sci Rep 2022; 12:8555. [PMID: 35595733 PMCID: PMC9122947 DOI: 10.1038/s41598-022-12118-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/29/2022] [Indexed: 11/09/2022] Open
Abstract
Retrieval of circulating tumor cells (CTC) has proven valuable for assessing a patient's cancer burden, evaluating response to therapy, and analyzing which drug might treat a cancer best. Although most isolation methods retrieve CTCs based on size, shape, or capture by tumor-specific antibodies, we explore here the use of small molecule tumor-specific ligands linked to magnetic beads for CTC capture. We have designed folic acid-biotin conjugates with different linkers for the capture of folate receptor (FR) + tumor cells spiked into whole blood, and application of the same technology to isolate FR + CTCs from the peripheral blood of both tumor-bearing mice and non-small cell lung patients. We demonstrate that folic acid linked via a rigid linker to a flexible PEG spacer that is in turn tethered to a magnetic bead enables optimal CTC retrieval, reaching nearly 100% capture when 100 cancer cells are spiked into 1 mL of aqueous buffer and ~ 90% capture when the same quantity of cells is diluted into whole blood. In a live animal model, the same methodology is shown to efficiently retrieve CTCs from tumor-bearing mice, yielding cancer cell counts that are proportional to total tumor burden. More importantly, the same method is shown to collect ~ 29 CTCs/8 mL peripheral blood from patients with non-small cell lung cancer. Since the ligand-presentation strategy optimized here should also prove useful in targeting other nanoparticles to other cells, the methods described below should have general applicability in the design of nanoparticles for cell-specific targeting.
Collapse
Affiliation(s)
- Yingwen Hu
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Danyang Chen
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - John V Napoleon
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Madduri Srinivasarao
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cagri A Savran
- School of Mechanical Engineering, Birck Nanotechnology Center, Purdue Center for Cancer Research, Purdue University, 1205 W. State St., West Lafayette, IN, 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
29
|
Dathathri E, Isebia KT, Abali F, Lolkema MP, Martens JWM, Terstappen LWMM, Bansal R. Liquid Biopsy Based Circulating Biomarkers in Metastatic Prostate Cancer. Front Oncol 2022; 12:863472. [PMID: 35669415 PMCID: PMC9165750 DOI: 10.3389/fonc.2022.863472] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the most dominant male malignancy worldwide. The clinical presentation of prostate cancer ranges from localized indolent to rapidly progressing lethal metastatic disease. Despite a decline in death rate over the past years, with the advent of early diagnosis and new treatment options, challenges remain towards the management of metastatic prostate cancer, particularly metastatic castration sensitive prostate cancer (mCSPC) and castration resistant prostate cancer (mCRPC). Current treatments involve a combination of chemotherapy with androgen deprivation therapy and/or androgen receptor signalling inhibitors. However, treatment outcomes are heterogeneous due to significant tumor heterogeneity indicating a need for better prognostic biomarkers to identify patients with poor outcomes. Liquid biopsy has opened a plethora of opportunities from early diagnosis to (personalized) therapeutic disease interventions. In this review, we first provide recent insights about (metastatic) prostate cancer and its current treatment landscape. We highlight recent studies involving various circulating biomarkers such as circulating tumor cells, genetic markers, circulating nucleic acids, extracellular vesicles, tumor-educated platelets, and the secretome from (circulating) tumor cells and tumor microenvironment in metastatic prostate cancer. The comprehensive array of biomarkers can provide a powerful approach to understanding the spectrum of prostate cancer disease and guide in developing improved and personalized treatments for patients.
Collapse
Affiliation(s)
- Eshwari Dathathri
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Khrystany T. Isebia
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - Fikri Abali
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Martijn P. Lolkema
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - John W. M. Martens
- Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Department of Medical Oncology, Rotterdam, Netherlands
| | - Leon W. M. M. Terstappen
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Ruchi Bansal
- Department of Medical Cell BioPhysics, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| |
Collapse
|
30
|
Liquid Biopsy and Dielectrophoretic Analysis—Complementary Methods in Skin Cancer Monitoring. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12073366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The incidence and prevalence of skin cancers is currently increasing worldwide, with early detection, adequate treatment, and prevention of recurrences being topics of great interest for researchers nowadays. Although tumor biopsy remains the gold standard of diagnosis, this technique cannot be performed in a significant proportion of cases, so that the use of alternative methods with high sensitivity and specificity is becoming increasingly desirable. In this context, liquid biopsy appears to be a feasible solution for the study of cellular and molecular markers relevant to different types of skin cancers. Circulating tumor cells are just one of the components of interest obtained from performing liquid biopsy, and their study by complementary methods, such as dielectrophoresis, could bring additional benefits in terms of characterizing skin tumors and subsequently applying personalized therapy. One purpose of this review is to demonstrate the utility of liquid biopsy primarily in monitoring the most common types of skin tumors: basal cell carcinoma, squamous cell carcinoma, and malign melanoma. In addition, the originality of the article is based on the detailed presentation of the dielectrophoretic analysis method of the most important elements obtained from liquid biopsy, with direct impact on the clinical and therapeutic approach of skin tumors.
Collapse
|
31
|
Ginghina O, Hudita A, Zamfir M, Spanu A, Mardare M, Bondoc I, Buburuzan L, Georgescu SE, Costache M, Negrei C, Nitipir C, Galateanu B. Liquid Biopsy and Artificial Intelligence as Tools to Detect Signatures of Colorectal Malignancies: A Modern Approach in Patient's Stratification. Front Oncol 2022; 12:856575. [PMID: 35356214 PMCID: PMC8959149 DOI: 10.3389/fonc.2022.856575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/16/2022] [Indexed: 01/19/2023] Open
Abstract
Colorectal cancer (CRC) is the second most frequently diagnosed type of cancer and a major worldwide public health concern. Despite the global efforts in the development of modern therapeutic strategies, CRC prognosis is strongly correlated with the stage of the disease at diagnosis. Early detection of CRC has a huge impact in decreasing mortality while pre-lesion detection significantly reduces the incidence of the pathology. Even though the management of CRC patients is based on robust diagnostic methods such as serum tumor markers analysis, colonoscopy, histopathological analysis of tumor tissue, and imaging methods (computer tomography or magnetic resonance), these strategies still have many limitations and do not fully satisfy clinical needs due to their lack of sensitivity and/or specificity. Therefore, improvements of the current practice would substantially impact the management of CRC patients. In this view, liquid biopsy is a promising approach that could help clinicians screen for disease, stratify patients to the best treatment, and monitor treatment response and resistance mechanisms in the tumor in a regular and minimally invasive manner. Liquid biopsies allow the detection and analysis of different tumor-derived circulating markers such as cell-free nucleic acids (cfNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs) in the bloodstream. The major advantage of this approach is its ability to trace and monitor the molecular profile of the patient's tumor and to predict personalized treatment in real-time. On the other hand, the prospective use of artificial intelligence (AI) in medicine holds great promise in oncology, for the diagnosis, treatment, and prognosis prediction of disease. AI has two main branches in the medical field: (i) a virtual branch that includes medical imaging, clinical assisted diagnosis, and treatment, as well as drug research, and (ii) a physical branch that includes surgical robots. This review summarizes findings relevant to liquid biopsy and AI in CRC for better management and stratification of CRC patients.
Collapse
Affiliation(s)
- Octav Ginghina
- Department II, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Marius Zamfir
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Andrada Spanu
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Mara Mardare
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | - Irina Bondoc
- Department of Surgery, “Sf. Ioan” Clinical Emergency Hospital, Bucharest, Romania
| | | | - Sergiu Emil Georgescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Carolina Negrei
- Department of Toxicology, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
| | - Cornelia Nitipir
- Department II, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, Bucharest, Romania
| | - Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
32
|
Deger T, Mendelaar PAJ, Kraan J, Prager-van der Smissen WJC, van der Vlugt-Daane M, Bindels EMJ, Sieuwerts AM, Sleijfer S, Wilting SM, Hollestelle A, Martens JWM. A pipeline for copy number profiling of single circulating tumor cells to assess intra-patient tumor heterogeneity. Mol Oncol 2021; 16:2981-3000. [PMID: 34964258 PMCID: PMC9394233 DOI: 10.1002/1878-0261.13174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/01/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022] Open
Abstract
Intrapatient tumour heterogeneity is likely a major determinant of clinical outcome in cancer patients. To assess heterogeneity in a minimally invasive manner, methods to perform single circulating tumour cell (CTC) genomics at high resolution are necessary. However, due to the rarity of CTCs, development of such methods is challenging. Here, we developed a modular single CTC analysis pipeline to assess intrapatient heterogeneity by copy number (CN) profiling. To optimize this pipeline, spike‐in experiments using MCF‐7 breast cancer cells were performed. The VyCAP puncher system was used to isolate single cells. The quality of whole genome amplification (WGA) products generated by REPLI‐g and Ampli1™ methods, as well as the results from the Illumina Truseq and the Ampli1™ LowPass library preparation techniques, was compared. Moreover, a bioinformatic pipeline was designed to generate CN profiles from single CTCs. The optimal combination of Ampli1™ WGA and Illumina Truseq library preparation was successfully validated on patient‐derived CTCs. In conclusion, we developed a novel modular pipeline to isolate single CTCs and subsequently generate detailed patient‐derived CN profiles that allow assessment of intrapatient heterogeneity in future studies.
Collapse
Affiliation(s)
- Teoman Deger
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Pauline A J Mendelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | | | - Eric M J Bindels
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
33
|
Fu X, Tao L, Zhang X. A chimeric virus-based probe unambiguously detects live circulating tumor cells with high specificity and sensitivity. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:78-86. [PMID: 34631928 PMCID: PMC8476710 DOI: 10.1016/j.omtm.2021.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
The current methods for detecting circulating tumor cells (CTCs) suffer from several drawbacks. We report a novel method that is based on a chimeric virus probe and can detect CTCs with extremely high specificity and sensitivity. Moreover, it exclusively detects live CTCs, and its detection efficacy is not impacted by the variation of epithelial cell adhesion molecule (EpCAM) expression. The chimeric virus probe is composed of a capsid from human papillomavirus that provides the detection with high specificity and an SV40-based genome that can amplify extensively inside CTCs and, hence, endows the detection with high sensitivity. Furthermore, different marker genes can be incorporated into the probe to provide detection with versatility. These unique capabilities will likely improve the validity and utility of this CTC detection in several clinical applications, which is one of the drawbacks suffered by many of the current CTC detection methods.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry and Center for Nuclear Receptor and Cell Signaling, University of Houston, Houston, TX 77204, USA
| | - Lihua Tao
- Department of Biology and Biochemistry and Center for Nuclear Receptor and Cell Signaling, University of Houston, Houston, TX 77204, USA
| | - Xiaoliu Zhang
- Department of Biology and Biochemistry and Center for Nuclear Receptor and Cell Signaling, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
34
|
Ostrikov K, Kashani MN, Vasilev K, MacGregor MN. Fluid Flow Dependency in Immunoselective Cell Capture via Liquid Biopsy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:12388-12396. [PMID: 34596407 DOI: 10.1021/acs.langmuir.1c01998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liquid biopsy targets rare cells that overexpress disease-specific membrane markers and capture these cells via immunoaffinity. The diagnosis efficiency of liquid biopsy can be impaired by the presence of healthy adherent cells also expressing the same biomarkers. Here, we investigated the effect of settling times and rinsing flow rates on the efficiency of EpCAM-based immunocapture using both simulation and experiments with three different cell types. Cell-surface adhesion forces and shear rates were calculated to define the range of rinsing flow rates to test experimentally. Healthy adherent cells did not adhere to blocked immunofunctionalized surfaces within the timeframe of the experiment; however, healthy EpCAM positive cells did bind to the surface to some extent. The greatest difference in capture efficiency was obtained using a high rinsing flow rate of 25 mL/min following 40 min static incubation, indicating that optimizing rinsing flow rates could be a viable option to capture, more specifically, cancer cells overexpressing EpCAM.
Collapse
Affiliation(s)
- Kola Ostrikov
- UniSA STEM, University of South Australia, Mawson Lakes 5095, Australia
| | - Moein Navvab Kashani
- UniSA STEM, University of South Australia, Mawson Lakes 5095, Australia
- South Australian Node of the Australian National Fabrication Facility, Mawson Lakes 5095, Australia
| | - Krasimir Vasilev
- UniSA STEM, University of South Australia, Mawson Lakes 5095, Australia
| | | |
Collapse
|
35
|
Ozimski LL, Gremmelspacher D, Aceto N. A fatal affair: Circulating tumor cell relationships that shape metastasis. iScience 2021; 24:103073. [PMID: 34568794 PMCID: PMC8449241 DOI: 10.1016/j.isci.2021.103073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells are metastatic precursors in several cancer types. Their biology and clinical utility are subject to numerous investigations, yet one aspect that is often neglected is their entanglement with the tumor microenvironment, namely the cross talk with stromal and immune cells and their relationships with other tumor-derived components such as circulating tumor DNA and extracellular vesicles in circulation. We will focus our short review specifically on these aspects, i.e., providing some examples of the liaison that circulating tumor cells have with stromal or immune cells and illustrating their relationship with other circulating tumor derivatives such as circulating tumor DNA and extracellular vesicles.
Collapse
Affiliation(s)
- Lauren L. Ozimski
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - David Gremmelspacher
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
36
|
Hyler AR, Hong D, Davalos RV, Swami NS, Schmelz EM. A novel ultralow conductivity electromanipulation buffer improves cell viability and enhances dielectrophoretic consistency. Electrophoresis 2021; 42:1366-1377. [PMID: 33687759 DOI: 10.1002/elps.202000324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/23/2021] [Accepted: 02/27/2021] [Indexed: 12/21/2022]
Abstract
Cell separation has become a critical diagnostic, research, and treatment tool for personalized medicine. Despite significant advances in cell separation, most widely used applications require the use of multiple, expensive antibodies to known markers in order to identify subpopulations of cells for separation. Dielectrophoresis (DEP) provides a biophysical separation technique that can target cell subpopulations based on phenotype without labels and return native cells for downstream analysis. One challenge in employing any DEP device is the sample being separated must be transferred into an ultralow conductivity medium, which can be detrimental in retaining cells' native phenotypes for separation. Here, we measured properties of traditional DEP reagents and determined that after just 1-2 h of exposure and subsequent culture, cells' viability was significantly reduced below 50%. We developed and tested a novel buffer (Cyto Buffer) that achieved 6 weeks of stable shelf-life and demonstrated significantly improved viability and physiological properties. We then determined the impact of Cyto Buffer on cells' dielectric properties and morphology and found that cells retained properties more similar to that of their native media. Finally, we vetted Cyto Buffer's usability on a cell separation platform (Cyto R1) to determine combined efficacy for cell separations. Here, more than 80% of cells from different cell lines were recovered and were determined to be >70% viable following exposure to Cyto Buffer, flow stimulation, electromanipulation, and downstream collection and growth. The developed buffer demonstrated improved opportunities for electrical cell manipulation, enrichment, and recovery for next generation cell separations.
Collapse
Affiliation(s)
| | - Daly Hong
- CytoRecovery, Inc., Blacksburg, VA, USA
| | - Rafael V Davalos
- Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Nathan S Swami
- Electrical and Computer Engineering, University of Virginia, Charlottesville, VA, USA
| | - Eva M Schmelz
- Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.,Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
37
|
Enhancing Prediction Performance by Add-On Combining Circulating Tumor Cell Count, CD45 neg EpCAM neg Cell Count on Colorectal Cancer, Advance, and Metastasis. Cancers (Basel) 2021; 13:cancers13112521. [PMID: 34063929 PMCID: PMC8196640 DOI: 10.3390/cancers13112521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Information describing circulating tumor cells (CTCs) holds promise for clinical applications. However, conventional CTCs enumeration could ignore the CTCs more relevant to cancer metastasis. Thus, negative selection CTC enumeration was proposed, by which information on the numbers of CTCs and CD45neg EpCAMneg cells can be obtained. By combining this approach with the conventional biomarker carcinoembryonic antigen (CEA), this study aimed to explore whether any combination of these biomarkers could improve the predictive performance for colorectal cancer (CRC) or its status. Results revealed that a combination of the two cell populations showed improved performance (AUROC: 0.893) for CRC prediction over the use of only one population. Compared with CEA alone, the combination of the three biomarkers increased the performance (AUROC) for advanced CRC prediction from 0.643 to 0.727. Compared with that of CEA alone for metastatic CRC prediction, the AUROC was increased from 0.780 to 0.837 when the CTC count was included. Abstract Conventional circulating tumor cell (CTC) enumeration could ignore the CTCs more relevant to cancer metastasis. Thus, negative selection CTC enumeration was proposed, by which information on two cellular biomarkers (numbers of CTCs and CD45neg EpCAMneg cells) can be obtained. By combining this approach with the conventional biomarker carcinoembryonic antigen (CEA), this study aimed to explore whether any combination of these biomarkers could improve the predictive performance for colorectal cancer (CRC) or its status. In this work, these two cell populations in healthy donors and CRC patients were quantified. Results revealed that enumeration of these two cell populations was able to discriminate healthy donors from CRC patients, even patients with non-advanced CRC. Moreover, the combination of the two cell populations showed improved performance (AUROC: 0.893) for CRC prediction over the use of only one population. Compared with CEA alone, the combination of the three biomarkers increased the performance (AUROC) for advanced CRC prediction from 0.643 to 0.727. Compared with that of CEA alone for metastatic CRC prediction, the AUROC was increased from 0.780 to 0.837 when the CTC count was included. Overall, this study demonstrated that the combination of these two cellular biomarkers with CEA improved the predictive performance for CRC and its status.
Collapse
|
38
|
Zhou X, Pu Q, Yu H, Peng Y, Li J, Yang Y, Chen H, Weng Y, Xie G. An electrochemical biosensor based on hemin/G-quadruplex DNAzyme and PdRu/Pt heterostructures as signal amplifier for circulating tumor cells detection. J Colloid Interface Sci 2021; 599:752-761. [PMID: 33989928 DOI: 10.1016/j.jcis.2021.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/21/2022]
Abstract
Metastasis due to circulating tumor cells (CTCs) shed from the original tumor accounts for the majority of cancer-related death. Efficient CTCs detection is pivotal to the diagnosis of early cancer metastasis. In this work, Platinum nanoparticles (PtNPs) decorated hyperbranched PdRu nanospines (PdRu/Pt) hierarchical structures were firstly synthesized to detect CTCs with the assistance of DNAzyme. Meanwhile, Super P and gold nanoparticles (AuNPs) acted as sensing medium to improve electrical conductivity and immobilization of anti-EpCAM antibody to specifically capture model CTCs. After immune-conjugation of anti-EpCAM-MCF-7-signal probes on the gold electrode, PtNPs, PdRu nanospines (PdRuNSs) and hemin/G-quadruplex co-catalyzed substrate H2O2 to realize multiplexed signal amplification, which significantly improves the analytical performance of the electrochemical biosensor. As-proposed biosensor reached a limit of detection (LOD) down to 2 cells mL-1 and showed a wide detection range of 2 to 106 cells mL-1. Application of the biosensor to detect MCF-7 cells spiked human blood samples further demonstrated the feasibility for early cancer evaluation in clinic.
Collapse
Affiliation(s)
- Xi Zhou
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China
| | - Qinli Pu
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China
| | - Hongyan Yu
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China
| | - Yang Peng
- Clinical Laboratory Medicine Center, Chongqing University Cancer Hospital, Chongqing 400030, PR China
| | - Junjie Li
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China
| | - Yujun Yang
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China
| | - Huajian Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China; Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yaguang Weng
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China.
| | - Guoming Xie
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), College of Laboratory Medicine, Chongqing Medical Laboratory Microfluidics and SPRi Engineering Research Center, Chongqing Medical University, No. 1 Yi Xue Yuan Road, Chongqing 400016, PR China.
| |
Collapse
|
39
|
Foy V, Lindsay CR, Carmel A, Fernandez-Gutierrez F, Krebs MG, Priest L, Carter M, Groen HJM, Hiltermann TJN, de Luca A, Farace F, Besse B, Terstappen L, Rossi E, Morabito A, Perrone F, Renehan A, Faivre-Finn C, Normanno N, Dive C, Blackhall F, Michiels S. EPAC-lung: European pooled analysis of the prognostic value of circulating tumour cells in small cell lung cancer. Transl Lung Cancer Res 2021; 10:1653-1665. [PMID: 34012782 PMCID: PMC8107738 DOI: 10.21037/tlcr-20-1061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/17/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circulating tumour cell (CTC) number is an independent prognostic factor in patients with small cell lung cancer (SCLC) but there is no consensus on the CTC threshold for prognostic significance. We undertook a pooled analysis of individual patient data to clinically validate CTC enumeration and threshold for prognostication. METHODS Four European cancer centres, experienced in CellSearch CTC enumeration for SCLC provided pseudo anonymised data for patients who had undergone pre-treatment CTC count. Data was collated, and Cox regression models, stratified by centre, explored the relationship between CTC count and survival. The added value of incorporating CTCs into clinico-pathological models was investigated using likelihood ratio tests. RESULTS A total of 367 patient records were evaluated. A one-unit increase in log-transformed CTC counts corresponded to an estimated hazard ratio (HR) of 1.24 (95% CI: 1.19-1.29, P<0.0001) for progression free survival (PFS) and 1.23 (95% CI: 1.18-1.28, P<0.0001) for overall survival (OS). CTC count of ≥15 or ≥50 was significantly associated with an increased risk of progression (CTC ≥15: HR 3.20, 95% CI: 2.50-4.09, P<0.001; CTC ≥50: HR 2.56, 95% CI: 2.01-3.25, P<0.001) and an increased risk of death (CTC ≥15: HR 2.90, 95% CI: 2.28-3.70, P<0.001; CTC ≥50: HR 2.47, 95% CI: 1.95-3.13, P<0.001). There was no significant inter-centre heterogeneity observed. Addition of CTC count to clinico-pathological models as a continuous log-transformed variable, offers further prognostic value (both likelihood ratio P<0.001 for OS and PFS). CONCLUSIONS Higher pre-treatment CTC counts are a negative independent prognostic factor in SCLC when considered as a continuous variable or dichotomised counts of ≥15 or ≥50. Incorporating CTC counts, as a continuous variable, improves clinic-pathological prognostic models.
Collapse
Affiliation(s)
- Victoria Foy
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Colin R Lindsay
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Alexandra Carmel
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM U1018 OncoStat, CESP, Université Paris-Sud, Université Paris-Saclay, labeled by Ligue Contre le Cancer, France
| | - Fabiola Fernandez-Gutierrez
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Matthew G Krebs
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Lynsey Priest
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Mathew Carter
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Harry J M Groen
- Department of Pulmonary Diseases, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - T Jeroen N Hiltermann
- Department of Pulmonary Diseases, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Antonella de Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francoise Farace
- INSERM, U981 "Predictive Biomarkers and New Therapeutics in Oncology", F-94805, Villejuif, France
- Gustave Roussy, Université Paris-Saclay. "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23, AMMICA, Villejuif, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France; Paris-Sud University, Orsay, France
| | - Leon Terstappen
- Department of Medical Cell BioPhysics, University of Twente, Enschede, The Netherlands
| | - Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Andrew Renehan
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
| | - Corinne Faivre-Finn
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Stefan Michiels
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM U1018 OncoStat, CESP, Université Paris-Sud, Université Paris-Saclay, labeled by Ligue Contre le Cancer, France
| |
Collapse
|
40
|
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers (Basel) 2021; 13:cancers13050970. [PMID: 33652649 PMCID: PMC7956528 DOI: 10.3390/cancers13050970] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Circulating tumour cells (CTCs) are cancer cells shed into the bloodstream from tumours and their analysis can provide important insights into cancer detection and monitoring, with the potential to direct personalised therapies for the patient. These CTCs are rare in the blood, which makes their detection and enrichment challenging and to date, only one technology (the CellSearch) has gained FDA approval for determining the prognosis of patients with advanced breast, prostate and colorectal cancers. Here, we review the wide range of enrichment technologies available to isolate CTCs from other blood components and highlight the important characteristics that new technologies should possess for routine clinical use. Abstract Circulating tumour cells (CTCs) are the precursor cells for the formation of metastatic disease. With a simple blood draw, liquid biopsies enable the non-invasive sampling of CTCs from the blood, which have the potential to provide important insights into cancer detection and monitoring. Since gaining FDA approval in 2004, the CellSearch system has been used to determine the prognosis of patients with metastatic breast, prostate and colorectal cancers. This utilises the cell surface marker Epithelial Cell Adhesion Molecule (EpCAM), to enrich CTCs, and many other technologies have adopted this approach. More recently, the role of mesenchymal-like CTCs in metastasis formation has come to light. It has been suggested that these cells are more aggressive metastatic precursors than their epithelial counterparts; however, mesenchymal CTCs remain undetected by EpCAM-based enrichment methods. This has prompted the development of a variety of ‘label free’ enrichment technologies, which exploit the unique physical properties of CTCs (such as size and deformability) compared to other blood components. Here, we review a wide range of both immunocapture and label free CTC enrichment technologies, summarising the most significant advantages and disadvantages of each. We also highlight the important characteristics that technologies should possess for routine clinical use, since future developments could have important clinical implications, with the potential to direct personalised therapies for patients with cancer.
Collapse
Affiliation(s)
- Amelia J. Rushton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
- Correspondence:
| | - Georgios Nteliopoulos
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| | - Jacqueline A. Shaw
- Leicester Cancer Research Centre, University of Leicester, Leicester LE2 7LX, UK;
| | - R. Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| |
Collapse
|
41
|
Neves RPL, Ammerlaan W, Andree KC, Bender S, Cayrefourcq L, Driemel C, Koch C, Luetke-Eversloh MV, Oulhen M, Rossi E, Alix-Panabières C, Betsou F, Farace F, Riethdorf S, Schlange T, Wikman H, Zamarchi R, Pantel K, Terstappen LWMM, Stoecklein NH. Proficiency Testing to Assess Technical Performance for CTC-Processing and Detection Methods in CANCER-ID. Clin Chem 2021; 67:631-641. [PMID: 33491069 DOI: 10.1093/clinchem/hvaa322] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/12/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Multiple technologies are available for detection of circulating tumor cells (CTCs), but standards to evaluate their technical performance are still lacking. This limits the applicability of CTC analysis in clinic routine. Therefore, in the context of the CANCER-ID consortium, we established a platform to assess technical validity of CTC detection methods in a European multi-center setting using non-small cell lung cancer (NSCLC) as a model. METHODS We characterized multiple NSCLC cell lines to define cellular models distinct in their phenotype and molecular characteristics. Standardized tumor-cell-bearing blood samples were prepared at a central laboratory and sent to multiple European laboratories for processing according to standard operating procedures. The data were submitted via an online tool and centrally evaluated. Five CTC-enrichment technologies were tested. RESULTS We could identify 2 cytokeratin expressing cell lines with distinct levels of EpCAM expression: NCI-H441 (EpCAMhigh, CKpos) and NCI-H1563 (EpCAMlow, CKpos). Both spiked tumor cell lines were detected by all technologies except for the CellSearch system that failed to enrich EpCAMlow NCI-H1563 cells. Mean recovery rates ranged between 49% and 75% for NCI-H411 and 32% and 76% for NCI-H1563 and significant differences were observed between the tested methods. CONCLUSIONS This multi-national proficiency testing of CTC-enrichment technologies has importance in the establishment of guidelines for clinically applicable (pre)analytical workflows and the definition of minimal performance qualification requirements prior to clinical validation of technologies. It will remain in operation beyond the funding period of CANCER-ID in the context of the European Liquid Biopsy Society (ELBS).
Collapse
Affiliation(s)
- Rui P L Neves
- Department of General, Visceral and Paediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Wim Ammerlaan
- Integrated BioBank of Luxembourg, Dudelange, Luxembourg
| | - Kiki C Andree
- Department of Medical Cell BioPhysics, University of Twente, Enschede, The Netherlands
| | | | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, Montpellier, France
| | - Christiane Driemel
- Department of General, Visceral and Paediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Claudia Koch
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Marianne Oulhen
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor Cells" Translational Platform, CNRS UMS3655-INSERM US23 AMMICA, Villejuif, France
| | - Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy.,Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, Montpellier, France
| | - Fay Betsou
- Integrated BioBank of Luxembourg, Dudelange, Luxembourg
| | - Françoise Farace
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor Cells" Translational Platform, CNRS UMS3655-INSERM US23 AMMICA, Villejuif, France
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Harriet Wikman
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rita Zamarchi
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon W M M Terstappen
- Department of Medical Cell BioPhysics, University of Twente, Enschede, The Netherlands
| | - Nikolas H Stoecklein
- Department of General, Visceral and Paediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
42
|
Mondelo-Macía P, García-González J, León-Mateos L, Castillo-García A, López-López R, Muinelo-Romay L, Díaz-Peña R. Current Status and Future Perspectives of Liquid Biopsy in Small Cell Lung Cancer. Biomedicines 2021; 9:48. [PMID: 33430290 PMCID: PMC7825645 DOI: 10.3390/biomedicines9010048] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 01/08/2023] Open
Abstract
Approximately 19% of all cancer-related deaths are due to lung cancer, which is the leading cause of mortality worldwide. Small cell lung cancer (SCLC) affects approximately 15% of patients diagnosed with lung cancer. SCLC is characterized by aggressiveness; the majority of SCLC patients present with metastatic disease, and less than 5% of patients are alive at 5 years. The gold standard of SCLC treatment is platinum and etoposide-based chemotherapy; however, its effects are short. In recent years, treatment for SCLC has changed; new drugs have been approved, and new biomarkers are needed for treatment selection. Liquid biopsy is a non-invasive, rapid, repeated and alternative tool to the traditional tumor biopsy that could allow the most personalized medicine into the management of SCLC patients. Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) are the most commonly used liquid biopsy biomarkers. Some studies have reported the prognostic factors of CTCs and cfDNA in SCLC patients, independent of the stage. In this review, we summarize the recent SCLC studies of CTCs, cfDNA and other liquid biopsy biomarkers, and we discuss the future utility of liquid biopsy in the clinical management of SCLC.
Collapse
Affiliation(s)
- Patricia Mondelo-Macía
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
| | - Jorge García-González
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Luis León-Mateos
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | | | - Rafael López-López
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (J.G.-G.); (L.L.-M.); (R.L.-L.)
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Roberto Díaz-Peña
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (P.M.-M.); (L.M.-R.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
43
|
Theil G, Fornara P, Bialek J. Position of Circulating Tumor Cells in the Clinical Routine in Prostate Cancer and Breast Cancer Patients. Cancers (Basel) 2020; 12:cancers12123782. [PMID: 33333999 PMCID: PMC7765455 DOI: 10.3390/cancers12123782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Many different therapies are applied to fight tumor disease. Blood-based biosources, like circulating tumor cells (CTCs), offer the opportunity to monitor the healing progression and the real-time response to the therapy. In this review, we analyze the outcomes of the clinical trials and scientific studies of prostate and breast cancer performed in the decade between April 2010 and April 2020. Additionally, we describe the abstracts from the 4th Advances in Circulating Tumor Cells (ACTC) meeting in 2019. We discuss the potential therapeutic opportunities related to the CTCs and the challenges ahead in the routine treatment of cancer. Abstract Prostate cancer and breast cancer are the most common cancers worldwide. Anti-tumor therapies are long and exhaustive for the patients. The real-time monitoring of the healing progression could be a useful tool to evaluate therapeutic response. Blood-based biosources like circulating tumor cells (CTCs) may offer this opportunity. Application of CTCs for the clinical diagnostics could improve the sequenced screening, provide additional valuable information of tumor dynamics, and help personalized management for the patients. In the past decade, CTCs as liquid biopsy (LB) has received tremendous attention. Many different isolation and characterization platforms are developed but the clinical validation is still missing. In this review, we focus on the clinical trials of circulating tumor cells that have the potential to monitor and stratify patients and lead to implementation into clinical practice.
Collapse
|
44
|
Liu D, Yin J, Liang S, Shi W, Jiang X, Gao Y. Enzyme-Regulated Peptide-Liquid Metal Hybrid Hydrogels as Cell Amber for Single-Cell Manipulation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:45807-45813. [PMID: 32951417 DOI: 10.1021/acsami.0c13334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Current strategies to construct cell-based bioartificial tissues largely remain on a multicell level. Taking cell diversity into account, single-cell manipulation is urgently needed for delicate bioartificial tissue construction. Current single-cell isolation and profiling techniques involve invasive processes and thus are not applicable for single-cell manipulation. Here, we managed to fabricate peptide-liquid metal hybrid hydrogels as "cell ambers" which were suitable for single-cell isolation as well as further handling. The successful preparation of uniform liquid metal nanoparticles allowed the fabrication of peptide-liquid metal hydrogel with excellent recovery property upon mechanical destruction. The alkaline phosphatase-instructed supramolecular self-assembly process allowed the formation of microhydrogel post-filling in the PDMS template. The co-culture of the hydrogel precursor and mammalian cells realized the embedding of cells into elastic hydrogels which were the so-called cell ambers. The cell ambers turned out to be biocompatible and capable of supporting cell survival. Aided with the micro-operating system and a laser scanning confocal microscope, we could arrange these as-prepared 3D single-cell ambers into various patterns as desired. Our strategy provided the possibility to manipulate a single cell, which served as a prototype of cell architecture toward cell-based bioartificial tissue construction.
Collapse
Affiliation(s)
- Dongdong Liu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxiang Yin
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Sen Liang
- The Technical Institute of Physics and Chemistry of the Chinese Academy of Sciences, Beijing 100190, China
| | - Wensheng Shi
- The Technical Institute of Physics and Chemistry of the Chinese Academy of Sciences, Beijing 100190, China
| | - Xingyu Jiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- Department of Biomedical Engineering, Southern University of Science & Technology, Shenzhen, Guangdong 518055, China
| | - Yuan Gao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
45
|
Lee LX, Li SC. Hunting down the dominating subclone of cancer stem cells as a potential new therapeutic target in multiple myeloma: An artificial intelligence perspective. World J Stem Cells 2020; 12:706-720. [PMID: 32952853 PMCID: PMC7477658 DOI: 10.4252/wjsc.v12.i8.706] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/08/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
The development of single-cell subclones, which can rapidly switch from dormant to dominant subclones, occur in the natural pathophysiology of multiple myeloma (MM) but is often "pressed" by the standard treatment of MM. These emerging subclones present a challenge, providing reservoirs for chemoresistant mutations. Technological advancement is required to track MM subclonal changes, as understanding MM's mechanism of evolution at the cellular level can prompt the development of new targeted ways of treating this disease. Current methods to study the evolution of subclones in MM rely on technologies capable of phenotypically and genotypically characterizing plasma cells, which include immunohistochemistry, flow cytometry, or cytogenetics. Still, all of these technologies may be limited by the sensitivity for picking up rare events. In contrast, more incisive methods such as RNA sequencing, comparative genomic hybridization, or whole-genome sequencing are not yet commonly used in clinical practice. Here we introduce the epidemiological diagnosis and prognosis of MM and review current methods for evaluating MM subclone evolution, such as minimal residual disease/multiparametric flow cytometry/next-generation sequencing, and their respective advantages and disadvantages. In addition, we propose our new single-cell method of evaluation to understand MM's mechanism of evolution at the molecular and cellular level and to prompt the development of new targeted ways of treating this disease, which has a broad prospect.
Collapse
Affiliation(s)
- Lisa X Lee
- Division of Hematology/Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, UCI Health, Orange, CA 92868, United States
| | - Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory, CHOC Children's Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
- Department of Neurology, University of California-Irvine School of Medicine, Orange, CA 92868, United States
| |
Collapse
|
46
|
Chu PY, Hsieh CH, Wu MH. The Combination of Immunomagnetic Bead-Based Cell Isolation and Optically Induced Dielectrophoresis (ODEP)-Based Microfluidic Device for the Negative Selection-Based Isolation of Circulating Tumor Cells (CTCs). Front Bioeng Biotechnol 2020; 8:921. [PMID: 32903713 PMCID: PMC7438881 DOI: 10.3389/fbioe.2020.00921] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/17/2020] [Indexed: 01/19/2023] Open
Abstract
Negative selection-based circulating tumor cell (CTC) isolation is able to harvest viable, label-free, and clinically meaningful CTCs from the cancer patients' blood. Nevertheless, its main shortcoming is its inability to isolate high-purity CTCs, restricting subsequent CTC-related analysis. To address this issue, this study proposed a two-step optically-induced dielectrophoresis (ODEP) cell manipulation to process the cell sample harvested by negative selection-/immunomagnetic microbeads-based CTC isolation. The working mechanism is that the ODEP force acting on the cells with and without magnetic microbeads binding is different. Accordingly, the use of ODEP cell manipulation in a microfluidic system was designed to first separate and then isolate the cancer cells from other magnetic microbead-bound cells. Immunofluorescent microscopic observation and ODEP cell manipulation were then performed to refine the purity of the cancer cells. In this study, the optimum operating conditions for effective cell isolation were determined experimentally. The results revealed that the presented method was able to further refine the purity of cancer cell in the sample obtained after negative selection-based CTC isolation with high cell purity (81.6~86.1%). Overall, this study proposed the combination of immunomagnetic bead-based cell isolation and ODEP cell manipulation for the negative selection-based isolation of CTCs.
Collapse
Affiliation(s)
- Po-Yu Chu
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Hsun Hsieh
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital (Linkou), Taoyuan City, Taiwan
- Division of Haematology-Oncology, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Min-Hsien Wu
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital (Linkou), Taoyuan City, Taiwan
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
47
|
Habli Z, AlChamaa W, Saab R, Kadara H, Khraiche ML. Circulating Tumor Cell Detection Technologies and Clinical Utility: Challenges and Opportunities. Cancers (Basel) 2020; 12:cancers12071930. [PMID: 32708837 PMCID: PMC7409125 DOI: 10.3390/cancers12071930] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/03/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
The potential clinical utility of circulating tumor cells (CTCs) in the diagnosis and management of cancer has drawn a lot of attention in the past 10 years. CTCs disseminate from tumors into the bloodstream and are believed to carry vital information about tumor onset, progression, and metastasis. In addition, CTCs reflect different biological aspects of the primary tumor they originate from, mainly in their genetic and protein expression. Moreover, emerging evidence indicates that CTC liquid biopsies can be extended beyond prognostication to pharmacodynamic and predictive biomarkers in cancer patient management. A key challenge in harnessing the clinical potential and utility of CTCs is enumerating and isolating these rare heterogeneous cells from a blood sample while allowing downstream CTC analysis. That being said, there have been serious doubts regarding the potential value of CTCs as clinical biomarkers for cancer due to the low number of promising outcomes in the published results. This review aims to present an overview of the current preclinical CTC detection technologies and the advantages and limitations of each sensing platform, while surveying and analyzing the published evidence of the clinical utility of CTCs.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Walid AlChamaa
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Raya Saab
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, 77030 TX, USA;
| | - Massoud L. Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
- Correspondence:
| |
Collapse
|
48
|
Li Y, Wu G, Yang W, Wang X, Duan L, Niu L, Zhang Y, Liu J, Hong L, Fan D. Prognostic value of circulating tumor cells detected with the CellSearch system in esophageal cancer patients: a systematic review and meta-analysis. BMC Cancer 2020; 20:581. [PMID: 32571299 PMCID: PMC7310134 DOI: 10.1186/s12885-020-07059-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/10/2020] [Indexed: 02/08/2023] Open
Abstract
Background Esophageal carcinoma (EC) is the seventh-most prevalent tumor in the world, which is still one of the primary causes of tumor-related death. Identifying noteworthy biomarkers for EC is particularly significant in guiding effective treatment. Recently, circulating tumor cells (CTCs) in peripheral blood (PB) were intensively discussed as prognostic markers in patients with EC. However, an ongoing controversy still exists regarding the prognostic significance of CTCs determined by the CellSearch system in EC sufferers. This meta-analysis was designed to approach this topic. Methods We systematically conducted searches using PubMed, Medline, Web of Science and the Cochrane Library for relevant studies, which were published through February 20, 2020. Using the random-effects model, our study was performed in Review Manager software, with odds ratios (ORs), risk ratios (RRs), hazard ratios (HRs) and 95% confidence intervals (CIs) as the effect values. Results Totally 7 articles were finally included in this study. For clinicopathological characteristics, the pooled results on TNM stage indicated that the III/IV group had higher rate of CTCs compared with the I/II group (OR = 1.36, 95% CI: 0.68–2.71, I2 = 0%). Incidence of CTCs was higher in patients with T3/T4 stage (OR = 2.92, 95% CI: 1.31–6.51, I2 = 0%) and distant metastasis group (OR = 5.18, 95% CI: 2.38–11.25, I2 = 0%) compared to patients with T1/T2 stage or non-metastatic group. The pooled analysis revealed that CTC positivity detected in EC patients was correlated with poor overall survival (OS) (HR = 2.83, 95% CI:1.99–4.03, I2 = 0%) and relapse-free survival (RFS) (HR = 4.71, 95% CI:2.73–8.13, I2 = 0%). When pooling the estimated RR, a poor therapeutic response to chemoradiotherapy was discovered in patients with CTC positivity (RR = 1.99, 95% CI:1.73–2.29, I2 = 60%). Conclusions In summary, our meta-analysis demonstrated that CTCs positivity determined by the CellSearch system are correlated with the prognosis of EC patients and might indicate a poor therapeutic response to chemotherapy in EC patients.
Collapse
Affiliation(s)
- Yiding Li
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Guiling Wu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wanli Yang
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Xiaoqian Wang
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Lili Duan
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Liaoran Niu
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Yujie Zhang
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Jinqiang Liu
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Liu Hong
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China.
| | - Daiming Fan
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi Province, 710032, PR China
| |
Collapse
|
49
|
Wu J, Raba K, Guglielmi R, Behrens B, Van Dalum G, Flügen G, Koch A, Patel S, Knoefel WT, Stoecklein NH, Neves RPL. Magnetic-Based Enrichment of Rare Cells from High Concentrated Blood Samples. Cancers (Basel) 2020; 12:E933. [PMID: 32290064 PMCID: PMC7225976 DOI: 10.3390/cancers12040933] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/30/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Here, we tested two magnetic-bead based systems for the enrichment and detection of rare tumor cells in concentrated blood products. For that, the defined numbers of cells from three pancreatic cancer cell lines were spiked in 108 peripheral blood mononuclear cells (PBMNCs) concentrated in 1 mL, mimicking diagnostic leukapheresis (DLA) samples, and samples were processed for circulating tumor cells (CTC) enrichment with the IsoFlux or the KingFisher systems, using different types of magnetic beads from the respective technology providers. Beads were conjugated with different anti-EpCAM and MUC-1 antibodies. Recovered cells were enumerated and documented by fluorescent microscopy. For the IsoFlux system, best performance was obtained with IsoFlux CTC enrichment kit, but these beads compromised the subsequent immunofluorescence staining. For the KingFisher system, best recoveries were obtained using Dynabeads Biotin Binder beads. These beads also allowed one to capture CTCs with different antibodies and the subsequent immunofluorescence staining. KingFisher instrument allowed a single and streamlined protocol for the enrichment and staining of CTCs that further prevented cell loss at the enrichment/staining interface. Both IsoFlux and KingFisher systems allowed the enrichment of cell line cells from the mimicked-DLA samples. However, in this particular experimental setting, the recovery rates obtained with the KingFisher system were globally higher, the system was more cost-effective, and it allowed higher throughput.
Collapse
Affiliation(s)
- Junhao Wu
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Katharina Raba
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Rosa Guglielmi
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Bianca Behrens
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Guus Van Dalum
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Georg Flügen
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Andreas Koch
- Thermo Fisher Scientific, Postfach 200152, Frankfurter Str. 129B, 64293 Darmstadt, Germany;
| | - Suraj Patel
- Thermo Fisher Scientific, 3 Fountain Drive, Inchinnan, Renfrew PA4 9RF, UK;
| | - Wolfram T. Knoefel
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Nikolas H. Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| | - Rui P. L. Neves
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; (J.W.); (R.G.); (B.B.); (G.V.D.); (G.F.); (W.T.K.)
| |
Collapse
|
50
|
Aghamir SMK, Heshmat R, Ebrahimi M, Khatami F. Liquid Biopsy: The Unique Test for Chasing the Genetics of Solid Tumors. Epigenet Insights 2020; 13:2516865720904052. [PMID: 32166219 PMCID: PMC7050026 DOI: 10.1177/2516865720904052] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
Blood test is a kind of liquid biopsy that checks cancer cells or cancer nucleic acids circulating freely from cells in the blood. A liquid biopsy may be used to distinguish cancer at early stages and it could be a game-changer for both cancer diagnosis and prognosis strategies. Liquid biopsy tests consider several tumor components, such as DNA, RNA, proteins, and the tiny vesicles originating from tumor cells. Actually, liquid biopsy signifies the genetic alterations of tumors through nucleic acids or cells in various body fluids, including blood, urine, cerebrospinal fluid, or saliva in a noninvasive manner. In this review, we present an overall description of liquid biopsy in which circulating tumor cells, cell-free nucleic acids, exosomes, and extrachromosomal circular DNA are included.
Collapse
Affiliation(s)
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ebrahimi
- Department of Internal Medicine, Faculty of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran Iran
| | - Fatemeh Khatami
- Urology Research Center (URC), Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|